Hepatic mitochondrial dysfunction is a feature of Glycogen Storage Disease Type Ia (GSDIa)

Benjamin L Farah, Rohit A Sinha, Yajun Wu, Brijesh K Singh, Andrea Lim, Masahiro Hirayama, Dustin J Landau, Boon Huat Bay, Dwight D Koeberl, Paul M Yen, Benjamin L Farah, Rohit A Sinha, Yajun Wu, Brijesh K Singh, Andrea Lim, Masahiro Hirayama, Dustin J Landau, Boon Huat Bay, Dwight D Koeberl, Paul M Yen

Abstract

Glycogen storage disease type Ia (GSDIa, von Gierke disease) is the most common glycogen storage disorder. It is caused by the deficiency of glucose-6-phosphatase, an enzyme which catalyses the final step of gluconeogenesis and glycogenolysis. Clinically, GSDIa is characterized by fasting hypoglycaemia and hepatic glycogen and triglyceride overaccumulation. The latter leads to steatohepatitis, cirrhosis, and the formation of hepatic adenomas and carcinomas. Currently, little is known about the function of various organelles and their impact on metabolism in GSDIa. Accordingly, we investigated mitochondrial function in cell culture and mouse models of GSDIa. We found impairments in oxidative phosphorylation and changes in TCA cycle metabolites, as well as decreased mitochondrial membrane potential and deranged mitochondrial ultra-structure in these model systems. Mitochondrial content also was decreased, likely secondary to decreased mitochondrial biogenesis. These deleterious effects culminated in the activation of the mitochondrial apoptosis pathway. Taken together, our results demonstrate a role for mitochondrial dysfunction in the pathogenesis of GSDIa, and identify a new potential target for the treatment of this disease. They also provide new insight into the role of carbohydrate overload on mitochondrial function in other hepatic diseases, such as non-alcoholic fatty liver disease.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Figure 1. Mitochondrial respiration is impaired in…
Figure 1. Mitochondrial respiration is impaired in G6PC KD versus control siRNA treated AML-12 cells.
G6PC was knocked-down in AML-12 cells for 96 hours, then mitochondrial oximetry analysis was performed using a Seahorse XF24 mitochondrial flux analyser. Basal respiration, ATP turnover, Maximal Respiration, and Spare Mitochondrial capacity were measured/calculated as described in the methods section. Oxygen consumption was normalised to total cellular protein content. For all parts, n = 6, error bars represent SEM, *represents p 

Figure 2. The tri-carboxylic acid cycle function…

Figure 2. The tri-carboxylic acid cycle function is impaired in G6PC KO mice.

( A…

Figure 2. The tri-carboxylic acid cycle function is impaired in G6PC KO mice.
(A) Organic acid analysis in the livers G6PC KO mice (KO, n = 6) compared to wild-type (WT, n = 10) mice. (B) Analysis of amino acid levels in the same samples. (C) Schematic of the changes of the species in the TCA cycle. Species in green were increased in G6PC KO mouse livers relative to WT livers, while those in red were decreased. Species in blue showed no significant change between groups, while those in black were not directly measured. For all parts, error bars represent SEM, *represents p < 0.05.

Figure 3. Mitochondrial morphology is deranged in…

Figure 3. Mitochondrial morphology is deranged in the livers of G6PC KO mice and G6PC…

Figure 3. Mitochondrial morphology is deranged in the livers of G6PC KO mice and G6PC KD AML-12 cells (96 hour knockdown).
Mitochondrial morphology was analysed in ultrathin sections of mouse liver (A) and AML-12 cells (B) by electron microscopy. Mitochondrial morphology in the wild-type mice and control cells were within normal limits, whereas in the KO mice and KD cells, the mitochondria were distended, and swollen, with effacement of the cristae, disruption of the mitochondrial membranes, and influx of cytoplasmic contents into the mitochondria. Representative images from the mouse liver (A) and KD cells (B) are displayed. Scale bars are marked on each image. Frames box areas shown at high magnification.

Figure 4. Mitochondrial content is reduced in…

Figure 4. Mitochondrial content is reduced in GSDIa models.

( A ) The protein levels…

Figure 4. Mitochondrial content is reduced in GSDIa models.
(A) The protein levels of various key mitochondrial proteins are reduced following G6PC KD for 96 hours in AML-12 cells (n = 3). (B) The copy number of mitochondrial DNA is also reduced following G6PC KD for 96 hours in AML-12 cells (n = 6). (C) G6PC KO mouse livers also show decreased levels of mitochondrial proteins (n = 3). *Represents p < 0.05, error bars represent SEM.

Figure 5. Key factors involved in mitochondrial…

Figure 5. Key factors involved in mitochondrial biogenesis are reduced following G6PC KD for 96…

Figure 5. Key factors involved in mitochondrial biogenesis are reduced following G6PC KD for 96 hours in AML-12 cells.
(A) Analysis of mRNA expression of genes involved in mitochondrial biogenesis, quality control and mitophagy shows a decrease in expression of genes related to mitochondrial biogenesis, with no change in expression of key fission and fusion genes. (n = 3). (B) Protein levels of TFAM are reduced following G6PC KD, however those of NRF1 show no significant change (n = 5). *Represents p < 0.05, error bars represent SEM.

Figure 6. Protein levels of key transcription…

Figure 6. Protein levels of key transcription factors involved in mitochondrial biogenesis.

( A )…

Figure 6. Protein levels of key transcription factors involved in mitochondrial biogenesis.
(A) PGC1α levels are reduced in G6PC KD AML-12 cells (n = 5). (B) PGC1α levels are also reduced in G6PC KO mouse livers (n = 3). (C) Levels of ERRα are unchanged in G6PC KO mouse livers as compared to WT livers (n = 3). *Represents p < 0.05, error bars represent SEM.

Figure 7. The mitochondrial apoptosis pathway is…

Figure 7. The mitochondrial apoptosis pathway is up-regulated in GSD1a.

( A ) Mitochondrial membrane…

Figure 7. The mitochondrial apoptosis pathway is up-regulated in GSD1a.
(A) Mitochondrial membrane potential is reduced in G6PC KD AML-12 cells 72 hours after knockdown. (B) Cytosolic cytochrome c is increased in G6PC KD AML-12 cells. (C) Caspase 9 and caspase 3 cleavage is increased in G6PC KD AML-12 cells. (D,E) Caspase 9 (D) and caspase 3 (E) cleavage is increasted in G6PC KO mice. For all parts except A, n = 3, error bars represent SEM, and *represents p < 0.05. For A, n = 6.
All figures (7)
Figure 2. The tri-carboxylic acid cycle function…
Figure 2. The tri-carboxylic acid cycle function is impaired in G6PC KO mice.
(A) Organic acid analysis in the livers G6PC KO mice (KO, n = 6) compared to wild-type (WT, n = 10) mice. (B) Analysis of amino acid levels in the same samples. (C) Schematic of the changes of the species in the TCA cycle. Species in green were increased in G6PC KO mouse livers relative to WT livers, while those in red were decreased. Species in blue showed no significant change between groups, while those in black were not directly measured. For all parts, error bars represent SEM, *represents p < 0.05.
Figure 3. Mitochondrial morphology is deranged in…
Figure 3. Mitochondrial morphology is deranged in the livers of G6PC KO mice and G6PC KD AML-12 cells (96 hour knockdown).
Mitochondrial morphology was analysed in ultrathin sections of mouse liver (A) and AML-12 cells (B) by electron microscopy. Mitochondrial morphology in the wild-type mice and control cells were within normal limits, whereas in the KO mice and KD cells, the mitochondria were distended, and swollen, with effacement of the cristae, disruption of the mitochondrial membranes, and influx of cytoplasmic contents into the mitochondria. Representative images from the mouse liver (A) and KD cells (B) are displayed. Scale bars are marked on each image. Frames box areas shown at high magnification.
Figure 4. Mitochondrial content is reduced in…
Figure 4. Mitochondrial content is reduced in GSDIa models.
(A) The protein levels of various key mitochondrial proteins are reduced following G6PC KD for 96 hours in AML-12 cells (n = 3). (B) The copy number of mitochondrial DNA is also reduced following G6PC KD for 96 hours in AML-12 cells (n = 6). (C) G6PC KO mouse livers also show decreased levels of mitochondrial proteins (n = 3). *Represents p < 0.05, error bars represent SEM.
Figure 5. Key factors involved in mitochondrial…
Figure 5. Key factors involved in mitochondrial biogenesis are reduced following G6PC KD for 96 hours in AML-12 cells.
(A) Analysis of mRNA expression of genes involved in mitochondrial biogenesis, quality control and mitophagy shows a decrease in expression of genes related to mitochondrial biogenesis, with no change in expression of key fission and fusion genes. (n = 3). (B) Protein levels of TFAM are reduced following G6PC KD, however those of NRF1 show no significant change (n = 5). *Represents p < 0.05, error bars represent SEM.
Figure 6. Protein levels of key transcription…
Figure 6. Protein levels of key transcription factors involved in mitochondrial biogenesis.
(A) PGC1α levels are reduced in G6PC KD AML-12 cells (n = 5). (B) PGC1α levels are also reduced in G6PC KO mouse livers (n = 3). (C) Levels of ERRα are unchanged in G6PC KO mouse livers as compared to WT livers (n = 3). *Represents p < 0.05, error bars represent SEM.
Figure 7. The mitochondrial apoptosis pathway is…
Figure 7. The mitochondrial apoptosis pathway is up-regulated in GSD1a.
(A) Mitochondrial membrane potential is reduced in G6PC KD AML-12 cells 72 hours after knockdown. (B) Cytosolic cytochrome c is increased in G6PC KD AML-12 cells. (C) Caspase 9 and caspase 3 cleavage is increased in G6PC KD AML-12 cells. (D,E) Caspase 9 (D) and caspase 3 (E) cleavage is increasted in G6PC KO mice. For all parts except A, n = 3, error bars represent SEM, and *represents p < 0.05. For A, n = 6.

References

    1. Chou J. Y., Matern D., Mansfield B. C. & Chen Y. T. Type I glycogen storage diseases: disorders of the glucose-6-phosphatase complex. Current molecular medicine 2, 121–143 (2002).
    1. von Gierke E. Hepato-nephromegalia glykogenica (Glykogenspeicherkrankheit der Leber und Nieren). Beiträge zur pathologischen Anatomie und zur allgemeinen Pathologie 82, 497–513 (1929).
    1. Chou J. Y., Jun H. S. & Mansfield B. C. Glycogen storage disease type I and G6Pase-beta deficiency: etiology and therapy. Nature reviews. Endocrinology 6, 676–688, doi: 10.1038/nrendo.2010.189 (2010).
    1. Bandsma R. H., Smit G. P. & Kuipers F. Disturbed lipid metabolism in glycogen storage disease type 1. European journal of pediatrics 161 Suppl 1, S65–69, doi: 10.1007/s00431-002-1007-8 (2002).
    1. Jones J. G., Garcia P., Barosa C., Delgado T. C. & Diogo L. Hepatic anaplerotic outflow fluxes are redirected from gluconeogenesis to lactate synthesis in patients with Type 1a glycogen storage disease. Metabolic engineering 11, 155–162, doi: 10.1016/j.ymben.2009.01.003 (2009).
    1. Froissart R. et al.. Glucose-6-phosphatase deficiency. Orphanet journal of rare diseases 6, 27, doi: 10.1186/1750-1172-6-27 (2011).
    1. Cori G. T. & Cori C. F. Glucose-6-phosphatase of the liver in glycogen storage disease. The Journal of biological chemistry 199, 661–667 (1952).
    1. Bandsma R. H. et al.. Increased de novo lipogenesis and delayed conversion of large VLDL into intermediate density lipoprotein particles contribute to hyperlipidemia in glycogen storage disease type 1a. Pediatric research 63, 702–707, doi: 10.1203/PDR.0b013e31816c9013 (2008).
    1. Yiu W. H. et al.. Angiotensin mediates renal fibrosis in the nephropathy of glycogen storage disease type Ia. Kidney international 73, 716–723, doi: 10.1038/sj.ki.5002718 (2008).
    1. Franco L. M. et al.. Hepatocellular carcinoma in glycogen storage disease type Ia: a case series. Journal of inherited metabolic disease 28, 153–162, doi: 10.1007/s10545-005-7500-2 (2005).
    1. Reddy S. K. et al.. Resection of hepatocellular adenoma in patients with glycogen storage disease type Ia. Journal of hepatology 47, 658–663, doi: 10.1016/j.jhep.2007.05.012 (2007).
    1. Liu P. P. et al.. Outcome of living donor liver transplantation for glycogen storage disease. Transplantation proceedings 35, 366–368 (2003).
    1. Koeberl D. D., Kishnani P. S. & Chen Y. T. Glycogen storage disease types I and II: treatment updates. Journal of inherited metabolic disease 30, 159–164, doi: 10.1007/s10545-007-0519-9 (2007).
    1. Grefhorst A. et al.. Carbohydrate-response-element-binding protein (ChREBP) and not the liver X receptor alpha (LXRalpha) mediates elevated hepatic lipogenic gene expression in a mouse model of glycogen storage disease type 1. The Biochemical journal 432, 249–254, doi: 10.1042/BJ20101225 (2010).
    1. Sun B. et al.. Activation of glycolysis and apoptosis in glycogen storage disease type Ia. Molecular genetics and metabolism 97, 267–271, doi: 10.1016/j.ymgme.2009.04.003 (2009).
    1. Farah B. L. et al.. Induction of autophagy improves hepatic lipid metabolism in glucose-6-phosphatase deficiency. Journal of hepatology, doi: 10.1016/j.jhep.2015.10.008 (2015).
    1. Vega R. B., Horton J. L. & Kelly D. P. Maintaining ancient organelles: mitochondrial biogenesis and maturation. Circulation research 116, 1820–1834, doi: 10.1161/circresaha.116.305420 (2015).
    1. Lightowlers R. N., Taylor R. W. & Turnbull D. M. Mutations causing mitochondrial disease: What is new and what challenges remain? Science 349, 1494–1499, doi: 10.1126/science.aac7516 (2015).
    1. Rieusset J. Contribution of mitochondria and endoplasmic reticulum dysfunction in insulin resistance: Distinct or interrelated roles? Diabetes & metabolism, doi: 10.1016/j.diabet.2015.02.006 (2015).
    1. Milane L., Trivedi M., Singh A., Talekar M. & Amiji M. Mitochondrial biology, targets, and drug delivery. Journal of controlled release: official journal of the Controlled Release Society 207, 40–58, doi: 10.1016/j.jconrel.2015.03.036 (2015).
    1. Estaquier J., Vallette F., Vayssiere J. L. & Mignotte B. The mitochondrial pathways of apoptosis. Advances in experimental medicine and biology 942, 157–183, doi: 10.1007/978-94-007-2869-1_7 (2012).
    1. Zamzami N. et al.. Reduction in mitochondrial potential constitutes an early irreversible step of programmed lymphocyte death in vivo. The Journal of experimental medicine 181, 1661–1672 (1995).
    1. Liu X., Kim C. N., Yang J., Jemmerson R. & Wang X. Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome c. Cell 86, 147–157 (1996).
    1. Kluck R. M. et al.. Cytochrome c activation of CPP32-like proteolysis plays a critical role in a Xenopus cell-free apoptosis system. The EMBO journal 16, 4639–4649, doi: 10.1093/emboj/16.15.4639 (1997).
    1. Riede U. N., Spycher M. A. & Gitzelmann R. Glycogenosis type I (glucose 6-phosphatase deficiency): I. Ultrastructural morphometric analysis of juvenile liver cells. Pathology, research and practice 167, 136–150, doi: 10.1016/S0344-0338(80)80185-3 (1980).
    1. Jun H. S. et al.. Lack of glucose recycling between endoplasmic reticulum and cytoplasm underlies cellular dysfunction in glucose-6-phosphatase-beta-deficient neutrophils in a congenital neutropenia syndrome. Blood 116, 2783–2792, doi: 10.1182/blood-2009-12-258491 (2010).
    1. Koeberl D. D. et al.. Early, sustained efficacy of adeno-associated virus vector-mediated gene therapy in glycogen storage disease type Ia. Gene therapy 13, 1281–1289, doi: 10.1038/sj.gt.3302774 (2006).
    1. Lei K. J. et al.. Glucose-6-phosphatase dependent substrate transport in the glycogen storage disease type-1a mouse. Nature genetics 13, 203–209, doi: 10.1038/ng0696-203 (1996).
    1. Hems D. A. & Brosnan J. T. Effects of ischaemia on content of metabolites in rat liver and kidney in vivo. The Biochemical journal 120, 105–111 (1970).
    1. Hackenbrock C. R. Ultrastructural bases for metabolically linked mechanical activity in mitochondria. II. Electron transport-linked ultrastructural transformations in mitochondria. The Journal of cell biology 37, 345–369 (1968).
    1. Heath-Engel H. M. & Shore G. C. Mitochondrial membrane dynamics, cristae remodelling and apoptosis. Biochimica et biophysica acta 1763, 549–560, doi: 10.1016/j.bbamcr.2006.02.006 (2006).
    1. Egan D. F. et al.. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science 331, 456–461, doi: 10.1126/science.1196371 (2011).
    1. Zhu J., Dagda R. K. & Chu C. T. Monitoring mitophagy in neuronal cell cultures. Methods in molecular biology 793, 325–339, doi: 10.1007/978-1-61779-328-8_21 (2011).
    1. Schreiber S. N., Knutti D., Brogli K., Uhlmann T. & Kralli A. The transcriptional coactivator PGC-1 regulates the expression and activity of the orphan nuclear receptor estrogen-related receptor alpha (ERRalpha). The Journal of biological chemistry 278, 9013–9018, doi: 10.1074/jbc.M212923200 (2003).
    1. Bhat A. H. et al.. Oxidative stress, mitochondrial dysfunction and neurodegenerative diseases; a mechanistic insight. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie 74, 101–110, doi: 10.1016/j.biopha.2015.07.025 (2015).
    1. Hirschey M. D. et al.. SIRT3 deficiency and mitochondrial protein hyperacetylation accelerate the development of the metabolic syndrome. Molecular cell 44, 177–190, doi: 10.1016/j.molcel.2011.07.019 (2011).
    1. Lozano L., Lara-Lemus R., Zenteno E. & Alvarado-Vasquez N. The mitochondrial O-linked N-acetylglucosamine transferase (mOGT) in the diabetic patient could be the initial trigger to develop Alzheimer disease. Experimental gerontology 58, 198–202, doi: 10.1016/j.exger.2014.08.008 (2014).
    1. Li Z., Berk M., McIntyre T. M., Gores G. J. & Feldstein A. E. The lysosomal-mitochondrial axis in free fatty acid-induced hepatic lipotoxicity. Hepatology 47, 1495–1503, doi: 10.1002/hep.22183 (2008).
    1. Summers S. A. Ceramides in insulin resistance and lipotoxicity. Progress in lipid research 45, 42–72, doi: 10.1016/j.plipres.2005.11.002 (2006).
    1. Komatsu M. et al.. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. The Journal of cell biology 169, 425–434, doi: 10.1083/jcb.200412022 (2005).
    1. Lesmana R. et al.. Thyroid Hormone Stimulation of Autophagy Is Essential for Mitochondrial Biogenesis and Activity in Skeletal Muscle. Endocrinology 157, 23–38, doi: 10.1210/en.2015-1632 (2016).
    1. Wei Y., Rector R. S., Thyfault J. P. & Ibdah J. A. Nonalcoholic fatty liver disease and mitochondrial dysfunction. World journal of gastroenterology 14, 193–199 (2008).
    1. Ibdah J. A. et al.. Mice heterozygous for a defect in mitochondrial trifunctional protein develop hepatic steatosis and insulin resistance. Gastroenterology 128, 1381–1390 (2005).
    1. Reddy S. K. et al.. Liver transplantation for glycogen storage disease type Ia. Journal of hepatology 51, 483–490, doi: 10.1016/j.jhep.2009.05.026 (2009).
    1. Finsterer J. & Bindu P. S. Therapeutic strategies for mitochondrial disorders. Pediatric neurology 52, 302–313, doi: 10.1016/j.pediatrneurol.2014.06.023 (2015).
    1. Demaster A. et al.. Long-term efficacy following readministration of an adeno-associated virus vector in dogs with glycogen storage disease type Ia. Human gene therapy 23, 407–418, doi: 10.1089/hum.2011.106 (2012).

Source: PubMed

3
Iratkozz fel