Inhibition of retrograde transport modulates misfolded protein accumulation and clearance in motoneuron diseases

Riccardo Cristofani, Valeria Crippa, Paola Rusmini, Maria Elena Cicardi, Marco Meroni, Nausicaa V Licata, Gessica Sala, Elisa Giorgetti, Christopher Grunseich, Mariarita Galbiati, Margherita Piccolella, Elio Messi, Carlo Ferrarese, Serena Carra, Angelo Poletti, Riccardo Cristofani, Valeria Crippa, Paola Rusmini, Maria Elena Cicardi, Marco Meroni, Nausicaa V Licata, Gessica Sala, Elisa Giorgetti, Christopher Grunseich, Mariarita Galbiati, Margherita Piccolella, Elio Messi, Carlo Ferrarese, Serena Carra, Angelo Poletti

Abstract

Motoneuron diseases, like spinal bulbar muscular atrophy (SBMA) and amyotrophic lateral sclerosis (ALS), are associated with proteins that because of gene mutation or peculiar structures, acquire aberrant (misfolded) conformations toxic to cells. To prevent misfolded protein toxicity, cells activate a protein quality control (PQC) system composed of chaperones and degradative pathways (proteasome and autophagy). Inefficient activation of the PQC system results in misfolded protein accumulation that ultimately leads to neuronal cell death, while efficient macroautophagy/autophagy-mediated degradation of aggregating proteins is beneficial. The latter relies on an active retrograde transport, mediated by dynein and specific chaperones, such as the HSPB8-BAG3-HSPA8 complex. Here, using cellular models expressing aggregate-prone proteins involved in SBMA and ALS, we demonstrate that inhibition of dynein-mediated retrograde transport, which impairs the targeting to autophagy of misfolded species, does not increase their aggregation. Rather, dynein inhibition correlates with a reduced accumulation and an increased clearance of mutant ARpolyQ, SOD1, truncated TARDBP/TDP-43 and expanded polyGP C9ORF72 products. The enhanced misfolded protein clearance is mediated by the proteasome, rather than by autophagy and correlates with the upregulation of the HSPA8 cochaperone BAG1. In line, overexpression of BAG1 increases the proteasome-mediated clearance of these misfolded proteins. Our data suggest that when the misfolded proteins cannot be efficiently transported toward the perinuclear region of the cells, where they are either degraded by autophagy or stored into the aggresome, the cells activate a compensatory mechanism that relies on the induction of BAG1 to target the HSPA8-bound cargo to the proteasome in a dynein-independent manner.

Keywords: BAG1; BAG3; HSPB8; aggregation; amyotrophic lateral sclerosis; autophagy; misfolded protein; proteasome; protein quality control.

Figures

Figure 1.
Figure 1.
Autophagic clearance of ARpolyQ. NSC34 cells cotransfected with AR.Q46 and pCDNA3 or HSPB8, and treated with vehicle (ethanol, EtOH) or 10 nM T, for 48 h. (A) Immunofluorescence microscopy (IF) analysis (63x magnification) of AR (green); nuclei were stained with DAPI (blue); scale bar: 20 μm. (B) WB and FRA show AR.Q46 total levels and AR.Q46 insoluble fraction respectively. TUBA was used as loading control. Bar graph represents the FRA mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD (* = P < 0.05, *** = P < 0.001; Two-Way ANOVA followed by the Uncorrected Fisher LSD test). (C) Phase contrast imaging and IF performed on NSC34 cells transfected with Dync1h1 siRNA show cell morphology and dynein distribution. Scale bar: 20 μm. (D) WB analysis of NSC34 cells transfected with Dync1h1 siRNA shows the levels of dynein heavy chain; TUBA was used as loading control. Bar graph represents the WB mean relative optical density measured over 3 independent biological samples for each condition (n = 3) ± SD (** = P<0.01, one-tailed unpaired Student t test). (E) FRA and WB of NSC34 stably expressing GFP-AR.Q39 and transfected with Dync1h1 siRNA show AR.Q39 insoluble fraction accumulation after 10 nM T treatment and reduction after dynein silencing. Bar graph represents the FRA mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD (* = P < 0.05, ** = P < 0.01; Two-Way ANOVA followed by the Uncorrected Fisher LSD test).
Figure 2.
Figure 2.
Effect of dynein-silencing on autophagy and ARpolyQ clearance. (A) Confocal microscopy analysis (40x magnification) performed on NSC34 cells transfected with Dync1h1 siRNA shows SQSTM1 or LC3 distribution (green); nuclei were stained with DAPI (blue); scale bars: 50 μm. Bar graphs represent the quantification of total green-fluorescence integrated density (IntDen) normalized on cells number. Six images were analyzed for each condition. (B) WB of LC3-I and LC3-II levels performed on NSC34 transfected with Dync1h1 siRNA and treated with 20 mM NH4Cl for the last 1.5 h. Nontargeting siRNA was used as control; TUBA was used as loading control. Bar graph represents the mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD (* = P < 0.05, ** = P < 0.01; Two-Way ANOVA followed by the Uncorrected Fisher LSD test); (C) WB of LC3-I and LC3-II levels performed on NSC34 treated with 100 μM EHNA and/or 20 mM NH4Cl for the last 1.5 h. Nontargeting siRNA was used as control; TUBA was used as loading control. Bar graphs represent the mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD (* = P < 0.05, ** = P < 0.01; Two-Way ANOVA followed by the Uncorrected Fisher LSD test).
Figure 3.
Figure 3.
Autophagy modulation by EHNA treatment. NSC34 cells treated with DMSO, 100 μM EHNA and/or 100 mM trehalose for 48 h; (A, B) the bar graph represents Sqstm1 and Lc3 mRNA relative levels normalized with Rplp0 mRNA levels. Analysis was conducted on 4 independent biological samples for each condition (n = 4) ± SD (** = P<0.01, *** = P<0.001; Two-Way ANOVA followed by the Uncorrected Fisher LSD test). (C) Confocal microscopy analysis shows SQSTM1 and LC3 localization (40x magnification); scale bars: 50 μm. Bar graph represents the quantification of total green fluorescence integrated density (IntDen) normalized on cells number. Six images were analyzed for each condition (n = 6) ± SD (** = P < 0.01, ** = P < 0.001; one-tailed unpaired Student t test). Lower gain was used to acquire trehalose-treated cells (low sensitivity [L.S.]) compared to control (high sensitivity [H.S.]). (D) WB analysis shows SQSTM1 and LC3 levels; TUBA was used as loading control. Bar graphs represent the mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD of SQSTM1:TUBA (middle inset) and LC3-II:TUBA (low inset) ratio (* = P < 0.05, ** = P < 0.01, *** = P < 0.001; One-Way ANOVA followed by the Uncorrected Fisher LSD test).
Figure 4.
Figure 4.
Effect of dynein ATPase activity-inhibition on ARpolyQ clearance. FRA and WB show ARpolyQ total levels and ARpolyQ insoluble fraction, respectively, of: (A) PC12 cells stably transfected with AR.Q10 and AR.Q112 induced with 1 μg/mL doxycycline for 72 h. Cells were treated with ethanol (EtOH) or 10 nM T and/or EHNA 100 μM for the last 48 h prior to analysis. TUBA was used as loading control and bar graphs represent the FRA mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD (*** = P<0.001; One-Way ANOVA followed by the Uncorrected Fisher LSD test); (B) TR4 NSC34 cells stably transfected with GFP-AR.Q0 induced with 1 μg/mL doxycycline for 72 h; (C) TR4 NSC34 cells stably transfected with GFP-AR.Q39 induced with 1 μg/mL doxycycline for 72 h; (D) NSC34 cells transiently transfected with AR.Q46 for 48 h. Cells were treated with ethanol (EtOH) or 10 nM T and/or EHNA 100 μM for the last 48 h prior to analysis. (C and D) TUBA was used as loading control and bar graphs represent the FRA mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD (* = P < 0.05, ** = P < 0.01; Two-Way ANOVA followed by the Uncorrected Fisher LSD test). (E) IF performed on NSC34 cells transfected with AR.Q46 and treated with 10 nM T or EtOH and 100 μM EHNA or DMSO for 48 h. AR was stained with AR H280 antibody (green), nuclei were stained with DAPI (blue) (63x magnification); scale bar: 20 μm. (F) Bar graph represents the mean percentage of transfected cells with inclusions computed over 3 independent biological samples for each condition (n = 3) ± SD (* = P < 0.05, *** = P < 0.001; Two-Way ANOVA followed by the Uncorrected Fisher LSD test). Ten fields for each biological sample were analyzed. (G) WB analysis of PBS, Triton X100, SDS and FA lysates fractions of NSC34 cells transfected with AR.Q46 and treated with 10 nM T or EtOH and 100 μM EHNA or DMSO for 48 h. GAPDH was used as loading control.
Figure 5.
Figure 5.
Dynein ATPase activity inhibition promotes ARpolyQ clearance via UPS. (A) NSC34 cells stably transfected with GFP-AR.Q39, treated with 10 nM T, 10 mM 3MA and/or 100 μM EHNA for the last 48 h and/or 10 μM MG132 for the last 16 h. WB shows ARpolyQ levels and FRA shows ARpolyQ insoluble fraction. Bar graph represents the FRA mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD (** = P < 0.01; *** = P < 0.001; one-tailed unpaired Student t test). (B) NSC34 cells transfected with GFPu reporter and/or ARpolyQ and treated with 10 nM T and/or 100 μM EHNA. WB shows ARpolyQ and GFPu bands; TUBA was used as loading control. Bar graph represents the GFP WB densitometric analysis of 3 independent samples for each condition (n = 3) ± SD (* = P < 0.05; *** = P < 0.001; Two-Way ANOVA followed by the Uncorrected Fisher LSD test); (C) Proteasome chymotryptic activity assay performed on NSC34 treated with DMSO or 100 μM EHNA and 10 μM MG132 used as negative control. Bar graph represents the mean relative fluorescence normalized on control group and computed over 4 independent biological samples for each condition (n = 4) ± SD (*** = P < 0.001, One-Way ANOVA followed by the Uncorrected Fisher LSD test). (D) Proteasome chymotryptic activity assay performed on NSC34 transfected with AR.Q46 and treated with 10 nM T, DMSO or 100 μM EHNA and 10 μM MG132 used as negative control. Bar graph represents the mean relative fluorescence normalized on control group and computed over 4 independent biological samples for each condition (n = 4) ± SD (*** = P < 0.001, One-Way ANOVA followed by the Uncorrected Fisher LSD test). (E) β-galactosidase assay performed on NSC34 cells transfected with pCMV-β and treated with DMSO or 100 μM EHNA for 24 or 48 h after transfection. Bar graph represents the mean relative absorbance normalized on transfected cells treated for 48 h and computed over 6 independent biological samples for each condition (n = 6) ± SD (Two-Way ANOVA, not significant). (F) MTT assay performed on NSC34 cells treated with DMSO or 100 μM EHNA for 24 or 48 h. Bar graph represents the mean relative absorbance normalized on untreated group and computed over 6 independent biological samples for each condition (n = 6) ± SD (One-Way ANOVA, not significant).
Figure 6.
Figure 6.
BAG1 upregulation reduces ARpolyQ accumulation. (A to C) NSC34 cells treated with DMSO or 100 μM EHNA for 48 h. The bar graphs represent Hspb8 (A), Bag3 (B) and Bag1 (C) mRNA levels normalized with Rplp0 mRNA levels. Four independent biological samples for each condition were analyzed (n = 4) ± SD (*** = P < 0.001; One-Way ANOVA followed by the Uncorrected Fisher LSD test). (D) NSC34 cells overexpressing AR.Q46 alone or coexpressing BAG1 and treated with T for last 48 h. FRA shows insoluble AR-polyQ levels (upper panel) and bar graph represents the FRA mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD (* = P < 0.05, *** = P < 0.001; Two-Way ANOVA followed by the Uncorrected Fisher LSD test). WB (lower panel) shows AR.Q46 and BAG1 protein levels. (E) NSC34 cells expressing AR.Q46 alone or coexpressing BAG1 and treated with 10 nM T for last 48 h and/or 10 μM MG132 for last 16 h. FRA shows insoluble AR-polyQ levels, bar graph represents the FRA mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD (* = P < 0.05, *** = P < 0.001; Two-Way ANOVA followed by the Uncorrected Fisher LSD test and one-tailed unpaired Student t test for control condition). (F) WB analysis of NSC34 cells transfected with Bag1 siRNA shows the levels of BAG1; TUBA was used as loading control. Bar graph represents the WB mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD (** = P < 0.01, one-tailed unpaired Student t test). (G) FRA and WB of NSC34 transfected with AR.Q46 and with Bag1 siRNA shows AR.Q46 insoluble fraction; TUBA was used as loading control. Bar graph represents the FRA densitometric analysis of 3 independent biological samples for each condition (n = 3) ± SD (** = P < 0.01; *** = P < 0.001; one-tailed unpaired Student t test).
Figure 7.
Figure 7.
EHNA effects on neuronal and motoneuronal cells obtained by differentiation of human iPSCs generated from SBMA patients. (A) IF analysis of neuronal and motoneuronal cells obtained by differentiation of human iPSCs generated from SBMA patients and treated with ethanol or 10 nM T (40x magnification). TUBB3 (green) and SMI312 (red) (upper inset), AR (green) and MNX1/HB9 (red) (lower inset). Nuclei were stained with DAPI (blue); scale bars: 30 μm. (B) Bar graph represents the percentage of MNX1/HB9-positive cells; 6 fields for each condition were analyzed (n = 6) ± SD (one-tailed unpaired Student t test, not significant). (C to I) RealTime PCR analyses performed on iPSCs differentiated to neuronal cells treated with 10 nM T and/or 100 μM EHNA for last 48 h. The bar graphs represent HSPB8 (C), BAG3 (D), BAG1 (E), SQSTM1 (F), LC3 (G), TFEB (H) and BECN1 (I) mRNA levels normalized with RPLP0 mRNA levels. Four independent samples for each condition were analyzed (n = 4) ± SD (* = P < 0.05, ** = P < 0.01, *** = P < 0.001; Two-Way ANOVA followed by the Uncorrected Fisher LSD test).
Figure 8.
Figure 8.
Effects of dynein inhibition on SOD1 and TARDBP insoluble species. (A) NSC34 cells expressing wtSOD1 and SOD1G93A treated with DMSO or 100 μM EHNA for last 48 h. FRA shows SOD1G93A accumulation compared to wtSOD1 and SOD1G93A reduction after 100 μM EHNA treatment. WB shows total level of transfected SOD1, TUBA was used as loading control. (B) NSC34 stably transfected with wtSOD1 or SOD1G93A induced with doxycycline and treated with DMSO or 100 μM EHNA for last 48 h. FRA shows SOD1G93A accumulation compared to wtSOD1 and SOD1G93A reduction after 100 μM EHNA treatment. WB shows total level of transfected SOD1, TUBA was used as loading control. (C) NSC34 expressing FL and TARDBPΔC treated with DMSO or 100 μM EHNA for last 48 h. FRA shows TARDBPΔC accumulation compared to FL TARDBP and TARDBPΔC reduction after 100 μM EHNA treatment. WB shows total level of transfected TARDBP, TUBA was used as loading control; (A to C) Bar graphs represent the FRA mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD (** = P < 0.01, *** = P < 0.001; Two-Way ANOVA followed by the Uncorrected Fisher LSD test). (D and E) NSC34 expressing SOD1G93A (D) or TARDBPΔC (E) and treated with DMSO, 100 μM EHNA and/or 10 mM 3MA for the last 48 h and 10 μM MG132 for the last 16 h. FRAs show SOD1G93A (D) or TARDBPΔC (E) accumulation after 3MA and MG132 treatments. EHNA treatment counteracts 3MA activity but not MG132 activity. WBs show total level of transfected SOD1 (D) or TARDBPΔC (E), TUBA or GAPDH was used as loading control. Bar graphs represent the FRA mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD (* = P < 0.05, ** = P < 0.01, *** = P < 0.001; Two-Way ANOVA followed by the Uncorrected Fisher LSD test and one-tailed unpaired Student t test for control conditions).
Figure 9.
Figure 9.
Effects of dynein inhibition on poly-GP insoluble fraction. (A) NSC34 cells expressing poly-GP (100 repeats) treated with 10 mM 3MA and/or 100 μM EHNA for the last 48 h prior to analysis and/or 10 μM MG132 for the last 16 h prior to analysis. FRA shows poly-GP insoluble fraction. Bar graphs represent the FRA mean relative optical density computed over 3 independent biological samples for each condition (n = 3) ± SD (** = P < 0.01; *** = P < 0.001; one-tailed unpaired Student t test); (B) the portion shown in A by the rectangle has been magnified to show the differences between short bars. WB shows total level of transfected poly-GP; TUBA was used as loading control.

References

    1. Cleveland DW, Rothstein JD. From Charcot to Lou Gehrig: deciphering selective motor neuron death in ALS. Nat Rev Neurosci 2001; 2:806-19; PMID:11715057;
    1. Boillée S, Vande Velde C, Cleveland DW. ALS: a disease of motor neurons and their nonneuronal neighbors. Neuron 2006; 52:39-59; PMID:17015226;
    1. Lobsiger CS, Boillee S, McAlonis-Downes M, Khan AM, Feltri ML, Yamanaka K, Cleveland DW. Schwann cells expressing dismutase active mutant SOD1 unexpectedly slow disease progression in ALS mice. Proc Natl Acad Sci U S A 2009; 106:4465-70; PMID:19251638;
    1. Cortes CJ, Ling SC, Guo LT, Hung G, Tsunemi T, Ly L, Tokunaga S, Lopez E, Sopher BL, Bennett CF, et al.. Muscle expression of mutant androgen receptor accounts for systemic and motor neuron disease phenotypes in spinal and bulbar muscular atrophy. Neuron 2014; 82:295-307; PMID:24742458;
    1. Lieberman AP, Yu Z, Murray S, Peralta R, Low A, Guo S, Yu XX, Cortes CJ, Bennett CF, Monia BP, et al.. Peripheral androgen receptor gene suppression rescues disease in mouse models of spinal and bulbar muscular atrophy. Cell Rep 2014; 7:774-84; PMID:24746732;
    1. Dobrowolny G, Aucello M, Rizzuto E, Beccafico S, Mammucari C, Bonconpagni S, Belia S, Wannenes F, Nicoletti C, Del Prete Z, et al.. Skeletal muscle is a primary target of SOD1G93A-mediated toxicity. Cell Metab 2008; 8:425-36; PMID:19046573;
    1. Halievski K, Henley CL, Domino L, Poort JE, Fu M, Katsuno M, Adachi H, Sobue G, Breedlove SM, Jordan CL. Androgen-dependent loss of muscle BDNF mRNA in two mouse models of SBMA. Exp Neurol 2015; 269:224-32; PMID:25929689;
    1. Monks DA, Johansen JA, Mo K, Rao P, Eagleson B, Yu Z, Lieberman AP, Breedlove SM, Jordan CL. Overexpression of wild-type androgen receptor in muscle recapitulates polyglutamine disease. Proc Natl Acad Sci U S A 2007; 104:18259-64; PMID:17984063;
    1. Jordan CL, Lieberman AP. Spinal and bulbar muscular atrophy: a motoneuron or muscle disease? Curr Opin Pharmacol 2008; 8:752-8; PMID:18775514;
    1. Johansen JA, Yu Z, Mo K, Monks DA, Lieberman AP, Breedlove SM, Jordan CL. Recovery of function in a myogenic mouse model of spinal bulbar muscular atrophy. Neurobiol Dis 2009; 34:113-20; PMID:19211034;
    1. Oki K, Wiseman RW, Breedlove SM, Jordan CL. Androgen receptors in muscle fibers induce rapid loss of force but not mass: implications for spinal bulbar muscular atrophy. Muscle Nerve 2013; 47:823-34; PMID:23629944;
    1. Galbiati M, Crippa V, Rusmini P, Cristofani R, Cicardi ME, Giorgetti E, Onesto E, Messi E, Poletti A. ALS-related misfolded protein management in motor neurons and muscle cells. Neurochem Int 2014; 79:70-8; PMID:25451799;
    1. Baralle M, Buratti E, Baralle FE. The role of TDP-43 in the pathogenesis of ALS and FTLD. Biochem Soc Trans 2013; 41:1536-40; PMID:24256250;
    1. Cozzolino M, Pesaresi MG, Gerbino V, Grosskreutz J, Carri MT. Amyotrophic lateral sclerosis: new insights into underlying molecular mechanisms and opportunities for therapeutic intervention. Antioxid Redox Signal 2012; 17:1277-330; PMID:22413952;
    1. Ciechanover A, Kwon YT. Degradation of misfolded proteins in neurodegenerative diseases: therapeutic targets and strategies. Exp Mol Med 2015; 47:e147; PMID:25766616;
    1. La Spada AR, Wilson EM, Lubahn DB, Harding AE, Fischbeck KH. Androgen receptor gene mutations in X-linked spinal and bulbar muscular atrophy. Nature 1991; 352:77-9; PMID:2062380;
    1. Katsuno M, Adachi H, Kume A, Li M, Nakagomi Y, Niwa H, Sang C, Kobayashi Y, Doyu M, Sobue G. Testosterone reduction prevents phenotypic expression in a transgenic mouse model of spinal and bulbar muscular atrophy. Neuron 2002; 35:843-54; PMID:12372280;
    1. Poletti A. The polyglutamine tract of androgen receptor: from functions to dysfunctions in motor neurons. Front Neuroendocrinol 2004; 25:1-26; PMID:15183036;
    1. Robberecht W, Philips T. The changing scene of amyotrophic lateral sclerosis. Nat Rev Neurosci 2013; 14:248-64; PMID:23463272;
    1. Simeoni S, Mancini MA, Stenoien DL, Marcelli M, Weigel NL, Zanisi M, Martini L, Poletti A. Motoneuronal cell death is not correlated with aggregate formation of androgen receptors containing an elongated polyglutamine tract. Hum Mol Genet 2000; 9:133-44; PMID:10587588;
    1. Saudou F, Finkbeiner S, Devys D, Greenberg ME. Huntingtin acts in the nucleus to induce apoptosis but death does not correlate with the formation of intranuclear inclusions. Cell 1998; 95:55-66; PMID:9778247;
    1. Klement IA, Skinner PJ, Kaytor MD, Yi H, Hersch SM, Clark HB, Zoghbi HY, Orr HT. Ataxin-1 nuclear localization and aggregation - role in polyglutamine-induced disease in SCA1 transgenic mice. Cell 1998; 95:41-53; PMID:9778246;
    1. Arrasate M, Mitra S, Schweitzer ES, Segal MR, Finkbeiner S. Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature 2004; 431:805-10; PMID:15483602;
    1. Montie HL, Cho MS, Holder L, Liu Y, Tsvetkov AS, Finkbeiner S, Merry DE. Cytoplasmic retention of polyglutamine-expanded androgen receptor ameliorates disease via autophagy in a mouse model of spinal and bulbar muscular atrophy. Hum Mol Genet 2009; 18:1937-50; PMID:19279159;
    1. Piccioni F, Pinton P, Simeoni S, Pozzi P, Fascio U, Vismara G, Martini L, Rizzuto R, Poletti A. Androgen receptor with elongated polyglutamine tract forms aggregates that alter axonal trafficking and mitochondrial distribution in motor neuronal processes. Faseb J 2002; 16:1418-20; PMID:12205033
    1. Johnston JA, Illing ME, Kopito RR. Cytoplasmic dynein/dynactin mediates the assembly of aggresomes. Cell Motil Cytoskeleton 2002; 53:26-38; PMID:12211113;
    1. Carra S, Crippa V, Rusmini P, Boncoraglio A, Minoia M, Giorgetti E, Kampinga HH, Poletti A. Alteration of protein folding and degradation in motor neuron diseases: Implications and protective functions of small heat shock proteins. Prog Neurobiol 2012; 97:83-100; PMID:21971574;
    1. Crippa V, Carra S, Rusmini P, Sau D, Bolzoni E, Bendotti C, De Biasi S, Poletti A. A role of small heat shock protein B8 (HspB8) in the autophagic removal of misfolded proteins responsible for neurodegenerative diseases. Autophagy 2010; 6:958-60; PMID:20699640;
    1. Crippa V, Sau D, Rusmini P, Boncoraglio A, Onesto E, Bolzoni E, Galbiati M, Fontana E, Marino M, Carra S, et al.. The small heat shock protein B8 (HspB8) promotes autophagic removal of misfolded proteins involved in amyotrophic lateral sclerosis (ALS). Hum Mol Genet 2010; 19:3440-56; PMID:20570967;
    1. Carra S, Sivilotti M, Chavez Zobel AT, Lambert H, Landry J. HspB8, a small heat shock protein mutated in human neuromuscular disorders, has in vivo chaperone activity in cultured cells. Hum Mol Genet 2005; 14:1659-69; PMID:15879436;
    1. Giorgetti E, Rusmini P, Crippa V, Cristofani R, Boncoraglio A, Cicardi ME, Galbiati M, Poletti A. Synergic prodegradative activity of Bicalutamide and trehalose on the mutant androgen receptor responsible for spinal and bulbar muscular atrophy. Hum Mol Genet 2015; 24:64-75; PMID:25122660;
    1. Rusmini P, Crippa V, Giorgetti E, Boncoraglio A, Cristofani R, Carra S, Poletti A. Clearance of the mutant androgen receptor in motoneuronal models of spinal and bulbar muscular atrophy. Neurobiol Aging 2013; 34:2585-603; PMID:23810450;
    1. Crippa V, D'Agostino VG, Cristofani R, Rusmini P, Cicardi ME, Messi E, Loffredo R, Pancher M, Piccolella M, Galbiati M, et al.. Transcriptional induction of the heat shock protein B8 mediates the clearance of misfolded proteins responsible for motor neuron diseases. Sci Rep 2016; 6:22827; PMID:26961006;
    1. Carra S, Seguin SJ, Landry J. HspB8 and Bag3: a new chaperone complex targeting misfolded proteins to macroautophagy. Autophagy 2008; 4:237-9; PMID:18094623;
    1. Carra S. The stress-inducible HspB8-Bag3 complex induces the eIF2alpha kinase pathway: implications for protein quality control and viral factory degradation? Autophagy 2009; 5:428-9; PMID:19202352;
    1. Gamerdinger M, Carra S, Behl C. Emerging roles of molecular chaperones and co-chaperones in selective autophagy: focus on BAG proteins. J Mol Med (Berl) 2011; 89:1175-82; PMID:21818581;
    1. Takayama S, Reed JC. Molecular chaperone targeting and regulation by BAG family proteins. Nat Cell Biol 2001; 3:E237-41; PMID:11584289;
    1. Minoia M, Boncoraglio A, Vinet J, Morelli FF, Brunsting JF, Poletti A, Krom S, Reits E, Kampinga HH, Carra S. BAG3 induces the sequestration of proteasomal clients into cytoplasmic puncta: Implications for a proteasome-to-autophagy switch. Autophagy 2014; 10:1603-21; PMID:25046115;
    1. Arndt V, Dick N, Tawo R, Dreiseidler M, Wenzel D, Hesse M, Furst DO, Saftig P, Saint R, Fleischmann BK, et al.. Chaperone-assisted selective autophagy is essential for muscle maintenance. Curr Biol 2010; 20:143-8; PMID:20060297;
    1. Klionsky DJ, Abdelmohsen K, Abe A, Abedin MJ, Abeliovich H, Acevedo Arozena A, Adachi H, Adams CM, Adams PD, Adeli K, et al.. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy 2016; 12:1-222; PMID:26799652;
    1. Fuchs M, Poirier DJ, Seguin SJ, Lambert H, Carra S, Charette SJ, Landry J. Identification of the key structural motifs involved in HspB8/HspB6-Bag3 interaction. Biochem J 2010; 425:245-55;
    1. Gamerdinger M, Kaya AM, Wolfrum U, Clement AM, Behl C. BAG3 mediates chaperone-based aggresome-targeting and selective autophagy of misfolded proteins. EMBO Rep 2011; 12:149-56; PMID:21252941;
    1. Behl C. BAG3 and friends: co-chaperones in selective autophagy during aging and disease. Autophagy 2011; 7:795-8; PMID:21681022;
    1. Sondermann H, Scheufler C, Schneider C, Hohfeld J, Hartl FU, Moarefi I. Structure of a Bag/Hsc70 complex: convergent functional evolution of Hsp70 nucleotide exchange factors. Science 2001; 291:1553-7; PMID:11222862;
    1. Rauch JN, Gestwicki JE. Binding of human nucleotide exchange factors to heat shock protein 70 (Hsp70) generates functionally distinct complexes in vitro. J Biol Chem 2014; 289:1402-14; PMID:24318877;
    1. Garcia-Mata R, Bebok Z, Sorscher EJ, Sztul ES. Characterization and dynamics of aggresome formation by a cytosolic GFP-chimera. J Cell Biol 1999; 146:1239-54; PMID:10491388;
    1. Kawaguchi Y, Kovacs JJ, McLaurin A, Vance JM, Ito A, Yao TP. The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 2003; 115:727-38; PMID:14675537;
    1. Iwata A, Riley BE, Johnston JA, Kopito RR. HDAC6 and Microtubules Are Required for Autophagic Degradation of Aggregated Huntingtin. J Biol Chem 2005; 280:40282-92; PMID:16192271;
    1. Dompierre JP, Godin JD, Charrin BC, Cordelieres FP, King SJ, Humbert S, Saudou F. Histone deacetylase 6 inhibition compensates for the transport deficit in Huntington's disease by increasing tubulin acetylation. J Neurosci 2007; 27:3571-83; PMID:17392473;
    1. Pandey UB, Nie Z, Batlevi Y, McCray BA, Ritson GP, Nedelsky NB, Schwartz SL, DiProspero NA, Knight MA, Schuldiner O, et al.. HDAC6 rescues neurodegeneration and provides an essential link between autophagy and the UPS. Nature 2007; 447:859-63; PMID:17568747;
    1. Lee JY, Koga H, Kawaguchi Y, Tang W, Wong E, Gao YS, Pandey UB, Kaushik S, Tresse E, Lu J, et al.. HDAC6 controls autophagosome maturation essential for ubiquitin-selective quality-control autophagy. EMBO J 2010; 29:969-80; PMID:20075865;
    1. Munoz-Moreno R, Barrado-Gil L, Galindo I, Alonso C. Analysis of HDAC6 and BAG3-aggresome pathways in African swine fever viral factory formation. Viruses 2015; 7:1823-31; PMID:25856634;
    1. Sha Y, Pandit L, Zeng S, Eissa NT. A critical role for CHIP in the aggresome pathway. Mol Cell Biol 2009; 29:116-28; PMID:18955503;
    1. Kraft C, Peter M, Hofmann K. Selective autophagy: ubiquitin-mediated recognition and beyond. Nat Cell Biol 2010; 12:836-41; PMID:20811356;
    1. Weedon MN, Hastings R, Caswell R, Xie W, Paszkiewicz K, Antoniadi T, Williams M, King C, Greenhalgh L, Newbury-Ecob R, et al.. Exome sequencing identifies a DYNC1H1 mutation in a large pedigree with dominant axonal Charcot-Marie-Tooth disease. Am J Hum Genet 2011; 89:308-12; PMID:21820100;
    1. Harms MB, Ori-McKenney KM, Scoto M, Tuck EP, Bell S, Ma D, Masi S, Allred P, Al-Lozi M, Reilly MM, et al.. Mutations in the tail domain of DYNC1H1 cause dominant spinal muscular atrophy. Neurology 2012; 78:1714-20; PMID:22459677;
    1. Strickland AV, Schabhuttl M, Offenbacher H, Synofzik M, Hauser NS, Brunner-Krainz M, Gruber-Sedlmayr U, Moore SA, Windhager R, Bender B, et al.. Mutation screen reveals novel variants and expands the phenotypes associated with DYNC1H1. J Neurol 2015; 262(9):2124-34; PMID:26100331;
    1. Scoto M, Rossor AM, Harms MB, Cirak S, Calissano M, Robb S, Manzur AY, Martinez Arroyo A, Rodriguez Sanz A, Mansour S, et al.. Novel mutations expand the clinical spectrum of DYNC1H1-associated spinal muscular atrophy. Neurology 2015; 84:668-79; PMID:25609763;
    1. LaMonte BH, Wallace KE, Holloway BA, Shelly SS, Ascano J, Tokito M, Van Winkle T, Howland DS, Holzbaur EL. Disruption of dynein/dynactin inhibits axonal transport in motor neurons causing late-onset progressive degeneration. Neuron 2002; 34:715-27; PMID:12062019;
    1. Hafezparast M, Klocke R, Ruhrberg C, Marquardt A, Ahmad-Annuar A, Bowen S, Lalli G, Witherden AS, Hummerich H, Nicholson S, et al.. Mutations in dynein link motor neuron degeneration to defects in retrograde transport. Science 2003; 300:808-12; PMID:12730604;
    1. Ravikumar B, Acevedo-Arozena A, Imarisio S, Berger Z, Vacher C, O'Kane CJ, Brown SD, Rubinsztein DC. Dynein mutations impair autophagic clearance of aggregate-prone proteins. Nat Genet 2005; 37:771-6; PMID:15980862;
    1. Sau D, Rusmini P, Crippa V, Onesto E, Bolzoni E, Ratti A, Poletti A. Dysregulation of axonal transport and motorneuron diseases. Biol Cell 2011; 103:87-107; PMID:21250942;
    1. Teuchert M, Fischer D, Schwalenstoecker B, Habisch HJ, Bockers TM, Ludolph AC. A dynein mutation attenuates motor neuron degeneration in SOD1(G93A) mice. Exp Neurol 2006; 198:271-4; PMID:16427626;
    1. Zhang F, Strom A-L, Fukada K, Lee S, Hayward LJ, Zhu H. Interaction between Familial Amyotrophic Lateral Sclerosis (ALS)-linked SOD1 mutants and the dynein complex. J Biol Chem 2007; 282:16691-9; PMID:17403682;
    1. El-Kadi AM, Bros-Facer V, Deng W, Philpott A, Stoddart E, Banks G, Jackson GS, Fisher EM, Duchen MR, Greensmith L, et al.. The Legs at odd angles (Loa) mutation in cytoplasmic dynein ameliorates mitochondrial function in SOD1G93A mouse model for motor neuron disease. J Biol Chem 2010; 285:18627-39; PMID:20382740
    1. Behl C. Breaking BAG: The Co-Chaperone BAG3 in Health and Disease. Trends Pharmacol Sci 2016; 37(8):672-88; PMID:27162137;
    1. Nollen EA, Kabakov AE, Brunsting JF, Kanon B, Hohfeld J, Kampinga HH. Modulation of in vivo HSP70 chaperone activity by Hip and Bag-1. J Biol Chem 2001; 276:4677-82; PMID:11076956;
    1. Stenoien DL, Cummings CJ, Adams HP, Mancini MG, Patel K, DeMartino G, Marcelli M, Weigel NL, Mancini MA. Polyglutamine-expanded androgen receptors form aggregates that sequester heat shock proteins, proteasome components and SRC-1, and are suppressed by the HDJ-2 chaperone. Hum Mol Genet 1999; 8:731-41; PMID:10196362;
    1. Rusmini P, Simonini F, Crippa V, Bolzoni E, Onesto E, Cagnin M, Sau D, Ferri N, Poletti A. 17-AAG increases autophagic removal of mutant androgen receptor in spinal and bulbar muscular atrophy. Neurobiol Dis 2011; 41:83-95; PMID:20816782;
    1. Carra S, Seguin SJ, Lambert H, Landry J. HspB8 chaperone activity toward poly(Q)-containing proteins depends on its association with Bag3, a stimulator of macroautophagy. J Biol Chem 2008; 283:1437-44; PMID:18006506;
    1. Strong MJ. The evidence for altered RNA metabolism in amyotrophic lateral sclerosis (ALS). J Neurol Sci 2010; 288:1-12; PMID:19840884;
    1. Volkening K, Leystra-Lantz C, Yang W, Jaffee H, Strong MJ. Tar DNA binding protein of 43 kDa (TDP-43), 14-3-3 proteins and copper/zinc superoxide dismutase (SOD1) interact to modulate NFL mRNA stability. Implications for altered RNA processing in amyotrophic lateral sclerosis (ALS). Brain Res 2009; 1305:168-82; PMID:19815002;
    1. Xu Z, Graham K, Foote M, Liang F, Rizkallah R, Hurt M, Wang Y, Wu Y, Zhou Y. 14-3-3 protein targets misfolded chaperone-associated proteins to aggresomes. J Cell Sci 2013; 126:4173-86; PMID:23843611;
    1. Jia B, Wu Y, Zhou Y. 14-3-3 and aggresome formation: implications in neurodegenerative diseases. Prion 2014; 8(2):173-7; PMID:24549097;
    1. Gennerich A, Carter AP, Reck-Peterson SL, Vale RD. Force-induced bidirectional stepping of cytoplasmic dynein. Cell 2007; 131:952-65; PMID:18045537;
    1. Penningroth SM, Cheung A, Bouchard P, Gagnon C, Bardin CW. Dynein ATPase is inhibited selectively in vitro by erythro-9-[3-2-(hydroxynonyl)]adenine. Biochem Biophys Res Commun 1982; 104:234-40; PMID:6462140;
    1. Kaplan A, Reiner O. Linking cytoplasmic dynein and transport of Rab8 vesicles to the midbody during cytokinesis by the doublecortin domain-containing 5 protein. J Cell Sci 2011; 124:3989-4000; PMID:22159412;
    1. Tanaka M, Machida Y, Niu S, Ikeda T, Jana NR, Doi H, Kurosawa M, Nekooki M, Nukina N. Trehalose alleviates polyglutamine-mediated pathology in a mouse model of Huntington disease. Nat Med 2004; 10:148-54; PMID:14730359;
    1. Sarkar S, Davies JE, Huang Z, Tunnacliffe A, Rubinsztein DC. Trehalose, a novel mTOR-independent autophagy enhancer, accelerates the clearance of mutant huntingtin and alpha-synuclein. J Biol Chem 2007; 282:5641-52; PMID:17182613;
    1. Zhang X, Chen S, Song L, Tang Y, Shen Y, Jia L, Le W. MTOR-independent, autophagic enhancer trehalose prolongs motor neuron survival and ameliorates the autophagic flux defect in a mouse model of amyotrophic lateral sclerosis. Autophagy 2014; 10:588-602; PMID:24441414;
    1. Berger TR, Montie HL, Jain P, Legleiter J, Merry DE. Identification of novel polyglutamine-expanded aggregation species in spinal and bulbar muscular atrophy. Brain Res 2015; 1628:254-64; PMID:26453288;
    1. Heine EM, Berger TR, Pluciennik A, Orr CR, Merry DE. Proteasome-mediated proteolysis of the Polyglutamine-expanded androgen receptor is a late event in SBMA pathogenesis. J Biol Chem 2015; 290(20):12572-84; PMID:25795778;
    1. Zboray L, Pluciennik A, Curtis D, Liu Y, Berman-Booty LD, Orr C, Kesler CT, Berger T, Gioeli D, Paschal BM, et al.. Preventing the Androgen Receptor N/C Interaction Delays Disease Onset in a Mouse Model of SBMA. Cell Rep 2015; 13:2312-23; PMID:26673324;
    1. Chua JP, Reddy SL, Yu Z, Giorgetti E, Montie HL, Mukherjee S, Higgins J, McEachin RC, Robins DM, Merry DE, et al.. Disrupting SUMOylation enhances transcriptional function and ameliorates polyglutamine androgen receptor-mediated disease. J Clin Invest 2015; 125:831-45; PMID:25607844;
    1. Palazzolo I, Nedelsky NB, Askew CE, Harmison GG, Kasantsev AG, Taylor JP, Fischbeck KH, Pennuto M. B2 attenuates polyglutamine-expanded androgen receptor toxicity in cell and fly models of spinal and bulbar muscular atrophy. J Neurosci Res 2010; 88:2207-16; PMID:20336775;
    1. Rusmini P, Sau D, Crippa V, Palazzolo I, Simonini F, Onesto E, Martini L, Poletti A. Aggregation and proteasome: the case of elongated polyglutamine aggregation in spinal and bulbar muscular atrophy. Neurobiol Aging 2007; 28:1099-111; PMID:16781019;
    1. Rusmini P, Bolzoni E, Crippa V, Onesto E, Sau D, Galbiati M, Piccolella M, Poletti A. Proteasomal and autophagic degradative activities in spinal and bulbar muscular atrophy. Neurobiol Dis 2010; 40:361-9; PMID:20621188;
    1. Dossena M, Bedini G, Rusmini P, Giorgetti E, Canazza A, Tosetti V, Salsano E, Sagnelli A, Mariotti C, Gellera C, et al.. Human adipose-derived mesenchymal stem cells as a new model of spinal and bulbar muscular atrophy. PLoS One 2014; 9:e112746; PMID:25392924;
    1. Walcott JL, Merry DE. Ligand promotes intranuclear inclusions in a novel cell model of spinal and bulbar muscular atrophy. J Biol Chem 2002; 277:50855-9; PMID:12388541;
    1. Montie HL, Merry DE. Autophagy and access: understanding the role of androgen receptor subcellular localization in SBMA. Autophagy 2009; 5:1194-7; PMID:19770590;
    1. Fischbeck KH. Kennedy disease. J Inher Metab Dis 1997; 20:152-8; PMID:9211187;
    1. Rhodes LE, Freeman BK, Auh S, Kokkinis AD, La Pean A, Chen C, Lehky TJ, Shrader JA, Levy EW, Harris-Love M, et al.. Clinical features of spinal and bulbar muscular atrophy. Brain 2009; 132:3242-51; PMID:19846582;
    1. Grunseich C, Rinaldi C, Fischbeck KH. Spinal and bulbar muscular atrophy: pathogenesis and clinical management. Oral Dis 2014; 20:6-9; PMID:23656576;
    1. Bence NF, Sampat RM, Kopito RR. Impairment of the ubiquitin-proteasome system by protein aggregation. Science 2001; 292:1552-5; PMID:11375494;
    1. Sau D, De Biasi S, Vitellaro-Zuccarello L, Riso P, Guarnieri S, Porrini M, Simeoni S, Crippa V, Onesto E, Palazzolo I, et al.. Mutation of SOD1 in ALS: a gain of a loss of function. Hum Mol Genet 2007; 16:1604-18; PMID:17504823;
    1. Carra S, Rusmini P, Crippa V, Giorgetti E, Boncoraglio A, Cristofani R, Naujock M, Meister M, Minoia M, Kampinga HH, et al.. Different anti-aggregation and pro-degradative functions of the members of the mammalian sHSP family in neurological disorders. Philos Trans R Soc Lond B Biol Sci 2013; 368:20110409; PMID:23530259;
    1. Luders J, Demand J, Hohfeld J. The ubiquitin-related BAG-1 provides a link between the molecular chaperones Hsc70/Hsp70 and the proteasome. J Biol Chem 2000; 275:4613-7; PMID:10671488;
    1. Grunseich C, Zukosky K, Kats IR, Ghosh L, Harmison GG, Bott LC, Rinaldi C, Chen KL, Chen G, Boehm M, et al.. Stem cell-derived motor neurons from spinal and bulbar muscular atrophy patients. Neurobiol Dis 2014; 70:12-20; PMID:24925468;
    1. Bendotti C, Marino M, Cheroni C, Fontana E, Crippa V, Poletti A, De Biasi S. Dysfunction of constitutive and inducible ubiquitin-proteasome system in amyotrophic lateral sclerosis: implication for protein aggregation and immune response. Prog Neurobiol 2012; 97:101-26; PMID:22033150;
    1. Al-Sarraj S, King A, Troakes C, Smith B, Maekawa S, Bodi I, Rogelj B, Al-Chalabi A, Hortobagyi T, Shaw CE. p62 positive, TDP-43 negative, neuronal cytoplasmic and intranuclear inclusions in the cerebellum and hippocampus define the pathology of C9orf72-linked FTLD and MND/ALS. Acta Neuropathol 2011; 122:691-702; PMID:22101323;
    1. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ, Nicholson AM, Finch NA, Flynn H, Adamson J, et al.. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 2011; 72:245-56; PMID:21944778;
    1. Renton AE, Majounie E, Waite A, Simon-Sanchez J, Rollinson S, Gibbs JR, Schymick JC, Laaksovirta H, van Swieten JC, Myllykangas L, et al.. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 2011; 72:257-68; PMID:21944779;
    1. Brettschneider J, Van Deerlin VM, Robinson JL, Kwong L, Lee EB, Ali YO, Safren N, Monteiro MJ, Toledo JB, Elman L, et al.. Pattern of ubiquilin pathology in ALS and FTLD indicates presence of C9ORF72 hexanucleotide expansion. Acta Neuropathol 2012; 123:825-39; PMID:22426854;
    1. Ash PE, Bieniek KF, Gendron TF, Caulfield T, Lin WL, Dejesus-Hernandez M, van Blitterswijk MM, Jansen-West K, Paul JW 3rd, Rademakers R, et al.. Unconventional translation of C9ORF72 GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS. Neuron 2013; 77:639-46; PMID:23415312;
    1. Cleary JD, Ranum LP. Repeat-associated non-ATG (RAN) translation in neurological disease. Hum Mol Genet 2013; 22(R1):R45-51; PMID:23918658;
    1. Lashley T, Hardy J, Isaacs AM. RANTing about C9orf72. Neuron 2013; 77:597-8; PMID:23439112;
    1. Yamakawa M, Ito D, Honda T, Kubo K, Noda M, Nakajima K, Suzuki N. Characterization of the dipeptide repeat protein in the molecular pathogenesis of c9FTD/ALS. Hum Mol Genet 2015; 24:1630-45; PMID:25398948;
    1. Alexandrova A, Petrov L, Georgieva A, Kirkova M, Kukan M. Effects of proteasome inhibitor, MG132, on proteasome activity and oxidative status of rat liver. Cell Biochem Funct 2008; 26:392-8; PMID:18236383;
    1. Onesto E, Rusmini P, Crippa V, Ferri N, Zito A, Galbiati M, Poletti A. Muscle cells and motoneurons differentially remove mutant SOD1 causing familial amyotrophic lateral sclerosis. J Neurochem 2011; 118:266-80; PMID:21554318;
    1. Rohde G, Kermer P, Reed JC, Bahr M, Weishaupt JH. Neuron-specific overexpression of the co-chaperone Bcl-2-associated athanogene-1 in superoxide dismutase 1(G93A)-transgenic mice. Neuroscience 2008; 157:844-9; PMID:18955116;
    1. Gamerdinger M, Hajieva P, Kaya AM, Wolfrum U, Hartl FU, Behl C. Protein quality control during aging involves recruitment of the macroautophagy pathway by BAG3. EMBO J 2009; 28:889-901; PMID:19229298;
    1. Demand J, Alberti S, Patterson C, Hohfeld J. Cooperation of a ubiquitin domain protein and an E3 ubiquitin ligase during chaperone/proteasome coupling. Curr Biol 2001; 11:1569-77; PMID:11676916;
    1. Alberti S, Demand J, Esser C, Emmerich N, Schild H, Hohfeld J. Ubiquitylation of BAG-1 suggests a novel regulatory mechanism during the sorting of chaperone substrates to the proteasome. J Biol Chem 2002; 277:45920-7; PMID:12297498;
    1. Bejarano E, Cuervo AM. Chaperone-mediated autophagy. Proc Am Thorac Soc 2010; 7:29-39; PMID:20160146;
    1. Takayama S, Ostuni E, LeDuc P, Naruse K, Ingber DE, Whitesides GM. Subcellular positioning of small molecules. Nature 2001; 411:1016; PMID:11429594;
    1. Tsukahara F, Maru Y. Bag1 directly routes immature BCR-ABL for proteasomal degradation. Blood 2010; 116:3582-92; PMID:20675402;
    1. Sroka K, Voigt A, Deeg S, Reed JC, Schulz JB, Bahr M, Kermer P. BAG1 modulates huntingtin toxicity, aggregation, degradation, and subcellular distribution. J Neurochem 2009; 111:801-7; PMID:19712056;
    1. Johnston JA, Ward CL, Kopito RR. Aggresomes: a cellular response to misfolded proteins. J Cell Biol 1998; 143:1883-98; PMID:9864362;
    1. Lamark T, Johansen T. Aggrephagy: selective disposal of protein aggregates by macroautophagy. Int J Cell Biol 2012; 2012:736905; PMID:22518139;
    1. Webb JL, Ravikumar B, Rubinsztein DC. Microtubule disruption inhibits autophagosome-lysosome fusion: implications for studying the roles of aggresomes in polyglutamine diseases. Int J Biochem Cell Biol 2004; 36:2541-50; PMID:15325591;
    1. Kopito RR. Aggresomes, inclusion bodies and protein aggregation. Trends Cell Biol 2000; 10:524-30; PMID:11121744;
    1. Kopito RR, Ron D. Conformational disease. Nat Cell Biol 2000; 2:E207-9; PMID:11056553;
    1. Kettern N, Rogon C, Limmer A, Schild H, Hohfeld J. The Hsc/Hsp70 co-chaperone network controls antigen aggregation and presentation during maturation of professional antigen presenting cells. PLoS One 2011; 6:e16398; PMID:21283720;
    1. Puls I, Jonnakuty C, LaMonte BH, Mann E, Floeter MK, Bidus K, Drayna D, Oh SJ, Brown RH Jr, Ludlow CL, et al.. Mutant dynactin in motor neuron disease. Nat Genet 2003; 33:455-6;
    1. Levy JR, Sumner CJ, Caviston JP, Tokito MK, Ranganathan S, Ligon LA, Wallace KE, LaMonte BH, Harmison GG, Puls I, et al.. A motor neuron disease-associated mutation in p150Glued perturbs dynactin function and induces protein aggregation. J Cell Biol 2006; 172:733-45; PMID:16505168;
    1. Kuzma-Kozakiewicz M, Usarek E, Ludolph AC, Baranczyk-Kuzma A. Mice with mutation in dynein heavy chain 1 do not share the same tau expression pattern with mice with SOD1-related motor neuron disease. Neurochem Res 2011; 36:978-85; PMID:21380844;
    1. Schiavo G, Greensmith L, Hafezparast M, Fisher EM. Cytoplasmic dynein heavy chain: the servant of many masters. Trends Neurosci 2013; 36:641-51; PMID:24035135;
    1. Kieran D, Hafezparast M, Bohnert S, Dick JR, Martin J, Schiavo G, Fisher EM, Greensmith L. A mutation in dynein rescues axonal transport defects and extends the life span of ALS mice. J Cell Biol 2005; 169:561-7; PMID:15911875;
    1. Teuling E, van Dis V, Wulf PS, Haasdijk ED, Akhmanova A, Hoogenraad CC, Jaarsma D. A novel mouse model with impaired dynein/dynactin function develops amyotrophic lateral sclerosis (ALS)-like features in motor neurons and improves lifespan in SOD1-ALS mice. Hum Mol Genet 2008; 17:2849-62; PMID:18579581;
    1. Nollen EA, Brunsting JF, Song J, Kampinga HH, Morimoto RI. Bag1 functions in vivo as a negative regulator of Hsp70 chaperone activity. Mol Cell Biol 2000; 20:1083-8; PMID:10629065;
    1. Tortarolo M, Crossthwaite AJ, Conforti L, Spencer JP, Williams RJ, Bendotti C, Rattray M. Expression of SOD1 G93A or wild-type SOD1 in primary cultures of astrocytes down-regulates the glutamate transporter GLT-1: lack of involvement of oxidative stress. J Neurochem 2004; 88:481-93; PMID:14690536;
    1. Ayala YM, Misteli T, Baralle FE. TDP-43 regulates retinoblastoma protein phosphorylation through the repression of cyclin-dependent kinase 6 expression. Proc Natl Acad Sci U S A 2008; 105:3785-9; PMID:18305152;
    1. Ayala YM, Zago P, D'Ambrogio A, Xu YF, Petrucelli L, Buratti E, Baralle FE. Structural determinants of the cellular localization and shuttling of TDP-43. J Cell Sci 2008; 121:3778-85; PMID:18957508;
    1. Cashman NR, Durham HD, Blusztajn JK, Oda K, Tabira T, Shaw IT, Dahrouge S, Antel JP. Neuroblastoma x spinal cord (NSC) hybrid cell lines resemble developing motor neurons. Dev Dyn 1992; 194:209-21; PMID:1467557;
    1. Durham HD, Dahrouge S, Cashman NR. Evaluation of the spinal cord neuron X neuroblastoma hybrid cell line NSC-34 as a model for neurotoxicity testing. Neurotoxicol 1992; 14:387-95.
    1. Marron TU, Guerini V, Rusmini P, Sau D, Brevini TAL, Martini L, Poletti A. Androgen-induced neurite outgrowth is mediated by neuritin in motor neurones. J Neurochem 2005; 92:10-20; PMID:15606892;
    1. Locatelli D, Terao M, Fratelli M, Zanetti A, Kurosaki M, Lupi M, Barzago MM, Uggetti A, Capra S, D'Errico P, et al.. Human axonal survival of motor neuron (a-SMN) protein stimulates axon growth, cell motility, C-C motif ligand 2 (CCL2), and insulin-like growth factor-1 (IGF1) production. J Biol Chem 2012; 287:25782-94; PMID:22669976;
    1. Ferri A, Cozzolino M, Crosio C, Nencini M, Casciati A, Gralla EB, Rotilio G, Valentine JS, Carri MT. Familial ALS-superoxide dismutases associate with mitochondria and shift their redox potentials. Proc Natl Acad Sci U S A 2006; 103:13860-5; PMID:16945901;
    1. Poletti A, Rampoldi A, Piccioni F, Volpi S, Simeoni S, Zanisi M, Martini L. 5Alpha-reductase type 2 and androgen receptor expression in gonadotropin releasing hormone GT1-1 cells. J Neuroendocrinol 2001; 13:353-7; PMID:11264723;
    1. Pozzi P, Bendotti C, Simeoni S, Piccioni F, Guerini V, Marron TU, Martini L, Poletti A. Androgen 5-alpha-reductase type 2 is highly expressed and active in rat spinal cord motor neurones. J Neuroendocrinol 2003; 15:882-7; PMID:12899683;
    1. Rusmini P, Polanco MJ, Cristofani R, Cicardi ME, Meroni M, Galbiati M, Piccolella M, Messi E, Giorgetti E, Lieberman AP, et al.. Aberrant autophagic response in the muscle of A knock-in mouse model of spinal and bulbar muscular atrophy. Sci Rep 2015; 5:15174; PMID:26490709;
    1. Crippa V, Boncoraglio A, Galbiati M, Aggarwal T, Rusmini P, Giorgetti E, Cristofani R, Carra S, Pennuto M, Poletti A. Differential autophagy power in the spinal cord and muscle of transgenic ALS mice. Front Cell Neurosci 2013; 7:234; PMID:24324403;
    1. Koyama S, Arawaka S, Chang-Hong R, Wada M, Kawanami T, Kurita K, Kato M, Nagai M, Aoki M, Itoyama Y, et al.. Alteration of familial ALS-linked mutant SOD1 solubility with disease progression: its modulation by the proteasome and Hsp70. Biochem Biophys Res Commun 2006; 343:719-30; PMID:16563356;
    1. Allen S, Heath PR, Kirby J, Wharton SB, Cookson MR, Menzies FM, Banks RE, Shaw PJ. Analysis of the cytosolic proteome in a cell culture model of familial amyotrophic lateral sclerosis reveals alterations to the proteasome, antioxidant defenses, and nitric oxide synthetic pathways. J Biol Chem 2003; 278:6371-83; PMID:12475980;

Source: PubMed

3
Prenumerera