Next Article in Journal
Development and Progression of Non-Alcoholic Fatty Liver Disease: The Role of Advanced Glycation End Products
Previous Article in Journal
Ischemia/Reperfusion Injury Revisited: An Overview of the Latest Pharmacological Strategies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Andrographolide Ameliorates Rheumatoid Arthritis by Regulating the Apoptosis–NETosis Balance of Neutrophils

1
School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
2
State Key Laboratory of Biocontrol, Department of Biochemistry, School of Life Sciences, Sun Yat-Sen (Zhongshan) University, Guangzhou 510275, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2019, 20(20), 5035; https://doi.org/10.3390/ijms20205035
Submission received: 3 August 2019 / Revised: 6 October 2019 / Accepted: 8 October 2019 / Published: 11 October 2019
(This article belongs to the Section Molecular Immunology)

Abstract

:
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by symmetric polyarthritis with swelling and pain at synovial joints. In RA patients, delayed neutrophil apoptosis amplifies the inflammatory response and massively released neutrophil extracellular traps (NETs) induce tissue damage and provide self-antigens. Andrographolide (AD) is the major active labdane diterpenoid derived from Andrographis paniculata, which has multiple pharmacological effects, including hepatoprotection, anti-angiogenesis, anti-thrombosis, and anti-inflammation. In the present study, we investigated the effect of AD on an adjuvant-induced arthritis (AA) murine model of RA and found that AD alleviated murine arthritis by reducing neutrophil infiltration and NETosis in the ankle joints and relieved the systematic inflammation. In vitro experiments showed that AD accelerated the apoptosis of lipopolysaccharide-activated neutrophils and inhibited autophagy-dependent extracellular traps formation of neutrophils. These findings suggest that AD has considerable potential for RA therapy.

Graphical Abstract

1. Introduction

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease characterized by symmetric polyarthritis with swelling and pain at synovial joints [1]. The incidence of RA is approximately 0.5–1% worldwide, with a higher incidence in women than in men [2]. Many types of immune cells have been proven to be involved in the pathogenesis of RA, including T cells, B cells, natural killer cells, neutrophils, macrophages, and dendritic cells [3]. For many years, the major contributions of neutrophils in RA were considered to be the releasing of numerous degradative enzymes and reactive oxygen species [3,4]. However, emerging studies have demonstrated that neutrophils also play critical roles in the initiation, progression, and perpetuation of RA [5].
Neutrophils granule proteins, such as myeloperoxidase (MPO) and neutrophil elastase (NE), are detected in high concentrations in RA synovial fluid, which are responsible for joint damage [6,7,8]. Synovial fluid neutrophils produce and secrete a variety of cytokines and chemokines, which are implicated in osteoclast activation and bone destruction [9]. Numerous studies have illustrated that neutrophil extracellular traps (NETs) can promote auto-immunity and aggravate tissue damage [8,9]. NETs are important sources of citrullinated autoantigens, which lead to an autoimmune response in RA [8,10]. Compared with neutrophils from healthy and osteoarthritis groups, enhanced NETosis was found in circulating and synovial fluid neutrophils from RA patients [11]. Neutrophils depletion or functional inhibition significantly ameliorates joint damage and inflammation in an arthritis murine model [12]. Therefore, neutrophils serve as an important target of RA treatment.
In the past few decades, great progress has been made in the management and treatment of RA, especially the application of biologic therapies such as tumor necrosis factor (TNF)-α-blockade agents, Janus kinase (JAK) inhibitors, and anti- interleukin (IL6) receptor antibody. However, despite the huge cost and high risk of adverse effects, only approximately 70% of RA patients get a satisfactory response to biologic therapies [13,14,15]. Thus, newer anti-arthritic therapeutic drugs are in urgent need, and natural products represent a lavish and promising source.
Andrographis paniculata has been used in traditional Chinese medicine for centuries. Andrographolide (AD) is the major diterpenoid bioactive compound derived from Andrographis paniculata, which has multiple pharmacological effects, including hepatoprotection [16], anti-angiogenesis [17], anti-thrombosis [18], and anti-inflammation [19], as well as anti-arthritis [20,21]. AD inhibits osteoclast differentiation [22], and triggers the apoptosis and cell cycle arrest of fibroblast-like synoviocytes [23]. AD also prevents oxygen radical production and the N-Formyl-l-methionyl-l-leucyl-l-phenylalanine (fMLP)-induced migration of neutrophils [24]. However, the underlying molecular mechanism of AD on RA remains largely unclear. In this study, we systemically investigated anti-inflammatory mechanism of AD on a murine RA model in vivo and on neutrophils in vitro.

2. Results

2.1. AD Ameliorated Adjuvant-Induced Arthritis (AA) in Mice

We first investigated whether AD can ameliorate Freund’s complete adjuvant-induced arthritis (AA) (Figure 1A). As shown in Figure 1B, AD treatments significantly relieved joint edema in AA mice. The ankle joint swelling was evaluated by using the ankle joint diameter and arthritis scores every 3 days from day 10 to day 37. There was no obvious paw and joint swelling in the control group. The adjuvant induced paw edema and the ankle joint diameter increased from 2.6 mm on day 0 to 4.5 mm on day 9 post induction (Figure 1C). Consistently, the arthritis scores increased from 0 on day 0 to 4.0 on day 19 (Figure 1D), and intraperitoneal injections of AD significantly reduced the ankle joint diameter (Figure 1C) and arthritis scores (Figure 1D) compared with PBS, especially in the 50 mg/kg group (p < 0.05).
The ameliorating effect of AD on AA was also confirmed using HE staining and Safranin O-fast staining of the ankle joints sections. Compared with the control group, the AA group showed a large amount of leukocyte infiltration, cartilage erosion, and synovial hyperplasia in the ankle joints. AD treatment significantly alleviated those joint symptoms (Figure 1E,F). AD treatment also attenuated the up-regulation of pro-inflammatory cytokines (TNF-α, interferon (IFN)-γ, IL-6, and IL-17A) and increased the expression level of anti-inflammatory cytokine IL-10 in the serum of AA mice (Figure 1G). There was no obvious effect of AD treatment alone on mouse ankle joint diameter, arthritis score, or the expression levels of plasma cytokines (Figure 1). These results suggest that AD treatment significantly relieved the inflammation in ankle joints, as well as the whole body.

2.2. AD Decreases Neutrophils Infiltration

Neutrophils play pivotal roles in the pathogenesis of RA [5], and MPO and NE are well-defined markers for neutrophil infiltration. To investigate whether AD treatment influences neutrophil infiltration, immunohistochemistry was performed to evaluate the expression levels of MPO and NE in RA mice joint tissue sections. As shown in Figure 2A,B, MPO and NE expression was significantly upregulated in the RA group and AD treatment downregulated MPO and NE expression. Thus, AD decreased neutrophils infiltration in RA mouse. Air pouch assay also showed that AD treatment significantly suppressed LPS-induced recruitment of neutrophil, as well as total leucocytes (Figure 2C,D). There was no obvious effect of AD treatment alone on neutrophil recruitment in the murine air pouch model (Figure 2C,D).

2.3. AD Accelerates Neutrophil Apoptosis in the Presence of LPS

Neutrophils are short-life leukocytes that undergo spontaneous apoptosis in the peripheral blood [25]. However, when they migrate into RA joints, the apoptosis of neutrophils is delayed, which prolongs the inflammation and increases the release of tissue-damage molecules, such as reactive oxygen species, elastase, and metalloproteases [26,27]. To investigate whether AD can reverse the neutrophil apoptosis delay, purified neutrophils were incubated with LPS or LPS + AD for 16 h, then were stained with Annexin V/PI and analyzed using flow cytometry. As shown in Figure 3A, the LPS treatment significantly reduced the early apoptosis (annexin V+, PI) and late apoptosis (annexin V+, PI+) of neutrophils. LPS + AD treatment significantly increased the late apoptosis of neutrophils and the number of late apoptosis cells was even more than that of the control group. Western blot analyses showed that compared with the LPS treatment, the LPS + AD treatment significantly reduced anti-apoptotic protein Bcl-2 expression and enhanced pro-apoptotic protein Bax expression, which lead to the cleavage and activation of caspase-3. The expression level of cleaved-caspase-3 in the LPS + AD treatment group was even higher than that of the control group (Figure 3B). There was no obvious effect of AD treatment alone on the apoptosis of neutrophils compared with the control group (Figure 3). These results showed that AD accelerated neutrophil apoptosis in the presence of LPS.

2.4. AD Inhibited PMA-Induced NETosis

NETs are extracellular web-like DNA decorated with MPO, NE, and other antimicrobial proteins. NETs are released from neutrophils during inflammation via a distinct process termed NETosis [7,28,29]. Besides granule enzymes, NETs also externalize various autoantigens, such as citrullinated histone 3 (CitH3), which induces specific autoantibodies and autoimmune responses in RA patients [30,31,32]. Peptidylarginine deiminase 4 (PAD4) is a nuclear citrullinating enzyme, which transforms protein l-arginine to l-citrulline and plays an important role in RA pathogenesis [33,34]. PAD4 is also a target of autoantibodies in a subgroup of RA patients and anti-PAD4 autoantibodies can serve as a severity biomarker of RA [35]. Immunostaining of RA mouse ankle joint tissue sections showed that PAD4 was up-regulated in RA mice and the AD treatment significantly reduced PAD4 expression (Figure 4A). Consistently, AD also suppressed CitH3 expression (Figure 4B). Then, purified neutrophils were used to test whether AD could directly suppress the expression of PAD4 and CitH3 in neutrophils. Western blot analyses showed that AD suppressed PMA-induced up-regulation of PAD4 (Figure 4C). Immunostaining analyses of CitH3 showed that AD treatment reversed PMA-induced histone 3 citrullination (Figure 4D).
Consistently, immunostaining analyses of MPO and NE showed that AD treatment reduced the PMA-induced NETosis of neutrophils (Figure 5A,C). Quantification of NETosis with relative DNA area showed that AD treatment significantly suppressed the PMA-induced nuclear de-condensation of neutrophils (Figure 5B,D). There was no obvious effect of AD treatment alone on the levels of PAD4, CitH3, and NETosis in neutrophils compared with the PBS treatment (Figure 4 and Figure 5).

2.5. AD Inhibited Neutrophil Autophagy

Autophagy is a vital cellular degradation mechanism responsible for the metabolism of the cells themselves and the renewal of certain organelles [36]. Previous studies have demonstrated that autophagy is required for NETs formation [37,38,39]. RA patients had significantly higher levels of autophagy in circulating leukocytes compared with healthy controls [40,41]. Immunostaining of purified neutrophils with anti-microtubule-associated protein 1A/1B-light chain 3 (LC3) antibody showed that AD treatment significantly reduced PMA–induced up-regulation of LC3 (Figure 6A,B). Western blot analyses showed that AD treatment down-regulated the expression levels of LC3-II and Beclin1, and up-regulated the expression level of p62 (Figure 6C). These results suggested that AD could significantly inhibit the autophagy of neutrophils.

3. Discussion

AD is a major bioactive constituent extracted from Andrographis paniculata, which belongs to the family of Acanthaceae. Despite recent progress in understanding the anti-arthritic effects of AD, the immunological mechanism that regulated neutrophils activities was unknown. In this study, we comprehensively investigated the role of AD on neutrophils in adjuvant-induced murine arthritis (AA) models. AD-treated AA mice exhibited a remarkable reduction in clinical arthritic scores, neutrophil infiltration, and bone destruction.
Neutrophils play important roles in the onset and perpetuation of RA [42]. The cytokines and mediators produced by neutrophils could amplify the inflammatory response in RA [5]. Previous studies demonstrated that inflammatory mediators, including cytokines, chemokines, and reactive oxygen species (ROS), have pleiotropic effects on inflammation progress [43,44]. TNF-α, IL-6, IL-17A, and IFN-γ have important effects in recruiting and activating neutrophils. These inflammatory cytokines could amplify the immune response by inducing the production of other cytokines and chemokines [45,46,47]. These cytokines are also involved in the pathogenesis of RA by activating chondrocytes and osteoclasts [48,49]. The IFN-γ expression level is significantly higher in RA patients after disease onset compared with health control [50]. As a factor of anti-inflammatory cytokines, IL-10 has the capacity to inhibit neutrophils activation, migration, and degranulation [51,52]. Targeting the inflammatory cytokines including TNF-α, IL-17A, and IL-6 are a pivotal strategy for RA treatment. TNF-α inhibitors (Adalimumab, Etanercept, Certolizumab) and IL-6 inhibitors (Tocilizumab) have been licensed by the Food and Drug Administration (FDA) for treating RA patients [53]. In this study, we found that AD treatment decreased the levels of pro-inflammatory cytokines (TNF-α, IFN-γ, IL-6, IL-17A), while it increased the level of anti-inflammatory cytokine IL-10 in the plasm of AA mice. These data indicated that AD had systematic anti-inflammatory effects in AA mice.
Furthermore, we investigated the mechanism of AD in the AA murine model. First, we discovered that AD inhibited the migration of neutrophils. The steps of neutrophils recruitment cascade include tethering, rolling, adhesion, crawling, and transmigration [54]. Neutrophil migration is an important stage in the inflammatory process of RA [27]. In the circulating blood cells, neutrophils are the first ones to infiltrate into the joints. They are the most plentiful cells in the synovial fluid. Several molecules, such as E-selectin, P-selectin, and IL-6, are involved in the massive migration of neutrophils into the joints. MPO and NE are regarded as markers for neutrophils infiltration in animal model. In the present study, the expression levels of MPO and NE were increased in the RA group compared with the control group. AD treatment significantly reduced MPO and NE expression compared with the RA group. The results indicated that AD could reduce neutrophil infiltration to the RA joints. We also found that AD inhibited LPS-induced recruitment of neutrophils in the air pouch assay. Taken together, AD could reduce neutrophils recruitment in RA.
Second, we demonstrated that AD accelerated neutrophil apoptosis in the presence of LPS. Naive neutrophils have a very short lifespan of 6 to 18 h before undergoing constitutive apoptosis [54]. However, in the synovial fluid of RA joints, the apoptosis of neutrophils are delayed [4]. Activated synovial fluid neutrophils could increase the inflammatory status and promote joint destruction. Neutrophils in the synovial fluid exhibited a higher expression of anti-apoptotic proteins (Bcl-2) and a lower expression of pro-apoptotic proteins (Bax, cleaved caspase-3) [27]. In our study, we showed that AD could reduce the expression of anti-apoptotic protein Bcl-2. Bcl-2 could be phosphorylated by activated extracellular signal-regulated kinase (ERK)1/2 on Serine 70, which blocks the ubiquitin-dependent proteasomal degradation of Bcl-2 [55]. AD inhibits the activation of ERK1/2 [20], which reduces the phosphorylation of Bcl-2 and promotes the degradation of Bcl-2. Additionally, AD increased pro-apoptotic protein Bax and cleaved caspase-3 expression. It is of note that the percentage of late apoptosis neutrophils and the expression level of cleaved-caspase-3 in the LPS + AD treatment group was even higher than the control group, which demonstrated the AD treatment accelerated neutrophil apoptosis in the presence of LPS.
Third, we explored the effects of AD in inhibiting PMA-induced NETosis. NETosis is a featured form of death of neutrophils [30]. NETs are extracellular fibrous DNA networks combined with granular and nuclear proteins. In recent years, researchers have proven that NETs play important roles in the pathogenesis of RA [56]. NETs are correlated with the production of anti-citrullinated protein antibodies (ACPAs) in RA [32]. NETs are an important source of autoantigens to stimulate ACPA production. ACPAs also facilitate the release of peptidyl arginine deiminases (PADs) of neutrophils, which transforms the protein L-arginine to L-citrulline and forms a vicious circle [33]. Apart from autoantibody production, NETs also induce inflammatory cytokine production to enlarge inflammatory response [56]. Recently, researchers reported that NETosis-derived products could be used to assess therapeutic effectiveness in RA patients [57]. MPO, NE, and citrullinated histone H3 (CitH3) are major component of NETs, which are considered biomarkers of NETosis [30]. In this study, we proved that AD significantly inhibited NETs formation and NETs-associated MPO, NE, and CitH3 release. PAD4 is an important enzyme that catalyzes protein citrullination. PAD4 is essential for the formation of NETs. PAD4-mediated histone citrullination is considered to promote NETs formation by facilitating chromosomal DNA expulsion and chromatin decondensation. Many studies have identified that PAD4 was a potential therapeutic target to treat RA [57]. PAD4 expression level is positively correlated with the severity of RA [58]. In our study, we found that AD significantly downregulated the expression of PAD4 in the ankles of RA mice.
Autophagy is a well-known intracellular mechanism for degradation and energy recycling [36]. It is an important regulatory mechanism in immune responses [38]. Neutrophil autophagy facilitates the formation of NETs [37]. LC3-II is the most important parameter to test autophagic flux, which is associated with the maturation of autophagosome [36]. Beclin1 regulates the autophagy and membrane trafficking involved in several pathological and physiological processes [59]. P62 is considered an autophagy substrate used as a reporter of autophagy activity [60]. In this study, we proved that AD could significantly inhibit the autophagy of neutrophils. We found that AD treatment down-regulated the expression levels of LC3-II and Beclin1, and up-regulated the expression of p62. AD treatment significantly reduced the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and ERK1/2 [20], which is required for the activation of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and ROS production [61], thus reducing the NET formation in a ROS-dependent manner. Autophagy is also responsible for the degradation of Bax protein under basal conditions [62], and the inhibition of neutrophil autophagy maybe the reason for the up-regulation of Bax after AD treatment.

4. Materials and Methods

4.1. Reagents and Antibodies

Complete Freund’s adjuvant (CFA), lipopolysaccharide (LPS), andrographolide, and phorbol 12-myristate 13-acetate (PMA) were purchased from Sigma-Aldrich (St Louis, MO, USA). 3-methyladenine (3-MA) was purchased from Selleck Chemicals LLC (Houston, TX, USA). Mouse Th1/Th2/Th17 cytometric bead array (CBA) kits were purchased from BD Biosciences (Franklin Lakes, NJ, USA). Anti-myeloperoxidase, anti-neutrophil elastase, anti-LC3, anti-Beclin1, anti-p62, anti- Bcl-2, anti-Bax, anti-rabbit-horseradish peroxidase, and immunoglobulin G antibodies were purchased from Abcam (Cambridge, MA, USA), and anti-cleaved caspase-3 was purchased from Cell Signaling Technology (Danvers, MA, USA).

4.2. Animals

C57BL/6 mice (female, 8 weeks old) were purchased from the Chinese Academy of Military Medical Sciences (Beijing, China). Mice were maintained in standard housing cages under specific pathogen-free conditions. All experimental procedures were reviewed and approved by the Animal Care and Use Committee of the Beijing University of Chinese Medicine (ethical approval number: BUCM-4-2018060416-2020, approved date: 4 June, 2018). All experiments were performed in accordance with the institutional guidelines for the Care and Use of Laboratory Animals.

4.3. Induction of Adjuvant-Induced Arthritis (AA)

Twenty microliters of CFA was injected into the joint space and four periarticular sites, respectively. Forty mice were randomly divided into four groups: control group (PBS), model group (AA), 25 mg/kg AD group (AD (25 mg/kg)), and 50 mg/kg AD group (AD (50 mg/kg)). To assess the ankle joint swelling, the joint diameters were measured with a pocket thickness gauge (Mitutoyo, Kawasaki, Japan). Arthritis severity was assessed using arthritis using a 0–4 scoring criterion scale: 0—normal, 1—slightly redness or swelling of the ankle joint, 2—moderate swelling and slightly activity limited, 3—obvious swelling and activity limited, and 4—severe swelling and activity disorder.

4.4. Histochemistry and Immunohistochemistry

Mice were sacrificed on day 37. The ankle joints were collected, fixed in 4% paraformaldehyde for 48 h, decalcified in 10% ethylenediaminetetraacetic acid solution, and embedded in paraffin. The tissue sections were stained with hematoxylin and eosin (HE), Weigert’s Iron Hematoxylin solution (Sigma-Aldrich) and Fast Green solution (Sigma-Aldrich) for histopathological analysis. The MPO and NE expression levels were examined using immunohistochemistry with anti-NE and anti-MPO antibodies in accordance with the instructions of the manufacturer.

4.5. Cytokine Analysis

Serum samples were obtained from mice’s blood in each group on day 37. The concentrations of cytokines (TNF-α, IFN-γ, IL-6, IL-2, IL-17A, IL-4, and IL-10) were measured with the Mouse Th1/Th2/Th17 Cytometric Bead Array (CBA) Human Chemokine Kit following the manufacturer’s introduction.

4.6. Air Pouch Experiments

C57BL/6 male mice (≥5/group) were anaesthetized using chloral hydrate, and 3 mL of sterilized air (filtered through a 0.22-μm filter; Millipore, Billerica, MA, USA) was injected into the skin on days 0 and 3 with a 26-gauge needle to produce an air pouch on the back. On day 6, 1 mL of buffer (control) or 1 μg/mL LPS with or without 25 μM AD was injected into the air pouches of mice 6 h before the mice were euthanized via CO2 asphyxiation. The air pouches were washed once with 1 mL and then twice with 2 mL of Hanks’s balanced salt solution (HBSS) containing 10 mM ethylenediamine tetraacetic acid (EDTA), and the exudates were centrifuged at 100× g for 10 min at room temperature. Cells were resuspended in 1 mL of HBSS containing 10 mM EDTA and stained with Wright’s stain to quantify the neutrophil populations.

4.7. Isolation of Mouse Neutrophils and Culture

Peritoneal exudate cells were collected from lavage fluid of mice inoculated intraperitoneally with 1 mL of 10% protease peptone 10–12 h previously. Cells were resuspended in 1 mL of RPMI-1640 supplemented with 10% fetal bovine serum (FBS), layered onto a two-step (54.8%/70.2%) discontinuous Percoll gradient, and centrifuged at 1500× g for 30 min at 22 °C. Neutrophils (≥95%, approximately 1 × 107 neutrophils/mouse) were recovered from the lower interface and were cultured at 37 °C with 5% CO2 in humidified incubator.

4.8. Western Blot Analysis

Neutrophils were collected and were lysed with radioimmunoprecipitation assay (RIPA) buffer containing protease inhibitor. Protein concentration was measured using a BCA Protein Assay Kit (Thermo Fisher Scientific, Tewksbury, MA, USA). Samples were separated using 10% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and were transferred to nitrocellulose membranes (Millipore, Billerica, MA, USA). The membranes were blocked with 5% non-fat dried milk for 1 h at room temperature and were incubated with primary antibodies against Bax, Bcl-2, cleaved-caspase-3, Beclin1, p62, LC3, or β-actin at 4 °C overnight. After three washes, the membranes were incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies for 1 h at room temperature. The proteins were detected using an enhanced chemiluminescence reagent (GE Healthcare, Chicago, IL, USA). The intensities of protein bands were quantified using ImageJ software (version 1.43 National Institutes of Health, Bethesda, MD, USA).

4.9. Immunofluorescence Assay

Neutrophils were seeded at a density of 1 × 106 cells/mL on glass coverslips in a six-well plate. The cells were incubated with or without AD (25 μM) and PMA (25 nM) for 4 h. Then, cells were fixed with 4% paraformaldehyde for 15 min and were permeabilized with 0.1% Triton X-100 in PBS for 10 min. After blocking with 5% BSA/PBS for 1 h, cells were incubated with anti-MPO, anti-NE, or anti-LC3 antibodies at 4 °C overnight and subsequently with Alexa-FluorVR-555-labeled secondary antibody (Invitrogen, Carlsbad, CA, USA) at room temperature for 1 h. Nuclei were counterstained with 4’,6-diamidino-2-phenylindole (DAPI) for 5 min and images were captured using a confocal microscope (FV1000, Olympus, Japan). Relative DNA area was measured with Image-Pro Plus software (version 6.0, Media Cybernetics Corporation, Rockville, MD, USA).

4.10. Flow Cytometry Analysis

Purified neutrophils were resuspended in culture medium at a density of 106 cells and 2 mL of cell suspension was plated in a six-well plate per well. Cells were treated with LPS and/or AD for 16 h. Cells were resuspended in 200 μL of 1× binding buffer after being washed twice with cold PBS. Fluorescein isothiocyanate (FITC)-annexin V and propidium iodide (PI) (5 μL each) were added and incubated for 15 min at room temperature (RT) (25 °C) in the dark with a gentle vortex. Then, cells were washed and analyzed using flow cytometry within 1 h. annexin V+ and PI cells were considered as early apoptosis cells and annexin V+ and PI+ cells were considered as late apoptosis cells.

4.11. Statistical Analysis

Data were expressed as the mean ± standard deviation (SD). Inter group comparisons were performed by using one-way analysis of variance. If the data did not satisfy a normal distribution, the rank sum test was used. A p-value less than 0.05 was considered to be significant.

5. Conclusions

The present study investigated the anti-arthritis mechanism of AD in a murine RA model and our results showed that AD alleviated murine arthritis by promoting neutrophil apoptosis and suppressed the autophagy-dependent NETosis of neutrophils (Figure 7). These findings suggest that AD has considerable pharmaceutical potential for RA treatment.

Author Contributions

Conceived and designed the experiments: A.X., G.H., and X.L. Performed the experiments: X.L., Q.Z., H.J., and M.Z. Analyzed the data: X.L., K.Y., Q.L., and G.H. Contributed reagents/materials/analysis tools: K.Y., G.H., and A.X. Wrote the paper: X.L., K.Y., G.H., and A.X. All authors have read and approved the manuscript.

Funding

This work was supported by projects of the National Natural Science Foundation of China (grant numbers 81430099 and 31500704), project of International Cooperation and Exchanges (grant number 2014DFA32950), and research projects from Beijing University of Chinese Medicine (grant number 1000041510049, BUCM-2019-JCRC006, and 2019-JYB-TD013).

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. McInnes, I.B.; Schett, G. The pathogenesis of rheumatoid arthritis. N Engl J. Med. 2011, 365, 2205–2219. [Google Scholar] [CrossRef] [PubMed]
  2. Smolen, J.S.; Aletaha, D.; McInnes, I.B. Rheumatoid arthritis. Lancet 2016, 388, 2023–2038. [Google Scholar] [CrossRef]
  3. Wright, H.L.; Moots, R.J.; Edwards, S.W. The multifactorial role of neutrophils in rheumatoid arthritis. Nat. Rev. Rheumatol. 2014, 10, 593–601. [Google Scholar] [CrossRef] [PubMed]
  4. Thieblemont, N.; Wright, H.L.; Edwards, S.W.; Witko-Sarsat, V. Human neutrophils in auto-immunity. Semin. Immunol. 2016, 28, 159–173. [Google Scholar] [CrossRef] [PubMed]
  5. Cecchi, I.; Arias de la Rosa, I.; Menegatti, E.; Roccatello, D.; Collantes-Estevez, E.; Lopez-Pedrera, C.; Barbarroja, N. Neutrophils: Novel key players in Rheumatoid Arthritis. Current and future therapeutic targets. Autoimmun. Rev. 2018, 17, 1138–1149. [Google Scholar] [CrossRef] [PubMed]
  6. Lefrancais, E.; Roga, S.; Gautier, V.; Gonzalez-de-Peredo, A.; Monsarrat, B.; Girard, J.P.; Cayrol, C. IL-33 is processed into mature bioactive forms by neutrophil elastase and cathepsin G. Proc. Natl. Acad. Sci. USA 2012, 109, 1673–1678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef] [PubMed]
  8. Khandpur, R.; Carmona-Rivera, C.; Vivekanandan-Giri, A.; Gizinski, A.; Yalavarthi, S.; Knight, J.S.; Friday, S.; Li, S.; Patel, R.M.; Subramanian, V.; et al. NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci. Transl. Med. 2013, 5, 178ra40. [Google Scholar] [CrossRef] [PubMed]
  9. Chakravarti, A.; Raquil, M.A.; Tessier, P.; Poubelle, P.E. Surface RANKL of Toll-like receptor 4-stimulated human neutrophils activates osteoclastic bone resorption. Blood 2009, 114, 1633–1644. [Google Scholar] [CrossRef] [PubMed]
  10. Chen, W.; Wang, Q.; Ke, Y.; Lin, J. Neutrophil Function in an Inflammatory Milieu of Rheumatoid Arthritis. J. Immunol. Res. 2018, 2018, 8549329. [Google Scholar] [CrossRef]
  11. Kaplan, M.J. Role of neutrophils in systemic autoimmune diseases. Arthritis Res. Ther. 2013, 15, 219. [Google Scholar] [CrossRef] [PubMed]
  12. Tanaka, D.; Kagari, T.; Doi, H.; Shimozato, T. Essential role of neutrophils in anti-type II collagen antibody and lipopolysaccharide-induced arthritis. Immunology 2006, 119, 195–202. [Google Scholar] [CrossRef] [PubMed]
  13. Smolen, J.S.; Kay, J.; Doyle, M.K.; Landewe, R.; Matteson, E.L.; Wollenhaupt, J.; Gaylis, N.; Murphy, F.T.; Neal, J.S.; Zhou, Y.; et al. Golimumab in patients with active rheumatoid arthritis after treatment with tumour necrosis factor alpha inhibitors (GO-AFTER study): A multicentre, randomised, double-blind, placebo-controlled, phase III trial. Lancet 2009, 374, 210–221. [Google Scholar] [CrossRef]
  14. Van Vollenhoven, R.F.; Fleischmann, R.; Cohen, S.; Lee, E.B.; Garcia Meijide, J.A.; Wagner, S.; Forejtova, S.; Zwillich, S.H.; Gruben, D.; Koncz, T.; et al. Tofacitinib or adalimumab versus placebo in rheumatoid arthritis. N Engl. J. Med. 2012, 367, 508–519. [Google Scholar] [CrossRef] [PubMed]
  15. Smolen, J.S.; Beaulieu, A.; Rubbert-Roth, A.; Ramos-Remus, C.; Rovensky, J.; Alecock, E.; Woodworth, T.; Alten, R.; Investigators, O. Effect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): A double-blind, placebo-controlled, randomised trial. Lancet 2008, 371, 987–997. [Google Scholar] [CrossRef]
  16. Handa, S.S.; Sharma, A. Hepatoprotective activity of andrographolide against galactosamine & paracetamol intoxication in rats. Indian J. Med. Res. 1990, 92, 284–292. [Google Scholar]
  17. Sheeja, K.; Guruvayoorappan, C.; Kuttan, G. Antiangiogenic activity of Andrographis paniculata extract and andrographolide. Int. Immunopharmacol. 2007, 7, 211–221. [Google Scholar] [CrossRef]
  18. Thisoda, P.; Rangkadilok, N.; Pholphana, N.; Worasuttayangkurn, L.; Ruchirawat, S.; Satayavivad, J. Inhibitory effect of Andrographis paniculata extract and its active diterpenoids on platelet aggregation. Eur. J. Pharmacol. 2006, 553, 39–45. [Google Scholar] [CrossRef]
  19. Hidalgo, M.A.; Romero, A.; Figueroa, J.; Cortes, P.; Concha, II; Hancke, J.L.; Burgos, R.A. Andrographolide interferes with binding of nuclear factor-kappaB to DNA in HL-60-derived neutrophilic cells. Br. J. Pharmacol. 2005, 144, 680–686. [Google Scholar] [CrossRef]
  20. Li, Z.Z.; Tan, J.P.; Wang, L.L.; Li, Q.H. Andrographolide Benefits Rheumatoid Arthritis via Inhibiting MAPK Pathways. Inflammation 2017, 40, 1599–1605. [Google Scholar] [CrossRef]
  21. Gupta, S.; Mishra, K.P.; Singh, S.B.; Ganju, L. Inhibitory effects of andrographolide on activated macrophages and adjuvant-induced arthritis. Inflammopharmacology 2018, 26, 447–456. [Google Scholar] [CrossRef] [PubMed]
  22. Zhai, Z.J.; Li, H.W.; Liu, G.W.; Qu, X.H.; Tian, B.; Yan, W.; Lin, Z.; Tang, T.T.; Qin, A.; Dai, K.R. Andrographolide suppresses RANKL-induced osteoclastogenesis in vitro and prevents inflammatory bone loss in vivo. Br. J. Pharmacol. 2014, 171, 663–675. [Google Scholar] [CrossRef]
  23. Yan, J.; Chen, Y.; He, C.; Yang, Z.Z.; Lu, C.; Chen, X.S. Andrographolide induces cell cycle arrest and apoptosis in human rheumatoid arthritis fibroblast-like synoviocytes. Cell Biol. Toxicol. 2012, 28, 47–56. [Google Scholar] [CrossRef] [PubMed]
  24. Shen, Y.C.; Chen, C.F.; Chiou, W.F. Andrographolide prevents oxygen radical production by human neutrophils: Possible mechanism(s) involved in its anti-inflammatory effect. Br. J. Pharmacol. 2002, 135, 399–406. [Google Scholar] [CrossRef] [PubMed]
  25. Ng, L.G.; Ostuni, R.; Hidalgo, A. Heterogeneity of neutrophils. Nat. Rev. Immunol. 2019, 19, 255–265. [Google Scholar] [CrossRef] [PubMed]
  26. Collison, J. Rheumatoid arthritis: Tipping the balance towards resolution. Nat. Rev. Rheumatol. 2016, 12, 622. [Google Scholar] [CrossRef] [PubMed]
  27. Smolen, J.S.; Aletaha, D.; Barton, A.; Burmester, G.R.; Emery, P.; Firestein, G.S.; Kavanaugh, A.; McInnes, I.B.; Solomon, D.H.; Strand, V.; et al. Rheumatoid arthritis. Nat. Rev. Dis. Primers 2018, 4, 18001. [Google Scholar] [CrossRef]
  28. Yousefi, S.; Gold, J.A.; Andina, N.; Lee, J.J.; Kelly, A.M.; Kozlowski, E.; Schmid, I.; Straumann, A.; Reichenbach, J.; Gleich, G.J.; et al. Catapult-like release of mitochondrial DNA by eosinophils contributes to antibacterial defense. Nat. Med. 2008, 14, 949–953. [Google Scholar] [CrossRef]
  29. Boeltz, S.; Amini, P.; Anders, H.J.; Andrade, F.; Bilyy, R.; Chatfield, S.; Cichon, I.; Clancy, D.M.; Desai, J.; Dumych, T.; et al. To NET or not to NET: Current opinions and state of the science regarding the formation of neutrophil extracellular traps. Cell Death Differ. 2019, 26, 395–408. [Google Scholar] [CrossRef]
  30. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef]
  31. Aleyd, E.; Al, M.; Tuk, C.W.; van der Laken, C.J.; van Egmond, M. IgA Complexes in Plasma and Synovial Fluid of Patients with Rheumatoid Arthritis Induce Neutrophil Extracellular Traps via FcalphaRI. J. Immunol. 2016, 197, 4552–4559. [Google Scholar] [CrossRef] [PubMed]
  32. Demoruelle, M.K.; Harrall, K.K.; Ho, L.; Purmalek, M.M.; Seto, N.L.; Rothfuss, H.M.; Weisman, M.H.; Solomon, J.J.; Fischer, A.; Okamoto, Y.; et al. Anti-Citrullinated Protein Antibodies Are Associated With Neutrophil Extracellular Traps in the Sputum in Relatives of Rheumatoid Arthritis Patients. Arthritis Rheumatol. 2017, 69, 1165–1175. [Google Scholar] [CrossRef] [PubMed]
  33. Harris, M.L.; Darrah, E.; Lam, G.K.; Bartlett, S.J.; Giles, J.T.; Grant, A.V.; Gao, P.; Scott, W.W., Jr.; El-Gabalawy, H.; Casciola-Rosen, L.; et al. Association of autoimmunity to peptidyl arginine deiminase type 4 with genotype and disease severity in rheumatoid arthritis. Arthritis Rheum. 2008, 58, 1958–1967. [Google Scholar] [CrossRef] [PubMed]
  34. Halvorsen, E.H.; Pollmann, S.; Gilboe, I.M.; van der Heijde, D.; Landewe, R.; Odegard, S.; Kvien, T.K.; Molberg, O. Serum IgG antibodies to peptidylarginine deiminase 4 in rheumatoid arthritis and associations with disease severity. Ann. Rheum. Dis. 2008, 67, 414–417. [Google Scholar] [CrossRef] [PubMed]
  35. Reyes-Castillo, Z.; Munoz-Valle, J.F.; Llamas-Covarrubias, M.A. Clinical and immunological aspects of anti-peptidylarginine deiminase type 4 (anti-PAD4) autoantibodies in rheumatoid arthritis. Autoimmun. Rev. 2018, 17, 94–102. [Google Scholar] [CrossRef] [PubMed]
  36. Doherty, J.; Baehrecke, E.H. Life, death and autophagy. Nat. Cell Biol. 2018, 20, 1110–1117. [Google Scholar] [CrossRef] [PubMed]
  37. Remijsen, Q.; Vanden Berghe, T.; Wirawan, E.; Asselbergh, B.; Parthoens, E.; De Rycke, R.; Noppen, S.; Delforge, M.; Willems, J.; Vandenabeele, P. Neutrophil extracellular trap cell death requires both autophagy and superoxide generation. Cell Res. 2011, 21, 290–304. [Google Scholar] [CrossRef] [PubMed]
  38. Bhattacharya, A.; Wei, Q.; Shin, J.N.; Abdel Fattah, E.; Bonilla, D.L.; Xiang, Q.; Eissa, N.T. Autophagy Is Required for Neutrophil-Mediated Inflammation. Cell Rep. 2015, 12, 1731–1739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Park, S.Y.; Shrestha, S.; Youn, Y.J.; Kim, J.K.; Kim, S.Y.; Kim, H.J.; Park, S.H.; Ahn, W.G.; Kim, S.; Lee, M.G.; et al. Autophagy Primes Neutrophils for Neutrophil Extracellular Trap Formation during Sepsis. Am. J. Respir. Crit. Care Med. 2017, 196, 577–589. [Google Scholar] [CrossRef]
  40. Chen, Y.M.; Chang, C.Y.; Chen, H.H.; Hsieh, C.W.; Tang, K.T.; Yang, M.C.; Lan, J.L.; Chen, D.Y. Association between autophagy and inflammation in patients with rheumatoid arthritis receiving biologic therapy. Arthritis Res. Ther. 2018, 20, 268. [Google Scholar] [CrossRef]
  41. Dai, Y.; Ding, J.; Yin, W.; He, Y.; Yu, F.; Ye, C.; Hu, S.; Yu, Y. Increased Autophagy Enhances the Resistance to Tumor Necrosis Factor-Alpha Treatment in Rheumatoid Arthritis Human Fibroblast-Like Synovial Cell. Biomed. Res. Int. 2018, 2018, 4941027. [Google Scholar] [CrossRef] [PubMed]
  42. Catrina, A.I.; Svensson, C.I.; Malmstrom, V.; Schett, G.; Klareskog, L. Mechanisms leading from systemic autoimmunity to joint-specific disease in rheumatoid arthritis. Nat. Rev. Rheumatol. 2017, 13, 79–86. [Google Scholar] [CrossRef] [PubMed]
  43. Dewas, C.; Dang, P.M.; Gougerot-Pocidalo, M.A.; El-Benna, J. TNF-alpha induces phosphorylation of p47(phox) in human neutrophils: partial phosphorylation of p47phox is a common event of priming of human neutrophils by TNF-alpha and granulocyte-macrophage colony-stimulating factor. J. Immunol. 2003, 171, 4392–4398. [Google Scholar] [CrossRef] [PubMed]
  44. Cross, A.; Moots, R.J.; Edwards, S.W. The dual effects of TNFalpha on neutrophil apoptosis are mediated via differential effects on expression of Mcl-1 and Bfl-1. Blood 2008, 111, 878–884. [Google Scholar] [CrossRef] [PubMed]
  45. Amarilyo, G.; Lourenco, E.V.; Shi, F.D.; La Cava, A. IL-17 promotes murine lupus. J. Immunol. 2014, 193, 540–543. [Google Scholar] [CrossRef] [PubMed]
  46. McInnes, I.B.; Buckley, C.D.; Isaacs, J.D. Cytokines in rheumatoid arthritis - shaping the immunological landscape. Nat. Rev. Rheumatol. 2016, 12, 63–68. [Google Scholar] [CrossRef]
  47. Taylor, P.R.; Roy, S.; Leal, S.M., Jr.; Sun, Y.; Howell, S.J.; Cobb, B.A.; Li, X.; Pearlman, E. Activation of neutrophils by autocrine IL-17A-IL-17RC interactions during fungal infection is regulated by IL-6, IL-23, RORgammat and dectin-2. Nat. Immunol. 2014, 15, 143–151. [Google Scholar] [CrossRef]
  48. Lubberts, E.; Koenders, M.I.; van den Berg, W.B. The role of T-cell interleukin-17 in conducting destructive arthritis: lessons from animal models. Arthritis Res. Ther. 2005, 7, 29–37. [Google Scholar] [CrossRef]
  49. Burska, A.; Boissinot, M.; Ponchel, F. Cytokines as biomarkers in rheumatoid arthritis. Mediators Inflamm. 2014, 2014, 545493. [Google Scholar] [CrossRef]
  50. Kokkonen, H.; Soderstrom, I.; Rocklov, J.; Hallmans, G.; Lejon, K.; Rantapaa Dahlqvist, S. Up-regulation of cytokines and chemokines predates the onset of rheumatoid arthritis. Arthritis Rheum. 2010, 62, 383–391. [Google Scholar] [CrossRef]
  51. Ouyang, W.; Rutz, S.; Crellin, N.K.; Valdez, P.A.; Hymowitz, S.G. Regulation and functions of the IL-10 family of cytokines in inflammation and disease. Annu. Rev. Immunol. 2011, 29, 71–109. [Google Scholar] [CrossRef] [PubMed]
  52. Kulkarni, U.; Karsten, C.M.; Kohler, T.; Hammerschmidt, S.; Bommert, K.; Tiburzy, B.; Meng, L.; Thieme, L.; Recke, A.; Ludwig, R.J.; et al. IL-10 mediates plasmacytosis-associated immunodeficiency by inhibiting complement-mediated neutrophil migration. J. Allergy Clin. Immunol. 2016, 137, 1487–1497.e6. [Google Scholar] [CrossRef] [PubMed]
  53. Siebert, S.; Tsoukas, A.; Robertson, J.; McInnes, I. Cytokines as therapeutic targets in rheumatoid arthritis and other inflammatory diseases. Pharmacol. Rev. 2015, 67, 280–309. [Google Scholar] [CrossRef] [PubMed]
  54. Liew, P.X.; Kubes, P. The Neutrophil’s Role During Health and Disease. Physiol. Rev. 2019, 99, 1223–1248. [Google Scholar] [CrossRef] [PubMed]
  55. Deng, X.; Ruvolo, P.; Carr, B.; May, W.S., Jr. Survival function of ERK1/2 as IL-3-activated, staurosporine-resistant Bcl2 kinases. Proc. Natl. Acad. Sci. USA 2000, 97, 1578–1583. [Google Scholar] [CrossRef] [Green Version]
  56. Perez-Sanchez, C.; Ruiz-Limon, P.; Aguirre, M.A.; Jimenez-Gomez, Y.; Arias-de la Rosa, I.; Abalos-Aguilera, M.C.; Rodriguez-Ariza, A.; Castro-Villegas, M.C.; Ortega-Castro, R.; Segui, P.; et al. Diagnostic potential of NETosis-derived products for disease activity, atherosclerosis and therapeutic effectiveness in Rheumatoid Arthritis patients. J. Autoimmun. 2017, 82, 31–40. [Google Scholar] [CrossRef]
  57. Koushik, S.; Joshi, N.; Nagaraju, S.; Mahmood, S.; Mudeenahally, K.; Padmavathy, R.; Jegatheesan, S.K.; Mullangi, R.; Rajagopal, S. PAD4: pathophysiology, current therapeutics and future perspective in rheumatoid arthritis. Expert. Opin. Ther. Targets 2017, 21, 433–447. [Google Scholar] [CrossRef]
  58. Turunen, S.; Huhtakangas, J.; Nousiainen, T.; Valkealahti, M.; Melkko, J.; Risteli, J.; Lehenkari, P. Rheumatoid arthritis antigens homocitrulline and citrulline are generated by local myeloperoxidase and peptidyl arginine deiminases 2, 3 and 4 in rheumatoid nodule and synovial tissue. Arthritis Res. Ther. 2016, 18, 239. [Google Scholar] [CrossRef]
  59. Guo, D.; Ma, J.; Yan, L.; Li, T.; Li, Z.; Han, X.; Shui, S. Down-Regulation of Lncrna MALAT1 Attenuates Neuronal Cell Death Through Suppressing Beclin1-Dependent Autophagy by Regulating Mir-30a in Cerebral Ischemic Stroke. Cell Physiol. Biochem. 2017, 43, 182–194. [Google Scholar] [CrossRef]
  60. Matsumoto, G.; Wada, K.; Okuno, M.; Kurosawa, M.; Nukina, N. Serine 403 phosphorylation of p62/SQSTM1 regulates selective autophagic clearance of ubiquitinated proteins. Mol. Cell 2011, 44, 279–289. [Google Scholar] [CrossRef]
  61. Ilatovskaya, D.V.; Pavlov, T.S.; Levchenko, V.; Staruschenko, A. ROS production as a common mechanism of ENaC regulation by EGF, insulin, and IGF-1. Am. J. Physiol. Cell Physiol. 2013, 304, C102–C111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Guan, J.J.; Zhang, X.D.; Sun, W.; Qi, L.; Wu, J.C.; Qin, Z.H. DRAM1 regulates apoptosis through increasing protein levels and lysosomal localization of BAX. Cell Death Dis. 2015, 6, e1624. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Andrographolide (AD) ameliorated adjuvant-induced arthritis in mice. (A) Schematic diagram of the study. (B) Representative pictures of the mouse hind paw on day 37. (C) Joint swelling was assessed by measuring the ankle joint diameter with a pocket thickness gauge (n = 10). Data are presented as the means ± SD. (D) The severity of arthritis was graded using a 0–4 arthritis scoring criterion: 0—normal, 1—slightly redness or swelling of the ankle joint, 2—moderate swelling and slightly activity limited, 3—obvious swelling and activity limited, and 4—severe swelling and activity disorder. Data are presented as the means ± SD (n = 10). (E) Hematoxylin and eosin staining of ankle joint sections of each treatment group on day 37. Images of representative sections are shown. (F) Assessment of articular cartilage damage by staining with Safranin O-fast green. Images of representative sections are shown. (G) The cytokine protein levels in the plasma of AA mice were measure using a Cytometric Bead Array (CBA) Human Chemokine Kit (n =10). Data represent mean ± SD (n = 10). * p < 0.05, ** p < 0.001. CFA: Complete Freund’s adjuvant; AA: adjuvant-induced arthritis; TNF: tumor necrosis factor, IFN: interferon; IL: interleukin.
Figure 1. Andrographolide (AD) ameliorated adjuvant-induced arthritis in mice. (A) Schematic diagram of the study. (B) Representative pictures of the mouse hind paw on day 37. (C) Joint swelling was assessed by measuring the ankle joint diameter with a pocket thickness gauge (n = 10). Data are presented as the means ± SD. (D) The severity of arthritis was graded using a 0–4 arthritis scoring criterion: 0—normal, 1—slightly redness or swelling of the ankle joint, 2—moderate swelling and slightly activity limited, 3—obvious swelling and activity limited, and 4—severe swelling and activity disorder. Data are presented as the means ± SD (n = 10). (E) Hematoxylin and eosin staining of ankle joint sections of each treatment group on day 37. Images of representative sections are shown. (F) Assessment of articular cartilage damage by staining with Safranin O-fast green. Images of representative sections are shown. (G) The cytokine protein levels in the plasma of AA mice were measure using a Cytometric Bead Array (CBA) Human Chemokine Kit (n =10). Data represent mean ± SD (n = 10). * p < 0.05, ** p < 0.001. CFA: Complete Freund’s adjuvant; AA: adjuvant-induced arthritis; TNF: tumor necrosis factor, IFN: interferon; IL: interleukin.
Ijms 20 05035 g001
Figure 2. AD significantly decreased neutrophil infiltration. (A,B) Immunohistochemical analysis was performed to detect myeloperoxidase (MPO) (A) and neutrophil elastase (NE) (B) expression in the ankle joint tissue sections of each treatment group on day 37 (n = 10). Representative images are shown. (C,D) The air pouch assay showed that the AD (25 μM) treatment significantly suppressed lipopolysaccharide (LPS)-induced recruitment of total leucocytes (C) and neutrophils (D). The numbers of neutrophils and total leukocytes in the air pouch are expressed as means ± SD. ** p < 0.01 (n ≥ 5).
Figure 2. AD significantly decreased neutrophil infiltration. (A,B) Immunohistochemical analysis was performed to detect myeloperoxidase (MPO) (A) and neutrophil elastase (NE) (B) expression in the ankle joint tissue sections of each treatment group on day 37 (n = 10). Representative images are shown. (C,D) The air pouch assay showed that the AD (25 μM) treatment significantly suppressed lipopolysaccharide (LPS)-induced recruitment of total leucocytes (C) and neutrophils (D). The numbers of neutrophils and total leukocytes in the air pouch are expressed as means ± SD. ** p < 0.01 (n ≥ 5).
Ijms 20 05035 g002
Figure 3. AD accelerates neutrophil apoptosis in the presence of LPS. Neutrophils were stimulated with LPS (10 ng/mL) or LPS + AD (25 μM) for 16 h, and then the cells were collected for flow cytometry or Western blot analysis. (A) The effects of AD on neutrophil apoptosis were analyzed using flow cytometry. (B) Western blot analyses of B cell lymphoma 2 (Bcl-2), Bcl-2-associated X (Bax), and caspase-3. These experiments were repeated three times and data are expressed as the mean ± SD. * p < 0.05, ** p < 0.01.
Figure 3. AD accelerates neutrophil apoptosis in the presence of LPS. Neutrophils were stimulated with LPS (10 ng/mL) or LPS + AD (25 μM) for 16 h, and then the cells were collected for flow cytometry or Western blot analysis. (A) The effects of AD on neutrophil apoptosis were analyzed using flow cytometry. (B) Western blot analyses of B cell lymphoma 2 (Bcl-2), Bcl-2-associated X (Bax), and caspase-3. These experiments were repeated three times and data are expressed as the mean ± SD. * p < 0.05, ** p < 0.01.
Ijms 20 05035 g003
Figure 4. AD inhibited peptidylarginine deiminase 4 (PAD4) expression and histone 3 citrullination. (A,B) Immunohistochemical analysis was performed to detect PAD4 (A) and citrullinated histone 3 (CitH3) (B) expression in the ankle joint tissue sections of each treatment group on day 37 (n = 10). Representative images are shown. (C) PAD4 expression levels were measured using Western blotting and the relative gray values were quantified using Image J. The β-actin was an internal control. Data indicate mean ± SD of three independent experiments, * p < 0.05, ** p < 0.01. (D) Phorbol 12-myristate 13-acetate (PMA)-induced neutrophil extracellular trap (NET) formation was visualized via staining neutrophils with 4’,6-diamidino-2-phenylindole (DAPI) (blue) and an anti-CitH3 antibody (red) and observed using confocal microscopy. Scale bar represents 10 μm. Representative images obtained from more than three experiments are shown.
Figure 4. AD inhibited peptidylarginine deiminase 4 (PAD4) expression and histone 3 citrullination. (A,B) Immunohistochemical analysis was performed to detect PAD4 (A) and citrullinated histone 3 (CitH3) (B) expression in the ankle joint tissue sections of each treatment group on day 37 (n = 10). Representative images are shown. (C) PAD4 expression levels were measured using Western blotting and the relative gray values were quantified using Image J. The β-actin was an internal control. Data indicate mean ± SD of three independent experiments, * p < 0.05, ** p < 0.01. (D) Phorbol 12-myristate 13-acetate (PMA)-induced neutrophil extracellular trap (NET) formation was visualized via staining neutrophils with 4’,6-diamidino-2-phenylindole (DAPI) (blue) and an anti-CitH3 antibody (red) and observed using confocal microscopy. Scale bar represents 10 μm. Representative images obtained from more than three experiments are shown.
Ijms 20 05035 g004
Figure 5. (A,C) PMA-induced NET formation was visualized via staining neutrophils with DAPI (blue) and an anti-MPO antibody ((A); red) or anti-NE antibody ((C); red) and observed using confocal microscopy, Scale bar represents 10 μm. Representative images obtained from more than three experiments are shown. (B,D) Relative DNA area of 100 neutrophils in A (B) and C (D). The data were obtained from three independent experiments.
Figure 5. (A,C) PMA-induced NET formation was visualized via staining neutrophils with DAPI (blue) and an anti-MPO antibody ((A); red) or anti-NE antibody ((C); red) and observed using confocal microscopy, Scale bar represents 10 μm. Representative images obtained from more than three experiments are shown. (B,D) Relative DNA area of 100 neutrophils in A (B) and C (D). The data were obtained from three independent experiments.
Ijms 20 05035 g005
Figure 6. AD inhibited neutrophil autophagy. (A) Immunostaining of purified neutrophils with anti- microtubule-associated protein 1A/1B-light chain 3 (LC3) antibody. Representative images of more than three experiments are shown. Scale bar represents 10 μm. (B) Western blot analyses of LC3, Beclin1, and p62 in neutrophils treated with PMA (25 nM) or PMA (25 nM) + AD (25 μM) for 4 h. The relative gray values were quantified using Image J. The β-actin was an internal control. Data indicate mean ± SD of three independent experiments, * p < 0.05, ** p < 0.01. 3-MA: 3-methyladenine.
Figure 6. AD inhibited neutrophil autophagy. (A) Immunostaining of purified neutrophils with anti- microtubule-associated protein 1A/1B-light chain 3 (LC3) antibody. Representative images of more than three experiments are shown. Scale bar represents 10 μm. (B) Western blot analyses of LC3, Beclin1, and p62 in neutrophils treated with PMA (25 nM) or PMA (25 nM) + AD (25 μM) for 4 h. The relative gray values were quantified using Image J. The β-actin was an internal control. Data indicate mean ± SD of three independent experiments, * p < 0.05, ** p < 0.01. 3-MA: 3-methyladenine.
Ijms 20 05035 g006
Figure 7. A graphical summary shows how andrographolide ameliorates rheumatoid arthritis by regulating the apoptosis–NETosis balance of neutrophils.
Figure 7. A graphical summary shows how andrographolide ameliorates rheumatoid arthritis by regulating the apoptosis–NETosis balance of neutrophils.
Ijms 20 05035 g007

Share and Cite

MDPI and ACS Style

Li, X.; Yuan, K.; Zhu, Q.; Lu, Q.; Jiang, H.; Zhu, M.; Huang, G.; Xu, A. Andrographolide Ameliorates Rheumatoid Arthritis by Regulating the Apoptosis–NETosis Balance of Neutrophils. Int. J. Mol. Sci. 2019, 20, 5035. https://doi.org/10.3390/ijms20205035

AMA Style

Li X, Yuan K, Zhu Q, Lu Q, Jiang H, Zhu M, Huang G, Xu A. Andrographolide Ameliorates Rheumatoid Arthritis by Regulating the Apoptosis–NETosis Balance of Neutrophils. International Journal of Molecular Sciences. 2019; 20(20):5035. https://doi.org/10.3390/ijms20205035

Chicago/Turabian Style

Li, Xiaohong, Kai Yuan, Qingqing Zhu, Qingyi Lu, Haixu Jiang, Mengmeng Zhu, Guangrui Huang, and Anlong Xu. 2019. "Andrographolide Ameliorates Rheumatoid Arthritis by Regulating the Apoptosis–NETosis Balance of Neutrophils" International Journal of Molecular Sciences 20, no. 20: 5035. https://doi.org/10.3390/ijms20205035

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop