24
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Mechanism of Action and Clinical Application of Tafamidis in Hereditary Transthyretin Amyloidosis

      review-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Transthyretin (TTR) transports the retinol-binding protein–vitamin A complex and is a minor transporter of thyroxine in blood. Its tetrameric structure undergoes rate-limiting dissociation and monomer misfolding, enabling TTR to aggregate or to become amyloidogenic. Mutations in the TTR gene generally destabilize the tetramer and/or accelerate tetramer dissociation, promoting amyloidogenesis. TTR-related amyloidoses are rare, fatal, protein-misfolding disorders, characterized by formation of soluble aggregates of variable structure and tissue deposition of amyloid. The TTR amyloidoses present with a spectrum of manifestations, encompassing progressive neuropathy and/or cardiomyopathy. Until recently, the only accepted treatment to halt progression of hereditary TTR amyloidosis was liver transplantation, which replaces the hepatic source of mutant TTR with the less amyloidogenic wild-type TTR. Tafamidis meglumine is a rationally designed, non-NSAID benzoxazole derivative that binds with high affinity and selectivity to TTR and kinetically stabilizes the tetramer, slowing monomer formation, misfolding, and amyloidogenesis. Tafamidis is the first pharmacotherapy approved to slow the progression of peripheral neurologic impairment in TTR familial amyloid polyneuropathy. Here we describe the mechanism of action of tafamidis and review the clinical data, demonstrating that tafamidis treatment slows neurologic deterioration and preserves nutritional status, as well as quality of life in patients with early-stage Val30Met amyloidosis.

          Electronic supplementary material

          The online version of this article (doi:10.1007/s40120-016-0040-x) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references95

          • Record: found
          • Abstract: found
          • Article: not found

          Rationalization of the effects of mutations on peptide and protein aggregation rates.

          In order for any biological system to function effectively, it is essential to avoid the inherent tendency of proteins to aggregate and form potentially harmful deposits. In each of the various pathological conditions associated with protein deposition, such as Alzheimer's and Parkinson's diseases, a specific peptide or protein that is normally soluble is deposited as insoluble aggregates generally referred to as amyloid. It is clear that the aggregation process is generally initiated from partially or completely unfolded forms of the peptides and proteins associated with each disease. Here we show that the intrinsic effects of specific mutations on the rates of aggregation of unfolded polypeptide chains can be correlated to a remarkable extent with changes in simple physicochemical properties such as hydrophobicity, secondary structure propensity and charge. This approach allows the pathogenic effects of mutations associated with known familial forms of protein deposition diseases to be rationalized, and more generally enables prediction of the effects of mutations on the aggregation propensity of any polypeptide chain.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Prediction of aggregation-prone regions in structured proteins.

            We present a method for predicting the regions of the sequences of peptides and proteins that are most important in promoting their aggregation and amyloid formation. The method extends previous approaches by allowing such predictions to be carried out for conditions under which the molecules concerned can be folded or contain a significant degree of persistent structure. In order to achieve this result, the method uses only knowledge of the sequence of amino acids to estimate simultaneously both the propensity for folding and aggregation and the way in which these two types of propensity compete. We illustrate the approach by its application to a set of peptides and proteins both associated and not associated with disease. Our results show not only that the regions of a protein with a high intrinsic aggregation propensity can be identified in a robust manner but also that the structural context of such regions in the monomeric form is crucial for determining their actual role in the aggregation process.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The acid-mediated denaturation pathway of transthyretin yields a conformational intermediate that can self-assemble into amyloid.

              Transthyretin (TTR) amyloid fibril formation is observed during partial acid denaturation and while refolding acid-denatured TTR, implying that amyloid fibril formation results from the self-assembly of a conformational intermediate. The acid denaturation pathway of TTR has been studied in detail herein employing a variety of biophysical methods to characterize the intermediate(s) capable of amyloid fibril formation. At physiological concentrations, tetrameric TTR remains associated from pH 7 to pH 5 and is incapable of amyloid fibril formation. Tetrameric TTR dissociates to a monomer in a process that is dependent on both pH and protein concentration below pH 5. The extent of amyloid fibril formation correlates with the concentration of the TTR monomer having an altered, but defined, tertiary structure over the pH range of 5.0-3.9. The inherent Trp fluorescence-monitored denaturation curve of TTR exhibits a plateau over the pH range where amyloid fibril formation is observed (albeit at a higher concentration), implying that a steady-state concentration of the amyloidogenic intermediate with an altered tertiary structure is being detected. Interestingly, 1-anilino-8-naphthalenesulfonate fluorescence is at a minimum at the pH associated with maximal amyloid fibril formation (pH 4.4), implying that the amyloidogenic intermediate does not have a high extent of hydrophobic surface area exposed, consistent with a defined tertiary structure. Transthyretin has two Trp residues in its primary structure, Trp-41 and Trp-79, which are conveniently located far apart in the tertiary structure of TTR. Replacement of each Trp with Phe affords two single Trp containing variants which were used to probe local pH-dependent tertiary structural changes proximal to these chromophores. The pH-dependent fluorescence behavior of the Trp-79-Phe mutant strongly suggests that Trp-41 is located near the site of the tertiary structural rearrangement that occurs in the formation of the monomeric amyloidogenic intermediate, likely involving the C-strand-loop-D-strand region. Upon further acidification of TTR (below pH 4.4), the structurally defined monomeric amyloidogenic intermediate begins to adopt alternative conformations that are not amyloidogenic, ultimately forming an A-state conformation below pH 3 which is also not amyloidogenic. In summary, analytical equilibrium ultracentrifugation, SDS-PAGE, far- and near-UV CD, fluorescence, and light scattering studies suggest that the amyloidogenic intermediate is a monomeric predominantly beta-sheet structure having a well-defined tertiary structure.
                Bookmark

                Author and article information

                Contributors
                tcoelho@netcabo.pt
                Journal
                Neurol Ther
                Neurol Ther
                Neurology and Therapy
                Springer Healthcare (Cheshire )
                2193-8253
                2193-6536
                19 February 2016
                19 February 2016
                June 2016
                : 5
                : 1
                : 1-25
                Affiliations
                [ ]Hospital de Santo António, Centro Hospitalar do Porto, Porto, Portugal
                [ ]Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
                [ ]Pfizer Rare Disease Research Unit, Cambridge, MA USA
                [ ]Johns Hopkins University School of Medicine, Baltimore, MD USA
                [ ]The Scripps Research Institute, La Jolla, CA USA
                [ ]Columbia University College of Physicians and Surgeons, New York, NY USA
                [ ]CHU Henri Mondor, Créteil, France
                [ ]Labaudiniere Consulting LLC, Lewes, DE USA
                [ ]Pfizer Inc., New York, NY USA
                [ ]Massachusetts General and McLean Hospitals, Harvard Medical School, Boston, MA USA
                [ ]FORUM Pharmaceuticals, Watertown, MA USA
                Article
                40
                10.1007/s40120-016-0040-x
                4919130
                26894299
                513edf4f-059e-4e82-a2a1-e9654592bf39
                © The Author(s) 2016
                History
                : 25 August 2015
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/100004319, Pfizer;
                Categories
                Review
                Custom metadata
                © Springer Healthcare 2016

                familial amyloid cardiomyopathy,familial amyloid polyneuropathy,hereditary ttr amyloid cardiomyopathy,pharmacology,senile systemic amyloidosis,therapeutic use,wild-type ttr amyloidosis

                Comments

                Comment on this article