Go to The Journal of Clinical Investigation
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Transfers
  • Advertising
  • Job board
  • Contact
  • Physician-Scientist Development
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Immunology
    • Metabolism
    • Nephrology
    • Oncology
    • Pulmonology
    • All ...
  • Videos
  • Collections
    • In-Press Preview
    • Resource and Technical Advances
    • Clinical Research and Public Health
    • Research Letters
    • Editorials
    • Perspectives
    • Physician-Scientist Development
    • Reviews
    • Top read articles

  • Current issue
  • Past issues
  • Specialties
  • In-Press Preview
  • Resource and Technical Advances
  • Clinical Research and Public Health
  • Research Letters
  • Editorials
  • Perspectives
  • Physician-Scientist Development
  • Reviews
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Transfers
  • Advertising
  • Job board
  • Contact

Autoimmunity

  • 308 Articles
  • 0 Posts
  • ← Previous
  • 1
  • 2
  • …
  • 24
  • 25
  • 26
  • …
  • 30
  • 31
  • Next →
CD19+IgM+ cells demonstrate enhanced therapeutic efficacy in type 1 diabetes mellitus
Andrew D. Vonberg, Maria Acevedo-Calado, Aaron R. Cox, Susan L. Pietropaolo, Roberto Gianani, Steven K. Lundy, Massimo Pietropaolo
Andrew D. Vonberg, Maria Acevedo-Calado, Aaron R. Cox, Susan L. Pietropaolo, Roberto Gianani, Steven K. Lundy, Massimo Pietropaolo
View: Text | PDF

CD19+IgM+ cells demonstrate enhanced therapeutic efficacy in type 1 diabetes mellitus

  • Text
  • PDF
Abstract

We describe a protective effect on autoimmune diabetes and reduced destructive insulitis in NOD.scid recipients following splenocyte injections from diabetic NOD donors and sorted CD19+ cells compared with NOD.scid recipients receiving splenocytes alone. This protective effect was age specific (only CD19+ cells from young NOD donors exerted this effect; P < 0.001). We found that the CD19+IgM+ cell is the primary subpopulation of B cells that delayed transfer of diabetes mediated by diabetogenic T cells from NOD mice (P = 0.002). Removal of IgM+ cells from the CD19+ pool did not result in protection. Notably, protection conferred by CD19+IgM+ cotransfers were not dependent on the presence of Tregs, as their depletion did not affect their ability to delay onset of diabetes. Blockade of IL-10 with neutralizing antibodies at the time of CD19+ cell cotransfers also abrogated the therapeutic effect, suggesting that IL-10 secretion was an important component of protection. These results were strengthened by ex vivo incubation of CD19+ cells with IL-5, resulting in enhanced proliferation and IL-10 production and equivalently delayed diabetes progression (P = 0.0005). The potential to expand CD19+IgM+ cells, especially in response to IL-5 stimulation or by pharmacologic agents, may be a new therapeutic option for type 1 diabetes.

Authors

Andrew D. Vonberg, Maria Acevedo-Calado, Aaron R. Cox, Susan L. Pietropaolo, Roberto Gianani, Steven K. Lundy, Massimo Pietropaolo

×

Dysregulated NK cell PLCγ2 signaling and activity in juvenile dermatomyositis
Allison A. Throm, Joshua B. Alinger, Jeanette T. Pingel, Allyssa L. Daugherty, Lauren M. Pachman, Anthony R. French
Allison A. Throm, Joshua B. Alinger, Jeanette T. Pingel, Allyssa L. Daugherty, Lauren M. Pachman, Anthony R. French
View: Text | PDF

Dysregulated NK cell PLCγ2 signaling and activity in juvenile dermatomyositis

  • Text
  • PDF
Abstract

Juvenile dermatomyositis (JDM) is a debilitating pediatric autoimmune disease manifesting with characteristic rash and muscle weakness. To delineate signaling abnormalities in JDM, mass cytometry was performed with PBMCs from treatment-naive JDM patients and controls. NK cell percentages were lower while frequencies of naive B cells and naive CD4+ T cells were higher in JDM patients than in controls. These cell frequency differences were attenuated with cessation of active disease. A large number of signaling differences were identified in treatment-naive JDM patients compared with controls. Classification models incorporating feature selection demonstrated that differences in phospholipase Cγ2 (PLCγ2) phosphorylation comprised 10 of 12 features (i.e., phosphoprotein in a specific immune cell subset) distinguishing the 2 groups. Because NK cells represented 5 of these 12 features, further studies focused on the PLCγ2 pathway in NK cells, which is responsible for stimulating calcium flux and cytotoxic granule movement. No differences were detected in upstream signaling or total PLCγ2 protein levels. Hypophosphorylation of PLCγ2 and downstream mitogen-activated protein kinase-activated protein kinase 2 were partially attenuated with cessation of active disease. PLCγ2 hypophosphorylation in treatment-naive JDM patients resulted in decreased calcium flux. The identification of dysregulation of PLCγ2 phosphorylation and decreased calcium flux in NK cells provides potential mechanistic insight into JDM pathogenesis.

Authors

Allison A. Throm, Joshua B. Alinger, Jeanette T. Pingel, Allyssa L. Daugherty, Lauren M. Pachman, Anthony R. French

×

The DILfrequency study is an adaptive trial to identify optimal IL-2 dosing in patients with type 1 diabetes
Eleonora Seelig, James Howlett, Linsey Porter, Lucy Truman, James Heywood, Jane Kennet, Emma L. Arbon, Katerina Anselmiova, Neil M. Walker, Ravinder Atkar, Marcin L. Pekalski, Ed Rytina, Mark Evans, Linda S. Wicker, John A. Todd, Adrian P. Mander, Simon Bond, Frank Waldron-Lynch
Eleonora Seelig, James Howlett, Linsey Porter, Lucy Truman, James Heywood, Jane Kennet, Emma L. Arbon, Katerina Anselmiova, Neil M. Walker, Ravinder Atkar, Marcin L. Pekalski, Ed Rytina, Mark Evans, Linda S. Wicker, John A. Todd, Adrian P. Mander, Simon Bond, Frank Waldron-Lynch
View: Text | PDF

The DILfrequency study is an adaptive trial to identify optimal IL-2 dosing in patients with type 1 diabetes

  • Text
  • PDF
Abstract

BACKGROUND. Type 1 diabetes (T1D) results from loss of immune regulation, leading to the development of autoimmunity to pancreatic β cells, involving autoreactive T effector cells (Teffs). Tregs, which prevent autoimmunity, require IL-2 for maintenance of immunosuppressive functions. Using a response-adaptive design, we aimed to determine the optimal regimen of aldesleukin (recombinant human IL-2) to physiologically enhance Tregs while limiting expansion of Teffs. METHODS. DILfrequency is a nonrandomized, open-label, response-adaptive study of participants, aged 18–70 years, with T1D. The initial learning phase allocated 12 participants to 6 different predefined regimens. Then, 3 cohorts of 8 participants were sequentially allocated dose frequencies, based on repeated interim analyses of all accumulated trial data. The coprimary endpoints were percentage change in Tregs and Teffs and CD25 (α subunit of the IL-2 receptor) expression by Tregs, from baseline to steady state. RESULTS. Thirty-eight participants were enrolled, with thirty-six completing treatment. The optimal regimen to maintain a steady-state increase in Tregs of 30% and CD25 expression of 25% without Teff expansion is 0.26 × 106 IU/m2 (95% CI –0.007 to 0.485) every 3 days. Tregs and CD25 were dose-frequency responsive, Teffs were not. The commonest adverse event was injection site reaction (464 of 694 events). CONCLUSIONS. Using a response-adaptive design, aldesleukin treatment can be optimized. Our methodology can generally be employed to immediately access proof of mechanism, thereby leading to more efficient and safe drug development. TRIAL REGISTRATION. International Standard Randomised Controlled Trial Number Register, ISRCTN40319192; ClinicalTrials.gov, NCT02265809. FUNDING. Sir Jules Thorn Trust, the Swiss National Science Foundation, Wellcome, JDRF, and NIHR Cambridge Biomedical Research Centre.

Authors

Eleonora Seelig, James Howlett, Linsey Porter, Lucy Truman, James Heywood, Jane Kennet, Emma L. Arbon, Katerina Anselmiova, Neil M. Walker, Ravinder Atkar, Marcin L. Pekalski, Ed Rytina, Mark Evans, Linda S. Wicker, John A. Todd, Adrian P. Mander, Simon Bond, Frank Waldron-Lynch

×

High-throughput T cell receptor sequencing identifies clonally expanded CD8+ T cell populations in alopecia areata
Annemieke de Jong, Ali Jabbari, Zhenpeng Dai, Luzhou Xing, Dustin Lee, Mei Mei Li, Madeleine Duvic, Maria Hordinsky, David A. Norris, Vera Price, Julian Mackay-Wiggan, Raphael Clynes, Angela M. Christiano
Annemieke de Jong, Ali Jabbari, Zhenpeng Dai, Luzhou Xing, Dustin Lee, Mei Mei Li, Madeleine Duvic, Maria Hordinsky, David A. Norris, Vera Price, Julian Mackay-Wiggan, Raphael Clynes, Angela M. Christiano
View: Text | PDF

High-throughput T cell receptor sequencing identifies clonally expanded CD8+ T cell populations in alopecia areata

  • Text
  • PDF
Abstract

Alopecia areata (AA) is an autoimmune disease in which cytotoxic T cells specifically target growing hair follicles. We used high-throughput TCR sequencing in the C3H/HeJ mouse model of AA and in human AA patients to gain insight into pathogenic T cell populations and their dynamics, which revealed clonal CD8+ T cell expansions in lesional skin. In the C3H/HeJ model, we observed interindividual sharing of TCRβ chain protein sequences, which strongly supports a model of antigenic drive in AA. The overlap between the lesional TCR repertoire and a population of CD8+NKG2D+ T cells in skin-draining lymph nodes identified this subset as pathogenic effectors. In AA patients, treatment with the oral JAK inhibitor tofacitinib resulted in a decrease in clonally expanded CD8+ T cells in the scalp but also revealed that many expanded lesional T cell clones do not completely disappear from either skin or blood during treatment with tofacitinib, which may explain in part the relapse of disease after stopping treatment.

Authors

Annemieke de Jong, Ali Jabbari, Zhenpeng Dai, Luzhou Xing, Dustin Lee, Mei Mei Li, Madeleine Duvic, Maria Hordinsky, David A. Norris, Vera Price, Julian Mackay-Wiggan, Raphael Clynes, Angela M. Christiano

×

IFI16 filament formation in salivary epithelial cells shapes the anti-IFI16 immune response in Sjögren’s syndrome
Brendan Antiochos, Mariusz Matyszewski, Jungsan Sohn, Livia Casciola-Rosen, Antony Rosen
Brendan Antiochos, Mariusz Matyszewski, Jungsan Sohn, Livia Casciola-Rosen, Antony Rosen
View: Text | PDF

IFI16 filament formation in salivary epithelial cells shapes the anti-IFI16 immune response in Sjögren’s syndrome

  • Text
  • PDF
Abstract

IFN-inducible protein 16 (IFI16) is an innate immune sensor that forms filamentous oligomers when activated by double-stranded DNA (dsDNA). Anti-IFI16 autoantibodies occur in patients with Sjögren’s syndrome (SS) and associate with severe phenotypic features. We undertook this study to determine whether the structural and functional properties of IFI16 play a role in its status as an SS autoantigen. IFI16 immunostaining in labial salivary glands (LSGs) yielded striking evidence of filamentous IFI16 structures in the cytoplasm of ductal epithelial cells, representing the first microscopic description of IFI16 oligomerization in human tissues, to our knowledge. Transfection of cultured epithelial cells with dsDNA triggered the formation of cytoplasmic IFI16 filaments with similar morphology to those observed in LSGs. We found that a majority of SS anti-IFI16 autoantibodies immunoprecipitate IFI16 more effectively in the oligomeric dsDNA-bound state. Epitopes in the C-terminus of IFI16 are accessible to antibodies in the DNA-bound oligomer and are preferentially targeted by SS sera. Furthermore, cytotoxic lymphocyte granule pathways (highly enriched in the SS gland) induce striking release of IFI16•dsDNA complexes from cultured cells. Our studies reveal that IFI16 is present in a filamentous state in the target tissue of SS and suggest that this property of DNA-induced filament formation contributes to its status as an autoantigen in SS. These studies highlight the role that tissue-specific modifications and immune effector pathways might play in the selection of autoantigens in rheumatic diseases.

Authors

Brendan Antiochos, Mariusz Matyszewski, Jungsan Sohn, Livia Casciola-Rosen, Antony Rosen

×

Abnormal neutrophil signature in the blood and pancreas of presymptomatic and symptomatic type 1 diabetes
Federica Vecchio, Nicola Lo Buono, Angela Stabilini, Laura Nigi, Matthew J. Dufort, Susan Geyer, Paola Maria Rancoita, Federica Cugnata, Alessandra Mandelli, Andrea Valle, Pia Leete, Francesca Mancarella, Peter S. Linsley, Lars Krogvold, Kevan C. Herold, Helena Elding Larsson, Sarah J. Richardson, Noel G. Morgan, Knut Dahl-Jørgensen, Guido Sebastiani, Francesco Dotta, Emanuele Bosi, the DRI_Biorepository Group, the Type 1 Diabetes TrialNet Study Group, Manuela Battaglia
Federica Vecchio, Nicola Lo Buono, Angela Stabilini, Laura Nigi, Matthew J. Dufort, Susan Geyer, Paola Maria Rancoita, Federica Cugnata, Alessandra Mandelli, Andrea Valle, Pia Leete, Francesca Mancarella, Peter S. Linsley, Lars Krogvold, Kevan C. Herold, Helena Elding Larsson, Sarah J. Richardson, Noel G. Morgan, Knut Dahl-Jørgensen, Guido Sebastiani, Francesco Dotta, Emanuele Bosi, the DRI_Biorepository Group, the Type 1 Diabetes TrialNet Study Group, Manuela Battaglia
View: Text | PDF

Abnormal neutrophil signature in the blood and pancreas of presymptomatic and symptomatic type 1 diabetes

  • Text
  • PDF
Abstract

BACKGROUND. Neutrophils and their inflammatory mediators are key pathogenic components in multiple autoimmune diseases, while their role in human type 1 diabetes (T1D), a disease that progresses sequentially through identifiable stages prior to the clinical onset, is not well understood. We previously reported that the number of circulating neutrophils is reduced in patients with T1D and in presymptomatic at-risk subjects. The aim of the present work was to identify possible changes in circulating and pancreas-residing neutrophils throughout the disease course to better elucidate neutrophil involvement in human T1D. METHODS. Data collected from 389 subjects at risk of developing T1D, and enrolled in 4 distinct studies performed by TrialNet, were analyzed with comprehensive statistical approaches to determine whether the number of circulating neutrophils correlates with pancreas function. To obtain a broad analysis of pancreas-infiltrating neutrophils throughout all disease stages, pancreas sections collected worldwide from 4 different cohorts (i.e., nPOD, DiViD, Siena, and Exeter) were analyzed by immunohistochemistry and immunofluorescence. Finally, circulating neutrophils were purified from unrelated nondiabetic subjects and donors at various T1D stages and their transcriptomic signature was determined by RNA sequencing. RESULTS. Here, we show that the decline in β cell function is greatest in individuals with the lowest peripheral neutrophil numbers. Neutrophils infiltrate the pancreas prior to the onset of symptoms and they continue to do so as the disease progresses. Of interest, a fraction of these pancreas-infiltrating neutrophils also extrudes neutrophil extracellular traps (NETs), suggesting a tissue-specific pathogenic role. Whole-transcriptome analysis of purified blood neutrophils revealed a unique molecular signature that is distinguished by an overabundance of IFN-associated genes; despite being healthy, said signature is already present in T1D-autoantibody-negative at-risk subjects. CONCLUSIONS. These results reveal an unexpected abnormality in neutrophil disposition both in the circulation and in the pancreas of presymptomatic and symptomatic T1D subjects, implying that targeting neutrophils might represent a previously unrecognized therapeutic modality. FUNDING. Juvenile Diabetes Research Foundation (JDRF), NIH, Diabetes UK.

Authors

Federica Vecchio, Nicola Lo Buono, Angela Stabilini, Laura Nigi, Matthew J. Dufort, Susan Geyer, Paola Maria Rancoita, Federica Cugnata, Alessandra Mandelli, Andrea Valle, Pia Leete, Francesca Mancarella, Peter S. Linsley, Lars Krogvold, Kevan C. Herold, Helena Elding Larsson, Sarah J. Richardson, Noel G. Morgan, Knut Dahl-Jørgensen, Guido Sebastiani, Francesco Dotta, Emanuele Bosi, the DRI_Biorepository Group, the Type 1 Diabetes TrialNet Study Group, Manuela Battaglia

×

Belimumab promotes negative selection of activated autoreactive B cells in systemic lupus erythematosus patients
Weiqing Huang, Tam D. Quach, Cosmin Dascalu, Zheng Liu, Tungming Leung, Miranda Byrne-Steele, Wenjing Pan, Qunying Yang, Jian Han, Martin Lesser, Thomas L. Rothstein, Richard Furie, Meggan Mackay, Cynthia Aranow, Anne Davidson
Weiqing Huang, Tam D. Quach, Cosmin Dascalu, Zheng Liu, Tungming Leung, Miranda Byrne-Steele, Wenjing Pan, Qunying Yang, Jian Han, Martin Lesser, Thomas L. Rothstein, Richard Furie, Meggan Mackay, Cynthia Aranow, Anne Davidson
View: Text | PDF

Belimumab promotes negative selection of activated autoreactive B cells in systemic lupus erythematosus patients

  • Text
  • PDF
Abstract

Belimumab has therapeutic benefit in active systemic lupus erythematosus (SLE), especially in patients with high-titer anti-dsDNA antibodies. We asked whether the profound B cell loss in belimumab-treated SLE patients is accompanied by shifts in the immunoglobulin repertoire. We enrolled 15 patients who had been continuously treated with belimumab for more than 7 years, 17 matched controls, and 5 patients who were studied before and after drug initiation. VH genes of sort-purified mature B cells and plasmablasts were subjected to next-generation sequencing. We found that B cell–activating factor (BAFF) regulates the transitional B cell checkpoint, with conservation of transitional 1 (T1) cells and approximately 90% loss of T3 and naive B cells after chronic belimumab treatment. Class-switched memory B cells, B1 B cells, and plasmablasts were also substantially depleted. Next-generation sequencing revealed no redistribution of VH, DH, or JH family usage and no effect of belimumab on representation of the autoreactive VH4-34 gene or CDR3 composition in unmutated IgM sequences, suggesting a minimal effect on selection of the naive B cell repertoire. Interestingly, a significantly greater loss of VH4-34 was observed among mutated IgM and plasmablast sequences in chronic belimumab–treated subjects than in controls, suggesting that belimumab promotes negative selection of activated autoreactive B cells.

Authors

Weiqing Huang, Tam D. Quach, Cosmin Dascalu, Zheng Liu, Tungming Leung, Miranda Byrne-Steele, Wenjing Pan, Qunying Yang, Jian Han, Martin Lesser, Thomas L. Rothstein, Richard Furie, Meggan Mackay, Cynthia Aranow, Anne Davidson

×

Precision DNA demethylation ameliorates disease in lupus-prone mice
Hao Li, Maria G. Tsokos, Sean Bickerton, Amir Sharabi, Yi Li, Vaishali R. Moulton, Philip Kong, Tarek M. Fahmy, George C. Tsokos
Hao Li, Maria G. Tsokos, Sean Bickerton, Amir Sharabi, Yi Li, Vaishali R. Moulton, Philip Kong, Tarek M. Fahmy, George C. Tsokos
View: Text | PDF

Precision DNA demethylation ameliorates disease in lupus-prone mice

  • Text
  • PDF
Abstract

Defective DNA methylation in T cells leads to a series of T cell abnormalities in lupus; however, the full effect of T cell lineage–specific DNA methylation on disease expression has not been explored. Here, we show that 5-azacytidine, a DNA methyltransferase inhibitor, targeted to either CD4 or CD8 T cells in mice with established disease using a nanolipogel delivery system dramatically ameliorates lupus-related pathology through distinct mechanisms. In vivo targeted delivery of 5-azacytidine into CD4 T cells favors the expansion and function of Foxp3+ Tregs, whereas targeted delivery to CD8 T cells enhances the cytotoxicity and restrains the expansion of pathogenic TCR-αβ+CD4–CD8– double-negative T cells. Our results signify the importance of cell-specific inhibition of DNA methylation in the treatment of established lupus.

Authors

Hao Li, Maria G. Tsokos, Sean Bickerton, Amir Sharabi, Yi Li, Vaishali R. Moulton, Philip Kong, Tarek M. Fahmy, George C. Tsokos

×

IL-16/miR-125a axis controls neutrophil recruitment in pristane-induced lung inflammation
Siobhan Smith, Pei Wen Wu, Jane J. Seo, Thilini Fernando, Mengyao Jin, Jorge Contreras, Erica N. Montano, Joan Ní Gabhann, Kyle Cunningham, Amro Widaa, Eoghan M. McCarthy, Eamonn S. Molloy, Grainne Kearns, Conor C. Murphy, Weiping Kong, Harry Björkbacka, Hardy Kornfeld, Lindsy Forbess, Swamy Venuturupalli, Mariko Ishimori, Daniel Wallace, Michael H. Weisman, Caroline A. Jefferies
Siobhan Smith, Pei Wen Wu, Jane J. Seo, Thilini Fernando, Mengyao Jin, Jorge Contreras, Erica N. Montano, Joan Ní Gabhann, Kyle Cunningham, Amro Widaa, Eoghan M. McCarthy, Eamonn S. Molloy, Grainne Kearns, Conor C. Murphy, Weiping Kong, Harry Björkbacka, Hardy Kornfeld, Lindsy Forbess, Swamy Venuturupalli, Mariko Ishimori, Daniel Wallace, Michael H. Weisman, Caroline A. Jefferies
View: Text | PDF

IL-16/miR-125a axis controls neutrophil recruitment in pristane-induced lung inflammation

  • Text
  • PDF
Abstract

Severe lung inflammation and alveolar hemorrhage can be life-threatening in systemic lupus erythematosus (SLE) patients if not treated early and aggressively. Neutrophil influx is the driver key of this pathology, but little is known regarding the molecular events regulating this recruitment. Here, we uncover a role for IL-16/mir-125a in this pathology and show not only that IL-16 is a target for miR-125a but that reduced miR-125a expression in SLE patients associates with lung involvement. Furthermore, in the pristane model of acute “SLE-like” lung inflammation and alveolar hemorrhage, we observed reduced pulmonary miR-125a and enhanced IL-16 expression. Neutrophil infiltration was markedly reduced in the peritoneal lavage of pristane-treated IL-16–deficient mice and elevated following i.n. delivery of IL-16. Moreover, a miR-125a mimic reduced pristane-induced IL-16 expression and neutrophil recruitment and rescued lung pathology. Mechanistically, IL-16 acts directly on the pulmonary epithelium and markedly enhances neutrophil chemoattractant expression both in vitro and in vivo, while the miR-125a mimic can prevent this. Our results reveal a role for miR-125a/IL-16 in regulating lung inflammation and suggest this axis may be a therapeutic target for management of acute lung injury in SLE.

Authors

Siobhan Smith, Pei Wen Wu, Jane J. Seo, Thilini Fernando, Mengyao Jin, Jorge Contreras, Erica N. Montano, Joan Ní Gabhann, Kyle Cunningham, Amro Widaa, Eoghan M. McCarthy, Eamonn S. Molloy, Grainne Kearns, Conor C. Murphy, Weiping Kong, Harry Björkbacka, Hardy Kornfeld, Lindsy Forbess, Swamy Venuturupalli, Mariko Ishimori, Daniel Wallace, Michael H. Weisman, Caroline A. Jefferies

×

Defective BTLA functionality is rescued by restoring lipid metabolism in lupus CD4+ T cells
Matthieu Sawaf, Jean-Daniel Fauny, Renaud Felten, Flora Sagez, Jacques-Eric Gottenberg, Hélène Dumortier, Fanny Monneaux
Matthieu Sawaf, Jean-Daniel Fauny, Renaud Felten, Flora Sagez, Jacques-Eric Gottenberg, Hélène Dumortier, Fanny Monneaux
View: Text | PDF

Defective BTLA functionality is rescued by restoring lipid metabolism in lupus CD4+ T cells

  • Text
  • PDF
Abstract

Coinhibitory receptors play an important role in the prevention of autoimmune diseases, such as systemic lupus erythematosus (SLE), by limiting T cell activation. B and T lymphocyte attenuator (BTLA) is an inhibitory receptor, similar to cytotoxic T lymphocyte–associated protein 4 (CTLA-4) and programmed death 1 (PD1), that negatively regulates the immune response. The role of BTLA in the pathogenesis of autoimmune diseases in humans and, more specifically, in SLE is largely unknown. We investigated BTLA expression on various T cell subsets, and we did not observe significant variations of BTLA expression between lupus patients and healthy controls. However, the enhancement of BTLA expression after activation was significantly lower in SLE patients compared with that in healthy controls. Furthermore, we found an impaired capacity of BTLA to inhibit T cell activation in SLE due to a poor BTLA recruitment to the immunological synapse following T cell stimulation. Finally, we demonstrated that defective BTLA function can be corrected by restoring intracellular trafficking and by normalizing the lipid metabolism in lupus CD4+ T cells. Collectively, our results evidence that the BTLA signaling pathway is altered in SLE T cells and highlight the potential of targeting this pathway for the development of new therapeutic strategies in lupus.

Authors

Matthieu Sawaf, Jean-Daniel Fauny, Renaud Felten, Flora Sagez, Jacques-Eric Gottenberg, Hélène Dumortier, Fanny Monneaux

×
  • ← Previous
  • 1
  • 2
  • …
  • 24
  • 25
  • 26
  • …
  • 30
  • 31
  • Next →

No posts were found with this tag.

Advertisement

Copyright © 2026 American Society for Clinical Investigation
ISSN 2379-3708

Sign up for email alerts