Introduction
The liver is essential for both glucose and fatty acid metabolisms, producing glucose in the fasting state via glycogenolysis and gluconeogenesis (
1- Petersen M.C.
- Vatner D.F.
- Shulman G.I.
Regulation of hepatic glucose metabolism in health and disease.
). Hepatic lipid accumulation characterizes nonalcoholic fatty liver disease (NAFLD)
3The abbreviations used are:
NAFLD
nonalcoholic fatty liver disease
IL
interleukin
TNF
tumor necrosis factor
eWAT
epididymal white adipose tissue
VLDL
very low-density lipoprotein
MTP
microsomal triglyceride transfer protein
GSK3
glycogen synthase kinase 3
GS
glycogen synthase
GCK
glucokinase
G6P
glucose 6-phosphate
PEPCK
phosphoenolpyruvate carboxykinase
G6Pase
glucose-6-phosphatase
ACC
acetyl-CoA carboxylase
FAS
fatty acid synthase
CCCP
carbonyl cyanide m-chlorophenyl hydrazone
FCCP
carbonyl cyanide p-trifluoromethoxyphenylhydrazone
Z
benzyloxycarbonyl
fmk
fluoromethyl ketone
LDH
lactate dehydrogenase
PC
pyruvate carboxylase
CPT
carnitine palmitoyltransferase
PPAR
peroxisome proliferator-activated receptor α
MOI
multiplicity of infection
AUC
area under the curve
H&E
hematoxylin and eosin
TMRM
tetramethylrhodamine methyl ester perchlorate.
that has become the most common chronic liver disease (
2- Bellentani S.
- Scaglioni F.
- Marino M.
- Bedogni G.
Epidemiology of non-alcoholic fatty liver disease.
,
3The global NAFLD epidemic.
). Mitochondria play an important role in liver metabolism, as they are major contributors to both gluconeogenesis and β-oxidation (
4Inhibition of mitochondrial β-oxidation as a mechanism of hepatotoxicity.
,
). In humans with NAFLD, mitochondrial failure has been reported (
6- Lane M.
- Boczonadi V.
- Bachtari S.
- Gomez-Duran A.
- Langer T.
- Griffiths A.
- Kleinle S.
- Dineiger C.
- Abicht A.
- Holinski-Feder E.
- Schara U.
- Gerner P.
- Horvath R.
Mitochondrial dysfunction in liver failure requiring transplantation.
) as well as excessive mitochondrial oxidative capacity and gluconeogenesis (
7- Sunny N.E.
- Parks E.J.
- Browning J.D.
- Burgess S.C.
Excessive hepatic mitochondrial TCA cycle and gluconeogenesis in humans with nonalcoholic fatty liver disease.
), leaving the role of mitochondria in these pathologies unclear.
Prohibitins are a family of proteins that consist of two members: prohibitin-1 (Phb1) and prohibitin-2 (Phb2) (
8- Back J.W.
- Sanz M.A.
- De Jong L.
- De Koning L.J.
- Nijtmans L.G.
- De Koster C.G.
- Grivell L.A.
- Van Der Spek H.
- Muijsers A.O.
A structure for the yeast prohibitin complex: structure prediction and evidence from chemical crosslinking and mass spectrometry.
,
9- McClung J.K.
- Jupe E.R.
- Liu X.T.
- Dell'Orco R.T.
Prohibitin: potential role in senescence, development, and tumor suppression.
). Prohibitins are predominately located in the mitochondrial inner membrane, where Phb1 and Phb2 form large interdependent ring-shaped heteromeric complexes (
8- Back J.W.
- Sanz M.A.
- De Jong L.
- De Koning L.J.
- Nijtmans L.G.
- De Koster C.G.
- Grivell L.A.
- Van Der Spek H.
- Muijsers A.O.
A structure for the yeast prohibitin complex: structure prediction and evidence from chemical crosslinking and mass spectrometry.
,
10- Tatsuta T.
- Model K.
- Langer T.
Formation of membrane-bound ring complexes by prohibitins in mitochondria.
). Mitochondrial prohibitins have diverse roles in cellular regulations, including mitochondrial dynamics, cell proliferation, and apoptosis (
11- Merkwirth C.
- Dargazanli S.
- Tatsuta T.
- Geimer S.
- Löwer B.
- Wunderlich F.T.
- von Kleist-Retzow J.C.
- Waisman A.
- Westermann B.
- Langer T.
Prohibitins control cell proliferation and apoptosis by regulating OPA1-dependent cristae morphogenesis in mitochondria.
12- Nijtmans L.G.
- de Jong L.
- Artal Sanz M.
- Coates P.J.
- Berden J.A.
- Back J.W.
- Muijsers A.O.
- van der Spek H.
- Grivell L.A.
Prohibitins act as a membrane-bound chaperone for the stabilization of mitochondrial proteins.
,
13- Steglich G.
- Neupert W.
- Langer T.
Prohibitins regulate membrane protein degradation by the m-AAA protease in mitochondria.
14- Wei Y.
- Chiang W.C.
- Sumpter Jr, R.
- Mishra P.
- Levine B.
Prohibitin 2 is an inner mitochondrial membrane mitophagy receptor.
). Prohibitin-deficient mouse embryonic fibroblasts (MEFs) show fragmented mitochondria, defective cristae, and excessive proteolytic cleavage of the dynamin-like GTPase OPA1 (
11- Merkwirth C.
- Dargazanli S.
- Tatsuta T.
- Geimer S.
- Löwer B.
- Wunderlich F.T.
- von Kleist-Retzow J.C.
- Waisman A.
- Westermann B.
- Langer T.
Prohibitins control cell proliferation and apoptosis by regulating OPA1-dependent cristae morphogenesis in mitochondria.
). The long form of OPA1 (L-OPA1) is essential for fusion, whereas the short form of OPA1 (S-OPA1) is associated with fission (
15- Ehses S.
- Raschke I.
- Mancuso G.
- Bernacchia A.
- Geimer S.
- Tondera D.
- Martinou J.C.
- Westermann B.
- Rugarli E.I.
- Langer T.
Regulation of OPA1 processing and mitochondrial fusion by m-AAA protease isoenzymes and OMA1.
,
16- Head B.
- Griparic L.
- Amiri M.
- Gandre-Babbe S.
- van der Bliek A.M.
Inducible proteolytic inactivation of OPA1 mediated by the OMA1 protease in mammalian cells.
17- Anand R.
- Wai T.
- Baker M.J.
- Kladt N.
- Schauss A.C.
- Rugarli E.
- Langer T.
The i-AAA protease YME1L and OMA1 cleave OPA1 to balance mitochondrial fusion and fission.
). In mice,
in vivo knockout of prohibitins specifically in neurons or β-cells alters cellular functions with severe specific phenotypes, neurodegeneration or diabetes, respectively (
18- Supale S.
- Thorel F.
- Merkwirth C.
- Gjinovci A.
- Herrera P.L.
- Scorrano L.
- Meda P.
- Langer T.
- Maechler P.
Loss of prohibitin induces mitochondrial damages altering beta-cell function and survival and is responsible for gradual diabetes development.
,
19- Merkwirth C.
- Martinelli P.
- Korwitz A.
- Morbin M.
- Brönneke H.S.
- Jordan S.D.
- Rugarli E.I.
- Langer T.
Loss of prohibitin membrane scaffolds impairs mitochondrial architecture and leads to tau hyperphosphorylation and neurodegeneration.
). Stabilization of OPA1 protects against apoptosis and tissue damage, leading to the amelioration of the mitochondrial function (
20- Korwitz A.
- Merkwirth C.
- Richter-Dennerlein R.
- Tröder S.E.
- Sprenger H.G.
- Quirós P.M.
- López-Otín C.
- Rugarli E.I.
- Langer T.
Loss of OMA1 delays neurodegeneration by preventing stress-induced OPA1 processing in mitochondria.
,
21- Civiletto G.
- Varanita T.
- Cerutti R.
- Gorletta T.
- Barbaro S.
- Marchet S.
- Lamperti C.
- Viscomi C.
- Scorrano L.
- Zeviani M.
Opa1 overexpression ameliorates the phenotype of two mitochondrial disease mouse models.
). However, to what extent OPA1 contributes to the phenotypic outcomes of mitochondrial dysfunction is unclear. Furthermore, it remains controversial whether altered mitochondrial morphology is the cause or the consequence of mitochondrial dysfunction and metabolic stress. In other words, whether or not morphological adaptation serves the mitochondrial function is an ongoing debate.
To assess the role of prohibitins and related mitochondrial integrity in the liver, we generated conditional tamoxifen-inducible hepatocyte-specific knockout mice. The loss of Phb2 in hepatocytes abolished gluconeogenesis and increased cell death. These effects were associated with mitochondrial fragmentation and loss of L-OPA1. To further define the role of OPA1-dependent fusion in mitochondrial liver disease, we expressed an isoform of cleavage-resistant OPA1 (L-OPA1Δ) by intravenous injection of recombinant adenovirus. Stabilization of L-OPA1 led to elongated mitochondria with higher respiration in control hepatocytes and to only the normalization of mitochondrial morphology in the absence of Phb2. Surprisingly, OPA1 stabilization in the control mice potentiated gluconeogenesis, pointing to mitochondrial oxidative capacity as a master regulator of hepatic glucose production.
Discussion
The present study demonstrates that in vivo deletion of Phb2 in hepatocytes rapidly leads to hypoglycemia and loss of body weight, accompanied by liver lipid accumulation and hypolipidemia. Although Hep-Phb2−/− mice had preserved glucose tolerance and insulin sensitivity, glycogen stores were depleted and gluconeogenesis was inefficient with down-regulation of hepatic PEPCK-c.
During the first phase of fasting, half of the hepatic glucose production is contributed by the breakdown of glycogen and the other half by gluconeogenesis (
45- Petersen K.F.
- Price T.
- Cline G.W.
- Rothman D.L.
- Shulman G.I.
Contribution of net hepatic glycogenolysis to glucose production during the early postprandial period.
). Upon prolonged fasting and exhaustion of hepatic glycogen, gluconeogenesis contributes to nearly all of the glucose production (
46- Rothman D.L.
- Magnusson I.
- Katz L.D.
- Shulman R.G.
- Shulman G.I.
Quantitation of hepatic glycogenolysis and gluconeogenesis in fasting humans with 13C NMR.
,
47- Landau B.R.
- Wahren J.
- Chandramouli V.
- Schumann W.C.
- Ekberg K.
- Kalhan S.C.
Contributions of gluconeogenesis to glucose production in the fasted state.
). Soon after deletion of liver Phb2 (1 week), Hep-Phb2
−/− mice were unable to produce hepatic glucose, and subsequently they rapidly became continuously hypoglycemic (about 4 m
m or below). With the chronicity of hypoglycemia, hepatic glycogen contents were substantially lowered. Thus, Hep-Phb2
−/− mice were severely ill and experienced continuous hypoglycemia even under the fed conditions, ultimately leading to severe weight loss and death. The etiology of the disease, induced by the knockout of liver Phb2, indicates that the abrogation of hepatic glucose production was an early defect caused by the loss of hepatic prohibitins, in turn driving the secondary peripheral dysregulations. Our group recently reported that the selective disruption of amino acid–derived gluconeogenesis in the hepatocytes (
48- Karaca M.
- Martin-Levilain J.
- Grimaldi M.
- Li L.
- Dizin E.
- Emre Y.
- Maechler P.
Liver glutamate dehydrogenase controls whole-body energy partitioning through amino acid-derived gluconeogenesis and ammonia homeostasis.
) is not sufficient to induce such a severe phenotype as the one observed in Hep-Phb2
−/− mice, pointing to additional liver failures, such as impaired mitochondrial oxidative activity. Lipid stores rerouted to the liver of Hep-Phb2
−/− mice lacking efficient β-oxidation contributed to the accumulation of hepatic triglycerides (
50- Donnelly K.L.
- Smith C.I.
- Schwarzenberg S.J.
- Jessurun J.
- Boldt M.D.
- Parks E.J.
Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease.
).
Ablation of Phb2 in hepatocytes led to excessive proteolytic cleavage of L-OPA1, increasing the ratio of short to long isoforms. This was associated with mitochondrial fragmentation and a dramatic increase in spontaneous cytochrome
c release. Loss of L-OPA1 accounts for the defects in Phb2-null cells (
11- Merkwirth C.
- Dargazanli S.
- Tatsuta T.
- Geimer S.
- Löwer B.
- Wunderlich F.T.
- von Kleist-Retzow J.C.
- Waisman A.
- Westermann B.
- Langer T.
Prohibitins control cell proliferation and apoptosis by regulating OPA1-dependent cristae morphogenesis in mitochondria.
). Our results show that the regulation of OPA1 cleavage is a central function of prohibitins in primary hepatocytes, linking mitochondrial morphology to liver function. In the liver of leptin-deficient ob/ob mice, leptin treatment reduces both lipid content and the OPA1 ratio of short to long isoforms, restoring this ratio to untreated lean control levels (
51- Holmström M.H.
- Tom R.Z.
- Björnholm M.
- Garcia-Roves P.M.
- Zierath J.R.
Effect of leptin treatment on mitochondrial function in obese leptin-deficient ob/ob mice.
).
The
in vivo stabilization of L-OPA1 did not improve the metabolic phenotype of Hep-Phb2
−/− mice despite efficient restoration of the mitochondrial morphology. Besides maintaining long-form OPA1 integrity, prohibitins protect mitochondrial respiratory chain complexes from degradation (
12- Nijtmans L.G.
- de Jong L.
- Artal Sanz M.
- Coates P.J.
- Berden J.A.
- Back J.W.
- Muijsers A.O.
- van der Spek H.
- Grivell L.A.
Prohibitins act as a membrane-bound chaperone for the stabilization of mitochondrial proteins.
,
13- Steglich G.
- Neupert W.
- Langer T.
Prohibitins regulate membrane protein degradation by the m-AAA protease in mitochondria.
) and facilitate mitophagy to remove dysfunctional mitochondria (
14- Wei Y.
- Chiang W.C.
- Sumpter Jr, R.
- Mishra P.
- Levine B.
Prohibitin 2 is an inner mitochondrial membrane mitophagy receptor.
). L-OPA1 expression in Phb2-deficient neurons does not restore impaired respiratory supercomplexes (
20- Korwitz A.
- Merkwirth C.
- Richter-Dennerlein R.
- Tröder S.E.
- Sprenger H.G.
- Quirós P.M.
- López-Otín C.
- Rugarli E.I.
- Langer T.
Loss of OMA1 delays neurodegeneration by preventing stress-induced OPA1 processing in mitochondria.
). Because the liver has a prominent metabolic activity largely relying on mitochondria, any dysfunction of this organelle may lead to metabolic alterations (
52The pathogenesis of insulin resistance: integrating signaling pathways and substrate flux.
). Our data emphasize the specific pathways by which prohibitins and OPA1 regulate mitochondrial integrity and cellular metabolism.
One intriguing observation made here was that up-regulation of long-form OPA1 and the associated hyperfused mitochondrial pattern resulted in enhanced hepatic glucose production in prohibitin-competent mice. Such an anabolic boost was associated with an increase of the lean mass. In prohibitin-null mice, the metabolic defects were not restored, pointing to other liver alterations not restored by L-OPA1. The usual view is that mitochondrial morphology is an adaptive response to cellular metabolic demands (
53Metabolic regulation of mitochondrial dynamics.
), and mitochondrial dynamics are associated with oxidative phosphorylation (
54- Rossignol R.
- Gilkerson R.
- Aggeler R.
- Yamagata K.
- Remington S.J.
- Capaldi R.A.
Energy substrate modulates mitochondrial structure and oxidative capacity in cancer cells.
,
55- Mishra P.
- Carelli V.
- Manfredi G.
- Chan D.C.
Proteolytic cleavage of Opa1 stimulates mitochondrial inner membrane fusion and couples fusion to oxidative phosphorylation.
). Starvation promotes an elongated pattern of mitochondria, presumably to maximize ATP production (
56- Rambold A.S.
- Kostelecky B.
- Elia N.
- Lippincott-Schwartz J.
Tubular network formation protects mitochondria from autophagosomal degradation during nutrient starvation.
), whereas high glucose conditions result in massive mitochondrial fragmentation (
57- Yu T.
- Robotham J.L.
- Yoon Y.
Increased production of reactive oxygen species in hyperglycemic conditions requires dynamic change of mitochondrial morphology.
). However, as most of the morphology-related studies were conducted on nutrient-consuming cell lines, there is no preceding report on the effects of hyperfused mitochondria in glucose production by hepatocytes. Mitochondria serve as a hub for metabolic enzymes, such as GCK (
33- Danial N.N.
- Gramm C.F.
- Scorrano L.
- Zhang C.Y.
- Krauss S.
- Ranger A.M.
- Datta S.R.
- Greenberg M.E.
- Licklider L.J.
- Lowell B.B.
- Gygi S.P.
- Korsmeyer S.J.
BAD and glucokinase reside in a mitochondrial complex that integrates glycolysis and apoptosis.
) and PEPCK-m (
58The mitochondrial isoform of phosphoenolpyruvate carboxykinase (PEPCK-M) and glucose homeostasis: has it been overlooked?.
). Interaction of GCK with mitochondria has been implicated in the control of glycolysis and apoptosis in hepatocytes (
33- Danial N.N.
- Gramm C.F.
- Scorrano L.
- Zhang C.Y.
- Krauss S.
- Ranger A.M.
- Datta S.R.
- Greenberg M.E.
- Licklider L.J.
- Lowell B.B.
- Gygi S.P.
- Korsmeyer S.J.
BAD and glucokinase reside in a mitochondrial complex that integrates glycolysis and apoptosis.
). Thus, the level of expression
per se of those enzymes is most likely not the key element for metabolic regulation. The interaction between metabolic enzymes and mitochondria seems to play a central role in the control of these pathways.
Increased hepatic glucose production is the primary contributor to fasting hyperglycemia (
59- Magnusson I.
- Rothman D.L.
- Katz L.D.
- Shulman R.G.
- Shulman G.I.
Increased rate of gluconeogenesis in type II diabetes mellitus: a 13C nuclear magnetic resonance study.
,
60- Wajngot A.
- Chandramouli V.
- Schumann W.C.
- Ekberg K.
- Jones P.K.
- Efendic S.
- Landau B.R.
Quantitative contributions of gluconeogenesis to glucose production during fasting in type 2 diabetes mellitus.
), which is a hallmark in the natural history of type 2 diabetes. However, neither PEPCK nor G6Pase are up-regulated in the liver of type 2 diabetic patients (
61- Samuel V.T.
- Beddow S.A.
- Iwasaki T.
- Zhang X.M.
- Chu X.
- Still C.D.
- Gerhard G.S.
- Shulman G.I.
Fasting hyperglycemia is not associated with increased expression of PEPCK or G6Pc in patients with type 2 diabetes.
), indicating that alternative molecular mechanisms account for excessive glucose production. The present data indicate that mitochondrial dynamics participate in hepatic glucose production. In particular, L-OPA1 not only elongated mitochondria but also rendered hepatocytes hyperresponsive in terms of mitochondrial respiration and glucose production (
Fig. 7C). This is reminiscent of the excessive mitochondrial oxidative capacity and gluconeogenesis observed in NAFLD humans (
6- Lane M.
- Boczonadi V.
- Bachtari S.
- Gomez-Duran A.
- Langer T.
- Griffiths A.
- Kleinle S.
- Dineiger C.
- Abicht A.
- Holinski-Feder E.
- Schara U.
- Gerner P.
- Horvath R.
Mitochondrial dysfunction in liver failure requiring transplantation.
,
7- Sunny N.E.
- Parks E.J.
- Browning J.D.
- Burgess S.C.
Excessive hepatic mitochondrial TCA cycle and gluconeogenesis in humans with nonalcoholic fatty liver disease.
). However, an elongated pattern of mitochondria is not sufficient for an overproduction of glucose, as shown in Phb2-null hepatocytes expressing L-OPA1.
In conclusion, our results establish an essential role for prohibitins in regulating hepatic metabolism and whole-body energy homeostasis. In the absence of prohibitins, L-OPA1 is sufficient to restore the morphology but not the function of liver mitochondria. In the presence of prohibitins, L-OPA1 promotes excessive mitochondrial respiration and glucose production, pointing to mitochondrial dynamics in the control of gluconeogenesis.
Experimental procedures
Generation of hepatocyte-specific prohibitin-2 knockout (Hep-Phb2−/−) mice
Phb2 floxed (Phb2
fl/fl) mice (
11- Merkwirth C.
- Dargazanli S.
- Tatsuta T.
- Geimer S.
- Löwer B.
- Wunderlich F.T.
- von Kleist-Retzow J.C.
- Waisman A.
- Westermann B.
- Langer T.
Prohibitins control cell proliferation and apoptosis by regulating OPA1-dependent cristae morphogenesis in mitochondria.
) were crossed with Alb-Cre-ER
T2 mice carrying the tamoxifen-dependent Cre-ER
T2 recombinase coding sequence preceded by an internal ribosomal entry site inserted in the 3′-UTR of the serum albumin gene (MGI:3052812) (
24- Schuler M.
- Dierich A.
- Chambon P.
- Metzger D.
Efficient temporally controlled targeted somatic mutagenesis in hepatocytes of the mouse.
). The
in vivo deletion of Phb2 in hepatocytes was induced at 8 weeks of age by subcutaneous implantation of tamoxifen pellets (tamoxifen free base, 25 mg/pellet, 21-day release, E-361; Innovative Research of America) in male Hep-Phb2
−/− mice. Animals were maintained on a mixed (C57BL/6J x 129/Sv) genetic background to avoid inbred strain-specific phenotypes. As control mice, we used male Phb2
fl/fl littermates, which were implanted with tamoxifen pellets at the same time as Hep-Phb2
−/− mice, to optimize standardization of the genetic background between the two groups. Mice were maintained in our certified animal facility on a 12-h dark-light cycle and were fed
ad libitum with standard chow diet (RM3-E-SQC catalog no. 811181, SDS Diets, Essex, UK) and water according to procedures that were approved by the animal care and experimentation authorities of the Canton of Geneva (GE/128/15, approval no. 27139).
Adenovirus construction
Recombinant adenovirus encoding for the FLAG-tagged cleavage-resistant isoform of rat OPA1 splice variant 7 (L-OPA1Δ) (
62- Ishihara N.
- Fujita Y.
- Oka T.
- Mihara K.
Regulation of mitochondrial morphology through proteolytic cleavage of OPA1.
) or mitochondrial targeted dsRED (mitoRFP) under the cytomegalovirus (CMV) promoter were generated using the Adeno-X Expression system (Clontech) according to the manufacturer's protocol and as described previously (
63- Grimaldi M.
- Karaca M.
- Latini L.
- Brioudes E.
- Schalch T.
- Maechler P.
Identification of the molecular dysfunction caused by glutamate dehydrogenase S445L mutation responsible for hyperinsulinism/hyperammonemia.
). A clonal stock was amplified and purified for its
in vivo use by Vector BioLabs (Malvern, PA). Adenovirus was administered
in vivo by tail vein injection of 4 × 10
10 pfu/kg for L-OPA1Δ expression or 3.2 × 10
10 pfu/kg for mitoRFP expression. For i
n vitro transduction, hepatocytes were treated with adenoviruses expressing either control MitoRFP or L-OPA1Δ at 5 MOI right after isolation and used 48 h later.
Isolation of primary mouse hepatocytes
Mice were anesthetized by intraperitoneal injection of Pentothal (50 μg/ml, 50 μl/25 g mouse body weight). Liver was perfused, and hepatocytes were isolated as described previously (
48- Karaca M.
- Martin-Levilain J.
- Grimaldi M.
- Li L.
- Dizin E.
- Emre Y.
- Maechler P.
Liver glutamate dehydrogenase controls whole-body energy partitioning through amino acid-derived gluconeogenesis and ammonia homeostasis.
). Collected cells were resuspended with 10 ml of culture medium (Williams E medium, 5% fetal calf serum, 10
−9 m insulin, 10
−6 m dexamethasone, penicillin/streptomycin, and 1% GlutaMAX). Hepatocytes were seeded at 50,000 cells/cm
2 on collagen-coated plates and incubated at 37 °C, 5% CO
2 until further experiments.
In vivo experiments
From the time of tamoxifen implantation, mice were separated from their cage mates and housed individually. Blood glucose levels were measured using an Accu-Check Aviva glucometer (Roche Diagnostics, Zürich, Switzerland) from the tail vein between 7:30 and 8:30 a.m. with body weight and food intake monitored at the same time. Body composition was assessed by an EchoMRI-700TM quantitative NMR analyzer (Echo Medical Systems, Houston, TX). For the glucose and pyruvate tolerance test, mice were fasted for 6 h. d-Glucose (2 g/kg body weight) or sodium pyruvate (2 g/kg body weight) was injected intraperitoneally. Glucose concentrations were measured from blood samples taken from the tail vein at time 0, 15, 30, 60, 90, and 120 min after injection using an Accu-Check Aviva glucometer. The total area under the curve (AUC) of glycemia was calculated using GraphPad Prism version 6 software. For the insulin tolerance test, mice under the fed conditions were injected with insulin (0.75 units/kg body weight, Actrapid HM, 100 units/ml, Novo Nordisk) intraperitoneally. Upon sacrifice, blood was collected into EDTA-coated tubes (Sarstedt catalog no. 20.1341) via retro-orbital bleeding and centrifuged at 2,000 rpm at 4 °C to separate plasma. Liver and epididymal adipose tissue were collected, weighed, and snap-frozen in liquid nitrogen. Tissues and plasma were stored at −80 °C. Pancreas was collected for fixation and further immunohistochemistry.
Determination of metabolic parameters
Alanine aminotransferase and aspartate aminotransferase, bilirubin, and cholesterol plasma levels were measured at Geneva University Hospitals. Plasma free fatty acids, plasma β-hydroxybutyrate, plasma and liver triglycerides, and liver glycogen content were measured by a colorimetric assay using adequate quantification kits (Biovision, Mountain View, CA) following the manufacturer's instructions. Under fed conditions, the activity of the MTP was measured in liver lysates by fluorimetric assay using the MTP Activity Assay Kit (Sigma). Insulin, glucagon, IL-6, and TNFα were measured using Luminex xMAPTM technology and commercially available kits (Bio-Plex Pro Diabetes Assays and Bio-Plex Pro Mouse Cytokine, Bio-Rad).
Glucose production by isolated hepatocytes
Glucose production was assessed as described previously (
48- Karaca M.
- Martin-Levilain J.
- Grimaldi M.
- Li L.
- Dizin E.
- Emre Y.
- Maechler P.
Liver glutamate dehydrogenase controls whole-body energy partitioning through amino acid-derived gluconeogenesis and ammonia homeostasis.
). In brief, isolated hepatocytes were fasted for 6 h in Dulbecco's modified Eagle's medium (without glucose, pyruvate, or phenol red) for glycogen depletion. The medium was replaced by Krebs-Ringer-Bicarbonate-Hepes (KRBH) buffer (140 m
m NaCl, 3.6 m
m KCl, 0.5 m
m NaH
2PO
4, 0.5 m
m MgSO
4, 2 m
m NaHCO
3, 1.5 m
m CaCl
2, and 10 m
m HEPES) alone for basal conditions or KRBH buffer containing 10 m
m lactate and 10 m
m pyruvate for stimulatory conditions. Cells were stimulated for 60 min at 37 °C. Supernatant was taken for glucose quantification with a glucose assay kit (Biovision) and later normalized by protein content.
Immunoblotting
Frozen liver tissues (30 mg) were lysed in 300 μl of radioimmune precipitation assay buffer in the presence of 1× protease inhibitor mix (Complete mini tablets, Roche Diagnostics) and 1× phosphatase inhibitor (PhosStop, Roche Diagnostics). Homogenate was collected after homogenization performed using a tissue lyser (Qiagen) and centrifugation at 14,000 rpm for 10 min at 4 °C. Proteins from liver extracts (20 mg) were resolved by gel electrophoresis, transferred on nitrocellulose membranes, and probed with specific antibodies (
Table S1). Band signals were detected by a horseradish peroxidase system, and quantifications were performed using the PXi gel imaging system (Syngene).
Histology
For morphology analyses, livers were fixed in 4% paraformaldehyde, dehydrated, and embedded in paraffin. Sections (5 μm) were stained with hematoxylin and eosin (H&E). For Oil Red O staining, frozen cryostat sections (5 μm) were fixed in ice-cold 10% formalin for 10 min, dried, washed, and incubated in propylene glycol for 5 min. The sections were then incubated with Oil Red O solution (Sigma catalog no. O1516) for 10 min at 60 °C and in 85% propylene glycol for 5 min. The sections were counterstained with hematoxylin. Sections were scanned by a widefield slide scanner (Axio Scan.Z1, Zeiss) at ×20 magnification. Images were visualized by ZEN software (Zeiss).
Immunofluorescence
For live imaging of primary hepatocytes, cells were cultured on collagen (Sigma C7661)-coated 35-mm diameter tissue culture dishes with a coverglass bottom (WPI) and incubated with MitoTracker Orange CMTMRos (100 n
m, Molecular Probes) and BODIPY dye (1 μg/ml; D3922, Molecular Probes). The preparation was put inside a temperature-controlled chamber (37 °C) during imaging, and images were acquired with an inverted Nikon A1r microscope. For quantification of mitochondrial morphology, hepatocytes were classified according three patterns of mitochondrial morphology (
i.e. fragmented, intermediate, and hyperfused (see
Fig. S8)). For insulin/glucagon staining on pancreas sections, pancreas was fixed for 2 h in 4% paraformaldehyde and finally embedded in paraffin. Sections of 5 μm were incubated with guinea pig anti-swine insulin (Dako 564) and mouse anti-glucagon (Sigma G2654) overnight at 4 °C. Secondary goat anti-guinea pig Alexa Fluor 488 antibody (Invitrogen A-11073) and goat anti-rabbit Alexa Fluor 546 antibody (Invitrogen A-11035) were added for 1 h. To monitor the
in vivo transduction efficiency of adenovirus, frozen cryostat sections were stained with DYKDDDDK tag mAb with Alexa Fluor 488 conjugate (Invitrogen MA1-142-A488) overnight at 4 °C. Images were captured by a Zeiss LSM 800 microscope.
Transmission EM
The livers from control and Hep-Phb2
−/− mice 2 weeks after tamoxifen-induced recombination were excised, cut into small pieces of 1 mm
2, and processed for EM. The tissue was fixed in 0.1
m sodium cacodylate buffer containing 2.5% glutaraldehyde at room temperature for 4 h. Samples were then washed three times with 0.1
m sodium cacodylate buffer and post-fixed with 1% osmium tetraoxide and 1.5% potassium ferrocyanide in 0.1
m cacodylate buffer for 1 h at room temperature. Samples were then dehydrated, embedded in epon resin, and processed for EM as described previously (
64- Foti M.
- Carpentier J.L.
- Aiken C.
- Trono D.
- Lew D.P.
- Krause K.H.
Second-messenger regulation of receptor association with clathrin-coated pits: a novel and selective mechanism in the control of CD4 endocytosis.
). Ultrathin sections were finally contrasted with uranyl acetate and lead citrate and observed with a Technai 20 electron microscope (FEI Co., Eindhoven, Netherlands). The same procedure was applied on isolated primary hepatocytes for EM analysis.
Mitochondrial membrane potential analysis
Cells were incubated at room temperature for 20 min with 20 nm TMRM, washed, and kept at 37 °C on the microscope until signal reached stability. CCCP (10 μm) was added, and sequential images of TMRM fluorescence were acquired every 3 s with an inverted Nikon A1r microscope. Analysis of TMRM fluorescence over mitochondrial regions of interest was performed using ImageJ software.
Cytochrome c release analysis
Hepatocytes were treated with adenovirus expressing L-OPA1Δ at 5 MOI right after isolation. After 48-h incubation at 37 °C, 5% CO2, general caspase inhibitor Z-VAD-fmk (50 μm; BD Biosciences 550377) was added to culture medium. After 3 h, cells were fixed and permeabilized. After blocking on the coverslips, cytochrome c was stained using mouse anti-cytochrome c antibody (BD Biosciences 556432), and mitochondria were stained using rabbit anti-Tom20 mAb (Cell Signaling 42406) overnight at 4 °C. Secondary goat anti-mouse Alexa Fluor 546 antibody (Invitrogen A-11030) and goat anti-rabbit Alexa Fluor 488 antibody (Invitrogen A-11034) were added for 1 h. The nucleus was stained with 4′,6-diamidino-2-phenylindole (Vector Laboratories H-1500). Images were acquired by a Nikon A1r microscope or Zeiss LSM 800 microscope. The number of cells releasing cytochrome c was determined using ImageJ.
Mitochondrial respiration measurement
The mitochondrial respiration on isolated hepatocytes was tested in a XF96 Seahorse apparatus (Agilent). Hepatocytes were plated at 10,000 cells/well in a Seahorse XF96 cell culture microplate. One h prior to the measurement, cells were washed and preincubated in PBS containing 2 mm CaCl2, 2 mm MgCl2, 5 mm glucose, and 5 mm sodium pyruvate in a non-CO2 37 °C incubator. Basal measurement was run for three cycles, followed by the sequential addition of 2 μm oligomycin (run for two cycles), 1 μm FCCP (two cycles), and 1 μm rotenone/antimycin A (two cycles). The calculation of various mitochondrial parameters was done as follows: ATP production = OCR (basal) − OCR (oligomycin-stimulated); total respiration = OCR (basal) − OCR (rotenone/antimycin A–stimulated); proton leak = OCR (total respiration) − OCR (ATP production); maximum respiration = OCR (FCCP) − OCR (rotenone/antimycin A–stimulated).
Plasma lactate measurement
Plasma samples were added to an assay buffer containing 37 mm glycine, 10 mm EDTA, 0.02% hydrazine hydrate, and 0.9 mm NAD+ and then stimulated with 2.75 units/ml l-lactate dehydrogenase (Roche Diagnostics catalog no. 000000010127230001). Lactate was measured as NADH fluorescence with a Fluostar Optima using an excitation wavelength of 340 nm and recording wavelength at 460 nm.
Gene expression analysis
Total RNA from frozen liver was isolated using TRIzol reagent (Invitrogen) and purified with Nucleospin RNA II kit (Macherey-Nagel, Duren, Germany). The primers were designed using the Primer Express Software (Applera Europe) (see
Table S2). Real-time PCR was performed using an ABI 7000 sequence detection system (Applera Europe), and PCR products were quantified fluorometrically using the SYBR Green core reagent kit (Life Technologies, Inc.). The values obtained were normalized to values of the housekeeping gene 18S rRNA.
Statistical analysis
Statistical analyses were performed using GraphPad Prism version 6 software, with one-way analysis of variance when more than two groups of data were compared and with nonparametric Mann–Whitney U test when only two groups of data were concerned. A p value < 0.05 was considered significant.
Author contributions
L. L., J. M.-L., M. F., J.-C. M., and P. M. conceived and designed the experiments. L. L., M. K., C. J. S., M. F., and J. M.-L. performed the experiments. L. L., J. M.-L., C. J. S., M. K., M. F., and P. M. performed data analysis. L. L., J. M.-L., and P. M. wrote the paper.
Article info
Publication history
Published online: July 08, 2019
Received in revised form:
June 14,
2019
Received:
January 17,
2019
Edited by Jeffrey E. Pessin
Footnotes
This work was supported by Swiss National Science Foundation Sinergia Grants CRSII3_147637 (to P. M. and J.-C. M.) and 310030_172862 (to M. F.) and by a Fellowship from the Fundación Alfonso Martín Escudero (to C. J. S). The authors declare that they have no conflicts of interest with the contents of this article.
This article was selected as one of our Editors' Picks.
This article contains Tables S1 and S2 and Figs. S1–S8.
Copyright
© 2019 Li et al.