Your browser doesn't support javascript.
loading
Fasting upregulates the monocarboxylate transporter MCT1 at the rat blood-brain barrier through PPAR δ activation.
Chasseigneaux, Stéphanie; Cochois-Guégan, Véronique; Lecorgne, Lucas; Lochus, Murielle; Nicolic, Sophie; Blugeon, Corinne; Jourdren, Laurent; Gomez-Zepeda, David; Tenzer, Stefan; Sanquer, Sylvia; Nivet-Antoine, Valérie; Menet, Marie-Claude; Laplanche, Jean-Louis; Declèves, Xavier; Cisternino, Salvatore; Saubaméa, Bruno.
Afiliación
  • Chasseigneaux S; Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France.
  • Cochois-Guégan V; Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France.
  • Lecorgne L; Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France.
  • Lochus M; Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France.
  • Nicolic S; Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France.
  • Blugeon C; Département de biologie, GenomiqueENS, Institut de Biologie de l'ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France.
  • Jourdren L; Département de biologie, GenomiqueENS, Institut de Biologie de l'ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France.
  • Gomez-Zepeda D; Helmholtz-Institute for Translational Oncology Mainz (HI-TRON Mainz), A Hemlholtz Institute of the DKFZ, Mainz, Germany.
  • Tenzer S; German Cancer Research Center (DKFZ) Heidelberg, Division 191, 69120, Heidelberg, Germany.
  • Sanquer S; Institute of Immunology, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany.
  • Nivet-Antoine V; Helmholtz-Institute for Translational Oncology Mainz (HI-TRON Mainz), A Hemlholtz Institute of the DKFZ, Mainz, Germany.
  • Menet MC; German Cancer Research Center (DKFZ) Heidelberg, Division 191, 69120, Heidelberg, Germany.
  • Laplanche JL; Institute of Immunology, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany.
  • Declèves X; Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University, Mainz, Germany.
  • Cisternino S; AP.HP.Centre, Université Paris Cité, Paris, France.
  • Saubaméa B; AP-HP Biochimie générale, Hôpital Necker Enfants Malades, Université Paris Cité, Inserm, Innovations Thérapeutiques en Hémostase, Paris, France.
Fluids Barriers CNS ; 21(1): 33, 2024 Apr 08.
Article en En | MEDLINE | ID: mdl-38589879
ABSTRACT

BACKGROUND:

The blood-brain barrier (BBB) is pivotal for the maintenance of brain homeostasis and it strictly regulates the cerebral transport of a wide range of endogenous compounds and drugs. While fasting is increasingly recognized as a potential therapeutic intervention in neurology and psychiatry, its impact upon the BBB has not been studied. This study was designed to assess the global impact of fasting upon the repertoire of BBB transporters.

METHODS:

We used a combination of in vivo and in vitro experiments to assess the response of the brain endothelium in male rats that were fed ad libitum or fasted for one to three days. Brain endothelial cells were acutely purified and transcriptionaly profiled using RNA-Seq. Isolated brain microvessels were used to assess the protein expression of selected BBB transporters through western blot. The molecular mechanisms involved in the adaptation to fasting were investigated in primary cultured rat brain endothelial cells. MCT1 activity was probed by in situ brain perfusion.

RESULTS:

Fasting did not change the expression of the main drug efflux ATP-binding cassette transporters or P-glycoprotein activity at the BBB but modulated a restrictive set of solute carrier transporters. These included the ketone bodies transporter MCT1, which is pivotal for the brain adaptation to fasting. Our findings in vivo suggested that PPAR δ, a major lipid sensor, was selectively activated in brain endothelial cells in response to fasting. This was confirmed in vitro where pharmacological agonists and free fatty acids selectively activated PPAR δ, resulting in the upregulation of MCT1 expression. Moreover, dosing rats with a specific PPAR δ antagonist blocked the upregulation of MCT1 expression and activity induced by fasting.

CONCLUSIONS:

Altogether, our study shows that fasting affects a selected set of BBB transporters which does not include the main drug efflux transporters. Moreover, we describe a previously unknown selective adaptive response of the brain vasculature to fasting which involves PPAR δ and is responsible for the up-regulation of MCT1 expression and activity. Our study opens new perspectives for the metabolic manipulation of the BBB in the healthy or diseased brain.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Barrera Hematoencefálica / PPAR delta Límite: Animals Idioma: En Año: 2024 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Barrera Hematoencefálica / PPAR delta Límite: Animals Idioma: En Año: 2024 Tipo del documento: Article