IL-6 family members contribute to host defense through the stimulation of acute-phase signaling, hematopoiesis, immune reactions, and regenerative processes. To investigate essential mechanisms that are linked toward a constitutively activated gp130 signaling, we generated and characterized a mouse model that reflects a constitutive and cytokine-independent activation of JAK/STAT3 signaling by Lgp130 in CD4- and CD8-positive T cells. Lgp130 is an engineered form of gp130 in which dimerization and activation are forced by a leucine zipper. T cell–specific Lgp130 activation resulted in massive phenotypical abnormalities, including splenomegaly, lymphadenopathy, and an upregulation of innate immune system components shown by hyperinflammatory signatures in several organs. Moreover, T cell–restricted expression of Lgp130 resulted in increased numbers of cytotoxic and regulatory T cells, especially in lymph nodes. Consistent with this, we found an elevated platelet production and increase in megakaryocytes in the spleen and bone marrow that are causative for an acute thrombocytosis accompanied by anemia. Due to a shortened life span of T cell–specific Lgp130 mice, we could also show that next to an overall increase in regulatory cell-cycle genes, an activation of p53 and increased expression of p21 provide evidence for a senescence-like phenotype. Together, these data suggest that T cell–restricted gp130 activation is not only involved in autoimmune processes but also in senescence-associated aging. Therefore, Lgp130 expression in T cells might be a suitable model to study inflammation and disease.

The IL-6 family of cytokines are crucial players in the coordinated activity of both the innate and the adaptive immune system and are involved especially in T and B cell responses (1, 2). Most of the known autoimmune, inflammatory, or myeloma diseases are linked to a dysregulation in the IL-6/JAK/STAT3 signaling axis (3–6). IL-6 binding to its IL-6R (IL-6Rα) forms an IL-6/IL-6R complex that leads to a dimerization of the signal-transducing receptor chain gp130, which activates several intracellular pathways, including the JAK/STAT3, Ras/MAPK, and PI3K/AKT signaling cascades (7–11). Importantly, the activation of STAT3 induces a multitude of target genes that orchestrate important physiological functions, such as the regulation of cell migration, proliferation and differentiation, angiogenesis, and cell survival (12, 13). Increasing evidence indicates that dysregulation of IL-6/gp130 formation and subsequent JAK/STAT3 signaling are not only associated with pathologic immune responses but also lead to chronic lymphoproliferative disorders and cancer (14–16). Consistently, permanent genetically driven IL-6 pathway activation (by gain-of-function mutations in gp130) leads to reactive oxygen species follwed by a strong development of premalignant cells and dysplastic intrahepatic nodules in chronic liver inflammation (17). In addition, senescence is initiated after an external stress imposed on the tissue. Senescent cells arrest in the cell cycle, encounter morphological changes, and develop a senescence-associated secretory phenotype (18, 19). Cytokines, such as IL-6, are a known senescence-associated secretory phenotype component that can promote or sustain senescence, including cytokine signaling pathways, such as JAK/STAT, that contribute to senescence (20–22). Therefore, a mechanistic and systemic understanding of the diverse functions of gp130 could help to better understand the development of several pathological diseases and their progression. In this study, we used a mouse model that reflects a constitutive and cytokine-independent activation of JAK/STAT3 in all T cells. Therefore, the extracellular domain of gp130 was replaced with the dimerizing human c-Jun leucine zipper, leading to forced gp130 dimerization. Lgp130 then was fused to a ZsGreen fluorescent reporter molecule, knocked into the ROSA26 locus, and crossed with CD4-Cre expressing mice (Lgp130CD4/wt). Activation of aberrant gp130/JAK/STAT3 signaling during the double-positive stage in T cells led in several organs to disease-associated hyperinflammatory signatures that mimic an autoimmune phenotype accompanied by a shortened life span and evidence of senescence. Taken together, this phenotype represents a perfect preclinical model that could be used to assess new candidate therapies in autoimmune diseases.

Lgp130fl/fl and CD4-Cre mice were originally obtained from one of the authors (S.R.-J., Kiel, Germany) and bred in the Animal Facility of the Heinrich-Heine-University of Düsseldorf, Germany. Both mouse lines were crossed under specific pathogen-free conditions, and experiments were performed with male Lgp130CD4/wt and Lgp130fl/wt control mice at the age of 8–12 wk, including one experimental setting until week 18. This study was carried out in accordance with the Declaration of Helsinki and the guidelines from Directive 2010/63/EU of the European Parliament on the protection of animals. The protocol was approved by the local authorities at Heinrich-Heine-University and by the Landesamtes für Natur- Umwelt und Verbraucherschutz (Germany, Nordrhein-Westfalen; Permit No. 81-02.04.2019, A064, O 117/11, O 23/19). Littermate controls (CD4-Cre–negative mice) were used as the control group. Physiological parameters such as total blood cell count, platelet isolation from the peripheral blood, body weight, and organ weight were determined, accompanied by histology, RNA, or protein analysis of spleen, liver, and heart tissue.

Histological analysis from snap-frozen tissue (liver, spleen) and paraffin-embedded heart tissue was performed with H&E staining (catalog no. GHS216; Sigma-Aldrich) and TUNEL (catalog no. 11684795910; Roche). For the analysis of platelet megakaryocytes (MKs) in the spleen and in the bone marrow (BM) of the femur of untreated mice, 5-μm sections of paraffin-embedded tissue were prepared followed by H&E staining. For each mouse/genotype, at least three spleen and BM sections were stained, three to five pictures per section were taken, and the MKs per visual field were counted. For immunohistochemistry, frozen lung tissues were prepared by inflating the lungs through the trachea with optimum cutting temperature compound, followed by tying off the trachea to maintain the fluid in the lung. The tissues were then slowly frozen in a vessel on 2-methyl-1-isopropanol on dry ice. Frozen sections were cut at 8 µm in a cryostat (CM3050S; Leica). The cut sections were treated with 4% formaldehyde for 15 min after blocking with 5% donkey serum for 1 h. Anti-CD45 primary Ab (catalog no. AF114; R&D Systems) was applied overnight at 4°C followed by anti-goat–NL493 secondary Ab (catalog no. NL003; R&D Systems) for 1 h.

Different immune populations were identified in single-cell solutions from naive spleen, thymus, and lymph node samples using several Abs (anti-CD3, CD4, CD8, CD25, CD44, and Foxp3; all from Thermo Fisher Scientific). For quantification of cell populations, calibration beads were added to access cell numbers (BD, San Diego, CA). Experiments were analyzed using FlowJo software. Murine platelet activation was performed using fluorophore-labeled Abs for P-selectin expression (Wug.E9-FITC; Emfret Analytics) and the active form of αIIbβ3 integrin (JON/A-PE; Emfret Analytics). Heparinized blood was diluted in Tyrode’s buffer and washed twice. Blood samples were mixed with Abs after addition of 2 mM CaCl2 and stimulated with indicated agonists for 15 min at room temperature. Reaction was stopped by the addition of PBS, and samples were analyzed on a FACSCalibur and LSRFortessa flow cytometer (BD Biosciences). For analysis of glycoprotein surface expression, blood samples were mixed with Abs (GPVI, JAQ1-FITC; GPIb/CD42, Xia.G5-PE; integrin α5/CD49e, Tap.A12-FITC; Emfret Analytics) and incubated for 15 min at room temperature. For determination of platelet size, the geometric mean of the CD42-positive platelets in their forward scatter profile was used.

RNA purification was performed according to the manufacturer’s instructions (TRIzol; Invitrogen). Gene expression of target genes (Supplemental Table I) was performed by iTaq Universal SYBR Green One-Step Kit (catalog no. 1725151; Bio-Rad). For analysis, the expression levels of all target genes were normalized to the expression of Lgp130fl/wt (Δ cycle threshold). Gene expression values were calculated based on the ΔΔCt method. Relative quantities (RQs) were determined using the equation: RQ = 2−ΔΔCt.

In brief, liver, spleen, and heart tissue were lysed in lysis buffer (50 mM Tris-HCl [pH 7.5], 150 mM NaCl, 2 mM EDTA [pH 8.0], 2 mM NaF, 1 mM Na3VO4, 1% Nonidet P-40, 1% Triton X-100, and one cOmplete protease inhibitor mixture tablet per 50 ml). After lysis, the protein content was measured by Bicinchoninic acid assay. A total protein amount of 50 µg was then loaded per lane followed by immunoblotting. Abs used for protein detection were as follows: anti–p-STAT3 (catalog no. 9145), anti–total-STAT3 (catalog no. 9139), anti–p-STAT1 (catalog no. 9167), anti–total-STAT1 (catalog no. 9172), anti–p-STAT5 (catalog no. 9359), anti–total-STAT5 (catalog no. 94205), anti–p-Akt (catalog no. 4060), anti–total-Akt (catalog no. 9272), anti–p-p44/42 (catalog no. 9106), anti–total-p44/42 (catalog no. 9102), and anti-GAPDH (catalog no. 2118) were purchased by Cell Signaling. Anti-p53 (catalog no. sc-126) was purchased from Santa Cruz Biotechnology.

Mouse IL-6 (catalog no. 88-7064-88; Invitrogen) and mouse IL-6R α DuoSet ELISA (soluble IL-6R [sIL-6R]; DY1830; R&D Systems) were measured according to the manufacturer’s protocols. Mouse anti-nuclear Abs, mouse anti-dsDNA Abs total Ig (catalog no. 5210 and 5110, respectively), and mouse Rheumatoid Factor (RF) Ig's [total (A+G+M) ELISA Kit; catalog no. 6200] were purchased by Alpha Diagnostic International, and assays were performed according to the manufacturer’s instruction.

Murine blood from retro-orbital plexus was collected in 300 µl heparin solution (20 U/ml in PBS), and total blood cell counts were analyzed by a hematology analyzer (Sysmex KX-N21 [Norderstedt, Germany] and VetScan HM5v2.31 [Lab Technologies Medizintechnik, Vienna, Austria]).

Aspartate aminotransferase (glutamate-oxalacetate transaminase [GOT]), alanine aminotransferase (glutamic-pyruvic transaminase), total bilirubin, total cholesterol, triglycerides, glucose, amylase (AMY), high-density lipoprotein (HDLc), albumin, lactate dehydrogenase, creatine phosphokinase, and alanine phosphatase were measured using the automated biochemical analyzer Spotchem EZ SP-4430 (Arkray, Amstelveen, the Netherlands) and the Spotchem EZ Reagent Single Stripes or Multi Stripes Liver-1, Kenshin-1.

Bile acids and their taurine derivatives in serum were analyzed by ultra-performance liquid chromatography–tandem mass spectrometry (UPLC-MS/MS). The system consists of an ACQUITY UPLC I-Class (Waters, U.K.) coupled to a Waters Xevo-TQS MS/MS equipped with an electrospray ionization source in the negative ion mode. Data were collected in the multiple reaction monitoring mode.

Senescence β-galactosidase activity was measured by an assay kit (catalog no. 23833S; Cell Signaling) according to the manufacturer’s protocol.

Data are provided as arithmetic means ± SEM using GraphPad Prism, Version 8. Statistically significant differences between two groups were determined with a Student t test, including Welch’s correction if indicated. Statistical analysis between several groups was determined using a two-way ANOVA including Tukey correction. Significance was calculated as follows: n.s.p > 0.05 (not significant), *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001.

Lgp130fl/wt mice carrying the CAG-(loxP)STOP(loxP)-Lgp130-2A-ZsGreen cDNA in the Rosa26 locus were crossed with heterozygous mice expressing the Cre recombinase under the control of the CD4 promoter (Fig. 1A, 1B) (23, 24). Expression of Lgp130 was induced after deletion of the (loxP)STOP(loxP)-cassette, placing Lgp130-2A-ZsGreen under the control of the CAG promoter. Coexpression of ZsGreen was used as marker for recombination efficiency and Lgp130 transgene expression. During thymic development, CD4 is first expressed in the double-positive CD4/CD8 state of all T cells, thus resulting in transgenic expression of Lgp130 in both CD4- and CD8-positive T cells on Cre-mediated recombination. Flow cytometric analysis of thymic T cells from Lgp130CD4/wt mice showed that CD4- and/or CD8-positive T cells, but not CD4/CD8-negative T cells, expressed ZsGreen (Fig. 1C), whereas control Lgp130fl/wt mice did not express ZsGreen (Fig. 1C). In the lymph nodes from Lgp130CD4/wt mice, but not Lgp130fl/wt mice, peripheral CD4- and CD8-positive T cells, but not peripheral CD19-positive B cells, were found to express ZsGreen (Fig. 1D, 1E, Supplemental Fig. 4). Consequently, ZsGreen fluorescence was detected in thymus, spleen, and lymph nodes but only minimally in the liver of Lgp130CD4/wt mice (Fig. 1F, Supplemental Fig. 4), which is consistent with the low amount of T cells in the liver (25, 26). Lgp130CD4/wt mice displayed significantly elevated transcript levels of lgp130 in total spleen in comparison with Lgp130fl/wt (Fig. 1G). The expression of Lgp130 in T cells goes along with statistically significant phenotypic abnormalities. Median survival of Lgp130CD4/wt mice was ∼45 d (Fig. 2A). In addition, Lgp130CD4/wt mice showed indications of embryonic lethality because of a reduced ratio of Lgp130CD4/wt over Lgp130fl/wt mice among offspring (Fig. 2B). Consistently, body weight at 6 wk of age was significantly lower (30%) in Lgp130CD4/wt compared with Lgp130fl/wt mice (17.5 versus 24.6 g) (Fig. 2C, 2D). Spleen-to-body weight ratio revealed splenomegaly in Lgp130CD4/wt mice accompanied by elevated heart-, lung-, lymph node-, spleen-, and thymus-to-body weight ratios, whereas the liver-to-body weight ratio was not altered (Fig. 2E, 2F). Surprisingly, the hearts of Lgp130fl/wt and Lgp130CD4/wt mice presented no signs of severe hypertrophy or dilation. For this reason, we would conclude that the effect of the increased heart weight might be a response to an increased immune cell infiltration (Supplemental Fig. 3E3G).

FIGURE 1.

Lgp130 and ZsGreen are expressed on CD4- and CD8-positive T cells. (A) Schematic view of the synthetic designer receptor Lgp130. IL-6 or IL-11 binds to its specific receptor. Homodimerization of gp130 leads to transphosphorylation of certain tyrosine residues by JAKs. p-STAT3 homodimers translocate into the nucleus, thereby increasing the expression of SOCS3 and SAA1, as well as cell proliferation. The exchange of the entire extracellular domain by a leucine zipper (PDB entry: 1JUN) has led to a constitutively active receptor complex, named Lgp130. (B) The expression strategy by generated mice carrying a CAG-(loxP)STOP(loxP)-Lgp130-2A-ZsGreen construct in the Rosa26 locus. (C) Cell-type–specific ZsGreen detection of CD4 and CD8 double-negative (black), CD4 and CD8 double-positive (red), CD8-positive (blue), and CD4-positive (green) cells by flow cytometry λex(488 nm) in thymus. Representative data from one Lgp130fl/wt and one Lgp130CD4/wt mouse are shown. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. (D) Cell-type–specific ZsGreen detection of CD4-positive, CD8-positive, and CD19-positive cells by flow cytometry excitation wavelength (488 nm). Lymph nodes derived from Lgp130CD4/wt mice. Representative data from one Lgp130fl/wt and one Lgp130CD4/wt mouse is shown. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. (E) Cell-type–specific ZsGreen analysis of CD4-positive, CD8-positive, and CD19-positive cells by flow cytometry λex(488 nm). Lymph nodes were derived from Lgp130fl/wt. Representative data from one Lgp130fl/wt and one Lgp130CD4/wt mouse are shown. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. (F) Detection of ZsGreen in liver (red), spleen (blue), thymus (orange), and lymph node (green) derived from Lgp130CD4/wt mice as analyzed by flow cytometry. Representative data from one Lgp130fl/wt and one Lgp130CD4/wt mouse are shown. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. (G) Relative Lgp130 mRNA expression in Lgp130fl/wt and Lgp130CD4/wt splenocytes. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. Data represent ± SEM of each group. The p value was determined by unpaired t test: **p < 0.01. Data presented in (C)–(E) represent one of two experiments, whereas data in (G) represent a single experiment with this mouse cohort.

FIGURE 1.

Lgp130 and ZsGreen are expressed on CD4- and CD8-positive T cells. (A) Schematic view of the synthetic designer receptor Lgp130. IL-6 or IL-11 binds to its specific receptor. Homodimerization of gp130 leads to transphosphorylation of certain tyrosine residues by JAKs. p-STAT3 homodimers translocate into the nucleus, thereby increasing the expression of SOCS3 and SAA1, as well as cell proliferation. The exchange of the entire extracellular domain by a leucine zipper (PDB entry: 1JUN) has led to a constitutively active receptor complex, named Lgp130. (B) The expression strategy by generated mice carrying a CAG-(loxP)STOP(loxP)-Lgp130-2A-ZsGreen construct in the Rosa26 locus. (C) Cell-type–specific ZsGreen detection of CD4 and CD8 double-negative (black), CD4 and CD8 double-positive (red), CD8-positive (blue), and CD4-positive (green) cells by flow cytometry λex(488 nm) in thymus. Representative data from one Lgp130fl/wt and one Lgp130CD4/wt mouse are shown. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. (D) Cell-type–specific ZsGreen detection of CD4-positive, CD8-positive, and CD19-positive cells by flow cytometry excitation wavelength (488 nm). Lymph nodes derived from Lgp130CD4/wt mice. Representative data from one Lgp130fl/wt and one Lgp130CD4/wt mouse is shown. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. (E) Cell-type–specific ZsGreen analysis of CD4-positive, CD8-positive, and CD19-positive cells by flow cytometry λex(488 nm). Lymph nodes were derived from Lgp130fl/wt. Representative data from one Lgp130fl/wt and one Lgp130CD4/wt mouse are shown. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. (F) Detection of ZsGreen in liver (red), spleen (blue), thymus (orange), and lymph node (green) derived from Lgp130CD4/wt mice as analyzed by flow cytometry. Representative data from one Lgp130fl/wt and one Lgp130CD4/wt mouse are shown. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. (G) Relative Lgp130 mRNA expression in Lgp130fl/wt and Lgp130CD4/wt splenocytes. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. Data represent ± SEM of each group. The p value was determined by unpaired t test: **p < 0.01. Data presented in (C)–(E) represent one of two experiments, whereas data in (G) represent a single experiment with this mouse cohort.

Close modal
FIGURE 2.

Lgp130 leads to prenatal death, weight loss, splenomegaly, and lymphadenopathy. (A) Kaplan–Meier curves depicting the survival of Lgp130CD4/wt mice (n = 17) and Lgp130fl/wt mice (n = 6). p value was determined by log-rank (Mantel–Cox) test. Median survival of 45 d (dotted line). (B) Ratio of the offspring: depicted are only male Lgp130CD4/wt (gray, n = 33) and Lgp130fl/wt (black, n = 62) mice. (C) Representative pictures from 8-wk-old Lgp130CD4/wt (top) and control mice (bottom). In total, Lgp130fl/wt (n = 5) and Lgp130CD4/wt (n = 5) mice were used. (D) Body weight of Lgp130 CD4/wt and control mice at the age of 8 wk. In total, Lgp130fl/wt (n = 12) and Lgp130CD4/wt (n = 12) mice were used. Data represent ± SEM of each group. p < 0.001 in an unpaired Student t test. (E) Ratios of heart weight, liver weight, lung weight, spleen weight, lymph node weight, and thymus weight divided by body weight of Lgp130CD4/wt and Lgp130fl/wt mice at the age of 8 wk. In total, Lgp130fl/wt (n = 5–8) and Lgp130CD4/wt (n = 5–8) mice were used. Data represent ± SEM of each group. p < 0.001 in an unpaired Student t test. (F) Representative pictures of heart, liver, lung, spleen, thymus, and lymph node from 8-wk-old Lgp130CD4/wt (n = 6) and Lgp130fl/wt mice (n = 6). Data presented in (A), (B), and (F) represent a single experiment with this mouse cohort, whereas data in (C)–(E) are aggregated from two experiments. n.s.p > 0.05, *p < 0.05, **p < 0.01, ****p < 0.0001. n.s., not significant.

FIGURE 2.

Lgp130 leads to prenatal death, weight loss, splenomegaly, and lymphadenopathy. (A) Kaplan–Meier curves depicting the survival of Lgp130CD4/wt mice (n = 17) and Lgp130fl/wt mice (n = 6). p value was determined by log-rank (Mantel–Cox) test. Median survival of 45 d (dotted line). (B) Ratio of the offspring: depicted are only male Lgp130CD4/wt (gray, n = 33) and Lgp130fl/wt (black, n = 62) mice. (C) Representative pictures from 8-wk-old Lgp130CD4/wt (top) and control mice (bottom). In total, Lgp130fl/wt (n = 5) and Lgp130CD4/wt (n = 5) mice were used. (D) Body weight of Lgp130 CD4/wt and control mice at the age of 8 wk. In total, Lgp130fl/wt (n = 12) and Lgp130CD4/wt (n = 12) mice were used. Data represent ± SEM of each group. p < 0.001 in an unpaired Student t test. (E) Ratios of heart weight, liver weight, lung weight, spleen weight, lymph node weight, and thymus weight divided by body weight of Lgp130CD4/wt and Lgp130fl/wt mice at the age of 8 wk. In total, Lgp130fl/wt (n = 5–8) and Lgp130CD4/wt (n = 5–8) mice were used. Data represent ± SEM of each group. p < 0.001 in an unpaired Student t test. (F) Representative pictures of heart, liver, lung, spleen, thymus, and lymph node from 8-wk-old Lgp130CD4/wt (n = 6) and Lgp130fl/wt mice (n = 6). Data presented in (A), (B), and (F) represent a single experiment with this mouse cohort, whereas data in (C)–(E) are aggregated from two experiments. n.s.p > 0.05, *p < 0.05, **p < 0.01, ****p < 0.0001. n.s., not significant.

Close modal

Blood analysis showed that there was an overall reduction in WBC counts with an increase of granulocytes and monocytes and a decrease of lymphocytes in Lgp130CD4/wt mice as compared with Lgp130fl/wt control mice (Fig. 3A–D). In addition, RBCs were affected in Lgp130CD4/wt mice by a significant decrease in total hemoglobin and the hematocrit (Fig. 3E, 3F). In regards to lymphocytes, Lgp130 expression resulted in dramatically reduced numbers of CD4/CD8 double-positive T cells, i.e., 80.6% in Lgp130fl/wt mice as compared with 9% in Lgp130CD4/wt mice (Fig. 3G). This was accompanied by a strong increase of CD4/CD8-negative T cells from 2.2 to 49.7% and CD8-positive cells from 6.1 to 25.7% in Lgp130CD4/wt mice compared with Lgp130fl/wt mice (Fig. 3H, Supplemental Fig. 6A). CD4/CD8/CD3 triple-negative thymocytes are subdivided into four subsets based on expression of CD44 and CD25, along their developmental stage from CD44+CD25 to CD44+CD25+, CD44CD25+, and CD44CD25 (27). In Lgp130CD4/wt mice, the least mature cell stage (CD44+CD25) was clearly overrepresented (Fig. 3I, Supplemental Fig. 6A). Based on the frequencies, we can conclude that Lgp130CD4/wt mice appear to arrest in the CD4CD8 and CD25CD44+ stage during T cell development within the thymus (Supplemental Fig. 1C, 1D). CD8-dominated CD4/CD8 T cell ratios were observed in the spleen and lymph nodes of Lgp130CD4/wt mice compared with control Lgp130fl/wt mice (Fig. 3J, Supplemental Fig. 6B). Moreover, we observed significantly increased numbers of regulatory T (Treg) cells in lymph nodes and a higher tendency in spleens of Lgp130CD4/wt mice (Fig. 3K, Supplemental Fig. 1E, 6C). Peripheral CD4+ T cells showed reduced surface CD44 expression and increased CD69 levels in the spleen. Similar findings were obtained for CD44 on CD8+ T cells in the lymph nodes and the spleen, although CD69 expression had only an increased tendency (Supplemental Fig. 1A, 1B). For B cells, we observed an increase of CD19-positive B cells in the lymph nodes, but not in the spleen, of Lgp130CD4/wt mice compared with Lgp130fl/wt mice (Fig. 3L, Supplemental Fig. 1F, 6B). Taken together, our data showed that T cell–restricted expression of Lgp130 resulted in increased levels of cytotoxic T and Treg cells. Importantly, due to the higher expression of CD69, peripheral T cells are in a more activated state compared with wild-type (wt) mice.

FIGURE 3.

Activated IL-6/STAT3 signaling results in altered immune cell counts and a Treg-driven immune response. Quantitative analysis of (A) WBC count, (B) granulocytes, (C) monocytes, (D) lymphocytes, (E) hemoglobin, and (F) hematocrit from Lgp130CD4/wt mice (n = 16–17) and Lgp130fl/wt (n = 8–12) mice. Data represent ± SEM. p values were determined by unpaired t test, including Welch’s correction. (G) CD4-positive and CD8-positive cells in thymocytes derived from Lgp130fl/wt (left) and Lgp130CD4/wt mice were analyzed by flow cytometry. Representative data from one Lgp130fl/wt and one Lgp130CD4/wt mouse are shown. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. (H) Cell counts of CD4 and CD8 double-negative, CD4 and CD8 double-positive, CD4-positive, and CD8-positive cells from Lgp130CD4/wt and Lgp130fl/wt mice as determined by flow cytometry. (I) Counts of CD44-positive, CD25 and CD44 double-positive, CD25-positive, and CD25 and CD44 double-negative thymocytes gated on the CD4 and CD8 double-negative cells from Lgp130CD4/wt and Lgp130fl/wt mice as determined by flow cytometry. In total for (H) and (I), Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. Data represent ± SEM of each group. p values were determined by unpaired t test. (J) Ratio of CD4- and CD8-positive cells of Lgp130CD4/wt and Lgp130fl/wt mice in lymph nodes and spleen as determined by flow cytometry. (K) Total counts of Treg cells analyzed by intracellular staining of Foxp3 and (L) total counts of CD19-positive cells of Lgp130CD4/wt and Lgp130fl/wt mice in lymph nodes and spleen as determined by flow cytometry. In total for (J)–(L), Lgp130fl/wt (n = 5) and Lgp130CD4/wt (n = 5) mice were used. Data represent ± SEM of each group. p values were determined by unpaired t test. Data presented in (A)–(F) represent aggregated data from three experiments, whereas data in (G)–(L) represent a single experiment with this mouse cohort. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 3.

Activated IL-6/STAT3 signaling results in altered immune cell counts and a Treg-driven immune response. Quantitative analysis of (A) WBC count, (B) granulocytes, (C) monocytes, (D) lymphocytes, (E) hemoglobin, and (F) hematocrit from Lgp130CD4/wt mice (n = 16–17) and Lgp130fl/wt (n = 8–12) mice. Data represent ± SEM. p values were determined by unpaired t test, including Welch’s correction. (G) CD4-positive and CD8-positive cells in thymocytes derived from Lgp130fl/wt (left) and Lgp130CD4/wt mice were analyzed by flow cytometry. Representative data from one Lgp130fl/wt and one Lgp130CD4/wt mouse are shown. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. (H) Cell counts of CD4 and CD8 double-negative, CD4 and CD8 double-positive, CD4-positive, and CD8-positive cells from Lgp130CD4/wt and Lgp130fl/wt mice as determined by flow cytometry. (I) Counts of CD44-positive, CD25 and CD44 double-positive, CD25-positive, and CD25 and CD44 double-negative thymocytes gated on the CD4 and CD8 double-negative cells from Lgp130CD4/wt and Lgp130fl/wt mice as determined by flow cytometry. In total for (H) and (I), Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. Data represent ± SEM of each group. p values were determined by unpaired t test. (J) Ratio of CD4- and CD8-positive cells of Lgp130CD4/wt and Lgp130fl/wt mice in lymph nodes and spleen as determined by flow cytometry. (K) Total counts of Treg cells analyzed by intracellular staining of Foxp3 and (L) total counts of CD19-positive cells of Lgp130CD4/wt and Lgp130fl/wt mice in lymph nodes and spleen as determined by flow cytometry. In total for (J)–(L), Lgp130fl/wt (n = 5) and Lgp130CD4/wt (n = 5) mice were used. Data represent ± SEM of each group. p values were determined by unpaired t test. Data presented in (A)–(F) represent aggregated data from three experiments, whereas data in (G)–(L) represent a single experiment with this mouse cohort. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

In the spleen and liver of Lgp130CD4/wt mice, we observed an increased STAT1 and STAT3 phosphorylation, with a tendency of higher STAT5 phosphorylation as compared with Lgp130fl/wt mice, whereas in the heart, STAT1 and STAT5, but not STAT3, phosphorylation was increased (Fig. 4A, Supplemental Fig. 2). As seen before for constitutively active gp130 (23, 28, 29), increased levels of p-ERK and p-AKT were not detected in Lgp130CD4/wt mice (Supplemental Fig. 2). Next, quantitative mRNA expression analysis revealed a significant increase of SOCS3 and a downregulation of TNF-α, IL-10, and CCL5/RANTES in the spleen of Lgp130CD4/wt mice (Fig. 4B). IL-6, TNF-α, IL-10, SOCS3, IFN-γ, CCL5/RANTES, and the acute-phase response protein SAA1 were highly upregulated in the liver of Lgp130CD4/wt mice as compared with Lgp130fl/wt control mice (Fig. 4B). In the heart of Lgp130CD4/wt mice, IL-6, TNF-α, IL-10, SOCS3, IFN-γ, and CCL5/RANTES mRNA were significantly elevated (Fig. 4B). In heart and spleen, α smooth muscle actin was significantly downregulated (Fig. 4B). IL-6 protein in the serum of Lgp130CD4/wt mice was found to be significantly increased compared with Lgp130fl/wt mice (Fig. 4C). Also, the sIL-6R serum level was upregulated in Lgp130fl/wt mice (Fig. 4D). Due to this overall proinflammatory phenotype in Lgp130fl/wt mice, we also found a strong upregulation of lupus-related autoantibodies against anti-nuclear IgG, dsDNA, and rheumatoid arthritis (RA)-related RF in the serum of Lgp130CD4/wt mice compared with Lgp130fl/wt control mice (Fig. 4E–G). Moreover, serum from Lgp130CD4/wt and Lgp130fl/wt control mice was applied to B cell–deficient spleen tissue to independently assess the degree of autoimmune Abs. As shown in Fig. 4H, almost no Abs from serum of Lgp130fl/wt control mice bound to B cell–deficient spleen tissue, whereas strong binding was observed for serum from Lgp130CD4/wt mice. Histological analysis of the liver and heart tissue from Lgp130CD4/wt mice revealed an increase in the infiltration of immune cells, whereas spleen tissue revealed a dramatic disruption of splenic architecture with loss of splenic marginal zones and the T cell/B cell organization (Fig. 5A). Increased apoptosis was, however, not observed by TUNEL staining of liver, heart, and spleen (Fig. 5B, 5C). Immunohistochemical staining for inflammatory (CD45) cell infiltration in lung tissue was significantly elevated in Lgp130CD4/wt mice compared with Lgp130fl/wt mice. This supports the overall polyautoimmune phenotype, but lung-specific changes in the architecture were not further investigated (Supplemental Fig. 3C, 3D). In contrast, expression of cell-cycle genes such as Cyclin A, Cyclin D, and Cyclin E were significantly upregulated in the liver of Lgp130CD4/wt mice as compared with Lgp130fl/wt control mice, accompanied by a small induction of the proliferation marker Ki67 (Fig. 5D). Analysis of serum parameters revealed increased GOT, AMY, and alkaline phosphatase (Fig. 5E) and decreased total cholesterol, HDLc, triglyceride, and Glu in Lgp130CD4/wt mice as compared with Lgp130fl/wt mice (Fig. 5F). Functional liver parameters such as albumin and total bilirubin were rather reduced in Lgp130CD4/wt compared with Lgp130fl/wt mice, whereas glutamic-pyruvic transaminase, LDH, and creatine phosphokinase were slightly increased in Lgp130CD4/wt mice (Supplemental Fig. 3A). Given that bile acids have been known to regulate cholesterol homeostasis and are recognized as hormones involved in the regulation of various metabolic processes (30), bile acid profiling in the serum was performed next. Lgp130CD4/wt mice revealed compared with Lgp130fl/wt controls a significant increase in the overall level of serum bile acids, especially taurine-conjugated and unconjugated bile acids (Supplemental Fig. 3B). Taken together, our data indicate massive liver damage accompanied by cholestasis and an overall metabolic imbalance.

FIGURE 4.

T cell–driven hyperinflammatory phenotype in Lgp130CD4/wt mice. (A) Quantification of STAT phosphorylation. Relative fluorescence intensity from the Western blots of heart, liver, and spleen of 8- to 12-wk-old Lgp130CD4/wt and Lgp130fl/wt mice. Equal amounts of total cell protein lysates (50 µg/lane) were analyzed using specific Abs detecting p-STAT1, GAPDH, STAT3, p-STAT3, STAT5, and p-STAT5. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. Data represent ± SEM of each group. p values were determined by unpaired t test. (B) Gene expression levels of IL-6, αSMA, TNF-α, IL-10, SOCS3, IFN-γ, CCL5/RANTES, and SAA1 in the spleen, liver, and heart tissue derived from 6- to 8-wk-old Lgp130CD4/wt and Lgp130fl/wt mice. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. Data represent ± SEM of each group. (C) ELISA against mouse IL-6 and (D) mouse IL-6R from isolated serum of Lgp130CD4/wt and Lgp130fl/wt mice. In total for (C), Lgp130fl/wt (n = 11) and Lgp130CD4/wt (n = 19) mice were used, and for (D), Lgp130fl/wt (n = 12) and Lgp130CD4/wt (n = 9) mice were used. Data represent ± SEM of each group. p values were determined by two-way ANOVA, including Tukey correction. (E) Total activity units of anti-nuclear Abs (total Ig) analyzed in Lgp130CD4/wt and Lgp130fl/wt mice. In total, Lgp130fl/wt (n = 7) and Lgp130CD4/wt (n = 8) mice were used. Data represent ± SEM of each group. p values were determined by unpaired t test including Welch’s correction. (F) Total activity units of mouse anti-dsDNA Abs total (Ig ELISA) from Lgp130CD4/wt and Lgp130fl/wt mice. In total, Lgp130fl/wt (n = 7) and Lgp130CD4/wt (n = 8) mice were used. Data represent ± SEM of each group. p value was determined by unpaired t test, including Welch’s correction. (G) Measurement of RF in Lgp130CD4/wt and Lgp130fl/wt mice by ELISA. In total, Lgp130fl/wt (n = 12) and Lgp130CD4/wt (n = 12) mice were used. Data represent ± SEM of each group. (H) Serum samples from Lgp130CD4/wt and Lgp130fl/wt mice were applied on 8-µm-thick frozen liver cryosections of B cell–deficient mice (Jh-mice) followed by staining with a Cy3-labeled Ab against mouse IgG (ThermoFisher Scientific, catalog no. M30010). Scale bars: 100 µm. Representative data from one Lgp130fl/wt and one Lgp130CD4/wt mouse are shown. In total, Lgp130fl/wt (n = 3) and Lgp130CD4/wt (n = 3) mice were used. Data presented in (A), (B), and (H) represent a single experiment with this mouse cohort, whereas data in (C) represent data aggregated from three experiments, and data in (D)–(G) represent data aggregated from two experiments. n.s.p > 0.05 (not significant), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 4.

T cell–driven hyperinflammatory phenotype in Lgp130CD4/wt mice. (A) Quantification of STAT phosphorylation. Relative fluorescence intensity from the Western blots of heart, liver, and spleen of 8- to 12-wk-old Lgp130CD4/wt and Lgp130fl/wt mice. Equal amounts of total cell protein lysates (50 µg/lane) were analyzed using specific Abs detecting p-STAT1, GAPDH, STAT3, p-STAT3, STAT5, and p-STAT5. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. Data represent ± SEM of each group. p values were determined by unpaired t test. (B) Gene expression levels of IL-6, αSMA, TNF-α, IL-10, SOCS3, IFN-γ, CCL5/RANTES, and SAA1 in the spleen, liver, and heart tissue derived from 6- to 8-wk-old Lgp130CD4/wt and Lgp130fl/wt mice. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used. Data represent ± SEM of each group. (C) ELISA against mouse IL-6 and (D) mouse IL-6R from isolated serum of Lgp130CD4/wt and Lgp130fl/wt mice. In total for (C), Lgp130fl/wt (n = 11) and Lgp130CD4/wt (n = 19) mice were used, and for (D), Lgp130fl/wt (n = 12) and Lgp130CD4/wt (n = 9) mice were used. Data represent ± SEM of each group. p values were determined by two-way ANOVA, including Tukey correction. (E) Total activity units of anti-nuclear Abs (total Ig) analyzed in Lgp130CD4/wt and Lgp130fl/wt mice. In total, Lgp130fl/wt (n = 7) and Lgp130CD4/wt (n = 8) mice were used. Data represent ± SEM of each group. p values were determined by unpaired t test including Welch’s correction. (F) Total activity units of mouse anti-dsDNA Abs total (Ig ELISA) from Lgp130CD4/wt and Lgp130fl/wt mice. In total, Lgp130fl/wt (n = 7) and Lgp130CD4/wt (n = 8) mice were used. Data represent ± SEM of each group. p value was determined by unpaired t test, including Welch’s correction. (G) Measurement of RF in Lgp130CD4/wt and Lgp130fl/wt mice by ELISA. In total, Lgp130fl/wt (n = 12) and Lgp130CD4/wt (n = 12) mice were used. Data represent ± SEM of each group. (H) Serum samples from Lgp130CD4/wt and Lgp130fl/wt mice were applied on 8-µm-thick frozen liver cryosections of B cell–deficient mice (Jh-mice) followed by staining with a Cy3-labeled Ab against mouse IgG (ThermoFisher Scientific, catalog no. M30010). Scale bars: 100 µm. Representative data from one Lgp130fl/wt and one Lgp130CD4/wt mouse are shown. In total, Lgp130fl/wt (n = 3) and Lgp130CD4/wt (n = 3) mice were used. Data presented in (A), (B), and (H) represent a single experiment with this mouse cohort, whereas data in (C) represent data aggregated from three experiments, and data in (D)–(G) represent data aggregated from two experiments. n.s.p > 0.05 (not significant), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal
FIGURE 5.

Metabolic imbalance and hyperactivated cell cycle in T cell–specific Lgp130 mice. (A) Representative pictures of H&E-stained liver, heart, and spleen (5 µm) of Lgp130CD4/wt (n = 5) and Lgp130fl/wt mice (n = 5) from two independent experiments. Arrows indicate accumulation of nuclei after inflammation (liver) and myocarditis (heart). Red pulp (RP) and white pulp (WP) in the spleen are highlighted in Lgp130fl/wt mice. Lgp130CD4/wt with disrupted splenic architecture. (B) TUNEL assay and DAPI staining from paraffin-embedded heart, liver, and spleen tissue sections (6 µm) of Lgp130CD4/wt and Lgp130fl/wt mice. Images demonstrate TUNEL-positive events (green) and nuclei (blue). Scale bars: 100 µm. Representative pictures from one Lgp130fl/wt and one Lgp130CD4/wt mouse is shown. In total, Lgp130fl/wt (n = 4) and Lgp130CD4/wt (n = 4) mice were used from two different experiments. (C) Quantification of TUNEL events in liver, heart, and spleen from (B) Lgp130CD4/wt and Lgp130fl/wt mice. (D) Gene expression of the cell-cycle markers: CyclinA2, CyclinD2, CyclinE1, and Ki67 in liver tissue from Lgp130CD4/wt-positive and control mice. In total, Lgp130fl/wt (n = 5) and Lgp130CD4/wt (n = 5) mice were used in a single experiment with this mouse cohort. Data represent ± SEM of each group. p values were determined by unpaired t test including Welch’s correction. (E) The activity of GOT, AMY, and alkaline phosphatase (ALP) was measured in serum samples of Lgp130CD4/wt-positive and Lgp130fl/wt mice. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used from two independent experiments. Data represent ± SEM of each group. p values were determined by unpaired t test including Welch’s correction. (F) Quantitative analysis of total cholesterin (TCho), HDLc, triglycerides, and Glu of Lgp130CD4/wt and Lgp130fl/wt mice. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used within two experiments. Data represent ± SEM of each group. p values were determined by unpaired t test including Welch’s correction. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 5.

Metabolic imbalance and hyperactivated cell cycle in T cell–specific Lgp130 mice. (A) Representative pictures of H&E-stained liver, heart, and spleen (5 µm) of Lgp130CD4/wt (n = 5) and Lgp130fl/wt mice (n = 5) from two independent experiments. Arrows indicate accumulation of nuclei after inflammation (liver) and myocarditis (heart). Red pulp (RP) and white pulp (WP) in the spleen are highlighted in Lgp130fl/wt mice. Lgp130CD4/wt with disrupted splenic architecture. (B) TUNEL assay and DAPI staining from paraffin-embedded heart, liver, and spleen tissue sections (6 µm) of Lgp130CD4/wt and Lgp130fl/wt mice. Images demonstrate TUNEL-positive events (green) and nuclei (blue). Scale bars: 100 µm. Representative pictures from one Lgp130fl/wt and one Lgp130CD4/wt mouse is shown. In total, Lgp130fl/wt (n = 4) and Lgp130CD4/wt (n = 4) mice were used from two different experiments. (C) Quantification of TUNEL events in liver, heart, and spleen from (B) Lgp130CD4/wt and Lgp130fl/wt mice. (D) Gene expression of the cell-cycle markers: CyclinA2, CyclinD2, CyclinE1, and Ki67 in liver tissue from Lgp130CD4/wt-positive and control mice. In total, Lgp130fl/wt (n = 5) and Lgp130CD4/wt (n = 5) mice were used in a single experiment with this mouse cohort. Data represent ± SEM of each group. p values were determined by unpaired t test including Welch’s correction. (E) The activity of GOT, AMY, and alkaline phosphatase (ALP) was measured in serum samples of Lgp130CD4/wt-positive and Lgp130fl/wt mice. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used from two independent experiments. Data represent ± SEM of each group. p values were determined by unpaired t test including Welch’s correction. (F) Quantitative analysis of total cholesterin (TCho), HDLc, triglycerides, and Glu of Lgp130CD4/wt and Lgp130fl/wt mice. In total, Lgp130fl/wt (n = 6) and Lgp130CD4/wt (n = 6) mice were used within two experiments. Data represent ± SEM of each group. p values were determined by unpaired t test including Welch’s correction. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

Platelets are essential mediators of hemostasis to avoid blood loss by controlling thrombus formation (31). Therefore, blood cell counts of RBCs and platelets, as well as platelet size, were analyzed in whole blood. In Lgp130CD4/wt mice, RBC count was significantly reduced by ∼20% (Fig. 6A), whereas platelet counts were massively increased from 500 × 103/µl in Lgp130fl/wt mice to 5000 × 103/µl in Lgp130CD4/wt mice (Fig. 6B). Moreover, a tendency for an increased platelet size was observed (Fig. 6C). In spleen and BM sections, we estimated platelet formation by megakaryopoiesis. The number of MKs was significantly elevated by 30% in spleen and 20% in BM of Lgp130CD4/wt compared with Lgp130fl/wt mice (Fig. 6D–F). Platelet activation capacity was analyzed by flow cytometry and Ab binding to activated integrin αIIbβ3 (fibrinogen receptor) and determination of P-selectin exposure as a marker for degranulation. Overexpression of Lg130 in T cells resulted in a significant reduced degranulation measured via P-selectin exposure on the surface of platelets by flow cytometry. Activation of the major ITAM-coupled collagen receptor (GPVI) via collagen-related peptide binding led to a reduced P-selectin degranulation with all tested concentrations. A reduced degranulation was also measured in combination of both autocrine agonists ADP and U46 (thromboxane A2 analogue) targeting G-protein–coupled receptors on the platelet surface. Only by trend, a reduced degranulation was measured upon PAR4-peptide binding, activating the thrombin-mediated pathway of platelet activation. Interestingly, a mild preactivated state regarding degranulation could be observed in unstimulated platelets (resting control [−]) that stayed unaffected upon stimulation with a mild agonist such as ADP alone (Fig. 6G, Supplemental Fig. 5D). As additional marker, integrin αIIbβ3 (fibrinogen receptor) activation was measured. Although platelet degranulation upon Lgp130 overexpression in T cells was highly deregulated, integrin activation stays unaffected in unstimulated control samples as with every tested agonist (Fig. 6H, Supplemental Fig. 5C). Moreover, platelets from Lgp130CD4/wt mice had reduced surface expression of the glycoprotein GPVI and integrin α5 compared with Lgp130fl/wt mice, whereas GPIbα and integrin β3 expression were not significantly changed (Fig. 6I, Supplemental Fig. 5A, 5B). A reduction of GPVI, as receptor for platelet activation and regulator of thromboinflammation (32), implicates an increase in shedding, but because of a basal preactivation, this defect in activation might only be a “secondary” effect. Aging platelets lose sialic acid, the terminal carbohydrate moiety that covers the underlying galactose residues, a ligand for the hepatic Ashwell-Morell receptor (AMR). Desialylated platelets are removed from the circulation via the AMR to regulate thrombopoietin (TPO) production by JAK2-STAT3 (33). To analyze whether platelet clearance in Lgp130CD4/wt mice is associated with a glycan-dependent mechanism via hepatocytes or Kupffer cells, gene expression of liver samples was investigated. Analysis of liver gene expression revealed enhanced TPO expression resulting in enhanced megakaryopoiesis. As already shown, enhanced JAK-STAT signaling occurs in liver tissue of Lgp130CD4/wt mice compared with wt mice (Fig. 4A). TPO gene expression is regulated upon different receptor pathways in the liver (34). First, IL-6R and AMR (consisting of asialoglycoprotein receptor [Asgr] 1 and Asgr2 subunits) signaling on hepatocytes induces TPO gene expression. Although overall enhanced TPO is observed, a reduction of the Asgr2 subunit with an overexpression of the IL-6R targets toward an IL-6–driven pathway. However, it was recently discovered that Kupffer cells form a platelet clearance receptor consisting of the Asgr1 and Mgl subunits. Because the Mgl subunit is overexpressed, a Kupffer cell–hepatocyte interaction might be relevant (Fig. 6J). Taken together, the increase in MKs in spleen and BM contributes to the permanently increased number of platelets, resulting in a reactive thrombocytosis in Lgp130CD4/wt mice compared with Lgp130fl/wt control mice.

FIGURE 6.

Lgp130CD4/wt mice suffer from altered platelet activation accompanied by anemia and thrombocytosis. (A and B) RBC count (A) and platelet count (B) of Lgp130CD4/wt and Lgp130fl/wt mice. In total for (A) and (B), Lgp130fl/wt (n = 12) and Lgp130CD4/wt (n = 8) mice were used in three independent experiments. Data represent ± SEM of each group. (C) Platelet size of Lgp130CD4/wt and Lgp130fl/wt mice as measured via flow cytometry using the geometric mean of GPIb-positive platelets. In total, Lgp130fl/wt (n = 9) and Lgp130CD4/wt (n = 7) mice were used from two experiments. Data represent ± SEM of each group. (D) Representative images from the spleen and BM tissue of Lgp130CD4/wt and Lgp130fl/wt mice were stained by H&E. Arrowheads indicate MKs. In total, Lgp130fl/wt (n = 5) and Lgp130CD4/wt (n = 5) mice were used in a single experiment with this mouse cohort. MK numbers were determined in paraffin-embedded (E) spleen tissue sections and (F) BM (femur) of Lgp130CD4/wt and Lgp130fl/wt mice. In total for (E) and (F), Lgp130fl/wt (n = 5) and Lgp130CD4/wt (n = 5) mice were used in a single experiment with this mouse cohort. Data represent ± SEM of each group. p values were determined by unpaired t test including Welch’s correction. (G) Analysis of P-selectin activation on the surface of platelets derived from Lgp130CD4/wt and Lgp130fl/wt mice upon treatment with the indicated agonists. In total, Lgp130fl/wt (n = 7) and Lgp130CD4/wt (n = 7) mice were used. Data represent ± SEM of each group. (H) Activation of αIIbβ3 integrin of Lgp130CD4/wt and Lgp130fl/wt mice on platelet surface with indicated agonists. In total, Lgp130fl/wt (n = 7) and Lgp130CD4/wt (n = 7) mice were used. Data represent ± SEM of each group. (I) Expression of indicated glycoproteins on the surface of platelets from Lgp130CD4/wt and Lgp130fl/wt mice measured via mean fluorescence intensity (MFI) using flow cytometric analysis. In total, Lgp130fl/wt (n = 7) and Lgp130CD4/wt (n = 7) mice were used. Data represent ± SEM of each group. (J) Gene expressions of MgI, IL-6R, Asgr1, Asgr2, and Tpo in liver samples from Lgp130CD4/wt and Lgp130fl/wt mice were analyzed using the 2−ΔΔCt method. In total, Lgp130fl/wt (n = 4–5) and Lgp130CD4/wt (n = 6–7) mice were used. Data represent ± SEM of each group. p values were determined by unpaired t test including Welch’s correction. Data in (G)–(J) represent data aggregated from two experiments. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 6.

Lgp130CD4/wt mice suffer from altered platelet activation accompanied by anemia and thrombocytosis. (A and B) RBC count (A) and platelet count (B) of Lgp130CD4/wt and Lgp130fl/wt mice. In total for (A) and (B), Lgp130fl/wt (n = 12) and Lgp130CD4/wt (n = 8) mice were used in three independent experiments. Data represent ± SEM of each group. (C) Platelet size of Lgp130CD4/wt and Lgp130fl/wt mice as measured via flow cytometry using the geometric mean of GPIb-positive platelets. In total, Lgp130fl/wt (n = 9) and Lgp130CD4/wt (n = 7) mice were used from two experiments. Data represent ± SEM of each group. (D) Representative images from the spleen and BM tissue of Lgp130CD4/wt and Lgp130fl/wt mice were stained by H&E. Arrowheads indicate MKs. In total, Lgp130fl/wt (n = 5) and Lgp130CD4/wt (n = 5) mice were used in a single experiment with this mouse cohort. MK numbers were determined in paraffin-embedded (E) spleen tissue sections and (F) BM (femur) of Lgp130CD4/wt and Lgp130fl/wt mice. In total for (E) and (F), Lgp130fl/wt (n = 5) and Lgp130CD4/wt (n = 5) mice were used in a single experiment with this mouse cohort. Data represent ± SEM of each group. p values were determined by unpaired t test including Welch’s correction. (G) Analysis of P-selectin activation on the surface of platelets derived from Lgp130CD4/wt and Lgp130fl/wt mice upon treatment with the indicated agonists. In total, Lgp130fl/wt (n = 7) and Lgp130CD4/wt (n = 7) mice were used. Data represent ± SEM of each group. (H) Activation of αIIbβ3 integrin of Lgp130CD4/wt and Lgp130fl/wt mice on platelet surface with indicated agonists. In total, Lgp130fl/wt (n = 7) and Lgp130CD4/wt (n = 7) mice were used. Data represent ± SEM of each group. (I) Expression of indicated glycoproteins on the surface of platelets from Lgp130CD4/wt and Lgp130fl/wt mice measured via mean fluorescence intensity (MFI) using flow cytometric analysis. In total, Lgp130fl/wt (n = 7) and Lgp130CD4/wt (n = 7) mice were used. Data represent ± SEM of each group. (J) Gene expressions of MgI, IL-6R, Asgr1, Asgr2, and Tpo in liver samples from Lgp130CD4/wt and Lgp130fl/wt mice were analyzed using the 2−ΔΔCt method. In total, Lgp130fl/wt (n = 4–5) and Lgp130CD4/wt (n = 6–7) mice were used. Data represent ± SEM of each group. p values were determined by unpaired t test including Welch’s correction. Data in (G)–(J) represent data aggregated from two experiments. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

The STAT3/IL-6 axis induces ROS in an IGFBP5-dependent manner (35, 36). As a consequence, ROS-induced DNA damage induces the senescence-induced secretory pathway and p53 stabilization, which resulted in cell-cycle arrest and ultimately in cellular senescence (35–37). IL-6 knockout mice have a reduced p53 level and show fewer signs of cellular senescence (38–40), and IL-6 in complex with the sIL-6R induces premature senescence in human fibroblasts (35). Apart from constitutive activation of STAT3 in Lgp130CD4/wt mice, these mice showed overall increased IL-6 mRNA levels and an increase of IL-6 and sIL-6R protein in the serum compared with Lgp130fl/wt control mice (Fig. 4B, 4C). Therefore, we analyzed p53 expression and senescence in Lgp130CD4/wt mice. By using Western blotting, we observed a strong increase of p53 levels in the heart, liver, and spleen of Lgp130CD4/wt mice as compared with Lgp130fl/wt mice (Fig. 7A). In line with this, also the senescence-associated cell-cycle inhibitor p21WAF was strongly upregulated on the mRNA level in heart, liver, and spleen of Lgp130CD4/wt mice as compared with Lgp130fl/wt mice (Fig. 7B). Senescence-associated β-galactosidase staining was performed using spleen and heart tissues. Whereas the spleen seemed to be rather unaffected, a significant increase in senescence-associated β-galactosidase staining of heart sections was found in Lgp130CD4/wt mice compared with Lgp130fl/wt mice (Fig. 7C). Taken together, our data indicate an increased senescence phenotype of Lgp130CD4/wt mice that likely contributes to premature aging and death of the mice.

FIGURE 7.

Senescence-like phenotype in mice with continuously T cell–directed gp130 activation. (A) Western blot against p53 and GAPDH in heart, spleen, and liver from 8- to 12-wk-old Lgp130CD4/wt (n = 5–6) and Lgp130fl/wt mice (n = 6). Equal amounts of proteins (50 µg/lane) were analyzed via specific Abs detecting p53. Evaluated with LiCor Image Studio Lite Ver 5.2. (B) Gene expression of p21 in heart, liver, and spleen from 8- to 12-wk-old Lgp130CD4/wt (n = 5) and Lgp130fl/wt mice (n = 5) was analyzed using the 2−ΔΔCt method. Data represent ± SEM of each group. (C) Mean fluorescence intensity of senescence β-galactosidase activity assay of spleen and heart from Lgp130CD4/wt and Lgp130fl/wt mice. β-Galactosidase activity was measured by fluorescence and plate-based assay kit (excitation wavelength: 360 nm; excitation wavelength: 465 nm). In total, Lgp130fl/wt (n = 9) and Lgp130CD4/wt (n = 7–9) mice were used. Data represent ± SEM of each group. p values were determined by two-way ANOVA, including Tukey correction. Data presented in (A) and (B) represent a single experiment with this mouse cohort, whereas data in (C) were performed in two independent experiments. *p < 0.05, ***p < 0.001.

FIGURE 7.

Senescence-like phenotype in mice with continuously T cell–directed gp130 activation. (A) Western blot against p53 and GAPDH in heart, spleen, and liver from 8- to 12-wk-old Lgp130CD4/wt (n = 5–6) and Lgp130fl/wt mice (n = 6). Equal amounts of proteins (50 µg/lane) were analyzed via specific Abs detecting p53. Evaluated with LiCor Image Studio Lite Ver 5.2. (B) Gene expression of p21 in heart, liver, and spleen from 8- to 12-wk-old Lgp130CD4/wt (n = 5) and Lgp130fl/wt mice (n = 5) was analyzed using the 2−ΔΔCt method. Data represent ± SEM of each group. (C) Mean fluorescence intensity of senescence β-galactosidase activity assay of spleen and heart from Lgp130CD4/wt and Lgp130fl/wt mice. β-Galactosidase activity was measured by fluorescence and plate-based assay kit (excitation wavelength: 360 nm; excitation wavelength: 465 nm). In total, Lgp130fl/wt (n = 9) and Lgp130CD4/wt (n = 7–9) mice were used. Data represent ± SEM of each group. p values were determined by two-way ANOVA, including Tukey correction. Data presented in (A) and (B) represent a single experiment with this mouse cohort, whereas data in (C) were performed in two independent experiments. *p < 0.05, ***p < 0.001.

Close modal

All known IL-6–type family members signal via the signal transducing β-subunit gp130 with different biological outcomes and efficacies (41). Several variants of constitutively active gp130 have been described in the literature combined with numerous diseases underlining the importance of the gp130/STAT3 axis. Downstream of gp130, the gain of function of gp130 was early described to develop autoimmune diseases such as RA in mice (42, 43). Recently, it was shown that a B cell–specific constitutively active gp130 variant induced mature lymphoma and plasmacytoma (24). Our study (to our knowledge) and the accompanying study by Heinig et al. (44) define a novel mouse model that mimics T cell–specific gp130/STAT3 signaling in a cytokine-independent manner. Lgp130CD4/wt mice show massive phenotypical abnormalities, including splenomegaly, lymphadenopathy, and an upregulation of hyperinflammatory and autoimmune-like signatures including autoantibodies (45, 46). Furthermore, permanent genetically driven JAK/STAT pathway activation in T cells showed premature aging based on upregulation of cell-cycle markers, accompanied by high levels of the tumor suppressor p53 and increased expression of the cyclin-dependent kinase inhibitor p21WAF. This accumulation of multiple senescence markers together with chronic inflammation might lead to the observed morphological changes in the organs, lastly resulting in multiorgan failure and the observed aging phenotype and strongly reduced survival. This hypothesis is supported by Kojima et al. (36), who already provided evidence that IL-6, and especially the IL-6/STAT3 axis, induced senescence in human fibroblasts. Our data are consistent with reports describing the complex role of Treg cells in immunity and aging, which is also in connection with increased numbers of cytotoxic and Treg cells seen in the T cell–restricted Lgp130 mice (47, 48). The accompanying phenomenon of inflammation and premature aging is summarized as inflammaging, which was first introduced by Franceschi et al. (49) in 2000 as a new branch of aging studies. Inflammaging itself is associated with many aging-associated diseases, such as Alzheimer’s, heart disease (i.e., myocardial infarction, aortic aneurysm), atherosclerosis, cancer, and type II diabetes, and is often linked to and potentially caused by immunosenescence. However, the underlying mechanisms are poorly understood (50–51). Moreover, T cell–restricted expression of Lgp130 and its permanent inflammatory signature showed severe effects on hemostasis. Besides an elevated platelet production and an increase in the number of MKs in spleen and BM, platelet clearance was permanently increased and accompanied by secondary anemia. Consistent with this, platelets seemed to be overreactive that manifested in a reactive thrombocytosis leading to defects in platelets activation and shortened life span in Lgp130CD4/wt mice. Collectively, our results are consistent with the results described by Kirito et al. (52) in 2002, as well as the indispensable role of STAT3 signaling in the early stage of megakaryopoiesis, presumably in the regulation of expansion of megakaryocytic progenitor cells. Abnormal thrombopoiesis via elevated IL-6 production, also seen in our study, results in thrombocytosis and contributes to a high platelet count, which is potentially associated with thrombosis and death (53, 54). In summary, to our knowledge, this study presents the first causal link between a hyperactivation of the STAT3/IL-6 axis in T cells and inflammaging. Moreover, the Lgp130CD4/wt mouse model can be used for further and detailed senescence studies, especially in connection with inflammaging. For the treatment of RA, TNF-α inhibitors, such as etanercept, were used until the IL-6R inhibitor tocilizumab outperformed anti–TNF-α agents in effectiveness (55–58). High levels of IL-6 and TNF-α are also associated in the elderly with increased risk of morbidity and mortality (59, 60). For these reasons, a treatment with an anti–IL-6 agent or JAK inhibitors to alleviate the inflammaging phenotype would be of interest as a novel proof-of-principle strategy to develop antiaging interventions. Another important strategy to be considered for further investigations would be to effectively eliminate senescent cells by compounds termed “senolytics.” Previous studies have shown that senolytics target antiapoptotic pathways and eliminate certain types of senescent cell with the potential to improve physiological functions in several tissues (61, 62).

The authors have no financial conflicts of interest.

We thank Martina Spelleken for technical assistance. We are grateful to S.R.-J. (Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany) for the provision of the Lgp130fl/fl mice. We thank SFB 1116 “Master Switches bei kardialer Ischämie” for funding this project.

This work was supported by the Deutsche Forschungsgemeinschaft (German Research Foundation; Grants RTG1949 and SFB 1116; Grant EL 651/8-1 to M.E.) and the Jürgen Manchot Foundation (MOI IV).

The online version of this article contains supplemental material.

AMR

Ashwell-Morell receptor

AAMY

amylase

Asgr

asialoglycoprotein receptor

BM

bone marrow

Glu

glucose

GOT

glutamate-oxalacetate transaminase

HDLc

high-density lipoprotein

MK

megakaryocyte

RA

rheumatoid arthritis

RF

rheumatoid factor

sIL-6R

soluble IL-6R

TPO

thrombopoietin

Treg

regulatory T

wt

wild-type

1
Hirano
,
T.
2021
.
IL-6 in inflammation, autoimmunity and cancer
.
Int. Immunol.
33
:
127
148
.
2
Kimura
,
A.
,
T.
Kishimoto
.
2010
.
IL-6: regulator of Treg/Th17 balance
.
Eur. J. Immunol.
40
:
1830
1835
.
3
Tanaka
,
T.
,
T.
Kishimoto
.
2014
.
The biology and medical implications of interleukin-6
.
Cancer Immunol. Res.
2
:
288
294
.
4
Burger
,
R.
2013
.
Impact of interleukin-6 in hematological malignancies
.
Transfus. Med. Hemother.
40
:
336
343
.
5
Gadó
,
K.
,
G.
Domján
,
H.
Hegyesi
,
A.
Falus
.
2000
.
Role of INTERLEUKIN-6 in the pathogenesis of multiple myeloma
.
Cell Biol. Int.
24
:
195
209
.
6
Giraldez
,
M. D.
,
D.
Carneros
,
C.
Garbers
,
S.
Rose-John
,
M.
Bustos
.
2021
.
New insights into IL-6 family cytokines in metabolism, hepatology and gastroenterology
.
Nat. Rev. Gastroenterol. Hepatol.
18
:
787
803
.
7
Garbers
,
C.
,
H. M.
Hermanns
,
F.
Schaper
,
G.
Müller-Newen
,
J.
Grötzinger
,
S.
Rose-John
,
J.
Scheller
.
2012
.
Plasticity and cross-talk of interleukin 6-type cytokines
.
Cytokine Growth Factor Rev.
23
:
85
97
.
8
Garbers
,
C.
,
J.
Scheller
.
2013
.
Interleukin-6 and interleukin-11: same same but different
.
Biol. Chem.
394
:
1145
1161
.
9
Grötzinger
,
J.
,
T.
Kernebeck
,
K. J.
Kallen
,
S.
Rose-John
.
1999
.
IL-6 type cytokine receptor complexes: hexamer, tetramer or both?
Biol. Chem.
380
:
803
813
.
10
Heinrich
,
P. C.
,
I.
Behrmann
,
G.
Müller-Newen
,
F.
Schaper
,
L.
Graeve
.
1998
.
Interleukin-6-type cytokine signalling through the gp130/Jak/STAT pathway
.
Biochem. J.
334
:
297
314
.
11
Taga
,
T.
,
T.
Kishimoto
.
1997
.
Gp130 and the interleukin-6 family of cytokines
.
Annu. Rev. Immunol.
15
:
797
819
.
12
Carpenter
,
R. L.
,
H. W.
Lo
.
2014
.
STAT3 target genes relevant to human cancers
.
Cancers (Basel)
6
:
897
925
.
13
Haghikia
,
A.
,
M.
Ricke-Hoch
,
B.
Stapel
,
I.
Gorst
,
D.
Hilfiker-Kleiner
.
2014
.
STAT3, a key regulator of cell-to-cell communication in the heart
.
Cardiovasc. Res.
102
:
281
289
.
14
Bromberg
,
J.
2002
.
Stat proteins and oncogenesis
.
J. Clin. Invest.
109
:
1139
1142
.
15
Ohgami
,
R. S.
,
L.
Ma
,
A.
Monabati
,
J. L.
Zehnder
,
D. A.
Arber
.
2014
.
STAT3 mutations are present in aggressive B-cell lymphomas including a subset of diffuse large B-cell lymphomas with CD30 expression
.
Haematologica
99
:
e105
e107
.
16
Jerez
,
A.
,
M. J.
Clemente
,
H.
Makishima
,
H.
Koskela
,
F.
Leblanc
,
K.
Peng Ng
,
T.
Olson
,
B.
Przychodzen
,
M.
Afable
,
I.
Gomez-Segui
, et al
.
2012
.
STAT3 mutations unify the pathogenesis of chronic lymphoproliferative disorders of NK cells and T-cell large granular lymphocyte leukemia
.
Blood
120
:
3048
3057
.
17
Heim
,
D.
,
I.
Gil-Ibanez
,
J.
Herden
,
A. C.
Parplys
,
K.
Borgmann
,
D.
Schmidt-Arras
,
A. W.
Lohse
,
S.
Rose-John
,
H.
Wege
.
2016
.
Constitutive gp130 activation rapidly accelerates the transformation of human hepatocytes via an impaired oxidative stress response
.
Oncotarget
7
:
55639
55648
.
18
Banerjee
,
P.
,
S.
Kotla
,
L.
Reddy Velatooru
,
R. J.
Abe
,
E. A.
Davis
,
J. P.
Cooke
,
K.
Schadler
,
A.
Deswal
,
J.
Herrmann
,
S. H.
Lin
, et al
.
2021
.
Senescence-associated secretory phenotype as a hinge between cardiovascular diseases and cancer
.
Front. Cardiovasc. Med.
8
:
763930
.
19
Gorgoulis
,
V.
,
P. D.
Adams
,
A.
Alimonti
,
D. C.
Bennett
,
O.
Bischof
,
C.
Bishop
,
J.
Campisi
,
M.
Collado
,
K.
Evangelou
,
G.
Ferbeyre
, et al
.
2019
.
Cellular senescence: defining a path forward
.
Cell
179
:
813
827
.
20
Chandra
,
A.
,
J.
Rajawat
.
2021
.
Skeletal aging and osteoporosis: mechanisms and therapeutics
.
Int. J. Mol. Sci.
22
:
3553
.
21
Kandhaya-Pillai
,
R.
,
F.
Miro-Mur
,
J.
Alijotas-Reig
,
T.
Tchkonia
,
J. L.
Kirkland
,
S.
Schwartz
.
2017
.
TNFα-senescence initiates a STAT-dependent positive feedback loop, leading to a sustained interferon signature, DNA damage, and cytokine secretion
.
Aging (Albany NY)
9
:
2411
2435
.
22
Orjalo
,
A. V.
,
D.
Bhaumik
,
B. K.
Gengler
,
G. K.
Scott
,
J.
Campisi
.
2009
.
Cell surface-bound IL-1alpha is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network
.
Proc. Natl. Acad. Sci. USA
106
:
17031
17036
.
23
Stuhlmann-Laeisz
,
C.
,
S.
Lang
,
A.
Chalaris
,
P.
Krzysztof
,
S.
Enge
,
J.
Eichler
,
U.
Klingmüller
,
M.
Samuel
,
M.
Ernst
,
S.
Rose-John
,
J.
Scheller
.
2006
.
Forced dimerization of gp130 leads to constitutive STAT3 activation, cytokine-independent growth, and blockade of differentiation of embryonic stem cells
.
Mol. Biol. Cell
17
:
2986
2995
.
24
Scherger
,
A. K.
,
M.
Al-Maarri
,
H. C.
Maurer
,
M.
Schick
,
S.
Maurer
,
R.
Öllinger
,
I.
Gonzalez-Menendez
,
M.
Martella
,
M.
Thaler
,
K.
Pechloff
, et al
.
2019
.
Activated gp130 signaling selectively targets B cell differentiation to induce mature lymphoma and plasmacytoma
.
JCI Insight
4
:
e128435
.
25
Bogdanos
,
D. P.
,
B.
Gao
,
M. E.
Gershwin
.
2013
.
Liver immunology
.
Compr. Physiol.
3
:
567
598
.
26
MacParland
,
S. A.
,
J. C.
Liu
,
X. Z.
Ma
,
B. T.
Innes
,
A. M.
Bartczak
,
B. K.
Gage
,
J.
Manuel
,
N.
Khuu
,
J.
Echeverri
,
I.
Linares
, et al
.
2018
.
Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations
.
Nat. Commun.
9
:
4383
.
27
Godfrey
,
D. I.
,
J.
Kennedy
,
T.
Suda
,
A.
Zlotnik
.
1993
.
A developmental pathway involving four phenotypically and functionally distinct subsets of CD3-CD4-CD8- triple-negative adult mouse thymocytes defined by CD44 and CD25 expression
.
J. Immunol.
150
:
4244
4252
.
28
Lamertz
,
L.
,
D. M.
Floss
,
J.
Scheller
.
2018
.
Combined deletion of the fibronectin-type III domains and the stalk region results in ligand-independent, constitutive activation of the Interleukin 6 signal-transducing receptor gp130
.
Cytokine
110
:
428
434
.
29
Rinis
,
N.
,
A.
Küster
,
H.
Schmitz-Van de Leur
,
A.
Mohr
,
G.
Müller-Newen
.
2014
.
Intracellular signaling prevents effective blockade of oncogenic gp130 mutants by neutralizing antibodies
.
Cell Commun. Signal.
12
:
14
.
30
Staels
,
B.
,
V. A.
Fonseca
.
2009
.
Bile acids and metabolic regulation: mechanisms and clinical responses to bile acid sequestration
.
Diabetes Care
32
(
Suppl. 2
):
S237
S245
.
31
Periayah
,
M. H.
,
A. S.
Halim
,
A. Z.
Mat Saad
.
2017
.
Mechanism action of platelets and crucial blood coagulation pathways in hemostasis
.
Int. J. Hematol. Oncol. Stem Cell Res.
11
:
319
327
.
32
Ahmed
,
M. U.
,
N.
Receveur
,
E.
Janus-Bell
,
C.
Mouriaux
,
C.
Gachet
,
M.
Jandrot-Perrus
,
B.
Hechler
,
E. E.
Gardiner
,
P. H.
Mangin
.
2021
.
Respective roles of glycoprotein VI and FcγRIIA in the regulation of αIIbβ3-mediated platelet activation to fibrinogen, thrombus buildup, and stability
.
Res. Pract. Thromb. Haemost.
5
:
e12551
.
33
Grozovsky
,
R.
,
A. J.
Begonja
,
K.
Liu
,
G.
Visner
,
J. H.
Hartwig
,
H.
Falet
,
K. M.
Hoffmeister
.
2015
.
The Ashwell-Morell receptor regulates hepatic thrombopoietin production via JAK2-STAT3 signaling
.
Nat. Med.
21
:
47
54
.
34
Kaser
,
A.
,
G.
Brandacher
,
W.
Steurer
,
S.
Kaser
,
F. A.
Offner
,
H.
Zoller
,
I.
Theurl
,
W.
Widder
,
C.
Molnar
,
O.
Ludwiczek
, et al
.
2001
.
Interleukin-6 stimulates thrombopoiesis through thrombopoietin: role in inflammatory thrombocytosis
.
Blood
98
:
2720
2725
.
35
Kojima
,
H.
,
H.
Kunimoto
,
T.
Inoue
,
K.
Nakajima
.
2012
.
The STAT3-IGFBP5 axis is critical for IL-6/gp130-induced premature senescence in human fibroblasts
.
Cell Cycle
11
:
730
739
.
36
Kojima
,
H.
,
T.
Inoue
,
H.
Kunimoto
,
K.
Nakajima
.
2013
.
IL-6-STAT3 signaling and premature senescence
.
JAK-STAT
2
:
e25763
.
37
Kim
,
Y. Y.
,
H. J.
Jee
,
J. H.
Um
,
Y. M.
Kim
,
S. S.
Bae
,
J.
Yun
.
2017
.
Cooperation between p21 and Akt is required for p53-dependent cellular senescence
.
Aging Cell
16
:
1094
1103
.
38
Bonda
,
T. A.
,
M.
Dziemidowicz
,
M.
Cieslinska
,
E.
Tarasiuk
,
N.
Wawrusiewicz-Kurylonek
,
I.
Bialuk
,
M. M.
Winnicka
,
K. A.
Kaminski
.
2019
.
Interleukin 6 knockout inhibits aging-related accumulation of p53 in the mouse myocardium
.
J. Gerontol. A Biol. Sci. Med. Sci.
74
:
176
182
.
39
Bialuk
,
I.
,
M.
Cieślińska
,
O.
Kowalczuk
,
T. A.
Bonda
,
J.
Nikliński
,
M. M.
Winnicka
.
2020
.
IL-6 deficiency attenuates p53 protein accumulation in aged male mouse hippocampus
.
Biogerontology
21
:
29
43
.
40
Li
,
Y.
,
L.
Lu
,
Y.
Xie
,
X.
Chen
,
L.
Tian
,
Y.
Liang
,
H.
Li
,
J.
Zhang
,
Y.
Liu
,
X.
Yu
.
2021
.
Interleukin-6 knockout inhibits senescence of bone mesenchymal stem cells in high-fat diet-induced bone loss
.
Front. Endocrinol. (Lausanne)
11
:
622950
.
41
Metcalfe
,
R. D.
,
T. L.
Putoczki
,
M. D. W.
Griffin
.
2020
.
Structural understanding of interleukin 6 family cytokine signaling and targeted therapies: focus on interleukin 11
.
Front. Immunol.
11
:
1424
.
42
Silver
,
J. S.
,
C. A.
Hunter
.
2010
.
gp130 at the nexus of inflammation, autoimmunity, and cancer
.
J. Leukoc. Biol.
88
:
1145
1156
.
43
Banerjee
,
S.
,
A.
Biehl
,
M.
Gadina
,
S.
Hasni
,
D. M.
Schwartz
.
2017
.
JAK-STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects. [Published errata appear in 2017 Drugs 77: 939 and 2017 Drugs 77: 1261.]
Drugs
77
:
521
546
.
44
Heinig
,
L. C.
,
E. V. M.
Huth
,
K.
Yan
,
N.
Schumacher
,
M.
Nawrocki
,
N. C.
Lory
,
P.
Bradtke
,
T.
Bertram
,
G.
Rattay
,
J.
Schmid
, et al
.
2023
.
Cell-autonomous constitutive gp130 signaling in T cells amplifies TH17 cell responses and causes severe lung inflammation
.
J. Immunol.
210
:
1717
1727
.
45
Auerbach
,
A.
,
T. A.
Summers
,
B.
Zhang
,
N. S.
Aguilera
.
2013
.
Splenic manifestations of chronic autoimmune disorder: a report of five cases with histiocytic necrotizing change in four cases
.
Histopathology
63
:
19
28
.
46
Prado
,
C.
,
B.
de Paz
,
J.
Gómez
,
P.
López
,
J.
Rodríguez-Carrio
,
A.
Suárez
.
2011
.
Glucocorticoids enhance Th17/Th1 imbalance and signal transducer and activator of transcription 3 expression in systemic lupus erythematosus patients
.
Rheumatology (Oxford)
50
:
1794
1801
.
47
Rocamora-Reverte
,
L.
,
F. L.
Melzer
,
R.
Würzner
,
B.
Weinberger
.
2021
.
The complex role of regulatory t cells in immunity and aging
.
Front. Immunol.
11
:
616949
.
48
Jagger
,
A.
,
Y.
Shimojima
,
J. J.
Goronzy
,
C. M.
Weyand
.
2014
.
Regulatory T cells and the immune aging process: a mini-review
.
Gerontology
60
:
130
137
.
49
Franceschi
,
C.
,
M.
Bonafè
,
S.
Valensin
,
F.
Olivieri
,
M.
De Luca
,
E.
Ottaviani
,
G.
De Benedictis
.
2000
.
Inflamm-aging. An evolutionary perspective on immunosenescence
.
Ann. N. Y. Acad. Sci.
908
:
244
254
.
50
Xia
,
S.
,
X.
Zhang
,
S.
Zheng
,
R.
Khanabdali
,
B.
Kalionis
,
J.
Wu
,
W.
Wan
,
X.
Tai
.
2016
.
An update on inflamm-aging: mechanisms, prevention, and treatment
.
J. Immunol. Res.
2016
:
8426874
.
51
Ferrucci
,
L.
,
E.
Fabbri
.
2018
.
Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty
.
Nat. Rev. Cardiol.
15
:
505
522
.
52
Kirito
,
K.
,
M.
Osawa
,
H.
Morita
,
R.
Shimizu
,
M.
Yamamoto
,
A.
Oda
,
H.
Fujita
,
M.
Tanaka
,
K.
Nakajima
,
Y.
Miura
, et al
.
2002
.
A functional role of Stat3 in in vivo megakaryopoiesis
.
Blood
99
:
3220
3227
.
53
Zhang
,
L.
,
R.
Lukowski
,
F.
Gaertner
,
M.
Lorenz
,
K. R.
Legate
,
K.
Domes
,
E.
Angermeier
,
F.
Hofmann
,
S.
Massberg
.
2013
.
Thrombocytosis as a response to high interleukin-6 levels in cGMP-dependent protein kinase I mutant mice
.
Arterioscler. Thromb. Vasc. Biol.
33
:
1820
1828
.
54
Ertenli
,
I.
,
S.
Kiraz
,
M. A.
Oztürk
,
I.
Haznedaroğlu
,
I.
Çelik
,
M.
Calgüneri
.
2003
.
Pathologic thrombopoiesis of rheumatoid arthritis
.
Rheumatol. Int.
23
:
49
60
.
55
Burmester
,
G. R.
,
J. E.
Pope
.
2017
.
Novel treatment strategies in rheumatoid arthritis
.
Lancet
389
:
2338
2348
.
56
Choy
,
E. H.
,
C.
Bernasconi
,
M.
Aassi
,
J. F.
Molina
,
O. M.
Epis
.
2017
.
Treatment of rheumatoid arthritis with anti-tumor necrosis factor or tocilizumab therapy as first biologic agent in a global comparative observational study
.
Arthritis Care Res. (Hoboken)
69
:
1484
1494
.
57
Ma
,
X.
,
S.
Xu
.
2013
.
TNF inhibitor therapy for rheumatoid arthritis
.
Biomed. Rep.
1
:
177
184
.
58
Biggioggero
,
M.
,
C.
Crotti
,
A.
Becciolini
,
E. G.
Favalli
.
2018
.
Tocilizumab in the treatment of rheumatoid arthritis: an evidence-based review and patient selection
.
Drug Des. Devel. Ther.
13
:
57
70
.
59
Michaud
,
M.
,
L.
Balardy
,
G.
Moulis
,
C.
Gaudin
,
C.
Peyrot
,
B.
Vellas
,
M.
Cesari
,
F.
Nourhashemi
.
2013
.
Proinflammatory cytokines, aging, and age-related diseases
.
J. Am. Med. Dir. Assoc.
14
:
877
882
.
60
Bruunsgaard
,
H.
,
S.
Ladelund
,
A. N.
Pedersen
,
M.
Schroll
,
T.
Jørgensen
,
B. K.
Pedersen
.
2003
.
Predicting death from tumour necrosis factor-alpha and interleukin-6 in 80-year-old people
.
Clin. Exp. Immunol.
132
:
24
31
.
61
Xu
,
M.
,
T.
Pirtskhalava
,
J. N.
Farr
,
B. M.
Weigand
,
A. K.
Palmer
,
M. M.
Weivoda
,
C. L.
Inman
,
M. B.
Ogrodnik
,
C. M.
Hachfeld
,
D. G.
Fraser
, et al
.
2018
.
Senolytics improve physical function and increase lifespan in old age
.
Nat. Med.
24
:
1246
1256
.
62
Yousefzadeh
,
M. J.
,
Y.
Zhu
,
S. J.
McGowan
,
L.
Angelini
,
H.
Fuhrmann-Stroissnigg
,
M.
Xu
,
Y. Y.
Ling
,
K. I.
Melos
,
T.
Pirtskhalava
,
C. L.
Inman
, et al
.
2018
.
Fisetin is a senotherapeutic that extends health and lifespan
.
EBioMedicine
36
:
18
28
.

Supplementary data