JAK-STAT signaling is activated in the kidney and peripheral blood cells of patients with focal segmental glomerulosclerosis

Jianling Tao, Laura Mariani, Sean Eddy, Holden Maecker, Neeraja Kambham, Kshama Mehta, John Hartman, Weiqi Wang, Matthias Kretzler, Richard A Lafayette, Jianling Tao, Laura Mariani, Sean Eddy, Holden Maecker, Neeraja Kambham, Kshama Mehta, John Hartman, Weiqi Wang, Matthias Kretzler, Richard A Lafayette

Abstract

Focal segmental glomerular sclerosis (FSGS) is a devastating disease with limited treatment options and poor prognosis. Activated JAK-STAT signaling has been implicated in other kidney diseases. Since new technologies allow us to better evaluate changes in systemic and renal JAK-STAT activity as it relates to kidney function, we examined this in 106 patients with biopsy-proven FSGS compared to 47 healthy control individuals. Peripheral immune function was assessed in peripheral blood mononuclear cells by phosphoflow studies before and after cytokine stimulation. Kidney JAK-STAT activity was measured by immunofluorescence and by transcriptomics. A STAT1 activity score was calculated by evaluating message status of downstream targets of pSTAT 1. Peripheral blood mononuclear cells were found to be upregulated in terms of pSTAT production at baseline in FSGS and to have limited reserve to respond to various cytokines. Increased staining for components of the JAK-STAT system in FSGS by microscopy was found. Furthermore, we found transcriptomic evidence for activation of JAK-STAT that increased pSTAT 1 and pSTAT 3 in glomerular and tubulointerstitial sections of the kidney. Some of these changes were associated with the likelihood of remission of proteinuria and progression of disease. JAK-STAT signaling is altered in patients with FSGS as compared to healthy controls with activated peripheral immune cells, increased message in the kidney and increased activated proteins in the kidney. Thus, our findings support immune activation in this disease and point to the JAK-STAT pathway as a potential target for treatment of FSGS.

Keywords: cell signaling; focal segmental glomerulosclerosis; gene expression; lymphocytes.

Copyright © 2018. Published by Elsevier Inc.

Figures

Figure 1.
Figure 1.
Work flow of FSGS JAK-STAT evaluation
Figure 2.
Figure 2.
Unstimulated phosphoflow. Baseline of pSTAT1 (2A), pSTAT3 (2B), and pSTAT5 (2C) in different cell subsets of B lymphocytes, CD4+, CD4+CD45RA+, CD4+CD45RA-, CD8+, CD8+CD45RA+, CD8+CD45RA-, NK cells, and monocytes in FSGS were compared with controls. * compared with control, p

Figure 3.

Changes of pSTAT1, and pSTAT3…

Figure 3.

Changes of pSTAT1, and pSTAT3 of B lymphocytes, CD4+, CD4+CD45RA+, CD4+CD45RA-, CD8+, CD8+CD45RA+,…

Figure 3.
Changes of pSTAT1, and pSTAT3 of B lymphocytes, CD4+, CD4+CD45RA+, CD4+CD45RA-, CD8+, CD8+CD45RA+, CD8+CD45RA-, NK cells, and monocytes stimulated by IFNα, IFNγ, IL-6, IL-7, IL-10, IL-2, and IL-21 were compared between FSGS and control. P values were determined by Wilcoxon rank sum test. P values lower than 0.05 were filtered and shown in a heatmap format (left panel). Direction of increased or decreased STAT phosphorylation in comparison to controls is represented by blue or red. Correlation analysis between 24 hour urine protein excretion, or serum creatinine with any fold change described above were conducted and the four significant correlations that overlapped with the significant change of pSTAT1 are shown in Figure 3A–D. Detailed raw data in change of pSTAT1, and pSTAT3 in any cell subset under any stimulant between control and FSGS were expressed as Mean±SEM in supplement file on request. P values from those comparisons are also provided in supplement file on request.

Figure 3.

Changes of pSTAT1, and pSTAT3…

Figure 3.

Changes of pSTAT1, and pSTAT3 of B lymphocytes, CD4+, CD4+CD45RA+, CD4+CD45RA-, CD8+, CD8+CD45RA+,…

Figure 3.
Changes of pSTAT1, and pSTAT3 of B lymphocytes, CD4+, CD4+CD45RA+, CD4+CD45RA-, CD8+, CD8+CD45RA+, CD8+CD45RA-, NK cells, and monocytes stimulated by IFNα, IFNγ, IL-6, IL-7, IL-10, IL-2, and IL-21 were compared between FSGS and control. P values were determined by Wilcoxon rank sum test. P values lower than 0.05 were filtered and shown in a heatmap format (left panel). Direction of increased or decreased STAT phosphorylation in comparison to controls is represented by blue or red. Correlation analysis between 24 hour urine protein excretion, or serum creatinine with any fold change described above were conducted and the four significant correlations that overlapped with the significant change of pSTAT1 are shown in Figure 3A–D. Detailed raw data in change of pSTAT1, and pSTAT3 in any cell subset under any stimulant between control and FSGS were expressed as Mean±SEM in supplement file on request. P values from those comparisons are also provided in supplement file on request.

Figure 4.

Representative images of IHC stain…

Figure 4.

Representative images of IHC stain of JAK1, JAK2, pSTAT1, STAT3 and pSTAT3 in…

Figure 4.
Representative images of IHC stain of JAK1, JAK2, pSTAT1, STAT3 and pSTAT3 in control kidney and FSGS patient. A: Control JAK1, B: Control JAK2, C: Control pSTAT1, D: Control STAT3, E: Control pSTAT3, F: FSGS JAK1, G: FSGS JAK2, H: FSGS pSTAT1, I: FSGS STAT3, J: FSGS pSTAT3. (200×) K: Control JAK1, L: Control JAK2, M: Control pSTAT1, N: Control STAT3, O: Control pSTAT3, P: FSGS JAK1, Q: FSGS JAK2, R: FSGS pSTAT1, S: FSGS STAT3, T: FSGS pSTAT3. (400×)

Figure 5.

Semi-quantification of the JAK2, pSTAT1,…

Figure 5.

Semi-quantification of the JAK2, pSTAT1, STAT3, and pSTAT3 immunohistochemistry staining in FSGS patients…

Figure 5.
Semi-quantification of the JAK2, pSTAT1, STAT3, and pSTAT3 immunohistochemistry staining in FSGS patients (n=9). Healthy non-neoplastic kidney dissected from tumor nephrectomy was used as control. For JAK2 (A), the scoring was used as 0: none; +1 75% cells positive with a strong intensity.

Figure 6.

Differentially expressed transcripts in FSGS…

Figure 6.

Differentially expressed transcripts in FSGS were enriched in the JAK-STAT pathway in the…

Figure 6.
Differentially expressed transcripts in FSGS were enriched in the JAK-STAT pathway in the tubulointerstitium. (A) Signaling network nodes highlighted in purple indicate those genes that were differentially expressed in FSGS relative to living donor controls, while shading of the nodes indicates up-regulation (red) or down-regulation (green) in FSGS relative to living donor controls. (B) Upstream regulator analysis identified a predicated activation of IFNG and STAT1 in the tubulointerstitium with evidence supporting activation of STAT1 indicated (C).

Figure 7.

Differentially expressed transcripts in FSGS…

Figure 7.

Differentially expressed transcripts in FSGS were enriched in the JAK-STAT pathway in the…

Figure 7.
Differentially expressed transcripts in FSGS were enriched in the JAK-STAT pathway in the glomeruli. (A) Signaling network nodes highlighted in purple indicate those genes that were differentially expressed in FSGS relative to living donor controls, while shading of the nodes indicates up-regulation (red) or down-regulation (green) in FSGS relative to living donor controls. (B) Predicated activation of STAT related signaling nodes in the glomeruli with evidence supporting activation of STAT1 indicated (C). Network supporting STAT1 activation in the glomeruli. Orange and blue connections indicate that STAT1 can induce or inhibit expression of downstream genes. These findings are consistent with STAT1 activation. Yellow connections indicate findings that are inconsistent with STAT1 activation, while grey lines are findings that neither support nor reject the predicated activation of STAT1.

Figure 8.

Dual IHC stain of pSTAT1…

Figure 8.

Dual IHC stain of pSTAT1 (red) and CD3 (grey) in 12 FSGS patient…

Figure 8.
Dual IHC stain of pSTAT1 (red) and CD3 (grey) in 12 FSGS patient from NEPTUNE cohort according to isolated glomerular genetic analysis results for JAK1, JAK2 or STAT1expression. A and G: two FSGS patients with low glomerular expression of JAK1; B and H: two FSGS patients with high glomerular expression of JAK1; C and I: two FSGS patients with low glomerular expression of JAK2; D and J: two FSGS patients with high glomerular expression of JAK2; E and K: two FSGS patients with low glomerular expression of STAT1; F and L: two FSGS patients with high glomerular expression of STAT1 (200×). A1 to L1 are corresponding images for the cases labeled as A to L (200×) under higher magnification (400×).

Figure 9.

Correlation of JAK-STAT1 activation score…

Figure 9.

Correlation of JAK-STAT1 activation score from glomerular compartment with baseline eGFR (panel A)…

Figure 9.
Correlation of JAK-STAT1 activation score from glomerular compartment with baseline eGFR (panel A) and baseline UPCR (panel B) and JAK-STAT activation score from tubular compartment with baseline eGFR (panel C) and baseline UPCR (panel D) in NEPTUNE patients.

Figure 10.

Adjusted Kaplan Meier survival curve…

Figure 10.

Adjusted Kaplan Meier survival curve for complete remission by tertile of JAK-STAT1 Activation…

Figure 10.
Adjusted Kaplan Meier survival curve for complete remission by tertile of JAK-STAT1 Activation z-score (Glomerular Compartment, panel A, p-value 0.14 and Tubular Compartment, panel B, p-value 0.08). Adjusted Kaplan Meier survival curve for Composite of 40% loss in eGFR and ESRD by tertile of JAK-STAT1 Activation z-score (Glomerular Compartment, panel C, p-value 0.41 and Tubular Compartment, panel D, p-value 0.02).
All figures (11)
Figure 3.
Figure 3.
Changes of pSTAT1, and pSTAT3 of B lymphocytes, CD4+, CD4+CD45RA+, CD4+CD45RA-, CD8+, CD8+CD45RA+, CD8+CD45RA-, NK cells, and monocytes stimulated by IFNα, IFNγ, IL-6, IL-7, IL-10, IL-2, and IL-21 were compared between FSGS and control. P values were determined by Wilcoxon rank sum test. P values lower than 0.05 were filtered and shown in a heatmap format (left panel). Direction of increased or decreased STAT phosphorylation in comparison to controls is represented by blue or red. Correlation analysis between 24 hour urine protein excretion, or serum creatinine with any fold change described above were conducted and the four significant correlations that overlapped with the significant change of pSTAT1 are shown in Figure 3A–D. Detailed raw data in change of pSTAT1, and pSTAT3 in any cell subset under any stimulant between control and FSGS were expressed as Mean±SEM in supplement file on request. P values from those comparisons are also provided in supplement file on request.
Figure 3.
Figure 3.
Changes of pSTAT1, and pSTAT3 of B lymphocytes, CD4+, CD4+CD45RA+, CD4+CD45RA-, CD8+, CD8+CD45RA+, CD8+CD45RA-, NK cells, and monocytes stimulated by IFNα, IFNγ, IL-6, IL-7, IL-10, IL-2, and IL-21 were compared between FSGS and control. P values were determined by Wilcoxon rank sum test. P values lower than 0.05 were filtered and shown in a heatmap format (left panel). Direction of increased or decreased STAT phosphorylation in comparison to controls is represented by blue or red. Correlation analysis between 24 hour urine protein excretion, or serum creatinine with any fold change described above were conducted and the four significant correlations that overlapped with the significant change of pSTAT1 are shown in Figure 3A–D. Detailed raw data in change of pSTAT1, and pSTAT3 in any cell subset under any stimulant between control and FSGS were expressed as Mean±SEM in supplement file on request. P values from those comparisons are also provided in supplement file on request.
Figure 4.
Figure 4.
Representative images of IHC stain of JAK1, JAK2, pSTAT1, STAT3 and pSTAT3 in control kidney and FSGS patient. A: Control JAK1, B: Control JAK2, C: Control pSTAT1, D: Control STAT3, E: Control pSTAT3, F: FSGS JAK1, G: FSGS JAK2, H: FSGS pSTAT1, I: FSGS STAT3, J: FSGS pSTAT3. (200×) K: Control JAK1, L: Control JAK2, M: Control pSTAT1, N: Control STAT3, O: Control pSTAT3, P: FSGS JAK1, Q: FSGS JAK2, R: FSGS pSTAT1, S: FSGS STAT3, T: FSGS pSTAT3. (400×)
Figure 5.
Figure 5.
Semi-quantification of the JAK2, pSTAT1, STAT3, and pSTAT3 immunohistochemistry staining in FSGS patients (n=9). Healthy non-neoplastic kidney dissected from tumor nephrectomy was used as control. For JAK2 (A), the scoring was used as 0: none; +1 75% cells positive with a strong intensity.
Figure 6.
Figure 6.
Differentially expressed transcripts in FSGS were enriched in the JAK-STAT pathway in the tubulointerstitium. (A) Signaling network nodes highlighted in purple indicate those genes that were differentially expressed in FSGS relative to living donor controls, while shading of the nodes indicates up-regulation (red) or down-regulation (green) in FSGS relative to living donor controls. (B) Upstream regulator analysis identified a predicated activation of IFNG and STAT1 in the tubulointerstitium with evidence supporting activation of STAT1 indicated (C).
Figure 7.
Figure 7.
Differentially expressed transcripts in FSGS were enriched in the JAK-STAT pathway in the glomeruli. (A) Signaling network nodes highlighted in purple indicate those genes that were differentially expressed in FSGS relative to living donor controls, while shading of the nodes indicates up-regulation (red) or down-regulation (green) in FSGS relative to living donor controls. (B) Predicated activation of STAT related signaling nodes in the glomeruli with evidence supporting activation of STAT1 indicated (C). Network supporting STAT1 activation in the glomeruli. Orange and blue connections indicate that STAT1 can induce or inhibit expression of downstream genes. These findings are consistent with STAT1 activation. Yellow connections indicate findings that are inconsistent with STAT1 activation, while grey lines are findings that neither support nor reject the predicated activation of STAT1.
Figure 8.
Figure 8.
Dual IHC stain of pSTAT1 (red) and CD3 (grey) in 12 FSGS patient from NEPTUNE cohort according to isolated glomerular genetic analysis results for JAK1, JAK2 or STAT1expression. A and G: two FSGS patients with low glomerular expression of JAK1; B and H: two FSGS patients with high glomerular expression of JAK1; C and I: two FSGS patients with low glomerular expression of JAK2; D and J: two FSGS patients with high glomerular expression of JAK2; E and K: two FSGS patients with low glomerular expression of STAT1; F and L: two FSGS patients with high glomerular expression of STAT1 (200×). A1 to L1 are corresponding images for the cases labeled as A to L (200×) under higher magnification (400×).
Figure 9.
Figure 9.
Correlation of JAK-STAT1 activation score from glomerular compartment with baseline eGFR (panel A) and baseline UPCR (panel B) and JAK-STAT activation score from tubular compartment with baseline eGFR (panel C) and baseline UPCR (panel D) in NEPTUNE patients.
Figure 10.
Figure 10.
Adjusted Kaplan Meier survival curve for complete remission by tertile of JAK-STAT1 Activation z-score (Glomerular Compartment, panel A, p-value 0.14 and Tubular Compartment, panel B, p-value 0.08). Adjusted Kaplan Meier survival curve for Composite of 40% loss in eGFR and ESRD by tertile of JAK-STAT1 Activation z-score (Glomerular Compartment, panel C, p-value 0.41 and Tubular Compartment, panel D, p-value 0.02).

References

    1. Malaga-Dieguez L, Bouhassira D, Gipson D, Trachtman H. Novel Therapies for FSGS: Preclinical and Clinical Studies. Advances in chronic kidney disease. 2015;22(2):e1–e6.
    1. Kitiyakara C, Kopp JB, Eggers P. Trends in the epidemiology of focal segmental glomerulosclerosis. Seminars in nephrology. 2003;23(2):172–182.
    1. McGrogan A, Franssen CF, de Vries CS. The incidence of primary glomerulonephritis worldwide: a systematic review of the literature. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association. 2011;26(2):414–430.
    1. Maisonneuve P, Agodoa L, Gellert R, et al. Distribution of primary renal diseases leading to end-stage renal failure in the United States, Europe, and Australia/New Zealand: results from an international comparative study. Am J Kidney Dis. 2000;35(1):157–165.
    1. Fogo AB. Causes and pathogenesis of focal segmental glomerulosclerosis. Nature reviews. Nephrology 2015;11(2):76–87.
    1. Bao L, Haas M, Pippin J, et al. Focal and segmental glomerulosclerosis induced in mice lacking decay-accelerating factor in T cells. J Clin Invest. 2009;119(5):1264–1274.
    1. Shalhoub RJ. Pathogenesis of lipoid nephrosis: a disorder of T-cell function. Lancet. 1974;2(7880):556–560.
    1. Wada T, Nangaku M. A circulating permeability factor in focal segmental glomerulosclerosis: the hunt continues. Clin Kidney J. 2015;8(6):708–715.
    1. Stachowski J, Barth C, Michalkiewicz J, et al. Th1/Th2 balance and CD45-positive T cell subsets in primary nephrotic syndrome. Pediatr Nephrol. 2000;14(8–9):779–785.
    1. Krutzik PO, Crane JM, Clutter MR, Nolan GP. High-content single-cell drug screening with phosphospecific flow cytometry. Nat Chem Biol. 2008;4(2):132–142.
    1. O’Shea JJ, Plenge R. JAK and STAT signaling molecules in immunoregulation and immune-mediated disease. Immunity. 2012;36(4):542–550.
    1. Brosius FC 3rd, He JC. JAK inhibition and progressive kidney disease. Curr Opin Nephrol Hypertens. 2015;24(1):88–95.
    1. Brosius FC, Tuttle KR, Kretzler M. JAK inhibition in the treatment of diabetic kidney disease. Diabetologia. 2016;59(8):1624–1627.
    1. Vincenti F, Silva HT, Busque S, et al. Evaluation of the effect of tofacitinib exposure on outcomes in kidney transplant patients. Am J Transplant. 2015;15(6):1644–1653.
    1. Cox SN, Sallustio F, Serino G, et al. Activated innate immunity and the involvement of CX3CR1-fractalkine in promoting hematuria in patients with IgA nephropathy. Kidney Int. 2012;82(5):548–560.
    1. Reggiani F, Ponticelli C. Focal segmental glomerular sclerosis: do not overlook the role of immune response. J Nephrol. 2016.
    1. Gadegbeku CA, Gipson DS, Holzman LB, et al. Design of the Nephrotic Syndrome Study Network (NEPTUNE) to evaluate primary glomerular nephropathy by a multidisciplinary approach. Kidney Int. 2013;83(4):749–756.
    1. Sampson MG, Robertson CC, Martini S, et al. Integrative Genomics Identifies Novel Associations with APOL1 Risk Genotypes in Black NEPTUNE Subjects. J Am Soc Nephrol. 2016;27(3):814–823.
    1. Sandberg R, Larsson O. Improved precision and accuracy for microarrays using updated probe set definitions. BMC Bioinformatics. 2007;8:48.
    1. Chen C, Grennan K, Badner J, et al. Removing batch effects in analysis of expression microarray data: an evaluation of six batch adjustment methods. PLoS One. 2011;6(2):e17238.
    1. Rapaport F, Khanin R, Liang Y, et al. Comprehensive evaluation of differential gene expression analysis methods for RNA-seq data. Genome Biol. 2013;14(9):R95.
    1. Edgar R, Domrachev M, Lash AE. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30(1):207–210.
    1. Shoguchi E, Tanaka M, Shinzato C, Kawashima T, Satoh N. A genome-wide survey of photoreceptor and circadian genes in the coral, Acropora digitifera. Gene. 2013;515(2):426–431.
    1. Popa C, Netea MG, Barrera P, et al. Cytokine production of stimulated whole blood cultures in rheumatoid arthritis patients receiving short-term infliximab therapy. Cytokine. 2005;30(2):72–77.
    1. Pavon EJ, Garcia-Rodriguez S, Zumaquero E, et al. Increased expression and phosphorylation of the two S100A9 isoforms in mononuclear cells from patients with systemic lupus erythematosus: a proteomic signature for circulating low-density granulocytes. J Proteomics. 2012;75(6):1778–1791.
    1. Ramirez-Velez G, Medina F, Ramirez-Montano L, et al. Constitutive phosphorylation of interferon receptor A-associated signaling proteins in systemic lupus erythematosus. PLoS One. 2012;7(7):e41414.
    1. Hale MB, Krutzik PO, Samra SS, Crane JM, Nolan GP. Stage dependent aberrant regulation of cytokine-STAT signaling in murine systemic lupus erythematosus. PLoS One. 2009;4(8):e6756.
    1. O’Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB, Laurence A. The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu Rev Med. 2015;66:311–328.
    1. Tuttle KBF, Adler S et al. Baricitinib in diabetic kidney disease: results from a phase 2, multi-center, randomized, double-blind, placebo-controlled study. Diabetes. 2015;64(Suppl 1):114–LB.
    1. Savin VJ, Sharma M, Zhou J, et al. Renal and Hematological Effects of CLCF-1, a B-Cell-Stimulating Cytokine of the IL-6 Family. J Immunol Res. 2015;2015:714964.
    1. McCarthy ET, Sharma M, Savin VJ. Circulating permeability factors in idiopathic nephrotic syndrome and focal segmental glomerulosclerosis. Clin J Am Soc Nephrol. 2010;5(11):2115–2121.

Source: PubMed

3
Tilaa