A Fine Balance of Dietary Lipids Improves Pathology of a Murine Model of VCP-Associated Multisystem Proteinopathy

Katrina J Llewellyn, Naomi Walker, Christopher Nguyen, Baichang Tan, Lbachir BenMohamed, Virginia E Kimonis, Angèle Nalbandian, Katrina J Llewellyn, Naomi Walker, Christopher Nguyen, Baichang Tan, Lbachir BenMohamed, Virginia E Kimonis, Angèle Nalbandian

Abstract

The discovery of effective therapies and of disease mechanisms underlying valosin containing protein (VCP)-associated myopathies and neurodegenerative disorders remains elusive. VCP disease, caused by mutations in the VCP gene, are a clinically and genetically heterogeneous group of disorders with manifestations varying from hereditary inclusion body myopathy, Paget's disease of bone, frontotemporal dementia (IBMPFD), and amyotrophic lateral sclerosis (ALS). In the present study, we examined the effects of higher dietary lipid percentages on VCPR155H/R155H, VCPR155H/+ and Wild Type (WT) mice from birth until 15 months of age by immunohistochemical and biochemical assays. Findings illustrated improvement in the muscle strength, histology, and autophagy signaling pathway in the heterozygote mice when fed 9% lipid-enriched diets (LED). However, increasing the LED by 12%, 30%, and 48% showed no improvement in homozygote and heterozygote survival, muscle pathology, lipid accumulation or the autophagy cascade. These findings suggest that a balanced lipid supplementation may have a therapeutic strategy for patients with VCP-associated multisystem proteinopathies.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Grip strength measurements and histological…
Fig 1. Grip strength measurements and histological analyses of WT and VCPR155H/+ mice on normal and 9% LED.
(A) Grip strength measurements for the WT (blue) and VCPR155H/+ (red) mice placed on the 6% ND from birth and VCPR155H/+ mice placed on the 9% LED from birth (purple) and at 7 months of age onwards (green). H&E staining on WT and VCPR155H/+ on (B) ND (pathology shown by white arrows, right panel), (C) 9% LED from birth, and (D) 9% LED beginning at 7 months of age onwards (pathology shown by white arrows, right panel). (E) Western blot analyses of WT and VCPR155H/+ animals fed either ND or 9% LED. (F) Densitometry analyses relative to loading control (Beta-actin). Statistical significance is denoted by *P ≤ 0.05.
Fig 2. Survival curves of WT, VCP…
Fig 2. Survival curves of WT, VCPR155H/+, and VCPR155H/R155H mice on normal and varying lipid-enriched diets.
Kaplan-Meier survival probability (%) analysis for the WT, VCPR155H/+, and VCPR155H/R155H pregnant dams placed on the (A) 6% normal diet, (B) 9% LED, (C) 12% LED, (D) 30% LED, and (E) 48% LED. (F) Combined survival curve of all the LED regimens for the WT, VCPR155H/+, and VCPR155H/R155H. The number of mice analyzed per experiment is 35–40.
Fig 3. Strength measurements and histological stainings…
Fig 3. Strength measurements and histological stainings of quadriceps muscle of WT and VCPR155H/+ mice on normal and different lipid-enriched diets.
(A,B) Grip strength and Rotarod measurements analyses at 15-months of age, (C,D) H&E and (E,F) Oil Red O staining of quadriceps muscle from WT and VCPR155H/+ animals placed on the 9%, 12%, 30%, and 48% LED from birth. The number of mice analyzed per experiment is 8–10. Insets show magnified pathology, including rimmed vacuoles (orange arrows), atrophy (blue arrows), centralized nuclei (green arrows), and increased lipid droplets (white arrows).
Fig 4. Autophagy influx in the quadriceps…
Fig 4. Autophagy influx in the quadriceps of WT and VCPR155H/+ mice on normal and different lipid-enriched diets.
Immunohistochemical histological analyses comparing WT and heterozygote VCPR155H/+ knock-in animals on varying 12%, 30%, and 48% lipid-enriched diets stained with (A) TDP-43 (white arrows), (B) p62/SQSTM1, and (C) LC3-I/II. (D) Western blot of WT and heterozygote VCPR155H/+ knock-in animals on varying 12%, 30%, and 48% lipid-enriched diets analyzed for ubiquitin, optineurin, p62/SQSTM1, TDP-43, LC3-I/II, and Mucolipin (TRPML1). Magnified insets and white arrows indicate increased p62/SQSTM1 autophagic puncta, LC3 and TDP-43 protein expression translocated from the nucleus to the cytoplasm. (E) Densitometry analyses relative to loading control (Beta-actin). Statistical significance is denoted by *P ≤ 0.05. The number of mice analyzed per experiment is 8–10.
Fig 5. Effects of LED on the…
Fig 5. Effects of LED on the expression of lysosomal enzymes in the quadriceps of WT and VCPR155H/+ mice on normal and different lipid-enriched diets.
(A) Western blot analyses of protein expression levels of acid phosphatase, lysosomal acid lipase in WT and VCPR155H/+ knock-in animals on varying ND (6%), 9%, 12%, 30%, and 48% lipid-enriched diets (LED). (B,C) Densitometry analyses of acid phosphatase and LAL from various diets relative to loading control (Beta-actin). Statistical significance is denoted by *P ≤ 0.05. The number of mice analyzed per experiment is 8–10.

References

    1. Nalbandian A, Donkervoort S, Dec E, Badadani M, Katheria V, Rana P, et al. The multiple faces of valosin-containing protein-associated diseases: inclusion body myopathy with Paget's disease of bone, frontotemporal dementia, and amyotrophic lateral sclerosis. J Mol Neurosci. 2011;45(3):522–31. Epub 2011/09/06. 10.1007/s12031-011-9627-y .
    1. Kimonis VE, Kovach MJ, Waggoner B, Leal S, Salam A, Rimer L, et al. Clinical and molecular studies in a unique family with autosomal dominant limb-girdle muscular dystrophy and Paget disease of bone. Genet Med. 2000;2(4):232–41. .
    1. Kovach MJ, Ruiz J, Kimonis K, Mueed S, Sinha S, Higgins C, et al. Genetic heterogeneity in autosomal dominant essential tremor. Genet Med. 2001;3(3):197–9. .
    1. Watts GD, Thorne M, Kovach MJ, Pestronk A, Kimonis VE. Clinical and genetic heterogeneity in chromosome 9p associated hereditary inclusion body myopathy: exclusion of GNE and three other candidate genes. Neuromuscul Disord. 2003;13(7–8):559–67. .
    1. Kimonis VE, Mehta SG, Fulchiero EC, Thomasova D, Pasquali M, Boycott K, et al. Clinical studies in familial VCP myopathy associated with Paget disease of bone and frontotemporal dementia. American journal of medical genetics. 2008;146(6):745–57. .
    1. Watts GD, Wymer J, Kovach MJ, Mehta SG, Mumm S, Darvish D, et al. Inclusion body myopathy associated with Paget disease of bone and frontotemporal dementia is caused by mutant valosin-containing protein. Nature genetics. 2004;36(4):377–81. .
    1. Kimonis VE, Fulchiero E, Vesa J, Watts G. VCP disease associated with myopathy, paget disease of bone and frontotemporal dementia: Review of a unique disorder. Biochimica et biophysica acta. 2008. .
    1. Ju JS, Weihl CC. Inclusion body myopathy, Paget's disease of the bone and fronto-temporal dementia: a disorder of autophagy. Human molecular genetics. 2010;19(R1):R38–45. 10.1093/hmg/ddq157
    1. Ju JS, Fuentealba RA, Miller SE, Jackson E, Piwnica-Worms D, Baloh RH, et al. Valosin-containing protein (VCP) is required for autophagy and is disrupted in VCP disease. The Journal of cell biology. 2009;187(6):875–88. 10.1083/jcb.200908115
    1. Vesa J, Su H, Watts GD, Krause S, Walter MC, Martin B, et al. Valosin containing protein associated inclusion body myopathy: abnormal vacuolization, autophagy and cell fusion in myoblasts. Neuromuscul Disord. 2009;19(11):766–72. 10.1016/j.nmd.2009.08.003
    1. Nalbandian A, Ghimbovschi S, Radom-Aizik S, Dec E, Vesa J, Martin B, et al. Global Gene Profiling of VCP-associated Inclusion Body Myopathy. Clin Transl Sci. 2012;5(3):226–34. Epub 2012/06/13. 10.1111/j.1752-8062.2012.00407.x
    1. Lee HS, Daniels BH, Salas E, Bollen AW, Debnath J, Margeta M. Clinical utility of LC3 and p62 immunohistochemistry in diagnosis of drug-induced autophagic vacuolar myopathies: a case-control study. PLoS ONE. 2012;7(4):e36221 Epub 2012/05/05. doi: [pii].
    1. Tung YT, Wang BJ, Hu MK, Hsu WM, Lee H, Huang WP, et al. Autophagy: a double-edged sword in Alzheimer's disease. J Biosci. 2012;37(1):157–65. Epub 2012/02/24. .
    1. Heng MY, Detloff PJ, Paulson HL, Albin RL. Early alterations of autophagy in Huntington disease-like mice. Autophagy. 2010;6(8):1206–8. Epub 2010/10/12. doi: 13617 [pii].
    1. Shea L, Raben N. Autophagy in skeletal muscle: implications for Pompe disease. International journal of clinical pharmacology and therapeutics. 2009;47 Suppl 1:S42–7. .
    1. Lunemann JD, Schmidt J, Dalakas MC, Munz C. Macroautophagy as a pathomechanism in sporadic inclusion body myositis. Autophagy. 2007;3(4):384–6. .
    1. Nalbandian A, Llewellyn K, Badadani M, Yin H, Nguyen C, Katheria V, et al. A Progressive Translational Mouse Model of Human VCP Disease: The VCP R155H/+ Mouse. Muscle & nerve. 2012;(in press).
    1. Yin HZ, Nalbandian A, Hsu CI, Li S, Llewellyn KJ, Mozaffar T, et al. Slow development of ALS-like spinal cord pathology in mutant valosin-containing protein gene knock-in mice. Cell Death Dis. 2012;3:e374. Epub 2012/08/18. doi: cddis2012115 [pii] 10.1038/cddis.2012.115
    1. Nalbandian A, Llewellyn KJ, Kitazawa M, Yin HZ, Badadani M, Khanlou N, et al. The homozygote VCP(R(1)(5)(5)H/R(1)(5)(5)H) mouse model exhibits accelerated human VCP-associated disease pathology. PLoS ONE. 2012;7(9):e46308 10.1371/journal.pone.0046308
    1. Nalbandian A, Llewellyn KJ, Kitazawa M, Yin HZ, Badadani M, Khanlou N, et al. The Homozygote VCP(R155H/R155H) Mouse Model Exhibits Accelerated Human VCP-Associated Disease Pathology. PLoS ONE. 2012;7(9):e46308 Epub 2012/10/03. doi: [pii].
    1. Llewellyn KJ, Nalbandian A, Jung KM, Nguyen C, Avanesian A, Mozaffar T, et al. Lipid-enriched diet rescues lethality and slows down progression in a murine model of VCP-associated disease. Human molecular genetics. 2014;23(5):1333–44. 10.1093/hmg/ddt523
    1. Badadani M, Nalbandian A, Watts GD, Vesa J, Kitazawa M, Su H, et al. VCP associated inclusion body myopathy and paget disease of bone knock-in mouse model exhibits tissue pathology typical of human disease. PLoS ONE. 2010;5(10). Epub 2010/10/20. 10.1371/journal.pone.0013183
    1. Nalbandian A, Llewellyn KJ, Badadani M, Yin HZ, Nguyen C, Katheria V, et al. A progressive translational mouse model of human valosin-containing protein disease: the VCP(R155H/+) mouse. Muscle & nerve. 2013;47(2):260–70. 10.1002/mus.23522
    1. Nalbandian A, Nguyen C, Katheria V, Llewellyn KJ, Badadani M, Caiozzo V, et al. Exercise training reverses skeletal muscle atrophy in an experimental model of VCP disease. PLoS ONE. 2013;8(10):e76187 10.1371/journal.pone.0076187
    1. Baranano KW, Hartman AL. The ketogenic diet: uses in epilepsy and other neurologic illnesses. Curr Treat Options Neurol. 2008;10(6):410–9. Epub 2008/11/08.
    1. Evangeliou A, Vlachonikolis I, Mihailidou H, Spilioti M, Skarpalezou A, Makaronas N, et al. Application of a ketogenic diet in children with autistic behavior: pilot study. Journal of child neurology. 2003;18(2):113–8. Epub 2003/04/16. .
    1. Gasior M, Rogawski MA, Hartman AL. Neuroprotective and disease-modifying effects of the ketogenic diet. Behav Pharmacol. 2006;17(5–6):431–9. Epub 2006/08/31. doi: 00008877-200609000-00009 [pii].
    1. Jozwiak S, Kossoff EH, Kotulska-Jozwiak K. Dietary treatment of epilepsy: rebirth of an ancient treatment. Neurol Neurochir Pol. 2011;45(4):370–8. Epub 2011/11/22. doi: 17519 [pii]. .
    1. Siva N. Can ketogenic diet slow progression of ALS? Lancet Neurol. 2006;5(6):476 Epub 2006/06/03. .
    1. Stafstrom CE, Rho JM. The ketogenic diet as a treatment paradigm for diverse neurological disorders. Front Pharmacol. 2012;3:59 Epub 2012/04/18. 10.3389/fphar.2012.00059
    1. Camargo N, Brouwers JF, Loos M, Gutmann DH, Smit AB, Verheijen MH. High-fat diet ameliorates neurological deficits caused by defective astrocyte lipid metabolism. FASEB J. 2012. Epub 2012/07/04. doi: fj.12-205807 [pii] 10.1096/fj.12-205807 .
    1. Moresi V, Carrer M, Grueter CE, Rifki OF, Shelton JM, Richardson JA, et al. Histone deacetylases 1 and 2 regulate autophagy flux and skeletal muscle homeostasis in mice. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(5):1649–54. Epub 2012/02/07. doi: 1121159109 [pii] 10.1073/pnas.1121159109
    1. Turner N, Bruce CR, Beale SM, Hoehn KL, So T, Rolph MS, et al. Excess lipid availability increases mitochondrial fatty acid oxidative capacity in muscle: evidence against a role for reduced fatty acid oxidation in lipid-induced insulin resistance in rodents. Diabetes. 2007;56(8):2085–92. Epub 2007/05/24. doi: db07-0093 [pii] 10.2337/db07-0093 .
    1. Li B, Tong L, Jia G, Sun R. Effects of ketogenic diet on the clinical and electroencephalographic features of children with drug therapy-resistant epilepsy. Experimental and therapeutic medicine. 2013;5(2):611–5. 10.3892/etm.2012.823
    1. Peuscher R, Dijsselhof ME, Abeling NG, Van Rijn M, Van Spronsen FJ, Bosch AM. The ketogenic diet is well tolerated and can be effective in patients with argininosuccinate lyase deficiency and refractory epilepsy. JIMD reports. 2012;5:127–30. 10.1007/8904_2011_115
    1. Luan G, Zhao Y, Zhai F, Chen Y, Li T. Ketogenic diet reduces Smac/Diablo and cytochrome c release and attenuates neuronal death in a mouse model of limbic epilepsy. Brain Res Bull. 2012;89(3–4):79–85. 10.1016/j.brainresbull.2012.07.002 .
    1. Noh HS, Kim YS, Choi WS. Neuroprotective effects of the ketogenic diet. Epilepsia. 2008;49 Suppl 8:120–3. 10.1111/j.1528-1167.2008.01855.x .
    1. Rabinowitz JD, White E. Autophagy and metabolism. Science (New York, NY. 2010;330(6009):1344–8. Epub 2010/12/04. doi: 330/6009/1344 [pii] 10.1126/science.1193497
    1. Schiaffino S, Mammucari C, Sandri M. The role of autophagy in neonatal tissues: just a response to amino acid starvation? Autophagy. 2008;4(5):727–30. Epub 2008/04/26. doi: 6143 [pii]. .
    1. Shen D, Wang X, Li X, Zhang X, Yao Z, Dibble S, et al. Lipid storage disorders block lysosomal trafficking by inhibiting a TRP channel and lysosomal calcium release. Nature communications. 2012;3:731 10.1038/ncomms1735
    1. Cheng X, Zhang X, Gao Q, Ali Samie M, Azar M, Tsang WL, et al. The intracellular Ca(2)(+) channel MCOLN1 is required for sarcolemma repair to prevent muscular dystrophy. Nature medicine. 2014;20(10):1187–92. 10.1038/nm.3611

Source: PubMed

3
Abonner