全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Administration of Recombinant Heat Shock Protein 70 Delays Peripheral Muscle Denervation in the SOD1G93A Mouse Model of Amyotrophic Lateral Sclerosis

DOI: 10.1155/2012/170426

Full-Text   Cite this paper   Add to My Lib

Abstract:

A prominent clinical feature of ALS is muscle weakness due to dysfunction, denervation and degeneration of motoneurons (MNs). While MN degeneration is a late stage event in the ALS mouse model, muscle denervation occurs significantly earlier in the disease. Strategies to prevent this early denervation may improve quality of life by maintaining muscle control and slowing disease progression. The precise cause of MN dysfunction and denervation is not known, but several mechanisms have been proposed that involve potentially toxic intra- and extracellular changes. Many cells confront these changes by mounting a stress response that includes increased expression of heat shock protein 70 (Hsp70). MNs do not upregulate Hsp70, and this may result in a potentially increased vulnerability. We previously reported that recombinant human hsp70 (rhHsp70) injections delayed symptom onset and increased lifespan in SOD1G93A mice. The exogenous rhHsp70 was localized to the muscle and not to spinal cord or brain suggesting it modulates peripheral pathophysiology. In the current study, we focused on earlier administration of Hsp70 and its effect on initial muscle denervation. Injections of the protein appeared to arrest denervation with preserved large myelinated peripheral axons, and reduced glial activation. 1. Introduction Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder affecting both upper and lower motoneurons (MNs), resulting in gradual muscle weakening and loss of MN function ultimately leading to paralysis and death of afflicted individuals. Much ALS research has focused on MN cell bodies and dendrites within the spinal cord and the role of local glial cells. Many therapeutic interventions have been designed to intervene in pathological events that occur in the anterior horn region with the goal of preventing MN cell body degeneration. However, the death of MNs occurs late in the disease process in mouse models, and even when physical degeneration of the cell is prevented, survival of the animal is only moderately prolonged [1]. More recently, research has focused attention on early events including muscle denervation that begins during the first postnatal month in the mouse [1–5]. Clinical onset of behavioral pathology is generally considered to occur during the third postnatal month; however, we, and others have observed more subtle behavior changes at earlier time points ([6]; unpublished observations). Furthermore, pathological events such as fragmentation of the Golgi apparatus, vacuolization of mitochondria, deficits in axonal transport,

References

[1]  T. W. Gould, R. R. Buss, S. Vinsant et al., “Complete dissociation of motor neuron death from motor dysfunction by Bax deletion in a mouse model of ALS,” Journal of Neuroscience, vol. 26, no. 34, pp. 8774–8786, 2006.
[2]  D. Frey, C. Schneider, L. Xu, J. Borg, W. Spooren, and P. Caroni, “Early and selective loss of neuromuscular synapse subtypes with low sprouting competence in motoneuron diseases,” Journal of Neuroscience, vol. 20, no. 7, pp. 2534–2542, 2000.
[3]  L. R. Fischer, D. G. Culver, P. Tennant et al., “Amyotrophic lateral sclerosis is a distal axonopathy: evidence in mice and man,” Experimental Neurology, vol. 185, no. 2, pp. 232–240, 2004.
[4]  A. M. Schaefer, J. R. Sanes, and J. W. Lichtman, “A compensatory subpopulation of motor neurons in a mouse model of amyotrophic lateral sclerosis,” Journal of Comparative Neurology, vol. 490, no. 3, pp. 209–219, 2005.
[5]  S. Saxena, E. Cabuy, and P. Caroni, “A role for motoneuron subtype-selective ER stress in disease manifestations of FALS mice,” Nature Neuroscience, vol. 12, no. 5, pp. 627–636, 2009.
[6]  J. Amendola, B. Verrier, P. Roubertoux, and J. Durand, “Altered sensorimotor development in a transgenic mouse model of amyotrophic lateral sclerosis,” European Journal of Neuroscience, vol. 20, no. 10, pp. 2822–2826, 2004.
[7]  Z. Mourelatos, N. K. Gonatas, A. Stieber, M. E. Gurney, and M. C. Dal Canto, “The Golgi apparatus of spinal cord motor neurons in transgenic mice expressing mutant Cu,Zn superoxide dismutase becomes fragmented in early, preclinical stages of the disease,” Proceedings of the National Academy of Sciences of the United States of America, vol. 93, no. 11, pp. 5472–5477, 1996.
[8]  A. Stieber, Y. Chen, S. Wei et al., “The fragmented neuronal Golgi apparatus in amyotrophic lateral sclerosis includes the trans-Golgi-network functional implications,” Acta Neuropathologica, vol. 95, no. 3, pp. 245–253, 1998.
[9]  C. Bendotti, N. Calvaresi, L. Chiveri et al., “Early vacuolization and mitochondrial damage in motor neurons of FALS mice are not associated with apoptosis or with changes in cytochrome oxidase histochemical reactivity,” Journal of the Neurological Sciences, vol. 191, no. 1-2, pp. 25–33, 2001.
[10]  N. K. Gonatas, A. Stieber, and J. O. Gonatas, “Fragmentation of the Golgi apparatus in neurodegenerative diseases and cell death,” Journal of the Neurological Sciences, vol. 246, no. 1-2, pp. 21–30, 2006.
[11]  H. Ilieva, M. Polymenidou, and D. W. Cleveland, “Non-cell autonomous toxicity in neurodegenerative disorders: ALS and beyond,” Journal of Cell Biology, vol. 187, no. 6, pp. 761–772, 2009.
[12]  F. Ritossa, “A new puffing pattern induced by temperature shock and DNP in drosophila,” Experientia, vol. 18, no. 12, pp. 571–573, 1962.
[13]  R. A. Stetler, Y. Gan, W. Zhang et al., “Heat shock proteins: cellular and molecular mechanisms in the central nervous system,” Progress in Neurobiology, vol. 92, no. 2, pp. 184–211, 2010.
[14]  C. Voisine, J. S. Pedersen, and R. I. Morimoto, “Chaperone networks: tipping the balance in protein folding diseases,” Neurobiology of Disease, vol. 40, no. 1, pp. 12–20, 2010.
[15]  D. J. Gifondorwa, M. B. Robinson, C. D. Hayes et al., “Exogenous delivery of heat shock protein 70 increases lifespan in a mouse model of amyotrophic lateral sclerosis,” Journal of Neuroscience, vol. 27, no. 48, pp. 13173–13180, 2007.
[16]  G. E. Truett, P. Heeger, R. L. Mynatt, A. A. Truett, J. A. Walker, and M. L. Warman, “Preparation of PCR-quality mouse genomic dna with hot sodium hydroxide and tris (HotSHOT),” BioTechniques, vol. 29, no. 1, pp. 52–54, 2000.
[17]  T. D. Heiman-Patterson, J. S. Deitch, E. P. Blankenhorn et al., “Background and gender effects on survival in the TgN(SOD1-G93A)1Gur mouse model of ALS,” Journal of the Neurological Sciences, vol. 236, no. 1-2, pp. 1–7, 2005.
[18]  D. Brown, J. Lydon, M. McLaughlin, A. Stuart-Tilley, R. Tyszkowski, and S. Alper, “Antigen retrieval in cryostat tissue sections and cultured cells by treatment with sodium dodecyl sulfate (SDS),” Histochemistry and Cell Biology, vol. 105, no. 4, pp. 261–267, 1996.
[19]  M. Maeda, N. Ohba, S. Nakagomi et al., “Vesicular acetylcholine transporter can be a morphological marker for the reinnervation to muscle of regenerating motor axons,” Neuroscience Research, vol. 48, no. 3, pp. 305–314, 2004.
[20]  N. J. Buchkovich, T. G. Maguire, Y. Yu, A. W. Paton, J. C. Paton, and J. C. Alwine, “Human cytomegalovirus specifically controls the levels of the endoplasmic reticulum chaperone BiP/GRP78, which is required for virion assembly,” Journal of Virology, vol. 82, no. 1, pp. 31–39, 2008.
[21]  D. Kieran, B. Kalmar, J. R. T. Dick, J. Riddoch-Contreras, G. Burnstock, and L. Greensmith, “Treatment with arimoclomol, a coinducer of heat shock proteins, delays disease progression in ALS mice,” Nature Medicine, vol. 10, no. 4, pp. 402–405, 2004.
[22]  S. Pun, M. Sigrist, A. F. Santos et al., “An intrinsic distinction in neuromuscular junction assembly and maintenance in different skeletal muscles,” Neuron, vol. 34, no. 3, pp. 357–370, 2002.
[23]  S. Pun, A. F. Santos, S. Saxena, L. Xu, and P. Caroni, “Selective vulnerability and pruning of phasic motoneuron axons in motoneuron disease alleviated by CNTF,” Nature Neuroscience, vol. 9, no. 3, pp. 408–419, 2006.
[24]  M. C. Raff, A. V. Whitmore, and J. T. Finn, “Neuroscience: axonal self-destruction and neurodegeneration,” Science, vol. 296, no. 5569, pp. 868–871, 2002.
[25]  I. M. Medana and M. M. Esiri, “Axonal damage: a key predictor of outcome in human CNS diseases,” Brain, vol. 126, no. 3, pp. 515–530, 2003.
[26]  J. J. Palop, J. Chin, and L. Mucke, “A network dysfunction perspective on neurodegenerative diseases,” Nature, vol. 443, no. 7113, pp. 768–773, 2006.
[27]  T. W. Gould and R. W. Oppenheim, “Synaptic dysfunction in disease and following injury in the developing and adult nervous system: caveats in the choice of therapeutic intervention,” Neuroscience and Biobehavioral Reviews, vol. 31, no. 8, pp. 1073–1087, 2007.
[28]  E. D. Hall, J. A. Oostveen, and M. E. Gurney, “Relationship of microglial and astrocytic activation to disease onset and progression in a transgenic model of familial ALS,” GLIA, vol. 23, no. 3, pp. 249–256, 1998.
[29]  M. B. Robinson, J. L. Tidwell, T. Gould et al., “Extracellular heat shock protein 70: a critical component for motoneuron survival,” Journal of Neuroscience, vol. 25, no. 42, pp. 9735–9745, 2005.
[30]  P. L. McGeer and E. G. McGeer, “Inflammatory processes in amyotrophic lateral sclerosis,” Muscle and Nerve, vol. 26, no. 4, pp. 459–470, 2002.
[31]  S. A. Sargsyan, P. N. Monk, and P. J. Shaw, “Microglia as potential contributors to motor neuron injury in amyotrophic lateral sclerosis,” GLIA, vol. 51, no. 4, pp. 241–253, 2005.
[32]  T. Magnus, M. Beck, R. Giess, I. Puls, M. Naumann, and K. V. Toyka, “Disease progression in amyotrophic lateral sclerosis: predictors of survival,” Muscle and Nerve, vol. 25, no. 5, pp. 709–714, 2002.
[33]  P. H. Gordon, B. Cheng, I. B. Katz, H. Mitsumoto, and L. P. Rowland, “Clinical features that distinguish PLS, upper motor neuron-dominant ALS, and typical ALS,” Neurology, vol. 72, no. 22, pp. 1948–1952, 2009.
[34]  J. R. Daube, “Electrophysiologic studies in the diagnosis and prognosis of motor neuron diseases,” Neurologic Clinics, vol. 3, no. 3, pp. 473–493, 1985.
[35]  M. Dadon-Nachum, E. Melamed, and D. Offen, “The “dying-back” phenomenon of motor neurons in ALS,” Journal of Molecular Neuroscience, vol. 43, no. 3, pp. 470–477, 2011.
[36]  L. Dupuis and J. P. Loeffler, “Neuromuscular junction destruction during amyotrophic lateral sclerosis: insights from transgenic models,” Current Opinion in Pharmacology, vol. 9, no. 3, pp. 341–346, 2009.
[37]  T. Gordon, J. Hegedus, and S. L. Tam, “Adaptive and maladaptive motor axonal sprouting in aging and motoneuron disease,” Neurological Research, vol. 26, no. 2, pp. 174–185, 2004.
[38]  T. Gordon, V. Ly, J. Hegedus, and N. Tyreman, “Early detection of denervated muscle fibers in hindlimb muscles after sciatic nerve transection in wild type mice and in the G93A mouse model of amyotrophic lateral sclerosis,” Neurological Research, vol. 31, no. 1, pp. 28–42, 2009.
[39]  J. Hegedus, C. T. Putman, and T. Gordon, “Time course of preferential motor unit loss in the mouse model of amyotrophic lateral sclerosis,” Neurobiology of Disease, vol. 28, no. 2, pp. 154–164, 2007.
[40]  J. Hegedus, C. T. Putman, N. Tyreman, and T. Gordon, “Preferential motor unit loss in the SOD1 G93A transgenic mouse model of amyotrophic lateral sclerosis,” Journal of Physiology, vol. 586, no. 14, pp. 3337–3351, 2008.
[41]  C. R. Hayworth and F. Gonzalez-Lima, “Pre-symptomatic detection of chronic motor deficits and genotype prediction in congenic B6. ALS mouse model,” Neuroscience, vol. 164, no. 3, pp. 975–985, 2009.
[42]  N. Kafkafi, D. Yekutieli, P. Yarowsky, and G. I. Elmer, “Data mining in a behavioral test detects early symptoms in a model of amyotrophic lateral sclerosis,” Behavioral Neuroscience, vol. 122, no. 4, pp. 777–787, 2008.
[43]  S. J. Feeney, P. A. McKelvie, L. Austin et al., “Presymptomatic motor neuron loss and reactive astrocytosis in the SOD1 mouse model of amyotrophic lateral sclerosis,” Muscle and Nerve, vol. 24, no. 11, pp. 1510–1519, 2001.
[44]  H. Guo, L. Lai, M. E. R. Butchbach et al., “Increased expression of the glial glutamate transporter EAAT2 modulates excitotoxicity and delays the onset but not the outcome of ALS in mice,” Human Molecular Genetics, vol. 12, no. 19, pp. 2519–2532, 2003.
[45]  J. S. Henkel, D. R. Beers, W. Zhao, and S. H. Appel, “Microglia in ALS: the good, the bad, and the resting,” Journal of Neuroimmune Pharmacology, vol. 4, no. 4, pp. 389–398, 2009.
[46]  J. B. Levine, J. Kong, M. Nadler, and Z. Xu, “Astrocytes interact intimately with degenerating motor neurons in mouse amyotrophic lateral sclerosis (ALS),” GLIA, vol. 28, no. 3, pp. 215–224, 1999.
[47]  C. Neusch, M. B?hr, and C. Schneider-Gold, “Glia cells in amyotrophic lateral sclerosis: new clues to understanding an old disease?” Muscle and Nerve, vol. 35, no. 6, pp. 712–724, 2007.
[48]  S. A. Sargsyan, D. J. Blackburn, S. C. Barber, P. N. Monk, and P. J. Shaw, “Mutant microglia have an inflammatory phenotype and elevated production of MCP-1,” NeuroReport, vol. 20, no. 16, pp. 1450–1455, 2009.
[49]  W. W. Yang, R. L. Sidman, T. V. Taksir, et al., “Relationship between neuropathology and disease progression in the ALS mouse,” Experimental Neurology, vol. 227, pp. 287–295, 2010.
[50]  D. Papadimitriou, V. Le Verche, A. Jacquier, B. Ikiz, S. Przedborski, and D. B. Re, “Inflammation in ALS and SMA: sorting out the good from the evil,” Neurobiology of Disease, vol. 37, no. 3, pp. 493–502, 2010.
[51]  J. M. Edgar and K. A. Nave, “The role of CNS glia in preserving axon function,” Current Opinion in Neurobiology, vol. 19, no. 5, pp. 498–504, 2009.
[52]  J. Kriz, M. D. Nguyen, and J. P. Julien, “Minocycline slows disease progression in a mouse model of amyotrophic lateral sclerosis,” Neurobiology of Disease, vol. 10, no. 3, pp. 268–278, 2002.
[53]  J. Kriz, G. Gowing, and J. P. Julien, “Efficient three-drug cocktail for disease induced by mutant superoxide dismutase,” Annals of Neurology, vol. 53, no. 4, pp. 429–436, 2003.
[54]  L. Van Den Bosch, P. Tilkin, G. Lemmens, and W. Robberecht, “Minocycline delays disease onset and mortality in a transgenic model of ALS,” NeuroReport, vol. 13, no. 8, pp. 1067–1070, 2002.
[55]  S. Zhu, I. G. Stavrovskaya, M. Drozda et al., “Minocycline inhibits cytochrome c release and delays progression of amyotrophic lateral sclerosis in mice,” Nature, vol. 417, no. 6884, pp. 74–78, 2002.
[56]  A. F. Keller, M. Gravel, and J. Kriz, “Treatment with minocycline after disease onset alters astrocyte reactivity and increases microgliosis in SOD1 mutant mice,” Experimental Neurology, vol. 228, no. 1, pp. 69–79, 2011.
[57]  L. H. Chamberlain and R. D. Burgoyne, “Cysteine string protein functions directly in regulated exocytosis,” Molecular Biology of the Cell, vol. 9, no. 8, pp. 2259–2267, 1998.
[58]  L. H. Chamberlain and R. D. Burgoyne, “Cysteine-string protein: the chaperone at the synapse,” Journal of Neurochemistry, vol. 74, no. 5, pp. 1781–1789, 2000.
[59]  Z. Ying, H. Wang, H. Fan et al., “Gp78, an ER associated E3, promotes SOD1 and ataxin-3 degradation,” Human Molecular Genetics, vol. 18, no. 22, pp. 4268–4281, 2009.
[60]  K. Vijayalakshmi, P. A. Alladi, S. Ghosh et al., “Evidence of endoplasmic reticular stress in the spinal motor neurons exposed to CSF from sporadic amyotrophic lateral sclerosis patients,” Neurobiology of Disease, vol. 41, no. 3, pp. 695–705, 2011.
[61]  M. Locke, E. G. Noble, and B. G. Atkinson, “Inducible isoform of HSP70 is constitutively expressed in a muscle fiber type specific pattern,” American Journal of Physiology, vol. 261, no. 5, pp. C774–C779, 1991.
[62]  M. Locke, B. G. Atkinson, R. M. Tanguay, and E. G. Noble, “Shifts in type I fiber proportion in rat hindlimb muscle are accompanied by changes in HSP72 content,” American Journal of Physiology, vol. 266, no. 5, pp. C1240–C1246, 1994.
[63]  T. Hoshino, N. Murao, T. Namba et al., “Suppression of Alzheimer's disease-related phenotypes by expression of heat shock protein 70 in mice,” Journal of Neuroscience, vol. 31, no. 14, pp. 5225–5234, 2011.
[64]  J. Liu, L. A. Shinobu, C. M. Ward, D. Young, and D. W. Cleveland, “Elevation of the Hsp70 chaperone does not effect toxicity in mouse models of familial amyotrophic lateral sclerosis,” Journal of Neurochemistry, vol. 93, no. 4, pp. 875–882, 2005.
[65]  K. H. J. Park and I. Vincent, “Presymptomatic biochemical changes in hindlimb muscle of G93A human Cu/Zn superoxide dismutase 1 transgenic mouse model of amyotrophic lateral sclerosis,” Biochimica et Biophysica Acta, vol. 1782, no. 7-8, pp. 462–468, 2008.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133