Early short-term PXT3003 combinational therapy delays disease onset in a transgenic rat model of Charcot-Marie-Tooth disease 1A (CMT1A)

Thomas Prukop, Jan Stenzel, Stephanie Wernick, Theresa Kungl, Magdalena Mroczek, Julia Adam, David Ewers, Serguei Nabirotchkin, Klaus-Armin Nave, Rodolphe Hajj, Daniel Cohen, Michael W Sereda, Thomas Prukop, Jan Stenzel, Stephanie Wernick, Theresa Kungl, Magdalena Mroczek, Julia Adam, David Ewers, Serguei Nabirotchkin, Klaus-Armin Nave, Rodolphe Hajj, Daniel Cohen, Michael W Sereda

Abstract

The most common type of Charcot-Marie-Tooth disease is caused by a duplication of PMP22 leading to dysmyelination, axonal loss and progressive muscle weakness (CMT1A). Currently, no approved therapy is available for CMT1A patients. A novel polytherapeutic proof-of-principle approach using PXT3003, a low-dose combination of baclofen, naltrexone and sorbitol, slowed disease progression after long-term dosing in adult Pmp22 transgenic rats, a known animal model of CMT1A. Here, we report an early postnatal, short-term treatment with PXT3003 in CMT1A rats that delays disease onset into adulthood. CMT1A rats were treated from postnatal day 6 to 18 with PXT3003. Behavioural, electrophysiological, histological and molecular analyses were performed until 12 weeks of age. Daily oral treatment for approximately 2 weeks ameliorated motor deficits of CMT1A rats reaching wildtype levels. Histologically, PXT3003 corrected the disturbed axon calibre distribution with a shift towards large motor axons. Despite dramatic clinical amelioration, only distal motor latencies were improved and correlated with phenotype performance. On the molecular level, PXT3003 reduced Pmp22 mRNA overexpression and improved the misbalanced downstream PI3K-AKT / MEK-ERK signalling pathway. The improved differentiation status of Schwann cells may have enabled better long-term axonal support function. We conclude that short-term treatment with PXT3003 during early development may partially prevent the clinical and molecular manifestations of CMT1A. Since PXT3003 has a strong safety profile and is currently undergoing a phase III trial in CMT1A patients, our results suggest that PXT3003 therapy may be a bona fide translatable therapy option for children and young adolescent patients suffering from CMT1A.

Conflict of interest statement

We have the following interests. This trial was financially supported by Pharnext, the employer of Serguei Nabirotchkin, Rodolphe Hajj and Daniel Cohen. DC, RH, MWS, TP and KAN submitted a patent based on this work: Full patent name: Early treatment of CMT disease Number: US2018/0000813. MWS, TP and KAN act as consultants to Pharnext. PXT3003 achieved a Phase 3 trial in CMT1A adult patients. There are no further patents related to this study, or marketed products to declare. This does not alter our adherence to all the PLOS ONE policies on sharing data and materials, as detailed online in the guide for authors.

Figures

Fig 1. Long-lasting restoration of phenotype deficits…
Fig 1. Long-lasting restoration of phenotype deficits after early short-term PXT3003 therapy.
Early short-term therapy regimen followed by a long-term observation of motor and sensory, histological and molecular effects (A). Early postnatally PXT3003 application dose-dependently improved the grip strength impairment at the forelimbs after the treatment phase in 3 weeks aged CMT1A rats (B; WT controls 3.66±0.09, CMT1A controls 2.64±0.06, CMT PXT3003-3 2.93±0.10). First grip strength improvement was observed at the hindlimbs in 9 weeks aged CMT1A rats (C; WT controls 3.64±0.16, CMT1A controls 3.06±0.12, CMT PXT3003-3 3.47±0.12) lasting until 12 weeks (D; WT controls 5.38±0.23, CMT1A controls 4.16±0.22, CMT PXT3003-3 5.10±0.19). PXT3003 long-term effects increased over time reaching wildtype levels in the long-term observation (E). PXT3003 early short-term therapy improved motor deficits on the inclined plane in 3 (F; WT controls 0.10±0.07, CMT1A controls 1.00±0.18, CMT PXT3003-3 0.48±0.13) and 12 weeks aged CMT1A rats (G; WT controls 0.07±0.04, CMT1A controls 0.33±0.11, CMT PXT3003-3 0.07±0.04). Although some training effects were observed in the inclined plane behaviour in CMT1A controls, PXT3003 effects increased over time and reached wildtype levels at the study end (H). (ns = not significant, * = p

Fig 2. Improved motor latency correlating with…

Fig 2. Improved motor latency correlating with the phenotype at study end.

Electrophysiological recordings including…

Fig 2. Improved motor latency correlating with the phenotype at study end.
Electrophysiological recordings including motor latency (red bar), nerve conduction velocity (NCV) and compound muscle potential (CMAP) analysis were performed as shown for representative measures (A). CMT1A controls showed prolonged motor latencies, which were dose-dependently improved by PXT3003 early short-term treatment (B; WT controls 1.66±0.10, CMT1A controls 3.35±0.22, CMT PXT3003-3 2.82±0.13). In long-term observation, the motor latency improvement correlated with the restored grip strength at the hindlimbs including all PXT3003 treated CMT1A rats (C). NCV and CMAP recordings were not affected by PXT3003 in long-term (D and F; WT controls 37.02±2.16, CMT1A controls 14.02±0,67, CMT PXT3003-3 14.86±0.53 and WT controls 5.31±0.47, CMT1A controls 1.49±0.25, CMT PXT3003-3 1.18±0.16, respectively), and did not correlate with the hindlimb grip strength of all PXT3003-treated CMT1A rats (E and G). (ns = not significant, * = p

Fig 3. Shift towards large-calibre axons in…

Fig 3. Shift towards large-calibre axons in the long-term observation.

Histological analyses were performed on…

Fig 3. Shift towards large-calibre axons in the long-term observation.
Histological analyses were performed on light microscopic level in the sciatic nerve as demonstrated in representative peripheral nerve cross sections. In contrast to WT controls, untreated CMT1A rats showed a loss of large-calibre axons (orange arrow) combined with a hypomyelination of predominantly large axons (green arrow) and a hypermyelination of predominantly small-calibre axons (blue arrow). A higher number of large-calibre axons appeared after PXT3003 treatment in CMT1A rats, without obvious differences between hypo- and hypermyelinated axons (A). Quantification of myelinated axons confirmed axonal loss in CMT1A controls not being affected by PXT3003 treatment (B; WT controls 9003±114, CMT1A controls 8654±74, CMT PXT3003-3 8727±68); however PXT3003 partially corrected the reduced mean axon diameter (C; WT controls 4.96±0.19, CMT1A controls 3.66±0.09, CMT PXT3003-2 3.97±0.08). More detailed analyses of axon calibre distribution confirmed an obvious loss of large-calibre axons in CMT1A rats, and a shift towards large-calibre axons after PXT3003 treatment (D). Mid- and large-calibre axons were corrected towards the wildtype situation as illustrated in more detail for 3–4μm and 5–6μm axons (E and F; WT controls 11.33±1.48, CMT1A controls 26.50±1.08, CMT PXT3003-3 22.44±1.43 and WT controls 21.83±1.54, CMT1A controls 11.11±1.12, CMT PXT3003-2 15.62±1.13, respectively). (ns = not significant, * = p

Fig 4. PXT3003 therapy acting on Pmp22…

Fig 4. PXT3003 therapy acting on Pmp22 overexpression and downstream signalling in long-term at 12…

Fig 4. PXT3003 therapy acting on Pmp22 overexpression and downstream signalling in long-term at 12 weeks.
Early short-term applied PXT3003 was in long-term effective on the downregulation of disease-causing Pmp22 mRNA overexpression in CMT1A rats in the sciatic nerve (A; WT controls 1.00±0.04, CMT1A controls 1.48±0.07, CMT PXT3003-3 1.33±0.04; Pmp22 splice variant normalised to Cyclophilin A). In the brachial plexus, mRNA expression of the differentiation marker Sox2 (B; WT controls 1.00±0.09, CMT1A controls 3.68±0.55, CMT PXT3003-3 2.22±0.28; normalised to Cyclophilin A and Rplp0) and cJun (C; WT controls 1.00±0.09, CMT1A controls 2.18±0.34, CMT PXT3003-3 1.14±0.10; normalised to Cyclophilin A and Rplp0), as well as the biomarker Ctsa were influenced by PXT3003 in long-term observation (D; WT controls 1.00±0.08, CMT1A controls 1.28±0.16, CMT PXT3003-3 0.66±0.06; normalised to Cyclophilin A and Rplp0). Downregulated Hmgcr and Prx mRNA expression in CMT1A rats were not affected by PXT3003 (E; WT controls 1.00±0.08, CMT1A controls 0.40±0.03, CMT PXT3003-3 0.35±0.05 and F; WT controls 1.00±0.07, CMT1A controls 0.67±0.05, CMT PXT3003-3 0.63±0.08; normalised to Cyclophilin A and Rplp0). Pmp22 downstream signalling was investigated by Western Blot analysis using antibodies against p-AKT/AKT and p-MAPK/MAPK (G). At the end of long-term observation, PXT3003 early short-term treatment restored the decreased p-AKT/AKT signalling in CMT1A rats (H; WT controls 1.00±0.17, CMT1A controls 0.38±0.08, CMT PXT3003-3 0.93±0.21) and dose-dependently corrected the disrupted ratio of p-AKT/AKT to p-MAPK/MAPK signalling towards the wildtype situation (I; CMT1A controls 0.30±0.08, CMT PXT3003-1 0.56±0.15, CMT PXT3003-2 0.85±0.13, CMT PXT3003-3 1.36±0.52), which is characterized by a higher relative amount of p-AKT versus p-MAPK. (ns = not significant, * = p<0.05, **p<0.01 and *** = p<0.001).
Similar articles
Cited by
References
    1. Skre H. Genetic and clinical aspects of Charcot-Marie-Tooth’s disease. Clin Genet. 1974; 6: 98–118. - PubMed
    1. Braathen GJ, Sand JC, Lobato A, Høyer H, Russell MB. Genetic epidemiology of Charcot-Marie-Tooth in the general population. Eur J Neurol. 2011; 18: 39–48. 10.1111/j.1468-1331.2010.03037.x - DOI - PubMed
    1. Timmerman V, Strickland AV, Züchner S. Genetics of Charcot-Marie-Tooth (CMT) Disease within the Frame of the Human Genome Project Success. Genes (Basel). 2014; 5: 13–32. 10.3390/genes5010013 - DOI - PMC - PubMed
    1. Rossor AM, Polke JM, Houlden H, Reilly MM. Clinical implications of genetic advances in Charcot-Marie-Tooth disease. Nat Rev Neurol. 2013; 9: 562–571. 10.1038/nrneurol.2013.179 - DOI - PubMed
    1. Rudnik-Schöneborn S, Tölle D, Senderek J, Eggermann K, Elbracht M, Kornak U, et al. Diagnostic algorithms in Charcot-Marie-Tooth neuropathies. Experiences from a German genetic laboratory on the basis of 1206 index patients. Clin Genet. 2016; 89: 34–43. 10.1111/cge.12594 - DOI - PubMed
Show all 40 references
Publication types
MeSH terms
Grant support
MWS was supported by the German Ministry of Education and Research (BMBF, CMT-BIO, FKZ: 01ES0812, CMT-NET, FKZ: 01GM1511C, CMT-NRG, ERA-NET ’ERARE3’, FKZ: 01GM1605). MWS holds a DFG Heisenberg Professorship (SE 1944/1-1). TP was supported by the European Leukodystrophie Society (ELA 2014-020I1 to MWS). KAN is supported by the DFG (SPP1757 and CNMPB) and holds an ERC Advanced Grant. This trial was financially supported by Pharnext who provided support in the form of salaries for authors SN, RH and DC, but did not have any additional role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript. The specific roles of these authors are articulated in the ‘author contributions’ section.
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig 2. Improved motor latency correlating with…
Fig 2. Improved motor latency correlating with the phenotype at study end.
Electrophysiological recordings including motor latency (red bar), nerve conduction velocity (NCV) and compound muscle potential (CMAP) analysis were performed as shown for representative measures (A). CMT1A controls showed prolonged motor latencies, which were dose-dependently improved by PXT3003 early short-term treatment (B; WT controls 1.66±0.10, CMT1A controls 3.35±0.22, CMT PXT3003-3 2.82±0.13). In long-term observation, the motor latency improvement correlated with the restored grip strength at the hindlimbs including all PXT3003 treated CMT1A rats (C). NCV and CMAP recordings were not affected by PXT3003 in long-term (D and F; WT controls 37.02±2.16, CMT1A controls 14.02±0,67, CMT PXT3003-3 14.86±0.53 and WT controls 5.31±0.47, CMT1A controls 1.49±0.25, CMT PXT3003-3 1.18±0.16, respectively), and did not correlate with the hindlimb grip strength of all PXT3003-treated CMT1A rats (E and G). (ns = not significant, * = p

Fig 3. Shift towards large-calibre axons in…

Fig 3. Shift towards large-calibre axons in the long-term observation.

Histological analyses were performed on…

Fig 3. Shift towards large-calibre axons in the long-term observation.
Histological analyses were performed on light microscopic level in the sciatic nerve as demonstrated in representative peripheral nerve cross sections. In contrast to WT controls, untreated CMT1A rats showed a loss of large-calibre axons (orange arrow) combined with a hypomyelination of predominantly large axons (green arrow) and a hypermyelination of predominantly small-calibre axons (blue arrow). A higher number of large-calibre axons appeared after PXT3003 treatment in CMT1A rats, without obvious differences between hypo- and hypermyelinated axons (A). Quantification of myelinated axons confirmed axonal loss in CMT1A controls not being affected by PXT3003 treatment (B; WT controls 9003±114, CMT1A controls 8654±74, CMT PXT3003-3 8727±68); however PXT3003 partially corrected the reduced mean axon diameter (C; WT controls 4.96±0.19, CMT1A controls 3.66±0.09, CMT PXT3003-2 3.97±0.08). More detailed analyses of axon calibre distribution confirmed an obvious loss of large-calibre axons in CMT1A rats, and a shift towards large-calibre axons after PXT3003 treatment (D). Mid- and large-calibre axons were corrected towards the wildtype situation as illustrated in more detail for 3–4μm and 5–6μm axons (E and F; WT controls 11.33±1.48, CMT1A controls 26.50±1.08, CMT PXT3003-3 22.44±1.43 and WT controls 21.83±1.54, CMT1A controls 11.11±1.12, CMT PXT3003-2 15.62±1.13, respectively). (ns = not significant, * = p

Fig 4. PXT3003 therapy acting on Pmp22…

Fig 4. PXT3003 therapy acting on Pmp22 overexpression and downstream signalling in long-term at 12…

Fig 4. PXT3003 therapy acting on Pmp22 overexpression and downstream signalling in long-term at 12 weeks.
Early short-term applied PXT3003 was in long-term effective on the downregulation of disease-causing Pmp22 mRNA overexpression in CMT1A rats in the sciatic nerve (A; WT controls 1.00±0.04, CMT1A controls 1.48±0.07, CMT PXT3003-3 1.33±0.04; Pmp22 splice variant normalised to Cyclophilin A). In the brachial plexus, mRNA expression of the differentiation marker Sox2 (B; WT controls 1.00±0.09, CMT1A controls 3.68±0.55, CMT PXT3003-3 2.22±0.28; normalised to Cyclophilin A and Rplp0) and cJun (C; WT controls 1.00±0.09, CMT1A controls 2.18±0.34, CMT PXT3003-3 1.14±0.10; normalised to Cyclophilin A and Rplp0), as well as the biomarker Ctsa were influenced by PXT3003 in long-term observation (D; WT controls 1.00±0.08, CMT1A controls 1.28±0.16, CMT PXT3003-3 0.66±0.06; normalised to Cyclophilin A and Rplp0). Downregulated Hmgcr and Prx mRNA expression in CMT1A rats were not affected by PXT3003 (E; WT controls 1.00±0.08, CMT1A controls 0.40±0.03, CMT PXT3003-3 0.35±0.05 and F; WT controls 1.00±0.07, CMT1A controls 0.67±0.05, CMT PXT3003-3 0.63±0.08; normalised to Cyclophilin A and Rplp0). Pmp22 downstream signalling was investigated by Western Blot analysis using antibodies against p-AKT/AKT and p-MAPK/MAPK (G). At the end of long-term observation, PXT3003 early short-term treatment restored the decreased p-AKT/AKT signalling in CMT1A rats (H; WT controls 1.00±0.17, CMT1A controls 0.38±0.08, CMT PXT3003-3 0.93±0.21) and dose-dependently corrected the disrupted ratio of p-AKT/AKT to p-MAPK/MAPK signalling towards the wildtype situation (I; CMT1A controls 0.30±0.08, CMT PXT3003-1 0.56±0.15, CMT PXT3003-2 0.85±0.13, CMT PXT3003-3 1.36±0.52), which is characterized by a higher relative amount of p-AKT versus p-MAPK. (ns = not significant, * = p<0.05, **p<0.01 and *** = p<0.001).
Similar articles
Cited by
References
    1. Skre H. Genetic and clinical aspects of Charcot-Marie-Tooth’s disease. Clin Genet. 1974; 6: 98–118. - PubMed
    1. Braathen GJ, Sand JC, Lobato A, Høyer H, Russell MB. Genetic epidemiology of Charcot-Marie-Tooth in the general population. Eur J Neurol. 2011; 18: 39–48. 10.1111/j.1468-1331.2010.03037.x - DOI - PubMed
    1. Timmerman V, Strickland AV, Züchner S. Genetics of Charcot-Marie-Tooth (CMT) Disease within the Frame of the Human Genome Project Success. Genes (Basel). 2014; 5: 13–32. 10.3390/genes5010013 - DOI - PMC - PubMed
    1. Rossor AM, Polke JM, Houlden H, Reilly MM. Clinical implications of genetic advances in Charcot-Marie-Tooth disease. Nat Rev Neurol. 2013; 9: 562–571. 10.1038/nrneurol.2013.179 - DOI - PubMed
    1. Rudnik-Schöneborn S, Tölle D, Senderek J, Eggermann K, Elbracht M, Kornak U, et al. Diagnostic algorithms in Charcot-Marie-Tooth neuropathies. Experiences from a German genetic laboratory on the basis of 1206 index patients. Clin Genet. 2016; 89: 34–43. 10.1111/cge.12594 - DOI - PubMed
Show all 40 references
Publication types
MeSH terms
Grant support
MWS was supported by the German Ministry of Education and Research (BMBF, CMT-BIO, FKZ: 01ES0812, CMT-NET, FKZ: 01GM1511C, CMT-NRG, ERA-NET ’ERARE3’, FKZ: 01GM1605). MWS holds a DFG Heisenberg Professorship (SE 1944/1-1). TP was supported by the European Leukodystrophie Society (ELA 2014-020I1 to MWS). KAN is supported by the DFG (SPP1757 and CNMPB) and holds an ERC Advanced Grant. This trial was financially supported by Pharnext who provided support in the form of salaries for authors SN, RH and DC, but did not have any additional role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript. The specific roles of these authors are articulated in the ‘author contributions’ section.
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Fig 3. Shift towards large-calibre axons in…
Fig 3. Shift towards large-calibre axons in the long-term observation.
Histological analyses were performed on light microscopic level in the sciatic nerve as demonstrated in representative peripheral nerve cross sections. In contrast to WT controls, untreated CMT1A rats showed a loss of large-calibre axons (orange arrow) combined with a hypomyelination of predominantly large axons (green arrow) and a hypermyelination of predominantly small-calibre axons (blue arrow). A higher number of large-calibre axons appeared after PXT3003 treatment in CMT1A rats, without obvious differences between hypo- and hypermyelinated axons (A). Quantification of myelinated axons confirmed axonal loss in CMT1A controls not being affected by PXT3003 treatment (B; WT controls 9003±114, CMT1A controls 8654±74, CMT PXT3003-3 8727±68); however PXT3003 partially corrected the reduced mean axon diameter (C; WT controls 4.96±0.19, CMT1A controls 3.66±0.09, CMT PXT3003-2 3.97±0.08). More detailed analyses of axon calibre distribution confirmed an obvious loss of large-calibre axons in CMT1A rats, and a shift towards large-calibre axons after PXT3003 treatment (D). Mid- and large-calibre axons were corrected towards the wildtype situation as illustrated in more detail for 3–4μm and 5–6μm axons (E and F; WT controls 11.33±1.48, CMT1A controls 26.50±1.08, CMT PXT3003-3 22.44±1.43 and WT controls 21.83±1.54, CMT1A controls 11.11±1.12, CMT PXT3003-2 15.62±1.13, respectively). (ns = not significant, * = p

Fig 4. PXT3003 therapy acting on Pmp22…

Fig 4. PXT3003 therapy acting on Pmp22 overexpression and downstream signalling in long-term at 12…

Fig 4. PXT3003 therapy acting on Pmp22 overexpression and downstream signalling in long-term at 12 weeks.
Early short-term applied PXT3003 was in long-term effective on the downregulation of disease-causing Pmp22 mRNA overexpression in CMT1A rats in the sciatic nerve (A; WT controls 1.00±0.04, CMT1A controls 1.48±0.07, CMT PXT3003-3 1.33±0.04; Pmp22 splice variant normalised to Cyclophilin A). In the brachial plexus, mRNA expression of the differentiation marker Sox2 (B; WT controls 1.00±0.09, CMT1A controls 3.68±0.55, CMT PXT3003-3 2.22±0.28; normalised to Cyclophilin A and Rplp0) and cJun (C; WT controls 1.00±0.09, CMT1A controls 2.18±0.34, CMT PXT3003-3 1.14±0.10; normalised to Cyclophilin A and Rplp0), as well as the biomarker Ctsa were influenced by PXT3003 in long-term observation (D; WT controls 1.00±0.08, CMT1A controls 1.28±0.16, CMT PXT3003-3 0.66±0.06; normalised to Cyclophilin A and Rplp0). Downregulated Hmgcr and Prx mRNA expression in CMT1A rats were not affected by PXT3003 (E; WT controls 1.00±0.08, CMT1A controls 0.40±0.03, CMT PXT3003-3 0.35±0.05 and F; WT controls 1.00±0.07, CMT1A controls 0.67±0.05, CMT PXT3003-3 0.63±0.08; normalised to Cyclophilin A and Rplp0). Pmp22 downstream signalling was investigated by Western Blot analysis using antibodies against p-AKT/AKT and p-MAPK/MAPK (G). At the end of long-term observation, PXT3003 early short-term treatment restored the decreased p-AKT/AKT signalling in CMT1A rats (H; WT controls 1.00±0.17, CMT1A controls 0.38±0.08, CMT PXT3003-3 0.93±0.21) and dose-dependently corrected the disrupted ratio of p-AKT/AKT to p-MAPK/MAPK signalling towards the wildtype situation (I; CMT1A controls 0.30±0.08, CMT PXT3003-1 0.56±0.15, CMT PXT3003-2 0.85±0.13, CMT PXT3003-3 1.36±0.52), which is characterized by a higher relative amount of p-AKT versus p-MAPK. (ns = not significant, * = p<0.05, **p<0.01 and *** = p<0.001).
Fig 4. PXT3003 therapy acting on Pmp22…
Fig 4. PXT3003 therapy acting on Pmp22 overexpression and downstream signalling in long-term at 12 weeks.
Early short-term applied PXT3003 was in long-term effective on the downregulation of disease-causing Pmp22 mRNA overexpression in CMT1A rats in the sciatic nerve (A; WT controls 1.00±0.04, CMT1A controls 1.48±0.07, CMT PXT3003-3 1.33±0.04; Pmp22 splice variant normalised to Cyclophilin A). In the brachial plexus, mRNA expression of the differentiation marker Sox2 (B; WT controls 1.00±0.09, CMT1A controls 3.68±0.55, CMT PXT3003-3 2.22±0.28; normalised to Cyclophilin A and Rplp0) and cJun (C; WT controls 1.00±0.09, CMT1A controls 2.18±0.34, CMT PXT3003-3 1.14±0.10; normalised to Cyclophilin A and Rplp0), as well as the biomarker Ctsa were influenced by PXT3003 in long-term observation (D; WT controls 1.00±0.08, CMT1A controls 1.28±0.16, CMT PXT3003-3 0.66±0.06; normalised to Cyclophilin A and Rplp0). Downregulated Hmgcr and Prx mRNA expression in CMT1A rats were not affected by PXT3003 (E; WT controls 1.00±0.08, CMT1A controls 0.40±0.03, CMT PXT3003-3 0.35±0.05 and F; WT controls 1.00±0.07, CMT1A controls 0.67±0.05, CMT PXT3003-3 0.63±0.08; normalised to Cyclophilin A and Rplp0). Pmp22 downstream signalling was investigated by Western Blot analysis using antibodies against p-AKT/AKT and p-MAPK/MAPK (G). At the end of long-term observation, PXT3003 early short-term treatment restored the decreased p-AKT/AKT signalling in CMT1A rats (H; WT controls 1.00±0.17, CMT1A controls 0.38±0.08, CMT PXT3003-3 0.93±0.21) and dose-dependently corrected the disrupted ratio of p-AKT/AKT to p-MAPK/MAPK signalling towards the wildtype situation (I; CMT1A controls 0.30±0.08, CMT PXT3003-1 0.56±0.15, CMT PXT3003-2 0.85±0.13, CMT PXT3003-3 1.36±0.52), which is characterized by a higher relative amount of p-AKT versus p-MAPK. (ns = not significant, * = p<0.05, **p<0.01 and *** = p<0.001).

References

    1. Skre H. Genetic and clinical aspects of Charcot-Marie-Tooth’s disease. Clin Genet. 1974; 6: 98–118.
    1. Braathen GJ, Sand JC, Lobato A, Høyer H, Russell MB. Genetic epidemiology of Charcot-Marie-Tooth in the general population. Eur J Neurol. 2011; 18: 39–48. 10.1111/j.1468-1331.2010.03037.x
    1. Timmerman V, Strickland AV, Züchner S. Genetics of Charcot-Marie-Tooth (CMT) Disease within the Frame of the Human Genome Project Success. Genes (Basel). 2014; 5: 13–32. 10.3390/genes5010013
    1. Rossor AM, Polke JM, Houlden H, Reilly MM. Clinical implications of genetic advances in Charcot-Marie-Tooth disease. Nat Rev Neurol. 2013; 9: 562–571. 10.1038/nrneurol.2013.179
    1. Rudnik-Schöneborn S, Tölle D, Senderek J, Eggermann K, Elbracht M, Kornak U, et al. Diagnostic algorithms in Charcot-Marie-Tooth neuropathies. Experiences from a German genetic laboratory on the basis of 1206 index patients. Clin Genet. 2016; 89: 34–43. 10.1111/cge.12594
    1. Murphy SM, Laura M, Fawcett K, Pandraud A, Liu Y-T, Davidson GL, et al. Charcot-Marie-Tooth disease. Frequency of genetic subtypes and guidelines for genetic testing. J Neurol Neurosurg Psychiatry. 2012; 83: 706–710. 10.1136/jnnp-2012-302451
    1. Lupski JR, Garcia CA. Molecular genetics and neuropathology of Charcot-Marie-Tooth disease type 1A. Brain Pathol. 1992; 2: 337–349.
    1. Raeymaekers P, Timmerman V, Nelis E, de Jonghe P, Hoogendijk JE, Baas F, et al. Duplication in chromosome 17p11.2 in Charcot-Marie-Tooth neuropathy type 1a (CMT 1a). The HMSN Collaborative Research Group. Neuromuscul Disord. 1991; 1: 93–97.
    1. Patel PI, Garcia C, Montes de Oca-Luna R, Malamut RI, Franco B, Slaugenhaupt S, et al. Isolation of a marker linked to the Charcot-Marie-Tooth disease type IA gene by differential Alu-PCR of human chromosome 17-retaining hybrids. Am J Hum Genet. 1990; 47: 926–934.
    1. Pareyson D, Saveri P, Piscosquito G. Charcot-Marie-Tooth Disease and Related Hereditary Neuropathies. From Gene Function to Associated Phenotypes. Curr Mol Med. 2014; 14: 1009–1033. 10.2174/1566524014666141010154205
    1. Berciano J, García A, Gallardo E, Ramón C, Combarros O. Phenotype and clinical evolution of Charcot-Marie-Tooth disease type 1A duplication. Adv Exp Med Biol. 2009; 652: 183–200. 10.1007/978-90-481-2813-6_12
    1. Rossor AM, Evans MRB, Reilly MM. A practical approach to the genetic neuropathies. Pract Neurol. 2015; 15: 187–198. 10.1136/practneurol-2015-001095
    1. Saporta ASD, Sottile SL, Miller LJ, Feely SME, Siskind CE, Shy ME. Charcot-Marie-Tooth disease subtypes and genetic testing strategies. Ann Neurol. 2011; 69: 22–33. 10.1002/ana.22166
    1. Fledrich R, Stassart RM, Sereda MW. Murine therapeutic models for Charcot-Marie-Tooth (CMT) disease. Br Med Bull. 2012; 102: 89–113. 10.1093/bmb/lds010
    1. Fledrich R, Schlotter-Weigel B, Schnizer TJ, Wichert SP, Stassart RM, Meyer zu Hörste G, et al. A rat model of Charcot-Marie-Tooth disease 1A recapitulates disease variability and supplies biomarkers of axonal loss in patients. Brain. 2012; 135: 72–87. 10.1093/brain/awr322
    1. Yiu EM, Burns J, Ryan MM, Ouvrier RA. Neurophysiologic abnormalities in children with Charcot-Marie-Tooth disease type 1A. J Peripher Nerv Syst. 2008; 13: 236–241. 10.1111/j.1529-8027.2008.00182.x
    1. Burns J, Raymond J, Ouvrier R. Feasibility of foot and ankle strength training in childhood Charcot-Marie-Tooth disease. Neuromuscul Disord. 2009; 19: 818–821. 10.1016/j.nmd.2009.09.007
    1. Cornett KMD, Menezes MP, Bray P, Halaki M, Shy RR, Yum SW, et al. Phenotypic Variability of Childhood Charcot-Marie-Tooth Disease. JAMA Neurol. 2016; 73: 645–651. 10.1001/jamaneurol.2016.0171
    1. Rossor AM, Tomaselli PJ, Reilly MM. Recent advances in the genetic neuropathies. Curr Opin Neurol. 2016; 29: 537–548. 10.1097/WCO.0000000000000373
    1. Young P, de Jonghe P, Stögbauer F, Butterfass-Bahloul T. Treatment for Charcot-Marie-Tooth disease. Cochrane Database Syst Rev. 2008: CD006052 10.1002/14651858.CD006052.pub2
    1. Micallef J, Attarian S, Dubourg O, Gonnaud P-M, Hogrel J-Y, Stojkovic T, et al. Effect of ascorbic acid in patients with Charcot-Marie-Tooth disease type 1A. A multicentre, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2009; 8: 1103–1110. 10.1016/S1474-4422(09)70260-1
    1. Verhamme C, de Haan RJ, Vermeulen M, Baas F, de Visser M, van Schaik IN. Oral high dose ascorbic acid treatment for one year in young CMT1A patients. A randomised, double-blind, placebo-controlled phase II trial. BMC Med. 2009; 7: 70 10.1186/1741-7015-7-70
    1. Pareyson D, Reilly MM, Schenone A, Fabrizi GM, Cavallaro T, Santoro L, et al. Ascorbic acid in Charcot-Marie-Tooth disease type 1A (CMT-TRIAAL and CMT-TRAUK). A double-blind randomised trial. Lancet Neurol. 2011; 10: 320–328. 10.1016/S1474-4422(11)70025-4
    1. Chumakov I, Milet A, Cholet N, Primas G, Boucard A, Pereira Y, et al. Polytherapy with a combination of three repurposed drugs (PXT3003) down-regulates Pmp22 over-expression and improves myelination, axonal and functional parameters in models of CMT1A neuropathy. Orphanet J Rare Dis. 2014; 9: 201 10.1186/s13023-014-0201-x
    1. Attarian S, Vallat J-M, Magy L, Funalot B, Gonnaud P-M, Lacour A, et al. An exploratory randomised double-blind and placebo-controlled phase 2 study of a combination of baclofen, naltrexone and sorbitol (PXT3003) in patients with Charcot-Marie-Tooth disease type 1A. Orphanet J Rare Dis. 2014; 9: 199 10.1186/s13023-014-0199-0
    1. Attarian S, Vallat J-M, Magy L, Funalot B, Gonnaud P-M, Lacour A, et al. Erratum to. An exploratory randomised double-blind and placebo-controlled phase 2 study of a combination of baclofen, naltrexone and sorbitol (PXT3003) in patients with Charcot-Marie-Tooth disease type 1A. Orphanet J Rare Dis. 2016; 11: 92 10.1186/s13023-016-0463-6
    1. Fledrich R, Stassart RM, Klink A, Rasch LM, Prukop T, Haag L, et al. Soluble neuregulin-1 modulates disease pathogenesis in rodent models of Charcot-Marie-Tooth disease 1A. Nat Med. 2014; 20: 1055–1061. 10.1038/nm.3664
    1. Jessen KR, Mirsky R. The origin and development of glial cells in peripheral nerves. Nat Rev Neurosci. 2005; 6: 671–682. 10.1038/nrn1746
    1. Michailov GV, Sereda MW, Brinkmann BG, Fischer TM, Haug B, Birchmeier C, et al. Axonal neuregulin-1 regulates myelin sheath thickness. Science. 2004; 304: 700–703. 10.1126/science.1095862
    1. Sereda M, Griffiths I, Pühlhofer A, Stewart H, Rossner MJ, Zimmerman F, et al. A transgenic rat model of Charcot-Marie-Tooth disease. Neuron. 1996; 16: 1049–1060.
    1. Rivlin AS, Tator CH. Objective clinical assessment of motor function after experimental spinal cord injury in the rat. J Neurosurg. 1977; 47: 577–581. 10.3171/jns.1977.47.4.0577
    1. Meyer zu Horste G, Prukop T, Liebetanz D, Mobius W, Nave K-A, Sereda MW. Antiprogesterone therapy uncouples axonal loss from demyelination in a transgenic rat model of CMT1A neuropathy. Ann Neurol. 2007; 61: 61–72. 10.1002/ana.21026
    1. Visigalli D, Castagnola P, Capodivento G, Geroldi A, Bellone E, Mancardi G, et al. Alternative Splicing in the Human PMP22 Gene. Implications in CMT1A Neuropathy. Hum Mutat. 2016; 37: 98–109. 10.1002/humu.22921
    1. Sereda MW, Meyer zu Hörste G, Suter U, Uzma N, Nave K-A. Therapeutic administration of progesterone antagonist in a model of Charcot-Marie-Tooth disease (CMT-1A). Nat Med. 2003; 9: 1533–1537. 10.1038/nm957
    1. Zhao HT, Damle S, Ikeda-Lee K, Kuntz S, Li J, Mohan A, et al. PMP22 antisense oligonucleotides reverse Charcot-Marie-Tooth disease type 1A features in rodent models. J Clin Invest. 2017. 10.1172/JCI96499
    1. Shy ME. Antisense oligonucleotides offer hope to patients with Charcot-Marie-Tooth disease type 1A. J Clin Invest. 2017. 10.1172/JCI98617
    1. Sociali G, Visigalli D, Prukop T, Cervellini I, Mannino E, Venturi C, et al. Tolerability and efficacy study of P2X7 inhibition in experimental Charcot-Marie-Tooth type 1A (CMT1A) neuropathy. Neurobiol Dis. 2016; 95: 145–157. 10.1016/j.nbd.2016.07.017
    1. Adlkofer K, Martini R, Aguzzi A, Zielasek J, Toyka KV, Suter U. Hypermyelination and demyelinating peripheral neuropathy in Pmp22-deficient mice. Nat Genet. 1995; 11: 274–280. 10.1038/ng1195-274
    1. Nave K-A. Myelination and support of axonal integrity by glia. Nature. 2010; 468: 244–252. 10.1038/nature09614
    1. Groh J, Heinl K, Kohl B, Wessig C, Greeske J, Fischer S, et al. Attenuation of MCP-1/CCL2 expression ameliorates neuropathy in a mouse model for Charcot-Marie-Tooth 1X. Hum Mol Genet. 2010; 19: 3530–3543. 10.1093/hmg/ddq269

Source: PubMed

3
Tilaa