Effect of n-3 PUFA on extracellular matrix protein turnover in patients with psoriatic arthritis: a randomized, double-blind, placebo-controlled trial

Signe Holm Nielsen, Samra Sardar, Anne Sofie Siebuhr, Annette Schlemmer, Erik Berg Schmidt, Anne-Christine Bay-Jensen, Morten A Karsdal, Jeppe Hagstrup Christensen, Salome Kristensen, Signe Holm Nielsen, Samra Sardar, Anne Sofie Siebuhr, Annette Schlemmer, Erik Berg Schmidt, Anne-Christine Bay-Jensen, Morten A Karsdal, Jeppe Hagstrup Christensen, Salome Kristensen

Abstract

Psoriatic arthritis (PsA) is a chronic inflammatory disease characterized by involvement of skin, axial and peripheral skeleton. An altered balance between extracellular matrix (ECM) formation and breakdown is a key event in PsA, and changes in ECM protein metabolites may provide insight to tissue changes. Dietary fish oils (n-3 PUFA) might affect the inflammation driven tissue turnover. The aim was to evaluate ECM metabolites in patients with PsA compared to healthy individuals and investigate the effects of n-3 PUFA. The 24-week randomized, double-blind, placebo-controlled trial of PUFA included 142 patients with PsA. Fifty-seven healthy individuals were included for comparison. This study is a sub-study investigating biomarkers of tissue remodelling as secondary outcomes. Serum samples at baseline and 24 weeks and healthy individuals were obtained, while a panel of ECM metabolites reflecting bone and soft tissue turnover were measured by ELISAs: PRO-C1, PRO-C3, PRO-C4, C1M, C3M, C4M, CTX-I and Osteocalcin (OC). C1M, PRO-C3, PRO-C4 and C4M was found to be elevated in PsA patients compared to the healthy individuals (from 56 to 792%, all p < 0.0001), where no differences were found for OC, CTX-I, PRO-C1 and C3M. PRO-C3 was increased by 7% in patients receiving n-3 PUFA after 24 weeks compared to baseline levels (p = 0.002). None of the other biomarkers was changed with n-3 PUFA treatment. This indicates that tissue turnover is increased in PsA patients compared to healthy individuals, while n-3 PUFA treatment for 24 weeks did not have an effect on tissue turnover. Trial registration NCT01818804. Registered 27 March 2013-Completed 18 February 2016. https://ichgcp.net/clinical-trials-registry/NCT01818804?term=NCT01818804&rank=1.

Keywords: Biomarkers; Collagens; Fish oil; Psoriatic arthritis.

Conflict of interest statement

SHN, ASS, ACBJ and MAK are full-time employees at Nordic Bioscience A/S. Nordic Bioscience is a privately-owned, small–medium size enterprise (SME) partly focused on the development of biomarkers. None of the authors received fees, bonuses or other benefits for the work described in the manuscript. MAK and ACBJ hold stocks in Nordic Bioscience A/S.

Figures

Fig. 1
Fig. 1
Flowchart of the trial
Fig. 2
Fig. 2
Levels of collagen formation and degradation biomarkers in serum from healthy controls (n = 57) and patients with PsA (n = 142). a. Serum levels of formation of collagen type I (PRO-C1), b. Serum levels of degradation of collagen type I (C1M), c. Serum levels of formation of collagen type III (PRO-C3), d. Serum levels of degradation of collagen type III, e. Serum levels of formation of collagen type VI (PRO-C4), b. Serum levels of degradation of collagen type IV (C4M). Statistical differences between the healthy controls and PsA patients were calculated using an unpaired t test for PRO-C3, and LOG10 transformed PRO-C1, C1M, C3M, PRO-C4 and C4M. Significance threshold was set at p < 0.05 and data is presented as Tukey boxplots. Significance levels: ****p < 0.0001
Fig. 3
Fig. 3
Levels of bone formation and degradation biomarkers in serum from healthy controls (n = 6) and patients with PsA (n = 142). a. Serum levels of bone formation measured by osteocalcin (OC) and b. Serum levels of bone degradation measured by CTX-I. Statistical differences between the healthy controls and PsA patients were calculated using an unpaired t test for OC, and LOG10 transformed CTX-I. Data is presented as Tukey boxplots
Fig. 4
Fig. 4
Biomarker levels in placebo (n = 57) and n-3 PUFA (n = 142) treated patients at baseline and 24 weeks. a Serum levels of the bone formation biomarker osteocalcin (OC), b Serum levels of the bone degradation biomarker CTX-I, c Serum levels of the collagen type I biomarker, PRO-C1, d Serum levels of the collagen type III formation biomarker, PRO-C3, e Serum levels of the collagen type IV formation biomarker, PRO-C4, f Serum levels of the collagen type I degradation biomarker, C1M, g Serum levels of the collagen type III degradation biomarker C3M, and h Serum levels of the collagen type IV degradation biomarker, C4M. Statistical differences between the baseline and 24 weeks of follow-up were calculated using a paired t test for OC and PRO-C3, and LOG10 transformed CTX-I PRO-C1, C1M, C3M, PRO-C4 and C4M. Significance threshold was set at p < 0.05 and data is presented as Tukey boxplots. Significance levels: **p < 0.01

References

    1. Mallbris L, Ritchlin CT, Ståhle M. Metabolic disorders in patients with psoriasis and psoriatic arthritis. Curr Rheumatol Rep. 2006;8:355–363. doi: 10.1007/s11926-006-0065-8.
    1. Veale DJ, Fearon U. The pathogenesis of psoriatic arthritis. Lancet. 2018;391:2273–2284. doi: 10.1016/S0140-6736(18)30830-4.
    1. Schett G, Coates LC, Ash ZR, et al. Structural damage in rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis: traditional views, novel insights gained from TNF blockade, and concepts for the future. Arthritis Res Ther. 2011 doi: 10.1186/1478-6354-13-S1-S4.
    1. Bartosińska J, Michalak-Stoma A, Juszkiewicz-Borowiec M, et al. The assessment of selected bone and cartilage biomarkers in psoriatic patients from Poland. Mediators Inflamm. 2015 doi: 10.1155/2015/194535.
    1. Karsdal MA, Nielsen MJ, Sand JM, et al. Extracellular matrix remodeling: the common denominator in connective tissue diseases possibilities for evaluation and current understanding of the matrix as more than a passive architecture, but a key player in tissue failure. Assay Drug Dev Technol. 2013;11:70–92. doi: 10.1089/adt.2012.474.
    1. Adair-Kirk TL, Senior RM. Fragments of extracellular matrix as mediators of inflammation. Int J Biochem Cell Biol. 2008;40:1101–1110. doi: 10.1016/j.biocel.2007.12.005.
    1. Moz S, Aita A, Basso D, et al. Spondyloarthritis: matrix metalloproteinasesas biomarkers of pathogenesis and response to tumor necrosis factor (TNF) inhibitors. Int J Mol Sci. 2017;18:830. doi: 10.3390/ijms18040830.
    1. Clore JN, Cohen IK, Diegelmann RF. Quantitation of collagen types I and III during wound healing in rat skin. Proc Soc Exp Biol Med. 1979;161:337–340. doi: 10.3181/00379727-161-40548.
    1. Volk SW, Wang Y, Mauldin EA, et al. Diminished type III collagen promotes myofibroblast differentiation and increases scar deposition in cutaneous wound healing. Cells Tissues Organs. 2011;194:25–37. doi: 10.1159/000322399.
    1. Poschl E, Schotzer-Schrehardt U, Brachvogel B, et al. Collagen IV is essential for basement membrane stability but dispensable for initiation of its assembly during early development. Development. 2004;131:1619–1628. doi: 10.1242/dev.01037.
    1. Christgau S, Bitsch-Jensen O, Hanover Bjarnason N, et al. Serum CrossLaps for monitoring the response in individuals undergoing antiresorptive therapy. Bone. 2000;26:505–511. doi: 10.1016/S8756-3282(00)00248-9.
    1. Baim S, Miller PD. Assessing the clinical utility of serum CTX in postmenopausal osteoporosis and its use in predicting risk of osteonecrosis of the jaw. J Bone Miner Res. 2009;24:561–574. doi: 10.1359/jbmr.090203.
    1. Szulc P. Bone turnover: Biology and assessment tools. Best Pract Res Clin Endocrinol Metab. 2018;32:725–738. doi: 10.1016/j.beem.2018.05.003.
    1. Chavassieux P, Portero-Muzy N, Roux JP, et al. Are biochemical markers of bone turnover representative of bone histomorphometry in 370 postmenopausal women? J Clin Endocrinol Metab. 2015;100:4662–4668. doi: 10.1210/jc.2015-2957.
    1. Jadon DR, Nightingale AL, McHugh NJ, et al. Serum soluble bone turnover biomarkers in psoriatic arthritis and psoriatic spondyloarthropathy. J Rheumatol. 2015;42:21–30. doi: 10.3899/jrheum.140223.
    1. D’Angelo S, Tramontano G, Gilio M, et al. Review of the treatment of psoriatic arthritis with biological agents: choice of drug for initial therapy and switch therapy for non-responders. Open Access Rheumatol Res Rev. 2017;9:21–28. doi: 10.2147/OARRR.S56073.
    1. Glintborg B, Ãstergaard M, Dreyer L, et al. Treatment response, drug survival, and predictors thereof in 764 patients with psoriatic arthritis treated with anti-tumor necrosis factor α therapy: results from the nationwide Danish DANBIO registry. Arthritis Rheum. 2011;63:382–390. doi: 10.1002/art.30117.
    1. Felquer MLA, Coates LC, Soriano ER, et al. Drug therapies for peripheral joint disease in psoriatic arthritis: a systematic review. J Rheumatol. 2014;41:2277–2285. doi: 10.3899/jrheum.140876.
    1. Calder PC. n-3 Polyunsaturated fatty acids, inflammation, and inflammatory diseases. Am J Clin Nutr. 2006;83:1505S–1519S. doi: 10.1093/ajcn/83.6.1505S.
    1. Calder PC. Omega-3 fatty acids and inflammatory processes: from molecules to man. Biochem Soc Trans. 2017;45:1105–1115. doi: 10.1111/j.1365-2125.2012.04374.x.
    1. Veselinovic M, Vasiljevic D, Vucic V, et al. Clinical benefits of n-3 PUFA and ɤ-linolenic acid in patients with rheumatoid arthritis. Nutrients. 2017 doi: 10.3390/nu9040325.
    1. Gioxari A, Kaliora AC, Marantidou F, Panagiotakos DP. Intake of ω-3 polyunsaturated fatty acids in patients with rheumatoid arthritis: a systematic review and meta-analysis. Nutrition. 2018;45:114–124.e4. doi: 10.1016/j.nut.2017.06.023.
    1. De Caterina R. N-3 fatty acids in cardiovascular disease. N Engl J Med. 2011;364:2439–2450. doi: 10.1056/NEJMra1008153.
    1. Michael-Titus AT. Omega-3 fatty acids and neurological injury. Prostaglandins Leukot Essent Fat Acids. 2007;77:295–300. doi: 10.1016/j.plefa.2007.10.021.
    1. Decker MJ, Jones K, Keating GL, et al. Maternal dietary supplementation with omega-3 polyunsaturated fatty acids confers neuroprotection to the newborn against hypoxia-induced dopamine dysfunction. Sleep Sci. 2016;9:94–99. doi: 10.1016/j.slsci.2016.05.007.
    1. Devassy JG, Leng S, Gabbs M, et al. Omega-3 polyunsaturated fatty acids and oxylipins in neuroinflammation and management of Alzheimer’s disease. Adv Nutr. 2016;7:905–916. doi: 10.3945/an.116.012187.of.
    1. Wann AKT, Mistry J, Blain EJ, et al. Eicosapentaenoic acid and docosahexaenoic acid reduce interleukin-1β-mediated cartilage degradation. Arthritis Res Ther. 2010 doi: 10.1186/ar3183.
    1. Knott L, Avery NC, Hollander AP, Tarlton JF. Regulation of osteoarthritis by omega-3 (n-3) polyunsaturated fatty acids in a naturally occurring model of disease. Osteoarthr Cartil. 2011;19:1150–1157. doi: 10.1016/j.joca.2011.06.005.
    1. Kristensen S, Schmidt EB, Schlemmer A, et al. Beneficial effect of n-3 polyunsaturated fatty acids on inflammation and analgesic use in psoriatic arthritis: a randomized, double blind, placebo-controlled trial. Scand J Rheumatol. 2018;47:27–36. doi: 10.1080/03009742.2017.1287304.
    1. Watkins BA, Li Y, Allen KG, et al. Dietary ratio of (n-6)/(n-3) polyunsaturated fatty acids alters the fatty acid composition of bone compartments and biomarkers of bone formation in rats. J Nutr. 2000;130:2274–2284. doi: 10.1016/j.jnutbio.2005.05.012.
    1. Watkins BA, Li Y, Lippman HE, Feng S. Modulatory effect of omega-3 polyunsaturated fatty acids on osteoblast function and bone metabolism. Prostaglandins Leukot Essent Fat Acids. 2003;68:387–398. doi: 10.1016/S0952-3278(03)00063-2.
    1. Yuan J, Akiyama M, Nakahama KI, et al. The effects of polyunsaturated fatty acids and their metabolites on osteoclastogenesis in vitro. Prostaglandins Other Lipid Mediat. 2010;92:85–90. doi: 10.1016/j.prostaglandins.2010.04.001.
    1. Wauquier F, Léotoing L, Philippe C, et al. Pros and cons of fatty acids in bone biology. Prog Lipid Res. 2015;58:121–145. doi: 10.1016/j.plipres.2015.03.001.
    1. Mangano KM, Sahni S, Kerstetter JE, et al. Polyunsaturated fatty acids and their relation with bone and muscle health in adults. Curr Osteoporos Rep. 2013;11:203–212. doi: 10.1007/s11914-013-0149-0.
    1. Lau BYY, Cohen DJA, Ward WE, Ma DWL. Investigating the role of polyunsaturated fatty acids in bone development using animal models. Molecules. 2013;18:14203–14227. doi: 10.3390/molecules181114203.
    1. Sharif PS, Asalforoush M, Ameri F, et al. The effect of n-3 fatty acids on bone biomarkers in Iranian postmenopausal osteoporotic women: a randomized clinical trial. Age (Omaha) 2010;32:179–186. doi: 10.1007/s11357-009-9122-3.
    1. Griel AE, Kris-Etherton PM, Hilpert KF, et al. An increase in dietary n-3 fatty acids decreases a marker of bone resorption in humans. Nutr J. 2007;6:4–11. doi: 10.1186/1475-2891-6-2.
    1. Rajaram S, Yip EL, Reghunathan R, et al. Effect of altering dietary n-6: n-3 polyunsaturated fatty acid ratio with plant and marine-based supplement on biomarkers of bone turnover in healthy adults. Nutrients. 2017 doi: 10.3390/nu9101162.
    1. Damsgaard CT, Mølgaard C, Matthiessen J, et al. The effects of n-3 long-chain polyunsaturated fatty acids on bone formation and growth factors in adolescent boys. Pediatr Res. 2012;71:713–719. doi: 10.1038/pr.2012.28.
    1. Serhan CN, Hong S, Gronert K, et al. Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. J Exp Med. 2002;196:1025–1037. doi: 10.1084/jem.20020760.
    1. Molfino A, Amabile MI, Monti M, Muscaritoli M. Omega-3 polyunsaturated fatty acids in critical illness: anti-inflammatory, proresolving, or both? Oxid Med Cell Longev. 2017;2017:1–6. doi: 10.1155/2017/5987082.
    1. Sundrarjun T, Komindr S, Archararit N, et al. Effects of n-3 fatty acids on serum interleukin-6, tumour necrosis factor-α and soluble tumour necrosis factor receptor p55 in active rheumatoid arthritis. J Int Med Res. 2004;32:443–454. doi: 10.1177/147323000403200501.
    1. Sköldstam L, Börjesson O, Kjällman A, et al. Effect of six months of fish oil supplementation in stable rheumatoid arthritis. a double-blind, controlled study. Scand J Rheumatol. 1992;21:178–185. doi: 10.3109/03009749209099218.
    1. Li K, Huang T, Zheng J, et al. Effect of marine-derived n-3 polyunsaturated fatty acids on C-reactive protein, interleukin 6 and tumor necrosis factor α: a meta-analysis. PLoS One. 2014 doi: 10.1371/journal.pone.0088103.
    1. Veale DJ, Torley HI, Richards IM, et al. A double-blind placebo controlled trial of efamol marine on skin and joint symptoms of psoriatic arthritis. Rheumatology. 1994;33:954–958. doi: 10.1093/rheumatology/33.10.954.
    1. Madland TM, Björkkjaer T, Brunborg LA, et al. Subjective improvement in patients with psoriatic arthritis after short-term oral treatment with seal oil. A pilot study with double blind comparison to soy oil. J Rheumatol. 2006;33:307–310.
    1. Lassus A, Dahlgren AL, Halpern MJ, et al. Effects of dietary supplementation with polyunsaturated ethyl ester lipids (Angiosan) in patients with psoriasis and psoriatic arthritis. J Int Med Res. 1990;18:68–73. doi: 10.1177/030006059001800109.
    1. Akhyani M, Ehsani AH, Robati RM, Robati AM. The lipid profile in psoriasis: a controlled study. J Eur Acad Dermatol Venereol. 2007;21:1330–1332. doi: 10.1111/j.1468-3083.2007.02260.x.
    1. Frommer KW, Schäffler A, Rehart S, et al. Free fatty acids: potential proinflammatory mediators in rheumatic diseases. Ann Rheum Dis. 2015;74:303–310. doi: 10.1136/annrheumdis-2013-203755.
    1. Brouwers H, Von Hegedus J, Toes R, et al. Lipid mediators of inflammation in rheumatoid arthritis and osteoarthritis. Best Pract Res Clin Rheumatol. 2015;29:741–755. doi: 10.1016/j.berh.2016.02.003.
    1. Mysliwiec H, Harasim-Symbor E, Baran A, et al. Abnormal serum fatty acid profile in psoriatic arthritis. Arch Med Sci. 2019;15:1407–1414. doi: 10.5114/aoms.2019.89451.
    1. Goldberg RJ, Katz J. A meta-analysis of the analgesic effects of omega-3 polyunsaturated fatty acid supplementation for inflammatory joint pain. Pain. 2007;129:210–223. doi: 10.1016/j.pain.2007.01.020.
    1. Kremer JM, Lawrence DA, Jubiz W, et al. Dietary fish oil and olive oil supplementation in patients with rheumatoid arthritis. Clin Immunologic Eff Arthritis Rheum. 1990;33:810–820. doi: 10.1002/art.1780330607.

Source: PubMed

3
Suscribir