Interspecies Variation Between Fish and Human Transthyretins in

Sep 18, 2018 - Here we explored the interspecies variation in TDC binding to human and fish TTR (exemplified by Gilthead seabream (Sparus aurata)). Th...
0 downloads 0 Views 1MB Size
Subscriber access provided by University of Sunderland

Ecotoxicology and Human Environmental Health

Interspecies Variation Between Fish and Human Transthyretins in Their Binding of Thyroid-Disrupting Chemicals Jin Zhang, Christin Grundström, Kristoffer Brannstrom, Irina Iakovleva, Mikael Lindberg, Anders Olofsson, Patrik L. Andersson, and A. Elisabeth Sauer-Eriksson Environ. Sci. Technol., Just Accepted Manuscript • DOI: 10.1021/acs.est.8b03581 • Publication Date (Web): 18 Sep 2018 Downloaded from http://pubs.acs.org on September 18, 2018

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 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 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.

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 31

Environmental Science & Technology

1

Interspecies Variation Between Fish and Human

2

Transthyretins in Their Binding of Thyroid-

3

Disrupting Chemicals

4

Jin Zhang†, Christin Grundström†, Kristoffer Brännström‡, Irina Iakovleva†, Mikael Lindberg†,

5

Anders Olofsson‡, Patrik L. Andersson†*, A. Elisabeth Sauer-Eriksson†*

6

†Department of Chemistry, Umeå University, SE-90187 Umeå, Sweden

7

‡Department of Medical Biochemistry and Biophysics, Umeå University, SE-90187 Umeå,

8

Sweden

9

Corresponding Authors

10

*Email: [email protected]. Phone: +46-(0)907865266

11

*Email: [email protected]. Phone: +46-(0)907865923

12

ACS Paragon Plus Environment

1

Environmental Science & Technology

Page 2 of 31

13

ABSTRACT

14

Thyroid-disrupting chemicals (TDCs) are xenobiotics that can interfere with the endocrine

15

system and cause adverse effects in organisms and their offspring. TDCs affect both the thyroid

16

gland and regulatory enzymes associated with thyroid hormone homeostasis. Transthyretin

17

(TTR) is found in serum and cerebrospinal fluid of vertebrates, where it transports thyroid

18

hormones. Here we explored the interspecies variation in TDC binding to human and fish TTR

19

(exemplified by Gilthead seabream (Sparus aurata)). The in vitro binding experiments showed

20

that TDCs bind with equal or weaker affinity to seabream TTR than to the human TTR,

21

particular, the polar TDCs (>500-fold lower affinity). Crystal structures of seabream TTR–TDC

22

complexes revealed that all TDCs bound at the thyroid binding sites. However, amino acid

23

substitution of Ser117 in human TTR to Thr117 in seabream prevented polar TDCs from binding

24

deep in the hormone binding cavity, which explains their low affinity to seabream TTR.

25

Molecular dynamics and in silico alanine scanning simulation also suggested that the protein

26

backbone of seabream TTR is more rigid than the human one and that Thr117 provides fewer

27

electrostatic contributions than Ser117 to ligand bindings. This provides an explanation for the

28

weaker affinities of the ligands that rely on electrostatic interactions with Thr117. The lower

29

affinities of TDCs to fish TTR, in particular the polar ones, could potentially lead to milder

30

thyroid-related effects in fish.

31 32 33 34 35

ACS Paragon Plus Environment

2

Page 3 of 31

Environmental Science & Technology

36 37

INTRODUCTION

38

Thyroid-disrupting chemicals (TDCs) are environmental xenobiotics that may induce significant

39

disorders in physiological processes in humans and wildlife, including macronutrient

40

metabolism, energy balance, brain development, and reproduction.1-3 They can alter the structure

41

or function of the thyroid gland, interfere with thyroid hormone signaling, and subsequently

42

disrupt its homeostasis.4 TDCs have multiple molecular mechanisms. For example, hydroxylated

43

polychlorinated biphenyls (OH-PCBs) bind to and disturb thyroid hormone receptors and

44

transthyretin,5-7 and polyhalogenated aromatic hydrocarbons accelerate the clearance of thyroid

45

hormones by inducing uridine diphosphoglucuronosyl transferases.8 Even low exposures to

46

TDCs are cause of concern as these can trigger non-monotonic dose-response relationships

47

resulting in severe adverse effects.9-12 In coastal regions with high fish consumption TDC levels

48

have been associated with increased incidence of thyroid cancer and thyroid autoimmune

49

disease.13, 14

50

Transthyretin (TTR) is a serum transport protein that delivers 3,3',5-triiodo-L-thyronine (T3)

51

and 3,3',5,5'-tetraiodo-L-thyronine (thyroxine T4) from the thyroid gland to target tissues in

52

vertebrates.15 TTR is a molecular transporter of TDCs that include per- and polyfluoroalkyl

53

substances (PFASs)16 and OH-PCBs.17 TDCs in the thyroxine-binding sites (TBSs) of TTR can

54

disrupt TTR-mediated thyroid hormone transport.18 The subsequent accumulation of TDCs in

55

vital organs19 by TTR-assisted delivery, may cause developmental disorders.20

56

The three-dimensional structure of transthyretin shows it to be a homotetramer with a central

57

hydrophobic channel in which two TBSs are situated.21, 22 Over 200 human TTR (hTTR) crystal

58

structures have been deposited in the Protein Data Bank (PDB).23,

24

Many of these are

ACS Paragon Plus Environment

3

Environmental Science & Technology

Page 4 of 31

59

complexes, of which some are complexed with environmental pollutants or their metabolites,

60

including

61

tetrahydroxybenzophenone (BP2), perfluorooctanoic acid (PFOA), perfluorooctanesulfonic acid

62

(PFOS), tetrabromobisphenol A (TBBPA), and hydroxylated polychlorinated biphenyls (OH-

63

PCBs)18, 25, 26, which are of relevance for this study.

hTTR

complexes

with

4,4'-sulfonyldiphenol

(bisphenol

S),

2,2’,4,4’-

64

TTR transports TH via the choroid plexus into the brain in fish27 and it is expressed in the

65

liver, brain, eye, and heart of fish larvae from the first day post-hatch.28 Data on TTR-TDC

66

complexes from fish species are scarce despite their importance in toxicity studies.29, 30 Gilthead

67

seabream (Sparus aurata) is an indicator species for the estimation of environmental pollutant

68

levels in aquatic environments.31 It is one of the most consumed fish species and it is widely

69

distributed in the Mediterranean sea and the eastern Atlantic ocean.32 Several classical TDCs

70

have been detected in seabream in concentrations ranging from 36.2 pg/g to 141 ng/g wet

71

weight, e.g. polychlorinated dibenzo-p-dioxins, dibenzofurans, PCBs, and polybrominated

72

diphenyl ethers (PBDEs).33 TDCs can disturb thyroid hormone homeostasis in seabream via

73

binding to TTR,34 the primary thyroid hormone transporter; this may cause underdevelopment of

74

the juveniles.35 More importantly, TDCs can accumulate in aquatic ecosystems and thereby

75

expose humans to high levels of TDCs.36

76

TTRs from fish have low sequence identity with hTTR.28, 37 Seabream TTR (SaTTR) has 54%

77

sequence identity with hTTR,28 but the only amino acid difference within the TBS is the

78

conservative substitution of Ser117 in hTTR to Thr117 in SaTTR.38 Crystal structures of wild-

79

type SaTTR38, 39, and SaTTR in complex with T3 and T4 are available.38 However, additional

80

crystal structures of the SaTTR protein in complex with diverse environmental pollutants would

81

improve our understanding of TDC interaction with SaTTR; interspecies variations of the TTR-

ACS Paragon Plus Environment

4

Page 5 of 31

Environmental Science & Technology

82

TDC molecular interactions; and a comparison of the chemical-induced thyroid disruptions in

83

fish vs. that in humans.

84

Six representative TDCs were selected for this study based on their use in consumer goods and

85

products, their structural diversity, and the environmental relevance.16, 25, 40, 41 The compounds

86

included TBBPA, BP2, PFOA, 3,5,6-trichloro-2-pyridinol (TC2P), 2,4,5-trichlorophenoxyacetic

87

acid (2,4,5-T), and 2-(3-chloro-2-methylanilino)pyridine-3-carboxylic acid (clonixin).16,

88

TBBPA is widely used as a flame retardant in electronics. It has been detected in the serum of IT

89

technicians and electronics recycling workers (500-fold, Table 1)

249

The TTR-TC2P complex structures are of particular interest. Two TC2P molecules bind

250

simultaneously within the TBSs in the hTTR-TC2P complex, i.e., one molecule forms a

251

hydrogen bond with Ser117 at the cavity bottom, and the second molecule forms a hydrogen

252

bond with the Lys15 at the cavity entrance (Figure 3e). This binding mode is similar to that of

253

2,6-dinitro-p-cresol (DNPC) in the hTTR,25 which is probably associated with their relatively

254

small molecular sizes and their polar nature. In SaTTR, TC2P only binds in the vicinity of the

255

side chain of Lys15 at the cavity entrance. The interspecies differences in binding modes are

256

consistent with the 10-fold weaker binding affinity of TC2P to SaTTR (Table 1).

257

Molecular dynamics simulations for TTR-TDCs interaction studies. We investigated the

258

TTR-TDCs molecular interactions by performing molecular dynamics simulations on the human

259

and seabream structures in complex with each of the six TDCs.25, 26 All co-ligands remained

260

bound in the thyroid-binding sites, and the root-mean-squared-deviations of the protein and

261

ligands were under 2 and 3 Å (except for TC2P in SaTTR) throughout the simulations (Figure

262

S2). The ligand binding free energies were calculated using the MM-PBSA method based on the

263

last 10 ns of the molecular dynamics trajectory.

264

We compared the calculated ligand binding free energy both intra- and interspecies. The

265

calculated binding free energies and the experimental binding free energy (∆G) are moderately

ACS Paragon Plus Environment

12

Page 13 of 31

Environmental Science & Technology

266

correlated yielding R2=0.56 for SaTTR and R2=0.62 for hTTR (Figure S3). This correlation

267

suggests that MM-PBSA could be used for estimating the ligand binding free energy of TDCs to

268

TTR and as a scoring function to identify novel TDCs and study their molecular interactions.67

269

The mean value (-24.4 kcal/mol) of calculated binding free energies of the six TDCs to SaTTR is

270

lower than those (-26.2 kcal/mol) for the hTTR (Figure S3), which agrees with the weaker in

271

vitro affinities of the ligands (except PFOA) to SaTTR compared to hTTR.

272

We tried to elucidate the contributions of residue 117 to the binding free energies by

273

performing in silico alanine scanning simulations on the residue in the TTR-TDC complexes.

274

TTR-PFOA and TTR-BP2 complexes from the two species were chosen as representatives for

275

the simulations, as they have the smallest and largest interspecies variations in in vitro ligand

276

binding affinities, respectively.

277

The in silico alanine scanning on TTR-PFOA complexes (Figure S4) showed that the

278

calculated binding free energy of PFOA to hTTR and SaTTR both became slightly stronger, at a

279

similar scale, when residue 117 was mutated to alanine. The slight increase might be due to the

280

improvement of the existing hydrophobic interactions between the fluorinated tail of PFOA and

281

the hydrophobic cavity of TTR.25 The similarity in scale increase may indicate that Thr117 in

282

SaTTR and Ser117 in hTTR provide a similar contribution to the binding of PFOA; this could

283

explain the interspecies similarities in in vitro ligand binding free energies. In silico alanine

284

scanning simulations of TTR-BP2 complexes resulted in clear interspecies variations in

285

mutation-caused shifts of the calculated ligand binding free energies (Figure S4). In the hTTR-

286

BP2 complex, we observed a significant decrease (4 kcal/mol) in calculated binding free energy

287

of the ligand to mutated hTTR, since mutation of Ser117 causes loss of important electrostatic

288

interactions for hTTR-BP2 recognition.25 In the SaTTR-BP2 complex, the mutation posed a

ACS Paragon Plus Environment

13

Environmental Science & Technology

Page 14 of 31

289

limited impact on the calculated binding free energy of BP2 to SaTTR, as Thr117 provides fewer

290

electrostatic contributions to the ligand binding due to the locked rotamer of its side chain.

291

Interspecies variations in mutation-caused shifts of calculated binding free energy could explain

292

the significantly weaker in vitro binding free energies of BP2 to SaTTR compared to hTTR

293

(Table 1). This supports our conclusions from the crystal structure analysis that Thr117 provides

294

fewer electrostatic contributions and that SaTTR has a clear preference to hydrophobic ligands.

295 296

DISCUSSION

297

In the present study, we aimed to elucidate the interspecies variations in human and seabream

298

TTR-TDC interactions using crystallographic, in vitro, and in silico data. The six TDCs studied

299

here have been detected in humans, wildlife, and the environment.

300

In vitro isothermal titration calorimetry binding experiments showed that all six TDCs bound

301

to SaTTR and hTTR. TBBPA was the strongest TTR binder, with nM affinity to hTTR.26, 68 With

302

the exception of PFOA, the TDC binding affinities to hTTR were significantly stronger than to

303

SaTTR. The sequence of hTTR and SaTTR differs at the thyroid-binding site, where it is Ser117

304

for hTTR and Thr117 for SaTTR. Serine and threonine are both slightly polar amino acids with a

305

hydroxyl group at the Cβ position, with the only difference between being that threonine has a

306

methyl group in place of the hydrogen group found in serine. A serine-to-threonine substitution

307

could therefore be considered as a fairly neutral substitution. However, the structural comparison

308

of the thyroid-binding sites in hTTR and SaTTR showed that the substitution has large functional

309

consequences for the two proteins. The side chain of residue 117 is situated at both the

310

monomer-monomer and the dimer-dimer interfaces. Superimposing all TTR-TDC complexes

311

shows that Ser117 in hTTR has high rotational freedom and that its site chain can occupy all

ACS Paragon Plus Environment

14

Page 15 of 31

Environmental Science & Technology

312

three possible rotamers (g+, t, and g). In contrast, the Thr117 in SaTTR is rigid and can only

313

occupy one rotamer, otherwise steric clashes will occur between the Cγ2 atoms of the rotamers

314

(Figure S5). Consequently, the mobility of Ser117 allows hTTR to make good binding

315

interactions with both polar and hydrophobic ligands, whereas SaTTR has a clear preference for

316

hydrophobic ones. As a consequence, larger ligands, such as T4, BP2 and clonixin, cannot reach

317

as deep into the thyroid-binding cavity of SaTTR as they can in hTTR, due to steric clashes with

318

the Cγ2 atom of Thr117.

319

In addition to this constraint, the rotamer of Lys15, positioned at entrance of the hormone-

320

binding channel, is also locked in SaTTR (Figure S5). Its ε-ammonium group forms a hydrogen

321

bond with the main chain carbonyl oxygen of Thr52 and a salt bridge with the side chain of

322

Glu54. These contacts are present in all monomers of all SaTTR structures solved to date. In

323

hTTR, however, the side chain of Lys15 has more rotational freedom (Figure S5). Inspection of

324

the hydrophobic core positioned directly underneath Lys15 suggests that the amino acid

325

substitutions Val16Ile and Ile73Phe in SaTTR shift the position of the β-strand A (residues

326

Leu12-Ala19) by up to 1 Å, which explains the different properties of the hTTR and SaTTR

327

Lys15 side chains. The significantly wider entrance of the hormone-binding channel in SaTTR,

328

in combination with its narrower cavity, provides an additional structural explanation for the

329

different binding affinities of human and seabream TTR to TDCs.38

330

The finding that Thr117 in SaTTR prevents high affinity binding to polar ligands could be

331

generalized to others piscine species, e.g. zebrafish (Danio rerio), Atlantic salmon (Salmo salar),

332

Rainbow trout (Oncorhynchus mykiss), and Channel catfish (Ictalurus punctatus). Like

333

seabream, they share high similarity in their thyroid-binding site residues and the same Thr117

ACS Paragon Plus Environment

15

Environmental Science & Technology

Page 16 of 31

334

residues.69 Here in silico molecular dynamics simulations could be used with advantage to

335

predict potential TDC binders to other fish species TTRs.

336

Of the six TDCs used here, BP2 was the most polar. We therefore used isothermal titration

337

calorimetry to determine binding affinities of three additional polar compounds to SaTTR. These

338

were two thyroid disrupting compounds 2,6-dinitro-p-cresol (DNPC) and bisphenol S (BPS), and

339

the flavonoid luteolin (Figure S6a). The binding affinities (Kd) and binding interactions to hTTR

340

are known: DNPC 1.3 µM, PDB code 5L4F; BPS 52 µM, PDB code 5L4J; and luteolin 0.07 µM

341

PDB code 4QXV.25, 70 These have 34-fold, 1.5-fold, and 250-fold higher affinities, respectively,

342

to hTTR than SaTTR. A close look at the hTTR-DNPC, hTTR-PBS, and hTTR-luteolin complex

343

structures explains the large variation in binding affinities of the three polar compounds to

344

SaTTR, in particular to the 250-fold higher affinity of luteolin to hTTR (Figure S6b). In

345

summary, isothermal titration calorimetry data from the three polar compounds support our

346

observation that Thr117 prevents high-affinity binding of polar ligands to SaTTR, and

347

presumably to other fish species that have a threonine at position 117. The ability of small,

348

strong binders adapting to multiple conformations in the thyroid-binding sites can explain the

349

interactions between TTR and such ligands, and provide guidance for molecular design and

350

hazard identification. Our results also highlight the difficulty in extrapolating data from one

351

species to another. Without structural data it would be difficult, if not impossible, to explain the

352

differences in the binding affinity data.

353 354

ENVIRONMENTAL IMPLICATIONS

355

The thyroid hormone system is vital for early development, metabolism, and growth in wildlife

356

and humans. In humans, thyroxine can be transported in the blood by albumin, thyroxine binding

ACS Paragon Plus Environment

16

Page 17 of 31

Environmental Science & Technology

357

globulin and TTR, whereas in fish TTR is the primary thyroid hormone transporter.71 It is well-

358

known that a large number of environmental pollutants bind to TTR and a few of these are even

359

stronger binders than the natural hormone thyroxine.41, 68, 72-74 This specific binding of organic

360

contaminants to TTR pose a risk of transport across biological barriers, e.g. the blood brain

361

barrier and placenta, and tissue specific accumulation, that could induce adverse effects in

362

vulnerable organs or developing organisms. Currently, no specific biomarker for thyroid

363

hormone disruption has been generally agreed upon even if effects on hormonal levels, gene

364

expression and histology are often being used.1 Binding to TTR is one potential marker of TDCs

365

which could be designed for high throughput screening.75 This study described on a molecular

366

level potential variation in interaction at the TTR binding site that that could cause species

367

variation in read-out from toxicity assays. In particular, these variations are expected for small

368

and polar chemicals. In assessing environmental and human health risks of industrial chemicals

369

and development of drug candidates, fish based assays is frequently being used motivated by the

370

high number of orthologs for human drug targets in e.g. zebrafish.76 Here we studied, seabream

371

but the amino acid sequence in the binding pocket of TTR is well conserved for several fish

372

species including zebrafish and rainbow trout which are frequently being used for toxicity tests.

373

Findings presented here may thus have an impact on data from fish tests for in particular small

374

and polar chemicals that could result in underestimating risks when extrapolated to the human

375

situation.

376 377

SUPPORTING INFORMATION

378

This information is available free of charge via the Internet at http://pubs.acs.org.

ACS Paragon Plus Environment

17

Environmental Science & Technology

Page 18 of 31

379 380

Details of the X-ray structures, in vitro isothermal titration calorimetry experiments; procedures

381

and results of the molecular dynamics simulations, MM-PBSA calculations and alanine scanning

382

simulations are in the supporting information.

383

ACKNOWLEDGEMENT

384

This study was financed by the MiSSE project through grants from the Swedish Research

385

Council for the Environment, Agricultural Sciences and Spatial Planning (Formas) (210-2012-

386

131) and by the Swedish Research Council (521-2011-6427 and 2015-03607). We also thank the

387

staff of the Protein Expertise Platform at Umeå University for cloning services. The molecular

388

dynamics simulations were conducted at the High Performance Computing Center North.

389

REFERENCES

390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411

1. Murk, A. J.; Rijntjes, E.; Blaauboer, B. J.; Clewell, R.; Crofton, K. M.; Dingemans, M. M.; Furlow, J. D.; Kavlock, R.; Kohrle, J.; Opitz, R.; Traas, T.; Visser, T. J.; Xia, M.; Gutleb, A. C., Mechanism-based testing strategy using in vitro approaches for identification of thyroid hormone disrupting chemicals. Toxicology in vitro : an international journal published in association with BIBRA 2013, 27, (4), 1320-46. 2. Jenssen, B. M., Endocrine-disrupting chemicals and climate change: A worst-case combination for arctic marine mammals and seabirds? Environmental health perspectives 2006, 114 Suppl 1, 76-80. 3. Rhind, S. M., Anthropogenic pollutants: a threat to ecosystem sustainability? Philosophical transactions of the Royal Society of London. Series B, Biological sciences 2009, 364, (1534), 3391-401. 4. Crofton, K. M.; Craft, E. S.; Hedge, J. M.; Gennings, C.; Simmons, J. E.; Carchman, R. A.; Carter, W. H., Jr.; DeVito, M. J., Thyroid-hormone-disrupting chemicals: evidence for dosedependent additivity or synergism. Environmental health perspectives 2005, 113, (11), 1549-54. 5. Meerts, I. A.; Assink, Y.; Cenijn, P. H.; Van Den Berg, J. H.; Weijers, B. M.; Bergman, A.; Koeman, J. H.; Brouwer, A., Placental transfer of a hydroxylated polychlorinated biphenyl and effects on fetal and maternal thyroid hormone homeostasis in the rat. Toxicological sciences : an official journal of the Society of Toxicology 2002, 68, (2), 361-71. 6. Freitas, J.; Cano, P.; Craig-Veit, C.; Goodson, M. L.; Furlow, J. D.; Murk, A. J., Detection of thyroid hormone receptor disruptors by a novel stable in vitro reporter gene assay. Toxicology in vitro : an international journal published in association with BIBRA 2011, 25, (1), 257-66.

ACS Paragon Plus Environment

18

Page 19 of 31

412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456

Environmental Science & Technology

7. Ishihara, A.; Nishiyama, N.; Sugiyama, S.; Yamauchi, K., The effect of endocrine disrupting chemicals on thyroid hormone binding to Japanese quail transthyretin and thyroid hormone receptor. General and comparative endocrinology 2003, 134, (1), 36-43. 8. Builee, T. L.; Hatherill, J. R., The role of polyhalogenated aromatic hydrocarbons on thyroid hormone disruption and cognitive function: a review. Drug and chemical toxicology 2004, 27, (4), 405-24. 9. Do, R. P.; Stahlhut, R. W.; Ponzi, D.; Vom Saal, F. S.; Taylor, J. A., Non-monotonic dose effects of in utero exposure to di(2-ethylhexyl) phthalate (DEHP) on testicular and serum testosterone and anogenital distance in male mouse fetuses. Reproductive toxicology 2012, 34, (4), 614-21. 10. Jenkins, S.; Wang, J.; Eltoum, I.; Desmond, R.; Lamartiniere, C. A., Chronic oral exposure to bisphenol A results in a nonmonotonic dose response in mammary carcinogenesis and metastasis in MMTV-erbB2 mice. Environmental health perspectives 2011, 119, (11), 16049. 11. Hayes, T. B.; Collins, A.; Lee, M.; Mendoza, M.; Noriega, N.; Stuart, A. A.; Vonk, A., Hermaphroditic, demasculinized frogs after exposure to the herbicide atrazine at low ecologically relevant doses. Proceedings of the National Academy of Sciences of the United States of America 2002, 99, (8), 5476-80. 12. Vandenberg, L. N.; Colborn, T.; Hayes, T. B.; Heindel, J. J.; Jacobs, D. R., Jr.; Lee, D. H.; Shioda, T.; Soto, A. M.; vom Saal, F. S.; Welshons, W. V.; Zoeller, R. T.; Myers, J. P., Hormones and endocrine-disrupting chemicals: low-dose effects and nonmonotonic dose responses. Endocrine reviews 2012, 33, (3), 378-455. 13. Patrick, L., Thyroid disruption: mechanism and clinical implications in human health. Alternative medicine review : a journal of clinical therapeutic 2009, 14, (4), 326-46. 14. Schnitzler, J. G.; Koutrakis, E.; Siebert, U.; Thome, J. P.; Das, K., Effects of persistent organic pollutants on the thyroid function of the European sea bass (Dicentrarchus labrax) from the Aegean sea, is it an endocrine disruption? Marine pollution bulletin 2008, 56, (10), 1755-64. 15. Bartalena, L.; Robbins, J., Thyroid hormone transport proteins. Clinics in laboratory medicine 1993, 13, (3), 583-98. 16. Weiss, J. M.; Andersson, P. L.; Lamoree, M. H.; Leonards, P. E.; van Leeuwen, S. P.; Hamers, T., Competitive binding of poly- and perfluorinated compounds to the thyroid hormone transport protein transthyretin. Toxicological sciences : an official journal of the Society of Toxicology 2009, 109, (2), 206-16. 17. Cheek, A. O.; Kow, K.; Chen, J.; McLachlan, J. A., Potential mechanisms of thyroid disruption in humans: interaction of organochlorine compounds with thyroid receptor, transthyretin, and thyroid-binding globulin. Environmental health perspectives 1999, 107, (4), 273-8. 18. Purkey, H. E.; Palaninathan, S. K.; Kent, K. C.; Smith, C.; Safe, S. H.; Sacchettini, J. C.; Kelly, J. W., Hydroxylated polychlorinated biphenyls selectively bind transthyretin in blood and inhibit amyloidogenesis: rationalizing rodent PCB toxicity. Chemistry & biology 2004, 11, (12), 1719-28. 19. Boas, M.; Feldt-Rasmussen, U.; Main, K. M., Thyroid effects of endocrine disrupting chemicals. Mol Cell Endocrinol 2012, 355, (2), 240-248. 20. Porterfield, S. P., Thyroidal dysfunction and environmental chemicals--potential impact on brain development. Environmental health perspectives 2000, 108 Suppl 3, 433-8.

ACS Paragon Plus Environment

19

Environmental Science & Technology

457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501

Page 20 of 31

21. Hamilton, J. A.; Steinrauf, L. K.; Braden, B. C.; Liepnieks, J.; Benson, M. D.; Holmgren, G.; Sandgren, O.; Steen, L., The x-ray crystal structure refinements of normal human transthyretin and the amyloidogenic Val-30-->Met variant to 1.7-A resolution. The Journal of biological chemistry 1993, 268, (4), 2416-24. 22. Hornberg, A.; Eneqvist, T.; Olofsson, A.; Lundgren, E.; Sauer-Eriksson, A. E., A comparative analysis of 23 structures of the amyloidogenic protein transthyretin. Journal of molecular biology 2000, 302, (3), 649-69. 23. RCSB-PDB, RCSB Protein Data Bank (PDB). http://www.rcsb.org/pdb/ 2017. 24. Palaninathan, S. K., Nearly 200 X-ray crystal structures of transthyretin: what do they tell us about this protein and the design of drugs for TTR amyloidoses? Current medicinal chemistry 2012, 19, (15), 2324-42. 25. Zhang, J.; Begum, A.; Brannstrom, K.; Grundstrom, C.; Iakovleva, I.; Olofsson, A.; Sauer-Eriksson, A. E.; Andersson, P. L., Structure-Based Virtual Screening Protocol for in Silico Identification of Potential Thyroid Disrupting Chemicals Targeting Transthyretin. Environmental science & technology 2016, 50, (21), 11984-11993. 26. Iakovleva, I.; Begum, A.; Brannstrom, K.; Wijsekera, A.; Nilsson, L.; Zhang, J.; Andersson, P. L.; Sauer-Eriksson, A. E.; Olofsson, A., Tetrabromobisphenol A Is an Efficient Stabilizer of the Transthyretin Tetramer. PloS one 2016, 11, (4), e0153529. 27. Richardson, S. J.; Wijayagunaratne, R. C.; D'Souza, D. G.; Darras, V. M.; Van Herck, S. L., Transport of thyroid hormones via the choroid plexus into the brain: the roles of transthyretin and thyroid hormone transmembrane transporters. Front. Neurosci. 2015, 9, 66. 28. Santos, C. R.; Power, D. M., Identification of transthyretin in fish (Sparus aurata): cDNA cloning and characterisation. Endocrinology 1999, 140, (5), 2430-3. 29. Abalos, M.; Abad, E.; Estevez, A.; Sole, M.; Buet, A.; Quiros, L.; Pina, B.; Rivera, J., Effects on growth and biochemical responses in juvenile gilthead seabream 'Sparus aurata' after long-term dietary exposure to low levels of dioxins. Chemosphere 2008, 73, (1 Suppl), S303-10. 30. Abalos, M.; Parera, J.; Estevez, A.; Sole, M.; Fabregat, M. C.; Pina, B.; Navarro, A.; Abad, E., Decontamination trends in the aquacultured fish gilthead seabream (Sparus aurata) after feeding long-term a PCDD/F spiked feed. Chemosphere 2011, 82, (1), 64-71. 31. Serrano, R.; Barreda, M.; Blanes, M. A., Investigating the presence of organochlorine pesticides and polychlorinated biphenyls in wild and farmed gilthead sea bream (Sparus aurata) from the Western Mediterranean sea. Marine pollution bulletin 2008, 56, (5), 963-72. 32. Sola, L.; Moretti, A.; Crosetti, D.; Karaiskou, N.; Magoulas, A.; Rossi, A.; Rye, M.; Triantafyllidis, A.; Tsigenopoulos, C. In Gilthead seabream—Sparus aurata, Proceedings of the WP1 workshop on Genetics of domestication, breeding and enhancement of performance of fish and shellfish, Viterbo, Italy, 2006; 2006; pp 12-17. 33. Parera, J.; Abalos, M.; Santos, F. J.; Galceran, M. T.; Abad, E., Polychlorinated dibenzop-dioxins, dibenzofurans, biphenyls, paraffins and polybrominated diphenyl ethers in marine fish species from Ebro River Delta (Spain). Chemosphere 2013, 93, (3), 499-505. 34. Morgado, I.; Hamers, T.; Van der Ven, L.; Power, D. M., Disruption of thyroid hormone binding to sea bream recombinant transthyretin by ioxinyl and polybrominated diphenyl ethers. Chemosphere 2007, 69, (1), 155-63. 35. Ribecco, C.; Baker, M. E.; Sasik, R.; Zuo, Y.; Hardiman, G.; Carnevali, O., Biological effects of marine contaminated sediments on Sparus aurata juveniles. Aquatic toxicology 2011, 104, (3-4), 308-16.

ACS Paragon Plus Environment

20

Page 21 of 31

502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547

Environmental Science & Technology

36. Costopoulou, D.; Vassiliadou, I.; Leondiadis, L., PCDDs, PCDFs and PCBs in farmed fish produced in Greece: Levels and human population exposure assessment. Chemosphere 2016, 146, 511-8. 37. Yamauchi, K.; Nakajima, J.; Hayashi, H.; Hara, A., Purification and characterization of thyroid-hormone-binding protein from masu salmon serum. A homolog of higher-vertebrate transthyretin. European journal of biochemistry 1999, 265, (3), 944-9. 38. Eneqvist, T.; Lundberg, E.; Karlsson, A.; Huang, S.; Santos, C. R.; Power, D. M.; SauerEriksson, A. E., High resolution crystal structures of piscine transthyretin reveal different binding modes for triiodothyronine and thyroxine. The Journal of biological chemistry 2004, 279, (25), 26411-6. 39. Folli, C.; Pasquato, N.; Ramazzina, I.; Battistutta, R.; Zanotti, G.; Berni, R., Distinctive binding and structural properties of piscine transthyretin. FEBS letters 2003, 555, (2), 279-84. 40. Zhang, J.; Kamstra, J. H.; Ghorbanzadeh, M.; Weiss, J. M.; Hamers, T.; Andersson, P. L., In Silico Approach To Identify Potential Thyroid Hormone Disruptors among Currently Known Dust Contaminants and Their Metabolites. Environmental science & technology 2015, 49, (16), 10099-107. 41. Hamers, T.; Kamstra, J. H.; Sonneveld, E.; Murk, A. J.; Kester, M. H.; Andersson, P. L.; Legler, J.; Brouwer, A., In vitro profiling of the endocrine-disrupting potency of brominated flame retardants. Toxicological sciences : an official journal of the Society of Toxicology 2006, 92, (1), 157-73. 42. Sjodin, A.; Patterson, D. G., Jr.; Bergman, A., A review on human exposure to brominated flame retardants--particularly polybrominated diphenyl ethers. Environment international 2003, 29, (6), 829-39. 43. Aznar-Alemany, O.; Trabalon, L.; Jacobs, S.; Barbosa, V. L.; Tejedor, M. F.; Granby, K.; Kwadijk, C.; Cunha, S. C.; Ferrari, F.; Vandermeersch, G.; Sioen, I.; Verbeke, W.; Vilavert, L.; Domingo, J. L.; Eljarrat, E.; Barcelo, D., Occurrence of halogenated flame retardants in commercial seafood species available in European markets. Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association 2017, 104, 35-47. 44. Wang, L.; Kannan, K., Characteristic profiles of benzonphenone-3 and its derivatives in urine of children and adults from the United States and China. Environmental science & technology 2013, 47, (21), 12532-8. 45. Schmutzler, C.; Bacinski, A.; Gotthardt, I.; Huhne, K.; Ambrugger, P.; Klammer, H.; Schlecht, C.; Hoang-Vu, C.; Gruters, A.; Wuttke, W.; Jarry, H.; Kohrle, J., The ultraviolet filter benzophenone 2 interferes with the thyroid hormone axis in rats and is a potent in vitro inhibitor of human recombinant thyroid peroxidase. Endocrinology 2007, 148, (6), 2835-44. 46. Karrman, A.; Ericson, I.; van Bavel, B.; Darnerud, P. O.; Aune, M.; Glynn, A.; Lignell, S.; Lindstrom, G., Exposure of perfluorinated chemicals through lactation: levels of matched human milk and serum and a temporal trend, 1996-2004, in Sweden. Environmental health perspectives 2007, 115, (2), 226-30. 47. Melzer, D.; Rice, N.; Depledge, M. H.; Henley, W. E.; Galloway, T. S., Association between serum perfluorooctanoic acid (PFOA) and thyroid disease in the U.S. National Health and Nutrition Examination Survey. Environmental health perspectives 2010, 118, (5), 686-92. 48. Jin, Y.; Saito, N.; Harada, K. H.; Inoue, K.; Koizumi, A., Historical trends in human serum levels of perfluorooctanoate and perfluorooctane sulfonate in Shenyang, China. The Tohoku journal of experimental medicine 2007, 212, (1), 63-70.

ACS Paragon Plus Environment

21

Environmental Science & Technology

548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592

Page 22 of 31

49. Fortenberry, G. Z.; Meeker, J. D.; Sanchez, B. N.; Barr, D. B.; Panuwet, P.; Bellinger, D.; Schnaas, L.; Solano-Gonzalez, M.; Ettinger, A. S.; Hernandez-Avila, M.; Hu, H.; Tellez-Rojo, M. M., Urinary 3,5,6-trichloro-2-pyridinol (TCPY) in pregnant women from Mexico City: distribution, temporal variability, and relationship with child attention and hyperactivity. International journal of hygiene and environmental health 2014, 217, (2-3), 405-12. 50. Fortenberry, G. Z.; Hu, H.; Turyk, M.; Barr, D. B.; Meeker, J. D., Association between urinary 3, 5, 6-trichloro-2-pyridinol, a metabolite of chlorpyrifos and chlorpyrifos-methyl, and serum T4 and TSH in NHANES 1999-2002. The Science of the total environment 2012, 424, 351-5. 51. Van den Berg, K. J.; van Raaij, J. A.; Bragt, P. C.; Notten, W. R., Interactions of halogenated industrial chemicals with transthyretin and effects on thyroid hormone levels in vivo. Archives of toxicology 1991, 65, (1), 15-9. 52. Sjöden, P.-O.; Söderberg, U., Long-lasting effects of prenatal 2, 4, 5trichlorophenoxyacetic acid on open-field behavior in rats: Pre- and postnatal mediation. Physiological Psychology 1975, 3, (2), 175-178. 53. Wojtczak, A.; Cody, V.; Luft, J. R.; Pangborn, W., Structures of human transthyretin complexed with thyroxine at 2.0 A resolution and 3',5'-dinitro-N-acetyl-L-thyronine at 2.2 A resolution. Acta crystallographica. Section D, Biological crystallography 1996, 52, (Pt 4), 75865. 54. Rice, P.; Longden, I.; Bleasby, A., EMBOSS: the European Molecular Biology Open Software Suite. Trends in genetics : TIG 2000, 16, (6), 276-7. 55. Schrödinger Multiple Sequence Viewer, Schrodinger, LLC, New York, NY., 2017. 56. Ferguson, R. N.; Edelhoch, H.; Saroff, H. A.; Robbins, J.; Cahnmann, H. J., Negative cooperativity in the binding of thyroxine to human serum prealbumin. Preparation of tritiumlabeled 8-anilino-1-naphthalenesulfonic acid. Biochemistry 1975, 14, (2), 282-9. 57. Cheng, S. Y.; Pages, R. A.; Saroff, H. A.; Edelhoch, H.; Robbins, J., Analysis of thyroid hormone binding to human serum prealbumin by 8-anilinonaphthalene-1-sulfonate fluorescence. Biochemistry 1977, 16, (16), 3707-13. 58. Kabsch, W., Xds. Acta crystallographica. Section D, Biological crystallography 2010, 66, (Pt 2), 125-32. 59. Winn, M. D.; Ballard, C. C.; Cowtan, K. D.; Dodson, E. J.; Emsley, P.; Evans, P. R.; Keegan, R. M.; Krissinel, E. B.; Leslie, A. G.; McCoy, A.; McNicholas, S. J.; Murshudov, G. N.; Pannu, N. S.; Potterton, E. A.; Powell, H. R.; Read, R. J.; Vagin, A.; Wilson, K. S., Overview of the CCP4 suite and current developments. Acta crystallographica. Section D, Biological crystallography 2011, 67, (Pt 4), 235-42. 60. McCoy, A. J.; Grosse-Kunstleve, R. W.; Adams, P. D.; Winn, M. D.; Storoni, L. C.; Read, R. J., Phaser crystallographic software. Journal of applied crystallography 2007, 40, (Pt 4), 658-674. 61. Adams, P. D.; Afonine, P. V.; Bunkoczi, G.; Chen, V. B.; Davis, I. W.; Echols, N.; Headd, J. J.; Hung, L. W.; Kapral, G. J.; Grosse-Kunstleve, R. W.; McCoy, A. J.; Moriarty, N. W.; Oeffner, R.; Read, R. J.; Richardson, D. C.; Richardson, J. S.; Terwilliger, T. C.; Zwart, P. H., PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta crystallographica. Section D, Biological crystallography 2010, 66, (Pt 2), 213-21. 62. Emsley, P.; Cowtan, K., Coot: model-building tools for molecular graphics. Acta crystallographica. Section D, Biological crystallography 2004, 60, (Pt 12 Pt 1), 2126-32.

ACS Paragon Plus Environment

22

Page 23 of 31

593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637

Environmental Science & Technology

63. McNicholas, S.; Potterton, E.; Wilson, K. S.; Noble, M. E., Presenting your structures: the CCP4mg molecular-graphics software. Acta crystallographica. Section D, Biological crystallography 2011, 67, (Pt 4), 386-94. 64. Case, D. A.; Darden, T. A.; Gusarov, S.; Kovalenko, A.; Kollman, P. A., Amber 2015 (AmberTools15 and Amber14), University of California, San Francisco. 2015. 65. Hou, T.; Wang, J.; Li, Y.; Wang, W., Assessing the performance of the MM/PBSA and MM/GBSA methods. 1. The accuracy of binding free energy calculations based on molecular dynamics simulations. Journal of chemical information and modeling 2011, 51, (1), 69-82. 66. Morgado, I.; Melo, E. P.; Lundberg, E.; Estrela, N. L.; Sauer-Eriksson, A. E.; Power, D. M., Hormone affinity and fibril formation of piscine transthyretin: the role of the N-terminal. Mol Cell Endocrinol 2008, 295, (1-2), 48-58. 67. Chen, Q.; Wang, X.; Shi, W.; Yu, H.; Zhang, X.; Giesy, J. P., Identification of Thyroid Hormone Disruptors among HO-PBDEs: In Vitro Investigations and Coregulator Involved Simulations. Environmental science & technology 2016, 50, (22), 12429-12438. 68. Meerts, I. A.; van Zanden, J. J.; Luijks, E. A.; van Leeuwen-Bol, I.; Marsh, G.; Jakobsson, E.; Bergman, A.; Brouwer, A., Potent competitive interactions of some brominated flame retardants and related compounds with human transthyretin in vitro. Toxicological sciences : an official journal of the Society of Toxicology 2000, 56, (1), 95-104. 69. Eneqvist, T.; Lundberg, E.; Nilsson, L.; Abagyan, R.; Sauer-Eriksson, A. E., The transthyretin-related protein family. European journal of biochemistry 2003, 270, (3), 518-32. 70. Iakovleva, I.; Begum, A.; Pokrzywa, M.; Walfridsson, M.; Sauer-Eriksson, A. E.; Olofsson, A., The flavonoid luteolin, but not luteolin-7-O-glucoside, prevents a transthyretin mediated toxic response. PloS one 2015, 10, (5), e0128222. 71. Power, D. M.; Llewellyn, L.; Faustino, M.; Nowell, M. A.; Bjornsson, B. T.; Einarsdottir, I. E.; Canario, A. V.; Sweeney, G. E., Thyroid hormones in growth and development of fish. Comparative biochemistry and physiology. Toxicology & pharmacology : CBP 2001, 130, (4), 447-59. 72. Ghosh, M.; Meerts, I. A.; Cook, A.; Bergman, A.; Brouwer, A.; Johnson, L. N., Structure of human transthyretin complexed with bromophenols: a new mode of binding. Acta crystallographica. Section D, Biological crystallography 2000, 56, (Pt 9), 1085-95. 73. Lans, M. C.; Klasson-Wehler, E.; Willemsen, M.; Meussen, E.; Safe, S.; Brouwer, A., Structure-dependent, competitive interaction of hydroxy-polychlorobiphenyls, -dibenzo-pdioxins and -dibenzofurans with human transthyretin. Chemico-biological interactions 1993, 88, (1), 7-21. 74. Weiss, J. M.; Andersson, P. L.; Zhang, J.; Simon, E.; Leonards, P. E.; Hamers, T.; Lamoree, M. H., Tracing thyroid hormone-disrupting compounds: database compilation and structure-activity evaluation for an effect-directed analysis of sediment. Analytical and bioanalytical chemistry 2015, 407, (19), 5625-34. 75. Montano, M.; Cocco, E.; Guignard, C.; Marsh, G.; Hoffmann, L.; Bergman, A.; Gutleb, A. C.; Murk, A. J., New approaches to assess the transthyretin binding capacity of bioactivated thyroid hormone disruptors. Toxicological sciences : an official journal of the Society of Toxicology 2012, 130, (1), 94-105. 76. Gunnarsson, L.; Jauhiainen, A.; Kristiansson, E.; Nerman, O.; Larsson, D. G., Evolutionary conservation of human drug targets in organisms used for environmental risk assessments. Environmental science & technology 2008, 42, (15), 5807-13.

ACS Paragon Plus Environment

23

Environmental Science & Technology

638 639 640 641 642 643 644

Page 24 of 31

77. Ucan-Marin, F.; Arukwe, A.; Mortensen, A.; Gabrielsen, G. W.; Fox, G. A.; Letcher, R. J., Recombinant transthyretin purification and competitive binding with organohalogen compounds in two gull species (Larus argentatus and Larus hyperboreus). Toxicol. Sci. 2009, 107, (2), 440-50. 78. Purkey, H. E.; Dorrell, M. I.; Kelly, J. W., Evaluating the binding selectivity of transthyretin amyloid fibril inhibitors in blood plasma. Proceedings of the National Academy of Sciences of the United States of America 2001, 98, (10), 5566-71.

645 646 647 648 649

ACS Paragon Plus Environment

24

Page 25 of 31

650

Environmental Science & Technology

GRAPHICAL ABSTRACT

651

652 653 654

ACS Paragon Plus Environment

25

Environmental Science & Technology

Page 26 of 31

655

Table 1. Studied compounds with abbreviations, chemical abstract services (CAS) registry

656

numbers, usage, and binding affinities (Kd) determined using isothermal titration calorimetry. Compounds (Abbreviations)

CAS number

Use

3,3',5,5'-tetraiodo-Lthyronine (T4)

51-48-9

Fish

Human

Ratio

Kd (µM)

Kd (µM)

Fish:Human Kd

Control

0.23a (0.01b66)

0.09a (0.015b77) (0.084b78)

~1:1-1:3

2,2',4,4'131-55-5 tetrahydroxybenzophe none (BP2)

UV absorber

54.61

0.10

1:500

2-(3-chloro-217737-65-4 methylanilino)pyridin e-3-carboxylic acid (Clonixin)

Pharmaceutical

3.61

0.52

1:7

335-67-1

Surfactant

2.16

2.19

1:1

2,2',6,6'tetrabromobisphenol A (TBBPA)

79-94-7

Flame retardant

0.18

0.02

1:9

3,5,6-Trichloro-2pyridinol (TC2P)

6515-38-4

Herbicide

28.41

3.34

1:9

2,4,5trichlorophenoxyaceti c acid (2,4,5-T)

93-76-5

Herbicide

10.20

3.19

1:3

pentadecafluorooctan oic acid (PFOA)

657 658

a

The binding affinity of T4 is only an approximate value due to its thermoinstability and poor solubility in the isothermal titration calorimetry buffer. bKd values reported in literatures.

659

ACS Paragon Plus Environment

26

Page 27 of 31

660

Environmental Science & Technology

FIGURE LEGENDS

661 662

Figure 1. Structural comparison of SaTTR (PDB code: 1SN0) and hTTR (PDB code: 2ROX). (a)

663

Ribbon drawing of the superimposed tetrameric structures, SaTTR in green and hTTR in gray.

664

SaTTR has a tetramer in the asymmetric unit; these are labelled A, B, C and D. hTTR has a

665

dimer in the asymmetric unit of the crystal; these are labelled A and B, and the symmetry-related

666

monomers in the hTTR tetramer are consequently labelled A’ and B’. T4 bound at the two

667

thyroxine binding sites are shown as sticks. (b) Close-up view of the thyroid-binding sites of

668

monomers B and B´ in hTTR and monomers B and D in SaTTR. Note that T4 in hTTR

669

penetrates deeper into the thyroid-binding site than the T4 in SaTTR. c) Global pair-wise

670

sequence alignment of SaTTR and hTTR. Residues constituting the thyroid-binding site are

671

highlighted in red.

672

Figure 2. Compound binding in the thyroid-binding site formed by monomers B and D in SaTTR

673

and monomers B and B´ in hTTR; (a) SaTTR-BP2, (b) SaTTR-clonixin (c) SaTTR-PFOA (d)

674

SaTTR-TBBPA (e) SaTTR-TC2P, (f) hTTR-TC2P, (g) SaTTR-2,4,5-T, (h) hTTR-2,4,5-T. The

675

refined (2m|Fo|−D|Fc|) electron density is shown in gray mesh at 1σ contour level. Water

676

molecules around the ligands are shown as red spheres.

677

Figure 3. Superimposed structures of SaTTR (green) and hTTR (gray) in complex with: (a) BP2

678

(hTTR-BP2, PDB code: 5JIQ), (b) clonixin (hTTR-clonixin, PDB code: 5L4I), (c) PFOA

679

(hTTR-PFOA, PDB code: 5JID), (d) TBBPA (hTTR-clonixin, PDB code: 5HJG), (e) TC2P, and

680

(f) 2,4,5-T. Selected water molecules are shown as red spheres. Hydrogen bonds are shown as

681

dashed lines.

ACS Paragon Plus Environment

27

Environmental Science & Technology

Figure1 349x240mm (96 x 96 DPI)

ACS Paragon Plus Environment

Page 28 of 31

Page 29 of 31

Environmental Science & Technology

Figure2 445x260mm (96 x 96 DPI)

ACS Paragon Plus Environment

Environmental Science & Technology

Figure3 372x293mm (96 x 96 DPI)

ACS Paragon Plus Environment

Page 30 of 31

Page 31 of 31

Environmental Science & Technology

TOC 202x83mm (96 x 96 DPI)

ACS Paragon Plus Environment