Publications

2022

Chen, P. M., Katsuyama, E., Satyam, A., Li, H., Rubio, J., Jung, S., Andrzejewski, S., Becherer, J. D., Tsokos, M. G., Abdi, R., & Tsokos, G. C. (2022). CD38 reduces mitochondrial fitness and cytotoxic T cell response against viral infection in lupus patients by suppressing mitophagy. Sci Adv, 8, Article 24. https://doi.org/10.1126/sciadv.abo4271eabo4271abo4271 [pii]
Infection is one of the major causes of mortality in patients with systemic lupus erythematosus (SLE). We previously found that CD38, an ectoenzyme that regulates the production of NAD(+), is up-regulated in CD8(+) T cells of SLE patients and correlates with the risk of infection. Here, we report that CD38 reduces CD8(+) T cell function by negatively affecting mitochondrial fitness through the inhibition of multiple steps of mitophagy, a process that is critical for mitochondria quality control. Using a murine lupus model, we found that administration of a CD38 inhibitor in a CD8(+) T cell-targeted manner reinvigorated their effector function, reversed the defects in autophagy and mitochondria, and improved viral clearance. We conclude that CD38 represents a target to mitigate infection rates in people with SLE.
Chen, P. M., & Tsokos, G. C. (2022). Mitochondria in the Pathogenesis of Systemic Lupus Erythematosus. Curr Rheumatol Rep, 24, Article 4. https://doi.org/10.1007/s11926-022-01063-910.1007/s11926-022-01063-9 [pii]
PURPOSE OF REVIEW: Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by autoantibody production and inflammation in multiple organs. In this article, we present data on how various mitochondria pathologies are involved in the pathogenesis of the disease including the fact that they serve as a reservoir of autoantigens which contribute to the upending of lymphocyte tolerance. RECENT FINDINGS: Mitochondrial DNA from various cell sources, including neutrophil extracellular traps, platelets, and red blood cells, elicits the production of type I interferon which contributes to breaking of peripheral tolerance. Mitochondrial DNA also serves as autoantigen targeted by autoantibodies. Mutations of mitochondrial DNA triggered by reactive oxygen species induce T cell cross-reactivity against self-antigens. Selective gene polymorphisms that regulate mitochondrial apoptosis in autoreactive B and T cells represent another key aspect in the induction of autoimmunity. Various mitochondrial abnormalities, including changes in mitochondrial function, oxidative stress, genetic polymorphism, mitochondrial DNA mutations, and apoptosis pathways, are each linked to different aspects of lupus pathogenesis. However, whether targeting these mitochondrial pathologies can be used to harness autoimmunity remains to be explored.
Dai, L., Uehara, M., Li, X., LaBarre, B. A., Banouni, N., Ichimura, T., Lee-Sundlov, M. M., Kasinath, V., Sullivan, J. A., Ni, H., Barone, F., Giannini, S., Bahmani, B., Sage, P. T., Patsopoulos, N. A., Tsokos, G. C., Bromberg, J. S., Hoffmeister, K., Jiang, L., & Abdi, R. (2022). Characterization of CD41(+) cells in the lymph node. Front Immunol, 13, 801945. https://doi.org/10.3389/fimmu.2022.801945801945
Lymph nodes (LNs) are the critical sites of immunity, and the stromal cells of LNs are crucial to their function. Our understanding of the stromal compartment of the LN has deepened recently with the characterization of nontraditional stromal cells. CD41 (integrin alphaIIb) is known to be expressed by platelets and hematolymphoid cells. We identified two distinct populations of CD41(+)Lyve1(+) and CD41(+)Lyve1(-) cells in the LNs. CD41(+)Lyve1(-) cells appear in the LN mostly at the later stages of the lives of mice. We identified CD41(+) cells in human LNs as well. We demonstrated that murine CD41(+) cells express mesodermal markers, such as Sca-1, CD105 and CD29, but lack platelet markers. We did not observe the presence of platelets around the HEVs or within proximity to fibroblastic reticular cells of the LN. Examination of thoracic duct lymph fluid showed the presence of CD41(+)Lyve1(-) cells, suggesting that these cells recirculate throughout the body. FTY720 reduced their trafficking to lymph fluid, suggesting that their egress is controlled by the S1P1 pathway. CD41(+)Lyve1(-) cells of the LNs were sensitive to radiation, suggestive of their replicative nature. Single cell RNA sequencing data showed that the CD41(+) cell population in naive mouse LNs expressed largely stromal cell markers. Further studies are required to examine more deeply the role of CD41(+) cells in the function of LNs.
Halasi, M., Nyska, A., Rubin, L., Tal, Y., Tsokos, G. C., & Adini, I. (2022). Melanocyte-secreted fibromodulin constrains skin inflammation in mice injected with lupus serum. Clin Immunol, 241, 109055. https://doi.org/S1521-6616(22)00136-X [pii]10.1016/j.clim.2022.109055
Skin pigmentation has been linked to the development, prevalence, and severity of several immune-mediated diseases such as SLE. Here, we asked whether fibromodulin (FMOD), which is highly expressed in skin with light complexion, can explain the known differences in the magnitude of inflammation. C57 mice with different levels of pigmentation and FMOD were injected with human lupus serum to induce skin inflammation. Histopathologic studies revealed that black C57 FMOD+/+ that produce low levels of FMOD and white C57 FMOD -/- mice develop more severe inflammation compared with white FMOD +/+ mice. This study also revealed that dark pigmentation and FMOD deletion correlates with the increased numbers of Langerhans cells. Altogether, we identify low pigmentation and FMOD are linked to low severity of inflammation and approaches to promote FMOD expression should offer clinical benefit.
Hisada, R., Yoshida, N., Orite, S. Y. K., Umeda, M., Burbano, C., Scherlinger, M., Kono, M., Krishfield, S., & Tsokos, G. C. (2022). Role of Glutaminase 2 in Promoting CD4+ T Cell Production of Interleukin-2 by Supporting Antioxidant Defense in Systemic Lupus Erythematosus. Arthritis Rheumatol, 74, Article 7. https://doi.org/10.1002/art.42112
OBJECTIVE: Glutaminase (GLS) isoenzymes GLS1 and GLS2 catalyze the first step of glutaminolysis. GLS1 is requisite for Th17 cell differentiation, and its inhibition suppresses autoimmune disease in animals, but the function of GLS2 is not known. The aim of this study was to investigate the role of GLS2 in CD4+ T cell function and systemic lupus erythematosus (SLE) pathogenesis. METHODS: We measured reactive oxygen species (ROS) levels, lipid peroxidation, and mitochondrial mass and polarization by flow cytometry, interleukin-2 (IL-2) production by a dual luciferase assay, and CpG DNA methylation of Il2 by a real-time polymerase chain reaction system. The impact of the overexpression of wild-type GLS1, wild-type GLS2, or mutated GLS2 at the PDZ domain-binding motif in CD4+ T cells was examined. Furthermore, GLS2 expression in CD4+ T cells from lupus-prone mice and patients with SLE was analyzed by Western blotting. RESULTS: GLS2, but not GLS1, reduced ROS levels and lipid peroxidation and restored mitochondrial function in T cells. GLS2 promoted IL-2 production through the demethylation of the Il2 promoter. Mutation of the PDZ domain-binding motif abated the ability of GLS2 to regulate IL-2 and ROS levels. In lupus-prone mice and patients with SLE, the expression of GLS2 was decreased in CD4+ T cells. Finally, GLS2 overexpression corrected ROS levels and restored IL-2 production by CD4+ T cells from lupus-prone mice and SLE patients. CONCLUSION: Our findings suggest that GLS2 has a crucial role in IL-2 production by CD4+ T cells by supporting antioxidant defense, and they offer a new approach to correcting IL-2 production by T cells in SLE.
Hisada, R., Yoshida, N., Umeda, M., Burbano, C., Bhargava, R., Scherlinger, M., Kono, M., Kyttaris, V. C., Krishfield, S., & Tsokos, G. C. (2022). The deacetylase SIRT2 contributes to autoimmune disease pathogenesis by modulating IL-17A and IL-2 transcription. Cell Mol Immunol, 19, Article 6. https://doi.org/10.1038/s41423-022-00874-110.1038/s41423-022-00874-1 [pii]874 [pii]
Aberrant IL-17A expression together with reduced IL-2 production by effector CD4(+) T cells contributes to the pathogenesis of systemic lupus erythematosus (SLE). Here, we report that Sirtuin 2 (SIRT2), a member of the family of NAD(+)-dependent histone deacetylases, suppresses IL-2 production by CD4(+) T cells while promoting their differentiation into Th17 cells. Mechanistically, we show that SIRT2 is responsible for the deacetylation of p70S6K, activation of the mTORC1/HIF-1alpha/RORgammat pathway and induction of Th17-cell differentiation. Additionally, SIRT2 was shown to be responsible for the deacetylation of c-Jun and histones at the Il-2 gene, resulting in decreased IL-2 production. We found that the transcription factor inducible cAMP early repressor (ICER), which is overexpressed in T cells from people with SLE and lupus-prone mice, bound directly to the Sirt2 promoter and promoted its transcription. AK-7, a SIRT2 inhibitor, limited the ability of adoptively transferred antigen-specific CD4(+) T cells to cause autoimmune encephalomyelitis in mice and limited disease in lupus-prone MRL/lpr mice. Finally, CD4(+) T cells from SLE patients exhibited increased expression of SIRT2, and pharmacological inhibition of SIRT2 in primary CD4(+) T cells from patients with SLE attenuated the ability of these cells to differentiate into Th17 cells and promoted the generation of IL-2-producing T cells. Collectively, these results suggest that SIRT2-mediated deacetylation is essential in the aberrant expression of IL-17A and IL-2 and that SIRT2 may be a promising molecular target for new SLE therapies.
Huang, C., Yi, P., Zhu, M., Zhou, W., Zhang, B., Yi, X., Long, H., Zhang, G., Wu, H., Tsokos, G. C., Zhao, M., & Lu, Q. (2022). Safety and efficacy of fecal microbiota transplantation for treatment of systemic lupus erythematosus: An EXPLORER trial. J Autoimmun, 130, 102844. https://doi.org/S0896-8411(22)00052-X [pii]10.1016/j.jaut.2022.102844
Gut microbiota dysbiosis is involved in the development of systemic lupus erythematosus (SLE). The safety and efficacy of fecal microbiota transplantation (FMT) for the treatment of SLE patients has not been explored. In this 12-week, single-arm pilot clinical trial of oral encapsulated fecal microbiome from healthy donors to patients with active SLE, we aimed to evaluate the safety and efficacy of FMT in patients with SLE (ChiCTR2000036352). 20 SLE patients with SLEDAI >/=6 were recruited. FMT was administered once a week for three consecutive weeks along with standard treatment and the patients were followed for 12 weeks. Safety was evaluated throughout the trial. The primary endpoint was the SLE Responder Index-4 (SRI-4) at week 12. Microbiome composition, levels of short chain fatty acids (SCFAs) in the gut and of cytokines in the sera were measured along with lymphocyte phenotyping. No serious adverse events were observed after FMT. At week 12, the SRI-4 response rate was 42.12%, and significant reductions in the SLEDAI-2K scores and the level of serum anti-dsDNA antibody were observed compared to baseline. Significant enrichment of SCFAs-producing bacterial taxa and reduction of inflammation-related bacterial taxa were observed, along with increased production of SCFAs in the gut and reduced levels of IL-6 and CD4(+) memory/naive ratio in the peripheral blood. Furthermore, SRI-4 responding patients displayed specific microbiota signatures both before and after FMT. The first clinical trial of FMT in active SLE patients provide supportive evidence that FMT might be a feasible, safe, and potentially effective therapy in SLE patients by modifying the gut microbiome and its metabolic profile.
Koga, T., Ichinose, K., & Tsokos, G. C. (2022). Tissue resident cell processes determine organ damage in systemic lupus erythematosus. Clin Immunol, 234, 108919. https://doi.org/S1521-6616(21)00256-4 [pii]10.1016/j.clim.2021.108919
Systemic lupus erythematosus (SLE) is an autoimmune disease that affects almost any organ. Multiple immunological abnormalities involving every domain of the immune system contribute to the expression of the disease. It is now recognized that elements of the immune system instigate processes in tissue resident cells which execute organ damage. Although correction of ongoing immune aberrations is important in the control of disease activity, targeting tissue specific injurious processes may prove desirable in limiting organ damage.