71 results on '"Berthier CC"'
Search Results
2. 41 Relevance of mouse lupus models of lupus nephritis to progression of ckd
- Author
-
Berthier, CC, primary, Kretzler, M, additional, and Davidson, A, additional
- Published
- 2017
- Full Text
- View/download PDF
3. 264 Pearl: pathway exploration and analysis in renal disease in the accelerating medicine partnership (amp) lupus network
- Author
-
Berthier, CC, primary, Rao, DA, additional, Arazi, A, additional, Browne, EP, additional, Eisenhaure, T, additional, Hacohen, N, additional, Lieb, D, additional, Diamond, B, additional, and Kretzler, M, additional
- Published
- 2017
- Full Text
- View/download PDF
4. Experimental heart transplantation: effect of cyclosporine on expression and activity of metzincins
- Author
-
Berthier, CC, primary, Pally, C, additional, Weckbecker, G, additional, Raulf, F, additional, Rehrauer, H, additional, Wagner, U, additional, and Le, Hir, additional
- Published
- 2009
- Full Text
- View/download PDF
5. Lupus-prone NZM2328 mice exhibit enhanced UV-induced myeloid cell recruitment and activation in a type I interferon dependent manner.
- Author
-
Maz MP, Reddy AL, Berthier CC, Tsoi LC, Colesa DJ, Wolf SJ, Shi H, Loftus SN, Moallemian R, Bogle R, Kretzler M, Jacob CO, Gudjonsson JE, and Kahlenberg JM
- Subjects
- Animals, Mice, Female, Mice, Knockout, Skin immunology, Skin pathology, Skin radiation effects, Skin metabolism, Mice, Inbred BALB C, Receptor, Interferon alpha-beta genetics, Receptor, Interferon alpha-beta metabolism, Ultraviolet Rays adverse effects, Interferon Type I metabolism, Lupus Erythematosus, Systemic etiology, Lupus Erythematosus, Systemic immunology, Myeloid Cells immunology, Myeloid Cells metabolism, Disease Models, Animal
- Abstract
Though the exact causes of systemic lupus erythematosus (SLE) remain unknown, exposure to ultraviolet (UV) light is one of the few well-known triggers of cutaneous inflammation in SLE. However, the precise cell types which contribute to the early cutaneous inflammatory response in lupus, and the ways that UV dosing and interferons modulate these findings, have not been thoroughly dissected. Here, we explore these questions using the NZM2328 spontaneous murine model of lupus. In addition, we use iNZM mice, which share the NZM2328 background but harbor a whole-body knockout of the type I interferon (IFN) receptor, and wild-type BALB/c mice. 10-13-week-old female mice of each strain were treated with acute (300 mJ/cm
2 x1), chronic (100 mJ/cm2 daily x5 days), or no UVB, and skin was harvested and processed for bulk RNA sequencing and flow cytometry. We identify that inflammatory pathways and gene signatures related to myeloid cells - namely neutrophils and monocyte-derived dendritic cells - are a shared feature of the acute and chronic UVB response in NZM skin greater than iNZM and wild-type skin. We also verify recruitment and activation of these cells by flow cytometry in both acutely and chronically irradiated NZM and WT mice and demonstrate that these processes are dependent on type I IFN signaling. Taken together, these data indicate a skewed IFN-driven inflammatory response to both acute and chronic UVB exposure in lupus-prone skin dominated by myeloid cells, suggesting both the importance of type I IFNs and myeloid cells as therapeutic targets for photosensitive patients and highlighting the risks of even moderate UV exposure in this patient population., (Copyright © 2024 Elsevier Ltd. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
6. Towards Risk Stratification in Clinical Care for IgA Nephropathy: Genetic Risk Scores: Comments on the Study "Clinical Application of Polygenic Risk Score in IgA Nephropathy".
- Author
-
Berthier CC and Ju W
- Abstract
Competing Interests: Conflict of InterestOn behalf of all authors, the corresponding author states that there is no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
7. Disease heterogeneity and molecular classification of inflammatory palmoplantar diseases.
- Author
-
van Straalen KR, Kirma J, Yee CM, Bugada LF, Rizvi SM, Wen F, Wasikowski R, Fox J, Do TH, Schuler CF 4th, Xing E, MacLeod AS, Harms PW, Berthier CC, Kahlenberg JM, Leung MWL, Tsoi LC, and Gudjonsson JE
- Subjects
- Humans, Male, Female, Adult, Middle Aged, Transcriptome, Cytokines immunology, Cytokines genetics, Skin pathology, Skin immunology, Aged, Eczema classification, Eczema immunology, Eczema genetics, Psoriasis immunology, Psoriasis genetics, Psoriasis classification
- Abstract
Background: Palmoplantar pustulosis (PPP) is an inflammatory disease characterized by relapsing eruptions of neutrophil-filled, sterile pustules on the palms and soles that can be clinically difficult to differentiate from non-pustular palmoplantar psoriasis (palmPP) and dyshidrotic palmoplantar eczema (DPE)., Objective: We sought to identify overlapping and unique PPP, palmPP, and DPE drivers to provide molecular insight into their pathogenesis., Methods: We performed bulk RNA sequencing of lesional PPP (n = 33), palmPP (n = 5), and DPE (n = 28) samples, as well as 5 healthy nonacral and 10 healthy acral skin samples., Results: Acral skin showed a unique immune environment, likely contributing to a unique niche for palmoplantar inflammatory diseases. Compared to healthy acral skin, PPP, palmPP, and DPE displayed a broad overlapping transcriptomic signature characterized by shared upregulation of proinflammatory cytokines (TNF, IL-36), chemokines, and T-cell-associated genes, along with unique disease features of each disease state, including enriched neutrophil processes in PPP and to a lesser extent in palmPP, and lipid antigen processing in DPE. Strikingly, unsupervised clustering and trajectory analyses demonstrated divergent inflammatory profiles within the 3 disease states. These identified putative key upstream immunologic switches, including eicosanoids, interferon responses, and neutrophil degranulation, contributing to disease heterogeneity., Conclusion: A molecular overlap exists between different inflammatory palmoplantar diseases that supersedes clinical and histologic assessment. This highlights the heterogeneity within each condition, suggesting limitations of current disease classification and the need to move toward a molecular classification of inflammatory acral diseases., Competing Interests: Disclosure statement Supported by an unrestricted research grant from Janssen. Additional support was provided by P30-AR075043 (J.E.G., L.C.T.), the Alfred A. Taubman Medical Research Institute (J.E.G., J.M.K.), and the George M. O’Brien Michigan Kidney Translational Research Core Center P30DK081943 (C.C.B.). C.F.S. is supported by K23–NIH/NIAID K23AI162661. F.W. is supported by the Taubman Institute Innovation Projects program. Disclosure of potential conflict of interest: K. R. van Straalen has received honoraria from Novartis, Boehringer-Ingelheim, and UCB. A. S. MacLeod is currently employed by Janssen Pharmaceuticals. J. M. Kahlenberg has received grant support from Q32 Bio, Celgene/Bristol Myers Squibb (BMS), Ventus Therapeutics, ROME Therapeutics, and Janssen; and has served on advisory boards for AstraZeneca, Eli Lilly, GlaxoSmithKline, BMS, Avion Pharmaceuticals, Provention Bio, Aurinia Pharmaceuticals, Ventus Therapeutics, Gilead, and EMD Serano. M. W. L. Leung is currently employed by Janssen R&D LLC, San Diego, Calif. J. E. Gudjonsson has received grant support from Celgene/BMS, Janssen, Eli Lilly, and Almirall and has served on advisory boards for AstraZeneca, Sanofi, Eli Lilly, Boehringer Ingelheim, Novartis, Janssen, Almirall, and BMS. The rest of the authors declare that they have no relevant conflicts of interest., (Copyright © 2024 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. A cluster of type I interferon-regulated genes associates with disease activity and prognosis in patients with IgA nephropathy.
- Author
-
Qu S, Gan T, Wang YN, Qi YY, Zhang YM, Berthier CC, Liu LJ, Shi SF, Lv JC, Zhang H, and Zhou XJ
- Subjects
- Humans, Cross-Sectional Studies, Prognosis, Kidney pathology, Glomerulonephritis, IGA genetics, Glomerulonephritis, IGA drug therapy, Interferon Type I genetics, Interferon Type I therapeutic use
- Abstract
The exact pathogenesis of IgA nephropathy (IgAN) is complex and so far, not well defined. Since it has been shown that microbial infections could induce high levels of type I interferon (IFN-I) and there is an evident link between mucosal infection and gross hematuria in IgAN, we hypothesized that IFN-I may play a role in the pathogenic process. In this study, we investigated the type I interferon status in IgAN based on the expression of 17 IFN-regulated genes (IRGs) in whole blood from 59 IgAN patients in a cross-sectional study, of which 34 patients followed longitudinally. Analysis of the IFN-score showed that there was a significant elevated IFN-score in the IgAN patients compared with healthy controls (n = 28, p = 9.80 × 10
-3 ), and we observed an elevated IFN-score in the group with less tubular atrophy/interstitial fibrosis (p = 1.07 × 10-2 ) and with a lower proportion of mesangial hypercellularity (p = 1.23 × 10-2 ). In the longitudinal analysis, Cox regression analysis revealed that a higher IFN level was associated with a better renal outcome in IgAN after adjustments for gender and age (hazard ratio, 0.90; 95 % confidence interval, 0.81 to 0.97; p = 4.20 × 10-2 ). In conclusion, our finding suggested that IFN score may represent a novel type of biomarker in IgAN, which requires further exploration on its mechanism and therapeutic targeting., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
9. Blood immunophenotyping identifies distinct kidney histopathology and outcomes in patients with lupus nephritis.
- Author
-
Horisberger A, Griffith A, Keegan J, Arazi A, Pulford J, Murzin E, Howard K, Hancock B, Fava A, Sasaki T, Ghosh T, Inamo J, Beuschel R, Cao Y, Preisinger K, Gutierrez-Arcelus M, Eisenhaure TM, Guthridge J, Hoover PJ, Dall'Era M, Wofsy D, Kamen DL, Kalunian KC, Furie R, Belmont M, Izmirly P, Clancy R, Hildeman D, Woodle ES, Apruzzese W, McMahon MA, Grossman J, Barnas JL, Payan-Schober F, Ishimori M, Weisman M, Kretzler M, Berthier CC, Hodgin JB, Demeke DS, Putterman C, Brenner MB, Anolik JH, Raychaudhuri S, Hacohen N, James JA, Davidson A, Petri MA, Buyon JP, Diamond B, Zhang F, Lederer JA, and Rao DA
- Abstract
Lupus nephritis (LN) is a frequent manifestation of systemic lupus erythematosus, and fewer than half of patients achieve complete renal response with standard immunosuppressants. Identifying non-invasive, blood-based pathologic immune alterations associated with renal injury could aid therapeutic decisions. Here, we used mass cytometry immunophenotyping of peripheral blood mononuclear cells in 145 patients with biopsy-proven LN and 40 healthy controls to evaluate the heterogeneity of immune activation in patients with LN and to identify correlates of renal parameters and treatment response. Unbiased analysis identified 3 immunologically distinct groups of patients with LN that were associated with different patterns of histopathology, renal cell infiltrates, urine proteomic profiles, and treatment response at one year. Patients with enriched circulating granzyme B+ T cells at baseline showed more severe disease and increased numbers of activated CD8 T cells in the kidney, yet they had the highest likelihood of treatment response. A second group characterized primarily by a high type I interferon signature had a lower likelihood of response to therapy, while a third group appeared immunologically inactive by immunophenotyping at enrollment but with chronic renal injuries. Main immune profiles could be distilled down to 5 simple cytometric parameters that recapitulate several of the associations, highlighting the potential for blood immune profiling to translate to clinically useful non-invasive metrics to assess immune-mediated disease in LN.
- Published
- 2024
- Full Text
- View/download PDF
10. Epidermal ZBP1 stabilizes mitochondrial Z-DNA to drive UV-induced IFN signaling in autoimmune photosensitivity.
- Author
-
Klein B, Reynolds MB, Xu B, Gharaee-Kermani M, Gao Y, Berthier CC, Henning S, Loftus SN, McNeely KE, Victory AM, Dobry C, Hile GA, Ma F, Turnier JL, Gudjonsson JE, O'Riordan MX, and Kahlenberg JM
- Abstract
Photosensitivity is observed in numerous autoimmune diseases and drives poor quality of life and disease flares. Elevated epidermal type I interferon (IFN) production primes for photosensitivity and enhanced inflammation, but the substrates that sustain and amplify this cycle remain undefined. Here, we show that IFN-induced Z-DNA binding protein 1 (ZBP1) stabilizes ultraviolet (UV)B-induced cytosolic Z-DNA derived from oxidized mitochondrial DNA. ZBP1 is significantly upregulated in the epidermis of adult and pediatric patients with autoimmune photosensitivity. Strikingly, lupus keratinocytes accumulate extensive cytosolic Z-DNA after UVB, and transfection of keratinocytes with Z-DNA results in stronger IFN production through cGAS-STING activation compared to B-DNA. ZBP1 knockdown abrogates UV-induced IFN responses, whereas overexpression results in a lupus-like phenotype with spontaneous Z-DNA accumulation and IFN production. Our results highlight Z-DNA and ZBP1 as critical mediators for UVB-induced inflammation and uncover how type I IFNs prime for cutaneous inflammation in photosensitivity., Competing Interests: Competing interests: JMK has received grant support from Q32 Bio, Celgene/Bristol-Myers Squibb, Ventus Therapeutics, Rome Therapeutics, and Janssen. JMK has served on advisory boards for AstraZeneca, Bristol-Myers Squibb, Eli Lilly, EMD serrano, Gilead, GlaxoSmithKline, Aurinia Pharmaceuticals, Rome Therapeutics, and Ventus Therapeutics. JEG has received support from Eli Lilly, Janssen, BMS, Sanofi, Prometheus, Almirall, Kyowa-Kirin, Novartis, AnaptysBio, Boehringer Ingelheim, Regeneron, AbbVie, and Galderma.
- Published
- 2024
- Full Text
- View/download PDF
11. Urine proteomic signatures of histological class, activity, chronicity, and treatment response in lupus nephritis.
- Author
-
Fava A, Buyon J, Magder L, Hodgin J, Rosenberg A, Demeke DS, Rao DA, Arazi A, Celia AI, Putterman C, Anolik JH, Barnas J, Dall'Era M, Wofsy D, Furie R, Kamen D, Kalunian K, James JA, Guthridge J, Atta MG, Monroy Trujillo J, Fine D, Clancy R, Belmont HM, Izmirly P, Apruzzese W, Goldman D, Berthier CC, Hoover P, Hacohen N, Raychaudhuri S, Davidson A, Diamond B, and Petri M
- Subjects
- Humans, Proteomics, Proteinuria, Inflammation, Aggression, Lupus Nephritis drug therapy
- Abstract
Lupus nephritis (LN) is a pathologically heterogenous autoimmune disease linked to end-stage kidney disease and mortality. Better therapeutic strategies are needed as only 30%-40% of patients completely respond to treatment. Noninvasive biomarkers of intrarenal inflammation may guide more precise approaches. Because urine collects the byproducts of kidney inflammation, we studied the urine proteomic profiles of 225 patients with LN (573 samples) in the longitudinal Accelerating Medicines Partnership in RA/SLE cohort. Urinary biomarkers of monocyte/neutrophil degranulation (i.e., PR3, S100A8, azurocidin, catalase, cathepsins, MMP8), macrophage activation (i.e., CD163, CD206, galectin-1), wound healing/matrix degradation (i.e., nidogen-1, decorin), and IL-16 characterized the aggressive proliferative LN classes and significantly correlated with histological activity. A decline of these biomarkers after 3 months of treatment predicted the 1-year response more robustly than proteinuria, the standard of care (AUC: CD206 0.91, EGFR 0.9, CD163 0.89, proteinuria 0.8). Candidate biomarkers were validated and provide potentially treatable targets. We propose these biomarkers of intrarenal immunological activity as noninvasive tools to diagnose LN and guide treatment and as surrogate endpoints for clinical trials. These findings provide insights into the processes involved in LN activity. This data set is a public resource to generate and test hypotheses and validate biomarkers.
- Published
- 2024
- Full Text
- View/download PDF
12. MTMR3 risk alleles enhance Toll Like Receptor 9-induced IgA immunity in IgA nephropathy.
- Author
-
Wang YN, Gan T, Qu S, Xu LL, Hu Y, Liu LJ, Shi SF, Lv JC, Tsoi LC, Patrick MT, He K, Berthier CC, Xu HJ, Zhou XJ, and Zhang H
- Subjects
- Animals, Humans, Mice, Alleles, Genome-Wide Association Study, Immunoglobulin A, Leukocytes, Mononuclear metabolism, Toll-Like Receptor 9, Glomerulonephritis, IGA pathology, Protein Tyrosine Phosphatases, Non-Receptor genetics, Protein Tyrosine Phosphatases, Non-Receptor metabolism
- Abstract
Multiple genome-wide association studies (GWASs) have reproducibly identified the MTMR3/HORMAD2/LIF/OSM locus to be associated with IgA nephropathy (IgAN). However, the causal variant(s), implicated gene(s), and altered mechanisms remain poorly understood. Here, we performed fine-mapping analyses based on GWAS datasets encompassing 2762 IgAN cases and 5803 control individuals, and identified rs4823074 as the candidate causal variant that intersects the MTMR3 promoter in B-lymphoblastoid cells. Mendelian randomization studies suggested the risk allele may modulate disease susceptibility by affecting serum IgA levels through increased MTMR3 expression. Consistently, elevated MTMR3 expression in peripheral blood mononuclear cells was observed in patients with IgAN. Further mechanistic studies in vitro demonstrated that MTMR3 increased IgA production dependent upon its phosphatidylinositol 3-phosphate binding domain. Moreover, our study provided the in vivo functional evidence that Mtmr3-/- mice exhibited defective Toll Like Receptor 9-induced IgA production, glomerular IgA deposition, as well as mesangial cell proliferation. RNA-seq and pathway analyses showed that MTMR3 deficiency resulted in an impaired intestinal immune network for IgA production. Thus, our results support the role of MTMR3 in IgAN pathogenesis by enhancing Toll Like Receptor 9-induced IgA immunity., (Copyright © 2023 International Society of Nephrology. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
13. An integrated organoid omics map extends modeling potential of kidney disease.
- Author
-
Lassé M, El Saghir J, Berthier CC, Eddy S, Fischer M, Laufer SD, Kylies D, Hutzfeldt A, Bonin LL, Dumoulin B, Menon R, Vega-Warner V, Eichinger F, Alakwaa F, Fermin D, Billing AM, Minakawa A, McCown PJ, Rose MP, Godfrey B, Meister E, Wiech T, Noriega M, Chrysopoulou M, Brandts P, Ju W, Reinhard L, Hoxha E, Grahammer F, Lindenmeyer MT, Huber TB, Schlüter H, Thiel S, Mariani LH, Puelles VG, Braun F, Kretzler M, Demir F, Harder JL, and Rinschen MM
- Subjects
- Humans, Proteome metabolism, Kidney, Organoids metabolism, Tumor Necrosis Factor-alpha metabolism, Kidney Diseases genetics, Kidney Diseases metabolism
- Abstract
Kidney organoids are a promising model to study kidney disease, but their use is constrained by limited knowledge of their functional protein expression profile. Here, we define the organoid proteome and transcriptome trajectories over culture duration and upon exposure to TNFα, a cytokine stressor. Older organoids increase deposition of extracellular matrix but decrease expression of glomerular proteins. Single cell transcriptome integration reveals that most proteome changes localize to podocytes, tubular and stromal cells. TNFα treatment of organoids results in 322 differentially expressed proteins, including cytokines and complement components. Transcript expression of these 322 proteins is significantly higher in individuals with poorer clinical outcomes in proteinuric kidney disease. Key TNFα-associated protein (C3 and VCAM1) expression is increased in both human tubular and organoid kidney cell populations, highlighting the potential for organoids to advance biomarker development. By integrating kidney organoid omic layers, incorporating a disease-relevant cytokine stressor and comparing with human data, we provide crucial evidence for the functional relevance of the kidney organoid model to human kidney disease., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
14. Regulation of Photosensitivity by the Hippo Pathway in Lupus Skin.
- Author
-
Hile GA, Coit P, Xu B, Victory AM, Gharaee-Kermani M, Estadt SN, Maz MP, Martens JWS, Wasikowski R, Dobry C, Tsoi LC, Iglesias-Bartolome R, Berthier CC, Billi AC, Gudjonsson JE, Sawalha AH, and Kahlenberg JM
- Subjects
- Humans, Keratinocytes metabolism, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, RNA, Small Interfering, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Hippo Signaling Pathway, Lupus Erythematosus, Systemic genetics
- Abstract
Objective: Photosensitivity is one of the most common manifestations of systemic lupus erythematosus (SLE), yet its pathogenesis is not well understood. The normal-appearing epidermis of patients with SLE exhibits increased ultraviolet B (UVB)-driven cell death that persists in cell culture. Here, we investigated the role of epigenetic modification and Hippo signaling in enhanced UVB-induced apoptosis seen in SLE keratinocytes., Methods: We analyzed DNA methylation in cultured keratinocytes from SLE patients compared to keratinocytes from healthy controls (n = 6/group). Protein expression was validated in cultured keratinocytes using immunoblotting and immunofluorescence. An immortalized keratinocyte line overexpressing WWC1 was generated via lentiviral vector. WWC1-driven changes were inhibited using a large tumor suppressor kinase 1/2 (LATS1/2) inhibitor (TRULI) and small interfering RNA (siRNA). The interaction between the Yes-associated protein (YAP) and the transcriptional enhancer associate domain (TEAD) was inhibited by overexpression of an N/TERT cell line expressing a tetracycline-inducible green fluorescent protein-tagged protein that inhibits YAP-TEAD binding (TEADi). Apoptosis was assessed using cleaved caspase 3/7 and TUNEL staining., Results: Hippo signaling was the top differentially methylated pathway in SLE versus control keratinocytes. SLE keratinocytes (n = 6) showed significant hypomethylation (Δβ = -0.153) and thus overexpression of the Hippo regulator WWC1 (P = 0.002). WWC1 overexpression increased LATS1/2 kinase activation, leading to YAP cytoplasmic retention and altered proapoptotic transcription in SLE keratinocytes. Accordingly, UVB-mediated apoptosis in keratinocytes could be enhanced by WWC1 overexpression or YAP-TEAD inhibition, mimicking SLE keratinocytes. Importantly, inhibition of LATS1/2 with either the chemical inhibitor TRULI or siRNA effectively eliminated enhanced UVB-apoptosis in SLE keratinocytes., Conclusion: Our work unravels a novel driver of photosensitivity in SLE: overactive Hippo signaling in SLE keratinocytes restricts YAP transcriptional activity, leading to shifts that promote UVB apoptosis., (© 2023 American College of Rheumatology.)
- Published
- 2023
- Full Text
- View/download PDF
15. Loss of interleukin-1 beta is not protective in the lupus-prone NZM2328 mouse model.
- Author
-
Loftus SN, Liu J, Berthier CC, Gudjonsson JE, Gharaee-Kermani M, Tsoi LC, and Kahlenberg JM
- Subjects
- Female, Animals, Mice, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Interleukin-1beta metabolism, Kidney pathology, Inflammasomes metabolism, Lupus Nephritis
- Abstract
Aberrant activation of the innate immune system is a known driver of lupus pathogenesis. Inhibition of the inflammasome and its downstream signaling components in murine models of lupus has been shown to reduce the severity of disease. Interleukin-1 beta (IL-1β) is a proinflammatory cytokine released from cells following inflammasome activation. Here, we examine how loss of IL-1β affects disease severity in the lupus-prone NZM2328 mouse model. We observed a sex-biased increase in immune complex deposition in the kidneys of female mice in the absence of IL-1β that corresponds to worsened proteinuria. Loss of IL-1β did not result in changes in overall survival, anti-dsDNA autoantibody production, or renal immune cell infiltration. RNA-sequencing analysis identified upregulation of TNF and IL-17 signaling pathways specifically in females lacking IL-1β. Increases in these signaling pathways were also found in female patients with lupus nephritis, suggesting clinical relevance for upregulation of these pathways. Together, these data suggest that inhibition of the inflammasome or its downstream elements that block IL-1β signaling may need to be approached with caution in SLE, especially in patients with renal involvement to prevent potential disease exacerbation., Competing Interests: JMK has received grant support from Q32 Bio, Celgene/Bristol-Myers Squibb, Ventus Therapeutics, Rome Therapeutics, and Janssen. JMK has served on advisory boards for AstraZeneca, Bristol-Myers Squibb, Eli Lilly, EMD serrano, Gilead, GlaxoSmithKline, Aurinia Pharmaceuticals, Rome Therapeutics, and Ventus Therapeutics. JEG has received support from Eli Lilly, Janssen, BMS, Sanofi, Prometheus, Almirall, Kyowa-Kirin, Novartis, AnaptysBio, Boehringer Ingelheim, Regeneron, AbbVie, and Galderma. LT has received support from Janssen, Novartis, and Galderma. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Loftus, Liu, Berthier, Gudjonsson, Gharaee-Kermani, Tsoi and Kahlenberg.)
- Published
- 2023
- Full Text
- View/download PDF
16. Genome-Wide Association Study in Acute Tubulointerstitial Nephritis.
- Author
-
Zhou XJ, Su T, Xie J, Xie QH, Wang LZ, Hu Y, Chen G, Jia Y, Huang JW, Li G, Liu Y, Yu XJ, Nath SK, Tsoi LC, Patrick MT, Berthier CC, Liu G, Wang SX, Xu H, Chen N, Hao CM, Zhang H, and Yang L
- Subjects
- Humans, HLA-DRB1 Chains genetics, Genotype, HLA-DQ alpha-Chains genetics, Haplotypes, Alleles, Genetic Predisposition to Disease, Genome-Wide Association Study, Nephritis, Interstitial genetics
- Abstract
Significance Statement: Polymorphisms of HLA genes may confer susceptibility to acute tubulointerstitial nephritis (ATIN), but small sample sizes and candidate gene design have hindered their investigation. The first genome-wide association study of ATIN identified two significant loci, risk haplotype DRB1*14-DQA1*0101-DQB1*0503 (DR14 serotype) and protective haplotype DRB1*1501-DQA1*0102-DQB1*0602 (DR15 serotype), with amino acid position 60 in the peptide-binding groove P10 of HLA-DR β 1 key. Risk alleles were shared among different causes of ATIN and HLA genotypes associated with kidney injury and immune therapy response. HLA alleles showed the strongest association. The findings suggest that a genetically conferred risk of immune dysregulation is part of the pathogenesis of ATIN., Background: Acute tubulointerstitial nephritis (ATIN) is a rare immune-related disease, accounting for approximately 10% of patients with unexplained AKI. Previous elucidation of the relationship between genetic factors that contribute to its pathogenesis was hampered because of small sample sizes and candidate gene design., Methods: We undertook the first two-stage genome-wide association study and meta-analysis involving 544 kidney biopsy-defined patients with ATIN and 2346 controls of Chinese ancestry. We conducted statistical fine-mapping analysis, provided functional annotations of significant variants, estimated single nucleotide polymorphism (SNP)-based heritability, and checked genotype and subphenotype correlations., Results: Two genome-wide significant loci, rs35087390 of HLA-DQA1 ( P =3.01×10 -39 ) on 6p21.32 and rs2417771 of PLEKHA5 on 12p12.3 ( P =2.14×10 -8 ), emerged from the analysis. HLA imputation using two reference panels suggested that HLA-DRB1*14 mainly drives the HLA risk association . HLA-DRB1 residue 60 belonging to pocket P10 was the key amino acid position. The SNP-based heritability estimates with and without the HLA locus were 20.43% and 10.35%, respectively. Different clinical subphenotypes (drug-related or tubulointerstitial nephritis and uveitis syndrome) seemed to share the same risk alleles. However, the HLA risk genotype was associated with disease severity and response rate to immunosuppressive therapy., Conclusions: We identified two candidate genome regions associated with susceptibility to ATIN. The findings suggest that a genetically conferred risk of immune dysregulation is involved in the pathogenesis of ATIN., (Copyright © 2023 by the American Society of Nephrology.)
- Published
- 2023
- Full Text
- View/download PDF
17. Imaging Mass Cytometry Reveals Predominant Innate Immune Signature and Endothelial-Immune Cell Interaction in Juvenile Myositis Compared to Lupus Skin.
- Author
-
Turnier JL, Yee CM, Madison JA, Rizvi SM, Berthier CC, Wen F, and Kahlenberg JM
- Subjects
- Humans, Child, Skin pathology, Cell Communication, Immunity, Innate, Endothelial Cells metabolism, Image Cytometry, Inflammation metabolism, Dermatomyositis metabolism, Lupus Erythematosus, Systemic metabolism
- Abstract
Objective: Cutaneous inflammation can signal disease in juvenile dermatomyositis (DM) and childhood-onset systemic lupus erythematosus (cSLE), but we do not fully understand cellular mechanisms of cutaneous inflammation. In this study, we used imaging mass cytometry to characterize cutaneous inflammatory cell populations and cell-cell interactions in juvenile DM as compared to cSLE., Methods: We performed imaging mass cytometry analysis on skin biopsy samples from juvenile DM patients (n = 6) and cSLE patients (n = 4). Tissue slides were processed and incubated with metal-tagged antibodies for CD14, CD15, CD16, CD56, CD68, CD11c, HLA-DR, blood dendritic cell antigen 2, CD20, CD27, CD138, CD4, CD8, E-cadherin, CD31, pan-keratin, and type I collagen. Stained tissue was ablated, and raw data were acquired using the Hyperion imaging system. We utilized the Phenograph unsupervised clustering algorithm to determine cell marker expression and permutation test by histoCAT to perform neighborhood analysis., Results: We identified 14 cell populations in juvenile DM and cSLE skin, including CD14+ and CD68+ macrophages, myeloid and plasmacytoid dendritic cells (pDCs), CD4+ and CD8+ T cells, and B cells. Overall, cSLE skin had a higher inflammatory cell infiltrate, with increased CD14+ macrophages, pDCs, and CD8+ T cells and immune cell-immune cell interactions. Juvenile DM skin displayed a stronger innate immune signature, with a higher overall percentage of CD14+ macrophages and prominent endothelial cell-immune cell interaction., Conclusion: Our findings identify immune cell population differences, including CD14+ macrophages, pDCs, and CD8+ T cells, in juvenile DM skin compared to cSLE skin, and highlight a predominant innate immune signature and endothelial cell-immune cell interaction in juvenile DM, providing insight into candidate cell populations and interactions to better understand disease-specific pathophysiology., (© 2022 The Authors. Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)
- Published
- 2022
- Full Text
- View/download PDF
18. Urine Proteomics and Renal Single-Cell Transcriptomics Implicate Interleukin-16 in Lupus Nephritis.
- Author
-
Fava A, Rao DA, Mohan C, Zhang T, Rosenberg A, Fenaroli P, Belmont HM, Izmirly P, Clancy R, Trujillo JM, Fine D, Arazi A, Berthier CC, Davidson A, James JA, Diamond B, Hacohen N, Wofsy D, Raychaudhuri S, Apruzzese W, Buyon J, and Petri M
- Subjects
- Biomarkers metabolism, Female, Humans, Interleukin-16 genetics, Kidney pathology, Male, Proteomics methods, Single-Cell Analysis, Transcriptome, Biological Products, Interleukin-16 metabolism, Lupus Nephritis pathology
- Abstract
Objective: Current lupus nephritis (LN) treatments are effective in only 30% of patients, emphasizing the need for novel therapeutic strategies. We undertook this study to develop mechanistic hypotheses and explore novel biomarkers by analyzing the longitudinal urinary proteomic profiles in LN patients undergoing treatment., Methods: We quantified 1,000 urinary proteins in 30 patients with LN at the time of the diagnostic renal biopsy and after 3, 6, and 12 months. The proteins and molecular pathways detected in the urine proteome were then analyzed with respect to baseline clinical features and longitudinal trajectories. The intrarenal expression of candidate biomarkers was evaluated using single-cell transcriptomics of renal biopsy sections from LN patients., Results: Our analysis revealed multiple biologic pathways, including chemotaxis, neutrophil activation, platelet degranulation, and extracellular matrix organization, which could be noninvasively quantified and monitored in the urine. We identified 237 urinary biomarkers associated with LN, as compared to controls without systemic lupus erythematosus. Interleukin-16 (IL-16), CD163, and transforming growth factor β mirrored intrarenal nephritis activity. Response to treatment was paralleled by a reduction in urinary IL-16, a CD4 ligand with proinflammatory and chemotactic properties. Single-cell RNA sequencing independently demonstrated that IL16 is the second most expressed cytokine by most infiltrating immune cells in LN kidneys. IL-16-producing cells were found at key sites of kidney injury., Conclusion: Urine proteomics may profoundly change the diagnosis and management of LN by noninvasively monitoring active intrarenal biologic pathways. These findings implicate IL-16 in LN pathogenesis, designating it as a potentially treatable target and biomarker., (© 2021 American College of Rheumatology.)
- Published
- 2022
- Full Text
- View/download PDF
19. Nonlesional lupus skin contributes to inflammatory education of myeloid cells and primes for cutaneous inflammation.
- Author
-
Billi AC, Ma F, Plazyo O, Gharaee-Kermani M, Wasikowski R, Hile GA, Xing X, Yee CM, Rizvi SM, Maz MP, Berthier CC, Wen F, Tsoi LC, Pellegrini M, Modlin RL, Gudjonsson JE, and Kahlenberg JM
- Subjects
- Humans, Inflammation pathology, Keratinocytes pathology, Myeloid Cells metabolism, Interferon Type I metabolism, Lupus Erythematosus, Cutaneous
- Abstract
Cutaneous lupus erythematosus (CLE) is a disfiguring and poorly understood condition frequently associated with systemic lupus. Previous studies suggest that nonlesional keratinocytes play a role in disease predisposition, but this has not been investigated in a comprehensive manner or in the context of other cell populations. To investigate CLE immunopathogenesis, normal-appearing skin, lesional skin, and circulating immune cells from lupus patients were analyzed via integrated single-cell RNA sequencing and spatial RNA sequencing. We demonstrate that normal-appearing skin of patients with lupus represents a type I interferon-rich, prelesional environment that skews gene transcription in all major skin cell types and markedly distorts predicted cell-cell communication networks. We also show that lupus-enriched CD16
+ dendritic cells undergo robust interferon education in the skin, thereby gaining proinflammatory phenotypes. Together, our data provide a comprehensive characterization of lesional and nonlesional skin in lupus and suggest a role for skin education of CD16+ dendritic cells in CLE pathogenesis.- Published
- 2022
- Full Text
- View/download PDF
20. Transcriptomic characterization of prurigo nodularis and the therapeutic response to nemolizumab.
- Author
-
Tsoi LC, Hacini-Rachinel F, Fogel P, Rousseau F, Xing X, Patrick MT, Billi AC, Berthier CC, Kahlenberg JM, Lazzari A, Wiegmann H, Ständer S, Piketty C, Julia V, Krishnaswamy JK, and Gudjonsson JE
- Subjects
- Antibodies, Monoclonal, Humanized pharmacology, Antibodies, Monoclonal, Humanized therapeutic use, Chronic Disease, Cytokines therapeutic use, Humans, Pruritus drug therapy, Pruritus genetics, Transcriptome, Prurigo drug therapy, Prurigo genetics
- Abstract
Background: Prurigo nodularis (PN) is a debilitating, difficult-to-treat, intensely pruritic, chronic inflammatory skin disease characterized by hyperkeratotic skin nodules. The pathogenesis of PN is not well understood but is believed to involve cross talk between sensory nerve fibers, immune cells, and the epidermis. It is centered around the neuroimmune cytokine IL-31, driving an intractable itch-scratch cycle., Objective: We sought to provide a comprehensive view of the transcriptomic changes in PN skin and characterize the mechanism of action of the anti-IL-31 receptor inhibitor nemolizumab., Method: RNA sequencing of biopsy samples obtained from a cohort of patients treated with the anti-IL-31 receptor inhibitor nemolizumab and taken at baseline and week 12. Generation and integration of patient data with RNA-Seq data generated from reconstructed human epidermis stimulated with IL-31 and other proinflammatory cytokines., Results: Our results demonstrate that nemolizumab effectively decreases IL-31 responses in PN skin, leading to effective suppression of downstream inflammatory responses including T
H 2/IL-13 and TH 17/IL-17 responses. This is accompanied by decreased keratinocyte proliferation and normalization of epidermal differentiation and function. Furthermore, our results demonstrate how transcriptomic changes associated with nemolizumab treatment correlate with improvement in lesions, pruritus, stabilization of extracellular matrix remodeling, and processes associated with cutaneous nerve function., Conclusion: These data demonstrate a broad response to IL-31 receptor inhibition with nemolizumab and confirm the critical upstream role of IL-31 in PN pathogenesis., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
21. Glomerular endothelial cell-podocyte stresses and crosstalk in structurally normal kidney transplants.
- Author
-
Menon R, Otto EA, Berthier CC, Nair V, Farkash EA, Hodgin JB, Yang Y, Luo J, Woodside KJ, Zamani H, Norman SP, Wiggins RC, Kretzler M, and Naik AS
- Subjects
- Endothelial Cells, Female, Glomerular Basement Membrane pathology, Humans, Hypertrophy, Integrin alpha3 metabolism, Male, Kidney Diseases pathology, Kidney Transplantation adverse effects, Podocytes pathology
- Abstract
Increased podocyte detachment begins immediately after kidney transplantation and is associated with long-term allograft failure. We hypothesized that cell-specific transcriptional changes in podocytes and glomerular endothelial cells after transplantation would offer mechanistic insights into the podocyte detachment process. To test this, we evaluated cell-specific transcriptional profiles of glomerular endothelial cells and podocytes from 14 patients of their first-year surveillance biopsies with normal histology from low immune risk recipients with no post-transplant complications and compared these to biopsies of 20 healthy living donor controls. Glomerular endothelial cells from these surveillance biopsies were enriched for genes related to fluid shear stress, angiogenesis, and interferon signaling. In podocytes, pathways were enriched for genes in response to growth factor signaling and actin cytoskeletal reorganization but also showed evidence of podocyte stress as indicated by reduced nephrin (adhesion protein) gene expression. In parallel, transcripts coding for proteins required to maintain podocyte adherence to the underlying glomerular basement membrane were downregulated, including the major glomerular podocyte integrin α3 and the actin cytoskeleton-related gene synaptopodin. The reduction in integrin α3 protein expression in surveillance biopsies was confirmed by immunoperoxidase staining. The combined growth and stress response of patient allografts post-transplantation paralleled similar changes in a rodent model of nephrectomy-induced glomerular hypertrophic stress that progress to develop proteinuria and glomerulosclerosis with shortened kidney life span. Thus, even among patients with apparently healthy allografts with no detectable histologic abnormality including alloimmune injury, transcriptomic changes reflecting cell stresses are already set in motion that could drive hypertrophy-associated glomerular disease progression., (Copyright © 2021 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
22. Urine Single-Cell RNA Sequencing in Focal Segmental Glomerulosclerosis Reveals Inflammatory Signatures.
- Author
-
Latt KZ, Heymann J, Jessee JH, Rosenberg AZ, Berthier CC, Arazi A, Eddy S, Yoshida T, Zhao Y, Chen V, Nelson GW, Cam M, Kumar P, Mehta M, Kelly MC, Kretzler M, Ray PE, Moxey-Mims M, Gorman GH, Lechner BL, Regunathan-Shenk R, Raj DS, Susztak K, Winkler CA, and Kopp JB
- Abstract
Introduction: Individuals with focal segmental glomerular sclerosis (FSGS) typically undergo kidney biopsy only once, which limits the ability to characterize kidney cell gene expression over time., Methods: We used single-cell RNA sequencing (scRNA-seq) to explore disease-related molecular signatures in urine cells from subjects with FSGS. We collected 17 urine samples from 12 FSGS subjects and captured these as 23 urine cell samples. The inflammatory signatures from renal epithelial and immune cells were evaluated in bulk gene expression data sets of FSGS and minimal change disease (MCD) (The Nephrotic Syndrome Study Network [NEPTUNE] study) and an immune single-cell data set from lupus nephritis (Accelerating Medicines Partnership)., Results: We identified immune cells, predominantly monocytes, and renal epithelial cells in the urine. Further analysis revealed 2 monocyte subtypes consistent with M1 and M2 monocytes. Shed podocytes in the urine had high expression of marker genes for epithelial-to-mesenchymal transition (EMT). We selected the 16 most highly expressed genes from urine immune cells and 10 most highly expressed EMT genes from urine podocytes as immune signatures and EMT signatures, respectively. Using kidney biopsy transcriptomic data from NEPTUNE, we found that urine cell immune signature and EMT signature genes were more highly expressed in FSGS biopsies compared with MCD biopsies., Conclusion: The identification of monocyte subsets and podocyte expression signatures in the urine samples of subjects with FSGS suggests that urine cell profiling might serve as a diagnostic and prognostic tool in nephrotic syndrome. Furthermore, this approach may aid in the development of novel biomarkers and identifying personalized therapies targeting particular molecular pathways in immune cells and podocytes., (© 2021 Published by Elsevier, Inc., on behalf of the International Society of Nephrology.)
- Published
- 2021
- Full Text
- View/download PDF
23. B Cell Signatures Distinguish Cutaneous Lupus Erythematosus Subtypes and the Presence of Systemic Disease Activity.
- Author
-
Abernathy-Close L, Lazar S, Stannard J, Tsoi LC, Eddy S, Rizvi SM, Yee CM, Myers EM, Namas R, Lowe L, Reed TJ, Wen F, Gudjonsson JE, Kahlenberg JM, and Berthier CC
- Subjects
- Computational Biology methods, Diagnosis, Differential, Gene Expression Profiling, Gene Expression Regulation, Gene Regulatory Networks, Humans, Immunoglobulins genetics, Immunohistochemistry, Lupus Erythematosus, Cutaneous diagnosis, Lupus Erythematosus, Systemic diagnosis, B-Lymphocytes immunology, B-Lymphocytes metabolism, Biomarkers, Disease Susceptibility, Lupus Erythematosus, Cutaneous etiology, Lupus Erythematosus, Cutaneous metabolism, Lupus Erythematosus, Systemic etiology, Lupus Erythematosus, Systemic metabolism
- Abstract
Cutaneous lupus erythematosus (CLE) is a chronic inflammatory skin disease characterized by a diverse cadre of clinical presentations. CLE commonly occurs in patients with systemic lupus erythematosus (SLE), and CLE can also develop in the absence of systemic disease. Although CLE is a complex and heterogeneous disease, several studies have identified common signaling pathways, including those of type I interferons (IFNs), that play a key role in driving cutaneous inflammation across all CLE subsets. However, discriminating factors that drive different phenotypes of skin lesions remain to be determined. Thus, we sought to understand the skin-associated cellular and transcriptional differences in CLE subsets and how the different types of cutaneous inflammation relate to the presence of systemic lupus disease. In this study, we utilized two distinct cohorts comprising a total of 150 CLE lesional biopsies to compare discoid lupus erythematosus (DLE), subacute cutaneous lupus erythematosus (SCLE), and acute cutaneous lupus erythematosus (ACLE) in patients with and without associated SLE. Using an unbiased approach, we demonstrated a CLE subtype-dependent gradient of B cell enrichment in the skin, with DLE lesions harboring a more dominant skin B cell transcriptional signature and enrichment of B cells on immunostaining compared to ACLE and SCLE. Additionally, we observed a significant increase in B cell signatures in the lesional skin from patients with isolated CLE compared with similar lesions from patients with systemic lupus. This trend was driven primarily by differences in the DLE subgroup. Our work thus shows that skin-associated B cell responses distinguish CLE subtypes in patients with and without associated SLE, suggesting that B cell function in skin may be an important link between cutaneous lupus and systemic disease activity., Competing Interests: JMK has received Grant support from Q32 Bio, Celgene/BMS, Ventus Therapeutics, and Janssen. JG has received Grant support from Celgene/BMS, Janssen, Eli Lilly, and Almirall. JMK has served on advisory boards for AstraZeneca, Eli Lilly, GlaxoSmithKline, Bristol Myers Squibb, Avion Pharmaceuticals, Provention Bio, Aurinia Pharmaceuticals, Ventus Therapeutics, and Boehringer Ingelheim. JG has served on advisory boards for AstraZeneca, Sanofi, Eli Lilly, Boehringer Ingelheim, Novartis, Janssen, Almirall, BMS. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Abernathy-Close, Lazar, Stannard, Tsoi, Eddy, Rizvi, Yee, Myers, Namas, Lowe, Reed, Wen, Gudjonsson, Kahlenberg and Berthier.)
- Published
- 2021
- Full Text
- View/download PDF
24. Reversible dysregulation of renal circadian rhythm in lupus nephritis.
- Author
-
Mishra R, Bethunaickan R, Berthier CC, Yi Z, Strohl JJ, Huerta PT, Zhang W, and Davidson A
- Subjects
- Animals, Computational Biology methods, Disease Models, Animal, Female, Gene Expression Profiling, Lupus Nephritis pathology, Mice, Transcriptome, Biomarkers, Circadian Rhythm genetics, Disease Susceptibility, Gene Expression Regulation, Lupus Nephritis etiology, Lupus Nephritis metabolism
- Abstract
Background: We have found disruption of expression of major transcriptional regulators of circadian rhythm in the kidneys of several mouse models of lupus nephritis. Here we define the consequence of this disturbance with respect to circadian gene expression and renal homeostatic function in a mouse model of lupus nephritis., Methods: Molecular profiling of kidneys from 47 young and 41 nephritic female NZB/W F1 mice was performed at 4 hourly intervals over a 24 h period. Disruption of major circadian transcriptional regulators was confirmed by qPCR. Molecular data was normalized and analyzed for rhythmicity using RAIN analysis. Serum aldosterone and glucose and urine sodium and potassium were measured at 4 hourly intervals in pre-nephritic and nephritic mice and blood pressure was measured every 4 h. Analyses were repeated after induction of complete remission of nephritis using combination cyclophosphamide and costimulatory blockade., Results: We show a profound alteration of renal circadian rhythms in mice with lupus nephritis affecting multiple renal pathways. Using Cosinor analysis we identified consequent alterations of renal homeostasis and metabolism as well as blood pressure dipper status. This circadian dysregulation was partially reversed by remission induction therapy., Conclusions: Our studies indicate the role of inflammation in causing the circadian disruption and suggest that screening for loss of normal blood pressure dipping should be incorporated into LN management. The data also suggest a potential role for circadian agonists in the treatment of lupus nephritis., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
25. Safety of procuring research tissue during a clinically indicated kidney biopsy from patients with lupus: data from the Accelerating Medicines Partnership RA/SLE Network.
- Author
-
Deonaraine KK, Carlucci PM, Fava A, Li J, Wofsy D, James JA, Putterman C, Diamond B, Davidson A, Fine DM, Monroy-Trujillo J, Atta MG, Haag K, Rao DA, Apruzzese W, Belmont HM, Izmirly PM, Wu M, Connery S, Payan-Schober F, Furie RA, Berthier CC, Dall'Era M, Cho K, Kamen DL, Kalunian K, Anolik J, Ishimori M, Weisman MH, Petri MA, and Buyon JP
- Subjects
- Biopsy, Hematoma, Humans, Kidney, United States, Arteriovenous Fistula, Lupus Nephritis drug therapy
- Abstract
Objectives: In lupus nephritis the pathological diagnosis from tissue retrieved during kidney biopsy drives treatment and management. Despite recent approval of new drugs, complete remission rates remain well under aspirational levels, necessitating identification of new therapeutic targets by greater dissection of the pathways to tissue inflammation and injury. This study assessed the safety of kidney biopsies in patients with SLE enrolled in the Accelerating Medicines Partnership, a consortium formed to molecularly deconstruct nephritis., Methods: 475 patients with SLE across 15 clinical sites in the USA consented to obtain tissue for research purposes during a clinically indicated kidney biopsy. Adverse events (AEs) were documented for 30 days following the procedure and were determined to be related or unrelated by all site investigators. Serious AEs were defined according to the National Institutes of Health reporting guidelines., Results: 34 patients (7.2%) experienced a procedure-related AE: 30 with haematoma, 2 with jets, 1 with pain and 1 with an arteriovenous fistula. Eighteen (3.8%) experienced a serious AE requiring hospitalisation; four patients (0.8%) required a blood transfusion related to the kidney biopsy. At one site where the number of cores retrieved during the biopsy was recorded, the mean was 3.4 for those who experienced a related AE (n=9) and 3.07 for those who did not experience any AE (n=140). All related AEs resolved., Conclusions: Procurement of research tissue should be considered feasible, accompanied by a complication risk likely no greater than that incurred for standard clinical purposes. In the quest for targeted treatments personalised based on molecular findings, enhanced diagnostics beyond histology will likely be required., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
26. Comparison of Lesional Juvenile Myositis and Lupus Skin Reveals Overlapping Yet Unique Disease Pathophysiology.
- Author
-
Turnier JL, Pachman LM, Lowe L, Tsoi LC, Elhaj S, Menon R, Amoruso MC, Morgan GA, Gudjonsson JE, Berthier CC, and Kahlenberg JM
- Subjects
- Adenosine Triphosphatases immunology, Adolescent, Antigens, Ly genetics, Autoantibodies immunology, Calcium-Binding Proteins genetics, Chemokine CXCL10 genetics, Chemokine CXCL9 genetics, Child, Choline Kinase genetics, DNA-Binding Proteins immunology, Dermatomyositis immunology, Dermatomyositis metabolism, Extracellular Matrix Proteins genetics, Female, Humans, Interferon Type I immunology, Interferons, Male, Myxovirus Resistance Proteins genetics, Myxovirus Resistance Proteins metabolism, Signal Transduction, Transcriptome, Tumor Suppressor Proteins genetics, Urokinase-Type Plasminogen Activator genetics, Dermatomyositis genetics, Lupus Erythematosus, Systemic genetics, Skin metabolism
- Abstract
Objective: Skin inflammation heralds systemic disease in juvenile myositis, yet we lack an understanding of pathogenic mechanisms driving skin inflammation in this disease. We undertook this study to define cutaneous gene expression signatures in juvenile myositis and identify key genes and pathways that differentiate skin disease in juvenile myositis from childhood-onset systemic lupus erythematosus (SLE)., Methods: We used formalin-fixed paraffin-embedded skin biopsy samples from 15 patients with juvenile myositis (9 lesional, 6 nonlesional), 5 patients with childhood-onset SLE, and 8 controls to perform transcriptomic analysis and identify significantly differentially expressed genes (DEGs; q ≤ 5%) between patient groups. We used Ingenuity Pathway Analysis (IPA) to highlight enriched biologic pathways and validated DEGs by immunohistochemistry and quantitative real-time polymerase chain reaction., Results: Comparison of lesional juvenile myositis to control samples revealed 221 DEGs, with the majority of up-regulated genes representing interferon (IFN)-stimulated genes. CXCL10, CXCL9, and IFI44L represented the top 3 DEGs (fold change 23.2, 13.3, and 13.0, respectively; q < 0.0001). IPA revealed IFN signaling as the top canonical pathway. When compared to childhood-onset SLE, lesional juvenile myositis skin shared a similar gene expression pattern, with only 28 unique DEGs, including FBLN2, CHKA, and SLURP1. Notably, patients with juvenile myositis who were positive for nuclear matrix protein 2 (NXP-2) autoantibodies exhibited the strongest IFN signature and also demonstrated the most extensive Mx-1 immunostaining, both in keratinocytes and perivascular regions., Conclusion: Lesional juvenile myositis skin demonstrates a striking IFN signature similar to that previously reported in juvenile myositis muscle and peripheral blood. Further investigation into the association of a higher IFN score with NXP-2 autoantibodies may provide insight into disease endotypes and pathogenesis., (© 2020, American College of Rheumatology.)
- Published
- 2021
- Full Text
- View/download PDF
27. Gene expression profiles of diabetic kidney disease and neuropathy in eNOS knockout mice: Predictors of pathology and RAS blockade effects.
- Author
-
Eid SA, Hinder LM, Zhang H, Eksi R, Nair V, Eddy S, Eichinger F, Park M, Saha J, Berthier CC, Jagadish HV, Guan Y, Pennathur S, Hur J, Kretzler M, Feldman EL, and Brosius FC
- Subjects
- Animals, Diabetic Nephropathies etiology, Diabetic Nephropathies metabolism, Diabetic Neuropathies etiology, Diabetic Neuropathies metabolism, Gene Expression Regulation, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Diabetes Mellitus, Experimental complications, Diabetes Mellitus, Type 2 complications, Diabetic Nephropathies pathology, Diabetic Neuropathies pathology, Nitric Oxide Synthase Type III physiology, Transcriptome, ras Proteins antagonists & inhibitors
- Abstract
Diabetic kidney disease (DKD) and diabetic peripheral neuropathy (DPN) are two common diabetic complications. However, their pathogenesis remains elusive and current therapies are only modestly effective. We evaluated genome-wide expression to identify pathways involved in DKD and DPN progression in db/db eNOS-/- mice receiving renin-angiotensin-aldosterone system (RAS)-blocking drugs to mimic the current standard of care for DKD patients. Diabetes and eNOS deletion worsened DKD, which improved with RAS treatment. Diabetes also induced DPN, which was not affected by eNOS deletion or RAS blockade. Given the multiple factors affecting DKD and the graded differences in disease severity across mouse groups, an automatic data analysis method, SOM, or self-organizing map was used to elucidate glomerular transcriptional changes associated with DKD, whereas pairwise bioinformatic analysis was used for DPN. These analyses revealed that enhanced gene expression in several pro-inflammatory networks and reduced expression of development genes correlated with worsening DKD. Although RAS treatment ameliorated the nephropathy phenotype, it did not alter the more abnormal gene expression changes in kidney. Moreover, RAS exacerbated expression of genes related to inflammation and oxidant generation in peripheral nerves. The graded increase in inflammatory gene expression and decrease in development gene expression with DKD progression underline the potentially important role of these pathways in DKD pathogenesis. Since RAS blockers worsened this gene expression pattern in both DKD and DPN, it may partly explain the inadequate therapeutic efficacy of such blockers., (© 2021 Federation of American Societies for Experimental Biology.)
- Published
- 2021
- Full Text
- View/download PDF
28. Exome Chip Analyses and Genetic Risk for IgA Nephropathy among Han Chinese.
- Author
-
Zhou XJ, Tsoi LC, Hu Y, Patrick MT, He K, Berthier CC, Li Y, Wang YN, Qi YY, Zhang YM, Gan T, Li Y, Hou P, Liu LJ, Shi SF, Lv JC, Xu HJ, and Zhang H
- Subjects
- Adult, Case-Control Studies, China, Complement Factor B genetics, Enhancer Elements, Genetic, Extracellular Matrix Proteins genetics, F-Box Proteins genetics, Female, Genotype, Glomerulonephritis, IGA ethnology, Humans, Male, Middle Aged, Oligonucleotide Array Sequence Analysis, Polymorphism, Single Nucleotide, Receptors, CCR6 genetics, Receptors, GABA-B genetics, STAT3 Transcription Factor genetics, Sequence Analysis, DNA, Transcriptome, Transforming Growth Factor beta genetics, Exome Sequencing, Young Adult, Asian People genetics, Genetic Predisposition to Disease genetics, Glomerulonephritis, IGA genetics
- Abstract
Background and Objectives: IgA nephropathy is the most common form of primary GN worldwide. The evidence of geographic and ethnic differences, as well as familial aggregation of the disease, supports a strong genetic contribution to IgA nephropathy. Evidence for genetic factors in IgA nephropathy comes also from genome-wide association patient-control studies. However, few studies have systematically evaluated the contribution of coding variation in IgA nephropathy., Design, Setting, Participants, & Measurements: We performed a two-stage exome chip-based association study in 13,242 samples, including 3363 patients with IgA nephropathy and 9879 healthy controls of Han Chinese ancestry. Common variant functional annotation, gene-based low-frequency variants analysis, differential mRNA expression, and gene network integration were also explored., Results: We identified three non-HLA gene regions ( FBXL21 , CCR6 , and STAT3 ) and one HLA gene region ( GABBR1 ) with suggestive significance ( P
meta <5×10-5 ) in single-variant associations. These novel non-HLA variants were annotated as expression-associated single-nucleotide polymorphisms and were located in enhancer regions enriched in histone marks H3K4me1 in primary B cells. Gene-based low-frequency variants analysis suggests CFB as another potential susceptibility gene. Further combined expression and network integration suggested that the five novel susceptibility genes, TGFBI , CCR6 , STAT3 , GABBR1 , and CFB , were involved in IgA nephropathy., Conclusions: Five novel gene regions with suggestive significance for IgA nephropathy were identified and shed new light for further mechanism investigation., (Copyright © 2021 by the American Society of Nephrology.)- Published
- 2021
- Full Text
- View/download PDF
29. SARS-CoV-2 receptor networks in diabetic and COVID-19-associated kidney disease.
- Author
-
Menon R, Otto EA, Sealfon R, Nair V, Wong AK, Theesfeld CL, Chen X, Wang Y, Boppana AS, Luo J, Yang Y, Kasson PM, Schaub JA, Berthier CC, Eddy S, Lienczewski CC, Godfrey B, Dagenais SL, Sohaney R, Hartman J, Fermin D, Subramanian L, Looker HC, Harder JL, Mariani LH, Hodgin JB, Sexton JZ, Wobus CE, Naik AS, Nelson RG, Troyanskaya OG, and Kretzler M
- Subjects
- Adult, Aged, Angiotensin Receptor Antagonists pharmacology, Angiotensin Receptor Antagonists therapeutic use, Angiotensin-Converting Enzyme Inhibitors pharmacology, Angiotensin-Converting Enzyme Inhibitors therapeutic use, COVID-19 complications, COVID-19 virology, Case-Control Studies, Diabetic Nephropathies drug therapy, Female, Gene Expression Profiling, Gene Regulatory Networks, Host-Pathogen Interactions, Humans, Kidney Tubules, Proximal drug effects, Male, Middle Aged, Angiotensin-Converting Enzyme 2 metabolism, COVID-19 metabolism, Diabetic Nephropathies metabolism, Kidney Tubules, Proximal metabolism, SARS-CoV-2 metabolism
- Abstract
COVID-19 morbidity and mortality are increased via unknown mechanisms in patients with diabetes and kidney disease. SARS-CoV-2 uses angiotensin-converting enzyme 2 (ACE2) for entry into host cells. Because ACE2 is a susceptibility factor for infection, we investigated how diabetic kidney disease and medications alter ACE2 receptor expression in kidneys. Single cell RNA profiling of kidney biopsies from healthy living donors and patients with diabetic kidney disease revealed ACE2 expression primarily in proximal tubular epithelial cells. This cell-specific localization was confirmed by in situ hybridization. ACE2 expression levels were unaltered by exposures to renin-angiotensin-aldosterone system inhibitors in diabetic kidney disease. Bayesian integrative analysis of a large compendium of public -omics datasets identified molecular network modules induced in ACE2-expressing proximal tubular epithelial cells in diabetic kidney disease (searchable at hb.flatironinstitute.org/covid-kidney) that were linked to viral entry, immune activation, endomembrane reorganization, and RNA processing. The diabetic kidney disease ACE2-positive proximal tubular epithelial cell module overlapped with expression patterns seen in SARS-CoV-2-infected cells. Similar cellular programs were seen in ACE2-positive proximal tubular epithelial cells obtained from urine samples of 13 hospitalized patients with COVID-19, suggesting a consistent ACE2-coregulated proximal tubular epithelial cell expression program that may interact with the SARS-CoV-2 infection processes. Thus SARS-CoV-2 receptor networks can seed further research into risk stratification and therapeutic strategies for COVID-19-related kidney damage., (Copyright © 2020 International Society of Nephrology. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
30. IL18-containing 5-gene signature distinguishes histologically identical dermatomyositis and lupus erythematosus skin lesions.
- Author
-
Tsoi LC, Gharaee-Kermani M, Berthier CC, Nault T, Hile GA, Estadt SN, Patrick MT, Wasikowski R, Billi AC, Lowe L, Reed TJ, Gudjonsson JE, and Kahlenberg JM
- Subjects
- Biopsy, Cohort Studies, Cornified Envelope Proline-Rich Proteins genetics, Dermatomyositis metabolism, Dermatomyositis pathology, Female, Humans, Interferons genetics, Interferons metabolism, Keratins, Type II genetics, Lupus Erythematosus, Cutaneous metabolism, Lupus Erythematosus, Cutaneous pathology, Male, Middle Aged, Myxovirus Resistance Proteins metabolism, Transcriptome, Tropomyosin genetics, Dermatomyositis genetics, Interleukin-18 genetics, Lupus Erythematosus, Cutaneous genetics
- Abstract
Skin lesions in dermatomyositis (DM) are common, are frequently refractory, and have prognostic significance. Histologically, DM lesions appear similar to cutaneous lupus erythematosus (CLE) lesions and frequently cannot be differentiated. We thus compared the transcriptional profile of DM biopsies with CLE lesions to identify unique features. Type I IFN signaling, including IFN-κ upregulation, was a common pathway in both DM and CLE; however, CLE also exhibited other inflammatory pathways. Notably, DM lesions could be distinguished from CLE by a 5-gene biomarker panel that included IL18 upregulation. Using single-cell RNA-sequencing, we further identified keratinocytes as the main source of increased IL-18 in DM skin. This study identifies a potentially novel molecular signature, with significant clinical implications for differentiating DM from CLE lesions, and highlights the potential role for IL-18 in the pathophysiology of DM skin disease.
- Published
- 2020
- Full Text
- View/download PDF
31. Integrated urine proteomics and renal single-cell genomics identify an IFN-γ response gradient in lupus nephritis.
- Author
-
Fava A, Buyon J, Mohan C, Zhang T, Belmont HM, Izmirly P, Clancy R, Trujillo JM, Fine D, Zhang Y, Magder L, Rao DA, Arazi A, Berthier CC, Davidson A, Diamond B, Hacohen N, Wofsy D, Apruzzese W, Raychaudhuri S, and Petri M
- Subjects
- Biomarkers analysis, CD8-Positive T-Lymphocytes metabolism, Chemokines metabolism, Humans, Kidney metabolism, Lupus Erythematosus, Systemic diagnosis, Lupus Erythematosus, Systemic urine, Lupus Nephritis diagnosis, Lupus Nephritis urine, Biomarkers urine, Kidney pathology, Lupus Erythematosus, Systemic metabolism, Lupus Nephritis immunology, Proteomics methods
- Abstract
Lupus nephritis, one of the most serious manifestations of systemic lupus erythematosus (SLE), has a heterogeneous clinical and pathological presentation. For example, proliferative nephritis identifies a more aggressive disease class that requires immunosuppression. However, the current classification system relies on the static appearance of histopathological morphology, which does not capture differences in the inflammatory response. Therefore, a biomarker grounded in the disease biology is needed in order to understand the molecular heterogeneity of lupus nephritis and identify immunologic mechanism and pathways. Here, we analyzed the patterns of 1000 urine protein biomarkers in 30 patients with active lupus nephritis. We found that patients stratify over a chemokine gradient inducible by IFN-γ. Higher values identified patients with proliferative lupus nephritis. After integrating the urine proteomics with the single-cell transcriptomics of kidney biopsies, we observed that the urinary chemokines defining the gradient were predominantly produced by infiltrating CD8+ T cells, along with natural killer and myeloid cells. The urine chemokine gradient significantly correlated with the number of kidney-infiltrating CD8+ cells. These findings suggest that urine proteomics can capture the complex biology of the kidney in lupus nephritis. Patient-specific pathways could be noninvasively tracked in the urine in real time, enabling diagnosis and personalized treatment.
- Published
- 2020
- Full Text
- View/download PDF
32. Staphylococcus aureus Colonization Is Increased on Lupus Skin Lesions and Is Promoted by IFN-Mediated Barrier Disruption.
- Author
-
Sirobhushanam S, Parsa N, Reed TJ, Berthier CC, Sarkar MK, Hile GA, Tsoi LC, Banfield J, Dobry C, Horswill AR, Gudjonsson JE, and Kahlenberg JM
- Subjects
- Adult, Cell Adhesion, Cell Movement, Cells, Cultured, Female, Filaggrin Proteins, Humans, Interferon Type I metabolism, Interferon-gamma metabolism, Intermediate Filament Proteins genetics, Intermediate Filament Proteins metabolism, Lupus Erythematosus, Systemic microbiology, Male, Membrane Proteins genetics, Membrane Proteins metabolism, Microarray Analysis, Middle Aged, Pregnancy, Sequence Analysis, RNA, Skin microbiology, Skin pathology, Staphylococcal Infections microbiology, Keratinocytes physiology, Lupus Erythematosus, Systemic immunology, Skin immunology, Staphylococcal Infections immunology, Staphylococcus aureus physiology
- Abstract
Cutaneous inflammation is recurrent in systemic lupus erythematosus (SLE), yet mechanisms that drive cutaneous inflammation in SLE are not well defined. Type I IFNs are elevated in nonlesional SLE skin and promote inflammatory responses. Staphylococcus aureus, known to induce IFN production, could play a role in cutaneous inflammation in SLE. We show here that active cutaneous lupus erythematosus lesions are highly colonized (∼50%) by S. aureus. To define the impact of IFNs on S. aureus colonization, we examined the effects of type I and type II IFNs on S. aureus adherence and invasion. An increase in adherent S. aureus was observed after exposure to both IFN-α and -γ, whereas IFN-γ appeared to inhibit invasion of S. aureus. Cutaneous lupus erythematosus lesional skin microarray data and RNA sequencing data from SLE keratinocytes identified repression of barrier gene expression, such as filaggrin and loricrin, and SLE keratinocytes exhibited increased S. aureus-binding integrins. These SLE-associated changes could be replicated by IFN treatment of keratinocytes. Further, SLE keratinocytes exhibited increased binding to S. aureus. Together, these data suggest that chronic exposure to IFNs induces barrier disruption that allows for higher S. aureus colonization in SLE skin., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
33. Single cell transcriptomics identifies focal segmental glomerulosclerosis remission endothelial biomarker.
- Author
-
Menon R, Otto EA, Hoover P, Eddy S, Mariani L, Godfrey B, Berthier CC, Eichinger F, Subramanian L, Harder J, Ju W, Nair V, Larkina M, Naik AS, Luo J, Jain S, Sealfon R, Troyanskaya O, Hacohen N, Hodgin JB, Kretzler M, and Kpmp KPMP
- Subjects
- Biomarkers analysis, Humans, Endothelial Cells pathology, Gene Expression Profiling methods, Glomerulosclerosis, Focal Segmental pathology
- Abstract
To define cellular mechanisms underlying kidney function and failure, the KPMP analyzes biopsy tissue in a multicenter research network to build cell-level process maps of the kidney. This study aimed to establish a single cell RNA sequencing strategy to use cell-level transcriptional profiles from kidney biopsies in KPMP to define molecular subtypes in glomerular diseases. Using multiple sources of adult human kidney reference tissue samples, 22,268 single cell profiles passed KPMP quality control parameters. Unbiased clustering resulted in 31 distinct cell clusters that were linked to kidney and immune cell types using specific cell markers. Focusing on endothelial cell phenotypes, in silico and in situ hybridization methods assigned 3 discrete endothelial cell clusters to distinct renal vascular beds. Transcripts defining glomerular endothelial cells (GEC) were evaluated in biopsies from patients with 10 different glomerular diseases in the NEPTUNE and European Renal cDNA Bank (ERCB) cohort studies. Highest GEC scores were observed in patients with focal segmental glomerulosclerosis (FSGS). Molecular endothelial signatures suggested 2 distinct FSGS patient subgroups with α-2 macroglobulin (A2M) as a key downstream mediator of the endothelial cell phenotype. Finally, glomerular A2M transcript levels associated with lower proteinuria remission rates, linking endothelial function with long-term outcome in FSGS.
- Published
- 2020
- Full Text
- View/download PDF
34. Accelerating Medicines Partnership: Organizational Structure and Preliminary Data From the Phase 1 Studies of Lupus Nephritis.
- Author
-
Hoover P, Der E, Berthier CC, Arazi A, Lederer JA, James JA, Buyon J, Petri M, Belmont HM, Izmirly P, Wofsy D, Hacohen N, Diamond B, Putterman C, and Davidson A
- Subjects
- Biomarkers metabolism, Humans, Lupus Nephritis epidemiology, Lupus Nephritis genetics, Sequence Analysis, RNA methods, United States epidemiology, Academic Medical Centers organization & administration, Clinical Trials, Phase I as Topic methods, Drug Industry organization & administration, Lupus Nephritis metabolism, National Institutes of Health (U.S.) organization & administration, Preliminary Data, Public-Private Sector Partnerships organization & administration
- Abstract
The Accelerating Medicines Partnership (AMP) Lupus Network was established as a partnership between the National Institutes of Health, pharmaceutical companies, nonprofit stakeholders, and lupus investigators across multiple academic centers to apply high-throughput technologies to the analysis of renal tissue, urine, and blood from patients with lupus nephritis (LN). The AMP network provides publicly accessible data to the community with the goal of generating new scientific hypotheses and improving diagnostic and therapeutic tools so as to improve disease outcomes. We present here a description of the structure of the AMP Lupus Network and a summary of the preliminary results from the phase 1 studies. The successful completion of phase 1 sets the stage for analysis of a large cohort of LN samples in phase 2 and provides a model for establishing similar discovery cohorts., (© 2019, American College of Rheumatology.)
- Published
- 2020
- Full Text
- View/download PDF
35. Publisher Correction: The immune cell landscape in kidneys of patients with lupus nephritis.
- Author
-
Arazi A, Rao DA, Berthier CC, Davidson A, Liu Y, Hoover PJ, Chicoine A, Eisenhaure TM, Jonsson AH, Li S, Lieb DJ, Zhang F, Slowikowski K, Browne EP, Noma A, Sutherby D, Steelman S, Smilek DE, Tosta P, Apruzzese W, Massarotti E, Dall'Era M, Park M, Kamen DL, Furie RA, Payan-Schober F, Pendergraft WF 3rd, McInnis EA, Buyon JP, Petri MA, Putterman C, Kalunian KC, Woodle ES, Lederer JA, Hildeman DA, Nusbaum C, Raychaudhuri S, Kretzler M, Anolik JH, Brenner MB, Wofsy D, Hacohen N, and Diamond B
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2019
- Full Text
- View/download PDF
36. IFN-γ enhances cell-mediated cytotoxicity against keratinocytes via JAK2/STAT1 in lichen planus.
- Author
-
Shao S, Tsoi LC, Sarkar MK, Xing X, Xue K, Uppala R, Berthier CC, Zeng C, Patrick M, Billi AC, Fullmer J, Beamer MA, Perez-White B, Getsios S, Schuler A, Voorhees JJ, Choi S, Harms P, Kahlenberg JM, and Gudjonsson JE
- Subjects
- Apoptosis drug effects, Epidermis pathology, Histocompatibility Antigens Class I metabolism, Humans, Inflammation pathology, Keratinocytes drug effects, Lichen Planus genetics, Signal Transduction drug effects, Transcriptome genetics, Cytotoxicity, Immunologic drug effects, Interferon-gamma pharmacology, Janus Kinase 2 metabolism, Keratinocytes immunology, Lichen Planus immunology, STAT1 Transcription Factor metabolism
- Abstract
Lichen planus (LP) is a chronic debilitating inflammatory disease of unknown etiology affecting the skin, nails, and mucosa with no current FDA-approved treatments. It is histologically characterized by dense infiltration of T cells and epidermal keratinocyte apoptosis. Using global transcriptomic profiling of patient skin samples, we demonstrate that LP is characterized by a type II interferon (IFN) inflammatory response. The type II IFN, IFN-γ, is demonstrated to prime keratinocytes and increase their susceptibility to CD8
+ T cell-mediated cytotoxic responses through MHC class I induction in a coculture model. We show that this process is dependent on Janus kinase 2 (JAK2) and signal transducer and activator of transcription 1 (STAT1), but not JAK1 or STAT2 signaling. Last, using drug prediction algorithms, we identify JAK inhibitors as promising therapeutic agents in LP and demonstrate that the JAK1/2 inhibitor baricitinib fully protects keratinocytes against cell-mediated cytotoxic responses in vitro. In summary, this work elucidates the role and mechanisms of IFN-γ in LP pathogenesis and provides evidence for the therapeutic use of JAK inhibitors to limit cell-mediated cytotoxicity in patients with LP., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2019
- Full Text
- View/download PDF
37. Molecular Profiling of Cutaneous Lupus Lesions Identifies Subgroups Distinct from Clinical Phenotypes.
- Author
-
Berthier CC, Tsoi LC, Reed TJ, Stannard JN, Myers EM, Namas R, Xing X, Lazar S, Lowe L, Kretzler M, Gudjonsson JE, and Kahlenberg JM
- Abstract
Cutaneous lupus erythematosus (CLE) is a common manifestation of systemic lupus erythematosus (SLE), and CLE can also develop without systemic involvement. CLE can be difficult to treat and negatively contributes to quality of life. Despite the importance of CLE, our knowledge of what differentiates cutaneous lupus subtypes is limited. Here, we utilized a large cohort of 90 CLE lesional biopsies to compare discoid lupus erythematosus (DLE) and subacute cutaneous lupus (SCLE) in patients with and without associated SLE in order to discern the drivers of disease activity and possibly uncover better treatment targets. Overall, we found that DLE and SCLE share many differentially expressed genes (DEG) reflecting type I interferon (IFN) signaling and repression of EGFR pathways. No differences between CLE only and SLE-associated CLE lesions were found. Of note, DLE uniquely expresses an IFN-γ node. Unbiased cluster analysis of the DEGs identified two groups separated by neutrophilic vs. monocytic signatures that did not sort the patients based on clinical phenotype or disease activity. This suggests that unbiased analysis of the pathobiology of CLE lesions may be important for personalized medicine and targeted therapeutic decision making.
- Published
- 2019
- Full Text
- View/download PDF
38. The immune cell landscape in kidneys of patients with lupus nephritis.
- Author
-
Arazi A, Rao DA, Berthier CC, Davidson A, Liu Y, Hoover PJ, Chicoine A, Eisenhaure TM, Jonsson AH, Li S, Lieb DJ, Zhang F, Slowikowski K, Browne EP, Noma A, Sutherby D, Steelman S, Smilek DE, Tosta P, Apruzzese W, Massarotti E, Dall'Era M, Park M, Kamen DL, Furie RA, Payan-Schober F, Pendergraft WF 3rd, McInnis EA, Buyon JP, Petri MA, Putterman C, Kalunian KC, Woodle ES, Lederer JA, Hildeman DA, Nusbaum C, Raychaudhuri S, Kretzler M, Anolik JH, Brenner MB, Wofsy D, Hacohen N, and Diamond B
- Subjects
- Biomarkers, Biopsy, Cluster Analysis, Computational Biology methods, Epithelial Cells metabolism, Flow Cytometry, Gene Expression Profiling, Gene Expression Regulation, Humans, Immunophenotyping, Interferons metabolism, Kidney metabolism, Kidney pathology, Leukocytes immunology, Leukocytes metabolism, Lupus Nephritis genetics, Lupus Nephritis metabolism, Lupus Nephritis pathology, Lymphocytes immunology, Lymphocytes metabolism, Molecular Sequence Annotation, Myeloid Cells immunology, Myeloid Cells metabolism, Single-Cell Analysis, Transcriptome, Kidney immunology, Lupus Nephritis immunology
- Abstract
Lupus nephritis is a potentially fatal autoimmune disease for which the current treatment is ineffective and often toxic. To develop mechanistic hypotheses of disease, we analyzed kidney samples from patients with lupus nephritis and from healthy control subjects using single-cell RNA sequencing. Our analysis revealed 21 subsets of leukocytes active in disease, including multiple populations of myeloid cells, T cells, natural killer cells and B cells that demonstrated both pro-inflammatory responses and inflammation-resolving responses. We found evidence of local activation of B cells correlated with an age-associated B-cell signature and evidence of progressive stages of monocyte differentiation within the kidney. A clear interferon response was observed in most cells. Two chemokine receptors, CXCR4 and CX3CR1, were broadly expressed, implying a potentially central role in cell trafficking. Gene expression of immune cells in urine and kidney was highly correlated, which would suggest that urine might serve as a surrogate for kidney biopsies.
- Published
- 2019
- Full Text
- View/download PDF
39. The female-biased factor VGLL3 drives cutaneous and systemic autoimmunity.
- Author
-
Billi AC, Gharaee-Kermani M, Fullmer J, Tsoi LC, Hill BD, Gruszka D, Ludwig J, Xing X, Estadt S, Wolf SJ, Rizvi SM, Berthier CC, Hodgin JB, Beamer MA, Sarkar MK, Liang Y, Uppala R, Shao S, Zeng C, Harms PW, Verhaegen ME, Voorhees JJ, Wen F, Ward NL, Dlugosz AA, Kahlenberg JM, and Gudjonsson JE
- Subjects
- Animals, Disease Models, Animal, Female, Humans, Lupus Erythematosus, Cutaneous genetics, Lupus Erythematosus, Cutaneous pathology, Lupus Erythematosus, Systemic genetics, Lupus Erythematosus, Systemic pathology, Male, Mice, Mice, Transgenic, Sex Factors, Skin immunology, Skin pathology, Transcription Factors genetics, Autoimmunity genetics, Gene Expression Regulation immunology, Lupus Erythematosus, Cutaneous immunology, Lupus Erythematosus, Systemic immunology, Transcription Factors metabolism
- Abstract
Autoimmune disease is 4 times more common in women than men. This bias is largely unexplained. Female skin is "autoimmunity prone," showing upregulation of many proinflammatory genes, even in healthy women. We previously identified VGLL3 as a putative transcription cofactor enriched in female skin. Here, we demonstrate that skin-directed overexpression of murine VGLL3 causes a severe lupus-like rash and systemic autoimmune disease that involves B cell expansion, autoantibody production, immune complex deposition, and end-organ damage. Excess epidermal VGLL3 drives a proinflammatory gene expression program that overlaps with both female skin and cutaneous lupus. This includes increased B cell-activating factor (BAFF), the only current biologic target in systemic lupus erythematosus (SLE); IFN-κ, a key inflammatory mediator in cutaneous lupus; and CXCL13, a biomarker of early-onset SLE and renal involvement. Our results demonstrate that skin-targeted overexpression of the female-biased factor VGLL3 is sufficient to drive cutaneous and systemic autoimmune disease that is strikingly similar to SLE. This work strongly implicates VGLL3 as a pivotal orchestrator of sex-biased autoimmunity.
- Published
- 2019
- Full Text
- View/download PDF
40. Hypersensitive IFN Responses in Lupus Keratinocytes Reveal Key Mechanistic Determinants in Cutaneous Lupus.
- Author
-
Tsoi LC, Hile GA, Berthier CC, Sarkar MK, Reed TJ, Liu J, Uppala R, Patrick M, Raja K, Xing X, Xing E, He K, Gudjonsson JE, and Kahlenberg JM
- Subjects
- Adult, Female, Humans, Male, Gene Expression Regulation immunology, Interferons immunology, Keratinocytes immunology, Lupus Erythematosus, Cutaneous immunology, Paired Box Transcription Factors immunology
- Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease in which 70% of patients experience disfiguring skin inflammation (grouped under the rubric of cutaneous lupus erythematosus [CLE]). There are limited treatment options for SLE and no Food and Drug Administration-approved therapies for CLE. Studies have revealed that IFNs are important mediators for SLE and CLE, but the mechanisms by which IFNs lead to disease are still poorly understood. We aimed to investigate how IFN responses in SLE keratinocytes contribute to development of CLE. A cohort of 72 RNA sequencing samples from 14 individuals (seven SLE and seven healthy controls) were analyzed to study the transcriptomic effects of type I and type II IFNs on SLE versus control keratinocytes. In-depth analysis of the IFN responses was conducted. Bioinformatics and functional assays were conducted to provide implications for the change of IFN response. A significant hypersensitive response to IFNs was identified in lupus keratinocytes, including genes ( IFIH1 , STAT1 , and IRF7 ) encompassed in SLE susceptibility loci. Binding sites for the transcription factor PITX1 were enriched in genes that exhibit IFN-sensitive responses. PITX1 expression was increased in CLE lesions based on immunohistochemistry, and by using small interfering RNA knockdown, we illustrated that PITX1 was required for upregulation of IFN-regulated genes in vitro. SLE patients exhibit increased IFN signatures in their skin secondary to increased production and a robust, skewed IFN response that is regulated by PITX1. Targeting these exaggerated pathways may prove to be beneficial to prevent and treat hyperinflammatory responses in SLE skin., (Copyright © 2019 by The American Association of Immunologists, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
41. Photosensitivity and type I IFN responses in cutaneous lupus are driven by epidermal-derived interferon kappa.
- Author
-
Sarkar MK, Hile GA, Tsoi LC, Xing X, Liu J, Liang Y, Berthier CC, Swindell WR, Patrick MT, Shao S, Tsou PS, Uppala R, Beamer MA, Srivastava A, Bielas SL, Harms PW, Getsios S, Elder JT, Voorhees JJ, Gudjonsson JE, and Kahlenberg JM
- Subjects
- Adult, Cells, Cultured, Dendritic Cells immunology, Female, Humans, Interferon Type I genetics, Interferon Type I immunology, Keratinocytes immunology, Lupus Erythematosus, Cutaneous immunology, Male, Middle Aged, Photosensitivity Disorders immunology, RNA, Messenger genetics, Skin immunology, TYK2 Kinase immunology, Up-Regulation immunology, Epidermis immunology, Interferon Type I biosynthesis, Lupus Erythematosus, Cutaneous complications, Photosensitivity Disorders etiology
- Abstract
Objective: Skin inflammation and photosensitivity are common in patients with cutaneous lupus erythematosus (CLE) and systemic lupus erythematosus (SLE), yet little is known about the mechanisms that regulate these traits. Here we investigate the role of interferon kappa (IFN-κ) in regulation of type I interferon (IFN) and photosensitive responses and examine its dysregulation in lupus skin., Methods: mRNA expression of type I IFN genes was analysed from microarray data of CLE lesions and healthy control skin. Similar expression in cultured primary keratinocytes, fibroblasts and endothelial cells was analysed via RNA-seq. IFNK knock-out (KO) keratinocytes were generated using CRISPR/Cas9. Keratinocytes stably overexpressing IFN-κ were created via G418 selection of transfected cells. IFN responses were assessed via phosphorylation of STAT1 and STAT2 and qRT-PCR for IFN-regulated genes. Ultraviolet B-mediated apoptosis was analysed via TUNEL staining. In vivo protein expression was assessed via immunofluorescent staining of normal and CLE lesional skin., Results: IFNK is one of two type I IFNs significantly increased (1.5-fold change, false discovery rate (FDR) q<0.001) in lesional CLE skin. Gene ontology (GO) analysis showed that type I IFN responses were enriched (FDR=6.8×10
-04 ) in keratinocytes not in fibroblast and endothelial cells, and this epithelial-derived IFN-κ is responsible for maintaining baseline type I IFN responses in healthy skin. Increased levels of IFN-κ, such as seen in SLE, amplify and accelerate responsiveness of epithelia to IFN-α and increase keratinocyte sensitivity to UV irradiation. Notably, KO of IFN-κ or inhibition of IFN signalling with baricitinib abrogates UVB-induced apoptosis., Conclusion: Collectively, our data identify IFN-κ as a critical IFN in CLE pathology via promotion of enhanced IFN responses and photosensitivity. IFN-κ is a potential novel target for UVB prophylaxis and CLE-directed therapy., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2018. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2018
- Full Text
- View/download PDF
42. ABIN1 Determines Severity of Glomerulonephritis via Activation of Intrinsic Glomerular Inflammation.
- Author
-
Korte EA, Caster DJ, Barati MT, Tan M, Zheng S, Berthier CC, Brosius FC 3rd, Vieyra MB, Sheehan RM, Kosiewicz M, Wysoczynski M, Gaffney PM, Salant DJ, McLeish KR, and Powell DW
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Glomerulonephritis metabolism, Humans, Inflammation metabolism, Inflammation pathology, Mice, Mice, Mutant Strains, DNA-Binding Proteins metabolism, Glomerulonephritis pathology, NF-kappa B metabolism, Podocytes metabolism
- Abstract
Transcription factor NF-κB regulates expression of numerous genes that control inflammation and is activated in glomerular cells in glomerulonephritis (GN). We previously identified genetic variants for a NF-κB regulatory, ubiquitin-binding protein ABIN1 as risk factors for GN in systemic autoimmunity. The goal was to define glomerular inflammatory events controlled by ABIN1 function in GN. Nephrotoxic serum nephritis was induced in wild-type (WT) and ubiquitin-binding deficient ABIN1[D485N] mice, and renal pathophysiology and glomerular inflammatory phenotypes were assessed. Proteinuria was also measured in ABIN1[D485N] mice transplanted with WT mouse bone marrow. Inflammatory activation of ABIN1[D472N] (D485N homolog) cultured human-derived podocytes, and interaction with primary human neutrophils were also assessed. Disruption of ABIN1 function exacerbated proteinuria, podocyte injury, glomerular NF-κB activity, glomerular expression of inflammatory mediators, and glomerular recruitment and retention of neutrophils in antibody-mediated nephritis. Transplantation of WT bone marrow did not prevent the increased proteinuria in ABIN1[D845N] mice. Tumor necrosis factor-stimulated enhanced expression and secretion of NF-κB-targeted proinflammatory mediators in ABIN1[D472N] cultured podocytes compared with WT cells. Supernatants from ABIN1[D472N] podocytes accelerated chemotaxis of human neutrophils, and ABIN1[D472N] podocytes displayed a greater susceptibility to injurious morphologic findings induced by neutrophil granule contents. These studies define a novel role for ABIN1 dysfunction and NF-κB in mediating GN through proinflammatory activation of podocytes., (Copyright © 2017 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
43. A systems approach to renal inflammation in SLE.
- Author
-
Berthier CC, Kretzler M, and Davidson A
- Subjects
- Animals, Genotype, Humans, Kidney, Lupus Nephritis complications, Phenotype, Renal Insufficiency, Chronic etiology, Renal Insufficiency, Chronic genetics, Lupus Nephritis genetics
- Abstract
Lupus disease and its complications including lupus nephritis (LN) are very disabling and significantly impact the quality of life and longevity of patients. Broadly immunosuppressive treatments do not always provide the expected clinical benefits and have significant side effects that contribute to patient morbidity. In the era of systems biology, new strategies are being deployed integrating diverse sources of information (molecular and clinical) so as to identify individual disease specificities and select less aggressive treatments. In this review, we summarize integrative approaches linking molecular disease profiles (mainly tissue transcriptomics) and clinical phenotypes. The main goals are to better understand the pathogenesis of lupus nephritis, to identify the risk factors for renal flare and to find the predictors of both short and long-term clinical outcome. Identification of common key drivers and additional patient-specific key drivers can open the door to improved and individualized therapy to prevent and treat LN., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
44. Scleroderma keratinocytes promote fibroblast activation independent of transforming growth factor beta.
- Author
-
McCoy SS, Reed TJ, Berthier CC, Tsou PS, Liu J, Gudjonsson JE, Khanna D, and Kahlenberg JM
- Subjects
- Actins metabolism, Adult, Aged, Cells, Cultured, Chemokine CCL5 genetics, Chemokine CCL5 metabolism, Collagen Type I genetics, Collagen Type I metabolism, Collagen Type I, alpha 1 Chain, Culture Media, Conditioned, Down-Regulation, Female, Fibroblasts metabolism, Fibrosis, Gene Expression Profiling, Humans, Immunohistochemistry, Keratinocytes metabolism, Male, Middle Aged, NF-kappa B metabolism, PPAR gamma metabolism, RNA, Messenger metabolism, Real-Time Polymerase Chain Reaction, Scleroderma, Diffuse, Scleroderma, Localized, Scleroderma, Systemic genetics, Scleroderma, Systemic pathology, Skin pathology, Up-Regulation, Cell Differentiation, Fibroblasts cytology, Keratinocytes cytology, Scleroderma, Systemic metabolism, Transforming Growth Factor beta metabolism
- Abstract
Objectives: SSc is a devastating disease that results in fibrosis of the skin and other organs. Fibroblasts are a key driver of the fibrotic process through deposition of extracellular matrix. The mechanisms by which fibroblasts are induced to become pro-fibrotic remain unclear. Thus, we examined the ability of SSc keratinocytes to promote fibroblast activation and the source of this effect., Methods: Keratinocytes were isolated from skin biopsies of 9 lcSSc, 10 dcSSc and 13 control patients. Conditioned media was saved from the cultures. Normal fresh primary fibroblasts were exposed to healthy control and SSc keratinocyte conditioned media in the presence or absence of neutralizing antibodies for TGF-β. Gene expression was assessed by microarrays and real-time PCR. Immunocytochemistry was performed for α-smooth muscle actin (α-SMA), collagen type 1 (COL1A1) and CCL5 expression., Results: SSc keratinocyte conditioned media promoted fibroblast activation, characterized by increased α-SMA and COL1A1 mRNA and protein expression. This effect was independent of TGF-β. Microarray analysis identified upregulation of nuclear factor κB (NF-κB) and downregulation of peroxisome proliferator-activated receptor γ (PPAR-γ) pathways in both SSc subtypes. Scleroderma keratinocytes exhibited increased expression of NF-κB-regulated cytokines and chemokines and lesional skin staining confirmed upregulation of CCL5 in basal keratinocytes., Conclusion: Scleroderma keratinocytes promote the activation of fibroblasts in a TGF-β-independent manner and demonstrate an imbalance in NF-κB1 and PPAR-γ expression leading to increased cytokine and CCL5 production. Further study of keratinocyte mediators of fibrosis, including CCL5, may provide novel targets for skin fibrosis therapy., (© The Author 2017. Published by Oxford University Press on behalf of the British Society for Rheumatology. All rights reserved. For Permissions, please email: journals.permissions@oup.com)
- Published
- 2017
- Full Text
- View/download PDF
45. Transcriptomic and Proteomic Profiling Provides Insight into Mesangial Cell Function in IgA Nephropathy.
- Author
-
Liu P, Lassén E, Nair V, Berthier CC, Suguro M, Sihlbom C, Kretzler M, Betsholtz C, Haraldsson B, Ju W, Ebefors K, and Nyström J
- Subjects
- Adult, Case-Control Studies, Cells, Cultured, Female, Gene Expression Profiling, Glomerulonephritis, IGA genetics, Humans, Male, Middle Aged, Proteome, Transcriptome, Glomerulonephritis, IGA metabolism, Mesangial Cells metabolism
- Abstract
IgA nephropathy (IgAN), the most common GN worldwide, is characterized by circulating galactose-deficient IgA (gd-IgA) that forms immune complexes. The immune complexes are deposited in the glomerular mesangium, leading to inflammation and loss of renal function, but the complete pathophysiology of the disease is not understood. Using an integrated global transcriptomic and proteomic profiling approach, we investigated the role of the mesangium in the onset and progression of IgAN. Global gene expression was investigated by microarray analysis of the glomerular compartment of renal biopsy specimens from patients with IgAN ( n =19) and controls ( n =22). Using curated glomerular cell type-specific genes from the published literature, we found differential expression of a much higher percentage of mesangial cell-positive standard genes than podocyte-positive standard genes in IgAN. Principal coordinate analysis of expression data revealed clear separation of patient and control samples on the basis of mesangial but not podocyte cell-positive standard genes. Additionally, patient clinical parameters (serum creatinine values and eGFRs) significantly correlated with Z scores derived from the expression profile of mesangial cell-positive standard genes. Among patients grouped according to Oxford MEST score, patients with segmental glomerulosclerosis had a significantly higher mesangial cell-positive standard gene Z score than patients without segmental glomerulosclerosis. By investigating mesangial cell proteomics and glomerular transcriptomics, we identified 22 common pathways induced in mesangial cells by gd-IgA, most of which mediate inflammation. The genes, proteins, and corresponding pathways identified provide novel insights into the pathophysiologic mechanisms leading to IgAN., (Copyright © 2017 by the American Society of Nephrology.)
- Published
- 2017
- Full Text
- View/download PDF
46. Enhanced Inflammasome Activity in Systemic Lupus Erythematosus Is Mediated via Type I Interferon-Induced Up-Regulation of Interferon Regulatory Factor 1.
- Author
-
Liu J, Berthier CC, and Kahlenberg JM
- Subjects
- Adult, Case-Control Studies, Enzyme-Linked Immunosorbent Assay, Female, Humans, Male, Middle Aged, Monocytes metabolism, Prospective Studies, Real-Time Polymerase Chain Reaction, Inflammasomes physiology, Interferon Regulatory Factor-1 physiology, Interferon Type I physiology, Lupus Erythematosus, Systemic physiopathology, Up-Regulation
- Abstract
Objective: The inflammasome complex is a driver of organ damage in patients with systemic lupus erythematosus (SLE). Although type I interferons (IFNs) are well established as mediators of SLE pathogenesis, their role in inflammasome activation in SLE has not been assessed. The aim of this study was to examine type I IFNs as regulators of the inflammasome., Methods: SLE patients fulfilled ≥4 American College of Rheumatology criteria and were recruited from the University of Michigan Lupus Cohort. Primary monocytes were isolated from SLE patients or healthy controls by negative selection, treated with inflammasome activators in the presence or absence of IFNα, and IL-1β secretion was measured by enzyme-linked immunosorbent assay. Expression levels of IFN and inflammasome-related molecules were assessed by real-time polymerase chain reaction and Western blotting. IFN regulatory factor 1 (IRF-1) expression was specifically down-regulated by small interfering RNA (siRNA) transfection and a chemical inhibitor., Results: Monocytes from patients with SLE exhibited increased expression and enhanced activation of the inflammasome by ATP when compared with control monocytes. Expression of inflammasome and IFN-regulated genes was significantly correlated in monocytes from SLE patients but not in control monocytes. Inflammasome activity was increased after prolonged exposure to IFNα. Reduction of IRF-1 expression via siRNA blocked caspase 1 up-regulation after treatment with IFNα. Importantly, hyperactivity of the inflammasome in the monocytes of SLE patients was significantly reduced after knockdown or inhibition of IRF-1., Conclusion: Prolonged type I IFN exposure, as seen in SLE patients, primes monocytes for robust inflammasome activation in an IRF-1-dependent manner. IRF-1 inhibition may serve as a novel target for treatment of SLE-associated inflammation and organ damage., (© 2017, American College of Rheumatology.)
- Published
- 2017
- Full Text
- View/download PDF
47. A gene network regulated by the transcription factor VGLL3 as a promoter of sex-biased autoimmune diseases.
- Author
-
Liang Y, Tsoi LC, Xing X, Beamer MA, Swindell WR, Sarkar MK, Berthier CC, Stuart PE, Harms PW, Nair RP, Elder JT, Voorhees JJ, Kahlenberg JM, and Gudjonsson JE
- Subjects
- Adult, Aged, Aged, 80 and over, Cells, Cultured, Female, Gene Expression Profiling, Genetic Association Studies, Genome-Wide Association Study, Humans, Male, Middle Aged, Quantitative Trait Loci, Transcription Factors genetics, Transcriptome, Young Adult, Gene Regulatory Networks, Keratinocytes physiology, Lupus Erythematosus, Cutaneous genetics, Scleroderma, Systemic genetics, Sex Factors, Sjogren's Syndrome genetics, Skin pathology, Transcription Factors metabolism
- Abstract
Autoimmune diseases affect 7.5% of the US population, and they are among the leading causes of death and disability. A notable feature of many autoimmune diseases is their greater prevalence in females than in males, but the underlying mechanisms of this have remained unclear. Through the use of high-resolution global transcriptome analyses, we demonstrated a female-biased molecular signature associated with susceptibility to autoimmune disease and linked this to extensive sex-dependent co-expression networks. This signature was independent of biological age and sex-hormone regulation and was regulated by the transcription factor VGLL3, which also had a strong female-biased expression. On a genome-wide level, VGLL3-regulated genes had a strong association with multiple autoimmune diseases, including lupus, scleroderma and Sjögren's syndrome, and had a prominent transcriptomic overlap with inflammatory processes in cutaneous lupus. These results identified a VGLL3-regulated network as a previously unknown inflammatory pathway that promotes female-biased autoimmunity. They demonstrate the importance of studying immunological processes in females and males separately and suggest new avenues for therapeutic development., Competing Interests: Competing Financial Interests. The authors declare no competing financial interests.
- Published
- 2017
- Full Text
- View/download PDF
48. Tissue transcriptome-driven identification of epidermal growth factor as a chronic kidney disease biomarker.
- Author
-
Ju W, Nair V, Smith S, Zhu L, Shedden K, Song PXK, Mariani LH, Eichinger FH, Berthier CC, Randolph A, Lai JY, Zhou Y, Hawkins JJ, Bitzer M, Sampson MG, Thier M, Solier C, Duran-Pacheco GC, Duchateau-Nguyen G, Essioux L, Schott B, Formentini I, Magnone MC, Bobadilla M, Cohen CD, Bagnasco SM, Barisoni L, Lv J, Zhang H, Wang HY, Brosius FC, Gadegbeku CA, and Kretzler M
- Subjects
- Adult, Aged, Biomarkers urine, Biopsy, Cell Differentiation, Cohort Studies, Disease Progression, Female, Glomerular Filtration Rate, Humans, Male, Middle Aged, Prognosis, Proteins chemistry, Regeneration, Renal Insufficiency, Chronic urine, Epidermal Growth Factor urine, Renal Insufficiency, Chronic diagnosis, Transcriptome
- Abstract
Chronic kidney disease (CKD) affects 8 to 16% people worldwide, with an increasing incidence and prevalence of end-stage kidney disease (ESKD). The effective management of CKD is confounded by the inability to identify patients at high risk of progression while in early stages of CKD. To address this challenge, a renal biopsy transcriptome-driven approach was applied to develop noninvasive prognostic biomarkers for CKD progression. Expression of intrarenal transcripts was correlated with the baseline estimated glomerular filtration rate (eGFR) in 261 patients. Proteins encoded by eGFR-associated transcripts were tested in urine for association with renal tissue injury and baseline eGFR. The ability to predict CKD progression, defined as the composite of ESKD or 40% reduction of baseline eGFR, was then determined in three independent CKD cohorts. A panel of intrarenal transcripts, including epidermal growth factor (EGF), a tubule-specific protein critical for cell differentiation and regeneration, predicted eGFR. The amount of EGF protein in urine (uEGF) showed significant correlation (P < 0.001) with intrarenal EGF mRNA, interstitial fibrosis/tubular atrophy, eGFR, and rate of eGFR loss. Prediction of the composite renal end point by age, gender, eGFR, and albuminuria was significantly (P < 0.001) improved by addition of uEGF, with an increase of the C-statistic from 0.75 to 0.87. Outcome predictions were replicated in two independent CKD cohorts. Our approach identified uEGF as an independent risk predictor of CKD progression. Addition of uEGF to standard clinical parameters improved the prediction of disease events in diverse CKD populations with a wide spectrum of causes and stages., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
49. Lupus nephritis susceptibility loci in women with systemic lupus erythematosus.
- Author
-
Chung SA, Brown EE, Williams AH, Ramos PS, Berthier CC, Bhangale T, Alarcon-Riquelme ME, Behrens TW, Criswell LA, Graham DC, Demirci FY, Edberg JC, Gaffney PM, Harley JB, Jacob CO, Kamboh MI, Kelly JA, Manzi S, Moser-Sivils KL, Russell LP, Petri M, Tsao BP, Vyse TJ, Zidovetzki R, Kretzler M, Kimberly RP, Freedman BI, Graham RR, and Langefeld CD
- Subjects
- Adolescent, Adult, Aged, Child, Female, Genome-Wide Association Study, Genotype, HLA-DR2 Antigen genetics, HLA-DR3 Antigen genetics, Humans, Lupus Erythematosus, Systemic epidemiology, Lupus Erythematosus, Systemic physiopathology, Middle Aged, Phenotype, Polymorphism, Single Nucleotide, RNA, Messenger metabolism, White People genetics, Young Adult, Genetic Predisposition to Disease, Lupus Erythematosus, Systemic genetics, Lupus Nephritis genetics
- Abstract
Lupus nephritis is a manifestation of SLE resulting from glomerular immune complex deposition and inflammation. Lupus nephritis demonstrates familial aggregation and accounts for significant morbidity and mortality. We completed a meta-analysis of three genome-wide association studies of SLE to identify lupus nephritis-predisposing loci. Through genotyping and imputation, >1.6 million markers were assessed in 2000 unrelated women of European descent with SLE (588 patients with lupus nephritis and 1412 patients with lupus without nephritis). Tests of association were computed using logistic regression adjusting for population substructure. The strongest evidence for association was observed outside the MHC and included markers localized to 4q11-q13 (PDGFRA, GSX2; P=4.5×10(-7)), 16p12 (SLC5A11; P=5.1×10(-7)), 6p22 (ID4; P=7.4×10(-7)), and 8q24.12 (HAS2, SNTB1; P=1.1×10(-6)). Both HLA-DR2 and HLA-DR3, two well established lupus susceptibility loci, showed evidence of association with lupus nephritis (P=0.06 and P=3.7×10(-5), respectively). Within the class I region, rs9263871 (C6orf15-HCG22) had the strongest evidence of association with lupus nephritis independent of HLA-DR2 and HLA-DR3 (P=8.5×10(-6)). Consistent with a functional role in lupus nephritis, intra-renal mRNA levels of PDGFRA and associated pathway members showed significant enrichment in patients with lupus nephritis (n=32) compared with controls (n=15). Results from this large-scale genome-wide investigation of lupus nephritis provide evidence of multiple biologically relevant lupus nephritis susceptibility loci., (Copyright © 2014 by the American Society of Nephrology.)
- Published
- 2014
- Full Text
- View/download PDF
50. The molecular phenotype of endocapillary proliferation: novel therapeutic targets for IgA nephropathy.
- Author
-
Hodgin JB, Berthier CC, John R, Grone E, Porubsky S, Gröne HJ, Herzenberg AM, Scholey JW, Hladunewich M, Cattran DC, Kretzler M, and Reich HN
- Subjects
- Adrenal Cortex Hormones therapeutic use, Adult, Computer Simulation, Drug Evaluation, Preclinical, Female, Glomerulonephritis, IGA drug therapy, Glomerulonephritis, IGA pathology, Humans, Kidney Glomerulus blood supply, Kidney Glomerulus pathology, Male, Middle Aged, Molecular Targeted Therapy, Neovascularization, Pathologic metabolism, Phenotype, Young Adult, Glomerulonephritis, IGA metabolism, Transcriptome
- Abstract
IgA nephropathy (IgAN) is a clinically and pathologically heterogeneous disease. Endocapillary proliferation is associated with higher risk of progressive disease, and clinical studies suggest that corticosteroids mitigate this risk. However, corticosteroids are associated with protean cellular effects and significant toxicity. Furthermore the precise mechanism by which they modulate kidney injury in IgAN is not well delineated. To better understand molecular pathways involved in the development of endocapillary proliferation and to identify novel specific therapeutic targets, we evaluated the glomerular transcriptome of microdissected kidney biopsies from 22 patients with IgAN. Endocapillary proliferation was defined according to the Oxford scoring system independently by 3 nephropathologists. We analyzed mRNA expression using microarrays and identified transcripts differentially expressed in patients with endocapillary proliferation compared to IgAN without endocapillary lesions. Next, we employed both transcription factor analysis and in silico drug screening and confirmed that the endocapillary proliferation transcriptome is significantly enriched with pathways that can be impacted by corticosteroids. With this approach we also identified novel therapeutic targets and bioactive small molecules that may be considered for therapeutic trials for the treatment of IgAN, including resveratrol and hydroquinine. In summary, we have defined the distinct molecular profile of a pathologic phenotype associated with progressive renal insufficiency in IgAN. Exploration of the pathways associated with endocapillary proliferation confirms a molecular basis for the clinical effectiveness of corticosteroids in this subgroup of IgAN, and elucidates new therapeutic strategies for IgAN.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.