Skip to main content

Vitamin D deficiency in patients with cystic fibrosis: a systematic review and meta-analysis

Abstract

Aim

Vitamin D is a prominent modulator of immunity and respiratory function. It plays a vital role in respiratory diseases such as cystic fibrosis (CF). S. However, there is a dearth of information on patients with CF. The purpose of the meta-analysis is to highlight the importance of following the existing guidelines regarding maintenance of Vitamin D serum levels in patients with CF.

Methods

The systematic search was conducted without utilizing any time or language limitations in original database from the beginning until March 2022. The meta-analysis was performed using a random-effects model. Heterogeneity was determined by I2 statistics and Cochrane Q test.

Results

Pooled analysis using the random-effects model of the 8 case–control studies with 13 effect sizes revealed that the serum 25-OH-vitamin D in participants with cystic fibrosis was significantly lower than controls in pediatrics and adolescences (WMD: − 3.41 ng/ml, 95% CI − 5.02, − 1.80, p =  < 0.001) and adults (WMD: − 2.60 ng/ml, 95% CI − 4.32, − 0.89, p = 0.003). Based on data from 12 studies (21 effect sizes) with a total of 1622 participants, the prevalence of vitamin D levels of 20–30 ng/ml in CF patients was 36% among pediatrics/adolescents and 63% among adults. In addition, 27% of pediatric/adolescent CF patients and 35% of adult CF patients had vitamin D levels of below 20 ng/ml.

Conclusions

As a result, according to the existing guidelines, our results proved the need to pay attention to the level of vitamin D in these patients.

Introduction

Cystic fibrosis (CF) is a disease caused by a mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene with an autosomal recessive pattern of inheritance. This mutation impairs chloride transport across the epithelial membranes, thickening the mucus layer on the surfaces of the lung, intestines, pancreas, and other organs' surfaces [1]. This results in persistent infection and airway inflammation, leading to respiratory failure [2,3,4]. The exocrine and endocrine pancreas, the gastrointestinal tract, and the lungs are just a few impacted by CF. This results in lipid malabsorption and lipid-soluble vitamin deficiencies [5].

Vitamin D deficiency is common in CF patients because of fat malabsorption due to exocrine pancreatic impairment, as well as inadequate dietary intake, alterations in vitamin D metabolism, and limited sun exposure [6,7,8,9]. Despite replacing vitamin D patients who are given oral supplements, vitamin D deficiency has been reported to affect up to 90% of CF patients at [5] and the majority of patients fail to achieve therapeutic levels vitamin D deficient (< 20 ng/ml) or insufficient (20–30 ng/ml) than healthy controls.

Lower levels of circulating 25-hydroxyvitamin D [25(OH)D], a determinant of vitamin D status, are associated with a high incidence of upper respiratory tract infections and chronic lung diseases [10]. According to clinical and in vitro studies, vitamin D increases the synthesis of cathelicidin (LL–37), a critical antimicrobial peptide, showing a molecular mechanism for its immunomodulatory functions [11,12,13]. Numerous studies have shown that children and adults affected by CF are more likely to experience lung exacerbations when they are vitamin D deficient [18,19,20]. Recent pilot studies showed that vitamin D supplementation in CF patients accelerated healing following a lung exacerbation of CF [14,15,16]. There is no clear evidence of a direct association between vitamin D levels in children and adolescence with CF; we conducted a systematic review to assess this association.

Methods

Search strategy

There is a comprehensive systematic search in PubMed/MEDLINE, Web of Science, SCOPUS, and Embase from inception until March 2022 without using time or language restrictions. Keywords from the medical subject headings (Mesh) database were used for this search as follows: (vitamin D OR vit D OR ergocalciferols OR calcifediol’ OR 25-hydroxyvitamin D) AND (Cystic Fibrosis OR CF). Additionally, the reference lists of the articles retrieved, dissertation of Phd or Masters, also the gray literature, and related review studies were also hand-screened to identify eligible publications that our search might have omitted.

Study selection

After excluding duplicate articles, two authors independently reviewed the titles, the abstracts, or the full text of the retrieved studies to detect eligible publications. Finally, original studies were included in the present meta-analysis if they met the following criteria: (1) population: adults and children of all ages and genders; (2) exposure: cystic fibrosis; (3); outcome: serum 25(OH) vitamin D; and (4) study design: cross-sectional, case–control, or cohort studies. Articles excluded from the analysis include: (1) studies not reporting the association between serum 25(OH) vitamin D and cystic fibrosis, (2) reviews, editorials, letters, or commentaries literature, (3) unpublished studies, (4) conference proceedings, (5) duplicates or irrelevant studies, and (6) animal, or in vitro or in vivo studies, and studies whose corresponding author did not offer any feedback after several emails.

Data extraction

Two independent researchers reviewed the data and an additional reviewer resolved any disagreements between the two. The following information was collected: author, year of publication, country, age, participants’ gender, sample size, mean and standard deviation of serum 25(OH) vitamin D, prevalence of serum vitamin D deficiency and/or insufficiency.

Quality assessment

Studies included in the meta-analysis were assessed using the Newcastle–Ottawa scale for cross-sectional and cohort studies. A quality score was specified on the basis of three major components: selection of study groups (0–5 points), adequacy of adjustment for confounding variables (0–2 points), and ascertainment of the outcome of interest (0–3 points). High-quality studies were defined as those that scored with at least seven stars on the Newcastle–Ottawa scale. Medium-quality studies received scores of five to six stars [17].

Data synthesis and statistical analysis

The statistical analysis was conducted using STATA software version 12.0 (Stata Corp, College Station, TX, USA). Data were combined, and if there were ≥ 3 case–controls, the random-effects model was used and reported as weighted mean differences (WMDs) [18]. The prevalence of serum vitamin D deficiency and insufficiency was expressed as proportions and 95% confidence intervals (95% CI) using the random-effects model and presented as forest plot. Heterogeneity was examined using the I-squared (I2) statistic, in which the source of heterogeneity was determined if the I2 value was > 50%. We assessed the presence of publication bias using the formal Egger’s test [19, 20].

Results

Figure 1 shows a flowchart of the study selection process and reasons for exclusion.. Following the systematic search, 713 publications were obtained and after excluding duplicate studies, a total of 517 publications were included. Then, we reviewed the title/abstract of the remaining studies and excluded 434 articles which did not meet the inclusion criteria. A total of 83 articles were retrieved during the secondary screening (by full text). Of these, 63 studies were discarded since they did not include data of interest. Finally, 20 studies (case–control = 8 and cross-sectional = 13) met the eligibility criteria and were included in the quantitative meta-analysis.

Fig. 1
figure 1

Flowchart of the selected studies, including identification, screening, eligibility and the final sample included

Study characteristics

The characteristics of the pooled studies are presented in Tables 1 and 2. All articles were published between 1981 and 2018. Among the studies integrated in this systematic review, 8 studies were case–control [21,22,23,24,25,26,27,28] and 12 studies had a cross-sectional design [29,30,31,32,33,34,35,36,37,38,39,40]. The total sample size was 1,157 in the case control studies and 1622 patients in the cross-sectional studies. Of the 20 papers, 10 were from children, 7 from adults, and 1 included all age groups (or both children and adults). Other variables are completely presented in Tables 1 and 2.

Table 1 Characteristics of included cross-sectional studies
Table 2 Characteristics of the included case–control studies

Meta-analysis results for case–control studies

Pooled analysis using the random-effects model of the 8 case–control studies (with 13 effect sizes) revealed that the serum 25-OH-vitamin D in participants with cystic fibrosis was significantly lower than controls in pediatrics and adolescences (WMD: − 3.41 ng/ml, 95% CI − 5.02, − 1.80, p ≤ 0.001) and adults (WMD: − 2.60 ng/ml, 95% CI − 4.32, − 0.89, p = 0.003) without a significant heterogeneity seen among the papers (I2 = 17.8%, p = 0.29 in pediatrics and adolescences and I2 = 0%, p = 0.55 in adults) (Fig. 2).

Fig. 2
figure 2

Pooled results of case–control studies for the analysis of relationship between serum 25-OH-vitamin D and cystic fibrosis

Meta-analysis results for cross-sectional studies

Based on data from 12 studies [21 effect sizes] with a total of 1622 participants, we detected an overall prevalence of vitamin D of 20–30 ng/ml in pediatrics/adolescences and adults with cystic fibrosis of 36% (95% CI 0.04, 0.68) and 63% (95% CI 0.57, 0.69), respectively, with significant heterogeneity noted among the included studies (I2 = 98.11%, P < 0.001; and I2 = 98.14%, P < 0.001; for pediatrics/adolescences and adults). The pooled prevalence of vitamin D < 20 ng/ml was 27% (95% CI 0.09, 0.45) in pediatrics/adolescences and 35% (95% CI 0.13, 0.57) in adults with cystic fibrosis (I2 = 95.42%, P < 0.001; and I2 = 98.16%, P < 0.001, respectively, for pediatrics/adolescences and adults) (Fig. 3).

Fig. 3
figure 3

This figure shows the pooled estimate of the prevalence of vitamin D deficiency and insufficiency in A pediatrics and adolescents and B adults with cystic fibrosis patients

Sensitivity analysis

The leave-one-out method was applied to assess the influence of each individual study on the pooled effect size. The findings remained robust after sequential elimination of studies (Additional file 1: Fig. S1).

Publication bias

The visual inspection of funnel plot revealed some evidence of publication bias regarding the association between serum vitamin D deficiency and insufficiency and cystic fibrosis. However, the Egger’s test also showed no evidence of bias for case–control (P = 0.59) or cross-sectional studies (P = 0.82) (Additional file 1: Fig. S2). Also, meta trim-and-fill analysis did not identify a study responsible for the publication bias (Table 3).

Table 3 Results of meta-analysis

Discussion

It is well recognized that vitamin D deficiency and insufficiency are common in the general population and significantly more prevalent among CF patients [41]. Both the Cystic Fibrosis Foundation's and the Endocrine Society's guidelines recommend 25(OH)D levels above 30 ng/ml [8, 42]. In the current meta-analysis, CF patients had significantly lower levels of vitamin D in comparison with healthy controls in both childhood/adolescence and adulthood. We also found that the prevalence of vitamin D levels of 20–30 ng/ml in CF patients was 36% among pediatrics/adolescents and 63% among adults. In addition, 27% of pediatric/adolescent CF patients and 35% of adult CF patients had vitamin D levels of below 20 ng/ml. These results were similar to the findings of Thursfield et al. who reported that 36% of the population of children and adults had vitamin D deficiency (< 50 nmol/L).

Guidelines for treating vitamin D deficiency have been published by the Cystic Fibrosis Foundation, which recommends serum levels of 25-hydroxyvitamin D of at least 30 ng/ml. The recommendations give age-specific rising dose regimes with monitoring at 12-week intervals after switching therapies. They discuss the research on appropriate formulations (cholecalciferol rather than ergocalciferol) and vehicles of administration, as well as alternative vitamin D sources like UV lamps. Despite these comprehensive recommendations patients are still deficient [43].

Rovner et al. [5] found a 20% increase in vitamin D insufficiency in children, adolescents, and young adults with CF who were receiving regular treatment with pancreatic enzyme replacement and vitamin D supplementation. The study also used a healthy reference group whose ethnicity and latitude of residence were similar to those of the CF group and adjusted for seasonal fluctuations. Since seasonal variation in serum vitamin D levels is well recognized, comparing the degree of vitamin D insufficiency in the CF population with controls can be challenging [44]. Many studies do not standardize or take seasonal variation into account. Acute changes in 25-hydroxyvitamin D levels can also result from pancreatic enzyme compliance and vitamin D supplementation regimens employing ergocalciferol (D2) or cholecalciferol (D3).

Compared to the general population, some factors can worsen vitamin D insufficiency in people with CF. Pancreatic insufficiency causes fat malabsorption in CF patients. Absorption may be reduced even with pancreatic enzyme supplementation [45] and poor diet and noncompliance with medication. Sunlight exposure was the most accurate predictor of vitamin D deficiency before lung exacerbation, according to one study [46]; however, some individuals with CF should stay out of the sun since their use of antibiotics causes photosensitivity [47]. On the other hand, many CF patients are underweight, their bodies may not keep as much vitamin D as persons with healthy body weight [47]. Furthermore, inadequate vitamin D 25-hydroxylation in the liver and rapid enterohepatic dumping might reduce total vitamin D storage [45, 48]. Due to lower amounts of vitamin D binding protein, the primary carrier in circulation, and aid in recovering 25-hydroxyvitamin D discharged in urine, patients with CF may have impaired vitamin D storage [49]. Additionally, exposure to glucocorticoids, rifampin, and isoniazid may cause patients with CF to catabolize vitamin D more quickly [50, 51].

Clinical studies and in vitro research have provided compelling evidence that vitamin D may be crucial to the innate immune system. The antimicrobial response to bacterial toll-like receptor activation in cultured human macrophages was initially documented by Liu et al. [12]. The results of subsequent human research showed that vitamin D administration increased the local mRNA expression of cathelicidin in peripheral blood monocytes but not the levels of circulating LL-37 (a cathelicidin cleaved product) [52]. Cathelicidin mRNA expression is elevated in cultured bronchial epithelial cells with the CFTR mutation F508 in response to vitamin D therapy [53]. Furthermore, Schögler et al. [54] showed that primary bronchial epithelial cells isolated from CF patients had higher cathelicidin mRNA expression when exposed to vitamin D. They did not observe changes in the circulating protein concentrations of LL-37 in response to vitamin D in the investigation of CF patients experiencing acute pulmonary exacerbation. The use of antibiotics during pulmonary exacerbation may interfere with the vitamin D's ability to induce LL-37, which could account for the absence of alterations observed in response to vitamin D in circulation [55]. Another option is that LL-37 interacts with host-derived glycosaminoglycan, neutrophil extracellular trap DNA, and anionic bacterial compounds such endotoxin (LPS) and capsular polysaccharides [56,57,58,59]. Particularly during pulmonary exacerbations, the CF host's high quantity of bacterial-derived anionic molecules inhibits LL-37 action and may lower plasma LL-37 concentrations. Additionally, the choice to begin IV antibiotics by clinicians during an acute pulmonary exacerbation is not well standardized, which may have had an impact on their results [60]. In addition, even receiving IV antibiotics, 25–50% CF patients do not regain their baseline lung function [61, 62]. Another explanation for the absence of alterations could be that it was not possible to determine any potential local changes in lung LL-37 concentrations. It may be possible to determine whether vitamin D improves these local cellular responses by looking at the local mRNA expression of cathelicidin in peripheral blood monocytes or in monocytes and epithelial cells isolated from bronchial alveolar lavage.

VD deficiency in CF patients is associated with several complications?. Insufficient vitamin D causes associated with calcium malabsorption, resulting in secondary hyperparathyroidism. Increased parathyroid hormone levels result in calcium resorption from bone, which weakens bones, causes skeletal losses, and accelerates the onset of osteoporosis [63]. CF patients have low bone mineral density and a relatively high fracture prevalence rate of 20%, which may be related to the high rates of vitamin D insufficiency [64]. Low vitamin D levels in patients without CF are linked to a higher risk of developing cancer, autoimmune disorders, infections, and cardiovascular disease [63, 65,66,67].

The CF population's ability to maintain lung function may be helped by vitamin D [68]. The forced expiratory volume in one second (FEV1) and forced vital capacity (FVC), which measure lung function, showed a significant connection with vitamin D status in the Third National Health and Nutrition Examination Survey (NHANES III) [69]. In a retrospective cohort, patients with CF showed a positive correlation between vitamin D levels and FEV1 [6]. Higher vitamin D levels are associated with Hetterlung function and lower rates of pulmonary exacerbation, according to several studies on CF populations [6, 70,71,72]. Based on research on CF and other chronic lung diseases, vitamin D may protect lung function through enhanced airway remodeling in response to injury, lower airway inflammation, and reduced airway bacterial colonization [68].

Patients with CF are more likely to develop lung infections, frequently requiring intravenous antibiotic therapy hospitalization. The innate immune system can be strengthened by vitamin D by upregulating antimicrobial peptides like human cathelicidin (hCAP18 or its cleaved protein LL-37) [12]. Toll-like receptors on alveolar macrophages can bind to invasive bacteria, which causes an upregulation of the 1-hydroxylase and increased production of the 1,25(OH)2D and the vitamin D receptor (VDR) [73]. To eradicate the infection caused by the invasive bacteria, the locally generated 1,25(OH)2D can promote the expression of cathelicidin by macrophages and monocytes [12, 73]. In CF, locally produced 1,25(OH)2D may increase LL-37 airway concentrations, preventing bacteria like Pseudomonas aeruginosa and Bordetella bronchiseptica from colonizing the airways [53]. Additionally, vitamin D can inhibit the production of pro-inflammatory cytokines by macrophages, which may potentially lessen inflammation in the CF airway [74]. Moreover, vitamin D may have favorable effects on the development of reactive nitrogen and oxygen intermediates and the stimulation of autophagy to assist in removing infections [75].

By the time they reach adulthood, up to half of those with cystic fibrosis (CF) also have diabetes. The main cause of CF-related diabetes (CFRD) is pancreatic dysfunction, which results in inadequate insulin release and/or insulin resistance. Peng et al. found that CF adults with vitamin D deficiency are at an increased risk of developing CFRD and its earlier onset. They found that maintaining serum levels of 25(OH)D above 20 ng/mL may diminish the risk of CF progression to CFRD [76].

It is nevertheless frequent for CF patients to experience pulmonary exacerbations, which are linked to higher mortality and deterioration of lung function [77]. Previous intravenous (IV) antibiotic therapy within the last year, the length of prior IV therapy, previous hospitalization, usage of inhaled aminoglycosides, leukotriene modifiers, and high-dose ibuprofen are all factors linked to a CF pulmonary exacerbation [78]. In both adults and children with CF, nutritional issues like a lack of vitamins A, E, and D have been linked to an increased risk of pulmonary exacerbation [15, 72, 79]. Early research revealed that vitamin D may have a positive effect on lung function, inflammatory markers, and innate immunity [14,15,16, 54]. In previous studies, oral treatment of 250,000 IU vitamin D3 increased 1-year survival and reduced blood levels of the pro-inflammatory cytokines, (tumor necrosis factor) TNF, and IL-6, with trends toward improvements in hospital-free days and lung function (FEV1%) as compared to placebo [14, 16]. However, despite the mentioned complications, a meta-analysis by Juhasz et al. on randomized controlled trials (RCTs) showed that CF patients receiving vitamin D supplementation compared with those who did not get supplementation had no significant difference in the incidence of progressive outcomes [80].

The limitation of our results includes: the low limited number of eligible studies in our meta-analysis. We could not consider confounding factors, including ethnicity and season of vitamin D levels measurement. Another limitation was the high heterogeneity among the included cross-sectional studies.

In conclusion, our results show that CF patients have lower vitamin D levels compared to healthy controls during childhood/adolescence and adulthood. Vitamin D deficiency in these patients can potentially lead to several complications; therefore, following the guidelines on serum vitamin D levels is critical. Further research is required to determine the prevalence of vitamin D deficiency among CF patients and to improve on the current treatment regimens to maintain serum targets and prevent potential complications.

Availability of data and materials

Data will not be made available in a public repository as we have not obtained ethical clearance to share data publicly. However, on request from corresponding author data could be provided while maintaining anonymity.

References

  1. Elborn JS. Cystic fibrosis. Lancet. 2016;388(10059):2519–31.

    Article  CAS  PubMed  Google Scholar 

  2. Waters V, Stanojevic S, Atenafu EG, Lu A, Yau Y, Tullis E, et al. Effect of pulmonary exacerbations on long-term lung function decline in cystic fibrosis. Eur Respir J. 2012;40(1):61–6.

    Article  PubMed  Google Scholar 

  3. Ferkol T, Rosenfeld M, Milla CE. Cystic fibrosis pulmonary exacerbations. J Pediatr. 2006;148(2):259–64.

    Article  PubMed  Google Scholar 

  4. Goss CH, Burns JL. Exacerbations in cystic fibrosis. 1: Epidemiology and pathogenesis. Thorax. 2007;62(4):360–7.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Rovner AJ, Stallings VA, Schall JI, Leonard MB, Zemel BS. Vitamin D insufficiency in children, adolescents, and young adults with cystic fibrosis despite routine oral supplementation. Am J Clin Nutr. 2007;86(6):1694–9.

    Article  CAS  PubMed  Google Scholar 

  6. Wolfenden LL, Judd SE, Shah R, Sanyal R, Ziegler TR, Tangpricha V. Vitamin D and bone health in adults with cystic fibrosis. Clin Endocrinol (Oxf). 2008;69(3):374–81.

    Article  CAS  PubMed  Google Scholar 

  7. Siwamogsatham O, Alvarez JA, Tangpricha V. Diagnosis and treatment of endocrine comorbidities in patients with cystic fibrosis. Curr Opin Endocrinol Diabetes Obes. 2014;21(5):422–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tangpricha V, Kelly A, Stephenson A, Maguiness K, Enders J, Robinson KA, et al. An update on the screening, diagnosis, management, and treatment of vitamin D deficiency in individuals with cystic fibrosis: evidence-based recommendations from the cystic fibrosis Foundation. J Clin Endocrinol Metab. 2012;97(4):1082–93.

    Article  CAS  PubMed  Google Scholar 

  9. Chesdachai S, Tangpricha V. Treatment of vitamin D deficiency in cystic fibrosis. J Steroid Biochem Mol Biol. 2016;164:36–9.

    Article  CAS  PubMed  Google Scholar 

  10. Martineau AR, Jolliffe DA, Hooper RL, Greenberg L, Aloia JF, Bergman P, et al. Vitamin D supplementation to prevent acute respiratory tract infections: systematic review and meta-analysis of individual participant data. BMJ. 2017;356:i6583.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Quraishi SA, De Pascale G, Needleman JS, Nakazawa H, Kaneki M, Bajwa EK, et al. Effect of cholecalciferol supplementation on vitamin D status and cathelicidin levels in sepsis: a randomized, placebo-controlled trial. Crit Care Med. 2015;43(9):1928–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Liu PT, Stenger S, Li H, Wenzel L, Tan BH, Krutzik SR, et al. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science. 2006;311(5768):1770–3.

    Article  CAS  PubMed  Google Scholar 

  13. Ramos-Martínez E, López-Vancell MR, Fernández de Córdova-Aguirre JC, Rojas-Serrano J, Chavarría A, Velasco-Medina A, et al. Reduction of respiratory infections in asthma patients supplemented with vitamin D is related to increased serum IL-10 and IFNγ levels and cathelicidin expression. Cytokine. 2018;108:239–46.

    Article  PubMed  Google Scholar 

  14. Grossmann RE, Zughaier SM, Kumari M, Seydafkan S, Lyles RH, Liu S, et al. Pilot study of vitamin D supplementation in adults with cystic fibrosis pulmonary exacerbation: A randomized, controlled trial. Dermatoendocrinol. 2012;4(2):191–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Pincikova T, Paquin-Proulx D, Sandberg JK, Flodström-Tullberg M, Hjelte L. Clinical impact of vitamin D treatment in cystic fibrosis: a pilot randomized, controlled trial. Eur J Clin Nutr. 2017;71(2):203–5.

    Article  CAS  PubMed  Google Scholar 

  16. Grossmann RE, Zughaier SM, Liu S, Lyles RH, Tangpricha V. Impact of vitamin D supplementation on markers of inflammation in adults with cystic fibrosis hospitalized for a pulmonary exacerbation. Eur J Clin Nutr. 2012;66(9):1072–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Stang A. Critical evaluation of the Newcastle–Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur J Epidemiol. 2010;25(9):603–5.

    Article  PubMed  Google Scholar 

  18. Borenstein M, Hedges LV, Higgins JP, Rothstein HR. Introduction to meta-analysis. New York: Wiley; 2021.

    Book  Google Scholar 

  19. Huedo-Medina TB, Sánchez-Meca J, Marín-Martínez F, Botella J. Assessing heterogeneity in meta-analysis: Q statistic or I2 index? Psychol Methods. 2006;11(2):193.

    Article  PubMed  Google Scholar 

  20. Egger M, Smith GD, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ. 1997;315(7109):629–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ambroszkiewicz J, Sands D, Gajewska J, Chelchowska M, Laskowska-Klita T. Bone turnover markers, osteoprotegerin and RANKL cytokines in children with cystic fibrosis. Adv Med Sci. 2013;58(2):338–43.

    Article  CAS  PubMed  Google Scholar 

  22. Buntain H, Greer RM, Schluter P, Wong J, Batch J, Potter J, et al. Bone mineral density in Australian children, adolescents and adults with cystic fibrosis: a controlled cross sectional study. Thorax. 2004;59(2):149–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Greer RM, Buntain HM, Potter JM, Wainwright CE, Wong JC, O’Rourke PK, et al. Abnormalities of the PTH-vitamin D axis and bone turnover markers in children, adolescents and adults with cystic fibrosis: comparison with healthy controls. Osteoporos Int. 2003;14(5):404–11.

    Article  CAS  PubMed  Google Scholar 

  24. Jakovska T. Analysis of bone metabolism in children with cystic fibrosis. Prilozi. 2018;39(1):151–5.

    Article  PubMed  Google Scholar 

  25. Jakovska-Maretti T, Meceska-Jovcevska J, Fustik S, Spirevska L. Bone turnover markers and vitamin D status in patients with cystic fibrosis. Eur Respiratory Soc; 2013.

  26. Solomons NW, Wagonfeld JB, Rieger C, Jacob RA, Bolt M, Horst J, et al. Some biochemical indices of nutrition in treated cystic fibrosis patients. Am J Clin Nutr. 1981;34(4):462–74.

    Article  CAS  PubMed  Google Scholar 

  27. Stead R, Houlder S, Agnew J, Thomas M, Hodson M, Batten J, et al. Vitamin D and parathyroid hormone and bone mineralisation in adults with cystic fibrosis. Thorax. 1988;43(3):190–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Thursfield RM, Naderi K, Leaver N, Rosenthal M, Alton EW, Bush A, et al. Children with cystic fibrosis demonstrate no respiratory immunological, infective or physiological, consequences of vitamin D deficiency. J Cyst Fibros. 2018;17(5):657–65.

    Article  PubMed  Google Scholar 

  29. Aziz DA, Fatima SK, Iftikhar H, Mir F. Vitamin D status and pulmonary exacerbations in children and adolescents with cystic fibrosis: experience from a tertiary care center. Lung India: Off Organ Indian Chest Soc. 2021;38(4):326.

    Article  Google Scholar 

  30. Bellini SG, Chapman P, Szendre K, McDonald C, Williams N, Hopkin L, et al. Changes in handgrip strength in children with cystic fibrosis compared to children without cystic fibrosis. Clin Nutr ESPEN. 2021;42:206–11.

    Article  PubMed  Google Scholar 

  31. Cemlyn-Jones J, Gamboa F, Loureiro M, Baganha MF. Evaluation of bone mineral density in cystic fibrosis patients. Rev Port Pneumol (Engl Edn). 2008;14(5):625–34.

    Article  Google Scholar 

  32. Chirita-Emandi A, Shepherd S, Kyriakou A, McNeilly JD, Dryden C, Corrigan D, et al. A retrospective analysis of longitudinal changes in bone mineral content in cystic fibrosis. J Pediatr Endocrinol Metab. 2017;30(8):807–14.

    Article  CAS  PubMed  Google Scholar 

  33. Ciuca IM, Pop LL, Dediu M, Tanasescu SA, Ardelean F, Iovanescu G, et al. Vitamin D (25-OH-cholecalciferol) in cystic fibrosis and the relations with cholesterol and proteins. Rev Chim. 2019;70:3185–7.

    Article  CAS  Google Scholar 

  34. Coriati A, Dubois CL, Phaneuf M, Mailhot M, Lavoie A, Berthiaume Y, et al. Relationship between vitamin D levels and glucose tolerance in an adult population with cystic fibrosis. Diabetes Metab. 2016;42(2):135–8.

    Article  CAS  PubMed  Google Scholar 

  35. Elkin S, Fairney A, Burnett S, Kemp M, Kyd P, Burgess J, et al. Vertebral deformities and low bone mineral density in adults with cystic fibrosis: a cross-sectional study. Osteoporos Int. 2001;12(5):366–72.

    Article  CAS  PubMed  Google Scholar 

  36. Gordon CM, Anderson EJ, Herlyn K, Hubbard JL, Pizzo A, Gelbard R, et al. Nutrient status of adults with cystic fibrosis. J Am Diet Assoc. 2007;107(12):2114–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Grey V, Atkinson S, Drury D, Casey L, Ferland G, Gundberg C, et al. Prevalence of low bone mass and deficiencies of vitamins D and K in pediatric patients with cystic fibrosis from 3 Canadian centers. Pediatrics. 2008;122(5):1014–20.

    Article  PubMed  Google Scholar 

  38. Gupta S, Mukherjee A, Khadgawat R, Kabra M, Lodha R, Kabra SK. Bone mineral density of Indian children and adolescents with cystic fibrosis. Indian Pediatr. 2017;54(7):545–9.

    Article  PubMed  Google Scholar 

  39. Haworth C, Selby P, Webb A, Dodd M, Musson H, Niven RM, et al. Low bone mineral density in adults with cystic fibrosis. Thorax. 1999;54(11):961–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. McCauley LA, Thomas W, Laguna TA, Regelmann WE, Moran A, Polgreen LE. Vitamin D deficiency is associated with pulmonary exacerbations in children with cystic fibrosis. Ann Am Thorac Soc. 2014;11(2):198–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Daley T, Hughan K, Rayas M, Kelly A, Tangpricha V. Vitamin D deficiency and its treatment in cystic fibrosis. J Cyst Fibros. 2019;18(Suppl 2):S66–73.

    Article  CAS  PubMed  Google Scholar 

  42. Holick MF, Binkley NC, Bischoff-Ferrari HA, Gordon CM, Hanley DA, Heaney RP, et al. Evaluation, treatment, and prevention of vitamin D deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2011;96(7):1911–30.

    Article  CAS  PubMed  Google Scholar 

  43. Wood C, Hasan S, Darukhanavala A, Tangpricha V. A Clinician’s guide to vitamin D supplementation for patients with cystic fibrosis. J Clin Transl Endocrinol. 2021;26:100273.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Robberecht E, Vandewalle S, Wehlou C, Kaufman JM, De Schepper J. Sunlight is an important determinant of vitamin D serum concentrations in cystic fibrosis. Eur J Clin Nutr. 2011;65(5):574–9.

    Article  CAS  PubMed  Google Scholar 

  45. Lark RK, Lester GE, Ontjes DA, Blackwood AD, Hollis BW, Hensler MM, et al. Diminished and erratic absorption of ergocalciferol in adult cystic fibrosis patients. Am J Clin Nutr. 2001;73(3):602–6.

    Article  CAS  PubMed  Google Scholar 

  46. Bhimavarapu A, Deng Q, Bean M, Lee N, Ziegler TR, Alvarez J, et al. Factors contributing to vitamin D status at hospital admission for pulmonary exacerbation in adults with cystic fibrosis. Am J Med Sci. 2021;361(1):75–82.

    Article  PubMed  Google Scholar 

  47. Hall WB, Sparks AA, Aris RM. Vitamin D deficiency in cystic fibrosis. Int J Endocrinol. 2010;2010:218691.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Hahn TJ, Squires AE, Halstead LR, Strominger DB. Reduced serum 25-hydroxyvitamin D concentration and disordered mineral metabolism in patients with cystic fibrosis. J Pediatr. 1979;94(1):38–42.

    Article  CAS  PubMed  Google Scholar 

  49. Coppenhaver D, Kueppers F, Schidlow D, Bee D, Isenburg JN, Barnett DR, et al. Serum concentrations of vitamin D-binding protein (group-specific component) in cystic fibrosis. Hum Genet. 1981;57(4):399–403.

    Article  CAS  PubMed  Google Scholar 

  50. Naik AL, Rajan MG, Manjrekar PA, Shenoy MT, Shreelata S, Srikantiah RM, et al. Effect of DOTS treatment on Vitamin D levels in pulmonary tuberculosis. J Clin Diagn Res. 2017;11(4):Bc18-bc22.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Khammissa RAG, Fourie J, Motswaledi MH, Ballyram R, Lemmer J, Feller L. The biological activities of vitamin D and its receptor in relation to calcium and bone homeostasis, cancer, immune and cardiovascular systems, skin biology, and oral health. Biomed Res Int. 2018;2018:9276380.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Adams JS, Ren S, Liu PT, Chun RF, Lagishetty V, Gombart AF, et al. Vitamin d-directed rheostatic regulation of monocyte antibacterial responses. J Immunol. 2009;182(7):4289–95.

    Article  CAS  PubMed  Google Scholar 

  53. Yim S, Dhawan P, Ragunath C, Christakos S, Diamond G. Induction of cathelicidin in normal and CF bronchial epithelial cells by 1,25-dihydroxyvitamin D(3). J Cyst Fibros. 2007;6(6):403–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Schögler A, Muster RJ, Kieninger E, Casaulta C, Tapparel C, Jung A, et al. Vitamin D represses rhinovirus replication in cystic fibrosis cells by inducing LL-37. Eur Respir J. 2016;47(2):520–30.

    Article  PubMed  Google Scholar 

  55. Chesdachai S, Zughaier SM, Hao L, Kempker RR, Blumberg HM, Ziegler TR, et al. The effects of first-line anti-tuberculosis drugs on the actions of vitamin D in human macrophages. J Clin Transl Endocrinol. 2016;6:23–9.

    PubMed  PubMed Central  Google Scholar 

  56. Zughaier SM, Svoboda P, Pohl J, Stephens DS, Shafer WM. The human host defense peptide LL-37 interacts with Neisseria meningitidis capsular polysaccharides and inhibits inflammatory mediators release. PLoS ONE. 2010;5(10):e13627.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Nagaoka I, Hirota S, Niyonsaba F, Hirata M, Adachi Y, Tamura H, et al. Augmentation of the lipopolysaccharide-neutralizing activities of human cathelicidin CAP18/LL-37-derived antimicrobial peptides by replacement with hydrophobic and cationic amino acid residues. Clin Diagn Lab Immunol. 2002;9(5):972–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Bergsson G, Reeves EP, McNally P, Chotirmall SH, Greene CM, Greally P, et al. LL-37 complexation with glycosaminoglycans in cystic fibrosis lungs inhibits antimicrobial activity, which can be restored by hypertonic saline. J Immunol. 2009;183(1):543–51.

    Article  CAS  PubMed  Google Scholar 

  59. Neumann A, Völlger L, Berends ET, Molhoek EM, Stapels DA, Midon M, et al. Novel role of the antimicrobial peptide LL-37 in the protection of neutrophil extracellular traps against degradation by bacterial nucleases. J Innate Immun. 2014;6(6):860–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Sanders DB, Solomon GM, Beckett VV, West NE, Daines CL, Heltshe SL, et al. Standardized Treatment of Pulmonary Exacerbations (STOP) study: observations at the initiation of intravenous antibiotics for cystic fibrosis pulmonary exacerbations. J Cyst Fibros. 2017;16(5):592–9.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Waters VJ, Stanojevic S, Sonneveld N, Klingel M, Grasemann H, Yau YC, et al. Factors associated with response to treatment of pulmonary exacerbations in cystic fibrosis patients. J Cyst Fibros. 2015;14(6):755–62.

    Article  CAS  PubMed  Google Scholar 

  62. Sanders DB, Bittner RC, Rosenfeld M, Hoffman LR, Redding GJ, Goss CH. Failure to recover to baseline pulmonary function after cystic fibrosis pulmonary exacerbation. Am J Respir Crit Care Med. 2010;182(5):627–32.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Holick MF. Vitamin D deficiency. N Engl J Med. 2007;357(3):266–81.

    Article  CAS  PubMed  Google Scholar 

  64. Haworth CS. Impact of cystic fibrosis on bone health. Curr Opin Pulm Med. 2010;16(6):616–22.

    CAS  PubMed  Google Scholar 

  65. Khazai N, Judd SE, Tangpricha V. Calcium and vitamin D: skeletal and extraskeletal health. Curr Rheumatol Rep. 2008;10(2):110–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Snijder M, van Dam R, Visser M, Deeg D, Seidell J, Lips P. To: Mathieu C, Gysemans C, Giulietti A, Bouillon R (2005) Vitamin D and diabetes. Diabetologia 48:1247-1257. Diabetologia. 2006;49(1):217–8.

    Article  CAS  PubMed  Google Scholar 

  67. Tangpricha V, Spina C, Yao M, Chen TC, Wolfe MM, Holick MF. Vitamin D deficiency enhances the growth of MC-26 colon cancer xenografts in Balb/c mice. J Nutr. 2005;135(10):2350–4.

    Article  CAS  PubMed  Google Scholar 

  68. Finklea JD, Grossmann RE, Tangpricha V. Vitamin D and chronic lung disease: a review of molecular mechanisms and clinical studies. Adv Nutr. 2011;2(3):244–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Black PN, Scragg R. Relationship between serum 25-hydroxyvitamin d and pulmonary function in the third national health and nutrition examination survey. Chest. 2005;128(6):3792–8.

    Article  CAS  PubMed  Google Scholar 

  70. Vanstone MB, Egan ME, Zhang JH, Carpenter TO. Association between serum 25-hydroxyvitamin D level and pulmonary exacerbations in cystic fibrosis. Pediatr Pulmonol. 2015;50(5):441–6.

    Article  PubMed  Google Scholar 

  71. Stephenson A, Brotherwood M, Robert R, Atenafu E, Corey M, Tullis E. Cholecalciferol significantly increases 25-hydroxyvitamin D concentrations in adults with cystic fibrosis. Am J Clin Nutr. 2007;85(5):1307–11.

    Article  CAS  PubMed  Google Scholar 

  72. Sexauer WP, Hadeh A, Ohman-Strickland PA, Zanni RL, Varlotta L, Holsclaw D, et al. Vitamin D deficiency is associated with pulmonary dysfunction in cystic fibrosis. J Cyst Fibros. 2015;14(4):497–506.

    Article  CAS  PubMed  Google Scholar 

  73. Hewison M. Vitamin D and the intracrinology of innate immunity. Mol Cell Endocrinol. 2010;321(2):103–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Herscovitch K, Dauletbaev N, Lands LC. Vitamin D as an anti-microbial and anti-inflammatory therapy for cystic fibrosis. Paediatr Respir Rev. 2014;15(2):154–62.

    CAS  PubMed  Google Scholar 

  75. Korf H, Decallonne B, Mathieu C. Vitamin D for infections. Curr Opin Endocrinol Diabetes Obes. 2014;21(6):431–6.

    Article  CAS  PubMed  Google Scholar 

  76. Peng Y, Wu M, Alvarez JA, Tangpricha V. Vitamin D status and risk of cystic fibrosis-related diabetes: a retrospective single center cohort study. Nutrients. 2021;13(11):4048.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Justicia JL, Solé A, Quintana-Gallego E, Gartner S, de Gracia J, Prados C, et al. Management of pulmonary exacerbations in cystic fibrosis: still an unmet medical need in clinical practice. Expert Rev Respir Med. 2015;9(2):183–94.

    Article  CAS  PubMed  Google Scholar 

  78. VanDevanter DR, Pasta DJ, Konstan MW. Treatment and demographic factors affecting time to next pulmonary exacerbation in cystic fibrosis. J Cyst Fibros. 2015;14(6):763–9.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Hakim F, Kerem E, Rivlin J, Bentur L, Stankiewicz H, Bdolach-Abram T, et al. Vitamins A and E and pulmonary exacerbations in patients with cystic fibrosis. J Pediatr Gastroenterol Nutr. 2007;45(3):347–53.

    Article  CAS  PubMed  Google Scholar 

  80. Juhász MF, Varannai O, Németh D, Szakács Z, Kiss S, Izsák VD, et al. Vitamin D supplementation in patients with cystic fibrosis: a systematic review and meta-analysis. J Cyst Fibros. 2021;20(5):729–36.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We express our gratitude to the participants of this study.

Funding

No funding.

Author information

Authors and Affiliations

Authors

Contributions

NF and AhH contributed to the conception, design, and statistical analysis. SF, ASh, MsM, MM, SAT, GK, ShY, and SZD contributed to data collection and manuscript draft. AhH supervised the study. All authors contributed to the manuscript draft and critical revision. All authors approved the final version of the manuscript.

Corresponding author

Correspondence to Amir hossein Hosseini.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

We, the authors, declare that we had no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Fig. S1.

The leave-one-out method on the pooled effect size for A) case control and , B) cross-sectional studies. Fig. S2. Funnel plot of the weighted mean difference (WMD) versus the standard error (s.e) for A) case control and , B)cross-sectional studies.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Farahbakhsh, N., Fatahi, S., Shirvani, A. et al. Vitamin D deficiency in patients with cystic fibrosis: a systematic review and meta-analysis. J Health Popul Nutr 43, 11 (2024). https://doi.org/10.1186/s41043-024-00499-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41043-024-00499-2

Keywords