Deutsch
 
Hilfe Datenschutzhinweis Impressum
  DetailsucheBrowse

Datensatz

DATENSATZ AKTIONENEXPORT
  Deletion of Tbc1d4/As160 abrogates cardiac glucose uptake and increases myocardial damage after ischemia/reperfusion

Binsch, C., Barbosa, D. M., Hansen-Dille, G., Hubert, M., Hodge, S. M., Kolasa, M., et al. (2023). Deletion of Tbc1d4/As160 abrogates cardiac glucose uptake and increases myocardial damage after ischemia/reperfusion. Cardiovascular Diabetology, 22: 17. doi:10.1186/s12933-023-01746-2.

Item is

Basisdaten

einblenden: ausblenden:
Genre: Zeitschriftenartikel

Dateien

einblenden: Dateien
ausblenden: Dateien
:
CardiovascDiabetol_Binsch et al_2023.pdf (Verlagsversion), 4MB
Name:
CardiovascDiabetol_Binsch et al_2023.pdf
Beschreibung:
-
OA-Status:
Gold
Sichtbarkeit:
Öffentlich
MIME-Typ / Prüfsumme:
application/pdf / [MD5]
Technische Metadaten:
Copyright Datum:
-
Copyright Info:
© 2023. The Author(s)

Externe Referenzen

einblenden:

Urheber

einblenden:
ausblenden:
 Urheber:
Binsch, C., Autor
Barbosa, D. M., Autor
Hansen-Dille, G., Autor
Hubert, M., Autor
Hodge, S. M., Autor
Kolasa, M., Autor
Jeruschke, K., Autor
Weiß, J., Autor
Springer, C., Autor
Gorressen, S., Autor
Fischer, J. W., Autor
Lienhard, M.1, Autor                 
Herwig, R.1, Autor                 
Börno, S.2, Autor                 
Timmermann, B.2, Autor           
Cremer, A. L., Autor
Backes, H., Autor
Chadt, A., Autor
Al-Hasani, H., Autor
Affiliations:
1Bioinformatics (Ralf Herwig), Dept. of Computational Molecular Biology (Head: Martin Vingron), Max Planck Institute for Molecular Genetics, Max Planck Society, ou_2385701              
2Sequencing, Scientific Service (Head: Claudia Thurow), Max Planck Institute for Molecular Genetics, Max Planck Society, ou_1479670              

Inhalt

einblenden:
ausblenden:
Schlagwörter: Ischemia/reperfusion; Metabolic flexibility; Myocardial infarction; TBC1D4
 Zusammenfassung:

Background: Type 2 Diabetes mellitus (T2DM) is a major risk factor for cardiovascular disease and associated with poor outcome after myocardial infarction (MI). In T2DM, cardiac metabolic flexibility, i.e. the switch between carbohydrates and lipids as energy source, is disturbed. The RabGTPase-activating protein TBC1D4 represents a crucial regulator of insulin-stimulated glucose uptake in skeletal muscle by controlling glucose transporter GLUT4 translocation. A human loss-of-function mutation in TBC1D4 is associated with impaired glycemic control and elevated T2DM risk. The study's aim was to investigate TBC1D4 function in cardiac substrate metabolism and adaptation to MI.

Methods: Cardiac glucose metabolism of male Tbc1d4-deficient (D4KO) and wild type (WT) mice was characterized using in vivo [18F]-FDG PET imaging after glucose injection and ex vivo basal/insulin-stimulated [3H]-2-deoxyglucose uptake in left ventricular (LV) papillary muscle. Mice were subjected to cardiac ischemia/reperfusion (I/R). Heart structure and function were analyzed until 3 weeks post-MI using echocardiography, morphometric and ultrastructural analysis of heart sections, complemented by whole heart transcriptome and protein measurements.

Results: Tbc1d4-knockout abolished insulin-stimulated glucose uptake in ex vivo LV papillary muscle and in vivo cardiac glucose uptake after glucose injection, accompanied by a marked reduction of GLUT4. Basal cardiac glucose uptake and GLUT1 abundance were not changed compared to WT controls. D4KO mice showed mild impairments in glycemia but normal cardiac function. However, after I/R D4KO mice showed progressively increased LV endsystolic volume and substantially increased infarction area compared to WT controls. Cardiac transcriptome analysis revealed upregulation of the unfolded protein response via ATF4/eIF2α in D4KO mice at baseline. Transmission electron microscopy revealed largely increased extracellular matrix (ECM) area, in line with decreased cardiac expression of matrix metalloproteinases of D4KO mice.

Conclusions: TBC1D4 is essential for insulin-stimulated cardiac glucose uptake and metabolic flexibility. Tbc1d4-deficiency results in elevated cardiac endoplasmic reticulum (ER)-stress response, increased deposition of ECM and aggravated cardiac damage following MI. Hence, impaired TBC1D4 signaling contributes to poor outcome after MI.

Details

einblenden:
ausblenden:
Sprache(n): eng - English
 Datum: 2023-01-172023-01-27
 Publikationsstatus: Online veröffentlicht
 Seiten: -
 Ort, Verlag, Ausgabe: -
 Inhaltsverzeichnis: -
 Art der Begutachtung: -
 Identifikatoren: DOI: 10.1186/s12933-023-01746-2
PMID: 36707786
 Art des Abschluß: -

Veranstaltung

einblenden:

Entscheidung

einblenden:

Projektinformation

einblenden:

Quelle 1

einblenden:
ausblenden:
Titel: Cardiovascular Diabetology
Genre der Quelle: Zeitschrift
 Urheber:
Affiliations:
Ort, Verlag, Ausgabe: London, UK : Biomed Central
Seiten: - Band / Heft: 22 Artikelnummer: 17 Start- / Endseite: - Identifikator: ISSN: 1475-2840