51
|
Chalmers SA, Ayilam Ramachandran R, Garcia SJ, Der E, Herlitz L, Ampudia J, Chu D, Jordan N, Zhang T, Parodis I, Gunnarsson I, Ding H, Shen N, Petri M, Mok CC, Saxena R, Polu KR, Connelly S, Ng CT, Mohan C, Putterman C. The CD6/ALCAM pathway promotes lupus nephritis via T cell-mediated responses. J Clin Invest 2022; 132:e147334. [PMID: 34981775 PMCID: PMC8718154 DOI: 10.1172/jci147334] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 11/10/2021] [Indexed: 12/18/2022] Open
Abstract
T cells are central to the pathogenesis of lupus nephritis (LN), a common complication of systemic lupus erythematosus (SLE). CD6 and its ligand, activated leukocyte cell adhesion molecule (ALCAM), are involved in T cell activation and trafficking. Previously, we showed that soluble ALCAM is increased in urine (uALCAM) of patients with LN, suggesting that this pathway contributes to disease. To investigate, uALCAM was examined in 1038 patients with SLE and LN from 5 ethnically diverse cohorts; CD6 and ALCAM expression was assessed in LN kidney cells; and disease contribution was tested via antibody blockade of CD6 in murine models of SLE and acute glomerulonephritis. Extended cohort analysis offered resounding validation of uALCAM as a biomarker that distinguishes active renal involvement in SLE, irrespective of ethnicity. ALCAM was expressed by renal structural cells whereas CD6 expression was exclusive to T cells, with elevated numbers of CD6+ and ALCAM+ cells in patients with LN. CD6 blockade in models of spontaneous lupus and immune-complex glomerulonephritis revealed significant decreases in immune cells, inflammatory markers, and disease measures. Our data demonstrate the contribution of the CD6/ALCAM pathway to LN and SLE, supporting its use as a disease biomarker and therapeutic target.
Collapse
Affiliation(s)
- Samantha A. Chalmers
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | - Sayra J. Garcia
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Evan Der
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Leal Herlitz
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Nicole Jordan
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ting Zhang
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institute and Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Iva Gunnarsson
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institute and Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Huihua Ding
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Shen
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Ramesh Saxena
- Division of Nephrology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | | | | | | | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Chaim Putterman
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
| |
Collapse
|
52
|
Axl regulated survival/proliferation network and its therapeutic intervention in mouse models of glomerulonephritis. Arthritis Res Ther 2022; 24:284. [PMID: 36578056 PMCID: PMC9795606 DOI: 10.1186/s13075-022-02965-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/02/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Lupus nephritis (LN) is the most common and serious complication of systemic lupus erythematosus (SLE). LN pathogenesis is not fully understood. Axl receptor tyrosine kinase is upregulated and contributes to the pathogenic progress in LN. We have reported that Axl disruption attenuates nephritis development in mice. METHODS In this study, we analyzed the gene expression profiles with RNA-seq using renal cortical samples from nephritic mice. Axl-KO mice were bred onto a B6.lpr spontaneous lupus background, and renal disease development was followed and compared to the Axl-sufficient B6.lpr mice. Finally, anti-glomerular basement membrane (anti-GBM) Ab-induced nephritic mice were treated with Axl small molecule inhibitor, R428, at different stages of nephritis development. Blood urine nitrogen levels and renal pathologies were evaluated. RESULTS Transcriptome analysis revealed that renal Axl activation contributed to cell proliferation, survival, and motility through regulation of the Akt, c-Jun, and actin pathways. Spontaneous lupus-prone B6.lpr mice with Axl deficiency showed significantly reduced kidney damages and decreased T cell infiltration compared to the renal damage and T cell infiltration in Axl-sufficient B6.lpr mice. The improved kidney function was independent of autoAb production. Moreover, R428 significantly reduced anti-GBM glomerulonephritis at different stages of GN development compared to the untreated nephritic control mice. R428 administration reduced inflammatory cytokine (IL-6) production, T cell infiltration, and nephritis disease activity. CONCLUSIONS Results from this study emphasize the important role of Axl signaling in LN and highlight Axl as an attractive target in LN.
Collapse
|
53
|
Van Laecke S, Van Damme K, Dendooven A. Immunosenescence: an unexplored role in glomerulonephritis. Clin Transl Immunology 2022; 11:e1427. [PMID: 36420421 PMCID: PMC9676375 DOI: 10.1002/cti2.1427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/15/2022] [Accepted: 10/16/2022] [Indexed: 11/22/2022] Open
Abstract
Immunosenescence is a natural ageing phenomenon with alterations in innate and especially adaptive immunity and contributes to reduced antimicrobial defence and chronic low‐grade inflammation. This is mostly reflected by an increase in organ‐directed and/or circulating reactive and cytolytic terminally differentiated T cells that have lost their expression of the costimulatory receptor CD28. Apart from being induced by a genetic predisposition, ageing or viral infections (particularly cytomegalovirus infection), immunosenescence is accelerated in many inflammatory diseases and uraemia. This translates into an enhancement of vascular inflammation and cardiovascular disease varying from endothelial dysfunction to plaque rupture. Emerging data point to a mechanistic role of CD28null T cells in glomerulonephritis, where they initiate and propagate local inflammation in concordance with dendritic cells and macrophages. They are suitably equipped to escape immunological dampening by the absence of homing to lymph nodes, anti‐apoptotic properties and resistance to suppression by regulatory T cells. Early accumulation of senescent CD28null T cells precedes glomerular or vascular injury, and targeting these cells could open avenues for early treatment interventions that aim at abrogating a detrimental vicious cycle.
Collapse
Affiliation(s)
| | - Karel Van Damme
- Renal Division Ghent University Hospital Ghent Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, Center for Inflammation Research VIB Center for Inflammation Research Ghent Belgium
| | | |
Collapse
|
54
|
Asada N, Ginsberg P, Gagliani N, Mittrücker HW, Panzer U. Tissue-resident memory T cells in the kidney. Semin Immunopathol 2022; 44:801-811. [PMID: 35411437 PMCID: PMC9708805 DOI: 10.1007/s00281-022-00927-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/28/2022] [Indexed: 12/15/2022]
Abstract
The identification of tissue-resident memory T cells (TRM cells) has significantly improved our understanding of immunity. In the last decade, studies have demonstrated that TRM cells are induced after an acute T-cell response, remain in peripheral organs for several years, and contribute to both an efficient host defense and autoimmune disease. TRM cells are found in the kidneys of healthy individuals and patients with various kidney diseases. A better understanding of these cells and their therapeutic targeting might provide new treatment options for infections, autoimmune diseases, graft rejection, and cancer. In this review, we address the definition, phenotype, and developmental mechanisms of TRM cells. Then, we further discuss the current understanding of TRM cells in kidney diseases, such as infection, autoimmune disease, cancer, and graft rejection after transplantation.
Collapse
Affiliation(s)
- Nariaki Asada
- grid.13648.380000 0001 2180 3484III. Department of Medicine, Division of Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pauline Ginsberg
- grid.13648.380000 0001 2180 3484III. Department of Medicine, Division of Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Gagliani
- grid.13648.380000 0001 2180 3484Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany ,grid.13648.380000 0001 2180 3484Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany ,grid.13648.380000 0001 2180 3484I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany ,grid.24381.3c0000 0000 9241 5705Immunology and Allergy Unit, Department of Medicine, SolnaKarolinska Institute and University Hospital, Stockholm, Sweden
| | - Hans-Willi Mittrücker
- grid.13648.380000 0001 2180 3484Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany ,grid.13648.380000 0001 2180 3484Institute for Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulf Panzer
- grid.13648.380000 0001 2180 3484III. Department of Medicine, Division of Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany ,grid.13648.380000 0001 2180 3484Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
55
|
Ye C, Yano H, Workman CJ, Vignali DAA. Interleukin-35: Structure, Function and Its Impact on Immune-Related Diseases. J Interferon Cytokine Res 2021; 41:391-406. [PMID: 34788131 DOI: 10.1089/jir.2021.0147] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The balance between inflammatory and anti-inflammatory immune responses is maintained through immunoregulatory cell populations and immunosuppressive cytokines. Interleukin-35 (IL-35), an inhibitory cytokine that belongs to the IL-12 family, is capable of potently suppressing T cell proliferation and inducing IL-35-producing induced regulatory T cells (iTr35) to limit inflammatory responses. Over the past decade, a growing number of studies have indicated that IL-35 plays an important role in controlling immune-related disorders, including autoimmune diseases, infectious diseases, and cancer. In this review, we summarize the current knowledge about the biology of IL-35 and its contribution in different diseases, and we discuss the potential of and barriers to harnessing IL-35 as a clinical biomarker or immunotherapy.
Collapse
Affiliation(s)
- Cheng Ye
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hiroshi Yano
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
56
|
Abstract
In this essay, we show that 3 distinct approaches to immunological exhaustion coexist and that they only partially overlap, generating potential misunderstandings. Exploring cases ranging from viral infections to cancer, we propose that it is crucial, for experimental and therapeutic purposes, to clarify these approaches and their interconnections so as to make the concept of exhaustion genuinely operational.
Collapse
Affiliation(s)
- Hannah Kaminski
- ImmunoConcept, CNRS & University of Bordeaux, Bordeaux, France
| | - Maël Lemoine
- ImmunoConcept, CNRS & University of Bordeaux, Bordeaux, France
| | - Thomas Pradeu
- ImmunoConcept, CNRS & University of Bordeaux, Bordeaux, France
| |
Collapse
|
57
|
Kingsmore KM, Bachali P, Catalina MD, Daamen AR, Heuer SE, Robl RD, Grammer AC, Lipsky PE. Altered expression of genes controlling metabolism characterizes the tissue response to immune injury in lupus. Sci Rep 2021; 11:14789. [PMID: 34285256 PMCID: PMC8292402 DOI: 10.1038/s41598-021-93034-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
To compare lupus pathogenesis in disparate tissues, we analyzed gene expression profiles of human discoid lupus erythematosus (DLE) and lupus nephritis (LN). We found common increases in myeloid cell-defining gene sets and decreases in genes controlling glucose and lipid metabolism in lupus-affected skin and kidney. Regression models in DLE indicated increased glycolysis was correlated with keratinocyte, endothelial, and inflammatory cell transcripts, and decreased tricarboxylic (TCA) cycle genes were correlated with the keratinocyte signature. In LN, regression models demonstrated decreased glycolysis and TCA cycle genes were correlated with increased endothelial or decreased kidney cell transcripts, respectively. Less severe glomerular LN exhibited similar alterations in metabolism and tissue cell transcripts before monocyte/myeloid cell infiltration in some patients. Additionally, changes to mitochondrial and peroxisomal transcripts were associated with specific cells rather than global signal changes. Examination of murine LN gene expression demonstrated metabolic changes were not driven by acute exposure to type I interferon and could be restored after immunosuppression. Finally, expression of HAVCR1, a tubule damage marker, was negatively correlated with the TCA cycle signature in LN models. These results indicate that altered metabolic dysfunction is a common, reversible change in lupus-affected tissues and appears to reflect damage downstream of immunologic processes.
Collapse
Affiliation(s)
- Kathryn M Kingsmore
- AMPEL BioSolutions, LLC and RILITE Research Institute, Charlottesville, VA, USA.
| | - Prathyusha Bachali
- AMPEL BioSolutions, LLC and RILITE Research Institute, Charlottesville, VA, USA
| | - Michelle D Catalina
- AMPEL BioSolutions, LLC and RILITE Research Institute, Charlottesville, VA, USA
- EMD Serono Research & Development Institute, 45 A Middlesex Turnpike, Billerica, MA, 01821, USA
| | - Andrea R Daamen
- AMPEL BioSolutions, LLC and RILITE Research Institute, Charlottesville, VA, USA
| | - Sarah E Heuer
- AMPEL BioSolutions, LLC and RILITE Research Institute, Charlottesville, VA, USA
- The Jackson Laboratory, Tufts Graduate School of Biomedical Sciences, 600 Main Street Bar, Harbor, ME, 04609, USA
| | - Robert D Robl
- AMPEL BioSolutions, LLC and RILITE Research Institute, Charlottesville, VA, USA
| | - Amrie C Grammer
- AMPEL BioSolutions, LLC and RILITE Research Institute, Charlottesville, VA, USA
| | - Peter E Lipsky
- AMPEL BioSolutions, LLC and RILITE Research Institute, Charlottesville, VA, USA
| |
Collapse
|
58
|
Baek WY, Lee SM, Lee SW, Son IO, Choi S, Suh CH. Intravenous Administration of Toll-Like Receptor Inhibitory Peptide 1 is Effective for the Treatment of Systemic Lupus Erythematosus in a Mus musculus Model. JOURNAL OF RHEUMATIC DISEASES 2021; 28:133-142. [PMID: 37475994 PMCID: PMC10324895 DOI: 10.4078/jrd.2021.28.3.133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 07/22/2023]
Abstract
Objective Systemic lupus erythematosus (SLE) is a common chronic autoimmune inflammatory disease According to recent studies, signaling through Toll-like receptor (TLR) protein, which promotes the production of inflammatory cytokines, leads to the development of SLE TLR-inhibitory peptide 1 (TIP1) has been newly identified for the treatment of autoimmune diseases. Methods The effect of TIP1 was analyzed in an SLE mouse model (MRL/lpr) The mice in the control treatment group (n=5) were administered an intravenous injection of phosphate-buffered saline twice weekly, whereas the mice in the TIP1 treatment group (n=6) were administered an intravenous injection of TIP1 (1 nmol/g) twice weekly MRL/mpj mice (n=5) were selected as normal controls The mice were injected for 4 weeks between 14 and 18 weeks of age, followed by assays of their spleen, kidneys, lymph nodes, serum, and urine. Results The antinuclear antibody and inflammatory cytokine (interferon-α) in the serum as well as levels of albumin in the urine of the mice in the TIP1 treatment group had decreased when compared to those of mice in the control treatment group Kidney inflammation in mice in the TIP1 treatment group was alleviated The mRNA expression levels of TLR7- or TLR9-related downstream signaling molecules also decreased in all organs of the mice in the TIP1 treatment group. Conclusion Intravenous treatment with TIP1 reduces symptoms and markers of inflammation in MRL/lpr mice Hence, TIP1 is a promising medication for the treatment of SLE.
Collapse
Affiliation(s)
- Wook-Young Baek
- Department of Rheumatology, Ajou University School of Medicine, Suwon, Korea
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Sung-Min Lee
- Department of Rheumatology, Ajou University School of Medicine, Suwon, Korea
| | - Sang-Won Lee
- Department of Rheumatology, Ajou University School of Medicine, Suwon, Korea
| | - In-Ok Son
- Department of Rheumatology, Ajou University School of Medicine, Suwon, Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Chang-Hee Suh
- Department of Rheumatology, Ajou University School of Medicine, Suwon, Korea
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| |
Collapse
|
59
|
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is a serious autoimmune disease with a wide range of organ involvement. In addition to aberrant B-cell responses leading to autoantibody production, T-cell abnormalities are important in the induction of autoimmunity and the ensuing downstream organ damage. In this article, we present an update on how subsets of CD8+ T cells contribute to SLE pathogenesis. RECENT FINDINGS Reduced cytolytic function of CD8+ T cells not only promotes systemic autoimmunity but also accounts for the increased risk of infections. Additional information suggests that effector functions of tissue CD8+ T cells contribute to organ damage. The phenotypic changes in tissue CD8+ T cells likely arise from exposure to tissue microenvironment and crosstalk with tissue resident cells. Research on pathogenic IL-17-producing double negative T cells also suggests their origin from autoreactive CD8+ T cells, which also contribute to the induction and maintenance of systemic autoimmunity. SUMMARY Reduced CD8+ T-cell effector function illustrates their role in peripheral tolerance in the control of autoimmunity and to the increased risk of infections. Inflammatory cytokine producing double negative T cells and functional defects of regulatory CD8+ T cell both contribute to SLE pathogenesis. Further in depth research on these phenotypic changes are warranted for the development of new therapeutics for people with SLE.
Collapse
|
60
|
Chen PM, Wilson PC, Shyer JA, Veselits M, Steach HR, Cui C, Moeckel G, Clark MR, Craft J. Kidney tissue hypoxia dictates T cell-mediated injury in murine lupus nephritis. Sci Transl Med 2021; 12:12/538/eaay1620. [PMID: 32269165 DOI: 10.1126/scitranslmed.aay1620] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 11/06/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022]
Abstract
The kidney is a frequent target of autoimmune injury, including in systemic lupus erythematosus; however, how immune cells adapt to kidney's unique environment and contribute to tissue damage is unknown. We found that renal tissue, which normally has low oxygen tension, becomes more hypoxic in lupus nephritis. In the injured mouse tissue, renal-infiltrating CD4+ and CD8+ T cells express hypoxia-inducible factor-1 (HIF-1), which alters their cellular metabolism and prevents their apoptosis in hypoxia. HIF-1-dependent gene-regulated pathways were also up-regulated in renal-infiltrating T cells in human lupus nephritis. Perturbation of these environmental adaptations by selective HIF-1 blockade inhibited infiltrating T cells and reversed tissue hypoxia and injury in murine models of lupus. The results suggest that targeting HIF-1 might be effective for treating renal injury in autoimmune diseases.
Collapse
Affiliation(s)
- Ping-Min Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Parker C Wilson
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Justin A Shyer
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Margaret Veselits
- Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, Departments of Medicine and Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Holly R Steach
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Can Cui
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gilbert Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Marcus R Clark
- Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, Departments of Medicine and Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Joe Craft
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA. .,Department of Internal Medicine (Rheumatology, Allergy and Immunology), Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
61
|
Paredes JL, Fernandez-Ruiz R, Niewold TB. T Cells in Systemic Lupus Erythematosus. Rheum Dis Clin North Am 2021; 47:379-393. [PMID: 34215369 DOI: 10.1016/j.rdc.2021.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
T-cell dysregulation has been implicated in the loss of tolerance and overactivation of B cells in systemic lupus erythematosus (SLE). Recent studies have identified T-cell subsets and genetic, epigenetic, and environmental factors that contribute to pathogenic T-cell differentiation, as well as disease pathogenesis and clinical phenotypes in SLE. Many therapeutics targeting T-cell pathways are under development, and although many have not progressed in clinical trials, the recent approval of the calcineurin inhibitor voclosporin is encouraging. Further study of T-cell subsets and biomarkers of T-cell action may pave the way for specific targeting of pathogenic T-cell populations in SLE.
Collapse
Affiliation(s)
- Jacqueline L Paredes
- Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Avenue, New York, NY 10016, USA
| | - Ruth Fernandez-Ruiz
- Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Avenue, New York, NY 10016, USA; Division of Rheumatology, NYU Grossman School of Medicine, 550 1st Avenue, New York, NY 10016, USA
| | - Timothy B Niewold
- Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Avenue, New York, NY 10016, USA.
| |
Collapse
|
62
|
Martins CP, New LA, O’Connor EC, Previte DM, Cargill KR, Tse IL, Sims- Lucas S, Piganelli JD. Glycolysis Inhibition Induces Functional and Metabolic Exhaustion of CD4 + T Cells in Type 1 Diabetes. Front Immunol 2021; 12:669456. [PMID: 34163475 PMCID: PMC8216385 DOI: 10.3389/fimmu.2021.669456] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/17/2021] [Indexed: 01/10/2023] Open
Abstract
In Type 1 Diabetes (T1D), CD4+ T cells initiate autoimmune attack of pancreatic islet β cells. Importantly, bioenergetic programs dictate T cell function, with specific pathways required for progression through the T cell lifecycle. During activation, CD4+ T cells undergo metabolic reprogramming to the less efficient aerobic glycolysis, similarly to highly proliferative cancer cells. In an effort to limit tumor growth in cancer, use of glycolytic inhibitors have been successfully employed in preclinical and clinical studies. This strategy has also been utilized to suppress T cell responses in autoimmune diseases like Systemic Lupus Erythematosus (SLE), Multiple Sclerosis (MS), and Rheumatoid Arthritis (RA). However, modulating T cell metabolism in the context of T1D has remained an understudied therapeutic opportunity. In this study, we utilized the small molecule PFK15, a competitive inhibitor of the rate limiting glycolysis enzyme 6-phosphofructo-2-kinase/fructose-2,6- biphosphatase 3 (PFKFB3). Our results confirmed PFK15 inhibited glycolysis utilization by diabetogenic CD4+ T cells and reduced T cell responses to β cell antigen in vitro. In an adoptive transfer model of T1D, PFK15 treatment delayed diabetes onset, with 57% of animals remaining euglycemic at the end of the study period. Protection was due to induction of a hyporesponsive T cell phenotype, characterized by increased and sustained expression of the checkpoint molecules PD-1 and LAG-3 and downstream functional and metabolic exhaustion. Glycolysis inhibition terminally exhausted diabetogenic CD4+ T cells, which was irreversible through restimulation or checkpoint blockade in vitro and in vivo. In sum, our results demonstrate a novel therapeutic strategy to control aberrant T cell responses by exploiting the metabolic reprogramming of these cells during T1D. Moreover, the data presented here highlight a key role for nutrient availability in fueling T cell function and has implications in our understanding of T cell biology in chronic infection, cancer, and autoimmunity.
Collapse
Affiliation(s)
- Christina P. Martins
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pediatric Surgery, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lee A. New
- Department of Pediatric Surgery, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Erin C. O’Connor
- Department of Pediatric Surgery, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Dana M. Previte
- Department of Pediatric Surgery, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kasey R. Cargill
- Department of Pediatrics, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Isabelle L. Tse
- Department of Pediatric Surgery, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sunder Sims- Lucas
- Department of Pediatrics, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jon D. Piganelli
- Department of Pediatric Surgery, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
63
|
Marinov AD, Wang H, Bastacky SI, van Puijenbroek E, Schindler T, Speziale D, Perro M, Klein C, Nickerson KM, Shlomchik MJ. The Type II Anti-CD20 Antibody Obinutuzumab (GA101) Is More Effective Than Rituximab at Depleting B Cells and Treating Disease in a Murine Lupus Model. Arthritis Rheumatol 2021; 73:826-836. [PMID: 33277983 DOI: 10.1002/art.41608] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 12/01/2020] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Depleting pathogenic B cells could treat systemic lupus erythematosus (SLE). However, depleting B cells in an inflammatory setting such as lupus is difficult. This study was undertaken to investigate whether a type II anti-CD20 monoclonal antibody (mAb) with a different mechanism of action, obinutuzumab (GA101), is more effective than a type I anti-CD20 mAb, rituximab (RTX), in B cell depletion in lupus, and whether efficient B cell depletion results in amelioration of disease. METHODS We treated lupus-prone MRL/lpr mice expressing human CD20 on B cells (hCD20 MRL/lpr mice) with either RTX or GA101 and measured B cell depletion under various conditions, as well as multiple clinical end points. RESULTS A single dose of GA101 was markedly more effective than RTX in depleting B cells in diseased MRL/lpr mice (P < 0.05). RTX overcame resistance to B cell depletion in diseased MRL/lpr mice with continuous treatments. GA101 was more effective in treating hCD20 MRL/lpr mice with early disease, as GA101-treated mice had reduced glomerulonephritis (P < 0.05), lower anti-RNA autoantibody titers (P < 0.05), and fewer activated CD4+ T cells (P < 0.0001) compared to RTX-treated mice. GA101 also treated advanced disease, and continual treatment prolonged survival. Using variants of GA101, we also elucidated B cell depletion mechanisms in vivo in mice with lupus. CONCLUSION Albeit both anti-CD20 antibodies ameliorated early disease, GA101 was more effective than RTX in important parameters, such as glomerulonephritis score. GA101 proved beneficial in an advanced disease model, where it prolonged survival. These data support clinical testing of GA101 in SLE and lupus nephritis.
Collapse
Affiliation(s)
- Anthony D Marinov
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | | | | | | | | | | | | | - Kevin M Nickerson
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Mark J Shlomchik
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
64
|
Wang J, Wu X, Tu Y, Dang J, Cai Z, Liao W, Quan W, Wei Y. An integrated analysis of lncRNA and mRNA expression profiles in the kidneys of mice with lupus nephritis. PeerJ 2021; 9:e10668. [PMID: 33628632 PMCID: PMC7894116 DOI: 10.7717/peerj.10668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/08/2020] [Indexed: 12/13/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are persistently expressed and have been described as potential biomarkers and therapeutic targets in various diseases. However, there is limited information regarding lncRNA expression in the tissue of kidney exhibiting lupus nephritis (LN)a serious complication of systemic lupus erythematosus (SLE). In this study, RNA sequencing (RNA-seq) was performed to characterize the lncRNA and mRNA expression in kidney tissues from LN (MRL/lpr) and control mice. We identified 12,979 novel lncRNAs in mouse. The expression profiles of both mRNAs and lncRNAs were differed significantly between LN and control mice. In particular, there were more upregulated lncRNAs and mRNAs than downregulated ones in the kidney tissues of LN mice. However, GO analysis showed that more downregulated genes were enriched in immune and inflammatory response-associated pathways. KEGG analysis showed that both downregulated and upregulated genes were enriched in a number of pathways, including the SLE pathway, and approximately half of these SLE-associated genes encoded inflammatory factors. Moreover, we observed that 2,181 DElncRNAs may have targeted and regulated the expression of 778 mRNAs in LN kidney tissues. The results of this study showed that 11 DElncRNAs targeted and were co-expressed with six immune and SLE-associated genes. qPCR analysis confirmed that lncRNA Gm20513 positively regulated the expression of the SLE-associated gene H2-Aa. In conclusion, the results of our study demonstrates that lncRNAs influence the progression of LN and provide some cues for further study of lncRNAs in LN. These results regarding the lncRNA-mRNAregulatory network may have important value in LN diagnosis and therapy.
Collapse
Affiliation(s)
- Juan Wang
- Nephrology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiongfei Wu
- Nephrology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yafang Tu
- Nephrology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jianzhong Dang
- Nephrology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhitao Cai
- Nephrology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wenjing Liao
- Nephrology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Weili Quan
- ABLife BioBigData Institute, Wuhan, Hubei, China
| | - Yaxun Wei
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China
| |
Collapse
|
65
|
Parikh SV, Malvar A, Shapiro J, Turman JM, Song H, Alberton V, Lococo B, Mejia-Vilet JM, Madhavan S, Zhang J, Yu L, Satoskar AA, Birmingham D, Jarjour WN, Rovin BH, Ganesan LP. A Novel Inflammatory Dendritic Cell That Is Abundant and Contiguous to T Cells in the Kidneys of Patients With Lupus Nephritis. Front Immunol 2021; 12:621039. [PMID: 33659005 PMCID: PMC7919935 DOI: 10.3389/fimmu.2021.621039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/25/2021] [Indexed: 01/06/2023] Open
Abstract
The mechanisms that promote local inflammatory injury during lupus nephritis (LN) flare are largely unknown. Understanding the key immune cells that drive intrarenal inflammation will advance our knowledge of disease pathogenesis and inform the development of new therapeutics for LN management. In this study, we analyzed kidney biopsies from patients with proliferative LN and identified a novel inflammatory dendritic cell (infDC) population that is highly expressed in the LN kidney, but minimally present in healthy human kidneys. During an agnostic evaluation of immune transcript expression in the kidneys of patients with proliferative LN, the most abundantly overexpressed transcript from isolated glomeruli was FCER1G, which encodes the Fc receptor gamma chain (FcRγ). To identify the cell types expressing FcRγ that infiltrate the kidney in LN, studies were done on kidney biopsies from patients with active LN using confocal immunofluorescence (IF) microscopy. This showed that FcRγ is abundantly present in the periglomerular (PG) region of the kidney and to a lesser extent in the tubulointerstitium (TI). Further investigation of the surface markers of these cells showed that they were FcRγ+, MHC II+, CD11c+, CD163+, CD5-, DC-SIGN+, CD64+, CD14+, CD16+, SIRPα+, CD206-, CD68-, CD123-, CD3-, and CD11b-, suggesting the cells were infDCs. Quantification of the infDCs showed an average 10-fold higher level of infDCs in the LN kidney compared to the healthy kidneys. Importantly, IF identified CD3+ T cells to be adjacent to these infDCs in the PG space of the LN kidney, whereas both cell types are minimally present in the healthy kidney. Thus, we have identified a previously undescribed DC in lupus kidneys that may interact with intrarenal T cells and play a role in the pathogenesis of kidney injury during LN flare.
Collapse
Affiliation(s)
- Samir V. Parikh
- Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Ana Malvar
- Nephrology Unit, Hospital Fernandez, Buenos Aires, Argentina
| | - John Shapiro
- Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - James M. Turman
- Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Huijuan Song
- Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Valeria Alberton
- Department of Pathology, Hospital Fernandez, Buenos Aires, Argentina
| | - Bruno Lococo
- Nephrology Unit, Hospital Fernandez, Buenos Aires, Argentina
| | - Juan M. Mejia-Vilet
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Sethu Madhavan
- Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jianying Zhang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States
| | - Lianbo Yu
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States
| | - Anjali A. Satoskar
- Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Dan Birmingham
- Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Wael N. Jarjour
- Division of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Brad H. Rovin
- Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Latha P. Ganesan
- Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Division of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
66
|
Viral Infections and Systemic Lupus Erythematosus: New Players in an Old Story. Viruses 2021; 13:v13020277. [PMID: 33670195 PMCID: PMC7916951 DOI: 10.3390/v13020277] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/06/2021] [Accepted: 02/07/2021] [Indexed: 02/07/2023] Open
Abstract
A causal link between viral infections and autoimmunity has been studied for a long time and the role of some viruses in the induction or exacerbation of systemic lupus erythematosus (SLE) in genetically predisposed patients has been proved. The strength of the association between different viral agents and SLE is variable. Epstein-Barr virus (EBV), parvovirus B19 (B19V), and human endogenous retroviruses (HERVs) are involved in SLE pathogenesis, whereas other viruses such as Cytomegalovirus (CMV) probably play a less prominent role. However, the mechanisms of viral-host interactions and the impact of viruses on disease course have yet to be elucidated. In addition to classical mechanisms of viral-triggered autoimmunity, such as molecular mimicry and epitope spreading, there has been a growing appreciation of the role of direct activation of innate response by viral nucleic acids and epigenetic modulation of interferon-related immune response. The latter is especially important for HERVs, which may represent the molecular link between environmental triggers and critical immune genes. Virus-specific proteins modulating interaction with the host immune system have been characterized especially for Epstein-Barr virus and explain immune evasion, persistent infection and self-reactive B-cell "immortalization". Knowledge has also been expanding on key viral proteins of B19-V and CMV and their possible association with specific phenotypes such as antiphospholipid syndrome. This progress may pave the way to new therapeutic perspectives, including the use of known or new antiviral drugs, postviral immune response modulation and innate immunity inhibition. We herein describe the state-of-the-art knowledge on the role of viral infections in SLE, with a focus on their mechanisms of action and potential therapeutic targets.
Collapse
|
67
|
McClung DM, Kalusche WJ, Jones KE, Ryan MJ, Taylor EB. Hypertension and endothelial dysfunction in the pristane model of systemic lupus erythematosus. Physiol Rep 2021; 9:e14734. [PMID: 33527772 PMCID: PMC7851437 DOI: 10.14814/phy2.14734] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 12/19/2020] [Indexed: 01/01/2023] Open
Abstract
Autoimmune diseases such as psoriasis, rheumatoid arthritis, and systemic lupus erythematosus (SLE) have high rates of hypertension and cardiovascular disease. Systemic lupus erythematosus is a prototypic autoimmune disorder that primarily affects women of childbearing age and is associated with a loss of self-tolerance, autoreactive B and T lymphocytes, and the production of autoantibodies, especially to nuclear components. In this study, we hypothesized that the pristane-inducible model of SLE would develop hypertension and vascular dysfunction as the disease progressed. To test this hypothesis, female C57BL/6 mice were administered PBS or pristane. Seven months after pristane administration, mice developed various autoantibodies, including anti-dsDNA IgG, anti-ssDNA IgG, and anti-nRNP IgG, as well as hypergammaglobulinemia. Several other immunological changes, including increased circulating neutrophils and increased CD4- CD8- (double negative) thymocytes were also detected. Mean arterial pressure (MAP) was elevated in pristane-treated mice when compared to PBS-treated mice. In addition, second-order mesenteric arteries from pristine-treated mice had impaired relaxation to the endothelium-dependent vasodilator acetylcholine compared to PBS-treated mice. These data suggest that the immune system dysfunction present in the pristane model of lupus contributes to the development of hypertension and vascular dysfunction.
Collapse
Affiliation(s)
- Daniel M. McClung
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| | - William J. Kalusche
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Katie E. Jones
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Michael J. Ryan
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
- G.V. (Sonny) Montgomery Veterans Affairs Medical CenterJacksonMSUSA
| | - Erin B. Taylor
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| |
Collapse
|
68
|
Chen PM, Tsokos GC. T Cell Abnormalities in the Pathogenesis of Systemic Lupus Erythematosus: an Update. Curr Rheumatol Rep 2021; 23:12. [PMID: 33512577 DOI: 10.1007/s11926-020-00978-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2020] [Indexed: 12/01/2022]
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus is a complex disease with broad spectrum of clinical manifestations. In addition to abnormal B cell responsive leading to autoantibody production, various T cells also play different roles in promoting systemic autoimmunity and end organ damage. We aim to provide a review on recent developments in how abnormalities in different T cells subsets contribute to systemic lupus erythematosus pathogenesis and how they inform the consideration of new promising therapeutics. RECENT FINDINGS Distinct subsets of T cells known as T follicular helper cells enable the production of pathogenic autoantibodies. Detailed understanding of the B cell helping T cell subsets should improve the performance of clinical trials targeting the cognate T:B cell interaction. CD8+ T cells play a role in peripheral tolerance and reversal of its exhausted phenotype could potentially alleviate both systemic autoimmunity and the risk of infection. Research on the abnormal lupus T cell signaling also leads to putative therapeutic targets able to restore interleukin-2 production and suppress the production of the pathogenic IL-17 cytokine. Recently, several studies have focused on dissecting T cell populations located in the damaged organs, aiming to target the pathogenic processes specific to each organ. Numerous T cell subsets play distinct roles in SLE pathogenesis and recent research in understanding abnormal signaling pathways, cellular metabolism, and environmental cues pave the way for the development of novel therapeutics.
Collapse
Affiliation(s)
- Ping-Min Chen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
69
|
Zhao X, Zhang L, Wang J, Zhang M, Song Z, Ni B, You Y. Identification of key biomarkers and immune infiltration in systemic lupus erythematosus by integrated bioinformatics analysis. J Transl Med 2021; 19:35. [PMID: 33468161 PMCID: PMC7814551 DOI: 10.1186/s12967-020-02698-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/31/2020] [Indexed: 02/07/2023] Open
Abstract
Background Systemic lupus erythematosus (SLE) is a multisystemic, chronic inflammatory disease characterized by destructive systemic organ involvement, which could cause the decreased functional capacity, increased morbidity and mortality. Previous studies show that SLE is characterized by autoimmune, inflammatory processes, and tissue destruction. Some seriously-ill patients could develop into lupus nephritis. However, the cause and underlying molecular events of SLE needs to be further resolved. Methods The expression profiles of GSE144390, GSE4588, GSE50772 and GSE81622 were downloaded from the Gene Expression Omnibus (GEO) database to obtain differentially expressed genes (DEGs) between SLE and healthy samples. The gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichments of DEGs were performed by metascape etc. online analyses. The protein–protein interaction (PPI) networks of the DEGs were constructed by GENEMANIA software. We performed Gene Set Enrichment Analysis (GSEA) to further understand the functions of the hub gene, Weighted gene co‐expression network analysis (WGCNA) would be utilized to build a gene co‐expression network, and the most significant module and hub genes was identified. CIBERSORT tools have facilitated the analysis of immune cell infiltration patterns of diseases. The receiver operating characteristic (ROC) analyses were conducted to explore the value of DEGs for SLE diagnosis. Results In total, 6 DEGs (IFI27, IFI44, IFI44L, IFI6, EPSTI1 and OAS1) were screened, Biological functions analysis identified key related pathways, gene modules and co‐expression networks in SLE. IFI27 may be closely correlated with the occurrence of SLE. We found that an increased infiltration of moncytes, while NK cells resting infiltrated less may be related to the occurrence of SLE. Conclusion IFI27 may be closely related pathogenesis of SLE, and represents a new candidate molecular marker of the occurrence and progression of SLE. Moreover immune cell infiltration plays important role in the progession of SLE.
Collapse
Affiliation(s)
- Xingwang Zhao
- Department of Dermatology, Southwest Hospital, Army Medical University, (Third Military Medical University), Chongqing, 400038, China
| | - Longlong Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Juan Wang
- Department of Dermatology, Southwest Hospital, Army Medical University, (Third Military Medical University), Chongqing, 400038, China
| | - Min Zhang
- Department of Dermatology, Southwest Hospital, Army Medical University, (Third Military Medical University), Chongqing, 400038, China
| | - Zhiqiang Song
- Department of Dermatology, Southwest Hospital, Army Medical University, (Third Military Medical University), Chongqing, 400038, China
| | - Bing Ni
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, (Third Military Medical University), Chongqing, China.
| | - Yi You
- Department of Dermatology, Southwest Hospital, Army Medical University, (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
70
|
Zhang L, Zhang M, Chen X, He Y, Chen R, Zhang J, Huang J, Ouyang C, Shi G. Identification of the tubulointerstitial infiltrating immune cell landscape and immune marker related molecular patterns in lupus nephritis using bioinformatics analysis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 8:1596. [PMID: 33437795 PMCID: PMC7791250 DOI: 10.21037/atm-20-7507] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease that commonly affects the kidneys. Research into markers that can predict the prognosis of tubulointerstitial lupus nephritis (LN) has been impeded by the lack of well-designed studies. Methods In this study, we selected and merged 3 sets of renal biopsy tubulointerstitial data from GSE32591, GSE69438, and GSE127797, including 95 LN and 15 living healthy donors. CIBERSORTx was utilized for differentially infiltrating immune cell (DIIC) analysis. Weighted Gene Co-Expression network analysis (WGCNA) was employed to explore differentially expressed gene (DEG) related modules. Combined WGCNA hub genes and protein-protein interaction (PPI) validation was used for immune marker identification. Lastly, unsupervised clustering was carried out to validate the correlation between these markers and clinical characteristics. Results Our findings unveiled TYROBP, C1QB, LAPTM5, CTSS, PTPRC as the 5 immune markers, which were negatively correlated with glomerular filtration rate (GFR). Specifically, the expression levels of TYROBP and C1QB were significantly different between proliferative LN (PLN) and membranous LN (MLN). Unsupervised clustering could aggregate LN by these immune marker expression spectrums. Conclusions This study is the first to identify infiltrating immune cells and associated molecular patterns in the tubulointerstitium of LN by utilizing bioinformatics methods. These findings contribute to a better understanding of the mechanisms behind LN, and promote more precise diagnosis.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Nephrology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,The Fifth Hospital of Xiamen, Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Mengqin Zhang
- Department of Rheumatology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xing Chen
- Department of Nephrology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,The Fifth Hospital of Xiamen, Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yan He
- Department of Rheumatology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Rongjuan Chen
- Department of Rheumatology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jun Zhang
- Department of Nephrology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,The Fifth Hospital of Xiamen, Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jiyi Huang
- Department of Nephrology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,The Fifth Hospital of Xiamen, Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Chun Ouyang
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guixiu Shi
- Department of Rheumatology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| |
Collapse
|
71
|
Psarras A, Alase A, Antanaviciute A, Carr IM, Md Yusof MY, Wittmann M, Emery P, Tsokos GC, Vital EM. Functionally impaired plasmacytoid dendritic cells and non-haematopoietic sources of type I interferon characterize human autoimmunity. Nat Commun 2020; 11:6149. [PMID: 33262343 PMCID: PMC7708979 DOI: 10.1038/s41467-020-19918-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 10/28/2020] [Indexed: 12/23/2022] Open
Abstract
Autoimmune connective tissue diseases arise in a stepwise fashion from asymptomatic preclinical autoimmunity. Type I interferons have a crucial role in the progression to established autoimmune diseases. The cellular source and regulation in disease initiation of these cytokines is not clear, but plasmacytoid dendritic cells have been thought to contribute to excessive type I interferon production. Here, we show that in preclinical autoimmunity and established systemic lupus erythematosus, plasmacytoid dendritic cells are not effector cells, have lost capacity for Toll-like-receptor-mediated cytokine production and do not induce T cell activation, independent of disease activity and the blood interferon signature. In addition, plasmacytoid dendritic cells have a transcriptional signature indicative of cellular stress and senescence accompanied by increased telomere erosion. In preclinical autoimmunity, we show a marked enrichment of an interferon signature in the skin without infiltrating immune cells, but with interferon-κ production by keratinocytes. In conclusion, non-hematopoietic cellular sources, rather than plasmacytoid dendritic cells, are responsible for interferon production prior to clinical autoimmunity.
Collapse
Affiliation(s)
- Antonios Psarras
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- National Institute for Health Research (NIHR), Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Adewonuola Alase
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | | | - Ian M Carr
- Leeds Institute for Data Analytics, University of Leeds, Leeds, UK
| | - Md Yuzaiful Md Yusof
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- National Institute for Health Research (NIHR), Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Miriam Wittmann
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- National Institute for Health Research (NIHR), Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- National Institute for Health Research (NIHR), Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - George C Tsokos
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Edward M Vital
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
- National Institute for Health Research (NIHR), Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| |
Collapse
|
72
|
Khodoun M, Chimote AA, Ilyas FZ, Duncan HJ, Moncrieffe H, Kant KS, Conforti L. Targeted knockdown of Kv1.3 channels in T lymphocytes corrects the disease manifestations associated with systemic lupus erythematosus. SCIENCE ADVANCES 2020; 6:6/47/eabd1471. [PMID: 33208373 PMCID: PMC7673800 DOI: 10.1126/sciadv.abd1471] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/02/2020] [Indexed: 05/16/2023]
Abstract
Lupus nephritis (LN) is an autoimmune disease with substantial morbidity/mortality and limited efficacy of available therapies. Memory T (Tm) lymphocytes infiltrate LN kidneys, contributing to organ damage. Analysis of LN, diabetic nephropathy, and healthy donor kidney biopsies revealed high infiltration of active CD8+ Tm cells expressing high voltage-dependent Kv1.3 potassium channels-key T cell function regulators-in LN. Nanoparticles that selectively down-regulate Kv1.3 in Tm cells (Kv1.3-NPs) reduced CD40L and interferon-γ (IFNγ) in Tm cells from LN patients in vitro. Kv1.3-NPs were tested in humanized LN mice obtained by engrafting peripheral blood mononuclear cells (PBMCs) from LN patients into immune-deficient mice. LN mice exhibited features of the disease: increased IFNγ and CD3+CD8+ T cell renal infiltration, and reduced survival versus healthy donor PBMC engrafted mice. Kv1.3-NP treatment of patient PBMCs before engraftment decreased CD40L/IFNγ and prolonged survival of LN mice. These data show the potential benefits of targeting Kv1.3 in LN.
Collapse
Affiliation(s)
- Marat Khodoun
- Division of Rheumatology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ameet A Chimote
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Farhan Z Ilyas
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Heather J Duncan
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Halima Moncrieffe
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| | - K Shashi Kant
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Laura Conforti
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
73
|
Nüssing S, Trapani JA, Parish IA. Revisiting T Cell Tolerance as a Checkpoint Target for Cancer Immunotherapy. Front Immunol 2020; 11:589641. [PMID: 33072137 PMCID: PMC7538772 DOI: 10.3389/fimmu.2020.589641] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/04/2020] [Indexed: 12/30/2022] Open
Abstract
Immunotherapy has revolutionized the treatment of cancer. Nevertheless, the majority of patients do not respond to therapy, meaning a deeper understanding of tumor immune evasion strategies is required to boost treatment efficacy. The vast majority of immunotherapy studies have focused on how treatment reinvigorates exhausted CD8+ T cells within the tumor. In contrast, how therapies influence regulatory processes within the draining lymph node is less well studied. In particular, relatively little has been done to examine how tumors may exploit peripheral CD8+ T cell tolerance, an under-studied immune checkpoint that under normal circumstances prevents detrimental autoimmune disease by blocking the initiation of T cell responses. Here we review the therapeutic potential of blocking peripheral CD8+ T cell tolerance for the treatment of cancer. We first comprehensively review what has been learnt about the regulation of CD8+ T cell peripheral tolerance from the non-tumor models in which peripheral tolerance was first defined. We next consider how the tolerant state differs from other states of negative regulation, such as T cell exhaustion and senescence. Finally, we describe how tumors hijack the peripheral tolerance immune checkpoint to prevent anti-tumor immune responses, and argue that disruption of peripheral tolerance may contribute to both the anti-cancer efficacy and autoimmune side-effects of immunotherapy. Overall, we propose that a deeper understanding of peripheral tolerance will ultimately enable the development of more targeted and refined cancer immunotherapy approaches.
Collapse
Affiliation(s)
- Simone Nüssing
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Joseph A Trapani
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Ian A Parish
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
74
|
Kim MS, Ma S, Chelariu-Raicu A, Leuschner C, Alila HW, Lee S, Coleman RL, Sood AK. Enhanced Immunotherapy with LHRH-R Targeted Lytic Peptide in Ovarian Cancer. Mol Cancer Ther 2020; 19:2396-2406. [PMID: 32943548 DOI: 10.1158/1535-7163.mct-20-0030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 06/08/2020] [Accepted: 08/26/2020] [Indexed: 11/16/2022]
Abstract
Here, we examined the role of EP-100 [luteinizing hormone-releasing hormone (LHRH) ligand joined to a lytic peptide], improving the efficacy of immune checkpoint blockade. LHRH-R-positive murine ovarian cancer cells (ID8, IG10, IF5, and 2C12) were sensitive to EP-100 and were specifically killed at low micromolar levels through LHRH-R. EP-100 increased PD-L1 levels on murine ovarian cancer cells. In vivo syngeneic mouse models (ID8 and IG10) demonstrated that single-agent EP-100 reduced tumor volume, tumor weight, and ascites volume. The greatest reductions in tumor and ascites volume were observed with the combination of EP-100 with an anti-PD-L1 antibody. Immune profiling analysis showed that the population of CD8+ T cells, natural killer cells, dendritic cells, and macrophages were significantly increased in tumor and ascitic fluid samples treated with anti-PD-L1, EP-100, and the combination. However, monocytic myeloid suppressor cells, B cells, and regulatory T cells were decreased in tumors treated with anti-PD-L1, EP-100, or the combination. In vitro cytokine arrays revealed that EP-100 induced IL1α, IL33, CCL20, VEGF, and Low-density lipoprotein receptor (LDLR) secretion. Of these, we validated increasing IL33 levels following EP-100 treatment in vitro and in vivo; we determined the specific biological role of CD8+ T-cell activation with IL33 gene silencing using siRNA and Cas9-CRISPR approaches. In addition, we found that CD8+ T cells expressed very low level of LHRH-R and were not affected by EP-100. Taken together, EP-100 treatment had a substantial antitumor efficacy, particularly in combination with an anti-PD-L1 antibody. These results warrant further clinical development of this combination.
Collapse
Affiliation(s)
- Mark Seungwook Kim
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Shaolin Ma
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Anca Chelariu-Raicu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Obstetrics and Gynecology, University of Hospital, LMU Munich, Germany
| | | | | | - Sanghoon Lee
- Department of System Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
75
|
Ostendorf L, Burns M, Durek P, Heinz GA, Heinrich F, Garantziotis P, Enghard P, Richter U, Biesen R, Schneider U, Knebel F, Burmester G, Radbruch A, Mei HE, Mashreghi MF, Hiepe F, Alexander T. Targeting CD38 with Daratumumab in Refractory Systemic Lupus Erythematosus. N Engl J Med 2020; 383:1149-1155. [PMID: 32937047 DOI: 10.1056/nejmoa2023325] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Daratumumab, a human monoclonal antibody that targets CD38, depletes plasma cells and is approved for the treatment of multiple myeloma. Long-lived plasma cells are implicated in the pathogenesis of systemic lupus erythematosus because they secrete autoantibodies, but they are unresponsive to standard immunosuppression. We describe the use of daratumumab that induced substantial clinical responses in two patients with life-threatening lupus, with the clinical responses sustained by maintenance therapy with belimumab, an antibody to B-cell activating factor. Significant depletion of long-lived plasma cells, reduction of interferon type I activity, and down-regulation of T-cell transcripts associated with chronic inflammation were documented. (Supported by the Deutsche Forschungsgemeinschaft and others.).
Collapse
Affiliation(s)
- Lennard Ostendorf
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Marie Burns
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Pawel Durek
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Gitta Anne Heinz
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Frederik Heinrich
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Panagiotis Garantziotis
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Philipp Enghard
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Ulrich Richter
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Robert Biesen
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Udo Schneider
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Fabian Knebel
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Gerd Burmester
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Andreas Radbruch
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Henrik E Mei
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Mir-Farzin Mashreghi
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Falk Hiepe
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| | - Tobias Alexander
- From the Departments of Rheumatology and Clinical Immunology (L.O., P.G., R.B., U.S., G.B., F. Hiepe, T.A.), Nephrology and Internal Intensive Care Unit (P.E.), Hematology, Oncology and Tumor Immunology (U.R.), and Cardiology and Angiology, Campus Mitte (F.K.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Deutsches Rheuma-Forschungszentrum (DRFZ) Institute of the Leibniz Association (L.O., M.B., P.D., G.A.H., F. Heinrich, A.R., H.E.M., M.-F.M., F. Hiepe, T.A.), German Center for Cardiovascular Research (DZHK) (F.K.), and BIH Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin (M.-F.M.) - all in Berlin; and the Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation, Academy of Athens, Athens (P.G.)
| |
Collapse
|
76
|
Dorraji SE, Kanapathippillai P, Hovd AMK, Stenersrød MR, Horvei KD, Ursvik A, Figenschau SL, Thiyagarajan D, Fenton CG, Pedersen HL, Fenton KA. Kidney Tertiary Lymphoid Structures in Lupus Nephritis Develop into Large Interconnected Networks and Resemble Lymph Nodes in Gene Signature. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2203-2225. [PMID: 32818496 DOI: 10.1016/j.ajpath.2020.07.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 06/30/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022]
Abstract
Immune aggregates organized as tertiary lymphoid structures (TLS) are observed within the kidneys of patients with systemic lupus erythematosus and lupus nephritis (LN). Renal TLS was characterized in lupus-prone New Zealand black × New Zealand white F1 mice analyzing cell composition and vessel formation. RNA sequencing was performed on transcriptomes isolated from lymph nodes, macrodissected TLS from kidneys, and total kidneys of mice at different disease stages by using a personal genome machine and RNA sequencing. Formation of TLS was found in anti-double-stranded DNA antibody-positive mice, and the structures were organized as interconnected large networks with distinct T/B cell zones with adjacent dendritic cells, macrophages, plasma cells, high endothelial venules, supporting follicular dendritic cells network, and functional germinal centers. Comparison of gene profiles of whole kidney, renal TLS, and lymph nodes revealed a similar gene signature of TLS and lymph nodes. The up-regulated genes within the kidneys of lupus-prone mice during LN development reflected TLS formation, whereas the down-regulated genes were involved in metabolic processes of the kidney cells. A comparison with human LN gene expression revealed similar up-regulated genes as observed during the development of murine LN and TLS. In conclusion, kidney TLS have a similar cell composition, structure, and gene signature as lymph nodes and therefore may function as a kidney-specific type of lymph node.
Collapse
Affiliation(s)
- Seyed Esmaeil Dorraji
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Premasany Kanapathippillai
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Aud-Malin Karlsson Hovd
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Mikael Ryan Stenersrød
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Kjersti Daae Horvei
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Anita Ursvik
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Stine Linn Figenschau
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Dhivya Thiyagarajan
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Christopher Graham Fenton
- Genomic Support Center, Department of Clinical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromso, Norway
| | - Hege Lynum Pedersen
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway
| | - Kristin Andreassen Fenton
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
77
|
Symonds AL, Zheng W, Miao T, Wang H, Wang T, Kiome R, Hou X, Li S, Wang P. Egr2 and 3 control inflammation, but maintain homeostasis, of PD-1 high memory phenotype CD4 T cells. Life Sci Alliance 2020; 3:3/9/e202000766. [PMID: 32709717 PMCID: PMC7391068 DOI: 10.26508/lsa.202000766] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 01/13/2023] Open
Abstract
PD-1high memory CD4 T cells are pathogenic in autoimmune disease; here they show their expression of Egr2 is defective in rheumatoid arthritis and Egr2 & 3 control their inflammation and homeostasis. The transcription factors Egr2 and 3 are essential for controlling inflammatory autoimmune responses of memory phenotype (MP) CD4 T cells. However, the mechanism is still unclear. We have now found that the Egr2+ subset (PD-1high MP) of MP CD4 T cells expresses high levels of checkpoint molecules (PD-1 and Lag3) and also markers of effector T cells (CXCR3 and ICAM-1). Egr2/3 are not required for PD-1high MP CD4 cell development but mediate a unique transcriptional programme that effectively controls their inflammatory responses, while promoting homeostatic proliferation and adaptive responses. Egr2 negative PD-1high MP CD4 T cells are impaired in homeostatic proliferation and adaptive responses against viral infection but display inflammatory responses to innate stimulation such as IL-12. PD-1high MP CD4 T cells have recently been implicated in rheumatoid arthritis pathogenesis, and we have now found that Egr2 expression is reduced in PD-1high MP CD4 T cells from patients with active rheumatoid arthritis compared with healthy controls. These findings demonstrate that Egr2/3 control the inflammatory responses of PD-1high MP CD4 T cells and maintain their adaptive immune fitness.
Collapse
Affiliation(s)
- Alistair Lj Symonds
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Wei Zheng
- Division of Rheumatology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tizong Miao
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Haiyu Wang
- Division of Rheumatology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - TieShang Wang
- Division of Rheumatology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ruth Kiome
- Bioscience, Brunel University, Uxbridge, UK
| | - Xiujuan Hou
- Division of Rheumatology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Suling Li
- Bioscience, Brunel University, Uxbridge, UK
| | - Ping Wang
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
78
|
Moore E, Huang MW, Jain S, Chalmers SA, Macian F, Putterman C. The T Cell Receptor Repertoire in Neuropsychiatric Systemic Lupus Erythematosus. Front Immunol 2020; 11:1476. [PMID: 32765512 PMCID: PMC7379895 DOI: 10.3389/fimmu.2020.01476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/05/2020] [Indexed: 11/13/2022] Open
Abstract
Objective: In systemic lupus erythematosus (SLE), widespread T cell infiltration into target organs contributes to inflammation and organ damage. Autoreactive T cells become aberrantly activated in this disease due to dysfunctional T cell receptor signaling that lowers the activation threshold. Characterizing the T cell repertoire can provide further insight into the specific homing and proliferation of these T cells into lupus target organs. In the spontaneous lupus model, MRL/lpr, the TCR repertoire has not been fully elucidated, especially for T cells infiltrating the brain. Our aim was to investigate and compare the TCR repertoire between MRL/lpr mice and its congenic controls, MRL/MpJ, and within MRL/lpr tissues. Methods: Spleen, salivary gland, and brain choroid plexus were isolated from female MRL/lpr mice and MRL/MpJ mice. The TCRβ CDR3 region was analyzed by multiplex PCRs and sequencing. Results: Significant differences were seen not only between the MRL/lpr and MRL/MpJ spleens, but also between MRL/lpr tissues. The TCR repertoire in MRL/lpr choroid plexus tissues had significantly increased clonality and sequence homology compared to MRL/lpr spleen and salivary gland. The consensus sequence, CASSQDWGGYEQYFF, was identified in the MRL/lpr choroid plexus repertoire. Conclusions: The TCR repertoire in lupus prone mice is not uniform between target organs, and suggests that T cells are specifically recruited into the choroid plexus of MRL/lpr mice. Further studies are needed to determine the antigen specificities for these infiltrating T cells in target organs of lupus mice, and their possible contribution to the pathogenesis of neuropsychiatric disease and other lupus manifestations.
Collapse
Affiliation(s)
- Erica Moore
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Michelle W Huang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Shweta Jain
- Early Discovery and Fundamental Research, Hansoh Bio, Rockville, MD, United States
| | - Samantha A Chalmers
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Fernando Macian
- Department of Pathology, Albert Einstein College of Medicine, New York, NY, United States
| | - Chaim Putterman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, United States.,Bar-Ilan University Azrieli Faculty of Medicine, Ramat Gan, Israel.,Galilee Medical Center, Nahariya, Israel
| |
Collapse
|
79
|
Chen W, Li W, Zhang Z, Tang X, Wu S, Yao G, Li K, Wang D, Xu Y, Feng R, Duan X, Fan X, Lu L, Chen W, Li C, Sun L. Lipocalin-2 Exacerbates Lupus Nephritis by Promoting Th1 Cell Differentiation. J Am Soc Nephrol 2020; 31:2263-2277. [PMID: 32646856 DOI: 10.1681/asn.2019090937] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 05/24/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Lipocalin-2 (LCN2) is an indicator of the severity of lupus nephritis (LN) and plays a pivotal role in immune responses, but it is not known if its effect on LN pathogenesis derives from regulating the immune imbalance of T lymphocyte subsets. METHODS The expression of LCN2 in T cells and kidneys was assessed in renal biopsies from patients with LN. We investigated the relationship between LCN2 levels and development of LN and systemic illness by injecting anti-LCN2 antibodies into MRL/lpr mice and analyzing pristane-treated LCN2 -/- mice. RESULTS LCN2 is highly expressed in CD4+ T cells and in renal tissues, and is associated with severe renal damage in patients with LN and in mice with experimental lupus. LCN2 promotes IFN-γ overexpression in CD4+ T cells through the IL-12/STAT4 pathway in an autocrine or paracrine manner. Both neutralization of LCN2 in MRL/lpr mice and genetic depletion of LCN2 in pristane-induced lupus mice greatly ameliorate nephritis. The frequency and number of splenic and renal Th1 cells decrease in proportion to LN disease activity. Conversely, administration of LCN2 exacerbates the disease with significantly higher renal activity scores and increased numbers of Th1 cells. CONCLUSIONS LCN2 plays a crucial role in Th1 cell differentiation, and may present a potential therapeutic target for LN.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Wenchao Li
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Zhuoya Zhang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Shufang Wu
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Genhong Yao
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Kang Li
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Dandan Wang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Yuemei Xu
- Department of Pathology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Ruihai Feng
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Xiaoxiao Duan
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Xiangshan Fan
- Department of Pathology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Liwei Lu
- Department of Pathology, Center of Infection and Immunology, University of Hong Kong, Hong Kong, China
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Chaojun Li
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China .,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China .,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| |
Collapse
|
80
|
Alrasheed N, Lee L, Ghorani E, Henry JY, Conde L, Chin M, Galas-Filipowicz D, Furness AJS, Chavda SJ, Richards H, De-Silva D, Cohen OC, Patel D, Brooks A, Rodriguez-Justo M, Pule M, Herrero J, Quezada SA, Yong KL. Marrow-Infiltrating Regulatory T Cells Correlate with the Presence of Dysfunctional CD4 +PD-1 + Cells and Inferior Survival in Patients with Newly Diagnosed Multiple Myeloma. Clin Cancer Res 2020; 26:3443-3454. [PMID: 32220887 DOI: 10.1158/1078-0432.ccr-19-1714] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/21/2019] [Accepted: 03/24/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Immune dysregulation is described in multiple myeloma. While preclinical models suggest a role for altered T-cell immunity in disease progression, the contribution of immune dysfunction to clinical outcomes remains unclear. We aimed to characterize marrow-infiltrating T cells in newly diagnosed patients and explore associations with outcomes of first-line therapy. EXPERIMENTAL DESIGN We undertook detailed characterization of T cells from bone marrow (BM) samples, focusing on immune checkpoints and features of immune dysfunction, correlating with clinical features and progression-free survival. RESULTS We found that patients with multiple myeloma had greater abundance of BM regulatory T cells (Tregs) which, in turn, expressed higher levels of the activation marker CD25 compared with healthy donors. Patients with higher frequencies of Tregs had shorter PFS and a distinct Treg immune checkpoint profile (increased PD-1, LAG-3) compared with patients with lower frequencies of Tregs. Analysis of CD4 and CD8 effectors revealed that low CD4effector (CD4eff):Treg ratio and increased frequency of PD-1-expressing CD4eff cells were independent predictors of early relapse over and above conventional risk factors, such as genetic risk and depth of response. Ex vivo functional analysis and RNA sequencing revealed that CD4 and CD8 cells from patients with greater abundance of CD4effPD-1+ cells displayed transcriptional and secretory features of dysfunction. CONCLUSIONS BM-infiltrating T-cell subsets, specifically Tregs and PD-1-expressing CD4 effectors, negatively influence clinical outcomes in newly diagnosed patients. Pending confirmation in larger cohorts and further mechanistic work, these immune parameters may inform new risk models, and present potential targets for immunotherapeutic strategies.
Collapse
Affiliation(s)
- Nouf Alrasheed
- Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Lydia Lee
- Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Ehsan Ghorani
- Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Jake Y Henry
- Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Lucia Conde
- Bill Lyons Informatics Centre, University College London Cancer Institute, London, United Kingdom
| | - Melody Chin
- Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Daria Galas-Filipowicz
- Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Andrew J S Furness
- Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Selina J Chavda
- Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Huw Richards
- Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Dunnya De-Silva
- Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Oliver C Cohen
- University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Dominic Patel
- Department of Histopathology, University College London, London, United Kingdom
| | - Anthony Brooks
- Institute of Child Health, University College London, London, United Kingdom
| | | | - Martin Pule
- Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Javier Herrero
- Bill Lyons Informatics Centre, University College London Cancer Institute, London, United Kingdom
| | - Sergio A Quezada
- Research Department of Haematology, University College London Cancer Institute, London, United Kingdom.
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Kwee L Yong
- Research Department of Haematology, University College London Cancer Institute, London, United Kingdom.
| |
Collapse
|
81
|
Olson WJ, Jakic B, Hermann‐Kleiter N. Regulation of the germinal center response by nuclear receptors and implications for autoimmune diseases. FEBS J 2020; 287:2866-2890. [PMID: 32246891 PMCID: PMC7497069 DOI: 10.1111/febs.15312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/15/2020] [Accepted: 03/26/2020] [Indexed: 01/09/2023]
Abstract
The immune system plays an essential role in protecting the host from infectious diseases and cancer. Notably, B and T lymphocytes from the adaptive arm of the immune system can co-operate to form long-lived antibody responses and are therefore the main target in vaccination approaches. Nevertheless, protective immune responses must be tightly regulated to avoid hyper-responsiveness and responses against self that can result in autoimmunity. Nuclear receptors (NRs) are perfectly adapted to rapidly alter transcriptional cellular responses to altered environmental settings. Their functional role is associated with both immune deficiencies and autoimmunity. Despite extensive linking of nuclear receptor function with specific CD4 T helper subsets, research on the functional roles and mechanisms of specific NRs in CD4 follicular T helper cells (Tfh) and germinal center (GC) B cells during the germinal center reaction is just emerging. We review recent advances in our understanding of NR regulation in specific cell types of the GC response and discuss their implications for autoimmune diseases such as systemic lupus erythematosus (SLE).
Collapse
Affiliation(s)
- William J. Olson
- Translational Cell GeneticsDepartment of Pharmacology and GeneticsMedical University of InnsbruckAustria
| | - Bojana Jakic
- Translational Cell GeneticsDepartment of Pharmacology and GeneticsMedical University of InnsbruckAustria
- Department of Immunology, Genetics and PathologyUppsala UniversitySweden
| | - Natascha Hermann‐Kleiter
- Translational Cell GeneticsDepartment of Pharmacology and GeneticsMedical University of InnsbruckAustria
| |
Collapse
|
82
|
Grebinoski S, Vignali DA. Inhibitory receptor agonists: the future of autoimmune disease therapeutics? Curr Opin Immunol 2020; 67:1-9. [PMID: 32619929 DOI: 10.1016/j.coi.2020.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/31/2022]
Abstract
Central and peripheral tolerance both contribute to protection against autoimmunity. The pathogenesis of autoimmunity, however, can result from critical deficits or limitations in peripheral and/or central tolerance mechanisms, presenting an opportunity for therapeutic intervention. Recent advances highlight the substantial impact of inhibitory receptors (IRs), which mediate peripheral tolerance, in autoimmunity. Deletion and blockade studies in mice, IR disruption in humans, and correlation with positive disease outcomes all highlight potential clinical benefits of enhancing IR signaling (agonism)-specifically CTLA4, PD1, LAG3, TIM3 and TIGIT-to treat autoimmune disease. Although critical questions remain, IR agonists represent an unappreciated and untapped opportunity for the treatment of autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Stephanie Grebinoski
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, 200 Lothrop St., Pittsburgh, PA 15213, USA
| | - Dario Aa Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA.
| |
Collapse
|
83
|
Tsokos GC. Autoimmunity and organ damage in systemic lupus erythematosus. Nat Immunol 2020; 21:605-614. [PMID: 32367037 PMCID: PMC8135909 DOI: 10.1038/s41590-020-0677-6] [Citation(s) in RCA: 280] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/31/2020] [Indexed: 01/07/2023]
Abstract
Impressive progress has been made over the last several years toward understanding how almost every aspect of the immune system contributes to the expression of systemic autoimmunity. In parallel, studies have shed light on the mechanisms that contribute to organ inflammation and damage. New approaches that address the complicated interaction between genetic variants, epigenetic processes, sex and the environment promise to enlighten the multitude of pathways that lead to what is clinically defined as systemic lupus erythematosus. It is expected that each patient owns a unique 'interactome', which will dictate specific treatment.
Collapse
Affiliation(s)
- George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA.
| |
Collapse
|
84
|
Gryzik S, Hoang Y, Lischke T, Mohr E, Venzke M, Kadner I, Poetzsch J, Groth D, Radbruch A, Hutloff A, Baumgrass R. Identification of a super-functional Tfh-like subpopulation in murine lupus by pattern perception. eLife 2020; 9:53226. [PMID: 32441253 PMCID: PMC7274784 DOI: 10.7554/elife.53226] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 05/20/2020] [Indexed: 01/20/2023] Open
Abstract
Dysregulated cytokine expression by T cells plays a pivotal role in the pathogenesis of autoimmune diseases. However, the identification of the corresponding pathogenic subpopulations is a challenge, since a distinction between physiological variation and a new quality in the expression of protein markers requires combinatorial evaluation. Here, we were able to identify a super-functional follicular helper T cell (Tfh)-like subpopulation in lupus-prone NZBxW mice with our binning approach "pattern recognition of immune cells (PRI)". PRI uncovered a subpopulation of IL-21+ IFN-γhigh PD-1low CD40Lhigh CXCR5- Bcl-6- T cells specifically expanded in diseased mice. In addition, these cells express high levels of TNF-α and IL-2, and provide B cell help for IgG production in an IL-21 and CD40L dependent manner. This super-functional T cell subset might be a superior driver of autoimmune processes due to a polyfunctional and high cytokine expression combined with Tfh-like properties.
Collapse
Affiliation(s)
- Stefanie Gryzik
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Yen Hoang
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| | - Timo Lischke
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Elodie Mohr
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Melanie Venzke
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Isabelle Kadner
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| | - Josephine Poetzsch
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| | | | - Andreas Radbruch
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,Charité, Campus Mitte, Berlin, Germany
| | - Andreas Hutloff
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Ria Baumgrass
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| |
Collapse
|
85
|
Ghorani E, Reading JL, Henry JY, Massy MRD, Rosenthal R, Turati V, Joshi K, Furness AJS, Ben Aissa A, Saini SK, Ramskov S, Georgiou A, Sunderland MW, Wong YNS, Mucha MVD, Day W, Galvez-Cancino F, Becker PD, Uddin I, Oakes T, Ismail M, Ronel T, Woolston A, Jamal-Hanjani M, Veeriah S, Birkbak NJ, Wilson GA, Litchfield K, Conde L, Guerra-Assunção JA, Blighe K, Biswas D, Salgado R, Lund T, Bakir MA, Moore DA, Hiley CT, Loi S, Sun Y, Yuan Y, AbdulJabbar K, Turajilic S, Herrero J, Enver T, Hadrup SR, Hackshaw A, Peggs KS, McGranahan N, Chain B, Swanton C, Quezada SA. The T cell differentiation landscape is shaped by tumour mutations in lung cancer. NATURE CANCER 2020; 1:546-561. [PMID: 32803172 PMCID: PMC7115931 DOI: 10.1038/s43018-020-0066-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/20/2020] [Indexed: 01/06/2023]
Abstract
Tumour mutational burden (TMB) predicts immunotherapy outcome in non-small cell lung cancer (NSCLC), consistent with immune recognition of tumour neoantigens. However, persistent antigen exposure is detrimental for T cell function. How TMB affects CD4 and CD8 T cell differentiation in untreated tumours, and whether this affects patient outcomes is unknown. Here we paired high-dimensional flow cytometry, exome, single-cell and bulk RNA sequencing from patients with resected, untreated NSCLC to examine these relationships. TMB was associated with compartment-wide T cell differentiation skewing, characterized by loss of TCF7-expressing progenitor-like CD4 T cells, and an increased abundance of dysfunctional CD8 and CD4 T cell subsets, with significant phenotypic and transcriptional similarity to neoantigen-reactive CD8 T cells. A gene signature of redistribution from progenitor-like to dysfunctional states associated with poor survival in lung and other cancer cohorts. Single-cell characterization of these populations informs potential strategies for therapeutic manipulation in NSCLC.
Collapse
Affiliation(s)
- Ehsan Ghorani
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - James L Reading
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| | - Jake Y Henry
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Marc Robert de Massy
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Rachel Rosenthal
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Virginia Turati
- Department of Cancer Biology, University College London Cancer Institute, London, UK
| | - Kroopa Joshi
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Andrew J S Furness
- Department of Medical Oncology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Assma Ben Aissa
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Sunil Kumar Saini
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Sofie Ramskov
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Andrew Georgiou
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Mariana Werner Sunderland
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Yien Ning Sophia Wong
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Maria Vila De Mucha
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - William Day
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Felipe Galvez-Cancino
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Pablo D Becker
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Imran Uddin
- Division of Infection and Immunity, University College London, London, UK
| | - Theres Oakes
- Division of Infection and Immunity, University College London, London, UK
| | - Mazlina Ismail
- Division of Infection and Immunity, University College London, London, UK
| | - Tahel Ronel
- Division of Infection and Immunity, University College London, London, UK
| | - Annemarie Woolston
- Division of Infection and Immunity, University College London, London, UK
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Nicolai J Birkbak
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Gareth A Wilson
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Kevin Litchfield
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Lucia Conde
- Bill Lyons Informatics Centre, University College London Cancer Institute, London, UK
| | | | - Kevin Blighe
- Bill Lyons Informatics Centre, University College London Cancer Institute, London, UK
| | - Dhruva Biswas
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | | | - Tom Lund
- Department of Medical Oncology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Maise Al Bakir
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - David A Moore
- Department of Pathology, University College London Cancer Institute, London, UK
| | - Crispin T Hiley
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Sherene Loi
- Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - Yuxin Sun
- Division of Infection and Immunity, University College London, London, UK
| | - Yinyin Yuan
- Department of Medical Oncology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Khalid AbdulJabbar
- Department of Medical Oncology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Samra Turajilic
- Department of Medical Oncology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Javier Herrero
- Bill Lyons Informatics Centre, University College London Cancer Institute, London, UK
| | - Tariq Enver
- Department of Cancer Biology, University College London Cancer Institute, London, UK
| | - Sine R Hadrup
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Allan Hackshaw
- Cancer Research UK and University College London Cancer Trials Centre, London, UK
| | - Karl S Peggs
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Benny Chain
- Division of Infection and Immunity, University College London, London, UK
- Department of Computer Sciences, University College London, London, UK
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- University College London Hospitals, London, UK.
| | - Sergio A Quezada
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| |
Collapse
|
86
|
Allocco JB, Alegre ML. Exploiting immunometabolism and T cell function for solid organ transplantation. Cell Immunol 2020; 351:104068. [PMID: 32139072 PMCID: PMC7150626 DOI: 10.1016/j.cellimm.2020.104068] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 12/25/2022]
Abstract
Cellular metabolism is central to T cell function and proliferation, with most of the research to date focusing on cancer and autoimmunity. Cellular metabolism is associated with a host of physiological phenomena, from epigenetic changes, to cellular function and fate. For the purpose of this review, we will discuss the metabolism of T cells relating to their differentiation and function. We will cover a variety of metabolic processes, ranging from glycolysis to amino acid metabolism. Understanding how T cell metabolism informs T cell function may be useful to understand alloimmune responses and design novel therapies to improve graft outcome.
Collapse
Affiliation(s)
- Jennifer B Allocco
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL 60637, United States
| | - Maria-Luisa Alegre
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL 60637, United States.
| |
Collapse
|
87
|
Update on the cellular and molecular aspects of lupus nephritis. Clin Immunol 2020; 216:108445. [PMID: 32344016 DOI: 10.1016/j.clim.2020.108445] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/26/2020] [Accepted: 04/23/2020] [Indexed: 12/17/2022]
Abstract
Recent progress has highlighted the involvement of a variety of innate and adaptive immune cells in lupus nephritis. These include activated neutrophils producing extracellular chromatin traps that induce type I interferon production and endothelial injury, metabolically-rewired IL-17-producing T-cells causing tissue inflammation, follicular and extra-follicular helper T-cells promoting the maturation of autoantibody-producing B-cells that may also sustain the formation of germinal centers, and alternatively activated monocytes/macrophages participating in tissue repair and remodeling. The role of resident cells such as podocytes and tubular epithelial cells is increasingly recognized in regulating the local immune responses and determining the kidney function and integrity. These findings are corroborated by advanced, high-throughput genomic studies, which have revealed an unprecedented amount of data highlighting the molecular heterogeneity of immune and non-immune cells implicated in lupus kidney disease. Importantly, this research has led to the discovery of putative pathogenic pathways, enabling the rationale design of novel treatments.
Collapse
|
88
|
Teng X, Brown J, Choi SC, Li W, Morel L. Metabolic determinants of lupus pathogenesis. Immunol Rev 2020; 295:167-186. [PMID: 32162304 DOI: 10.1111/imr.12847] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/20/2020] [Accepted: 02/24/2020] [Indexed: 12/12/2022]
Abstract
The metabolism of healthy murine and more recently human immune cells has been investigated with an increasing amount of details. These studies have revealed the challenges presented by immune cells to respond rapidly to a wide variety of triggers by adjusting the amount, type, and utilization of the nutrients they import. A concept has emerged that cellular metabolic programs regulate the size of the immune response and the plasticity of its effector functions. This has generated a lot of enthusiasm with the prediction that cellular metabolism could be manipulated to either enhance or limit an immune response. In support of this hypothesis, studies in animal models as well as human subjects have shown that the dysregulation of the immune system in autoimmune diseases is associated with a skewing of the immunometabolic programs. These studies have been mostly conducted on autoimmune CD4+ T cells, with the metabolism of other immune cells in autoimmune settings still being understudied. Here we discuss systemic metabolism as well as cellular immunometabolism as novel tools to decipher fundamental mechanisms of autoimmunity. We review the contribution of each major metabolic pathway to autoimmune diseases, with a focus on systemic lupus erythematosus (SLE), with the relevant translational opportunities, existing or predicted from results obtained with healthy immune cells. Finally, we review how targeting metabolic programs may present novel therapeutic venues.
Collapse
Affiliation(s)
- Xiangyu Teng
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Josephine Brown
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Seung-Chul Choi
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Wei Li
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
89
|
Zhou M, Guo C, Li X, Huang Y, Li M, Zhang T, Zhao S, Wang S, Zhang H, Yang N. JAK/STAT signaling controls the fate of CD8 +CD103 + tissue-resident memory T cell in lupus nephritis. J Autoimmun 2020; 109:102424. [PMID: 32085893 DOI: 10.1016/j.jaut.2020.102424] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/28/2020] [Accepted: 02/04/2020] [Indexed: 12/21/2022]
Abstract
Autoimmune mediated inflammation and renal damage in lupus nephritis (LN) depends partly on the infiltration of lymphocytes in glomeruli and renal interstitium. Here we identified a population of CD8+ T cells with a CD103+-phenotype in the healthy kidneys of human and mouse. These cells were typically CD69+CD103+ tissue-resident memory T cells (TRM) in the kidney. CD8+ TRM cells were expanded in the kidneys of patients with LN or MRL/lpr mice. The expansion of renal CD8+ TRM cells correlated significantly with kidney disease activity. These cells were active in producing cytokines, perforin and granzyme B in the kidney of MRL/lpr mice. Importantly, renal CD8+ TRM cells underwent proliferation and self-renewal to maintain a stable TRM pool in the kidney of MRL/lpr mice, contributing to renal inflammation and damage. JAK/STAT signaling in the MRL/lpr mice was required for renal TRM self-renewal as well as maintenance of effector functions. Targeting JAK/STAT signaling by tofacitinib effectively suppressed effector functions and impaired the survival of renal TRM cells in the kidney, contributing to improved kidney function in MRL/lpr mice. These results provided evidences that renal CD8+ TRM cells play a role in the pathogenesis of LN. They could serve as a therapeutic target for LN.
Collapse
Affiliation(s)
- Mianjing Zhou
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chaohuan Guo
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xue Li
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuefang Huang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mengyuan Li
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Tengyue Zhang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Siyuan Zhao
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuang Wang
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hui Zhang
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Niansheng Yang
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
90
|
Del Bello A, Kamar N, Treiner E. T cell reconstitution after lymphocyte depletion features a different pattern of inhibitory receptor expression in ABO- versus HLA-incompatible kidney transplant recipients. Clin Exp Immunol 2019; 200:89-104. [PMID: 31869432 DOI: 10.1111/cei.13412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
Chronic antigen stimulation can lead to immune exhaustion (a state of T cell dysfunction). Several phenotypical signatures of T cell exhaustion have been described in various pathological situations, characterized by aberrant expression of multiple inhibitory receptors (IR). This signature has been barely studied in the context of allogenic organ transplantation. We undertook a cross-sectional analysis of the expression of IR [CD244, CD279, T cell immunoreceptor with immunoglobulin (Ig) and immunoreceptor tyrosine-based inhibition motif (ITIM) domains (TIGIT) and CD57] and their correlation with cytokine-producing functions in T cells reconstituting after lymphocyte depletion in patients transplanted from living donors, with preformed donor-specific antibodies. After ABO incompatible transplantation, T cells progressively acquired a phenotype similar to healthy donors and the expression of several IR marked cells with increased functions, with the exception of TIGIT, which was associated with decreased cytokine production. In stark contrast, T cell reconstitution in patients with anti-human leukocyte antigen (HLA) antibodies was characterized with an increased co-expression of IR by T cells, and specifically by an increased expression of TIGIT. Furthermore, expression of these receptors was no longer directly correlated to cytokine production. These results suggest that T cell alloreactivity in HLA-incompatible kidney transplantation drives an aberrant T cell reconstitution with respect to IR profile, which could have an impact on the transplantation outcome.
Collapse
Affiliation(s)
- A Del Bello
- Nephrology and Organ Transplant Department, CHU de Toulouse, Toulouse, France.,Université Paul Sabatier Toulouse III, Toulouse, France.,Centre de Physiopathologie de Toulouse-Purpan (CPTP), Toulouse, France
| | - N Kamar
- Nephrology and Organ Transplant Department, CHU de Toulouse, Toulouse, France.,Université Paul Sabatier Toulouse III, Toulouse, France.,Centre de Physiopathologie de Toulouse-Purpan (CPTP), Toulouse, France
| | - E Treiner
- Université Paul Sabatier Toulouse III, Toulouse, France.,Centre de Physiopathologie de Toulouse-Purpan (CPTP), Toulouse, France.,Laboratory of Immunology, Biology Department, CHU de Toulouse, Toulouse, France
| |
Collapse
|
91
|
Rao DA, Arazi A, Wofsy D, Diamond B. Design and application of single-cell RNA sequencing to study kidney immune cells in lupus nephritis. Nat Rev Nephrol 2019; 16:238-250. [PMID: 31853010 DOI: 10.1038/s41581-019-0232-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2019] [Indexed: 11/09/2022]
Abstract
The immune mechanisms that cause tissue injury in lupus nephritis have been challenging to define. The advent of high-dimensional cellular analyses, such as single-cell RNA sequencing, has enabled detailed characterization of the cell populations present in small biopsy samples of kidney tissue. In parallel, the development of methods that cryopreserve kidney biopsy specimens in a manner that preserves intact, viable cells, has enabled the uniform analysis of tissue samples collected at multiple sites and across many geographic areas and demographic cohorts with high-dimensional platforms. The application of these methods to kidney biopsy samples from patients with lupus nephritis has begun to define the phenotypes of both infiltrating and resident immune cells, as well as parenchymal cells, present in nephritic kidneys. The detection of similar immune cell populations in urine suggests that it might be possible to non-invasively monitor immune activation in kidneys. Once applied to large patient cohorts, these high-dimensional studies might enable patient stratification according to patterns of immune cell activation in the kidney or identify disease features that can be used as surrogate measures of efficacy in clinical trials. Applied broadly across multiple inflammatory kidney diseases, these studies promise to enormously expand our understanding of renal inflammation in the next decade.
Collapse
Affiliation(s)
- Deepak A Rao
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Arnon Arazi
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David Wofsy
- Rheumatology Division and Russell/Engleman Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Betty Diamond
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA.
| |
Collapse
|
92
|
Wang H, Lu M, Zhai S, Wu K, Peng L, Yang J, Xia Y. ALW peptide ameliorates lupus nephritis in MRL/lpr mice. Arthritis Res Ther 2019; 21:261. [PMID: 31791413 PMCID: PMC6889545 DOI: 10.1186/s13075-019-2038-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023] Open
Abstract
Background Lupus nephritis (LN) is a common and serious complication of systemic lupus erythematosus. Anti-double-stranded (ds) DNA immunoglobulin G (IgG) plays a pivotal role in the pathogenesis of LN. Currently, there are various therapies for patients with LN; however, most of them are associated with considerable side effects. We confirmed previously that ALW (ALWPPNLHAWVP), a 12-amino acid peptide, inhibited the binding of polyclonal anti-dsDNA antibodies to mesangial cells and isolated glomeruli in vitro. In this study, we further investigate whether the administration of ALW peptide decreases renal IgG deposition and relevant damage in MRL/lpr lupus-prone mice. Methods Forty female MRL/lpr mice were randomly divided into four groups. The mice were intravenously injected with D-form ALW peptide (ALW group), scrambled peptide (PLP group), and normal saline (NaCl group) or were not treated (blank group). The IgG deposition, the histopathologic changes, and the expressions of profibrotic factors were analyzed in the kidney of MRL/lpr mice. Results Compared with the other groups, glomerular deposition of IgG, IgG2a, IgG2b, and IgG3 was decreased in the ALW group. Moreover, ALW administration attenuated renal histopathologic changes in MRL/lpr mice, including mesangial proliferation and infiltration of inflammatory cells. Furthermore, the expressions of profibrotic cytokines, such as transforming growth factor-beta1 (TGF-β1) and platelet-derived growth factor B (PDGF-B), decreased in the serum and kidney tissue of ALW-treated mice. Conclusions Our study demonstrated that ALW peptide ameliorates the murine model of LN, possibly through inhibiting renal IgG deposition and relevant tissue inflammation and fibrosis.
Collapse
Affiliation(s)
- Huixia Wang
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Mei Lu
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Siyue Zhai
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Kunyi Wu
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Lingling Peng
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jie Yang
- Department of Nephrology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
93
|
Abstract
PURPOSE OF REVIEW The management of lupus nephritis remains unsatisfactory due to insufficiently effective treatment regimens and the dearth of reliable predictors of disease onset or progression to guide individualized therapeutic decisions. This review summarizes new findings related to lupus nephritis over the last 18 months and discusses clinical needs that should be considered to advance trials of mechanism-based therapeutic strategies. RECENT FINDINGS Collaborative teams are addressing how to improve disease definitions and are developing predictive models for disease onset, disease response and risk of flare in individual patients. More attention is being paid to clinical trial design. Advanced technologic approaches are allowing the analysis of small amounts of human tissue and urine in unprecedented detail so as to discover new pathogenic mechanisms and identify disease biomarkers. Novel therapies continue to be tested in disease models and include new strategies to protect renal tissue from cell damage and fibrosis. SUMMARY The collaborative efforts of patients, clinical and translational researchers, the pharmaceutical industry and funding sources are needed to advance therapies for lupus nephritis. Specialized clinical centers can then deliver optimal and more personalized patient care that will improve patient outcomes.
Collapse
Affiliation(s)
- Anne Davidson
- Center for Autoimmunity, Musculoskeletal and Hematologic Diseases, Feinstein Institute for Medical Research, New York, New York, USA
| | | | | |
Collapse
|
94
|
Teng X, Cornaby C, Li W, Morel L. Metabolic regulation of pathogenic autoimmunity: therapeutic targeting. Curr Opin Immunol 2019; 61:10-16. [PMID: 31422315 DOI: 10.1016/j.coi.2019.07.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/05/2019] [Accepted: 07/14/2019] [Indexed: 12/12/2022]
Abstract
Metabolism has recently emerged as an essential mechanism that regulates the immune system. Studies in healthy mice and, to a lesser extent, humans, have defined the metabolism of immune cells in response to various challenges. It is increasingly recognized that the overactive immune system that drives autoimmune diseases presents metabolic abnormalities that offer therapeutic opportunities. These novel therapeutic venues are supported by a few studies using metabolic inhibitors in mouse models and in small clinical trials. Reaching the full potential of targeting immuno-metabolism in autoimmune diseases requires a systemic cell-specific characterization of metabolic pathways in mouse models and cells from patients. Here, we review recent reports of immuno-metabolic alterations in autoimmune diseases, as well as alterations in immune effector pathways that have been implicated in autoimmunity, with a focus on systemic lupus erythematosus.
Collapse
Affiliation(s)
- Xiangyu Teng
- Department of Pathology, Immunology, and Laboratory Medicine, 1395 Center Drive, University of Florida, Gainesville, FL, 32610, USA
| | - Caleb Cornaby
- Department of Pathology, Immunology, and Laboratory Medicine, 1395 Center Drive, University of Florida, Gainesville, FL, 32610, USA
| | - Wei Li
- Department of Pathology, Immunology, and Laboratory Medicine, 1395 Center Drive, University of Florida, Gainesville, FL, 32610, USA
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, 1395 Center Drive, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
95
|
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, 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, Diamond B. The immune cell landscape in kidneys of patients with lupus nephritis. Nat Immunol 2019; 20:902-914. [PMID: 31209404 PMCID: PMC6726437 DOI: 10.1038/s41590-019-0398-x] [Citation(s) in RCA: 454] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 04/05/2019] [Indexed: 02/07/2023]
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.
Collapse
Affiliation(s)
- Arnon Arazi
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Deepak A Rao
- Division of Rheumatology, Immunology, Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Celine C Berthier
- Internal Medicine, Department of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Anne Davidson
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Yanyan Liu
- Division of Rheumatology, Immunology, Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul J Hoover
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Adam Chicoine
- Division of Rheumatology, Immunology, Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - A Helena Jonsson
- Division of Rheumatology, Immunology, Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shuqiang Li
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David J Lieb
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Fan Zhang
- Division of Rheumatology, Immunology, Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kamil Slowikowski
- Division of Rheumatology, Immunology, Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Edward P Browne
- UNC HIV Cure Center and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Akiko Noma
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Dawn E Smilek
- Lupus Nephritis Trials Network, University of California San Francisco, San Francisco, CA, USA
- Immune Tolerance Network, University of California San Francisco, San Francisco, CA, USA
| | - Patti Tosta
- Lupus Nephritis Trials Network, University of California San Francisco, San Francisco, CA, USA
- Immune Tolerance Network, University of California San Francisco, San Francisco, CA, USA
| | - William Apruzzese
- Division of Rheumatology, Immunology, Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Elena Massarotti
- Division of Rheumatology, Immunology, Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria Dall'Era
- Rheumatology Division, University of California San Francisco, San Francisco, CA, USA
| | - Meyeon Park
- Division of Nephrology, University of California San Francisco, San Francisco, CA, USA
| | - Diane L Kamen
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Richard A Furie
- Division of Rheumatology, Northwell Health, Great Neck, NY, USA
| | - Fernanda Payan-Schober
- Department of Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | | | - Elizabeth A McInnis
- University of North Carolina Kidney Center, Division of Nephrology and Hypertension, Department of Medicine, UNC School of Medicine, Chapel Hill, NC, USA
| | - Jill P Buyon
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, NY, USA
| | - Michelle A Petri
- Division of Rheumatology, Johns Hopkins University, Baltimore, MD, USA
| | - Chaim Putterman
- Division of Rheumatology and Department of Microbiology and Immunology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Kenneth C Kalunian
- University of California San Diego School of Medicine, La Jolla, CA, USA
| | - E Steve Woodle
- Division of Transplantation, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - James A Lederer
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David A Hildeman
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Soumya Raychaudhuri
- Division of Rheumatology, Immunology, Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthias Kretzler
- Internal Medicine, Department of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer H Anolik
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Michael B Brenner
- Division of Rheumatology, Immunology, Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David Wofsy
- Rheumatology Division, University of California San Francisco, San Francisco, CA, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Betty Diamond
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA.
| |
Collapse
|
96
|
Yang L, Gong NR, Zhang Q, Ma YB, Zhou H. Apparent Correlations Between AMPK Expression and Brain Inflammatory Response and Neurological Function Factors in Rats with Chronic Renal Failure. J Mol Neurosci 2019; 68:204-213. [PMID: 30919248 DOI: 10.1007/s12031-019-01299-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/12/2019] [Indexed: 01/23/2023]
Abstract
To explore the correlations between AMP-activated protein kinase (AMPK) expression and brain inflammatory response and neurological function factors in rats with chronic renal failure. Chronic renal failure models in rats were established, and the healthy control group (normal group) was set. Chronic renal failure model rats were divided into model group (without any treatment), control group (intraperitoneal injection of normal saline), A-769662 group (intraperitoneal injection of AMPK specific activator), and compound C group (intraperitoneal injection of AMPK specific inhibitor). The results of HE staining showed renal tissue enlargement, and significant pathological changes. Compared with the normal group, AMPK level in peripheral blood and AMPK mRNA and protein expressions in brain tissue were significantly reduced, and AMPK pathway activation was significantly inhibited in other groups. Compared with the model group, rats in the A-769662 group had significantly decreased serum creatinine (Scr) and blood urea nitrogen (BUN) levels and γ-aminobutyric acid (γ-GABA) content, significantly increased brain-derived neurotrophic factor (BDNF) positive expressions and 5-hydroxytryptamine (5-HT) content, and decreased interleukin-1 (IL-1), tumor necrosis factor-α (TNF-α), and intercellular adhesion molecule 1 (ICAM-1) expressions (all P < 0.05), while it was just the opposite in compound C group (all P < 0.05). There is an apparent correlation between AMPK expression and brain inflammatory response in chronic renal failure rats. AMPK is expected to be an important pathway in the treatment of uremic encephalopathy.
Collapse
Affiliation(s)
- Li Yang
- Department of Nephrology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong Province, China
| | - Ni-Rong Gong
- Department of Nephrology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong Province, China
| | - Qin Zhang
- Department of Nephrology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong Province, China
| | - Ya-Bin Ma
- Department of Neurosurgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19 Nonglin Xia Road, Yuexiu District, Guangzhou, 510080, Guangdong Province, China
| | - Hui Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19 Nonglin Xia Road, Yuexiu District, Guangzhou, 510080, Guangdong Province, China.
| |
Collapse
|
97
|
Mijnheer G, van Wijk F. T-Cell Compartmentalization and Functional Adaptation in Autoimmune Inflammation: Lessons From Pediatric Rheumatic Diseases. Front Immunol 2019; 10:940. [PMID: 31143175 PMCID: PMC6520654 DOI: 10.3389/fimmu.2019.00940] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/11/2019] [Indexed: 12/30/2022] Open
Abstract
Chronic inflammatory diseases are characterized by a disturbed immune balance leading to recurring episodes of inflammation in specific target tissues, such as the joints in juvenile idiopathic arthritis. The tissue becomes infiltrated by multiple types of immune cell, including high numbers of CD4 and CD8 T-cells, which are mostly effector memory cells. Locally, these T-cells display an environment-adapted phenotype, induced by inflammation- and tissue-specific instructions. Some of the infiltrated T-cells may become tissue resident and play a role in relapses of inflammation. Adaptation to the environment may lead to functional (re)programming of cells and altered cellular interactions and responses. For example, specifically at the site of inflammation both CD4 and CD8 T-cells can become resistant to regulatory T-cell-mediated regulation. In addition, CD8 and CD4 T-cells show a unique profile with pro- and anti-inflammatory features coexisting in the same compartment. Also regulatory T-cells are neither homogeneous nor static in nature and show features of functional differentiation, and plasticity in inflammatory environments. Here we will discuss the recent insights in T-cell functional specialization, regulation, and clonal expansion in local (tissue) inflammation.
Collapse
Affiliation(s)
- Gerdien Mijnheer
- Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Femke van Wijk
- Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
98
|
Teng X, Li W, Cornaby C, Morel L. Immune cell metabolism in autoimmunity. Clin Exp Immunol 2019; 197:181-192. [PMID: 30770544 DOI: 10.1111/cei.13277] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2019] [Indexed: 12/13/2022] Open
Abstract
Immune metabolism is a rapidly moving field. While most of the research has been conducted to define the metabolism of healthy immune cells in the mouse, it is recognized that the overactive immune system that drives autoimmune diseases presents metabolic abnormalities that provide therapeutic opportunities, as well as a means to understand the fundamental mechanisms of autoimmune activation more clearly. Here, we review recent publications that have reported how the major metabolic pathways are affected in autoimmune diseases, with a focus on rheumatic diseases.
Collapse
Affiliation(s)
- X Teng
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - W Li
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - C Cornaby
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - L Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
99
|
Alperin JM, Ortiz-Fernández L, Sawalha AH. Monogenic Lupus: A Developing Paradigm of Disease. Front Immunol 2018; 9:2496. [PMID: 30459768 PMCID: PMC6232876 DOI: 10.3389/fimmu.2018.02496] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 10/09/2018] [Indexed: 12/13/2022] Open
Abstract
Monogenic lupus is a form of systemic lupus erythematosus (SLE) that occurs in patients with a single gene defect. This rare variant of lupus generally presents with early onset severe disease, especially affecting the kidneys and central nervous system. To date, a significant number of genes have been implicated in monogenic lupus, providing valuable insights into a very complex disease process. Throughout this review, we will summarize the genes reported to be associated with monogenic lupus or lupus-like diseases, and the pathogenic mechanisms affected by the mutations involved upon inducing autoimmunity.
Collapse
Affiliation(s)
- Jessie M Alperin
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Lourdes Ortiz-Fernández
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Amr H Sawalha
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States.,Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|