TY - JOUR A1 - Mayer, Alexander E. A1 - Löffler, Mona C. A1 - Loza Valdés, Angel E. A1 - Schmitz, Werner A1 - El-Merahbi, Rabih A1 - Trujillo-Viera, Jonathan A1 - Erk, Manuela A1 - Zhang, Thianzhou A1 - Braun, Ursula A1 - Heikenwalder, Mathias A1 - Leitges, Michael A1 - Schulze, Almut A1 - Sumara, Grzegorz T1 - The kinase PKD3 provides negative feedback on cholesterol and triglyceride synthesis by suppressing insulin signaling JF - Science Signaling N2 - Hepatic activation of protein kinase C (PKC) isoforms by diacylglycerol (DAG) promotes insulin resistance and contributes to the development of type 2 diabetes (T2D). The closely related protein kinase D (PKD) isoforms act as effectors for DAG and PKC. Here, we showed that PKD3 was the predominant PKD isoform expressed in hepatocytes and was activated by lipid overload. PKD3 suppressed the activity of downstream insulin effectors including the kinase AKT and mechanistic target of rapamycin complex 1 and 2 (mTORC1 and mTORC2). Hepatic deletion of PKD3 in mice improved insulin-induced glucose tolerance. However, increased insulin signaling in the absence of PKD3 promoted lipogenesis mediated by SREBP (sterol regulatory element-binding protein) and consequently increased triglyceride and cholesterol content in the livers of PKD3-deficient mice fed a high-fat diet. Conversely, hepatic-specific overexpression of a constitutively active PKD3 mutant suppressed insulin-induced signaling and caused insulin resistance. Our results indicate that PKD3 provides feedback on hepatic lipid production and suppresses insulin signaling. Therefore, manipulation of PKD3 activity could be used to decrease hepatic lipid content or improve hepatic insulin sensitivity. KW - Protein kinase D3 (PKD3) KW - cholesterol KW - diacylglycerol (DAG) KW - liver KW - metabolism Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-250025 ET - accepted manuscript ER - TY - THES A1 - Mayer, Alexander E. T1 - Protein kinase D3 signaling in the regulation of liver metabolism T1 - Proteinkinase D3 Signalwirkung in der Regulation des Leberstoffwechsels N2 - The liver plays a pivotal role in maintaining energy homeostasis. Hepatic carbohydrate and lipid metabolism are tightly regulated in order to adapt quickly to changes in nutrient availability. Postprandially, the liver lowers the blood glucose levels and stores nutrients in form of glycogen and triglycerides (TG). In contrast, upon fasting, the liver provides glucose, TG, and ketone bodies. However, obesity resulting from a discrepancy in food intake and energy expenditure leads to abnormal fat accumulation in the liver, which is associated with the development of hepatic insulin resistance, non-alcoholic fatty liver disease, and diabetes. In this context, hepatic insulin resistance is directly linked to the accumulation of diacylglycerol (DAG) in the liver. Besides being an intermediate product of TG synthesis, DAG serves as second messenger in response to G-protein coupled receptor signaling. Protein kinase D (PKD) family members are DAG effectors that integrate multiple metabolic inputs. However, the impact of PKD signaling on liver physiology has not been studied so far. In this thesis, PKD3 was identified as the predominantly expressed isoform in liver. Stimulation of primary hepatocytes with DAG as well as high-fat diet (HFD) feeding of mice led to an activation of PKD3, indicating its relevance during obesity. HFD-fed mice lacking PKD3 specifically in hepatocytes displayed significantly improved glucose tolerance and insulin sensitivity. However, at the same time, hepatic deletion of PKD3 in mice resulted in elevated liver weight as a consequence of increased hepatic lipid accumulation. Lack of PKD3 in hepatocytes promoted sterol regulatory element-binding protein (SREBP)-mediated de novo lipogenesis in vitro and in vivo, and thus increased hepatic triglyceride and cholesterol content. Furthermore, PKD3 suppressed the activation of SREBP by impairing the activity of the insulin effectors protein kinase B (AKT) and mechanistic target of rapamycin complexes (mTORC) 1 and 2. In contrast, liver-specific overexpression of constitutive active PKD3 promoted glucose intolerance and insulin resistance. Taken together, lack of PKD3 improves hepatic insulin sensitivity but promotes hepatic lipid accumulation. For this reason, manipulating PKD3 signaling might be a valid strategy to improve hepatic lipid content or insulin sensitivity. However, the exact molecular mechanism by which PKD3 regulates hepatocytes metabolism remains unclear. Unbiased proteomic approaches were performed in order to identify PKD3 phosphorylation targets. In this process, numerous potential targets of PKD3 were detected, which are implicated in different aspects of cellular metabolism. Among other hits, phenylalanine hydroxylase (PAH) was identified as a target of PKD3 in hepatocytes. PAH is the enzyme that is responsible for the conversion of phenylalanine to tyrosine. In fact, manipulation of PKD3 activity using genetic tools confirmed that PKD3 promotes PAH-dependent conversion of phenylalanine to tyrosine. Therefore, the data in this thesis suggests that PKD3 coordinates lipid and amino acid metabolism in the liver and contributes to the development of hepatic dysfunction. N2 - Die Leber spielt eine zentrale Rolle bei der Aufrechterhaltung der Energiehomöostase. Der hepatische Kohlenhydrat- und Fettstoffwechsel ist stark reguliert, um sich schnell an Veränderungen in der Nährstoffverfügbarkeit anzupassen. Die Leber senkt postprandial den Blutzuckerspiegel und speichert Nährstoffe in Form von Glykogen und Triglyzeriden (TG). Im Gegensatz dazu stellt die Leber beim Fasten Glukose, TG und Ketonkörper bereit. Fettleibigkeit, welche aus einer Diskrepanz zwischen Nahrungsaufnahme und Energieaufwand resultiert, führt allerdings zu einer abnormalen Fettansammlung in der Leber, die mit der Entwicklung von Leberinsulinresistenz, nicht-alkoholischen Fettlebererkrankungen und Diabetes einhergeht. Hepatische Insulinresistenz steht dabei in direktem Zusammenhang mit der Akkumulation von Diacylglycerol (DAG) in der Leber. DAG ist nicht nur ein Zwischenprodukt der TG-Synthese, sondern dient auch als sekundärer Messenger im G-Protein-gekoppelten Rezeptor-Signalweg. Die Mitglieder der Proteinkinase D (PKD)-Familie sind DAG-Effektoren, die vielfache metabolische Inputs integrieren. Jedoch wurden die Auswirkungen der PKD-Signalwirkung auf die Leberphysiologie bisher nicht untersucht. Im Rahmen dieser Thesis wurde PKD3 als die in der Leber überwiegend exprimierte Isoform identifiziert. Die Stimulation von primären Hepatozyten mit DAG sowie die Fütterung von Mäusen mit fettreicher Nahrung (HFD) führte zu einer Aktivierung von PKD3, was auf eine Relevanz von PKD3 bei Fettleibigkeit hinweist. Mäusen, welchen PKD3 spezifisch in Hepatozyten fehlte und mit HFD gefüttert wurden, zeigten eine deutlich verbesserte Glukosetoleranz und Insulinsensitivität. Gleichzeitig führte jedoch die hepatische Deletion von PKD3 bei Mäusen zu einem erhöhten Lebergewicht in Folge einer erhöhten Lipidakkumulation in der Leber. Das Fehlen von PKD3 in Hepatozyten förderte die Sterol Regulatory Element-Binding Protein (SREBP)-vermittelte de novo Lipogenese in vitro und in vivo und erhöhte damit den Gehalt an Triglyceriden und Cholesterol in der Leber. Darüber hinaus supprimierte PKD3 die Aktivierung von SREBP, indem es die Aktivität der Insulin-Effektoren Proteinkinase B (AKT) und mechanistisches Ziel von Rapamycin- Komplexen (mTORC) 1 und 2 verminderte. Im Gegensatz dazu förderte die leberspezifische Überexpression von konstitutiv aktiver PKD3 die Glukoseintoleranz und Insulinresistenz. Zusammenfassend verbessert der Mangel an PKD3 die hepatische Insulinempfindlichkeit, aber fördert gleichzeitig die Akkumulation von Lipiden in der Leber. Aus diesem Grund könnte das Eingreifen in den PKD3-Signalweg eine gute Strategie zur Verbesserung des hepatischen Lipidgehalts oder der Insulinempfindlichkeit sein. Allerdings bleibt der genaue molekulare Mechanismus, mit dem PKD3 den Stoffwechsel von Hepatozyten reguliert, unklar. Es wurden unvoreingenommene proteomische Ansätze durchgeführt, um PKD3- Phosphorylierungsziele zu identifizieren. In diesem Prozess wurden zahlreiche potenzielle Ziele von PKD3 entdeckt, welche in den verschiedensten Aspekten des Zellstoffwechsels involviert sind. Unter anderem wurde Phenylalaninhydroxylase (PAH) als Ziel von PKD3 in Hepatozyten identifiziert. PAH ist das Enzym, welches für die Umwandlung von Phenylalanin in Tyrosin verantwortlich ist. Tatsächlich bestätigte die Manipulation der PKD3-Aktivität mit Hilfe von genetischen Werkzeugen, dass PKD3 die PAH-abhängige Umwandlung von Phenylalanin in Tyrosin fördert. Deswegen legen die Daten in dieser Arbeit nahe, dass PKD3 den Lipid- und Aminosäurestoffwechsel in der Leber koordiniert und zur Entwicklung von Leber- Dysfunktion beiträgt. KW - Metabolismus KW - Proteinkinase D KW - Leber-Metabolismus Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-207978 ER - TY - INPR A1 - Löffler, Mona C. A1 - Mayer, Alexander E. A1 - Trujillo Viera, Jonathan A1 - Loza Valdes, Angel A1 - El-Merahib, Rabih A1 - Ade, Carsten P. A1 - Karwen, Till A1 - Schmitz, Werner A1 - Slotta, Anja A1 - Erk, Manuela A1 - Janaki-Raman, Sudha A1 - Matesanz, Nuria A1 - Torres, Jorge L. A1 - Marcos, Miguel A1 - Sabio, Guadalupe A1 - Eilers, Martin A1 - Schulze, Almut A1 - Sumara, Grzegorz T1 - Protein kinase D1 deletion in adipocytes enhances energy dissipation and protects against adiposity T2 - The EMBO Journal N2 - Nutrient overload in combination with decreased energy dissipation promotes obesity and diabetes. Obesity results in a hormonal imbalance, which among others, activates G-protein coupled receptors utilizing diacylglycerol (DAG) as secondary messenger. Protein kinase D1 (PKD1) is a DAG effector which integrates multiple nutritional and hormonal inputs, but its physiological role in adipocytes is unknown. Here, we show that PKD1 promotes lipogenesis and suppresses mitochondrial fragmentation, biogenesis, respiration, and energy dissipation in an AMP-activated protein kinase (AMPK)-dependent manner. Moreover, mice lacking PKD1 in adipocytes are resistant to diet-induced obesity due to elevated energy expenditure. Beiging of adipocytes promotes energy expenditure and counteracts obesity. Consistently, deletion of PKD1 promotes expression of the β3-adrenergic receptor (ADRB3) in a CCAAT/enhancerbinding protein (C/EBP)-α and δ-dependent manner, which leads to the elevated expression of beige markers in adipocytes and subcutaneous adipose tissue. Finally, deletion of PKD1 in adipocytes improves insulin sensitivity and ameliorates liver steatosis. Thus, loss of PKD1 in adipocytes increases energy dissipation by several complementary mechanisms and might represent an attractive strategy to treat obesity and its related complications. KW - AMP-activated protein kinase (AMPK) KW - Beige adipocytes KW - β3 adrenergic receptor (ADRB3) KW - C/EBP KW - Protein kinase D1 (PKD1) Y1 - 2018 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-176093 ER - TY - JOUR A1 - Cai, Kai A1 - El-Merahbi, Rabih A1 - Loeffler, Mona A1 - Mayer, Alexander E. A1 - Sumara, Grzegorz T1 - Ndrg1 promotes adipocyte differentiation and sustains their function JF - Scientific Reports N2 - Adipocytes play a central role in maintaining metabolic homeostasis in the body. Differentiation of adipocyte precursor cells requires the transcriptional activity of peroxisome proliferator-activated receptor-γ (Pparγ) and CCAAT/enhancer binding proteins (C/Ebps). Transcriptional activity is regulated by signaling modules activated by a plethora of hormones and nutrients. Mechanistic target of rapamacin complexes (mTORC) 1 and 2 are central for the coordination of hormonal and nutritional inputs in cells and are essential for adipogenesis. Serum glucocorticoid kinase 1 (Sgk1)-dependent phosphorylation of N-Myc downstream-regulated gene 1 (Ndrg1) is a hallmark of mTORC2 activation in cells. Moreover, Pparγ activation promotes Ndrg1 expression. However, the impact of Ndrg1 on adipocyte differentiation and function has not yet been defined. Here, we show that Ndrg1 expression and its Sgk1-dependent phosphorylation are induced during adipogenesis. Consistently, we demonstrate that Ndrg1 promotes adipocyte differentiation and function by inducing Pparγ expression. Additionally, our results indicate that Ndrg1 is required for C/Ebpα phosphorylation. Moreover, we found that Ndrg1 phosphorylation by Sgk1 promotes adipocyte formation. Taken together, we show that induction of Ndrg1 expression by Pparγ and its phosphorylation by Sgk1 kinase are required for the acquisition of adipocyte characteristics by precursor cells. KW - differentiation KW - cell signalling KW - adipocytes KW - Ndrg1 Y1 - 2017 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-170565 VL - 7 IS - 7191 ER - TY - JOUR A1 - Trujillo‐Viera, Jonathan A1 - El‐Merahbi, Rabih A1 - Schmidt, Vanessa A1 - Karwen, Till A1 - Loza‐Valdes, Angel A1 - Strohmeyer, Akim A1 - Reuter, Saskia A1 - Noh, Minhee A1 - Wit, Magdalena A1 - Hawro, Izabela A1 - Mocek, Sabine A1 - Fey, Christina A1 - Mayer, Alexander E. A1 - Löffler, Mona C. A1 - Wilhelmi, Ilka A1 - Metzger, Marco A1 - Ishikawa, Eri A1 - Yamasaki, Sho A1 - Rau, Monika A1 - Geier, Andreas A1 - Hankir, Mohammed A1 - Seyfried, Florian A1 - Klingenspor, Martin A1 - Sumara, Grzegorz T1 - Protein Kinase D2 drives chylomicron‐mediated lipid transport in the intestine and promotes obesity JF - EMBO Molecular Medicine N2 - Lipids are the most energy‐dense components of the diet, and their overconsumption promotes obesity and diabetes. Dietary fat content has been linked to the lipid processing activity by the intestine and its overall capacity to absorb triglycerides (TG). However, the signaling cascades driving intestinal lipid absorption in response to elevated dietary fat are largely unknown. Here, we describe an unexpected role of the protein kinase D2 (PKD2) in lipid homeostasis. We demonstrate that PKD2 activity promotes chylomicron‐mediated TG transfer in enterocytes. PKD2 increases chylomicron size to enhance the TG secretion on the basolateral side of the mouse and human enterocytes, which is associated with decreased abundance of APOA4. PKD2 activation in intestine also correlates positively with circulating TG in obese human patients. Importantly, deletion, inactivation, or inhibition of PKD2 ameliorates high‐fat diet‐induced obesity and diabetes and improves gut microbiota profile in mice. Taken together, our findings suggest that PKD2 represents a key signaling node promoting dietary fat absorption and may serve as an attractive target for the treatment of obesity. KW - chylomicron KW - fat absorption KW - intestine KW - obesity KW - protein kinase D2/PKD2/PRKD2 Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-239018 VL - 13 IS - 5 ER -