Suberoylanilide Hydroxamic Acid as a Neuroactive Compound: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Suberoylanilide hydroxamic acid (SAHA) was reported as a promising therapeutic molecule for several neurological disorders that still lack effective treatments.

  • neuro-epigenetics
  • deacetylase inhibitor
  • protein acetylation
  • in vitro and in vivo treatments

1. Introduction

Neuroplasticity is a fundamental capacity of neurons to reorganize themselves in response to new information and/or sensory stimulation changing connections in the neuronal network. This complex process occurs most profoundly in the first few years of life as neurons grow very rapidly and send out multiple branches, ultimately forming many connections. Defects in this process may compromise brain development and functioning, as observed in several neurodevelopmental disorders (NDDs).
In the last decade, many studies have been aimed at identifying neuroactive molecules capable of exerting a positive effect on the ability of neurons to reorganize the synaptic network [1]. This branch of research is considered a gold mine in drug discovery applicable to a huge spectrum of neurological disorders with neuroplasticity impairment, including NDDs, psychiatric and neurodegenerative disorders.
Originally approved by the Food and Drug Administration (FDA) for the treatment of T-cell lymphoma [2] and permeable to the blood-brain barrier [3], SAHA was reported as a promising therapeutic molecule for several neurological disorders that still lack effective treatments [4][5]. Remarkably, most of the disease genes involved in these pathologies encode splicing factors and structural proteins implicated in neurogenesis and synaptogenesis. Consequently, mutations in these genes cause aberrant functional processes that can lead to the onset and exacerbation of clinical symptoms, including intellectual disability (ID) and epilepsy severity [6][7]. SAHA is a pan-HDAC inhibitor that inhibits both Class I and Class II HDACs. It belongs to a wide group of HDACI that vary in structures, target selectivity, and biological activities. In recent years, their HDAC inhibitor activity has been exploited as antiproliferative and proapoptotic agents in the treatments of cancer and as neuroprotective and neurotrophic molecules in non-malignant conditions of the nervous system [5][8][9][10][11][12]. This dual activity mirrors the lack of molecular specificity and indicates that the pharmacological effects of SAHA and other HDACIs are based on a wide range of mechanisms of action—a feature common to several drugs—whose identification requires further studies. Of course, given the global effects of SAHA and other HDACIs, it is fundamental to distinguish benefits and detrimental effects, both in vitro and in vivo. Several studies highlight that, similarly to other HDACIs, the therapeutic outcome of SAHA—effect, efficacy, effectiveness, and benefit—may vary depending on the dosage used and the biological context analyzed (e.g., cell type, tissue specificity, developmental window, etc.) [4][8][13][14][15][16][17][18]. Although future research may elucidate the downstream effects of SAHA, there is a clear trend toward its applicability with new drug repurposing opportunities because (i) it has a good blood-brain barrier permeability [3]; (ii) It does not lead to massive transcriptional changes; and (iii) compared to other HDACi, it is active at nanomolar concentrations in a variety of cellular and animal models [4][5][15][16][17][18]. Future multi-omic studies in in vivo models coupled with fast transcriptome and proteome screenings may help to figure out the pharmacology response of SAHA to distinguish at molecular levels the advantages and disadvantages of SAHA, also compared to other HDAC inhibitors, allowing the adoption of personalized applicability of HDAC activity inhibition. Although pharmacogenomic studies for HDAC inhibitors are still scarce, genotype-directed dosing could improve pharmacotherapy of SAHA, reducing toxicity risk or suboptimal dosage.

2. The Role of SAHA as a Neuroactive Compound

Neuroprotective effects refer to the ability of a compound to protect neurons from damage or degeneration, thereby preserving their structure and function. SAHA has been demonstrated to have the potential to exert such neuroprotective effects, which could be beneficial in preventing cognitive decline and maintaining cognitive function. Broader assessments of in vivo and in vitro treatments have shown that this activity has multiple downstream effects and is not restricted to a particular signaling pathway (Table 1).
Table 1. Molecular functions of SAHA as a neuroactive compound.
Additionally, SAHA has shown neuritogenic effects, meaning it can promote the growth and development of neurites. Neurites are essential for establishing neural connections and facilitating proper communication between neurons. By enhancing neurite growth, SAHA could potentially aid in the restoration of neuronal connectivity, which may be disrupted in cognitive impairment and epilepsy.
By enhancing neurite growth, SAHA could potentially aid in the restoration of neuronal connectivity, which may be disrupted in cognitive impairment and epilepsy. The combination of both neuroprotective and neuritogenic effects of SAHA strengthens its potential for drug repurposing in the treatment of cognition impairment and epilepsy.

2.1. Neuronal Maturation and Plasticity

Morphological and molecular data indicate that SAHA-treated ESCs are committed toward neural differentiation with a dose-dependent effect that is cytotoxic at higher doses and pro-proliferative at lower concentrations [13]. Furthermore, it has been shown that SAHA promotes a significant increase in mean neurite length, number of neurites/cell, and neurite length/cell via activation of the ERK pathway [11]. This activity is extremely interesting because in some instances ERK activation may lead to detrimental effects and contribute to neurodegeneration; in other cases, it fosters survival and differentiation or counteracts pathogenic effects of mutations in Huntington’s disease models (HD). Indeed, several studies have reported the dual role of ERK1/2 signaling, as a double-edged sword [29], that in response to specific signals may lead to beneficial effects on long-term and short-term memory formation and neuronal protective activity in Huntington’s disease models (HD) [30][31] and to detrimental effects that induce neurotoxicity [32]. This double role of ERK1/2 signaling may reflect differences in the functional response to SAHA, depending on target cells and tissue, as well as the dosage used [33][34].
Supporting this evidence of SAHA as a proneuronal molecule, researchers previously showed the activity of this epidrug counteracting the defective neuronal differentiation and maturation in animal and cellular models ablated for aristaless homeobox-related gene (ARX), a well-conserved disease gene involved in multiple NDDs [14]. Specifically, SAHA is able to recover KDM5C-H3K4me3 signaling and improve neuronal differentiation both in Arx-KO murine ES-derived neurons and ARX/alr-1-deficient Caenorhabditis elegans animals. Behavioral phenotype analysis of alr-1-KO worm mutants revealed that SAHA rescued defective touch response, a behavioral defect affecting mechanosensory neurons [14]. Consistent with these findings, it has been shown that SAHA stimulates the expression of BDNF in cultured rat neurons [23] and controls BDNF release in different brain cell populations contributing to memory formation [24]. Furthermore, several investigations proved the association of SAHA with long-lasting chromatin modifications regulating the expression levels of plasticity-related genes or involved in signaling networks that regulate neuronal morphogenesis [25][26][27]. Noteworthy, in human iPSC-derived neurons and mouse embryonic cortical cells, SAHA upregulates—in a dose-dependent manner—the expression of evolutionarily conserved gene networks with key roles in synaptic maintenance and function [35]. Further studies are required to establish in detail the molecular benefits, as well as to balance the possible installation of inappropriate histone modifications.

2.2. Activation of Autophagy

Emerging evidence suggests that neuronal autophagy regulates the refinement of the polarized morphology of neurons (e.g., axonal outgrowth, dendritic complexity, and spine pruning) and the formation and maintenance of synapses. Mechanistically, it is a complex catabolic process that delivers cytosolic components to lysosomes for degradation. SAHA has been shown to influence the autophagy pathway at various points and to protect the aged brain against plasticity impairment. This is what emerges from two recent studies showing that cognitive dysfunction induced by the anesthetic sevoflurane or insulin resistance can be counteracted by SAHA, through the expression control of autophagy-related gene markers, such as LC3 and P62 [19][20]. As for the mechanism of action, previous studies carried out in MEF cells demonstrated that SAHA induces autophagy by activating the ULK1 complex, which is the most upstream component in the core autophagy pathway, and by suppressing the mammalian target of rapamycin (mTOR), which in turn inhibits autophagy induction [21]. Moreover, several studies have shown that perturbations affecting autophagy may cause NDDs. For example, mutations in genes encoding regulatory autophagic proteins have been detected in ASD patients characterized by elevated mTOR activity (mTORopathy) and defective autophagy [36]. Elevated mTOR signaling activity has been also detected in patients with fragile X syndrome [37] and in congenital forms of epileptogenesis and cortical malformations [38]. Given the activity of SAHA as an mTOR inhibitor, this epidrug might be a promising therapy to correct neuronal autophagy in mTOR-related NDDs. However, further studies are required to identify the autophagy-related pathways involved in neurodevelopmental processes in order to identify an appropriate time window for SAHA administration.

2.3. Microtubule Organization

Microtubules (MTs) are cylindrical cytoskeletal structures constituted by polymers of αβ-tubulin that play essential roles in many essential mammalian cell functions, such as cytoskeletal support and transportation of intracellular cargo, thus regulating neuroplasticity and synaptogenesis. The dynamic changes regulating MT structure and stability are mediated by acetylation of K40 in α-tubulin, a posttranslational modification, which marks the luminal surface of microtubules and is fundamental for microtubule stabilization and vesicle transportation. The dynamics between acetylation/deacetylation of α-tubulin is mediated by the action of acetyltransferases and deacetylases, respectively. Because of its HDACi activity, SAHA induces hyperacetylation of α-tubulin, facilitating sliding between filaments and making MTs more mechanically stable [11][22]. A defective level in the α-tubulin acetylation has been detected in animal models for different neurological disorders, including cortical malformations [39], Rett syndrome [40], and various neurodegenerative pathologies [41]. For some of these pathologies, treatments with other HDAC inhibitors rescue the defective α-tubulin acetylation and thus correct a number of MT abnormalities, such as the defective axonal transport observed in Charcot–Marie–Tooth primary neurons [42] and the aberrant BDNF vesicle trafficking observed in Rett patient fibroblasts [40]. Taking this evidence into account, the applicability of SAHA to counteract cytoskeleton malfunctioning in NDDs seems to be very promising to be addressed.
Related to the acetylation/deacetylation effect of SAHA, Luo et al. (2022) underlined a new mechanism of action preventing axonal damage and neurological dysfunction in a rat model of subarachnoid hemorrhage (SAH). This SAHA-induced response is mediated by heat shock protein (HSP) co-inducer activity via the acetylation of HSP70, which in turn induces the degradation of TDP-43 aggregates [28].

2.4. Regulation of Alternative Splicing Switch

Alternative splicing (AS) is a critical process of posttranscriptional gene expression that diversifies and expands the proteome and increases the functional diversity of molecules implicated in mammalian brain development and plasticity [43][44]. Characterized by high flexibility, AS is a complex mechanism controlled via RNA-binding proteins (RBPs) forming a megadalton complex named spliceosome [43].
In relation to the levels of histone acetylation, the opening and closing states of chromatin have the ability to influence exon inclusion and exclusion by recruiting directly splicing factors to sites of specific exons [45]. Indeed, histone hyperacetylation, which creates a more open chromatin structure, leads to a faster elongation rate phase allowing for the recruitment of splicing factors to a strong splicing site, resulting in exon skipping. Conversely, a slower elongation rate can recruit splicing factors to weak upstream splice sites, resulting in exon inclusion [45]. In these frameworks, splicing defects can be overcome by manipulating HDACs. In addition to that, a recent study proved that SAHA is also capable to counteract splicing abnormalities by rescuing RNA foci, as demonstrated in a mouse model of myotonic dystrophy type 1 (DM1) [46]. DM1 is the most common form of adult neuromuscular dystrophy caused by the abnormal expansion of CTG repeats in the 3′ untranslated region of the dystrophia myotonic protein kinase (DMPK) gene. These expanded repeats lead to the formation of hairpin structures affecting the DMPK mRNA (GC-rich foci), which in turn causes the sequestration of splicing regulators such as MBLN1, resulting in aberrant alternative splicing of a variety of mRNAs [16]. The authors proved that SAHA is able to reduce RNA foci formation.
More interestingly, the daily intraperitoneal injections of SAHA in DM1 mice and the daily administration in DM1 patient-derived myoblasts counteract the aberrant splicing affecting myotonia genes (e.g., chloride channel genes) [46]. Moreover, Nakano et al. [47] proved the efficacy of SAHA to counteract aberrant gene expression caused by the defective splicing of the RE1 silencing transcription factor (REST) gene. This gene encodes a transcriptional repressor that forms a protein complex with HDAC1 and HDAC2. Since HDACs are critical for REST-dependent gene expression, the authors demonstrated that the subcutaneous injection of SAHA in Rest+/ΔEx4 mice induces histone hyperacetylation, rescues defective gene expression of REST target genes, and subsequently induces improvement of cochlear defects [47].

This entry is adapted from the peer-reviewed paper 10.3390/biom13091301

References

  1. von Bernhardi, R.; Bernhardi, L.E.; Eugenín, J. What Is Neural Plasticity? Adv. Exp. Med. Biol. 2017, 1015, 1–15.
  2. Bondarev, A.D.; Attwood, M.M.; Jonsson, J.; Chubarev, V.N.; Tarasov, V.V.; Schiöth, H.B. Recent developments of HDAC inhibitors: Emerging indications and novel molecules. Br. J. Clin. Pharmacol. 2021, 87, 4577–4597.
  3. Guntner, A.S.; Peyrl, A.; Mayr, L.; Englinger, B.; Berger, W.; Slavc, I.; Buchberger, W.; Gojo, J. Cerebrospinal fluid penetration of targeted therapeutics in pediatric brain tumor patients. Acta Neuropathol. Commun. 2020, 8, 78.
  4. Gottesfeld, J.M.; Pandolfo, M. Development of histone deacetylase inhibitors as therapeutics for neurological disease. Future Neurol. 2009, 4, 775–784.
  5. Athira, K.V.; Sadanandan, P.; Chakravarty, S. Repurposing Vorinostat for the Treatment of Disorders Affecting Brain. Neuromolecular Med. 2021, 23, 449–465.
  6. Ciptasari, U.; van Bokhoven, H. The phenomenal epigenome in neurodevelopmental disorders. Hum. Mol. Genet. 2020, 29, R42–R50.
  7. Mossink, B.; Negwer, M.; Schubert, D.; Nadif Kasri, N. The emerging role of chromatin remodelers in neurodevelopmental disorders: A developmental perspective. Cell Mol. Life Sci. 2021, 78, 2517–2563.
  8. Carafa, V.; Nebbioso, A.; Altucci, L. Histone deacetylase inhibitors: Recent insights from basic to clinical knowledge & patenting of anti-cancer actions. Recent Pat. Anticancer Drug Discov. 2011, 6, 131–145.
  9. Goey, A.K.; Sissung, T.M.; Peer, C.J.; Figg, W.D. Pharmacogenomics and histone deacetylase inhibitors. Pharmacogenomics 2016, 17, 1807–1815.
  10. Suresh, P.S.; Devaraj, V.C.; Srinivas, N.R.; Mullangi, R. Review of bioanalytical assays for the quantitation of various HDAC inhibitors such as vorinostat, belinostat, panobinostat, romidepsin and chidamine. Biomed. Chromatogr. 2017, 31, 3807.
  11. Shukla, S.; Shariat-Madar, Z.; Walker, L.A.; Tekwani, B.L. Mechanism for neurotropic action of vorinostat; a pan histone deacetylase inhibitor. Mol. Cell. Neurosci. 2016, 77, 11–20.
  12. Shanmugam, G.; Rakshit, S.; Sarkar, K. HDAC inhibitors: Targets for tumor therapy, immune modulation and lung diseases. Transl. Oncol. 2022, 16, 101312.
  13. Franci, G.; Casalino, L.; Petraglia, F.; Miceli, M.; Menafra, R.; Radic, B.; Tarallo, V.; Vitale, M.; Scarfò, M.; Pocsfalvi, G.; et al. The class I-specific HDAC inhibitor MS-275 modulates the differentiation potential of mouse embryonic stem cells. Biol. Open 2013, 22, 1070–1077.
  14. Poeta, L.; Padula, A.; Attianese, B.; Valentino, M.; Verrillo, L.; Filosa, S.; Shoubridge, C.; Barra, A.; Schwartz, C.E.; Christensen, J.; et al. Histone demethylase KDM5C is a SAHA-sensitive central hub at the crossroads of transcriptional axes involved in multiple neurodevelopmental disorders. Hum. Mol. Genet. 2019, 28, 4089–4102.
  15. Hockly, E.; Richon, V.M.; Woodman, B.; Smith, D.L.; Zhou, X.; Rosa, E.; Sathasivam, K.; Ghazi-Noori, S.; Mahal, A.; Lowden, P.A.S.; et al. Suberoylanilide hydroxamic acid; a histone deacetylase inhibitor; ameliorates motor deficits in a mouse model of Huntington’s disease. Proc. Natl. Acad. Sci. USA 2003, 100, 2041–2046.
  16. Benito, E.; Urbanke, H.; Ramachandran, B.; Barth, J.; Halder, R.; Awasthi, A.; Jain, G.; Capece, V.; Burkhardt, S.; Navarro-Sala, M.; et al. HDAC inhibitor-dependent transcriptome and memory reinstatement in cognitive decline models. J. Clin. Investig. 2015, 125, 3572–3584.
  17. Elvir, L.; Duclot, F.; Wang, Z.; Kabbaj, M. Epigenetic regulation of motivated behaviors by histone deacetylase inhibitors. Neurosci. Biobehav. Rev. 2019, 105, 305–317.
  18. Lunke, S.; Maxwell, S.; Khurana, I.; Kaipananickal, H.; Okabe, J.; Al-Hasani, K.; El-Osta, A. Epigenetic evidence of an Ac/Dc axis by VPA and SAHA. Clin. Epigenet. 2021, 13, 58.
  19. Kakoty, V.; Sarathlal, K.C.; Dubey, S.K.; Yang, C.H.; Marathe, S.A.; Taliyan, R. Epigenetic regulation and autophagy modulation debilitates insulin resistance associated Alzheimer’s disease condition in rats. Metab. Brain Dis. 2022, 37, 927–944.
  20. Fang, P.; Chen, C.; Zheng, F.; Jia, J.; Chen, T.; Zhu, J.; Chang, J.; Zhang, Z. NLRP3 inflammasome inhibition by histone acetylation ameliorates sevoflurane-induced cognitive impairment in aged mice by activating the autophagy pathway. Brain Res. Bull. 2021, 172, 79–88.
  21. Gammoh, N.; Lam, D.; Puente, C.; Ganley, I.; Marks, P.A.; Jiang, X. Role of autophagy in histone deacetylase inhibitor-induced apoptotic and nonapoptotic cell death. Proc. Natl. Acad. Sci. USA 2012, 109, 6561–6565.
  22. Dompierre, J.P.; Godin, J.D.; Charrin, B.C.; Cordelières, F.P.; King, S.J.; 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, 3571–3583.
  23. Koppel, I.; Timmusk, T. Differential regulation of Bdnf expression in cortical neurons by class-selective histone deacetylase inhibitors. Neuropharmacology 2013, 75, 106–115.
  24. Hossain, M.S.; Oomura, Y.; Katafuchi, T. Glucose Can Epigenetically Alter the Gene Expression of Neurotrophic Factors in the Murine Brain Cells. Mol. Neurobiol. 2018, 55, 3408–3425.
  25. Rusconi, F.; Grillo, B.; Ponzoni, L.; Bassani, S.; Toffolo, E.; Paganini, L.; Mallei, A.; Braida, D.; Passafaro, M.; Popoli, M.; et al. LSD1 modulates stress-evoked transcription of immediate early genes and emotional behavior. Proc. Natl. Acad. Sci. USA 2016, 113, 3651–3656.
  26. de la Fuente Revenga, M.; Ibi, D.; Saunders, J.M.; Cuddy, T.; Ijaz, M.K.; Toneatti, R.; Kurita, M.; Holloway, T.; Shen, L.; Seto, J.; et al. HDAC2-dependent Antipsychotic-like Effects of Chronic Treatment with the HDAC Inhibitor SAHA in Mice. Neuroscience 2018, 388, 102–117.
  27. Cavallo, F.; Troglio, F.; Fagà, G.; Fancelli, D.; Shyti, R.; Trattaro, S.; Zanella, M.; D’Agostino, G.; Hughes, J.M.; Cera, M.R.; et al. High-throughput screening identifies histone deacetylase inhibitors that modulate GTF2I expression in 7q11.23 microduplication autism spectrum disorder patient-derived cortical neurons. Mol. Autism 2020, 11, 88.
  28. Luo, K.; Wang, Z.; Zhuang, K.; Yuan, S.; Liu, F.; Liu, A. Suberoylanilide hydroxamic acid suppresses axonal damage and neurological dysfunction after subarachnoid hemorrhage via the HDAC1/HSP70/TDP-43 axis. Exp. Mol. Med. 2022, 54, 1423–1433.
  29. Colucci-D’Amato, L.; Perrone-Capano, C.; di Porzio, U. Chronic activation of ERK and neurodegenerative diseases. Bioessays 2003, 25, 1085–1095.
  30. Medina, J.H.; Viola, H. ERK1/2: A Key Cellular Component for the Formation, Retrieval, Reconsolidation and Persistence of Memory. Front. Mol. Neurosci. 2018, 11, 361.
  31. Fusco, F.R.; Anzilotti, S.; Giampà, C.; Dato, C.; Laurenti, D.; Leuti, A.; Colucci D’Amato, L.; Perrone, L.; Bernardi, G.; Melone, M.A. Changes in the expression of extracellular regulated kinase (ERK 1/2) in the R6/2 mouse model of Huntington’s disease after phosphodiesterase IV inhibition. Neurobiol. Dis. 2012, 46, 225–233.
  32. Nizzari, M.; Barbieri, F.; Gentile, M.T.; Passarella, D.; Caorsi, C.; Diaspro, A.; Taglialatela, M.; Pagano, A.; Colucci-D’Amato, L.; Florio, T.; et al. Amyloid-β protein precursor regulates phosphorylation and cellular compartmentalization of microtubule associated protein tau. J. Alzheimer’s Dis. 2012, 29, 211–227.
  33. Chen, S.H.; Wu, H.M.; Ossola, B.; Schendzielorz, N.; Wilson, B.C.; Chu, C.H.; Chen, S.L.; Wang, Q.; Zhang, D.; Qian, L.; et al. Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, protects dopaminergic neurons from neurotoxin-induced damage. Br. J. Pharmacol. 2012, 165, 494–505.
  34. Kurundkar, D.; Srivastava, R.K.; Chaudhary, S.C.; Ballestas, M.E.; Kopelovich, L.; Elmets, C.A.; Athar, M. Vorinostat, an HDAC inhibitor attenuates epidermoid squamous cell carcinoma growth by dampening mTOR signaling pathway in a human xenograft murine model. Toxicol. Appl. Pharmacol. 2013, 266, 233–244.
  35. Swarup, V.; Hinz, F.I.; Rexach, J.E.; Noguchi, K.I.; Toyoshiba, H.; Oda, A.; Hirai, K.; Sarkar, A.; Seyfried, N.T.; Cheng, C.; et al. Identification of evolutionarily conserved gene networks mediating neurodegenerative dementia. Nat. Med. 2019, 25, 152–164.
  36. Thomas, S.D.; Jha, N.K.; Ojha, S.; Sadek, B. mTOR Signaling Disruption and Its Association with the Development of Autism Spectrum Disorder. Molecules 2023, 28, 1889.
  37. Hoeffer, C.A.; Sanchez, E.; Hagerman, R.J.; Mu, Y.; Nguyen, D.V.; Wong, H.; Whelan, A.M.; Zukin, R.S.; Klann, E.; Tassone, F. Altered mTOR signaling and enhanced CYFIP2 expression levels in subjects with fragile X syndrome. Genes Brain Behav. 2012, 11, 332–341.
  38. Moloney, P.B.; Cavalleri, G.L.; Delanty, N. Epilepsy in the mTORopathies: Opportunities for precision medicine. Brain Commun. 2021, 3, 4.
  39. Drongitis, D.; Caterino, M.; Verrillo, L.; Santonicola, P.; Costanzo, M.; Poeta, L.; Attianese, B.; Barra, A.; Terrone, G.; Lioi, M.B.; et al. Deregulation of microtubule organization and RNA metabolism in Arx models for lissencephaly and developmental epileptic encephalopathy. Hum. Mol. Genet. 2022, 31, 1884–1908.
  40. Gold, W.A.; Lacina, T.A.; Cantrill, L.C.; Christodoulou, J. MeCP2 deficiency is associated with reduced levels of tubulin acetylation and can be restored using HDAC6 inhibitors. J. Mol. Med. 2015, 93, 63–72.
  41. Cappelletti, G.; Calogero, A.M.; Rolando, C. Microtubule acetylation: A reading key to neural physiology and degeneration. Neurosci. Lett. 2021, 755, 135900.
  42. Adalbert, R.; Kaieda, A.; Antoniou, C.; Loreto, A.; Yang, X.; Gilley, J.; Hoshino, T.; Uga, K.; Makhija, M.T.; Coleman, M.P. Novel HDAC6 Inhibitors Increase Tubulin Acetylation and Rescue Axonal Transport of Mitochondria in a Model of Charcot-Marie-Tooth Type 2F. ACS Chem. Neurosci. 2020, 11, 258–267.
  43. Su, C.H.; Dhananjaya, D.; Tarn, W.Y. Alternative Splicing in Neurogenesis and Brain Development. Front. Mol. Biosci. 2018, 12, 12.
  44. Joglekar, A.; Prjibelski, A.; Mahfouz, A.; Collier, P.; Lin, S.; Schlusche, A.K.; Marrocco, J.; Williams, S.R.; Haase, B.; Hayes, A.; et al. A spatially resolved brain region- and cell type-specific isoform atlas of the postnatal mouse brain. Nat. Commun. 2021, 12, 463.
  45. Rahhal, R.; Seto, E. Emerging roles of histone modifications and HDACs in RNA splicing. Nucleic Acids Res. 2019, 47, 4911–4926.
  46. Neault, N.; Ravel-Chapuis, A.; Baird, S.D.; Lunde, J.A.; Poirier, M.; Staykov, E.; Plaza-Diaz, J.; Medina, G.; Abadía-Molina, F.; Jasmin, B.J.; et al. Vorinostat Improves Myotonic Dystrophy Type 1 Splicing Abnormalities in DM1 Muscle Cell Lines and Skeletal Muscle from a DM1 Mouse Model. Int. J. Mol. Sci. 2023, 24, 3794.
  47. Nakano, Y.; Kelly, M.C.; Rehman, A.U.; Boger, E.T.; Morell, R.J.; Kelley, M.W.; Friedman, T.B.; Bánfi, B. Defects in the Alternative Splicing-Dependent Regulation of REST Cause Deafness. Cell 2018, 174, 536–548.
More
This entry is offline, you can click here to edit this entry!
ScholarVision Creations