Archives

  • 2018-07
  • 2018-10
  • 2018-11
  • 2019-04
  • 2019-05
  • 2019-06
  • 2019-07
  • 2019-08
  • 2019-09
  • 2019-10
  • 2019-11
  • 2019-12
  • 2020-01
  • 2020-02
  • 2020-03
  • 2020-04
  • 2020-05
  • 2020-06
  • 2020-07
  • 2020-08
  • 2020-09
  • 2020-10
  • 2020-11
  • 2020-12
  • 2021-01
  • 2021-02
  • 2021-03
  • 2021-04
  • 2021-05
  • 2021-06
  • 2021-07
  • 2021-08
  • 2021-09
  • 2021-10
  • 2021-11
  • 2021-12
  • 2022-01
  • 2022-02
  • 2022-03
  • 2022-04
  • 2022-05
  • 2022-06
  • 2022-07
  • 2022-08
  • 2022-09
  • 2022-10
  • 2022-11
  • 2022-12
  • 2023-01
  • 2023-02
  • 2023-03
  • 2023-04
  • 2023-05
  • 2023-06
  • 2023-08
  • 2023-09
  • 2023-10
  • 2023-11
  • 2023-12
  • 2024-01
  • 2024-02
  • 2024-03
  • Hypertriglyceridemia is associated with an

    2021-09-17

    Hypertriglyceridemia is associated with an overproduction and secretion of triglyceride-rich lipoproteins (TRLs), due to increased liver lipid substrate availability (Adiels et al., 2005, Choi and Ginsberg, 2011), and/or reduced catabolism of TRLs and their remnants, due to reduced lipoprotein lipase (LPL) activity, insufficient hepatic remnant receptors, or nigericin of dietary and hepatic-derived lipoproteins for a common clearance pathway (Ayyobi and Brunzell, 2003, Bishop et al., 2008, Mamo et al., 2001). Abnormal TRL remnant catabolism may also be related to various apolipoproteins found on TRLs—of these, the best-studied is apolipoprotein C3 (ApoC3) (Jong et al., 1999). People with complete absence of ApoC3 have very low TG levels associated with rapid plasma TG clearance (Ginsberg et al., 1986, Norum et al., 1982), and decreased risk of CHD in Amish and Ashkenazi Jewish populations has been observed with genetic variants that confer partial ApoC3 deficiency (Pollin et al., 2008). Similarly, ApoC3 knockout mice demonstrate markedly lower plasma TG levels (Maeda et al., 1994), while ApoC3 transgenic mice show hypertriglyceridemia (Ito et al., 1990) and increased atherosclerosis (Masucci-Magoulas et al., 1997, Zheng, 2014). But as plasma TGs and ApoC3 are highly correlated (Le et al., 1988, Schonfeld et al., 1979), the causal factor in altered atherosclerosis risk in mouse and man cannot be disentangled; in fact, these data have led to a renewed push to identify novel therapeutic targets to reduce CHD risk in hypertriglyceridemic patients. The γ-secretase is a multiprotein complex consisting of redundant catalytic (Presenilin 1 or 2) and regulatory (Aph-1a or -1b) subunits as well as unique targeting (Nicastrin) and enhancer (PEN2) components that regulate intramembrane proteolysis of type 1 transmembrane proteins (Wolfe, 2006). As γ-secretase mediates the pathologic cleavage of Alzheimer’s precursor protein (APP) to generate amyloid β protein (Aβ), γ-secretase inhibitors (GSIs) have been proposed as Alzheimer’s disease (AD) therapeutics (Selkoe, 2001). Unfortunately, a lack of efficacy has plagued GSIs in clinical trials for AD (Doody et al., 2013), but their antagonistic effects on Notch receptors have led to efforts to repurpose these therapeutics as antineoplastic agents (De Jesus-Acosta et al., 2014, Wei et al., 2010) and, more recently, for metabolic disease (Bi and Kuang, 2015, Pajvani et al., 2011, Sparling et al., 2015). For instance, we found that GSI treatment of diet-induced or genetic mouse models of obesity improved hepatic insulin sensitivity, likely through inhibition of Notch co-activation of FoxO1-mediated hepatic glucose production (Pajvani et al., 2011). Here, we describe our finding that GSIs reduce plasma TGs and non-HDL cholesterol, independent of liver Notch signaling. To elucidate the mechanism of this unexpected result, we created hepatocyte-specific γ-secretase knockout (albumin-Cre:Nicastrinflox/flox, henceforth, L-Ncst) mice, which, similar to GSI treatment, also show improved glucose tolerance and reduced plasma TGs due to increased hepatocyte TRL uptake. These data suggest that liver-specific γ-secretase inhibition has therapeutic potential to protect from hypertriglyceridemia without known gastrointestinal (GI) toxicity of GSIs (Real et al., 2009). To exploit this potential, we developed liver-selective Ncst antisense oligonucleotides (ASOs) and found that Ncst ASO-treated mice also show lower plasma TGs. These parallel pharmacologic and genetic approaches suggest a non-Notch, hepatocyte γ-secretase target that regulates plasma TGs. In fact, beyond APP and Notch, an increasing number of additional putative γ-secretase type 1 transmembrane protein targets have been identified (De Strooper, 2003, Shih and Wang, 2007, Wolfe, 2006, Wolfe and Kopan, 2004). To this end, an unbiased proteomics screen (Hemming et al., 2008) identified, but did not experimentally validate, a potential candidate for the γ-secretase effect on hepatocyte TRL uptake: the LDL receptor (LDLR). Indeed, we find that Nicastrin binds the C-terminal domain of LDLR, targeting LDLR for γ-secretase-mediated cleavage, which in turn induces LDLR lysosomal degradation. Thus, Ncst ASO treatment fails to lower plasma TGs in Ldlr−/− or Ldlr ASO-treated mice. These data uncover the novel role of hepatic γ-secretase to regulate LDLR and highlight the potential of liver-specific γ-secretase inhibitors to simultaneously ameliorate obesity-induced glucose intolerance and hypertriglyceridemia.