Loading

Review Article Open Access
Volume 5 | Issue 4

Protein Citrullination in Amyotrophic Lateral Sclerosis and Other Neurodegenerative Diseases

  • 1Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
  • 2Program in Chemical Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605 USA
+ Affiliations - Affiliations

*Corresponding Author

Zuoshang Xu, zuoshang.xu@umassmed.edu

Received Date: October 05, 2024

Accepted Date: November 13, 2024

Abstract

Protein citrullination (PC) is a posttranslational modification (PTM) that converts a peptidyl arginine into a peptidyl citrulline. Aberrant PC is a hallmark of neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), Alzheimer’s disease, Parkinson’s disease, prion disease, and multiple sclerosis. Common among these diseases is a dramatic increase of PC in reactive astrocytes. Some citrullinated proteins have been identified. The most prominent are astrocytic cytoskeletal proteins such as GFAP and vimentin, and myelin protein MBP. Recent investigation in ALS has revealed new changes, including a decreased PC in neurons and an association of PC with myelin protein aggregates. These findings suggest that PC contributes to protein aggregation, neuronal dysfunction, neuroinflammation, and axonal degeneration. However, how PC impact neurodegeneration remains to be understood. Further studies are needed to understand a range of questions, from how PC modulates individual protein functions to its impact on diseases. Because of the PC’s robust changes in neurodegenerative diseases, there are also prospects that this PTM may be harnessed as biomarkers, and modulation of this PTM may be an avenue for therapy. In this review, we summarize the current understanding of PC in ALS and other neurodegenerative diseases, the investigative methods for PC, and PC’s potential as a biomarker and a therapeutic target.

Keywords

Alzheimer’s disease, Neurodegenerative disease, Parkinson’s disease

Introduction 

Proteins are critical cellular machines that control cell growth, replication, and biological function. However, these proteins are encoded by a limited number of genes in our genome. To increase both the structural and functional diversity of the proteome to meet the demands in various environments, individual proteins can be posttranslationally modified in specific regions after synthesis. Several hundred posttranslational modifications (PTMs) are currently known, including phosphorylation, methylation, acetylation, glycosylation, ubiquitination, and SUMOylation [1]. Compared to these well-known PTMs, protein citrullination (PC) is relatively less studied, but has emerged to be a critical player in innate immunity. A well-established example is the formation of neutrophil extracellular traps (NETs), which trap pathogens (bacteria, viruses and parasites) for destruction during infections. Citrullination of histones decondenses chromatin and releases DNA strands to form NETs [2]. PC converts arginine residues in proteins (peptidyl-arginine) into citrullines (peptidyl-citrulline). Since the side chain of arginine is positively charged at physiological pH, this hydrolysis reaction results in loss of a positive charge, thereby altering the protein’s overall charge distribution. This PTM also alters the hydrogen bonding characteristics of the modified arginine, leading to protein structural and functional changes, including its activity and interactions with other proteins and nucleic acids [3-5]. In the case of NETs, PC weakens interaction between histones and DNA, thereby facilitating the unraveling of chromatin and DNA release.

PC is catalyzed by a family of enzymes referred to as the protein arginine deiminases (PADs). Mammals express five PADs, namely PAD1-4 (active), and PAD6 (inactive) [5]. All PAD isozymes show extensive sequence homology conserved across species. They are, however, differentially expressed in tissues, with PAD2 being the most ubiquitously expressed, and most dominant in the central nervous system (CNS) [5,6]. PAD activity is tightly regulated by calcium levels. Under normal cellular calcium concentrations (~100 nM), the PADs do not bind calcium and adopt a conformation that is catalytically inactive. Upon pathological or pharmacological stimulation, the intracellular concentrations of calcium rise, and the PADs bind to four to six calcium ions depending on the specific PAD. Calcium binding triggers a series of conformational changes that form and stabilize the active site enabling catalysis [7]. Once activated, PADs citrullinate a wide range of protein substrates, and this modification regulates several cellular processes, including cell signaling, immune responses, and gene regulation [5,8]. PADs and citrullination are involved in many inflammatory pathologies, including atherosclerosis, rheumatoid arthritis, COVID-19, lupus, cancers, and neurodegenerative diseases [3,9,10]. In neurodegenerative diseases, aberrant PAD2 activity and protein citrullination have been shown in Alzheimer’s disease (AD), Parkinson’s disease (PD), prion diseases, multiple sclerosis (MS), ischemic and traumatic brain injuries [11-16], and recently, in ALS [17,18]. In this review, we will summarize the current understanding of PC in ALS and other neurodegenerative diseases and its potential as both a biomarker and therapeutic target.

Protein Citrullination Changes are a Common Pathological Hallmark of Neurodegenerative Diseases

Neurodegenerative diseases are a diverse group of disorders of the nervous system that are caused by a progressive loss of one or multiple groups of neurons leading to manifestation of specific clinical symptoms that are associated with the loss of neurons. These diseases increasingly affect people globally as the population ages. Pathologically, these diseases share common characteristics such as unique genetic mutations as well as sporadic cases, relentless protein aggregation, aberrant RNA processing, cytoskeletal disruption, oxidative stress, altered energy metabolism, neuroinflammation, and neuronal cell death [19]. Additionally, significant changes in PTMs and their contribution to pathogenesis are widely recognized [1]. Compared with other PTM changes, PC and PAD dysregulation are relatively understudied, but are increasingly recognized as hallmarks of these diseases (Table 1).

Table 1: Overview of PADs and PC Changes in Neurodegenerative Diseases.

 

Amyotrophic Lateral Sclerosis

Alzheimer’s Disease

Parkinson’s Disease

Multiple Sclerosis

Prion disease

Disease features

Progressive motor neuron loss; motor dysfunction and paralysis; aggregation of TDP-43 and other proteins [20].

Loss of cortical and hippocampal neurons; memory loss and dementia; accumulation of senile plaques and neurofibrillary tangles [23].

Progressive loss of dopaminergic neurons; motor dysfunction; neuronal α-synuclein inclusions [29].

Chronic inflammatory demyelination and axonal degeneration; functional impairment in sensory, motor, visual, and autonomic nervous systems [40].

Wildspread neurodegeneration in the CNS; accumulation and spreading of misfolded and pathogenic prion proteins in the nervous system [37].

CNS regions with altered PADs and/or PC

Motor cortex, Spinal cord and brainstem.

Hippocampus, frontal cortex, retina.

Substantia nigra, Prefrontal cortex, Cingulate cortex, and hippocampus.

Medulla, spinal cord, brain white matter.

Hippocampus, brain stem, striatum, thalamus, Frontal cortex.

PADs and PC associated cell type changes and pathology

↑ PAD2 and PC in reactive astrocytes in motor cortex and spinal cord [17,18].

 

↓ PAD2 and PC in neurons [18].

 

↑ PC in white matter aggregates [17,18].

 

PC enriched in myelin proteins MBP and PLP aggregates [17,18].

↑ PAD2 and PC in reactive astrocytes [24-26].

 

↑ PAD4 and PC in cortical pyramidal and hippocampal hilar neurons [26].

 

↑ PC in nucleus of neurons with cytoplasmic p-Tau [25].

 

PC colocalizes with Aβ42 in extracellular plaque [25].

 

↑ citAβ in sporadic and familial AD [27].

 

↑ PAD2, PAD4, citTau in AD models and postmortem brain and retinal tissues [70,94].

 

↑ PC in reactive astrocytes in substantia nigra in PD

[16].

 

↑ PC in dopamine neurons in substantia nigra in PD [16].

 

↑ PAD2, PAD4, and citH3 in prefrontal cortex of X-linked Dystonia Parkinsonism (XDP) [32].

 

↑ PADs and PC in anterior cingulate cortex and hippocampus in earlier PD stages [33].

 

↑ PC in the vasculature of cortex and hippocampus in premotor PD rat model [34].

↑ PC in reactive astrocytes in areas of ongoing demyelination in chronic active lesions [42].

 

↑ PC at plaque interfaces and gray-white matter junctions in MS brains [49].

 

↑ PAD2, PAD4 and PC in MS brain normal-appearing white matter (NAWM) [14].

 

↑ PAD activity and citMBP in MS mouse model [15].

 

↑ PAD2, PAD4, and PC in myelin of MS brain [43].

 

↑ PC stained as puncta in acute and relapse EAE mouse model in medulla and spinal cord [59].

 

↑ PAD2 and PC in reactive astrocytes in the brain of sCJD and ME7 scrapie-infected mice [11,38].

 

↑ citVimentin accumulates in reactive astrocytes in sCJD and ME7 scrapie-infected mice [39].

 

↑ citMBP stained as puncta in scrapie infected mice brain white matter [58].

Common modified proteins

PC overlaps with GFAP, and MBP and PLP aggregates [17,18].

Vimentin, GFAP, HSP90A, NFL, MBP, CNP [24,26,28].

NFM, NFH, HSP70, H2B, H4 [34]. PC overlaps with GFAP [16].

H3, MBP, GFAP, Vimentin, CN37, NFM [42,49,69].

Vimentin, GFAP, MBP [11].

Models studied

Mouse models, SOD1G93A and PFN1C7G. Sporadic ALS postmortem tissues.

AD postmortem brain and retinal tissues. AD tauopathy mouse model.

PD postmortem brain tissues. XDP post-mortem tissues. Rat model of pre-motor PD.

DM-20 overexpressing mice, PAD2 overexpressing mice, EAE mouse model, MS postmortem tissues.

sCJD postmortem brain and ME7 scrapie-infected mice.

 

ALS is a deadly neurodegenerative disease that causes progressive motor neuron loss, motor dysfunction, and paralysis [20]. The role for PC in ALS has only recently been appreciated. In 2018, Tanikawa and colleagues showed that a group of RNA binding proteins that are known to be mutated in ALS, i.e., FUS, EWS, TAF15, and hnRNPA1, are citrullinated in PAD4-overexpressing cancer cells [21]. Notably, mutations in these proteins are known to enhance their propensity to aggregate but citrullination inhibited their aggregation. Furthermore, a polymorphism linked to reduced PAD4 expression appeared to increase the risk of ALS. This study suggests that a reduction in PAD4 activity could promote the onset of ALS [21]. However, these observations remain to be confirmed in ALS animal models and patients.

More recently, alterations in PC were found in both human postmortem tissues and two mouse models of ALS that express ALS-causing mutants SOD1G93A and PFN1C71G, respectively. In both models, PC was greatly increased in astrocytes and this increase correlates spatially with regions of active neurodegeneration, and temporally with disease progression. In the spinal cord white matter, PC is present prominently in protein aggregates that colocalize with the myelin proteins PLP and MBP. Interestingly, the increase in PC is not uniform in all cells. In neurons, PC is decreased at disease onset, and this decrease persists during disease progression [18]. These findings were validated in a follow-up study on human sporadic ALS (sALS) [17]. These results suggest that changes in PC play a role in ALS disease progression, possibly by contributing to neuronal dysfunction, protein aggregation, and neuroinflammation.

AD is the most common aging-associated neurodegenerative disease characterized by loss of neurons in the cortex and hippocampus, and accumulation of senile plaques and neurofibrillary tangles in the affected brain areas. Clinically, AD manifests as progressive loss of memory and dementia [22,23]. Ishigami and colleagues examined PC in AD and showed increased PAD2 expression and PC in reactive astrocytes in the hippocampus of AD brains [24]. Since then, several studies have further characterized aberrant citrullination in AD brains. Nicholas showed that citrullinated proteins are enriched in the nucleus of neurons with cytoplasmic phosphorylated tau and in reactive astrocytes surrounding extracellular amyloid plaque in the frontal cortex [25]. Acharya and colleagues reported that PAD4 and citrullinated proteins accumulate in the cytoplasm of cortical pyramidal and hippocampal hilar neurons expressing Aβ42 [26]. Mukerjee et al., showed that Aβ is citrullinated in sporadic and familial AD brains at arginine 5 [27]. Increased citrullination was also observed in the brains of AD with cerebrovascular disease [28]. The significance of the increased PC is yet to be established but likely contributes to neuroinflammation [28].

PD is a chronic, progressive neurodegenerative disease characterized by loss of dopaminergic neurons in the substantia nigra pars compacta, neuronal α-synuclein inclusions, and the resulting motor symptoms including bradykinesia, static tremor, and motor rigidity. PD is also associated with cognitive impairments [29-31]. In the PD brain, citrullinated proteins were detected in astrocytes in the substantia nigra (SN), but appeared more prominent in surviving dopamine neurons, where citrullinated proteins are diffusely distributed in the cytoplasm predominantly in the cell bodies and occasionally in proximal dendrites [16]. In the prefrontal cortex of X-linked Dystonia Parkinsonism, PAD2 and PAD4 are increased together with histone H3 citrullination [32]. A significant increase in PC and PAD expression levels was also observed in the anterior cingulate cortex and hippocampus in the earlier stages of PD [33]. In a rat model of pre-motor PD, PC was increased in the vasculature of cortex and hippocampus. Furthermore, a significantly high number of proteins were citrullinated in the plasma and plasma extracellular vesicles [34]. The early detection of PC in different models of PD suggests that aberrant citrullination is involved in the pathogenesis of this disease.

Prion diseases are a group of incurable neurodegenerative conditions that display a wide range of neurological symptoms including dementia, motor dysfunction, psychiatric problems and others [35]. The disease is caused by the replication, accumulation and spreading of misfolded and pathogenic prion proteins (PrPSC) in the nervous systems [36,37]. The main histopathological features are spongiform degeneration of the CNS, reactive gliosis, neuronal loss, chronic inflammation, and, in some cases, the formation of amyloid plaques. In ME7 scrapie-infected mice and Sporadic Creutzfeldt-Jakob disease (sCJD), the most common human prion disease, PAD2 and protein citrullination are significantly increased in reactive astrocytes in the brain. Several cytoskeletal, energy metabolism-associated, and myelin proteins were found to be citrullinated including vimentin, GFAP, enolase 1, aldolase A, MBP, cyclophilin A, and phosphoglycerate kinase [11,38,39].

MS is a chronic neurodegenerative disease characterized by inflammatory demyelination and axonal degeneration leading to a variety of neurological impairments to the visual, sensory, motor, cognitive and autonomic nervous systems [40]. These functional problems result from impaired neuronal function due to demyelination and axon degeneration. MS is generally thought to be an autoimmune disease mediated by T and B cell dysfunction leading to targeted destruction of myelin and axons [40,41]. The exact mechanism by which autoimmunity is triggered remains unknown. However, PAD2 and PAD4, and citrullinated proteins are elevated in myelin from MS patients and in active lesions with ongoing demyelination and surrounding reactive astrocytes [42,43]. Similarly, animal models of the disease show increased PAD2 and citrullinated MBP in the brain, particularly in the white matter [44,45]. Furthermore, MBP citrullination correlates to disease severity. In normal human brain, ~20% of MBP is citrullinated. In chronic MS, the citrullinated MBP increases to 45%. In the fulminating MS (Marburg disease), the citrullinated MBP accounted for 90% of the total [45,46].

PAD2 and PC as Hallmarks of Reactive Astrogliosis in Neurodegenerative Diseases

An increase in PAD2 expression and PC in astrocytes is a common pathological phenomenon in neurodegenerative diseases. The colocalization of this increase with areas of neurodegeneration or injury suggests that the increase is an insult-induced response and may serve as markers of reactive astrogliosis [18]. What role PC plays in astrocytes and how PC contributes to neurodegeneration is unknown. Nonetheless, PC could impact astrocyte functions and exacerbate neurodegeneration. First, citrullination induces depolymerization and fragmentation of GFAP and other astrocytic intermediate filaments like vimentin [47-49]. This could interfere with the cytoskeletal architecture of the astrocytes, compromising its functions including structural and nutritional supports for neurons, and synaptic formations. Second, a byproduct of citrullination reaction is ammonia. Ammonia is implicated as a neurotoxicant in many diseases [50,51]. The overwhelming astrocytic protein citrullination may constitute an ammonia build-up that could impair the astrocyte and/or spill over to exacerbate neuronal damage.

PC in Myelin Protein Aggregation and Myelin Degeneration

Protein aggregation is a prominent feature in neurodegenerative disease. Different protein aggregates not only differentiate distinct neurodegenerative diseases but also are thought to drive distinct neuropathology in each disease [52]. PTMs may modulate protein aggregation. For example, phosphorylation at specific sites of various proteins is enriched in disease-specific protein aggregates. In ALS and frontotemporal lobar degeneration (FTLD), phosphorylation of TDP-43 at S409 and S410 is enriched in TDP-43 aggregates [53,54]. Antibodies against the phosphorylation sites are widely used to detect the aggregates and as a neuropathological diagnostic marker. In PD and Lewy body dementia, phosphorylation of α-synuclein at S129 promotes aggregation, whereas phosphorylation at Y136 inhibits aggregation [55,56]. In AD and FTD-tau, hyperphosphorylated tau is enriched in tau aggregates and may facilitate and/or stabilize the aggregates and mediate neuronal toxicity [57].

Compared to phosphorylation, the relationship between PC and protein aggregation is much less studied and has only recently come to light. Yusuf and colleagues reported a progressive increase in citrullinated protein aggregates in white matter, starting at disease onset and peaking at the paralysis stage in ALS mouse models [18]. They also confirmed the existence of citrullinated protein aggregates in human ALS spinal cord and motor cortex [17]. The citrullinated aggregates colocalize with MBP and PLP aggregates, suggesting that PC may drive myelin protein aggregation. Previous literature suggests that myelin protein citrullination and aggregation may not be unique to ALS and may be relevant to other neurodegenerative diseases. In prion disease, an antibody raised against citrullinated MBP showed a punctate staining pattern in white matter [58], which could be MBP aggregates similar to what was observed in ALS. In an experimental autoimmune encephalomyelitis model (a model for MS), aggregate-like and PC-positive structures similar to those observed in ALS were detected in white matter, and increased levels of citrullinated MBP were detected in the insoluble fractions of brain and spinal cord proteins [59]. These results suggest that citrullinated protein aggregates that are enriched in myelin proteins may be common amongst various neurodegenerative conditions.

How PC impacts neurodegeneration is yet to be defined. In MS, citrullinated myelin proteins may represent autoantigens, thereby exacerbating autoimmunity against myelin and accelerate myelin destruction [3]. Moscarello and colleagues proposed that high levels of MBP citrullination disrupt compact myelin by diminishing the electrostatic interactions between the positively charged arginines in MBP and the negatively charged phospholipids [60]. The citrullinated MBP becomes membrane-unbound, has an open and unfolded structure and is vulnerable to proteolysis, which releases immunodominant peptides, triggering the immune response and destruction of myelin [61]. In ALS, increased MBP citrullination may result in its dissociation from the myelin membrane, misfolding, and subsequently aggregating. PC may also directly promote protein aggregation, as has been demonstrated with the myelin protein MOG, where citrullination promoted amyloid formation [62]. PC in myelin proteins may contribute to myelin degeneration in neurodegenerative diseases. However, PC may not just negatively impact neurodegeneration. Because numerous proteins are citrullinated and show complex changes in disease conditions, it may not be surprising that in some situations, citrullination in some proteins may be beneficial. Further experiments will be needed to tease out the significance of PC in neurodegenerative diseases.

Citrullinated Proteins as Potential Biomarkers in ALS

Biomarkers are critical for ALS diagnosis and over the last decade, several biomarkers for ALS have been identified [63]. These include neuronal cytoskeleton specific proteins such as neurofilaments (NFs) that are released into the CSF and blood due to axon degeneration [64]. Currently, neurofilament light chain (NfL) is the leading biomarker for monitoring the clinical progression of ALS [65]. However, some reports have shown that phosphorylated NfH (pNfH) has more diagnostic sensitivity and specificity than NfL [66-68], suggesting that measuring protein PTMs may be advantageous for better diagnosing the disease. NfL and NfH as well as other neuronal skeletons, i.e. α-internexin and the microtubule-associated protein tau, are known to be citrullinated [28,69,70]. Furthermore, neuronal PC is decreased in ALS, and studies in ALS mouse models suggest that the decrease starts at disease onset [17,18]. The decreased PC may be an early signal for compromised neuronal function before neuronal loss, raising the possibility that measuring neuronal protein citrullination (e.g. citNfL, citNfH) from biofluids may improve the accuracy of current biomarkers for ALS diagnosis. There is also a possibility that citrullination of other proteins or peptides may be altered and could be biomarkers for ALS, even if their protein levels do not change in biofluids.

Tools for Identifying Citrullinated Proteins

As changes in PC are being increasingly documented in neurodegenerative diseases, identifying citrullinated proteins and the changes in their citrullination sites has become increasingly important for understanding how citrullination modulates protein function and contributes to diseases. Several techniques have been developed in recent years to detect citrullinated proteins [8]. The easiest and quickest techniques are antibody-based. Senshu and colleagues developed an anti-citrulline modified (ACM) assay where a polyclonal antibody was raised against citrullinated proteins that were modified with 2,3-butanedione and antipyrine [71]. Nicholas and Whitaker developed another monoclonal antibody, F95, against a deca-citrullinated peptide [72]. Both antibodies recognize multiple citrullinated proteins and are widely used to detect citrullinated proteins in Western blots, immunohistochemistry, and immunofluorescence assays. These antibody assays detect the most abundant citrullinated proteins such as GFAP, vimentin, MBP and keratins when combined with other specific protein identification techniques, but they do not detect low-abundant citrullinated proteins nor the specific citrullination sites.

To identify citrullinated proteins efficiently, Lewallen and colleagues developed rhodamine- phenylglyoxal (RhPG) and biotin-PG (BPG) probes [73,74]. RhPG can be used in a high throughput assay format and can directly visualize protein citrullination in a complex biological proteome on an SDS-PAGE gel without transfer to a membrane. BPG can be used to enrich citrullinated proteins from cells or tissues, and the proteins can be visualized in western blots with streptavidin as a surrogate antibody. Furthermore, citrullinated proteins can be identified by liquid chromatography and tandem mass spectrometry (LC-MS/MS) [74]. By this approach, the whole citrullinome can be determined for both normal as well as diseased tissues [75].

A limitation of the BPG-enrichment coupled with LC-MS/MS method is that it does not pinpoint citrullinated amino acid residues in proteins. To solve this problem, several obstacles need to be overcome using standard mass spectrometry approaches. For example, it is difficult to identify deimination events, because the mass change is small (0.98 Da). Additionally, trypsin does not cleave after citrulline leading to longer peptides that are difficult to detect by MS/MS. These problems are further exaggerated when attempting to analyze low-abundance proteins in a complex biological sample. Lee and colleagues used LC-MS/MS to identify citrullination sites from various human tissues, but their work required manual inspection of mass spectra [76]. This approach is labor intensive, thereby limiting its wide use. Several researchers have tried to overcome this limitation by chemically modifying citrulline and detecting the mass shift of each adduct [77,78]. Recently, Maurais, Salinger and colleagues developed a software solution called ionFinder to automate this process [79]. Ionfinder was designed to unambiguously identify citrullinated peptides using a decision tree algorithm that takes into account the presence of neutral loss with the corresponding -43 Dalton shift on the mass spectra. The input data must be non-enriched proteomic data with protein coverage that spans the sites in question. Generally, it identifies highly abundant citrullinated proteins. There remains an opportunity to develop a MS spectrometry-based method that identifies the sites of citrullination on low abundant proteins.

PAD2 as a Potential Therapeutic Target in ALS

Research has shown that motor neuron degeneration in ALS is triggered by multiple distinct and sometimes overlapping pathways. Therefore, treatments targeting upstream causes for all ALS including sporadic and familial cases may be difficult if not impossible. Consequently, targeting a downstream convergent pathway of this disease may have advantages as to the broad relevance. Our recent studies indicate that an increase in protein citrullination is a common pathway downstream of diverse neurodegenerative conditions, including ALS [17,18]. Preclinical studies targeting PAD2, the most abundant enzyme that catalyzes protein citrullination in the brain, has shown benefits in some disease models, thereby highlighting the therapeutic potential of PAD2 inhibition [12,80-84]. PAD inhibitors which are derivatives of benzoylated arginines include the pan-PAD inhibitors Cl-amidine and BB-Cl-amidine, and isoform-specific inhibitors AFM-30a and GSK199 for PAD2 and PAD4, respectively [85,86]. These compounds have shown efficacy in multiple inflammatory disease models [82,83,87-93]. Whether PAD2 inhibition is therapeutically beneficial in ALS and other neurodegenerative diseases remains to be investigated. Considering the similarities in PC between ALS and MS [17,18,42], and PAD2 targeting reduces citrullination and disease phenotypes in the latter [81], PAD inhibition may elicit similar beneficial outcomes in ALS. Further experiments will be needed to test this hypothesis.

Conclusion

PC is a widespread PTM in various tissues and its alterations have been reported in the CNS of multiple neurodegenerative diseases. Detailed studies in ALS have demonstrated that PC is increased spatially and temporally in correlation with neurodegeneration. The upregulation of PC is mostly in astrocytes. By contrast, PC is decreased in neurons. Additionally, PC is enriched in myelin protein aggregates containing PLP and MBP. These observations, along with the observations in other neurodegenerative diseases, indicate that aberrant PC is a common hallmark in neurodegenerative diseases. To understand the significance of PC changes, future experiments will need to focus on the impact of these changes on the diseases. To pave the way for future experiments, the citrullinated proteins and the citrullination sites on these proteins need to be identified, and their changes in the diseases determined. To accomplish this, continuing efforts are needed to develop more powerful methods to efficiently unveil the citrullinome.

Funding

This work was supported by grants from the NIH/NINDS RO1-NS118145 to ZX and PRT, and NIH/NIGMS R35 GM118112 to PRT.

Competing Interest

PRT holds equity in Padlock Therapeutics, a subsidiary of Bristol Myers Squibb. The other authors have no competing interests to declare that are relevant to this article

References

1. Zhong Q, Xiao X, Qiu Y, Xu Z, Chen C, Chong B, et al. Protein posttranslational modifications in health and diseases: Functions, regulatory mechanisms, and therapeutic implications. MedComm (2020). 2023 May 2;4(3):e261.

2. Poli V, Zanoni I. Neutrophil intrinsic and extrinsic regulation of NETosis in health and disease. Trends Microbiol. 2023 Mar;31(3):280-93.

3. Ciesielski O, Biesiekierska M, Panthu B, Soszyński M, Pirola L, Balcerczyk A. Citrullination in the pathology of inflammatory and autoimmune disorders: recent advances and future perspectives. Cell Mol Life Sci. 2022 Jan 25;79(2):94.

4. Fuhrmann J, Clancy KW, Thompson PR. Chemical biology of protein arginine modifications in epigenetic regulation. Chem Rev. 2015 Jun 10;115(11):5413-61.

5. Yu K, Proost P. Insights into peptidylarginine deiminase expression and citrullination pathways. Trends Cell Biol. 2022 Sep;32(9):746-61.

6. Shimada N, Handa S, Uchida Y, Fukuda M, Maruyama N, Asaga H, et al. Developmental and age-related changes of peptidylarginine deiminase 2 in the mouse brain. J Neurosci Res. 2010 Mar;88(4):798-806.

7. Slade DJ, Fang P, Dreyton CJ, Zhang Y, Fuhrmann J, Rempel D, et al. Protein arginine deiminase 2 binds calcium in an ordered fashion: implications for inhibitor design. ACS Chem Biol. 2015 Apr 17;10(4):1043-53.

8. Tilvawala R, Thompson PR. Peptidyl arginine deiminases: detection and functional analysis of protein citrullination. Curr Opin Struct Biol. 2019 Dec;59:205-15.

9. Song S, Yu Y. Progression on Citrullination of Proteins in Gastrointestinal Cancers. Front Oncol. 2019 Jan 23;9:15.

10. Witalison EE, Thompson PR, Hofseth LJ. Protein Arginine Deiminases and Associated Citrullination: Physiological Functions and Diseases Associated with Dysregulation. Curr Drug Targets. 2015;16(7):700-10.

11. Jang B, Jin JK, Jeon YC, Cho HJ, Ishigami A, Choi KC, Carp RI, Maruyama N, Kim YS, Choi EK. Involvement of peptidylarginine deiminase-mediated post-translational citrullination in pathogenesis of sporadic Creutzfeldt-Jakob disease. Acta Neuropathol. 2010 Feb;119(2):199-210.

12. Lange S, Rocha-Ferreira E, Thei L, Mawjee P, Bennett K, Thompson PR, et al. Peptidylarginine deiminases: novel drug targets for prevention of neuronal damage following hypoxic ischemic insult (HI) in neonates. J Neurochem. 2014 Aug;130(4):555-62.

13. Lazarus RC, Buonora JE, Flora MN, Freedy JG, Holstein GR, Martinelli GP, et al. Protein Citrullination: A Proposed Mechanism for Pathology in Traumatic Brain Injury. Front Neurol. 2015 Sep 22;6:204.

14. Mastronardi FG, Wood DD, Mei J, Raijmakers R, Tseveleki V, Dosch HM, et al. Increased citrullination of histone H3 in multiple sclerosis brain and animal models of demyelination: a role for tumor necrosis factor-induced peptidylarginine deiminase 4 translocation. J Neurosci. 2006 Nov 1;26(44):11387-96.

15. Moscarello MA, Pritzker L, Mastronardi FG, Wood DD. Peptidylarginine deiminase: a candidate factor in demyelinating disease. J Neurochem. 2002 Apr;81(2):335-43.

16. Nicholas AP. Dual immunofluorescence study of citrullinated proteins in Parkinson diseased substantia nigra. Neurosci Lett. 2011 May 9;495(1):26-9.

17. Yusuf IO, Parsi S, Ostrow LW, Brown RH, Thompson PR, Xu Z. PAD2 dysregulation and aberrant protein citrullination feature prominently in reactive astrogliosis and myelin protein aggregation in sporadic ALS. Neurobiol Dis. 2024 Mar;192:106414.

18. Yusuf IO, Qiao T, Parsi S, Tilvawala R, Thompson PR, Xu Z. Protein citrullination marks myelin protein aggregation and disease progression in mouse ALS models. Acta Neuropathol Commun. 2022 Sep 8;10(1):135.

19. Wilson DM 3rd, Cookson MR, Van Den Bosch L, Zetterberg H, Holtzman DM, Dewachter I. Hallmarks of neurodegenerative diseases. Cell. 2023 Feb 16;186(4):693-714.

20. Younger DS, Brown RH Jr. Amyotrophic lateral sclerosis. Handb Clin Neurol. 2023;196:203-29.

21. Tanikawa C, Ueda K, Suzuki A, Iida A, Nakamura R, Atsuta N, et al. Citrullination of RGG Motifs in FET Proteins by PAD4 Regulates Protein Aggregation and ALS Susceptibility. Cell Rep. 2018 Feb 6;22(6):1473-83.

22. DeTure MA, Dickson DW. The neuropathological diagnosis of Alzheimer's disease. Mol Neurodegener. 2019 Aug 2;14(1):32.

23. Wu BS, Zhang YR, Li HQ, Kuo K, Chen SD, Dong Q, et al. Cortical structure and the risk for Alzheimer's disease: a bidirectional Mendelian randomization study. Transl Psychiatry. 2021 Sep 15;11(1):476.

24. Ishigami A, Ohsawa T, Hiratsuka M, Taguchi H, Kobayashi S, Saito Y, et al. Abnormal accumulation of citrullinated proteins catalyzed by peptidylarginine deiminase in hippocampal extracts from patients with Alzheimer's disease. J Neurosci Res. 2005 Apr 1;80(1):120-8.

25. Nicholas AP. Dual immunofluorescence study of citrullinated proteins in Alzheimer diseased frontal cortex. Neurosci Lett. 2013 Jun 17;545:107-11.

26. Acharya NK, Nagele EP, Han M, Coretti NJ, DeMarshall C, Kosciuk MC, et al. Neuronal PAD4 expression and protein citrullination: possible role in production of autoantibodies associated with neurodegenerative disease. J Autoimmun. 2012 Jun;38(4):369-80.

27. Mukherjee S, Perez KA, Dubois C, Nisbet RM, Li QX, Varghese S, et al. Citrullination of Amyloid-β Peptides in Alzheimer's Disease. ACS Chem Neurosci. 2021 Oct 6;12(19):3719-32.

28. Gallart-Palau X, Serra A, Lee BST, Guo X, Sze SK. Brain ureido degenerative protein modifications are associated with neuroinflammation and proteinopathy in Alzheimer's disease with cerebrovascular disease. J Neuroinflammation. 2017 Sep 2;14(1):175.

29. Aarsland D, Batzu L, Halliday GM, Geurtsen GJ, Ballard C, Ray Chaudhuri K, et al. Parkinson disease-associated cognitive impairment. Nat Rev Dis Primers. 2021 Jul 1;7(1):47.

30. DeMaagd G, Philip A. Parkinson's Disease and Its Management: Part 1: Disease Entity, Risk Factors, Pathophysiology, Clinical Presentation, and Diagnosis. P T. 2015 Aug;40(8):504-32.

31. Zeng XS, Geng WS, Jia JJ, Chen L, Zhang PP. Cellular and Molecular Basis of Neurodegeneration in Parkinson Disease. Front Aging Neurosci. 2018 Apr 17;10:109.

32. Petrozziello T, Mills AN, Vaine CA, Penney EB, Fernandez-Cerado C, Legarda GPA, et al. Neuroinflammation and histone H3 citrullination are increased in X-linked Dystonia Parkinsonism post-mortem prefrontal cortex. Neurobiol Dis. 2020 Oct;144:105032.

33. Mercer A, Jaunmuktane Z, Hristova M, Lange S. Differential, Stage Dependent Detection of Peptidylarginine Deiminases and Protein Deimination in Lewy Body Diseases-Findings from a Pilot Study. Int J Mol Sci. 2022 Oct 28;23(21):13117.

34. Sancandi M, Uysal-Onganer P, Kraev I, Mercer A, Lange S. Protein Deimination Signatures in Plasma and Plasma-EVs and Protein Deimination in the Brain Vasculature in a Rat Model of Pre-Motor Parkinson's Disease. Int J Mol Sci. 2020 Apr 15;21(8):2743.

35. Baldwin KJ, Correll CM. Prion Disease. Semin Neurol. 2019 Aug;39(4):428-39.

36. McKintosh E, Tabrizi SJ, Collinge J. Prion diseases. J Neurovirol. 2003 Apr;9(2):183-93.

37. Sigurdson CJ, Bartz JC, Glatzel M. Cellular and Molecular Mechanisms of Prion Disease. Annu Rev Pathol. 2019 Jan 24;14:497-516.

38. Jang B, Kim E, Choi JK, Jin JK, Kim JI, Ishigami A, et al. Accumulation of citrullinated proteins by up-regulated peptidylarginine deiminase 2 in brains of scrapie-infected mice: a possible role in pathogenesis. Am J Pathol. 2008 Oct;173(4):1129-42.

39. Jang B, Kim MJ, Lee YJ, Ishigami A, Kim YS, Choi EK. Vimentin citrullination probed by a novel monoclonal antibody serves as a specific indicator for reactive astrocytes in neurodegeneration. Neuropathol Appl Neurobiol. 2020 Dec;46(7):751-69.

40. Dobson R, Giovannoni G. Multiple sclerosis - a review. Eur J Neurol. 2019 Jan;26(1):27-40.

41. Herz J, Zipp F, Siffrin V. Neurodegeneration in autoimmune CNS inflammation. Exp Neurol. 2010 Sep;225(1):9-17.

42. Bradford CM, Ramos I, Cross AK, Haddock G, McQuaid S, Nicholas AP, et al. Localisation of citrullinated proteins in normal appearing white matter and lesions in the central nervous system in multiple sclerosis. J Neuroimmunol. 2014 Aug 15;273(1-2):85-95.

43. Wood DD, Ackerley CA, Brand Bv, Zhang L, Raijmakers R, Mastronardi FG, et al. Myelin localization of peptidylarginine deiminases 2 and 4: comparison of PAD2 and PAD4 activities. Lab Invest. 2008 Apr;88(4):354-64.

44. Musse AA, Li Z, Ackerley CA, Bienzle D, Lei H, Poma R, et al. Peptidylarginine deiminase 2 (PAD2) overexpression in transgenic mice leads to myelin loss in the central nervous system. Dis Model Mech. 2008 Nov-Dec;1(4-5):229-40.

45. Moscarello MA, Wood DD, Ackerley C, Boulias C. Myelin in multiple sclerosis is developmentally immature. J Clin Invest. 1994 Jul;94(1):146-54.

46. Wood DD, Bilbao JM, O'Connors P, Moscarello MA. Acute multiple sclerosis (Marburg type) is associated with developmentally immature myelin basic protein. Ann Neurol. 1996 Jul;40(1):18-24.

47. Briot J, Simon M, Méchin MC. Deimination, Intermediate Filaments and Associated Proteins. Int J Mol Sci. 2020 Nov 19;21(22):8746.

48. Inagaki M, Takahara H, Nishi Y, Sugawara K, Sato C. Ca2+-dependent deimination-induced disassembly of intermediate filaments involves specific modification of the amino-terminal head domain. J Biol Chem. 1989 Oct 25;264(30):18119-27.

49. Nicholas AP, Sambandam T, Echols JD, Tourtellotte WW. Increased citrullinated glial fibrillary acidic protein in secondary progressive multiple sclerosis. J Comp Neurol. 2004 May 17;473(1):128-36.

50. Adlimoghaddam A, Sabbir MG, Albensi BC. Ammonia as a Potential Neurotoxic Factor in Alzheimer's Disease. Front Mol Neurosci. 2016 Aug 8;9:57.

51. Eid T, Lee TS. Reassessing the role of astrocytes in ammonia neurotoxicity. Nat Med. 2013 Dec;19(12):1572-4.

52. Soto C, Pritzkow S. Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat Neurosci. 2018 Oct;21(10):1332-40.

53. Hasegawa M, Arai T, Nonaka T, Kametani F, Yoshida M, Hashizume Y, et al. Phosphorylated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Ann Neurol. 2008 Jul;64(1):60-70.

54. Inukai Y, Nonaka T, Arai T, Yoshida M, Hashizume Y, Beach TG, et al. Abnormal phosphorylation of Ser409/410 of TDP-43 in FTLD-U and ALS. FEBS Lett. 2008 Aug 20;582(19):2899-904.

55. Samuel F, Flavin WP, Iqbal S, Pacelli C, Sri Renganathan SD, Trudeau LE, et al. Effects of Serine 129 Phosphorylation on α-Synuclein Aggregation, Membrane Association, and Internalization. J Biol Chem. 2016 Feb 26;291(9):4374-85.

56. Sano K, Iwasaki Y, Yamashita Y, Irie K, Hosokawa M, Satoh K, et al. Tyrosine 136 phosphorylation of α-synuclein aggregates in the Lewy body dementia brain: involvement of serine 129 phosphorylation by casein kinase 2. Acta Neuropathol Commun. 2021 Nov 12;9(1):182.

57. Liu M, Dexheimer T, Sui D, Hovde S, Deng X, Kwok R, et al. Hyperphosphorylated tau aggregation and cytotoxicity modulators screen identified prescription drugs linked to Alzheimer's disease and cognitive functions. Sci Rep. 2020 Oct 6;10(1):16551.

58. Jang B, Jeon YC, Shin HY, Lee YJ, Kim H, Kondo Y, et al. Myelin Basic Protein Citrullination, a Hallmark of Central Nervous System Demyelination, Assessed by Novel Monoclonal Antibodies in Prion Diseases. Mol Neurobiol. 2018 Apr;55(4):3172-84.

59. Raijmakers R, Vogelzangs J, Croxford JL, Wesseling P, van Venrooij WJ, Pruijn GJ. Citrullination of central nervous system proteins during the development of experimental autoimmune encephalomyelitis. J Comp Neurol. 2005 Jun 6;486(3):243-53.

60. Moscarello MA, Mastronardi FG, Wood DD. The role of citrullinated proteins suggests a novel mechanism in the pathogenesis of multiple sclerosis. Neurochem Res. 2007 Feb;32(2):251-6.

61. Cao L, Goodin R, Wood D, Moscarello MA, Whitaker JN. Rapid release and unusual stability of immunodominant peptide 45-89 from citrullinated myelin basic protein. Biochemistry. 1999 May 11;38(19):6157-63.

62. Araman C, van Gent ME, Meeuwenoord NJ, Heijmans N, Marqvorsen MHS, Doelman W, et al. Amyloid-like Behavior of Site-Specifically Citrullinated Myelin Oligodendrocyte Protein (MOG) Peptide Fragments inside EBV-Infected B-Cells Influences Their Cytotoxicity and Autoimmunogenicity. Biochemistry. 2019 Feb 12;58(6):763-75.

63. Benatar M, Boylan K, Jeromin A, Rutkove SB, Berry J, Atassi N, et al. ALS biomarkers for therapy development: State of the field and future directions. Muscle Nerve. 2016 Feb;53(2):169-82.

64. Gordon BA. Neurofilaments in disease: what do we know? Curr Opin Neurobiol. 2020 Apr;61:105-15.

65. Benatar M, Wuu J, Turner MR. Neurofilament light chain in drug development for amyotrophic lateral sclerosis: a critical appraisal. Brain. 2023 Jul 3;146(7):2711-6.

66. Heckler I, Venkataraman I. Phosphorylated neurofilament heavy chain: a potential diagnostic biomarker in amyotrophic lateral sclerosis. J Neurophysiol. 2022 Mar 1;127(3):737-45.

67. Li DW, Ren H, Jeromin A, Liu M, Shen D, Tai H, et al. Diagnostic Performance of Neurofilaments in Chinese Patients With Amyotrophic Lateral Sclerosis: A Prospective Study. Front Neurol. 2018 Aug 28;9:726.

68. Poesen K, De Schaepdryver M, Stubendorff B, Gille B, Muckova P, Wendler S, et al. Neurofilament markers for ALS correlate with extent of upper and lower motor neuron disease. Neurology. 2017 Jun 13;88(24):2302-9.

69. Faigle W, Cruciani C, Wolski W, Roschitzki B, Puthenparampil M, Tomas-Ojer P, et al. Brain Citrullination Patterns and T Cell Reactivity of Cerebrospinal Fluid-Derived CD4+ T Cells in Multiple Sclerosis. Front Immunol. 2019 Apr 10;10:540.

70. Shi H, Mirzaei N, Koronyo Y, Davis MR, Robinson E, Braun GM, et al. Identification of retinal oligomeric, citrullinated, and other tau isoforms in early and advanced AD and relations to disease status. Acta Neuropathol. 2024 Jul 9;148(1):3.

71. Senshu T, Sato T, Inoue T, Akiyama K, Asaga H. Detection of citrulline residues in deiminated proteins on polyvinylidene difluoride membrane. Anal Biochem. 1992 May 15;203(1):94-100.

72. Nicholas AP, Whitaker JN. Preparation of a monoclonal antibody to citrullinated epitopes: its characterization and some applications to immunohistochemistry in human brain. Glia. 2002 Mar 15;37(4):328-36.

73. Bicker KL, Subramanian V, Chumanevich AA, Hofseth LJ, Thompson PR. Seeing citrulline: development of a phenylglyoxal-based probe to visualize protein citrullination. J Am Chem Soc. 2012 Oct 17;134(41):17015-8.

74. Lewallen DM, Bicker KL, Subramanian V, Clancy KW, Slade DJ, Martell J, et al. Chemical Proteomic Platform To Identify Citrullinated Proteins. ACS Chem Biol. 2015 Nov 20;10(11):2520-8.

75. Tilvawala R, Nguyen SH, Maurais AJ, Nemmara VV, Nagar M, Salinger AJ, et al. The Rheumatoid Arthritis-Associated Citrullinome. Cell Chem Biol. 2018 Jun 21;25(6):691-704.e6.

76. Lee CY, Wang D, Wilhelm M, Zolg DP, Schmidt T, Schnatbaum K, et al. Mining the Human Tissue Proteome for Protein Citrullination. Mol Cell Proteomics. 2018 Jul;17(7):1378-91.

77. Choi M, Song JS, Kim HJ, Cha S, Lee EY. Matrix-assisted laser desorption ionization-time of flight mass spectrometry identification of peptide citrullination site using Br signature. Anal Biochem. 2013 Jun 1;437(1):62-7.

78. Holm A, Rise F, Sessler N, Sollid LM, Undheim K, Fleckenstein B. Specific modification of peptide-bound citrulline residues. Anal Biochem. 2006 May 1;352(1):68-76.

79. Maurais AJ, Salinger AJ, Tobin M, Shaffer SA, Weerapana E, Thompson PR. A Streamlined Data Analysis Pipeline for the Identification of Sites of Citrullination. Biochemistry. 2021 Sep 28;60(38):2902-14.

80. Kim S, Song YK, Cho CS, Kim HJ, Fang S, Jo DH, et al. Inhibition of protein arginine deiminase II suppresses retinoblastoma in orthotopic transplantation in mice. Oncol Rep. 2023 Jul;50(1):146.

81. Moscarello MA, Lei H, Mastronardi FG, Winer S, Tsui H, Li Z, et al. Inhibition of peptidyl-arginine deiminases reverses protein-hypercitrullination and disease in mouse models of multiple sclerosis. Dis Model Mech. 2013 Mar;6(2):467-78.

82. Sodré FMC, Bissenova S, Bruggeman Y, Tilvawala R, Cook DP, Berthault C, et al. Peptidylarginine Deiminase Inhibition Prevents Diabetes Development in NOD Mice. Diabetes. 2021 Feb;70(2):516-28.

83. Wu Z, Deng Q, Pan B, Alam HB, Tian Y, Bhatti UF, et al. Inhibition of PAD2 Improves Survival in a Mouse Model of Lethal LPS-Induced Endotoxic Shock. Inflammation. 2020 Aug;43(4):1436-45.

84. Zhou J, Biesterveld BE, Li Y, Wu Z, Tian Y, Williams AM, et al. Peptidylarginine Deiminase 2 Knockout Improves Survival in hemorrhagic shock. Shock. 2020 Oct;54(4):458-63.

85. Martín Monreal MT, Rebak AS, Massarenti L, Mondal S, Šenolt L, Ødum N, et al. Applicability of Small-Molecule Inhibitors in the Study of Peptidyl Arginine Deiminase 2 (PAD2) and PAD4. Front Immunol. 2021 Oct 19;12:716250

86. Muth A, Subramanian V, Beaumont E, Nagar M, Kerry P, McEwan P, et al. Development of a Selective Inhibitor of Protein Arginine Deiminase 2. J Med Chem. 2017 Apr 13;60(7):3198-11.

87. Biron BM, Chung CS, Chen Y, Wilson Z, Fallon EA, Reichner JS, et al. PAD4 Deficiency Leads to Decreased Organ Dysfunction and Improved Survival in a Dual Insult Model of Hemorrhagic Shock and Sepsis. J Immunol. 2018 Mar 1;200(5):1817-28.

88. Chumanevich AA, Causey CP, Knuckley BA, Jones JE, Poudyal D, Chumanevich AP, et al. Suppression of colitis in mice by Cl-amidine: a novel peptidylarginine deiminase inhibitor. Am J Physiol Gastrointest Liver Physiol. 2011 Jun;300(6):G929-38.

89. Domiciano TP, Lee Y, Carvalho TT, Wakita D, Martinon D, Jena PK, et al. Redundant role of PAD2 and PAD4 in the development of cardiovascular lesions in a mouse model of Kawasaki disease vasculitis. Clin Exp Immunol. 2024 Nov 12;218(3):314-28.

90. Knight JS, Subramanian V, O'Dell AA, Yalavarthi S, Zhao W, Smith CK, et al. Peptidylarginine deiminase inhibition disrupts NET formation and protects against kidney, skin and vascular disease in lupus-prone MRL/lpr mice. Ann Rheum Dis. 2015 Dec;74(12):2199-206.

91. Lewis HD, Liddle J, Coote JE, Atkinson SJ, Barker MD, Bax BD, et al. Inhibition of PAD4 activity is sufficient to disrupt mouse and human NET formation. Nat Chem Biol. 2015 Mar;11(3):189-91.

92. Shen Y, You Q, Wu Y, Wu J. Inhibition of PAD4-mediated NET formation by cl-amidine prevents diabetes development in nonobese diabetic mice. Eur J Pharmacol. 2022 Feb 5;916:174623.

93. Willis VC, Banda NK, Cordova KN, Chandra PE, Robinson WH, Cooper DC, et al. Protein arginine deiminase 4 inhibition is sufficient for the amelioration of collagen-induced arthritis. Clin Exp Immunol. 2017 May;188(2):263-74.

94. Liang H, Hunt Jr JB, Ma C, Kovalenko A, Calahatian J, Pedersen C, et al. Probing tau citrullination in Alzheimer's disease brains and mouse models of tauopathy. bioRxiv preprint. 2024 July 9. doi: https://doi.org/10.1101/2024.07.06.601399.

Author Information X