Skip to main content
medRxiv
  • Home
  • About
  • Submit
  • ALERTS / RSS
Advanced Search

The Gut Metabolism is altered in patients with CRB1-associated inherited retinal degeneration

View ORCID ProfileLude Moekotte, Joke H. de Boer, Sanne Hiddingh, Bram Gerritsen, Jutta Lintelmann, Alexander Cecil, L. Ingeborgh van den Born, Xuan-Thanh-An Nguyen, Camiel J.F. Boon, Maria M. van Genderen, Jonas J.W. Kuiper
doi: https://doi.org/10.1101/2024.02.22.24303210
Lude Moekotte
1Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Lude Moekotte
  • For correspondence: l.moekotte{at}umcutrecht.nl
Joke H. de Boer
1Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sanne Hiddingh
1Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Bram Gerritsen
1Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jutta Lintelmann
2Metabolomics and Proteomics Core (MPC), Helmholtz, Munich, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Alexander Cecil
2Metabolomics and Proteomics Core (MPC), Helmholtz, Munich, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
L. Ingeborgh van den Born
3The Rotterdam Eye Hospital, Rotterdam, the Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Xuan-Thanh-An Nguyen
4Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Camiel J.F. Boon
4Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
5Department of Ophthalmology, Amsterdam UMC, Amsterdam, the Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Maria M. van Genderen
1Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
6Bartiméus, Diagnostic Centre for complex visual disorders, Zeist, the Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jonas J.W. Kuiper
1Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Abstract
  • Full Text
  • Info/History
  • Metrics
  • Supplementary material
  • Data/Code
  • Preview PDF
Loading

Abstract

Purpose To compare the plasma metabolic profile of patients with a CRB1-associated inherited retinal degeneration (CRB1-IRD) with healthy controls (HCs).

Design A case-control study.

Methods Plasma concentration of 619 metabolites was measured with the MxP® Quant 500 Kit in 30 patients with a CRB1-IRD and 29 HCs. We fitted a linear regression model with adjustments for age and sex based on the concentration of metabolites in µM (µmol/L), or on the sums and ratios of metabolites, to determine differences between patients and controls.

Results Over-representation of pathways among metabolites associated strongest to CRB1-IRDs (P < 0.05, n = 62) identified amino acid pathways (such as beta-alanine, histidine, and glycine/serine) and bile acid biosynthesis, driven by a decrease in deoxycholic acid derivatives produced by gut microbiota. Enrichment analysis of metabolic classes across the plasma metabolic profile further identified significant positive enrichment for lipid metabolites glycerophospholipids, cholesterol esters, and ceramides, and significant depletion for bile acid metabolites. Further investigation of the sums and ratios (i.e., metabolism indicators) ascertained a significant decrease in intestinal microbial-dependent secondary bile acid classes.

Conclusions Lipid metabolic alterations and decreased microbiota-related secondary bile acid concentrations indicate significant alterations in gut metabolism in patients with a CRB1-IRD.

Introduction

Inherited retinal degenerations (IRDs) comprise a clinically and genetically heterogeneous group of monogenic retinal disorders, characterized by progressive loss of photoreceptors leading to severe visual impairment. Pathogenic variants in the Crumbs homolog 1 (CRB1) gene are associated with different IRD phenotypes, such as Leber congenital amaurosis, Severe Early Childhood Onset Retinal Dystrophy (SECORD), and cone-rod dystrophies.1 CRB1 is highly expressed in retinal photoreceptors, as well as cells of the ciliary body that are involved in maintaining the blood-aqueous barrier.2 As compared to other retinal degenerations, CRB1-IRDs are more frequently associated with ocular inflammation or uveitis, suggesting that the disease may progress through inflammatory responses, and with loss of immune regulatory functions in the eye.3–7 This is supported by molecular studies, that show pro-inflammatory changes in peripheral blood T cell and dendritic cell populations, and elevated plasma complement components in peripheral blood.8,9 Interestingly, blood microbiota-produced metabolites detectable in blood, such as secondary bile acids, are associated with the severity of autoimmune uveitis.10 Retinal degeneration models also showed gut dysbiosis, which correlated with visual decline.11,12 The altered plasma metabolome might reflect differences in the gut microbiome, and can be used to probe disease mechanisms that involve gut metabolism and retinal function.

The retina is one of the most metabolic active areas in the body,13,14 of which photoreceptors have the highest energy demand.15,16 In retinal degenerations, a disturbance in glucose metabolism and oxidative damage contribute to photoreceptor death. This means that elucidating metabolic pathways responsible for the degeneration of photoreceptors is a major topic of interest.17–19 Previous studies in animal models of retinal degeneration showed that a metabolic switch in retinal cells leads to the release of neurotoxic inflammatory factors that damage photoreceptors.20 Other observations in animal models include decreased fatty acids in the retina which are correlated with photoreceptor degeneration.21 Also, the metabolism of amino acids, carbohydrates, nucleotides, and lipids has been proposed to play an important role in retinitis pigmentosa, the most common form of retinal degeneration.22–26 Oral supplementation to correct these metabolic changes proved to have a neuroprotective effect on the photoreceptor cells in mouse models of retinal degeneration.27 Studies of human retinal degeneration patients showed abnormalities in copper metabolisms,28 and lipid and amino acid metabolisms,29–31 suggesting that metabolic pathways may play a role in CRB1-IRDs.

Metabolomics is the multidisciplinary field studying the composition of metabolites (i.e., small molecules involved in cell metabolism) and has provided significant insights into immunomodulation and identification of metabolic and functional pathways that modulate clinical phenotypes.32–35 Application of metabolomics to retinal diseases has identified dysregulated functional pathways in proliferative diabetic retinopathy, age-related macular degeneration, and proliferative vitreoretinopathy,22,36,37 including alterations in fatty acids, amino acids, and nucleosides.38–49 Combining the current knowledge of immune system changes in CRB1-IRDs with new metabolic findings in patients can aid in better understanding the pathophysiological mechanisms of CRB1-IRDs. The purpose of this study was to elucidate the plasma metabolic profile of patients with a CRB1-IRD compared to healthy controls.

Methods

Patients

This study was performed in compliance with the guidelines of the Declaration of Helsinki and has the approval of the local Institutional Review Board (University Medical Center Utrecht (UMCU)). In total, 30 patients with a CRB1-IRD were included in the UMCU. Diagnosis was based on ophthalmic examination, imaging, and full field electroretinography. The diagnosis was confirmed with next-generation sequencing or whole-exome sequencing, displaying 2 or more pathogenic variants in the CRB1 gene (Supplementary Table 1). All patients with systemic inflammatory conditions at the time of inclusion and/or patients with systemic immunomodulatory treatment were excluded from participation. On the day of inclusion, slit lamp examination was performed by an experienced uveitis specialist for assessment of vitreous cells and vitreous haze, and visual acuity was measured.

Twenty nine age matched healthy controls (HCs) with no known ophthalmic history or inflammatory disease were included through the research blood bank of the UMCU (known as the “Mini Donor Dienst”).

Metabolomics

Venous blood samples were collected in Lavender Top Tubes ((#362084, BD Biosciences, Franklin Lakes, USA) of all patients and HCs on the day of inclusion (Supplementary Table 2). First, the tubes were centrifuged at 400g for 10 minutes. Plasma was transferred to a new tube, which was centrifuged at 1500g for 10 minutes. Next, the centrifuged plasma was collected in Micronic 1.4 mL round bottom tubes (#MP32022, Micronic, Lelystad, the Netherlands) and stored at −80 °C until measurement. For metabolomics measurement, the samples were shipped on dry ice to the Metabolomics and Proteomics Core (MPC) of the Helmholtz Facility München (Germany). Samples were randomized over one 96-wells plate and analyzed with the MxP® Quant 500 Kit (#21094.12, Biocrates life sciences ag, Innsbruck, Austria). This kit is based on liquid chromatography-electrospray ionization-tandem mass spectrometric (LC-MS/MS) and flow injection-electrospray ionization-tandem mass spectrometric (FIA) measurements. In total, 5 aliquots of a plasma sample of an in-house plasma pool were used for quality control.

Statistical analyses

Principal component analysis was performed with the prcomp function with the factoextra package.50 Metabolite concentrations of all plasma samples were expressed in units of µM (µmol/L). Individual metabolites and MetaboINDICATOR™ data – metabolite sums and ratios (Biocrates life sciences ag) – were analyzed separately. In case more than half of the data points of a metabolite were zero or a metabolite had zero variance, the metabolite was removed from the dataset. A Box Cox transformation was used to better approximate Gaussian distributions, after which all metabolite values were standardized relative to healthy controls. Then, a linear regression model was fitted with the lm function on each metabolite to analyze differences between patients with a CRB1-IRD and HCs, after which volcano plots were made with the ggplot2 package to visualize the results.51 Metabolite classes were tested through a metabolite class enrichment analysis with the GSEA function of the clusterProfiler package.52 The classes were categorized as by the MxP® Quant 500 Kit (#21094.12, Biocrates life sciences ag, Innsbruck, Austria) (Figure 1A and Supplementary Table 2). A P value adjusted with Benjamini Hochberg (BH) of < 0.05 was considered statistically significant. Over-representation analysis was performed with the MetaboAnalystR package v4.0.0. Other figures were generated with the pie function and ggplot2 package.

Figure 1.
  • Download figure
  • Open in new tab
Figure 1. Overview and quality check.

A) Pie chart representing coverage of the main classes of metabolites measured in plasma in this study. B) The first two principal components of metabolic profiles (n = 612 analytes; 7 analytes were removed in advance because more than half of the values were zero) in 30 patients with a CRB1-IRD and 29 healthy controls [HCs], and 5 (closely clustered) reference plasma samples.

Results

We determined the concentrations of 619 metabolites across 23 metabolite classes in the peripheral blood plasma of 30 patients with a CRB1-IRD and 29 controls (Figure 1A). There were 7 metabolites in total that had more than half of their values zero, and they were removed. First, to visualize the variation of (reference) samples across the remaining 612 metabolites, we performed a principal component analysis, which showed close clustering of the reference plasma samples (Figure 1B).

Using a linear model, we compared patients with a CRB1-IRD and HCs, corrected for age and sex. We detected 62 lipids and small molecules that showed a nominal significant difference in concentration (Figure 2A and C). We found that patients had elevated levels of lipid metabolites, such as Cholesteryl Esters [CE(18:0), and CE(18:1)], sphingomyelins [SM C24:1] (Figure 2A), as well as amino acid derivatives Anserine and biogenic amine β-Alanine (Figure 2C). We further noted a decrease in plasma secondary bile acids (Glycodeoxycholic acid [GDCA], Deoxycholic acid [DCA], Taurodeoxycholic acid [TDCA], and Glycolithocholic acid [GLCA]) produced by the gut microbiota,53 cortisol (corticosteroid hormone), and nonessential amino acids, such as Alanine and Glycine (Figure 2C). Statistical differences at the level of individual metabolites did not exceed the threshold for statistical significance after correction for multiple testing (Supplementary Table 3). A statistical analysis of over-representation was performed to determine if metabolites associated with CRB1-IRDs were associated with specific metabolic pathways (49/62 nominal P < 0.05 present in libraries used in MetaboAnalystR analysis). This analysis revealed a significant enrichment for Bile Acid Biosynthesis, and amino acid metabolism pathways (i.e., Beta-Alanine Metabolism, Glycine and Serine Metabolism, Histidine Metabolism, and Ammonia Recycling) (Figure 3A and Supplementary Table 4).

Figure 2.
  • Download figure
  • Open in new tab
Figure 2. Changes in lipids and small molecules in plasma of patients with a CRB1-IRD.

A and C) Volcano plot of the results from the comparison of patients versus controls. The normalized regression coefficient and the −log10 of the nominal P value are shown. The metabolites are colored per class of lipid as shown in the legend. The horizontal line represents a P value cutoff of 0.05. B and D) Boxplots of the top5 most significant metabolites (smallest P values). All top 5 lipid metabolites were increased in plasma of patients compared to HCs, while all top 5 small molecule metabolites were decreased.

Figure 3.
  • Download figure
  • Open in new tab
Figure 3. Enrichment and over-representation analysis.

A) Over-representation analysis of 64 nominal significant metabolites. B-E) A metabolite class enrichment analysis performed on a ranked metabolite list. The top graph of each figure shows where the metabolite is located relative to the ranked list, as well as the distribution. The bottom graph of each figure visualizes the enrichment score. Graph B till E show the top 4 most significant metabolite classes. Glycerophospholipids (graph C) and Glycosylceramides (graph D) are positively enriched in patients, while the classes Aminoacids (graph B) and Bile Acids (graph E) are negatively enriched in patients.

Figure 4.
  • Download figure
  • Open in new tab
Figure 4. Linear model shows a significant difference in metabolic indicators associated with gut metabolism (MetaboINDICATOR™).

A) Volcano plot visualizing the significance of metabolic indicators tested with a linear model. The normalized regression coefficient and the −log10 of the adjusted P value (FDR) is shown. The horizontal line represents a Padj value cutoff of 0.05. B) Boxplots of the significant metabolic indicators. All metabolic indicators are decreased in patients compared to HCs.

Next, we were interested to assess the enrichment for metabolic classes. Enrichment analysis considers the metabolic profile as a whole using differential data from every measured metabolite and searches for classes that display significantly coordinated changes in metabolite levels. To this end, we ranked the metabolic profiles (n = 612 metabolites) based on the statistics from the group comparison and performed metabolite class enrichment analysis for the 23 probed classes. After correcting for multiple testing, Bile Acids and Aminoacids were significantly and negatively enriched in the metabolome of patients (Figure 3B and Figure 3E). In addition, we detected a significant enrichment for Glycerophospholipids, Glycosylceramides, Sphingolipids, Cholesterol Esters, and Ceramides (Figure 3B-E for top 4 most significant and Supplementary Table 5).

Finally, we used the predefined sums and ratios of metabolites reported in clinical chemistry and biomedical phenotypes (MetaboINDICATOR™ data). Using a linear model corrected for sex and age, we detected a significant decrease in several metabolites produced by the intestinal microbiota, including Sum of Secondary Bile-Acids (Padj = 0.030),54–57 Sum of 12a-OH Bile-Acids (Padj = 0.047),58–60 Sum of Conjugated Secondary Bile-Acids (Padj = 0.047),61,62 and Sum of Carboxylic Acids (Padj = 0.047),63 as well as decreased Lactate dehydrogenase [LDH] Activity (Padj = 0.019) (Supplementary Table 6 and 7 for description and references).64,65

Discussion

In this study, we show differences in plasma metabolic profiles in patients with a CRB1-IRD compared to HCs. We detected enrichment for amino acid pathways, changes in lipid metabolites, and evidence for a decrease in secondary bile acids.

Bile acids are derivatives of cholesterol,66–68 and interest on bile acids in ocular diseases has peaked in the last decade. Bile acids have neuroprotective properties and protect against photoreceptor degeneration,69–73 mainly acting on oxidative stress and apoptosis.70,74 Primary and secondary bile acids influence metabolism and inflammation, and the gut microbiome is required to metabolize secondary bile acids from primary bile acids produced by the liver. Some intestinal species in the genus Clostridium metabolize the secondary bile acids we found to be reduced in patients with a CRB1-IRD, including deoxycholic acid (DCA) and their derivatives.75 DCA and derivatives of DCA play important roles in immune cells by activation of bile acid receptors and are known to regulate innate immunity and T cell activation.76 This is of interest because we have shown substantial activation of innate immune pathways and changes in T cell populations in patients with a CRB1-IRD.8 Perhaps, patients with a CRB1-IRD may exhibit changes in the gut microbiome, which are proxied by the changes in metabolites, such as secondary bile acids. Gut dysbiosis has also been linked to visual dysfunction in animal models of retinal degeneration.11 Interestingly, treatment with the closely related isomers of secondary bile acids protects against retinal degeneration in a widely used mouse model for retinal degeneration.73,77,78 CRB1-IRDs are characterized by inflammation and involve both innate and adaptive pathways, with intraocular inflammation (or uveitis) being an unusually frequent manifestation in this type of retinal degeneration.8,9 In models of non-infectious uveitis, microbiome changes are a key pathological trigger.79 This is important, because experimental autoimmune uveitis is characterized by low microbiota-related secondary bile acids in blood, and selective restoration of the gut bile acid pool attenuates the severity of uveitis.10 The anti-inflammatory mechanism of bile acids involves deoxycholic acid [DCA], which directly inhibits dendritic cell activation in uveitis.10 Dendritic cell alterations have also been reported in CRB1-IRDs, and are shown to reflect disease activity.5,8 Whether gut dysbiosis, DCA, and dendritic cell activation play a role in disease progression or if this relates to the inflammatory processes observed in selected patients with CRB1-IRDs exhibiting intraocular inflammation requires further investigation.

In the current cohort of patients with a CRB1-IRD, we also found evidence for changes in other lipid metabolites, which is in line with other studies on retinal degenerations.80–83 Blood metabolites, including lipids, enter the eye through two diffusion barriers: the outer limiting membrane and the outer blood-retina barrier (BRB).83,84 The RPE and the Bruch’s membrane mediate the metabolite exchange between photoreceptors and the blood, of which even large metabolites can cross and expand to all neural retinal layers.83,85–87 One notable finding in our study is the increase of cholesteryl esters in patients, which is the inactive form of cholesterol that transports and stores cholesterol.88–92 Cholesterol has toxic effects if not esterified,91 and higher concentrations of cholesteryl esters have been described in neurodegeneration after induced excitotoxic injury, Huntington’s disease, ALS (amyotrophic lateral sclerosis), MS (multiple sclerosis), and Alzheimer’s disease.92–96 When cholesterol excess occurs, esterification seems to occur to prevent further degeneration. In AMD patients, cholesteryl esters accumulate in the Bruch’s membrane and, together with unesterified cholesterol, represent >40% hard druse volume.97 It is unclear what the role of cholesteryl esters are in CRB1-IRDs, but with its protective effects it is possible that esterification of cholesterol occurs as a reaction to photoreceptor damage to limit further degeneration of photoreceptors.

Another significantly positively enriched lipid class in patients is Glycerophospholipids. In our metabolomics panel, all glycerophospholipids were lysophosphatidylcholines (LPCs). LPCs result from inflammatory processes and have been contributed to pro-inflammatory, as well as anti-inflammatory effects.98,99 They can increase the phagocytic activity of macrophages,98 increase genes involved in cholesterol biosynthesis,100 and increase the reactive oxygen production in polymorphonuclear leukocytes.101 A decrease of Lpcat1 expression, coding for the protein lysophosphatidylcholine acyltransferase 1, is linked to photoreceptor degeneration in rd11 mice.102 In diabetes type 1 and type 2 mice, Lpcat1 is downregulated.103 Treating hyperlipidemic patients with an antioxidant significantly reduced LPCs in low-density lipoprotein compared to controls.104

In our study we included 30 CRB1-IRD patients and 29 HCs, limiting the power of our analyses. After correcting for multiple testing, the differences for individual metabolites did not exceed the statistical significance threshold. However, reflecting the physiological function of multiple metabolites functioning in a particular metabolic pathway, testing for grouped metabolic classes clearly showed significant enrichment after multiple testing correction for several pathways. The design of this cross-sectional study does, however, limit our ability to establish causality.

In conclusion, patients with a CRB1-IRD show an increase of lipid metabolites that are linked to photoreceptor degeneration, as well as a decrease of neuroprotective secondary bile acid concentrations, which indicate significant changes in the gut metabolism of patients with a CRB1-IRD. In the future, our findings combined with a larger prospective cohort study could aid us to elucidate causality and determine possible metabolic targets for therapy.

Data Availability

All data produced in the present study are available upon reasonable request to the authors

Acknowledgements

We would like to thank Dr. N.H. ten Dam-van Loon, Dr. J. Ossewaarde-van Norel, and Dr. V. Koopman-Kalinina Ayuso for their assistance performing slit lamp examination and assessment of vitreous cells and vitreous haze on all included patients.

Footnotes

  • Funding: Bartimeus Foundation

References

  1. 1.↵
    Zahid S, Branham K, Schlegel D, Pennesi ME, Michaelides M, Heckenlively J, et al. CRB1. In: Retinal Dystrophy Gene Atlas. Cham: Springer International Publishing; 2018. p. 75–8.
  2. 2.↵
    Van Zyl T, Yan W, McAdams AM, Monavarfeshani A, Hageman GS, Sanes JR. Cell atlas of the human ocular anterior segment: Tissue-specific and shared cell types. Proc Natl Acad Sci U S A. 2022;119(29).
  3. 3.↵
    Li AS, Pasricha MV, Mishra K, Nguyen QD, Beres SJ, Wood EH. CRB1-associated retinal dystrophy presenting as self-resolving opsoclonus and posterior uveitis. Am J Ophthalmol Case Rep. 2022;26.
  4. 4.
    Hettinga YM, van Genderen MM, Wieringa W, Ossewaarde-van Norel J, de Boer JH. Retinal Dystrophy in 6 Young Patients Who Presented with Intermediate Uveitis. Ophthalmology. 2016;123(9):2043–6.
    OpenUrl
  5. 5.↵
    Verhagen F, Kuiper J, Nierkens S, Imhof SM, Radstake T, de Boer J. Systemic inflammatory immune signatures in a patient with CRB1 linked retinal dystrophy. Expert Rev Clin Immunol. 2016;12(12):1359–62.
    OpenUrl
  6. 6.
    Strong S, Liew G, Michaelides M. Retinitis pigmentosa-associated cystoid macular oedema: pathogenesis and avenues of intervention. Br J Ophthalmol. 2017;101(1):31–7.
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    Murro V, Mucciolo DP, Sodi A, Vannozzi L, De Libero C, Simonini G, et al. Retinal capillaritis in a CRB1-associated retinal dystrophy. Ophthalmic Genet. 2017;38(6):555–8.
    OpenUrlCrossRef
  8. 8.↵
    Moekotte L, Kuiper JJW, Hiddingh S, Nguyen XTA, Boon CJF, van den Born LI, et al. CRB1-Associated Retinal Dystrophy Patients Have Expanded Lewis Glycoantigen-Positive T Cells. Invest Ophthalmol Vis Sci. 2023;64(13):6.
    OpenUrl
  9. 9.↵
    Moekotte L, Boer JH de, Hiddingh S, Ligt A de, Nguyen XTA, Hoyng CB, et al. Elevated Plasma Complement Factors in CRB1-associated Inherited Retinal Dystrophies. medRxiv. 2023;10(11):2023.11.10.23298334.
    OpenUrl
  10. 10.↵
    Hu J, Wang C, Huang X, Yi S, Pan S, Zhang Y, et al. Gut microbiota-mediated secondary bile acids regulate dendritic cells to attenuate autoimmune uveitis through TGR5 signaling. Cell Rep. 2021;36(12).
  11. 11.↵
    Kutsyr O, Maestre-Carballa L, Lluesma-Gomez M, Martinez-Garcia M, Cuenca N, Lax P. Retinitis pigmentosa is associated with shifts in the gut microbiome. Sci Rep. 2021;11(1).
  12. 12.↵
    Bringer MA, Gabrielle PH, Bron AM, Creuzot-Garcher C, Acar N. The gut microbiota in retinal diseases. Exp Eye Res. 2022;214:108867.
    OpenUrl
  13. 13.↵
    Wong-Riley M. Energy metabolism of the visual system. Eye Brain. 2010;2:99.
    OpenUrlCrossRefPubMed
  14. 14.↵
    Country MW. Retinal metabolism: A comparative look at energetics in the retina. Brain Res. 2017;1672:50–7.
    OpenUrlCrossRefPubMed
  15. 15.↵
    Yu DY, Cringle SJ. Oxygen distribution and consumption within the retina in vascularised and avascular retinas and in animal models of retinal disease. Prog Retin Eye Res. 2001;20(2):175–208.
    OpenUrlCrossRefPubMedWeb of Science
  16. 16.↵
    Wangsa-Wirawan ND, Linsenmeier RA. Retinal oxygen: fundamental and clinical aspects. Arch Ophthalmol. 2003;121(4):547–57.
    OpenUrlCrossRefPubMedWeb of Science
  17. 17.↵
    Usui S, Oveson BC, Lee SY, Jo YJ, Yoshida T, Miki A, et al. NADPH oxidase plays a central role in cone cell death in retinitis pigmentosa. J Neurochem. 2009;110(3):1028–37.
    OpenUrlCrossRefPubMedWeb of Science
  18. 18.
    Xue Y, Cepko CL. Gene Therapies for Retinitis Pigmentosa that Target Glucose Metabolism. Cold Spring Harb Perspect Med. 2023;a041289.
  19. 19.↵
    Wang W, Kini A, Wang Y, Liu T, Chen Y, Vukmanic E, et al. Metabolic Deregulation of the Blood-Outer Retinal Barrier in Retinitis Pigmentosa. Cell Rep. 2019;28(5):1323–1334.e4.
    OpenUrlCrossRef
  20. 20.↵
    Wang SK, Xue Y, Cepko CL. Microglia modulation by TGF-β1 protects cones in mouse models of retinal degeneration. J Clin Invest. 2020;130(8):4360–9.
    OpenUrlCrossRef
  21. 21.↵
    Ruiz-Pastor MJ, Kutsyr O, Lax P, Cuenca N. Decrease in DHA and other fatty acids correlates with photoreceptor degeneration in retinitis pigmentosa. Exp Eye Res. 2021;209.
  22. 22.↵
    Li X, Cai S, He Z, Reilly J, Zeng Z, Strang N, et al. Metabolomics in Retinal Diseases: An Update. Biology (Basel). 2021;10(10).
  23. 23.
    Zhang E, Ryu J, Levi SR, Oh JK, Hsu CW, Cui X, et al. PKM2 ablation enhanced retinal function and survival in a preclinical model of retinitis pigmentosa. Mammalian Genome. 2020;31(3–4):77–85.
    OpenUrlCrossRef
  24. 24.
    Weiss ER, Osawa S, Xiong Y, Dhungana S, Carlson J, McRitchie S, et al. Broad spectrum metabolomics for detection of abnormal metabolic pathways in a mouse model for retinitis pigmentosa. Exp Eye Res. 2019;184:135–45.
    OpenUrl
  25. 25.
    Wert KJ, Velez G, Kanchustambham VL, Shankar V, Evans LP, Sengillo JD, et al. Metabolite therapy guided by liquid biopsy proteomics delays retinal neurodegeneration. EBioMedicine. 2020;52:102636.
  26. 26.↵
    Osada H, Toda E, Homma K, Guzman NA, Nagai N, Ogawa M, et al. ADIPOR1 deficiency-induced suppression of retinal ELOVL2 and docosahexaenoic acid levels during photoreceptor degeneration and visual loss. Cell Death & Disease 2021 12:5. 2021;12(5):1–13.
    OpenUrl
  27. 27.↵
    Wert KJ, Velez G, Kanchustambham VL, Shankar V, Evans LP, Sengillo JD, et al. Metabolite therapy guided by liquid biopsy proteomics delays retinal neurodegeneration. EBioMedicine. 2020;52.
  28. 28.↵
    Rao SS, Satapathy M, Sitaramayya A. Copper metabolism in retinitis pigmentosa patients. Br J Ophthalmol. 1981;65(2):127–30.
    OpenUrlAbstract/FREE Full Text
  29. 29.↵
    Hoffman DR, Uauy R, Birch DG. Metabolism of omega-3 fatty acids in patients with autosomal dominant retinitis pigmentosa. Exp Eye Res. 1995;60(3):279–89.
    OpenUrlCrossRefPubMedWeb of Science
  30. 30.
    Gong J, Rosner B, Rees DG, Berson EL, Weigel-DiFranco CA, Schaefer EJ. Plasma docosahexaenoic acid levels in various genetic forms of retinitis pigmentosa. Invest Ophthalmol Vis Sci. 1992;33(9):2596–602.
    OpenUrlAbstract/FREE Full Text
  31. 31.↵
    Berson EL, Schmidt SY, Rabin AR. Plasma amino-acids in hereditary retinal disease. Ornithine, lysine, and taurine. Br J Ophthalmol. 1976;60(2):142–7.
    OpenUrlAbstract/FREE Full Text
  32. 32.↵
    Guijas C, Montenegro-Burke JR, Warth B, Spilker ME, Siuzdak G. Metabolomics activity screening for identifying metabolites that modulate phenotype. Nat Biotechnol. 2018;36(4):316–20.
    OpenUrlCrossRefPubMed
  33. 33.
    Johnson CH, Ivanisevic J, Siuzdak G. Metabolomics: beyond biomarkers and towards mechanisms. Nat Rev Mol Cell Biol. 2016;17(7):451–9.
    OpenUrlCrossRefPubMed
  34. 34.
    Patti GJ, Yanes O, Siuzdak G. Innovation: Metabolomics: the apogee of the omics trilogy. Nat Rev Mol Cell Biol. 2012;13(4):263–9.
    OpenUrlCrossRefPubMed
  35. 35.↵
    Schrimpe-Rutledge AC, Codreanu SG, Sherrod SD, McLean JA. Untargeted Metabolomics Strategies-Challenges and Emerging Directions. J Am Soc Mass Spectrom. 2016;27(12):1897–905.
    OpenUrlCrossRef
  36. 36.↵
    Li M, Li H, Jiang P, Liu X, Xu D, Wang F. Investigating the pathological processes of rhegmatogenous retinal detachment and proliferative vitreoretinopathy with metabolomics analysis. Mol Biosyst. 2014;10(5):1055–62.
    OpenUrl
  37. 37.↵
    Dilworth L, Facey A, Omoruyi F. Diabetes Mellitus and Its Metabolic Complications: The Role of Adipose Tissues. Int J Mol Sci. 2021;22(14).
  38. 38.↵
    Du X, Yang L, Kong L, Sun Y, Shen K, Cai Y, et al. Metabolomics of various samples advancing biomarker discovery and pathogenesis elucidation for diabetic retinopathy. Front Endocrinol (Lausanne). 2022;13.
  39. 39.
    Xia JF, Wang Z hua, Liang QL, Wang YM, Li P, Luo GA. Correlations of six related pyrimidine metabolites and diabetic retinopathy in Chinese type 2 diabetic patients. Clin Chim Acta. 2011;412(11–12):940–5.
    OpenUrlCrossRefPubMed
  40. 40.
    Rhee SY, Jung ES, Park HM, Jeong SJ, Kim K, Chon S, et al. Plasma glutamine and glutamic acid are potential biomarkers for predicting diabetic retinopathy. Metabolomics. 2018;14(7).
  41. 41.
    Sun Y, Zou H, Li X, Xu S, Liu C. Plasma Metabolomics Reveals Metabolic Profiling For Diabetic Retinopathy and Disease Progression. Front Endocrinol (Lausanne). 2021;12.
  42. 42.
    Acar İE, Lores-Motta L, Colijn JM, Meester-Smoor MA, Verzijden T, Cougnard-Gregoire A, et al. Integrating Metabolomics, Genomics, and Disease Pathways in Age-Related Macular Degeneration: The EYE-RISK Consortium. Ophthalmology. 2020;127(12):1693–709.
    OpenUrl
  43. 43.
    Kersten E, Paun CC, Schellevis RL, Hoyng CB, Delcourt C, Lengyel I, et al. Systemic and ocular fluid compounds as potential biomarkers in age-related macular degeneration. Surv Ophthalmol. 2018;63(1):9–39.
    OpenUrlCrossRefPubMed
  44. 44.
    Ambreen F, Khan WA, Qureshi N, Qureshi IZ. Assessment of serum lipids in patients with age related macular degeneration from Pakistan. The Journal of the Pakistan Medical Association. 2014;64(6):664–9.
    OpenUrl
  45. 45.
    Čolak E, Kosanović-Jaković N, Žorić L, Radosavljević A, Stanković S, Majkić-Singh N. The association of lipoprotein parameters and C-reactive protein in patients with age-related macular degeneration. Ophthalmic Res. 2011;46(3):125–32.
    OpenUrlCrossRefPubMedWeb of Science
  46. 46.
    Javadzadeh A, Ghorbanihaghjo A, Javadzadeh A, Rashtchizadeh N, Sorkhabi R, Khalili H, et al. Osteoprotegerin and Soluble Receptor Activator of Nuclear Factor-Kappa B Ligand in Exudative Age-Related Macular Degeneration. Acta Med Iran. 2014;52(4):265–70.
    OpenUrl
  47. 47.
    Orban T, Johnson WM, Dong Z, Maeda T, Maeda A, Sakai T, et al. Serum levels of lipid metabolites in age-related macular degeneration. FASEB J. 2015;29(11):4579– 88.
    OpenUrlCrossRefPubMed
  48. 48.
    Laíns I, Duarte D, Barros AS, Martins AS, Gil J, Miller JB, et al. Human plasma metabolomics in age-related macular degeneration (AMD) using nuclear magnetic resonance spectroscopy. PLoS One. 2017;12(5).
  49. 49.↵
    Laíns I, Chung W, Kelly RS, Gil J, Marques M, Barreto P, et al. Human Plasma Metabolomics in Age-Related Macular Degeneration: Meta-Analysis of Two Cohorts. Metabolites. 2019;9(7).
  50. 50.↵
    factoextra: Extract and Visualize the Results of Multivariate Data Analyses [R package factoextra version 1.0.7]. 2020;
  51. 51.↵
    Wickham H. ggplot2: Elegant Graphics for Data Analysis. Springer-Verlag New York; 2016.
  52. 52.↵
    Guangchuang Yu M. Package “clusterProfiler” Type Package Title A universal enrichment tool for interpreting omics data. 2023;
  53. 53.↵
    Funabashi M, Grove TL, Wang M, Varma Y, McFadden ME, Brown LC, et al. A metabolic pathway for bile acid dehydroxylation by the gut microbiome. Nature. 2020;582(7813):566–70.
    OpenUrlCrossRefPubMed
  54. 54.↵
    Martinot E, Sèdes L, Baptissart M, Lobaccaro JM, Caira F, Beaudoin C, et al. Bile acids and their receptors. Mol Aspects Med. 2017;56:2–9.
    OpenUrl
  55. 55.
    Wahlström A, Sayin SI, Marschall HU, Bäckhed F. Intestinal Crosstalk between Bile Acids and Microbiota and Its Impact on Host Metabolism. Cell Metab. 2016;24(1):41– 50.
    OpenUrlCrossRefPubMed
  56. 56.
    Ajouz H, Mukherji D, Shamseddine A. Secondary bile acids: an underrecognized cause of colon cancer. World J Surg Oncol. 2014;12(1).
  57. 57.↵
    Bayerdörffer E, Mannes GA, Ochsenkühn T, Dirschedl P, Wiebecke B, Paumgartner G. Unconjugated secondary bile acids in the serum of patients with colorectal adenomas. Gut. 1995;36(2):268–73.
    OpenUrlAbstract/FREE Full Text
  58. 58.↵
    Chiang JYL. Linking Sex Differences in Non-Alcoholic Fatty Liver Disease to Bile Acid Signaling, Gut Microbiota, and High Fat Diet. Am J Pathol. 2017;187(8):1658–9.
    OpenUrl
  59. 59.
    Tomkin GH, Owens D. Obesity diabetes and the role of bile acids in metabolism. J Transl Int Med. 2016;4(2):73–80.
    OpenUrl
  60. 60.↵
    Sonne DP, Hansen M, Knop FK. Bile acid sequestrants in type 2 diabetes: potential effects on GLP1 secretion. Eur J Endocrinol. 2014;171(2).
  61. 61.↵
    Behr C, Slopianka M, Haake V, Strauss V, Sperber S, Kamp H, et al. Analysis of metabolome changes in the bile acid pool in feces and plasma of antibiotic-treated rats. Toxicol Appl Pharmacol. 2019;363:79–87.
    OpenUrl
  62. 62.↵
    Shonsey EM, Sfakianos M, Johnson M, He D, Falany CN, Falany J, et al. Bile acid coenzyme A: amino acid N-acyltransferase in the amino acid conjugation of bile acids. Methods Enzymol. 2005;400:374–94.
    OpenUrlCrossRefPubMed
  63. 63.↵
    Neusüß C, Gnauk T, Plewka A, Herrmann H, Quinn PK. Carbonaceous aerosol over the Indian Ocean: OC/EC fractions and selected specifications from size-segregated onboard samples. Journal of Geophysical Research: Atmospheres. 2002;107(D19):INX2 30–1.
    OpenUrl
  64. 64.↵
    Li XB, Gu JD, Zhou QH. Review of aerobic glycolysis and its key enzymes - new targets for lung cancer therapy. Thorac Cancer. 2015;6(1):17–24.
    OpenUrl
  65. 65.↵
    Ganapathy-Kanniappan S, Geschwind JFH. Tumor glycolysis as a target for cancer therapy: progress and prospects. Mol Cancer. 2013;12(1).
  66. 66.↵
    Russell DW, Setchell KDR. Bile acid biosynthesis. Biochemistry. 1992;31(20):4737– 49.
    OpenUrlCrossRefPubMedWeb of Science
  67. 67.
    Chiang JY. Regulation of bile acid synthesis. Front Biosci. 1998;3.
  68. 68.↵
    Chiang JYL, Li T. Regulation of bile acid and cholesterol metabolism by PPARs. PPAR Res. 2009;2009.
  69. 69.↵
    Daruich A, Picard E, Boatright JH, Behar-Cohen F. Review: The bile acids urso- and tauroursodeoxycholic acid as neuroprotective therapies in retinal disease. Mol Vis. 2019;25:610.
  70. 70.↵
    Pardue MT, Allen RS. Neuroprotective strategies for retinal disease. Prog Retin Eye Res. 2018;65:50–76.
    OpenUrlPubMed
  71. 71.
    Ackerman HD, Gerhard GS. Bile Acids in Neurodegenerative Disorders. Front Aging Neurosci. 2016;8(NOV).
  72. 72.
    Fernández-Sánchez L, Lax P, Noailles A, Angulo A, Maneu V, Cuenca N. Natural Compounds from Saffron and Bear Bile Prevent Vision Loss and Retinal Degeneration. Molecules. 2015;20(8):13875–93.
    OpenUrl
  73. 73.↵
    Win A, Delgado A, Jadeja RN, Martin PM, Bartoli M, Thounaojam MC. Pharmacological and Metabolic Significance of Bile Acids in Retinal Diseases. Biomolecules. 2021;11(2):1–18.
    OpenUrlCrossRef
  74. 74.↵
    Gaspar JM, Martins A, Cruz R, Rodrigues CMP, Ambrósio AF, Santiago AR. Tauroursodeoxycholic acid protects retinal neural cells from cell death induced by prolonged exposure to elevated glucose. Neuroscience. 2013;253:380–8.
    OpenUrlCrossRefPubMed
  75. 75.↵
    Ridlon JM, Kang DJ, Hylemon PB. Bile salt biotransformations by human intestinal bacteria. J Lipid Res. 2006;47(2):241–59.
    OpenUrlAbstract/FREE Full Text
  76. 76.↵
    Kiriyama Y, Nochi H. The Role of Gut Microbiota-Derived Lithocholic Acid, Deoxycholic Acid and Their Derivatives on the Function and Differentiation of Immune Cells. Microorganisms. 2023;11(11):2730.
    OpenUrl
  77. 77.↵
    Phillips MJ, Walker TA, Choi HY, Faulkner AE, Kim MK, Sidney SS, et al. Tauroursodeoxycholic acid preservation of photoreceptor structure and function in the rd10 mouse through postnatal day 30. Invest Ophthalmol Vis Sci. 2008;49(5):2148– 55.
    OpenUrlAbstract/FREE Full Text
  78. 78.↵
    Oveson BC, Iwase T, Hackett SF, Lee SY, Usui S, Sedlak TW, et al. Constituents of bile, bilirubin and TUDCA, protect against oxidative stress-induced retinal degeneration. J Neurochem. 2011;116(1):144–53.
    OpenUrlCrossRefPubMedWeb of Science
  79. 79.↵
    Salvador R, Zhang A, Horai R, Caspi RR. Microbiota as Drivers and as Therapeutic Targets in Ocular and Tissue Specific Autoimmunity. Front Cell Dev Biol. 2021;8.
  80. 80.↵
    Newsome DA, Anderson RE, May JG, McKay TA, Maude M. Clinical and serum lipid findings in a large family with autosomal dominant retinitis pigmentosa. Ophthalmology. 1988;95(12):1691–5.
    OpenUrl
  81. 81.
    Converse CA, Hammer HM, Packard CJ, Shepherd J. Plasma lipid abnormalities in retinitis pigmentosa and related conditions. Trans Ophthalmol Soc U K. 1983;103 ( Pt 5):508–12.
    OpenUrlPubMedWeb of Science
  82. 82.
    McColl AJ, Converse CA. Lipid studies in retinitis pigmentosa. Prog Lipid Res. 1995;34(1):1–16.
    OpenUrlCrossRefPubMedWeb of Science
  83. 83.↵
    Lewandowski D, Sander CL, Tworak A, Gao F, Xu Q, Skowronska-Krawczyk D. Dynamic lipid turnover in photoreceptors and retinal pigment epithelium throughout life. Prog Retin Eye Res. 2022;89.
  84. 84.↵
    Finkelstein S, Gospe SM, Schuhmann K, Shevchenko A, Arshavsky VY, Lobanova ES. Phosphoinositide Profile of the Mouse Retina. Cells. 2020;9(6).
  85. 85.↵
    Elner VM. Retinal pigment epithelial acid lipase activity and lipoprotein receptors: effects of dietary omega-3 fatty acids. Trans Am Ophthalmol Soc. 2002;100:301.
  86. 86.
    Fliesler SJ, Bretillon L. The ins and outs of cholesterol in the vertebrate retina. J Lipid Res. 2010;51(12):3399–413.
    OpenUrlAbstract/FREE Full Text
  87. 87.↵
    Gabrielle PH. Lipid metabolism and retinal diseases. Acta Ophthalmol. 2022;100 Suppl 269(S269):3–43.
    OpenUrl
  88. 88.↵
    Homma Y, Ozawa H, Kobayashi T, Yamaguchi H, Sakane H, Nakamura H. Effects of simvastatin on plasma lipoprotein subfractions, cholesterol esterification rate, and cholesteryl ester transfer protein in type II hyperlipoproteinemia. Atherosclerosis. 1995;114(2):223–34.
    OpenUrlCrossRefPubMedWeb of Science
  89. 89.
    Zhang J, Liu Q. Cholesterol metabolism and homeostasis in the brain. Protein Cell. 2015;6(4):254–64.
    OpenUrlCrossRefPubMed
  90. 90.
    Gulati S, Liu Y, Munkacsi AB, Wilcox L, Sturley SL. Sterols and sphingolipids: dynamic duo or partners in crime? Prog Lipid Res. 2010;49(4):353–65.
    OpenUrlCrossRefPubMed
  91. 91.↵
    Gonen A, Miller YI. From Inert Storage to Biological Activity-In Search of Identity for Oxidized Cholesteryl Esters. Front Endocrinol (Lausanne). 2020;11.
  92. 92.↵
    Phillips GR, Hancock SE, Brown SHJ, Jenner AM, Kreilaus F, Newell KA, et al. Cholesteryl ester levels are elevated in the caudate and putamen of Huntington’s disease patients. Sci Rep. 2020;10(1).
  93. 93.
    Kim JH, Ee SM, Jittiwat J, Ong ES, Farooqui AA, Jenner AM, et al. Increased expression of acyl-coenzyme A: cholesterol acyltransferase-1 and elevated cholesteryl esters in the hippocampus after excitotoxic injury. Neuroscience. 2011;185:125–34.
    OpenUrlCrossRef
  94. 94.
    Cutler RG, Pedersen WA, Camandola S, Rothstein JD, Mattson MP. Evidence that accumulation of ceramides and cholesterol esters mediates oxidative stress-induced death of motor neurons in amyotrophic lateral sclerosis. Ann Neurol. 2002;52(4):448– 57.
    OpenUrlCrossRefPubMedWeb of Science
  95. 95.
    Shah SN, Johnson RC. Activity levels of cholesterol ester metabolizing enzymes in brain in multiple sclerosis: correlation with cholesterol ester concentrations. Exp Neurol. 1980;68(3):601–4.
    OpenUrlPubMed
  96. 96.↵
    Astarita G, Jung KM, Vasilevko V, DiPatrizio N V., Martin SK, Cribbs DH, et al. Elevated stearoyl-CoA desaturase in brains of patients with Alzheimer’s disease. PLoS One. 2011;6(10).
  97. 97.↵
    Curcio CA, Johnson M, Rudolf M, Huang JD. The oil spill in ageing Bruch membrane. Br J Ophthalmol. 2011;95(12):1638–45.
    OpenUrlAbstract/FREE Full Text
  98. 98.↵
    Ngwenya BZ, Yamamoto N. Activation of peritoneal macrophages by lysophosphatidylcholine. Biochim Biophys Acta. 1985;839(1):9–15.
    OpenUrlCrossRefPubMed
  99. 99.↵
    Knuplez E, Marsche G. An Updated Review of Pro- and Anti-Inflammatory Properties of Plasma Lysophosphatidylcholines in the Vascular System. Int J Mol Sci. 2020;21(12):1–18.
    OpenUrlCrossRefPubMed
  100. 100.↵
    Cha MH, Lee SM, Jung J. Lysophosphatidylcholine induces expression of genes involved in cholesterol biosynthesis in THP-1 derived macrophages. Steroids. 2018;139:28–34.
    OpenUrl
  101. 101.↵
    Englberger W, Bitter-Suermann D, Hadding U. Influence of lysophospholipids and PAF on the oxidative burst of PMNL. Int J Immunopharmacol. 1987;9(3):275–82.
    OpenUrlCrossRefPubMedWeb of Science
  102. 102.↵
    Friedman JS, Chang B, Krauth DS, Lopez I, Waseem NH, Hurd RE, et al. Loss of lysophosphatidylcholine acyltransferase 1 leads to photoreceptor degeneration in rd11 mice. Proc Natl Acad Sci U S A. 2010;107(35):15523–8.
    OpenUrlAbstract/FREE Full Text
  103. 103.↵
    Cheng L, Han X, Shi Y. A regulatory role of LPCAT1 in the synthesis of inflammatory lipids, PAF and LPC, in the retina of diabetic mice. Am J Physiol Endocrinol Metab. 2009;297(6).
  104. 104.↵
    Yoshinari M, Shi AH, Yoshizumi H, Wakisaka M, Iwase M, Fujishima M. Probucol reduces lysophosphatidylcholines in low-density lipoprotein. Eur J Clin Pharmacol. 2000;55(11–12):787–92.
    OpenUrlCrossRefPubMedWeb of Science
Back to top
PreviousNext
Posted February 23, 2024.
Download PDF

Supplementary Material

Data/Code
Email

Thank you for your interest in spreading the word about medRxiv.

NOTE: Your email address is requested solely to identify you as the sender of this article.

Enter multiple addresses on separate lines or separate them with commas.
The Gut Metabolism is altered in patients with CRB1-associated inherited retinal degeneration
(Your Name) has forwarded a page to you from medRxiv
(Your Name) thought you would like to see this page from the medRxiv website.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
The Gut Metabolism is altered in patients with CRB1-associated inherited retinal degeneration
Lude Moekotte, Joke H. de Boer, Sanne Hiddingh, Bram Gerritsen, Jutta Lintelmann, Alexander Cecil, L. Ingeborgh van den Born, Xuan-Thanh-An Nguyen, Camiel J.F. Boon, Maria M. van Genderen, Jonas J.W. Kuiper
medRxiv 2024.02.22.24303210; doi: https://doi.org/10.1101/2024.02.22.24303210
Twitter logo Facebook logo LinkedIn logo Mendeley logo
Citation Tools
The Gut Metabolism is altered in patients with CRB1-associated inherited retinal degeneration
Lude Moekotte, Joke H. de Boer, Sanne Hiddingh, Bram Gerritsen, Jutta Lintelmann, Alexander Cecil, L. Ingeborgh van den Born, Xuan-Thanh-An Nguyen, Camiel J.F. Boon, Maria M. van Genderen, Jonas J.W. Kuiper
medRxiv 2024.02.22.24303210; doi: https://doi.org/10.1101/2024.02.22.24303210

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Subject Area

  • Ophthalmology
Subject Areas
All Articles
  • Addiction Medicine (349)
  • Allergy and Immunology (668)
  • Allergy and Immunology (668)
  • Anesthesia (181)
  • Cardiovascular Medicine (2648)
  • Dentistry and Oral Medicine (316)
  • Dermatology (223)
  • Emergency Medicine (399)
  • Endocrinology (including Diabetes Mellitus and Metabolic Disease) (942)
  • Epidemiology (12228)
  • Forensic Medicine (10)
  • Gastroenterology (759)
  • Genetic and Genomic Medicine (4103)
  • Geriatric Medicine (387)
  • Health Economics (680)
  • Health Informatics (2657)
  • Health Policy (1005)
  • Health Systems and Quality Improvement (985)
  • Hematology (363)
  • HIV/AIDS (851)
  • Infectious Diseases (except HIV/AIDS) (13695)
  • Intensive Care and Critical Care Medicine (797)
  • Medical Education (399)
  • Medical Ethics (109)
  • Nephrology (436)
  • Neurology (3882)
  • Nursing (209)
  • Nutrition (577)
  • Obstetrics and Gynecology (739)
  • Occupational and Environmental Health (695)
  • Oncology (2030)
  • Ophthalmology (585)
  • Orthopedics (240)
  • Otolaryngology (306)
  • Pain Medicine (250)
  • Palliative Medicine (75)
  • Pathology (473)
  • Pediatrics (1115)
  • Pharmacology and Therapeutics (466)
  • Primary Care Research (452)
  • Psychiatry and Clinical Psychology (3432)
  • Public and Global Health (6527)
  • Radiology and Imaging (1403)
  • Rehabilitation Medicine and Physical Therapy (814)
  • Respiratory Medicine (871)
  • Rheumatology (409)
  • Sexual and Reproductive Health (410)
  • Sports Medicine (342)
  • Surgery (448)
  • Toxicology (53)
  • Transplantation (185)
  • Urology (165)