Next Article in Journal
Neural Regeneration in Dry Eye Secondary to Systemic Lupus Erythematosus Is Also Disrupted like in Rheumatoid Arthritis, but in a Progressive Fashion
Next Article in Special Issue
A Proteomic Survey of the Cystic Fibrosis Transmembrane Conductance Regulator Surfaceome
Previous Article in Journal
Plasma Metabolomic Profiling after Feeding Dried Distiller’s Grains with Solubles in Different Cattle Breeds
Previous Article in Special Issue
It Takes Two to Tango! Protein–Protein Interactions behind cAMP-Mediated CFTR Regulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tissue-Specific Regulation of CFTR Gene Expression

1
Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200 Brest, France
2
Laboratoire de Génétique Moléculaire et d’Histocompatibilité, CHU Brest, F-29200 Brest, France
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(13), 10678; https://doi.org/10.3390/ijms241310678
Submission received: 22 May 2023 / Revised: 21 June 2023 / Accepted: 23 June 2023 / Published: 26 June 2023
(This article belongs to the Special Issue Cystic Fibrosis and CFTR Interactions 2.0)

Abstract

:
More than 2000 variations are described within the CFTR (Cystic Fibrosis Transmembrane Regulator) gene and related to large clinical issues from cystic fibrosis to mono-organ diseases. Although these CFTR-associated diseases have been well documented, a large phenotype spectrum is observed and correlations between phenotypes and genotypes are still not well established. To address this issue, we present several regulatory elements that can modulate CFTR gene expression in a tissue-specific manner. Among them, cis-regulatory elements act through chromatin loopings and take part in three-dimensional structured organization. With tissue-specific transcription factors, they form chromatin modules and can regulate gene expression. Alterations of specific regulations can impact and modulate disease expressions. Understanding all those mechanisms highlights the need to expand research outside the gene to enhance our knowledge.

1. Introduction

In 1595, the first historical record of Cystic Fibrosis (CF) was made. CF was described as a nutritional disturbance with diarrhea, steatorrhea, growth failure, salty sweat, bronchopneumonia and pulmonary infection [1,2]. Based on family studies, Andersen and May defined CF as a genetic disorder with autosomal recessive transmission [3,4]. In 1989, molecular biology development led to the discovery of the CFTR (Cystic Fibrosis Transmembrane Regulator) gene and also rapidly to the first report of a variation, F508del, which is now identified in 70% of people with CF (pwCF) [5,6,7,8]. The CFTR protein structure is described as an ion channel capable of modulating NaCl efflux. Genotyping in pwCF led to the identification of 2114 variations in the CFTR gene (genet.sickkids.on.ca, accessed on 21 May 2023). However, it is still very difficult to establish a link between the phenotype of individuals with CFTR-associated diseases and their genotype. Much remains to be done in the field to better understand the implication of the CFTR gene.
This review aims to decipher the implication of regulatory elements in the CFTR gene expression and hence the modulation of disease expression. Cis- and trans-regulations are two mechanisms discussed here.

2. Clinical Issues

Cystic fibrosis is a monogenic disorder with autosomal recessive transmission. It is caused by alterations of the CFTR gene, which spans over 189 kilobases with 27 exons and encodes a protein of 1480 amino acids. The CFTR protein belongs to the ATP-binding cassette (ABC) transporter family, which acts as an AMPc regulated ion channel. Many variants are found within the CFTR gene and are classified in seven classes depending on their effect on the protein (no protein, less protein, impaired gating, etc.) [9]. The most common variation found in 70% of pwCF is the F508del.
Defects of the CFTR protein lead to dysfunctions of the channel in epithelial cells. Ion transports are affected, resulting in water absorption into mucus. Obstruction of organs expressing CFTR is the consequence of mucus thickening.
pwCF are mainly affected by respiratory tract damage. In addition to the obstructive aspect, mucus thickening promotes bacterial development leading to important pulmonary infection, which is the major cause of mortality. Cystic fibrosis is a multi-organ disorder. pwCF are also affected by exocrine pancreatic damage, intestinal obstruction, infertility and liver and bone diseases [10].
However, the clinical spectrum is complex as variations in the CFTR gene also cause several mono-organ disorders, such as pancreatitis or congenital bilateral absence of the vas deferens (CBAVD) [10]. CBAVD represents 3% of male infertility and is the most frequent CFTR-related disorder (CFTR-RD) [11]. Two genes have been implicated, the CFTR gene in 80% of cases and ADGRG2 [12]. ADGRG2, a G protein-coupled receptor, regulates fluid reabsorption in efferent ducts through the activation of CFTR [13]. Variants in CFTR lead to a defect in HCO3 secretion, which has been shown to be critical for fertilizing the capacity of sperm [14]. Pancreatitis is also caused by a defect of HCO3 secretion, essential to solubilize mucins and avoid the plugging of pancreatic duct [15].
The CFTR gene is expressed in many organs with different levels of expression, and molecular consequences of a defect are variable across tissues. We, therefore, have a complex picture with a variable genotype–phenotype relationship.

3. Tissue Expression

Clinical issues of CFTR-associated diseases are directly correlated with the gene expression pattern. Indeed, a very clear tissue-specificity expression is observed but is also a temporal aspect. In-situ hybridizations show that CFTR is expressed in particular in epithelial cells with a very great level in the pancreatic duct and nasal polyps and to a lesser extent in lungs, gut, sweat glands, placenta, liver and male genital ducts as shown in Figure 1 [6]. In the male reproductive system, the CFTR gene is in majority expressed in vas deferens [16]. After birth the CFTR mRNA level is low, in particular in lungs, which is surprising in view of lung disease lethality. In fact, unique rare epithelial cells within the lung express the highest level of CFTR, the ionocytes [17,18]. More recently, secretory cells have been identified as the most important cells that express CFTR in lungs [19].

4. Characteristics of the CFTR Locus

4.1. Promoter as a Housekeeping Gene

For the majority of genes, the regulation of its transcription relies on the binding of specific transcription factors, activators or repressors, to the promoter. Surprisingly, while the CFTR gene expression is strictly regulated both spatially and temporally, its promoter has many features of a housekeeping gene. The CFTR promoter has no TATA box and possesses high GC content and multiple transcription start sites [20,21]. Several binding sites for transcription factors are observed as activator protein 1 (AP-1), Sp1, cAMP response element binding protein (CREB), an inverted CCAAT element (Y-box) and a CArG-like motif [22]. Also, an iκβ-like motif allows NF-κB binding next to inflammatory context and a HIF Responsive Element (HRE) motif in case of hypoxia (Figure 2) [23,24].
However, these elements alone cannot explain the precise tissue-specificity of the CFTR gene.

4.2. Cis-Regulation of the CFTR Gene

While elements within the CFTR promoter cannot explain its regulation, some research teams decided to study long range elements to provide a clearer picture. Chromatin in the nucleus is highly organized in different compartments. Long-distance elements can interact with each other through chromatin looping, notably within topologically associated domains (TAD). A huge effort to understand the non-coding DNA has been made through the ENCODE project, leading to a better characterization of cis-regulatory elements (CREs) [25]. CREs are regularly identified within DNase I hypersensitive sites (DHS); overlap biochemical marks (H3K27ac, activator or H3K27me3, repressor); and bind functional elements. They can interact with promoter or other CREs [26]. If functional characterizations do not provide all features, an element is just described as candidate CRE (cCRE). Activities are context-dependent as cis-regulation is tissue-specific.
The cis-regulation of the CFTR locus began to be explored since the 90s, and several elements are well described. Some elements are common to all cell types expressing CFTR and are considered more as structural elements, and some are more related to tissue-specific expression. A CFTR TAD was described as spanning 317 kb (chr7:117,039,878–117,356,812, hg19) and including the neighboring genes ASZ1 and CTTNBP2. Boundaries are delineated by two CREs identified as barrier insulators illustrated in Figure 3, in 5′ at −80.1 kb from the transcription start site (TSS) and in 3′ at +48.9 kb from the last codon of CFTR [27,28]. Another type of insulator exists: the enhancer-blocking insulator. Its role is to block interactions between promoter and CREs. Three enhancer-blocking insulators are described in the TAD; at −20.9 kb and +6.8 kb the activity is related to the recruitment of the CTCF protein and at +15.6 kb by the recruitment of C/EBP, CREB, AP-1, ARP-1 and HNF-4 [29,30].

4.2.1. Airway

Several DHS have been identified in different pulmonary cells across the CFTR locus and have been shown to interact with the promoter. DHS at −35 kb from the TSS and DHS at −44 kb are the most evident regions that interact with the promoter in airway epithelial cells [28,31]. H3K27ac marks correlating with transcriptionally active chromatin overlap with these two regions. In the presence of both elements, the CFTR promoter highly increases its activity (×30), demonstrating a cooperative effect [32]. Furthermore, RNA polymerase II binds these elements, which may imply a production of enhancer RNA (eRNA) [33]. eRNA are unstable transcripts emerging from active enhancer and acting as transcriptional regulators [34]. Identifying transcription factors that bind CREs helps to explain the three-dimensional organization and the formation of DNA looping. At −35 kb, a complex of transcription factors has been identified by ChIP-seq (Chromatin Immunoprecipitation Sequencing), including BAF155 (subunit of nucleosome remodeling complex SWI/SNF), IRF1 (interferon regulatory factor 1), NF-Y (Nuclear transcription factor Y), EHF (ETS Homologous Factor) and KLF5 (Kruppel-like factor 5) [35,36,37]. A nucleosome depletion of this region has also been shown, confirming its accessibility [35]. At −44 kb, recruitment of BACH1 (BTB Domain and CNC Homolog 1), BRD8 (Bromodomain Containing 8) and CTCF have been described [38,39]. Those two CREs are essential, a dramatic loss of CFTR expression is observed after their depletion as well as a loss of three-dimensional organization of the locus [33].
In addition to these two CREs, CREs at −3.4 kb from the TSS and in the intron 26 (chr7:117,305,325–117,307,149, intron 23 legacy name) show an increase of 3-fold of promotor activity in a cell-type specific manner [31]. A cooperative effect is observed when CREs −35 kb, −3.4 kb and intron 26 are associated. A weak enhancer effect is observed in the presence of another region at +36.6 kb from the last codon, but it increases in association with CRE −44 kb [40]. Some other regions are implicated in the maintenance of the locus architecture, for example a CTCF site at −20.9 kb [27,29]. An element in intron 22 (chr7:117,280,467–117,283,006) interacts with the promoter but without effect on the promoter activity [40].
Hence, many CREs appear to be involved in the correct expression of the CFTR gene in airway epithelial cells. For a correct level of expression, H3K4 mono-methylation by methyltransferase SEDT7 (SET Domain Containing 7) is required at CRE −35 kb for the binding of NF-Y. Depletion of SETD7 shows a loss of NF-Y binding and prevents occupation of SIN3a (SIN3 Transcription Regulator Family Member A) causing an enrichment of p300 (p300 histones acetyltransferase) at multiple sites including −44 kb and thus an increase of CFTR expression [36]. These observations suggest the presence of control mechanisms when a CRE is defective. When the KLF5 repressor is depleted, CFTR expression increases by six-fold as well as the activity of the ion channel [37]. Conversely, when IRF1 is depleted, the expression of the CFTR gene decreases [36].
Briefly, CREs at −44 kb and −35 kb are two strong enhancers which interact with the promoter and other CREs at −3.4 kb, in intron 26 and in 5′ of the gene at +36.6 kb to accent the effect. A complex of transcription factors binds these elements to ensure the correct level of gene expression. To maintain this organization, structural elements are used such as the −80.1 kb and +48.9 kb TAD boundaries and the −20.9 kb CTCF site (Figure 4).

4.2.2. Intestine

In intestinal cells, the same type of specific cis-regulations are observed. DHS are observed along the locus, and cell type-specific elements have been identified. By chromatin conformation capture studies, DHS in the introns 1, 11 and 12 (chr7:117,129,649–117,130,749; chr7:117,212,364–117,213,789; chr7:117,227,802–117,229,475, respectively), overlapping H3K27ac marks, have been shown to interact with the CFTR promoter [41,42,43]. DHS in 5′ of the gene, −80.1 kb and −20.9 kb as well as downstream in 3′, +48.9 kb, also interact with the promoter [27,28]. DHS in introns 1 and 12 are the most obvious CREs implicated in the CFTR cis-regulation in the small intestine. CRE in intron 12 is a strong enhancer, and in combination with CRE in intron 1, the effect is doubled (×33) [41,44]. Assuming that cooperation is frequently used, other combinations have been made in Caco2 cells, the most frequent intestinal cell line used. When CRE in intron 12 and the enhancer-blocking insulator at +15.6 kb are combined, a more impressive effect is observed [41]. DHS in intron 24 and 26 (chr7:117,299,158–117,301,081; chr7:117,305,325–117,307,149) are present but have weak enhancer activities on the promoter. However, if they are combined with intron 12, the enhancer activity increases by 57-fold and 34-fold, respectively [44]. When the three regions are combined, the same effect as CREs 12–24 is observed. Many transcription factors bind the different CREs notably because sequences in intron 1 and 12 are nucleosome-free regions [35]. Intron 12 is bound by RNA polymerase II [27]. Transcription factors HNF1α (hepatocyte nuclear factor 1 homeobox A), CDX2 (Caudal Type Homeobox 2), HNF4 (Hepatocyte Nuclear Factor 4) and FOXA2 (Forkhead Box A2) form a complex and bind CREs 1, 11, 12, 24 and 26 [44]. This high local concentration of transcription factors can illustrate a chromatin module [45]. HNF1α acts in this case as a master regulator and certainly stabilizes the binding of transcription factors. Besides, its expression correlates with the CFTR gene expression [46]. Hence, a variation in the HNF1α binding site leads to a loss of binding of the protein complex; notably it is observed that, in absence of HNF1α, a loss of acetylation occurs, leading to a decrease of CFTR expression [42,47]. In addition, TCF4 (Transcription Factor 4) binds CREs in intron 1 and 24, and its deletion also leads to a decrease of CFTR expression [47]. A loss of FOXA2 perturbs chromatin interactions, indeed its role is to maintain open chromatin [48].
If CRE in intron 1 is depleted, the CFTR gene expression decreases by 60% in vivo [49]. When the CREs 1 and 12 are depleted, the CFTR gene expression is almost undetectable [48]. After CRISPR/Cas9 deletions of CRE in intron 1 and 12, great changes in the three-dimensional organization are observed [48]. A gain of interactions between the CFTR promoter and many regions of the locus is observed, in particular with TAD boundaries, CTCF sites at −20.9 kb and +6.8 kb and intronic CREs as intron 4, 12 and 26 next to the deletion of intron 1. In the case of a loss of intron 12, interactions decrease between promoter and introns 1 and 11. A double deletion leads to the loss of all interactions between promoter and regions around both CREs and significantly disrupts the higher order chromatin organization, suggesting the very important role of these elements [48].
CRE in intron 1 is a weak enhancer but has a more important role in maintaining the locus architecture, while CRE in intron 12 is a strong enhancer and acts directly on CFTR expression (Figure 5).

4.2.3. Epididymis

In epididymis cells several DHS have been described in 5′ at −20.9 kb, in introns 1, 23 and 26 and in the 3′ region at +15.6 kb and +6.8 kb [29,41,50,51]. Also, a more distal region at the 3′ end of WNT2 gene is described. Active chromatin marks as H3K4me1 and H3K27ac are particularly present within DHS23 [52]. Nucleosome depletion occurs also at this site and c-Fos, c-Jun, JunD and C/EBP are predicted to bind the region [52]. Moreover, this region slightly interacts with the promoter. In addition, DHS 2 and 4 as well as 5′ regions and DHS + 48.9 kb interact with the promoter [27,53]. The +6.8 kb region binds CTCF and acts as an enhancer-blocking insulator as well as the +15.6 kb but without recruitment of CTCF protein [29]. Unlike the other cell types, the strongest interactions with the promoter are in 5′ regions at −80.1 kb and −20.9 kb. Reporter gene assay has not been performed; the majority of the data being produced with primary cells and no relevant cell lines are available.
HNF1α seems to have an important role in the regulation of genes expressed in the epididymis including CFTR. Indeed, a depletion by siRNA induces a decrease of genes implicated in ion transports, such as the solute carrier family. Perturbation of the regulation via HNF1α disturbs the intracellular pH, and finally, the luminal environment is modified [54]. ChIP-seq data indicate the binding of HNF1α at −44 kb and +15.6 kb [54].
Regions upstream and downstream the CFTR gene seem to have an important role as well as the region in intron 23 in regulating expression in epididymis cells (Figure 6).

4.2.4. Pancreas

The cis-regulation of the CFTR gene in the pancreas is less well-known, certainly because of the lack of relevant cell models. In fact, the duct pancreatic cells highly express the CFTR gene while cell lines available express very low levels of it. Hence, only candidate CREs have been highlighted. DNase-seq data indicate DHS in 5′ end of the gene, at −80.1 kb, −44 kb, −35 kb and in the 3′ at +6.8 kb and +15.6 kb [55]. The most important regions seem to be intronic. DHS have been observed at introns 2, 18, 19, 21 and 23. By EMSA (Electrophoretic Mobility Shift Assay) bindings of HNF1α, CDX2 and PBX1 (PBX Homeobox 1) have been shown on DHS 18 and 19, but only PBX1 has been confirmed in vivo [56]. Nevertheless, HNF1α depletion leads to CFTR mRNA decrease. Another transcription factor positively impacts the CFTR expression, BAF155, involved in nucleosome remodeling [57]. DHS 21 activates CFTR transcription next to mitomycine C treatment, an activator of CFTR [58].
To go farther, Smith et al. have achieved 5C (Chromosome Conformation Capture Carbon Copy) experiments on Capan-1 (pancreatic cell line) and showed a unique important interaction between the CFTR promoter and the region in intron 11. The interaction profile is rather low and looks like interaction profiles of cells that do not express CFTR. However, TAD boundaries are conserved [28]. This confirms that TADs are conserved between cell types but that intra-TAD structures vary according to the context.
Only candidate CREs have been described in pancreatic cells with assumptions allowing to implement a three-dimensional cis-regulatory model (Figure 7). Studies have to be carried out to obtain an accurate model.

4.3. Impact of CFTR Cis-Regulatory Variants?

It is now well-known that, in presence of alterations within CREs, diseases can occur; this is defined by the term enhanceropathies [59].
Next to the analysis of 210 kb across the CFTR locus on F508del homozygous patients, several variants have been newly identified, and some of them are found within CREs as −80.1 kb, −44 kb, −35 kb, −20.9 kb, intron 12 and +48.9 kb [60]. Association studies on lung function or sweat chloride concentrations have highlighted that common or rare variants could modulate the CF phenotype (positively or negatively). The most striking effect for both conditions is caused by variants within −80.1 kb, which corresponds to a CFTR TAD boundary. Important structural modifications can induce a change of expression level and partially recover a sufficient amount of CFTR at the membrane. Not surprisingly, variants present in CREs, interacting with the promoter only in epididymis cells, have no significant effect on lung function or sweat chloride level.
In 2019, Kerschner et al. studied 80 patients with 1 or 2 unknown CFTR variants and performed a resequencing of a region of 463 kb [61]. Variants (total: 1737) have been identified, and one-half of the studied alleles contains unknown pathogenic variants. Variants (51) are localized among 17 CREs (37 substitutions, 11 indels). Some of them are present in addition to two causal variants. In regions −44 kb, −35 kb, introns 1, 12 and 26, variants have been found within transcription factor binding sites. Four variants localized in CRE of intron 11 induced a decrease from 37% to 63% of promoter activity by luciferase assays. More functional tests are required to validate the impact of identifying variants, but this study demonstrates the importance of variants in non-coding DNA.
Several variants identified within CREs have led to modification in chromatin organization or in promoter activity in a tissue specific manner, showing the importance of investigating these regions.

4.4. microRNA and lncRNA

MicroRNA or miRNA are small non-coding RNA capable of modulating gene expression through the binding of 3′UTR of the targeted gene. As well as CREs, miRNA are expressed in a tissue-specific manner. Several miRNA have been highlighted to impact CFTR expression directly or indirectly by binding genes regulating CFTR [62].
miR-145, miR-331-3p and miR-494 are the first described miRNA to regulate CFTR in pulmonary cells [63]. More precise study of temporal regulation shows that miR-145 and miR-101, actually, have no effect in fetal lung cells but regulate negatively the level of CFTR mRNA in adult lung cells [64]. Blocking both miRNAs’ binding sites allows the stabilization of CFTR mRNA, hence increased channel activity of F508del homozygous patients [64]. miR-145 and miR-494 are also involved in CFTR regulation in intestinal cells by repressing by 40% CFTR mRNA [63]. In pancreatic cells, miR-1246, miR-1290 and miR-1827 repress CFTR levels [63].
The miR-888 cluster is exclusively expressed in the reproductive tract, in particular in epididymis, but without link with CFTR [65]. Other miRNA have also been described in different parts of the epididymis as miR-573, miR-155, miR-1204 and miR-770 [66].
In contrast to microRNA, long non-coding RNA (lncRNA) are made up of more than 200 nucleotides and constitute a group of non-coding DNA which is not yet well described [67]. A study comparing bronchial brushings from pwCF and non-CF identifies 1063 lncRNAs differently expressed [68]. Next to Pseudomonas aeruginosa (PsA) infection, a modification in several lncRNA as MEG9 (Maternally Expressed 9), a positive co-regulator of inflammatory pathways in CF lung, is also observed [69]. Functional tests on BGas lncRNA, described by Saayman et al., show that its transcription is initiate from intron 11 of the CFTR gene and regulates negatively its expression by acting with other protein and modifying local chromatin conformation [70].
Both microRNA and lncRNA belong to the family of non-coding DNA, previously named “Junk DNA”. Here, we report few examples that highlight the importance of those elements that are capable of regulating the CFTR gene with varying degrees of tissue specificity.

4.5. Modifier Genes

Other genetic factors can impact the CFTR gene. In fact, each individual is characterized by a specific polymorphism pattern. Polymorphism is a common variant in DNA sequence with an allele frequency of at least 1% in the general population. The presence of a single nucleotide polymorphism (SNP) alone is not pathogenic, but in a specific gene it can modulate the severity of a disease caused by another gene; they are referred as modifier genes. In CF and CFTR-RD, several genes have been implicated to modify lung function, the bacterial infection and inflammation and the severity of the intestinal obstruction.

4.5.1. Pulmonary

Huge genome wide association study (GWAS) identifies several locus/modifier genes that could impact lung function [71,72]. For example, natural host defense barrier can be altered in case of variations within MUC4/MUC20. These are glycoproteins, mucins, expressed on the surface of epithelial cells from gastrointestinal and respiratory tracts that prevent mucus accumulation in periciliary layer. Hence, they represent a host defense barrier and are essential to maintaining equilibrium [73]. SLC9A3 is an ion transporter acting on pH regulation and is involved in neonatal intestinal obstruction. Early infection by PsA is related with SLC9A3 variants [74,75]. Defect on pH regulation leads to depletion of airway liquid surface representing a supportive environment to bacterial growth. On the same locus, EXOC3 is part of post-Golgi trafficking. Another locus contains the modifier gene HLA class II involved in immune response, which is associated with asthma and altered lung function. AGTR2 is associated with lung fibrosis and SLC6A14 with lung severity and appearance of PsA [76]. EHF has an important role in lung epithelial function by regulating inflammation and pathways in response to injury [77]. CE72/TPPP plays a role in microtubule organization, which is disturbed in CF [71]. Cytokines as TGFβ1, IL-8, Il1β, TNFα are related to inflammation in CF [78].

4.5.2. Intestines

Meconium ileus, which presents in ~15% of pwCF, is also highly related to modifier genes, such as the solute carrier family as SLC26A9, SLC9A3, SLC4A4 and SLC6A14, ATP12A and PRSS1 [78,79,80]. Several proteins from the solute carrier family have been shown to interact with CFTR and to impact its activity. We can suppose that the presence of SNPs within a gene mentioned above can modify this interaction. In the case of ATP12A, a proton exchanger, minor changes in the ion transport could affect viscosity [80].

4.5.3. Epididymis

In the case of infertility due to absence of vas deferens, fewer studies have been performed. TGFβ1, EDNRA and SLC9A3 were only suggested to impact the correct development of the reproductive male tracts [81,82,83].

4.5.4. Pancreas

Pancreatic insufficiency in CF is also probably related to modifier genes, and some of them have been predicted but have not been confirmed by functional studies. CTNNB1, IRF5, EPHX1, PRSS1, CASR, CTRC and KRT8 are some of them [84,85,86,87].
Modifier genes seem to have an important role in the different tissues to modify the phenotype in addition to the CFTR defect. Huge efforts need to be taken to accurately understand the functional linkage with phenotype.

4.6. Complex Alleles

In 2018, the Cystic Fibrosis Foundation Patient Registry reported that 3.1% of pwCF carry more than two CFTR variants. Complex allele is defined by the presence of an additional variation on the same parental allele. The presence of one more variant can modulate the CFTR function, and several combinations are reported in literature. For example, a severe effect on CFTR function is observed in the presence of complex allele c.[1397C  >  A;3209G  >  A] [88]. In the case of c.3469-1304C > G, all the patients present early symptoms with PsA colonization and pancreatic insufficiency [89]. Conversely, patients with c.3874-4522A > G present fewer symptoms with moderate effect, corresponding to CFTR-RD characteristics [89]. To gain insight into the understanding of complex alleles a recent protocol of Targeted Locus Amplification and Haplotyping applied to the CFTR gene has been published [90].
It is important to gain insight about complex alleles. Complex alleles present within CREs can modify a phenotype due to tissue-specific activation of them. If we limit ourselves to identifying just two variations, we may miss the element explaining different phenotypes. Complex alleles must be taken into serious consideration, in particular for the implementation of treatment.

5. Conclusions

Care for pwCF has largely improved these past years through the development of modulators (elexacaftor/tezacaftor/ivacaftor) treatment [91,92]. Nonetheless, some patients are still not eligible or not responding. Moreover, some moderate CFTR disease cases remain misunderstood. These different findings highlight the necessity to understand the precise mechanism underlying the different patterns of disease’s expression and the relationship between genotype and phenotype.
Multiple elements are implicated in the modulation of diseases in a tissue-specific manner but also in the response of treatment. Actually, numerous pathways can impact the CFTR expression through indirect cellular regulations as inflammation, hypoxia, oxidative stress and endoplasmic reticulum stress or through transcriptional regulation [23,24,93]. Here, we decided to list some elements that have been demonstrated to impact CFTR transcriptional expression and detail different regulations in few cell types. We have not mentioned CFTR expression in non-epithelial tissues such as neutrophils, macrophages, brain, bone due to the lack of available data. We present modifier genes as an important mechanism that can modify specific clinical manifestations, in particular according to the tissue expression of the modifier gene. We refer to the presence of complex alleles that are probably underestimated but which must definitely affect responses to treatments [60]. Some complex alleles have been identified as non-responder [94]. Also, we highlight the importance of knowing their presence and thus explaining phenotype if, for example, active CREs are affected by them. This leads to the last element described in the review, CREs. They act at distance from the gene to regulate its expression, and variants can lead to dysfunction. Functional tests have to be performed to confirm effects of the cis-regulatory variants found in patients. This also highlights the need to perform a resequencing on larger regions in unresolved cases. Also, we stress that three-dimensional organization is very important and highly tissue-related depending on individual transcription factors recruitment. Alterations of cis-regulation mechanism lead to various clinical manifestations.
This review states diverse gene regulation systems and outlines the importance to understand basal gene expression in the different tissues and to transfer this knowledge to the clinical management.

Funding

This research was funded by Vaincre la Mucoviscidose grant number [RF20210502832] and Association Gaétan Saleün.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Andersen, D.H. Cystic Fibrosis of the Pancreas and Its Relation to Celiac Disease: A Clinical and Pathologic Study. Am. J. Dis. Child. 1938, 56, 344. [Google Scholar] [CrossRef]
  2. Fanconi, G.; Uehlinger, E.; Knauer, C. Das Coeliakiesyndrom Bei Angeborenerzysticher Pankreasfibromatose Und Bronchiektasien. Wien. Med. Wochenschr. 1936, 86, 753–756. [Google Scholar]
  3. Andersen, D.H.; Hodges, R. Celiac Syndrome: V. Genetics of Cystic Fibrosis of the Pancreas with a Consideration of Etiology. Am. J. Dis. Child. 1946, 72, 62–80. [Google Scholar] [CrossRef] [PubMed]
  4. May, C.D. Fibrosis of the Pancreas in Infants and Children. J. Pediatr. 1949, 34, 663–687. [Google Scholar] [CrossRef] [Green Version]
  5. Kerem, B.-S.; Rommens, J.M.; Buchanan, J.A.; Markiewicz, D.; Cox, T.K.; Chakravarti, A.; Buchwald, M.; Tsui, L.-C. Identification of the Cystic Fibrosis Gene: Genetic Analysis. Science 1989, 245, 1073–1080. [Google Scholar] [CrossRef] [Green Version]
  6. Riordan, J.R.; Rommens, J.M.; Kerem, B.-S.; Alon, N.; Rozmahel, R.; Grzelczak, Z.; Zielenski, J.; Lok, S.; Plavsic, N.; Chou, J.-L.; et al. Identification of the Cystic Fibrosis Gene: Cloning and Characterization of Complementary DNA. Science 1989, 245, 1066–1073. [Google Scholar] [CrossRef]
  7. Rommens, J.M.; Iannuzzi, M.C.; Kerem, B.; Drumm, M.L.; Melmer, G.; Dean, M.; Rozmahel, R.; Cole, J.L.; Kennedy, D.; Hidaka, N. Identification of the Cystic Fibrosis Gene: Chromosome Walking and Jumping. Science 1989, 245, 1059–1065. [Google Scholar] [CrossRef]
  8. Becq, F. CFTR et mucoviscidose, une histoire cinquantenaire. Med. Sci. 2021, 37, 654–659. [Google Scholar] [CrossRef]
  9. De Boeck, K.; Amaral, M.D. Progress in Therapies for Cystic Fibrosis. Lancet Respir. Med. 2016, 4, 662–674. [Google Scholar] [CrossRef]
  10. Castellani, C.; Assael, B.M. Cystic Fibrosis: A Clinical View. Cell. Mol. Life Sci. 2017, 74, 129–140. [Google Scholar] [CrossRef]
  11. Bombieri, C.; Claustres, M.; De Boeck, K.; Derichs, N.; Dodge, J.; Girodon, E.; Sermet, I.; Schwarz, M.; Tzetis, M.; Wilschanski, M.; et al. Recommendations for the Classification of Diseases as CFTR-Related Disorders. J. Cyst. Fibros. 2011, 10 (Suppl. S2), S86–S102. [Google Scholar] [CrossRef] [Green Version]
  12. Bieth, E.; Hamdi, S.M.; Mieusset, R. Genetics of the Congenital Absence of the Vas Deferens. Hum. Genet. 2021, 140, 59–76. [Google Scholar] [CrossRef] [Green Version]
  13. Zhang, D.-L.; Sun, Y.-J.; Ma, M.-L.; Wang, Y.; Lin, H.; Li, R.-R.; Liang, Z.-L.; Gao, Y.; Yang, Z.; He, D.-F.; et al. Gq Activity- and β-Arrestin-1 Scaffolding-Mediated ADGRG2/CFTR Coupling Are Required for Male Fertility. eLife 2018, 7, e33432. [Google Scholar] [CrossRef]
  14. Xu, W.M.; Shi, Q.X.; Chen, W.Y.; Zhou, C.X.; Ni, Y.; Rowlands, D.K.; Yi Liu, G.; Zhu, H.; Ma, Z.G.; Wang, X.F.; et al. Cystic Fibrosis Transmembrane Conductance Regulator Is Vital to Sperm Fertilizing Capacity and Male Fertility. Proc. Natl. Acad. Sci. USA 2007, 104, 9816–9821. [Google Scholar] [CrossRef] [Green Version]
  15. Angyal, D.; Bijvelds, M.J.C.; Bruno, M.J.; Peppelenbosch, M.P.; de Jonge, H.R. Bicarbonate Transport in Cystic Fibrosis and Pancreatitis. Cells 2021, 11, 54. [Google Scholar] [CrossRef]
  16. Harris, A.; Chalkley, G.; Goodman, S.; Coleman, L. Expression of the Cystic Fibrosis Gene in Human Development. Development 1991, 113, 305–310. [Google Scholar] [CrossRef]
  17. Montoro, D.T.; Haber, A.L.; Biton, M.; Vinarsky, V.; Lin, B.; Birket, S.E.; Yuan, F.; Chen, S.; Leung, H.M.; Villoria, J.; et al. A Revised Airway Epithelial Hierarchy Includes CFTR-Expressing Ionocytes. Nature 2018, 560, 319–324. [Google Scholar] [CrossRef]
  18. Plasschaert, L.W.; Žilionis, R.; Choo-Wing, R.; Savova, V.; Knehr, J.; Roma, G.; Klein, A.M.; Jaffe, A.B. A Single-Cell Atlas of the Airway Epithelium Reveals the CFTR-Rich Pulmonary Ionocyte. Nature 2018, 560, 377–381. [Google Scholar] [CrossRef]
  19. Okuda, K.; Dang, H.; Kobayashi, Y.; Carraro, G.; Nakano, S.; Chen, G.; Kato, T.; Asakura, T.; Gilmore, R.C.; Morton, L.C.; et al. Secretory Cells Dominate Airway CFTR Expression and Function in Human Airway Superficial Epithelia. Am. J. Respir. Crit. Care Med. 2021, 203, 1275–1289. [Google Scholar] [CrossRef]
  20. Yoshimura, K.; Nakamura, H.; Trapnell, B.C.; Dalemans, W.; Pavirani, A.; Lecocq, J.P.; Crystal, R.G. The Cystic Fibrosis Gene Has a “Housekeeping”-Type Promoter and Is Expressed at Low Levels in Cells of Epithelial Origin. J. Biol. Chem. 1991, 266, 9140–9144. [Google Scholar] [CrossRef]
  21. Chou, J.L.; Rozmahel, R.; Tsui, L.C. Characterization of the Promoter Region of the Cystic Fibrosis Transmembrane Conductance Regulator Gene. J. Biol. Chem. 1991, 266, 24471–24476. [Google Scholar] [CrossRef] [PubMed]
  22. Nuthall, H.N.; Moulin, D.S.; Huxley, C.; Harris, A. Analysis of DNase-I-Hypersensitive Sites at the 3’ End of the Cystic Fibrosis Transmembrane Conductance Regulator Gene (CFTR). Biochem. J. 1999, 341, 601–611. [Google Scholar] [CrossRef] [PubMed]
  23. Brouillard, F.; Bouthier, M.; Leclerc, T.; Clement, A.; Baudouin-Legros, M.; Edelman, A. NF-ΚB Mediates Up-Regulation of CFTR Gene Expression in Calu-3 Cells by Interleukin-1β. J. Biol. Chem. 2001, 276, 9486–9491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zheng, W.; Kuhlicke, J.; Jäckel, K.; Eltzschig, H.K.; Singh, A.; Sjöblom, M.; Riederer, B.; Weinhold, C.; Seidler, U.; Colgan, S.P.; et al. Hypoxia Inducible Factor-1 (HIF-1)-Mediated Repression of Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) in the Intestinal Epithelium. FASEB J. 2009, 23, 204–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. ENCODE Project Consortium. The ENCODE (ENCyclopedia of DNA Elements) Project. Science 2004, 306, 636–640. [Google Scholar] [CrossRef] [Green Version]
  26. Gasperini, M.; Tome, J.M.; Shendure, J. Towards a Comprehensive Catalogue of Validated and Target-Linked Human Enhancers. Nat. Rev. Genet. 2020, 21, 292–310. [Google Scholar] [CrossRef]
  27. Yang, R.; Kerschner, J.L.; Gosalia, N.; Neems, D.; Gorsic, L.K.; Safi, A.; Crawford, G.E.; Kosak, S.T.; Leir, S.-H.; Harris, A. Differential Contribution of Cis-Regulatory Elements to Higher Order Chromatin Structure and Expression of the CFTR Locus. Nucleic Acids Res. 2016, 44, 3082–3094. [Google Scholar] [CrossRef] [Green Version]
  28. Smith, E.M.; Lajoie, B.R.; Jain, G.; Dekker, J. Invariant TAD Boundaries Constrain Cell-Type-Specific Looping Interactions between Promoters and Distal Elements around the CFTR Locus. Am. J. Hum. Genet. 2016, 98, 185–201. [Google Scholar] [CrossRef] [Green Version]
  29. Blackledge, N.P.; Carter, E.J.; Evans, J.R.; Lawson, V.; Rowntree, R.K.; Harris, A. CTCF Mediates Insulator Function at the CFTR Locus. Biochem. J. 2007, 408, 267–275. [Google Scholar] [CrossRef] [Green Version]
  30. Rowntree, R.; Harris, A. DNA Polymorphisms in Potential Regulatory Elements of the CFTR Gene Alter Transcription Factor Binding. Hum. Genet. 2002, 111, 66–74. [Google Scholar] [CrossRef]
  31. Zhang, Z.; Ott, C.J.; Lewandowska, M.A.; Leir, S.-H.; Harris, A. Molecular Mechanisms Controlling CFTR Gene Expression in the Airway. J. Cell. Mol. Med. 2012, 16, 1321–1330. [Google Scholar] [CrossRef]
  32. Zhang, Z.; Leir, S.-H.; Harris, A. Oxidative Stress Regulates CFTR Gene Expression in Human Airway Epithelial Cells through a Distal Antioxidant Response Element. Am. J. Respir. Cell. Mol. Biol. 2015, 52, 387–396. [Google Scholar] [CrossRef] [Green Version]
  33. NandyMazumdar, M.; Yin, S.; Paranjapye, A.; Kerschner, J.L.; Swahn, H.; Ge, A.; Leir, S.-H.; Harris, A. Looping of Upstream Cis-Regulatory Elements Is Required for CFTR Expression in Human Airway Epithelial Cells. Nucleic Acids Res. 2020, 48, 3513–3524. [Google Scholar] [CrossRef] [Green Version]
  34. Harrison, L.J.; Bose, D. Enhancer RNAs Step Forward: New Insights into Enhancer Function. Development 2022, 149, dev200398. [Google Scholar] [CrossRef]
  35. Yigit, E.; Bischof, J.M.; Zhang, Z.; Ott, C.J.; Kerschner, J.L.; Leir, S.-H.; Buitrago-Delgado, E.; Zhang, Q.; Wang, J.-P.Z.; Widom, J.; et al. Nucleosome Mapping across the CFTR Locus Identifies Novel Regulatory Factors. Nucleic Acids Res. 2013, 41, 2857–2868. [Google Scholar] [CrossRef] [Green Version]
  36. Zhang, Z.; Leir, S.-H.; Harris, A. Immune Mediators Regulate CFTR Expression through a Bifunctional Airway-Selective Enhancer. Mol. Cell. Biol. 2013, 33, 2843–2853. [Google Scholar] [CrossRef] [Green Version]
  37. Mutolo, M.J.; Leir, S.-H.; Fossum, S.L.; Browne, J.A.; Harris, A. A Transcription Factor Network Represses CFTR Gene Expression in Airway Epithelial Cells. Biochem. J. 2018, 475, 1323–1334. [Google Scholar] [CrossRef]
  38. NandyMazumdar, M.; Paranjapye, A.; Browne, J.; Yin, S.; Leir, S.-H.; Harris, A. BACH1, the Master Regulator of Oxidative Stress, has a Dual Effect on CFTR Expression. Biochem. J. 2021, 478, 3741–3756. [Google Scholar] [CrossRef]
  39. Browne, J.A.; NandyMazumdar, M.; Paranjapye, A.; Leir, S.-H.; Harris, A. The Bromodomain Containing 8 (BRD8) Transcriptional Network in Human Lung Epithelial Cells. Mol. Cell. Endocrinol. 2021, 524, 111169. [Google Scholar] [CrossRef]
  40. Moisan, S.; Berlivet, S.; Ka, C.; Le Gac, G.; Dostie, J.; Férec, C. Analysis of Long-Range Interactions in Primary Human Cells Identifies Cooperative CFTR Regulatory Elements. Nucleic Acids Res. 2016, 44, 2564–2576. [Google Scholar] [CrossRef] [Green Version]
  41. Ott, C.J.; Blackledge, N.P.; Kerschner, J.L.; Leir, S.-H.; Crawford, G.E.; Cotton, C.U.; Harris, A. Intronic Enhancers Coordinate Epithelial-Specific Looping of the Active CFTR Locus. Proc. Natl. Acad. Sci. USA 2009, 106, 19934–19939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Ott, C.J.; Suszko, M.; Blackledge, N.P.; Wright, J.E.; Crawford, G.E.; Harris, A. A Complex Intronic Enhancer Regulates Expression of the CFTR Gene by Direct Interaction with the Promoter. J. Cell. Mol. Med. 2009, 13, 680–692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Gosalia, N.; Neems, D.; Kerschner, J.L.; Kosak, S.T.; Harris, A. Architectural Proteins CTCF and Cohesin Have Distinct Roles in Modulating the Higher Order Structure and Expression of the CFTR Locus. Nucleic Acids Res. 2014, 42, 9612–9622. [Google Scholar] [CrossRef] [PubMed]
  44. Collobert, M.; Bocher, O.; Le Nabec, A.; Génin, E.; Férec, C.; Moisan, S. CFTR Cooperative Cis-Regulatory Elements in Intestinal Cells. Int. J. Mol. Sci. 2021, 22, 2599. [Google Scholar] [CrossRef] [PubMed]
  45. Van Mierlo, G.; Pushkarev, O.; Kribelbauer, J.F.; Deplancke, B. Chromatin Modules and Their Implication in Genomic Organization and Gene Regulation. Trends Genet. 2022, 39, 140–153. [Google Scholar] [CrossRef] [PubMed]
  46. Mouchel, N.; Henstra, S.A.; McCarthy, V.A.; Williams, S.H.; Phylactides, M.; Harris, A. HNF1alpha Is Involved in Tissue-Specific Regulation of CFTR Gene Expression. Biochem. J. 2004, 378, 909–918. [Google Scholar] [CrossRef] [Green Version]
  47. Paul, T.; Li, S.; Khurana, S.; Leleiko, N.S.; Walsh, M.J. The Epigenetic Signature of CFTR Expression Is Co-Ordinated via Chromatin Acetylation through a Complex Intronic Element. Biochem. J. 2007, 408, 317–326. [Google Scholar] [CrossRef] [Green Version]
  48. Yin, S.; NandyMazumdar, M.; Paranjapye, A.; Harris, A. Cross-Talk between Enhancers, Structural Elements and Activating Transcription Factors Maintains the 3D Architecture and Expression of the CFTR Gene. Genomics 2022, 114, 110350. [Google Scholar] [CrossRef]
  49. Rowntree, R.K.; Vassaux, G.; McDowell, T.L.; Howe, S.; McGuigan, A.; Phylactides, M.; Huxley, C.; Harris, A. An Element in Intron 1 of the CFTR Gene Augments Intestinal Expression in Vivo. Hum. Mol. Genet. 2001, 10, 1455–1464. [Google Scholar] [CrossRef] [Green Version]
  50. Blackledge, N.P.; Ott, C.J.; Gillen, A.E.; Harris, A. An Insulator Element 3’ to the CFTR Gene Binds CTCF and Reveals an Active Chromatin Hub in Primary Cells. Nucleic Acids Res. 2009, 37, 1086–1094. [Google Scholar] [CrossRef] [Green Version]
  51. Smith, D.J.; Nuthall, H.N.; Majetti, M.E.; Harris, A. Multiple Potential Intragenic Regulatory Elements in the CFTR Gene. Genomics 2000, 64, 90–96. [Google Scholar] [CrossRef]
  52. Bischof, J.M.; Gillen, A.E.; Song, L.; Gosalia, N.; London, D.; Furey, T.S.; Crawford, G.E.; Harris, A. A Genome-Wide Analysis of Open Chromatin in Human Epididymis Epithelial Cells Reveals Candidate Regulatory Elements for Genes Coordinating Epididymal Function. Biol. Reprod. 2013, 89, 104. [Google Scholar] [CrossRef]
  53. Kerschner, J.L.; Gosalia, N.; Leir, S.-H.; Harris, A. Chromatin Remodeling Mediated by the FOXA1/A2 Transcription Factors Activates CFTR Expression in Intestinal Epithelial Cells. Epigenetics 2014, 9, 557–565. [Google Scholar] [CrossRef] [Green Version]
  54. Browne, J.A.; Yang, R.; Eggener, S.E.; Leir, S.-H.; Harris, A. HNF1 Regulates Critical Processes in the Human Epididymis Epithelium. Mol. Cell. Endocrinol. 2016, 425, 94–102. [Google Scholar] [CrossRef] [Green Version]
  55. ENCODE Project Consortium. An Integrated Encyclopedia of DNA Elements in the Human Genome. Nature 2012, 489, 57–74. [Google Scholar] [CrossRef] [Green Version]
  56. McCarthy, V.A.; Ott, C.J.; Phylactides, M.; Harris, A. Interaction of Intestinal and Pancreatic Transcription Factors in the Regulation of CFTR Gene Expression. Biochim. Biophys. Acta 2009, 1789, 709–718. [Google Scholar] [CrossRef] [Green Version]
  57. Sancho, A.; Li, S.; Paul, T.; Zhang, F.; Aguilo, F.; Vashisht, A.; Balasubramaniyan, N.; Leleiko, N.S.; Suchy, F.J.; Wohlschlegel, J.A.; et al. CHD6 Regulates the Topological Arrangement of the CFTR Locus. Hum. Mol. Genet. 2015, 24, 2724–2732. [Google Scholar] [CrossRef] [Green Version]
  58. Phylactides, M.; Rowntree, R.; Nuthall, H.; Ussery, D.; Wheeler, A.; Harris, A. Evaluation of Potential Regulatory Elements Identified as DNase I Hypersensitive Sites in the CFTR Gene. Eur. J. Biochem. 2002, 269, 553–559. [Google Scholar] [CrossRef]
  59. Zaugg, J.B.; Sahlén, P.; Andersson, R.; Alberich-Jorda, M.; de Laat, W.; Deplancke, B.; Ferrer, J.; Mandrup, S.; Natoli, G.; Plewczynski, D.; et al. Current Challenges in Understanding the Role of Enhancers in Disease. Nat. Struct. Mol. Biol. 2022, 29, 1148–1158. [Google Scholar] [CrossRef]
  60. Vecchio-Pagán, B.; Blackman, S.M.; Lee, M.; Atalar, M.; Pellicore, M.J.; Pace, R.G.; Franca, A.L.; Raraigh, K.S.; Sharma, N.; Knowles, M.R.; et al. Deep Resequencing of CFTR in 762 F508del Homozygotes Reveals Clusters of Non-Coding Variants Associated with Cystic Fibrosis Disease Traits. Hum. Genome Var. 2016, 3, 16038. [Google Scholar] [CrossRef] [Green Version]
  61. Kerschner, J.L.; Ghosh, S.; Paranjapye, A.; Cosme, W.R.; Audrézet, M.-P.; Nakakuki, M.; Ishiguro, H.; Férec, C.; Rommens, J.; Harris, A. Screening for Regulatory Variants in 460 Kb Encompassing the CFTR Locus in Cystic Fibrosis Patients. J. Mol. Diagn. 2019, 21, 70–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Ramachandran, S.; Karp, P.H.; Osterhaus, S.R.; Jiang, P.; Wohlford-Lenane, C.; Lennox, K.A.; Jacobi, A.M.; Praekh, K.; Rose, S.D.; Behlke, M.A.; et al. Post-Transcriptional Regulation of Cystic Fibrosis Transmembrane Conductance Regulator Expression and Function by MicroRNAs. Am. J. Respir. Cell. Mol. Biol. 2013, 49, 544–551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Gillen, A.E.; Gosalia, N.; Leir, S.-H.; Harris, A. MicroRNA Regulation of Expression of the Cystic Fibrosis Transmembrane Conductance Regulator Gene. Biochem. J. 2011, 438, 25–32. [Google Scholar] [CrossRef] [Green Version]
  64. Viart, V.; Bergougnoux, A.; Bonini, J.; Varilh, J.; Chiron, R.; Tabary, O.; Molinari, N.; Claustres, M.; Taulan-Cadars, M. Transcription Factors and MiRNAs That Regulate Fetal to Adult CFTR Expression Change Are New Targets for Cystic Fibrosis. Eur. Respir. J. 2015, 45, 116–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Landgraf, P.; Rusu, M.; Sheridan, R.; Sewer, A.; Iovino, N.; Aravin, A.; Pfeffer, S.; Rice, A.; Kamphorst, A.O.; Landthaler, M.; et al. A Mammalian MicroRNA Expression Atlas Based on Small RNA Library Sequencing. Cell 2007, 129, 1401–1414. [Google Scholar] [CrossRef] [Green Version]
  66. Browne, J.A.; Leir, S.-H.; Eggener, S.E.; Harris, A. Region-Specific MicroRNA Signatures in the Human Epididymis. Asian J. 2018, 20, 539–544. [Google Scholar] [CrossRef]
  67. Glasgow, A.M.A.; De Santi, C.; Greene, C.M. Non-Coding RNA in Cystic Fibrosis. Biochem. Soc. Trans. 2018, 46, 619–630. [Google Scholar] [CrossRef]
  68. McKiernan, P.J.; Molloy, K.; Cryan, S.A.; McElvaney, N.G.; Greene, C.M. Long Noncoding RNA Are Aberrantly Expressed in Vivo in the Cystic Fibrosis Bronchial Epithelium. Int. J. Biochem. Cell. Biol. 2014, 52, 184–191. [Google Scholar] [CrossRef]
  69. Balloy, V.; Koshy, R.; Perra, L.; Corvol, H.; Chignard, M.; Guillot, L.; Scaria, V. Bronchial Epithelial Cells from Cystic Fibrosis Patients Express a Specific Long Non-Coding RNA Signature upon Pseudomonas Aeruginosa Infection. Front. Cell. Infect. Microbiol. 2017, 7, 218. [Google Scholar] [CrossRef] [Green Version]
  70. Saayman, S.M.; Ackley, A.; Burdach, J.; Clemson, M.; Gruenert, D.C.; Tachikawa, K.; Chivukula, P.; Weinberg, M.S.; Morris, K.V. Long Non-Coding RNA BGas Regulates the Cystic Fibrosis Transmembrane Conductance Regulator. Mol. Ther. 2016, 24, 1351–1357. [Google Scholar] [CrossRef] [Green Version]
  71. Corvol, H.; Blackman, S.M.; Boëlle, P.-Y.; Gallins, P.J.; Pace, R.G.; Stonebraker, J.R.; Accurso, F.J.; Clement, A.; Collaco, J.M.; Dang, H.; et al. Genome-Wide Association Meta-Analysis Identifies Five Modifier Loci of Lung Disease Severity in Cystic Fibrosis. Nat. Commun. 2015, 6, 8382. [Google Scholar] [CrossRef] [Green Version]
  72. Zhou, Y.-H.; Gallins, P.J.; Pace, R.G.; Dang, H.; Aksit, M.A.; Blue, E.E.; Buckingham, K.J.; Collaco, J.M.; Faino, A.V.; Gordon, W.W.; et al. Genetic Modifiers of Cystic Fibrosis Lung Disease Severity: Whole Genome Analysis of 7840 Patients. Am. J. Respir. Crit. Care Med. 2023, 207, 1324–1333. [Google Scholar] [CrossRef]
  73. Jonckheere, N.; Van Seuningen, I. Integrative Analysis of the Cancer Genome Atlas and Cancer Cell Lines Encyclopedia Large-Scale Genomic Databases: MUC4/MUC16/MUC20 Signature Is Associated with Poor Survival in Human Carcinomas. J. Transl. Med. 2018, 16, 259. [Google Scholar] [CrossRef]
  74. Dorfman, R.; Taylor, C.; Lin, F.; Sun, L.; Sandford, A.; Paré, P.; Berthiaume, Y.; Corey, M.; Durie, P.; Zielenski, J.; et al. Modulatory Effect of the SLC9A3 Gene on Susceptibility to Infections and Pulmonary Function in Children with Cystic Fibrosis. Pediatr. Pulmonol. 2011, 46, 385–392. [Google Scholar] [CrossRef]
  75. Zhong, L.; Strug, L.J. RoPE: A Robust Profile Likelihood Method for Differential Gene Expression Analysis. Genet. Epidemiol. 2023, 47, 379–393. [Google Scholar] [CrossRef]
  76. Mercier, J.; Calmel, C.; Mésinèle, J.; Sutanto, E.; Merabtene, F.; Longchampt, E.; Sage, E.; Kicic, A.; Boëlle, P.-Y.; Corvol, H.; et al. SLC6A14 Impacts Cystic Fibrosis Lung Disease Severity via MTOR and Epithelial Repair Modulation. Front. Mol. Biosci. 2022, 9, 850261. [Google Scholar] [CrossRef]
  77. Fossum, S.L.; Mutolo, M.J.; Tugores, A.; Ghosh, S.; Randell, S.H.; Jones, L.C.; Leir, S.-H.; Harris, A. Ets Homologous Factor (EHF) Has Critical Roles in Epithelial Dysfunction in Airway Disease. J. Biol. Chem. 2017, 292, 10938–10949. [Google Scholar] [CrossRef] [Green Version]
  78. Butnariu, L.I.; Țarcă, E.; Cojocaru, E.; Rusu, C.; Moisă, Ș.M.; Leon Constantin, M.-M.; Gorduza, E.V.; Trandafir, L.M. Genetic Modifying Factors of Cystic Fibrosis Phenotype: A Challenge for Modern Medicine. J. Clin. Med. 2021, 10, 5821. [Google Scholar] [CrossRef]
  79. Sun, L.; Rommens, J.M.; Corvol, H.; Li, W.; Li, X.; Chiang, T.A.; Lin, F.; Dorfman, R.; Busson, P.-F.; Parekh, R.V.; et al. Multiple Apical Plasma Membrane Constituents Are Associated with Susceptibility to Meconium Ileus in Individuals with Cystic Fibrosis. Nat. Genet. 2012, 44, 562–569. [Google Scholar] [CrossRef]
  80. Gong, J.; Wang, F.; Xiao, B.; Panjwani, N.; Lin, F.; Keenan, K.; Avolio, J.; Esmaeili, M.; Zhang, L.; He, G.; et al. Genetic Association and Transcriptome Integration Identify Contributing Genes and Tissues at Cystic Fibrosis Modifier Loci. PLoS Genet. 2019, 15, e1008007. [Google Scholar] [CrossRef]
  81. Wang, Y.-Y.; Lin, Y.-H.; Wu, Y.-N.; Chen, Y.-L.; Lin, Y.-C.; Cheng, C.-Y.; Chiang, H.-S. Loss of SLC9A3 Decreases CFTR Protein and Causes Obstructed Azoospermia in Mice. PLoS Genet. 2017, 13, e1006715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Sharma, H.; Mavuduru, R.S.; Singh, S.K.; Prasad, R. Heterogeneous Spectrum of Mutations in CFTR Gene from Indian Patients with Congenital Absence of the Vas Deferens and Their Association with Cystic Fibrosis Genetic Modifiers. Mol. Hum. Reprod. 2014, 20, 827–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Yi, S.; Pierucci-Alves, F.; Schultz, B.D. Transforming Growth Factor-Β1 Impairs CFTR-Mediated Anion Secretion across Cultured Porcine Vas Deferens Epithelial Monolayer via the P38 MAPK Pathway. Am. J. Physiol.-Cell. Physiol. 2013, 305, C867–C876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Trouvé, P.; Génin, E.; Férec, C. In Silico Search for Modifier Genes Associated with Pancreatic and Liver Disease in Cystic Fibrosis. PLoS ONE 2017, 12, e0173822. [Google Scholar] [CrossRef]
  85. Felderbauer, P.; Hoffmann, P.; Einwächter, H.; Bulut, K.; Ansorge, N.; Schmitz, F.; Schmidt, W.E. A Novel Mutation of the Calcium Sensing Receptor Gene Is Associated with Chronic Pancreatitis in a Family with Heterozygous SPINK1 Mutations. BMC Gastroenterol. 2003, 3, 34. [Google Scholar] [CrossRef] [Green Version]
  86. Szmola, R.; Sahin-Toth, M. Pancreatitis-Associated Chymotrypsinogen C (CTRC) Mutant Elicits Endoplasmic Reticulum Stress in Pancreatic Acinar Cells. Gut 2010, 59, 365–372. [Google Scholar] [CrossRef]
  87. Sofia, V.M.; Surace, C.; Terlizzi, V.; Da Sacco, L.; Alghisi, F.; Angiolillo, A.; Braggion, C.; Cirilli, N.; Colombo, C.; Di Lullo, A.; et al. Trans-Heterozygosity for Mutations Enhances the Risk of Recurrent/Chronic Pancreatitis in Patients with Cystic Fibrosis. Mol. Med. 2018, 24, 38. [Google Scholar] [CrossRef] [Green Version]
  88. Pereira, S.V.-N.; Ribeiro, J.D.; Ribeiro, A.F.; Bertuzzo, C.S.; Marson, F.A.L. Novel, Rare and Common Pathogenic Variants in the CFTR Gene Screened by High-Throughput Sequencing Technology and Predicted by in Silico Tools. Sci. Rep. 2019, 9, 6234. [Google Scholar] [CrossRef] [Green Version]
  89. Bergougnoux, A.; Deletang, K.; Varilh, J.; Houriez, F.; Altieri, J.-P.; Koenig, M.; Férec, C.; Mireille, C.; Bienvenu, T.; Audrezet, M.-P.; et al. Large Phenotypic Spectrum Associated with Two New Deep Intronic Variants on the CFTR Gene. Eur. Respir. J. 2018, 52, PA3422. [Google Scholar] [CrossRef]
  90. Lefferts, J.W.; Boersma, V.; Hagemeijer, M.C.; Hajo, K.; Beekman, J.M.; Splinter, E. Targeted Locus Amplification and Haplotyping. In Haplotyping: Methods and Protocols; Peters, B.A., Drmanac, R., Eds.; Methods in Molecular Biology; Springer US: New York, NY, USA, 2023; pp. 31–48. ISBN 978-1-07-162819-5. [Google Scholar]
  91. Keating, D.; Marigowda, G.; Burr, L.; Daines, C.; Mall, M.A.; McKone, E.F.; Ramsey, B.W.; Rowe, S.M.; Sass, L.A.; Tullis, E.; et al. VX-445-Tezacaftor-Ivacaftor in Patients with Cystic Fibrosis and One or Two Phe508del Alleles. N. Engl. J. Med. 2018, 379, 1612–1620. [Google Scholar] [CrossRef]
  92. Davies, J.C.; Moskowitz, S.M.; Brown, C.; Horsley, A.; Mall, M.A.; McKone, E.F.; Plant, B.J.; Prais, D.; Ramsey, B.W.; Taylor-Cousar, J.L.; et al. VX-659–Tezacaftor–Ivacaftor in Patients with Cystic Fibrosis and One or Two Phe508del Alleles. N. Engl. J. Med. 2018, 379, 1599–1611. [Google Scholar] [CrossRef]
  93. Trouvé, P.; Férec, C.; Génin, E. The Interplay between the Unfolded Protein Response, Inflammation and Infection in Cystic Fibrosis. Cells 2021, 10, 2980. [Google Scholar] [CrossRef]
  94. Chevalier, B.; Hinzpeter, A. The Influence of CFTR Complex Alleles on Precision Therapy of Cystic Fibrosis. J. Cyst. Fibros. 2020, 19, S15–S18. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Differential expressions of the CFTR gene in a human. High CFTR expressions are detected in reproductive tract, pancreas, small intestine and salivary glands. Lower levels are observed in liver and lungs.
Figure 1. Differential expressions of the CFTR gene in a human. High CFTR expressions are detected in reproductive tract, pancreas, small intestine and salivary glands. Lower levels are observed in liver and lungs.
Ijms 24 10678 g001
Figure 2. Proximal regulatory elements of the CFTR promoter. The CFTR translation start site is represented by the ATG codon, and upstream different motifs are present. The minimal promoter starts at −34 bp from the ATG codon to −358 bp. To allow correct expression of the CFTR gene, the promoter is composed of several regulatory elements, such as a Cyclic AMP Response Element motif, a Y-box motif (inverted CCAAT element), a CArG-like motif, binding motifs for Activator Protein-1 (AP-1), Specificity Protein 1 (SP1), an iκβ-like motif and a HIF Responsive Element (HRE) motif.
Figure 2. Proximal regulatory elements of the CFTR promoter. The CFTR translation start site is represented by the ATG codon, and upstream different motifs are present. The minimal promoter starts at −34 bp from the ATG codon to −358 bp. To allow correct expression of the CFTR gene, the promoter is composed of several regulatory elements, such as a Cyclic AMP Response Element motif, a Y-box motif (inverted CCAAT element), a CArG-like motif, binding motifs for Activator Protein-1 (AP-1), Specificity Protein 1 (SP1), an iκβ-like motif and a HIF Responsive Element (HRE) motif.
Ijms 24 10678 g002
Figure 3. Cis-regulatory elements across the CFTR locus. Hi-C data from UCSC genome browser (IMR90 cells—hg19) show a CFTR TAD. This TAD is delineated by boundaries at −80.1 kb of TSS of the CFTR gene in ASZ1 gene and +48.9 kb of the last codon of CFTR gene in CTTNBP2 gene. Boundaries are represented by red squares and CREs by black squares.
Figure 3. Cis-regulatory elements across the CFTR locus. Hi-C data from UCSC genome browser (IMR90 cells—hg19) show a CFTR TAD. This TAD is delineated by boundaries at −80.1 kb of TSS of the CFTR gene in ASZ1 gene and +48.9 kb of the last codon of CFTR gene in CTTNBP2 gene. Boundaries are represented by red squares and CREs by black squares.
Ijms 24 10678 g003
Figure 4. Cis-regulation of the CFTR gene in airway epithelial cells. Three-dimensional representation of the CFTR locus (A). Chromatin loopings allow CREs to interact with the CFTR promoter. Coding sequences are represented in thick lines. Chromatin module (B) with multiple transcription factors binding specific CREs. −44 kb and −35 kb are the most implicated CREs in airway epithelial cells.
Figure 4. Cis-regulation of the CFTR gene in airway epithelial cells. Three-dimensional representation of the CFTR locus (A). Chromatin loopings allow CREs to interact with the CFTR promoter. Coding sequences are represented in thick lines. Chromatin module (B) with multiple transcription factors binding specific CREs. −44 kb and −35 kb are the most implicated CREs in airway epithelial cells.
Ijms 24 10678 g004
Figure 5. Cis-regulation of the CFTR gene in intestinal cells. Three-dimensional representation of the CFTR locus (A). Chromatin loopings allow CREs to interact with the CFTR promoter. Coding sequences are represented in thick lines. Chromatin module (B) with multiple transcription factors binding specific CREs. CREs in introns 1 and 12 are the most implicated CREs in intestinal epithelial cells.
Figure 5. Cis-regulation of the CFTR gene in intestinal cells. Three-dimensional representation of the CFTR locus (A). Chromatin loopings allow CREs to interact with the CFTR promoter. Coding sequences are represented in thick lines. Chromatin module (B) with multiple transcription factors binding specific CREs. CREs in introns 1 and 12 are the most implicated CREs in intestinal epithelial cells.
Ijms 24 10678 g005
Figure 6. Cis-regulation of the CFTR gene in epididymis cells. Three-dimensional representation of the CFTR locus (A). Chromatin loopings allow CREs to interact with the CFTR promoter. Coding sequences are represented in thick lines. Chromatin module (B) with multiple transcription factors binding specific cCREs. Only predicted regions are highlighted.
Figure 6. Cis-regulation of the CFTR gene in epididymis cells. Three-dimensional representation of the CFTR locus (A). Chromatin loopings allow CREs to interact with the CFTR promoter. Coding sequences are represented in thick lines. Chromatin module (B) with multiple transcription factors binding specific cCREs. Only predicted regions are highlighted.
Ijms 24 10678 g006
Figure 7. Cis-regulation of the CFTR gene in pancreatic cells. Three-dimensional representation of the CFTR locus (A). Chromatin loopings allow CREs to interact with the CFTR promoter. Coding sequences are represented in thick lines. Chromatin module (B) with multiple transcription factors binding specific cCREs. Only predicted regions are highlighted.
Figure 7. Cis-regulation of the CFTR gene in pancreatic cells. Three-dimensional representation of the CFTR locus (A). Chromatin loopings allow CREs to interact with the CFTR promoter. Coding sequences are represented in thick lines. Chromatin module (B) with multiple transcription factors binding specific cCREs. Only predicted regions are highlighted.
Ijms 24 10678 g007
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Blotas, C.; Férec, C.; Moisan, S. Tissue-Specific Regulation of CFTR Gene Expression. Int. J. Mol. Sci. 2023, 24, 10678. https://doi.org/10.3390/ijms241310678

AMA Style

Blotas C, Férec C, Moisan S. Tissue-Specific Regulation of CFTR Gene Expression. International Journal of Molecular Sciences. 2023; 24(13):10678. https://doi.org/10.3390/ijms241310678

Chicago/Turabian Style

Blotas, Clara, Claude Férec, and Stéphanie Moisan. 2023. "Tissue-Specific Regulation of CFTR Gene Expression" International Journal of Molecular Sciences 24, no. 13: 10678. https://doi.org/10.3390/ijms241310678

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop