In vivo stabilization of OPA1 in hepatocytes potentiates mitochondrial respiration and gluconeogenesis in a prohibitin-dependent way
←
→
Page content transcription
If your browser does not render page correctly, please read the page content below
JBC Papers in Press. Published on July 8, 2019 as Manuscript RA119.007601 The latest version is at http://www.jbc.org/cgi/doi/10.1074/jbc.RA119.007601 Mitochondrial dynamics and liver gluconeogenesis In vivo stabilization of OPA1 in hepatocytes potentiates mitochondrial respiration and gluconeogenesis in a prohibitin-dependent way Lingzi Li1,2,#, Juliette Martin-Levilain1,2,#, Cecilia Jiménez-Sánchez1,2, Melis Karaca1,2, Michelangelo Foti1,2, Jean-Claude Martinou3, Pierre Maechler1,2* 1 Department of Cell Physiology and Metabolism & 2Faculty Diabetes Centre, University of Geneva Medical Centre, Geneva, Switzerland; 3Cell Biology Department, Faculty of Sciences, University of Geneva, Geneva, Switzerland Running title: mitochondrial dynamics and liver gluconeogenesis # These authors contributed equally to this work *Correspondence to: Pierre Maechler, Department of Cell Physiology and Metabolism, CMU, rue Michel-Servet 1, 1211 Geneva 4, Switzerland; Pierre.Maechler@unige.ch; Tel: +41 22 379 55 54 Keywords: liver; gluconeogenesis; mitochondria; prohibitins; OPA1 Downloaded from http://www.jbc.org/ by guest on July 18, 2019 _______________________________________________________________________________ ABSTRACT participates in the control of hepatic glucose production. Patients with fatty liver diseases present altered mitochondrial morphology and impaired metabolic _________________________________________ function. Mitochondrial dynamics and related cell function require the uncleaved form of the The liver is essential for both glucose and fatty dynamin-like GTPase OPA1. Stabilization of acid metabolisms, producing glucose in the fasting OPA1 might then confer protective mechanism state via glycogenolysis and gluconeogenesis (1). against stress-induced tissue damages. In order to Hepatic lipid accumulation characterizes non- study the putative role of hepatic mitochondrial alcoholic fatty liver disease (NAFLD) that has morphology in a sick liver, we expressed a become the most common chronic liver disease cleavage-resistant long form of OPA1 (L-OPA1Δ) (2,3). Mitochondria play an important role in liver in the liver of a mouse model with mitochondrial metabolism as they are major contributors to both liver dysfunction, i.e. the hepatocyte-specific gluconeogenesis and ß-oxidation (4,5). In humans prohibitin-2 knockout (Hep-Phb2-/-) mice. Liver with NAFLD, mitochondrial failure has been prohibitin-2 deficiency caused excessive reported (6) as well as excessive mitochondrial proteolytic cleavage of L-OPA1, mitochondrial oxidative capacity and gluconeogenesis (7), fragmentation, and increased apoptosis. These leaving the role of mitochondria in these molecular alterations were associated with lipid pathologies unclear. accumulation, abolished gluconeogenesis and extensive liver damages. Such liver dysfunction Prohibitins are a family of proteins that consist of was associated with severe hypoglycaemia. In two members: prohibitin-1 (Phb1) and prohibitin-2 prohibitin-2 knockout mice, expression of L- (Phb2) (8,9). Prohibitins are predominately located OPA1Δ by in vivo adenovirus delivery restored the in the mitochondrial inner membrane, where Phb1 morphology but not the function of mitochondria and Phb2 form large interdependent ring-shaped in hepatocytes. In prohibitin-competent mice, heteromeric complexes (8,10). Mitochondrial elongation of liver mitochondria by expression of prohibitins have diverse roles in cellular L-OPA1Δ resulted in excessive glucose production regulations including mitochondrial dynamics, cell associated with increased mitochondrial proliferation, and apoptosis (11-14). Prohibitin- respiration. In conclusion, mitochondrial dynamics deficient mouse embryonic fibroblasts (MEFs) 1
Mitochondrial dynamics and liver gluconeogenesis show fragmented mitochondria, defective cristae with mice expressing a tamoxifen-inducible Cre and excessive proteolytic cleavage of the dynamin- recombinase under the control of the albumin like GTPase OPA1 (11). The long form of OPA1 promoter (Alb-Cre-ERT2) (24). Non-recombined (L-OPA1) is essential for fusion, whereas the short Phb2fl/fl;Alb-Cre-ERT2 mice were bred at the form of OPA1 (S-OPA1) is associated with fission expected Mendelian ratio with normal gross (15-17). In mice, in vivo knockout of prohibitins anatomy and fertility. We used floxed littermates specifically in neurons or ß-cells alter cellular without the Cre transgene (Phb2fl/fl) as controls for functions with severe specific phenotypes, experiments in order to homogenize the genetic respectively neurodegeneration or diabetes (18,19). background between the groups. Tamoxifen Stabilization of OPA1 protects against apoptosis treatment was initiated at the age of 8 weeks on and tissue damage, leading to the amelioration of both Phb2fl/fl controls and Phb2fl/fl;Alb-Cre-ERT2 the mitochondrial function (20,21). However, to mice by one-time subcutaneous implantation of a which extent OPA1 contributes to the phenotypic tamoxifen pellet, the latter producing hepatocyte- outcomes of mitochondrial dysfunction is unclear. specific Phb2 knockout (Hep-Phb2-/-) mice. Two Furthermore, it remains controversial whether weeks after tamoxifen administration, altered mitochondrial morphology is the cause or immunoblotting on extracts of isolated hepatocytes the consequence of mitochondrial dysfunction and revealed close to complete deletion of Phb2 in metabolic stress. In other words, whether or not Hep-Phb2-/- mice (Figure 1A). Consistent with morphological adaptation serves the mitochondrial previous findings in mice and cell lines function is an ongoing debate. (11,18,19,25), loss of Phb2 was accompanied by Downloaded from http://www.jbc.org/ by guest on July 18, 2019 depletion of Phb1. This observation indicates that, In order to assess the role of prohibitins and as for other tissues, Phb1 and Phb2 are related mitochondrial integrity in the liver, we functionally interdependent in hepatocytes. generated conditional tamoxifen-inducible Immunoblotting of liver lysates from Hep-Phb2-/- hepatocyte-specific knockout mice. The loss of mice showed residual Phb2, which is presumably Phb2 in hepatocytes abolished gluconeogenesis explained by expression of the gene in non- and increased cell death. These effects were parenchymal liver cells and the persistence of associated with mitochondrial fragmentation and some hepatocytes that eventually escaped loss of L-OPA1. To further define the role of recombination. No recombination was observed in OPA1-dependent fusion in the mitochondrial liver the pancreas, skeletal muscles, the kidney and the disease, we expressed an isoform of cleavage- brain of Hep-Phb2-/- mice (Figure 1A and Suppl. resistant OPA1 (L-OPA1Δ) by intravenous Figure S1A). injection of recombinant adenovirus. Stabilization of L-OPA1 led to elongated mitochondria with About 1 week after tamoxifen-induced higher respiration in control hepatocytes and to recombination was initiated, Hep-Phb2-/- mice only the normalization of mitochondrial showed a progressive decline of random plasma morphology in the absence of Phb2. Surprisingly, glucose levels and of their body weights (Figure OPA1 stabilization in the control mice potentiated 1B-C). At 2 weeks post-recombination, food gluconeogenesis, pointing to mitochondrial consumption (Suppl. Figure S1B) was lower (- oxidative capacity as a master regulator of hepatic 25%) in knockout animals and by 3 weeks they glucose production. were severely sick and consumed approximately 63% less food than their controls. The pro- inflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor (TNF)-α were not Results significantly different in Hep-Phb2-/- mice at 2- weeks post-recombination despite of a trend for Generation and phenotype of hepatocyte-specific elevated plasma levels (Suppl. Figure S1C-D), while at 3-weeks IL-6 and TNF-α levels were Phb2 knockout mice dramatically higher compared to controls (TNF-α concentrations were out of range). Hep-Phb2-/- Global deletion of prohibitins is embryonically mice died at around 3-4 weeks after tamoxifen- lethal in mice (22,23). To achieve post-natal gene induced deletion of prohibitins. Liver weights of ablation specifically in mouse hepatocytes, Hep-Phb2-/- mice were reduced by 15% after 3 conditional Phb2 mice (Phb2fl/fl) (11) were bred 2
Mitochondrial dynamics and liver gluconeogenesis weeks of recombination versus controls (Figure microsomal triglyceride transfer protein (MTP) 1D-E). There was a marked reduction of the activity as a rate-limiting factor (28). Under fed epididymal white adipose tissue (eWAT) in Hep- condition, Hep-Phb2-/- mice had 1.5-fold higher Phb2-/- mice, being 55% and 85% lower compared MTP activity than the control mice, probably to control mice 2 and 3 weeks after recombination, driven by over-sufficient supply of hepatic respectively (Figure 1F). Histology on liver triglycerides (Figure 2G). Considering the marked sections (Figure 1G) of Hep-Phb2-/- mice revealed reduction of eWAT (a sign of increased lipolysis) disorganized hepatic structures and small lipid and potential higher hepatic VLDL formation, one droplets, reminiscent of NAFLD. On the other would expect plasma lipid levels to be increased in hand, steatotic liver of ob/ob mice did not exhibit Hep-Phb2-/- mice. On the contrary, Hep-Phb2-/- changes in Phb2 (Suppl. Figure S1E), while others mice exhibited hypolipidemia, suggesting reported reduced levels of prohibitin complex in increased clearance by peripheral tissues such as liver mitochondria of ob/ob mice (26). Oil Red O the skeletal muscles. Although the liver normally staining further confirmed the accumulation of increases the production of ketone bodies from lipids in the livers of Hep-Phb2-/- mice. It has been fatty acids under low blood glucose conditions, reported that high dose of tamoxifen induces plasma levels of ß-hydroxybutyrate were similar hepatotoxicity (27). However, we observed no between the knockout and control mice (Figure change in liver histology 3 weeks after tamoxifen 2H). Overall, loss of prohibitins induced liver administration on the Cre-negative control floxed failure and a dramatic imbalance of lipid storage mice, which disqualifies tamoxifen as a from adipose tissue to the liver. Downloaded from http://www.jbc.org/ by guest on July 18, 2019 confounding factor for the observed phenotype. This set of data shows that the loss of hepatic Loss of liver prohibitins altered glucose prohibitins resulted in hypoglycaemia, reduction of metabolism and whole-body energy homeostasis body weight as well as altered liver mass and histology. Consistent with their low glycemia, Hep-Phb2-/- mice displayed 70% reduction of hepatic glycogen Loss of prohibitins induced liver injury and content (fed condition) compared with the control altered lipid metabolism mice (Figure 2I). In parallel, plasma insulin levels were reduced by 58% in Hep-Phb2-/- mice, along Due to the severity of the phenotype observed in with a trend for increased glucagon levels (Figure Hep-Phb2-/- mice at about 3-4 weeks after 2J-K). As insulin and glucagon are produced by recombination, we then centred the study when the pancreatic islets, we examined the islets by disease was less severe, i.e. 2 weeks after immunohistochemistry. We observed no recombination. Potential hepatic damages due to significant changes in the knockout mice regarding Phb2 deficiency were evaluated by the analysis of neither the pancreatic distribution of islets nor the liver enzymes and bilirubin in plasma, which were density of endocrine cells within the islets with the markedly increased in Hep-Phb2-/- mice versus expected organisation of α-cells at the periphery controls (Figure 2A, B). These data indicate and ß-cells predominately in the centre (Suppl. significant liver damages in Hep-Phb2-/- mice Figure S2). before the induction of cytokines (Suppl. Figure S1), illness appearing already 2 weeks after the Despite of the low plasma glucose and insulin loss of hepatic Phb2. levels, Hep-Phb2-/- mice showed similar glucose tolerance in response to an intraperitoneal glucose We next analysed plasma and hepatic lipid profiles. tolerance test (ipGTT) and robust insulin The plasma concentrations of free fatty acid (FFA), sensitivity upon intraperitoneal insulin tolerance total cholesterol and triglycerides (Figure 2C-E) test (ipITT), compared with the control mice were lower in Hep-Phb2-/- mice compared to (Figure 3A-B). Remarkably, starting from 1-week controls. Conversely, liver triglyceride contents post recombination, gluconeogenesis was (Figure 2F) were markedly increased in Hep-Phb2- completely abolished in Hep-Phb2-/- mice as /- mice versus controls, confirming the lipid indicated by in vivo pyruvate challenges (Figure accumulation revealed by histology. Hepatic 3C-E), concomitant with the appearance of triglycerides are released into the bloodstream via hypoglycaemia (Figure 1B). Of note, the in vivo packaging with apolipoprotein B into VLDL, with pyruvate challenge may be influenced by the 3
Mitochondrial dynamics and liver gluconeogenesis uptake of lactate/pyruvate by peripheral tissues Next, we examined the protein levels of lipogenic competing for the substrate (29). However, the enzymes in the liver. Both acetyl-CoA carboxylase impaired gluconeogenesis was also observed in (ACC) and fatty acid synthase (FAS) were vitro on hepatocytes isolated from Hep-Phb2-/- markedly reduced in the liver of Hep-Phb2-/- mice mice (see later). (Figure 3F). This shows defective capacity for de novo lipogenesis in Phb2-deficient liver despite the We next analysed the proteins implicated in observed lipid accumulation, pointing to a non- glucose and lipid metabolisms in liver extracts by hepatic source contributing to increased immunoblotting (Figure 3F). Insulin signalling triglyceride storage. Given that Hep-Phb2-/- mice pathway was first investigated for its role in eat less (Suppl. Figure S1B), the few dietary lipids hepatic steatosis (30). Binding of insulin to its from their chow diet hardly account for increased receptor initiates a signalling cascade that diverges hepatic lipid content. As insulin inhibits lipase in at protein kinase Akt. The absence of Phb2 caused white adipose tissue, the low plasma insulin levels a 50% reduction of phospho-Akt Ser473, while (Figure 2J) might have contributed to promote phospho-Akt Thr308 was unchanged. Downstream lipolysis in adipose tissue (Figure 1F), resulting in of insulin-Akt signalling, glycogen synthase kinase elevated supply of hepatic lipids in Hep-Phb2-/- 3 (GSK3) inhibits glycogen synthase (GS) activity liver (Figure 2F). by phosphorylation, whereas Akt promotes GS activity by inhibitory phosphorylation of GSK3. PEPCK and ACC directly rely on mitochondrial Expression of GSK3ß in Hep-Phb2-/- liver was activity for the supply of their respective substrates Downloaded from http://www.jbc.org/ by guest on July 18, 2019 markedly increased, associated with higher and GCK is associated with liver mitochondria, phosphorylated GSK3. Glucokinase (GCK) is integrating glucose metabolism and apoptosis (33). induced by insulin and converts glucose to Since these proteins were markedly downregulated glucose-6-phosphate (G6P) used for both in prohibitin-deficient liver, we investigated the glycolysis and glycogen synthesis. In the liver of integrity of mitochondria normally contributed by Hep-Phb2-/- mice, GCK protein levels were prohibitins. Electron microscopy showed the loss decreased, possibly as a consequence of low of an elongated ultrastructure resulting in a insulin levels. In Phb2-deficient livers, GS was globular pattern in Hep-Phb2-/- liver (Figure 3G). markedly downregulated and the remaining GS This might indicate a link between the structure of was phosphorylated to levels similar to those of mitochondria and the levels of proteins associated control liver, further reducing its overall activity. with this organelle. Regarding glycogenolysis, expression of glycogen phosphorylase was also markedly reduced. Overall, L-OPA1 restored mitochondrial morphology and while the signalling pathway for glycogen improved resistance to spontaneous apoptosis in production was upregulated, the enzymatic Phb2-deficient hepatocytes machinery was markedly downregulated in Phb2- deficient liver. Moreover, the scarce availability of Deletion of Phb2 in MEFs, neurons and pancreatic the necessary substrates rendered glycogen storage ß-cells leads to excessive proteolytic cleavage of inoperative (31). L-OPA1, which is accompanied by mitochondrial fragmentation (11,18,19) associated with the Although not rate-limiting (32), observed globular pattern (Figure 3G). Re- phosphoenolpyruvate carboxykinase (PEPCK) introducing L-OPA1 into Phb2 null cells promotes significantly contributes to hepatic fusion and therefore recovery of the mitochondrial gluconeogenesis and is localized mainly in the morphology (11). To understand whether loss of cytosol (PEPCK-c) and partly in mitochondria L-OPA1 and the associated disrupted (PEPCK-m) according to its two isoforms. mitochondrial dynamics were responsible for the Expression of PEPCK-c is stimulated by glucagon functional defect of Phb2-deficient hepatocytes, and repressed by insulin, whereas PEPCK-m is we used a cleavage-resistant L-OPA1 isoform (L- constitutively expressed. Phb2-deficient liver had OPA1Δ). Flag-tagged isoform of L-OPA1Δ was almost complete suppression of PEPCK-c protein introduced by adenoviral gene transfer into (Figure 3F), while glucose-6-phosphatase (G6Pase) isolated hepatocytes and its expression was was similarly expressed in the knockout and detected by immunoblotting (Figure 4A). As control livers. expected, we detected 5 different OPA1 cleavage 4
Mitochondrial dynamics and liver gluconeogenesis forms in mouse hepatocytes: long (L) forms a and which was prevented by the expression of L- b derived from splice variants 7 and 1, respectively, OPA1Δ (Figure 4E). We also examined the and short forms c, d and e produced by proteolytic production of reactive oxygen species (ROS) by cleavage at S1 and S2 sites. Following L-OPA1Δ measurement of lipid peroxidation derived 4- transduction of isolated hepatocytes, we observed hydroxynonenal (4-HNE) and found no difference the restoration of the L-OPA1 a isoform that was between Phb2-deficient livers and controls (Suppl. absent in Phb2 null hepatocytes. In the control Figure S5). Overall, L-OPA1Δ restored hepatocytes, protein levels of L-OPA1 a isoform mitochondrial morphology in Phb2-deficient was further elevated. Imaging of hepatocytes hepatocytes and conferred protection against showed that the loss of prohibitins led to a high apoptosis induced by the deletion of prohibitins. percentage of fragmented and ring-shaped mitochondria (Figure 4B, Suppl. Figure S3), in In vivo expression of L-OPA1Δ in the liver was agreement with the globular pattern revealed by not sufficient to rescue metabolic defects in Hep- electron microscopy (Figure 3G). Expression of L- Phb2-/- mice OPA1Δ restored an elongated mitochondrial morphology in most of the Phb2 null hepatocytes Diet-induced obesity and steatosis can be and promoted hyperfused mitochondria in control prevented by hepatic deletion or dominant hepatocytes (Figure 4B). Electron microscopy negative mutation of dynamin-related protein 1 performed on isolated hepatocytes (Figure 4C) (Drp1), a mitochondrial fission machinery (39). confirmed the globular pattern of mitochondria Since L-OPA1Δ was able to enhance Downloaded from http://www.jbc.org/ by guest on July 18, 2019 observed in Hep-Phb2-/- liver (Figure 3G) and mitochondrial fusion in isolated hepatocytes, we showed rescued mitochondrial morphology tested its potential effects in vivo in Hep-Phb2-/- following L-OPA1Δ transduction. mice. Adenovirus expressing L-OPA1Δ was injected in the tail of knockout and control mice on Phb2-deficient cells are prone to apoptosis, while the same day of tamoxifen implantation for L-OPA1 is able to promote cell survival upon simultaneous induction of liver Phb2 deletion Phb2 depletion (11,19,34). Given that apoptosis is (Figure 5A). Adenovirus expressing a key pathological feature of NAFLD (35), we mitochondrion-targeted dsRED (mitoRFP) was tested in vitro expression of L-OPA1Δ in isolated used as control adenovirus. Additionally, we tested hepatocytes and checked dissipation of the if delivery of L-OPA1Δ postponed 1 week after mitochondrial membrane potential (Δψm) as an tamoxifen-induced deletion of Phb2 could prevent early event of apoptosis (36); analysed by the the severe metabolic defects of Hep-Phb2-/- mice sensitivity to the mitochondrial uncoupler CCCP (Suppl. Figure S6). (carbonyl cyanide m-chlorophenyl hydrazone) (Suppl. Figure S4A). Consistent with Phb2- Two weeks after the simultaneous deletion of liver deficient MEFs (11), Phb2-null hepatocytes Phb2 and induction of L-OPA1Δ expression, we exhibited a similar degree of vulnerability to isolated hepatocytes and analysed their CCCP-induced mitochondrial membrane mitochondrial morphology (Figure 5B). In depolarization as the control hepatocytes (Figure agreement with the in vitro results (Figure 4B), 4D). L-OPA1Δ expression delayed the dissipation hepatocytes isolated from Hep-Phb2-/- mice and of Δψm in control hepatocytes, while it did not in expressing L-OPA1Δ showed less fragmented Phb2-deficient hepatocytes, indicating mitochondria and increased elongated ones. morphology-independent underlying defects Hepatocytes from control mice expressing L- caused by Phb2 deletion. Associated with the OPA1Δ also exhibited a marked increase in dissipation of Δψm, the intrinsic pathway of elongated mitochondria. However, hepatocytes apoptosis is triggered via the release of isolated from Hep-Phb2-/- mice following in vivo cytochrome c from mitochondria to activate L-OPA1Δ expression were not protected from caspases in the cytosol (37). We monitored the spontaneous apoptosis (Figure 5C, Suppl. Figure release of cytochrome c in isolated hepatocytes S7). Of note, compared to the in vitro experiments treated with the general caspase inhibitor z-VAD- performed 48h post-transduction (Figure 4E), we fmk to prevent cell detachment (38) (Suppl. Figure observed higher cytochrome c release in the S4B). Loss of prohibitins in hepatocytes caused a hepatocytes of the control mice 2 weeks after substantial release of cytochrome c into the cytosol, adenoviral injection (Figure 5C). 5
Mitochondrial dynamics and liver gluconeogenesis Immunofluorescence on the Flag tag of L-OPA1Δ make sure that the availability of circulating construct revealed more than 90% of transduction lactate was not rate limiting in knockout animals, efficiency in hepatocytes (Figure 5D). Some we measured the lactatemia that was in the control hepatocytes displayed abnormal nuclei as a physiological range for all of the groups (Figure sign of apoptosis (40), possibly contributed by 6E). Other enzymes involved in gluconeogenesis, sporadic adenovirus-induced inflammation (41). such as G6Pase and pyruvate carboxylase (PC), were similarly expressed regardless of the Regarding the phenotype of the animals, L- experimental conditions (Figure 6C). OPA1Δ expression failed to rescue the glycaemia and body weight of Hep-Phb2-/- mice (Figure 5E- Insulin signalling results in the phosphorylation of F). Intriguingly, in control mice, after 1-week of the transcription factor FOXO1 and its exclusion L-OPA1Δ expression there was a significant from the nucleus, thereby reducing the expression increase of blood glucose levels (Suppl. Figure of PEPCK-c and G6Pase. In Phb2-deficient liver, S6B). Neither the fat nor the lean masses were we observed an increase in the protein level of corrected in Hep-Phb2-/- mice following L-OPA1Δ FOXO1, not of its phosphorylated form, consistent expression in the liver (Figure 5G, Suppl. Figure with the inhibition of Akt phosphorylation on S6C), while the lean mass was increased by 14.4% Ser473 (Figure 3F). This led us to investigate the in control mice expressing L-OPA1Δ (Figure 5G). key gluconeogenic enzymes at the transcript level L-OPA1Δ expression normalized neither liver (Figure 6D). There was no change between the architecture nor hepatic lipid contents in Hep- knockout and control mice for PEPCK-c, G6Pase Downloaded from http://www.jbc.org/ by guest on July 18, 2019 Phb2-/- mice (Figure 5H, Suppl. Figure S7B). In and PC, while glucose transporter Glut2 summary, in vivo restoration of mitochondrial expression was reduced in Phb2-deficient liver. In morphology by L-OPA1Δ expression failed to control mice, the expression of L-OPA1Δ did not protect mice from illness induced by loss of modify the expression of these gluconeogenic hepatic prohibitins. genes. This indicates that these enzymes were not rate-limiting, pointing to alternative effectors In vivo expression of L-OPA1Δ in liver mediating L-OPA1-induced gluconeogenesis. potentiated gluconeogenesis Regarding fatty acid metabolism, carnitine palmitoyltransferase (CPT) in Phb2-dificient liver Consistent with the persistence of hypoglycaemia was upregulated at the mRNA level (Figure 6D), in Phb2 knockout animals expressing L-OPA1Δ, a while slightly reduced at the protein level (Figure pyruvate challenge (Figure 6A) on 6h-fasted Hep- 6C). A discrepancy between the levels of mRNA Phb2-/- mice transduced with L-OPA1Δ showed no and protein has already been reported by us and restoration of gluconeogenesis. In line with the others regarding CPT1, in particular higher mRNA unexpected increase in blood glucose (Suppl. not translated at the protein level in mouse liver Figure S6B), L-OPA1Δ expression in control mice (43,44). Expression of the key transcription factor enhanced endogenous glucose production as for liver ß-oxidation, namely peroxisome observed 2 weeks after in vivo adenoviral delivery proliferator-activated receptor α (PPARα), was (Figure 6B). The same effect was observed when markedly downregulated in the absence of Phb2, L-OPA1Δ was administered only 1 week before not rescued by the introduction of L-OPA1Δ. This the challenge (Suppl. Figure S6E). points to repressed ß-oxidation in knockout hepatocytes and substantiates the observed lipid Because PEPCK-c was downregulated in Phb2- accumulation. Overall, the absence of prohibitins deficient liver (Figure 3F), we checked if L- supressed gluconeogenesis independently of the OPA1Δ would increase the gluconeogenic presence of L-OPA1 and in vivo expression of L- PEPCK-c, which was not the case (Figure 6C). OPA1Δ in the liver of prohibitin-competent However, PEPCK-m was upregulated in Phb2-null control mice enhanced endogenous glucose liver, potentially as a compensatory mechanism production. (Figure 6C). Alike PEPCK-m, lactate dehydrogenase (LDH), that provides the In isolated hepatocytes, L-OPA1Δ enhances gluconeogenic substrate pyruvate, was upregulated mitochondrial respiration and glucose production in Phb2-deficient liver but unchanged by the expression L-OPA1Δ (Figure 6D). In order to 6
Mitochondrial dynamics and liver gluconeogenesis In order to substantiate the in vivo observation below). With the chronicity of hypoglycaemia, (Figure 6A-B), we assessed gluconeogenesis in hepatic glycogen contents were substantially vitro on hepatocytes isolated from control and lowered. Thus, Hep-Phb2-/- mice were severely ill Hep-Phb2-/- mice transduced either with control and experienced continuous hypoglycaemia even (MitoRFP) or L-OPA1Δ expressing adenoviruses, under the fed conditions, ultimately leading to then stimulated with pyruvate and lactate. Hep- severe weight loss and death. The aetiology of the Phb2-/- hepatocytes were unable to produce disease, induced by the knockout of liver Phb2, glucose, with or without the introduction of L- indicates that the abrogation of hepatic glucose OPA1Δ. However, consistent with the in vivo production was an early defect caused by the loss situation, expression of L-OPA1Δ conferred of hepatic prohibitins, in turn driving the hyperresponsiveness to control hepatocytes secondary peripheral dysregulations. Our group (Figure 7A). recently reported that the selective disruption of amino acid-derived gluconeogenesis in the As gluconeogenesis is an energy-consuming hepatocytes (48) is not sufficient to induce such a process relying on mitochondrial oxidative severe phenotype as the one observed in Hep- capacity (7), we finally tested mitochondrial Phb2-/- mice, pointing to additional liver failures respiration in isolated hepatocytes (Figure 7B). such as impaired mitochondrial oxidative activity. Loss of Phb2 in hepatocytes reduced oxygen Lipid stores rerouted to the liver of Hep-Phb2-/- consumption and ATP production (Figure 7C-E). mice lacking efficient ß-oxidation contributed to Conversely, expression of L-OPA1Δ enhanced the accumulation of hepatic triglycerides (50). Downloaded from http://www.jbc.org/ by guest on July 18, 2019 both respiration and ATP generation in control hepatocytes. Such functions were not restored in Ablation of Phb2 in hepatocytes led to excessive hepatocytes lacking prohibitins. These results not proteolytic cleavage of L-OPA1, increasing the only suggest that a hyperfused pattern of ratio of short to long isoforms. This was associated mitochondria could increase their energetic with mitochondrial fragmentation and a dramatic efficiency, pending integrity of prohibitins, but increase in spontaneous cytochrome c release. that gluconeogenesis tightly relies on Loss of L-OPA1 has been accounted for the mitochondrial respiration. defects in Phb2 null cells (11). Our results show that the regulation of OPA1 cleavage is a central function of prohibitins in primary hepatocytes, linking mitochondrial morphology to liver Discussion function. In the liver of leptin-deficient ob/ob mice, leptin treatment reduces both lipid content and the The present study demonstrates that in vivo OPA1 ratio of short to long isoforms, restoring deletion of Phb2 in hepatocytes rapidly leads to such ratio to untreated lean control levels (51). hypoglycaemia and loss of body weight, accompanied by liver lipid accumulation and The in vivo stabilization of L-OPA1 did not hypolipidemia. Although Hep-Phb2-/- mice had improve of the metabolic phenotype of Hep-Phb2-/- preserved glucose tolerance and insulin sensitivity, mice despite efficient restoration of the glycogen stores were depleted and mitochondrial morphology. Besides maintaining gluconeogenesis was inefficient with long-form OPA1 integrity, prohibitins protect downregulation of hepatic PEPCK-c. mitochondrial respiratory chain complexes from degradation (12,13) and facilitate mitophagy to During the first phase of fasting, half of the hepatic remove dysfunctional mitochondria (14). L-OPA1 glucose production is contributed by the expression in Phb2-deficient neurons does not breakdown of glycogen and the other half by restore impaired respiratory supercomplexes (20). gluconeogenesis (45). Upon prolonged fasting and Because the liver has a prominent metabolic exhaustion of hepatic glycogen, gluconeogenesis activity largely relying on mitochondria, any contributes to nearly all of the glucose production dysfunction of this organelle may lead to (46,47). Soon after deletion of liver Phb2 (1 week), metabolic alterations (52). Our data emphasize the Hep-Phb2-/- mice were unable to produce hepatic specific pathways by which prohibitins and OPA1 glucose and subsequently they rapidly became regulate mitochondrial integrity and cellular continuously hypoglycaemic (about 4 mM or metabolism. 7
Mitochondrial dynamics and liver gluconeogenesis One intriguing observation made here was that In conclusion, our results establish an essential upregulation of long-form OPA1, and the role for prohibitins in regulating hepatic associated hyperfused mitochondrial pattern, metabolism and whole-body energy homeostasis. resulted in enhanced hepatic glucose production in In the absence of prohibitins, L-OPA1 is sufficient prohibitin-competent mice. Such an anabolic boost to restore the morphology but not the function of was associated with an increase of the lean mass. liver mitochondria. In the presence of prohibitins, In prohibitin null mice, the metabolic defects were L-OPA1 promotes excessive mitochondrial not restored, pointing to other liver alterations not respiration and glucose production, pointing to restored by L-OPA1. The usual view is that mitochondrial dynamics in the control of mitochondrial morphology is an adaptive response gluconeogenesis. to cellular metabolic demands (53) and mitochondrial dynamics are associated with oxidative phosphorylation (54,55). Starvation promotes an elongated pattern of mitochondria, Experimental Procedures presumably to maximize ATP production (56), whereas high glucose condition results in massive Generation of hepatocyte-specific prohibitin-2 mitochondrial fragmentation (57). However, as knockout (Hep-Phb2-/-) mice most of the morphology-related studies were conducted on nutrient-consuming cell lines, there Phb2 floxed (Phb2fl/fl) mice (11) were crossed with is no preceding report on the effects of hyperfused Downloaded from http://www.jbc.org/ by guest on July 18, 2019 Alb-Cre-ERT2 mice carrying the tamoxifen- mitochondria in glucose production by hepatocytes. dependent Cre-ERT2 recombinase coding sequence Mitochondria serve as a hub for metabolic preceded by an internal ribosomal entry site enzymes such as GCK (33) and PEPCK-m (58). inserted in the 3 untranslated region of the serum Interaction of GCK with mitochondria has been albumin gene (MGI:3052812) (24). The in vivo implicated in the control of glycolysis and deletion of Phb2 in hepatocytes was induced at 8 apoptosis in hepatocytes (33). Thus, the level of weeks of age by subcutaneous implantation of expression per se of those enzymes is most likely tamoxifen pellets (Tamoxifen free base, 25 not the key element for metabolic regulation. The mg/pellet, 21 day release, E-361; Innovative interaction between metabolic enzymes and Research of America) in male Hep-Phb2-/- mice. mitochondria seems to play a central role in the Animals were maintained on a mixed (C57BL/6J x control of these pathways. 129/Sv) genetic background to avoid inbred strain- specific phenotypes. As control mice, we used Increased hepatic glucose production is the male Phb2fl/fl littermates, which were implanted primary contributor to fasting hyperglycaemia with tamoxifen pellets at the same time as Hep- (59,60), which is a hallmark in the natural history Phb2-/- mice, in order to optimize standardization of type 2 diabetes. However, neither PEPCK nor of the genetic background between the two groups. G6Pase are upregulated in the liver of type 2 Mice were maintained in our certified animal diabetic patients (61), indicating that alternative facility on 12-h dark-light cycle and were fed ad molecular mechanisms account for excessive libitum with standard chow diet (RM3-E-SQC glucose production. The present data indicates that #811181, SDS Diets, Essex, UK) and water mitochondrial dynamics participate to hepatic according to procedures that were approved by the glucose production. In particular, L-OPA1 not animal care and experimentation authorities of the only elongated mitochondria but also rendered Canton of Geneva (GE/128/15, #27139). hepatocytes hyperresponsive in terms of mitochondrial respiration and glucose production Adenovirus construction (Figure 7C). This is reminiscent of the excessive mitochondrial oxidative capacity and gluconeogenesis observed in NAFLD humans Recombinant adenovirus encoding for Flag tagged (6,7). However, an elongated pattern of cleavage-resistant isoform of rat OPA1 splice mitochondria is not sufficient for an over- variant 7 (L-OPA1∆) (62) or mitochondrial production of glucose, as shown in Phb2 null targeted dsRED (mitoRFP) under the hepatocytes expressing L-OPA1. cytomegalovirus (CMV) promoter were generated using the Adeno-X Expression system (Clontech, CA, USA) according to the manufacturer’s 8
Mitochondrial dynamics and liver gluconeogenesis protocol and as described previously (63). A clonal centrifuged at 2,000 rpm at 4°C to separate plasma. stock was amplified and purified for its in vivo use Liver and epididymal adipose tissue were collected, by Vector BioLabs (Malvern, PA, USA). weighed, snap-frozen in liquid nitrogen. Tissues Adenovirus was administered in vivo by tail vein and plasma were stored at -80°C. Pancreas was injection of 4 x 1010 plaque-forming units (pfu)/kg collected for fixation and further for L-OPA1∆ expression, or 3.2 x 1010 plaque- immunohistochemistry. forming units (pfu)/kg for mitoRFP expression. For in vitro transduction, hepatocytes were treated Determination of metabolic parameters with adenoviruses expressing either control MitoRFP or L-OPA1∆ at 5 MOI right after Alanine aminotransferase (ALT) and aspartate isolation and used 48h later. aminotransferase (AST), bilirubin and cholesterol plasma levels were measured at Geneva University Isolation of primary mouse hepatocytes Hospitals. Plasma free fatty acids, plasma ß- hydroxybutyrate, plasma and liver triglycerides, Mice were anesthetized by intraperitoneal and liver glycogen content were measured by injection of Pentothal (50 μg/ml, 50μl/25g of colorimetric assay using adequate quantification mouse body weight). Liver was perfused and kits (Biovision, Mountain View, CA) following hepatocytes were isolated as previously described manufacturer’s instructions. Under fed conditions, (48). Collected cells were re-suspended with 10 ml the activity of the microsomal triglyceride transfer protein (MTP) was measured in liver lysates by Downloaded from http://www.jbc.org/ by guest on July 18, 2019 of culture medium (Williams E medium, 5% FCS, 10-9 M insulin, 10-6 M dexamethasone, Pen/Strep fluorimetric assay using MTP Activity Assay Kit and 1% Glutamax). Hepatocytes were seeded at (Sigma). Insulin, glucagon, IL-6 and TNF-α were 50,000 cells/cm2 on collagen-coated plates and measured using Luminex xMAPTM technology and incubated at 37°C, 5% CO2 until further commercially available kits (Bio-Plex Pro experiments. Diabetes Assays and Bio-Plex Pro Mouse Cytokine, Biorad). In vivo experiments Glucose production by isolated hepatocytes From the time of tamoxifen implantation, mice were separated from their cage mates and housed Glucose production was assessed as previously individually. Blood glucose levels were measured described (48). In brief, isolated hepatocytes were using Accu-Check Aviva glucometer (Roche fasted for 6h in DMEM (without glucose, pyruvate Diagnostics, Switzerland) from the tail vein or phenol red) for glycogen depletion. The between 7:30 to 8:30 a.m. with body weight and medium was replaced by Krebs-Ringer- food intake monitored at the same time. Body Bicarbonate-Hepes (KRBH) buffer (140 mM NaCl, composition was assessed by an EchoMRI-700™ 3.6 mM KCl, 0.5 mM NaH2PO4, 0.5 mM MgSO4, quantitative nuclear magnetic resonance analyser 2 mM NaHCO3, 1.5 mM CaCl2, and 10 mM (Echo Medical Systems, Houston, TX). For HEPES) alone for basal condition, or KRBH glucose and pyruvate tolerance test, mice were buffer containing 10 mM lactate and 10 mM fasted for 6 h. D-Glucose (2 g/kg body weight) or pyruvate for stimulatory condition. Cells were sodium pyruvate (2 g/kg body weight) were stimulated for 60 min at 37°C. Supernatant was injected intraperitoneally. Glucose concentrations taken for glucose quantification with glucose assay were measured from blood samples taken from the kit (Biovision) and later normalized by protein tail vein at time 0, 15, 30, 60, 90 and 120 min after content. injection using Accu-Check Aviva glucometer. The total area under the curve (AUC) of glycaemia Immunoblotting was calculated using GraphPad Prism 6 software. For insulin tolerance test, mice under the fed Frozen liver tissues (30 mg) were lysed in 300 μl condition were injection with insulin (0.75 U/kg RIPA buffer in presence of 1x protease inhibitor body weight, Actrapid HM, 100 U/ml, Novo mix (Complete mini tablets, Roche) and 1x Nordisk) intraperitoneally. Upon sacrifice, blood phosphatase inhibitor (PhosStop, Roche). was collected into EDTA-coated tubes (Sarstedt Homogenate was collected after homogenization #20.1341) via retro-orbital bleeding and performed using Tissue Lyser (Qiagen) and 9
Mitochondrial dynamics and liver gluconeogenesis centrifugation at 14,000rpm for 10 min at 4°C. cryostat sections were stained with DYKDDDDK Proteins from liver extracts (20 mg) were resolved tag monoclonal antibody with Alexa Fluor 488 by gel electrophoresis, transferred on conjugate (Invitrogen MA1-142-A488) overnight nitrocellulose membranes and probed with specific at 4 °C. Images were captured by Zeiss LSM 800 antibodies (Suppl. Table S1). Band signals were microscope. detected by horseradish peroxidase peroxidase system and quantifications were performed using Transmission electron microscopy (TEM) the PXi gel imaging system (Syngene). The livers from control and Hep-Phb2-/- mice 2 Histology weeks after tamoxifen-induced recombination were excised, cut in small pieces of 1 mm2 and For morphology analyses, livers were fixed in 4% processed for electron microscopy. The tissue was paraformaldehyde (PFA), dehydrated and fixed in 0.1M sodium cacodylate buffer containing embedded in paraffin. Sections (5 μm) were 2.5% glutaraldehyde at room temperature for 4h. stained with hematoxylin and eosin (H&E). For Samples were then washed three times with 0.1M Oil Red O staining, frozen cryostat sections (5 μm) sodium cacodylate buffer and post-fixed with 1% were fixed in ice cold 10% formalin for 10 min, osmium tetraoxide and 1.5% potassium dried, washed and incubated in propylene glycol ferrocynide in 0.1M cacodylate buffer for 1h at for 5 min. The sections were then incubated with room temperature. Samples were then dehydrated, Oil Red O solution (Sigma #O1516) for 10 min at embedded in epon resin and processed for electron Downloaded from http://www.jbc.org/ by guest on July 18, 2019 60°C and in 85% propylene glycol for 5 min. The microscopy as previously described (64). Ultrathin sections were counterstained with haematoxylin. sections were finally contrasted with uranyl acetate Sections were scanned by widefield slide scanner and lead citrate and observed with a Technai 20 (Axio Scan.Z1, Zeiss) at 20 x magnification. electron microscope (FEI Company, Eindhoven, Images were visualized by software ZEN (Zeiss). Netherlands). The same procedure was applied on isolated primary hepatocytes for EM analysis. Immunofluorescence Mitochondrial membrane potential analysis For live imaging of primary hepatocytes, cells were cultured on collagen (Sigma C7661) coated Cells were incubated at room temperature for 35 mm diameter tissue culture dishes with cover 20 min with 20 nM TMRM, washed, and kept at glass bottom (WPI) and incubated with 37°C on the microscope until signal reached MitoTracker Orange CMTMRos (100 nM, stability. CCCP (10 μM, carbonyl cyanide m- Molecular Probes) and Bodipy dye (1 μg/mL, chlorophenyl hydrazone) was added and sequential D3922, Molecular Probes). The preparation was images of TMRM fluorescence were acquired put inside a temperature-controlled chamber every 3s with an inverted Nikon A1r microscope. (37 °C) during imaging, and images were acquired Analysis of TMRM fluorescence over with an inverted Nikon A1r microscope. For mitochondrial regions of interest was performed quantification of mitochondrial morphology, using ImageJ software. hepatocytes were classified according three patterns of mitochondrial morphology: i.e. Cytochrome c release analysis fragmented, intermediate, and hyperfused (see Suppl. Figure S8). For insulin/glucagon staining Hepatocytes were treated with adenovirus on pancreas sections, pancreas was fixed for 2 h in expressing L-OPA1∆ at 5 MOI right after isolation. 4% PFA, and finally embedded in paraffin. After 48h incubation at 37°C, 5% CO2, general Sections of 5 µm were incubated with guinea pig caspase inhibitor z-VAD-fmk (50 μM, BD anti-swine insulin (Dako 564) and mouse anti- Biosciences 550377) was added to culture medium. glucagon (Sigma G2654) overnight at 4 °C. After 3h, cells were fixed and permeabilized. After Secondary goat anti-guinea pig Alexa Fluor 488 blocking on the coverslips, cytochrome c was antibody (Invitrogen A-11073) and goat anti-rabbit stained using mouse anti-cytochrome c antibody Alexa Fluor 546 antibody (Invitrogen A-11035) (BD Biosciences 556432) and mitochondria were were added for 1h. To monitor the in vivo stained using rabbit anti-Tom20 mAb (Cell transduction efficiency of adenovirus, frozen Signaling 42406) overnight at 4 °C. Secondary 10
Mitochondrial dynamics and liver gluconeogenesis goat anti-mouse Alexa Fluor 546 antibody Plasma samples were added to an assay buffer (Invitrogen A-11030) and goat anti-rabbit Alexa containing 37 mM glycine, 10 mM EDTA, 0.02% Fluor 488 antibody (Invitrogen A-11034) were hydrazine hydrate and 0.9 mM NAD+, and then added for 1h. The nucleus was stained with DAPI stimulated with 2.75 U/ml L-Lactate (Vector Laboratories H-1500). Images were Dehydrogenase (Roche 000000010127230001). acquired by Nikon A1r microscope or Zeiss LSM Lactate was measured as NADH fluorescence with 800 microscope. The number of cells releasing a Fluostar Optima using excitation wavelength of cytochrome c was determined using ImageJ. 340 nm and recording wavelength at 460 nm. Mitochondrial respiration measurement Gene expression analysis The mitochondrial respiration on isolated Total RNA from frozen liver was isolated using hepatocytes was tested in XF96 Seahorse TRIzol reagent (Invitrogen) and purified with apparatus (Agilent). Hepatocytes were plated at Nucleospin RNA II kit (Macherey-Nagel, Duren, 10,000 cells/well in a Seahorse XF96 cell culture Germany). The primers were designed using the microplate. One h prior to the measurement, cells Primer Express Software (Applera Europe), see were washed and pre-incubated in PBS containing Suppl. Table S2. Real time PCR was performed 2 mM CaCl2, 2 mM MgCl2, 5 mM glucose and 5 using an ABI 7000 sequence detection system mM sodium pyruvate in a non-CO2 37°C incubator. (Applera Europe), and PCR products were Basal measurement was run for 3 cycles followed quantified fluorometrically using the SYBR Green Downloaded from http://www.jbc.org/ by guest on July 18, 2019 by the sequential addition of 2 μM oligomycin core reagent kit (Life Technologies). The values (run for 2 cycles), 1 μM FCCP (2 cycles) and 1 obtained were normalized to values of the μM rotenone/antimycin A (2 cycles). The housekeeping genes 18S rRNA. calculation of various mitochondrial parameters was done as follows: ATP production = OCR Statistical analysis (basal) - OCR (oligomycin stimulated); Total respiration = OCR (basal) - OCR Statistical analyses were performed using (rotenone/antimycin A stimulated); Proton leak = GraphPad Prism 6 software, with one-way OCR (total respiration) - OCR (ATP production); ANOVA when more than two groups of data were Max. respiration = OCR (FCCP) - OCR compared and with non-parametric Mann-Whitney (rotenone/antimycin A stimulated). U test when only two groups of data were concerned. A p value < 0.05 was considered Plasma lactate measurement significant. Acknowledgements: We are grateful to Dr. P. Chambon and Dr. D. Metzger (IGBMC, Strasbourg) for sharing the SA-Cre-ERT2 mice and to Dr. T. Langer and Dr. C. Merkwirth (University of Cologne) for the Phb2fl/fl mice. We thank Christian Vesin and Florian Visentin for expert animal surgery and sample collections, Clarissa Bartley for analyses and technical help, Dr. Manon Rosselin for support with TMRM experiments, Dr. Yalin Emre for help with cytokine measurements, Dr. Sachin Supale for preliminary investigations, Dr. Cyril Sobolewski for ob/ob samples, and Dr. Thierry Brun for helpful discussions (University of Geneva). We are grateful to Dr. G. Mithieux’s laboratory (Inserm U855, Lyon) for sharing G6Pase antibody. We thank the members of bioimaging and histology core facilities (University of Geneva) for technical support. Conflict of interest: The authors have declared that no conflict of interest exists. Author contribution: Conceived and designed the experiments: LL, JM-L, MF, J-CM, and PM; performed the experiments: LL, MK, CJS, MF and JM-L; performed data analysis: LL, JM-L, CJS, MK, MF and PM; wrote the paper: LL, JM-L and PM. 11
Mitochondrial dynamics and liver gluconeogenesis REFERENCES 1. Petersen, M. C., Vatner, D. F., and Shulman, G. I. (2017) Regulation of hepatic glucose metabolism in health and disease. Nat Rev Endocrinol 13, 572-587 2. Bellentani, S., Scaglioni, F., Marino, M., and Bedogni, G. (2010) Epidemiology of non-alcoholic fatty liver disease. Dig Dis 28, 155-161 3. Loomba, R., and Sanyal, A. J. (2013) The global NAFLD epidemic. Nat Rev Gastroenterol Hepatol 10, 686-690 4. Fromenty, B., and Pessayre, D. (1995) Inhibition of mitochondrial beta-oxidation as a mechanism of hepatotoxicity. Pharmacol Ther 67, 101-154 5. Exton, J. H. (1972) Gluconeogenesis. Metabolism: clinical and experimental 21, 945-990 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., and Horvath, R. (2016) Mitochondrial dysfunction in liver failure requiring transplantation. Journal of inherited metabolic disease 39, 427-436 7. Sunny, N. E., Parks, E. J., Browning, J. D., and Burgess, S. C. (2011) Excessive hepatic mitochondrial TCA cycle and gluconeogenesis in humans with nonalcoholic fatty liver disease. Cell Metab 14, 804- 810 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., and Muijsers, A. O. (2002) A structure for the yeast prohibitin complex: Downloaded from http://www.jbc.org/ by guest on July 18, 2019 Structure prediction and evidence from chemical crosslinking and mass spectrometry. Protein Sci 11, 2471-2478 9. McClung, J. K., Jupe, E. R., Liu, X. T., and Dell'Orco, R. T. (1995) Prohibitin: potential role in senescence, development, and tumor suppression. Exp Gerontol 30, 99-124 10. Tatsuta, T., Model, K., and Langer, T. (2005) Formation of membrane-bound ring complexes by prohibitins in mitochondria. Mol Biol Cell 16, 248-259 11. Merkwirth, C., Dargazanli, S., Tatsuta, T., Geimer, S., Lower, B., Wunderlich, F. T., von Kleist- Retzow, J. C., Waisman, A., Westermann, B., and Langer, T. (2008) Prohibitins control cell proliferation and apoptosis by regulating OPA1-dependent cristae morphogenesis in mitochondria. Genes Dev 22, 476-488 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., and Grivell, L. A. (2000) Prohibitins act as a membrane-bound chaperone for the stabilization of mitochondrial proteins. EMBO J 19, 2444-2451 13. Steglich, G., Neupert, W., and Langer, T. (1999) Prohibitins regulate membrane protein degradation by the m-AAA protease in mitochondria. Mol Cell Biol 19, 3435-3442 14. Wei, Y., Chiang, W. C., Sumpter, R., Jr., Mishra, P., and Levine, B. (2017) Prohibitin 2 Is an Inner Mitochondrial Membrane Mitophagy Receptor. Cell 168, 224-238 e210 15. Ehses, S., Raschke, I., Mancuso, G., Bernacchia, A., Geimer, S., Tondera, D., Martinou, J. C., Westermann, B., Rugarli, E. I., and Langer, T. (2009) Regulation of OPA1 processing and mitochondrial fusion by m-AAA protease isoenzymes and OMA1. J Cell Biol 187, 1023-1036 16. Head, B., Griparic, L., Amiri, M., Gandre-Babbe, S., and van der Bliek, A. M. (2009) Inducible proteolytic inactivation of OPA1 mediated by the OMA1 protease in mammalian cells. J Cell Biol 187, 959-966 17. Anand, R., Wai, T., Baker, M. J., Kladt, N., Schauss, A. C., Rugarli, E., and Langer, T. (2014) The i- AAA protease YME1L and OMA1 cleave OPA1 to balance mitochondrial fusion and fission. J Cell Biol 204, 919-929 18. Supale, S., Thorel, F., Merkwirth, C., Gjinovci, A., Herrera, P. L., Scorrano, L., Meda, P., Langer, T., and Maechler, P. (2013) Loss of prohibitin induces mitochondrial damages altering beta-cell function and survival and is responsible for gradual diabetes development. Diabetes 62, 3488-3499 19. Merkwirth, C., Martinelli, P., Korwitz, A., Morbin, M., Bronneke, H. S., Jordan, S. D., Rugarli, E. I., and Langer, T. (2012) Loss of prohibitin membrane scaffolds impairs mitochondrial architecture and leads to tau hyperphosphorylation and neurodegeneration. PLoS Genet 8, e1003021 2
Mitochondrial dynamics and liver gluconeogenesis 20. Korwitz, A., Merkwirth, C., Richter-Dennerlein, R., Troder, S. E., Sprenger, H. G., Quiros, P. M., Lopez-Otin, C., Rugarli, E. I., and Langer, T. (2016) Loss of OMA1 delays neurodegeneration by preventing stress-induced OPA1 processing in mitochondria. J Cell Biol 212, 157-166 21. Civiletto, G., Varanita, T., Cerutti, R., Gorletta, T., Barbaro, S., Marchet, S., Lamperti, C., Viscomi, C., Scorrano, L., and Zeviani, M. (2015) Opa1 overexpression ameliorates the phenotype of two mitochondrial disease mouse models. Cell Metab 21, 845-854 22. He, B., Feng, Q., Mukherjee, A., Lonard, D. M., DeMayo, F. J., Katzenellenbogen, B. S., Lydon, J. P., and O'Malley, B. W. (2008) A repressive role for prohibitin in estrogen signaling. Mol Endocrinol 22, 344-360 23. Park, S. E., Xu, J., Frolova, A., Liao, L., O'Malley, B. W., and Katzenellenbogen, B. S. (2005) Genetic deletion of the repressor of estrogen receptor activity (REA) enhances the response to estrogen in target tissues in vivo. Mol Cell Biol 25, 1989-1999 24. Schuler, M., Dierich, A., Chambon, P., and Metzger, D. (2004) Efficient temporally controlled targeted somatic mutagenesis in hepatocytes of the mouse. Genesis 39, 167-172 25. Ross, J. A., Nagy, Z. S., and Kirken, R. A. (2008) The PHB1/2 phosphocomplex is required for mitochondrial homeostasis and survival of human T cells. J Biol Chem 283, 4699-4713 26. Garcia-Ruiz, I., Fernandez-Moreira, D., Solis-Munoz, P., Rodriguez-Juan, C., Diaz-Sanjuan, T., Munoz-Yague, T., and Solis-Herruzo, J. A. (2010) Mitochondrial complex I subunits are decreased in murine nonalcoholic fatty liver disease: implication of peroxynitrite. J Proteome Res 9, 2450-2459 27. Parvez, S., Tabassum, H., Rehman, H., Banerjee, B. D., Athar, M., and Raisuddin, S. (2006) Catechin Downloaded from http://www.jbc.org/ by guest on July 18, 2019 prevents tamoxifen-induced oxidative stress and biochemical perturbations in mice. Toxicology 225, 109-118 28. Tietge, U. J., Bakillah, A., Maugeais, C., Tsukamoto, K., Hussain, M., and Rader, D. J. (1999) Hepatic overexpression of microsomal triglyceride transfer protein (MTP) results in increased in vivo secretion of VLDL triglycerides and apolipoprotein B. J Lipid Res 40, 2134-2139 29. Hui, S., Ghergurovich, J. M., Morscher, R. J., Jang, C., Teng, X., Lu, W., Esparza, L. A., Reya, T., Le, Z., Yanxiang Guo, J., White, E., and Rabinowitz, J. D. (2017) Glucose feeds the TCA cycle via circulating lactate. Nature 551, 115-118 30. Farese, R. V., Sajan, M. P., and Standaert, M. L. (2005) Insulin-sensitive protein kinases (atypical protein kinase C and protein kinase B/Akt): actions and defects in obesity and type II diabetes. Exp Biol Med (Maywood) 230, 593-605 31. von Wilamowitz-Moellendorff, A., Hunter, R. W., Garcia-Rocha, M., Kang, L., Lopez-Soldado, I., Lantier, L., Patel, K., Peggie, M. W., Martinez-Pons, C., Voss, M., Calbo, J., Cohen, P. T., Wasserman, D. H., Guinovart, J. J., and Sakamoto, K. (2013) Glucose-6-phosphate-mediated activation of liver glycogen synthase plays a key role in hepatic glycogen synthesis. Diabetes 62, 4070-4082 32. Burgess, S. C., He, T., Yan, Z., Lindner, J., Sherry, A. D., Malloy, C. R., Browning, J. D., and Magnuson, M. A. (2007) Cytosolic phosphoenolpyruvate carboxykinase does not solely control the rate of hepatic gluconeogenesis in the intact mouse liver. Cell Metab 5, 313-320 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., and Korsmeyer, S. J. (2003) BAD and glucokinase reside in a mitochondrial complex that integrates glycolysis and apoptosis. Nature 424, 952-956 34. Ross, J. A., Robles-Escajeda, E., Oaxaca, D. M., Padilla, D. L., and Kirken, R. A. (2017) The prohibitin protein complex promotes mitochondrial stabilization and cell survival in hematologic malignancies. Oncotarget 8, 65445-65456 35. Wieckowska, A., Zein, N. N., Yerian, L. M., Lopez, A. R., McCullough, A. J., and Feldstein, A. E. (2006) In vivo assessment of liver cell apoptosis as a novel biomarker of disease severity in nonalcoholic fatty liver disease. Hepatology 44, 27-33 36. Ly, J. D., Grubb, D. R., and Lawen, A. (2003) The mitochondrial membrane potential (deltapsi(m)) in apoptosis; an update. Apoptosis : an international journal on programmed cell death 8, 115-128 37. Desagher, S., and Martinou, J. C. (2000) Mitochondria as the central control point of apoptosis. Trends Cell Biol 10, 369-377 3
You can also read