Myogenic differentiation of VCP disease-induced pluripotent stem cells: A novel platform for drug discovery

Katrina J Llewellyn, Angèle Nalbandian, Lan N Weiss, Isabela Chang, Howard Yu, Bibo Khatib, Baichang Tan, Vanessa Scarfone, Virginia E Kimonis, Katrina J Llewellyn, Angèle Nalbandian, Lan N Weiss, Isabela Chang, Howard Yu, Bibo Khatib, Baichang Tan, Vanessa Scarfone, Virginia E Kimonis

Abstract

Valosin Containing Protein (VCP) disease is an autosomal dominant multisystem proteinopathy caused by mutations in the VCP gene, and is primarily associated with progressive muscle weakness, including atrophy of the pelvic and shoulder girdle muscles. Currently, no treatments are available and cardiac and respiratory failures can lead to mortality at an early age. VCP is an AAA ATPase multifunction complex protein and mutations in the VCP gene resulting in disrupted autophagic clearance. Due to the rarity of the disease, the myopathic nature of the disorder, ethical and practical considerations, VCP disease muscle biopsies are difficult to obtain. Thus, disease-specific human induced pluripotent stem cells (hiPSCs) now provide a valuable resource for the research owing to their renewable and pluripotent nature. In the present study, we report the differentiation and characterization of a VCP disease-specific hiPSCs into precursors expressing myogenic markers including desmin, myogenic factor 5 (MYF5), myosin and heavy chain 2 (MYH2). VCP disease phenotype is characterized by high expression of TAR DNA Binding Protein-43 (TDP-43), ubiquitin (Ub), Light Chain 3-I/II protein (LC3-I/II), and p62/SQSTM1 (p62) protein indicating disruption of the autophagy cascade. Treatment of hiPSC precursors with autophagy stimulators Rapamycin, Perifosine, or AT101 showed reduction in VCP pathology markers TDP-43, LC3-I/II and p62/SQSTM1. Conversely, autophagy inhibitors chloroquine had no beneficial effect, and Spautin-1 or MHY1485 had modest effects. Our results illustrate that hiPSC technology provide a useful platform for a rapid drug discovery and hence constitutes a bridge between clinical and bench research in VCP and related diseases.

Conflict of interest statement

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

Figures

Fig 1. Differentiation of patient and control…
Fig 1. Differentiation of patient and control VCP iPSC into myogenic lineages.
(A) Schematic of early hiPSCs and embryoid bodies commitment (Days 0–50) into early mesenchymal stem/stromal cells (MSC) and differentiation into myoblasts (Day 64). (B) Myogenic differentiation of human iPSC at Days 0, 21, 50, and 69 with (C) primary myoblast cells from a 57-year old patient diagnosed with IBMPFD. (D) Control derived myogenic precursors at Days 0, 21, 50, and 69 with (E) primary myoblast cells from age matched healthy control. Differential interference contrast (DIC) microscopy images of differentiated primary mature myoblasts from iPSCs. Scale: Bar = 1000 μm.
Fig 2. Validation of myogenic differentiation in…
Fig 2. Validation of myogenic differentiation in patient and control-derived iPSCs.
Human iPSC from a 57-year old patient diagnosed with VCP disease-derived myogenic precursors were stained at Day 21 and Day 50 with (A-B) MYF-5, desmin, and Pax7; (C-D) MyoD and MYH2. Representative merged overlay images of stained iPSC with DAPI. Scale: Bar = 50 μM. (E) Western blot analysis of myoblast differentiation markers at Day 0, 7, 21, 35, and 50 with anti-Oct3/4, Nanog, desmin, MYF5, MyoD, MyoG and MYH2. GAPDH was used as a positive loading control. (F) FACS analysis of iPSC-derived MSCs with pluripotent marker (CD34) and myoblast marker (CD56). (G) CD34 isotype control. (H) CD65 isotype control.
Fig 3. Characterization of autophagy signaling cascade…
Fig 3. Characterization of autophagy signaling cascade in control and patient VCP iPSC-derived myoblast lineages.
Differentiated (A) control and (B) iPSC-derived myoblast lineages were immunostained with TDP-43, LC3, and p62/SQSTM1. Representative merged overlay images of stained iPSC with DAPI. Scale: Bar = 50 μM. (C) Western blot analysis of iPSC-derived control and patient myoblasts with anti-TDP-43, LC3I/II, p62/SQSTM1, and ubiquitin. GAPDH was used as a positive loading control. (D) Densitometry analyses confirmed these Western blot results. Statistical significance is denoted by *p<0.05, **p<0.005 and ***p<0.001. (E) Western blot analysis of cytoplasmic (Cy) and nuclear (Nu) factions of iPSC-derived control and patient myoblasts with anti-TDP-43.
Fig 4. Drug screening with autophagy inducers…
Fig 4. Drug screening with autophagy inducers Rapamycin, Perifosine and AT101 in patient VCP iPSC-derived myogenic lineages.
(A) Schematic of intervention with autophagy modifying agents. Green arrows show the active location of autophagy activators Rapamycin, Perifosine and AT101. Red arrows show the active location of autophagy inhibitors chloroquine, Spautin-1 and MHY1485. VCP is indicated to have an interactive role with Akt and as a chaperone protein with ubiquitin. (B) Untreated differentiated control and (C) untreated patient derived myogenic lineages. Patient derived myogenic lineage were treated with either (D) Rapamycin (10 μM), (E) Perifosine (80 μM) or (F) AT101 (10 μM) for 24 hours. Subsequently, cells were stained with TDP-43, LC3 or p62/SQSTM1 antibodies. Representative merged overlay images of stained iPSC with DAPI. Scale: Bar = 25 μm. White dotted lines represent areas of increased or decreased expressions. (G) Western blot analysis of iPSC-derived control (C) and patient (P) myoblasts with mTOR, TDP-43, LC3I/II and p62/SQSTM1. GAPDH was used as a positive loading control. (H) Densitometry analyses of the Western blot. Black dotted line indicates expression over baseline control sample. Statistical significance is denoted by *p<0.05, **p<0.005 and ***p<0.001.
Fig 5. Drug screening with autophagy inhibitors…
Fig 5. Drug screening with autophagy inhibitors chloroquine, Spautin-1, and MHY1485 shows in patient VCP iPSC-derived myoblast lineages.
(A) Untreated differentiated control and (B) untreated patient derived myogenic lineages. Patient derived myogenic lineage were treated with either (C) chloroquine (10 μM), (D) Spautin-1 (10 μM) or (E) MYH1485 (2 μM) for 24 hours. Subsequently, cells were stained with TDP-43, LC3 or p62/SQSTM1 antibodies. Representative merged overlay images of stained iPSC with DAPI. Scale: Bar = 25 μm. White dotted lines represent areas of increased or decreased expressions. (F) Western blot analysis of iPSC-derived control and patient myoblasts probed against TDP-43, LC3-I/II, and p62/SQSTM1 antibodies. GAPDH was used as a positive loading control. (G) Densitometry analyses from Western blot. Black dotted line indicates expression over baseline control sample.

References

    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 Genet. 2004;36(4):377–81. doi:
    1. Kimonis VE, Watts GD. Autosomal dominant inclusion body myopathy, Paget disease of bone, and frontotemporal dementia. Alzheimer disease and associated disorders. 2005;19 Suppl 1:S44–7.
    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. doi:
    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;1782(12):744–8. doi:
    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. doi:
    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. Am J Med Genet. 2008;146A(6):745–57. doi:
    1. Ralston SH, Langston AL, Reid IR. Pathogenesis and management of Paget's disease of bone. Lancet. 2008;372(9633):155–63. doi:
    1. Ralston SH. Pathogenesis of Paget's disease of bone. Bone. 2008;43(5):819–25. doi:
    1. Mirra JM, Brien EW, Tehranzadeh J. Paget's disease of bone: review with emphasis on radiologic features, Part I. Skeletal radiology. 1995;24(3):163–71.
    1. Kimonis V, Donkervoort S, Watts G. Inclusion Body Myopathy Associated with Paget Disease of Bone and/or Frontotemporal Dementia. Gene Reviews. University of Washington, Seattle: 2007. [updated 2011];1993–2017.
    1. Rosso SM, Kamphorst W, de Graaf B, Willemsen R, Ravid R, Niermeijer MF, et al. Familial frontotemporal dementia with ubiquitin-positive inclusions is linked to chromosome 17q21-22. Brain. 2001;124(Pt 10):1948–57.
    1. Neumann M, Mackenzie IR, Cairns NJ, Boyer PJ, Markesbery WR, Smith CD, et al. TDP-43 in the ubiquitin pathology of frontotemporal dementia with VCP gene mutations. J Neuropathol Exp Neurol. 2007;66(2):152–7. doi:
    1. Forman MS, Mackenzie IR, Cairns NJ, Swanson E, Boyer PJ, Drachman DA, et al. Novel ubiquitin neuropathology in frontotemporal dementia with valosin-containing protein gene mutations. J Neuropathol Exp Neurol. 2006;65(6):571–81.
    1. Majounie E, Traynor BJ, Chio A, Restagno G, Mandrioli J, Benatar M, et al. Mutational analysis of the VCP gene in Parkinson's disease. Neurobiol Aging. 2012;33(1):209 e1–2.
    1. Nacmias B, Piaceri I, Bagnoli S, Tedde A, Piacentini S, Sorbi S. Genetics of Alzheimer's Disease and Frontotemporal Dementia. Curr Mol Med. 2014;14(8):993–1000. doi:
    1. Shamirian S, Nalbandian A, Khare M, Castellani R, Kim R, Kimonis VE. Early-onset Alzheimers and cortical vision impairment in a woman with valosin-containing protein disease associated with 2 APOE epsilon4/APOE epsilon4 genotype. Alzheimer disease and associated disorders. 2015;29(1):90–3. doi:
    1. Williams KL, Solski JA, Nicholson GA, Blair IP. Mutation analysis of VCP in familial and sporadic amyotrophic lateral sclerosis. Neurobiol Aging. 2012;33(7)1488.e15–1488.e16.
    1. Van Langenhove T, Van der Zee J, Van Broeckhoven C. The molecular basis of the frontotemporal lobar degeneration-amyotrophic lateral sclerosis spectrum. Ann Med. 2012; 44(8):817–28. doi:
    1. Djamshidian A, Schaefer J, Haubenberger D, Stogmann E, Zimprich F, Auff E, et al. A novel mutation in the VCP gene (G157R) in a German family with inclusion-body myopathy with Paget disease of bone and frontotemporal dementia. Muscle & nerve. 2009;39(3):389–91.
    1. Schroder R, Watts GD, Mehta SG, Evert BO, Broich P, Fliessbach K, et al. Mutant valosin-containing protein causes a novel type of frontotemporal dementia. Annals of neurology. 2005;57(3):457–61. doi:
    1. Stojkovic T, Hammouda el H, Richard P, Lopez de Munain A, Ruiz-Martinez J, Gonzalez PC, et al. Clinical outcome in 19 French and Spanish patients with valosin-containing protein myopathy associated with Paget's disease of bone and frontotemporal dementia. Neuromuscul Disord. 2009;19(5):316–23. doi:
    1. Haubenberger D, Bittner RE, Rauch-Shorny S, Zimprich F, Mannhalter C, Wagner L, et al. Inclusion body myopathy and Paget disease is linked to a novel mutation in the VCP gene. Neurology. 2005;65(8):1304–5. doi:
    1. Bersano A, Del Bo R, Lamperti C, Ghezzi S, Fagiolari G, Fortunato F, et al. Inclusion body myopathy and frontotemporal dementia caused by a novel VCP mutation. Neurobiol Aging. 2009;30(5):752–8. doi:
    1. Miller TD, Jackson AP, Barresi R, Smart CM, Eugenicos M, Summers D, et al. Inclusion body myopathy with Paget disease and frontotemporal dementia (IBMPFD): clinical features including sphincter disturbance in a large pedigree. J Neurol Neurosurg Psychiatry. 2009;80(5):583–4. doi:
    1. Kumar KR, Needham M, Mina K, Davis M, Brewer J, Staples C, et al. Two Australian families with inclusion-body myopathy, Paget's disease of bone and frontotemporal dementia: novel clinical and genetic findings. Neuromuscul Disord. 2010;20(5):330–4. doi:
    1. Kim EJ, Park YE, Kim DS, Ahn BY, Kim HS, Chang YH, et al. Inclusion body myopathy with Paget disease of bone and frontotemporal dementia linked to VCP p.Arg155Cys in a Korean family. Arch Neurol. 2011;68(6):787–96. doi:
    1. Weihl CC, Pestronk A, Kimonis VE. Valosin-containing protein disease: inclusion body myopathy with Paget's disease of the bone and fronto-temporal dementia. Neuromuscul Disord. 2009;19(5):308–15. doi:
    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. Watts GD, Thomasova D, Ramdeen SK, Fulchiero EC, Mehta SG, Drachman DA, et al. Novel VCP mutations in inclusion body myopathy associated with Paget disease of bone and frontotemporal dementia. Clin Genet. 2007;72(5):420–6. doi:
    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. doi:
    1. Mehta SG, Khare M, Ramani R, Watts GD, Simon M, Osann KE, et al. Genotype-phenotype studies of VCP-associated inclusion body myopathy with Paget disease of bone and/or frontotemporal dementia. Clin Genet. 2013;83(5):422–31. doi:
    1. Braun RJ, Zischka H. Mechanisms of Cdc48/VCP-mediated cell death: from yeast apoptosis to human disease. Biochimica et biophysica acta. 2008;1783(7):1418–35. doi:
    1. Meyer H, Bug M, Bremer S. Emerging functions of the VCP/p97 AAA-ATPase in the ubiquitin system. Nat Cell Biol. 2012;14(2):117–23. doi:
    1. Fujita E, Kouroku Y, Isoai A, Kumagai H, Misutani A, Matsuda C, et al. Two endoplasmic reticulum-associated degradation (ERAD) systems for the novel variant of the mutant dysferlin: ubiquitin/proteasome ERAD(I) and autophagy/lysosome ERAD(II). Hum Mol Genet. 2007;16(6):618–29. doi:
    1. Kim NC, Tresse E, Kolaitis RM, Molliex A, Thomas RE, Alami NH, et al. VCP is essential for mitochondrial quality control by PINK1/Parkin and this function is impaired by VCP mutations. Neuron. 2013;78(1):65–80. doi:
    1. Braun RJ, Zischka H, Madeo F, Eisenberg T, Wissing S, Buttner S, et al. Crucial mitochondrial impairment upon CDC48 mutation in apoptotic yeast. The Journal of biological chemistry. 2006;281(35):25757–67. doi:
    1. Bartolome F, Wu HC, Burchell VS, Preza E, Wray S, Mahoney CJ, et al. Pathogenic VCP mutations induce mitochondrial uncoupling and reduced ATP levels. Neuron. 2013;78(1):57–64. doi:
    1. Angelos MG, Kaufman DS. Pluripotent stem cell applications for regenerative medicine. Curr Opin Organ Transplant. 2015;20(6):663–70. doi:
    1. Abramson S, Miller RG, Phillips RA. The identification in adult bone marrow of pluripotent and restricted stem cells of the myeloid and lymphoid systems. The Journal of experimental medicine. 1977;145(6):1567–79.
    1. Moharil J, Lei P, Tian J, Gaile DP, Andreadis ST. Lentivirus Live Cell Array for Quantitative Assessment of Gene and Pathway Activation during Myogenic Differentiation of Mesenchymal Stem Cells. PLoS ONE. 2015;10(10):e0141365 doi:
    1. Tzatzalos E, Abilez OJ, Shukla P, Wu JC. Engineered heart tissues and induced pluripotent stem cells: Macro- and microstructures for disease modeling, drug screening, and translational studies. Adv Drug Deliv Rev. 2015.
    1. Haston KM, Finkbeiner S. Clinical Trials in a Dish: The Potential of Pluripotent Stem Cells to Develop Therapies for Neurodegenerative Diseases. Annu Rev Pharmacol Toxicol. 2015.
    1. Grimm FA, Iwata Y, Sirenko O, Bittner M, Rusyn I. High-Content Assay Multiplexing for Toxicity Screening in Induced Pluripotent Stem Cell-Derived Cardiomyocytes and Hepatocytes. Assay Drug Dev Technol. 2015.
    1. Young-Pearse TL, Morrow EM. Modeling developmental neuropsychiatric disorders with iPSC technology: challenges and opportunities. Curr Opin Neurobiol. 2015;36:66–73. doi:
    1. Stoccoa G, Lanzib G, Yuec F, Gilianib S, Sasakic K, Tommasinid A, et al. Patients' induced pluripotent stem cells to model drug induced adverse events: a role in predicting thiopurine induced pancreatitis? Curr Drug Metab. 2015.
    1. de Boer AS, Eggan K. A perspective on stem cell modeling of Amyotrophic Lateral Sclerosis. Cell cycle. 2015:0.
    1. Darabi R, Arpke RW, Irion S, Dimos JT, Grskovic M, Kyba M, et al. Human ES- and iPS-derived myogenic progenitors restore DYSTROPHIN and improve contractility upon transplantation in dystrophic mice. Cell Stem Cell. 2012;10(5):610–9. doi:
    1. Takahashi J. [Cell replacement therapy for Parkinson's disease using iPS cells]. Rinsho Shinkeigaku. 2013;53(11):1009–12.
    1. Okano H, Yamanaka S. iPS cell technologies: significance and applications to CNS regeneration and disease. Mol Brain. 2014;7:22 doi:
    1. Nieweg K, Andreyeva A, van Stegen B, Tanriover G, Gottmann K. Alzheimer's disease-related amyloid-beta induces synaptotoxicity in human iPS cell-derived neurons. Cell Death Dis. 2015;6:e1709 doi:
    1. Singh R, Shen W, Kuai D, Martin JM, Guo X, Smith MA, et al. iPS cell modeling of Best disease: insights into the pathophysiology of an inherited macular degeneration. Human molecular genetics. 2013;22(3):593–607. doi:
    1. Singh R, Kuai D, Guziewicz KE, Meyer J, Wilson M, Lu J, et al. Pharmacological Modulation of Photoreceptor Outer Segment Degradation in a Human iPS Cell Model of Inherited Macular Degeneration. Mol Ther. 2015.
    1. Raikwar SP, Kim EM, Sivitz WI, Allamargot C, Thedens DR, Zavazava N. Human iPS cell-derived insulin producing cells form vascularized organoids under the kidney capsules of diabetic mice. PLoS ONE. 2015;10(1):e0116582 doi:
    1. Maehr R. iPS cells in type 1 diabetes research and treatment. Clin Pharmacol Ther. 2011;89(5):750–3. doi:
    1. Dec E FD, Nalbandian A, Gargus M, Katheria V, Rana P, Ibrahim A, Hatch M, Lan M, Llewellyn KJ, Keirstead H and Kimonis VE. Disease-Specific Induced Pluripotent Stem Cell Modeling: Insights into the Pathophysiology of Valosin Containing Protein (VCP) Disease. Journal of Stem Cell Research & Therapy. 2014;4(2).
    1. Mizuno Y, Chang H, Umeda K, Niwa A, Iwasa T, Awaya T, et al. Generation of skeletal muscle stem/progenitor cells from murine induced pluripotent stem cells. FASEB J. 2010;24(7):2245–53. doi:
    1. Lal H, Emmett-Oglesby MW. Behavioral analogues of anxiety. Animal models. Neuropharmacology. 1983;22(12B):1423–41.
    1. Roca I, Requena J, Edel MJ, Alvarez-Palomo AB. Myogenic Precursors from iPS Cells for Skeletal Muscle Cell Replacement Therapy. J Clin Med. 2015;4(2):243–59. doi:
    1. Meola G, Velicogna M, Brigato C, Pizzul S, Rotondo G, Scarlato G. Growth and differentiation of myogenic clones from adult human muscle cell cultures. Eur J Basic Appl Histochem. 1991;35(3):219–31.
    1. Kaufman SJ, George-Weinstein M, Foster RF. In vitro development of precursor cells in the myogenic lineage. Dev Biol. 1991;146(1):228–38.
    1. Quinn LS, Holtzer H, Nameroff M. Generation of chick skeletal muscle cells in groups of 16 from stem cells. Nature. 1985;313(6004):692–4.
    1. Schultz E, Jaryszak DL. Effects of skeletal muscle regeneration on the proliferation potential of satellite cells. Mech Ageing Dev. 1985;30(1):63–72.
    1. Bischoff R, Holtzer H. Mitosis and the processes of differentiation of myogenic cells in vitro. The Journal of cell biology. 1969;41(1):188–200.
    1. Shelton M, Kocharyan A, Liu J, Skerjanc IS, Stanford WL. Robust generation and expansion of skeletal muscle progenitors and myocytes from human pluripotent stem cells. Methods. 2015.
    1. Chal J, Oginuma M, Al Tanoury Z, Gobert B, Sumara O, Hick A, et al. Differentiation of pluripotent stem cells to muscle fiber to model Duchenne muscular dystrophy. Nature biotechnology. 2015;33(9):962–9. doi:
    1. Salani S, Donadoni C, Rizzo F, Bresolin N, Comi GP, Corti S. Generation of skeletal muscle cells from embryonic and induced pluripotent stem cells as an in vitro model and for therapy of muscular dystrophies. Journal of cellular and molecular medicine. 2012;16(7):1353–64. doi:
    1. Borchin B, Chen J, Barberi T. Derivation and FACS-mediated purification of PAX3+/PAX7+ skeletal muscle precursors from human pluripotent stem cells. Stem cell reports. 2013;1(6):620–31. doi:
    1. Abujarour R, Valamehr B. Generation of skeletal muscle cells from pluripotent stem cells: advances and challenges. Front Cell Dev Biol. 2015;3:29 doi:
    1. Awaya T, Kato T, Mizuno Y, Chang H, Niwa A, Umeda K, et al. Selective development of myogenic mesenchymal cells from human embryonic and induced pluripotent stem cells. PLoS ONE. 2012;7(12):e51638 doi:
    1. Zhou YP, Rochat A, Hatzfeld A, Peiffer I, Barbet R, Hatzfeld J, et al. [bFGF-stimulated MEF-conditioned medium is capable of maintaining human embryonic stem cells]. Fen Zi Xi Bao Sheng Wu Xue Bao. 2009;42(3–4):193–9.
    1. Siddiqui A, Bhaumik D, Chinta SJ, Rane A, Rajagopalan S, Lieu CA, et al. Mitochondrial Quality Control via the PGC1alpha-TFEB Signaling Pathway Is Compromised by Parkin Q311X Mutation But Independently Restored by Rapamycin. J Neurosci. 2015;35(37):12833–44. doi:
    1. Bove J, Martinez-Vicente M, Vila M. Fighting neurodegeneration with rapamycin: mechanistic insights. Nature reviews Neuroscience. 2011;12(8):437–52. doi:
    1. Richardson PG, Eng C, Kolesar J, Hideshima T, Anderson KC. Perifosine, an oral, anti-cancer agent and inhibitor of the Akt pathway: mechanistic actions, pharmacodynamics, pharmacokinetics, and clinical activity. Expert Opin Drug Metab Toxicol. 2012;8(5):623–33. doi:
    1. Balakrishnan K, Burger JA, Wierda WG, Gandhi V. AT-101 induces apoptosis in CLL B cells and overcomes stromal cell-mediated Mcl-1 induction and drug resistance. Blood. 2009;113(1):149–53. doi:
    1. Mei H, Lin Z, Wang Y, Wu G, Song Y. Autophagy inhibition enhances pan-Bcl-2 inhibitor AT-101-induced apoptosis in non-small cell lung cancer. Neoplasma. 2014;61(2):186–92. doi:
    1. Moore BR, Page-Sharp M, Stoney JR, Ilett KF, Jago JD, Batty KT. Pharmacokinetics, pharmacodynamics, and allometric scaling of chloroquine in a murine malaria model. Antimicrob Agents Chemother. 2011;55(8):3899–907. doi:
    1. Zirin J, Nieuwenhuis J, Perrimon N. Role of autophagy in glycogen breakdown and its relevance to chloroquine myopathy. PLoS Biol. 2013;11(11):e1001708 doi:
    1. Liu J, Xia H, Kim M, Xu L, Li Y, Zhang L, et al. Beclin1 controls the levels of p53 by regulating the deubiquitination activity of USP10 and USP13. Cell. 2011;147(1):223–34. doi:
    1. Choi YJ, Park YJ, Park JY, Jeong HO, Kim DH, Ha YM, et al. Inhibitory effect of mTOR activator MHY1485 on autophagy: suppression of lysosomal fusion. PLoS ONE. 2012;7(8):e43418 doi:
    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).
    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.
    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. doi:
    1. Tresse E, Salomons FA, Vesa J, Bott LC, Kimonis V, Yao TP, et al. VCP/p97 is essential for maturation of ubiquitin-containing autophagosomes and this function is impaired by mutations that cause IBMPFD. Autophagy. 2010;6(2):217–27.
    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.19(R1):R38–45. doi:
    1. Cai Z, Yan LJ. Rapamycin, Autophagy, and Alzheimer's Disease. J Biochem Pharmacol Res. 2013;1(2):84–90.
    1. Proud CG. Amino acids and mTOR signalling in anabolic function. Biochemical Society transactions. 2007;35(Pt 5):1187–90. doi:
    1. Klein JB, Barati MT, Wu R, Gozal D, Sachleben LR Jr., Kausar H, et al. Akt-mediated valosin-containing protein 97 phosphorylation regulates its association with ubiquitinated proteins. The Journal of biological chemistry. 2005;280(36):31870–81. doi:
    1. Vandermoere F, El Yazidi-Belkoura I, Slomianny C, Demont Y, Bidaux G, Adriaenssens E, et al. The valosin-containing protein (VCP) is a target of Akt signaling required for cell survival. The Journal of biological chemistry. 2006;281(20):14307–13. doi:
    1. Steinman RM, Mellman IS, Muller WA, Cohn ZA. Endocytosis and the recycling of plasma membrane. The Journal of cell biology. 1983;96(1):1–27.
    1. Liang X, Tang J, Liang Y, Jin R, Cai X. Suppression of autophagy by chloroquine sensitizes 5-fluorouracil-mediated cell death in gallbladder carcinoma cells. Cell Biosci. 2014;4(1):10 doi:
    1. Shintani T, Klionsky DJ. Autophagy in health and disease: a double-edged sword. Science (New York, NY. 2004;306(5698):990–5.
    1. Day K, Paterson B, Yablonka-Reuveni Z. A distinct profile of myogenic regulatory factor detection within Pax7+ cells at S phase supports a unique role of Myf5 during posthatch chicken myogenesis. Dev Dyn. 2009;238(4):1001–9. doi:
    1. Knappe S, Zammit PS, Knight RD. A population of Pax7-expressing muscle progenitor cells show differential responses to muscle injury dependent on developmental stage and injury extent. Frontiers in aging neuroscience. 2015;7:161 doi:
    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. doi:
    1. Huang S, Bjornsti MA, Houghton PJ. Rapamycins: mechanism of action and cellular resistance. Cancer Biol Ther. 2003;2(3):222–32.
    1. Zhang J, Hong Y, Shen J. Combination treatment with perifosine and MEK-162 demonstrates synergism against lung cancer cells in vitro and in vivo. Tumour Biol. 2015;36(7):5699–706. doi:
    1. Kim MN, Ro SW, Kim do Y, Kim da Y, Cho KJ, Park JH, et al. Efficacy of perifosine alone and in combination with sorafenib in an HrasG12V plus shp53 transgenic mouse model of hepatocellular carcinoma. Cancer chemotherapy and pharmacology. 2015;76(2):257–67. doi:
    1. Amantini C, Morelli MB, Santoni M, Soriani A, Cardinali C, Farfariello V, et al. Sorafenib induces cathepsin B-mediated apoptosis of bladder cancer cells by regulating the Akt/PTEN pathway. The Akt inhibitor, perifosine, enhances the sorafenib-induced cytotoxicity against bladder cancer cells. Oncoscience. 2015;2(4):395–409. doi:

Source: PubMed

3
Prenumerera