Systemic lupus erythematosus is an autoimmune disease characterized by excessive inflammation and production of pathogenic Abs. Histone deacetylase 6 (HDAC6) is a class IIb histone deacetylase. It has been reported that selective HDAC6 inhibition decreases inflammation in lupus mouse models. In this study, sex- and age-matched wild-type (WT) and HDAC6−/− mice on the C57BL/6 background were administered 0.5 ml of pristane or PBS i.p. at 8–12 wk of age and were euthanized 10 d later. At sacrifice, body weight and spleen weight were measured, sera were collected, and splenocytes and peritoneal cells were harvested for flow cytometry. We found pristane administration increased the spleen weight with no difference between WT and HDAC6−/− mice. Pristane administration promoted the population of CD11b+Ly6C++ inflammatory monocytes and CD11b+Ly6G+ neutrophils. Peritoneal recruitment of these inflammatory monocytes and neutrophils was significantly decreased in HDAC6−/− mice compared with the WT mice. Flow cytometry results showed that the number of CD69+ T and B cells was increased in HDAC6−/− mice. Pristane administration also induced the IFN signature genes as determined by RT-qPCR. Furthermore, IFN signature genes were not affected in HDAC6−/− mice compared with the WT mice. In vitro studies in J774A.1 cells revealed that the selective HDAC6 inhibitor (ACY-738) increased acetylation of NF-κB while increasing Stat1 phosphorylation, which resulted in inducible NO synthase production in LPS/IFN-γ–stimulated cells. Taken together, these results demonstrate that although HDAC6 inhibition may inhibit some inflammatory pathways, others remain unaffected.

Systemic lupus erythematosus (SLE) is an autoimmune disease in which body tissues and organs are attacked by their own immune system (1). In SLE, autoantibodies are generated, resulting in the formation of circulating Ag–Ab complexes that become lodged in tiny capillaries and different organ systems, including the kidney and skin (2). Histone acetylation has been shown to play a critical role in controlling gene expression (3, 4). The process of acetylation of histone proteins frequently leads to enhanced transcription, whereas deacetylation is linked to gene repression (5). We and others have previously reported that HDAC inhibitors showed efficacy in the treatment of allergy, cancer, and autoimmune diseases, in particular SLE (6). However, the use of pan-HDAC inhibitors has shown significant adverse effects limiting the use for chronic conditions (7). HDAC6 modulates protein degradation by deacetylating nonhistone proteins, such as heat shock protein 90, α-tubulin, and NF-κB, and has been shown to play a role in the modulation of immune response (8). It has been demonstrated that overexpression of HDAC6 in macrophages significantly increased the expression of proinflammatory cytokines, including IL-6, IL-1β, and TNF-α, via the upregulation of AP-1 and NF-κB signaling pathways (9). HDAC6 inhibition has been shown to interfere with the NF-κB signaling pathway and decrease inflammation by increasing acetylation of NF-κB, which results in decreased nuclear translocation (10, 11).

One commonly used animal model to study lupus is the pristane-induced disease model. Pristane (2, 6, 10, and 14 tetramethylpentadecane) is a naturally occurring branched alkane found in mineral oil and in low concentration in vegetables (12). When injected into the peritoneum in mice, pristane induces lupus-like symptoms over time, including the activation of immune cells and the production of autoantibodies against the host’s own DNA and nuclear Ags (13). This resembles the autoimmune response observed in human SLE. Interestingly, after pristane administration, in the first 2 wk, severe alveolar hemorrhage occurs in C57BL/6 (B6) mice; symptoms include endothelial damage, hemorrhage, and alveolar and perivascular inflammation (small vessel vasculitis and capillaritis) (14). This leads to a mortality rate as high as 20% in the pristane-treated B6 mice (15). In our current studies, we sought to determine whether HADC6 deletion would decrease the inflammation seen with pristane administration. Furthermore, we sought to determine the mechanism by which HADC6 inhibition alters macrophage activation.

Female wild-type (WT) B6 mice and HDAC6 knockout (KO) mice on B6 background were purchased from The Jackson Lab (Bar Harbor, ME). The animals were kept in accordance with the Institutional Animal Care and Use Committee at Virginia Tech College of Veterinary Medicine, in a pathogen-free environment with a regular 12-h light/dark cycle. All animal procedures were performed in compliance with the Institutional Animal Care and Use Committee–approved guidelines. WT and HDAC6 KO mice on the B6 background were administered a single dose of 0.5 ml of pristane (Sigma Aldrich, St. Louis, MO) or PBS i.p. at 8–12 wk of age. Mice were sacrificed at 10 d after pristane injection. At sacrifice, body weight and spleen weight were measured, sera were collected, and splenocytes and peritoneal cells were harvested for flow cytometry.

At sacrifice, spleens were collected and minced into small pieces, then transferred on the 70-μm cell strainer over a 50-ml conical tube. The spleen pieces were minced and pressed through the strainer with the plunger end of a syringe. The cells were washed through the strainer with PBS and then collected after centrifuge. The cell pellet was resuspended in 2–5 ml of cold 1× RBC Lysis buffer (eBioscience, San Diego, CA) for 5 min on ice to lyse the RBCs. The cell suspension was washed with cold PBS and centrifuged. Splenocytes were collected for flow cytometry analysis. Mouse peritoneal cells were extracted as previously published (16). In brief, each mouse’s abdominal skin was covered in 70% alcohol after euthanasia. A tiny incision was created so that the intact peritoneal wall could be seen. In the peritoneal cavity, 5 ml of ice-cold PBS was injected. Peritoneal fluid was collected after a little massage and centrifuged at 500 × g for 10 min at 4°C. The cells were then resuspended for flow cytometry.

Attune NxT (Thermo Fisher Scientific, Waltham, MA) or BD FACSAria II (BD Biosciences, San Jose, CA) flow cytometer was used for flow cytometry; peritoneal cells and splenocytes were extracted, blocked with anti-mouse CD16/32 (eBioscience), and stained with fluorochrome-conjugated Abs. Anti-mouse Abs used in this study included PerCP/Cy5.5 CD19 (115534; BioLegend), FITC CD3 (100204; BioLegend), allophycocyanin CD69 (104514; BioLegend), PE CD4 (BD 553730), PE/Cy7 CD138 (142513; BioLegend), PerCp/Cy5.5 CD11b (101227; BioLegend), allophycocyanin/Cy7 Ly6c (128025; BioLegend), and BV421 Ly6G (127627; BioLegend). The gating strategy for splenocytes is in Supplemental Fig. 1.

Using the RNeasy mini kit (Qiagen, Hilden, Germany) as directed by the manufacturer, total RNA was extracted, purified, and then reverse transcribed using the iScript cDNA Synthesis Kit (Bio-Rad, Hercules, CA). Real-time PCR was performed using SYBR Green master mix on an ABI 7500 fast system (Applied Biosystems, Foster City, CA). The endogenous reference genes were GAPDH. The comparative ΔCt method was used to calculate the relative mRNA expression levels. Every experiment’s samples were conducted in technical triplicates, with each sample consisting of three reaction wells that produced an average threshold cycle value. Primer sequences are listed as follows: Isg15, forward 5′-GGT GTC CGT GAC TAA CTC CAT-3′, reverse 5′-CTG TAC CAC TAG CAT CAC TGT G-3′; Mx1, forward 5′-GAT CCG ACT TCA CTT CCA GAT GG-3′, reverse 5′-CAT CTC AGT GGT AGT CAA CCC-3′; Irf9, forward 5′-TGT CTG GAA GAC TCG CCT AC-3′, reverse 5′-GCA ACA TCC ATA CGA CCT CTC T-3′; Ifnb, forward 5′-CAG CTC CAA GAA AGG ACG AAC-3′, reverse 5′-GGC AGT GTA ACT CTT CTG CAT-3′; Irf7, forward 5′-TGC TGT TTG GAG ACT GGC TAT-3′, reverse 5′-TCC AAG CTC CCG GCT AAG T-3′; Oas1a, forward 5′-GCC TGA TCC CAG AAT CTA TGC-3′, reverse 5′-GAG CAA CTC TAG GGC GTA CTG-3′; Cxcl10, forward 5′-CCA AGT GCT GCC GTC ATT TTC-3′, reverse 5′-GGC TCG CAG GGA TGA TTT CAA-3′; Gapdh, forward 5′-AGG TCG GTG TGA ACG GAT TTG-3′, reverse 5′-TGT AGA CCA TGT AGT TGA GGT CA-3′.

J774A.1 cells were purchased from ATCC (TIB-67) and cultured in DMEM supplemented with 10% heat-inactivated FBS, penicillin, and streptomycin (100 μg/ml) at 37°C in a 5% CO2 humidified incubator. When cells were 80% confluent, cells were treated with the selective HDAC6 inhibitor ACY-738 (Adooq Bioscience, Irvine, CA) and stimulated with LPS (Sigma Aldrich) and IFN-γ (R&D Systems, Minneapolis, MN) for 24 h.

BCA protein assay kit (Thermo Fisher Scientific) was used to measure protein concentrations after total protein was extracted and lysed in radioimmunoprecipitation buffer containing 1% protease and phosphatase inhibitor mixture (Thermo Fisher Scientific). Equal amounts of protein in the lysates were mixed with SDS loading buffer and boiled for 5 min. The protein samples were separated by electrophoresis on 4–12% SDS-PAGE gel (Bio-Rad) and transferred to polyvinylidenedifluoride membranes. The membrane was then blocked with 0.1% Tween 20 in TBS containing 5% BSA. The membranes were then probed with primary and secondary Abs. The blot was scanned on an Odyssey Clx Imager (LI-COR Biosciences, Lincoln, NE). All Abs were purchased from Cell Signaling Technology (Danvers, MA). The primary Abs dilution ratio is 1:1000. The Western blot Abs and catalog number are as follows: NF-κB p65 (8242S), acetyl-NF-κB p65 (Lys310) (12629S), inducible NO synthase (iNOS; 13120S), Histone H3 (4499T), acetyl-Histone H3 (Lys9) (9649T), acetyl-α-Tubulin (Lys40) (5335T), α-Tubulin (3873T), phospho-Stat1 (Tyr701) (9167T), Stat1 (14994T), phospho-NF-κB p65 (Ser536) (3033T), anti-mouse IgG (H + L) (5257P), and anti-rabbit IgG (H + L) (5151P). Using ImageJ software and densitometric analysis, we quantified and normalized the amounts of proteins to housekeeping protein.

The one-way ANOVA for multiple comparisons or the Student t test for single comparisons were used to evaluate statistical differences between sample groups. Data are shown as mean ± SEM. Statistical significance was determined by p value (*p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001).

The splenomegaly is one of the indicators of inflammation. To determine the role of HDAC6 in splenomegaly, we compared HDAC6−/− mice with WT mice 10 d after pristane injection. WT and HDAC6−/− mice on the B6 background were administered 0.5 ml of pristane or PBS at 12 wk of age. After 10 d of pristane injection, the mice were euthanized, and we measured body weight (Fig. 1A), spleen weight (Fig. 1B), and spleen-to-body weight ratio (Fig. 1C). Although body weights were not significantly altered in any group, spleen weights were significantly increased in the pristane-treated HDAC6−/− mice compared with the PBS-treated HDAC6−/− mice. In addition, the spleen-to-body weight ratio was significantly increased in the pristane-treated HDAC6−/− mice compared with the PBS-treated HDAC6−/− mice. The results show that HDAC6 deletion does not affect splenomegaly induced by pristane.

FIGURE 1.

HDAC6 KO did not alter the body weight and spleen weight after pristane treatment.

WT and HDAC6−/− mice on the B6 background were administered 0.5 ml of pristane or PBS at 8–12 wk of age and were euthanized 10 d later. (A and B) Body weight and spleen weight were measured. (C) The ratio of spleen weight to body weight. Mice number (WT PBS, n = 3; WT pristane, n = 5; KO PBS, n = 4; KO pristane, n = 4). ***p < 0.001, ****p < 0.0001.

FIGURE 1.

HDAC6 KO did not alter the body weight and spleen weight after pristane treatment.

WT and HDAC6−/− mice on the B6 background were administered 0.5 ml of pristane or PBS at 8–12 wk of age and were euthanized 10 d later. (A and B) Body weight and spleen weight were measured. (C) The ratio of spleen weight to body weight. Mice number (WT PBS, n = 3; WT pristane, n = 5; KO PBS, n = 4; KO pristane, n = 4). ***p < 0.001, ****p < 0.0001.

Close modal

The inflammation caused by pristane is defined by the ongoing recruitment of leukocytes, such as lymphocytes, macrophages, and neutrophils, to the peritoneal cavity (17, 18). To assess the impact of HDAC6 deletion on early inflammation in pristane-induced lupus, we compared peritoneal cells in WT and HDAC6−/− mice 10 d after pristane administration. At sacrifice, cells were collected from the peritoneum and assessed by flow cytometry. Inflammatory monocytes were identified as CD11b+Ly6C++ (Fig. 2A), and neutrophils were identified as CD11b+Ly6G+ (Fig. 2B). In both WT and HDAC6−/− mice, pristane administration increased the number of inflammatory monocytes and neutrophils compared with PBS controls. In the pristane-treated HDAC6−/− mice, there was significantly less recruitment of inflammatory monocytes (Fig. 2C) and neutrophils (Fig. 2D) compared with the pristane-treated WT mice. Taken together, these data demonstrated that HDAC6 deletion decreased the early inflammatory response to pristane.

FIGURE 2.

HDAC6 KO inhibited peritoneal recruitment of inflammatory monocytes and neutrophils after pristane treatment.

WT and HDAC6−/− mice on the B6 background were administered 0.5 ml of pristane or PBS at 8–12 wk of age and were euthanized 10 d later. Peritoneal cells were isolated and stained with CD11b, Ly6C, and Ly6G Abs. (A and B) Representative flow cytometry picture of inflammatory monocytes (CD11b+Ly6C++) and neutrophils (CD11b+Ly6G+). (C and D) The percent of inflammatory monocytes (CD11b+Ly6C++) and neutrophils (CD11b+Ly6G+) to the live cells. Mice number (WT PBS, n = 4; WT pristane, n = 5; KO PBS, n = 4; KO pristane, n = 4). *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 2.

HDAC6 KO inhibited peritoneal recruitment of inflammatory monocytes and neutrophils after pristane treatment.

WT and HDAC6−/− mice on the B6 background were administered 0.5 ml of pristane or PBS at 8–12 wk of age and were euthanized 10 d later. Peritoneal cells were isolated and stained with CD11b, Ly6C, and Ly6G Abs. (A and B) Representative flow cytometry picture of inflammatory monocytes (CD11b+Ly6C++) and neutrophils (CD11b+Ly6G+). (C and D) The percent of inflammatory monocytes (CD11b+Ly6C++) and neutrophils (CD11b+Ly6G+) to the live cells. Mice number (WT PBS, n = 4; WT pristane, n = 5; KO PBS, n = 4; KO pristane, n = 4). *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

The administration of pristane via peritoneal injection can elicit a localized inflammatory reaction and induce lipogranuloma (19). Pristane-induced inflammation is correlated with the continuous recruitment of leukocytes in the spleen, which are subsequently directed toward the site of inflammation (17). To examine the responses of splenic leukocytes to pristane, we compared immune cells in WT and HDAC6−/− mice 10 d after pristane administration. Ten days after pristane administration, the spleens were removed and splenocytes were isolated. Flow cytometry was used to assess whole splenic cells. The percent of CD4+ T cells (Fig. 3B), CD8+ T cells (Fig. 3C), and CD19+ B cells (Fig. 3G) in the spleen was unchanged after pristane treatment. The percent of CD3+ T cells was decreased in pristane-treated HDAC6−/− mice compared with PBS-treated HDAC6−/− mice (Fig. 3A). The number of CD3+ T cells was reduced in PBS-treated HDAC6−/− mice compared with PBS-treated WT mice (Fig. 3D). CD69 is an early activation marker in immune cells and is implicated in T cell differentiation. In T cells, the number of CD69+ cells was significantly increased in the pristane-treated HDAC6−/− mice compared with the pristane-treated WT mice (Fig. 3E, 3F). In B cells, the number of CD69+ cells was increased by pristane treatment, and the number of CD69+ cells was significantly increased in the pristane-treated HDAC6−/− mice compared with the pristane-treated WT mice (Fig. 3H). Furthermore, the percentages of CD19+CD138+ plasmablasts (Fig. 3I), as well as CD19CD138+ plasma cells (Fig. 3J), were significantly increased in the pristane-treated HDAC6−/− mice compared with PBS-treated HDAC6−/− mice. CD69 can regulate regulatory T (Treg)/Th17 differentiation and is crucial for immunosuppression activity (20). In summary, these data indicate that HDAC6 KO mice are shielded from early inflammation induced by pristane injection.

FIGURE 3.

HDAC6 KO promoted the number of CD69+ T and B cells after pristane treatment.

WT and HDAC6−/− mice on the B6 background were administered 0.5 ml of pristane or PBS at 8–12 wk of age and were euthanized 10 d later. Splenocytes were isolated and stained with CD3, CD4, CD8, CD19, CD69, and CD138 Abs. (A) The percent of CD3+ T cells to the live cells. (B) The percent of CD3+CD4+ Th cells. (C) The percent of CD3+CD8+ cytotoxic T cells. (D) The total number of CD3+ T cells. (E) The percent of CD3+CD4+CD69+ Th cells. (F) The percent of CD3+CD8+CD69+ cytotoxic T cells. (G) The percent of CD19+ B cells to the live cells. (H) The percent of CD19+CD69+ B cells. (I) The percent of CD19+CD138+ plasmablasts. (J) The percent of CD19CD138+ plasma cells. Mice number (WT PBS, n = 4; WT pristane, n = 5; KO PBS, n = 4; KO pristane, n = 4). *p < 0.05, **p < 0.01, ****p < 0.0001.

FIGURE 3.

HDAC6 KO promoted the number of CD69+ T and B cells after pristane treatment.

WT and HDAC6−/− mice on the B6 background were administered 0.5 ml of pristane or PBS at 8–12 wk of age and were euthanized 10 d later. Splenocytes were isolated and stained with CD3, CD4, CD8, CD19, CD69, and CD138 Abs. (A) The percent of CD3+ T cells to the live cells. (B) The percent of CD3+CD4+ Th cells. (C) The percent of CD3+CD8+ cytotoxic T cells. (D) The total number of CD3+ T cells. (E) The percent of CD3+CD4+CD69+ Th cells. (F) The percent of CD3+CD8+CD69+ cytotoxic T cells. (G) The percent of CD19+ B cells to the live cells. (H) The percent of CD19+CD69+ B cells. (I) The percent of CD19+CD138+ plasmablasts. (J) The percent of CD19CD138+ plasma cells. Mice number (WT PBS, n = 4; WT pristane, n = 5; KO PBS, n = 4; KO pristane, n = 4). *p < 0.05, **p < 0.01, ****p < 0.0001.

Close modal

Type I IFNs have been identified as crucial pathogenic cytokines in SLE and are closely associated with the severity of the disease (21). To determine the potential involvement of type I IFN in the initial inflammatory response in mice treated with pristane, we evaluated the expression of IFN-stimulated genes in the peritoneum of these mice. After 10 d of pristane treatment, a portion of the peritoneal cells was collected, and mRNA was isolated. We used real-time quantitative PCR to assess several IFN signature genes. We examined the expression of the IFN signature genes Ifnb (Fig. 4A), Mx1 (Fig. 4B), Oas1a (Fig. 4C), Irf7 (Fig. 4D), Irf9 (Fig. 4E), Cxcl10 (Fig. 4F), and Isg15 (Fig. 4G) and normalized the expression to the housekeeping gene GAPDH. Pristane treatment increased type I IFN signature genes except for IFN-β, which was decreased. Furthermore, there was no significant difference between pristane-treated HDAC6−/− mice and pristane-treated WT mice. Taken together, these results indicate that HDAC6 deletion has little effect on pristane-induced IFN signature genes expression.

FIGURE 4.

HDAC6 KO did not affect IFN signature genes expression induced by pristane treatment.

WT and HDAC6−/− mice on the B6 background were administered 0.5 ml of pristane or PBS at 8–12 wk of age and were euthanized 10 d later. Peritoneal cells were isolated, and RNA was extracted. (AG) Ifnb, Mx1, Oas1a, Irf7, Irf9, Cxcl10, and Isg15 mRNA levels were determined by real-time quantitative PCR and normalized to the GAPDH levels. Mice number (WT PBS, n = 4; WT pristane, n = 5; KO PBS, n = 4; KO pristane, n = 4). *p < 0.05, **p < 0.01, ****p < 0.0001.

FIGURE 4.

HDAC6 KO did not affect IFN signature genes expression induced by pristane treatment.

WT and HDAC6−/− mice on the B6 background were administered 0.5 ml of pristane or PBS at 8–12 wk of age and were euthanized 10 d later. Peritoneal cells were isolated, and RNA was extracted. (AG) Ifnb, Mx1, Oas1a, Irf7, Irf9, Cxcl10, and Isg15 mRNA levels were determined by real-time quantitative PCR and normalized to the GAPDH levels. Mice number (WT PBS, n = 4; WT pristane, n = 5; KO PBS, n = 4; KO pristane, n = 4). *p < 0.05, **p < 0.01, ****p < 0.0001.

Close modal

We have demonstrated that HDAC6 deletion inhibited the peritoneal recruitment of inflammatory monocytes after pristane treatment. Inflammatory monocytes possess a high degree of infiltration capability and can transform into inflammatory macrophages. To investigate further the role of HDAC6 inhibition on macrophage function, we used the J774A.1 cell line. Initially, experiments were conducted to determine the proper concertation of the selective HDAC6 inhibitor ACY-738 that would increase α-tubulin acetylation without increasing histone acetylation. We found that with 1 μM ACY-738 treatment, α-tubulin was acetylated, but not histone H3 (Fig. 5A). When J774A.1 cells were stimulated with a mixture of LPS/IFN-γ, we found that 1 μM concertation of ACY-738 increased NF-κB acetylation and decreased total NF-κB protein level without affecting NF-κB phosphorylation (Fig. 5B). Surprisingly, when macrophages were treated with ACY-738 and stimulated with LPS/IFN-γ, we measured an increase in iNOS expression (Fig. 5B). Furthermore, STAT1 phosphorylation was significantly increased by ACY-738 with LPS/IFN-γ stimulation (Fig. 5C). These results suggest that HDAC6 inhibitor ACY-738 downregulates total NF-κB protein level while upregulating iNOS expression via the STAT1 pathway.

FIGURE 5.

HDAC6 inhibitor ACY-738 inhibited the NF-κB signaling in vitro.

J774A.1 mouse macrophage cells were treated with ACY-738 and LPS/IFN-γ for 24 h. (A) Western blot analysis showed that 1 µM ACY-738 was able to significantly increase the level of α-tubulin acetylation while not affecting the acetylation of histone H3. (B) J774A.1 mouse macrophage cells were treated with 1 µM ACY-738, as well as LPS (1 µg/ml) and IFN-γ (100 ng/ml) for 24 h. Western blot analysis showed that with LPS/IFN-γ stimulation, 1 µM ACY-738 was able to decrease the protein level of NF-κB and increase NF-κB acetylation. (C) Protein quantification of NF-κB, acetyl-NF-κB, p-NF-κB, iNOS, STAT1, and p-STAT1 by using ImageJ software. The experiments were repeated three times independently. *p < 0.05, ***p < 0.001.

FIGURE 5.

HDAC6 inhibitor ACY-738 inhibited the NF-κB signaling in vitro.

J774A.1 mouse macrophage cells were treated with ACY-738 and LPS/IFN-γ for 24 h. (A) Western blot analysis showed that 1 µM ACY-738 was able to significantly increase the level of α-tubulin acetylation while not affecting the acetylation of histone H3. (B) J774A.1 mouse macrophage cells were treated with 1 µM ACY-738, as well as LPS (1 µg/ml) and IFN-γ (100 ng/ml) for 24 h. Western blot analysis showed that with LPS/IFN-γ stimulation, 1 µM ACY-738 was able to decrease the protein level of NF-κB and increase NF-κB acetylation. (C) Protein quantification of NF-κB, acetyl-NF-κB, p-NF-κB, iNOS, STAT1, and p-STAT1 by using ImageJ software. The experiments were repeated three times independently. *p < 0.05, ***p < 0.001.

Close modal

HDAC6 dysregulation plays a significant role in the development and progression of various human diseases (22, 23). HDAC6 is an enzyme responsible for removing acetyl groups from various cytosolic proteins and transcription factors that influence cell signaling and gene expression (24, 25). Dysregulation of HDAC6 has been linked to several pathological conditions, including cancer, neurodegenerative disorders, autoimmune diseases, and cardiovascular ailments. In cancer, overexpression of HDAC6 can promote uncontrolled cell growth and inhibit cell death, contributing to tumorigenesis and tumor progression (26–28). In neurodegenerative diseases such as Alzheimer’s and Parkinson’s, altered HDAC6 activity has been associated with abnormal protein aggregates and impaired cellular clearance mechanisms, contributing to neuronal damage and degeneration (29–34). In inflammation, and in particular SLE, we have previously reported that selective HDAC6 inhibition can decrease disease in lupus mice (6, 35–37). The pristane-induced lupus model is one frequently used animal model for lupus research. The B6 pristane-induced lupus mouse model does not develop lupus-like disease until several months after the initial injection (13). However, these mice often develop severe alveolar hemorrhage that may result in up to 20% mortality by 2 wk after injection (15). In our present studies, we sought to determine whether HDAC6 deletion would decrease early inflammation in this lupus mouse model.

We found that gene deletion of HDAC6 significantly decreased inflammatory monocytes and neutrophils accumulated in the peritoneum 10 d after pristane injection compared with WT mice. This was particularly interesting because HDAC6 inhibition has been shown to decrease neutrophil activation in Pseudomonas aeruginosa–induced inflammation and infection in cystic fibrosis (38). Moreover, studies by Yan et al. (39) found differing effects of HADC6 gene deletion in the LPS-induced model of acute peritonitis. They found that neutrophil activation was largely unaffected and macrophage activation was enhanced in the WT animals, whereas it was blunted in the HDAC6−/− animals. This apparent dichotomy could be because of the different mechanisms by which the immune system is activated.

In contrast, the intracellular mechanisms of pristane-induced inflammation are complex and involve the activation of immune cells and various signaling pathways. Although the exact mechanisms may vary depending on the specific context and cell types involved, pristane is believed to act through both pattern recognition receptors, such as TLRs, and NOD-like receptors (NLRs) on the surface or within the cytoplasm of immune cells (40). TLR activation, in particular, can trigger intracellular signaling cascades, including NF-κB and MAPK pathways, which are central to the regulation of inflammatory gene expression (41–46).

In addition, pristane-induced inflammation has been associated with the activation of inflammasomes and multiprotein complexes that regulate the processing and release of proinflammatory cytokines, especially IL-1β (47). Furthermore, the NLRP3 (NOD-like receptor family, pyrin domain-containing inflammasome) has been implicated in pristane-induced inflammation (48). Through these mechanisms, pristane has been linked to the development of autoimmune diseases and can lead to the production of autoantibodies (49).

In regard to T and B cell activation, pristane has been shown to activate B cells and promote the production of Abs. It induces the formation of long-lived Ab-secreting cells in the peritoneal cavity of mice. We found that there was an increase in B cell activation with pristane treatment, and the increase in B cell activation was greater in the HDAC6−/− animals compared with the WT mice. Furthermore, there was an increase in the plasma cells in the pristane-treated HDAC6−/− mice compared with the pristane-treated WT mice. Similarly, pristane has been reported to activate certain subsets of T cells (50). It can influence the differentiation and activation of CD4+ Th cells (51). We found increased activation markers of T cells, along with increased B cell differentiation to plasma cells in HDAC6−/− mice treated with pristane. It has been reported that pristane increases autoantibody production and nephritis through TLR7 or IFNR-mediated type I IFN production (52).

CD69 is a classic early marker of lymphocyte activation because of its rapid expression on the plasma membrane after stimulation (53). Apart from its inherent significance as an activation marker, contemporary data indicate that CD69 plays a crucial role in regulating immunological responses. In particular, CD69 controls the secretion of IL-10, IL-17, IL-22, and IFN-γ, as well as the differentiation of Treg cells (53). CD69 KO mice show enhanced Th17 proinflammatory responses (54). Yu et al. (55) found that CD69 stimulates the production of IL-10, which is crucial for the suppressive role of Tregs. Specifically, they found the proliferation of CD4+ T cells is considerably inhibited by CD69+ Tregs from IL10+/+ mice as opposed to IL10−/− mice. They also found the development of inflammatory bowel disease in mice was significantly prevented by the adoptive transfer of CD69+ Tregs, but not CD69 Tregs or CD69+ Tregs deficient in IL-10. In the mouse model of collagen-induced arthritis, the lack of CD69 intensifies the inflammatory and destructive process in the afflicted joints (56). CD69 functions as a suppressor of autoimmune reactivity and inflammation by controlling the production of TGF-β, a cytokine that subsequently reduces the production of other proinflammatory substances (56). In a cardiomyopathy model, the absence of CD69 amplifies the inflammatory process and exacerbates Th17-mediated cardiac tissue damage, which promotes heart failure (57). According to their research, CD69 negatively controls cardiac inflammation, heart-specific Th17 responses, and the progression of heart failure in experimental autoimmune myocarditis (57). Another group studying myocardial infarction found that Cd69−/− mice exhibited robust IL-17+ γδ T cell responses after ischemia, which exacerbated myocardial inflammation and, as a result, impaired cardiac function (58). They also found the recruitment of IL-17+ γδ T cells was reduced by the adoptive transfer of CD69+ Tregs into Cd69−/− mice after left anterior descending ligation, thereby increasing survival (58). The transplantation of CD4+ T cells from CD69 KO mice into Rag-1−/− immunodeficient animals results in an accelerated form of colitis, characterized by an increased production of IL-17, IFN-γ, and TNF-α (59). The absence of CD69 resulted in heightened synthesis of proinflammatory cytokines, diminished induction of Foxp3+ Treg cells, compromised development of oral tolerance, and exacerbated colitis (59). CD69-deficient animals have been seen to exhibit heightened tissue inflammation in the initial phase of Listeria monocytogenes infection (60). In (NZB × NZW)F1 mice, a model of SLE, the number of CD4+CD69+ T cells increases, which leads to a suppressive effect in vitro by inhibiting the synthesis of IL-2 (61). In summary, in addition to serving as an activation marker, CD69 also participates in regulating Treg/Th17 differentiation and is crucial for immunosuppression (20, 62). Our result shows that the number of CD69+ T and B cells was increased in HDAC6−/− mice after pristane treatment, which indicates that HDAC6 deletion protects mice from early inflammation.

Inflammatory monocytes exhibit a specific pattern of movement toward areas of inflammation, where they generate inflammatory cytokines and contribute to both local and systemic inflammation (63). Inflammatory monocytes can transform into inflammatory macrophages. Based on the result that HDAC6 deletion inhibited the peritoneal recruitment of inflammatory monocytes, we conducted in vitro studies to further characterize the mechanism of activation of the monocytes. J774A.1 is a mouse monocyte/macrophage cell line and can be used in immunology research. We found that HDAC6 inhibitor ACY-738 inhibited the NF-κB signaling in stimulated J774A.1 cells. NF-κB triggers the expression of many genes that promote inflammation. Our results of in vitro experiments provide more evidence that HDAC6 promotes inflammation. Surprisingly, we found HDAC6 inhibition did not suppress iNOS expression in LPS/IFN-γ–stimulated J774A.1 cells. IFN-γ interacted with LPS synergistically to induce iNOS expression; we also found that neither LPS nor IFN-γ alone was sufficient to induce iNOS expression in J774A.1 cells (Supplemental Fig. 2). Wang et al. (64) have previously reported that macrophages from HDAC6−/− mice have decreased iNOS production with LPS stimulation. Because LPS acts predominantly through NF-κB to induce iNOS, the IFN-γ pathway acting through STAT1 to induce iNOS expression can act independently from NF-κB. IFN-γ can potentiate proinflammatory signaling by priming macrophages for antimicrobial actions because it induces NO production and inhibits NLRP3 inflammasome activation. Furthermore, the contribution by p38 to the induction of iNOS and apoptosis is independent of NF-κB nuclear translocation (65). Discrepancies in our results from Wang and coworkers (64) could be because of many factors, including using a different inhibitor, different cell type, different concentration, and not pretreating the cells before stimulation. In our studies, we added ACY-738 concurrent with LPS/IFN-γ for 24 h. In their studies, they pretreated the cells for 3 h before 12 h of LPS stimulation.

In summary, we discovered that in the early inflammatory response to pristane, HDAC6 deletion inhibited the recruitment of inflammatory monocytes and neutrophils in the peritoneum. HDAC6 deletion promoted the number of CD69+ T and B cells, and CD69 was reported to function as a suppressor of inflammation. The HDAC6 inhibitor was found to increase NF-κB acetylation and decrease total NF-κB protein level in in vitro investigations using J774A.1 cells.

The authors have no financial conflicts of interest.

We appreciate Melissa Makris for allowing use of Virginia Tech’s flow cytometry core laboratory.

This work was supported by National Institutes of Health Grant 1R15AI152022-01.

The online version of this article contains supplemental material.

B6

C57BL/6

HDAC6

histone deacetylase 6

iNOS

inducible NO synthase

KO

knockout

SLE

systemic lupus erythematosus

Treg

regulatory T

WT

wild-type

1
Tsokos
,
G. C.
2020
.
Autoimmunity and organ damage in systemic lupus erythematosus
.
Nat. Immunol.
21
:
605
614
.
2
Tsokos
,
G. C.
2011
.
Systemic lupus erythematosus
.
N. Engl. J. Med.
365
:
2110
2121
.
3
Haberland
,
M.
,
R. L.
Montgomery
,
E. N.
Olson
.
2009
.
The many roles of histone deacetylases in development and physiology: implications for disease and therapy
.
Nat. Rev. Genet.
10
:
32
42
.
4
Verdone
,
L.
,
E.
Agricola
,
M.
Caserta
,
E.
Di Mauro
.
2006
.
Histone acetylation in gene regulation
.
Brief. Funct. Genomic. Proteomic.
5
:
209
221
.
5
Gallinari
,
P.
,
S.
Di Marco
,
P.
Jones
,
M.
Pallaoro
,
C.
Steinkühler
.
2007
.
HDACs, histone deacetylation and gene transcription: from molecular biology to cancer therapeutics
.
Cell. Res.
17
:
195
211
.
6
Regna
,
N. L.
,
M. D.
Vieson
,
X. M.
Luo
,
C. B.
Chafin
,
A. G.
Puthiyaveetil
,
S. E.
Hammond
,
D. L.
Caudell
,
M. B.
Jarpe
,
C. M.
Reilly
.
2016
.
Specific HDAC6 inhibition by ACY-738 reduces SLE pathogenesis in NZB/W mice
.
Clin. Immunol.
162
:
58
73
.
7
Li
,
Y.
,
E.
Seto
.
2016
.
HDACs and HDAC inhibitors in cancer development and therapy
.
Cold Spring Harb. Perspect. Med.
6
:
a026831
.
8
Banik
,
D.
,
S.
Noonepalle
,
M.
Hadley
,
E.
Palmer
,
M.
Gracia-Hernandez
,
C.
Zevallos-Delgado
,
N.
Manhas
,
H.
Simonyan
,
C. N.
Young
,
A.
Popratiloff
, et al
2020
.
HDAC6 plays a noncanonical role in the regulation of antitumor immune responses, dissemination, and invasiveness of breast cancer
.
Cancer Res.
80
:
3649
3662
.
9
Youn
,
G. S.
,
K. W.
Lee
,
S. Y.
Choi
,
J.
Park
.
2016
.
Overexpression of HDAC6 induces pro-inflammatory responses by regulating ROS-MAPK-NF-κB/AP-1 signaling pathways in macrophages
.
Free Radic. Biol. Med.
97
:
14
23
.
10
Yang
,
C. J.
,
Y. P.
Liu
,
H. Y.
Dai
,
Y. L.
Shiue
,
C. J.
Tsai
,
M. S.
Huang
,
Y. T.
Yeh
.
2015
.
Nuclear HDAC6 inhibits invasion by suppressing NF-κB/MMP2 and is inversely correlated with metastasis of non-small cell lung cancer
.
Oncotarget
6
:
30263
30276
.
11
Zhang
,
W. B.
,
F.
Yang
,
Y.
Wang
,
F. Z.
Jiao
,
H. Y.
Zhang
,
L. W.
Wang
,
Z. J.
Gong
.
2019
.
Inhibition of HDAC6 attenuates LPS-induced inflammation in macrophages by regulating oxidative stress and suppressing the TLR4-MAPK/NF-κB pathways
.
Biomed. Pharmacother.
117
:
109166
.
12
Reeves
,
W. H.
,
P. Y.
Lee
,
J. S.
Weinstein
,
M.
Satoh
,
L.
Lu
.
2009
.
Induction of autoimmunity by pristane and other naturally occurring hydrocarbons
.
Trends. Immunol.
30
:
455
464
.
13
Leiss
,
H.
,
B.
Niederreiter
,
T.
Bandur
,
B.
Schwarzecker
,
S.
Blüml
,
G.
Steiner
,
W.
Ulrich
,
J. S.
Smolen
,
G. H.
Stummvoll
.
2013
.
Pristane-induced lupus as a model of human lupus arthritis: evolvement of autoantibodies, internal organ and joint inflammation
.
Lupus
22
:
778
792
.
14
Zhuang
,
H. Y.
,
S. H.
Han
,
P. Y.
Lee
,
R.
Khaybullin
,
S.
Shumyak
,
L.
Lu
,
A.
Chatha
,
A.
Afaneh
,
Y.
Zhang
,
C.
Xie
, et al
2017
.
Pathogenesis of diffuse alveolar hemorrhage in murine lupus
.
Arthritis Rheumatol.
69
:
1280
1293
.
15
Barker
,
T. T.
,
P. Y.
Lee
,
K. M.
Kelly-Scumpia
,
J. S.
Weinstein
,
D. C.
Nacionales
,
Y.
Kumagai
,
S.
Akira
,
B. P.
Croker
,
E. S.
Sobel
,
W. H.
Reeves
,
M.
Satoh
.
2011
.
Pathogenic role of B cells in the development of diffuse alveolar hemorrhage induced by pristane
.
Lab. Invest.
91
:
1540
1550
.
16
Zhang
,
X.
,
R.
Goncalves
,
D. M.
Mosser
.
2008
.
The isolation and characterization of murine macrophages
.
Curr. Protoc. Immunol.
14
:
14.1.1
14.1.14
.
17
Han
,
S.
,
H.
Zhuang
,
S.
Shumyak
,
J.
Wu
,
H.
Li
,
L. J.
Yang
,
W. H.
Reeves
.
2017
.
A novel subset of anti-inflammatory CD138(+) macrophages is deficient in mice with experimental lupus
.
J. Immunol.
199
:
1261
1274
.
18
Lee
,
P. Y.
,
Y.
Li
,
Y.
Kumagai
,
Y.
Xu
,
J. S.
Weinstein
,
E. S.
Kellner
,
D. C.
Nacionales
,
E. J.
Butfiloski
,
N.
van Rooijen
,
S.
Akira
, et al
2009
.
Type I interferon modulates monocyte recruitment and maturation in chronic inflammation
.
Am. J. Pathol.
175
:
2023
2033
.
19
Chen
,
H.
,
D.
Liao
,
D.
Cain
,
I.
McLeod
,
Y.
Ueda
,
Z.
Guan
,
C.
Raetz
,
G.
Kelsoe
.
2010
.
Distinct granuloma responses in C57BL/6J and BALB/cByJ mice in response to pristane
.
Int. J. Exp. Pathol.
91
:
460
471
.
20
González-Amaro
,
R.
,
J. R.
Cortés
,
F.
Sánchez-Madrid
,
P.
Martín
.
2013
.
Is CD69 an effective brake to control inflammatory diseases?
Trends Mol. Med.
19
:
625
632
.
21
Kirou
,
K. A.
,
C. P.
Mavragani
,
M. K.
Crow
.
2007
.
Activation of type I interferon in systemic lupus erythematosus
.
Expert. Rev. Clin. Immunol.
3
:
579
588
.
22
Li
,
Y.
,
D.
Shin
,
S. H.
Kwon
.
2013
.
Histone deacetylase 6 plays a role as a distinct regulator of diverse cellular processes
.
FEBS J.
280
:
775
793
.
23
Dallavalle
,
S.
,
C.
Pisano
,
F.
Zunino
.
2012
.
Development and therapeutic impact of HDAC6-selective inhibitors
.
Biochem. Pharmacol.
84
:
756
765
.
24
Hubbert
,
C.
,
A.
Guardiola
,
R.
Shao
,
Y.
Kawaguchi
,
A.
Ito
,
A.
Nixon
,
M.
Yoshida
,
X. F.
Wang
,
T. P.
Yao
.
2002
.
HDAC6 is a microtubule-associated deacetylase
.
Nature
417
:
455
458
.
25
Kaur
,
S.
,
P.
Rajoria
,
M.
Chopra
.
2022
.
HDAC6: a unique HDAC family member as a cancer target
.
Cell. Oncol. (Dordr).
45
:
779
829
.
26
Aldana-Masangkay
,
G. I.
,
K. M.
Sakamoto
.
2011
.
The role of HDAC6 in cancer
.
J. Biomed. Biotechnol.
2011
:
875824
.
27
Yang
,
P. H.
,
L.
Zhang
,
Y. J.
Zhang
,
J.
Zhang
,
W. F.
Xu
.
2013
.
HDAC6: physiological function and its selective inhibitors for cancer treatment
.
Drug. Discov. Ther.
7
:
233
242
.
28
Li
,
T.
,
C.
Zhang
,
S.
Hassan
,
X. Y.
Liu
,
F. J.
Song
,
K. X.
Chen
,
W.
Zhang
,
J. L.
Yang
.
2018
.
Histone deacetylase 6 in cancer
.
J. Hematol. Oncol.
11
:
111
.
29
Van Helleputte
,
L.
,
V.
Benoy
,
L.
Van Den Bosch
.
2014
.
The role of histone deacetylase 6 (HDAC6) in neurodegeneration
.
Res. Rep. Biol.
5
:
1
13
.
30
Simões-Pires
,
C.
,
V.
Zwick
,
A.
Nurisso
,
E.
Schenker
,
P. A.
Carrupt
,
M.
Cuendet
.
2013
.
HDAC6 as a target for neurodegenerative diseases: what makes it different from the other HDACs?
Mol. Neurodegener.
8
:
7
.
31
LoPresti
,
P.
2021
.
HDAC6 in diseases of cognition and of neurons
.
Cells
10
:
12
.
32
Shen
,
S. D.
,
A. P.
Kozikowski
.
2020
.
A patent review of histone deacetylase 6 inhibitors in neurodegenerative diseases (2014-2019)
.
Expert. Opin. Ther. Pat.
30
:
121
136
.
33
Mondal
,
P.
,
P.
Bai
,
A.
Gomm
,
G.
Bakiasi
,
C.-C. J.
Lin
,
Y.
Wang
,
S. H.
Choi
,
R. E.
Tanzi
,
C. N.
Wang
,
C.
Zhang
.
2023
.
Structure-based discovery of a small molecule inhibitor of histone deacetylase 6 (HDAC6) that significantly reduces Alzheimer’s disease neuropathology
.
Adv. Sci. (Weinh).
11
:
e2304545
.
34
Zhang
,
L.
,
S. L.
Sheng
,
C.
Qin
.
2013
.
The role of HDAC6 in Alzheimer’s disease
.
J. Alzheimers. Dis.
33
:
283
295
.
35
Vieson
,
M. D.
,
A. M.
Gojmerac
,
D.
Khan
,
R. J.
Dai
,
J. H.
van Duzer
,
R.
Mazitschek
,
D. L.
Caudell
,
X. F.
Liao
,
X. M.
Luo
,
C. M.
Reilly
.
2017
.
Treatment with a selective histone deacetylase 6 inhibitor decreases lupus nephritis in NZB/W mice
.
Histol. Histopathol.
32
:
1317
1332
.
36
Ren
,
J.
,
X.
Liao
,
M. D.
Vieson
,
M.
Chen
,
R.
Scott
,
J.
Kazmierczak
,
X. M.
Luo
,
C. M.
Reilly
.
2018
.
Selective HDAC6 inhibition decreases early stage of lupus nephritis by down-regulating both innate and adaptive immune responses
.
Clin. Exp. Immunol.
191
:
19
31
.
37
Ren
,
J. J.
,
M. D.
Catalina
,
K.
Eden
,
X. F.
Liao
,
K. A.
Read
,
X.
Luo
,
R. P.
McMillan
,
M. W.
Hulver
,
M.
Jarpe
,
P.
Bachali
, et al
2019
.
Selective histone deacetylase 6 inhibition normalizes B cell activation and germinal center formation in a model of systemic lupus erythematosus
.
Front. Immunol.
10
:
2512
.
38
Brindisi
,
M.
,
S.
Barone
,
A.
Rossi
,
E.
Cassese
,
N.
Del Gaudio
,
A. J.
Feliz Morel
,
G.
Filocamo
,
A.
Alberico
,
I.
De Fino
,
D.
Gugliandolo
, et al
2022
.
Efficacy of selective histone deacetylase 6 inhibition in mouse models of Pseudomonas aeruginosa infection: a new glimpse for reducing inflammation and infection in cystic fibrosis
.
Eur. J. Pharmacol.
936
:
175349
.
39
Yan
,
B.
,
S. B.
Xie
,
Y.
Liu
,
W. X.
Liu
,
D. W.
Li
,
M.
Liu
,
H. B. R.
Luo
,
J.
Zhou
.
2018
.
Histone deacetylase 6 modulates macrophage infiltration during inflammation
.
Theranostics
8
:
2927
2938
.
40
Summers
,
S. A.
,
A.
Hoi
,
O. M.
Steinmetz
,
K. M.
O’Sullivan
,
J. D.
Ooi
,
D.
Odobasic
,
S.
Akira
,
A. R.
Kitching
,
S. R.
Holdsworth
.
2010
.
TLR9 and TLR4 are required for the development of autoimmunity and lupus nephritis in pristane nephropathy
.
J. Autoimmun.
35
:
291
298
.
41
Sameer
,
A. S.
,
S.
Nissar
.
2021
.
Toll-like receptors (TLRs): structure, functions, signaling, and role of their polymorphisms in colorectal cancer susceptibility
.
Biomed. Res. Int.
2021
:
1157023
.
42
Kawai
,
T.
,
S.
Akira
.
2007
.
Signaling to NF-κB by Toll-like receptors
.
Trends. Mol. Med.
13
:
460
469
.
43
Fitzgerald
,
K. A.
,
J. C.
Kagan
.
2020
.
Toll-like receptors and the control of immunity
.
Cell
180
:
1044
1066
.
44
Duan
,
T. H.
,
Y.
Du
,
C. S.
Xing
,
H. Y. Y.
Wang
,
R. F.
Wang
.
2022
.
Toll-like receptor signaling and its role in cell-mediated immunity
.
Front. Immunol.
13
:
812774
.
45
Lind
,
N. A.
,
V. E.
Rael
,
K.
Pestal
,
B.
Liu
,
G. M.
Barton
.
2022
.
Regulation of the nucleic acid-sensing Toll-like receptors
.
Nat. Rev. Immunol.
22
:
224
235
.
46
Fillatreau
,
S.
,
B.
Manfroi
,
T.
Dörner
.
2021
.
Toll-like receptor signalling in B cells during systemic lupus erythematosus
.
Nat. Rev. Rheumatol.
17
:
98
108
.
47
Bonomini
,
F.
,
M.
Dos Santos
,
F. V.
Veronese
,
R.
Rezzani
.
2019
.
NLRP3 inflammasome modulation by melatonin supplementation in chronic pristane-induced lupus nephritis
.
Int. J. Mol. Sci.
20
:
3466
.
48
Lu.
,
A.
,
H.
Li
,
J. L.
Niu
,
S. X.
Wu
,
G.
Xue
,
X. M.
Yao
,
Q. H.
Guo
,
N. H.
Wan
,
P.
Abliz
,
G. W.
Yang
, et al
2017
.
Hyperactivation of the NLRP3 inflammasome in myeloid cells leads to severe organ damage in experimental lupus
.
J. Immunol.
198
:
1119
1129
.
49
Richard
,
M. L.
,
G.
Gilkeson
.
2018
.
Mouse models of lupus: what they tell us and what they don't
.
Lupus Sci. Med.
5
:
e000199
.
50
Richards
,
H. B.
,
M.
Satoh
,
J. C.
Jennette
,
T.
Okano
,
Y. S.
Kanwar
,
W. H.
Reeves
.
1999
.
Disparate T cell requirements of two subsets of lupus-specific autoantibodies in pristane-treated mice
.
Clin. Exp. Immunol.
115
:
547
553
.
51
Liu
,
S.
,
Y.-M.
Li
,
J.-Z.
Li
,
S.-J.
Wang
,
P.
Ji
,
M.-Y.
Zhang
,
Y.
Wang
.
2022
.
CD4 [Database] T cells promote IgG production in MHC-independent and ICAM-1-dependent manners in pristane-induced lupus mice
.
Mediat. Inflamm.
2022
:
e968847
.
52
Han
,
S. H.
,
H. Y.
Zhuang
,
Y.
Xu
,
P.
Lee
,
Y.
Li
,
J. C.
Wilson
,
O.
Vidal
,
H. S.
Choi
,
Y.
Sun
,
L. J.
Yang
,
W. H.
Reeves
.
2015
.
Maintenance of autoantibody production in pristane-induced murine lupus
.
Arthritis Res. Ther.
17
:
384
.
53
Cibrían
,
D.
,
F.
Sánchez-Madrid
.
2017
.
CD69: from activation marker to metabolic gatekeeper
.
Eur. J. Immunol.
47
:
946
953
.
54
Martín
,
P.
,
M.
Gómez
,
A.
Lamana
,
A.
Cruz-Adalia
,
M.
Ramírez-Huesca
,
M. A.
Ursa
,
M.
Yáñez-Mo
,
F.
Sánchez-Madrid
.
2010
.
CD69 association with Jak3/Stat5 proteins regulates Th17 cell differentiation
.
Mol. Cell. Biol.
30
:
4877
4889
.
55
Yu
,
L.
,
F.
Yang
,
F.
Zhang
,
D.
Guo
,
L.
Li
,
X.
Wang
,
T.
Liang
,
J.
Wang
,
Z.
Cai
,
H.
Jin
.
2018
.
CD69 enhances immunosuppressive function of regulatory T-cells and attenuates colitis by prompting IL-10 production
.
Cell Death Dis.
9
:
905
.
56
Sancho
,
D.
,
M.
Gómez
,
F.
Viedma
,
E.
Esplugues
,
M.
Górdon-Alonso
,
M. A.
García-López
,
H.
de la Fuente
,
A. C.
Martínez
,
P.
Lauzurica
,
F.
Sánchez-Madrid
.
2003
.
CD69 downregulates autoimmune reactivity through active transforming growth factor-β production in collagen-induced arthritis
.
J. Clin. Invest.
112
:
872
882
.
57
Cruz-Adalia
,
A.
,
L. J.
Jiménez-Borreguero
,
M.
Ramírez-Huesca
,
I.
Chico-Calero
,
O.
Barreiro
,
E.
López-Conesa
,
M.
Fresno
,
F.
Sánchez-Madrid
,
P.
Martín
.
2010
.
CD69 limits the severity of cardiomyopathy after autoimmune myocarditis
.
Circulation
122
:
1396
1404
.
58
Blanco-Domínguez
,
R.
,
H.
de la Fuente
,
C.
Rodríguez
,
L.
Martín-Aguado
,
R.
Sánchez-Díaz
,
R.
Jiménez-Alejandre
,
I.
Rodríguez-Arabaolaza
,
A.
Curtabbi
,
M. M.
García-Guimaraes
,
A.
Vera
, et al
2022
.
CD69 expression on regulatory T cells protects from immune damage after myocardial infarction
.
J. Clin. Invest.
132
:
e152418
.
59
Radulovic
,
K.
,
C.
Manta
,
V.
Rossini
,
K.
Holzmann
,
H. A.
Kestler
,
U. M.
Wegenka
,
T.
Nakayama
,
J. H.
Niess
.
2012
.
CD69 regulates type I IFN-induced tolerogenic signals to mucosal CD4 T cells that attenuate their colitogenic potential
.
J. Immunol.
188
:
2001
2013
.
60
Vega-Ramos
,
J.
,
E.
Alari-Pahissa
,
J. D.
Valle
,
E.
Carrasco-Marín
,
E.
Esplugues
,
M.
Borràs
,
A. C.
Martínez
,
P.
Lauzurica
.
2010
.
CD69 limits early inflammatory diseases associated with immune response to Listeria monocytogenes infection
.
Immunol. Cell. Biol.
88
:
707
715
.
61
Ishikawa
,
S.
,
S.
Akakura
,
M.
Abe
,
K.
Terashima
,
K.
Chijiiwa
,
H.
Nishimura
,
S.
Hirose
,
T.
Shirai
.
1998
.
A subset of CD4+ T cells expressing early activation antigen CD69 in murine lupus: possible abnormal regulatory role for cytokine imbalance
.
J. Immunol.
161
:
1267
1273
.
62
Li
,
Y.
,
Y.
Gu
,
P.
Yang
,
Y.
Wang
,
X.
Yu
,
Y.
Li
,
Z.
Jin
,
L.
Xu
.
2024
.
CD69 is a promising immunotherapy and prognosis prediction target in cancer
.
Immunotargets Ther.
13
:
1
14
.
63
Yang
,
J.
,
L.
Zhang
,
C.
Yu
,
X. F.
Yang
,
H.
Wang
.
2014
.
Monocyte and macrophage differentiation: circulation inflammatory monocyte as biomarker for inflammatory diseases
.
Biomark. Res.
2
:
1
.
64
Wang
,
Y.
,
K.
Wang
,
J.
Fu
.
2020
.
HDAC6 mediates macrophage iNOS expression and excessive nitric oxide production in the blood during endotoxemia
.
Front. Immunol.
11
:
1893
.
65
Saldeen
,
J.
,
N.
Welsh
.
2004
.
p38 MAPK inhibits JNK2 and mediates cytokine-activated iNOS induction and apoptosis independently of NF-κB translocation in insulin-producing cells
.
Eur. Cytokine Netw.
15
:
47
52
.
This article is distributed under the terms of the CC BY-NC 4.0 Unported license.

Supplementary data