Κυριακή 18 Οκτωβρίου 2020

A synergistic effect of Ambroxol and Beta-Glucosylceramide in alleviating immune-mediated hepatitis: A novel immunomodulatory non-immunosuppressive formulation for treatment of immune-mediated disorders



A synergistic effect of Ambroxol and Beta-Glucosylceramide in alleviating immune-mediated hepatitis: A novel immunomodulatory non-immunosuppressive formulation for treatment of immune-mediated disorders:



Publication date: December 2020

Source: Biomedicine & Pharmacotherapy, Volume 132

Author(s): Tawfik Khoury, Yuval Ishay, Devorah Rotnemer-Golinkin, Lidya Zolotarovya, David Arkadir, Ari Zimran, Yaron Ilan




Figures (5)








Biomedicine & Pharmacotherapy
Volume 132, December 2020, 110890

A synergistic effect of Ambroxol and Beta-Glucosylceramide in alleviating immune-mediated hepatitis: A novel immunomodulatory non-immunosuppressive formulation for treatment of immune-mediated disorders
Author links open overlay panelTawfikKhouryaYuvalIshayaDevorahRotnemer-GolinkinaLidyaZolotarovyaaDavidArkadirbAriZimrancYaronIlanaShow more
https://doi.org/10.1016/j.biopha.2020.110890Get rights and content
Under a Creative Commons license
open access


Highlights




Ambroxol hydrochloride is used in respiratory diseases as a broncholytic therapy.•

Beta-Glucosylceramide (GC) is a glycosphingolipid that exerts an immune protective effect.•

Coadministration of Ambroxol and GC significantly alleviated immune-mediated liver damage.•

A synergistic effect on suppression of intrahepatic CD8+CD25+ was shown.•

Synergism was documented for an increase in the CD4/CD8 lymphocyte ratio.



Abstract

Background

Ambroxol hydrochloride is being used in respiratory diseases as a broncholytic therapy. Beta-Glucosylceramide (GC) is a naturally occurring glycosphingolipid that exerts an immune protective effect. The aim of the present study was to determine the synergistic immunomodulatory effect between these two compounds.

Methods

Immune-mediated hepatitis was induced in the mice by administration of Con A. Mice were treated with either Ambroxol or GC alone or with the combination of both. Mice were followed for their effect on the liver injury, cytokine profile, and the immune system.

Results

Coadministration of Ambroxol and GC significantly alleviated the liver injury induced by ConA, as demonstrated by the decreased liver enzymes. The combined treatment had a statistically significant synergistic effect on the suppression of intrahepatic CD8+CD25+, an increase in the CD4/CD8 lymphocyte ratio and in the CD8+ intrahepatic lymphocyte trapping, as well as on change of serum in the IL4 levels. The beneficial effect was associated with the promotion of regulatory T lymphocytes subsets, and with a trend for a pro-inflammatory to an anti-inflammatory cytokine shift.

Conclusions

Coadministration of Ambroxol with GC exerted a synergistic immunoprotective effect in a model of immune-mediated acute liver damage. Considering the high safety profile of both agents, the combination may become a novel immunomodulatory non-immunosuppressive therapeutic agent.

Significance statement

Coadministration of Ambroxol with glucocerebroside exerted a synergistic immunoprotective effect in a model of immune-mediated acute liver damage.

Graphical abstract



Download : Download high-res image (84KB)
Download : Download full-size image


Previous article in issue
Next article in issue

Abbreviations
GC
Beta- Glucosylceramide
ConA
Concanavalin A
PMNLs
polymorphonuclear leucocytes
IL
interleukin
TNF
tumor necrosis factor
DC
dendritic-cells
NKT
natural killer T cells
IFN
interferon
PBS
phosphate-buffered saline
AST
aspartate aminotransferase
ALT
alanine aminotransferase
FACS
Fluorescence-activated cell sorting
TGF
transforming growth factor

Keywords
Ambroxol
immune mediated hepatitis
glucocerebroside

1. Introduction

The Current methods for the treatment of immune-mediated and autoimmune disorders usually involve the use of immunosuppressive agents. These drugs are associated with side effects, many of which are related to a direct suppressive effect on various arms of the immune system [1]. Therefore, there is a need to develop the immunomodulatory agents, which will not exert immunosuppressive effects.

Ambroxol hydrochloride is being used in several countries as an over-the-counter bronchosecretolytic and expectorant drug [2,3]. In clinical practice, it has been proved to facilitate the repair of bronchial epithelium and to accelerate mucociliary transport [2,3]. Ambroxol stimulates the formation and release of surfactant by type II pneumocytes [4] and is an effective inhibitor of free radical-mediated processes in the lungs [5]. Ambroxol protects α1-proteinase inhibitor from oxidative inactivation and inhibits the chemotactic response and spontaneous migration of human polymorphonuclear leucocytes (PMNLs). It inhibits the interleukin-1 (IL-1) and tumor necrosis factor alpha (TNFα) production by mononuclear cells stimulated with lipopolysaccharide (LPS) [6]. Intraperitoneal administration of Ambroxol protects the lung and heart lipids from oxidative stress provoked by intravenous injection of endotoxin in mice [7]. Ambroxol inhibits cigarette smoke-induced acute lung injury in a by inhibiting the Erk pathway, TNF-alpha, and IL-1beta [8]. Ambroxol prevents airway inflammation through alteration of cytokines and protection from oxidative stress. Levels of IL-5 and IL-13 in BAL fluid were decreased, along with increased levels of IL-10 and IL-12 [9].

Beta- glucosylceramide (GC) has been previously shown to exert a beneficial effect on a variety of immune-mediated disorders [[10], [11], [12], [13], [14], [15], [16], [17], [18], [19], [20], [21]]. Preliminary data suggest that it exerts similar effects in humans [22]. GC affects cell membranes and the crosstalk between dendritic cells (DCs) and natural killer T (NKT) cells, which underlie some of its beneficial effects [11,[14], [15], [16], [17],19,23,24].

Concanavalin A (ConA), plant lectin and T cell mitogen, rapidly induces severe immune-mediated hepatitis in the mice that is associated with increased pro-inflammatory cytokines, IFN-γ, IL-12, IL-18, and IL-4 expression [25], and in which NKT lymphocytes, CD4 + T cells, and Kupffer cells have a contributory role [26,27].

The aim of our study was to examine the potential to develop Ambroxol with GC-synergistic combination that will have an immunomodulatory role without immunosuppression.
2. Materials and Methods
2.1. Animals

Male C57Bl/6 mice (12 weeks old) were obtained from Harlan Laboratories (Jerusalem, Israel) and maintained in the Animal Core of the Hadassah-Hebrew University Medical School. The Mice were kept at 12 weeks of age and maintained in the Animal Core of the Hebrew University Medical School. All the mice were administered standard laboratory chow and water ad libitum and kept in a 12 -h light/dark cycle. All the experiments were performed in accordance with the guidelines of the Institutional Committee for Care and Use of Laboratory Animals (IACUC protocol number: MD-16-14986-3).
2.2. Oral administration of Ambroxol hydrochloride

Ambroxol was dissolved in distilled water by vigorous shaking. The Ambroxol solution was orally administered to the mice.
2.3. Preparation of Glycolipids

β-glucosylceramide (GC) was purchased from Avanti Polar Lipids (Alabaster, AL, USA). GC was dissolved in a mixture of 30% Cremophor EL (Sigma, Rehovot, Israel) and ethanol (1:1) in PBS.
2.4. Experimental groups

Four groups of mice (n = 4) were studied. Mice in all the groups were injected with Concanavalin A (Con A, 500 mg/mouse). Mice in the control group A were treated with PBS. The Mice in group B were orally administered 6 mg of GC, per mouse daily for 5 days prior to the ConA injection. Mice in group C were orally administered 1.4 mg of Ambroxol per mouse daily for 5 days. Mice in group D were orally administered the combination of 6 mg of GC and 1.4 mg of Ambroxol per mouse daily for 5 days.
2.5. Histological examination of the liver

Livers of all the mice in all the experimental group was cut into 5 μm thin slices, fixed in formaldehyde solution (10%), and kept at room temperature. The Tissue blocks were embedded in paraffin. The sections were stained with hematoxylin-eosin (H&E) for morphological examination.
2.6. Liver enzymes

The serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels were determined with an automatic analyzer on all the mice in all groups.
2.7. Assessment of the effect on the systemic immune system

The immune modulatory effect determined by the FACS analysis and serum cytokines.
2.8. Flow cytometry on isolated splenocytes and hepatic lymphocytes

Spleen and liver-derived lymphocytes were isolated as previously described [28,29]. Flow cytometry was performed on lymphocytes resuspended in 1 mL of the FACS buffer (PBS + 1% BSA + 0.1% sodium azide). Cells were stained with the diluted anti-LAP antibody (50 μL /sample), FITC-conjugated anti-CD4/CD8 (0.5 μL per sample), PE-conjugated anti-CD25/NK1.1 FITC–conjugated anti-CD3 (1 μL per sample), and PerCP-conjugated anti-CD45 (2 μL per sample). All stains were performed after blocking the Fc receptor with anti-mouse CD16/CD32 (BD Fc Block). The flow cytometry was performed using an LSR-II flow cytometer and FCS express software.
2.9. Cytokine measurement

Cytokine assessment was performed on serum samples by the MILLIPLEX® Analytes (EMD Millipore Corporation, Missouri 63304 U.S.A.) based on the Luminex xMAP® technology for performing immunoassays on the surface of the fluorescent-coded magnetic beads MagPlex®-C microspheres. The Acquiring and analyzing the data were performed using the Luminex analyzer (MAGPIX®) software. Cytokines assessment was measured by the mean fluorescence intensity (MFI).
2.10. Statistical analysis

SAS Vs 9.14 was used for statistical analysis. A one-way analysis of variance (ANOVA) model was used to analyze differences between groups. For significant ANOVA models, post-hoc comparisons were performed, using the Tukey-Kramer adjustment for multiple comparisons. Statistical analysis was performed by an experienced statistician.
3. Results
3.1. A synergistic effect of oral administration of Ambroxol and GC on


Fig. 1 shows the effects of the treatment on different subsets of lymphocytes. Panel A1 shows a decrease in the hepatic CD8+CD25+ lymphocytes which were noted only in mice treated with the combination of Ambroxol and GC in group D compared to the untreated mice in group A (6.09% vs. 11.25% for groups D and A, respectively). Panel B shows a significant effect on the redistribution of NK1.1 positive cells, both in the spleen and in the liver of the mice in the treated group. There was a decrease in the splenic to the hepatic ratio of NK1. Cells in group B, C and D as compared to group A (2.2% vs. 0.55% vs. 0.13% vs. 0.38%, for groups A, B, C, and D, respectively).
Download : Download high-res image (201KB)
Download : Download full-size image

Fig. 1. Ambroxol and GC alter the systemic and hepatic immune system. FACS analysis was performed on lymphocytes subsets harvested from both the liver and spleen of mice in all groups for CD8+CD25+ (A); Ratio of spleen/liver NK1.1 (B); the ratio of CD4 to CD8 is the spleen and in the liver (C); ratio of splenic CD4/CD8 to hepatic CD4/CD8 ratio (D); and CD3+FoxP3 (E).

Panel C shows a synergistic effect of the combination of Ambroxol and GC on the CD4/CD8 lymphocyte ratio. The CD4\CD8 ratio in the spleen increased in the combination treated group compared to the other groups. No significant effect was noted for the CD4/CD8 ratio in the liver.

Panel D shows the intrasplenic/ intrahepatic ratio of the CD4/CD8 ratios. This ratio was markedly increased only in the combination treated group D compared to the other groups suggesting the sequestration of CD8+ positive cells in the liver.

Panel E shows an increase in the FoxP3 positive CD3 lymphocytes in all the treated groups compared to group A (1.76% vs. 4.85% vs. 3.015% vs. 4.14 % for groups A, B, C and D respectively).
3.2. Oral administration of Ambroxol with GC was associated with a pro- to an anti-inflammatory cascade shift


Fig. 2A shows that the beneficial effects on the liver damage associated with a pro-inflammatory to anti-inflammatory cytokine shift in the serum. There was a trend for decrease in the level of serum IL-1a in the treated groups as compared to the control group A (206.5 vs. 48 vs. 131 vs. 42 pg/ml for groups A, B, C and D respectively, P = 0.09 for A vs. D). However, the decrease in group B and C compared to group A wasn’t statistically significant (P = 0.11 for group A vs. B and P = 0.63 for group A vs. C).
Download : Download high-res image (200KB)
Download : Download full-size image

Fig. 2. Effects of Ambroxol and GC on serum cytokine profile. Serum cytokines were measured at the end of the study for IL-1a (A); IL-6 (B); IFN-y (C); IL-12 (D); IL-4 (E).

Similarly, Fig. 2B showed a significant decrease of serum IL-6 in the treated group (11768 vs. 984 vs. 9504 vs. 567 pg/ml for groups A, B, C and D respectively, P < 0.0001 for group D vs. A, P = 0.98 for group D vs. B and P < 0.0001 for group D vs. C). However, the decrease in group C compared to group A didn’t reach a statistical significance (P = 0.38).

Fig. 2C showed a trend for decrease in the pro-inflammatory cytokine INF-γ in groups, B and D compared to group A (36 and 19 vs. 643 pg/ml, respectively, P = 0.07 and P = 0.06 for group B and D compared to group A). Similarly, there was a statistically significant decrease in group D compared to C (P = 0.01). However, there was no decrease in INF-g in group C compared to group A (P = 0.99).

Fig. 2D showed the effects on the serum level of IL-12. There was a decrease in all the treated groups as compared to the control group A. The difference was statistically significant for group A vs. B (P = 0.03), however the change between the other groups didn’t reach statistical significance (P = 0.53 for C vs. A, P = 0.34 for group D vs. B and P = 0.91 for group C vs. D).

Fig. 2E showed a trend for increase in the serum level of the anti-inflammatory cytokine IL-4, mainly in group D (32 pg/ml) as compared to group A (22 pg/ml), group B (8.4 pg/ml) and group C (8.7 pg/ml). The difference didn’t reach statistical significance (P = 0.1 for group D vs. B and group D vs. C).

No statistically significant effects were noted in the serum levels of IL10, Il-17, and TGF-β (data were not shown).
3.3. Oral administration of Ambroxol and GC alleviated immune-mediated liver injury


Fig. 3 shows the effect of oral administration of the either the Ambroxol, GC, or a combination of Ambroxol and GC on the immune-mediated liver injury as measured by the effect on liver enzymes (ALT and AST). The oral administration of the combine treatment of Ambroxol and GC was associated with a significant alleviation of the liver injury compared to the untreated controls in group A (24040 vs. 4198 vs. 16455 vs. 2235 IU for ALT levels; and 9140 vs. 2939 vs. 8516 vs. 1470 IU for AST levels, for groups A, B, C, and D, respectively. For ALT, P = 0.005 for group B vs. A, P = 0.003 for group D vs. A, and P = 0.01 for group D vs. C. For AST, P = 0.01 for group B vs. A, P = 0.004 for group D vs. A, and P = 0.001 for group D vs. C.
Download : Download high-res image (55KB)
Download : Download full-size image

Fig. 3. Effect of oral administration of Ambroxol and GC on liver enzymes. There was a significant decrease in liver enzymes (ALT and AST) in all treated groups.

Notably, the decrease in the liver enzymes was more profound in group D compared to group B, however, the difference didn’t reach any statistical significance (P = 0.94 for ALT and P = 0.7 for AST). Moreover, the decrease in group C wasn’t statistically significant compared to group A.


Fig. 4 shows the representative sections from the liver biopsies performed at the end of the treatment period. The Alleviation of the liver apoptosis and improved hepatocyte architecture were noted in the mice in all the treated groups compared to the untreated controls
Download : Download high-res image (1MB)
Download : Download full-size image

Fig. 4. Effects of Ambroxol and GC on liver histology at the end of the study. Hematoxylin Eosin staining was performed on liver sections from treated and control mice. Amelioration of hepatocytes apoptosis coupled with improved liver architecture was noted in treated groups B, C and D compared with control group A.
4. Discussion

The results of the present study show that the co-administration of Ambroxol and GC exert a synergistic effect on the immune system, alleviating the immune-mediated liver damage in a ConA hepatitis model. A significant synergistic effect on the suppression of intrahepatic CD8+CD25+, which increase in the CD8 lymphocyte trapping, and increase in the serum IL4 levels was shown for the Ambroxol with the GC-treated group.

CD8+CD25+ lymphocytes exert immunosuppressive functions in various organs both in the animal models and humans [30,31]. This subset of cells exerts an atheroprotective effect in experimental atherosclerosis [32]. The manipulation of these cells prevented chronic allergic inflammation and improve lung function in allergic asthma exacerbation [33]. The data of the present study support the redistribution of this subset of regulatory lymphocytes, as part of the overall regulatory immunomodulation exerted by the treatment. The increase in the FoxP3 cells further supports the promotion of regulatory lymphocytes in the treated groups.

A synergistic effect of the combination of Ambroxol with GC was noted for the CD4/CD8 lymphocyte ratio. The CD4/CD8 ratio in the spleen increased in the combination-treated group compared with other groups. Interestingly, the intrasplenic/ intrahepatic ratio of the CD4/CD8 ratios was markedly increased only in the combination-treated group D compared to all other groups suggesting a sequestration of CD8+ cells in the liver. Intrahepatic lymphocyte trapping was described as a mechanism for the immune regulation [15,17,[34], [35], [36], [37], [38]]. Antigen presentation by the liver cells controls intrahepatic T cell trapping [39]. In the immune-mediated colitis model, the liver is an important site for CD8+ accumulation during tolerance induction [34]. A similar effect was shown in the models of fatty liver disease and diabetes [40].

A synergistic effect of the combination of Ambroxol and GC was also noted for the increase in the serum IL4 levels. While IL4 was suggested to be associated with the ConA liver damage [41,42], other studies show that its role may be somehow diverse [43]. An altered pro to anti-inflammatory cytokine balance, or different types of effects on the NKT cells which are associated with the IL4 secretion in this model, may partially explain these differences [16,17,[43], [44], [45]]. An altered distribution of the NK1.1 cells and suppression of pro-inflammatory cytokines, IL1a, IFNγ, IL6, and IL12 was shown in the treated groups, further supporting an anti-inflammatory cytokine shift. These effects on the immune system were associated with marked alleviation of the liver damage demonstrated by a decrease in the liver enzymes and amelioration of the histological liver damage.

Ambroxol hydrochloride has been extensively investigated in pulmonary diseases and is mostly prescribed as a mucolytic therapy. In an LPS-mediated acute lung injury model, Ambroxol alleviated lung inflammation and decreased IL-6, TNF-α, and TGF-β pro-inflammatory cytokines secretion [46]. Ambroxol was shown to exert an anti-oxidative and anti-inflammatory effect in vitro and in vivo [[47], [48], [49]], and an organ preservation effect in a transplantation model [50].

Glycosphingolipids are being explored as secondary messengers in various systems [21,24,51]. Their protective immune effect was described both in animal models and humans [13,15,18,20,23,45,[52], [53], [54]]. GC serves as a potent immune adjuvant in the gut for oral immunotherapy [[55], [56], [57], [58]]. Part of the beneficial effect of the GC is associated with the alteration of the NKT-mediated immune balance, and by the alteration of the NKT-dendritic cells cross talk [13,19,23,44]. The patients with Gaucher disease have high levels of GC. Both the clinical and laboratory data suggested a potential beneficial effect for GC in these [59].

Recently the combination of Ambroxol with GC was suggested as a possible mode of therapy for the patients with Parkinson's disease [60]. The patients with non-neuronopathic Gaucher disease and heterozygous GBA mutation carrier are at increased risk for Parkinson disease [61,62]. However, the risk of Parkinson does not increase with accumulation of GC, and is not higher in heterozygous GBA mutation carrier relative to Gaucher patients with two mutated alleles [63]. This observation suggests that the risk for PD is not correlated with the acid beta-glucocerebrosidase (GCase) activity. It further supports that extracellular GC may have beneficial anti-inflammatory properties. Based on these, administration of GC to GBA carriers at risk for PD was suggested as a means to slow inflammatory-driven secondary neuronal death.

Ambroxol is a pharmacological chaperone which reduces endoplasmic reticulum stress induced by the accumulation of mutant misfold GCase. It may prevent the primary pathologic process that leads to cell death [9,64]. GC may act synergistically with Ambroxol in GBA carriers [60]. The present study further supports a synergism of GC and Ambroxol.

Immunosuppression is a major obstacle for long-term use of most immunomodulatory agents including biologicals [65]. Both the Amobroxol and GC have a high safety profile. The data of the present study suggest that their co-administration may provide a mean for a safe immunomodulation which does not induce immunosuppression. Further studies are required to test the effect of therapy on functional assays on CD8+ and CD4 + T cells, as well to elaborate on the potential effect of therapy on macrophages and dendritic cells and on liver NK1.1 and CD4+Foxp3 + .

In summary, the co-administration of the Ambroxol with GC exerted a synergistic immunoprotective effect in a model of the immune-mediated acute liver damage. Bearing in mind the high safety profile of both agents, this combination may become a novel immunomodulatory non-immunosuppressive therapeutic platform.
Authorship Contributions

Tawfik Khoury, Devorah Rotnemer-Golinkin, Lidya Zolotarovya, Yuval Ishay: conducted the studies, and analyzed the data.

David Arkadir, Ari Zimran, Yaron Ilan: Designed the study and wrote the manuscript.

All authors reviewed the final version.
Declaration of Competing Interest

The authors report no declarations of interest.
Acknowledgments

NA.

References[1]
C. Burkhart, C. Heusser, R.E. Morris, F. Raulf, G. Weckbecker, G. Weitz-Schmidt, K. WelzenbachPharmacodynamics in the development of new immunosuppressive drugs
Ther Drug Monit, 26 (2004), pp. 588-592
CrossRefView Record in ScopusGoogle Scholar[2]
M. Rojpibulstit, S. Kasiwong, S. Juthong, N. Phadoongsombat, D. FaroongsarngAmbroxol lozenge bioavailability : an open-label, two-way crossover study of the comparative bioavailability of ambroxol lozenges and commercial tablets in healthy thai volunteers
Clin Drug Investig, 23 (2003), pp. 273-280
CrossRefView Record in ScopusGoogle Scholar[3]
M. Qi, P. Wang, R. Cong, J. YangSimultaneous determination of roxithromycin and ambroxol hydrochloride in a new tablet formulation by liquid chromatography
J Pharm Biomed Anal, 35 (2004), pp. 1287-1291
ArticleDownload PDFView Record in ScopusGoogle Scholar[4]
P. Cerutti, Y. KapanciEffects of metabolite VIII of bromexine (Na 872) on type II epithelium of the lung: an experimental and morphological study with reference to surfactant secretion
Respiration, 37 (1979), pp. 241-251
CrossRefView Record in ScopusGoogle Scholar[5]
K. WinselThe antioxidative and inflammation inhibiting properties of ambroxol
Pneumologie, 46 (1992), pp. 461-475
View Record in ScopusGoogle Scholar[6]
M. Bianchi, A. Mantovani, A. Erroi, C.A. Dinarello, P. GhezziAmbroxol inhibits interleukin 1 and tumor necrosis factor production in human mononuclear cells
Agents Actions, 31 (1990), pp. 275-279
View Record in ScopusGoogle Scholar[7]
D. Nowak, T. Pietras, A. Antczak, M. Krol, G. PiaseckaAmbroxol inhibits endotoxin-induced lipid peroxidation in mice
Pol J Pharmacol, 45 (1993), pp. 317-322
View Record in ScopusGoogle Scholar[8]
L.T. Ge, Y.N. Liu, X.X. Lin, H.J. Shen, Y.L. Jia, X.W. Dong, Y. Sun, Q.M. XieInhalation of ambroxol inhibits cigarette smoke-induced acute lung injury in a mouse model by inhibiting the Erk pathway
International immunopharmacology, 33 (2016), pp. 90-98
ArticleDownload PDFView Record in ScopusGoogle Scholar[9]
K. Takeda, N. Miyahara, S. Matsubara, C. Taube, K. Kitamura, A. Hirano, M. Tanimoto, E.W. GelfandImmunomodulatory Effects of Ambroxol on Airway Hyperresponsiveness and Inflammation
Immune network, 16 (2016), pp. 165-175
CrossRefView Record in ScopusGoogle Scholar[10]
M. Margalit, Z. Shalev, O. Pappo, M. Sklair-Levy, R. Alper, M. Gomori, D. Engelhardt, E. Rabbani, Y. IlanGlucocerebroside ameliorates the metabolic syndrome in OB/OB mice
The Journal of pharmacology and experimental therapeutics, 319 (2006), pp. 105-110
CrossRefView Record in ScopusGoogle Scholar[11]
M. El Haj, A. Ben Ya’acov, G. Lalazar, Y. IlanPotential role of NKT regulatory cell ligands for the treatment of immune mediated colitis
World J Gastroenterol, 13 (2007), pp. 5799-5804
CrossRefGoogle Scholar[12]
Y. Ilan, M. Ohana, O. Pappo, M. Margalit, G. Lalazar, D. Engelhardt, E. Rabbani, A. NaglerAlleviation of acute and chronic graft-versus-host disease in a murine model is associated with glucocerebroside-enhanced natural killer T lymphocyte plasticity
Transplantation, 83 (2007), pp. 458-467
CrossRefView Record in ScopusGoogle Scholar[13]
E. Zigmond, S. Preston, O. Pappo, G. Lalazar, M. Margalit, Z. Shalev, L. Zolotarov, D. Friedman, R. Alper, Y. IlanBeta-glucosylceramide: a novel method for enhancement of natural killer T lymphoycte plasticity in murine models of immune-mediated disorders
Gut, 56 (2007), pp. 82-89
CrossRefView Record in ScopusGoogle Scholar[14]
T. Adar, Y. IlanBeta-Glycosphingolipids as immune modulators
J Immunotoxicol, 5 (2008), pp. 209-220
CrossRefView Record in ScopusGoogle Scholar[15]
G. Lalazar, A. Ben Ya’acov, N. Eliakim-Raz, D.M. Livovsky, O. Pappo, S. Preston, L. Zolotarov, Y. IlanBeta-glycosphingolipids-mediated lipid raft alteration is associated with redistribution of NKT cells and increased intrahepatic CD8+ T lymphocyte trapping
Journal of lipid research, 49 (2008), pp. 1884-1893
CrossRefView Record in ScopusGoogle Scholar[16]
G. Lalazar, A. Ben Ya’acov, A. Lador, D.M. Livovsky, O. Pappo, S. Preston, M. Hareati, Y. IlanModulation of intracellular machinery by beta-glycolipids is associated with alteration of NKT lipid rafts and amelioration of concanavalin-induced hepatitis
Mol Immunol, 45 (2008), pp. 3517-3525
ArticleDownload PDFView Record in ScopusGoogle Scholar[17]
D.M. Livovsky, G. Lalazar, A. Ben Ya’acov, O. Pappo, S. Preston, L. Zolotaryova, Y. IlanAdministration of beta-glycolipids overcomes an unfavorable nutritional dependent host milieu: a role for a soy-free diet and natural ligands in intrahepatic CD8+ lymphocyte trapping and NKT cell redistribution
International immunopharmacology, 8 (2008), pp. 1298-1305
ArticleDownload PDFView Record in ScopusGoogle Scholar[18]
M. Mizrahi, G. Lalazar, A. Ben Ya’acov, D.M. Livovsky, Y. Horowitz, L. Zolotarov, R. Adler, D. Shouval, Y. IlanBeta-glycoglycosphingolipid-induced augmentation of the anti-HBV immune response is associated with altered CD8 and NKT lymphocyte distribution: a novel adjuvant for HBV vaccination
Vaccine, 26 (2008), pp. 2589-2595
ArticleDownload PDFView Record in ScopusGoogle Scholar[19]
Y. IlanAlpha versus beta: are we on the way to resolve the mystery as to which is the endogenous ligand for natural killer T cells?
Clinical and experimental immunology, 158 (2009), pp. 300-307
CrossRefView Record in ScopusGoogle Scholar[20]
W. Zhang, Y. Moritoki, K. Tsuneyama, G.X. Yang, Y. Ilan, Z.X. Lian, M.E. GershwinBeta-glucosylceramide ameliorates liver inflammation in murine autoimmune cholangitis
Clin Exp Immunol, 157 (2009), pp. 359-364
CrossRefView Record in ScopusGoogle Scholar[21]
Y. IlanCompounds of the sphingomyelin-ceramide-glycosphingolipid pathways as secondary messenger molecules: new targets for novel therapies for fatty liver disease and insulin resistance
Am J Physiol Gastrointest Liver Physiol, 310 (2016), pp. G1102-1117
CrossRefView Record in ScopusGoogle Scholar[22]
E. Zigmond, G. Lalazar, O. Pappo, S.W. Zangen, M. Levy Sklair, N. Hemed, E. Rabbani, R. Itamar, Y. Ilan, M. MargalitTreatment of non-alcoholic steatohepatitis by B-glucosylceramide: A phase I/II clinical study
Hepatology, 44 (2006), p. 180A
Google Scholar[23]
A. Ben Ya’acov, G. Lalazar, D.M. Livovsky, D. Kanovich, E. Axelrod, S. Preston, G. Schwarzmann, Y. IlanDecreased STAT-1 phosphorylation by a thio analogue of beta-D-glucosylceramide is associated with altered NKT lymphocyte polarization
Molecular immunology, 47 (2009), pp. 526-533
ArticleDownload PDFView Record in ScopusGoogle Scholar[24]
E. Zigmond, O. Tayer-Shifman, G. Lalazar, A. Ben Ya’acov, S. Weksler-Zangen, D. Shasha, M. Sklair-Levy, L. Zolotarov, Z. Shalev, R. Kalman, E. Ziv, I. Raz, Y. IlanBeta-glycosphingolipids ameliorated non-alcoholic steatohepatitis in the Psammomys obesus model
Journal of inflammation research, 7 (2014), pp. 151-158
View Record in ScopusGoogle Scholar[25]
R. Faggioni, J. Jones-Carson, D.A. Reed, C.A. Dinarello, K.R. Feingold, C. Grunfeld, G. FantuzziLeptin-deficient (ob/ob) mice are protected from T cell-mediated hepatotoxicity: role of tumor necrosis factor alpha and IL-18
Proc Natl Acad Sci U S A, 97 (2000), pp. 2367-2372
View Record in ScopusGoogle Scholar[26]
F. Gantner, M. Leist, A.W. Lohse, P.G. Germann, G. TiegsConcanavalin A-induced T-cell-mediated hepatic injury in mice: the role of tumor necrosis factor
Hepatology, 21 (1995), pp. 190-198
ArticleDownload PDFView Record in ScopusGoogle Scholar[27]
J.E. Gumperz, M.B. BrennerCD1-specific T cells in microbial immunity
Curr Opin Immunol, 13 (2001), pp. 471-478
ArticleDownload PDFView Record in ScopusGoogle Scholar[28]
S. Trop, D. Samsonov, I. Gotsman, R. Alper, J. Diment, Y. IlanLiver-associated lymphocytes expressing NK1.1 are essential for oral immune tolerance induction in a murine model
Hepatology (Baltimore, Md), 29 (1999), pp. 746-755
View Record in ScopusGoogle Scholar[29]
M. Falcone, F. Facciotti, N. Ghidoli, P. Monti, S. Olivieri, L. Zaccagnino, E. Bonifacio, G. Casorati, F. Sanvito, N. SarvetnickUp-regulation of CD1d expression restores the immunoregulatory function of NKT cells and prevents autoimmune diabetes in nonobese diabetic mice
J Immunol, 172 (2004), pp. 5908-5916
CrossRefView Record in ScopusGoogle Scholar[30]
G. Churlaud, F. Pitoiset, F. Jebbawi, R. Lorenzon, B. Bellier, M. Rosenzwajg, D. KlatzmannHuman and Mouse CD8(+)CD25(+)FOXP3(+) Regulatory T Cells at Steady State and during Interleukin-2 Therapy
Front Immunol, 6 (2015), p. 171
Google Scholar[31]
M. Eusebio, P. Kuna, L. Kraszula, M. Kupczyk, M. PietruczukThe relative values of CD8+CD25+Foxp3brigh Treg cells correlate with selected lung function parameters in asthma
Int J Immunopathol Pharmacol, 28 (2015), pp. 218-226
CrossRefView Record in ScopusGoogle Scholar[32]
J. Zhou, P.C. Dimayuga, X. Zhao, J. Yano, W.M. Lio, P. Trinidad, T. Honjo, B. Cercek, P.K. Shah, K.Y. ChyuCD8(+)CD25(+) T cells reduce atherosclerosis in apoE(-/-) mice
Biochemical and biophysical research communications, 443 (2014), pp. 864-870
ArticleDownload PDFView Record in ScopusGoogle Scholar[33]
M. Eusebio, L. Kraszula, M. Kupczyk, P. Kuna, M. PietruczukLow frequency of CD8+CD25+FOXP3(BRIGHT) T cells and FOXP3 mRNA expression in the peripheral blood of allergic asthma patients
J Biol Regul Homeost Agents, 26 (2012), pp. 211-220
View Record in ScopusGoogle Scholar[34]
O. Shibolet, R. Alper, L. Zolotarov, S. Trop, B. Thalenfeld, D. Engelhardt, E. Rabbani, Y. IlanThe role of intrahepatic CD8+ T cell trapping and NK1.1+ cells in liver-mediated immune regulation
Clinical immunology (Orlando, Fla), 111 (2004), pp. 82-92
ArticleDownload PDFView Record in ScopusGoogle Scholar[35]
M. Shuvy, T. Hershcovici, C. Lull-Noguera, H. Wichers, O. Danay, D. Levanon, L. Zolotarov, Y. IlanIntrahepatic CD8(+) lymphocyte trapping during tolerance induction using mushroom derived formulations: a possible role for liver in tolerance induction
World J Gastroenterol, 14 (2008), pp. 3872-3878
CrossRefView Record in ScopusGoogle Scholar[36]
S. Bancos, Q. Cao, W.J. Bowers, I.N. CrispeDysfunctional memory CD8+ T cells after priming in the absence of the cell cycle regulator E2F4
Cell Immunol, 257 (2009), pp. 44-54
ArticleDownload PDFView Record in ScopusGoogle Scholar[37]
I.N. CrispeThe liver as a lymphoid organ
Annual review of immunology, 27 (2009), pp. 147-163
CrossRefView Record in ScopusGoogle Scholar[38]
I.N. CrispeImmune tolerance in liver disease
Hepatology, 60 (2014), pp. 2109-2117
CrossRefView Record in ScopusGoogle Scholar[39]
W.Z. Mehal, F. Azzaroli, I.N. CrispeAntigen presentation by liver cells controls intrahepatic T cell trapping, whereas bone marrow-derived cells preferentially promote intrahepatic T cell apoptosis
J Immunol, 167 (2001), pp. 667-673
CrossRefView Record in ScopusGoogle Scholar[40]
E. Elinav, O. Pappo, M. Sklair-Levy, M. Margalit, O. Shibolet, M. Gomori, R. Alper, B. Thalenfeld, D. Engelhardt, E. Rabbani, Y. IlanAdoptive transfer of regulatory NKT lymphocytes ameliorates non-alcoholic steatohepatitis and glucose intolerance in ob/ob mice and is associated with intrahepatic CD8 trapping
The Journal of pathology, 209 (2006), pp. 121-128
CrossRefView Record in ScopusGoogle Scholar[41]
S. Toyabe, S. Seki, T. Iiai, K. Takeda, K. Shirai, H. Watanabe, H. Hiraide, M. Uchiyama, T. AboRequirement of IL-4 and liver NK1+ T cells for concanavalin A-induced hepatic injury in mice
J Immunol, 159 (1997), pp. 1537-1542
View Record in ScopusGoogle Scholar[42]
T. Nishikage, S. Seki, S. Toyabe, T. Abo, Y. Kagata, T. Iwai, H. HiraideInhibition of concanavalin A-induced hepatic injury of mice by bacterial lipopolysaccharide via the induction of IL-6 and the subsequent reduction of IL-4: the cytokine milieu of concanavalin A hepatitis
Journal of hepatology, 31 (1999), pp. 18-26
ArticleDownload PDFCrossRefView Record in ScopusGoogle Scholar[43]
M. Margalit, S. Abu Gazala, R. Alper, E. Elinav, A. Klein, V. Doviner, Y. Sherman, B. Thalenfeld, D. Engelhardt, E. Rabbani, Y. IlanGlucocerebroside treatment ameliorates ConA hepatitis by inhibition of NKT lymphocytes
Am J Physiol Gastrointest Liver Physiol, 289 (2005), pp. G917-925
CrossRefView Record in ScopusGoogle Scholar[44]
G. Lalazar, S. Preston, E. Zigmond, A. Ben Yaacov, Y. IlanGlycolipids as immune modulatory tools
Mini reviews in medicinal chemistry, 6 (2006), pp. 1249-1253
CrossRefView Record in ScopusGoogle Scholar[45]
E. Zigmond, Z. Shalev, O. Pappo, R. Alper, L. Zolotarov, Y. IlanNKT lymphocyte polarization determined by microenvironment signaling: a role for CD8+ lymphocytes and beta-glycosphingolipids
Journal of autoimmunity, 31 (2008), pp. 188-195
ArticleDownload PDFView Record in ScopusGoogle Scholar[46]
X. Su, L. Wang, Y. Song, C. BaiInhibition of inflammatory responses by ambroxol, a mucolytic agent, in a murine model of acute lung injury induced by lipopolysaccharide
Intensive Care Med, 30 (2004), pp. 133-140
View Record in ScopusGoogle Scholar[47]
V. Stetinova, V. Herout, J. KvetinaIn vitro and in vivo antioxidant activity of ambroxol
Clin Exp Med, 4 (2004), pp. 152-158
CrossRefView Record in ScopusGoogle Scholar[48]
A. Gillissen, A. Bartling, S. Schoen, G. Schultze-WerninghausAntioxidant function of ambroxol in mononuclear and polymorphonuclear cells in vitro
Lung, 175 (1997), pp. 235-242
View Record in ScopusGoogle Scholar[49]
A. Gillissen, B. Scharling, M. Jaworska, A. Bartling, K. Rasche, G. Schultze-WerninghausOxidant scavenger function of ambroxol in vitro: a comparison with N-acetylcysteine
Res Exp Med (Berl), 196 (1997), pp. 389-398
View Record in ScopusGoogle Scholar[50]
G. Drews, A. Tannapfel, H.G. Gnauk, D. Palmes, R. Martin, J. Hauss, H.U. SpiegelProtective effects of ambroxol in hypothermic liver preservation: a transplant study
J Invest Surg, 13 (2000), pp. 197-202
View Record in ScopusGoogle Scholar[51]
A. Ben Ya’acov, G. Lalazar, L. Zolotaryova, Y. Steinhardt, Y. Lichtentein, Y. Ilan, E. ShteyerImpaired liver regeneration by beta-glucosylceramide is associated with decreased fat accumulation
J Dig Dis, 14 (2013), pp. 425-432
CrossRefView Record in ScopusGoogle Scholar[52]
G. Lalazar, A. Ben Ya’acov, D.M. Livovsky, M. El Haj, O. Pappo, S. Preston, L. Zolotarov, Y. IlanBeta-glycoglycosphingolipid-induced alterations of the STAT signaling pathways are dependent on CD1d and the lipid raft protein flotillin-2
The American journal of pathology, 174 (2009), pp. 1390-1399
ArticleDownload PDFCrossRefView Record in ScopusGoogle Scholar[53]
M. Shuvy, A. Ben Ya’acov, L. Zolotarov, C. Lotan, Y. IlanBeta glycosphingolipids suppress rank expression and inhibit natural killer T cell and CD8+ accumulation in alleviating aortic valve calcification
Int J Immunopathol Pharmacol, 22 (2009), pp. 911-918
CrossRefView Record in ScopusGoogle Scholar[54]
E. Zigmond, S.W. Zangen, O. Pappo, M. Sklair-Levy, G. Lalazar, L. Zolotaryova, I. Raz, Y. IlanBeta-glycosphingolipids improve glucose intolerance and hepatic steatosis of the Cohen diabetic rat
Am J Physiol Endocrinol Metab, 296 (2009), pp. E72-78
CrossRefView Record in ScopusGoogle Scholar[55]
Y. Ilan, R. Maron, A.M. Tukpah, T.U. Maioli, G. Murugaiyan, K. Yang, H.Y. Wu, H.L. WeinerInduction of regulatory T cells decreases adipose inflammation and alleviates insulin resistance in ob/ob mice
Proc Natl Acad Sci U S A, 107 (2010), pp. 9765-9770
CrossRefView Record in ScopusGoogle Scholar[56]
Y. Ilan, E. Zigmond, G. Lalazar, A. Dembinsky, A. Ben Ya’acov, N. Hemed, I. Kasis, E. Axelrod, L. Zolotarov, A. Klein, M. El Haj, R. Gandhi, C. Baecher-Allan, H. Wu, G. Murugaiyan, P. Kivisakk, M.F. Farez, F.J. Quintana, S.J. Khoury, H.L. WeinerOral administration of OKT3 monoclonal antibody to human subjects induces a dose-dependent immunologic effect in T cells and dendritic cells
Journal of clinical immunology, 30 (2010), pp. 167-177
CrossRefView Record in ScopusGoogle Scholar[57]
Y. IlanReview article: novel methods for the treatment of non-alcoholic steatohepatitis - targeting the gut immune system to decrease the systemic inflammatory response without immune suppression
Alimentary pharmacology & therapeutics, 44 (2016), pp. 1168-1182
CrossRefView Record in ScopusGoogle Scholar[58]
Y. IlanOral immune therapy: targeting the systemic immune system via the gut immune system for the treatment of inflammatory bowel disease
Clinical & translational immunology, 5 (2016), p. e60
CrossRefGoogle Scholar[59]
Y. Ilan, D. Elstein, A. ZimranGlucocerebroside: an evolutionary advantage for patients with Gaucher disease and a new immunomodulatory agent
Immunology and cell biology, 87 (2009), pp. 514-524
CrossRefView Record in ScopusGoogle Scholar[60]
Y. Ishay, A. Zimran, J. Szer, T. Dinur, Y. Ilan, D. ArkadirCombined beta-glucosylceramide and ambroxol hydrochloride in patients with Gaucher related Parkinson disease: From clinical observations to drug development
Blood Cells Mol Dis. (2016)
Google Scholar[61]
D.G. Hernandez, X. Reed, A.B. SingletonGenetics in Parkinson disease: Mendelian versus non-Mendelian inheritance
Journal of neurochemistry, 139 (Suppl 1) (2016), pp. 59-74
CrossRefView Record in ScopusGoogle Scholar[62]
T. Moors, S. Paciotti, D. Chiasserini, P. Calabresi, L. Parnetti, T. Beccari, W.D. van de BergLysosomal Dysfunction and alpha-Synuclein Aggregation in Parkinson’s Disease: Diagnostic Links
Movement disorders : official journal of the Movement Disorder Society, 31 (2016), pp. 791-801
CrossRefView Record in ScopusGoogle Scholar[63]
R.N. Alcalay, T. Dinur, T. Quinn, K. Sakanaka, O. Levy, C. Waters, S. Fahn, T. Dorovski, W.K. Chung, M. Pauciulo, W. Nichols, H.Q. Rana, M. Balwani, L. Bier, D. Elstein, A. ZimranComparison of Parkinson risk in Ashkenazi Jewish patients with Gaucher disease and GBA heterozygotes
JAMA Neurol, 71 (2014), pp. 752-757
CrossRefView Record in ScopusGoogle Scholar[64]
D.G. Peroni, S. Moser, G. Gallo, R. Pigozzi, L. Tenero, L. Zanoni, A.L. Boner, G.L. PiacentiniAmbroxol inhibits neutrophil respiratory burst activated by alpha chain integrin adhesion
Int J Immunopathol Pharmacol, 26 (2013), pp. 883-887
CrossRefView Record in ScopusGoogle Scholar[65]
A. Mika, P. StepnowskiCurrent methods of the analysis of immunosuppressive agents in clinical materials: A review
Journal of pharmaceutical and biomedical analysis, 127 (2016), pp. 207-231
ArticleDownload PDFView Record in ScopusGoogle ScholarView Abstract
© 2020 Published by Elsevier Masson SAS.

Δεν υπάρχουν σχόλια:

Δημοσίευση σχολίου

Αρχειοθήκη ιστολογίου