Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.101
Filter
1.
Nat Commun ; 12(1): 2141, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33837218

ABSTRACT

Molecular characterization of the individual cell types in human kidney as well as model organisms are critical in defining organ function and understanding translational aspects of biomedical research. Previous studies have uncovered gene expression profiles of several kidney glomerular cell types, however, important cells, including mesangial (MCs) and glomerular parietal epithelial cells (PECs), are missing or incompletely described, and a systematic comparison between mouse and human kidney is lacking. To this end, we use Smart-seq2 to profile 4332 individual glomerulus-associated cells isolated from human living donor renal biopsies and mouse kidney. The analysis reveals genetic programs for all four glomerular cell types (podocytes, glomerular endothelial cells, MCs and PECs) as well as rare glomerulus-associated macula densa cells. Importantly, we detect heterogeneity in glomerulus-associated Pdgfrb-expressing cells, including bona fide intraglomerular MCs with the functionally active phagocytic molecular machinery, as well as a unique mural cell type located in the central stalk region of the glomerulus tuft. Furthermore, we observe remarkable species differences in the individual gene expression profiles of defined glomerular cell types that highlight translational challenges in the field and provide a guide to design translational studies.


Subject(s)
Endothelial Cells/metabolism , Glomerular Mesangium/metabolism , Podocytes/metabolism , Protein Biosynthesis/genetics , Transcriptome/physiology , Animals , Cell Separation , Computational Biology , Flow Cytometry , Genetic Heterogeneity , Glomerular Mesangium/cytology , Humans , Male , Mice , RNA-Seq , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptors, Phospholipase A2/genetics , Single-Cell Analysis , Species Specificity
2.
Biomolecules ; 11(2)2021 02 13.
Article in English | MEDLINE | ID: mdl-33668480

ABSTRACT

Prostaglandin E2 (PGE2) is a key mediator of inflammation, and consequently huge efforts have been devoted to the development of novel agents able to regulate its formation. In this work, we present the synthesis of various α-ketoheterocycles and a study of their ability to inhibit the formation of PGE2 at a cellular level. A series of α-ketobenzothiazoles, α-ketobenzoxazoles, α-ketobenzimidazoles, and α-keto-1,2,4-oxadiazoles were synthesized and chemically characterized. Evaluation of their ability to suppress the generation of PGE2 in interleukin-1ß plus forskolin-stimulated mesangial cells led to the identification of one α-ketobenzothiazole (GK181) and one α-ketobenzoxazole (GK491), which are able to suppress the PGE2 generation at a nanomolar level.


Subject(s)
Dinoprostone/antagonists & inhibitors , Glomerular Mesangium/drug effects , Heterocyclic Compounds/pharmacology , Prostaglandin Antagonists/pharmacology , Animals , Cells, Cultured , Dinoprostone/biosynthesis , Glomerular Mesangium/cytology , Glomerular Mesangium/metabolism , Molecular Docking Simulation , Rats , Spectrum Analysis/methods
3.
Biosci Rep ; 41(2)2021 02 26.
Article in English | MEDLINE | ID: mdl-33565572

ABSTRACT

Ferroptosis, a novel type of programmed cell death, is involved in inflammation and oxidation of various human diseases, including diabetic kidney disease. The present study explored the role of high-mobility group box-1 (HMGB1) on the regulation of ferroptosis in mesangial cells in response to high glucose. Compared with healthy control, levels of serum ferritin, lactate dehydrogenase (LDH), reactive oxygen species (ROS), malonaldehyde (MDA), and HMGB1 were significantly elevated in diabetic nephropathy (DN) patients, accompanied with deregulated ferroptosis-related molecules, including long-chain acyl-CoA synthetase 4 (ACSL4), prostaglandin-endoperoxide synthase 2 (PTGS2), NADPH oxidase 1 (NOX1), and glutathione peroxidase 4 (GPX4). In vitro assay revealed that erastin and high glucose both induced ferroptosis in mesangial cells. Suppression of HMGB1 restored cellular proliferation, prevented ROS and LDH generation, decreased ACSL4, PTGS2, and NOX1, and increased GPX4 levels in mesangial cells. Furthermore, nuclear factor E2-related factor 2 (Nrf2) was decreased in DN patients and high glucose-mediated translocation of HMGB1 in mesangial cells. Knockdown of HMGB1 suppressed high glucose-induced activation of TLR4/NF-κB axis and promoted Nrf2 expression as well as its downstream targets including HO-1, NQO-1, GCLC, and GCLM. Collectively, these findings suggest that HMGB1 regulates glucose-induced ferroptosis via Nrf2 pathway in mesangial cells.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/metabolism , Ferroptosis/physiology , Glomerular Mesangium/metabolism , Glucose/metabolism , HMGB1 Protein/physiology , NF-E2-Related Factor 2/metabolism , Cell Line , Female , Glomerular Mesangium/cytology , Humans , Male , Middle Aged , Oxidative Stress/physiology
4.
Am J Physiol Renal Physiol ; 320(3): F492-F504, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33491562

ABSTRACT

Although macula densa (MD) cells are chief regulatory cells in the nephron with unique microanatomical features, they have been difficult to study in full detail due to their inaccessibility and limitations in earlier microscopy techniques. The present study used a new mouse model with a comprehensive imaging approach to visualize so far unexplored microanatomical features of MD cells, their regulation, and functional relevance. MD-GFP mice with conditional and partial induction of green fluorescent protein (GFP) expression, which specifically and intensely illuminated only single MD cells, were used with fluorescence microscopy of fixed tissue and live MD cells in vitro and in vivo with complementary electron microscopy of the rat, rabbit, and human kidney. An elaborate network of major and minor cell processes, here named maculapodia, were found at the cell base, projecting toward other MD cells and the glomerular vascular pole. The extent of maculapodia showed upregulation by low dietary salt intake and the female sex. Time-lapse imaging of maculapodia revealed highly dynamic features including rapid outgrowth and an extensive vesicular transport system. Electron microscopy of rat, rabbit, and human kidneys and three-dimensional volume reconstruction in optically cleared whole-mount MD-GFP mouse kidneys further confirmed the presence and projections of maculapodia into the extraglomerular mesangium and afferent and efferent arterioles. The newly identified dynamic and secretory features of MD cells suggest the presence of novel functional and molecular pathways of cell-to-cell communication in the juxtaglomerular apparatus between MD cells and between MD and other target cells.NEW & NOTEWORTHY This study illuminated a physiologically regulated dense network of basal cell major and minor processes (maculapodia) in macula densa (MD) cells. The newly identified dynamic and secretory features of these microanatomical structures suggest the presence of novel functional and molecular pathways of cell-to-cell communication in the juxtaglomerular apparatus between MD and other target cells. Detailed characterization of the function and molecular details of MD cell intercellular communications and their role in physiology and disease warrant further studies.


Subject(s)
Glomerular Mesangium/ultrastructure , Juxtaglomerular Apparatus/ultrastructure , Kidney Glomerulus/ultrastructure , Kidney Tubules/ultrastructure , Animals , Cell Communication/physiology , Epithelial Cells/cytology , Epithelial Cells/ultrastructure , Glomerular Mesangium/cytology , Kidney Glomerulus/cytology , Kidney Tubules/cytology , Mice , Rabbits , Rats
5.
Int J Mol Sci ; 20(21)2019 Oct 28.
Article in English | MEDLINE | ID: mdl-31661773

ABSTRACT

The alteration of mesangial matrix (MM) components in mesangium, such as type IV collagen (COL4) and type I collagen (COL1), is commonly found in progressive glomerular disease. Mesangial cells (MCs) responding to altered MM, show critical changes in cell function. This suggests that the diseased MM structure could play an important role in MC behavior. To investigate how MC behavior is influenced by the diseased MM 3D nanostructure, we fabricated the titanium dioxide (TiO2)-based nanopatterns that mimic diseased MM nanostructures. Immortalized mouse MCs were used to assess the influence of disease-mimic nanopatterns on cell functions, and were compared with a normal-mimic nanopattern. The results showed that the disease-mimic nanopattern induced disease-like behavior, including increased proliferation, excessive production of abnormal MM components (COL1 and fibronectin) and decreased normal MM components (COL4 and laminin α1). In contrast, the normal-mimic nanopattern actually resulted in cells displaying normal proliferation and the production of normal MM components. In addition, increased expressions of α-smooth muscle actin (α-SMA), transforming growth factor ß1 (TGF-ß1) and integrin α5ß1 were detected in cells grown on the disease-mimic nanopattern. These results indicated that the disease-mimic nanopattern induced disease-like cell behavior. These findings will help further establish a disease model that mimics abnormal MM nanostructures and also to elucidate the molecular mechanisms underlying glomerular disease.


Subject(s)
Kidney Diseases/metabolism , Kidney Diseases/pathology , Mesangial Cells/cytology , Mesangial Cells/metabolism , Actins/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/metabolism , Collagen Type IV/metabolism , Fibronectins/metabolism , Glomerular Mesangium/cytology , Integrins/metabolism , Laminin/metabolism , Mesangial Cells/pathology , Mesangial Cells/ultrastructure , Mice , Nanostructures/chemistry , Nanostructures/toxicity , Nanostructures/ultrastructure , Titanium/chemistry , Transforming Growth Factor beta/metabolism
6.
Adv Exp Med Biol ; 1165: 165-194, 2019.
Article in English | MEDLINE | ID: mdl-31399966

ABSTRACT

The main cellular constituents in glomerular mesangium are mesangial cells, which account for approximately 30-40% of the total cells in the glomerulus. Together with the mesangial matrix, mesangial cells form the glomerular basement membrane (GBM) in the glomerulus, whose main function is to perform the filtration. Under the pathologic conditions, mesangial cells are activated, leading to hyperproliferation and excess extracellular matrix (ECM). Moreover, mesangial cells also secrete several kinds of inflammatory cytokines, adhesion molecules, chemokines, and enzymes, all of which participate in the process of renal glomerular fibrosis. During the past years, researchers have revealed the roles of mesangial cells and the associated signal pathways involved in renal fibrosis. In this section, we will discuss how mesangial cells are activated and its contributions to renal fibrosis, as well as the molecular mechanisms and novel anti-fibrotic agents. Full understanding of the contributions of mesangial cells to renal fibrosis will benefit the clinical drug developing.


Subject(s)
Glomerular Mesangium/cytology , Kidney Diseases/physiopathology , Mesangial Cells/cytology , Cell Adhesion Molecules , Chemokines , Cytokines , Extracellular Matrix , Fibrosis , Humans , Kidney/pathology , Kidney Glomerulus
7.
Am J Physiol Renal Physiol ; 317(4): F839-F851, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31364374

ABSTRACT

The small GTPase Rho and its effector Rho kinase (ROCK) are involved in the pathogenesis of diabetic kidney disease. Rho kinase has two isoforms: ROCK1 and ROCK2. However, it remains unclear which is mainly involved in the progression of diabetic glomerulosclerosis and the regulation of profibrotic mediators. Glomeruli isolated from type 2 diabetic db/db mice demonstrated increased gene expression of transforming growth factor (TGF)-ß and its downstream profibrotic mediators. Chemical inhibition of ROCK suppressed the expression of profibrotic mediators in both isolated glomeruli and cultured mesangial cells. An investigation of mechanisms underlying this observation revealed activated ROCK functions through the phosphorylation of JNK and Erk and the nuclear translocation of NF-κB via actin dynamics. Knockdown by siRNA against ROCK1 and ROCK2 showed that ROCK2 but not ROCK1 controls this fibrotic machinery. Further in vivo experiments showed that ROCK2 activity in the renal cortex of db/db mice was elevated compared with control db/m mice. Importantly, oral administration of ROCK2 inhibitor attenuated renal ROCK2 activity, albuminuria, and glomerular fibrosis in db/db mice. These observations indicate that ROCK2 is a key player in the development of diabetic renal injury. Glomerular ROCK2 may be a potential therapeutic target for the treatment of diabetic kidney disease.


Subject(s)
Connective Tissue Growth Factor/biosynthesis , Cytoskeleton/metabolism , Fibrosis/genetics , Glomerular Mesangium/metabolism , NF-kappa B/biosynthesis , Transforming Growth Factor beta/pharmacology , rho-Associated Kinases/metabolism , Actins/metabolism , Animals , Diabetic Nephropathies/metabolism , Enzyme Activation , Glomerular Mesangium/cytology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred NOD , rho-Associated Kinases/antagonists & inhibitors
8.
Endocrine ; 63(1): 70-78, 2019 01.
Article in English | MEDLINE | ID: mdl-30128961

ABSTRACT

PURPOSE: Oxidative stress is an important mechanism for diabetic nephropathy. Studies showed that hemo oxygenase-1 (HO-1) expression in renal tissue of patients with diabetic nephropathy has upregulated, while the HO-1 can protect the body through anti-oxidative stress. The study aimed to preliminarily explore the molecular mechanism by observing the effect of Sitagliptin on HO-1 expression in renal tissue of rats with diabetic nephropathy. METHODS: The diabetic nephropathy rat model was established by STZ injection followed by intraperitoneal injection of sitagliptin with different concentrations. The mRNA expressions of HO-1 were detected by real-time PCR and Western blot and HO-1 enzyme activity change was detected by colorimetry. Human renal mesangial cell (HRMC) were cultured in vitro with high glucose concentration (30 µmol/L), phosphatidylinositol-3-kinase (PI3K) level and nuclear factor erythroid-2-related factor (Nrf2) content in cytoplasm and cell nucleus were observed before and after treatment with sitagliptin, as well as the action of in meditating HO-1 expression. RESULTS: HO-1 mRNA, protein level, and HO-1 enzyme activity in renal tissue of rats with diabetic nephropathy were significantly increased after treatment with sitagliptin (P < 0.05). As comparison, the 24 h urinary microalbumin, creatinine, and boold urea nitrogen were all decreased after treatment of sitagliptin (P < 0.05). Similar results were observed after CoPP (an agonist of HO-1) treatment (P < 0.05). In contrast, ZnPP, an inhibitor of HO-1, significantly abrogated the inhibitory effect of sitagliptin (P < 0.05). Phosphorylation of PI3K and Nrf2 nuclear translocation under high-glucose concentration condition was induced by sitagliptin in HRMC. HO-1 expression was suppressed by pretreating HRMC with PI3K inhibitor or RNA interference. CONCLUSIONS: Sitagliptin may induce HO-1 expression via activation of PI3K and Nrf2 in rats with diabetic nephropathy; HO-1 can improve the oxidative stress of diabetic nephropathy, eventually protect from diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/enzymology , Heme Oxygenase (Decyclizing)/biosynthesis , Hypoglycemic Agents/therapeutic use , Sitagliptin Phosphate/therapeutic use , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/pathology , Glomerular Mesangium/cytology , Glomerular Mesangium/drug effects , Glucose/pharmacology , Heme Oxygenase (Decyclizing)/drug effects , Humans , Kidney/pathology , Kidney Function Tests , Male , NF-E2-Related Factor 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
9.
J Interferon Cytokine Res ; 38(12): 566-577, 2018 12.
Article in English | MEDLINE | ID: mdl-30523751

ABSTRACT

Mesangial cell (MC) activation and macrophage infiltration are 2 major events closely related with each other in mesangial proliferative glomerulonephritis. In the anti-Thy 1 nephritis model, macrophages mediate the damage and also the expansion of mesangium through secreting various inflammatory factors; however, in glomerular microenvironment how MCs affect macrophage activity in the presence of various stimuli have not yet been understood. In the present study, we found that resting human MCs (HMCs) constitutively expressed chemokine [C-C motif] ligand 2 (CCL-2) and interleukin (IL)-6 and induced M2 polarization of macrophages in the coculture system. HMC proliferation and migration and expression of IL-6, CCL-2, and macrophage colony-stimulating factor in HMCs were enhanced after platelet-derived growth factor (PDGF)-BB stimulation, among which CCL-2 was responsible for inducing the M2 polarization of macrophages. Furthermore, PDGF-BB-stimulated HMCs alleviated the classical activation of macrophages and drove more intensified M2 polarization of macrophages than resting HMCs did. However, lipopolysaccharide and interferon-γ (IFN-γ) stimulated HMCs maintained the M1 phenotype of cocultured macrophages. In conclusion, MCs actively participated in glomerular inflammation through influencing macrophage polarization. The interplay between MCs and infiltrated macrophages is finely modulated by secretory factors such as PDGF-BB and IFN-γ in response to the renal inflammatory microenvironment.


Subject(s)
Glomerular Mesangium/cytology , Glomerular Mesangium/metabolism , Glomerulonephritis/pathology , Macrophage Activation/immunology , Macrophages/immunology , Animals , Becaplermin/metabolism , Cell Movement/immunology , Cell Polarity/immunology , Cell Proliferation/physiology , Cells, Cultured , Chemokine CCL2/biosynthesis , Coculture Techniques , Humans , Inflammation/pathology , Interferon-gamma/metabolism , Interleukin-6/biosynthesis , Macrophage Colony-Stimulating Factor/biosynthesis , Rats , Rats, Wistar
10.
Sci Rep ; 8(1): 10548, 2018 Jul 12.
Article in English | MEDLINE | ID: mdl-30002389

ABSTRACT

Diabetic nephropathy (DN) is the major cause of end-stage renal failure and is associated with increased morbidity and mortality compared with other causes of renal diseases. We previously found that Smad1 plays a critical role in the development of DN both in vitro and in vivo. However, functional interaction between Smad1 and Smad3 signaling in DN is unclear. Here, we addressed the molecular interplay between Smad1 and Smad3 signaling under a diabetic condition by using Smad3-knockout diabetic mice. Extracellular matrix (ECM) protein overexpression and Smad1 activation were observed in the glomeruli of db/db mice but were suppressed in the glomeruli of Smad3+/-; db/db mice. Smad3 activation enhanced the phosphorylation of Smad1 C-terminal domain but decreased the phosphorylation of linker domain, thus regulating Smad1 activation in advanced glycation end product-treated mesangial cells (MCs). However, forced phosphorylation of the Smad1 linker domain did not affect Smad3 activation in MCs. Phosphorylation of the Smad1 linker domain increased in Smad3+/-; db/db mice and probucol-treated db/db mice, which was consistent with the attenuation of ECM overproduction. These results indicate that Smad3 expression and activation or probucol treatment alters Smad1 phosphorylation, thus suggesting new molecular mechanisms underlying DN development and progression.


Subject(s)
Diabetic Nephropathies/pathology , Glycation End Products, Advanced/metabolism , Smad1 Protein/metabolism , Smad3 Protein/metabolism , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cells, Cultured , Diabetic Nephropathies/blood , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/genetics , Disease Models, Animal , Disease Progression , Extracellular Matrix Proteins/metabolism , Female , Glomerular Mesangium/cytology , Glomerular Mesangium/pathology , Glycation End Products, Advanced/blood , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Primary Cell Culture , Probucol/pharmacology , Probucol/therapeutic use , Protein Domains , Smad3 Protein/genetics
11.
Cell Physiol Biochem ; 45(6): 2161-2173, 2018.
Article in English | MEDLINE | ID: mdl-29544224

ABSTRACT

BACKGROUND/AIMS: Healing of mesangioproliferative glomerulonephritis involves degradation of excess extracellular matrix, resolution of hypercellularity by apoptosis and phagocytosis of apoptotic cells. Integrin receptors participate in the regulation of phagocytosis. In mice deficient for alpha8 integrin (Itga8-/-) healing of glomerulonephritis is delayed. As Itga8 is abundant in mesangial cells (MC) which are non-professional phagocytes, we hypothesized that Itga8 facilitates phagocytosis of apoptotic cells and matrix components by MC. METHODS: MC were isolated from wild type (WT) and Itga8-/- mice. Latex beads were coated with matrix components. Apoptosis was induced by cisplatin in macrophages and in DiI-stained MC. After coincubation of latex beads or apoptotic cells with MC, the phagocytosis rate was detected in WT and Itga8-/- MC via fluorescence microscopy and FACS analysis. RESULTS: Itga8-/- MC showed reduced phagocytosis of matrix-coated beads and apoptotic cells compared to WT MC. Reduction of stress fibers was observed in Itga8-/- compared to WT MC. Inhibition of cytoskeletal reorganization by inhibition of Rac1 or ROCK during phagocytosis significantly decreased the rate of phagocytosis by WT MC but not by Itga8-/- MC. CONCLUSION: The expression of Itga8 facilitates phagocytosis in MC, likely mediated by Itga8-cytoskeleton interactions. An impairment of MC phagocytosis might thus contribute to a delayed glomerular regeneration in Itga8-/- mice.


Subject(s)
Glomerular Mesangium/cytology , Integrin alpha Chains/genetics , Mesangial Cells/immunology , Phagocytosis , Animals , Apoptosis , Cells, Cultured , Gene Deletion , Gene Expression , Glomerular Mesangium/immunology , Glomerular Mesangium/metabolism , HEK293 Cells , Humans , Integrin alpha Chains/immunology , Mesangial Cells/metabolism , Mice , RAW 264.7 Cells , Rats, Sprague-Dawley
12.
J Am Soc Nephrol ; 29(4): 1128-1140, 2018 04.
Article in English | MEDLINE | ID: mdl-29335243

ABSTRACT

A critical aspect of kidney function occurs at the glomerulus, the capillary network that filters the blood. The glomerular basement membrane (GBM) is a key component of filtration, yet our understanding of GBM interactions with mesangial cells, specialized pericytes that provide structural stability to glomeruli, is limited. We investigated the role of nephronectin (Npnt), a GBM component and known ligand of α8ß1 integrin. Immunolocalization and in situ hybridization studies in kidneys of adult mice revealed that nephronectin is produced by podocytes and deposited into the GBM. Conditional deletion of Npnt from nephron progenitors caused a pronounced increase in mesangial cell number and mesangial sclerosis. Nephronectin colocalized with α8ß1 integrin to novel, specialized adhesion structures that occurred at sites of mesangial cell protrusion at the base of the capillary loops. Absence of nephronectin disrupted these adhesion structures, leading to mislocalization of α8ß1. Podocyte-specific deletion of Npnt also led to mesangial sclerosis in mice. These results demonstrate a novel role for nephronectin and α8ß1 integrin in a newly described adhesion complex and begin to uncover the molecular interactions between the GBM and mesangial cells, which govern mesangial cell behavior and may have a role in pathologic states.


Subject(s)
Extracellular Matrix Proteins/physiology , Glomerular Basement Membrane/physiology , Glomerular Mesangium/cytology , Pericytes/cytology , Podocytes/metabolism , Animals , Cell Adhesion/physiology , Cell Count , Epithelial Cells/metabolism , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/deficiency , Female , Focal Adhesions , Gene Deletion , Glomerular Mesangium/abnormalities , Integrins/metabolism , Kidney Glomerulus/abnormalities , Male , Mice , Mice, Mutant Strains , Organ Specificity , Pericytes/metabolism
13.
J Histochem Cytochem ; 66(1): 7-22, 2018 01.
Article in English | MEDLINE | ID: mdl-29077526

ABSTRACT

The homeostatic chemokine receptor CCR7 serves as key molecule in lymphocyte homing into secondary lymphoid tissues. Previous experiments from our group identified CCR7 also to be expressed by human mesangial cells. Exposing cultured human mesangial cells to the receptor ligand CCL21 revealed a positive effect on these cells regarding proliferation, migration, and survival. In the present study, we localized CCR7 and CCL21 during murine nephrogenesis. Analyzing wild-type and CCR7 deficient (CCR7-/-) mice, we observed a retarded glomerulogenesis during renal development and a significantly decreased mesangial cellularity in adult CCR7-/- mice, as a consequence of less mesangial cell proliferation between embryonic day E17.5 and week 5 postpartum. Cell proliferation assays and cell-wounding experiments confirmed reduced proliferative and migratory properties of mesangial cells cultured from CCR7-/- kidneys. To further emphasize the role of CCR7 as important factor for mesangial biology, we examined the chemokine receptor expression in rats after induction of a mesangioproliferative glomerulonephritis. Here, we demonstrated for the first time that extra- and intraglomerular mesangial cells that were CCR7-negative in control rats exhibited a strong CCR7 expression during the phase of mesangial repopulation and proliferation.


Subject(s)
Glomerular Mesangium/growth & development , Glomerular Mesangium/pathology , Glomerulonephritis/pathology , Receptors, CCR7/analysis , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Gene Deletion , Gene Expression Regulation, Developmental , Glomerular Mesangium/cytology , Glomerular Mesangium/ultrastructure , Glomerulonephritis/genetics , Kidney/cytology , Kidney/growth & development , Kidney/pathology , Kidney/ultrastructure , Male , Mice, Inbred C57BL , Rats, Wistar , Receptors, CCR7/genetics
14.
Kidney Int ; 92(6): 1419-1432, 2017 12.
Article in English | MEDLINE | ID: mdl-28688581

ABSTRACT

Renin lineage cells (RLCs) serve as a progenitor cell reservoir during nephrogenesis and after renal injury. The maintenance mechanisms of the RLC pool are still poorly understood. Since RLCs were also identified as a progenitor cell population in bone marrow we first considered that these may be their source in the kidney. However, transplantation experiments in adult mice demonstrated that bone marrow-derived cells do not give rise to RLCs in the kidney indicating their non-hematopoietic origin. Therefore we tested whether RLCs develop in the kidney through neogenesis (de novo differentiation) from cells that have never expressed renin before. We used a murine model to track neogenesis of RLCs by flow cytometry, histochemistry, and intravital kidney imaging. During nephrogenesis RLCs first appear at e14, form a distinct population at e16, and expand to reach a steady state level of 8-10% of all kidney cells in adulthood. De novo differentiated RLCs persist as a clearly detectable population through embryogenesis until at least eight months after birth. Pharmacologic stimulation of renin production with enalapril or glomerular injury induced the rate of RLC neogenesis in the adult mouse kidney by 14% or more than three-fold, respectively. Thus, the renal RLC niche is constantly filled by local de novo differentiation. This process could be stimulated consequently representing a new potential target to beneficially influence repair and regeneration after kidney injury.


Subject(s)
Acute Kidney Injury/pathology , Cell Differentiation/physiology , Glomerular Mesangium/physiology , Regeneration/drug effects , Renin/metabolism , Stem Cells/physiology , Acute Kidney Injury/chemically induced , Animals , Biopsy , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Bone Marrow Transplantation/methods , Cell Lineage/drug effects , Cell Lineage/physiology , Enalapril/pharmacology , Glomerular Mesangium/cytology , Glomerular Mesangium/drug effects , Glomerular Mesangium/pathology , Humans , Lipopolysaccharides/toxicity , Mesangial Cells/drug effects , Mesangial Cells/pathology , Mesangial Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Renin/genetics , Stem Cells/drug effects
15.
Exp Biol Med (Maywood) ; 242(2): 160-168, 2017 01.
Article in English | MEDLINE | ID: mdl-27590500

ABSTRACT

In the present study, the effect and mechanism of periostin on renal proliferation and extracellular matrix accumulation of lupus mice were investigated. MRL /lpr mice, known as lupus mice, were revealed to show enhanced periostin, proliferating cell nuclear antigen (PCNA), and extracellular matrix accumulation in the kidney accompanied by increased serum platelet-derived growth factor (PDGF). Again, cultured mouse mesangial cells (MMCs) were treated with PDGF, then periostin, and PCNA and secreted fibronectin were detected. The results showed that intracellular periostin and PCNA were respectively enhanced by 2.691 and 2.308 times in PDGF-treated MMC cells at 6 h after stimulation. In addition, secreted fibronectin was increased by 1.442 times. Next, the transfection of periostin shRNA vector in PDGF-stimulated MMC cells effectively suppressed periostin, PCNA and secreted fibronectin by 45.27%, 47.75%, and 39.95%, compared with PDGF-stimulated cells transfected with control vector. Furthermore, it was found that PDGF increased the expression of phospho-Akt (Ser 473) from 30 min to 6 h in MMCs. LY294002 effectively inhibited phospho-Akt (Ser 473) expression caused by PDGF stimulation. Then, periostin, PCNA, and fibronectin were respectively decreased by 69.61%, 46.00%, and 46.20%. In the end, phosphoinositide 3-kinase/protein kinase B/periostin was suggested to mediate PDGF-induced cell proliferation and extracellular matrix production in lupus nephritis.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Proliferation/physiology , Extracellular Matrix/metabolism , Lupus Nephritis/pathology , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Adhesion Molecules/genetics , Cells, Cultured , Chromones/pharmacology , Female , Fibronectins/metabolism , Glomerular Mesangium/cytology , Glomerular Mesangium/metabolism , Kidney/metabolism , Mesangial Cells/metabolism , Mice , Mice, Transgenic , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics
16.
PLoS One ; 11(2): e0150083, 2016.
Article in English | MEDLINE | ID: mdl-26919698

ABSTRACT

Human familial lecithin:cholesterol acyltransferase (LCAT) deficiency (FLD) is characterized by low HDL, accumulation of an abnormal cholesterol-rich multilamellar particle called lipoprotein-X (LpX) in plasma, and renal disease. The aim of our study was to determine if LpX is nephrotoxic and to gain insight into the pathogenesis of FLD renal disease. We administered a synthetic LpX, nearly identical to endogenous LpX in its physical, chemical and biologic characteristics, to wild-type and Lcat-/- mice. Our in vitro and in vivo studies demonstrated an apoA-I and LCAT-dependent pathway for LpX conversion to HDL-like particles, which likely mediates normal plasma clearance of LpX. Plasma clearance of exogenous LpX was markedly delayed in Lcat-/- mice, which have low HDL, but only minimal amounts of endogenous LpX and do not spontaneously develop renal disease. Chronically administered exogenous LpX deposited in all renal glomerular cellular and matrical compartments of Lcat-/- mice, and induced proteinuria and nephrotoxic gene changes, as well as all of the hallmarks of FLD renal disease as assessed by histological, TEM, and SEM analyses. Extensive in vivo EM studies revealed LpX uptake by macropinocytosis into mouse glomerular endothelial cells, podocytes, and mesangial cells and delivery to lysosomes where it was degraded. Endocytosed LpX appeared to be degraded by both human podocyte and mesangial cell lysosomal PLA2 and induced podocyte secretion of pro-inflammatory IL-6 in vitro and renal Cxl10 expression in Lcat-/- mice. In conclusion, LpX is a nephrotoxic particle that in the absence of Lcat induces all of the histological and functional hallmarks of FLD and hence may serve as a biomarker for monitoring recombinant LCAT therapy. In addition, our studies suggest that LpX-induced loss of endothelial barrier function and release of cytokines by renal glomerular cells likely plays a role in the initiation and progression of FLD nephrosis.


Subject(s)
Kidney Glomerulus/drug effects , Lecithin Cholesterol Acyltransferase Deficiency/metabolism , Lipoprotein-X/toxicity , Proteinuria/etiology , Animals , Apolipoprotein A-I/metabolism , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , Endothelial Cells/metabolism , Endothelial Cells/pathology , Extracellular Matrix/metabolism , Gene Expression Profiling , Glomerular Basement Membrane/drug effects , Glomerular Basement Membrane/pathology , Glomerular Mesangium/cytology , Glomerular Mesangium/metabolism , Glomerular Mesangium/pathology , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-6/metabolism , Kidney Glomerulus/pathology , Lecithin Cholesterol Acyltransferase Deficiency/pathology , Lipoprotein-X/metabolism , Lipoprotein-X/pharmacokinetics , Lipoproteins, HDL/metabolism , Lysosomes/metabolism , Metabolic Clearance Rate , Mice , Mice, Inbred C57BL , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Phospholipases A2/metabolism , Pinocytosis , Podocytes/metabolism , Podocytes/pathology , Proteinuria/chemically induced , Proteinuria/genetics , Proteinuria/pathology
17.
Ren Fail ; 38(3): 458-64, 2016.
Article in English | MEDLINE | ID: mdl-26837331

ABSTRACT

Cytotoxin-associated antigen A (CagA), a major virulence factor of Helicobacter pylori (Hp), is associated with the pathogenesis of peptic ulcer and gastric cancer. Recent researches demonstrated that Hp exists in palatine tonsil in all studied IgA nephropathy (IgAN) patients, most of which were CagA-positive, suggesting that CagA may be a causative pathogenic factor of IgAN. However, the underlying molecular mechanisms and signaling pathway are still largely unclear. In the present study, CCK8 assay, enzyme-linked immunosorbent assay, and immunohistochemistry were performed to investigate the effect of CagA on cell proliferation and extracellular matrix secretion in rat glomerular mesangial cells. RT-PCR and western blotting were used to reveal the potential signaling pathway. Rat glomerular mesangial cells were treated with recombinant CagA protein for 72 h, in a dose- and time-dependent manner. We found that CagA promoted cell proliferation and extracellular matrix secretion by inhibiting signaling pathway of apoptosis. Taken together, these findings suggested that CagA induced cellular injury in glomerular mesangium by proliferation and secretion of extracellular matrix, and may play an important role in pathogenesis of IgAN.


Subject(s)
Antigens, Bacterial/pharmacology , Apoptosis , Bacterial Proteins/pharmacology , Cell Proliferation , Extracellular Matrix/metabolism , Glomerular Mesangium/cytology , Signal Transduction , Animals , Cell Culture Techniques , Cell Line , Glomerulonephritis, IGA/physiopathology , Rats
18.
Biochim Biophys Acta ; 1850(12): 2506-17, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26423448

ABSTRACT

The phenomenon that heme oxygenase-1 (HO-1) protects cell from injury yet its enzymatic product, iron, may facilitate generation of free radical has been long puzzling. Here we establish a functional connection between ferritin heavy chain (FHC) and HO-1. In human lupus nephritis HO-1 and FHC are colocalized within the glomeruli. In rodent anti-Thy1 (thymocyte antigen 1) induced glomerulonephritis, heme oxygenase blockade lowers the expression of FHC and accelerates mesangial cell death. Stimulation of heme oxygenase in cultured rat mesangial cell enhances its resistance to hydrogen peroxide, whereas FHC knockdown by RNA interference compromises this salutary effect. RNA interference of HO-1 makes the cell more susceptible to hydrogen peroxide, which can be rescued by forced expression of wild-type FHC but not mutants that lose the capacity of iron storage and ferroxidase activity. Phosphorylation of JunD was not sustained in these cells. Microarray analysis identifies four candidate transcriptional factors that may regulate the HO-1-induced transcription of FHC. Our results support the role of FHC in neutralizing the iron toxicity as well as mediating the protective effect of HO-1 in response to oxidative stress.


Subject(s)
Apoferritins/physiology , Heme Oxygenase-1/physiology , Oxidative Stress , Animals , Glomerular Mesangium/cytology , Glomerular Mesangium/metabolism , Rats
19.
Cell Physiol Biochem ; 36(5): 1793-1808, 2015.
Article in English | MEDLINE | ID: mdl-26184511

ABSTRACT

BACKGROUND/AIMS: IgA nephropathy (IgAN) is the most common form of primary glomerulonephritis, and often aggravates by mucosal infection. Secretory IgA (SIgA) is the dominant immunoglobulin in mucosal immunity, and is deposited in the mesangium in IgAN. The biological effects of SIgA on mesangial cells are poorly understood. METHODS: Deposition of SIgA in frozen renal sections from IgAN patients was detected and the association between deposition of SIgA and patients characteristics was analyzed. The biological effects of SIgA and polymeric IgA (pIgA) on human renal mesangial cells were compared. We also studied the molecular mechanism of microRNA regulating the inflammatory effects of SIgA on mesangial cells. RESULTS: Fifty-five of 176 patients had SIgA deposition with higher incidence of infection history and hematuria, lower serum cystatin C, ß2 microglobulin, blood urea nitrogen and T-grade in the Oxford classification, compared with patients without SIgA deposition. SIgA stimulated mesangial cells at a higher ratio of proliferation and higher production of interleukin (IL)-6, IL-8, monocyte chemotactic protein 1, transforming growth factor-ß1 and fibronectin, compared with SIgA from healthy volunteers. The proliferation and cytokines production in mesangial cells stimulated by SIgA were significantly lower than that stimulated by pIgA. miR- 16 targeted the 3'-untranslated region of IL-6 and suppressed its translation in mesangial cells induced by SIgA. CONCLUSIONS: The biological effects of SIgA on mesangial cells differ from those of pIgA. SIgA stimulates mesangial cell proliferation and production of proinflammatory cytokines. IL-6 production is regulated by miR-16 in mesangial cells.


Subject(s)
Cytokines/biosynthesis , Glomerular Mesangium/metabolism , Glomerulonephritis, IGA/blood , Immunoglobulin A, Secretory/pharmacology , Adult , Case-Control Studies , Cells, Cultured , Female , Glomerular Mesangium/cytology , Humans , Immunoglobulin A, Secretory/isolation & purification , Male , Middle Aged
20.
Cell Physiol Biochem ; 36(5): 1928-38, 2015.
Article in English | MEDLINE | ID: mdl-26202353

ABSTRACT

BACKGROUND AND AIMS: There is accumulating evidence that sympathetic nervous hyperactivity contributes to the pathogenesis of glomerular sclerosis independent of blood pressure effects. A previous study showed that α1-adrenoceptor (α1-AR) antagonists inhibit mesangial cell (MC) proliferation. However, the underlying mechanism remains unclear. METHODS AND RESULTS: We found that α1-AR is expressed in a human mesangial cell line. The α1-AR agonist phenylephrine (PE) induced Ca(2+) influx as well as release from intracellular Ca(2+) stores. Blockade of TRPC6 with siRNA, anti-TRPC6 antibodies and a TRPC blocker attenuated the PE-induced [Ca(2+)]i increase. Additionally, the PE-induced [Ca(2+)]i increase was phospholipase C dependent. Furthermore, PE induced a [Ca(2+)]i increase even when the intracellular Ca(2+) stores were already depleted. This effect was mimicked by an analog of diacylglycerol. These results suggested that, upon α1-AR stimulation, TRPC6 mediates Ca(2+) influx via a receptor-operated Ca(2+) entry mechanism. Finally, TRPC6 contributes to the PE-induced MC proliferation. The mechanisms are associated with the extracellular signal-regulated kinase (ERK) signaling pathway because blockade of TRPC6 and chelation of extracellular Ca(2+) abrogated PE-induced ERK1/2 abrogated PE-induced ERK1/2 phosphorylation. CONCLUSION: TRPC6 channels are involved in α1-AR activation-induced Ca(2+) entry, which mediates proliferation via ERK signaling in human MCs.


Subject(s)
Calcium/metabolism , Cell Proliferation , Glomerular Mesangium/metabolism , Receptors, Adrenergic, alpha-1/drug effects , TRPC Cation Channels/physiology , Adrenergic alpha-Agonists/pharmacology , Cell Line , Glomerular Mesangium/cytology , Humans , Ion Transport , MAP Kinase Signaling System , Receptors, Adrenergic, alpha-1/metabolism , TRPC6 Cation Channel
SELECTION OF CITATIONS
SEARCH DETAIL
...