Next Article in Journal
Effect of Sex Differences in Silicotic Mice
Previous Article in Journal
Comparative Analysis of Alternative Splicing in Two Contrasting Apple Cultivars Defense against Alternaria alternata Apple Pathotype Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Modifier Factors of Cystic Fibrosis Phenotypes: A Focus on Modifier Genes

1
Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France
2
Inovarion, 75005 Paris, France
3
Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Trousseau, Service de Pneumologie Pédiatrique, 75012 Paris, France
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(22), 14205; https://doi.org/10.3390/ijms232214205
Submission received: 18 October 2022 / Revised: 7 November 2022 / Accepted: 11 November 2022 / Published: 17 November 2022
(This article belongs to the Topic Cystic Fibrosis)

Abstract

:
Although cystic fibrosis (CF) is recognized as a monogenic disease, due to variants within the CFTR (Cystic Fibrosis Transmembrane Regulator) gene, an extreme clinical heterogeneity is described among people with CF (pwCF). Apart from the exocrine pancreatic status, most studies agree that there is little association between CFTR variants and disease phenotypes. Environmental factors have been shown to contribute to this heterogeneity, accounting for almost 50% of the variability of the lung function of pwCF. Nevertheless, pwCF with similar CFTR variants and sharing the same environment (such as in siblings) may have highly variable clinical manifestations not explained by CFTR variants, and only partly explained by environmental factors. It is recognized that genetic variants located outside the CFTR locus, named “modifier genes”, influence the clinical expression of the disease. This short review discusses the latest studies that have described modifier factors associated with the various CF phenotypes as well as the response to the recent CFTR modulator therapies.

1. Introduction

Cystic fibrosis (CF) affects about 80,000 patients worldwide. It is caused by variants in the gene coding for the chloride channel CFTR (Cystic Fibrosis Transmembrane conductance Regulator) ubiquitously expressed in epithelia [1]. CFTR gene variants can cause defects of synthesis or of function of the CFTR protein in epithelial cells, leading to a reduction in the permeability to chloride ions and a lack of hydration of secretions, which become viscous and sticky [2,3]. The disease affects predominately the lungs, gastrointestinal tract, pancreas, liver and biliary tract, and also the sweat glands, genital tract and osteoarticular system.
More than 2000 variants in CFTR have been described and assembled into six classes according to their functional consequences on the protein, with alteration of either: (I) its production; (II) its addressing to the plasma membrane; (III) its regulation (gating variant); (IV) its conduction; and (V and VI) the stability of the mRNA or of the mature protein [2,3,4]. Depending on the impact on the protein, these classes are further described as ‘minimal function variant’ (classes I, II and III) or ‘residual function variant’ (classes IV, V and VI). This classification has therapeutic implications as CFTR modulator therapies target some specific classes [2,3,4]. Exocrine pancreatic function status, whether deficient or preserved, is also associated with CFTR variants. Indeed, people with CF (pwCF) with pancreatic insufficiency are those carrying two minimal function variants and are more prone to suffer a severe CF phenotype, whereas pwCF with normal pancreatic function carry at least one residual function variant and have less clinical impairment [2,3,4].
Since the description of the disease, numerous observations have documented the extreme clinical heterogeneity of the disease. The identification of the CFTR gene and its principal variants has led to many studies aimed at establishing relationships between genotypic characteristics and clinical manifestations [5,6,7]. Apart from the exocrine pancreatic status, most studies, including those carried out in twins and siblings, agree in concluding that there is little association between CFTR variants and disease phenotypes [7,8,9]. Environmental factors have been shown to contribute to this heterogeneity, accounting for almost 50% of the variability of the lung function of pwCF [10]. Nevertheless, pwCF with similar CFTR variants and sharing the same environment (such as in siblings) may have highly variable clinical manifestations [11] not explained by CFTR variants, and only partly explained by environmental factors. It is recognized that genetic variants located outside the CFTR locus and called “modifier genes” influence the clinical expression of the disease [6,12,13]. This short review discusses the latest studies that have described modifier factors associated with the various CF phenotypes as well as the response to the recent CFTR modulator therapies.

2. Socio-Demographic and Environmental Modifier Factors

Several studies have highlighted the contribution of demographic and clinical factors in the variability of respiratory disease. It has been shown that women and pwCF with poor nutritional status are at greater risk of severe disease [14,15]; pwCF with CF-related diabetes (CFRD) have a more rapid decline in respiratory function [16]; CF-liver disease (CFLD) increases susceptibility to infection [17]; and the age at chronic colonization with Pseudomonas aeruginosa (Pa) decreases in the most recent cohorts [18,19]. Nevertheless, many of these factors have been questioned and remain controversial [20,21].
Regarding environmental factors, the heterogeneity of phenotypes could be modulated by passive smoking [22,23], temperature [24], outdoor [25,26,27,28] and indoor air pollution [29], seasonality [30], climate and geography [31], socioeconomic status and access to health care [32,33]. The contribution of environmental factors is nevertheless variable across the phenotypes, ranging from 40% for variability in airway obstruction to 20% for nutritional status [5].

3. Modifier Genes

3.1. Genetic Polymorphism

Modifier genes are genetic variants or polymorphisms, i.e., individual variations in the sequence of a gene, which are not pathological in themselves but can modulate disease severity [5,6]. A genetic polymorphism is determined by the coexistence of several alleles for a given gene or locus. A gene is said to be polymorphic if at least two alleles exist at a frequency greater than 1% in the population. Several types of polymorphisms can be distinguished, including minisatellite and microsatellite markers, deletions or insertions of short nucleotide sequences and single nucleotide polymorphisms (SNPs). SNPs correspond to a substitution of one purine or pyrimidine base for another one. When they are located in the coding region of a gene, they may lead to a modification of function due to the modification of the transcription of the mRNA (messenger ribonucleic acid) stability or of the protein conformation. SNPs located outside of genes have less obvious functional consequences and are generally used as markers for genetic linkage studies. SNPs are widespread, with an estimated frequency of one SNP in every 300 to 1000 base pairs [34], so more than 84.7 million SNPs are in the human genome [35]. These variations are mostly silent but are thought to be involved in the susceptibility to certain diseases, their severity, or the response to treatments.

3.2. A Priori and Non-A Priori Approaches

Two approaches have been applied to search for modifier genes in CF: an a priori or candidate-gene approach based on the known pathophysiology of the phenotypic trait; and a non-a priori approach carried out by whole genome analysis (GWAS for genome wide association study or WGS for whole genome sequencing) and by exome sequencing (WES for whole exome sequencing). To provide convincing statistical evidence for association and to rule out associations due to biases, these types of analyses need to be conducted in large cohorts and then replicated in independent cohorts. Thus, consortia were formed in France and North America. In France, our team has been coordinating a national study since 2006, in which nearly 5000 French pwCF are participating out of 7500 followed up in CF expert centers [36]. The merger of these two consortia has led to the creation of an international consortium that initially analyzed the GWAS of 5000 North American and 1250 French pwCF. Several regions of the genome have been shown to be associated with severity of the respiratory disease [37], risk of meconium ileus [38,39], exocrine [40] and endocrine pancreatic status [41,42], sweat chloride levels [43] and immune response [44].

3.3. Modifier Genes of the Lung Function

Lung function is the most studied phenotype in the search for modifier genes in CF lung disease, and multiple factors of variability have been suggested. Lung function is usually estimated by measurements of the forced expiratory volume in one second (FEV1) expressed as percent predicted of reference values (ppFEV1). It has been estimated that the lung function severity would have a heritability of more than 50% independent of CFTR [11]. The gene-candidate approach identified four main families of modifier genes of lung function based on their physiological role: genes involved in either tissue repair, host defense and inflammation, epithelial surface ion transport and mucus secretion, and response to drug therapy (detailed in the reviews of Butnariu et al. [45] and Sepahzad et al. [46]). Regarding the non-a priori approach, three (meta) GWAS were conducted, which identified seven genomic regions of interest associated with the severity of lung function (Table 1) [37,47,48].

3.4. Modifier Genes of Pseudomonas aeruginosa Infection

CF lung disease is caused by structural lung damage due to an exacerbated inflammation associated with recurrent bacterial infections. Pa is one of the most commonly isolated pathogens in the airways of pwCF. After Pa initial acquisition (Pa-IA), and despite effective eradication therapies, persistent infection may occur leading to Pa chronic colonization (Pa-CC), which is extremely difficult, if not impossible, to eradicate.
Studies conducted in twins and siblings with CF estimate that Pa-CC is highly heritable, up to 85% [49], with a contribution of modifier genes thought to be around 55% compared to ~23% for CFTR variants and ~23% for environmental factors [5]. These data attest the important role of genetic factors in the variability of the infectious phenotype of pwCF. Two non-a priori studies (WES) identified Caveolin 2 (CAV2), Dynactin subunit 4 (DCTN4) and Transmembrane channel like 6 (TMC6) as modifier genes of age in Pa-CC [50,51]. Several candidate gene studies highlighted the role of modifiers of Pa-IA (as Solute carrier family 9 member 3 (SLC9A3) and Solute carrier family 6 member 14 (SLC6A14) [52]); Pa-CC (as Taste receptor 2 member 38 (TAS2R38) [53] and Interleukin-10 (IL10) [54]); and both Pa-IA and Pa-CC (as Tumor necrosis factor (TNF) [55] and Mannose-binding lectin 2 (MBL2) [56,57]).
We recently analyzed a large cohort of 1231 French children with CF to identify risk factors and genetic modifiers for Pa-IA, Pa-CC and progression from Pa-IA to Pa-CC [19].We found that Pa infection occurred early in childhood, with the median age of patients being 5.1 years at Pa-IA. One-quarter of the pwCF were chronically colonized with Pa by the age of 14.7 years, within 6.3 years after Pa-IA. We highlighted that Pa-CC occurs later in life for pwCF in the most recent birth cohorts, which may reflect improvements in the standardization of therapeutic care, especially since the establishment of specialized CF care centers. Interestingly, we observed that CFRD and CFLD were risk factors for Pa, while gender, CFTR variants and CF center size were not. Moreover, we confirmed that genetic variants of TNF, DCTN4, SLC9A3 and CAV2 were associated with Pa.

3.5. Modifier Genes of Meconium Ileus

Around 15% of pwCF are born with an intestinal obstruction at birth named meconium ileus (MI). A first GWAS identified several genes associated with MI susceptibility, especially SLC6A14, SLC26A9 (Solute carrier family 26 member 9) and SLC9A3 [38]. These genes belong to the SLC (solute carriers) family, which encodes membrane transport proteins. The different members of the SLC family transport a large panel of substances including charged and uncharged organic molecules as well as inorganic ions. A more recent meta-GWAS for MI susceptibility confirmed the previously identified SLC26A9 and SLC6A14 and found two new loci, ATP12A (ATPase H+/K+ transporting non-gastric alpha2 subunit) and PRSS1 (Serine protease 1) [39].

3.6. Modifier Genes of CF-Related Diabetes

CFRD is a frequent complication of CF. Its prevalence increases with age, affecting more than 90% of pwCF carrying two minimal function CFTR variants by their sixth decade [41]. Age at onset varies considerably (e.g., from 10 to 50 years old) and has been shown to be highly heritable but independent of CFTR genotype, suggesting an important role for modifiers genes.
An initial gene-candidate study highlighted that the TCF7L2 (Transcription factor 7-like 2) gene, known to be associated with type 2 diabetes in the general population, was also associated with CFRD [58]. This association was confirmed in an early GWAS, which also identified the SLC26A9 gene and genomic regions next to the CDKAL1 (Cyclin dependent kinase 5 regulatory associated protein 1-like 1), CDKN2A/B (Cyclin dependent kinase inhibitor 2A/B) and IGF2BP2 (Insulin-like growth factor-binding protein 2) genes [59]. More recently, the SLC26A9 gene has been confirmed in a candidate gene study [60] as well as a meta-GWAS [41]. The later meta-GWAS also replicated the involvement of the TCF7L2 gene and identified the PTMA (Prothymosin alpha) gene [41].
Innovatively, using genetic and clinical measures available at birth, a CFRD prediction model has been recently constructed [42]. This model found that the strongest CFRD predictors included sex, CFTR severity score and several genetic variants. A web-based application was developed to provide practitioners with patient-specific CFRD risk to guide monitoring and treatment.

3.7. Modifier Genes of CF-Liver Disease

CFLD includes various hepatobiliary abnormalities. Focal biliary cirrhosis is the most clinically relevant in CFLD, and extension of the initially focal fibrogenic process may indeed lead to multilobular biliary cirrhosis with subsequent portal hypertension and related complications [61,62,63]. Modifier genes of CFLD have so far only been investigated by gene-candidate approaches. Studies have explored the potential role of SERPINA1 (Alpha-1 Antitrypsine) [64], ACE (Angiotensin I-converting enzyme) [65], GSTP1 (Glutathione S-transferases P1) [66], MBL2 [67,68], TGFβ1 (Transforming growth factor beta 1) [65] and ABCB4 (ATP Binding cassette subfamily B member 4) [68]. The association between the SERPINA1 gene and CFLD severity was the only one to be successfully replicated, showing a highly increased risk for pwCF carrying the Z allele [69,70]. To date, no GWAS analysis has been conducted. Nevertheless, one study has highlighted the probable association between CFLD and six new genes using a system biology approach to investigate the functional relationship between the previously studied genes [71]. This particular methodology combines database and literature mining, gene expression study and network analysis as well as pathway enrichment analysis and protein–protein interactions.

4. Modifier Factors of CFTR Modulators Response

The management of CF, which was essentially symptomatic until recently, has led to a marked improvement in survival, which now exceeds 50 years [36]. These symptomatic treatments target the side effects of CFTR protein dysfunction. For respiratory disease, for example, treatments consist of improving secretion drainage, mucus hydration and/or mucociliary clearance, as well as treating recurrent infections or exacerbated inflammation [72]. In the last decade, considerable efforts have led to the development of therapies that target the CFTR proteins named CFTR modulators [73]. Interpatient variability in the response to these modulators has been shown to be partly explained by modifier genes.

4.1. Modifier Genes and Response to Ivacaftor

Since 2012, pwCF carrying various CFTR gating variants could be treated with ivacaftor, a potentiator therapy that increases the open probability of CFTR-channels. Its efficacy was initially demonstrated in pwCF carrying at least one G551D variant and was later extended to other less common gating variants [74]. Ivacaftor was shown to allow the recovery of 30 to 40% of ‘normal’ CFTR function and is associated with significant clinical benefits, such as gain of lung function and reduced number of pulmonary exacerbations and hospitalizations [75]. Significant inter-patient variability in response to ivacaftor is observed, particularly with regard to lung response. Initiation of ivacaftor treatment is indeed associated with a mean ppFEV1 improvement of 10.4% at 24 weeks of treatment with a variability of ±15.5% between pwCF [76]. In French pwCF, we have shown that the variability of the respiratory response to ivacaftor was associated with the SLC26A9 gene [77], confirming the results observed in Canadians [78]. However, these results were recently controverted in pwCF from the US [79].

4.2. Modifier Genes and Response to Lumacaftor-Ivacaftor

Lumacaftor, a CFTR corrector that improves the processing and trafficking of the Phe508del-CFTR protein, was combined with ivacaftor to treat pwCF homozygous for the Phe508del-CFTR variant. A phase 2 trial of this combined therapy, lumacaftor–ivacaftor (LUMA–IVA), demonstrated improvements in lung function, measured by an increase of the ppFEV1, and nutritional status, evaluated by the body mass index (BMI) [80]. Since the marketing authorizations granted in 2015, the clinical benefits of LUMA–IVA have been questioned, particularly because of its high cost [81]. In addition, phase 3 trials and real-life studies over the first 2 years of treatment demonstrated highly heterogeneous responses in lung function and nutritional status [82,83,84,85,86,87,88]. These studies also highlighted the inter-individual variability of the airway response and the limited tolerance to treatment, with high discontinuation rates ranging from 17.2% to 28.9%. Predisposing factors for interruption and for the response variability were shown to be baseline lung function, age at treatment initiation and gender [82,84,85,86,87,88].
Our team has very recently been able to identify factors, including modifier genes, influencing the respiratory and nutritional response to LUMA–IVA [89]. Among the 4981 pwCF of our gene-modifiers cohort, 878 were eligible for analysis of the respiratory and nutritional response to LUMA–IVA, and 765 were included. We evaluated the respiratory and nutritional responses after 1 to 2 years of LUMA–IVA and then looked for clinical and genetic factors involved in their variability. We analyzed SLC variants that had been implicated in the variability of response to ivacaftor [77,78] and/or in the severity of several CF phenotypes [37,38,52,90,91,92]. Gains in lung function and nutritional status were observed after 6 months of treatment (on average 2.11 ± 7.81% for ppFEV1 and 0.44 ± 0.77 kg/m2 for BMI) and sustained over the 2 years. We observed that the more severe pwCF gained the most in lung function and nutritional status. While females started with a nutritional status more impaired than males, they had a larger response and regained BMI Z-score values similar to men after 2 years of treatment. We observed no association between SLC variants and the respiratory function response to LUMA–IVA, but SLC6A14 rs12839137 was associated with the nutritional response.

4.3. Modifier Genes and Response to Tezacaftor-Ivacaftor and Elexacaftor-Tezacaftor-Ivacaftor

Therapies combining the modulators tezacaftor–ivacaftor (TEZ–IVA) and elexacaftor–tezacaftor–ivacaftor (ETI) have been marketed more recently. Huge improvements in lung function and nutritional status have been shown in treated patients [93,94,95,96]. So far, no phenotype/genotype association study has investigated the role of modifier genes in the variability of the response to these therapies. Nevertheless, two studies suggest SLC34A2 (Solute carrier family 34 member 2) and SLC26A9 as potential modifiers [97,98].

5. Conclusions and Perspectives

Advances in the understanding of CFTR functions have led to amazing therapeutic progresses. Nevertheless, the observed phenotypic variability highlights the need to better understand the differences between subgroups of pwCF in order to identify prognostic indicators for optimal patient management. More specifically, the identification of modifier genes and a better knowledge of their functions in CF could allow the development of personalized, predictive, preventive and participatory medicine [4]. Several modifier genes of different CF phenotypes have been identified so far, as detailed in this short review (Figure 1).
However, many studies have been carried out using candidate gene studies, which have not been replicated and for which the functionality of the identified genes or variants has not been deeply investigated. The challenge for the future is, as it was initiated for lung function, CFRD and MI, to perform studies without preconceptions to identify modifier genes and variants involved in CFLD infections, as well as validate these modifiers at the functional level. This is necessary to move on to the next step which will consist of establishing polygenic risk scores (PRS) that could predict the degree of severity of the various CF disorders, as realized with CFRD [42], and thus allow targeted and individualized management of pwCF.
To conclude, genomic knowledge will enable individualized medicine through the development of new therapies or by adapting existing ones more specifically. Furthermore, it will facilitate predictive and preventive medicine through the evaluation of the risks of developing complications, which is equivalent to a proactive approach to patient management and oriented towards the preservation of health capital.

Author Contributions

Conceptualization, J.M., M.R., L.G. and H.C.; writing—original draft preparation, J.M., M.R., L.G. and H.C.; writing—review and editing, J.M., M.R., L.G. and H.C. All authors have read and agreed to the published version of the manuscript.

Funding

The French CF Modifier Gene Study was funded by the Fondation pour la Recherche Médicale (grant number “PMJ201810007620” to Julie Mésinèle), Vaincre la Mucoviscidose, Blanche pour Vaincre la Mucoviscidose, Agir et Informer Contre la Mucoviscidose, Institut National de la Santé et de la Recherche Médicale.

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. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Riordan, J.R.; Rommens, J.M.; Kerem, B.; 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] [PubMed]
  2. Corvol, H.; Taytard, J.; Tabary, O.; Le Rouzic, P.; Guillot, L.; Clement, A. Challenges of personalized medicine for cystic fibrosis. Arch. Pediatr. 2015, 22, 778–786. [Google Scholar] [CrossRef] [PubMed]
  3. Elborn, J.S. Cystic fibrosis. Lancet 2016, 388, 2519–2531. [Google Scholar] [CrossRef]
  4. Corvol, H.; Thompson, K.E.; Tabary, O.; le Rouzic, P.; Guillot, L. Translating the genetics of cystic fibrosis to personalized medicine. Transl. Res. 2015, 168, 40–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Cutting, G.R. Cystic fibrosis genetics: From molecular understanding to clinical application. Nat. Rev. Genet. 2014, 16, 45–56. [Google Scholar] [CrossRef] [Green Version]
  6. Paranjapye, A.; Ruffin, M.; Harris, A.; Corvol, H. Genetic variation in CFTR and modifier loci may modulate cystic fibrosis disease severity. J. Cyst. Fibros. 2020, 19 (Suppl. 1), S10–S14. [Google Scholar] [CrossRef] [Green Version]
  7. Rowntree, R.K.; Harris, A. The phenotypic consequences of CFTR mutations. Ann. Hum. Genet. 2003, 67, 471–485. [Google Scholar] [CrossRef]
  8. Santis, G.; Osborne, L.; Knight, R.A.; Hodson, M.E. Independent genetic determinants of pancreatic and pulmonary status in cystic fibrosis. Lancet 1990, 336, 1081–1084. [Google Scholar] [CrossRef]
  9. Mekus, F.; Ballmann, M.; Bronsveld, I.; Bijman, J.; Veeze, H.; Tummler, B. Categories of deltaF508 homozygous cystic fibrosis twin and sibling pairs with distinct phenotypic characteristics. Twin Res. 2000, 3, 277–293. [Google Scholar] [CrossRef]
  10. Collaco, J.M.; Blackman, S.M.; McGready, J.; Naughton, K.M.; Cutting, G.R. Quantification of the relative contribution of environmental and genetic factors to variation in cystic fibrosis lung function. J. Pediatr. 2010, 157, 802–807.e3. [Google Scholar] [CrossRef]
  11. Vanscoy, L.L.; Blackman, S.M.; Collaco, J.M.; Bowers, A.; Lai, T.; Naughton, K.; Algire, M.; McWilliams, R.; Beck, S.; Hoover-Fong, J.; et al. Heritability of lung disease severity in cystic fibrosis. Am. J. Respir. Crit. Care Med. 2007, 175, 1036–1043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Cutting, G.R. Modifier genes in Mendelian disorders: The example of cystic fibrosis. Ann. N. Y. Acad. Sci. 2010, 1214, 57–69. [Google Scholar] [CrossRef] [PubMed]
  13. Ferec, C.; Scotet, V.; Beucher, J.; Corvol, H. Genetics and modifier genes, atypical and rare forms. Arch. Pediatr. 2012, 19 (Suppl. 1), S3–S7. [Google Scholar] [CrossRef] [PubMed]
  14. Corey, M.; Edwards, L.; Levison, H.; Knowles, M. Longitudinal analysis of pulmonary function decline in patients with cystic fibrosis. J. Pediatr. 1997, 131, 809–814. [Google Scholar] [CrossRef]
  15. Konstan, M.W.; Morgan, W.J.; Butler, S.M.; Pasta, D.J.; Craib, M.L.; Silva, S.J.; Stokes, D.C.; Wohl, M.E.; Wagener, J.S.; Regelmann, W.E.; et al. Risk factors for rate of decline in forced expiratory volume in one second in children and adolescents with cystic fibrosis. J. Pediatr. 2007, 151, 134–139.e1. [Google Scholar] [CrossRef]
  16. Schaedel, C.; de Monestrol, I.; Hjelte, L.; Johannesson, M.; Kornfalt, R.; Lindblad, A.; Strandvik, B.; Wahlgren, L.; Holmberg, L. Predictors of deterioration of lung function in cystic fibrosis. Pediatr. Pulmonol. 2002, 33, 483–491. [Google Scholar] [CrossRef]
  17. Toledano, M.B.; Mukherjee, S.K.; Howell, J.; Westaby, D.; Khan, S.A.; Bilton, D.; Simmonds, N.J. The emerging burden of liver disease in cystic fibrosis patients: A UK nationwide study. PLoS ONE 2019, 14, e0212779. [Google Scholar] [CrossRef]
  18. Crull, M.R.; Somayaji, R.; Ramos, K.J.; Caldwell, E.; Mayer-Hamblett, N.; Aitken, M.L.; Nichols, D.P.; Rowhani-Rahbar, A.; Goss, C.H. Changing Rates of Chronic Pseudomonas aeruginosa Infections in Cystic Fibrosis: A Population-Based Cohort Study. Clin. Infect. Dis. 2018, 67, 1089–1095. [Google Scholar] [CrossRef] [Green Version]
  19. Mésinèle, J.; Ruffin, M.; Kemgang, A.; Guillot, L.; Boëlle, P.Y.; Corvol, H. Risk factors for Pseudomonas aeruginosa airway infection and lung function decline in children with cystic fibrosis. J. Cyst. Fibros 2022, 21, 45–51. [Google Scholar] [CrossRef]
  20. van Horck, M.; van de Kant, K.; Winkens, B.; Wesseling, G.; Gulmans, V.; Hendriks, H.; van der Grinten, C.; Jobsis, Q.; Dompeling, E. Risk factors for lung disease progression in children with cystic fibrosis. Eur. Respir. J. 2018, 51, 1702509. [Google Scholar] [CrossRef]
  21. Pittman, J.E.; Noah, H.; Calloway, H.E.; Davis, S.D.; Leigh, M.W.; Drumm, M.; Sagel, S.D.; Accurso, F.J.; Knowles, M.R.; Sontag, M.K. Early childhood lung function is a stronger predictor of adolescent lung function in cystic fibrosis than early Pseudomonas aeruginosa infection. PLoS ONE 2017, 12, e0177215. [Google Scholar] [CrossRef] [PubMed]
  22. Smyth, A.; O’Hea, U.; Williams, G.; Smyth, R.; Heaf, D. Passive smoking and impaired lung function in cystic fibrosis. Arch. Dis. Child 1994, 71, 353–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Collaco, J.M.; Vanscoy, L.; Bremer, L.; McDougal, K.; Blackman, S.M.; Bowers, A.; Naughton, K.; Jennings, J.; Ellen, J.; Cutting, G.R. Interactions between secondhand smoke and genes that affect cystic fibrosis lung disease. JAMA 2008, 299, 417–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Collaco, J.M.; McGready, J.; Green, D.M.; Naughton, K.M.; Watson, C.P.; Shields, T.; Bell, S.C.; Wainwright, C.E.; Group, A.S.; Cutting, G.R. Effect of temperature on cystic fibrosis lung disease and infections: A replicated cohort study. PLoS ONE 2011, 6, e27784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Goss, C.H.; Newsom, S.A.; Schildcrout, J.S.; Sheppard, L.; Kaufman, J.D. Effect of ambient air pollution on pulmonary exacerbations and lung function in cystic fibrosis. Am. J. Respir. Crit. Care Med. 2004, 169, 816–821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Psoter, K.J.; De Roos, A.J.; Mayer, J.D.; Kaufman, J.D.; Wakefield, J.; Rosenfeld, M. Fine particulate matter exposure and initial Pseudomonas aeruginosa acquisition in cystic fibrosis. Ann. Am. Thorac. Soc. 2015, 12, 385–391. [Google Scholar] [CrossRef]
  27. Goeminne, P.C.; Kicinski, M.; Vermeulen, F.; Fierens, F.; De Boeck, K.; Nemery, B.; Nawrot, T.S.; Dupont, L.J. Impact of air pollution on cystic fibrosis pulmonary exacerbations: A case-crossover analysis. Chest 2013, 143, 946–954. [Google Scholar] [CrossRef]
  28. Psoter, K.J.; De Roos, A.J.; Wakefield, J.; Mayer, J.D.; Rosenfeld, M. Air pollution exposure is associated with MRSA acquisition in young U.S. children with cystic fibrosis. BMC Pulm. Med. 2017, 17, 106. [Google Scholar] [CrossRef]
  29. Carson, S.W.; Psoter, K.; Koehler, K.; Siklosi, K.R.; Montemayor, K.; Toporek, A.; West, N.E.; Lechtzin, N.; Hansel, N.N.; Collaco, J.M.; et al. Indoor air pollution exposure is associated with greater morbidity in cystic fibrosis. J. Cyst. Fibros. 2021, 21, e129–e135. [Google Scholar] [CrossRef]
  30. Psoter, K.J.; De Roos, A.J.; Wakefield, J.; Mayer, J.D.; Rosenfeld, M. Seasonality of acquisition of respiratory bacterial pathogens in young children with cystic fibrosis. BMC Infect. Dis. 2017, 17, 411. [Google Scholar] [CrossRef]
  31. Psoter, K.J.; DE Roos, A.J.; Wakefield, J.; Mayer, J.D.; Bryan, M.; Rosenfeld, M. Association of meteorological and geographical factors and risk of initial Pseudomonas aeruginosa acquisition in young children with cystic fibrosis. Epidemiol. Infect. 2015, 144, 1075–1083. [Google Scholar] [CrossRef]
  32. Curtis, J.R.; Burke, W.; Kassner, A.W.; Aitken, M.L. Absence of health insurance is associated with decreased life expectancy in patients with cystic fibrosis. Am. J. Respir. Crit. Care Med. 1997, 155, 1921–1924. [Google Scholar] [CrossRef]
  33. Schechter, M.S.; Shelton, B.J.; Margolis, P.A.; Fitzsimmons, S.C. The Association of Socioeconomic Status with Outcomes in Cystic Fibrosis Patients in the United States. Am. J. Respir. Crit. Care Med. 2001, 163, 1331–1337. [Google Scholar] [CrossRef]
  34. Lander, E.S.; Linton, L.M.; Birren, B.; Nusbaum, C.; Zody, M.C.; Baldwin, J.; Devon, K.; Dewar, K.; Doyle, M.; FitzHugh, W.; et al. Initial sequencing and analysis of the human genome. Nature 2001, 409, 860–921. [Google Scholar] [CrossRef] [Green Version]
  35. 1000 Genomes Project Consortium. A global reference for human genetic variation. Nature 2015, 526, 68–74. [Google Scholar] [CrossRef] [Green Version]
  36. Vaincre-la-Mucoviscidose. Registre Français de la Mucoviscidose—Bilan des Données 2020; Vaincre La Mucovisidose: Paris, France, 2022; Available online: https://www.vaincrelamuco.org/sites/default/files/registre_francais_de_la_mucoviscidose_bilan_2020.pdf (accessed on 17 October 2022).
  37. Corvol, H.; Blackman, S.M.; Boelle, 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]
  38. 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]
  39. 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]
  40. Miller, M.R.; Soave, D.; Li, W.; Gong, J.; Pace, R.G.; Boelle, P.Y.; Cutting, G.R.; Drumm, M.L.; Knowles, M.R.; Sun, L.; et al. Variants in Solute Carrier SLC26A9 Modify Prenatal Exocrine Pancreatic Damage in Cystic Fibrosis. J. Pediatr. 2015, 166, 1152–1157.e6. [Google Scholar] [CrossRef] [Green Version]
  41. Aksit, M.A.; Pace, R.G.; Vecchio-Pagán, B.; Ling, H.; Rommens, J.M.; Boelle, P.-Y.; Guillot, L.; Raraigh, K.S.; Pugh, E.; Zhang, P.; et al. Genetic Modifiers of Cystic Fibrosis-Related Diabetes Have Extensive Overlap with Type 2 Diabetes and Related Traits. J. Clin. Endocrinol. Metab. 2020, 105, 1401–1415. [Google Scholar] [CrossRef]
  42. Lin, Y.-C.; Keenan, K.; Gong, J.; Panjwani, N.; Avolio, J.; Lin, F.; Adam, D.; Barrett, P.; Bégin, S.; Berthiaume, Y.; et al. Cystic fibrosis–related diabetes onset can be predicted using biomarkers measured at birth. Genet. Med. 2021, 23, 927–933. [Google Scholar] [CrossRef]
  43. Collaco, J.M.; Blackman, S.M.; Raraigh, K.S.; Corvol, H.; Rommens, J.M.; Pace, R.G.; Boelle, P.-Y.; McGready, J.; Sosnay, P.R.; Strug, L.J.; et al. Sources of Variation in Sweat Chloride Measurements in Cystic Fibrosis. Am. J. Respir. Crit. Care Med. 2016, 194, 1375–1382. [Google Scholar] [CrossRef] [Green Version]
  44. Polineni, D.; Dang, H.; Gallins, P.J.; Jones, L.C.; Pace, R.G.; Stonebraker, J.R.; Commander, L.A.; Krenicky, J.E.; Zhou, Y.-H.; Corvol, H.; et al. Airway Mucosal Host Defense Is Key to Genomic Regulation of Cystic Fibrosis Lung Disease Severity. Am. J. Respir. Crit. Care Med. 2018, 197, 79–93. [Google Scholar] [CrossRef]
  45. Butnariu, L.I.; Țarcă, E.; Cojocaru, E.; Rusu, C.; Moisă, M.; Constantin, M.-M.L.; 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]
  46. Sepahzad, A.; Morris-Rosendahl, D.; Davies, J. Cystic Fibrosis Lung Disease Modifiers and Their Relevance in the New Era of Precision Medicine. Genes 2021, 12, 562. [Google Scholar] [CrossRef]
  47. Wright, F.A.; Strug, L.J.; Doshi, V.K.; Commander, C.; Blackman, S.; Sun, L.; Berthiaume, Y.; Cutler, D.M.; Cojocaru, A.; Collaco, J.M.; et al. Genome-wide association and linkage identify modifier loci of lung disease severity in cystic fibrosis at 11p13 and 20q13.2. Nat. Genet. 2011, 43, 539–546. [Google Scholar] [CrossRef]
  48. Gu, Y.; Harley, I.T.W.; Henderson, L.B.; Aronow, B.J.; Vietor, I.; Huber, L.A.; Harley, J.B.; Kilpatrick, J.R.; Langefeld, C.D.; Williams, A.H.; et al. Identification of IFRD1 as a modifier gene for cystic fibrosis lung disease. Nature 2009, 458, 1039–1042. [Google Scholar] [CrossRef] [Green Version]
  49. Green, D.M.; Collaco, J.M.; McDougal, K.E.; Naughton, K.M.; Blackman, S.M.; Cutting, G.R. Heritability of respiratory infection with Pseudomonas aeruginosa in cystic fibrosis. J. Pediatr. 2012, 161, 290–295.e1. [Google Scholar] [CrossRef] [Green Version]
  50. Emond, M.J.; Louie, T.; Emerson, J.; Chong, J.X.; Mathias, R.A.; Knowles, M.R.; Rieder, M.J.; Tabor, H.K.; Nickerson, D.A.; Barnes, K.C.; et al. Exome Sequencing of Phenotypic Extremes Identifies CAV2 and TMC6 as Interacting Modifiers of Chronic Pseudomonas aeruginosa Infection in Cystic Fibrosis. PLoS Genet. 2015, 11, e1005273. [Google Scholar] [CrossRef] [Green Version]
  51. Emond, M.J.; Louie, T.; Emerson, J.; Zhao, W.; Mathias, R.A.; Knowles, M.R.; Wright, F.A.; Rieder, M.J.; Tabor, H.K.; Nickerson, D.A.; et al. Exome sequencing of extreme phenotypes identifies DCTN4 as a modifier of chronic Pseudomonas aeruginosa infection in cystic fibrosis. Nat. Genet. 2012, 44, 886–889. [Google Scholar] [CrossRef]
  52. Li, W.; Soave, D.; Miller, M.R.; Keenan, K.; Lin, F.; Gong, J.; Chiang, T.; Stephenson, A.L.; Durie, P.; Rommens, J.; et al. Unraveling the complex genetic model for cystic fibrosis: Pleiotropic effects of modifier genes on early cystic fibrosis-related morbidities. Hum. Genet. 2014, 133, 151–161. [Google Scholar] [CrossRef]
  53. Castaldo, A.; Cernera, G.; Iacotucci, P.; Cimbalo, C.; Gelzo, M.; Comegna, M.; Di Lullo, A.M.; Tosco, A.; Carnovale, V.; Raia, V.; et al. TAS2R38 is a novel modifier gene in patients with cystic fibrosis. Sci. Rep. 2020, 10, 5806. [Google Scholar] [CrossRef] [Green Version]
  54. Tesse, R.; Cardinale, F.; Santostasi, T.; Polizzi, A.; Mappa, L.; Manca, A.; De Robertis, F.; Silecchia, O.; Armenio, L. Association of interleukin-10 gene haplotypes with Pseudomonas aeruginosa airway colonization in cystic fibrosis. J. Cyst. Fibros. 2008, 7, 329–332. [Google Scholar] [CrossRef] [Green Version]
  55. Coutinho, C.A.; Al Marson, F.; Marcelino, A.R.; Bonadia, L.C.; Carlin, M.P.; Ribeiro, A.F.; Ribeiro, J.D.; Bertuzzo, C.S. TNF-alpha polymorphisms as a potential modifier gene in the cystic fibrosis. Int. J. Mol. Epidemiol. Genet. 2014, 5, 87–99. [Google Scholar]
  56. Trevisiol, C.; Boniotto, M.; Giglio, L.; Poli, F.; Morgutti, M.; Crovella, S. MBL2 polymorphisms screening in a regional Italian CF Center. J. Cyst. Fibros. 2005, 4, 189–191. [Google Scholar] [CrossRef] [Green Version]
  57. McDougal, K.E.; Green, D.M.; Vanscoy, L.L.; Fallin, M.D.; Grow, M.; Cheng, S.; Blackman, S.M.; Collaco, J.M.; Henderson, L.B.; Naughton, K.; et al. Use of a modeling framework to evaluate the effect of a modifier gene (MBL2) on variation in cystic fibrosis. Eur. J. Hum. Genet. 2010, 18, 680–684. [Google Scholar] [CrossRef] [Green Version]
  58. Blackman, S.M.; Hsu, S.; Ritter, S.E.; Naughton, K.M.; Wright, F.A.; Drumm, M.L.; Knowles, M.R.; Cutting, G.R. A susceptibility gene for type 2 diabetes confers substantial risk for diabetes complicating cystic fibrosis. Diabetologia 2009, 52, 1858–1865. [Google Scholar] [CrossRef] [Green Version]
  59. Blackman, S.M.; Commander, C.W.; Watson, C.; Arcara, K.M.; Strug, L.J.; Stonebraker, J.R.; Wright, F.A.; Rommens, J.M.; Sun, L.; Pace, R.G.; et al. Genetic modifiers of cystic fibrosis-related diabetes. Diabetes 2013, 62, 3627–3635. [Google Scholar] [CrossRef] [Green Version]
  60. Lam, A.N.; Aksit, M.A.; Vecchio-Pagan, B.; Shelton, C.A.; Osorio, D.L.; Anzmann, A.F.; Goff, L.A.; Whitcomb, D.C.; Blackman, S.M.; Cutting, G.R. Increased expression of anion transporter SLC26A9 delays diabetes onset in cystic fibrosis. J. Clin. Investig. 2020, 130, 272–286. [Google Scholar] [CrossRef] [Green Version]
  61. Stonebraker, J.R.; Ooi, C.Y.; Pace, R.G.; Corvol, H.; Knowles, M.R.; Durie, P.R.; Ling, S.C. Features of severe liver disease with portal hypertension in patients with cystic fibrosis. Clin. Gastroenterol. Hepatol. 2016, 14, 1207–1215.e3. [Google Scholar] [CrossRef] [Green Version]
  62. Debray, D.; Kelly, D.; Houwen, R.; Strandvik, B.; Colombo, C. Best practice guidance for the diagnosis and management of cystic fibrosis-associated liver disease. J. Cyst. Fibros. 2011, 10 (Suppl. 2), S29–S36. [Google Scholar] [CrossRef]
  63. Boelle, P.Y.; Debray, D.; Guillot, L.; Clement, A.; Corvol, H.; on behalf of the French CF Modifier Gene Study Investigators. Cystic Fibrosis Liver Disease: Outcomes and Risk Factors in a Large Cohort of French Patients. Hepatology 2019, 69, 1648–1656. [Google Scholar] [CrossRef] [Green Version]
  64. Frangolias, D.D.; Ruan, J.; Wilcox, P.J.; Davidson, A.G.; Wong, L.T.; Berthiaume, Y.; Hennessey, R.; Freitag, A.; Pedder, L.; Corey, M.; et al. Alpha 1-antitrypsin deficiency alleles in cystic fibrosis lung disease. Am. J. Respir. Cell Mol. Biol. 2003, 29, 390–396. [Google Scholar] [CrossRef] [PubMed]
  65. Arkwright, P.D.; Pravica, V.; Geraghty, P.J.; Super, M.; Webb, A.K.; Schwarz, M.; Hutchinson, I.V. End-organ dysfunction in cystic fibrosis: Association with angiotensin I converting enzyme and cytokine gene polymorphisms. Am. J. Respir. Crit. Care Med. 2003, 167, 384–389. [Google Scholar] [CrossRef] [PubMed]
  66. Henrion-Caude, A.; Flamant, C.; Roussey, M.; Housset, C.; Flahault, A.; Fryer, A.A.; Chadelat, K.; Strange, R.C.; Clement, A. Liver disease in pediatric patients with cystic fibrosis is associated with glutathione S-transferase P1 polymorphism. Hepatology 2002, 36, 913–917. [Google Scholar] [CrossRef]
  67. Gabolde, M.; Hubert, D.; Guilloud-Bataille, M.; Lenaerts, C.; Feingold, J.; Besmond, C. The mannose binding lectin gene influences the severity of chronic liver disease in cystic fibrosis. J. Med. Genet. 2001, 38, 310–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Tomaiuolo, R.; Degiorgio, D.; Coviello, D.A.; Baccarelli, A.; Elce, A.; Raia, V.; Motta, V.; Seia, M.; Castaldo, G.; Colombo, C. An MBL2 haplotype and ABCB4 variants modulate the risk of liver disease in cystic fibrosis patients: A multicentre study. Dig. Liver Dis. 2009, 41, 817–822. [Google Scholar] [CrossRef]
  69. Bartlett, J.R.; Friedman, K.J.; Ling, S.C.; Pace, R.G.; Bell, S.C.; Bourke, B.; Castaldo, G.; Castellani, C.; Cipolli, M.; Colombo, C.; et al. Genetic modifiers of liver disease in cystic fibrosis. JAMA 2009, 302, 1076–1083. [Google Scholar] [CrossRef]
  70. Boelle, P.Y.; Debray, D.; Guillot, L.; Corvol, H.; on behalf of the French CF Modifier Gene Study Investigators. SERPINA1 Z allele is associated with cystic fibrosis liver disease. Genet. Med. 2019, 21, 2151–2155. [Google Scholar] [CrossRef]
  71. Trouve, P.; Genin, E.; Ferec, C. In silico search for modifier genes associated with pancreatic and liver disease in Cystic Fibrosis. PLoS ONE 2017, 12, e0173822. [Google Scholar] [CrossRef]
  72. HAS. Protocole national de diagnostic et de soins (PNDS) Muocviscidose. In Guide—Affection de Longue Durée; HAS: Paris, France, 2017; Available online: https://www.has-sante.fr/jcms/c_2792719/fr/mucoviscidose (accessed on 17 October 2022).
  73. Hubert, D.; Bui, S.; Marguet, C.; Colomb-Jung, V.; Murris-Espin, M.; Corvol, H.; Munck, A. New therapies for cystic fibrosis targeting the CFTR gene or the CFTR protein. Rev. Mal. Respir. 2016, 33, 658–665. [Google Scholar] [CrossRef] [PubMed]
  74. Van Goor, F.; Hadida, S.; Grootenhuis, P.D.; Burton, B.; Cao, D.; Neuberger, T.; Turnbull, A.; Singh, A.; Joubran, J.; Hazlewood, A.; et al. Rescue of CF airway epithelial cell function in vitro by a CFTR potentiator, VX-770. Proc. Natl. Acad. Sci. USA 2009, 106, 18825–18830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Ramsey, B.W.; Davies, J.; McElvaney, N.G.; Tullis, E.; Bell, S.C.; Drevinek, P.; Griese, M.; McKone, E.F.; Wainwright, C.E.; Konstan, M.W.; et al. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N. Engl. J. Med. 2011, 365, 1663–1672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. McKone, E.F.; Borowitz, D.; Drevinek, P.; Griese, M.; Konstan, M.W.; Wainwright, C.; Ratjen, F.; Sermet-Gaudelus, I.; Plant, B.; Munck, A.; et al. Long-term safety and efficacy of ivacaftor in patients with cystic fibrosis who have the Gly551Asp-CFTR mutation: A phase 3, open-label extension study (PERSIST). Lancet Respir. Med. 2014, 2, 902–910. [Google Scholar] [CrossRef]
  77. Corvol, H.; Mésinèle, J.; Douksieh, I.-H.; Strug, L.J.; Boëlle, P.-Y.; Guillot, L. SLC26A9 Gene Is Associated With Lung Function Response to Ivacaftor in Patients With Cystic Fibrosis. Front. Pharmacol. 2018, 9, 828. [Google Scholar] [CrossRef]
  78. Strug, L.J.; Gonska, T.; He, G.; Keenan, K.; Ip, W.; Boelle, P.Y.; Lin, F.; Panjwani, N.; Gong, J.; Li, W.; et al. Cystic fibrosis gene modifier SLC26A9 modulates airway response to CFTR-directed therapeutics. Hum. Mol. Genet. 2016, 25, 4590–4600. [Google Scholar] [CrossRef] [Green Version]
  79. Eastman, A.C.; Pace, R.G.; Dang, H.; Aksit, M.A.; Vecchio-Pagan, B.; Lam, A.N.; O’Neal, W.K.; Blackman, S.M.; Knowles, M.R.; Cutting, G.R. SLC26A9 SNP rs7512462 is not associated with lung disease severity or lung function response to ivacaftor in cystic fibrosis patients with G551D-CFTR. J. Cyst. Fibros. 2021, 20, 851–856. [Google Scholar] [CrossRef]
  80. Wainwright, C.E.; Elborn, J.S.; Ramsey, B.W.; Marigowda, G.; Huang, X.; Cipolli, M.; Colombo, C.; Davies, J.C.; De Boeck, K.; Flume, P.A.; et al. Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del CFTR. N. Engl. J. Med. 2015, 373, 220–231. [Google Scholar] [CrossRef] [Green Version]
  81. Jones, A.M.; Barry, P.J. Lumacaftor/ivacaftor for patients homozygous for Phe508del-CFTR: Should we curb our enthusiasm? Thorax 2015, 70, 615–616. [Google Scholar] [CrossRef] [Green Version]
  82. Jennings, M.T.; Dezube, R.; Paranjape, S.; West, N.E.; Hong, G.; Braun, A.; Grant, J.; Merlo, C.A.; Lechtzin, N. An Observational Study of Outcomes and Tolerances in Patients with Cystic Fibrosis Initiated on Lumacaftor/Ivacaftor. Ann. Am. Thorac. Soc. 2017, 14, 1662–1666. [Google Scholar] [CrossRef]
  83. Konstan, M.W.; McKone, E.F.; Moss, R.B.; Marigowda, G.; Tian, S.; Waltz, D.; Huang, X.; Lubarsky, B.; Rubin, J.; Millar, S.J.; et al. Assessment of safety and efficacy of long-term treatment with combination lumacaftor and ivacaftor therapy in patients with cystic fibrosis homozygous for the F508del-CFTR mutation (PROGRESS): A phase 3, extension study. Lancet Respir. Med. 2017, 5, 107–118. [Google Scholar] [CrossRef]
  84. Burgel, P.-R.; Munck, A.; Durieu, I.; Chiron, R.; Mely, L.; Prevotat, A.; Murris-Espin, M.; Porzio, M.; Abely, M.; Reix, P.; et al. Real-Life Safety and Effectiveness of Lumacaftor–Ivacaftor in Patients with Cystic Fibrosis. Am. J. Respir. Crit. Care Med. 2020, 201, 188–197. [Google Scholar] [CrossRef] [PubMed]
  85. Taylor-Cousar, J.L.; Jain, M.; Barto, T.L.; Haddad, T.; Atkinson, J.; Tian, S.; Tang, R.; Marigowda, G.; Waltz, D.; Pilewski, J. Lumacaftor/ivacaftor in patients with cystic fibrosis and advanced lung disease homozygous for F508del-CFTR. J. Cyst. Fibros. 2018, 17, 228–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Burgel, P.-R.; Durieu, I.; Chiron, R.; Mely, L.; Prevotat, A.; Murris-Espin, M.; Porzio, M.; Abely, M.; Reix, P.; Marguet, C.; et al. Clinical response to lumacaftor-ivacaftor in patients with cystic fibrosis according to baseline lung function. J. Cyst. Fibros. 2020, 20, 220–227. [Google Scholar] [CrossRef] [PubMed]
  87. Aalbers, B.; Groot, K.D.W.-D.; Arets, H.; Hofland, R.; de Kiviet, A.; Ven, M.V.O.-V.D.; Kruijswijk, M.; Schotman, S.; Michel, S.; van der Ent, C.; et al. Clinical effect of lumacaftor/ivacaftor in F508del homozygous CF patients with FEV1 ≥ 90% predicted at baseline. J. Cyst. Fibros. 2020, 19, 654–658. [Google Scholar] [CrossRef] [PubMed]
  88. Elborn, J.S.; Ramsey, B.W.; Boyle, M.P.; Konstan, M.W.; Huang, X.; Marigowda, G.; Waltz, D.; Wainwright, C.E. Efficacy and safety of lumacaftor/ivacaftor combination therapy in patients with cystic fibrosis homozygous for Phe508del CFTR by pulmonary function subgroup: A pooled analysis. Lancet Respir. Med. 2016, 4, 617–626. [Google Scholar] [CrossRef] [Green Version]
  89. Mésinèle, J.; Ruffin, M.; Guillot, L.; Boëlle, P.-Y.; Corvol, H.; on behalf of the French CF Modifier Gene Study Investigators. Factors Predisposing the Response to Lumacaftor/Ivacaftor in People with Cystic Fibrosis. J. Pers. Med. 2022, 12, 252. [Google Scholar] [CrossRef]
  90. Pereira, S.V.-N.; Ribeiro, J.D.; Bertuzzo, C.S.; Marson, F.A.L. Association of clinical severity of cystic fibrosis with variants in the SLC gene family (SLC6A14, SLC26A9, SLC11A1 and SLC9A3). Gene 2017, 629, 117–126. [Google Scholar] [CrossRef]
  91. Ruffin, M.; Mercier, J.; Calmel, C.; Mésinèle, J.; Bigot, J.; Sutanto, E.N.; Kicic, A.; Corvol, H.; Guillot, L. Update on SLC6A14 in lung and gastrointestinal physiology and physiopathology: Focus on cystic fibrosis. Cell. Mol. Life Sci. 2020, 77, 3311–3323. [Google Scholar] [CrossRef] [Green Version]
  92. Ruffin, M.; Mercier, J.; Calmel, C.; Mésinèle, J.; Corvol, H.; Guillot, L. SLC6A14, un gène modificateur dans la mucoviscidose. Rev. Mal. Respir. 2020, 37, 218–221. [Google Scholar] [CrossRef]
  93. Rowe, S.M.; Daines, C.; Ringshausen, F.C.; Kerem, E.; Wilson, J.; Tullis, E.; Nair, N.; Simard, C.; Han, L.; Ingenito, E.P.; et al. Tezacaftor–Ivacaftor in Residual-Function Heterozygotes with Cystic Fibrosis. N. Engl. J. Med. 2017, 377, 2024–2035. [Google Scholar] [CrossRef] [PubMed]
  94. Taylor-Cousar, J.L.; Munck, A.; McKone, E.F.; Van Der Ent, C.K.; Moeller, A.; Simard, C.; Wang, L.T.; Ingenito, E.P.; McKee, C.; Lu, Y.; et al. Tezacaftor–Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del. N. Engl. J. Med. 2017, 377, 2013–2023. [Google Scholar] [CrossRef] [PubMed]
  95. Middleton, P.G.; Mall, M.A.; Dřevínek, P.; Lands, L.C.; McKone, E.F.; Polineni, D.; Ramsey, B.W.; Taylor-Cousar, J.L.; Tullis, E.; Vermeulen, F.; et al. Elexacaftor–Tezacaftor–Ivacaftor for Cystic Fibrosis with a Single Phe508del Allele. N. Engl. J. Med. 2019, 381, 1809–1819. [Google Scholar] [CrossRef] [PubMed]
  96. Burgel, P.-R.; Durieu, I.; Chiron, R.; Ramel, S.; Danner-Boucher, I.; Prevotat, A.; Grenet, D.; Marguet, C.; Reynaud-Gaubert, M.; Macey, J.; et al. Rapid Improvement after Starting Elexacaftor–Tezacaftor–Ivacaftor in Patients with Cystic Fibrosis and Advanced Pulmonary Disease. Am. J. Respir. Crit. Care Med. 2021, 204, 64–73. [Google Scholar] [CrossRef] [PubMed]
  97. Saint-Criq, V.; Wang, Y.; Delpiano, L.; Lin, J.; Sheppard, D.N.; Gray, M.A. Extracellular phosphate enhances the function of F508del-CFTR rescued by CFTR correctors. J. Cyst. Fibros. 2021, 20, 843–850. [Google Scholar] [CrossRef]
  98. Pinto, M.C.; Quaresma, M.C.; Silva, I.A.L.; Railean, V.; Ramalho, S.S.; Amaral, M.D. Synergy in Cystic Fibrosis Therapies: Targeting SLC26A9. Int. J. Mol. Sci. 2021, 22, 13064. [Google Scholar] [CrossRef]
Figure 1. Genetic modifiers of cystic fibrosis (CF) phenotypic traits. Schematic representation of genes identified by genome wide association studies (GWAS) and whole exome sequencing (WES) as associated with the heterogeneity of CF complications (lung disease severity, Pseudomonas. aeruginosa lung infection, meconium ileus susceptibility and CF-related diabetes). Abbreviations (alphabetic order): angiotensin II receptor type 2 (AGTR2), Apaf-1 interacting protein (APIP), ATPase H+/K+ transporting non-gastric alpha2 subunit (ATP12A), Caveolin 2 (CAV2), class II major histocompatibility complex (HLA Class II), cyclin dependent kinase 5 regulatory associated protein 1-like 1 (CDKAL1), cyclin dependent kinase inhibitor 2A/B (CDKN2A/B), Dynactin subunit 4 (DCTN4), Ets Homologous Factor (EHF), Interferon Related Developmental Regulator 1 (IFRD1), insulin-like growth factor-binding protein 2 (IGFBP2), mucin 4 (MUC4), mucin 20 (MUC20), prothymosin alpha (PTMA), serine protease 1 (PRSS1), solute carrier family 26 member 9 (SLC26A9), solute carrier family 6 member 14 (SLC6A14), solute carrier family 9 member A3 (SLC9A3), transcription factor 7-like 2 (TCF7L2) and Transmembrane channel like 6 (TMC6). Created with BioRender.com.
Figure 1. Genetic modifiers of cystic fibrosis (CF) phenotypic traits. Schematic representation of genes identified by genome wide association studies (GWAS) and whole exome sequencing (WES) as associated with the heterogeneity of CF complications (lung disease severity, Pseudomonas. aeruginosa lung infection, meconium ileus susceptibility and CF-related diabetes). Abbreviations (alphabetic order): angiotensin II receptor type 2 (AGTR2), Apaf-1 interacting protein (APIP), ATPase H+/K+ transporting non-gastric alpha2 subunit (ATP12A), Caveolin 2 (CAV2), class II major histocompatibility complex (HLA Class II), cyclin dependent kinase 5 regulatory associated protein 1-like 1 (CDKAL1), cyclin dependent kinase inhibitor 2A/B (CDKN2A/B), Dynactin subunit 4 (DCTN4), Ets Homologous Factor (EHF), Interferon Related Developmental Regulator 1 (IFRD1), insulin-like growth factor-binding protein 2 (IGFBP2), mucin 4 (MUC4), mucin 20 (MUC20), prothymosin alpha (PTMA), serine protease 1 (PRSS1), solute carrier family 26 member 9 (SLC26A9), solute carrier family 6 member 14 (SLC6A14), solute carrier family 9 member A3 (SLC9A3), transcription factor 7-like 2 (TCF7L2) and Transmembrane channel like 6 (TMC6). Created with BioRender.com.
Ijms 23 14205 g001
Table 1. Modifier genes associated with the inter-individual variability of lung function in people with cystic fibrosis and identified by genome wide association studies (sorted alphabetically).
Table 1. Modifier genes associated with the inter-individual variability of lung function in people with cystic fibrosis and identified by genome wide association studies (sorted alphabetically).
Modifier Gene
or Locus
Physiological RoleRef.
Angiotensin II receptor type 2 (AGTR2)/Solute carrier family 6 member A14 (SLC6A14)
Locus chrXq22-q23
AGTR2 encodes the angiotensin, which is part of the renin-angiotensin system, a major control system for blood pressure and fluid balance.
SLC6A14 imports and concentrates neutral amino acids and the two cationic acids lysine and arginine into the cytoplasm of different cell types.
[37]
ETS homologous factor (EHF)/APAF1 interacting protein (APIP)
Locus chr11p12-p13
EHF transcription factor modulates epithelial tight junctions and wound repair. APIP is a methionine salvage pathway enzyme associated with apoptosis and systemic inflammatory responses.[37]
[47]
Human leukocyte antigen (HLA) Class II
Locus chr6p21.3
HLA plays a central role in immunity by presenting peptides derived from extracellular proteins.[37]
Interferon related developmental regulator 1 (IFRD1)
Locus chr11p12-p13
IFRD1 is as a transcriptional co-activator/repressor regulating cellular growth and differentiation during tissue development and regeneration.[48]
Locus chr 20q13.2The five genes located in this locus are expressed in respiratory epithelial cells. Among these genes, the melanocortin 3 receptor (MC3R) is implicated in weight maintenance and regulation of energy balance. Some of these genes encode proteins involved in cell adhesion, migration and phagocytosis of bacterial pathogens.[47]
Mucin 4 (MUC4)/Mucin 20 (MUC20)
Locus chr3q29
Mucins are highly glycosylated mucus proteins.[37]
Solute carrier family 9 member A3 (SLC9A3)SLC9A3 is a sodium–hydrogen exchanger.[37]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mésinèle, J.; Ruffin, M.; Guillot, L.; Corvol, H. Modifier Factors of Cystic Fibrosis Phenotypes: A Focus on Modifier Genes. Int. J. Mol. Sci. 2022, 23, 14205. https://doi.org/10.3390/ijms232214205

AMA Style

Mésinèle J, Ruffin M, Guillot L, Corvol H. Modifier Factors of Cystic Fibrosis Phenotypes: A Focus on Modifier Genes. International Journal of Molecular Sciences. 2022; 23(22):14205. https://doi.org/10.3390/ijms232214205

Chicago/Turabian Style

Mésinèle, Julie, Manon Ruffin, Loïc Guillot, and Harriet Corvol. 2022. "Modifier Factors of Cystic Fibrosis Phenotypes: A Focus on Modifier Genes" International Journal of Molecular Sciences 23, no. 22: 14205. https://doi.org/10.3390/ijms232214205

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