Publications by Year: 2021

2021

Koga, T., Kawakami, A., & Tsokos, G. C. (2021). Current insights and future prospects for the pathogenesis and treatment for rheumatoid arthritis. Clin Immunol, 225, 108680. https://doi.org/S1521-6616(21)00017-6 [pii]10.1016/j.clim.2021.108680
Genetic, environmental, and epigenetic factors simultaneously or serially contribute to immune cells and resident joint tissue cell abnormalities, invariably leading to joint destruction. Understanding the immune cell dysfunction in earlier years has brought forward life-changing therapeutics to patients with rheumatoid arthritis (RA). Further advances in the understanding of the immune and joint tissue-resident cell signaling and metabolic defects should produce additional tools to treat people with RA and foretell those who will respond to each biological or small drug. This review presents the latest evidence on RA pathogenesis and outlines the prospects for achieving precision medicine.
Kolios, A. G. A., & Tsokos, G. C. (2021). Skin-kidney crosstalk in SLE. Nat Rev Rheumatol, 17, Article 5. https://doi.org/10.1038/s41584-021-00588-010.1038/s41584-021-00588-0 [pii]
Kolios, A. G. A., Tsokos, G. C., & Klatzmann, D. (2021). Interleukin-2 and regulatory T cells in rheumatic diseases. Nat Rev Rheumatol, 17, Article 12. https://doi.org/10.1038/s41584-021-00707-x10.1038/s41584-021-00707-x [pii]
Failure of regulatory T (T(reg)) cells to properly control immune responses leads invariably to autoimmunity and organ damage. Decreased numbers or impaired function of T(reg) cells, especially in the context of inflammation, has been documented in many human autoimmune diseases. Restoration of T(reg) cell fitness and/or expansion of their numbers using low-dose natural IL-2, the main cytokine driving T(reg) cell survival and function, has demonstrated clinical efficacy in early clinical trials. Genetically modified IL-2 with an extended half-life and increased selectivity for T(reg) cells is now in clinical development. Administration of IL-2 combined with therapies targeting other pathways involved in the expression of autoimmune diseases should further enhance its therapeutic potential. Ongoing clinical efforts that capitalize on the early clinical success of IL-2 treatment should bring the use of this cytokine to the forefront of biological treatments for autoimmune diseases.
Kolios, A. G. A., Yoshida, N., & Tsokos, G. C. (2021). New therapeutic approaches in systemic lupus erythematosus. Curr Opin Rheumatol, 33, Article 2. https://doi.org/10.1097/BOR.000000000000077200002281-202103000-00011 [pii]
PURPOSE OF REVIEW: This review gives an overview of the recently published clinical trials in systemic lupus erythematosus (SLE). RECENT FINDINGS: Our continuously improving understanding of the cellular and molecular mechanisms, which are involved in the pathogenesis of SLE, has inspired the performance of multiple clinical trials in an attempt to modify recognized targets. Here, we summarize results obtained from recent trials, which used monoclonal antibodies blocking cytokines, blockers of costimulatory molecules or deleting immune cells, small drug inhibitors of kinases and replenishment of cytokines. SUMMARY: The therapeutic options for patients with SLE grow continuously and in parallel it raises the need for pathogenetic mechanism-based precision medicine so that we may select the right treatment for the right patient.
Kono, M., Yoshida, N., & Tsokos, G. C. (2021). Amino Acid Metabolism in Lupus. Front Immunol, 12, 623844. https://doi.org/10.3389/fimmu.2021.623844623844
T cell metabolism is central to cell proliferation, survival, differentiation, and aberrations have been linked to the pathophysiology of systemic autoimmune diseases. Besides glycolysis and fatty acid oxidation/synthesis, amino acid metabolism is also crucial in T cell metabolism. It appears that each T cell subset favors a unique metabolic process and that metabolic reprogramming changes cell fate. Here, we review the mechanisms whereby amino acid transport and metabolism affects T cell activation, differentiation and function in T cells in the prototype systemic autoimmune disease systemic lupus erythematosus. New insights in amino acid handling by T cells should guide approaches to correct T cell abnormalities and disease pathology.
Li, H., & Tsokos, G. C. (2021). Double-negative T cells in autoimmune diseases. Curr Opin Rheumatol, 33, Article 2. https://doi.org/10.1097/BOR.000000000000077800002281-202103000-00009 [pii]
PURPOSE OF REVIEW: TCRalphabeta+CD4-CD8- double-negative T (DNT) cells, a principal subset of mature T lymphocytes, have been closely linked with autoimmune/inflammatory conditions. However, controversy persists regarding their ontogeny and function. Here, we present an overview on DNT cells in different autoimmune diseases to advance a deeper understanding of the contribution of this population to disease pathogenesis. RECENT FINDINGS: DNT cells have been characterized in various chronic inflammatory diseases and they have been proposed to display pathogenic or regulatory function. The tissue location of DNT cells and the effector cytokines they produce bespeak to their active involvement in chronic inflammatory diseases. SUMMARY: By producing various cytokines, expanded DNT cells in inflamed tissues contribute to the pathogenesis of a variety of autoimmune inflammatory diseases. However, it is unclear whether this population represents a stable lineage consisting of different subsets similar to CD4+ T helper cell subset. Better understanding of the possible heterogeneity and plasticity of DNT cells is needed to reveal interventional therapeutic opportunities.
Li, H., & Tsokos, G. C. (2021). IL-23/IL-17 Axis in Inflammatory Rheumatic Diseases. Clin Rev Allergy Immunol, 60, Article 1. https://doi.org/10.1007/s12016-020-08823-410.1007/s12016-020-08823-4 [pii]
In inflammatory rheumatic disorders, the immune system attacks and damages the connective tissues and invariably internal organs. During the past decade, remarkable advances having been made towards our understanding on the cellular and molecular mechanisms involved in rheumatic diseases. The discovery of IL-23/IL-17 axis and the delineation of its important role in the inflammation led to the introduction of many needed new therapeutic tools. We will present an overview of the rationale for targeting therapeutically the IL-23/IL-17 axis in rheumatic diseases and the clinical benefit which has been realized so far. Finally, we will discuss the complex interrelationship between IL-23 and IL-17 and the possible uncoupling in certain disease settings.
Li, H., Tsokos, M. G., Bhargava, R., Adamopoulos, I. E., Menn-Josephy, H., Stillman, I. E., Rosenstiel, P., Jordan, J., & Tsokos, G. C. (2021). IL-23 reshapes kidney resident cell metabolism and promotes local kidney inflammation. J Clin Invest, 131, Article 12. https://doi.org/142428 [pii]10.1172/JCI142428e142428
Interstitial kidney inflammation is present in various nephritides in which serum interleukin 23 (IL-23) is elevated. Here we showed that murine and human renal tubular epithelial cells (TECs) expressing the IL-23 receptor (IL-23R) responded to IL-23 by inducing intracellular calcium flux, enhancing glycolysis, and upregulating calcium/calmodulin kinase IV (CaMK4), which resulted in suppression of the expression of the arginine-degrading enzyme arginase 1 (ARG1), thus increasing in situ levels of free L-arginine. Limited availability of arginine suppressed the ability of infiltrating T cells to proliferate and produce inflammatory cytokines. TECs from humans and mice with nephritis expressed increased levels of IL-23R and CaMK4 but reduced levels of ARG1. TEC-specific deletion of Il23r or Camk4 suppressed inflammation, whereas deletion of Arg1 exacerbated inflammation in different murine disease models. Finally, TEC-specific delivery of a CaMK4 inhibitor specifically curbed renal inflammation in lupus-prone mice without affecting systemic inflammation. Our data offer the first evidence to our knowledge of the immunosuppressive capacity of TECs through a mechanism that involves competitive uptake of arginine and signify the importance of modulation of an inflammatory cytokine in the function of nonlymphoid cells, which leads to the establishment of an inflammatory microenvironment. New approaches to treat kidney inflammation should consider restoring the immunosuppressive capacity of TECs.
Li, P., Jiang, M., Li, K., Li, H., Zhou, Y., Xiao, X., Xu, Y., Krishfield, S., Lipsky, P. E., Tsokos, G. C., & Zhang, X. (2021). Glutathione peroxidase 4-regulated neutrophil ferroptosis induces systemic autoimmunity. Nat Immunol, 22, Article 9. https://doi.org/10.1038/s41590-021-00993-310.1038/s41590-021-00993-3 [pii]
The linkage between neutrophil death and the development of autoimmunity has not been thoroughly explored. Here, we show that neutrophils from either lupus-prone mice or patients with systemic lupus erythematosus (SLE) undergo ferroptosis. Mechanistically, autoantibodies and interferon-alpha present in the serum induce neutrophil ferroptosis through enhanced binding of the transcriptional repressor CREMalpha to the glutathione peroxidase 4 (Gpx4, the key ferroptosis regulator) promoter, which leads to suppressed expression of Gpx4 and subsequent elevation of lipid-reactive oxygen species. Moreover, the findings that mice with neutrophil-specific Gpx4 haploinsufficiency recapitulate key clinical features of human SLE, including autoantibodies, neutropenia, skin lesions and proteinuria, and that the treatment with a specific ferroptosis inhibitor significantly ameliorates disease severity in lupus-prone mice reveal the role of neutrophil ferroptosis in lupus pathogenesis. Together, our data demonstrate that neutrophil ferroptosis is an important driver of neutropenia in SLE and heavily contributes to disease manifestations.
Li, X., Zhao, J., Naini, S. M., Sabiu, G., Tullius, S. G., Shin, S. R., Bromberg, J. S., Fiorina, P., Tsokos, G. C., Abdi, R., & Kasinath, V. (2021). Kidney-Draining Lymph Node Fibrosis Following Unilateral Ureteral Obstruction. Front Immunol, 12, 768412. https://doi.org/10.3389/fimmu.2021.768412768412
Although the primary organ has been the subject of intense investigation in the field of organ fibrosis over the past several decades, the presence of lymph node fibrosis due to persistent activation of the immune response in its partner organ remains largely unknown. Previously, we demonstrated that activation of the immune response following ischemia-reperfusion injury (IRI) and crescentic glomerulonephritis (CGN) in the kidney was associated with extracellular matrix (ECM) production by fibroblastic reticular cells (FRCs) of the kidney-draining lymph node (KLN). Here, we sought to determine whether FRCs in the KLN become similarly fibrogenic following unilateral ureteral obstruction (UUO) of the kidney. We subjected 6-8-week-old C57BL/6J mice to UUO for 2, 7, and 14 days. We examined the microarchitecture of the kidney and KLN by immunofluorescence staining at each timepoint, and we quantified immune cell populations in the KLN by flow cytometry. The contralateral kidney unaffected by UUO and its partner KLN were used as controls. We found through immunofluorescence staining that FRCs increased production of ECM fibers and remodeled the microarchitecture of the UUO KLN, contributing to fibrosis that mirrored the changes in the kidney. We also observed by flow cytometry that the populations of CD11b(+) antigen-presenting cells, CD11c(+) dendritic cells, and activated CD4(+) and CD8(+) T cells were significantly higher in the UUO KLN than the KLN draining the unaffected contralateral kidney. Expression of the TGFbeta/TGFbetaR signaling pathway was upregulated and colocalized with FRCs in the UUO KLNs, suggesting a possible mechanism behind the fibrosis. Both release of ureteral ligation at 2 days following UUO and depletion of FRCs at the time of injury onset halted the progression of fibrosis in both the kidney and the KLN. These findings for the first time highlight the association between fibrosis both in the kidney and the KLN during UUO, and they lay the groundwork for future studies that will investigate more deeply the mechanisms behind the connection between FRCs and KLN fibrosis.