Abstract
Cardiovascular disease is a pressing health problem with significant global health, societal, and financial burdens. Understanding the molecular basis of polygenic cardiac pathology is thus essential to devising novel approaches for management and treatment. Recent identification of uncharacterized regulatory functions for a class of nuclear envelope proteins called nucleoporins offers the opportunity to understand novel putative mechanisms of cardiac disease development and progression. Consistent reports of nucleoporin deregulation associated with ischemic and dilated cardiomyopathies, arrhythmias and valvular disorders suggests that nucleoporin impairment may be a significant but understudied variable in cardiopathologic disorders. This review discusses and converges existing literature regarding nuclear pore complex proteins and their association with cardiac pathologies, and proposes a role for nucleoporins as facilitators of cardiac disease
Keywords: cardiopathologies, nucleoporin, nuclear pore complex, arrhythmia, cardiomyopathy
1. Introduction
New light is being shed on the etiology of cardiac pathologies by studies focused on the role of discrete nuclear envelope components [1, 2]. Specifically, the role of proteins within the nuclear pore complex (NPC) called nucleoporins (NUPs) have been noted as potential causes for a variety of cardiac diseases ranging from atrial fibrillation to cardiomyopathy [3–6]. NPCs, first identified in the 1950s by Callan and Tomlin [7], are comprised of multiple subcomplexes that form a physical channel between the nucleus and cytoplasm to regulate macromolecular transport across the nuclear envelope [8, 9]. The canonical role of NPCs in nucleocytoplasmic trafficking is well characterized, in which cargoes bound for either nuclear or cytoplasmic compartments are respectively dependent on specific nuclear localization or export signaling sequences [10]. This requires coordination of multiple soluble transport receptors, cofactors, and nuclear envelope bound components that work synergistically to facilitate transport [11], in addition to employing a GTP gradient across the nuclear envelope that provides the driving force for nucleocytoplasmic translocation [11, 12]. Furthermore, phosphorylating enzymes such as MAP kinases, CaM kinases, and lipid dependent kinases have been reported to regulate transport processes and nuclear pore expression [13].
Recent high resolution molecular mapping studies of NPC anatomy have advanced the knowledge of how subcomplexes that comprise the NPC are oriented in three dimensional space as well as their intra- and inter- modular interactions [14]. Each of these subcomplexes are composed of discrete NUPs localized to specific regions of the NPC [15, 16] (Figure 1). In the mammalian NPC, NUP45/58, NUP54, and NUP62 are part of a trimeric complex that make up the central channel of the NPC [17]. POM121, NDC1, and NUP210 are integral membrane bound NUPs that anchor the NPC to the nuclear membrane. TPR, NUP153, and NUP50 comprise the nuclear basket found on the nucleoplasmic face, with NUP358, NUP214, NUP88, ALADIN and NUP42/hCG1 located on the cytoplasmic side. The inner ring (NUP35/53, NUP93, NUP155, NUP188, NUP205) [18] and Y-subcomplex (NUP37, NUP43, NUP75/85, NUP96, NUP107, NUP133, NUP160, SEC13, SEH1L and ELYS [8] provide structural support to the NPC, while NUP98 is found on both cytoplasmic and nucleoplasmic sides of the NPC [16, 19, 20]. In addition, each of these subcomplexes possess distinct prioritized functions (Table 1). For example, the cytoplasmic, central channel, and nuclear basket regions mediate nucleocytoplasmic trafficking and harbor phenylalanineglycine (FG) rich NUPs that define the transport dynamics of the NPC [8, 21]. Considering the nature of the molecular cargo moving in and out of the nucleus, it is noteworthy that the cytoplasmic regions contains NUPs that specialize in finalizing mRNA export [22], while the nuclear face of the NPC possesses NUPs optimized for chromatin binding and remodeling [3, 23–26]. The Y-complex and inner ring complexes are composed of scaffold NUPs that do not possess these FG repeats [27], while the membrane bound NUPs are characterized by transmembrane domains that define their localization within the NPC [28–30]. In total, ~30 different NUPs are accepted as building blocks of the contemporary NPC [8, 31].
Figure 1.
Schematic illustration of the Nuclear Pore Complex (NPC) depicting the location of known nucleoporins, and their subcomplexes, within the NPC. Highlighted (*) are the mobile nucleoporins. CCR = central channel region, IR = inner ring, NB = nuclear basket, OR = outer rings.
Table 1.
Nucleoporin functions.
NPC region | Protein Symbol | Description | Function | References |
---|---|---|---|---|
Cytoplasmic | NUP358 | Nucleoporin 358; RAN binding protein 2 (RANBP2) | SUMOylation enhancer | [60–62] |
NUP214 | Nucleoporin 214 | Regulation of protein activity | [63] | |
NUP88 | Nucleoporin 88 | Chromatin binding | [20] | |
ALADIN | ALADIN/ADRACALIN | Spindle formation and chromosomal alignment | [64] | |
NUP42 | Nucleoporin 42; NUPL2; HCG1; NLP1/2 | RNA export/enhances RNA helicase activity | [65] | |
NUP98* | Nucleoporin 98* | Chromatin binding, mRNA export | [20, 66, 67] | |
Y-complex | NUP37 | Nucleoporin 37 | NPC stabilization and assembly during mitosis | [68, 69] |
NUP43 | Nucleoporin 43 | NPC assembly during mitosis | [70, 71] | |
NUP85 | Nucleoporin 85 | NPC assembly during mitosis | [70, 71] | |
NUP96* | Nucleoporin 96* | mRNA export, role in immune response mediated by interferons | [72–75] | |
NUP107 | Nucleoporin 107 | NPC assembly during mitosis | [70, 71] | |
NUP133 | Nucleoporin 133 | mRNA export, NPC assembly during mitosis | [71, 76] | |
NUP160 | Nucleoporin 160 | mRNA export, NPC assembly during mitosis | [70, 71, 76] | |
SEC13 | SEC13 Homolog, Nuclear Pore and COPII Complex Component | NPC assembly | [77] | |
SEH1L | SEH1 Like Nucleoporin | Activation of mTOR signaling, NPC assembly during mitosis | [70, 78] | |
ELYS | AHCTF1 gene, AT-Hook Containing Transcription Factor 1 | NPC stabilization and assembly during mitosis | [68] | |
Inner Ring | NUP35 | Nucleoporin 53 | Post-transcriptional regulation; oocyte meiotic spindle formation | [79, 80] |
NUP93 | Nucleoporin 93 | Gene repression | [47, 81] | |
NUP155 | Nucleoporin 155 | Chromatin binding/tethering and accessibility | [43, 51, 82, 83] | |
NUP188 | Nucleoporin 188 | Gene repression | [81] | |
NUP205 | Nucleoporin 205 | Gene repression | [81] | |
Central channel | NUP45/58 | Nucleoporin 58; NUPL1 | Centrosome regulation | [84] |
NUP54 | Nucleoporin 54 | Double strand break repair | [85] | |
NUP62 | Nucleoporin 62 | Imprinted domain regulation; chromatin tethering; centrosome homeostasis | [51, 86, 87] | |
Transmembrane | POM121 | Nuclear pore membrane protein 121 kDa | Transcriptional regulation; NPC assembly | [88–90] |
NUP210/GP210 | Nuclear pore membrane glycoprotein 210 | NE/ER homeostasis; gene regulation | [91–93] | |
NDC1/TMEM48 | Nuclear Division Cycle 1 homolog; Transmembrane protein 48 | NPC assembly | [94] | |
Nucleoplasmic | NUP50 | Nucleoporin 50 | Chromatin binding | [19, 92] |
NUP153 | Nucleoporin 153 | Chromatin binding, RNA export | [3, 44, 45, 92, 95] | |
TPR | Translocated Promoter Region | Heterochromatin exclusion | [48] |
Provided here are functions/biological contexts documented for NUPs. In bold are NUPs with reported high mobility (NUP98, NUP210, NUP50, and NUP153) that can be found distal from the NPC within the nuclear envelope and/or the nucleoplasm.
NUP96 and NUP98 are translated from the same transcript, and post-translational cleavage of the protein product generates both.
In addition to the canonical transport function of the NPC, other constitutive roles include NPC biogenesis, nuclear envelope assembly, as well as functional roles in cell cycling and mitosis that are the subject of excellent studies (Table 1). Individual NUPs have other cellular roles including chromatin organization, gene expression regulation, and DNA repair, among others [32–35]. Furthermore, NUPs may temporally recruit a diversity of protein-binding partners to form distinct signaling complexes, and this may occur at the nuclear envelope or within the nucleoplasm [36–40]. In tandem with these individual functions for NUPs, a consistent and recognized role for NUPs in development is emerging [9, 41]. Indeed, evidence for NUPs directly and indirectly binding to chromatin to regulate gene expression has been demonstrated [42–47].
The ability of NUPs to spatially organize chromatin underlies their capacity as gene regulators, and several modes of dynamic chromatin regulation have been reported for specific NUPs. For example, the maintenance of heterochromatin exclusion zones within the nucleoplasm proximal to the nuclear basket of the NPC is a function of TPR [48], and was proposed to exist as a means of maintaining a transcriptionally permissive microenvironment. This notion is supported by work in the yeast homologue of TPR, Mlp1, that maintains chromatin loops as a form of transcriptional memory. This was elegantly demonstrated by work in which Mlp1 was found to bind the 5’ and 3’ ends of the HXK1 gene to promote rapid metabolic responses to substrate switching [49]. The ability for NUPs to dynamically interact with chromatin appears to be broadly conserved [50], and is not exclusive to TPR. In a model of stem cell-derived neurogenesis, the identification of NUP153 as a regulator of superenhancer activity confirmed its function as a developmental regulator with clear demonstration of its ability to mediate chromatin interactions between distal gene elements [44, 50]. Complementary to loop formation, regulation of chromatin tethering in somatic and female germline Drosophila cells has been reported for the insect homologues of NUP155, NUP93, and NUP62 [51]. In their proposed model, chromatin is constitutively tethered to the nuclear periphery via interactions with NUP155; this interaction is fully or partially released upon interaction with NUP62 or NUP93, respectively. The area of chromatin regulation and NUP function is an exciting field, yet the large-scale chromatin anchoring and release dynamics reported for NUPs is unsurprising given their ability to effect individual gene regulation.
NUP153 was shown to regulate the cardiopathogenic expression of several genes in the setting of muscular dystrophy [3]. Specifically, NUP153 was localized to promoters of specific genes in cardiac tissue to facilitate recruitment of the activating lysine acetylase CBP/p300 [3]. This was confirmed by gene NUP153 overexpression and knockdown in which respective increases and decreases of p300 and p300/CBP loci interaction and enrichment was observed. In support of a broader gene regulatory function for NUP153, recent independent work demonstrated that NUP153 also acts in concert with SOX2 to regulate superenhancer dynamics during neurogenesis [44]. Importantly, this latest evidence in cardiac and neural contexts has emerged after investigation of NUP153 chromatin binding in other metazoan systems [52], thus highlighting the prevalence and conservation of this mode of gene regulation which may serve as a paradigm for understanding other NUPs with similar mobility and DNA interaction profiles.
NUP98 was shown to recruit methylating SET1-containing complexes to promoter regions as a paradigm of gene activation in the setting of hematopoiesis [53], and was further supported by independent demonstration of NUP98-mediated enhancer-promoter looping occurring at activated genes [54]. Indirect modes of chromatin interaction by NUPs have also been reported. Earlier work by Kehat et al sought to identify binding partners of HDAC4, given its role in cardiac hypertrophy [55]. In this manner, NUP155 was identified as a direct binding partner of HDAC4, and demonstrated the ability to control chromatin localization and regulate both activation and repression of key genes associated with cardiac hypertrophy [43].
While it is recognized that disruption of homeostatic nuclear transport underlies a diversity of pathophysiological conditions [56], recognition of the expanded functional repertoire for other transport machinery like NUPs is critical for understanding their potential pathognomonic role. This is of particular importance for polygenic disorders such as cardiovascular pathologies, as improved knowledge of disease etiology may provide novel opportunities for therapeutic intervention [57–59]. Here we provide an overview regarding the role of NUPs in normal cardiac development, and summarize dysregulated dynamics for NUPs that have been associated with diverse cardiopathologies.
2. NUPs in Cardiovascular Development
The first data suggesting a role for NUPs in cardiac development was provided by reports of congenital heart defects associated with specific NUP expression disorders. Specifically, NUPs can act distal and independent of the nuclear pore affecting the establishment of cardiac asymmetry during embryonic development [96]. In a study focused on examining copy number variations in patients with heterotaxia and cardiac asymmetry disorders, a gene duplication of NUP188 was discovered that demonstrated significant heart looping defects when modeled in Xenopus [97]. Subsequent work identified impairment of ciliogenesis dependent on NUP188 and its binding partner NUP93. As cardiac looping is critically dependent on proper ciliogenic dynamics [98], the role of the NUP93/NUP188 complex in determining vertebrate laterality and proper asymmetric morphogenesis through its effects on cilia regulation established a precedent for consideration of NUPs in cardiac development. This was followed by the identification and investigation of other NUPs, such as NUP98, NUP205, and NUP210 in establishing proper cardiac asymmetry [96, 99–101].
Among these NUPs, mechanisms have been elucidated in part for NUP188, its binding partner NUP93, and NUP98. For the former, genetic and super resolution imaging studies identified NUP93/NUP188 organization into barrel structures comprising the pericentriolar material that encases the centriolar core at the ciliary base [96]. Furthermore, the observation that knockdown of NUP93 and/or NUP188 led to embryo-wide cilia loss provided evidence in support of a role for both NUPs in ciliogenesis and ultimately on cardiac laterality [96]. NUP98 has an active role in controlling ciliary length by actively gating soluble tubulin efflux out of the ciliary base [101]. In addition, localization of NUP98 to the ciliary base is controlled by NEK2-mediated phosphorylation, such that phosphorylation insensitive NUP98 mutants were found with higher frequency in the ciliary base [100]. Altered NUP98 concentrations at this location ultimately affects tubulin transport dynamics into and out of the ciliary matrix that regulates cilia length [101]. These data are currently the only evidence for a specific function for NUPs in developmental cardiac phenotypes, and remains an exciting and emerging field. Intriguingly, the bulk of existing and contemporary data has pointed to potential roles for NUPs in cardiac diseases (Figure 2), allowing for studies of a novel etiology for cardiopathogenesis.
Figure 2.
Nucleoporins associated with cardiac disorders. Representative illustration of nucleoporins associated with cardiac disorders and their localization within the nuclear pore complex, with each group highlighted in different colors. Nucleoporins associated with dilated and ischemic cardiomyopathy are highlighted in pink (NUP358, NUP62, NDC1, NUP93, NUP160, NUP153), valve disorders in blue (NUP43, NUP188), congestive heart failure (HF) in green (NUP85), heterotaxy and situs inversus in purple (NUP188, NUP205, NUP210) and arrhythmias in orange (NUP155, NUP107, NUP37, NUP153)
3. Cardiovascular Diseases and Nucleoporin Dynamics
3.1. Nucleoporins in Arrhythmias
The role of nucleoporins in arrhythmogenesis has been implicated by clinical as well as basic research studies, as shown in Figure 2 and Table 2 [5, 102, 103]. The first evidence of nucleoporins being involved in arrhythmogenesis was suggested by linkage mapping studies on an isolated South American community in which sudden cardiac death associated with atrial fibrillation (AF) was observed for pediatric cohorts from several generations within the same family [5]. Analysis revealed mutation in the NUP155 gene that resulted in an Arg to His substitution at amino acid position 391 (R391H). While heterozygous expression of this mutation in carriers were reported to have normal phenotypes without any cardiac complications, offspring with homozygous expression died perinatally from arrhythmogenic impairment [5, 108]. Follow up studies in mouse models demonstrated comparable electrophysiological abnormalities heterozygously expressing a truncated form of NUP155 [5]. Furthermore, cursory examination of cellular phenotypes revealed dysregulated NUP155 localization, suggesting that its function as part of the nuclear transport machinery could be compromised. Nuclear import was impaired in these cells, thus it was concluded that deficiencies in the nuclear transport of HSP70 and its cognate mRNA could be responsible for the electrophysiological deficits observed both in the experimental model and in the patient cohort [5]. In another clinical report investigating the molecular pathology underlying sudden unexplained nocturnal death syndrome (SUNDS), a rare NUP155 variant of uncertain significance resulting in a Leu to Phe change (L503F) was discovered in a 26 year old male reported to have died from atrial fibrillation associated with SUNDS [102]. Postmortem autopsy revealed no significant morphological differences between case and control hearts, aside from a slight but statistically relevant increase in myocardial size for SUNDS patients. Of note, for both studies that reported AF-associated NUP155 gene disruption, no evidence of abnormal myocardial geometry or deficits in cardiomyocyte ultrastructure were reported.
Table 2.
List of nucleoporins associated with cardiovascular disorders. Each nucleoporin is depicted with their respective gene and/or protein alteration associated with the respective cardiopathology and related reference.
Cardiopathology | Gene Name | Description | Gene/protein alteration | Reference |
---|---|---|---|---|
Arrhythmias | NUP155 | Nucleoporin 155 | missense mutation (R391H, L503F) | [5, 42, 102] |
NUP107 | Nucleoporin 107 | disrupted export of scn5a | [103] | |
NUP37 | Nucleoporin 37 | nonsense mutation | [2] | |
NUP153 | Nucleoporin 153 | protein overexpression | [3] | |
Dilated Cardiomyopathy | NDC1 | NDC1 transmembrane nucleoporin | protein overexpression | [4] |
NUP160 | Nucleoporin 160 | protein overexpression | [4] | |
NUP93 | Nucleoporin 93 | protein overexpression | [4] | |
NUP153 | Nucleoporin 153 | protein overexpression | [3, 4, 104] | |
NUP358/RANBP2 | RAN binding protein 2 | gene down regulation | [104] | |
Ischemic Cardiomyopathy | NDC1 | NDC1 transmembrane nucleoporin | protein overexpression | [4] |
NUP160 | Nucleoporin 160 | protein overexpression | [4] | |
NUP153 | Nucleoporin 153 | protein overexpression | [3, 4, 104] | |
NUP358/RANBP2 | RAN binding protein 2 | gene down regulation | [104] | |
NUP62 | Nucleoporin 62 | protein changes | [105, 106] | |
Congestive Heart Failure | NUP85/FROUNT | Nucleoporin 85 | protein overexpression and cytosolic mislocalization | [107] |
Valve, vascular disorders | NUP188 | Nucleoporin 188 | Splice-donor site mutation, gene duplication | [2, 96] |
NUP43 | Nucleoporin 43 | nonsense mutation | [2] | |
Heterotaxy/Situs inversus | NUP205 | Nucleoporin 205 | missense mutation | [99] |
NUP210 | Nucleoporin 210 | missense mutation | [99] | |
NUP188 | Nucleoporin 188 | gene duplication | [97] |
Systems biology analysis of a heterozygous nup155 exon-trapped mouse embryonic stem (mES) cell line revealed potential developmental susceptibility to developing AF [42]. In this study, transcriptome remodeling occurred in mES cells that expressed truncated NUP155 as a result of the heterozygous nup155 exon trap and significant re-prioritization of Gene Ontology Consortium (GOC)-defined cell membrane interactions and functions occurred in compromised cells. Furthermore, differentiation of the NUP155 truncated mES cells into beating embryoid bodies revealed cardiomyocyte-containing contractile foci that recapitulated the electrophysiological deficits previously reported [5, 42]. Significantly, NUP155-deficient cells exhibited severely dysregulated Ca2+-handling, and failed to mount a proper physiological response to β-agonist treatment [42]. This systems biology analysis, together with previous clinical reports as well as independent mechanistic evidence implicating the role of NUP155 in regulating cardiac hypertrophy [43], provide intriguing evidence in support of future work to discern the potential role of NUP155 in cardiac biology. Identifying other cardiopathological variants of NUP155, as well as recapitulating these in translational human induced pluripotent stem cell models would provide clear evidence of a role for NUP155 in electrical cardiac disorders. Broadly, this would have implications for understanding idiopathic cases of AF as well as defining a novel etiology for arrhythmogenesis.
A recent animal model study by Guan and colleagues showed that increased mRNA expression of Nup107 was associated with an enhanced susceptibility to postmyocardial infarction cardiac arrhythmias [103]. They demonstrated that Nup107 regulates cardiac electrical activity by facilitating the posttranscriptional transport of Scn5a (sodium voltage-gated channel alpha subunit 5) mRNA to increase Nav1.5 protein after cardiac injury, without any effect seen in other ion channels. The increase in expression of Nup107 in diseased cardiac tissue was also reported by a previous study documenting significant Y-complex overexpression in infarcted rat myocardial tissue [80]. The Y-complex consists of NUP160, NUP133, NUP107, NUP98/96, NUP85, NUP43, NUP37, SEC13, and SEH1, and is essential for NPC assembly and mRNA transport as well as regulating microtubule polymerization at kinetochores [9, 109–112].
Whole exome sequencing (WES) analyses in patients with severe cardiovascular disease identified novel disease-causing variants in NUP37, NUP43 and NUP188 [2]. Namely, this study associated clinical atrial fibrillation and sudden cardiac death with a heterozygous nonsense variant of NUP37. Functional examination of this mutation in a zebrafish model revealed development of arrhythmias, heart failure and heart chamber abnormalities including an abnormal atrium. The other two identified variants, NUP43 and NUP188, were associated with a strong family history of valve and vascular disorders that included mitral regurgitation, mitral valve prolapse and thoracic aortic dilation [2].
3.2. Nucleoporins in Cardiomyopathy
Evidence also indicates potential associations of several NUPs with inherited cardiomyopathies (Figure 2 and Table 2). Tarazon and colleagues published work that reported patients with heart failure (HF) with specific NUP expression changes [4]. In their study, they reported on NUPs localized to distinct regions within the NPC, i.e. TPR and NUP153 for the nucleoplasmic region, NUP93 and NUP155 that represented the inner ring subcomplex, NUP160 from the Y-subcomplex, and NDC1 from the integral membrane proteins [4], and identified increased protein expression of NDC1, NUP160, NUP153, and NUP93 in HF patients. In addition to significant increases in NUP protein expression, NDC1 was observed to distribute within the nucleoplasm under cardiomyopathic conditions [4], in contrast to its well-established localization as a transmembrane NUP anchoring the NPC to the nuclear envelope [113].
In recent studies, a novel correlation between increased expression of NUP153 has been associated with compromised ventricular function in ischemic cardiomyopathy [114]. Other studies have identified NUP153 transcripts as deregulated in dilated cardiomyopathy [104] though the function of up regulated NUP153 in cardiac ischemia is not well understood [4]. Some mechanistic clues have been provided by a study carried out by Nanni et al in which NUP153 and its potential role as a mediator of nitric oxide (NO) altered signing in the setting of cardiac dysfunction was examined [3]. The authors hypothesized that NUP153-regulated NO signaling would drive epigenetic alterations that contribute to development of cardiac dysfunction. The authors discovered that post NO deregulation, acetylated regulation of NUP153 overexpression occurred in dystrophic cardiac muscle. Furthermore, this process controlled a set of genes involved in cardiac dysfunction that included: nexilin (nexn), versican (vcan), adrenergic receptor alpha 2a (adra2), and cAMP responsive element modulator (crem). This study was the first to identify novel roles for NUP153 in dystrophic cardiac pathology. Mechanistically, the authors reported several novel findings in support of their conclusion that post-translational acetylation of NUP153 is regulated due to NO and oxidative stress. First, NUP153 modification significantly reduced P300/CBP-associated factor (PCAF) and p300 specific activities [3]. This stress-mediated regulation of NUP153, and its role as an epigenetic regulator, was a novel characteristic of the dystrophic heart. Second, the study reported lysine acetyltransferases (KATs) and NUP153 as binding partners in hearts of Dmdmdx mice, an established model of Duchenne’s muscular dystrophy (DMD). In comparison, the expression of vcan, adra2, nexn, and crem were all deregulated in the presence of acetylated NUP153 in 5-month old Dmdmdx mice. This finding suggests that these conditions could be associated with a pathological program of cardiac remodeling. Third, Nanni et al used cardiomyocytes derived from induced pluripotent stem cells (iPSC) isolated from DMD patients. Their finding showed that cardiomyocytes derived iPSC had increased NUP153 protein expression and acetylation, plus abnormal expression and distribution levels of nexilin. With this, Nanni et al provided evidence for NUP153 and its role in altered NO signaling, with important epigenetic implications for gene regulation in dystrophic hearts. Importantly, this study identifies a potential therapeutic target for cardiomyopathy and other cardiac disease related to NO dysfunction [3].
3.3. Nucleoporins in other cardiovascular diseases
3.3.1. Cardiac Disorders
Other cardiovascular disorders, besides arrhythmias and cardiomyopathies, have been linked to nucleoporin dysregulation. Congestive heart failure (CHF) has been clinically associated with nucleoporin 85 (FROUNT) [107]. NUP85 was thought to be correlated with C-C chemokine receptor 2 (CCR2) and was associated with a function in the chemokine system, which is involved in the development of CHF [107]. In the study presented by Satoh and colleagues, they examined biopsied tissues obtained from CHF and non-HF patients. The team found that the levels of CCR2 and CCL2 mRNA were closely correlated to NUP85 mRNA levels in patients with CHF. The levels of NUP85 mRNA in relation to CCR2 and CCL2 were also found to be related to left ventricle systolic dysfunction [107]. They concluded that the levels of NUP85, CCR2, and CCL2 were higher in patients with severe, compared to mild, CHF.
Recently, clinical mutations in NUP205 and NUP210 were identified as potential etiological determinants underlying heterotaxy and situs inversus totalis (SIT) [99]. In this study, whole exome and genome sequencing was performed using samples from patients with heterotaxy and SIT in which the authors reported several bi-allelic missense mutations for NUP205 and NUP210 [99]. Generation of induced pluripotent stem cells for functional analysis from the patient expressing the bi-allelic NUP205 T1044M/P1610R mutation revealed that NUP205 did not localize to the nuclear envelope. Furthermore, mutant NUP205 demonstrated impaired binding with its cognate inner ring subcomplex partners as well as a diminished interaction with the nuclear export factor XPO1 [99].
Left-right asymmetry deficits have been associated with dysregulated nucleoporin-driven mechanisms. In this regard, NUP98 interacts with NEK2 kinase to regulate cilia synthesis and resorption dynamics that ultimately affects proper cardiac looping [100, 101]. In a departure from nuclear transport-related modes of action, NUP98 located at the ciliary base stabilizes the cilium and prevents NEK2-mediated resorption. Phosphorylation of NUP98 by NEK2 is proposed to be the main protein modification of NUP98 that regulates both cilia resorption and nuclear envelope breakdown at the G2/M transition that ultimately affects cardiac looping.
Other NUPs at the nuclear envelope also contribute to proper myocardial asymmetry, with a number of clinical instances underscoring the role of NUPs in normal cardiac development. Within the context of cardiopathology, a duplication of NUP188 was reported for patients who presented with heterotaxy sampled from a population that presented phenotypes of right atrial isomerism, left atrial isomerism, and transposition of the great arteries [97]. When modeled in Xenopus, NUP188 depletion, or knockdown of its binding partner NUP93, led to impaired cilia formation and abnormal heart patterning in the embryos downstream of aberrant cardiac looping [96]. Though the experimental approach diminishing Xenopus NUP188 expression did not recapitulate clinical NUP188 overexpression, the authors concluded that abnormal NUP188 expression in general impaired ciliogenesis that disrupted cardiac looping and proper establishment of left-right asymmetry [96].
The association of cardiac asymmetry defects with NUP205, NUP188, and NUP93 is unsurprising given the interaction of these proteins in the NPC. [18, 81] Along with NUP155 and NUP35, these five nucleoporins form the inner ring subcomplex of the NPC. [18] Interestingly, although no cardiac developmental symmetry problems have been reported for NUP155 or NUP35, there have been cardiac phenotypes associated with these NUPs. Arrhythmogenic deficits are associated with NUP155 as noted in the present review, while cardiac ischemia reperfusion injury has been reported to involve dysregulation of NUP35 via its ability to regulate expression of the sodium-hydrogen exchanger NHE1 in cardiomyocytes [80].
NHE1 is critical to the pathogenesis of cardiac ischemia-reperfusion injury [115]. In brief, increased cytosolic acidification during cardiac ischemia stimulates sodium hydrogen exchange, bringing in one Na+ while extruding one H+, with acute increases in activity triggered by internal pH levels less than 7.2 [116]. This sodium influx is functionally coupled to the electrogenic Na+/Ca2+ exchanger that moves three Na+ out of the cell for uptake of one Ca2+ and whose different isoforms can confer variable susceptibilities to calcium overload in the subsequent reperfusion phase [117, 118]. This can lead to paradoxical increases in diastolic length of the cardiomyocyte, impaired contractility, and cardiomyocyte death [119–122]. Within the molecular sequelae of ischemia reperfusion injury, it was recently discovered that NUP35 post-transcriptionally regulates NHE1 expression [80]. Mechanistically this occurs by interaction of an Nterminal RNA recognition motif (RRM) within NUP35 with the 5’-UTR of NHE1 mRNA to promote its nuclear export efficiency and availability for translation. It was also observed that in an animal model of acute myocardial infarction, concomitant down regulation of NUP35 and NHE1 was observed 3 days following following ischemic injury. Overexpression of NUP35 rescued ischemia-induced downregulation, in further support of a potential role for NUP35 in regulating cardiomyocyte pH homeostasis. Indeed, other work has identified NUP35 as a bona fide cardiac RNA binding protein defined by a classic RNA binding domain within its RRM [123]. Given this, it is likely that NUP35 has the ability to bind a variety of isoforms and RNAs other than NHE1, and may function in this capacity in other tissues.
Cardiac disorders that manifest as secondary phenotypes within a syndrome related to nucleoporin expression disorders have been reported. For example, dysregulation or mutation in AAAS leads to Triple A syndrome in which patients present with poor heart rate variability that is a result of overall dysautonomia in affected individuals [124]. In another example, myocarditis associated with Coxsackievirus B3 (CVB3) infection was proposed to occur as a result of virus-mediated inhibition of NUP98 expression that stimulated Th17 cell differentiation [125]. It is clear from these examples that the causal relationship between heart disease and nucleoporin biology is more complicated than a source-to-sink model of disease etiology. With recognition of the expanded functional repertoire of individual NUPs, it is thus critical to include investigations of non-transport modes of regulation as potential mechanisms that when disrupted give rise to heart disease.
3.3.2. Vascular Disorders
A role for NUPs in vascular disorders has been reported, where they may function to drive vascular smooth muscle cell proliferation (VSMC) in the setting of neointimal hyperplasia [126]. Similar to nucleoporin dynamics in cases of clinical cardiac disease, overexpression of NUP62 and NUP153 were observed in an animal model of stress-induced VSMC proliferation. This is corrobrated by independent work showing that inhibition of NUP153 inhibits angiogenesis in a HUVEC culture model [127]. Additional supporting evidence demonstrates that increases in VSMC growth, import dynamics and NUP expression occurs in response to mechanical stimulation [13] and oxidized LDL exposure [128, 129]. Furthermore, mislocalized NUP153 is associated with aging VSMCs [130] that may be facilitated in part by NUP-mediated effects on the RAN gradient critical for nucleocytoplasmic transport [131]. In these aging cells, NUP153 was observed to relocalize away from the nuclear envelope into the nucleoplasmic interior. It is unclear if the loss of critical interactions at the nuclear envelope or binding to chromatin or protein within the nuclear interior is the main consequence of increased nucleoplasmic localization. Given that intranuclear mobility is a normal function and characteristic of NUP153, it may reflect a dysregulation in what NUP153 interacts with. These emerging data point to potential and specific functional roles for nucleoporins in vascular development and disease that remains to be studied further.
4. NUPs, -omics, and cardiac systems biology
Pioneering DamID studies were among the first high throughput approaches to identify chromatin wide occupation of model genomes by various NUPs. Early work in Drosophila revealed that NUPs bound genes discretely and distally from the nuclear periphery, indicating a genomic occupancy function for each of these NUPs [20, 52]. As an example, SEC13 and NUP98 enriched at transcriptionally active sites in accordance with developmental timing, while NUP88 occupied silent loci, supporting the notion of NUP-specific functions in the context of genome dynamics [20]. Since these initial reports, independent work has identified chromatin looping mechanisms [49, 51], nucleoporin mobility [23, 53, 132], and NUP-mediated complex recruitment and formation [44, 53] related to development and disease signaling. It is conceivable that NUPs engage a diversity of regulatory epigenomic and transcriptomic modalities, thus offering researchers the opportunity to leverage the power of high throughput technology to understand the global effects of NUP function on gene and protein expression.
In this regard, present work has examined NUPs in the context of epigenomic regulation, where NUPs bind to and regulate chromatin access via direct or indirect interactions. ChIP-seq, or variations thereof, are widely used in these studies, which has helped to identify genomic enrichment of a variety of NUPs in different developmental contexts [23, 43–45, 53, 88]. Importantly, technical and analytical caveats associated with earlier approaches can be addressed and refined with advancements in sample preparation and bioinformatic algorithms [133–135]. In one approach, to circumvent limitations associated with a lack of antibody specificity for myocardin, CRISPR-based CETCh-seq was used to identify myocardin genome occupancy profiles [135]. This is particularly powerful when applied to non-canonical genomic regulators such as NUPs that may not have optimized antibodies for ChIP-seq. Aligned with these emerging epigenomic studies are transcriptome remodeling investigations, in which differential transcriptome signatures can be profiled to detect systemic RNA expression differences. RNA based approaches, e.g. RNA-seq, PAR-CLIP, and GRO-seq, have been used to provide insight into cardiac specific transcriptome signatures, as well as sub-transcriptome composition and dynamics [136–139]. In light of recent studies that identify a subset of NUPs with defined RNA binding domains, these approaches are relevant as they may further define the role of NUPs in differential RNA metabolism [123, 140].
Complementing these genomic and transcriptomic approaches are proteomic studies that inform on the cellular machinery coded by the DNA/RNA milieu. In this regard, governing proteomic composition is in line with the canonical functions of NUPs as regulators of nucleocytoplasmic transport. Changes in NUP expression or functionality directly impact the nuclear pore complex and its function as a macromolecular transporter, and by extension molecular cargo dynamics within the cell. In support of this, the discovery of cell- and tissue- specific NPC composition and NUP isoforms argues in favor of differential transport that can facilitate proteomic remodeling [141, 142].
Integrating these multi-omic datasets is fundamental to understanding the global regulatory roles of candidate molecules in the context of cardiac development and disease. The application of network theory in this context offers opportunities to consolidate disparate (epi)genomic, transcriptomic, and proteomic datasets for a comprehensive understanding of cardiac health and disease systems biology [143–146]. Network, or graph theory, facilitates identification of molecules key to the function of the overall system, with the potential to prioritize therapeutically tractable targets [147–151]. This is particularly relevant for individualized medicine approaches, where patient variability can render standard treatments less effective [152]. In the context of investigating potential substrates for idiopathic cardiovascular disease such as NUPs, systems biology approaches identify global nuances that cannot be detected by traditional single pathway analyses. Ultimately, understanding these molecular processes from an integrated perspective unifies existing reductionist knowledge that facilitates the identification of potentially powerful interventional targets and advances overall understanding of the system as a whole [153, 154].
5. Conclusion
The NPC regulates development and disease of multiple systems via a variety of potential mechanisms. Novel studies are improving our knowledge of these processes in the context of cardiovascular disease, although many aspects of how the NPC does so, and to what extent, remain uncharacterized. To date, the dozen or so clinically reported NUP dysregulations associated with cardiac disorders may act through canonical transport dysfunction. However, evidence suggests that other epigenomic mechanisms may facilitate the development and progression of disease and understanding these will be critical to devising novel therapeutic measures that address the polygenic etiology of cardiovascular disorders. Furthermore, NUP dynamics and their association with regulatory networks is facilitated by the ongoing development of increasingly sophisticated approaches for high throughput molecular profiling. This will be key to understanding the intricate molecular foundation that predisposes and facilitates cardiopathogenesis.
Cardiovascular disease is a confounding disorder with a multigenic basis. While a number of clinical studies have demonstrated consistent nucleoporin derangement associated with disease, it remains uncharacterized whether or not nucleoporin dysregulation occurs as a cause or effect of cardiovascular pathology. Evidence for NUP malfunction as the underlying cause for cardiac disease is strongly supported by current data. However, considering the spectrum of inherited and acquired cardiac disorders associated with nucleoporin dysfunction, the possibility that NUP derangements follow cardiac insult cannot be ruled out. NUPs may act within a feedback loop as both effector and marker of cardiac disease, with an increased susceptibility to cardiopathology imposed by NUPs harboring genetic lesions. More studies in this regard, with experiments designed to specifically target NUP function in an exclusive cardiac or vascular context will be required to move forward in this field. Ultimately, identification and characterization of NUPs as regulators of CVD will provide insights into the intricate mechanisms that gives rise to cardiac and vascular pathologies, and will provide novel targets with which to treat cardiovascular disorders. Understanding NUP-driven mechanisms that contribute to disease development is paramount to developing advanced therapeutic strategies for improvement of patient longevity and quality of life, with concomitant mitigation of the social and financial burden of pervasive cardiovascular disease.
Highlights.
The nuclear pore complex regulates development and disease of multiple systems via a variety of mechanisms.
Identification and consistent association of nucleoporin dysfunction with clinical cardiac diseases parallels nucleoporin roles in cardiac disease etiology.
Nucleoporins have potential roles in cardiomyopathy, arrhythmogensis, and congenital heart disease.
High throughput molecular profiling is paramount to understanding nucleoporin association substrates that cause cardiopathogenesis and other disorders.
Acknowledgements
This work was supported by funding from Sanford Research and the NIH Center for Pediatric Research COBRE grant (P20GM103620).
Footnotes
Conflict of Interest Statement
The authors declare no conflict of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Puckelwartz MJ The Missing LINC for Genetic Cardiovascular Disease? Circ Cardiovasc Genet 2017; 10: e001793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Haskell GT, Jensen BC, Samsa LA, Marchuk D, Huang W, Skrzynia C, et al. Whole Exome Sequencing Identifies Truncating Variants in Nuclear Envelope Genes in Patients With Cardiovascular Disease. Circ Cardiovasc Genet 2017; 10: e001443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Nanni S, Re A, Ripoli C, Gowran A, Nigro P, D’Amario D, et al. The nuclear pore protein Nup153 associates with chromatin and regulates cardiac gene expression in dystrophic mdx hearts. Cardiovasc Res. 2016; 112: 555–567. [DOI] [PubMed] [Google Scholar]
- [4].Tarazon E, Rivera M, Rosello-Lleti E, Molina-Navarro MM, Sanchez-Lazaro IJ, Espana F, et al. Heart failure induces significant changes in nuclear pore complex of human cardiomyocytes. PLoS One. 2012; 7: e48957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Zhang X, Chen S, Yoo S, Chakrabarti S, Zhang T, Ke T, et al. Mutation in nuclear pore component NUP155 leads to atrial fibrillation and early sudden cardiac death. Cell. 2008; 135: 1017–27. [DOI] [PubMed] [Google Scholar]
- [6].Donnaloja F, Jacchetti E, Soncini M, Raimondi MT Mechanosensing at the Nuclear Envelope by Nuclear Pore Complex Stretch Activation and Its Effect in Physiology and Pathology. Front Physiol 2019; 10: 896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Callan HG, Tomlin SG Experimental studies on amphibian oocyte nuclei. I. Investigation of the structure of the nuclear membrane by means of the electron microscope. Proc R Soc Lond B Biol Sci. 1950; 137: 367–78. [DOI] [PubMed] [Google Scholar]
- [8].von Appen A, Beck M Structure Determination of the Nuclear Pore Complex with ThreeDimensional Cryo electron Microscopy. J Mol Biol. 2016; 428: 2001–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].D’Angelo MA, Hetzer MW Structure, dynamics and function of nuclear pore complexes. Trends Cell Biol. 2008; 18: 456–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Perez-Terzic C, Faustino RS, Boorsma BJ, Arrell DK, Niederlander NJ, Behfar A, et al. Stem cells transform into a cardiac phenotype with remodeling of the nuclear transport machinery. Nat Clin Pract Cardiovasc Med. 2007; 4 Suppl 1: S68–76. [DOI] [PubMed] [Google Scholar]
- [11].Faustino RS, Pierce GN Nucleocytoplasmic trafficking. Curr Res Cardiol. 2016; 3: 43–52. [Google Scholar]
- [12].Kapinos LE, Huang B, Rencurel C, Lim RYH Karyopherins regulate nuclear pore complex barrier and transport function. J Cell Biol. 2017; 216: 3609–3624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Richard MN, Deniset JF, Kneesh AL, Blackwood D, Pierce GN Mechanical stretching stimulates smooth muscle cell growth, nuclear protein import, and nuclear pore expression through mitogen-activated protein kinase activation. J Biol Chem. 2007; 282: 23081–8. [DOI] [PubMed] [Google Scholar]
- [14].Kim SJ, Fernandez-Martinez J, Nudelman I, Shi Y, Zhang W, Raveh B, et al. Integrative structure and functional anatomy of a nuclear pore complex. Nature. 2018; 555: 475–482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Hampoelz B, Andres-Pons A, Kastritis P, Beck M Structure and Assembly of the Nuclear Pore Complex. Annu Rev Biophys. 2019; 48: 515–536. [DOI] [PubMed] [Google Scholar]
- [16].von Appen A, Kosinski J, Sparks L, Ori A, DiGuilio AL, Vollmer B, et al. In situ structural analysis of the human nuclear pore complex. Nature. 2015; 526: 140–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Chug H, Trakhanov S, Hulsmann BB, Pleiner T, Gorlich D Crystal structure of the metazoan Nup62*Nup58*Nup54 nucleoporin complex. Science. 2015; 350: 106–10. [DOI] [PubMed] [Google Scholar]
- [18].Kosinski J, Mosalaganti S, von Appen A, Teimer R, DiGuilio AL, Wan W, et al. Molecular architecture of the inner ring scaffold of the human nuclear pore complex. Science.2016; 352: 363–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Kalverda B, Pickersgill H, Shloma VV, Fornerod M Nucleoporins directly stimulate expression of developmental and cell-cycle genes inside the nucleoplasm. Cell. 2010; 140: 360–71. [DOI] [PubMed] [Google Scholar]
- [20].Capelson M, Liang Y, Schulte R, Mair W, Wagner U, Hetzer MW Chromatin-bound nuclear pore components regulate gene expression in higher eukaryotes. Cell. 2010; 140: 372–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Hampoelz B, Andres-Pons A, Kastritis P, Beck M Structure and Assembly of the Nuclear Pore Complex. Annu Rev Biophys. 2019. [DOI] [PubMed] [Google Scholar]
- [22].Fernandez-Martinez J, Kim SJ, Shi Y, Upla P, Pellarin R, Gagnon M, et al. Structure and Function of the Nuclear Pore Complex Cytoplasmic mRNA Export Platform. Cell. 2016; 167: 1215–1228 e25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Buchwalter AL, Liang Y, Hetzer MW Nup50 is required for cell differentiation and exhibits transcription-dependent dynamics. Mol Biol Cell. 2014; 25: 2472–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Lelek M, Casartelli N, Pellin D, Rizzi E, Souque P, Severgnini M, et al. Chromatin organization at the nuclear pore favours HIV replication. Nat Commun. 2015; 6: 6483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Boumendil C, Hari P, Olsen KCF, Acosta JC, Bickmore WA Nuclear pore density controls heterochromatin reorganization during senescence. Genes Dev. 2019; 33: 144–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Perez-Garrastachu M, Arluzea J, Andrade R, Diez-Torre A, Urtizberea M, Silio M, et al. Nucleoporins redistribute inside the nucleus after cell cycle arrest induced by histone deacetylases inhibition. Nucleus. 2017; 8: 515–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Terry LJ, Wente SR Flexible gates: dynamic topologies and functions for FG nucleoporins in nucleocytoplasmic transport. Eukaryot Cell. 2009; 8: 1814–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Favreau C, Bastos R, Cartaud J, Courvalin JC, Mustonen P Biochemical characterization of nuclear pore complex protein gp210 oligomers. Eur J Biochem. 2001; 268: 3883–9. [DOI] [PubMed] [Google Scholar]
- [29].Yamazumi Y, Kamiya A, Nishida A, Nishihara A, Iemura S, Natsume T, et al. The transmembrane nucleoporin NDC1 is required for targeting of ALADIN to nuclear pore complexes. Biochem Biophys Res Commun. 2009; 389: 100–4. [DOI] [PubMed] [Google Scholar]
- [30].Soderqvist H, Hallberg E The large C-terminal region of the integral pore membrane protein, POM121, is facing the nuclear pore complex. Eur J Cell Biol. 1994; 64: 186–91. [PubMed] [Google Scholar]
- [31].Raices M, D’Angelo MA Nuclear pore complex composition: a new regulator of tissue-specific and developmental functions. Nat Rev Mol Cell Biol. 2012; 13: 687–99. [DOI] [PubMed] [Google Scholar]
- [32].Beck M, Hurt E The nuclear pore complex: understanding its function through structural insight. Nat Rev Mol Cell Biol. 2017; 18: 73–89. [DOI] [PubMed] [Google Scholar]
- [33].Prunuske AJ, Liu J, Elgort S, Joseph J, Dasso M, Ullman KS Nuclear envelope breakdown is coordinated by both Nup358/RanBP2 and Nup153, two nucleoporins with zinc finger modules. Mol Biol Cell. 2006; 17: 760–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Galy V, Antonin W, Jaedicke A, Sachse M, Santarella R, Haselmann U, et al. A role for gp210 in mitotic nuclear-envelope breakdown. J Cell Sci. 2008; 121: 317–28. [DOI] [PubMed] [Google Scholar]
- [35].Salina D, Enarson P, Rattner JB, Burke B Nup358 integrates nuclear envelope breakdown with kinetochore assembly. J Cell Biol. 2003; 162: 991–1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Romana S, Radford-Weiss I, Lapierre JM, Doye V, Geoffroy MC Formation of Nup98-containing nuclear bodies in HeLa sublines is linked to genomic rearrangements affecting chromosome 11. Chromosoma. 2016; 125: 789–805. [DOI] [PubMed] [Google Scholar]
- [37].Lang A, Eriksson J, Schink KO, Lang E, Blicher P, Polec A, et al. Visualization of PML nuclear import complexes reveals FG-repeat nucleoporins at cargo retrieval sites. Nucleus. 2017; 8: 404–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Baffet AD, Hu DJ, Vallee RB Cdk1 Activates Pre-mitotic Nuclear Envelope Dynein Recruitment and Apical Nuclear Migration in Neural Stem Cells. Dev Cell. 2015; 33: 703–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Jodoin JN, Shboul M, Albrecht TR, Lee E, Wagner EJ, Reversade B, et al. The snRNA-processing complex, Integrator, is required for ciliogenesis and dynein recruitment to the nuclear envelope via distinct mechanisms. Biol Open. 2013; 2: 1390–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Hu DJ, Baffet AD, Nayak T, Akhmanova A, Doye V, Vallee RB Dynein recruitment to nuclear pores activates apical nuclear migration and mitotic entry in brain progenitor cells. Cell. 2013; 154: 1300–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].D’Angelo MA, Gomez-Cavazos JS, Mei A, Lackner DH, Hetzer MW A change in nuclear pore complex composition regulates cell differentiation. Dev Cell. 2012; 22: 446–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Preston CC, Wyles SP, Reyes S, Storm EC, Eckloff BW, Faustino RS NUP155 insufficiency recalibrates a pluripotent transcriptome with network remodeling of a cardiogenic signaling module. BMC Syst Biol. 2018; 12: 62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Kehat I, Accornero F, Aronow BJ, Molkentin JD Modulation of chromatin position and gene expression by HDAC4 interaction with nucleoporins. J Cell Biol. 2011; 193: 21–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Toda T, Hsu JY, Linker SB, Hu L, Schafer ST, Mertens J, et al. Nup153 Interacts with Sox2 to Enable Bimodal Gene Regulation and Maintenance of Neural Progenitor Cells. Cell Stem Cell. 2017; 21: 618–634 e7. [DOI] [PubMed] [Google Scholar]
- [45].Jacinto FV, Benner C, Hetzer MW The nucleoporin Nup153 regulates embryonic stem cell pluripotency through gene silencing. Genes Dev. 2015; 29: 1224–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Kuhn TM, Capelson M Nuclear Pore Proteins in Regulation of Chromatin State. Cells. 2019; 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Gozalo A, Duke A, Lan Y, Pascual-Garcia P, Talamas JA, Nguyen SC, et al. Core Components of the Nuclear Pore Bind Distinct States of Chromatin and Contribute to Polycomb Repression. Mol Cell. 2020; 77: 67–81 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Krull S, Dorries J, Boysen B, Reidenbach S, Magnius L, Norder H, et al. Protein Tpr is required for establishing nuclear pore-associated zones of heterochromatin exclusion. EMBO J. 2010; 29: 1659–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Tan-Wong SM, Wijayatilake HD, Proudfoot NJ Gene loops function to maintain transcriptional memory through interaction with the nuclear pore complex. Genes Dev. 2009; 23: 2610–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Ibarra A, Benner C, Tyagi S, Cool J, Hetzer MW Nucleoporin-mediated regulation of cell identity genes. Genes Dev. 2016; 30: 2253–2258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Breuer M, Ohkura H A negative loop within the nuclear pore complex controls global chromatin organization. Genes Dev. 2015; 29: 1789–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Vaquerizas JM, Suyama R, Kind J, Miura K, Luscombe NM, Akhtar A Nuclear pore proteins nup153 and megator define transcriptionally active regions in the Drosophila genome. PLoS Genet. 2010; 6: e1000846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Franks TM, McCloskey A, Shokirev MN, Benner C, Rathore A, Hetzer MW Nup98 recruits the Wdr82-Set1A/COMPASS complex to promoters to regulate H3K4 trimethylation in hematopoietic progenitor cells. Genes Dev. 2017; 31: 2222–2234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Pascual-Garcia P, Debo B, Aleman JR, Talamas JA, Lan Y, Nguyen NH, et al. Metazoan Nuclear Pores Provide a Scaffold for Poised Genes and Mediate Induced Enhancer-Promoter Contacts. Mol Cell. 2017; 66: 63–76 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Weeks KL, Avkiran M Roles and post-translational regulation of cardiac class IIa histone deacetylase isoforms. J Physiol. 2015; 593: 1785–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Faustino RS, Stronger LN, Richard MN, Czubryt MP, Ford DA, Prociuk MA, et al. RanGAP-mediated nuclear protein import in vascular smooth muscle cells is augmented by lysophosphatidylcholine. Mol Pharmacol. 2007; 71: 438–45. [DOI] [PubMed] [Google Scholar]
- [57].Chahine MN, Pierce GN Therapeutic targeting of nuclear protein import in pathological cell conditions. Pharmacol Rev. 2009; 61: 358–72. [DOI] [PubMed] [Google Scholar]
- [58].Faustino RS, Rousseau DC, Landry MN, Kostenuk AL, Pierce GN Effects of mitogen-activated protein kinases on nuclear protein import. Can J Physiol Pharmacol. 2006; 84: 469–75. [DOI] [PubMed] [Google Scholar]
- [59].Faustino RS, Nelson TJ, Terzic A, Perez-Terzic C Nuclear transport: target for therapy. Clin Pharmacol Ther. 2007; 81: 880–6. [DOI] [PubMed] [Google Scholar]
- [60].Pichler A, Knipscheer P, Saitoh H, Sixma TK, Melchior F The RanBP2 SUMO E3 ligase is neither HECT- nor RING-type. Nat Struct Mol Biol. 2004; 11: 984–91. [DOI] [PubMed] [Google Scholar]
- [61].Kirsh O, Seeler JS, Pichler A, Gast A, Muller S, Miska E, et al. The SUMO E3 ligase RanBP2 promotes modification of the HDAC4 deacetylase. EMBO J. 2002; 21: 2682–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Pichler A, Gast A, Seeler JS, Dejean A, Melchior F The nucleoporin RanBP2 has SUMO1 E3 ligase activity. Cell. 2002; 108: 109–20. [DOI] [PubMed] [Google Scholar]
- [63].Miyauchi Y, Sakaguchi N, Okada T, Makishima M, Ozono K, Michigami T Oncogenic nucleoporin CAN/Nup214 interacts with vitamin D receptor and modulates its function. J Cell Biochem. 2009; 106: 1090–101. [DOI] [PubMed] [Google Scholar]
- [64].Carvalhal S, Ribeiro SA, Arocena M, Kasciukovic T, Temme A, Koehler K, et al. The nucleoporin ALADIN regulates Aurora A localization to ensure robust mitotic spindle formation. Mol Biol Cell. 2015; 26: 3424–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Adams RL, Mason AC, Glass L, Aditi, Wente S.R. Nup42 and IP6 coordinate Gle1 stimulation of Dbp5/DDX19B for mRNA export in yeast and human cells. Traffic. 2017; 18: 776–790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].von Kobbe C, van Deursen JM, Rodrigues JP, Sitterlin D, Bachi A, Wu X, et al. Vesicular stomatitis virus matrix protein inhibits host cell gene expression by targeting the nucleoporin Nup98. Mol Cell. 2000; 6: 1243–52. [DOI] [PubMed] [Google Scholar]
- [67].Griffis ER, Altan N, Lippincott-Schwartz J, Powers MA Nup98 is a mobile nucleoporin with transcription-dependent dynamics. Mol Biol Cell. 2002; 13: 1282–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Liu HL, De Souza CP, Osmani AH, Osmani SA The three fungal transmembrane nuclear pore complex proteins of Aspergillus nidulans are dispensable in the presence of an intact An-Nup84–120 complex. Mol Biol Cell. 2009; 20: 616–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Bilokapic S, Schwartz TU Molecular basis for Nup37 and ELY5/ELYS recruitment to the nuclear pore complex. Proc Natl Acad Sci U S A. 2012; 109: 15241–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Loiodice I, Alves A, Rabut G, Van Overbeek M, Ellenberg J, Sibarita JB, et al. The entire Nup107–160 complex, including three new members, is targeted as one entity to kinetochores in mitosis. Mol Biol Cell. 2004; 15: 3333–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Walther TC, Alves A, Pickersgill H, Loiodice I, Hetzer M, Galy V, et al. The conserved Nup107–160 complex is critical for nuclear pore complex assembly. Cell. 2003; 113: 195–206. [DOI] [PubMed] [Google Scholar]
- [72].Faria AM, Levay A, Wang Y, Kamphorst AO, Rosa ML, Nussenzveig DR, et al. The nucleoporin Nup96 is required for proper expression of interferon-regulated proteins and functions. Immunity. 2006; 24: 295–304. [DOI] [PubMed] [Google Scholar]
- [73].Zhang Y, Li X A putative nucleoporin 96 Is required for both basal defense and constitutive resistance responses mediated by suppressor of npr1–1,constitutive 1. Plant Cell. 2005; 17: 1306–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Enninga J, Levy DE, Blobel G, Fontoura BM Role of nucleoporin induction in releasing an mRNA nuclear export block. Science. 2002; 295: 1523–5. [DOI] [PubMed] [Google Scholar]
- [75].Wozniak RW, Goldfarb DS Cyclin-like oscillations in levels of the nucleoporin Nup96 control G1/S progression. Dev Cell. 2008; 15: 643–4. [DOI] [PubMed] [Google Scholar]
- [76].Vasu S, Shah S, Orjalo A, Park M, Fischer WH, Forbes DJ Novel vertebrate nucleoporins Nup133 and Nup160 play a role in mRNA export. J Cell Biol. 2001; 155: 339–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Niu X, Hong J, Zheng X, Melville DB, Knapik EW, Meng A, et al. The nuclear pore complex function of Sec13 protein is required for cell survival during retinal development. J Biol Chem. 2014; 289: 11971–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Platani M, Samejima I, Samejima K, Kanemaki MT, Earnshaw WC Seh1 targets GATOR2 and Nup153 to mitotic chromosomes. J Cell Sci. 2018; 131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Chen F, Jiao XF, Zhang JY, Wu D, Ding ZM, Wang YS, et al. Nucleoporin35 is a novel microtubule associated protein functioning in oocyte meiotic spindle architecture. Exp Cell Res. 2018; 371: 435–443. [DOI] [PubMed] [Google Scholar]
- [80].Xu L, Pan L, Li J, Huang B, Feng J, Li C, et al. Nucleoporin 35 regulates cardiomyocyte pH homeostasis by controlling Na+-H+ exchanger-1 expression. J Mol Cell Biol. 2015; 7: 476–85. [DOI] [PubMed] [Google Scholar]
- [81].Labade AS, Karmodiya K, Sengupta K HOXA repression is mediated by nucleoporin Nup93 assisted by its interactors Nup188 and Nup205. Epigenetics Chromatin. 2016; 9: 54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Seo HS, Blus BJ, Jankovic NZ, Blobel G Structure and nucleic acid binding activity of the nucleoporin Nup157. Proc Natl Acad Sci U S A. 2013; 110: 16450–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Van de Vosse DW, Wan Y, Lapetina DL, Chen WM, Chiang JH, Aitchison JD, et al. A role for the nucleoporin Nup170p in chromatin structure and gene silencing. Cell. 2013; 152: 969–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Hartono Hazawa M., Lim K.S., Dewi F.R.P., Kobayashi A., Wong R.W. Nucleoporin Nup58 localizes to centrosomes and mid-bodies during mitosis. Cell Div. 2019; 14: 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Rodriguez-Berriguete G, Granata G, Puliyadi R, Tiwana G, Prevo R, Wilson RS, et al. Nucleoporin 54 contributes to homologous recombination repair and post-replicative DNA integrity. Nucleic Acids Res. 2018; 46: 7731–7746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Sachani SS, Landschoot LS, Zhang L, White CR, MacDonald WA, Golding MC, et al. Nucleoporin 107, 62 and 153 mediate Kcnq1ot1 imprinted domain regulation in extraembryonic endoderm stem cells. Nat Commun. 2018; 9: 2795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Hashizume C, Moyori A, Kobayashi A, Yamakoshi N, Endo A, Wong RW Nucleoporin Nup62 maintains centrosome homeostasis. Cell Cycle. 2013; 12: 3804–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Franks TM, Benner C, Narvaiza I, Marchetto MC, Young JM, Malik HS, et al. Evolution of a transcriptional regulator from a transmembrane nucleoporin. Genes Dev. 2016; 30: 1155–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Funakoshi T, Clever M, Watanabe A, Imamoto N Localization of Pom121 to the inner nuclear membrane is required for an early step of interphase nuclear pore complex assembly. Mol Biol Cell. 2011; 22: 1058–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Kralt A, Jagalur NB, van den Boom V, Lokareddy RK, Steen A, Cingolani G, et al. Conservation of inner nuclear membrane targeting sequences in mammalian Pom121 and yeast Heh2 membrane proteins. Mol Biol Cell. 2015; 26: 3301–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Gomez-Cavazos JS, Hetzer MW The nucleoporin gp210/Nup210 controls muscle differentiation by regulating nuclear envelope/ER homeostasis. J Cell Biol. 2015; 208: 671–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Rabut G, Doye V, Ellenberg J Mapping the dynamic organization of the nuclear pore complex inside single living cells. Nat Cell Biol. 2004; 6: 1114–21. [DOI] [PubMed] [Google Scholar]
- [93].Raices M, Bukata L, Sakuma S, Borlido J, Hernandez LS, Hart DO, et al. Nuclear Pores Regulate Muscle Development and Maintenance by Assembling a Localized Mef2C Complex. Dev Cell. 2017; 41: 540–554 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Madrid AS, Mancuso J, Cande WZ, Weis K The role of the integral membrane nucleoporins Ndc1p and Pom152p in nuclear pore complex assembly and function. J Cell Biol. 2006; 173: 361–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Ullman KS, Shah S, Powers MA, Forbes DJ The nucleoporin nup153 plays a critical role in multiple types of nuclear export. Mol Biol Cell. 1999; 10: 649–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Del Viso F, Huang F, Myers J, Chalfant M, Zhang Y, Reza N, et al. Congenital Heart Disease Genetics Uncovers Context-Dependent Organization and Function of Nucleoporins at Cilia. Dev Cell. 2016; 38: 478–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Fakhro KA, Choi M, Ware SM, Belmont JW, Towbin JA, Lifton RP, et al. Rare copy number variations in congenital heart disease patients identify unique genes in left-right patterning. Proc Natl Acad Sci U S A. 2011; 108: 2915–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Dasgupta A, Amack JD Cilia in vertebrate left-right patterning. Philos Trans R Soc Lond B Biol Sci. 2016; 371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Chen W, Zhang Y, Yang S, Shi Z, Zeng W, Lu Z, et al. Bi-Allelic Mutations in NUP205 and NUP210 Are Associated With Abnormal Cardiac Left-Right Patterning. Circ Genom Precis Med. 2019; 12: e002492. [DOI] [PubMed] [Google Scholar]
- [100].Endicott SJ, Basu B, Khokha M, Brueckner M The NIMA-like kinase Nek2 is a key switch balancing cilia biogenesis and resorption in the development of left-right asymmetry. Development. 2015; 142: 4068–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Endicott SJ, Brueckner M NUP98 Sets the Size-Exclusion Diffusion Limit through the Ciliary Base. Curr Biol. 2018; 28: 1643–1650 e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Zhang L, Tester DJ, Lang D, Chen Y, Zheng J, Gao R, et al. Does Sudden Unexplained Nocturnal Death Syndrome Remain the Autopsy-Negative Disorder: A Gross, Microscopic, and Molecular Autopsy Investigation in Southern China. Mayo Clin Proc. 2016; 91: 1503–1514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Guan Y, Gao X, Tang Q, Huang L, Gao S, Yu S, et al. Nucleoporin 107 facilitates the nuclear export of Scn5a mRNA to regulate cardiac bioelectricity. J Cell Mol Med. 2019; 23: 1448–1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Molina-Navarro MM, Trivino JC, Martinez-Dolz L, Lago F, Gonzalez-Juanatey JR, Portoles M, et al. Functional networks of nucleocytoplasmic transport-related genes differentiate ischemic and dilated cardiomyopathies. A new therapeutic opportunity. PLoS One. 2014; 9: e104709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Chahine MN, Mioulane M, Sikkel MB, O’Gara P, Dos Remedios CG, Pierce GN, et al. Nuclear pore rearrangements and nuclear trafficking in cardiomyocytes from rat and human failing hearts. Cardiovasc Res. 2015; 105: 31–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Cortes R, Portoles M, Rosello-Lleti E, Martinez-Dolz L, Almenar L, Salvador A, et al. [Nuclear changes and p62 expression in ischemic and dilated cardiomyopathy]. Rev Esp Cardiol. 2007; 60: 1319–23. [DOI] [PubMed] [Google Scholar]
- [107].Satoh M, Akatsu T, Ishkawa Y, Minami Y, Nakamura M A novel activator of C-C chemokine, FROUNT, is expressed with C-C chemokine receptor 2 and its ligand in failing human heart. J Card Fail. 2007; 13: 114–9. [DOI] [PubMed] [Google Scholar]
- [108].Oberti C, Wang L, Li L, Dong J, Rao S, Du W, et al. Genome-wide linkage scan identifies a novel genetic locus on chromosome 5p13 for neonatal atrial fibrillation associated with sudden death and variable cardiomyopathy. Circulation. 2004; 110: 3753–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Morchoisne-Bolhy S, Geoffroy MC, Bouhlel IB, Alves A, Auduge N, Baudin X, et al. Intranuclear dynamics of the Nup107–160 complex. Mol Biol Cell. 2015; 26: 2343–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Mishra RK, Chakraborty P, Arnaoutov A, Fontoura BM, Dasso M The Nup107–160 complex and gamma-TuRC regulate microtubule polymerization at kinetochores. Nat Cell Biol. 2010; 12: 164–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Yang HJ, Haraguchi T, Hiraoka Y A nucleoporin that facilitates meiotic kinetochore reorganization. Cell Cycle. 2016; 15: 307–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Zuccolo M, Alves A, Galy V, Bolhy S, Formstecher E, Racine V, et al. The human Nup107–160 nuclear pore subcomplex contributes to proper kinetochore functions. EMBO J. 2007; 26: 1853–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Marelli M, Lusk CP, Chan H, Aitchison JD, Wozniak RW A link between the synthesis of nucleoporins and the biogenesis of the nuclear envelope. J Cell Biol. 2001; 153: 709–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Omar SA, Webb AJ, Lundberg JO, Weitzberg E Therapeutic effects of inorganic nitrate and nitrite in cardiovascular and metabolic diseases. J Intern Med. 2016; 279: 315–36. [DOI] [PubMed] [Google Scholar]
- [115].Hurtado C, Pierce GN Inhibition of Na(+)/H(+) exchange at the beginning of reperfusion is cardioprotective in isolated, beating adult cardiomyocytes. J Mol Cell Cardiol. 2000; 32: 1897–907. [DOI] [PubMed] [Google Scholar]
- [116].Garciarena CD, Youm JB, Swietach P, Vaughan-Jones RD. H(+)-activated Na(+) influx in the ventricular myocyte couples Ca(2)(+)-signalling to intracellular pH. J Mol Cell Cardiol. 2013; 61: 51–9. [DOI] [PubMed] [Google Scholar]
- [117].Ander BP, Hurtado C, Raposo CS, Maddaford TG, Deniset JF, Hryshko LV, et al. Differential sensitivities of the NCX1.1 and NCX1.3 isoforms of the Na+-Ca2+ exchanger to alpha-linolenic acid. Cardiovasc Res. 2007; 73: 395–403. [DOI] [PubMed] [Google Scholar]
- [118].Hurtado C, Prociuk M, Maddaford TG, Dibrov E, Mesaeli N, Hryshko LV, et al. Cells expressing unique Na+/Ca2+ exchange (NCX1) splice variants exhibit different susceptibilities to Ca2+ overload. Am J Physiol Heart Circ Physiol. 2006; 290: H2155–62. [DOI] [PubMed] [Google Scholar]
- [119].Hurtado C, Ander BP, Maddaford TG, Lukas A, Hryshko LV, Pierce GN Adenovirally delivered shRNA strongly inhibits Na+-Ca2+ exchanger expression but does not prevent contraction of neonatal cardiomyocytes. J Mol Cell Cardiol. 2005; 38: 647–54. [DOI] [PubMed] [Google Scholar]
- [120].Vassalle M, Lin CI Calcium overload and cardiac function. J Biomed Sci. 2004; 11: 542–65. [DOI] [PubMed] [Google Scholar]
- [121].Kalogeris T, Baines CP, Krenz M, Korthuis RJ Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol. 2012; 298: 229–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Ganguly R, Hasanally D, Stamenkovic A, Maddaford TG, Chaudhary R, Pierce GN, et al. Alpha linolenic acid decreases apoptosis and oxidized phospholipids in cardiomyocytes during ischemia/reperfusion. Mol Cell Biochem. 2018; 437: 163–175. [DOI] [PubMed] [Google Scholar]
- [123].Liao Y, Castello A, Fischer B, Leicht S, Foehr S, Frese CK, et al. The Cardiomyocyte RNA-Binding Proteome: Links to Intermediary Metabolism and Heart Disease. Cell Rep. 2016; 16: 1456–1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Gazarian M, Cowell CT, Bonney M, Grigor WG The “4A” syndrome: adrenocortical insufficiency associated with achalasia, alacrima, autonomic and other neurological abnormalities. Eur J Pediatr. 1995; 154: 18–23. [DOI] [PubMed] [Google Scholar]
- [125].Long Q, Liao YH, Xie Y, Liang W, Cheng X, Yuan J, et al. Coxsackievirus B3 Directly Induced Th17 Cell Differentiation by Inhibiting Nup98 Expression in Patients with Acute Viral Myocarditis. Front Cell Infect Microbiol 2016; 6: 171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Deniset JF, Hedley TE, Hlavackova M, Chahine MN, Dibrov E, O’Hara K, et al. Heat shock protein 60 involvement in vascular smooth muscle cell proliferation. Cell Signal. 2018; 47: 44–51. [DOI] [PubMed] [Google Scholar]
- [127].Kim NH, Pham NB, Quinn RJ, Shim JS, Cho H, Cho SM, et al. The Small Molecule R-(−)-beta-O-Methylsynephrine Binds to Nucleoporin 153 kDa and Inhibits Angiogenesis. Int J Biol Sci. 2015; 11: 1088–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Chahine MN, Blackwood DP, Dibrov E, Richard MN, Pierce GN Oxidized LDL affects smooth muscle cell growth through MAPK-mediated actions on nuclear protein import. J Mol Cell Cardiol. 2009; 46: 431–41. [DOI] [PubMed] [Google Scholar]
- [129].Chahine MN, Dibrov E, Blackwood DP, Pierce GN Oxidized LDL enhances stretch-induced smooth muscle cell proliferation through alterations in nuclear protein import. Can J Physiol Pharmacol. 2012; 90: 1559–68. [DOI] [PubMed] [Google Scholar]
- [130].Cobb AM, Larrieu D, Warren DT, Liu Y, Srivastava S, Smith AJO, et al. Prelamin A impairs 53BP1 nuclear entry by mislocalizing NUP153 and disrupting the Ran gradient. Aging Cell. 2016; 15: 1039–1050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Vorpahl M, Schonhofer-Merl S, Michaelis C, Flotho A, Melchior F, Wessely R The Ran GTPase-activating protein (RanGAP1) is critically involved in smooth muscle cell differentiation, proliferation and migration following vascular injury: implications for neointima formation and restenosis. PLoS One. 2014; 9: e101519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Franks TM, Hetzer MW The role of Nup98 in transcription regulation in healthy and diseased cells. Trends Cell Biol. 2013; 23: 112–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Hainer SJ, Fazzio TG High-Resolution Chromatin Profiling Using CUT&RUN. Curr Protoc Mol Biol. 2019; 126: e85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].Savic D, Partridge EC, Newberry KM, Smith SB, Meadows SK, Roberts BS, et al. CETCh-seq: CRISPR epitope tagging ChIP-seq of DNA-binding proteins. Genome Res. 2015; 25: 1581–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Lyu Q, Dhagia V, Han Y, Guo B, Wines-Samuelson ME, Christie CK, et al. CRISPR-Cas9-Mediated Epitope Tagging Provides Accurate and Versatile Assessment of Myocardin-Brief Report. Arterioscler Thromb Vasc Biol. 2018; 38: 2184–2190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Garzia A, Morozov P, Sajek M, Meyer C, Tuschl T PAR-CLIP for Discovering Target Sites of RNA-Binding Proteins. Methods Mol Biol. 2018; 1720: 55–75. [DOI] [PubMed] [Google Scholar]
- [137].Kim KK, Yang Y, Zhu J, Adelstein RS, Kawamoto S Rbfox3 controls the biogenesis of a subset of microRNAs. Nat Struct Mol Biol. 2014; 21: 901–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Luo X, Chae M, Krishnakumar R, Danko CG, Kraus WL Dynamic reorganization of the AC16 cardiomyocyte transcriptome in response to TNFalpha signaling revealed by integrated genomic analyses. BMC Genomics. 2014; 15: 155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Faustino RS, Behfar A, Perez-Terzic C, Terzic A Genomic chart guiding embryonic stem cell cardiopoiesis. Genome Biol. 2008; 9: R6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Capitanio JS, Montpetit B, Wozniak RW Nucleoplasmic Nup98 controls gene expression by regulating a DExH/D-box protein. Nucleus. 2018; 9: 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Ori A, Banterle N, Iskar M, Andres-Pons A, Escher C, Khanh Bui H, et al. Cell type-specific nuclear pores: a case in point for context-dependent stoichiometry of molecular machines. Mol Syst Biol. 2013; 9: 648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Capitanchik C, Dixon CR, Swanson SK, Florens L, Kerr ARW, Schirmer EC Analysis of RNA-Seq datasets reveals enrichment of tissue-specific splice variants for nuclear envelope proteins. Nucleus. 2018; 9: 410–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Perrino C, Barabasi AL, Condorelli G, Davidson SM, De Windt L, Dimmeler S, et al. Epigenomic and transcriptomic approaches in the post-genomic era: path to novel targets for diagnosis and therapy of the ischaemic heart? Position Paper of the European Society of Cardiology Working Group on Cellular Biology of the Heart. Cardiovasc Res. 2017; 113: 725–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].Wijetunga NA, Johnston AD, Maekawa R, Delahaye F, Ulahannan N, Kim K, et al. SMITE: an R/Bioconductor package that identifies network modules by integrating genomic and epigenomic information. BMC Bioinformatics. 2017; 18: 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Meng C, Zeleznik OA, Thallinger GG, Kuster B, Gholami AM, Culhane AC Dimension reduction techniques for the integrative analysis of multi-omics data. Brief Bioinform. 2016; 17: 628–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Sun YV, Hu YJ Integrative Analysis of Multi-omics Data for Discovery and Functional Studies of Complex Human Diseases. Adv Genet. 2016; 93: 147–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [147].Recamonde-Mendoza M, Werhli AV, Biolo A Systems biology approach identifies key regulators and the interplay between miRNAs and transcription factors for pathological cardiac hypertrophy. Gene. 2019; 698: 157–169. [DOI] [PubMed] [Google Scholar]
- [148].Harrington J, Fillmore N, Gao S, Yang Y, Zhang X, Liu P, et al. A Systems Biology Approach to Investigating Sex Differences in Cardiac Hypertrophy. J Am Heart Assoc. 2017; 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Louridas GE, Lourida KG Heart failure: a complex clinical process interpreted by systems biology approach and network medicine. Anadolu Kardiyol Derg. 2014; 14: 178–85. [DOI] [PubMed] [Google Scholar]
- [150].Preston CC, Storm EC, Burdine RD, Bradley TA, Uttecht AD, Faustino RS Nucleoporin insufficiency disrupts a pluripotent regulatory circuit in a pro-arrhythmogenic stem cell line. Sci Rep. 2019; 9: 12691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Zhang Q, Li J, Xue H, Kong L, Wang Y Network-based methods for identifying critical pathways of complex diseases: a survey. Mol Biosyst. 2016; 12: 1082–9. [DOI] [PubMed] [Google Scholar]
- [152].Leopold JA, Loscalzo J Emerging Role of Precision Medicine in Cardiovascular Disease. Circ Res. 2018; 122: 1302–1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Lambert JP, Picaud S, Fujisawa T, Hou H, Savitsky P, Uuskula-Reimand L, et al. Interactome Rewiring Following Pharmacological Targeting of BET Bromodomains. Mol Cell. 2019; 73: 621–638 e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Hu JX, Thomas CE, Brunak S Network biology concepts in complex disease comorbidities. Nat Rev Genet. 2016; 17: 615–29. [DOI] [PubMed] [Google Scholar]