Open access peer-reviewed chapter

The Role of TDP-43 in Genome Repair and beyond in Amyotrophic Lateral Sclerosis

Written By

Joy Mitra and Muralidhar L. Hegde

Submitted: 10 March 2020 Reviewed: 29 April 2020 Published: 03 June 2020

DOI: 10.5772/intechopen.92696

From the Edited Volume

Amyotrophic Lateral Sclerosis - Recent Advances and Therapeutic Challenges

Edited by Muralidhar L. Hegde

Chapter metrics overview

914 Chapter Downloads

View Full Metrics

Abstract

The pathology of the RNA-/DNA-binding protein TDP-43, first implicated a decade ago in the motor neuron disease amyotrophic lateral sclerosis (ALS), has been subsequently linked to a wide spectrum of neurodegenerative diseases, including frontotemporal dementia (FTD), Alzheimer’s disease (AD), and related dementia-associated disorders. ALS, also known as Lou Gehrig’s disease, is a progressive, degenerative motor neuron disorder, characterized by a diverse etiopathology. TDP-43 pathology, mediated by a combination of several mutations in the TARDBP gene and stress factors, has been linked to more than 97% of ALS patients. We recently identified, for the first time, the critical involvement of TDP-43 in neuronal genome maintenance and the repair of DNA double-strand breaks (DSBs). Our studies showed that TDP-43 regulates the DNA break-sealing activities of the XRCC4-DNA Ligase 4 (LIG4) complex in DSB repair, suggesting that loss of genomic integrity in TDP-43-associated neurodegeneration may be amenable to a DNA repair-based intervention. In this chapter, we discuss the broader aspects of TDP-43 toxicity-induced pathomechanisms, including the emerging role of TDP-43 in neuronal DSB repair and its synergistic genotoxic effects with other neurodegeneration-associated etiologies that contribute significantly to neuronal dysfunction. We also discuss potential future perspectives and underscore how unraveling the molecular basis and implications of TDP-43-induced genome instability in ALS could guide the development of neuroprotective therapies.

Keywords

  • TDP-43
  • DNA damage
  • DNA double-strand breaks
  • nonhomologous end joining
  • XRCC4-DNA ligase 4
  • ALS
  • stress granule
  • Rab11
  • neurodegeneration

1. TDP-43 pathology: a predominant player in ALS

Transactive response DNA-binding protein 43 (TDP-43) is a versatile 43 kDa DNA-/RNA-binding protein of the heterogeneous nuclear ribonucleoprotein (hnRNP) family. TDP-43’s cytosolic aggregation and inclusion body formation are the key pathologic hallmarks of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) [1, 2, 3]. In these progressive motor neuron diseases, TDP-43 pathology manifests as the tau- and synuclein- negative and ubiquitin-positive inclusions in the anterior horn, spinal cord, neocortex, and hippocampus regions of the brain. In the last decade, tremendous scientific investigations have been carried out to understand the etiopathologies of TDP-43 toxicity in ALS and FTD. These studies demonstrated that TDP-43 pathology-linked ALS is one of the most complex neurological diseases due to TDP-43 pathomechanisms overlapping with other ALS-causing genes, such as C9orf72 [4, 5, 6], valosin-containing protein (VCP) [7, 8, 9], fused in sarcoma (FUS) [10], optineurin (OPTN) [11], ubiquilin 2 (UBQLN2) [12, 13], and ataxin 2 (ATXN2) [14, 15]. Strikingly, TDP-43 pathology has been detected in more than 97% of sporadic ALS cases (>90%), making it a predominant player in most ALS patients.

1.1 Familial and sporadic ALS

Due to highly overlapping symptoms, determining the sporadic or familial nature of the ALS disease at its time of onset is difficult based on clinical features alone. Familial ALS (FALS) is predicted based on genetic screening for TARDBP mutation(s) in the patient and matching the same mutation in the family member(s) with a positive clinical/medical history of neurodegeneration and dementia in the juvenile or early stage of life. Race, age, and gender are risk factors for progression of FALS. FALS incidences vary significantly from 5 to 10% among all ALS cases. The disease is more common in Caucasian men and women than African, Asian, and Hispanic populations [16]. Notably, clusters of ALS incidences have been associated with socio-economic conditions along with race. According to Centers for Disease Control and Prevention, ALS prevalence is highest in the Midwest and Northeast of US main land (5.7 and 5.5 per 100,000 population, respectively) [17]. In Europe, the prevalence of FALS was lower in the southern part [18]. Studies show that men are at slightly higher risk than women, with an average age of onset ~60 years. Patients usually survive for 1.5–4 years after diagnosis, depending on the disease aggressiveness [19].

FALS associated pathology affects anterior horn motor neurons in the cervical and lumbosacral regions in the spinal cord, frontal cortex, and cranial nerve motor neurons in the pons and medulla segments of brainstem. Both sporadic ALS (SALS) and FALS pathologies are associated with upper and/or lower motor neurons of the spinal cord. Upper motor neuron diseases accompany degeneration of lateral corticospinal tracts leading to hyperreflexia/spasticity and cardiac arrest. Conversely, lower motor neuron death leads to muscle atrophy and denervation.

FALS cases fall into to three categories, namely, autosomal dominant, autosomal recessive, and X-linked dominant. Autosomal dominance is the most common inherited pattern among the FALS cases, where a mutation in one copy of the ALS-linked gene is sufficient to develop ALS conditions. These patients usually have a strong positive family history and their children bear 50% risk of developing ALS. However, autosomal dominance can also happen in apparently sporadic ALS (because the genetic inheritance is unknown or poor medical history) with de novo dominant mutation(s). In such cases, patients’ siblings may have very low risk but children may have up to 50% risk of developing ALS. To date, more than 50 mutations in TARDBP gene have been identified in ALS etiopathologies [20]. In addition, a study on a German cohort of non-SOD1 FALS patients revealed mutations in TARDBP, suggesting TARDBP gene mutation screening should be crucial among non-SOD1 FALS patients [21]. The known TARDBP mutations in FALS are G290A, G298S, A315T, M337V, N345K, A382T, and I383V; some of the common mutations identified in SALS are D169G, G287S, G294A, Q331K, G348C, R361S, and N390S/D (Figure 1) [21, 22, 23, 24, 25, 26, 27]. Most of these TARDBP mutations are located within exon 6, as commonly found in Caucasian and European ALS patients; however, a large cohort of Han Chinese population with non-SOD1 SALS did not show any such mutations in exon 6 [28].

Figure 1.

TDP-43 protein’s structural organization. Schematic representation of TDP-43 protein structure shows N-terminal domain of TDP-43 consists of highly ordered nucleic acid binding domains, namely, RNA recognition motifs (RRM) 1 and 2. RRM1 domain follows a bifurcated nuclear localization signal sequence (82–98aa)—NLS1 and NLS2. RRM1 domain spans from 104 to 176aa. RRM2 domain ranges from 192aa to 262aa including bifurcated nuclear export signal sequence (239–250aa)—NES1 and NES2. Later part in the C-terminal domain is mostly disordered and contains majority of the pathogenic mutational hotspots. Familial ALS-related mutations are indicated in red, and sporadic ALS-linked mutations are in blue color.

SALS has been reported in an increasing number among people exposed to lead or other heavy metals [29, 30, 31], smoking [32, 33], and pesticides [34]. Increased exposure to organochlorine pesticides, biphenyls, and brominated flame retardants used in household furniture is associated with higher risk for ALS [35, 36, 37]. Interestingly, while smoking has been shown to predispose individuals to ALS risk, alcohol consumption did not show any effect on ALS development [38]. This suggests that diet and exposure to various environmental toxins could increase the ALS susceptibility in the vulnerable population. Most notably, US veterans from the Gulf War [39, 40] and those with head injury [41, 42] are also at a greater risk for developing SALS. By targeted sequencing of exon 6 of TARDBP from brain samples of US veterans with SALS, we observed several missense mutations (both reported and novel) and the association of TDP-43 Q331K mutation with DNA DSB repair impairment [22]. Another well-known incidence of sporadic ALS is the Guamanian ALS, which falls on the Parkinsonism-dementia complex spectrum (ALS/PDC). This particular type of ALS in people living in Guam and Rota islands is caused by the consumption of cyanobacteria-infested cycad fruits containing the neurotoxin β-methylamino-L-alanine (BMAA) [43, 44]. BMAA exerts neurotoxicity by incorporating into amino acid sequences and inducing tau pathology, oxidative stress, glutathione depletion, and protein aggregation [45, 46, 47, 48]. Genome-wide analyses for ALS-/PDC-associated chromosomal loci revealed three disease-specific loci—two regions on chromosome 12 and the MAPT region on chromosome 17—in these populations [49].

1.2 Unique feature of TDP-43 pathology: dual role of loss of function and gain of toxicity

Given that TDP-43 contains a prion-like domain, its cellular content and distribution are two contributing factors for preventing the protein aggregation leading to the onset of disease. These two conditions are maintained by TDP-43’s autoregulatory feedback loop mechanism. TDP-43 is mainly a nuclear protein, but it also shuttles to the cytosol, mitochondria, and other cellular organelles in response to various stimuli. In ALS/FTD pathology, TDP-43 mislocalization, due to pathogenic mutations and/or protein aggregation, is known as the primary culprit for the onset of neurodegeneration. Studies suggest that initiation of caspase-3/7 activation by TDP-43 toxicity facilitates the disease progression by cleaving mislocalized TDP-43 into 35 and 25 kDa fragments. These highly aggregation-prone fragments promote inclusion body formations with the hyperphosphorylated and polyubiquitinated C-terminal domain of TDP-43 [50, 51]. In addition to the C-terminal fragment, full-length and homo-dimerized TDP-43 also has been identified in the spinal cord and brain of ALS patients [52, 53]. Furthermore, recent findings have highlighted that this protein aggregation enhances the aggregation propensities of other prion-like domain containing proteins. The interaction of these prion-like proteins increasing the disease complexity severalfold higher than that of single-protein toxicities [54]. TDP-43’s cellular shuttling is regulated by the bifurcated nuclear localization signal (NLS) and nuclear export signal (NES) sequences (Figure 1), and loss of either of these signal sequences could be deleterious for cell survival. In vitro studies expressing NLS-deleted TDP-43 found accelerated cytoplasmic aggregate formation along with aberrant RNA processing defects [55, 56]. This phenomenon was further supported by in vivo studies with a transgenic mouse model and transcriptomic analysis in ALS/FTD human brain samples, which revealed significantly altered mRNA splicing of histones and aberrant chromatin remodeling following cytosolic accumulation of toxic TDP-43 [57]. Almost all of the pathogenic sporadic and familial mutations in TDP-43 induce its nuclear clearance and cytosolic sequestration to varying extents. However, in vitro studies with the FALS-linked, NLS-specific A90V mutation in TDP-43 found disease-like detergent insoluble cytosolic aggregates, confirming the crucial role of NLS/NES sequences in TDP-43 homeostasis, and further suggesting cytosolic aggregation of TDP-43 is a determining factor in motor neuron death [58, 59].

Persistent neuroinflammation is considered one of the leading causes for motor neuron death in ALS/FTD and also affects patients’ therapeutic response. Innate immune responses, including microglial hyperactivation and astrogliosis, were consistently documented in human postmortem ALS human brain and spinal cord, as well as brains from ALS-model mice [60, 61, 62, 63]. TDP-43 plays a vital role in regulation of neuroinflammation by inhibiting NF-κB activation, possibly through competitive binding to importin α3 [64]. TDP-43 depletion or nuclear loss induces extracellular secretion of TNF-α and enhanced nuclear localization of NF-κB p65 in glia and neurons [65, 66, 67]. Furthermore, inflammatory responses increase nuclear loss of TDP-43 and progression of ALS/FTD pathology.

Advertisement

2. Broad functions of TDP-43 in central nervous system (CNS)

2.1 RNA transactions

Extensive research in the last decade on TDP-43 toxicity in ALS has established that TDP-43 has multiple functions, including autoregulation of its levels through a negative feedback loop and controlling a diverse set of RNA-associated mechanisms, including pre-mRNA processing and splicing, micro RNA biogenesis, and RNA transport, transcription, and translation. Importantly, TDP-43 also regulates protein levels of critical RNA-binding proteins involved in RNA splicing, including SRSF1, PTB, and hnRNP L [68, 69]. TDP-43’s autoregulation is solely controlled by the TDP-43-binding region (TDPBR) domain, which is comprised of several polyadenylation (pA) sites. The silent intron-7 linked to the TDPBR regulates the alternative splicing of pA sites based on the TDP-43 cellular concentration. In the steady state, pA1 allows cytoplasmic shuttling of TDP-43 mRNA for the appropriate amount of protein production. When overexpressed, TDP-43 binds threefold more to TDPBR and increases polymerase II binding at this sequence, which then processes intron-7 to remove the pA1 signal sequence and enforces the use of pA2 and pA4 sites [70]. The use of pA2/A4 sites makes the TDP-43 mRNA partially retained in the nucleus and leads to reduced production of TDP-43 protein in the cell [71]. Moreover, a recent study on TDP-43 autoregulation using the Drosophila model has identified human homolog transcription elongation regulator 1 (TCGER1) of Drosophila protein GC42724 as having a role in TDP-43 mRNA alternative splicing and nucleo-cytoplasmic shuttling. TCGER1 regulates TDP-43 production through its TDPBR region [72].

TDP-43 contains two RNA recognition motifs (RRMs): RRM1 and RRM2 (Figure 1). Each RRM contains two highly conserved RNA recognition segments: octameric ribonucleoprotein 1 and hexameric ribonucleoprotein 2 [73]. TDP-43 RRMs bind a minimum of six UG/TG repeats, and its binding affinity increases with increasing numbers of repeats [74]. However, the binding preference is quite different between two motifs; unlike RRM1, RRM2 prefers a short stretch of UG repeats over long repeats ((UG)3 > (UG)6) [75].

Amino acid sequence analysis reveals a high degree of sequence similarities in the N-terminal domains of TDP-43 among human, mouse, rat, Drosophila melanogaster, and Caenorhabditis elegans, suggesting that TDP-43’s function is crucial in these organisms. The C-terminal region of TDP-43 is predominantly disordered in its native structure, comprising a prion-like domain and several glycine-asparagine rich patches that contribute to the exon-skipping activity of TDP-43 (Figure 1). In vivo studies have revealed that TDP-43 regulates its splicing mechanism via its C-terminal glycine-rich domains [76]. A recent study of TDP-43 knockdown in a Drosophila model expressing chimeric repressor proteins demonstrated that TDP-43 in motor neurons regulates RNA splicing fidelity through splicing repression [77]. These findings are in agreement with previous studies reporting TDP-43’s interaction with more than 6000 mRNAs in the mouse brain and TDP-43 depletion-induced altered splicing of ~900 mRNAs causing neuronal vulnerability (Figure 2) [78]. Furthermore, a recent study with ALS-TDP-43 M337V mutant knock-in mouse model has revealed mRNA splicing deregulation as the major pathological hallmark of mutant TDP-43, even in the absence of any noticeable motor deficits [79]. The mammalian TDP-43 primary transcript produces 11 alternatively spliced variants of mRNA, further supporting the functional complexity of TDP-43 [80]. As part of the hnRNP family, TDP-43 has a similar domain organization to hnRNP A1 and A2/B1 [81]. TDP-43’s C-terminal domain interacts with a number of hnRNP family proteins, particularly hnRNP A2/B1 and A1, to form the hnRNP rich complex that is crucial for splicing inhibition [82]. HnRNP A1, a 34 kDa protein, is abundant in motor neurons and has been implicated in the pathomechanism of spino-muscular atrophy [83]. Mislocalized TDP-43 modulates the inclusion of exon 7B in the alternatively spliced hnRNP A1 transcript, leading to the production of an isoform with an extended prion-like domain [84, 85, 86]. Because TDP-43 and hnRNP A1 interact directly, increasing the aggregation propensity of hnRNP A1 could strongly influence TDP-43’s aggregation in a synergistic pattern [87].

Figure 2.

TDP-43 proteinopathy causes RNA processing defects. TDP-43 being a major functional subunit of spliceosomal complex regulates the RNA maturation processes of hundreds of target genes. Many of which are directly associated with amyotrophic lateral sclerosis (ALS) and FTD. TDP-43, through its RRM domains, binds mainly to intronic and 3ʹ UTR-located UG-repeat sequences of pre-mRNA to modulate their splicing events. In healthy condition, TDP-43 strongly promotes normal splicing events and inhibits disease-causing aberrant alternative splicing. However, ALS/FTD-TDP-43 pathology induces aberrant splicing several folds higher than normal splicing leading to mRNA dysregulation-associated proteinopathies in motor neurons and glia. Moreover, exposure to various environmental toxins like heavy metals (Lead, iron, zinc), pesticides (organochloride compounds), and food toxins (BMAA) could cause pathogenic alterations in TDP-43 leading to its mislocalization and dysregulation of its splicing activity as a result causing sporadic ALS.

In addition to pre-mRNA splicing regulation, the N-terminal domain (NTD) of TDP-43 plays an essential role in protein stabilization and prevention of pathogenic cytoplasmic aggregation. In vitro studies have shown that N-terminal Leu71 and Val72 in the β7 strand region at the interface are crucial for its homodimerization into dimers and/or tetramers in a concentration-dependent manner [88]. Furthermore, single amino acid substitutions at V31R and T32R disrupt TDP-43’s splicing activity and induce aggregation. The L28A mutation strongly destabilizes the TDP-43 NTD and promotes its mislocalization, and the L27A mutation increases its monomeric forms [89]. Recent findings by Chen et al. show that the K181E mutation near the TDP-43 RRM1 domain disrupts TDP-43’s ability to process mRNA, induces mutant TDP-43 hyperphosphorylation, enhances detergent-resistant aggregation propensity by several fold, and leads to more wild-type TDP-43 in the nuclear and cytoplasmic inclusion bodies [90]. TDP-43 oligomerization has been reported to be the first toxic intermediate in TDP-43 proteinopathies [91]. An RNA-mediated intervention strategy showed inhibition of TDP-43 misfolding in wild-type and pathogenic ALS-TDP-43 mutants, further suggesting a critical role for RRM domains in TDP-43 pathology.

MicroRNAs (miRNAs) are the master regulators of a number of vital cellular mechanisms and diseases, including neurodegeneration and genomic instability. TDP-43 regulates a subset of miRNAs by direct interaction and modulates their biogenesis through Drosha and Dicer complexes [92]. In the brain, neurexin-1 (NRXN1) controls vesicular trafficking between synaptic junctions. TDP-43 has been shown to bind mir-NID1 to suppress NRXN1 expression and thereby inhibit neuronal development and functionality [93]. Given that long noncoding RNA (lncRNA) confers a higher degree of gene expression regulation, TDP-43’s increased interaction with two crucial lncRNAs, MALAT1 and NEAT1, possibly modulates the RNA metabolism dysregulation of ALS- and FTD-associated TDP-43 pathology [94, 95]. Furthermore, lncRNA gadd7, which is involved in cell cycle regulation and DDR signaling, orchestrates TDP-43’s interaction with CDK6 mRNA that leads to its controlled decay [96].

2.2 Stress granules

Stress granule (SG) assembly is a dynamic, reversible process that promotes cell survival when stress factors are present. Membraneless SG organelles vary in their morphology and building composition in a cell type-specific manner [97, 98]. As long as the assembly/disassembly ratio is maintained in healthy cells, SGs act as the emergency store house for certain classes of RNA and protein molecules to protect them from the various stress stimuli (Figure 3). However, altered SG assembly processes are associated with a number of human diseases, including neurodegenerative disorders and dementia [99]. Recent discoveries indicate SGs are critical players in modulating signal circuits determining cell death versus survival in response to stress exposure.

Figure 3.

TDP-43 is a critical component of stress granule. Stress granule formation, in normal condition, is a very important cellular defense mechanism to overcome stress responses. Stress granule assembly and disassembly is a reversible process in healthy cells. It’s a membraneless structure consisting of stress granule marker proteins TIA-1, G3BP-1, and TDP-43 as the major structural component. Given that ALS-/FTD-associated TDP-43 pathology induces aberrant cytosolic sequestration of toxic TDP-43, it impairs the reversible nature of stress granule formation mechanism, thereby inducing the seeding mechanism for protein aggregation.

Emerging evidence suggests that TDP-43, but not FUS, regulates SG dynamics and secondary polymerization of TIA-1, which is essential for SG assembly (Figure 3) [100]. TIA-1 is an RNA-binding protein and classical SG marker and has more recently been considered as a candidate gene for ALS and ALS/FTD due to its rare heterozygous mutations (P362L and E384K) in the conserved amino acid residues within its low complexity domain [101]. TDP-43 increases its association with TIA-1 in a time-dependent manner in response to the osmotic and oxidative stressor, Sorbitol, in primary glial cells or other stressor-induced SGs in primary cortical neurons and astrocytes [102]. Studies have also shown aging as a crucial modulator of SG dynamics in neurodegenerative conditions. Notably, wild-type and ALS-linked pathogenic TDP-43 mutants show distinct patterns of stress formation rates and morphology. Mutant TDP-43 exhibits faster stress granule incorporation along with rapid increases in granule-size, while wild-type TDP-43 forms increasing numbers of granules with consistent size over time [99]. Apart from the TIA-1 aggregation regulation, TDP-43 also controls the mRNA level of G3BP1, an essential SG initiation factor. The loss of functional TDP-43 or pathogenic TDP-43-mediated G3BP1 mRNA depletion perturbs the interaction between SG components and processing bodies, leading to impaired storage of polyadenylated mRNAs [103]. However, different TDP-43 ALS-mutant variants exhibit differential regulation mechanisms. For instance, TDP-43 D169G does not affect SG formation mechanism, but R361S and A382T variants show loss-of-function phenotypes with respect to SG dynamics [104, 105]. Interestingly, chronic TDP-43 liquid-liquid phase separation (LLPS) has been observed in neuronal cells and ALS-iPSc-derived motor neurons, even with only a transient stress induction from sonicated amyloid fibrils or arsenite. Early stage LLPS TDP-43 droplets are formed in the nucleus independent of conventional SG mechanisms. Cytosolic TDP-43 droplets formed by transient stress induction gradually incorporate importin-α and Nup62, leading to the mislocalization of nuclear pore complex proteins Nup107, Ran, and RanGap1. This situation inhibits nucleo-cytoplasmic shuttling and clearance of toxic TDP-43 and results in neuronal cell death [106]. Hans et al. have shown that TDP-43 hyperubiquitylation-mediated insolubility could happen by multiple distinct mechanisms independent of its translocation to cytosolic SGs [107]. This study shows neither endoplasmic reticulum kinase inhibitors nor translation blockers could prevent TDP-43 ubiquitylation. Moreover, the sorbitol-induced stress response involves impaired TDP-43 splicing activity, whereas sodium arsenite-induced SG formation occurs through oxidative stress, which can be quenched by the treatment with antioxidant like N-acetylcysteine. High-content screening of inhibitors for blocking pathogenic TDP-43 accumulation in SGs from ALS patient-derived iPSc-motor neurons has identified planar aromatic moieties with DNA intercalation properties as the potent small molecule therapy for ALS/FTD-TDP-43 pathology [108]. In this context, it is important to mention that TDP-43 aggregates/inclusions do not completely overlap with TDP-43 associated SGs, rather a subpopulation of those TDP-43 aggregates enter into SGs, and ALS-pathology-linked TDP-43 inclusion bodies are devoid of SGs. Notably, SGs indirectly exhibit positive feedback regulation of TDP-43 aggregation by disrupting HDAC6-mediated pathogenic TDP-43 clearance from ALS neurons and thereby accelerating TDP-43’s cytosolic aggregation [109].

2.3 Novel roles of GTPases: Rab11 and RGNEF in neurodegeneration

A common feature in a number of neurodegenerative diseases, including ALS, Alzheimer’s, and Parkinson’s disease, is the impaired clearance and recycling of damaged and aggregated proteins from cells. Moreover, perturbation of physiological vesicle trafficking systems affects several vital mechanisms in the cells, such as efficient nutrient absorption, failure of cell-to-cell communication, the immune response, and loss of synaptic transmission [110, 111]. Rab-GTPases, first discovered in brain tissue, belong to the major subset of the Ras superfamily [112]. In neurons, Rab-GTPases primarily orchestrate vesicle sorting and trafficking between target membranes through their interactions with effector proteins (coat proteins, kinesins, and dyneins), in addition to tethering and SNARE complexes [113, 114]. Among the Rab-GTPases, Rab11 plays critical roles in trafficking, sorting, and recycling endosomal vesicles around the perinuclear region. Rab11 is transported to the cellular periphery via recycling vesicles traveling along the microtubules and directly regulates vesicular exocytosis at the plasma membrane (Figure 4) [115, 116]. Furthermore, Rab11 has been identified as the master regulator for the transport of neurotrophin receptors and β-integrin via axonal junctions in dorsal root ganglion neurons and is critical for their maturation, functionality, and survival [117]. Rab11 participates in different cell survival pathways in neurodegenerative diseases in response prion/prion-like protein toxicity. In the ALS-TDP-43 Drosophila model, TDP-43 toxicity reduces levels of the synaptic growth promoting bone morphogenetic protein (BMP) at the neuromuscular junction and increases BMP receptors in recycling endosomes and at the neuromuscular junction. This pathogenic condition leads to larval crawling defects in ALS fruit flies, which are rescued by the overexpression of Rab11 [118]. Schwenk et al. has shown that TDP-43 regulates the number of Rab11-positive recycling endosomes in dendrites (Figure 4) [119]. These recycling defects in turn affect transferrin recycling in neurons and its decrease in cerebrospinal fluid of patients. In Drosophila, a Rab11 mutation induces JNK and MAPK/ERK signaling activation and apoptosis, resulting in defective eye and wing phenotypes [120, 121]. TDP-43 can be phosphorylated at threonine 153 and tyrosine 155 by MEK kinase, a central player in MAPK/ERK signaling pathway, in response to heat shock [122]. However, this unique phospho-TDP-43 variant is not involved in aggregate formation but is instead recruited to nucleoli for processing nucleolar-associated RNA. Further supporting the Drosophila model results, we have observed that sporadic ALS-TDP-43 mutations induce the loss of Rab11 in patients’ brain and spinal cord compared to age-matched control samples [123]. In contrast to MAPK/ERK and Akt signaling pathways operating in parallel to activate mTORC signaling [124], we have shown that Akt and ERK signaling pathways work in a competitive manner to determine the cell fate. Moreover, in Parkinson’s disease, Rab11 co-localizes with toxic α-synuclein inclusion bodies in dopaminergic neurons, and overexpression of Rab11 prevents the affected neurons from degeneration and rescues behavioral deficits [125]. In Huntington’s disease, Rab11 overexpression restores synaptic dysfunction and prevents glutamate-induced cell death in neurons [126, 127]. Rab11 has also been found to co-localize with Rab7 and C9orf72 in postmortem ALS brain samples and primary cortical neurons exhibiting C9orf72-induced disrupted vesicular trafficking system in ALS [128]. These findings suggest a broader impact of TDP-43 mutation-induced Rab11 dysfunction on cellular function and survival signaling cascades.

Figure 4.

TDP-43 modulates synaptic vesicle trafficking. Synaptic vesicle trafficking is a crucial mechanism for transporting macromolecules and neurotransmitters across the neuronal axon junctions. Defect in this mechanism may lead to accumulation of toxic waste products in the cells and activation of neurodegenerative conditions. Ras-GTPases like Rab11 plays a critical role in endosomal vesicle trafficking, sorting, and recycling. A study with sporadic ALS patients’ brain and spinal cord samples show loss of Rab11 could be closely linked to TDP-43 proteinopathies in ALS and FTD. Loss of Rab11 affects the cargo load capacity and the size of the vesicles as well. This condition in turn activates JNK and MAPK/ERK signaling cascades leading to neuronal apoptosis.

Apart from the Rab-GTPases of Ras superfamily, small RhoA GTPases also regulate broad spectrum of cellular mechanisms including cell-to-cell communication, migration, and proliferation. Given that the mode of GTPase functionality relies on the rate of GTP to GDP turn over and vice versa, dysregulation of guanine nucleotide exchange factor (GNEF) that is responsible for such turnover has been linked to a number of diseases including neurodegeneration. Among this class of proteins with enzymatic activity, Rho GNEF (RGNEF/p190) is directly associated with neurodegenerative disease like ALS, where it not only regulates the function of GTPases for stress survival [129] but also acts as the RNA-binding protein to modulate the stability of the crucial low molecular weight cytoskeleton protein neurofilament’s mRNA by binding to its 3′ untranslated region [130]. Emerging studies on GNEF’s involvement in ALS has revealed that RGNEF co-exists with TDP-43 and FUS in the neuronal cytoplasmic inclusion bodies in spinal motor neurons indicating a cross talk of protein aggregation and dysregulated cell signaling pathways in the ALS pathogenesis [131]. Micronuclei structures, containing small DNA fragments with clustered DSBs and surrounded by one lipid layer, are a strong hallmark of cellular metabolic stress, exposure to genotoxic agents, and genomic instability leading to apoptosis [132, 133, 134]. Notably, micronuclei have been observed in the ALS patient brain and spinal cord tissue samples. Studies have shown that TDP-43 interacts with RGNEF through its leucine-rich domain and forms co-aggregated structures [135]. Taken together, these findings further support the fact that genomic instability is one of the major outcomes in ALS-TDP-43 pathology leading to neuronal death.

Advertisement

3. DNA binding and role in DNA transactions

TDP-43 was first identified and characterized as the transcriptional regulator of HIV-1 gene expression by binding to long terminal repeat TAR DNA motifs [136]. In contrast, a separate study found that when exposed to HIV-1 infection, HIV-1 viral production could occur in T cells and macrophages, even in the absence of TDP-43 protein [137]. As an RNA-/DNA-binding protein, TDP-43 also interacts with specific TG and non-TG repeat containing DNA sequences through its RRM domains, with each domain having specific interaction affinities and DNA conformation requirements [138]. TDP-43 has been found to regulate the cyclin-dependent kinase 6 (CDK6) transcript and protein levels through its binding to the highly conserved long-stretch of GT repeats in CDK6 gene sequence. This binding leads to CDK6 upregulation and thereby increases phosphorylation of retinoblastoma proteins pRb and pRb2/p130 [139]. TDP-43 also acts as the transcriptional repressor and/or insulation regulator for the spatiotemporal regulation of the ACRV1 (SP-10) gene [140, 141]. TDP-43 binds to two GTGTGT motifs in the promoter core region through its N-terminal RRM1 domain during spermatogenesis. In vivo studies reveal that, unlike the wild-type variant, mutations in the GTGTGT motifs in the −186/+28 promoter region leads to premature reporter gene expression in the meiotic spermatocytes [142]. Previously, TDP-43 has been shown to bind both double-stranded DNA as well as single-stranded DNA, with a higher affinity toward single-stranded DNA through binding its RRM1 domain [143, 144]. Qin et al. reported that TDP-43’s N-terminal domain is indispensable for its physiological and proteinopathy functions and showed that the N-terminus maintains a highly ordered structure that equilibrates the C-terminal disordered structure by acquiring a novel ubiquitin-like fold that directly binds single-strand DNA [145]. More recently, we also documented in vitro studies on TDP-43’s affinity for binding free double-stranded DNA ends, instead of binding a partially or completely blocked terminus [146].

Advertisement

4. TDP-43 in DNA damage response (DDR) and repair

Induction of DNA damage and dysregulated damage response are critical factors for neuronal death in ALS and other neurodegenerative diseases [147, 148]. Studies also suggest endogenous DNA breaks, including DSBs, are routinely generated and repaired in healthy neurons and are essential for the regulation of neuronal gene expression [149, 150]. Besides the transcriptional regulation by RRM domains of TDP-43, no other DNA interactions have been reported for TDP-43. Based on an interactome study targeting TDP-43 in human cells, Freibaum et al. identified the DNA repair protein, Ku70, as one of the interacting partners of TDP-43 [151]. Furthermore, we found that TDP-43’s interaction with Ku70 was modulated by DNA damage induction in neuronal cells. Notably, TDP-43 showed pathway-specific roles in DNA DSB repair via direct interaction with the classical nonhomologous end joining (NHEJ) factors XRCC4 and DNA Ligase 4 (LIG4) complex, but not with single-strand break repair factors XRCC1 and DNA Ligase 3 (LIG3) complex. Further experimental studies revealed TDP-43’s interaction with a number of NHEJ proteins, including DNA-PKcs, 53BP1, polymerase lambda, XRCC4-like factor (XLF), and the DNA damage response (DDR) factors phosphorylated ATM and histone H2AX (γH2AX). Interestingly, TDP-43 depletion in neurons elicited hyperactivation of DDR signaling without affecting the recruitment of activated DDR proteins to genome damage sites and inhibited the docking, but not assembly, of DNA ligation complex factors (XRCC4, LIG4, and XLF) at the break sites. This suggests that TDP-43 has a crucial role in maintenance of genomic integrity by efficiently completing the rate-limiting DSB sealing step (Figure 5). Consistent with these results, neuronal cells with TDP-43 downregulation exhibited delayed DSB repair kinetics and a higher population of apoptotic cells due to the persistent accumulation of unrepaired DSBs than controls. In a correlative study of postmortem human sporadic ALS brain and cervical spinal cord samples, we determined that samples with extensive TDP-43 aggregation and/or fragmentation had greater staining for DSB markers (γH2AX and TUNEL) than their age-matched controls [146].

Figure 5.

TDP-43’s scaffolding role in NHEJ repair mechanism of nuclear genome. In response to DNA damage induction, TDP-43 gets recruited at the DSB sites and participates in relaying DDR signaling through interaction with p-ATM (Serine1981), p-histone H2AX (Serine139), p-53BP1 (Serine1778), and Ku70/80 heterodimer. More importantly, TDP-43 regulates the most rate-limiting step of the DNA DSB repair pathway—DNA DSB end ligation, through its interaction with DNA ligation complex XRCC4-ligase 4 at the break sites. However, pathogenic mutations in TDP-43 related to ALS and FTD cause enhanced cytosolic mislocalization of TDP-43, thereby inhibiting the DNA damage-induced translocation of XRCC4-ligase 4 complex from cytosol to nucleus. Either of TDP-43 loss-of-function or ligation complex translocation inhibition causes DNA DSB repair impairment leading to nuclear genome instability in ALS/FTD motor neurons.

4.1 Mutant TDP-43 and defective DSB repair in neurons

Efficient sealing of DNA DSBs, which are the most lethal form of DNA damage, is critical for maintaining genome integrity and fidelity. In the majority of the SALS cases, the total amount of TDP-43 does not change significantly, but a significant proportion of the protein mislocalizes to the cytosol and causes ALS/FTD pathophysiology similar to disease-linked mutant TDP-43. We recently linked the TDP-43 Q331K mutation with genome instability and DSB repair defects. In studies with conditionally expressed TDP-43 Q331K mutant, cultured neurons exhibited a strong nuclear clearance phenotype and a higher amount of DNA damage at basal level without the effect of any external DNA damaging agents than control cells. Further investigations showed that TDP-43 Q331K trapped XRCC4 and LIG4 in the cytosol and inhibited their translocation to the nucleus in response to DNA damage induction. These findings suggest that TDP-43 is not only involved in the recruitment of the DNA ligation complex at genomic break sites but also regulates the damage-dependent nuclear translocation of DNA repair proteins (Figure 5). These impaired TDP-43 functions in the disease condition leading to genomic instability and neurodegeneration [22].

Advertisement

5. A double whammy of DNA damage induction and defects in their repair in TDP-43-ALS

We and others have observed that the most salient hallmarks of ALS/FTD-TDP-43 pathology, the protein aggregation and inclusion body formation in the cytosol, increase oxidative stress in affected neurons [22, 48, 152, 153]. 4-hydroxynonenal (HNE) increases as an oxidative stress indicator in sporadic ALS patients’ brain, spinal cord, and serum. HNE induces oxidative damage to a broad range of cysteine-containing proteins, including TDP-43, through cysteine oxidation. Oxidized TDP-43 mislocalizes from the nucleus to form insoluble cytosolic aggregates in ALS [154]. Such nuclear clearance and cytosolic increase may cause a gain-of-toxicity, leading to aberrant mRNA processing, which in turn exerts a direct DNA destabilization through impaired DSB repair and an indirect repair inhibition by negatively regulating mRNA splicing of DNA repair proteins. Furthermore, the unrepaired DNA DSBs leads to persistent DDR signaling activation through an ATM-mediated signaling cascade that promotes neuroinflammation. This inflammatory response may induce oxidative genome damage and enhanced TDP-43’s nuclear clearance, exacerbating the ALS/FTD conditions in a feed forward pattern. Moreover, dysregulation of metal homeostasis has been observed in ALS patients’ brain/spinal cord tissues. In the presence of cellular oxidative stress, zinc induces TDP-43’s mislocalization and cytoplasmic inclusions [155, 156]. Given that zinc is a crucial metal co-activator for several transcription factors, trapping of zinc in TDP-43 aggregates could globally affect gene activation patterns, including those for DNA repair and response-associated factors, leading to genomic instability.

Advertisement

6. Conclusions: TDP-43-ALS, a case for DNA repair-targeted therapy?

Emerging studies from our laboratory and other groups have shown that in addition to its RNA processing and miRNA biogenesis functions, TDP-43 acts as a key component of the NHEJ pathway for DSB repair in neurons, and its pathological clearance from the nucleus leads to the DSB repair defects seen in ALS and other TDP-43-linked neurodegenerative diseases. These newly discovered paradigms link TDP-43 pathology to impaired DNA repair and suggest potential avenues for DNA repair-targeted therapies for TDP-43-ALS and related motor neuron diseases. Future studies should focus on a comprehensive delineation of the molecular mechanisms involved in order to develop efficiently targeted interventions. In addition, the implications of these defects in neuronal and glial functions in the CNS of TDP-43-ALS patients and the role of TDP-43 in maintaining genome integrity in non-neuronal brain cells, including glia and astrocytes, are important lines of investigation. By comparing the DNA repair role of TDP-43 in post-mitotic vs. cycling cells, we could learn important mechanistic insights on the selective vulnerability of neurons in ALS.

Recently, an increasing number of studies have demonstrated the role of RNA/DNA-binding proteins in DNA repair. We previously documented the involvement of hnRNP-U, a member of the hnRNP family, in oxidative damage repair in the human genome and its role as a molecular switch between DSB repair and oxidative damage repair when these complex damages occur in a clustered fashion [157, 158]. We also discovered that another RNA-/DNA-binding protein, fused in sarcoma/translocated in liposarcoma (FUS/TLS), linked to the ALS-FUS subtype, participates in break sealing during DNA SSB repair [10]. Because both TDP-43 and FUS influence the final DNA break/sealing step of repair, further investigations in DNA repair mechanisms are critical to developing clinically effective strategies for ameliorating the genome instability of ALS-TDP-43.

Advertisement

Acknowledgments

Research in Hegde laboratory is supported by grants from the National Institute of Neurological Disorders and Stroke (NINDS) and National Institute of Aging (NIA) of the National Institute of Health (NIH) under the award numbers R01NS088645, RF1NS112719, and R03AG064266 and the Houston Methodist Research Institute funds. The content is solely the responsibility of the authors and does not necessarily represent the official views of the funding agencies. The authors thank members of the Hegde Laboratory—Haibo Wang, Manohar Kodavati, Pavana Hegde, Velmarini Vasquez, and Erika Guerrero for assistance.

References

  1. 1. Neumann M et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314(5796):130-133
  2. 2. Arai T et al. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochemical and Biophysical Research Communications. 2006;351(3):602-611
  3. 3. Maekawa S et al. TDP-43 is consistently co-localized with ubiquitinated inclusions in sporadic and Guam amyotrophic lateral sclerosis but not in familial amyotrophic lateral sclerosis with and without SOD1 mutations. Neuropathology. 2009;29(6):672-683
  4. 4. Al-Sarraj S et al. 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 Neuropathologica. 2011;122(6):691-702
  5. 5. DeJesus-Hernandez M et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 2011;72(2):245-256
  6. 6. Chew J et al. Neurodegeneration. C9ORF72 repeat expansions in mice cause TDP-43 pathology, neuronal loss, and behavioral deficits. Science. 2015;348(6239):1151-1154
  7. 7. Gitcho MA et al. VCP mutations causing frontotemporal lobar degeneration disrupt localization of TDP-43 and induce cell death. The Journal of Biological Chemistry. 2009;284(18):12384-12398
  8. 8. Liu G et al. Cdc48/VCP and endocytosis regulate TDP-43 and FUS toxicity and turnover. Molecular and Cellular Biology. 2020:40(4):e00256-19
  9. 9. Neumann M et al. TDP-43 in the ubiquitin pathology of frontotemporal dementia with VCP gene mutations. Journal of Neuropathology and Experimental Neurology. 2007;66(2):152-157
  10. 10. Wang H et al. Mutant FUS causes DNA ligation defects to inhibit oxidative damage repair in amyotrophic lateral sclerosis. Nature Communications. 2018;9(1):3683
  11. 11. Hortobagyi T et al. Optineurin inclusions occur in a minority of TDP-43 positive ALS and FTLD-TDP cases and are rarely observed in other neurodegenerative disorders. Acta Neuropathologica. 2011;121(4):519-527
  12. 12. Deng HX et al. Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia. Nature. 2011;477(7363):211-215
  13. 13. Williams KL et al. UBQLN2/ubiquilin 2 mutation and pathology in familial amyotrophic lateral sclerosis. Neurobiology of Aging. 2012;33(10):2527.e3-2527.10
  14. 14. Elden AC et al. Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS. Nature. 2010;466(7310):1069-1075
  15. 15. Farg MA et al. Ataxin-2 interacts with FUS and intermediate-length polyglutamine expansions enhance FUS-related pathology in amyotrophic lateral sclerosis. Human Molecular Genetics. 2013;22(4):717-728
  16. 16. Henry KA et al. Geographic variation of amyotrophic lateral sclerosis incidence in New Jersey, 2009-2011. American Journal of Epidemiology. 2015;182(6):512-519
  17. 17. Mehta P et al. Prevalence of amyotrophic lateral sclerosis - United States, 2015. MMWR. Morbidity and Mortality Weekly Report. 2018;67(46):1285-1289
  18. 18. Byrne S et al. Rate of familial amyotrophic lateral sclerosis: A systematic review and meta-analysis. Journal of Neurology, Neurosurgery, and Psychiatry. 2011;82(6):623-627
  19. 19. Chio A et al. Prognostic factors in ALS: A critical review. Amyotrophic Lateral Sclerosis. 2009;10(5-6):310-323
  20. 20. Buratti E. Functional significance of TDP-43 mutations in disease. Advances in Genetics. 2015;91:1-53
  21. 21. Kuhnlein P et al. Two German kindreds with familial amyotrophic lateral sclerosis due to TARDBP mutations. Archives of Neurology. 2008;65(9):1185-1189
  22. 22. Guerrero EN et al. Amyotrophic lateral sclerosis-associated TDP-43 mutation Q331K prevents nuclear translocation of XRCC4-DNA ligase 4 complex and is linked to genome damage-mediated neuronal apoptosis. Human Molecular Genetics. 2019;28(5):2459-2476
  23. 23. Rutherford NJ et al. Novel mutations in TARDBP (TDP-43) in patients with familial amyotrophic lateral sclerosis. PLoS Genetics. 2008;4(9):e1000193
  24. 24. Gitcho MA et al. TDP-43 A315T mutation in familial motor neuron disease. Annals of Neurology. 2008;63(4):535-538
  25. 25. Sreedharan J et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science. 2008;319(5870):1668-1672
  26. 26. Van Deerlin VM et al. TARDBP mutations in amyotrophic lateral sclerosis with TDP-43 neuropathology: A genetic and histopathological analysis. Lancet Neurology. 2008;7(5):409-416
  27. 27. Kabashi E et al. TARDBP mutations in individuals with sporadic and familial amyotrophic lateral sclerosis. Nature Genetics. 2008;40(5):572-574
  28. 28. Ye CH et al. Absence of mutations in exon 6 of the TARDBP gene in 207 Chinese patients with sporadic amyotrohic lateral sclerosis. PLoS One. 2013;8(7):e68106
  29. 29. Fang F et al. Association between blood lead and the risk of amyotrophic lateral sclerosis. American Journal of Epidemiology. 2010;171(10):1126-1133
  30. 30. Campbell AM, Williams ER, Barltrop D. Motor neurone disease and exposure to lead. Journal of Neurology, Neurosurgery, and Psychiatry. 1970;33(6):877-885
  31. 31. Gresham LS et al. Amyotrophic lateral sclerosis and occupational heavy metal exposure: A case-control study. Neuroepidemiology. 1986;5(1):29-38
  32. 32. Wang H et al. Smoking and risk of amyotrophic lateral sclerosis: A pooled analysis of 5 prospective cohorts. Archives of Neurology. 2011;68(2):207-213
  33. 33. Gallo V et al. Smoking and risk for amyotrophic lateral sclerosis: Analysis of the EPIC cohort. Annals of Neurology. 2009;65(4):378-385
  34. 34. Bonvicini F et al. Exposure to pesticides and risk of amyotrophic lateral sclerosis: A population-based case-control study. Annali dell’Istituto Superiore di Sanità. 2010;46(3):284-287
  35. 35. Su FC et al. Association of environmental toxins with amyotrophic lateral sclerosis. JAMA Neurology. 2016;73(7):803-811
  36. 36. Malek AM et al. Exposure to hazardous air pollutants and the risk of amyotrophic lateral sclerosis. Environmental Pollution. 2015;197:181-186
  37. 37. Yu Y et al. Environmental risk factors and amyotrophic lateral sclerosis (ALS): A case-control study of ALS in Michigan. PLoS One. 2014;9(6):e101186
  38. 38. de Jong SW et al. Smoking, alcohol consumption, and the risk of amyotrophic lateral sclerosis: A population-based study. American Journal of Epidemiology. 2012;176(3):233-239
  39. 39. Haley RW. Excess incidence of ALS in young gulf war veterans. Neurology. 2003;61(6):750-756
  40. 40. Weisskopf MG et al. Prospective study of military service and mortality from ALS. Neurology. 2005;64(1):32-37
  41. 41. Chen H et al. Head injury and amyotrophic lateral sclerosis. American Journal of Epidemiology. 2007;166(7):810-816
  42. 42. Schmidt S et al. Association of ALS with head injury, cigarette smoking and APOE genotypes. Journal of the Neurological Sciences. 2010;291(1-2):22-29
  43. 43. Cox PA, Sacks OW. Cycad neurotoxins, consumption of flying foxes, and ALS-PDC disease in Guam. Neurology. 2002;58(6):956-959
  44. 44. Garruto RM, Gajdusek C, Chen KM. Amyotrophic lateral sclerosis among Chamorro migrants from Guam. Annals of Neurology. 1980;8(6):612-619
  45. 45. Arif M et al. Tau pathology involves protein phosphatase 2A in parkinsonism-dementia of Guam. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(3):1144-1149
  46. 46. Weiss JH, Koh JY, Choi DW. Neurotoxicity of beta-N-methylamino-L-alanine (BMAA) and beta-N-oxalylamino-L-alanine (BOAA) on cultured cortical neurons. Brain Research. 1989;497(1):64-71
  47. 47. Yin HZ et al. Intrathecal infusion of BMAA induces selective motor neuron damage and astrogliosis in the ventral horn of the spinal cord. Experimental Neurology. 2014;261:1-9
  48. 48. Guerrero EN et al. TDP-43/FUS in motor neuron disease: Complexity and challenges. Progress in Neurobiology. 2016;145-146:78-97
  49. 49. Sieh W et al. Identification of novel susceptibility loci for Guam neurodegenerative disease: Challenges of genome scans in genetic isolates. Human Molecular Genetics. 2009;18(19):3725-3738
  50. 50. Li Q et al. The cleavage pattern of TDP-43 determines its rate of clearance and cytotoxicity. Nature Communications. 2015;6:6183
  51. 51. Asakawa K, Handa H, Kawakami K. Optogenetic modulation of TDP-43 oligomerization accelerates ALS-related pathologies in the spinal motor neurons. Nature Communications. 2020;11(1):1004
  52. 52. Zhang YJ et al. Aberrant cleavage of TDP-43 enhances aggregation and cellular toxicity. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(18):7607-7612
  53. 53. Shiina Y et al. TDP-43 dimerizes in human cells in culture. Cellular and Molecular Neurobiology. 2010;30(4):641-652
  54. 54. Hergesheimer RC et al. The debated toxic role of aggregated TDP-43 in amyotrophic lateral sclerosis: A resolution in sight? Brain. 2019;142(5):1176-1194
  55. 55. Winton MJ et al. Disturbance of nuclear and cytoplasmic TAR DNA-binding protein (TDP-43) induces disease-like redistribution, sequestration, and aggregate formation. The Journal of Biological Chemistry. 2008;283(19):13302-13309
  56. 56. Shodai A et al. Conserved acidic amino acid residues in a second RNA recognition motif regulate assembly and function of TDP-43. PLoS One. 2012;7(12):e52776
  57. 57. Amlie-Wolf A et al. Transcriptomic changes due to cytoplasmic TDP-43 expression reveal Dysregulation of histone transcripts and nuclear chromatin. PLoS One. 2015;10(10):e0141836
  58. 58. Winton MJ et al. A90V TDP-43 variant results in the aberrant localization of TDP-43 in vitro. FEBS Letters. 2008;582(15):2252-2256
  59. 59. Barmada SJ et al. Cytoplasmic mislocalization of TDP-43 is toxic to neurons and enhanced by a mutation associated with familial amyotrophic lateral sclerosis. The Journal of Neuroscience. 2010;30(2):639-649
  60. 60. Jara JH et al. Evidence for an early innate immune response in the motor cortex of ALS. Journal of Neuroinflammation. 2017;14(1):129
  61. 61. Jara JH et al. MCP1-CCR2 and neuroinflammation in the ALS motor cortex with TDP-43 pathology. Journal of Neuroinflammation. 2019;16(1):196
  62. 62. Beers DR et al. Neuroinflammation modulates distinct regional and temporal clinical responses in ALS mice. Brain, Behavior, and Immunity. 2011;25(5):1025-1035
  63. 63. Puentes F et al. Non-neuronal cells in ALS: Role of glial, immune cells and blood-CNS barriers. Brain Pathology. 2016;26(2):248-257
  64. 64. Zhu J, Cynader MS, Jia W. TDP-43 inhibits NF-kappaB activity by blocking p65 nuclear translocation. PLoS One. 2015;10(11):e0142296
  65. 65. Frakes AE et al. Microglia induce motor neuron death via the classical NF-kappaB pathway in amyotrophic lateral sclerosis. Neuron. 2014;81(5):1009-1023
  66. 66. Correia AS et al. Inflammation induces TDP-43 Mislocalization and aggregation. PLoS One. 2015;10(10):e0140248
  67. 67. Swarup V et al. Deregulation of TDP-43 in amyotrophic lateral sclerosis triggers nuclear factor kappaB-mediated pathogenic pathways. The Journal of Experimental Medicine. 2011;208(12):2429-2447
  68. 68. Buratti E, Baralle FE. Multiple roles of TDP-43 in gene expression, splicing regulation, and human disease. Frontiers in Bioscience. 2008;13:867-878
  69. 69. Buratti E, Baralle FE. TDP-43: Gumming up neurons through protein-protein and protein-RNA interactions. Trends in Biochemical Sciences. 2012;37(6):237-247
  70. 70. Avendano-Vazquez SE et al. Autoregulation of TDP-43 mRNA levels involves interplay between transcription, splicing, and alternative polyA site selection. Genes & Development. 2012;26(15):1679-1684
  71. 71. Bembich S et al. Predominance of spliceosomal complex formation over polyadenylation site selection in TDP-43 autoregulation. Nucleic Acids Research. 2014;42(5):3362-3371
  72. 72. Pons M et al. Identification of TCERG1 as a new genetic modulator of TDP-43 production in drosophila. Acta Neuropathologica Communications. 2018;6(1):138
  73. 73. Maris C, Dominguez C, Allain FH. The RNA recognition motif, a plastic RNA-binding platform to regulate post-transcriptional gene expression. The FEBS Journal. 2005;272(9):2118-2131
  74. 74. Buratti E, Baralle FE. Characterization and functional implications of the RNA binding properties of nuclear factor TDP-43, a novel splicing regulator of CFTR exon 9. The Journal of Biological Chemistry. 2001;276(39):36337-36343
  75. 75. Kuo PH et al. Structural insights into TDP-43 in nucleic-acid binding and domain interactions. Nucleic Acids Research. 2009;37(6):1799-1808
  76. 76. Ayala YM et al. Human, drosophila, and C.elegans TDP43: Nucleic acid binding properties and splicing regulatory function. Journal of Molecular Biology. 2005;348(3):575-588
  77. 77. Donde A et al. Splicing repression is a major function of TDP-43 in motor neurons. Acta Neuropathologica. 2019;138(5):813-826
  78. 78. Polymenidou M et al. Long pre-mRNA depletion and RNA missplicing contribute to neuronal vulnerability from loss of TDP-43. Nature Neuroscience. 2011;14(4):459-468
  79. 79. Watanabe S et al. ALS-linked TDP-43(M337V) knock-in mice exhibit splicing deregulation without neurodegeneration. Molecular Brain. 2020;13(1):8
  80. 80. Wang HY et al. Structural diversity and functional implications of the eukaryotic TDP gene family. Genomics. 2004;83(1):130-139
  81. 81. Dreyfuss G et al. hnRNP proteins and the biogenesis of mRNA. Annual Review of Biochemistry. 1993;62:289-321
  82. 82. Buratti E et al. TDP-43 binds heterogeneous nuclear ribonucleoprotein a/B through its C-terminal tail: An important region for the inhibition of cystic fibrosis transmembrane conductance regulator exon 9 splicing. The Journal of Biological Chemistry. 2005;280(45):37572-37584
  83. 83. Kashima T, Manley JL. A negative element in SMN2 exon 7 inhibits splicing in spinal muscular atrophy. Nature Genetics. 2003;34(4):460-463
  84. 84. Buvoli M et al. Alternative splicing in the human gene for the core protein A1 generates another hnRNP protein. The EMBO Journal. 1990;9(4):1229-1235
  85. 85. Bekenstein U, Soreq H. Heterogeneous nuclear ribonucleoprotein A1 in health and neurodegenerative disease: From structural insights to post-transcriptional regulatory roles. Molecular and Cellular Neurosciences. 2013;56:436-446
  86. 86. Deshaies JE et al. TDP-43 regulates the alternative splicing of hnRNP A1 to yield an aggregation-prone variant in amyotrophic lateral sclerosis. Brain. 2018;141(5):1320-1333
  87. 87. Honda H et al. Loss of hnRNPA1 in ALS spinal cord motor neurons with TDP-43-positive inclusions. Neuropathology. 2015;35(1):37-43
  88. 88. Jiang LL et al. The N-terminal dimerization is required for TDP-43 splicing activity. Scientific Reports. 2017;7(1):6196
  89. 89. Mompean M et al. Point mutations in the N-terminal domain of transactive response DNA-binding protein 43 kDa (TDP-43) compromise its stability, dimerization, and functions. The Journal of Biological Chemistry. 2017;292(28):11992-12006
  90. 90. Chen HJ et al. RRM adjacent TARDBP mutations disrupt RNA binding and enhance TDP-43 proteinopathy. Brain. 2019;142(12):3753-3770
  91. 91. French RL et al. Detection of TAR DNA-binding protein 43 (TDP-43) oligomers as initial intermediate species during aggregate formation. The Journal of Biological Chemistry. 2019;294(17):6696-6709
  92. 92. Kawahara Y, Mieda-Sato A. TDP-43 promotes microRNA biogenesis as a component of the Drosha and dicer complexes. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(9):3347-3352
  93. 93. Fan Z, Chen X, Chen R. Transcriptome-wide analysis of TDP-43 binding small RNAs identifies miR-NID1 (miR-8485), a novel miRNA that represses NRXN1 expression. Genomics. 2014;103(1):76-82
  94. 94. Tollervey JR et al. Characterizing the RNA targets and position-dependent splicing regulation by TDP-43. Nature Neuroscience. 2011;14(4):452-458
  95. 95. Beltran M, Garcia de Herreros A. Antisense non-coding RNAs and regulation of gene transcription. Transcription. 2016;7(2):39-43
  96. 96. Liu X et al. Long non-coding RNA gadd7 interacts with TDP-43 and regulates Cdk6 mRNA decay. The EMBO Journal. 2012;31(23):4415-4427
  97. 97. Anderson P, Kedersha N. Stress granules: The Tao of RNA triage. Trends in Biochemical Sciences. 2008;33(3):141-150
  98. 98. Guil S, Long JC, Caceres JF. hnRNP A1 relocalization to the stress granules reflects a role in the stress response. Molecular and Cellular Biology. 2006;26(15):5744-5758
  99. 99. Dewey CM et al. TDP-43 is directed to stress granules by sorbitol, a novel physiological osmotic and oxidative stressor. Molecular and Cellular Biology. 2011;31(5):1098-1108
  100. 100. Aulas A, Stabile S, Vande Velde C. Endogenous TDP-43, but not FUS, contributes to stress granule assembly via G3BP. Molecular Neurodegeneration. 2012;7:54
  101. 101. Mackenzie IR et al. TIA1 mutations in amyotrophic lateral sclerosis and Frontotemporal dementia promote phase separation and Alter stress granule dynamics. Neuron. 2017;95(4):808-816.e9
  102. 102. Khalfallah Y et al. TDP-43 regulation of stress granule dynamics in neurodegenerative disease-relevant cell types. Scientific Reports. 2018;8(1):7551
  103. 103. Aulas A et al. G3BP1 promotes stress-induced RNA granule interactions to preserve polyadenylated mRNA. The Journal of Cell Biology. 2015;209(1):73-84
  104. 104. McDonald KK et al. TAR DNA-binding protein 43 (TDP-43) regulates stress granule dynamics via differential regulation of G3BP and TIA-1. Human Molecular Genetics. 2011;20(7):1400-1410
  105. 105. Orru S et al. Reduced stress granule formation and cell death in fibroblasts with the A382T mutation of TARDBP gene: Evidence for loss of TDP-43 nuclear function. Human Molecular Genetics. 2016;25(20):4473-4483
  106. 106. Gasset-Rosa F et al. Cytoplasmic TDP-43 De-mixing independent of stress granules drives inhibition of nuclear import, loss of nuclear TDP-43, and cell death. Neuron. 2019;102(2):339-357.e7
  107. 107. Hans F, Glasebach H, Kahle PJ. Multiple distinct pathways lead to hyperubiquitylated insoluble TDP-43 protein independent of its translocation into stress granules. The Journal of Biological Chemistry. Jan 17 2020;295(3):673-689
  108. 108. Fang MY et al. Small-molecule modulation of TDP-43 recruitment to stress granules prevents persistent TDP-43 accumulation in ALS/FTD. Neuron. 2019;103(5):802-819.e11
  109. 109. Chen Y, Cohen TJ. Aggregation of the nucleic acid-binding protein TDP-43 occurs via distinct routes that are coordinated with stress granule formation. The Journal of Biological Chemistry. 2019;294(10):3696-3706
  110. 110. Vagnozzi AN, Pratico D. Endosomal sorting and trafficking, the retromer complex and neurodegeneration. Molecular Psychiatry. 2019;24(6):857-868
  111. 111. Bouchet J et al. Rab11-FIP3 regulation of Lck Endosomal traffic controls TCR signal transduction. Journal of Immunology. 2017;198(7):2967-2978
  112. 112. Touchot N, Chardin P, Tavitian A. Four additional members of the ras gene superfamily isolated by an oligonucleotide strategy: Molecular cloning of YPT-related cDNAs from a rat brain library. Proceedings of the National Academy of Sciences of the United States of America. 1987;84(23):8210-8214
  113. 113. Cai H, Reinisch K, Ferro-Novick S. Coats, tethers, Rabs, and SNAREs work together to mediate the intracellular destination of a transport vesicle. Developmental Cell. 2007;12(5):671-682
  114. 114. Grosshans BL, Ortiz D, Novick P. Rabs and their effectors: Achieving specificity in membrane traffic. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(32):11821-11827
  115. 115. Takahashi S et al. Rab11 regulates exocytosis of recycling vesicles at the plasma membrane. Journal of Cell Science. 2012;125(Pt 17):4049-4057
  116. 116. Goldenring JR. Recycling endosomes. Current Opinion in Cell Biology. 2015;35:117-122
  117. 117. Bucci C, Alifano P, Cogli L. The role of Rab proteins in neuronal cells and in the trafficking of neurotrophin receptors. Membranes. 2014;4(4):642-677
  118. 118. Deshpande M et al. Role of BMP receptor traffic in synaptic growth defects in an ALS model. Molecular Biology of the Cell. 2016;27(19):2898-2910
  119. 119. Schwenk BM et al. TDP-43 loss of function inhibits endosomal trafficking and alters trophic signaling in neurons. The EMBO Journal. 2016;35(21):2350-2370
  120. 120. Tiwari AK, Roy JK. Mutation in Rab11 results in abnormal organization of ommatidial cells and activation of JNK signaling in the drosophila eye. European Journal of Cell Biology. 2009;88(8):445-460
  121. 121. Bhuin T, Roy JK. Rab11 regulates JNK and Raf/MAPK-ERK signalling pathways during drosophila wing development. Cell Biology International. 2010;34(11):1113-1118
  122. 122. Li W et al. Heat shock-induced phosphorylation of TAR DNA-binding protein 43 (TDP-43) by MAPK/ERK kinase regulates TDP-43 function. The Journal of Biological Chemistry. 2017;292(12):5089-5100
  123. 123. Mitra J, Hegde PM, Hegde ML. Loss of endosomal recycling factor RAB11 coupled with complex regulation of MAPK/ERK/AKT signaling in postmortem spinal cord specimens of sporadic amyotrophic lateral sclerosis patients. Molecular Brain. 2019;12(1):55
  124. 124. Winter JN, Jefferson LS, Kimball SR. ERK and Akt signaling pathways function through parallel mechanisms to promote mTORC1 signaling. American Journal of Physiology. Cell Physiology. 2011;300(5):C1172-C1180
  125. 125. Breda C et al. Rab11 modulates alpha-synuclein-mediated defects in synaptic transmission and behaviour. Human Molecular Genetics. 2015;24(4):1077-1091
  126. 126. Steinert JR et al. Rab11 rescues synaptic dysfunction and behavioural deficits in a drosophila model of Huntington’s disease. Human Molecular Genetics. 2012;21(13):2912-2922
  127. 127. Li X et al. Disruption of Rab11 activity in a knock-in mouse model of Huntington’s disease. Neurobiology of Disease. 2009;36(2):374-383
  128. 128. Farg MA et al. C9ORF72, implicated in amytrophic lateral sclerosis and frontotemporal dementia, regulates endosomal trafficking. Human Molecular Genetics. 2017;26(20):4093-4094
  129. 129. Cheung K et al. Rho guanine nucleotide exchange factor (RGNEF) is a prosurvival factor under stress conditions. Molecular and Cellular Neurosciences. 2017;82:88-95
  130. 130. Droppelmann CA et al. Rho guanine nucleotide exchange factor is an NFL mRNA destabilizing factor that forms cytoplasmic inclusions in amyotrophic lateral sclerosis. Neurobiology of Aging. 2013;34(1):248-262
  131. 131. Keller BA et al. Co-aggregation of RNA binding proteins in ALS spinal motor neurons: Evidence of a common pathogenic mechanism. Acta Neuropathologica. 2012;124(5):733-747
  132. 132. Utani K et al. Emergence of micronuclei and their effects on the fate of cells under replication stress. PLoS One. 2010;5(4):e10089
  133. 133. Kiraly G et al. Micronucleus formation during chromatin condensation and under apoptotic conditions. Apoptosis. 2017;22(2):207-219
  134. 134. Xu B et al. Replication stress induces micronuclei comprising of aggregated DNA double-strand breaks. PLoS One. 2011;6(4):e18618
  135. 135. Droppelmann CA et al. TDP-43 aggregation inside micronuclei reveals a potential mechanism for protein inclusion formation in ALS. Scientific Reports. 2019;9(1):19928
  136. 136. Ou SH et al. Cloning and characterization of a novel cellular protein, TDP-43, that binds to human immunodeficiency virus type 1 TAR DNA sequence motifs. Journal of Virology. 1995;69(6):3584-3596
  137. 137. Nehls J et al. HIV-1 replication in human immune cells is independent of TAR DNA binding protein 43 (TDP-43) expression. PLoS One. 2014;9(8):e105478
  138. 138. Furukawa Y et al. A molecular mechanism realizing sequence-specific recognition of nucleic acids by TDP-43. Scientific Reports. 2016;6:20576
  139. 139. Ayala YM, Misteli T, Baralle FE. TDP-43 regulates retinoblastoma protein phosphorylation through the repression of cyclin-dependent kinase 6 expression. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(10):3785-3789
  140. 140. Lalmansingh AS, Urekar CJ, Reddi PP. TDP-43 is a transcriptional repressor: The testis-specific mouse acrv1 gene is a TDP-43 target in vivo. The Journal of Biological Chemistry. 2011;286(13):10970-10982
  141. 141. Abhyankar MM, Urekar C, Reddi PP. A novel CpG-free vertebrate insulator silences the testis-specific SP-10 gene in somatic tissues: Role for TDP-43 in insulator function. The Journal of Biological Chemistry. 2007;282(50):36143-36154
  142. 142. Acharya KK et al. Cis-requirement for the maintenance of round spermatid-specific transcription. Developmental Biology. 2006;295(2):781-790
  143. 143. Kitamura A et al. Analysis of the substrate recognition state of TDP-43 to single-stranded DNA using fluorescence correlation spectroscopy. Biochemistry and Biophysics Reports. 2018;14:58-63
  144. 144. Kuo PH et al. The crystal structure of TDP-43 RRM1-DNA complex reveals the specific recognition for UG- and TG-rich nucleic acids. Nucleic Acids Research. 2014;42(7):4712-4722
  145. 145. Qin H et al. TDP-43 N terminus encodes a novel ubiquitin-like fold and its unfolded form in equilibrium that can be shifted by binding to ssDNA. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(52):18619-18624
  146. 146. Mitra J et al. Motor neuron disease-associated loss of nuclear TDP-43 is linked to DNA double-strand break repair defects. Proceedings of the National Academy of Sciences of the United States of America. 2019;116(10):4696-4705
  147. 147. Vasquez V et al. Chromatin-bound oxidized alpha-Synuclein causes Strand breaks in neuronal genomes in in vitro models of Parkinson’s disease. Journal of Alzheimer’s Disease. 2017;60(s1):S133-s150
  148. 148. Hou Y et al. NAD(+) supplementation normalizes key Alzheimer’s features and DNA damage responses in a new AD mouse model with introduced DNA repair deficiency. Proceedings of the National Academy of Sciences of the United States of America. 2018;115(8):E1876-e1885
  149. 149. Madabhushi R et al. Activity-induced DNA breaks govern the expression of neuronal early-response genes. Cell. 2015;161(7):1592-1605
  150. 150. Su Y, Ming GL, Song H. DNA damage and repair regulate neuronal gene expression. Cell Research. 2015;25(9):993-994
  151. 151. Freibaum BD et al. Global analysis of TDP-43 interacting proteins reveals strong association with RNA splicing and translation machinery. Journal of Proteome Research. 2010;9(2):1104-1120
  152. 152. Nagase M et al. Increased oxidative stress in patients with amyotrophic lateral sclerosis and the effect of edaravone administration. Redox Report. 2016;21(3):104-112
  153. 153. Niedzielska E et al. Oxidative stress in neurodegenerative diseases. Molecular Neurobiology. 2016;53(6):4094-4125
  154. 154. Kabuta C et al. 4-Hydroxynonenal induces persistent insolubilization of TDP-43 and alters its intracellular localization. Biochemical and Biophysical Research Communications. 2015;463(1-2):82-87
  155. 155. Zinszner H, Albalat R, Ron D. A novel effector domain from the RNA-binding protein TLS or EWS is required for oncogenic transformation by CHOP. Genes & Development. 1994;8(21):2513-2526
  156. 156. Caragounis A et al. Zinc induces depletion and aggregation of endogenous TDP-43. Free Radical Biology & Medicine. 2010;48(9):1152-1161
  157. 157. Hegde ML et al. Scaffold attachment factor a (SAF-A) and Ku temporally regulate repair of radiation-induced clustered genome lesions. Oncotarget. 2016;7(34):54430-54444
  158. 158. Hegde ML et al. Enhancement of NEIL1 protein-initiated oxidized DNA base excision repair by heterogeneous nuclear ribonucleoprotein U (hnRNP-U) via direct interaction. The Journal of Biological Chemistry. 2012;287(41):34202-34211

Written By

Joy Mitra and Muralidhar L. Hegde

Submitted: 10 March 2020 Reviewed: 29 April 2020 Published: 03 June 2020