scholarly journals THU0014 COMPARATIVE TRANSCRIPTOME ANALYSES ACROSS TISSUES AND SPECIES IDENTIFY TARGETABLE GENES FOR HUMAN SYSTEMIC LUPUS ERYTHEMATOSUS (SLE) AND LUPUS NEPHRITIS (LN)

2020 ◽  
Vol 79 (Suppl 1) ◽  
pp. 220.1-220
Author(s):  
E. Frangou ◽  
P. Garantziotis ◽  
M. Grigoriou ◽  
A. Banos ◽  
N. Panousis ◽  
...  

Background:Systemic Lupus Erythematosus (SLE) is a complex disease associated with the dysfunction of multiple tissues and cells. The causal tissue for each disease phenotype is not known a priori. Despite improvements in diagnosis and treatment, major organ involvement (such as the kidneys) contributes significantly to morbidity and mortality that still remain increased. There is an unmet need for timely targeted therapy.Objectives:RNA-sequencing was performed to investigate the patterns of transcription variation across tissues between healthy and lupus-prone mice at different stages of lupus, and how these patterns associate with human Systemic Lupus Erythematosus (SLE).Methods:NZB/W-F1 lupus prone mice were sacrificed at the pre-puberty, pre-autoimmunity and nephritic stage. Age-matched C57BL/6 were used as controls. An “effector” tissue (spleen) and “end-organs” (kidneys, brain) were collected. Total RNA was isolated, and mRNA-sequencing was performed. A time-series analysis was developed and differentially expressed genes (DEGs) were analyzed with DESeq. Hierarchical clustering and functional enrichment analysis were performed with gProfiler. Human orthologs of mouse tissue DEGs were identified in the whole-blood RNA-sequencing dataset comprised of 55 lupus-nephritis (LN), 65 non-LN SLE patients and 58 healthy individuals (HI). Human orthologs were compared to human DEGs. Using machine learning, human orthologs identified in the mouse dataset were used to predict kidney involvement in the human dataset, which was split in training and validation sets.Results:Lupus susceptibility and progression signatures at different tissues and different stages of the disease were identified. Tissue-specific signatures and a common cross-tissue signature were also described. Previously described and novel biological processes and pathways were revealed. The comparative murine-human transcriptome analysis identified human orthologs from the mouse spleen-signature (including CCL5, IFIT and HLA genes) that are involved in systemic autoimmunity. It also identified human orthologs from the kidney- and brain-signature (including FCGR2A, C1Q, JAK1 and APOA2) that are involved in major “end-organ” damage and response mechanisms. Using a neural network model, 193 human orthologs accurately predicted LN patients vs HI (accuracy=0.86, sensitivity=0.82, specificity=0.91 in the validation set). Using a support vector machine model, 30 human orthologs and age and gender were the best predictors of LN vs non-LN SLE patients (accuracy=0.71, sensitivity=0.73, specificity=0.69 in the validation set).Conclusion:Murine tissue gene signatures identified by RNA-sequencing analysis revealed biological processes and pathways that could be potentially used as biomarkers or therapeutic targets in human SLE. Comparison of the murine tissue-transcriptome with the whole-blood human-transcriptome revealed common gene signatures, demonstrating similar biological processes and pathways. Machine learning identified a murine kidney lupus signature that can accurately predict kidney involvement in human SLE. Validation in other datasets is ongoing.References:[1]Panousis NI, et al. Ann Rheum Dis 2019;78:1079Acknowledgments:This work was supported by FOREUM, SYSCID and ERC -Advanced GrantDisclosure of Interests:Eleni Frangou: None declared, Panayiotis Garantziotis: None declared, Maria Grigoriou: None declared, Aggelos Banos: None declared, Nikolaos Panousis: None declared, Emmanouil Dermitzakis: None declared, George Bertsias Grant/research support from: GSK, Consultant of: Novartis, Dimitrios Boumpas: None declared, Anastasia Filia: None declared

2020 ◽  
Vol 35 (Supplement_3) ◽  
Author(s):  
Eleni Frangou ◽  
Panagiotis Garantziotis ◽  
Maria Grigoriou ◽  
Angelos Banos ◽  
Nikolaos Panousis ◽  
...  

Abstract Background and Aims Systemic Lupus Erythematosus (SLE) is a complex disease associated with non-synchronous multi-tissue dysfunction of varying severity. Involvement of major organs, such as kidneys, contributes significantly to morbidity and mortality. We sought to unravel new pathways to be used as potential biomarkers and therapeutic targets. To this end, we compared the patterns of gene transcription -as determined by the use of RNA-sequencing- across tissues between healthy and lupus-prone mice at different stages of the disease, and explored their implications for human disease and prediction of kidney involvement, in our whole blood RNA sequencing dataset comprised of 120 SLE [55 lupus nephritis (LN), 65 non-LN] patients and 58 healthy individuals (HI). Method NZB/W-F1 lupus-prone mice were sacrificed at the pre-puberty, pre-autoimmunity and nephritic stage. Age-matched C57BL/6 mice were used as controls. An “effector” tissue (spleen) and major end-organ tissues (kidneys, brain) were collected. Total RNA was isolated, and mRNA-sequencing was performed. Differential expression and time-series analyses were performed using DESeq2. Differentially expressed genes (DEGs) were hierarchically clustered and functionally interpreted using gProfiler enrichment analysis. Human orthologous genes of mouse common DEGs in each tissue and across all disease stages, were compared to human DEGs. Using machine learning techniques, human orthologs identified in the common DEGs across all stages in the kidneys of lupus-prone vs healthy mice were used to predict kidney involvement in the human dataset, which was split in training and validation sets. Results: Gene signatures: the common cross-tissue signature was identified by the comparison of DEGs between tissues of lupus-prone vs healthy mice at each stage of the disease. A total 134 genes (including C4A, LYRM7 and HDDC3) were found, suggesting their involvement in a common pathogenic mechanism across “effector” and end-organ tissues. Tissue-specific signatures showed enrichment of FCERI mediated NF-kB activation pathway in the spleen, steroid hormone biosynthesis pathway in the kidney and phosphatidylcholine metabolic process in the brain, suggesting distinct pathways implicated in end-organ injury. Comparative murine-human transcriptome analysis: 76 human orthologs (including CCL5, IFIT and HLA genes) identified in the murine spleen signature were also differentially expressed in SLE patients vs HI, suggesting their involvement in systemic autoimmunity. A total of 68 human orthologs (including FCGR2A, C1R and JAK1) identified in the mouse kidney-signature and 25 human orthologs (including APOA2) identified in the mouse brain-signature, were differentially expressed in LN patients vs HI and neuropsychiatric SLE patients vs HI, respectively. Kidney involvement prediction in human SLE: using a neural network model, 193 human orthologs predicted LN patients vs HI with high accuracy (accuracy=0.86, sensitivity=0.82, specificity=0.91 in the validation set). Using a support vector machine model, 30 human orthologs and age and gender were the best predictors of LN vs non-LN SLE patients (accuracy=0.71, sensitivity=0.73, specificity=0.69 in the validation set). Conclusion Murine RNA-sequencing uncovered both shared cross-tissue and tissue-specific gene signatures that could be potentially targeted in SLE. Murine-human comparative transcriptome analysis revealed common gene signatures suggesting that similar biological processes and pathways are disturbed across species, with murine kidney lupus signature predicting kidney involvement in human SLE. Validation in other datasets is ongoing.


Author(s):  
Francis R. Comerford ◽  
Alan S. Cohen

Mice of the inbred NZB strain develop a spontaneous disease characterized by autoimmune hemolytic anemia, positive lupus erythematosus cell tests and antinuclear antibodies and nephritis. This disease is analogous to human systemic lupus erythematosus. In ultrastructural studies of the glomerular lesion in NZB mice, intraglomerular dense deposits in mesangial, subepithelial and subendothelial locations were described. In common with the findings in many examples of human and experimental nephritis, including many cases of human lupus nephritis, these deposits were amorphous or slightly granular in appearance with no definable substructure.We have recently observed structured deposits in the glomeruli of NZB mice. They were uncommon and were found in older animals with severe glomerular lesions by morphologic criteria. They were seen most commonly as extracellular elements in subendothelial and mesangial regions. The deposits ranged up to 3 microns in greatest dimension and were often adjacent to deposits of lipid-like round particles of 30 to 250 millimicrons in diameter and with amorphous dense deposits.


Lupus ◽  
2021 ◽  
pp. 096120332098345
Author(s):  
Alessandra Ida Celia ◽  
Roberta Priori ◽  
Bruna Cerbelli ◽  
Francesca Diomedi-Camassei ◽  
Vincenzo Leuzzi ◽  
...  

Proteinuria is one of the most typical manifestations of kidney involvement in Systemic Lupus Erythematosus (SLE). We report the case of a 23-year-old woman with a 6-year-long history of SLE presenting with proteinuria after a three-year remission on hydroxychloroquine. Kidney histological examination showed alterations inconsistent with lupus nephritis and suggestive of hydroxychloroquine toxicity or Fabry disease. The latter was confirmed by genetic assay.


2011 ◽  
Vol 32 (11) ◽  
pp. 1268-1277 ◽  
Author(s):  
Jianming Wu ◽  
Fenglong Xie ◽  
Kun Qian ◽  
Andrew W. Gibson ◽  
Jeffrey C. Edberg ◽  
...  

2021 ◽  
Vol 11 ◽  
Author(s):  
Benoit Brilland ◽  
Emeline Vinatier ◽  
Jean-François Subra ◽  
Pascale Jeannin ◽  
Jean-François Augusto ◽  
...  

Pentraxins are soluble innate immunity receptors involved in sensing danger molecules. They are classified as short (CRP, SAP) and long pentraxin subfamilies, including the prototypic long pentraxin PTX3. Pentraxins act mainly as bridging molecules favoring the clearance of microbes and dead cells. They are also involved in many other biological processes, such as regulation of complement activation, inflammation and tissue homeostasis. Autoantibodies directed against pentraxins have been reported in various autoimmune diseases, especially in systemic lupus erythematosus and ANCA-associated vasculitis. In this review, we review the main biological characteristics and functions of pentraxins and summarize data concerning autoantibodies directed against pentraxins in the context of autoimmune diseases and discuss their potential pathological role.


1999 ◽  
Vol 42 (9) ◽  
pp. 1902-1907 ◽  
Author(s):  
Kathy L. Moser ◽  
Courtney Gray-McGuire ◽  
Jennifer Kelly ◽  
Neeraj Asundi ◽  
Hua Yu ◽  
...  

Lupus ◽  
2004 ◽  
Vol 13 (8) ◽  
pp. 575-583 ◽  
Author(s):  
M García ◽  
M E Colombani-Vidal ◽  
C C Zylbersztein ◽  
A Testi ◽  
J Marcos ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document