Abstract
Autoimmune uveitis (AU) is a sight-threatening ocular inflammatory disorder, characterized by massive retinal vascular leakage and inflamed lesions with infiltration of the uveitogenic T cells in the retina and disorders of the T cell–related immune response in the system. Stimulation of TCRs can trigger calcium release and influx via Ca2+ channels and then transmit signals from the surface to the nucleus, which are important for energy metabolism, proliferation, activation, and differentiation. Inhibition of Ca2+ influx by pharmacological modulation of Ca2+ channels may suppress T cell function, representing a novel anti-inflammatory strategy in the treatment of AU. This study investigated the effects of the l-type voltage-gated calcium channel blocker nimodipine in experimental AU (EAU). Nimodipine was found to not only decrease the clinical and histopathological inflammation score of EAU (C57BL/6J mice) but also dwindle the infiltration of uveitogenic CD4+ T cells into the retina. Moreover, nimodipine decreased the effector T cells and increased the regulatory T cells in the immune system. In vitro, nimodipine reduced the effector T cell differentiation of the IRBP1–20–specific CD4+ T cells of EAU mice and LPS-stimulated PBMCs of uveitis patients. Meanwhile, nimodipine suppressed the energy metabolism, proliferation, activation, and Th1 cell differentiation of T cells. Further studies on RNA sequencing and molecular mechanisms have established that nimodipine alleviates EAU by regulating T cells response through the p38–MAPK pathway signaling. Taken together, our data reveal a novel therapeutic potential of the l-type Ca2+ channels antagonist nimodipine in AU by regulating the balance of T cell subsets.
Visual Abstract
Introduction
Autoimmune uveitis (AU) is one of the sight-threatening ocular inflammatory disorders characterized by massive retinal vascular leakage, inflamed lesions, and macular edema (1, 2). Timely suppression of inflammation and prevention of recurrence is essential to avoid irreversible tissue damage and permanent vision loss. The proliferation, activation, and differentiation of the effector T cells (Teffs) contribute to ocular impairments by secreting inflammatory cytokines such as IL-17A, IFN-γ, and TNF-α (3, 4). Regulation of the T cells is a crucial therapeutic strategy for AU. Calcium is one of the most important second messengers maintaining the physiological and biochemical processes, such as muscle contraction and neurotransmitter release, in many types of cells. Recently, lymphocytes, including T cells, have been demonstrated to express Ca2+ permeable channels and transporters, and the activation of TCRs followed by calcium entry via calcium channels is important for energy metabolism, proliferation, activation, and differentiation of T cells (5). The altered intracellular Ca2+ concentration regulates T cells function and is related to various autoimmune or inflammatory diseases (6). Targeted modulation of Ca2+ channels or Ca2+-conducted TCR-activation signals might have an anti-inflammatory effect.
As an l-type (long-lasting) voltage-activated Ca2+ (CaV) channels antagonist, nimodipine can inhibit contractions of vascular smooth muscle and often can be used to lower blood pressure with favorable clinical tolerability and safety. Studies have reported that nimodipine demonstrates positive effects in CNS diseases of patients and animal models. It has been approved for the prevention of vasospasms in subarachnoid hemorrhage, neuroprotection, and remyelination in multiple sclerosis (7–10). The protective properties of nimodipine are presumed to be associated with the modulation of calcium homeostasis and prevention of intracellular calcium overload. However, the underlying molecular mechanism is still elusive. In vitro studies suggest that l-type calcium channels played a fundamental role in the induction and/or proliferation of reactive astrocytes and the inhibition of microglial activation (11, 12). However, whether nimodipine could regulate lymphocytes, especially T cells remains to be uncovered.
As described above, calcium channels may be implicated in signal transduction of lymphocytes and play a potential role in shaping the immune response. Based on this report, we assessed the potential influence of l-type Ca2+ channels inhibitor nimodipine therapy on autoimmune inflammation in experimental AU (EAU) mice.
Materials and Methods
EAU induction and treatment
Animals
Female wild type C57BL/6J mice (6–8 wk old) were purchased from Guangzhou Animal Testing Center and maintained in specific pathogen–free conditions with a 12 h light–dark cycle with stable temperature (23 ± 2°C) and humidity (55 ± 10%). All animal experiments were performed according to a protocol approved by the Institutional Animal Care and Use Committee of Zhongshan Ophthalmic Center, Sun Yat-sen University, and all procedures were performed in compliance with the Association for Research in Vision and Ophthalmology Statement for the Use of Animals in Ophthalmic and Vision Research.
EAU induction by active immunization
Human IRBP1–20 (GPTHLFQPSLVLDMAKVLLD, Shanghai Shengong, Shanghai, China) was emulsified with CFA containing 5 mg/ml heat-denatured Mycobacterium tuberculosis (Chondrex, Seattle, WA) in a 1:1 volume ratio (v/v). C57BL/6J mice were immunized via s.c. injection of 200 µl of emulsion (IRBP1–20 200 µg/mouse) at the base of the tail and two thighs; 200 ng of pertussis toxin (List Biological Laboratories, Campbell, CA) was injected i.p. in each mouse on days 0 and 2 after immunization.
EAU induction by adoptive transfer experiment
T cells isolated from the draining lymph nodes (DLNs) and spleen of EAU mice (day 14) were stimulated by IRBP1–20 (20 μg/ml) for 72 h. The cells were washed with PBS three times. Then, C57BL/6J mice were injected with IRBP1–20–specific CD4+ T cells (20 million living cells/mice) through the tail vein.
Treatment of EAU mice
Nimodipine (chemical abstract service number [CAS]: 66085-59-4, Selleck Chemicals, Houston, TX) was dissolved in the vehicle solution (2% dimethyl sulfoxide, 5% Tween 80, 10% polyethene glycol, and PBS) and kept in the dark. Mice were injected with nimodipine daily (10 mg/kg), as previously reported, starting on the same day of immunization till sacrifice. The vehicle solution without nimodipine was injected as a control group (vehicle) with the same protocol.
Clinical and histopathologic evaluation of EAU retina inflammation
The photos of the fundus (fundus camera: Phoenix Technology, Campbell, CA) were taken for evaluating retinal inflammation on day 21. Eyes were dissected, fixed, and stained with H&E to evaluate the pathological score. The clinical and pathological score was graded on a scale from 0 to 4 according to the previously described criteria (13).
Cell isolation and treatment
Isolation of DLNs, spleen cells, and intraocular cells
For in vitro assays, the cells were isolated from the DLNs and spleen of C57BL/6J mice on day 21 after immunization. Intraocular infiltrated cells were isolated from the eyes of EAU mice as described previously (14). The ocular tissues were minced and dispersed in RPMI-1640 medium containing collagenase D (1 mg/ml, Roche, Basel, Switzerland) and DNase I (100 µg/ml, Sigma-Aldrich) and incubated for 15 min at 37°C. The samples were then filtered through a 70-µm disposable sieve, washed, and resuspended in a culture medium. Flow cytometry was used for the analysis of naive CD4+ T (CD4+CD62L+CD44−), Th1 (CD4+IFN-γ+), Th17 (CD4+IL-17A+), and regulatory T cells (Tregs) (CD4+CD25+Foxp3+).
T cell recall assays
The T cell recall assays were performed on day 21 after immunization. The cells were seeded into 96-well plates at a density of 2 × 105 cells per well and cultured in complete RPMI-1640 medium (with 10% FBS, 1% penicillin–streptomycin, 1% sodium pyruvate, and 0.1% β-mercaptoethanol) supplemented with 20 µg/ml hIRBP1–20 at 37°C and 5% CO2 and treated with nimodipine (CAS no. 66085-59-4, Selleck Chemicals) at different concentrations (0, 10, 20, 40, or 80 µM). After 72 h, the cells were harvested for intracellular cytokine analysis by flow cytometry. The IFN-γ, IL-17A, and TNF-α concentrations in the cell culture supernatants were determined using ELISA kits (Invitrogen, Carlsbad, CA).
T cell activation and Th1 cell differentiation assay
Naive CD4+ T cells were isolated from DLNs and spleen with EasySepMouse Naive CD4+ T Cell Isolation Kit (STEMCELL Technologies, Vancouver, BC, Canada) and then cultured in 96-well plates at a density of 2 × 105 cells/well adding 1 × 105/well anti-CD3/CD28 beads (Invitrogen), both with and without nimodipine (0, 10, 20, or 40 µM). The cultures were also supplemented with Th1 differentiation condition (ImmunoCult mouse Th1 differentiation supplement, STEMCELL Technologies, Vancouver, BC, Canada) for Th1 conversion. The cells were harvested for flow cytometry assay of CD69 expression after 6 h culture and intracellular cytokine of Th1 cells after 4 d, respectively.
T cell proliferation assay
The cells were first labeled with CFSE (BioLegend, San Diego, CA), and then the CD3+ T cells were obtained using Dynabeads Untouched Mouse T Cells Kit (Invitrogen). The labeled CD3+ T cells were cultured in 96-well plates at a density of 2 × 105 cells per well for 4 d, adding 1 × 105/well anti-CD3/CD28 beads both with and without nimodipine (0, 10, 20, or 40 µM).
Reactive oxygen species production assay of T cell
The identification of reactive oxygen species (ROS) by using oxidized 2-7-dichlorodihydro-fluoresceindiacetate by flow cytometry was performed as previously reported (15). The cells were harvested and incubated with 2-7-dichlorodihydro-fluoresceindiacetate for 20–30 min. The cells were washed with PBS three times before the assay. The accumulation of dichlorofluorescein in cells was measured by flow cytometry.
Isolation of PBMCs and cytokine production assay
The peripheral venous blood (10 ml) of five patients with active Behcet uveitis were collected into a heparinized tube. The PBMCs were isolated immediately by density gradient centrifugation (Ficoll-Hypaque; Pharmacia Biotech, Shanghai, China). Written informed consent was collected from each patient, the study was performed in accordance with the tenets of the Declaration of Helsinki, and institutional review board approval was obtained. Fresh PBMCs (2 × 105 cells per well) were added to 96-well plates and incubated with LPS (100 ng/ml) and nimodipine at concentrations of 0, 20, 40, and 80 µM for 72 h. The inflammatory cells were analyzed by flow cytometry.
RNA sequencing
The CD3+ T cells were isolated from DLNs of EAU mice both with and without nimodipine treatment (n = 3, each group). The total RNAs were isolated with Direct-zol RNA MicroPrep kits (Zymo Research, R2062) processed. The total RNA was processed by the Yale Center for Genome Analysis using the Ribo-Zero rRNA Removal Kit, and the libraries were constructed and subjected to standard Illumina HiSeq2000 sequencing and obtained >40 million reads for each sample.
Differentially expressed genes analysis
The transcripts were analyzed by R (4.0.3). The R package limma was employed to identify the differentially expressed genes (DEGs) with a fold change >2 and p value <0.05. The statistical significance was determined following the Hochberg–Benjamini method.
Gene ontology and Kyoto Encyclopedia of Genes and Genomes analyses
The genes up- or downregulated were assigned biological functions according to the Database for Annotation, Visualization, and Integrated Discovery. The enrichment score >1.0 and p value <0.05 were set for the identification of gene ontology (GO) terms. Kyoto Encyclopedia of Genes and Genomes (KEGG) was applied for pathway enrichment analyses, and p < 0.05 was set as the cutoff value.
Gene set enrichment
The gene set variation analyses (GSVA) were performed for the analysis of the gene set enrichment, and the cutoff value was set at enrichment score change >1.0 and p value <0.05. The R package GSVA was employed for GSVA.
Flow cytometry
The cells were incubated with Fc block (clone 2.4G2, Bio X Cell) and stained with the following Abs from BioLegend: anti-CD45 (BV510), anti-CD3 (BV421), anti-CD4 (Percp-cy5.5), anti-CD25 (PE-cy7), anti-CD44 (allophycocyanin), anti-CD62L (FITC), and anti-CD69 (PE). For intracellular cytokine staining, the cells were pulsed with PMA (50 ng/ml; Sigma), ionomycin (500 ng/ml; Sigma), and brefeldin A (1 µg/ml; Sigma) for 4 h. After further fixation and permeabilization, the intracellular cytokine staining for IFN-γ (BV786), IL-17A (BV650), and Foxp3 (FITC) was performed. For p-p38 (PE) staining, the cells were fixed with 4% paraformaldehyde and permeabilized with methanol at −20°C. The stained cells were collected on an LSR Fortessa (BD Biosciences). Data were analyzed using FlowJo software 10.0 (Tree Star, Ashland, OR).
Gene expression by real-time PCR
The retinal tissues were isolated from the EAU treated both with and without nimodipine. The total RNA was isolated using the RNeasy Mini Kit (QIAGEN, Valencia, CA), and cDNA was synthesized using the PrimeScript RT Master Mix (Perfect Real Time, Takara Bio, Shiga, Japan). Real-time quantitative PCR was performed using SYBR Premix Ex Taq II (Takara Bio), according to the manufacturer’s instructions. The relative expression levels of IL-17A and IFN-γ were analyzed by the ΔΔCT method.
Statistical analysis
The Student t test (for parametric data) or Mann–Whitney U test (for nonparametric data) was used for the two-group comparisons. A p value < 0.05 was considered statistically significant. Data are displayed as mean ± SD. The experiments were repeated no fewer than three times.
Results
Nimodipine attenuates retinal inflammation in EAU
To determine the therapeutic effect of nimodipine on EAU, nimodipine (10 mg/kg) or vehicle were i.p. injected into the mice daily following immunization. No abnormality such as weight loss, lethargy, or depilation was observed in any of the mice. On day 21, the fundus was photographed to evaluate the clinical score of retinal inflammation. The EAU mice treated with vehicle developed severe uveitis with higher clinical scores evident as the swollen retina, tortuous and dilated vessels, and patchy inflammatory infiltration. The EAU mice treated with nimodipine exhibited an almost normal retina (Fig. 1A). The eyeball sections were prepared for pathological evaluation. Histopathological manifestations were consistent with clinical changes, and nimodipine treatment decreased the retina folds, retinal lesions, and inflammatory cell infiltration (Fig. 1B).
Nimodipine attenuates retinal inflammation in EAU mice. (A) Representative fundus photos showed that nimodipine decreased retinal inflammation, characterized by less vasculitis, fewer linear lesions, and minimal confluent tissue destruction of the retinas (n = 10). (B) On day 21, the eyes were enucleated and prepared for histopathology. Nimodipine treatment reduced the inflamed lesions greatly. The histopathologic scores were statistically different (n = 10). (C–E) The intraocular infiltrated cells were isolated and measured by flow cytometry on day 21 after immunization. Nimodipine decreased CD4+ T cell infiltration into the eyes in EAU mice, manifesting as decreased Th17 (IL-17A+CD4+) cells and Th1 (IFN-γ+CD4+) cells (n = 5). (F and G) The mRNA of IL-17A and IFN-γ in retinas was assessed by real-time quantitative PCR on day 21 after immunization. Nimodipine suppressed the mRNA expression of IL-17A and IFN-γ greatly (n = 5). The data are presented as the mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
Nimodipine attenuates retinal inflammation in EAU mice. (A) Representative fundus photos showed that nimodipine decreased retinal inflammation, characterized by less vasculitis, fewer linear lesions, and minimal confluent tissue destruction of the retinas (n = 10). (B) On day 21, the eyes were enucleated and prepared for histopathology. Nimodipine treatment reduced the inflamed lesions greatly. The histopathologic scores were statistically different (n = 10). (C–E) The intraocular infiltrated cells were isolated and measured by flow cytometry on day 21 after immunization. Nimodipine decreased CD4+ T cell infiltration into the eyes in EAU mice, manifesting as decreased Th17 (IL-17A+CD4+) cells and Th1 (IFN-γ+CD4+) cells (n = 5). (F and G) The mRNA of IL-17A and IFN-γ in retinas was assessed by real-time quantitative PCR on day 21 after immunization. Nimodipine suppressed the mRNA expression of IL-17A and IFN-γ greatly (n = 5). The data are presented as the mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
The Teffs, especially Th17 and Th1 cells, contribute to the autoimmune inflammatory damage. Therefore, the effect of nimodipine on the frequency of Teffs in the uveitic eyes was investigated. Nimodipine prevented the CD4+ T cells infiltration into the eyes of EAU mice and decreased the frequencies of Th17 and Th1 significantly (Fig. 1C–E). In the meantime, the inflammatory genes in the eyes were measured using quantitative PCR. Significantly decreased mRNA expressions of IL-17A and IFN-γ were detected in the nimodipine-treated group compared with the vehicle group (Fig. 1F, 1G). Our results demonstrated that nimodipine protects the EAU mice from retinal inflammation by inhibiting local inflammatory cells infiltration and inflammatory gene expression.
Nimodipine modulates the systemic immune profiles
Naive T cells can be activated and differentiated into Teffs and Tregs under different polarizing conditions. Teffs, including Th1 and Th17 cells, produce proinflammatory cytokines to initiate tissue inflammation and contribute to damage. Tregs, also known as suppressor T cells, can suppress Teffs to maintain tolerance and prevent autoimmune disease. To explore the effect of nimodipine on the systemic immune response, the effect of nimodipine on the frequency of naive T cells, Teffs, and Tregs were investigated in DLNs of EAU. The cells were collected from the EAU mice treated both with and without nimodipine on day 21 after immunization and analyzed by flow cytometry. Compared with the EAU mice treated with vehicle, cells from EAU mice treated with nimodipine exhibited less activation, manifesting as increased naive CD4+ T cells (CD4+CD62+CD44−) and decreased memory CD4+ T cells (CD4+CD62−CD44+) (Fig. 2A). Reduced Th17/Th1 and upregulated Tregs were detected in the nimodipine-treated group (Fig. 2B, 2C). These results indicated that nimodipine suppresses the systemic immunological response by downregulating the CD4+ T activation, decreasing Th1 and Th17 cells, and potentiating the Treg.
Nimodipine modulates the systemic immune profiles. (A) Representative flow cytometry analysis of the naive and memory CD4+ T cells. Nimodipine inhibited the naive CD4+ T cells (CD44−CD62L+) differentiation into memory CD4+ T cells (CD44+CD62L−). (B) Nimodipine decreased the levels of the Th1 and Th17 cells of the DLNs. (C) The proportion of Tregs in DLNs were increased more significantly in the nimodipine-treated mice than in the vehicle-treated mice. Flow cytometry experiments above were gated on CD4. Data are representative of at least six independent experiments and presented as the mean ± SD. *p < 0.05, **p < 0.01.
Nimodipine modulates the systemic immune profiles. (A) Representative flow cytometry analysis of the naive and memory CD4+ T cells. Nimodipine inhibited the naive CD4+ T cells (CD44−CD62L+) differentiation into memory CD4+ T cells (CD44+CD62L−). (B) Nimodipine decreased the levels of the Th1 and Th17 cells of the DLNs. (C) The proportion of Tregs in DLNs were increased more significantly in the nimodipine-treated mice than in the vehicle-treated mice. Flow cytometry experiments above were gated on CD4. Data are representative of at least six independent experiments and presented as the mean ± SD. *p < 0.05, **p < 0.01.
Nimodipine decreases IRBP1–20–specific Teffs in vitro
To address whether nimodipine could shape IRBP1–20–specific Teff in vitro, we collected lymphocytes from the DLNs of EAU mice and stimulated them with IRBP1–20 in the presence of nimodipine at different concentrations (0, 20, 40, and 80 µM). Three days later, we analyzed the populations of IRBP1–20–stimulated CD4+ T cells by flow cytometry and the secretion of cytokines in the supernatants by ELISA. The flow cytometry results showed that cell viabilities were similar at the concentration of 0, 20, and 40 µM, whereas at 80 µM, nimodipine decreased the viability of the cells greatly (Fig. 3A). Therefore, we chose 20 and 40 µM as the optimal concentrations for in vitro studies.
Nimodipine decreases IRBP1–20–specific immunological response in vitro. The lymphocytes from DLNs of EAU mice were collected and cultured in vitro with IRBP1–20 for 3 d. (A) The percentages of viable cells (zombie−) were similar at concentrations of 0, 20, and 40 μM of nimodipine, whereas 80 μM nimodipine treatment decreased the percentage. (B–D) Nimodipine (20 and 40 μM) suppressed the differentiation of Th17 and Th1 cells in a dose-dependent manner with no effect on TNF-α−secreting cells (n = 7). (E and F) ELISA results showed that nimodipine reduced the releases of the IL-17A and IFN-γ in a dose-dependent manner (n = 7). (G) Expression of TNF-α showed no statistical difference between nimodipine and vehicle groups (n = 7). Flow cytometry experiments above were gated on CD4. The values represent the mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
Nimodipine decreases IRBP1–20–specific immunological response in vitro. The lymphocytes from DLNs of EAU mice were collected and cultured in vitro with IRBP1–20 for 3 d. (A) The percentages of viable cells (zombie−) were similar at concentrations of 0, 20, and 40 μM of nimodipine, whereas 80 μM nimodipine treatment decreased the percentage. (B–D) Nimodipine (20 and 40 μM) suppressed the differentiation of Th17 and Th1 cells in a dose-dependent manner with no effect on TNF-α−secreting cells (n = 7). (E and F) ELISA results showed that nimodipine reduced the releases of the IL-17A and IFN-γ in a dose-dependent manner (n = 7). (G) Expression of TNF-α showed no statistical difference between nimodipine and vehicle groups (n = 7). Flow cytometry experiments above were gated on CD4. The values represent the mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
Compared with the vehicle group, the proportions of Th17 and Th1 cells were notably lowered by nimodipine in a dose-dependent manner (Fig. 3B, 3C). The reduced release of IL-17A and IFN-γ were also detected in the supernatants of nimodipine-treated cells (Fig. 3E, 3F). Although the expression of TNF-α was also reduced by nimodipine in a dose-dependent manner, there was no statistical difference between the nimodipine and vehicle groups (Fig. 3D, 3G). These results demonstrated that nimodipine could suppress IRBP1–20–specific immunological responses, which was confirmed by diminished Teff differentiation and inflammatory cytokine release.
Nimodipine suppresses LPS-stimulated immunological responses in vitro
Nimodipine presented a prominent inhibitory effect on Teff of EAU mice in vivo. To determine the clinical relevance of this finding, we extended the study from mouse cells to human cells. Therefore, we explored whether nimodipine has the potential of shaping human Th17 and Th1 cells, two major immunopathogenic lymphocyte populations in uveitis patients. Five active uveitis patients of Behcet disease (two females and three males) were enrolled in this study with a median age of 32 y old, ranging from 24 to 36. PBMCs from these patients were isolated and stimulated with LPS (100 ng/ml) in the presence of nimodipine at different dosages (0, 20, 40, and 80 µM) for 3 d. The percentages of viable cells were not greatly reduced by nimodipine at concentrations of 20 and 40 µM. However, nimodipine at a concentration of 80 µM increased cell death significantly. Therefore, nimodipine at concentrations of 20 and 40 µM was chosen for the subsequent experiments (Fig. 4A). The expressions of IL-17A, IFN-γ, and TNF-α of CD4+ T cells were measured by flow cytometry. The Th17 and Th1 cells were significantly suppressed by nimodipine at concentrations of 20 and 40 µM (Fig. 4B, 4C). The TNF-α expression was not greatly changed (Fig. 4D). Our results suggested that nimodipine can regulate human T cell pathogenicity, offering a potential choice for clinical therapy in future.
Nimodipine inhibits LPS-stimulated immunological responses in vitro. The PBMCs from uveitis patients were collected and cultured in vitro with LPS (100 ng/ml) for 3 d. (A) The viability of cells was not greatly affected by nimodipine at the concentrations of 0, 10, 20 and 40 μM. However, nimodipine (80 μM) decreased viable cells significantly. (B and C) Nimodipine (20 and 40 μM) decreased the expression of IL-17A and IFN-γ of CD4+ T cells (n = 5). (D) The TNF-α secretion of LPS-stimulated PBMCs was not influenced by nimodipine (n = 5). Flow cytometry experiments above were gated on CD3+CD8−. The values represent the mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
Nimodipine inhibits LPS-stimulated immunological responses in vitro. The PBMCs from uveitis patients were collected and cultured in vitro with LPS (100 ng/ml) for 3 d. (A) The viability of cells was not greatly affected by nimodipine at the concentrations of 0, 10, 20 and 40 μM. However, nimodipine (80 μM) decreased viable cells significantly. (B and C) Nimodipine (20 and 40 μM) decreased the expression of IL-17A and IFN-γ of CD4+ T cells (n = 5). (D) The TNF-α secretion of LPS-stimulated PBMCs was not influenced by nimodipine (n = 5). Flow cytometry experiments above were gated on CD3+CD8−. The values represent the mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
Nimodipine suppresses the energy metabolism, proliferation, activation, and differentiation of T cells in vitro
Recent advances have revealed that the fundamental processes in T cell biology, such as proliferation, TCR-mediated activation, and Th lineage differentiation, are closely linked to changes in the cellular metabolic programs (16). The dynamic regulation of energy metabolism plays an active role in shaping T cell responses. As both calcium and Ca2+ channels are involved in the cellular metabolism and TCR-activated signal (17), we explored whether nimodipine altered the energy metabolism and downstream TCR signal transduction events, such as proliferation, activation, and differentiation of T cells.
The CD3+ T cells labeled with CFSE were cultured with anti-CD3/CD28 beads in the presence of nimodipine (0, 10, 20, and 40 µM) for 4 d. The intracellular fluorescence of dichlorofluorescein and CFSE was detected by flow cytometry, which represented the ROS production (an indicator of cell energy metabolism) and proliferation, respectively. The results showed that the metabolism and proliferation of CD4+ T cells were reduced by nimodipine (20 and 40 μM) (Fig. 5A, 5B).
Nimodipine suppresses the energy metabolism, proliferation, activation, and differentiation of T cells. (A) Energy metabolism was evaluated by ROS production. Nimodipine decreased the ROS production of CD4+ T cells (n = 6). (B) Nimodipine inhibited the proliferation of CD4+ T cells dose dependently (n = 6). (C) Nimodipine (20 and 40 μM) suppressed the early activation of the naive CD4+ T cells characterized by the downregulated CD69 expression (cultured for 6 h) (n = 6). (D) The Th1 polarization characterized by IFN-γ expression was inhibited by nimodipine (20 and 40 μM) with a statistical difference (n = 6). Flow cytometry experiments above were gated on CD4. The values represent the mean ± SD. ****p < 0.0001.
Nimodipine suppresses the energy metabolism, proliferation, activation, and differentiation of T cells. (A) Energy metabolism was evaluated by ROS production. Nimodipine decreased the ROS production of CD4+ T cells (n = 6). (B) Nimodipine inhibited the proliferation of CD4+ T cells dose dependently (n = 6). (C) Nimodipine (20 and 40 μM) suppressed the early activation of the naive CD4+ T cells characterized by the downregulated CD69 expression (cultured for 6 h) (n = 6). (D) The Th1 polarization characterized by IFN-γ expression was inhibited by nimodipine (20 and 40 μM) with a statistical difference (n = 6). Flow cytometry experiments above were gated on CD4. The values represent the mean ± SD. ****p < 0.0001.
Further, the naive CD4+ T cells were isolated from DLNs and spleen and then cultured in a 96-well plate with anti-CD3/CD28 beads under Th1 cell differentiation condition with or without nimodipine. The expression of CD69 was assayed by flow cytometry after 6 h of culture. The upregulation of CD69 provided a quantitative measure of T cell activation and occurred within 6–12 h following anti-TCR stimulation. Treating cells with nimodipine resulted in a 20–30% reduction of CD69 in a dose-dependent manner (Fig. 5C). Meanwhile, the addition of nimodipine to the cultures significantly decreased the frequency of IFN-γ–expressing CD4+ T cells. Nimodipine inhibited the polarization of Th1 cells (Fig. 5D). Overall, nimodipine could regulate T cell biology, including energy metabolism, proliferation, activation, and differentiation.
Nimodipine alleviates EAU through the p38–MAPK pathway signaling
Next, to explore the potential molecular mechanism involved in nimodipine-mediated immunoregulation, we performed the RNA sequencing of the isolated CD3+ T cells from DLNs of EAU mice both with and without nimodipine treatment (n = 3, each group). The data had been deposited in the National Center for Biotechnology Information Gene Expression Omnibus under accession code GSE183165, which can be accessed in https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE183165. We sought to determine the differences that existed in the T cells of EAU treated with nimodipine and those treated with vehicle. Abundant DEGs were detected between the two groups (Fig. 6A). Enrichment analyses including KEGG and GO showed significant differences in the p38–MAPK signaling pathway and T cell immune responses characterized by cells proliferation, activation, differentiation, and cytokines production. Both the p38–MAPK signaling pathway and T cell immune responses were negatively regulated by nimodipine in the EAU mice (Fig. 6B–D).
Nimodipine alleviates EAU through p38–MAPK signaling. (A) DEGs were explored and presented in a heatmap of mRNA abundance. Rows denote RNA expression according to their enrichment in EAU vehicle versus EAU + nimodipine (n = 3). (B) The bubble plot from the KEGG analysis showed a significant difference in the MAPK signaling pathway between the two groups. (C) Circle plot for GO analysis of pathways that significantly enriched in EAU + nimodipine group. The z-score meant the changes in the pathways. The red or blue points referred to the DEGs that contributed to the pathways shown in the right sheet. (D) Heatmap for GSVA analysis that identified the differentially expressed pathways in each sample. (E) The flow cytometry analysis showed that the expression of p-p38 in CD4+ T cells of DLNs was higher in the EAU vehicle group than the nonimmunization group, and nimodipine treatment significantly downregulated the p-p38 expression (n = 6). (F) Lymphocytes from DLNs of EAU mice were stimulated with IRBP1–20 for 3 d in the presence of nimodipine or vehicle. Nimodipine (20 and 40 µM) treatment downregulated the p-p38 expression significantly (n = 6). (G) Nimodipine suppressed the CD69 expression at 6 h, which could be compromised by asiatic acid (20 µM), a p38 activator (n = 6). Flow cytometry experiments above were gated on CD4. The values represent the mean ± SD. **p < 0.01, ****p < 0.0001.
Nimodipine alleviates EAU through p38–MAPK signaling. (A) DEGs were explored and presented in a heatmap of mRNA abundance. Rows denote RNA expression according to their enrichment in EAU vehicle versus EAU + nimodipine (n = 3). (B) The bubble plot from the KEGG analysis showed a significant difference in the MAPK signaling pathway between the two groups. (C) Circle plot for GO analysis of pathways that significantly enriched in EAU + nimodipine group. The z-score meant the changes in the pathways. The red or blue points referred to the DEGs that contributed to the pathways shown in the right sheet. (D) Heatmap for GSVA analysis that identified the differentially expressed pathways in each sample. (E) The flow cytometry analysis showed that the expression of p-p38 in CD4+ T cells of DLNs was higher in the EAU vehicle group than the nonimmunization group, and nimodipine treatment significantly downregulated the p-p38 expression (n = 6). (F) Lymphocytes from DLNs of EAU mice were stimulated with IRBP1–20 for 3 d in the presence of nimodipine or vehicle. Nimodipine (20 and 40 µM) treatment downregulated the p-p38 expression significantly (n = 6). (G) Nimodipine suppressed the CD69 expression at 6 h, which could be compromised by asiatic acid (20 µM), a p38 activator (n = 6). Flow cytometry experiments above were gated on CD4. The values represent the mean ± SD. **p < 0.01, ****p < 0.0001.
To further corroborate whether the p38–MAPK pathway was involved in the mechanism of nimodipine treatment on EAU, the cells were collected from the C57BL/6J mice without immunization and EAU mice both with and without nimodipine at day 21 after immunization and stained for CD4 and p-p38. The flow cytometry results showed that nimodipine treatment significantly downregulated the p38–MAPK signaling by inhibiting the p-p38 expression in the CD4+ T cells (Fig. 6E). The inhibition on p-p38 was also verified in vitro. The lymphocytes from DLNs of EAU mice were stimulated with IRBP1–20 both with and without nimodipine at dosages of 20 and 40 µM for 3-d culture. Nimodipine (20 and 40 µM) treatment downregulated the p38–MAPK signaling in the IRBP1–20–specific CD4+ T cells (Fig. 6F).
In addition, as shown in (Fig. 6G), the flow cytometry results showed that nimodipine suppressed the CD69 expression of the naive CD4+ T cells under Th1 polarization condition at 6 h. To determine whether the p38–MAPK pathway signaling mediated the inhibition of nimodipine in T cell activation, asiatic acid (CAS no. 464-92-6, Selleck Chemicals), a reported p38 activator, was added into the culture system, and the CD69 expression was examined. Results showed that asiatic acid (20 µM) led to nimodipine’s inhibition on CD69. To sum up, nimodipine alleviated EAU by regulating T cells through p38–MAPK pathway signaling.
The p38–MAPK signaling is involved in inhibiting the effect of nimodipine on the pathogenicity of T cells
To further prove the inhibitory capacity of nimodipine on the pathogenicity of IRBP1–20–specific T cells, we performed the adoptive transfer experiment. The spleens and DLNs of EAU mice were isolated to make single cells suspension, which was stimulated by IRBP1–20 (20 μg/ml), separately, with a vehicle, nimodipine (20 µM), asiatic acid (20 µM), and both nimodipine (20 µM) and asiatic acid (20 µM) for 3 d. Equal numbers of IRBP1–20–specific T cells pretreated with different conditions were adoptively transferred to the C57BL/6J mice through the tail vein. The mice were evaluated 2 wk later. The photos of the fundus showed mice receiving IRBP1–20–specific T cells with vehicle, and those with asiatic acid alone developed severe ocular inflammatory features at day 14, including obvious chorioretinal lesions, significant vascular leakage, and vasculitis. However, nimodipine (20 µM)–treated IRBP1–20–specific T cells failed to induce uveitis, and the protective effect was faded by asiatic acid at 20 µM concentration (Fig. 7A, 7B). IRBP1–20–specific T cells pretreated with both nimodipine and asiatic acid induced a retinal inflammation with small retinal folds and slight cell infiltration. The results further revealed that p38–MAPK signaling was involved in the nimodipine-mediated effect on pathogenic T cells in EAU.
The p38–MAPK signaling is involved in the inhibitory effect of nimodipine on the pathogenicity of T cell. (A) The fundus photo showed that IRBP1–20–treated T cells induced uveitis successfully, and T cells pretreated with nimodipine failed to induce uveitis. IRBP1–20–treated T cells in the presence of p38 activator asiatic acid (AA) resulted in severe uveitis, characterized by the prominent swelling of retina, tortuous and dilated vessels, and more patchy inflammatory infiltration. Nimodipine only partially blocked the pathogenicity of T cells caused by AA, manifesting as mild uveitis. Clinical scores of the nimodipine group had a significant difference with vehicle group and nimodipine plus AA group (n = 6). (B) On day 14, the eyes were enucleated and prepared for histopathology. The histopathologic scores were statistically different (n = 6). The representative results are presented. The values represent the mean ± SD. *p < 0.05, ****p < 0.0001.
The p38–MAPK signaling is involved in the inhibitory effect of nimodipine on the pathogenicity of T cell. (A) The fundus photo showed that IRBP1–20–treated T cells induced uveitis successfully, and T cells pretreated with nimodipine failed to induce uveitis. IRBP1–20–treated T cells in the presence of p38 activator asiatic acid (AA) resulted in severe uveitis, characterized by the prominent swelling of retina, tortuous and dilated vessels, and more patchy inflammatory infiltration. Nimodipine only partially blocked the pathogenicity of T cells caused by AA, manifesting as mild uveitis. Clinical scores of the nimodipine group had a significant difference with vehicle group and nimodipine plus AA group (n = 6). (B) On day 14, the eyes were enucleated and prepared for histopathology. The histopathologic scores were statistically different (n = 6). The representative results are presented. The values represent the mean ± SD. *p < 0.05, ****p < 0.0001.
Discussion
In the present research, we explored the effect of nimodipine on EAU, a classic animal model representative of human uveitis. We demonstrated that exogenous nimodipine treatment attenuated the severity of EAU mice. Histopathological analysis showed alleviated vasculitis and reduced inflammatory changes in the eye of nimodipine-treated EAU mice. Moreover, nimodipine was proved to be able to regulate Teff/Treg balance by suppressing the energy metabolism, proliferation, activation, and Th1 cell differentiation of the T cells. Further, the RNA sequencing and molecular mechanism studies established that nimodipine alleviates EAU by regulating T cells through the p38–MAPK pathway signaling.
Autoreactive CD4+ T cells play a major role in the initiation and orchestration of EAU (3, 4). The activated CD4+ T cells migrate from the peripheral circulation and lymphatic organs to the retina, where the cascade of inflammatory reactions is initiated by secreted cytokines and chemokines. To mount effective immune responses, stimulation of the TCRs by specific Ags can trigger calcium release and influx via Ca2+ channels, and then the intracellular messenger molecules, Ca2+ ions, transmit signals from the surface to the nucleus, which activates the transcription factors such as CREB, MEF, and the NF of activated T cell. These transcription factors are important for the cellular metabolism, proliferation, activation, and differentiation of T cells (5, 18, 19). Previous studies presented that the deletion of subunits β3 in mice led to a reduction in Ca2+ channels expression and partial inhibition of activation of the T cells. The β-subunit–deficient CD4+ T cells displayed defects in cytokine production (IL-2, IL-4, and IFN-γ) (20). Based on the above, it can be inferred those drugs contributing to the calcium homeostasis in the regulation of the functions of T cells may have therapeutic potential in autoimmune and inflammatory diseases.
Nimodipine is an antagonist of the l-type Ca2+ channels, which are broadly expressed in the mouse and human cells including T cells. Several decades ago, studies demonstrated the potency of nimodipine in vasodilation and neuroprotection, thereby improving neurologic function because of intracranial hemorrhage and protecting the retina during retinal ischemia–reperfusion (8, 21–23). The majority of studies have focused on subarachnoid hemorrhage and cerebral vasospasm, but few studies exploited the anti-inflammatory properties, especially those of nimodipine, on the T cells. Sanz et al. (24) showed that nimodipine inhibited the Aβ-stimulated IL-1β synthesis and dose-dependent release from the primary microglia in Alzheimer disease. Zamora et al. (25) reported that the production of proinflammatory cytokines such as TNF-α, IL-1β, and TGF-β1 were significantly decreased in the brain by demyelination because of nimodipine treatment. A retrospective clinical study found that nimodipine could effectively reduce inflammatory cytokines in patients of hypertensive cerebral vasospasm compared with amlodipine (9). However, the regulation of T cell biology and the molecular mechanism of anti-inflammatory and immunoregulatory properties of nimodipine has not been well determined. To the best of our knowledge, this is the first study to attempt to explore the possibility of using the Ca2+ channel inhibitor, nimodipine, to alleviate EAU by regulating T cell biology. Because EAU is initiated and orchestrated by autoreactive CD4+ T cells, we focused on the CD4+ T cells in both in vivo and in vitro experiments. The analysis of lymphocytes in lymph nodes of EAU mice showed that nimodipine decreased the Teffs (both Th17 and Th1) differentiation, suppressed the activation of naive CD4+ T cells, and resumed the balance of Teff/Treg both locally and systemically protecting the mice from EAU. Wang et al. (26) reported that mice deficient for CaV3.1 were resistant to the induction of experimental autoimmune encephalomyelitis and showed reduced productions of the GM-CSF by the CNS-infiltrating Th1 and Th17 cells. Similarly, results from our study support these findings that the inhibition of Ca2+ channels by nimodipine could regulate the balance of Teff/Treg and protect the mice from uveitis.
In vitro, nimodipine reduced the IL-17A and IFN-γ expression in IRBP1–20–specific CD4+ T cells of EAU mice. Interestingly, the inhibitory effect of nimodipine on CD4+ T cells was also proved in the LPS-stimulated PBMCs of uveitis patients, which laid a theoretical foundation for future clinical application. The subsequent study revealed that the inhibition of Ca2+ signals by nimodipine was accompanied by the reduced production of ROS of energy metabolism, proliferation, CD69 expression of early activation, and Th1 cell differentiation of T cells. The results were consistent with certain previous studies. The CaV1.4-deficient mice showed a reduction in the levels of mature thymocytes and peripheral CD4+ and CD8+ T cells, suggesting the role of CaV1.4 in promoting T cell survival and expansion (27). However, Ingwersen et al. (28) demonstrated that nimodipine improved the clinical outcome in experimental autoimmune encephalomyelitis but did not alter the lymphocyte subtypes (proportion of CD4+, CD8+, and CD19+ cells), proliferation, and T cell activation markers of lymphocytes in vitro. In their study, the proportion of CD4+ T cells was not altered by nimodipine, but the proportions of CD4+ T cell subsets, including Th1/Th17, Treg, and naive T cells, had not been subdivided and analyzed. As for the activation pattern of lymphocytes, they used cells from immunized mice, which were already mostly activated. Research from Ingwersen et al. (28) demonstrated that nimodipine could not reverse the activated lymphocytes. In our study, we evaluated the effect of nimodipine on naive T cells polarization into activated T cells and proved nimodipine could inhibit the activation. Taken together, we and Ingwersen et al. (28) investigated different aspects of T cells influenced by nimodipine.
Although nimodipine can mitigate EAU, the underlying mechanisms remain to be fully elucidated. Then, we explored the mechanism involved in TCR-activated Ca2+-related signals in the T cells of EAU mice. The RNA sequencing and functional enrichment analysis of T cells hinted that nimodipine shaped the uveitogenic T cells through the p38–MAPK signaling pathway. The family of MAPKs, including p38, ERK, and JNK, regulate a large number of extracellular signals involving cytokine secretion, proliferation, survival, and apoptosis (29). The p38–MAPK signals have been reported to be potentiated in the mast cell–mediated inflammation (30) and the blockade of p38 attenuated IL-1β, TNF-α, and IL-6 in astrocytoma and microglial cells (31, 32). The p38–MAPK interacted with the l-type CaV channels in multiple ways. Liu et al. (33) reported that TGF-β1 regulated l-type CaV channels through a mechanism dependent on MEK, JNK1/2, and p38–MAPK signal pathways in the cortical neurons. The l-type CaV channels upregulated p38–MAPK activation in CXCL12-induced neuronal death (34). Nimodipine can protect the retina through the downregulation of p38–MAPK and caspase-3 expression in a mice retinal ischemia–reperfusion model (21). However, all of the above experiments were conducted using neurocytes. The alteration of p38–MAPK signals of T cells because of nimodipine has not been reported yet.
The p38 activation is tightly regulated by phosphorylation/dephosphorylation events. Thus, we assayed the expression of p-p38 by flow cytometry and found that cells isolated from EAU showed higher p38 phosphorylation compared with the cells from the nonimmunized mice, whereas the nimodipine treatment prevented the p38 from phosphorylation. This effect was also found in the cultured IRBP1–20–specific CD4+ T cells in a dose-dependent manner. Moreover, the inhibition of nimodipine on the CD4+ T cells could be rescued by p38 activator asiatic acid. The adoptive transfer experiments demonstrated partial rescue of nimodipine by asiatic acid in the clinical score of recipient mice, presenting as mild inflammation of retina. The mild inflammatory changes like tortuous and dilated vessels could not be detected in histopathological examination occasionally, which contributed to no significant difference in pathological scores between nimodipine group and nimodipine plus asiatic acid group. To our knowledge, this is the first study to demonstrate that the nimodipine shaped the T cells to immune response and reduced inflammation through the p38–MAPK signaling pathway.
Nimodipine has been proven to dilate the cerebral blood vessels and increase the cerebral blood flow in animals and humans. Preliminary findings reveal its potential benefit for the treatment of a wide range of cerebrovascular disorders and an inhibitory effect on age-related neurocytes degeneration (7, 8, 22). As nimodipine is commonly used and well tolerated clinically, the successful application of nimodipine in the EAU mice may contribute to other autoimmune diseases. Several comorbidities such as hypertension, coronary heart disease, and cerebral disorders often concur in patients with autoimmune diseases, including multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, and uveitis, limiting the choice of medication and compromising the life quality. Thus, drugs like nimodipine that possess both anti-inflammatory and comorbidity alleviation properties may benefit these patients the most. However, for autoimmune diseases and patients without cardiovascular and cerebrovascular disorders, the indication of nimodipine is considered off label at present. Clinicians and patients should collaboratively assess the risks and benefits before initiating treatment. The efficacy and safety of nimodipine in the treatment of these patients needs clinical validation, which calls for randomized controlled trials.
In summary, our study was the first, to our knowledge, to demonstrate the therapeutic effects of nimodipine on experimental uveitis. Nimodipine alleviated the local and systemic inflammation significantly and shaped the uveitogenic T cells through the p38–MAPK signaling pathway. These results enriched our understanding of the pathogenic mechanism of EAU and expanded the potential clinical use of Ca2+ channel inhibitors. It provided a new promising option for the treatment of AU, further promoting the value of nimodipine in the therapy of other autoimmune diseases especially those with cardio–cerebrovascular comorbidities.
Footnotes
This work was supported by grants from the National Science Foundation of China (81870649, Guangzhou, Guangdong, China) and Guangdong Basic and Applied Basic Research Foundation (2019A1515110093).
Y.H. and G.C were responsible for the conception and design of the study, experiments, data collection, data analysis, and manuscript writing. J.H., Zhuang Li, and H.L. provided guide of experiments. Zhuang Li, H.L., and W.S. helped in the experiment design. Y.X., Zuoyi Li, and Y.C. were helpful in manuscript writing. X.C., and D.L. were responsible for conception and design, revision of the manuscript, and final manuscript approval. All authors contributed to the article and approved the submitted version.
The sequences presented in this article have been submitted to National Center for Biotechnology Information Gene Expression Omnibus under GSE183165.
Abbreviations used in this article
- AU
autoimmune uveitis
- CAS, chemical abstract service number; CaV
voltage-activated Ca2+
- DEG
differentially expressed gene
- DLN
draining lymph node
- EAU
experimental AU
- GO
gene ontology
- GSVA
gene set variation analysis
- KEGG
Kyoto Encyclopedia of Genes and Genomes
- ROS
reactive oxygen species
- Teff
effector T cell
- Treg
regulatory T cell
References
Disclosures
The authors have no financial conflicts of interest.