Elsevier

Tetrahedron

Volume 72, Issues 27–28, 7 July 2016, Pages 3898-3904
Tetrahedron

Stereocontrolled synthesis of (S)-9-cis- and (S)-11-cis-13,14-dihydroretinoic acid

https://doi.org/10.1016/j.tet.2016.05.006Get rights and content

Abstract

The 9-cis and 11-cis stereoisomers of 13,14-dihydroretinoic acid with S configuration, (S)-7 and (S)-9, respectively, have been synthesized stereoselectively. The former has been recently characterized as the first endogenous natural ligand of the retinoid X receptor (RXR). The Julia-Kocienski reaction of allyl sulfones and aldehydes was used as connective step and afforded the Z isomer of a trienyl ester accounting for the entire side chain of the targets. A highly selective and unidirectional iodine-induced isomerization of a Z,Z,E triene to the desired E,Z,E isomer was required prior to the synthesis of (S)-7 via a Suzuki cross-coupling. The same approach to (S)-9 led to substantial isomerization when the Suzuki cross-coupling was used as the last bond-forming reaction. As alternative, the two bond-forming steps were exchanged and the synthesis of (S)-9 was completed using the Z-selective Julia-Kocienski reaction.

Introduction

Vitamin A (retinol, 1) and its metabolites (native retinoids) are essential in many physiological processes, such as vision, immunity, the regulation of embryonic development, the control of cell differentiation, cell proliferation and apoptosis.1 With the exception of vision, most of these cellular processes have been traditionally considered to be mediated by the binding (and activation) of vitamin A metabolites all-trans-retinoic acid 2 and its 9-cis isomer 3 to retinoic acid receptors (RARs) and retinoid X receptors (RXRs), which are members of the nuclear receptor superfamily.2, 3, 4, 5

In addition to all-trans-retinoic acid 2, vitamin A is transformed into additional metabolites that collectively help establish a delicate control of the homeostasis of retinoid levels.1, 6 In this regard, a group of endogenous dihydroretinoids, including all-trans-13,14-dihydroretinoic acid 5, have been recently added to the vitamin A metabolite pool.7, 8, 9 It has been shown that the formation of 5 involves the formal hydrogenation of all-trans-retinol 1 mediated by the enzyme retinol saturase (RetSat)10 and further two-stage oxidation of the polar group of 4 to metabolite 5 mediated by retinol dehydrogenases (ADH/SDR) and retinal dehydrogenases (RADH).8, 9 Naturally-occurring 5 was shown to be the R enantiomer, which is consistent with its higher binding affinity to RARs and transactivation activity in comparison with its antipode.8

Some double bond stereoisomers of the parent polyene6 also play fundamental roles in biological systems. The best known of those is 11-cis-retinal 6, the chromophore of the visual pigments, which binds to the apoprotein opsin as a protonated Schiff base to trigger the visual cycle upon light absorption.11 Fig 1.

More controversial is the endogenous presence of 9-cis-retinoic acid 3 in cells and tissues.12 Although originally described in the liver and kidney,13 and later in rat epididymal tissue and spermatozoa,14 its detection in various organs has been questioned,15, 16, 17, 18, 19, 20 perhaps with the exception of pancreas.21, 22 Pharmacological and genetic studies in mouse epidermis keratinocytes23 and mouse embryos24, 25 added functional evidence to the analytical data and together concurred that 9-cis-retinoic acid 3 is unlikely a bona fide physiological ligand but rather a pharmacological ligand for both RARs and RXRs.12

Studies of retinoid metabolism and gene expression are of major interest to aid in the identification of novel pathways regulated by yet uncharacterized endogenous ligands that can also alter gene expression.6 Retinoid metabolites with 9-cis configuration, namely 9-cis-13,14-dihydroretinoic acid (S)-726 and its 4-oxo derivative (S)-827, 28 have been recently isolated from several organs and characterized as retinoid receptor ligands. Although compounds with the structure of 9-cis-13,14-dihydroretinoic acid 7 (its absolute configuration was not determined) and its taurine conjugate were detected in rats, the animals had been fed with high doses of 9-cis-retinoic acid 3, making interpretation of their putative role as an endogenous retinoid doubtful.29 In contrast 9-cis-13,14-dihydroretinoic acid 7 was detected in normally fed animals, and found to bind and transactivate RXR (as well as RAR) in various assays and to display similar transcriptional activity as other RXR ligands in cultured human dendritic cells.26 Intriguingly (S)-8 was reported as an endogenous ligand for at least two RAR subtypes (α and β) but not for RXR. This compound induced in vivo morphological changes in chicken limb buds similarly to all-trans-retinoic acid 2 and regulated gene transcription in the same organ although with lower potency than 2.27, 28 Thus far, the detailed metabolic pathway for formation of 9-cis-13,14-dihydroretinoids is unknown.

In order to aid in the characterization of endogenous retinoids, their metabolic formation and their function, we started a program aimed at the development of stereoselective approaches to dihydroretinoids of potential biological interest. We have reported the synthesis of all-trans-13,14-dihydroretinoic acid 5 in both enantiomeric forms and showed that the R enantiomer is the endogenous ligand.8 The stereoselective synthesis of the S enantiomers (the configuration of the presumably oxidation metabolite 8) of the 9-cis and 11-cis stereoisomers of this compound (7 and 9) is shown below.

Section snippets

Results and discussion

Central to the construction of the polyene side chain of retinoids1a is the choice of the synthetic tactic. This may involve either the use of monofunctionalized components to perform Csp2double bondCsp2 bond construction by carbonyl condensation reactions (Wittig, HWE, Julia) or the use of mono and bis-functionalized alkenyl linchpins30 to connect components of complementary reactivity by palladium-catalyzed Csp2–Csp2 cross-coupling reactions.31 In practice, functionalized alkenyl reagents (as halogens

Experimental section

For general procedures, see Supplementary data.

Acknowledgements

This work was supported by funds from the Spanish MINECO (SAF2013-48397-R-FEDER), Xunta de Galicia (Grant 08CSA052383PR from DXI+D+i; Consolidación 2013/007 from DXPCTSUG; INBIOMED-FEDER ‘Unha maneira de facer Europa’).

References and notes (50)

  • R. Alvarez et al.

    Chem. Rev.

    (2014)
  • P. Huang et al.

    Annu. Rev. Physiol.

    (2010)
  • A.R. Moise et al.

    Biochemistry

    (2007)
  • P.D. Kiser et al.

    Chem. Rev.

    (2014)
  • R.S. Pappas et al.

    Biol. Reprod.

    (1993)
  • Y. Yamakoshi et al.

    Biol. Pharm. Bull.

    (2002)
  • R. Rühl

    Rapid Commun. Mass Spectrom.

    (2006)
  • C. Calléja et al.

    Genes Dev.

    (2006)
  • R. Rühl et al.

    PLoS Genet.

    (2015)
  • J.P. Schuchardt et al.

    FEBS J.

    (2009)
  • A. Torrado et al.

    Tetrahedron

    (1995)
  • A. Torrado et al.

    Synthesis

    (1995)
  • Y. Pazos et al.

    J. Org. Chem.

    (2001)
  • B. Domínguez et al.

    J. Org. Chem.

    (2000)
  • B. Vaz et al.

    J. Org. Chem.

    (2002)
  • R.M. Evans et al.

    Cell

    (2014)
  • F. Rastinejad et al.

    J. Mol. Endocrinol.

    (2013)
  • P. Huang et al.

    Chem. Rev.

    (2014)
  • O. Ziouzenkova et al.

    FEBS Lett.

    (2008)
  • A.R. Moise et al.

    J. Am. Chem. Soc.

    (2008)
  • A.R. Moise et al.

    Mol. Pharmacol.

    (2009)
  • A.R. Moise et al.

    J. Biol. Chem.

    (2004)
  • G. Wolf

    Nutr. Rev.

    (2006)
  • R.A. Heyman et al.

    Cell

    (1992)
  • Cited by (4)

    View full text