Skip to main content
Top
Published in: Arthritis Research & Therapy 1/2020

01-12-2020 | Systemic Lupus Erythematosus | Review

Caution in studying and interpreting the lupus metabolome

Authors: Ting Zhang, Chandra Mohan

Published in: Arthritis Research & Therapy | Issue 1/2020

Login to get access

Abstract

Several metabolomics studies have shed substantial light on the pathophysiological pathways underlying multiple diseases including systemic lupus erythematosus (SLE). This review takes stock of our current understanding of this field. We compare, collate, and investigate the metabolites in SLE patients and healthy volunteers, as gleaned from published metabolomics studies on SLE. In the surveyed primary reports, serum or plasma samples from SLE patients and healthy controls were assayed using mass spectrometry or nuclear magnetic resonance spectroscopy, and metabolites differentiating SLE from controls were identified. Collectively, the circulating metabolome in SLE is characterized by reduced energy substrates from glycolysis, Krebs cycle, fatty acid β oxidation, and glucogenic and ketogenic amino acid metabolism; enhanced activity of the urea cycle; decreased long-chain fatty acids; increased medium-chain and free fatty acids; and augmented peroxidation and inflammation. However, these findings should be interpreted with caution because several of the same metabolic pathways are also significantly influenced by the medications commonly used in SLE patients, common co-morbidities, and other factors including smoking and diet. In particular, whereas the metabolic alterations relating to inflammation, oxidative stress, lipid peroxidation, and glutathione generation do not appear to be steroid-dependent, the other metabolic changes may in part be influenced by steroids. To conclude, metabolomics studies of SLE and other rheumatic diseases ought to factor in the potential contributions of confounders such as medications, co-morbidities, smoking, and diet.
Literature
1.
go back to reference Tsokos GC. Systemic lupus erythematosus. N Engl J Med. 2011;365:2110–21.PubMed Tsokos GC. Systemic lupus erythematosus. N Engl J Med. 2011;365:2110–21.PubMed
2.
go back to reference Assmann N, Finlay DK. Metabolic regulation of immune responses: therapeutic opportunities. J Clin Invest. 2016;126:2031–9.PubMedPubMedCentral Assmann N, Finlay DK. Metabolic regulation of immune responses: therapeutic opportunities. J Clin Invest. 2016;126:2031–9.PubMedPubMedCentral
3.
4.
go back to reference Morel L. Immunometabolism in systemic lupus erythematosus. Nat Rev Rheumatol. 2017;13:280–90.PubMed Morel L. Immunometabolism in systemic lupus erythematosus. Nat Rev Rheumatol. 2017;13:280–90.PubMed
5.
go back to reference Ouyang X, Dai Y, Wen JL, Wang LX. (1) H NMR-based metabolomic study of metabolic profiling for systemic lupus erythematosus. Lupus. 2011;20:1411–20.PubMed Ouyang X, Dai Y, Wen JL, Wang LX. (1) H NMR-based metabolomic study of metabolic profiling for systemic lupus erythematosus. Lupus. 2011;20:1411–20.PubMed
6.
go back to reference Wu T, Xie C, Han J, Ye Y, Weiel J, Li Q, et al. Metabolic disturbances associated with systemic lupus erythematosus. PLoS One. 2012;7:e37210.PubMedPubMedCentral Wu T, Xie C, Han J, Ye Y, Weiel J, Li Q, et al. Metabolic disturbances associated with systemic lupus erythematosus. PLoS One. 2012;7:e37210.PubMedPubMedCentral
7.
go back to reference Bengtsson AA, Trygg J, Wuttge DM, Sturfelt G, Theander E, Donten M, et al. Metabolic profiling of systemic lupus erythematosus and comparison with primary Sjogren’s syndrome and systemic sclerosis. PLoS One. 2016;11:e0159384.PubMedPubMedCentral Bengtsson AA, Trygg J, Wuttge DM, Sturfelt G, Theander E, Donten M, et al. Metabolic profiling of systemic lupus erythematosus and comparison with primary Sjogren’s syndrome and systemic sclerosis. PLoS One. 2016;11:e0159384.PubMedPubMedCentral
8.
go back to reference Guleria A, Pratap A, Dubey D, Rawat A, Chaurasia S, Sukesh E, et al. NMR based serum metabolomics reveals a distinctive signature in patients with lupus nephritis. Sci Rep. 2016;6:35309.PubMedPubMedCentral Guleria A, Pratap A, Dubey D, Rawat A, Chaurasia S, Sukesh E, et al. NMR based serum metabolomics reveals a distinctive signature in patients with lupus nephritis. Sci Rep. 2016;6:35309.PubMedPubMedCentral
9.
go back to reference Yan B, Huang J, Zhang C, Hu X, Gao M, Shi A, et al. Serum metabolomic profiling in patients with systemic lupus erythematosus by GC/MS. Mod Rheumatol. 2016;26:914–22.PubMed Yan B, Huang J, Zhang C, Hu X, Gao M, Shi A, et al. Serum metabolomic profiling in patients with systemic lupus erythematosus by GC/MS. Mod Rheumatol. 2016;26:914–22.PubMed
10.
go back to reference Shin TH, Kim H-A, Jung J-Y, Baek W-Y, Lee H-S, Park HJ, et al. Analysis of the free fatty acid metabolome in the plasma of patients with systemic lupus erythematosus and fever. Metabolomics. 2017;14. Shin TH, Kim H-A, Jung J-Y, Baek W-Y, Lee H-S, Park HJ, et al. Analysis of the free fatty acid metabolome in the plasma of patients with systemic lupus erythematosus and fever. Metabolomics. 2017;14.
11.
go back to reference Guleria A, Phatak S, Dubey D, Kumar S, Zanwar A, Chaurasia S, et al. NMR-based serum metabolomics reveals reprogramming of lipid dysregulation following cyclophosphamide-based induction therapy in lupus nephritis. J Proteome Res. 2018;17:2440–8.PubMed Guleria A, Phatak S, Dubey D, Kumar S, Zanwar A, Chaurasia S, et al. NMR-based serum metabolomics reveals reprogramming of lipid dysregulation following cyclophosphamide-based induction therapy in lupus nephritis. J Proteome Res. 2018;17:2440–8.PubMed
12.
go back to reference Li JXX, Zhou H, Wang B, Zhang MJ, Tang FY. Metabolic profiling reveals new serum biomarkers of lupus nephritis. Lupus. 2017;26:1166–73.PubMed Li JXX, Zhou H, Wang B, Zhang MJ, Tang FY. Metabolic profiling reveals new serum biomarkers of lupus nephritis. Lupus. 2017;26:1166–73.PubMed
13.
go back to reference Bellocchi C, Fernandez-Ochoa A, Montanelli G, Vigone B, Santaniello A, Quirantes-Pine R, et al. Identification of a shared microbiomic and metabolomic profile in systemic autoimmune diseases. J Clin Med. 2019;8. Bellocchi C, Fernandez-Ochoa A, Montanelli G, Vigone B, Santaniello A, Quirantes-Pine R, et al. Identification of a shared microbiomic and metabolomic profile in systemic autoimmune diseases. J Clin Med. 2019;8.
14.
go back to reference Li YLL, Deng X, Zhong L. Lipidomic and metabolomic profiling reveals novel candidate biomarkers in active systemic lupus erythematosus. Int J Clin Exp Pathol. 2019;12:857–66.PubMedPubMedCentral Li YLL, Deng X, Zhong L. Lipidomic and metabolomic profiling reveals novel candidate biomarkers in active systemic lupus erythematosus. Int J Clin Exp Pathol. 2019;12:857–66.PubMedPubMedCentral
15.
go back to reference Zhang Q, Li X, Yin X, Wang H, Fu C, Wang H, et al. Metabolomic profiling reveals serum L-pyroglutamic acid as a potential diagnostic biomarker for systemic lupus erythematosus. Rheumatology (Oxford) 2020. Zhang Q, Li X, Yin X, Wang H, Fu C, Wang H, et al. Metabolomic profiling reveals serum L-pyroglutamic acid as a potential diagnostic biomarker for systemic lupus erythematosus. Rheumatology (Oxford) 2020.
16.
go back to reference Kim HALH, Shin TH, Jung JY, Baek WY, Park HJ, Lee G, Paik MJ, Suh CH. Polyamine patterns in plasma of patients with systemic lupus erythematosus and fever. Lupus. 2018;27:930–8.PubMed Kim HALH, Shin TH, Jung JY, Baek WY, Park HJ, Lee G, Paik MJ, Suh CH. Polyamine patterns in plasma of patients with systemic lupus erythematosus and fever. Lupus. 2018;27:930–8.PubMed
17.
go back to reference Lee SM, Lee EM, Park JK, Jeon HS, Oh S, Hong S, et al. Metabolic biomarkers in midtrimester maternal plasma can accurately predict adverse pregnancy outcome in patients with SLE. Sci Rep. 2019;9:15169.PubMedPubMedCentral Lee SM, Lee EM, Park JK, Jeon HS, Oh S, Hong S, et al. Metabolic biomarkers in midtrimester maternal plasma can accurately predict adverse pregnancy outcome in patients with SLE. Sci Rep. 2019;9:15169.PubMedPubMedCentral
18.
go back to reference Malkawi AK, Alzoubi KH, Jacob M, Matic G, Ali A, Al Faraj A, et al. Metabolomics based profiling of dexamethasone side effects in rats. Front Pharmacol. 2018;9:46.PubMedPubMedCentral Malkawi AK, Alzoubi KH, Jacob M, Matic G, Ali A, Al Faraj A, et al. Metabolomics based profiling of dexamethasone side effects in rats. Front Pharmacol. 2018;9:46.PubMedPubMedCentral
19.
go back to reference Bordag N, Klie S, Jurchott K, Vierheller J, Schiewe H, Albrecht V, et al. Glucocorticoid (dexamethasone)-induced metabolome changes in healthy males suggest prediction of response and side effects. Sci Rep. 2015;5:15954.PubMedPubMedCentral Bordag N, Klie S, Jurchott K, Vierheller J, Schiewe H, Albrecht V, et al. Glucocorticoid (dexamethasone)-induced metabolome changes in healthy males suggest prediction of response and side effects. Sci Rep. 2015;5:15954.PubMedPubMedCentral
20.
go back to reference Di Dalmazi G, Quinkler M, Deutschbein T, Prehn C, Rayes N, Kroiss M, et al. Cortisol-related metabolic alterations assessed by mass spectrometry assay in patients with Cushing’s syndrome. Eur J Endocrinol. 2017;177:227–37.PubMed Di Dalmazi G, Quinkler M, Deutschbein T, Prehn C, Rayes N, Kroiss M, et al. Cortisol-related metabolic alterations assessed by mass spectrometry assay in patients with Cushing’s syndrome. Eur J Endocrinol. 2017;177:227–37.PubMed
21.
go back to reference Gropper S SJ, Groff J. Carbohydrates. In: P A, editor. Advanced nutrition and human metabolism. Belmont: Cengage; 2009. p. 63–105. Gropper S SJ, Groff J. Carbohydrates. In: P A, editor. Advanced nutrition and human metabolism. Belmont: Cengage; 2009. p. 63–105.
22.
go back to reference Wu G. Amino acids: metabolism, functions, and nutrition. Amino Acids. 2009;37:1–17.PubMed Wu G. Amino acids: metabolism, functions, and nutrition. Amino Acids. 2009;37:1–17.PubMed
23.
go back to reference CM NDL. Amino acid oxidation and the production of urea. In: LS, editor. Lehninger principles of biochemistry. New York: W.H. Freeman; 2013. p. 695–730. CM NDL. Amino acid oxidation and the production of urea. In: LS, editor. Lehninger principles of biochemistry. New York: W.H. Freeman; 2013. p. 695–730.
24.
go back to reference CM NDL. Lipids. In: LS, editor. Lehninger principles of biochemistry. New York: W.H. Freeman; 2013. p. 357–84. CM NDL. Lipids. In: LS, editor. Lehninger principles of biochemistry. New York: W.H. Freeman; 2013. p. 357–84.
25.
go back to reference Scavuzzi BM, Simao ANC, Iriyoda TMV, Lozovoy MAB, Stadtlober NP, Franchi Santos L, et al. Increased lipid and protein oxidation and lowered anti-oxidant defenses in systemic lupus erythematosus are associated with severity of illness, autoimmunity, increased adhesion molecules, and Th1 and Th17 immune shift. Immunol Res. 2018;66:158–71.PubMed Scavuzzi BM, Simao ANC, Iriyoda TMV, Lozovoy MAB, Stadtlober NP, Franchi Santos L, et al. Increased lipid and protein oxidation and lowered anti-oxidant defenses in systemic lupus erythematosus are associated with severity of illness, autoimmunity, increased adhesion molecules, and Th1 and Th17 immune shift. Immunol Res. 2018;66:158–71.PubMed
26.
go back to reference Lu L, Hu C, Zhao Y, He L, Zhou J, Li H, et al. Shotgun lipidomics revealed altered profiles of serum lipids in systemic lupus erythematosus closely associated with disease activity. Biomolecules. 2018;8. Lu L, Hu C, Zhao Y, He L, Zhou J, Li H, et al. Shotgun lipidomics revealed altered profiles of serum lipids in systemic lupus erythematosus closely associated with disease activity. Biomolecules. 2018;8.
27.
go back to reference Shah D, Mahajan N, Sah S, Nath SK, Paudyal B. Oxidative stress and its biomarkers in systemic lupus erythematosus. J Biomed Sci. 2014;21. Shah D, Mahajan N, Sah S, Nath SK, Paudyal B. Oxidative stress and its biomarkers in systemic lupus erythematosus. J Biomed Sci. 2014;21.
28.
go back to reference Frostegard J, Svenungsson E, Wu RH, Gunnarsson I, Lundberg IE, Klareskog L, et al. Lipid peroxidation is enhanced in patients with systemic lupus erythematosus and is associated with arterial and renal disease manifestations. Arthritis Rheum-Us. 2005;52:192–200. Frostegard J, Svenungsson E, Wu RH, Gunnarsson I, Lundberg IE, Klareskog L, et al. Lipid peroxidation is enhanced in patients with systemic lupus erythematosus and is associated with arterial and renal disease manifestations. Arthritis Rheum-Us. 2005;52:192–200.
29.
go back to reference Hage MP, Al-Badri MR, Azar ST. A favorable effect of hydroxychloroquine on glucose and lipid metabolism beyond its anti-inflammatory role. Ther Adv Endocrinol Metab. 2014;5:77–85.PubMedPubMedCentral Hage MP, Al-Badri MR, Azar ST. A favorable effect of hydroxychloroquine on glucose and lipid metabolism beyond its anti-inflammatory role. Ther Adv Endocrinol Metab. 2014;5:77–85.PubMedPubMedCentral
30.
go back to reference Babary H, Liu X, Ayatollahi Y, Chen XP, Doo L, Uppaluru LK, et al. Favorable effects of hydroxychloroquine on serum low density lipid in patients with systemic lupus erythematosus: a systematic review and meta-analysis. Int J Rheum Dis. 2018;21:84–92.PubMed Babary H, Liu X, Ayatollahi Y, Chen XP, Doo L, Uppaluru LK, et al. Favorable effects of hydroxychloroquine on serum low density lipid in patients with systemic lupus erythematosus: a systematic review and meta-analysis. Int J Rheum Dis. 2018;21:84–92.PubMed
31.
go back to reference Durcan L, Winegar DA, Connelly MA, Otvos JD, Magder LS, Petri M. Longitudinal evaluation of lipoprotein variables in systemic lupus erythematosus reveals adverse changes with disease activity and prednisone and more favorable profiles with hydroxychloroquine therapy. J Rheumatol. 2016;43:745–50.PubMedPubMedCentral Durcan L, Winegar DA, Connelly MA, Otvos JD, Magder LS, Petri M. Longitudinal evaluation of lipoprotein variables in systemic lupus erythematosus reveals adverse changes with disease activity and prednisone and more favorable profiles with hydroxychloroquine therapy. J Rheumatol. 2016;43:745–50.PubMedPubMedCentral
32.
go back to reference Pereira MJ, Palming J, Rizell M, Aureliano M, Carvalho E, Svensson MK, et al. The immunosuppressive agents rapamycin, cyclosporin A and tacrolimus increase lipolysis, inhibit lipid storage and alter expression of genes involved in lipid metabolism in human adipose tissue. Mol Cell Endocrinol. 2013;365:260–9.PubMed Pereira MJ, Palming J, Rizell M, Aureliano M, Carvalho E, Svensson MK, et al. The immunosuppressive agents rapamycin, cyclosporin A and tacrolimus increase lipolysis, inhibit lipid storage and alter expression of genes involved in lipid metabolism in human adipose tissue. Mol Cell Endocrinol. 2013;365:260–9.PubMed
33.
go back to reference Subramanian S, Trence DL. Immunosuppressive agents: effects on glucose and lipid metabolism. Endocrinol Metab Clin N Am. 2007;36:891–905 vii. Subramanian S, Trence DL. Immunosuppressive agents: effects on glucose and lipid metabolism. Endocrinol Metab Clin N Am. 2007;36:891–905 vii.
34.
go back to reference Nieto MF, Jayne DR. Con: the use of calcineurin inhibitors in the treatment of lupus nephritis. Nephrol Dial Transpl. 2016;31:1567–71. Nieto MF, Jayne DR. Con: the use of calcineurin inhibitors in the treatment of lupus nephritis. Nephrol Dial Transpl. 2016;31:1567–71.
35.
go back to reference Xu T, Holzapfel C, Dong X, Bader E, Yu Z, Prehn C, et al. Effects of smoking and smoking cessation on human serum metabolite profile: results from the KORA cohort study. BMC Med. 2013;11:60.PubMedPubMedCentral Xu T, Holzapfel C, Dong X, Bader E, Yu Z, Prehn C, et al. Effects of smoking and smoking cessation on human serum metabolite profile: results from the KORA cohort study. BMC Med. 2013;11:60.PubMedPubMedCentral
36.
go back to reference Thomson NC, Chaudhuri R, Spears M, Messow CM, Jelinsky S, Miele G, et al. Arachidonic acid metabolites and enzyme transcripts in asthma are altered by cigarette smoking. Allergy. 2014;69:527–36.PubMed Thomson NC, Chaudhuri R, Spears M, Messow CM, Jelinsky S, Miele G, et al. Arachidonic acid metabolites and enzyme transcripts in asthma are altered by cigarette smoking. Allergy. 2014;69:527–36.PubMed
37.
go back to reference Floegel A, Stefan N, Yu Z, Muhlenbruch K, Drogan D, Joost HG, et al. Identification of serum metabolites associated with risk of type 2 diabetes using a targeted metabolomic approach. Diabetes. 2013;62:639–48.PubMedPubMedCentral Floegel A, Stefan N, Yu Z, Muhlenbruch K, Drogan D, Joost HG, et al. Identification of serum metabolites associated with risk of type 2 diabetes using a targeted metabolomic approach. Diabetes. 2013;62:639–48.PubMedPubMedCentral
38.
go back to reference Strand E, Rebnord EW, Flygel MR, Lysne V, Svingen GFT, Tell GS, et al. Serum carnitine metabolites and incident type 2 diabetes mellitus in patients with suspected stable angina pectoris. J Clin Endocrinol Metab. 2018;103:1033–41.PubMed Strand E, Rebnord EW, Flygel MR, Lysne V, Svingen GFT, Tell GS, et al. Serum carnitine metabolites and incident type 2 diabetes mellitus in patients with suspected stable angina pectoris. J Clin Endocrinol Metab. 2018;103:1033–41.PubMed
39.
go back to reference Drogan D, Dunn WB, Lin W, Buijsse B, Schulze MB, Langenberg C, et al. Untargeted metabolic profiling identifies altered serum metabolites of type 2 diabetes mellitus in a prospective, nested case control study. Clin Chem. 2015;61:487–97.PubMed Drogan D, Dunn WB, Lin W, Buijsse B, Schulze MB, Langenberg C, et al. Untargeted metabolic profiling identifies altered serum metabolites of type 2 diabetes mellitus in a prospective, nested case control study. Clin Chem. 2015;61:487–97.PubMed
40.
go back to reference Wu T, Xie G, Ni Y, Liu T, Yang M, Wei H, et al. Serum metabolite signatures of type 2 diabetes mellitus complications. J Proteome Res. 2015;14:447–56.PubMed Wu T, Xie G, Ni Y, Liu T, Yang M, Wei H, et al. Serum metabolite signatures of type 2 diabetes mellitus complications. J Proteome Res. 2015;14:447–56.PubMed
41.
go back to reference Wang TJ, Gupta DK. Metabolite profiles in heart failure: looking for unique signatures in a heterogeneous syndrome. J Am Coll Cardiol. 2015;65:1521–4.PubMed Wang TJ, Gupta DK. Metabolite profiles in heart failure: looking for unique signatures in a heterogeneous syndrome. J Am Coll Cardiol. 2015;65:1521–4.PubMed
42.
go back to reference Dietrich S, Floegel A, Weikert C, Prehn C, Adamski J, Pischon T, et al. Identification of serum metabolites associated with incident hypertension in the European Prospective Investigation into Cancer and Nutrition-Potsdam Study. Hypertension. 2016;68:471–7.PubMed Dietrich S, Floegel A, Weikert C, Prehn C, Adamski J, Pischon T, et al. Identification of serum metabolites associated with incident hypertension in the European Prospective Investigation into Cancer and Nutrition-Potsdam Study. Hypertension. 2016;68:471–7.PubMed
43.
go back to reference Eudy AM, Siega-Riz AM, Engel SM, Franceschini N, Howard AG, Clowse MEB, et al. Effect of pregnancy on disease flares in patients with systemic lupus erythematosus. Ann Rheum Dis. 2018;77:855–60.PubMedPubMedCentral Eudy AM, Siega-Riz AM, Engel SM, Franceschini N, Howard AG, Clowse MEB, et al. Effect of pregnancy on disease flares in patients with systemic lupus erythematosus. Ann Rheum Dis. 2018;77:855–60.PubMedPubMedCentral
44.
go back to reference Yacoub R, Jacob A, Wlaschin J, McGregor M, Quigg RJ, Alexander JJ. Lupus: the microbiome angle. Immunobiology. 2018;223:460–5.PubMed Yacoub R, Jacob A, Wlaschin J, McGregor M, Quigg RJ, Alexander JJ. Lupus: the microbiome angle. Immunobiology. 2018;223:460–5.PubMed
45.
go back to reference Manfredo Vieira S, Hiltensperger M, Kumar V, Zegarra-Ruiz D, Dehner C, Khan N, et al. Translocation of a gut pathobiont drives autoimmunity in mice and humans. Science. 2018;359:1156–61.PubMed Manfredo Vieira S, Hiltensperger M, Kumar V, Zegarra-Ruiz D, Dehner C, Khan N, et al. Translocation of a gut pathobiont drives autoimmunity in mice and humans. Science. 2018;359:1156–61.PubMed
46.
47.
go back to reference Li Y, Wang HF, Li X, Li HX, Zhang Q, Zhou HW, et al. Disordered intestinal microbes are associated with the activity of Systemic Lupus Erythematosus. Clin Sci (Lond). 2019;133:821–38. Li Y, Wang HF, Li X, Li HX, Zhang Q, Zhou HW, et al. Disordered intestinal microbes are associated with the activity of Systemic Lupus Erythematosus. Clin Sci (Lond). 2019;133:821–38.
48.
go back to reference Azzouz D, Omarbekova A, Heguy A, Schwudke D, Gisch N, Rovin BH, et al. Lupus nephritis is linked to disease-activity associated expansions and immunity to a gut commensal. Ann Rheum Dis. 2019;78:947–56.PubMedPubMedCentral Azzouz D, Omarbekova A, Heguy A, Schwudke D, Gisch N, Rovin BH, et al. Lupus nephritis is linked to disease-activity associated expansions and immunity to a gut commensal. Ann Rheum Dis. 2019;78:947–56.PubMedPubMedCentral
49.
go back to reference Mu Q, Zhang H, Liao X, Lin K, Liu H, Edwards MR, et al. Control of lupus nephritis by changes of gut microbiota. Microbiome. 2017;5:73.PubMedPubMedCentral Mu Q, Zhang H, Liao X, Lin K, Liu H, Edwards MR, et al. Control of lupus nephritis by changes of gut microbiota. Microbiome. 2017;5:73.PubMedPubMedCentral
50.
go back to reference Gudi R, Suber J, Brown R, Johnson BM, Vasu C. Pretreatment with yeast-derived complex dietary polysaccharides suppresses gut inflammation, alters the microbiota composition, and increases immune regulatory short-chain fatty acid production in C57BL/6 mice. J Nutr. 2020;150:1291–302.PubMed Gudi R, Suber J, Brown R, Johnson BM, Vasu C. Pretreatment with yeast-derived complex dietary polysaccharides suppresses gut inflammation, alters the microbiota composition, and increases immune regulatory short-chain fatty acid production in C57BL/6 mice. J Nutr. 2020;150:1291–302.PubMed
51.
go back to reference Brestoff JR, Artis D. Commensal bacteria at the interface of host metabolism and the immune system. Nat Immunol. 2013;14:676–84.PubMedPubMedCentral Brestoff JR, Artis D. Commensal bacteria at the interface of host metabolism and the immune system. Nat Immunol. 2013;14:676–84.PubMedPubMedCentral
52.
go back to reference Rodriguez-Carrio J, Lopez P, Sanchez B, Gonzalez S, Gueimonde M, Margolles A, et al. Intestinal dysbiosis is associated with altered short-chain fatty acids and serum-free fatty acids in systemic lupus erythematosus. Front Immunol. 2017;8. Rodriguez-Carrio J, Lopez P, Sanchez B, Gonzalez S, Gueimonde M, Margolles A, et al. Intestinal dysbiosis is associated with altered short-chain fatty acids and serum-free fatty acids in systemic lupus erythematosus. Front Immunol. 2017;8.
53.
go back to reference Dodd D, Spitzer MH, Van Treuren W, Merrill BD, Hryckowian AJ, Higginbottom SK, et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature. 2017;551:648.PubMedPubMedCentral Dodd D, Spitzer MH, Van Treuren W, Merrill BD, Hryckowian AJ, Higginbottom SK, et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature. 2017;551:648.PubMedPubMedCentral
54.
go back to reference Wang J, Wang Y, Zhang X, Liu JQ, Zhang QP, Zhao Y, et al. Gut microbial dysbiosis is associated with altered hepatic functions and serum metabolites in chronic hepatitis B patients. Front Microbiol. 2017;8. Wang J, Wang Y, Zhang X, Liu JQ, Zhang QP, Zhao Y, et al. Gut microbial dysbiosis is associated with altered hepatic functions and serum metabolites in chronic hepatitis B patients. Front Microbiol. 2017;8.
55.
go back to reference Liu RX, Hong J, Xu XQ, Feng Q, Zhang DY, Gu YY, et al. Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention. Nat Med. 2017;23:859.PubMed Liu RX, Hong J, Xu XQ, Feng Q, Zhang DY, Gu YY, et al. Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention. Nat Med. 2017;23:859.PubMed
56.
go back to reference Org E, Blum Y, Kasela S, Mehrabian M, Kuusisto J, Kangas AJ, et al. Relationships between gut microbiota, plasma metabolites, and metabolic syndrome traits in the METSIM cohort. Genome Biol. 2017;18. Org E, Blum Y, Kasela S, Mehrabian M, Kuusisto J, Kangas AJ, et al. Relationships between gut microbiota, plasma metabolites, and metabolic syndrome traits in the METSIM cohort. Genome Biol. 2017;18.
57.
go back to reference Ryan PM, Patterson E, Kent RM, Stack H, O'Connor PM, Murphy K, et al. Recombinant incretin-secreting microbe improves metabolic dysfunction in high-fat diet fed rodents. Sci Rep. 2017;7:13523.PubMedPubMedCentral Ryan PM, Patterson E, Kent RM, Stack H, O'Connor PM, Murphy K, et al. Recombinant incretin-secreting microbe improves metabolic dysfunction in high-fat diet fed rodents. Sci Rep. 2017;7:13523.PubMedPubMedCentral
58.
go back to reference Maukonen J, Saarela M. Human gut microbiota: does diet matter? Proc Nutr Soc. 2015;74:23–36.PubMed Maukonen J, Saarela M. Human gut microbiota: does diet matter? Proc Nutr Soc. 2015;74:23–36.PubMed
59.
go back to reference Caesar R, Fak F, Backhed F. Effects of gut microbiota on obesity and atherosclerosis via modulation of inflammation and lipid metabolism. J Intern Med. 2010;268:320–8.PubMed Caesar R, Fak F, Backhed F. Effects of gut microbiota on obesity and atherosclerosis via modulation of inflammation and lipid metabolism. J Intern Med. 2010;268:320–8.PubMed
60.
go back to reference Floegel A, von Ruesten A, Drogan D, Schulze MB, Prehn C, Adamski J, et al. Variation of serum metabolites related to habitual diet: a targeted metabolomic approach in EPIC-Potsdam. Eur J Clin Nutr. 2013;67:1100–8.PubMed Floegel A, von Ruesten A, Drogan D, Schulze MB, Prehn C, Adamski J, et al. Variation of serum metabolites related to habitual diet: a targeted metabolomic approach in EPIC-Potsdam. Eur J Clin Nutr. 2013;67:1100–8.PubMed
61.
go back to reference Akbaraly T, Wurtz P, Singh-Manoux A, Shipley MJ, Haapakoski R, Lehto M, et al. Association of circulating metabolites with healthy diet and risk of cardiovascular disease: analysis of two cohort studies. Sci Rep. 2018;8:8620.PubMedPubMedCentral Akbaraly T, Wurtz P, Singh-Manoux A, Shipley MJ, Haapakoski R, Lehto M, et al. Association of circulating metabolites with healthy diet and risk of cardiovascular disease: analysis of two cohort studies. Sci Rep. 2018;8:8620.PubMedPubMedCentral
62.
go back to reference Rebholz CM, Zheng Z, Grams ME, Appel LJ, Sarnak MJ, Inker LA, et al. Serum metabolites associated with dietary protein intake: results from the Modification of Diet in Renal Disease (MDRD) randomized clinical trial. Am J Clin Nutr. 2019;109:517–25.PubMedPubMedCentral Rebholz CM, Zheng Z, Grams ME, Appel LJ, Sarnak MJ, Inker LA, et al. Serum metabolites associated with dietary protein intake: results from the Modification of Diet in Renal Disease (MDRD) randomized clinical trial. Am J Clin Nutr. 2019;109:517–25.PubMedPubMedCentral
63.
go back to reference Perl A, Hanczko R, Lai ZW, Oaks Z, Kelly R, Borsuk R, et al. Comprehensive metabolome analyses reveal N-acetylcysteine-responsive accumulation of kynurenine in systemic lupus erythematosus: implications for activation of the mechanistic target of rapamycin. Metabolomics. 2015;11:1157–74.PubMedPubMedCentral Perl A, Hanczko R, Lai ZW, Oaks Z, Kelly R, Borsuk R, et al. Comprehensive metabolome analyses reveal N-acetylcysteine-responsive accumulation of kynurenine in systemic lupus erythematosus: implications for activation of the mechanistic target of rapamycin. Metabolomics. 2015;11:1157–74.PubMedPubMedCentral
64.
go back to reference Sipka SSS, Szucs K, Kovács I, Kiss E, Antal-Szamás P, Lakos G, Aleksza M, Illés A, Gergely P, Szegedi G. Decreased arachidonic acid release in peripheral blood monocytes of patients with systemic lupus erythematosus. J Rheumatol. 2001;28:2012–7.PubMed Sipka SSS, Szucs K, Kovács I, Kiss E, Antal-Szamás P, Lakos G, Aleksza M, Illés A, Gergely P, Szegedi G. Decreased arachidonic acid release in peripheral blood monocytes of patients with systemic lupus erythematosus. J Rheumatol. 2001;28:2012–7.PubMed
65.
go back to reference Coras R, Murillo-Saich JD, Guma M. Circulating pro- and anti-inflammatory metabolites and its potential role in rheumatoid arthritis pathogenesis. Cells. 2020;9. Coras R, Murillo-Saich JD, Guma M. Circulating pro- and anti-inflammatory metabolites and its potential role in rheumatoid arthritis pathogenesis. Cells. 2020;9.
66.
go back to reference Narasimhan R, Coras R, Rosenthal SB, Sweeney SR, Lodi A, Tiziani S, et al. Serum metabolomic profiling predicts synovial gene expression in rheumatoid arthritis. Arthritis Res Ther. 2018;20:164.PubMedPubMedCentral Narasimhan R, Coras R, Rosenthal SB, Sweeney SR, Lodi A, Tiziani S, et al. Serum metabolomic profiling predicts synovial gene expression in rheumatoid arthritis. Arthritis Res Ther. 2018;20:164.PubMedPubMedCentral
67.
go back to reference Anderson JR, Chokesuwattanaskul S, Phelan MM, Welting TJM, Lian LY, Peffers MJ, et al. (1) H NMR metabolomics identifies underlying inflammatory pathology in osteoarthritis and rheumatoid arthritis synovial joints. J Proteome Res. 2018;17:3780–90.PubMedPubMedCentral Anderson JR, Chokesuwattanaskul S, Phelan MM, Welting TJM, Lian LY, Peffers MJ, et al. (1) H NMR metabolomics identifies underlying inflammatory pathology in osteoarthritis and rheumatoid arthritis synovial joints. J Proteome Res. 2018;17:3780–90.PubMedPubMedCentral
68.
go back to reference Kageyama G, Saegusa J, Irino Y, Tanaka S, Tsuda K, Takahashi S, et al. Metabolomics analysis of saliva from patients with primary Sjogren’s syndrome. Clin Exp Immunol. 2015;182:149–53.PubMedPubMedCentral Kageyama G, Saegusa J, Irino Y, Tanaka S, Tsuda K, Takahashi S, et al. Metabolomics analysis of saliva from patients with primary Sjogren’s syndrome. Clin Exp Immunol. 2015;182:149–53.PubMedPubMedCentral
69.
go back to reference Fernandez-Ochoa A, Brunius C, Borras-Linares I, Quirantes-Pine R, Cadiz-Gurrea ML, Precisesads Clinical C, et al. Metabolic disturbances in urinary and plasma samples from seven different systemic autoimmune diseases detected by HPLC-ESI-QTOF-MS. J Proteome Res 2020. Fernandez-Ochoa A, Brunius C, Borras-Linares I, Quirantes-Pine R, Cadiz-Gurrea ML, Precisesads Clinical C, et al. Metabolic disturbances in urinary and plasma samples from seven different systemic autoimmune diseases detected by HPLC-ESI-QTOF-MS. J Proteome Res 2020.
70.
go back to reference Carlson AKRR, Wallace CW, Adams E, Greenwood MC, Bothner B, June RK. Global metabolomic profiling of human synovial fluid for rheumatoid arthritis biomarkers. Clin Exp Rheumatol. 2019;37:393–9.PubMed Carlson AKRR, Wallace CW, Adams E, Greenwood MC, Bothner B, June RK. Global metabolomic profiling of human synovial fluid for rheumatoid arthritis biomarkers. Clin Exp Rheumatol. 2019;37:393–9.PubMed
71.
go back to reference Fernandez-Ochoa A, Borras-Linares I, Quirantes-Pine R, Alarcon-Riquelme ME, Beretta L, Segura-Carretero A, et al. Discovering new metabolite alterations in primary Sjogren’s syndrome in urinary and plasma samples using an HPLC-ESI-QTOF-MS methodology. J Pharm Biomed Anal. 2020;179:112999.PubMed Fernandez-Ochoa A, Borras-Linares I, Quirantes-Pine R, Alarcon-Riquelme ME, Beretta L, Segura-Carretero A, et al. Discovering new metabolite alterations in primary Sjogren’s syndrome in urinary and plasma samples using an HPLC-ESI-QTOF-MS methodology. J Pharm Biomed Anal. 2020;179:112999.PubMed
72.
go back to reference Souto-Carneiro M, Toth L, Behnisch R, Urbach K, Klika KD, Carvalho RA, et al. Differences in the serum metabolome and lipidome identify potential biomarkers for seronegative rheumatoid arthritis versus psoriatic arthritis. Ann Rheum Dis. 2020;79:499–506.PubMedPubMedCentral Souto-Carneiro M, Toth L, Behnisch R, Urbach K, Klika KD, Carvalho RA, et al. Differences in the serum metabolome and lipidome identify potential biomarkers for seronegative rheumatoid arthritis versus psoriatic arthritis. Ann Rheum Dis. 2020;79:499–506.PubMedPubMedCentral
Metadata
Title
Caution in studying and interpreting the lupus metabolome
Authors
Ting Zhang
Chandra Mohan
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Arthritis Research & Therapy / Issue 1/2020
Electronic ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-020-02264-2

Other articles of this Issue 1/2020

Arthritis Research & Therapy 1/2020 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine