Umeå University's logo

umu.sePublications
Change search
CiteExportLink to record
Permanent link

Direct link
Cite
Citation style
  • apa
  • ieee
  • modern-language-association-8th-edition
  • vancouver
  • Other style
More styles
Language
  • de-DE
  • en-GB
  • en-US
  • fi-FI
  • nn-NO
  • nn-NB
  • sv-SE
  • Other locale
More languages
Output format
  • html
  • text
  • asciidoc
  • rtf
Tetrabromobisphenol A Is an Efficient Stabilizer of the Transthyretin Tetramer
Umeå University, Faculty of Medicine, Department of Medical Biochemistry and Biophysics.ORCID iD: 0000-0002-9500-5917
Umeå University, Faculty of Science and Technology, Department of Chemistry.
Umeå University, Faculty of Medicine, Department of Medical Biochemistry and Biophysics.
Umeå University, Faculty of Medicine, Department of Medical Biochemistry and Biophysics.
Show others and affiliations
2016 (English)In: PLOS ONE, E-ISSN 1932-6203, Vol. 11, no 4, article id e0153529Article in journal (Refereed) Published
Abstract [en]

Amyloid formation of the human plasma protein transthyretin (TTR) is associated with several human disorders, including familial amyloidotic polyneuropathy (FAP) and senile systemic amyloidosis. Dissociation of TTR’s native tetrameric assembly is the rate-limiting step in the conversion into amyloid, and this feature presents an avenue for intervention because binding of an appropriate ligand to the thyroxin hormone binding sites of TTR stabilizes the native tetrameric assembly and impairs conversion into amyloid. The desired features for an effective TTR stabilizer include high affinity for TTR, high selectivity in the presence of other proteins, no adverse side effects at the effective concentrations, and a long half-life in the body. In this study we show that the commonly used flame retardant tetrabromobisphenol A (TBBPA) efficiently stabilizes the tetrameric structure of TTR. The X-ray crystal structure shows TBBPA binding in the thyroxine binding pocket with bromines occupying two of the three halogen binding sites. Interestingly, TBBPA binds TTR with an extremely high selectivity in human plasma, and the effect is equal to the recently approved drug tafamidis and better than diflunisal, both of which have shown therapeutic effects against FAP. TBBPA consequently present an interesting scaffold for drug design. Its absorption, metabolism, and potential side-effects are discussed.

Place, publisher, year, edition, pages
Public Library Science , 2016. Vol. 11, no 4, article id e0153529
National Category
Chemical Sciences
Identifiers
URN: urn:nbn:se:umu:diva-119999DOI: 10.1371/journal.pone.0153529ISI: 000374541200025PubMedID: 27093678Scopus ID: 2-s2.0-84964658661OAI: oai:DiVA.org:umu-119999DiVA, id: diva2:926128
Available from: 2016-05-04 Created: 2016-05-04 Last updated: 2023-03-23Bibliographically approved
In thesis
1. Selection of transthyretin amyloid inhibitors
Open this publication in new window or tab >>Selection of transthyretin amyloid inhibitors
2016 (English)Doctoral thesis, comprehensive summary (Other academic)
Abstract [en]

Amyloidosis is a group of clinical disorders caused by the aggregation of specific proteins into abnormal extracellular deposits. Today, 31 different proteins have been linked to amyloid diseases including transthyretin-related amyloidosis (ATTR). ATTR occurs through the aggregation of either wild-type plasma protein transthyretin (TTR) or a mutated form. TTR is a homotetramer that under normal circumstances functions as a carrier of thyroxine and retinol binding protein. The aggregation cascade requires dissociation of the tetramer into monomers, and preventing this dissociation represents a potential mode of intervention. Interestingly, small molecules, referred as kinetic stabilizers, can bind to TTR’s thyroxine-binding site (TBS) and such molecules are currently being used as a therapeutic approach to impair tetramer dissociation.

The efficacy of TTR stabilization is directly correlated to the binding affinity of the ligand to TBS. However, the binding of the ligand to TTR in vivo can be affected by other plasma components resulting in poor efficacy. Thus, the selectivity of ligands is an important parameter. We have designed an assay where the ability to stabilize TTR can be directly evaluated in plasma and we have investigated the stabilizing effect of nine potential TTR binders (Paper I). The results, surprisingly, revealed that the binding affinity of molecules has a poor correlation to its selectivity. However, the nature of protein-ligand complex formation can also be described by enthalpic (∆H) and entropic (∆S) energy contributions. ∆H represents the change in chemical bonds and frequently requires a higher order of orientation compared to the ∆S component, which mainly represents the hydrophobic effect via the exclusion of water. We hypothesized that ligands possessing high ΔH in binding to their co-partner would also be more specific in a complex environment such as plasma. By applying a thermodynamic analysis using isothermal titration calorimetry, we found that the selectivity in plasma correlates well with the ∆H contribution and might, therefore, be a better predictor for selectivity.

Luteolin was found to be a highly selective stabilizer of TTR and was investigated further (Paper II). The ligand displayed a significant rescuing effect in both cell culture and animal models. However, luteolin undergoes rapid enzymatic degradation in the liver and this impairs its use as a potential therapeutic drug. To attempt to circumvent this issue, we modified the most exposed hydroxyl group thus rendering the molecule inert towards glucuronidation (Paper III). The substitutions resulted in higher stability in the face of hepatic degradation molecules, but they also affected the selectivity in a negative manner.

The screening for new TTR stabilizers resulted in the discovery of tetrabromobisphenol A, which displayed a very high selectivity (Paper IV). This study also included a comparison with the drug Vyndaqel™ which currently is in clinically use, and showed how the dosage could be altered to acquire a better level of saturation and possibly also a better clinical effect.

Taken together we present new molecules with the ability to stabilize TTR, and these can serve as scaffolds for the design of new drugs. We present a method to measure the efficacy of a TTR-stabilizing drugs in a complex matrix and as well as a way to adjust the dosage of existing drugs. We also show that the selectivity of a drug is affected by the relative proportion of ∆H and ∆S, and this is of interest for drug design in general.

Place, publisher, year, edition, pages
Umeå: Umeå University, 2016. p. 72
Series
Umeå University medical dissertations, ISSN 0346-6612 ; 1826
Keywords
Transthyretin, TTR, ATTR, TTR-stabilizing drugs, selectivity
National Category
Other Basic Medicine
Research subject
Medical Biochemistry
Identifiers
urn:nbn:se:umu:diva-123939 (URN)978-91-7601-528-5 (ISBN)
Public defence
2016-09-09, KB3A9, byggnad KBC, Umeå, 09:00 (English)
Opponent
Supervisors
Available from: 2016-08-18 Created: 2016-07-06 Last updated: 2021-12-20Bibliographically approved
2. Kinetic stabilization of transthyretin and its role as an inhibitor of Aβ amyloid formation
Open this publication in new window or tab >>Kinetic stabilization of transthyretin and its role as an inhibitor of Aβ amyloid formation
2017 (English)Doctoral thesis, comprehensive summary (Other academic)
Abstract [en]

Amyloid formation occurs when normally soluble proteins and peptides misfold and aggregate into intractable threadlike structures called fibrils. There are currently more than 30 proteins associated with this aberrant structure, including the Aβ peptide in Alzheimer’s disease (AD) and transthyretin (TTR) in TTR amyloidosis. TTR is a homotetrameric transporter protein present in both cerebrospinal fluid and plasma. Dissociation of its tetrameric structure is required for the formation of amyloid fibrils. Small molecule ligands able to bind and stabilize the tetrameric structure of TTR thus represent a potential therapeutic intervention. Interestingly, apart from TTR’s role as a toxic agent in TTR amyloidosis, it also has a role as an inhibitor of the Aβ toxicity associated with AD. The work presented in this thesis focused on small molecules that have the potential ability to prevent TTR amyloidosis. We also sought to gain a greater understanding of the interaction between TTR and the Aβ peptide with respect to Aβ fibril formation.

The ability of a drug to stabilize TTR is directly correlated to its binding affinity. However, since TTR is a plasma protein, it is of great importance that the drug binds selectively to TTR. In paper I, we used a newly developed urea denaturation assay, in combination with isothermal titration calorimetry, to show that, in a complex environment such as plasma, the enthalpy of binding correlates better with a drug’s ability to stabilize TTR than the binding affinity. In paper II, we modified the highly selective but rapidly degraded TTR ligand luteolin in order to increase its resistance against biotransformation. Using a liver-based microsome assay, in combination with HPLC, we show how the luteolin analogues have gained increased stability. However, using the urea assay, we also show that the analogues have lost much of luteolin’s selectivity. In paper III, we show that tetrabromobisphenol A is a highly selective binder of TTR in plasma and is able to rescue cells from TTR-induced toxicity. In paper IV, we studied the interaction of TTR with Aβ and its effect on Aβ fibril formation. We used a ThT fluorescence-based assay and dot blotting to show that TTR inhibits Aβ amyloid formation and promotes the formation of high molecular weight assemblies with an open N-terminus. Using surface plasmon resonance, we further show how TTR is unable to inhibit fibril elongation and instead targets the nucleation processes, both primary and fibril-catalyzed secondary nucleation. To conclude, we present new molecules with the ability to selectively stabilize TTR that can serve as scaffolds in drug design. We also elucidate TTR’s inhibiting effects on toxic Aβ amyloid formation.

Place, publisher, year, edition, pages
Umeå: Umeå Universitet, 2017. p. 55
Keywords
Amyloid, transthyretin, amyloid beta, kinetic stabilizer
National Category
Biochemistry and Molecular Biology
Research subject
Biochemistry
Identifiers
urn:nbn:se:umu:diva-131776 (URN)978-91-7601-676-3 (ISBN)
Public defence
2017-03-17, Lilla hörsalen, KBC-huset, Linnaeusväg 6, 90756 Umeå, Umeå, 10:00 (English)
Opponent
Supervisors
Available from: 2017-02-23 Created: 2017-02-21 Last updated: 2018-06-09Bibliographically approved

Open Access in DiVA

fulltext(1210 kB)362 downloads
File information
File name FULLTEXT01.pdfFile size 1210 kBChecksum SHA-512
f3857346922db77d3a2ec74844a79f91837d0956038fa80ed66ba828c37f37edebf16a425838f95c7ceb36c1758ebe17cd9ca73d0ff51133cc81c61f4f86b110
Type fulltextMimetype application/pdf

Other links

Publisher's full textPubMedScopus

Authority records

Iakovleva, IrinaBegum, AfshanBrännström, KristofferNilsson, LinaZhang, JinAndersson, Patrik L.Sauer-Eriksson, A. ElisabethOlofsson, Anders

Search in DiVA

By author/editor
Iakovleva, IrinaBegum, AfshanBrännström, KristofferNilsson, LinaZhang, JinAndersson, Patrik L.Sauer-Eriksson, A. ElisabethOlofsson, Anders
By organisation
Department of Medical Biochemistry and BiophysicsDepartment of Chemistry
In the same journal
PLOS ONE
Chemical Sciences

Search outside of DiVA

GoogleGoogle Scholar
Total: 362 downloads
The number of downloads is the sum of all downloads of full texts. It may include eg previous versions that are now no longer available

doi
pubmed
urn-nbn

Altmetric score

doi
pubmed
urn-nbn
Total: 777 hits
CiteExportLink to record
Permanent link

Direct link
Cite
Citation style
  • apa
  • ieee
  • modern-language-association-8th-edition
  • vancouver
  • Other style
More styles
Language
  • de-DE
  • en-GB
  • en-US
  • fi-FI
  • nn-NO
  • nn-NB
  • sv-SE
  • Other locale
More languages
Output format
  • html
  • text
  • asciidoc
  • rtf