Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cells ; 10(11)2021 11 06.
Article in English | MEDLINE | ID: mdl-34831283

ABSTRACT

Renal tubulointerstitial lesions (TILs), a common pathologic hallmark of chronic kidney disease that evolves to end-stage renal disease, is characterized by progressive inflammation and pronounced fibrosis of the kidney. However, current therapeutic approaches to treat these lesions remain largely ineffectual. Previously, we demonstrated that elevated IL-36α levels in human renal tissue and urine are implicated in impaired renal function, and IL-36 signaling enhances activation of NLRP3 inflammasome in a mouse model of TILs. Recently, we synthesized NSC828779, a salicylanilide derivative (protected by U.S. patents with US 8975255 B2 and US 9162993 B2), which inhibits activation of NF-κB signaling with high immunomodulatory potency and low IC50, and we hypothesized that it would be a potential drug candidate for renal TILs. The current study validated the therapeutic effects of NSC828779 on TILs using a mouse model of unilateral ureteral obstruction (UUO) and relevant cell models, including renal tubular epithelial cells under mechanically induced constant pressure. Treatment with NSC828779 improved renal lesions, as demonstrated by dramatically reduced severity of renal inflammation and fibrosis and decreased urinary cytokine levels in UUO mice. This small molecule specifically inhibits the IL-36α/NLRP3 inflammasome pathway. Based on these results, the beneficial outcome represents synergistic suppression of both the IL-36α-activated MAPK/NLRP3 inflammasome and STAT3- and Smad2/3-dependent fibrogenic signaling. NSC828779 appears justified as a new drug candidate to treat renal progressive inflammation and fibrosis.


Subject(s)
Interleukin-1/metabolism , Nephritis, Interstitial/metabolism , Salicylanilides/pharmacology , Signal Transduction , Animals , Cell Line , Cytokines/urine , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Hydrogen Peroxide , Inflammasomes/metabolism , Lipopolysaccharides , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Molecular Docking Simulation , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephritis, Interstitial/complications , Nephritis, Interstitial/pathology , Nephritis, Interstitial/urine , STAT3 Transcription Factor/metabolism , Ureteral Obstruction/complications
2.
FASEB J ; 35(8): e21785, 2021 08.
Article in English | MEDLINE | ID: mdl-34314075

ABSTRACT

In the present study, acute onset of severe lupus nephritis was successfully treated in mice using a new, benzamide-linked, small molecule that targets immune modulation and the NLRP3 inflammasome. Specifically, 6-(2,4-difluorophenyl)-3-(3-(trifluoromethyl)phenyl)-2H-benzo[e][1,3]oxazine-2,4(3H)-dione (Cf-02) (a) reduced serum levels of IgG anti-dsDNA, IL-1ß, IL-6, and TNF-α, (b) inhibited activation of dendritic cells and differentially regulated T cell functions, and (c) suppressed the NF-κB/NLRP3 inflammasome axis, targeting priming and activating signals of the inflammasome. Moreover, treatment with Cf-02 significantly inhibited secretion of IL-1ß in lipopolysaccharide-stimulated macrophages, but this effect was abolished by autophagy induction. These results recommend Cf-02 as a promising drug candidate for the serious renal conditions associated with systemic lupus erythematosus. Future investigations should examine whether Cf-02 may also be therapeutic in other types of chronic kidney disease involving NLRP3 inflammasome-driven signaling.


Subject(s)
Autophagy/drug effects , Immunologic Factors/pharmacology , Interleukin-1beta/immunology , Lupus Nephritis/drug therapy , NF-kappa B/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Animals , Case-Control Studies , Cells, Cultured , Dendritic Cells , Female , Humans , Macrophages , Mice , Mice, Inbred C57BL , Sjogren's Syndrome
3.
J Pathol ; 253(4): 427-441, 2021 04.
Article in English | MEDLINE | ID: mdl-33373038

ABSTRACT

IgA nephropathy (IgAN), an immune complex-mediated process and the most common primary glomerulonephritis, can progress to end-stage renal disease in up to 40% of patients. Accordingly, a therapeutic strategy targeting a specific molecular pathway is urgently warranted. Aided by structure characterisation and target identification, we predicted that a novel ring-fused 6-(2,4-difluorophenyl)-3-(3-(trifluoromethyl)phenyl)-2H-benzo[e][1,3]oxazine-2,4(3H)-dione (LCC18) targets the NLRP3 inflammasome, which participates in IgAN pathogenesis. We further developed biomarkers for the disease. We used two complementary IgAN models in C57BL/6 mice, involving TEPC-15 hybridoma-derived IgA, and in gddY mice. Moreover, we created specific cell models to validate therapeutic effects of LCC18 on IgAN and to explain its underlying mechanisms. IgAN mice benefited significantly from treatment with LCC18, showing dramatically improved renal function, including greatly reduced proteinuria and renal pathology. Mechanistic studies showed that the mode of action specifically involved: (1) blocking of the MAPKs/COX-2 axis-mediated priming of the NLRP3 inflammasome; (2) inhibition of ASC oligomerisation and NLRP3 inflammasome assembly by inhibiting NLRP3 binding to PKR, NEK7 and ASC; and (3) activation of autophagy. LCC18 exerts therapeutic effects on murine IgAN by differentially regulating NLRP3 inflammasome activation and autophagy induction, suggesting this new compound as a promising drug candidate to treat IgAN. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Benzamides/pharmacology , Glomerulonephritis, IGA/pathology , Immunologic Factors/pharmacology , Inflammasomes/drug effects , Animals , Autophagy/drug effects , Benzamides/chemistry , Disease Models, Animal , Female , Immunologic Factors/chemistry , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
4.
J Cell Mol Med ; 24(23): 13609-13622, 2020 12.
Article in English | MEDLINE | ID: mdl-33135320

ABSTRACT

Tris (dibenzylideneacetone) dipalladium (Tris DBA), a small-molecule palladium complex, can inhibit cell growth and proliferation in pancreatic cancer, lymphocytic leukaemia and multiple myeloma. Given that this compound is particularly active against B-cell malignancies, we have been suggested that it can alleviate immune complexes (ICs)-mediated conditions, especially IgA nephropathy (IgAN). The therapeutic effects of Tris DBA on glomerular cell proliferation and renal inflammation and mechanism of action were examined in a mouse model of IgAN. Treatment of IgAN mice with Tris DBA resulted in markedly improved renal function, albuminuria and renal pathology, including glomerular cell proliferation, neutrophil infiltration, sclerosis and periglomerular inflammation in the renal interstitium, together with (Clin J Am Soc Nephrol. 2011, 6, 1301-1307) reduced mitochondrial ROS generation; (Am J Physiol-Renal Physiol. 2011. 301, F1218-F1230) differentially regulated autophagy and NLRP3 inflammasome; (Clin J Am Soc Nephrol. 2012, 7, 427-436) inhibited phosphorylation of JNK, ERK and p38 MAPK signalling pathways, and priming signal of the NLRP3 inflammasome; and (Free Radic Biol Med. 2013, 61, 285-297) blunted NLRP3 inflammasome activation through SIRT1- and SIRT3-mediated autophagy induction, in renal tissues or cultured macrophages. In conclusion, Tris DBA effectively ameliorated the mouse IgAN model and targeted signalling pathways downstream of ICs-mediated interaction, which is a novel immunomodulatory strategy. Further development of Tris DBA as a therapeutic candidate for IgAN is warranted.


Subject(s)
Autophagy/drug effects , Glomerulonephritis, IGA/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Organometallic Compounds/pharmacology , Signal Transduction/drug effects , Sirtuin 1/metabolism , Sirtuin 3/metabolism , Animals , Autophagy/genetics , Biomarkers , Biopsy , Disease Models, Animal , Glomerulonephritis, IGA/diagnosis , Glomerulonephritis, IGA/drug therapy , Glomerulonephritis, IGA/etiology , Immunohistochemistry , Kidney Function Tests , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Models, Biological , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Sirtuin 1/genetics , Sirtuin 3/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
5.
FASEB J ; 34(10): 13284-13299, 2020 10.
Article in English | MEDLINE | ID: mdl-32813287

ABSTRACT

Using honokiol (HNK), a major anti-inflammatory bioactive compound in Magnolia officinalis, we show a potent therapeutic outcome against an accelerated, severe form of lupus nephritis (ASLN). The latter may follow infectious insults that act as environmental triggers in the patients. In the current study, an ASLN model in NZB/W F1 mice was treated with HNK by daily gavage after onset of the disease. We show that HNK ameliorated the ASLN by improving renal function, albuminuria, and renal pathology, especially reducing cellular crescents, neutrophil influx, fibrinoid necrosis in glomeruli, and glomerulonephritis activity scores. Meanwhile, HNK differentially regulated T cell functions, reduced serum anti-dsDNA autoantibodies, and inhibited NLRP3 inflammasome activation in the mice. The latter involved: (a) suppressed production of reactive oxygen species and NF-κB activation-mediated priming signal of the inflammasome, (b) reduced mitochondrial damage, and (c) enhanced sirtuin 1 (SIRT1)/autophagy axis activation. In conclusion, HNK represents a new drug candidate for acute, severe episodes of LN capable of alleviating renal lesions in ASLN mice by negatively regulating T cell functions and by enhancing SIRT1/autophagy axis-lessened NLRP3 inflammasome activation.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Autophagy , Biphenyl Compounds/therapeutic use , Inflammasomes/metabolism , Lignans/therapeutic use , Lupus Nephritis/drug therapy , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Cells, Cultured , Female , Inflammasomes/drug effects , Kidney/drug effects , Kidney/metabolism , Lupus Nephritis/metabolism , Mice , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism , T-Lymphocytes/drug effects
6.
Kidney Int ; 98(2): 378-390, 2020 08.
Article in English | MEDLINE | ID: mdl-32622527

ABSTRACT

Xenon, an inert anesthetic gas, is increasingly recognized to possess desirable properties including cytoprotective and anti-inflammatory effects. Here we evaluated the effects of xenon on the progression of lupus nephritis (LN) in a mouse model. A two hour exposure of either 70% xenon or 70% nitrogen balanced with oxygen was administered daily for five weeks to female NZB/W F1 mice that had been induced to develop accelerated and severe LN. Xenon treatment improved kidney function and renal histology, and decreased the renal expression of neutrophil chemoattractants, thereby attenuating glomerular neutrophil infiltration. The effects of xenon were mediated primarily by deceasing serum levels of anti-double stranded DNA autoantibody, inhibiting reactive oxygen species production, NF-κB/NLRP3 inflammasome activation, ICAM-1 expression, glomerular deposition of IgG and C3 and apoptosis, in the kidney; and enhancing renal hypoxia inducible factor 1-α expression. Proteomic analysis revealed that the treatment with xenon downregulated renal NLRP3 inflammasome-mediated cellular signaling. Similarly, xenon was effective in improving renal pathology and function in a spontaneous LN model in female NZB/W F1 mice. Thus, xenon may have a therapeutic role in treating LN but further studies are warranted to determine applicability to patients.


Subject(s)
Lupus Nephritis , Animals , Female , Inflammasomes , Kidney , Lupus Nephritis/drug therapy , Mice , Mice, Inbred NZB , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Proteomics , Xenon
7.
J Immunol ; 204(6): 1448-1461, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32060137

ABSTRACT

Tris (dibenzylideneacetone) dipalladium (Tris DBA), a small-molecule palladium complex, has been shown to inhibit cell growth and proliferation in pancreatic cancer, lymphocytic leukemia, and multiple myeloma. In the current study, we examined the therapeutic effects of Tris DBA on glomerular cell proliferation, renal inflammation, and immune cells. Treatment of accelerated and severe lupus nephritis (ASLN) mice with Tris DBA resulted in improved renal function, albuminuria, and pathology, including measurements of glomerular cell proliferation, cellular crescents, neutrophils, fibrinoid necrosis, and tubulointerstitial inflammation in the kidneys as well as scoring for glomerulonephritis activity. The treated ASLN mice also showed significantly decreased glomerular IgG, IgM, and C3 deposits. Furthermore, the compound was able to 1) inhibit bone marrow-derived dendritic cell-mediated T cell functions and reduce serum anti-dsDNA autoantibody levels; 2) differentially regulate autophagy and both the priming and activation signals of the NLRP3 inflammasome; and 3) suppress the phosphorylation of JNK, ERK, and p38 MAPK signaling pathways. Tris DBA improved ASLN in mice through immunoregulation by blunting the MAPK (ERK, JNK)-mediated priming signal of the NLRP3 inflammasome and by regulating the autophagy/NLRP3 inflammasome axis. These results suggest that the pure compound may be a drug candidate for treating the accelerated and deteriorated type of lupus nephritis.


Subject(s)
Inflammasomes/antagonists & inhibitors , Lupus Nephritis/drug therapy , Lymphocyte Activation/drug effects , Organometallic Compounds/pharmacology , T-Lymphocytes, Regulatory/immunology , Animals , Autophagy/drug effects , Cell Communication/drug effects , Cell Communication/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Kidney Glomerulus/immunology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lupus Nephritis/diagnosis , Lupus Nephritis/immunology , Lupus Nephritis/pathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Organometallic Compounds/therapeutic use , Severity of Illness Index , T-Lymphocytes, Regulatory/drug effects
8.
Front Immunol ; 10: 1951, 2019.
Article in English | MEDLINE | ID: mdl-31475012

ABSTRACT

Chinese herbal medicines used in combination have long-term been shown to be mild remedies with "integrated effects." However, our study provides the first demonstration that M1, an active metabolite of ginsenoside, exerted its dramatic therapeutic effects on accelerated and severe lupus nephritis (ASLN) mice, featuring acute renal function impairment, heavy proteinuria, high serum levels of anti-dsDNA, and high-grade, diffuse proliferative renal lesions. In the present study, NZB/WF1 mice were given injections of lipopolysaccharide to induce the ASLN model. M1 (30 mg/kg) was then administered to the mice by gavage daily, and the mice were sacrificed on week 3 and week 5 after the induction of disease. To identify the potential mechanism of action for the pure compound, levels of NLRP3 inflammasome activation in bone marrow-derived dendritic cells (BMDCs), podocytes and macrophages, and antigen-specific T cell activation in BMDCs were determined in addition to mechanistic experiments in vivo. Treatment with M1 dramatically improved renal function, albuminuria and renal lesions and reduced serum levels of anti-dsDNA in the ASLN mice. These beneficial effects with M1 treatment involved the following cellular and molecular mechanistic events: [1] inhibition of NLRP3 inflammasome associated with autophagy induction, [2] modulation of T help cell activation, and [3] induction of regulatory T cell differentiation. M1 improved the ASLN mice by blunting NLRP3 inflammasome activation and differentially regulating T cell functions, and the results support M1 as a new therapeutic candidate for LN patients with a status of abrupt transformation of lower-grade (mesangial) to higher-grade (diffuse proliferative) nephritis.


Subject(s)
Ginsenosides/pharmacology , Inflammasomes/drug effects , Lupus Nephritis/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , T-Lymphocytes/drug effects , Animals , Antibodies, Antinuclear/blood , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Inflammasomes/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/physiology , Lipopolysaccharides , Lupus Nephritis/chemically induced , Lupus Nephritis/metabolism , Lymphocyte Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred NZB , Podocytes/drug effects , Podocytes/metabolism , Signal Transduction/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...