Elsevier

Metabolism

Volume 93, April 2019, Pages 33-43
Metabolism

Basic Science
Hyperinsulinemia drives hepatic insulin resistance in male mice with liver-specific Ceacam1 deletion independently of lipolysis

https://doi.org/10.1016/j.metabol.2019.01.008Get rights and content

Highlights

  • Mice with liver-specific Ceacam1 deletion manifest impaired insulin clearance followed by progressive insulin resistance

Abstract

Background

CEACAM1 regulates insulin sensitivity by promoting insulin clearance. Accordingly, global C57BL/6J.Cc1−/− null mice display hyperinsulinemia due to impaired insulin clearance at 2 months of age, followed by insulin resistance, steatohepatitis, visceral obesity and leptin resistance at 6 months. The study aimed at investigating the primary role of hepatic CEACAM1 in insulin and lipid homeostasis independently of its metabolic effect in extra-hepatic tissues.

Methods

Liver-specific C57BL/6J.AlbCre+Cc1fl/fl mice were generated and their metabolic phenotype was characterized by comparison to that of their littermate controls at 2–9 months of age, using hyperinsulinemic-euglycemic clamp analysis and indirect calorimetry. The effect of hyperphagia on insulin resistance was assessed by pair-feeding experiments.

Results

Liver-specific AlbCre+Cc1fl/fl mutants exhibited impaired insulin clearance and hyperinsulinemia at 2 months, followed by hepatic insulin resistance (assessed by hyperinsulinemic-euglycemic clamp analysis) and steatohepatitis at ~ 7 months of age, at which point visceral obesity and hyperphagia developed, in parallel to hyperleptinemia and blunted hypothalamic STAT3 phosphorylation in response to an intraperitoneal injection of leptin. Hyperinsulinemia caused hypothalamic insulin resistance, followed by increased fatty acid synthase activity, which together with defective hypothalamic leptin signaling contributed to hyperphagia and reduced physical activity. Pair-feeding experiment showed that hyperphagia caused systemic insulin resistance, including blunted insulin signaling in white adipose tissue and lipolysis, at 8–9 months of age.

Conclusion

AlbCre+Cc1fl/fl mutants provide an in vivo demonstration of the key role of impaired hepatic insulin clearance and hyperinsulinemia in the pathogenesis of secondary hepatic insulin resistance independently of lipolysis. They also reveal an important role for the liver-hypothalamic axis in the regulation of energy balance and subsequently, systemic insulin sensitivity.

Introduction

Insulin clearance occurs mostly in the liver and to a lower extent in kidneys and other peripheral tissues [1]. Upon its pulsatile release from pancreatic β-cells into the portal vein [2,3], insulin crosses the sinusoidal endothelium to reach and activate its receptor on the hepatocytic surface membrane [4]. This phosphorylates other substrates and delivers insulin to early/late endosomes for degradation before the receptor recycles back to the surface membrane [5]. In this manner, excess insulin is rapidly removed and maintained at a physiologically higher concentration in the portal vein than in the systemic circulation [6]. Under hyperinsulinemic conditions, the receptor is diverted to lysosomal degradation [7,8] to cause cellular insulin resistance.

Chronic hyperinsulinemia also induces hepatic de novo lipogenesis by activating SREBP1c, a master transcriptional regulator of lipogenic genes [9]. Thus, impaired hepatic insulin clearance leads to a concomitant increase in hepatic steatosis and hepatic insulin resistance.

Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), a surface membrane substrate of the insulin receptor, promotes insulin clearance [10,11] by taking part of the insulin-insulin receptor complex and increasing its rate of uptake and endosomal targeting [12,13]. Subsequently, it binds to cytosolic fatty acid synthase (FASN), an event that detaches it from the complex to facilitate the dissociation of insulin from its receptor while mediating an inhibitory effect of insulin on FASN [14]. This restricts hepatic de novo lipogenesis and protects the liver against high portal insulin levels. Consistently, mice with null deletion of Ceacam1 gene (Cc1−/−) manifested impaired insulin clearance and chronic hyperinsulinemia at 2 months, followed by insulin resistance, primarily hepatic, at ~6 months of age when propagated on C57BL/6J background [11]. Deletion of Ceacam1 gene induced triacylglycerol production and redistribution to white adipose tissue (WAT). This resulted in hepatic steatosis and visceral obesity at 2 months, followed by lipolysis and mobilization of non-esterified free fatty acids (NEFA) at ~6 months of age [11].

Because of the regulatory effect of CEACAM1 on lipid production (by preventing hyperinsulinemia or mediating insulin downregulation of FASN activity), it is possible that deleting Ceacam1 primarily increased FASN synthesis and hepatic de novo lipogenesis, independently of hyperinsulinemia, to lead to visceral obesity and NEFA mobilization, which causes hepatic insulin resistance (portal hypothesis) [15] that in turn induces a compensatory increase in insulin secretion and ensuing hyperinsulinemia. This model would be consistent with the commonly accepted paradigm of primary insulin resistance causing chronic hyperinsulinemia mainly by inducing a compensatory increase in insulin secretion. However, blocking lipolysis with nicotinic acid did not restore insulin clearance or sensitivity [16]. Together with intact β-cell mass [11], this indicates that hyperinsulinemia in Cc1−/− mice resulted primarily from impaired insulin clearance rather than increased insulin secretion, and that it caused insulin resistance independently of lipolysis.

Cc1−/− mice also developed leptin resistance concomitantly with insulin resistance, basal hyperphagia and decreased spontaneous locomotor activity [17]. Whereas hyperinsulinemia-driven insulin resistance in the hypothalamus caused energy imbalance, as shown by liver-specific rescuing of Ceacam1 in Cc1−/− mice [16], deleting CEACAM1 from the arcuate nucleus region of the hypothalamus, in particular pro-opiomelanocortin (POMC)-expressing neurons, may contribute to the regulation of energy imbalance in Cc1−/− mutants [17].

Given the multiple factors that could contribute to insulin resistance in Cc1−/− mice, including impaired insulin clearance, lipolysis and leptin resistance, it became imperative to identify its primary underlying mechanism with the overarching goal to determine whether it caused or resulted from hyperinsulinemia. To this end, we generated a liver-specific C57BL/6J.Ceacam1 knockout mouse (AlbCre+Cc1fl/fl) and characterized the hepatic and extra-hepatic mechanisms that could be implicated in its altered metabolic phenotype.

Section snippets

Generation of Liver-specific AlbCre+Cc1fl/fl Mice

As in the conditional T cell-specific null mouse [18], the targeting construct inserted a loxP-neo cassette in intron 6 and a loxP fragment in intron 9, deleting a sequence that encodes the cytoplasmic domain [19]. We crossed Cc1loxp/loxp mice with transgenic mice expressing Cre under the transcriptional control of the albumin gene promoter (AlbCre) on C57BL/6J background (Jackson Laboratories, Bar Harbor, ME) (Fig. S1). Heterozygotes were backcrossed >6× with C57BL/6J mice. Offsprings were

Liver-specific Deletion of CEACAM1

qRT-PCR analysis demonstrated that mouse Ceacam1 mRNA (mCc1) was absent in primary hepatocytes from AlbCre+Cc1fl/fl mice (Alb+Cc1fl/fl for simplicity) (Fig. 1A). Immunoblotting with mouse α-CEACAM1 (Ib:α-mCC1) detected CEACAM1 protein in the livers of controls (Alb–Cc1+/+, Alb+Cc1+/+ and Alb–Cc1fl/fl), but not Alb+Cc1fl/fl mutants (Fig. 1B.i). CEACAM1 protein content was intact in other tissues from mutant mice, including kidney, hypothalamus and heart (Fig. 1B.i-ii).

Early Onset of Impaired Insulin Clearance and Hyperinsulinemia in Alb+Cc1fl/fl Mice

Like Cc1−/− [11], Alb+Cc1

Discussion

The cause-effect relationship between insulin resistance and hyperinsulinemia remains elusive [37]. Whereas primary insulin resistance causes hyperinsulinemia mainly by inducing a compensatory increase in insulin secretion [38], evidence in support of the causative role of chronic hyperinsulinemia in insulin resistance is mounting [39,40], in particular when hyperinsulinemia results from impaired insulin clearance [41]. This is underlined by several mechanisms, including downregulation of the

Acknowledgments

We thank M. Kopfman at the Najjar laboratory for her technical assistance in maintaining the mouse lines and assisting in genotyping and phenotyping. We also thank the Osteopathic Heritage Foundation for its John J. Kopchick, PhD, Eminent Research Chair fund to SMN.

Funding

This work was supported by NIH grants: R01-DK054254, R01-DK083850, and R01-HL112248 to SMN, R01-HD081792 to JWH, and 5U2C-DK093000 to JKK. The work was also supported by the Middle-East Diabetes Research Center to HEG and SSG, and the American Heart Association (14POST20480294) to LR.

Duality of Interest

The authors declare no duality of interest.

Author Contributions

HEG, LR, HTM, SSG, IHM, JEH, HLN, and SS researched data. HEG coordinated the research among co-authors, analyzed data and wrote the first draft of the manuscript. JKK analyzed the hyperinsulinemic-euglycemic clamp data. SMN analyzed data and reviewed and revised the manuscript. SMN had full access to all the data of the study and takes responsibility for the conception of the studies and for the integrity and accuracy of data analysis. Original data are available upon request from the senior

References (50)

  • S.J. Koopmans et al.

    Chronic physiologic hyperinsulinemia impairs suppression of plasma free fatty acids and increases de novo lipogenesis but does not cause dyslipidemia in conscious normal rats

    Metabolism

    (1999)
  • T. Dai et al.

    Interaction between altered insulin and lipid metabolism in CEACAM1-inactive transgenic mice

    J Biol Chem

    (2004)
  • D.C. Polidori et al.

    Hepatic and extrahepatic insulin clearance are differentially regulated: results from a novel model-based analysis of intravenous glucose tolerance data

    Diabetes

    (2016)
  • A.V. Matveyenko et al.

    Pulsatile portal vein insulin delivery enhances hepatic insulin action and signaling

    Diabetes

    (2012)
  • N. Porksen et al.

    Pulsatile insulin secretion accounts for 70% of total insulin secretion during fasting

    Am J Physiol Endocrinol Metab

    (1995)
  • M. Mohamad et al.

    Ultrastructure of the liver microcirculation influences hepatic and systemic insulin activity and provides a mechanism for age-related insulin resistance

    Aging Cell

    (2016)
  • V.L. Tokarz et al.

    The cell biology of systemic insulin function

    J Cell Biol

    (2018)
  • G.M. Ward et al.

    Effects of prolonged pulsatile hyperinsulinemia in humans. Enhancement of insulin sensitivity

    Diabetes

    (1990)
  • J.S. Flier

    Insulin receptors and insulin resistance

    Annu Rev Med

    (1983)
  • J.D. Horton et al.

    SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver

    J Clin Invest

    (2002)
  • M.N. Poy et al.

    CEACAM1 regulates insulin clearance in liver

    Nat Genet

    (2002)
  • A.M. DeAngelis et al.

    Carcinoembryonic antigen-related cell adhesion molecule 1: a link between insulin and lipid metabolism

    Diabetes

    (2008)
  • C.V. Choice et al.

    Comparison of the intracellular trafficking of two alternatively spliced isoforms of pp120, a substrate of the insulin receptor tyrosine kinase

    J Cell Biochem

    (1999)
  • M. Kabir et al.

    Molecular evidence supporting the portal theory: a causative link between visceral adiposity and hepatic insulin resistance

    Am J Physiol Endocrinol Metab

    (2005)
  • L. Russo et al.

    Liver-specific reconstitution of CEACAM1 reverses the metabolic abnormalities caused by its global deletion in male mice

    Diabetologia

    (2017)
  • Cited by (35)

    • Regulation of hepatic fibrosis by carcinoembryonic antigen-related cell adhesion molecule 1

      2021, Metabolism: Clinical and Experimental
      Citation Excerpt :

      Moreover, insulin-resistant obese subjects with NAFLD manifested reduced hepatic levels of Carcinoembryonic Antigen-Related Cell Adhesion molecule 1 (CEACAM1) [12,13], a plasma membrane glycoprotein that promotes receptor-mediated insulin uptake in hepatocytes followed by its degradation [14]. Consistently, mice with global [15,16] and liver-specific [17] deletion of Ceacam1 gene, and L-SACC1 mice with its liver-specific inactivation [18] manifested chronic hyperinsulinemia driven by impaired insulin clearance. This in turn, caused insulin resistance (due to downregulation of the insulin receptor) and hepatic steatosis (owing to increased hyperinsulinemia-driven SREBP-1c activation of lipogenic genes transcription and to the loss of insulin's repression of fatty acid synthase activity mediated by CEACAM1 phosphorylation [19]).

    • Reduced insulin sensitivity and increased β/α cell mass is associated with reduced hepatic insulin-degrading enzyme activity in pregnant rats

      2021, Life Sciences
      Citation Excerpt :

      AlbCre+CEACAM fl/fl mice [31]. In this study, the authors demonstrated that insulin clearance impairment results in chronic hyperinsulinemia with consequent development of numerous metabolic dysfunctions typic of prediabetes [31]. Although the precise role of IDE on hepatic insulin degradation is not completely established, its absence exacerbates the metabolic outcomes from a high-fat diet and its functional reconstitution normalizes such parameters, suggesting the relevance of hepatic IDE activity for glucose and lipid homeostasis [15].

    View all citing articles on Scopus
    View full text