Next Article in Journal
Understanding Dietary Habit Changes during the COVID-19 Pandemic in Italy
Previous Article in Journal
Use of Nutritional Supplements Based on L-Theanine and Vitamin B6 in Children with Tourette Syndrome, with Anxiety Disorders: A Pilot Study
Previous Article in Special Issue
Breastfeeding in Cystic Fibrosis: A Systematic Review on Prevalence and Potential Benefits
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

Nutrition in Cystic Fibrosis—Some Notes on the Fat Recommendations

by
Birgitta Strandvik
Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14183 Stockholm, Sweden
Submission received: 14 January 2022 / Revised: 13 February 2022 / Accepted: 14 February 2022 / Published: 18 February 2022
(This article belongs to the Special Issue Nutrition in Cystic Fibrosis)

Abstract

:
Nutrition is important in cystic fibrosis (CF) because the disease is associated with a higher energy consumption, special nutritional deficiencies, and malabsorption mainly related to pancreatic insufficiency. The clinical course with deterioration of lung function has been shown to relate to nutrition. Despite general recommendation of high energy intake, the clinical deterioration is difficult to restrain suggesting that special needs have not been identified and specified. It is well-known that the CF phenotype is associated with lipid abnormalities, especially in the essential or conditionally essential fatty acids. This review will concentrate on the qualitative aspects of fat metabolism, which has mainly been neglected in dietary fat recommendations focusing on fat quantity. For more than 60 years it has been known and confirmed that the patients have a deficiency of linoleic acid, an n-6 essential fatty acid of importance for membrane structure and function. The ratio between arachidonic acid and docosahexaenoic acid, conditionally essential fatty acids of the n-6 and n-3 series, respectively, is often increased. The recently discovered relations between the CFTR modulators and lipid metabolism raise new interests in this field and together with new technology provide possibilities to specify further specify personalized therapy.

1. Introduction

Cystic fibrosis (CF), also known as mucoviscidosis, was identified in 1936–1938 as a special disease, cystic fibrosis of the pancreas, distinguished from celiac disease due to its characteristic pancreatic involvement and rapidly progressive pulmonary changes [1,2]. Before efficient pancreatic enzymes and a wide spectrum of antibiotics were available, most patients died in early childhood. The impressive progress of care for these patients during the latest decades [3,4], has now brought the adult to exceed the child patients in developed countries [5,6,7,8]), can be related to better clinical treatment, development of drugs and organ transplantation. The most common cause of death is the pulmonary destruction caused by the disease. The importance of nutrition for the pulmonary function is well documented [9,10,11].
Many papers have been published regarding nutritional recommendations for CF patients, as well as during recent years [12,13,14,15,16,17]. These documents unanimously recommend high energy, 110–200% of recommended daily intake (RDI), often with higher protein and fat contributions. This review will not further repeat these mostly concordant recommendations but discuss some special issues regarding the energy-rich macronutrient, the fat, whose quality has been given less attention in the guidelines as well as in evaluating the clinical results of the recommendations. Our knowledge about the well-known lipid abnormalities in CF will improve with the available new techniques giving possibilities to extensive lipidomics [18,19,20,21,22], expanding the knowledge about the role of lipids in the disease. In this context, the quality of fat intake is important since it is reflected in cell membranes influencing transport and metabolism [23,24,25].

2. Fat Recommendations and Intake

A difference in patient status between Boston and Toronto despite general similarities in treatment was reported by Corey et al. in 1988 [26]. The US, like many European countries, but differently from Toronto, recommended a low-fat diet due to the problems with steatorrhoea, probably at least partly related to less-efficient pancreatic enzyme restitution therapy (PERT). That paper resulted in a general recommendation of high-fat diet to patients with CF, and recent comparisons between the countries showed that the differences in patient status decreased [27] and were recently mainly omitted [28]. These results suggest that higher lipid intake is a stronger factor than only high energy intake.
A higher fat intake usually results in a high intake of saturated fat [29,30,31,32], which might be a factor explaining why many studies report that the high fat recommendations are not followed, sometimes only in half of the patient population [33,34]. It is difficult to obtain the high recommended energy intake without increasing the fat, it being the most energy dense food. In a study by Stallings et al. a structured lipid supplement with 54% polyunsaturated fat showed clinical improvement compared to a calorie- and macronu-trient-matched placebo group after 3 months treatment [35]. Of interest are a few studies showing that normal weight gain can be achieved in CF without increased energy intake if the essential fatty acid deficiency (EFAD) was treated [36,37]. So, although the high energy demand is multifactorial, the quality of the fat should command more interest than just the amount.
Patients with CF have a higher need (see below) for EFA, e.g., the fatty acids of the omega-6 (n-6) and omega-3 (n-3) series, which we cannot synthesize. A high fat intake usually also increases the intake of EFA [38,39]. That connection has generally not been commented on in the general recommendations but is of interest when comparing the intake of different fatty acids intake in different countries (Table 1). These intakes reflect also that of the children with the disease. It is well-known that the intake of n-6 fatty acids is high in the US [40] and that it also differs between European countries, as reflected in maternal plasma during pregnancy [41,42]. In this context it is of interest that the most recent dietary recommendations for CF from the US show no indications for even discussing recommendations of EFA [14]. To the contrary, it is usually emphasized that an increased intake of EFA cannot be recommended due to the shortcoming of good controlled randomized studies, which is true, but the low levels related to the severe CF phenotype cannot just be neglected [43]. Only a few recommendations comment on the possibility that EFA may be of value [17,44,45]. This is a little surprising since lipid abnormalities with deficiency of especially the n-6 fatty acid linoleic acid (LA, 18:2n-6) has been known for more than 60 years in patients with CF [46]. Importantly, any comments regarding EFA in the recommendations only refer to the n-3 fatty acids, although no recommendation is given because no convincing clinical effects are shown in many studies supplementing n-3 fatty acids [12,47,48,49,50].

3. Fatty Acid Abnormalities in CF

The old report about LA deficiency in CF has been confirmed in different blood compartments [51,52,53] and in various tissues [54,55,56,57]. Many studies of supplementation with LA have been performed, but were often of relatively short duration and, lacking in stable study conditions, not resulting in clear outcomes and motivating general recommendations. Some studies have shown an important influence on growth [37,58,59] and some observation studies have shown associations between lung function and serum phospholipid concentrations of LA [44,60,61,62,63]. In a longitudinal prospective observation study of neonatally screened patients, the Wisconsin group has shown a strong relation between the LA concentration and growth and pulmonary function up to 12 years of age [64,65,66]. Some other longitudinal studies have shown an influence on growth, renal and liver function [55,58,67,68]. The progress of CF-related liver disease has shown an association to low LA [69,70], and Sweden, where the largest centres had provided LA supplementation for decades [4], had the lowest percentage of portal hypertension and cirrhosis compared to 10 other countries worldwide [71].
LA is an important constituent of cell membranes and together with the ratio of n-6/n-3 fatty acids and cholesterol influences the function of transmembrane proteins, including ion channels and enzymes [23,72]. In the body, LA is transformed to arachidonic acid (20:4n-6, ARA) (Figure 1), which is the substrate for the pro-inflammatory eicosanoids, increased in CF [73,74], and important in the defense of infections but also involved in many metabolic processes. The synthesis of ARA is the rate-limiting step in eicosanoid production [75]. There seems to be a feedback mechanism since the relation between ARA and LA is inverted [20], signaling possible importance in the context of prostaglandin production. This inverse relation was observed in cell systems and related to an inhibition of FADS2 by LA [76]. That is supported by studies of rats showing that EFA deficiency increases FADS 2 activity [77]. The enzyme activities involved in the transformation of LA are increased in CF, suggesting it to be more basically related to CFTR [78] than being an effect of the increased AA release [79]. Since the AA release in CF blood cells could not be inhibited by dexamethasone [80] and not by bradykinin in cell systems [81,82], it is an open question if the high turnover is an effect of, or a cause for, the highly increased AA release [83,84,85] on the one hand, or on the other if the LA deficiency itself is increasing the turnover [77]. Algorithms have been presented to suggest relations between the disturbed lipid metabolism and the clinical symptoms, which later clinical investigations have partly supported [86,87].
The most important EFA of the n-3 fatty acid series is docosahexaenoic acid (22:6n-3, DHA). DHA is often decreased in CF, and low concentrations are related to the liver disease [88,89]. It is of interest that in CF mice receiving long-term treatment with high doses of DHA, liver disease could be attenuated [90]. The improvement of ileal mucosa and pancreas pathology in CF mice by high doses of DHA has not been confirmed [91] but has increased the interest for studies of supplying DHA to patients but without resulting in clear clinical improvements [92]. This long-chain n-3 PUFA also contributes to membrane function by interfering with raft formation important for transport function in membranes [93]. DHA, like other long-chain n-3 fatty acids, are alo transferred to special pro-solving lipid mediators (SPM), e.g., the resolvins, maresins and protectins, of importance to balance inflammation provided by the ARA metabolites [94]. The role of SPM in regulating autophagy, which is defective in CF, may be a possible therapeutic option against 136 the persistent inflammation [95,96,97]. That was supported by adding resolvin D1 to infected CF KO mice and in studies of cells from CF patients [98]. Since long-term administration of DHA did not improve clinical status [99], it has been suggested that impaired production of SPM in CF is associated with abnormal expression of lipoxygenases [95]. Furthermore, lipoxin A4, an anti-inflammatory SPM from ARA, is deficient in CF [100]. Altogether, the lipid metabolism of polyunsaturated fatty acids is unbalanced in CF [20,51].
Reasons for the low interest in these well-known fatty acid abnormalities in CF might be multiplex: (1) the unclear connection to the genetic defect, the defective chloride and bicarbonate channel CFTR (Cystic fibrosis transmembrane conductance regulator) [101] compounded by the fact the lipid abnormalities are related to a more severe phenotype [43,102]. Today, more than 2000 mutations are identified, but only a minority are related to the CF phenotype, although classified in six types, some with very minor clinical relevance [103,104]. New technologies show different lipid patterns related to type of mutations [105], and correlations between different phospholipid abnormalities and mild and severe clinical disease [106]. Since severe disease is related to mutations with more bacterial infections some of these changes can be due to bacterial interactions, which needs further studies for differentiation. (2) The levels or concentrations in the serum/plasma of LA are often low but seldom at a real deficient status, and tissue levels/concentrations are usually only obtained at autopsy [55,57], but can be shown also in nasal and intestinal biopsies [54]. Studies with contemporary examinations of plasma and tissues show that tissues can have varying and much lower concentrations than in plasma [55,107], suggesting that relatively low blood concentrations might also be of clinical relevance. Lower concentrations and values in different tissues would also explain why the EFA index (the ratio between Mead acid (20:3n-9) to ARA, i.e., the triene/tetraene (T:T) ratio) may remain pathological, when plasma levels are normalized. This is reported from Belgium [108,109], which has one of the highest fat intakes in Europe (Table 1). (3) The relation to other fatty acid abnormalities is partly unclear, e.g., the high ratio between ARA and DHA, although not always related to low DHA or high ARA, is not fully explained. Nor is the probably compensatory increase in the monounsaturated fatty acids palmitoleic acid (16:1n-7), oleic acid (18:1n-9) and Mead acid [53,78,110,111]. (4) The relation to other lipid abnormalities such as low cholesterol and different pattern of the lipoproteins [112,113,114] are incompletely understood. (5) The difficulty to identify clinically the deficiency in relation to symptoms, such as growth failure, pancreatic insufficiency increased resting respiratory expenditure, etc. and not least. (6) the omission to check the concentration in blood, which recently was shown only to be performed only in 15% of centers caring for CF patients according to the DIGEST (Developing Innovative Gastro Enterology Special Training) program [115].
The increased activity of the epithelial sodium channel (ENaC), which is considered to contribute to the pulmonary symptoms, have been suggested to be influenced by LA [116]. ENaC are expressed in the kidney, and the renal excretion of sodium was restored after long-term LA supplementation [67]. Sodium excretion in sweat was also decreased after long-term LA supplementation [59,74,117]. Other transporters and membrane-bound activities might also be influenced, since the Na(+)–K+ ratio in erythrocytes was normalized by regular LA supplementation [118].

4. Is Dietary Qualitative Intervention Justified?

Altogether, few dietary recommendations in CF mention the well-known deficiencies of EFA and the different needs they satisfy, e.g., the n-6 for the possible membrane physiology and its products in balancing inflammation and infections and the n-3 also involved in membrane physiology and for anti-inflammatory balancing function via SPM [92]. At least three studies use n-6-rich oil as control when supplying n-3 fatty acids and see no differences between the groups [119,120,121]. In a recent systematic review of supplementations in CF, LA supplementation was summarized to influence nutritional status and n-3 supplementation to reduce inflammatory markers [122], supporting the theoretical concept that both n-6 and n-3 fatty acids might be valuable in CF nutrition.
Despite extensive research during more than 30 years since CFTR was detected, no cure of the disease has been developed. In the search for therapy, the high-throughput technique had identified correctors in the synthesis of some mutations and especially one potentiator, ivacaftor, for the action of some mutated proteins making it possible to improve the status of several patients [123]. Combinations of the potentiator and correctors have further increased the potential target of mutations and shown to influence the lung function, sweat test and weight gain but not less the chronic inflammation [124,125]. Of certain interest is that the modulators are recommended to be ingested together with lipids [126]. The real influence of CFTR mutations on clinical pathophysiology is still inconclusive, and many other types of modulators are under investigation [127], which illustrates the persistent challenges of clinical treatment. It is also important to realize that a general clinical improvement in lung function with modern treatment was obvious before the recent development of modulators [128].
To ignore the deficiency of fatty acids, as being of well-documented importance for cell physiology, is hopefully coming to an end since recent research has shown a strong difference in the lipid pattern related to the type of mutation [105]. Furthermore, it is now shown that the modulators interfere with the membrane lipid structure, concerning the glycerophospholipids, the sphingomyelins, and also the ceramides [18,19,21]. All repeatedly shown involvement in the CF context [106,129,130]. An influence on the prostanoids has also been suggested [131]. These findings open a link between the CFTR and the lipid abnormality, which has been lacking and probably contributed to the neglect of treating the EFAD in CF. A further reason for considering the EFA abnormalities is that there might be rather extensive deficiency in different organs since serum concentrations of the fatty acids are less reflecting the total body metabolism, and it is therefore a need for longer treatment periods to influence organ symptoms [67,68].
An ongoing multicenter European study blindly supplying either LA or oleic acid for one year might hopefully contribute to an answer on the putative beneficial effect of LA supplementation in CF (NCT 04531410) and, by using metabolomics, further add to the knowledge of the pathophysiology [132]. More studies are warranted.

5. Conclusions

The general guidelines worldwide recommend high energy intake with special attention to protein and fat, but the fat recommendations are seldom achieved. The type of fat should be given more attention in relation to the known deficiencies and the conditions of the patients. More normal energy recommendations can probably be obtained if the quality of the fat supply is changed with greater focus on linoleic-rich oils, such as sunflower oil and corn oil, as opposed to the general recommendation of canola oil and olive oil for the normal population. Fish or omega-3 supplementation should also be encouraged potentially to hamper the inflammation. Such recommendations would be valuable non-toxic additives to the modulator therapy in waiting for long-term controlled studies.

Funding

The Swedish Heart and Lung Association.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Andersen, D.H. Cystic fibrosis of the pancreas and its relation to celiac disease. A clinical and pathological study. Am. J. Dis. Child 1938, 56, 344–399. [Google Scholar] [CrossRef]
  2. Fanconi, G.; Uehlinger, E.; Knauer, C. Das Coelioksyndrome bei Angeborener zystischer Pankreas fibromatose und Bronkiektasien. Wien. Med. Wochenschr. 1936, 86, 753–756. [Google Scholar]
  3. Shteinberg, M.; Haq, I.J.; Polineni, D.; Davies, J.C. Cystic fibrosis. Lancet 2021, 397, 2195–2211. [Google Scholar] [CrossRef]
  4. Strandvik, B. Care of patients with cystic fibrosis. Treatment, screening and clinical outcome. Ann. Nestlé 2006, 64, 131–140. [Google Scholar]
  5. Earnest, A.; Salimi, F.; Wainwright, C.E.; Bell, S.C.; Ruseckaite, R.; Ranger, T.; Kotsimbos, T.; Ahern, S. Lung function over the life course of paediatric and adult patients with cystic fibrosis from a large multi-centre registry. Sci. Rep. 2020, 10, 17421. [Google Scholar] [CrossRef]
  6. North American Cystic Fibrosis Foundation. 2019 Patient Registry Annual Data Report; Cystic Fibrosis Foundation: Bethesda, MD, USA, 2021. [Google Scholar]
  7. Cystic Fibrosis Canada. 2018 Canadian CF Registry Annual Data Report; Cystic Fibrosis Canada: Toronto, ON, Canada, 2021. [Google Scholar]
  8. Orenti, A.; Zolin, A.; Jung, A.; van Rens, J. ECFS Patient Registry Annual Report 2019; European Cystic Fibrosis Society: Karup, Denmark, 2021. [Google Scholar]
  9. Steinkamp, G.; Wiedemann, B. Relationship between nutritional status and lung function in cystic fibrosis: Cross sectional and longitudinal analyses from the German CF quality assurance (CFQA) project. Thorax 2002, 57, 596–601. [Google Scholar] [CrossRef] [Green Version]
  10. Altman, K.; McDonald, C.M.; Michel, S.H.; Maguiness, K. Nutrition in cystic fibrosis: From the past to the present and into the future. Pediatric Pulmonol. 2019, 54 (Suppl. S3), S56–S73. [Google Scholar] [CrossRef] [Green Version]
  11. Ashkenazi, M.; Nathan, N.; Sarouk, I.; Aluma, B.E.B.; Dagan, A.; Bezalel, Y.; Keler, S.; Vilozni, D.; Efrati, O. Nutritional Status in Childhood as a Prognostic Factor in Patients with Cystic Fibrosis. Lung 2019, 197, 371–376. [Google Scholar] [CrossRef]
  12. Van der Haak, N.; King, S.J.; Crowder, T.; Kench, A.; Painter, C.; Saxby, N. Highlights from the nutrition guidelines for cystic fibrosis in Australia and New Zealand. J. Cyst. Fibros. 2020, 19, 16–25. [Google Scholar] [CrossRef]
  13. Wilschanski, M.; Braegger, C.P.; Colombo, C.; Declercq, D.; Morton, A.; Pancheva, R.; Robberecht, E.; Stern, M.; Strandvik, B.; Wolfe, S.; et al. Highlights of the ESPEN-ESPGHAN-ECFS Guidelines on Nutrition Care for Infants and Children with Cystic Fibrosis. J. Pediatric Gastroenterol. Nutr. 2016, 63, 671–675. [Google Scholar] [CrossRef] [Green Version]
  14. McDonald, C.M.; Alvarez, J.A.; Bailey, J.; Bowser, E.K.; Farnham, K.; Mangus, M.; Padula, L.; Porco, K.; Rozga, M. Academy of Nutrition and Dietetics: 2020 Cystic Fibrosis Evidence Analysis Center Evidence-Based Nutrition Practice Guideline. J. Acad. Nutr. Diet. 2021, 121, 1591–1636.e3. [Google Scholar] [CrossRef] [PubMed]
  15. Lahiri, T.; Hempstead, S.E.; Brady, C.; Cannon, C.L.; Clark, K.; Condren, M.E.; Guill, M.F.; Guillerman, R.P.; Leone, C.G.; Maguiness, K.; et al. Clinical Practice Guidelines from the Cystic Fibrosis Foundation for Preschoolers with Cystic Fibrosis. Pediatrics 2016, 137, e20151784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Saxby, N.; Painter, C.; Kench, A.; King, S.; Crowder, T.; van der Haak, N. Nutrition Guidelines for Cystic Fibrosis in Australia and New Zealand; Thoracic Society of Australia and New Zealand: Sydney, Australia, 2017. [Google Scholar]
  17. Turck, D.; Braegger, C.P.; Colombo, C.; Declercq, D.; Morton, A.; Pancheva, R.; Robberecht, E.; Stern, M.; Strandvik, B.; Wolfe, S.; et al. ESPEN-ESPGHAN-ECFS guidelines on nutrition care for infants, children, and adults with cystic fibrosis. Clin. Nutr. 2016, 35, 557–577. [Google Scholar] [CrossRef] [Green Version]
  18. Kopp, B.T.; Joseloff, E.; Goetz, D.; Ingram, B.; Heltshe, S.L.; Leung, D.H.; Ramsey, B.W.; McCoy, K.; Borowitz, D. Urinary metabolomics reveals unique metabolic signatures in infants with cystic fibrosis. J. Cyst. Fibros. 2019, 18, 507–515. [Google Scholar] [CrossRef]
  19. Liessi, N.; Pedemonte, N.; Armirotti, A.; Braccia, C. Proteomics and Metabolomics for Cystic Fibrosis Research. Int. J. Mol. Sci. 2020, 21, 5439. [Google Scholar] [CrossRef]
  20. Wheelock, C.E.; Strandvik, B. Abnormal n-6 fatty acid metabolism in cystic fibrosis contributes to pulmonary symptoms. Prostaglandins Leukot. Essent. Fat. Acids 2020, 160, 102156. [Google Scholar] [CrossRef]
  21. Dei Cas, M.; Zulueta, A.; Mingione, A.; Caretti, A.; Ghidoni, R.; Signorelli, P.; Paroni, R. An Innovative Lipidomic Workflow to Investigate the Lipid Profile in a Cystic Fibrosis Cell Line. Cells 2020, 9, 1197. [Google Scholar] [CrossRef]
  22. Pacetti, D.; Malavolta, M.; Bocci, F.; Boselli, E.; Frega, N.G. High-performance liquid chromatography/electrospray ionization ion-trap tandem mass spectrometric analysis and quantification of phosphatidylcholine molecular species in the serum of cystic fibrosis subjects supplemented with docosahexaenoic acid. Rapid Commun. Mass Spectrom. 2004, 18, 2395–2400. [Google Scholar] [CrossRef]
  23. Hulbert, A.J.; Turner, N.; Storlien, L.H.; Else, P.L. Dietary fats and membrane function: Implications for metabolism and disease. Biol. Rev. Camb. Philos. Soc. 2005, 80, 155–169. [Google Scholar] [CrossRef]
  24. Ulven, S.M.; Holven, K.B. Metabolomic and gene expression analysis to study the effects of dietary saturated and polyunsaturated fats. Curr. Opin. Lipidol. 2020, 31, 15–19. [Google Scholar] [CrossRef]
  25. Gimenez, M.S.; Oliveros, L.B.; Gomez, N.N. Nutritional deficiencies and phospholipid metabolism. Int. J. Mol. Sci. 2011, 12, 2408–2433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Corey, M.; McLaughlin, F.J.; Williams, M.; Levison, H. A comparison of survival, growth, and pulmonary function in patients with cystic fibrosis in Boston and Toronto. J. Clin. Epidemiol. 1988, 41, 583–591. [Google Scholar] [CrossRef]
  27. Lai, H.C.; Corey, M.; FitzSimmons, S.; Kosorok, M.R.; Farrell, P.M. Comparison of growth status of patients with cystic fibrosis between the United States and Canada. Am. J. Clin. Nutr. 1999, 69, 531–538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Goss, C.H.; Sykes, J.; Stanojevic, S.; Marshall, B.; Petren, K.; Ostrenga, J.; Fink, A.; Elbert, A.; Quon, B.S.; Stephenson, A.L. Comparison of Nutrition and Lung Function Outcomes in Patients with Cystic Fibrosis Living in Canada and the United States. Am. J. Respir. Crit. Care Med. 2018, 197, 768–775. [Google Scholar] [CrossRef] [PubMed]
  29. Poulimeneas, D.; Grammatikopoulou, M.G.; Devetzi, P.; Petrocheilou, A.; Kaditis, A.G.; Papamitsou, T.; Doudounakis, S.E.; Vassilakou, T. Adherence to Dietary Recommendations, Nutrient Intake Adequacy and Diet Quality among Pediatric Cystic Fibrosis Patients: Results from the GreeCF Study. Nutrients 2020, 12, 3126. [Google Scholar] [CrossRef]
  30. Moukarzel, S.; Dyer, R.A.; Innis, S.M. Complex Relation Between Diet and Phospholipid Fatty Acids in Children with Cystic Fibrosis. J. Pediatric Gastroenterol. Nutr. 2017, 64, 598–604. [Google Scholar] [CrossRef]
  31. Smith, C.; Winn, A.; Seddon, P.; Ranganathan, S. A fat lot of good: Balance and trends in fat intake in children with cystic fibrosis. J. Cyst. Fibros. 2012, 11, 154–157. [Google Scholar] [CrossRef] [Green Version]
  32. Woestenenk, J.W.; Castelijns, S.J.; van der Ent, C.K.; Houwen, R.H. Dietary intake in children and adolescents with cystic fibrosis. Clin. Nutr. 2014, 33, 528–532. [Google Scholar] [CrossRef]
  33. Filigno, S.S.; Robson, S.M.; Szczesniak, R.D.; Chamberlin, L.A.; Baker, M.A.; Sullivan, S.M.; Kroner, J.; Powers, S.W. Macronutrient intake in preschoolers with cystic fibrosis and the relationship between macronutrients and growth. J. Cyst. Fibros. 2017, 16, 519–524. [Google Scholar] [CrossRef] [Green Version]
  34. Calvo-Lerma, J.; Hulst, J.M.; Asseiceira, I.; Claes, I.; Garriga, M.; Colombo, C.; Fornés, V.; Woodcock, S.; Martins, T.; Boon, M.; et al. Nutritional status, nutrient intake and use of enzyme supplements in paediatric patients with Cystic Fibrosis; a European multicentre study with reference to current guidelines. J. Cyst. Fibros. 2017, 16, 510–518. [Google Scholar] [CrossRef] [Green Version]
  35. Stallings, V.A.; Tindall, A.M.; Mascarenhas, M.R.; Maqbool, A.; Schall, J.I. Improved residual fat malabsorption and growth in children with cystic fibrosis treated with a novel oral structured lipid supplement: A randomized controlled trial. PLoS ONE 2020, 15, e0232685. [Google Scholar] [CrossRef]
  36. Kindstedt-Arfwidson, K.; Strandvik, B. Food intake in patients with cystic fibrosis on an ordinary diet. Scand. J. Gastroenterol. Suppl. 1988, 143, 160–162. [Google Scholar] [CrossRef]
  37. Van Egmond, A.W.; Kosorok, M.R.; Koscik, R.; Laxova, A.; Farrell, P.M. Effect of linoleic acid intake on growth of infants with cystic fibrosis. Am. J. Clin. Nutr. 1996, 63, 746–752. [Google Scholar] [CrossRef] [Green Version]
  38. Walkowiak, J.; Przyslawski, J. Five-year prospective analysis of dietary intake and clinical status in malnourished cystic fibrosis patients. J. Hum. Nutr. Diet 2003, 16, 225–231. [Google Scholar] [CrossRef]
  39. Strandvik, B.; Lundquist-Persson, C.; Sabel, K.-G. Early behavior and development are influenced by the n-6 and n-3 status in prematures. Oleagineux Corps Gras Lipides 2011, 18, 297–300. [Google Scholar] [CrossRef] [Green Version]
  40. Blasbalg, T.L.; Hibbeln, J.R.; Ramsden, C.E.; Majchrzak, S.F.; Rawlings, R.R. Changes in consumption of omega-3 and omega-6 fatty acids in the United States during the 20th century. Am. J. Clin. Nutr. 2011, 93, 950–962. [Google Scholar] [CrossRef] [Green Version]
  41. Heird, W.C. The role of polyunsaturated fatty acids in term and preterm infants and breastfeeding mothers. Pediatric Clin. N. Am. 2001, 48, 173–188. [Google Scholar] [CrossRef]
  42. Otto, S.J.; Houwelingen, A.C.; Antal, M.; Manninen, A.; Godfrey, K.; López-Jaramillo, P.; Hornstra, G. Maternal and neonatal essential fatty acid status in phospholipids: An international comparative study. Eur. J. Clin. Nutr. 1997, 51, 232–242. [Google Scholar] [CrossRef] [Green Version]
  43. Strandvik, B.; Gronowitz, E.; Enlund, F.; Martinsson, T.; Wahlström, J. Essential fatty acid deficiency in relation to genotype in patients with cystic fibrosis. J. Pediatric 2001, 139, 650–655. [Google Scholar] [CrossRef]
  44. Walkowiak, J.; Lisowska, A.; Blaszczynski, M.; Przyslawski, J.; Walczak, M. Polyunsaturated fatty acids in cystic fibrosis are related to nutrition and clinical expression of the disease. J. Pediatric Gastroenterol. Nutr. 2007, 45, 488–489. [Google Scholar] [CrossRef]
  45. Sinaasappel, M.; Stern, M.; Littlewood, J.; Wolfe, S.; Steinkamp, G.; Heijerman, H.G.; Robberecht, E.; Döring, G. Nutrition in patients with cystic fibrosis: A European Consensus. J. Cyst. Fibros. 2002, 1, 51–75. [Google Scholar] [CrossRef] [Green Version]
  46. Kuo, P.T.; Huang, N.N.; Bassett, D.R. The fatty acid composition of the serum chylomicrons and adipose tissue of children with cystic fibrosis of the pancreas. J. Pediatric 1962, 60, 394–403. [Google Scholar] [CrossRef]
  47. Colombo, C.; Nobili, R.M.; Alicandro, G. Challenges with optimizing nutrition in cystic fibrosis. Expert Rev. Respir. Med. 2019, 13, 533–544. [Google Scholar] [CrossRef]
  48. Hankard, R.; Munck, A.; Navarro, J. Nutrition and growth in cystic fibrosis. Horm. Res. 2002, 58 (Suppl. S1), 16–20. [Google Scholar] [CrossRef]
  49. Munck, A. Nutritional considerations in patients with cystic fibrosis. Expert Rev. Respir. Med. 2010, 4, 47–56. [Google Scholar] [CrossRef]
  50. Borowitz, D.; Baker, R.D.; Stallings, V. Consensus report on nutrition for pediatric patients with cystic fibrosis. J. Pediatric Gastroenterol. Nutr. 2002, 35, 246–259. [Google Scholar] [CrossRef] [Green Version]
  51. Strandvik, B. Fatty acid metabolism in cystic fibrosis. Prostaglandins Leukot. Essent. Fat. Acids 2010, 83, 121–129. [Google Scholar] [CrossRef]
  52. Gibson, R.A.; Teubner, J.K.; Haines, K.; Cooper, D.M.; Davidson, G.P. Relationships between pulmonary function and plasma fatty acid levels in cystic fibrosis patients. J. Pediatric Gastroenterol. Nutr. 1986, 5, 408–415. [Google Scholar] [CrossRef]
  53. Rogiers, V.; Vercruysse, A.; Dab, I.; Crokaert, R.; Vis, H.L. Fatty acid pattern of platelet phospholipids in cystic fibrosis. Eur. J. Pediatric 1984, 142, 305–306. [Google Scholar] [CrossRef]
  54. Freedman, S.D.; Blanco, P.G.; Zaman, M.M.; Shea, J.C.; Ollero, M.; Hopper, I.K.; Weed, D.A.; Gelrud, A.; Regan, M.M.; Laposata, M.; et al. Association of cystic fibrosis with abnormalities in fatty acid metabolism. N. Engl. J. Med. 2004, 350, 560–569. [Google Scholar] [CrossRef]
  55. Mischler, E.H.; Parrell, S.W.; Farrell, P.M.; Raynor, W.J.; Lemen, R.J. Correction of linoleic acid deficiency in cystic fibrosis. Pediatric Res. 1986, 20, 36–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Moonen, C.T.; Dimand, R.J.; Cox, K.L. The noninvasive determination of linoleic acid content of human adipose tissue by natural abundance carbon-13 nuclear magnetic resonance. Magn. Reason. Med. 1988, 6, 140–157. [Google Scholar] [CrossRef] [PubMed]
  57. Underwood, B.A.; Denning, C.R.; Navab, M. Polyunsaturated fatty acids and tocopherol levels in patients with cystic fibrosis. Ann. N. Y. Acad. Sci. 1972, 203, 237–247. [Google Scholar] [CrossRef] [PubMed]
  58. Chase, H.P.; Cotton, E.K.; Elliott, R.B. Intravenous linoleic acid supplementation in children with cystic fibrosis. Pediatrics 1979, 64, 207–213. [Google Scholar] [CrossRef]
  59. Elliott, R.B. A therapeutic trial of fatty acid supplementation in cystic fibrosis. Pediatrics 1976, 57, 474–479. [Google Scholar] [CrossRef]
  60. Hjelte, L.; Nilsson, K.; Moen, I.E.; Lindblad, A.; Mared, L.; Pressler, T.; Fluge, G. Linoleic acid but not EPA and DHA correlates to prognostic markers in Scandinavian CF patients. J. Cyst. Fibros. 2008, 7, S93. [Google Scholar]
  61. Lloyd-Still, J.D.; Bibus, D.M.; Powers, C.A.; Johnson, S.B.; Holman, R.T. Essential fatty acid deficiency and predisposition to lung disease in cystic fibrosis. Acta Paediatr. 1996, 85, 1426–1432. [Google Scholar] [CrossRef]
  62. Maqbool, A.; Schall, J.I.; Garcia-Espana, J.F.; Zemel, B.S.; Strandvik, B.; Stallings, V.A. Serum linoleic acid status as a clinical indicator of essential fatty acid status in children with cystic fibrosis. J. Pediatric Gastroenterol. Nutr. 2008, 47, 635–644. [Google Scholar] [CrossRef] [Green Version]
  63. Risé, P.; Volpi, S.; Colombo, C.; Padoan, R.F.; D’Orazio, C.; Ghezzi, S.; Melotti, P.; Bennato, V.; Agostoni, C.; Assael, B.M.; et al. Whole blood fatty acid analysis with micromethod in cystic fibrosis and pulmonary disease. J. Cyst. Fibros. 2010, 9, 228–233. [Google Scholar] [CrossRef] [Green Version]
  64. Lai, H.C.; Kosorok, M.R.; Laxova, A.; Davis, L.A.; FitzSimmon, S.C.; Farrell, P.M. Nutritional status of patients with cystic fibrosis with meconium ileus: A comparison with patients without meconium ileus and diagnosed early through neonatal screening. Pediatrics 2000, 105, 53–61. [Google Scholar] [CrossRef]
  65. Shoff, S.M.; Ahn, H.Y.; Davis, L.; Lai, H. Temporal associations among energy intake, plasma linoleic acid, and growth improvement in response to treatment initiation after diagnosis of cystic fibrosis. Pediatrics 2006, 117, 391–400. [Google Scholar] [CrossRef]
  66. Sanders, D.B.; Zhang, Z.; Farrell, P.M.; Lai, H.J. Early life growth patterns persist for 12 years and impact pulmonary outcomes in cystic fibrosis. J. Cyst. Fibros. 2018, 17, 528–535. [Google Scholar] [CrossRef]
  67. Strandvik, B.; Berg, U.; Kallner, A.; Kusoffsky, E. Effect on renal function of essential fatty acid supplementation in cystic fibrosis. J. Pediatric 1989, 115, 242–250. [Google Scholar] [CrossRef]
  68. Strandvik, B.; Hultcrantz, R. Liver function and morphology during long-term fatty acid supplementation in cystic fibrosis. Liver 1994, 14, 32–36. [Google Scholar] [CrossRef]
  69. Aldámiz-Echevarría, L.; Prieto, J.A.; Andrade, F.; Elorz, J.; Sojo, A.; Lage, S.; Sanjurjo, P.; Vázquez, C.; Rodríguez-Soriano, J. Persistence of essential fatty acid deficiency in cystic fibrosis despite nutritional therapy. Pediatric Res. 2009, 66, 585–589. [Google Scholar] [CrossRef] [Green Version]
  70. Lindblad, A.; Glaumann, H.; Strandvik, B. Natural history of liver disease in cystic fibrosis. Hepatology 1999, 30, 1151–1158. [Google Scholar] [CrossRef]
  71. Colombo, C.; Alicandro, G.; Oliver, M.; Lewindon, P.J.; Ramm, G.A.; Ooi, C.Y.; Alghisi, F.; Kashirskaya, N.; Kondratyeva, E.; Corti, F.; et al. Ursodeoxycholic acid and liver disease associated with cystic fibrosis: A multicenter cohort study. J. Cyst. Fibros. 2021, in press. [CrossRef]
  72. Harayama, T.; Riezman, H. Understanding the diversity of membrane lipid composition. Nat. Rev. Mol. Cell Biol. 2018, 19, 281–296. [Google Scholar] [CrossRef]
  73. Strandvik, B.; Svensson, E.; Seyberth, H.W. Prostanoid biosynthesis in patients with cystic fibrosis. Prostaglandins Leukot. Essent. Fat. Acids 1996, 55, 419–425. [Google Scholar] [CrossRef]
  74. Dodge, J.A.; Custance, J.M.; Goodchild, M.C.; Laing, S.C.; Vaughan, M. Paradoxical effects of essential fatty acid supplementation on lipid profiles and sweat electrolytes in cystic fibrosis. Br. J. Nutr. 1990, 63, 259–271. [Google Scholar] [CrossRef] [Green Version]
  75. Van, D.; Beerthuis, R.K.; Nugteren, D.H.; Vonkeman, H. The biosynthesis of prostaglandins. Biochim. Biophys. Acta 1964, 90, 204–207. [Google Scholar] [CrossRef]
  76. Brenner, R.R.; Peluffo, R.O.; Nervi, A.M.; De Thomas, M.E. Competitive effect of alpha- and gamma-lionlenyl-CoA in linoleyl-CoA desaturation to gamma-linolenyl-CoA. Biochim. Biophys. Acta 1969, 176, 420–422. [Google Scholar] [CrossRef]
  77. Peluffo, R.O.; Nervi, A.M.; Brenner, R.R. Linoleic acid desaturation activity of liver microsomes of essential fatty acid deficient and sufficient rats. Biochim. Biophys. Acta 1976, 441, 25–31. [Google Scholar] [CrossRef]
  78. Seegmiller, A.C. Abnormal unsaturated fatty acid metabolism in cystic fibrosis: Biochemical mechanisms and clinical implications. Int. J. Mol. Sci. 2014, 15, 16083–16099. [Google Scholar] [CrossRef] [Green Version]
  79. Strandvik, B.; Brönnegård, M.; Gilljam, H.; Carlstedt-Duke, J. Relation between defective regulation of arachidonic acid release and symptoms in cystic fibrosis. Scand. J. Gastroenterol. Suppl. 1988, 143, 1–4. [Google Scholar] [CrossRef]
  80. Carlstedt-Duke, J.; Brönnegård, M.; Strandvik, B. Pathological regulation of arachidonic acid release in cystic fibrosis: The putative basic defect. Proc. Natl. Acad. Sci. USA 1986, 83, 9202–9206. [Google Scholar] [CrossRef] [Green Version]
  81. Levistre, R.; Lemnaouar, M.; Rybkine, T.; Béréziat, G.; Masliah, J. Increase of bradykinin-stimulated arachidonic acid release in a delta F508 cystic fibrosis epithelial cell line. Biochim. Biophys. Acta 1993, 1181, 233–239. [Google Scholar] [CrossRef]
  82. Berguerand, M.; Klapisz, E.; Thomas, G.; Humbert, L.; Jouniaux, A.M.; Olivier, J.L.; Béréziat, G.; Masliah, J. Differential stimulation of cytosolic phospholipase A2 by bradykinin in human cystic fibrosis cell lines. Am. J. Respir. Cell Mol. Biol. 1997, 17, 481–490. [Google Scholar] [CrossRef] [Green Version]
  83. Bhura-Bandali, F.N.; Suh, M.; Man, S.F.; Clandinin, M.T. The deltaF508 mutation in the cystic fibrosis transmembrane conductance regulator alters control of essential fatty acid utilization in epithelial cells. J. Nutr. 2000, 130, 2870–2875. [Google Scholar] [CrossRef] [Green Version]
  84. Miele, L.; Cordella-Miele, E.; Xing, M.; Frizzell, R.; Mukherjee, A.B. Cystic fibrosis gene mutation (deltaF508) is associated with an intrinsic abnormality in Ca2+-induced arachidonic acid release by epithelial cells. DNA Cell Biol. 1997, 16, 749–759. [Google Scholar] [CrossRef]
  85. Ulane, M.M.; Butler, J.D.; Peri, A.; Miele, L.; Ulane, R.E.; Hubbard, V.S. Cystic fibrosis and phosphatidylcholine biosynthesis. Clin. Chim. Acta 1994, 230, 109–116. [Google Scholar] [CrossRef]
  86. Strandvik, B. Long Chain Fatty Acid Metabolism and Essential Fatty Acid Deficiency with Special Emphasis on Cystic Fibrosis; Bracco, U., Deckelbaum, R., Eds.; Raven Press: New York, NY, USA, 1992; Volume 28, pp. 159–167. [Google Scholar]
  87. Strandvik, B. Relation between essential fatty acid metabolism and gastrointestinal symptoms in cystic fibrosis. Acta Paediatr. Scand. Suppl. 1989, 363, 58–63. [Google Scholar] [CrossRef]
  88. Van Biervliet, S.; Van Biervliet, J.P.; Robberecht, E.; Christophe, A. Fatty acid composition of serum phospholipids in cystic fibrosis (CF) patients with or without CF related liver disease. Clin. Chem. Lab. Med. 2010, 48, 1751–1755. [Google Scholar] [CrossRef]
  89. Drzymała-Czyż, S.; Szczepanik, M.; Krzyżanowska, P.; Duś-Żuchowska, M.; Pogorzelski, A.; Sapiejka, E.; Juszczak, P.; Lisowska, A.; Koletzko, B.; Walkowiak, J. Serum Phospholipid Fatty Acid Composition in Cystic Fibrosis Patients with and without Liver Cirrhosis. Ann. Nutr. Metab. 2017, 71, 91–98. [Google Scholar] [CrossRef]
  90. Beharry, S.; Ackerley, C.; Corey, M.; Kent, G.; Heng, Y.M.; Christensen, H.; Luk, C.; Yantiss, R.K.; Nasser, I.A.; Zaman, M.; et al. Long-term docosahexaenoic acid therapy in a congenic murine model of cystic fibrosis. Am. J. Physiol. Gastrointest. Liver Physiol. 2007, 292, G839–G848. [Google Scholar] [CrossRef]
  91. Freedman, S.D.; Katz, M.H.; Parker, E.M.; Laposata, M.; Urman, M.Y.; Alvarez, J.G. A membrane lipid imbalance plays a role in the phenotypic expression of cystic fibrosis in cftr(−/−) mice. Proc. Natl. Acad. Sci. USA 1999, 96, 13995–14000. [Google Scholar] [CrossRef] [Green Version]
  92. Watson, H.; Stackhouse, C. Omega-3 fatty acid supplementation for cystic fibrosis. Cochrane Database Syst. Rev. 2020, 4, Cd002201. [Google Scholar] [CrossRef]
  93. Wassall, S.R.; Leng, X.; Canner, S.W.; Pennington, E.R.; Kinnun, J.J.; Cavazos, A.T.; Dadoo, S.; Johnson, D.; Heberle, F.A.; Katsaras, J.; et al. Docosahexaenoic acid regulates the formation of lipid rafts: A unified view from experiment and simulation. Biochim. Biophys. Acta Biomembr. 2018, 1860, 1985–1993. [Google Scholar] [CrossRef]
  94. Chiang, N.; Serhan, C.N. Specialized pro-resolving mediator network: An update on production and actions. Essays Biochem. 2020, 64, 443–462. [Google Scholar] [CrossRef]
  95. Philippe, R.; Urbach, V. Specialized Pro-Resolving Lipid Mediators in Cystic Fibrosis. Int. J. Mol. Sci. 2018, 19, 2865. [Google Scholar] [CrossRef] [Green Version]
  96. Recchiuti, A.; Isopi, E.; Romano, M.; Mattoscio, D. Roles of Specialized Pro-Resolving Lipid Mediators in Autophagy and Inflammation. Int. J. Mol. Sci. 2020, 21, 6637. [Google Scholar] [CrossRef]
  97. Mingione, A.; Ottaviano, E.; Barcella, M.; Merelli, I.; Rosso, L.; Armeni, T.; Cirilli, N.; Ghidoni, R.; Borghi, E.; Signorelli, P. Cystic Fibrosis Defective Response to Infection Involves Autophagy and Lipid Metabolism. Cells 2020, 9, 1845. [Google Scholar] [CrossRef] [PubMed]
  98. Isopi, E.; Mattoscio, D.; Codagnone, M.; Mari, V.C.; Lamolinara, A.; Patruno, S.; D’Aurora, M.; Cianci, E.; Nespoli, A.; Franchi, S.; et al. Resolvin D1 Reduces Lung Infection and Inflammation Activating Resolution in Cystic Fibrosis. Front. Immunol. 2020, 11, 581. [Google Scholar] [CrossRef] [PubMed]
  99. López-Neyra, A.; Suárez, L.; Muñoz, M.; de Blas, A.; Ruiz de Valbuena, M.; Garriga, M.; Calvo, J.; Ribes, C.; Girón Moreno, R.; Máiz, L.; et al. Long-term docosahexaenoic acid (DHA) supplementation in cystic fibrosis patients: A randomized, multi-center, double-blind, placebo-controlled trial. Prostaglandins Leukot. Essent. Fat. Acids 2020, 162, 102186. [Google Scholar] [CrossRef] [PubMed]
  100. Urbach, V.; Higgins, G.; Buchanan, P.; Ringholz, F. The role of Lipoxin A4 in Cystic Fibrosis Lung Disease. Comput. Struct. Biotechnol. J. 2013, 6, e201303018. [Google Scholar] [CrossRef] [Green Version]
  101. 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]
  102. Van Biervliet, S.; Vanbillemont, G.; Van Biervliet, J.P.; Declercq, D.; Robberecht, E.; Christophe, A. Relation between fatty acid composition and clinical status or genotype in cystic fibrosis patients. Ann. Nutr. Metab. 2007, 51, 541–549. [Google Scholar] [CrossRef] [PubMed]
  103. Welsh, M.J.; Smith, A.E. Molecular mechanisms of CFTR chloride channel dysfunction in cystic fibrosis. Cell 1993, 73, 1251–1254. [Google Scholar] [CrossRef]
  104. De Boeck, K.; Amaral, M.D. Progress in therapies for cystic fibrosis. Lancet Respir. Med. 2016, 4, 662–674. [Google Scholar] [CrossRef]
  105. Zardini Buzatto, A.; Abdel Jabar, M.; Nizami, I.; Dasouki, M.; Li, L.; Abdel Rahman, A.M. Lipidome Alterations Induced by Cystic Fibrosis, CFTR Mutation, and Lung Function. J. Proteome Res. 2021, 20, 549–564. [Google Scholar] [CrossRef]
  106. Guerrera, I.C.; Astarita, G.; Jais, J.P.; Sands, D.; Nowakowska, A.; Colas, J.; Sermet-Gaudelus, I.; Schuerenberg, M.; Piomelli, D.; Edelman, A.; et al. A novel lipidomic strategy reveals plasma phospholipid signatures associated with respiratory disease severity in cystic fibrosis patients. PLoS ONE 2009, 4, e7735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Korotkova, M.; Strandvik, B. Essential fatty acid deficiency affects the fatty acid composition of the rat small intestinal and colonic mucosa differently. Biochim. Biophys. Acta 2000, 1487, 319–325. [Google Scholar] [CrossRef]
  108. Witters, P.; Dupont, L.; Vermeulen, F.; Proesmans, M.; Cassiman, D.; Wallemacq, P.; De Boeck, K. Lung transplantation in cystic fibrosis normalizes essential fatty acid profiles. J. Cyst. Fibros. 2013, 12, 222–228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Hanssens, L.; Duchateau, J.; Namane, S.A.; Malfroot, A.; Knoop, C.; Casimir, G. Influence of lung transplantation on the essential fatty acid profile in cystic fibrosis. Prostaglandins Leukot. Essent. Fat. Acids 2020, 158, 102060. [Google Scholar] [CrossRef]
  110. Parsons, H.G.; O’Loughlin, E.V.; Forbes, D.; Cooper, D.; Gall, D.G. Supplemental calories improve essential fatty acid deficiency in cystic fibrosis patients. Pediatric Res. 1988, 24, 353–356. [Google Scholar] [CrossRef] [Green Version]
  111. Rogiers, V.; Dab, I.; Crokaert, R.; Vis, H.L. Long chain non-esterified fatty acid pattern in plasma of cystic fibrosis patients and their parents. Pediatric Res. 1980, 14, 1088–1091. [Google Scholar] [CrossRef] [Green Version]
  112. Harindhanavudhi, T.; Wang, Q.; Dunitz, J.; Moran, A.; Moheet, A. Prevalence and factors associated with overweight and obesity in adults with cystic fibrosis: A single-center analysis. J. Cyst. Fibros. 2020, 19, 139–145. [Google Scholar] [CrossRef] [Green Version]
  113. Hanssens, L.S.; Duchateau, J.; Casimir, G.J. CFTR Protein: Not Just a Chloride Channel? Cells 2021, 10, 2844. [Google Scholar] [CrossRef]
  114. Nowak, J.K.; Szczepanik, M.; Wojsyk-Banaszak, I.; Mądry, E.; Wykrętowicz, A.; Krzyżanowska-Jankowska, P.; Drzymała-Czyż, S.; Nowicka, A.; Pogorzelski, A.; Sapiejka, E.; et al. Cystic fibrosis dyslipidaemia: A cross-sectional study. J. Cyst. Fibros. 2019, 18, 566–571. [Google Scholar] [CrossRef]
  115. Shaikhkhalil, A.K.; Freeman, A.J.; Sathe, M. Variations in Nutrition Practices in Cystic Fibrosis: A Survey of the DIGEST Program. Nutr. Clin. Pract. 2021, 36, 1247–1251. [Google Scholar] [CrossRef]
  116. Strandvik, B. Is the ENaC Dysregulation in CF an Effect of Protein-Lipid Interaction in the Membranes? Int. J. Mol. Sci. 2021, 22, 2739. [Google Scholar] [CrossRef] [PubMed]
  117. Rosenlund, M.L.; Selekman, J.A.; Kim, H.K.; Kritchevsky, D. Dietary essential fatty acids in cystic fibrosis. Pediatrics 1977, 59, 428–432. [Google Scholar] [CrossRef] [PubMed]
  118. Sigström, L.; Strandvik, B. Erythrocyte sodium-potassium transport in cystic fibrosis. Pediatric Res. 1992, 31, 425–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Lloyd-Still, J.D.; Powers, C.A.; Hoffman, D.R.; Boyd-Trull, K.; Lester, L.A.; Benisek, D.C.; Arterburn, L.M. Bioavailability and safety of a high dose of docosahexaenoic acid triacylglycerol of algal origin in cystic fibrosis patients: A randomized, controlled study. Nutrition 2006, 22, 36–46. [Google Scholar] [CrossRef] [PubMed]
  120. Alicandro, G.; Faelli, N.; Gagliardini, R.; Santini, B.; Magazzù, G.; Biffi, A.; Risé, P.; Galli, C.; Tirelli, A.S.; Loi, S.; et al. A randomized placebo-controlled study on high-dose oral algal docosahexaenoic acid supplementation in children with cystic fibrosis. Prostaglandins Leukot. Essent. Fat. Acids 2013, 88, 163–169. [Google Scholar] [CrossRef]
  121. Van Biervliet, S.; Devos, M.; Delhaye, T.; Van Biervliet, J.P.; Robberecht, E.; Christophe, A. Oral DHA supplementation in DeltaF508 homozygous cystic fibrosis patients. Prostaglandins Leukot. Essent. Fat. Acids 2008, 78, 109–115. [Google Scholar] [CrossRef]
  122. Simon, M.; Dalle Molle, R.; Silva, F.M.; Rodrigues, T.W.; Feldmann, M.; Forte, G.C.; Marostica, P.J.C. Antioxidant Micronutrients and Essential Fatty Acids Supplementation on Cystic Fibrosis Outcomes: A Systematic Review. J. Acad. Nutr. Diet. 2020, 120, 1016–1033. [Google Scholar] [CrossRef]
  123. Bass, R.; Brownell, J.N.; Stallings, V.A. The Impact of Highly Effective CFTR Modulators on Growth and Nutrition Status. Nutrients 2021, 13, 2907. [Google Scholar] [CrossRef]
  124. Harris, J.K.; Wagner, B.D.; Zemanick, E.T.; Robertson, C.E.; Stevens, M.J.; Heltshe, S.L.; Rowe, S.M.; Sagel, S.D. Changes in Airway Microbiome and Inflammation with Ivacaftor Treatment in Patients with Cystic Fibrosis and the G551D Mutation. Ann. Am. Thorac. Soc. 2020, 17, 212–220. [Google Scholar] [CrossRef]
  125. Veltman, M.; De Sanctis, J.B.; Stolarczyk, M.; Klymiuk, N.; Bähr, A.; Brouwer, R.W.; Oole, E.; Shah, J.; Ozdian, T.; Liao, J.; et al. CFTR Correctors and Antioxidants Partially Normalize Lipid Imbalance but not Abnormal Basal Inflammatory Cytokine Profile in CF Bronchial Epithelial Cells. Front. Physiol. 2021, 12, 619442. [Google Scholar] [CrossRef]
  126. Vertex. Pharmaceuticals; Incorporated. Prescribing Information. TRIKAFTA (Elexaccaftor/Tezacaftor/Ivacaftor; Ivacaftor Tablets. Table 4: Pharmacokinetic Parameters of TRIKAFTA Components. 2019. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/212273s000lbl.pdf (accessed on 10 February 2022).
  127. Laselva, O.; Guerra, L.; Castellani, S.; Favia, M.; Di Gioia, S.; Conese, M. Small-molecule drugs for cystic fibrosis: Where are we now? Pulm. Pharmacol. Ther. 2021, 72, 102098. [Google Scholar] [CrossRef] [PubMed]
  128. Hansen, C.R.; Gilljam, M.; Olesen, H.V.; Høiby, N.; Karpati, F.; Johansson, E.; Krantz, C.; Skov, M.; Pressler, T.; Lindblad, A. Maintaining normal lung function in children with cystic fibrosis is possible with aggressive treatment regardless of Pseudomonas aeruginosa infections. Acta Paediatr. 2021, 110, 2607–2609. [Google Scholar] [CrossRef] [PubMed]
  129. Garić, D.; Dumut, D.C.; Shah, J.; De Sanctis, J.B.; Radzioch, D. The role of essential fatty acids in cystic fibrosis and normalizing effect of fenretinide. Cell. Mol. Life Sci. 2020, 77, 4255–4267. [Google Scholar] [CrossRef] [PubMed]
  130. Vandebrouck, C.; Ferreira, T. Glued in lipids: Lipointoxication in cystic fibrosis. EBioMedicine 2020, 61, 103038. [Google Scholar] [CrossRef] [PubMed]
  131. O’Connor, M.G.; Seegmiller, A. The effects of ivacaftor on CF fatty acid metabolism: An analysis from the GOAL study. J. Cyst. Fibros. 2017, 16, 132–138. [Google Scholar] [CrossRef] [Green Version]
  132. Strandvik, B.; Walkowiak, J.; Drzymala-Czyz, S.; Colombo, C.; Alicandro, A.; Bakkeheim, E.; Badolato, R.; Hansen, C. A double-blind randomised multi-centre European study of linoleic acid supplementation for one year in patients with cystic fibrosis. J. Cyst. Fibros. 2021, 20, S56. [Google Scholar] [CrossRef]
Figure 1. Major pathways of polyunsaturated fatty acid synthesis. Simplified major transformation steps to long-chain polyunsaturated fatty acids from the essential fatty acids, linoleic (LA) and α-linolenic (ALA) acids and the endogenously synthesized oleic acid (OA), which all compete for the same desaturases (FADS1 and FADS2) and elongases. The major metabolites are indicated in boxes, proinflammatory and ellipses (anti-inflammatory) lipid mediators. ARA, arachidonic acid; EPA, eicosapentaenoic acid; DHA, docosahexaenoic acid; LT, leukotrienes; TX, tromboxanes.
Figure 1. Major pathways of polyunsaturated fatty acid synthesis. Simplified major transformation steps to long-chain polyunsaturated fatty acids from the essential fatty acids, linoleic (LA) and α-linolenic (ALA) acids and the endogenously synthesized oleic acid (OA), which all compete for the same desaturases (FADS1 and FADS2) and elongases. The major metabolites are indicated in boxes, proinflammatory and ellipses (anti-inflammatory) lipid mediators. ARA, arachidonic acid; EPA, eicosapentaenoic acid; DHA, docosahexaenoic acid; LT, leukotrienes; TX, tromboxanes.
Nutrients 14 00853 g001
Table 1. Calculated fat intake in g/capita/day of commonly used vegetable oils, dairy fats, raw animal fat and fish (sum of demersal, pelagic and other marine fishes) in the USA, Canada, the United Kingdom, Belgium, The Netherlands, Spain, Italy and Sweden according to the Food and Agriculture Organization of the United Nations (FAO) 2019 (www.fao.org/faostat/en/#data/FBS, accessed on: 9 February 2022). + indicates that information aout butter is missing.
Table 1. Calculated fat intake in g/capita/day of commonly used vegetable oils, dairy fats, raw animal fat and fish (sum of demersal, pelagic and other marine fishes) in the USA, Canada, the United Kingdom, Belgium, The Netherlands, Spain, Italy and Sweden according to the Food and Agriculture Organization of the United Nations (FAO) 2019 (www.fao.org/faostat/en/#data/FBS, accessed on: 9 February 2022). + indicates that information aout butter is missing.
FatUSACanadaUKBelgiumNetherlandsItalySpainSweden
Soybean oil61.81.7913.511.611.610.614.10.16
Sunflower oil0.520.947.079.541.6931.611.76.99
Corn oil6.672.791.254.821.831.612.880.16
Olive oil2.673.002.673.422.3030.927.22.48
Ratio n-6 richoils/olive oil25.81.848.177.606.731.421.052.95
Cream + milk24.219.621.233.633.217.019.228.6
Butter5.418.276.43ND13.315.52.7811.3
Animal fat, raw5.8412.95.6626.710.82.478.455.87
Fish1.171.122.291.851.512.942.141.50
Ratio n-6 rich oils/
dairy + animal fat
2.300.170.810.430.352.251.040.21
Ratio n-6 rich oils/
dairy + butter + animal fat
1.950.140.66ND0.271.750.940.16
Total fat, (based on presented fat categories)10647.457.488.1+74.371.761.354.6
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Strandvik, B. Nutrition in Cystic Fibrosis—Some Notes on the Fat Recommendations. Nutrients 2022, 14, 853. https://0-doi-org.brum.beds.ac.uk/10.3390/nu14040853

AMA Style

Strandvik B. Nutrition in Cystic Fibrosis—Some Notes on the Fat Recommendations. Nutrients. 2022; 14(4):853. https://0-doi-org.brum.beds.ac.uk/10.3390/nu14040853

Chicago/Turabian Style

Strandvik, Birgitta. 2022. "Nutrition in Cystic Fibrosis—Some Notes on the Fat Recommendations" Nutrients 14, no. 4: 853. https://0-doi-org.brum.beds.ac.uk/10.3390/nu14040853

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