Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1865(10): 158776, 2020 10.
Article in English | MEDLINE | ID: mdl-32738301

ABSTRACT

Palmitoleic acid (POA, 16:1n-7) is a lipokine that has potential nutraceutical use to treat non-alcoholic fatty liver disease. We tested the effects of POA supplementation (daily oral gavage, 300 mg/Kg, 15 days) on murine liver inflammation induced by a high fat diet (HFD, 59% fat, 12 weeks). In HFD-fed mice, POA supplementation reduced serum insulin and improved insulin tolerance compared with oleic acid (OA, 300 mg/Kg). The livers of POA-treated mice exhibited less steatosis and inflammation than those of OA-treated mice with lower inflammatory cytokine levels and reduced toll-like receptor 4 protein content. The anti-inflammatory effects of POA in the liver were accompanied by a reduction in liver macrophages (LM, CD11c+; F4/80+; CD86+), an effect that could be triggered by peroxisome proliferator activated receptor (PPAR)-γ, a lipogenic transcription factor upregulated in livers of POA-treated mice. We also used HFD-fed mice with selective deletion of PPAR-γ in myeloid cells (PPAR-γ KOLyzCre+) to test whether the beneficial anti-inflammatory effects of POA are dependent on macrophages PPAR-γ. POA-mediated improvement of insulin tolerance was tightly dependent on myeloid PPAR-γ, while POA anti-inflammatory actions including the reduction in liver inflammatory cytokines were preserved in mice bearing myeloid cells deficient in PPAR-γ. This overlapped with increased CD206+ (M2a) cells and downregulation of CD86+ and CD11c+ liver macrophages. Moreover, POA supplementation increased hepatic AMPK activity and decreased expression of the fatty acid binding scavenger receptor, CD36. We conclude that POA controls liver inflammation triggered by fat accumulation through induction of M2a macrophages independently of myeloid cell PPAR-γ.


Subject(s)
Fatty Acids, Monounsaturated/pharmacology , Inflammation/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , PPAR gamma/genetics , AMP-Activated Protein Kinase Kinases , Animals , B7-2 Antigen/genetics , CD11c Antigen/genetics , Diet, High-Fat/adverse effects , Fatty Acids, Monounsaturated/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Insulin Resistance/genetics , Lectins, C-Type/genetics , Liver/drug effects , Liver/metabolism , Liver/pathology , Mannose Receptor , Mannose-Binding Lectins/genetics , Mice , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Oleic Acid/metabolism , Oleic Acid/pharmacology , Protein Kinases/genetics , Receptors, Cell Surface/genetics
2.
FASEB J ; 33(11): 11894-11908, 2019 11.
Article in English | MEDLINE | ID: mdl-31366236

ABSTRACT

Butyrate is a short-chain fatty acid derived from the metabolism of indigestible carbohydrates by the gut microbiota. Butyrate contributes to gut homeostasis, but it may also control inflammatory responses and host physiology in other tissues. Butyrate inhibits histone deacetylases, thereby affecting gene transcription, and also signals through the metabolite-sensing G protein receptor (GPR)109a. We produced an mAb to mouse GPR109a and found high expression on podocytes in the kidney. Wild-type and Gpr109a-/- mice were induced to develop nephropathy by a single injection of Adriamycin and treated with sodium butyrate or high butyrate-releasing high-amylose maize starch diet. Butyrate improved proteinuria by preserving podocyte at glomerular basement membrane and attenuated glomerulosclerosis and tissue inflammation. This protective phenotype was associated with increased podocyte-related proteins and a normalized pattern of acetylation and methylation at promoter sites of genes essential for podocyte function. We found that GPR109a is expressed by podocytes, and the use of Gpr109a-/- mice showed that the protective effects of butyrate depended on GPR109a expression. A prebiotic diet that releases high amounts of butyrate also proved highly effective for protection against kidney disease. Butyrate and GPR109a play a role in the pathogenesis of kidney disease and provide one of the important molecular connections between diet, the gut microbiota, and kidney disease.-Felizardo, R. J. F., de Almeida, D. C., Pereira, R. L., Watanabe, I. K. M., Doimo, N. T. S., Ribeiro, W. R., Cenedeze, M. A., Hiyane, M. I., Amano, M. T., Braga, T. T., Ferreira, C. M., Parmigiani, R. B., Andrade-Oliveira, V., Volpini, R. A., Vinolo, M. A. R., Mariño, E., Robert, R., Mackay, C. R., Camara, N. O. S. Gut microbial metabolite butyrate protects against proteinuric kidney disease through epigenetic- and GPR109a-mediated mechanisms.


Subject(s)
Butyrates/pharmacology , Epigenesis, Genetic , Gastrointestinal Microbiome/physiology , Kidney Diseases/prevention & control , Proteinuria/prevention & control , Receptors, G-Protein-Coupled/genetics , Animals , Bacteria/metabolism , Butyrates/metabolism , Cells, Cultured , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Podocytes/drug effects , Podocytes/metabolism , Protective Agents/metabolism , Protective Agents/pharmacology , Receptors, G-Protein-Coupled/metabolism
3.
Oncotarget ; 9(40): 25808-25825, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29899823

ABSTRACT

The antitumor effect of metformin has been demonstrated in several types of cancer; however, the mechanisms involved are incompletely understood. In this study, we showed that metformin acts directly on melanoma cells as well as on the tumor microenvironment, particularly in the context of the immune response. In vitro, metformin induces a complex interplay between apoptosis and autophagy in melanoma cells. The anti-metastatic activity of metformin in vivo was assessed in several mouse models challenged with B16F10 cells. Metformin's activity was, in part, immune system-dependent, whereas its antitumor properties were abrogated in immunodeficient (NSG) mice. Metformin treatment increased the number of lung CD8-effector-memory T and CD4+Foxp3+IL-10+ T cells in B16F10-transplanted mice. It also decreased the levels of Gr-1+CD11b+ and RORγ+ IL17+CD4+ cells in B16F10-injected mice and the anti-metastatic effect was impaired in RAG-1-/- mice challenged with B16F10 cells, suggesting an important role for T cells in the protection induced by metformin. Finally, metformin in combination with the clinical metabolic agents rapamycin and sitagliptin showed a higher antitumor effect. The metformin/sitagliptin combination was effective in a BRAFV600E/PTEN tamoxifen-inducible murine melanoma model. Taken together, these results suggest that metformin has a pronounced effect on melanoma cells, including the induction of a strong protective immune response in the tumor microenvironment, leading to tumor growth control, and the combination with other metabolic agents may increase this effect.

4.
Methods Mol Biol ; 1714: 149-165, 2018.
Article in English | MEDLINE | ID: mdl-29177861

ABSTRACT

Assembly of a relatively large protein aggregate or "speck" formed by the adaptor protein ASC is a common downstream step in the activation of most inflammasomes. This unique feature of ASC allows its visualization by several imaging techniques and constitutes a reliable and feasible readout for inflammasome activation in cells and tissues. We have previously described step-by-step protocols to generate immortalized cell lines stably expressing ASC fused to a fluorescent protein for measuring inflammasome activation by confocal microscopy, and immunofluorescence of endogenous ASC in primary cells. Here, we present two more methods to detect ASC speck formation: (1) Assessment of ASC speck formation by flow cytometry; and (2) Chemical cross-linking of ASC followed by immunoblotting. These methods allow for the discrimination of inflammasome-activated versus non-activated cells, the identification of lineage-specific inflammasome activation in complex cell mixtures, and sorting of inflammasome-activated cells for further analysis.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Cross-Linking Reagents/metabolism , Flow Cytometry/methods , Fluorescent Antibody Technique/methods , HEK293 Cells , Humans , Image Processing, Computer-Assisted/methods , Immunoblotting/methods , Inflammasomes/analysis , Inflammasomes/metabolism , Microscopy, Confocal/methods
5.
Cell Rep ; 20(11): 2678-2692, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28903047

ABSTRACT

Caloric restriction (CR) is the most effective intervention known to enhance lifespan, but its effect on the skin is poorly understood. Here, we show that CR mice display fur coat remodeling associated with an expansion of the hair follicle stem cell (HFSC) pool. We also find that the dermal adipocyte depot (dWAT) is underdeveloped in CR animals. The dermal/vennule annulus vasculature is enlarged, and a vascular endothelial growth factor (VEGF) switch and metabolic reprogramming in both the dermis and the epidermis are observed. When the fur coat is removed, CR mice display increased energy expenditure associated with lean weight loss and locomotion impairment. Our findings indicate that CR promotes extensive skin and fur remodeling. These changes are necessary for thermal homeostasis and metabolic fitness under conditions of limited energy intake, suggesting a potential adaptive mechanism.


Subject(s)
Caloric Restriction , Skin/anatomy & histology , Skin/metabolism , Adipose Tissue, White/anatomy & histology , Animals , Body Temperature Regulation , Body Weight , Dermis/anatomy & histology , Epidermis/anatomy & histology , Hair Follicle/cytology , Hair Follicle/growth & development , Locomotion , Mice , Oxidation-Reduction , Skin/blood supply , Skin/ultrastructure , Stem Cells/cytology , Time Factors
6.
Front Microbiol ; 8: 1732, 2017.
Article in English | MEDLINE | ID: mdl-28959241

ABSTRACT

Asthma is a chronic inflammatory disease that affects more females than males after puberty, and its symptoms and severity in women change during menstruation and menopause. Recently, evidence has demonstrated that interactions among the microbiota, female sex hormones, and immunity are associated with the development of autoimmune diseases. However, no studies have investigated if therapeutic gut microbiota modulation strategies could affect asthma exacerbation during menstruation and menopause. Here we aimed to examine the preventive effects of a probiotic, Bifidobacterium longum 51A, on airway inflammation exacerbation in allergic ovariectomized mice. We first evaluated the gut microbiota composition and diversity in mice 10 days after ovariectomy. Next, we examined whether re-exposure of ovariectomized allergic mice to antigen (ovalbumin) would lead to exacerbation of lung inflammation. Finally, we evaluated the preventive and treatment effect of B. longum 51A on lung inflammation and airway hyperresponsiveness. Our results showed that whereas ovariectomy caused no alterations in the gut microbiota composition and diversity in this animal model, 10 days after ovariectomy, preventive use administration of B. longum 51A, rather than its use after surgery was capable of attenuate the exacerbated lung inflammation and hyperresponsiveness in ovariectomized allergic mice. This prophylactic effect of B. longum 51A involves acetate production, which led to increased fecal acetate levels and, consequently, increased Treg cells in ovariectomized allergic mice.

7.
Front Immunol ; 8: 881, 2017.
Article in English | MEDLINE | ID: mdl-28824619

ABSTRACT

Mesenchymal stromal cells (MSCs) are multipotent cells with abilities to exert immunosuppressive response promoting tissue repair. Studies have shown that MSCs can secrete extracellular vesicles (MVs-MSCs) with similar regulatory functions to the parental cells. Furthermore, strong evidence suggesting that MVs-MSCs can modulate several immune cells (i.e., Th1, Th17, and Foxp3+ T cells). However, their precise effect on macrophages (Mϕs) remains unexplored. We investigated the immunoregulatory effect of MVs-MSCs on activated M1-Mϕs in vitro and in vivo using differentiated bone marrow Mϕs and an acute experimental model of thioglycollate-induced peritonitis, respectively. We observed that MVs-MSCs shared surface molecules with MSCs (CD44, CD105, CD90, CD73) and expressed classical microvesicle markers (Annexin V and CD9). The in vitro treatment with MVs-MSCs exerted a regulatory-like phenotype in M1-Mϕs, which showed higher CD206 level and reduced CCR7 expression. This was associated with decreased levels of inflammatory molecules (IL-1ß, IL-6, nitric oxide) and increased immunoregulatory markers (IL-10 and Arginase) in M1-Mϕs. In addition, we detected that MVs-MSCs promoted the downregulation of inflammatory miRNAs (miR-155 and miR-21), as well as, upregulated its predicted target gene SOCS3 in activated M1-Mϕs. In vivo MVs-MSCs treatment reduced the Mϕs infiltrate in the peritoneal cavity inducing a M2-like regulatory phenotype in peritoneal Mϕs (higher arginase activity and reduced expression of CD86, iNOS, IFN-γ, IL-1ß, TNF-α, IL-1α, and IL-6 molecules). This in vivo immunomodulatory effect of MVs-MSCs on M1-Mϕs was partially associated with the upregulation of CX3CR1 in F4/80+/Ly6C+/CCR2+ Mϕs subsets. In summary, our findings indicate that MVs-MSCs can modulate an internal program in activated Mϕs establishing an alternative regulatory-like phenotype.

8.
Am J Physiol Gastrointest Liver Physiol ; 311(4): G724-G733, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27634010

ABSTRACT

Nonalcoholic steatohepatitis (NASH), especially as part of the metabolic syndrome (MS), is an increasing burden in Western countries. Statins are already used in MS and seem to be beneficial in liver diseases. The aim of this study was to investigate the molecular mechanisms underlying pleiotropic effects on small GTPases of statins in NASH. NASH within MS was induced in 12-wk-old apoE-/- mice after 7 wk of Western diet (NASH mice). Small GTPases were inhibited by activated simvastatin (SMV), NSC23766 (NSC), or Clostridium sordellii lethal toxin (LT) by using subcutaneous osmotic minipumps. Hepatic steatosis, inflammation, and fibrosis were assessed by histology, Western blot, and RT-PCR measurements of cholesterol and hydroxyproline content. SMV treatment significantly decreased hepatic inflammation and fibrosis, but had no significant effect on steatosis and hepatic cholesterol content in NASH. SMV blunted fibrosis due to inhibition of both RhoA/Rho kinase and Ras/ERK pathways. Interestingly, inhibition of RAC1 and Ras (by LT) failed to decrease fibrosis to the same extent. Inhibition of RAC1 (by NSC) showed no significant effect at all. Inhibition of RhoA and Ras downstream signaling by statins is responsible for the beneficial hepatic effects in NASH.


Subject(s)
Cholesterol/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Simvastatin/therapeutic use , ras Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Diet, Western , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liver/metabolism , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction/drug effects , Simvastatin/pharmacology
9.
Mol Cell Biochem ; 421(1-2): 19-28, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27535240

ABSTRACT

Deficient wound healing is a common multifactorial complication in diabetic patients, but the cellular and molecular mechanisms involved are poorly defined. In the present study, we analyzed the effects of hyperglycemia on integrins expression in rat dermal fibroblasts and addressed its role in cell adhesion and migration. Diabetes Mellitus was induced in rats by streptozotocin injection and maintained for 30 days. Primary cultures of dermal fibroblasts from control and diabetic rats were maintained under low glucose (5 mM D-glucose) or high glucose (30 mM D-glucose) for 7 days. Cell adhesion and migration were studied by kymography, transwell, and time-lapse assays, and the expressions of integrin subunits αv and α5 were studied by immunocytochemistry and western blotting. Fibroblasts derived from diabetic rats confirmed a reduced migration speed and delayed spreading compared to fibroblasts derived from control rats. The membrane fraction of diabetic-derived fibroblasts showed a decrease of integrin subunits α5 and αv, which was confirmed by immunocytochemistry assays. A reduction in the pericellular fibronectin matrix was also observed. The exposure of diabetic-derived cells to a higher concentration of exogenous fibronectin improved migration velocity and the expression of αv but did not completely restore their migration capacity. In conclusion, the mechanisms involved in the deleterious effects of Diabetes Mellitus on wound healing include the ability of fibroblasts to secrete and to adhere to fibronectin.


Subject(s)
Cell Movement , Dermis/metabolism , Diabetes Mellitus, Experimental/metabolism , Fibroblasts/metabolism , Hyperglycemia/metabolism , Integrin alphaV/metabolism , Animals , Dermis/pathology , Diabetes Mellitus, Experimental/pathology , Fibroblasts/pathology , Hyperglycemia/chemically induced , Hyperglycemia/pathology , Male , Rats , Rats, Wistar
10.
Cell Rep ; 12(5): 760-73, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26212322

ABSTRACT

The evolutionarily conserved miR-302 family of microRNAs is expressed during early mammalian embryonic development. Here, we report that deletion of miR-302a-d in mice results in a fully penetrant late embryonic lethal phenotype. Knockout embryos have an anterior neural tube closure defect associated with a thickened neuroepithelium. The neuroepithelium shows increased progenitor proliferation, decreased cell death, and precocious neuronal differentiation. mRNA profiling at multiple time points during neurulation uncovers a complex pattern of changing targets over time. Overexpression of one of these targets, Fgf15, in the neuroepithelium of the chick embryo induces precocious neuronal differentiation. Compound mutants between mir-302 and the related mir-290 locus have a synthetic lethal phenotype prior to neurulation. Our results show that mir-302 helps regulate neurulation by suppressing neural progenitor expansion and precocious differentiation. Furthermore, these results uncover redundant roles for mir-290 and mir-302 early in development.


Subject(s)
Cell Differentiation/physiology , Embryo, Mammalian/embryology , MicroRNAs/biosynthesis , Neural Stem Cells/metabolism , Neural Tube/embryology , Animals , Chick Embryo , Embryo, Mammalian/cytology , Mice , MicroRNAs/genetics , Neural Stem Cells/cytology , Neural Tube/cytology
11.
Mol Med ; 21: 553-62, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26101952

ABSTRACT

Natural killer T (NKT) cells are a subset of lymphocytes that reacts to glycolipids presented by CD1d. Invariant NKT cells (iNKT) correspond to >90% of the total population of NKTs and reacts to α-galactosylceramide (αGalCer). αGalCer promotes a complex mixture of Th1 and Th2 cytokines, as interferon (IFN)-γ and interleukin (IL)-4. NKT cells and IFN-γ are known to participate in some models of renal diseases, but further studies are still necessary to elucidate their mechanisms. The aim of our study was to analyze the participation of iNKT cells in an experimental model of tubule-interstitial nephritis. We used 8-wk-old C57BL/6j, Jα18KO and IFN-γKO mice. They were fed a 0.25% adenine diet for 10 d. Both adenine-fed wild-type (WT) and Jα18KO mice exhibited renal dysfunction, but adenine-fed Jα18KO mice presented higher expression of kidney injury molecule-1 (KIM-1), tumor necrosis factor (TNF)-α and type I collagen. To analyze the role of activated iNKT cells in our model, we administered αGalCer in WT mice during adenine ingestion. After αGalCer injection, we observed a significant reduction in serum creatinine, proinflammatory cytokines and renal fibrosis. However, this improvement in renal function was not observed in IFN-γKO mice after αGalCer treatment and adenine feeding, illustrating that this cytokine plays a role in our model. Our findings may suggest that IFN-γ production is one of the factors contributing to improved renal function after αGalCer administration.


Subject(s)
Galactosylceramides/administration & dosage , Interferon-gamma/genetics , Nephritis/drug therapy , Renal Insufficiency/drug therapy , Adenine/toxicity , Animals , Antigens, CD1d/biosynthesis , Antigens, CD1d/genetics , Collagen Type I/biosynthesis , Hepatitis A Virus Cellular Receptor 1 , Humans , Interleukin-4/biosynthesis , Interleukin-4/genetics , Kidney Tubules/drug effects , Kidney Tubules/pathology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Knockout , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Nephritis/chemically induced , Nephritis/genetics , Nephritis/pathology , Renal Insufficiency/chemically induced , Renal Insufficiency/genetics , Renal Insufficiency/pathology , Tumor Necrosis Factor-alpha/biosynthesis
12.
Lab Invest ; 94(4): 455-66, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24492283

ABSTRACT

Platelet-activating factor (PAF) is a lipid mediator with important pro-inflammatory effects, being synthesized by several cell types including kidney cells. Although there is evidence of its involvement in acute renal dysfunction, its role in progressive kidney injury is not completely known. In the present study, we investigated the role of PAF receptor (PAFR) in an experimental model of chronic renal disease. Wild-type (WT) and PAFR knockout (KO) mice underwent unilateral ureter obstruction (UUO), and at kill time, urine and kidney tissue was collected. PAFR KO animals compared with WT mice present: (a) less renal dysfunction, evaluated by urine protein/creatinine ratio; (b) less fibrosis evaluated by collagen deposition, type I collagen, Lysyl Oxidase-1 (LOX-1) and transforming growth factor ß (TGF-ß) gene expression, and higher expression of bone morphogenetic protein 7 (BMP-7) (3.3-fold lower TGF-ß/BMP-7 ratio); (c) downregulation of extracellular matrix (ECM) and adhesion molecule-related machinery genes; and (d) lower levels of pro-inflammatory cytokines. These indicate that PAFR engagement by PAF or PAF-like molecules generated during UUO potentiates renal dysfunction and fibrosis and might promote epithelial-to-mesenchymal transition (EMT). Also, early blockade of PAFR after UUO leads to a protective effect, with less fibrosis deposition. In conclusion, PAFR signaling contributes to a pro-inflammatory environment in the model of obstructive nephropathy, favoring the fibrotic process, which lately will generate renal dysfunction and progressive organ failure.


Subject(s)
Kidney/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Azepines , Collagen/metabolism , Disease Models, Animal , Fibrosis , Kidney/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Nephritis/metabolism , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Renal Insufficiency, Chronic/pathology , Triazoles , Ureteral Obstruction
13.
Kidney Int ; 79(11): 1217-27, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21412216

ABSTRACT

Focal and segmental glomerulosclerosis (FSGS) is one of the most important causes of end-stage renal failure. The bradykinin B1 receptor has been associated with tissue inflammation and renal fibrosis. To test for a role of the bradykinin B1 receptor in podocyte injury, we pharmacologically modulated its activity at different time points in an adriamycin-induced mouse model of FSGS. Estimated albuminuria and urinary protein to creatinine ratios correlated with podocytopathy. Adriamycin injection led to loss of body weight, proteinuria, and upregulation of B1 receptor mRNA. Early treatment with a B1 antagonist reduced albuminuria and glomerulosclerosis, and inhibited the adriamycin-induced downregulation of podocin, nephrin, and α-actinin-4 expression. Moreover, delayed treatment with antagonist also induced podocyte protection. Conversely, a B1 agonist aggravated renal dysfunction and even further suppressed the levels of podocyte-related molecules. Thus, we propose that kinin has a crucial role in the pathogenesis of FSGS operating through bradykinin B1 receptor signaling.


Subject(s)
Bradykinin/analogs & derivatives , Glomerulosclerosis, Focal Segmental/chemically induced , Podocytes/drug effects , Receptor, Bradykinin B1/agonists , Signal Transduction/drug effects , Actinin/metabolism , Albuminuria/chemically induced , Albuminuria/metabolism , Albuminuria/prevention & control , Animals , Bradykinin/pharmacology , Bradykinin/toxicity , Bradykinin B1 Receptor Antagonists , Disease Models, Animal , Doxorubicin , Gene Expression Regulation/drug effects , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/prevention & control , Heme Oxygenase-1/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Podocytes/metabolism , Podocytes/pathology , RNA, Messenger/metabolism , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B1/metabolism , Time Factors , Tissue Inhibitor of Metalloproteinase-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...