Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Kidney Int ; 72(4): 489-98, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17554257

ABSTRACT

Detachment or apoptosis of podocytes leads to proteinuria and glomerulosclerosis. There are no current interventions for diabetic or non-diabetic glomerular diseases specifically preventing podocyte apoptosis. Binding of erythropoiesis stimulating proteins (ESPs) to receptors on non-hematopoietic cells has been shown to have anti-apoptotic effects in vitro, in vivo, and in preliminary human studies. Recently, erythropoietin receptors were identified on podocytes; therefore, we tested effects of darbepoetin alfa in preventing podocyte apoptosis. Cultured immortalized mouse podocytes were treated with low-dose ultraviolet-C (uv-C) irradiation to induce apoptosis in the absence or presence of darbepoetin alfa. Apoptosis was quantified by Hoechst staining and by caspase 3 cleavage assessed by Western blots. Pretreatment with darbepoetin alfa significantly reduced podocyte apoptosis with this effect involving intact Janus family protein kinase-2 (JAK2) and AKT signaling pathways. Additionally, darbepoetin alfa was found protective against transforming growth factor-beta1 but not puromycin aminonucleoside induced apoptosis. Mice with anti-glomerular antibody induced glomerulonephritis had significantly less proteinuria, glomerulosclerosis, and podocyte apoptosis when treated with darbepoetin alfa. Our studies show that treatment of progressive renal diseases characterized by podocyte apoptosis with ESPs may be beneficial in slowing progression of chronic kidney disease.


Subject(s)
Apoptosis/drug effects , Erythropoietin/analogs & derivatives , Glomerulonephritis/prevention & control , Podocytes/drug effects , Protective Agents/pharmacology , Signal Transduction/drug effects , Animals , Antibodies , Apoptosis/radiation effects , Autoantibodies , Cell Proliferation/drug effects , Cells, Cultured , Darbepoetin alfa , Disease Models, Animal , Disease Progression , Erythropoietin/pharmacology , Erythropoietin/therapeutic use , Glomerulonephritis/complications , Glomerulonephritis/immunology , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/prevention & control , Janus Kinase 2/metabolism , Mice , Podocytes/metabolism , Podocytes/pathology , Podocytes/radiation effects , Protective Agents/therapeutic use , Proteinuria/etiology , Proteinuria/pathology , Proteinuria/prevention & control , Proto-Oncogene Proteins c-akt/metabolism , Puromycin Aminonucleoside/pharmacology , Receptors, Erythropoietin/drug effects , Receptors, Erythropoietin/metabolism , Signal Transduction/radiation effects , Transforming Growth Factor beta1/metabolism , Ultraviolet Rays
2.
Kidney Int ; 72(1): 26-36, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17457377

ABSTRACT

Human genetic and in vivo animal studies have helped to define the critical importance of podocytes for kidney function in health and disease. However, as in any other research area, by default these approaches do not allow for mechanistic studies. Such mechanistic studies require the availability of cells grown ex vivo (i.e., in culture) with the ability to directly study mechanistic events and control the environment such that specific hypotheses can be tested. A seminal breakthrough came about a decade ago with the documentation of differentiation in culture of primary rat and human podocytes and the subsequent development of conditionally immortalized differentiated podocyte cell lines that allow deciphering the decisive steps of differentiation and function of 'in vivo' podocytes. Although this paper is not intended to provide a comprehensive review of podocyte biology, nor their role in proteinuric renal diseases or progressive glomerulosclerosis, it will focus specifically on several aspects of podocytes in culture. In particular, we will discuss the scientific and research rationale and need for cultured podocytes, how podocyte cell-culture evolved, and how cultured podocytes are currently being used to uncover novel functions of podocytes that can then be validated in vivo in animal or human studies. In addition, we provide a detailed description of how to properly culture and characterize podocytes to avoid potential pitfalls.


Subject(s)
Cell Culture Techniques/trends , Podocytes/cytology , Animals , Cell Differentiation/physiology , Cells, Cultured , Humans , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Mice , Phenotype , Podocytes/physiology , Rats
3.
Kidney Int ; 70(11): 1962-73, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17035936

ABSTRACT

A decline in podocyte number correlates with progression to glomerulosclerosis. A mechanism underlying reduced podocyte number is the podocyte's relative inability to proliferate in response to injury. Injury by the podocyte toxin puromycin aminonucleoside (PA) is mediated via reactive oxygen species (ROS). The precise role of ROS in the pathogenesis of PA-induced glomerulosclerosis remains to be determined. We sought to examine whether PA-induced ROS caused podocyte DNA damage, possibly accounting for the podocyte's inability to proliferate in response to PA. In vitro, podocytes were exposed to PA, with or without the radical scavenger 1,3-dimethyl-2-thiourea (DMTU). In vivo, male Sprague-Dawley rats were divided into experimental groups (n = 6/group/time point): PA, PA with DMTU, and control, killed at days 1.5, 3, or 7. DNA damage was measured by DNA precipitation, apurinic/apyrimidinic site, Comet, and 8-hydroxydeoxyguanosine assays. Cell cycle checkpoint protein upregulation (by immunostaining and Western blotting), histopathology, and biochemical parameters were examined. DNA damage was increased in cultured podocytes that received PA, but not PA with DMTU. PA exposure activated specific cell cycle checkpoint proteins, with attenuation by DMTU. DNA repair enzymes were activated, providing evidence for attempted DNA repair. The PA-treated animals developed worse proteinuria and histopathologic disease and exhibited more DNA damage than the DMTU pretreated group. No significant apoptosis was detected by terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling staining. A mechanism underlying the lack of podocyte proliferation following PA-induced injury in vitro and in vivo may be ROS-mediated DNA damage, with upregulation of specific cell cycle checkpoints leading to cell cycle arrest.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , DNA Damage , Podocytes/drug effects , Puromycin Aminonucleoside/pharmacology , Animals , Apoptosis , Cell Cycle Proteins/drug effects , Cells, Cultured , DNA Repair Enzymes/drug effects , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Male , Mice , Oxidative Stress/drug effects , Proteinuria , Rats , Rats, Sprague-Dawley
4.
Oncogene ; 21(11): 1750-8, 2002 Mar 07.
Article in English | MEDLINE | ID: mdl-11896606

ABSTRACT

Apoptosis is closely linked to proliferation. In this study we showed that inducing apoptosis in mouse mesangial cells with ultraviolet (UV) irradiation was associated with increased cyclin A-cyclin dependent kinase (CDK) 2 activity. Inhibiting CDK2 activity with Roscovitine or dominant negative mutant reduced apoptosis. Because apoptosis typically begins in the cytoplasm, we tested the hypothesis that the subcellular localization of CDK2 determines the proliferative or apoptotic fate of the cell. Our results showed that cyclin A-CDK2 was nuclear in proliferating cells. However, inducing apoptosis in proliferating cells with UV irradiation was associated with a decrease in nuclear cyclin A and CDK2 protein levels. This coincided with an increase in protein and kinase activity for cyclin A-CDK2 in the cytoplasm. Translocation of cyclin A-CDK2 also occurred in p53-/- mesangial cells. Finally, we showed that caspase-3 activity was significantly reduced by inhibiting CDK2 activity with Roscovitine. In summary, our results show that apoptosis is associated with an increase in cytoplasmic cyclin A-CDK2 activity, which is p53 independent and upstream of caspase-3. We propose that the subcellular localization of CDK2 determines the proliferative or apoptotic fate of the cell.


Subject(s)
Apoptosis , CDC2-CDC28 Kinases , Cyclin-Dependent Kinases/physiology , Glomerular Mesangium/cytology , Protein Serine-Threonine Kinases/physiology , Animals , Biological Transport , Caspase 3 , Caspases/physiology , Cell Division , Cells, Cultured , Cyclin A/physiology , Cyclin E/physiology , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/analysis , Cytoplasm/enzymology , Glomerular Mesangium/enzymology , Glomerular Mesangium/ultrastructure , Mice , Nuclear Envelope/enzymology , Protein Serine-Threonine Kinases/analysis , Tumor Suppressor Protein p53/physiology
5.
Kidney Int ; 59(3): 905-12, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231345

ABSTRACT

BACKGROUND: The membrane attack complex C5b-9 causes injury in many forms of immune-mediated glomerular diseases characterized by mesangial cell (MC) proliferation and inhibiting C5b-9 decreases MC proliferation in vivo. Membrane insertion of sublytic quantities of the membrane attack complex of complement (C5b-9) is a potent stimulus for cell activation and the production of a variety of cytokines, growth factors, oxidants, matrix components, and other nephritogenic molecules. In vivo, a common response of MC to C5b-9--mediated injury is cell proliferation, an event closely linked to matrix expansion and sclerosis. In this study, we tested the hypothesis that C5b-9 might also serve as a mitogenic stimulus for MCs. METHODS: Rat MCs in vitro were exposed anti-Thy1 antibody and 2% normal PVG serum (a complement source) to induce sublytic C5b-9 attack and DNA synthesis and cell number were measured. Control MCs were exposed to antibody and C6-deficient PVG serum. RESULTS: Sublytic C5b-9--induced injury to MCs is sufficient to induce DNA synthesis. Furthermore, C5b-9 augmented DNA synthesis induced by platelet-derived growth factor (PDGF) and 5% fetal calf serum. C5b-9--induced DNA synthesis was reduced by inhibiting reactive oxygen species (ROS) with superoxide dismutase and catalase, but not by neutralizing the mitogenic growth factors PDGF and basic fibroblast growth factor (bFGF). CONCLUSIONS: This study demonstrates that C5b-9 may directly increase DNA synthesis in cultured MCs, which are mediated in part by the release of ROS, and that C5b-9 also augments DNA synthesis induced in MCs by other known mitogens.


Subject(s)
Complement Membrane Attack Complex/pharmacology , DNA/biosynthesis , Glomerular Mesangium/drug effects , Glomerular Mesangium/metabolism , Animals , Catalase/pharmacology , Cell Count , Cells, Cultured , Fibroblast Growth Factor 2/physiology , Glomerular Mesangium/cytology , Osmolar Concentration , Platelet-Derived Growth Factor/pharmacology , Platelet-Derived Growth Factor/physiology , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/pharmacology
6.
Kidney Int ; 56(2): 538-48, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10432393

ABSTRACT

BACKGROUND: The C5b-9 membrane attack complex of complement is the principal mediator of injury induced experimentally by antibodies directed at glomerular cell membranes. In experimental membranous nephropathy, C5b-9 induced injury to the glomerular visceral epithelial cell (VEC) is associated with DNA synthesis, but not cytokinesis. In the current study we determined if C5b-9 increases DNA synthesis in VEC in vitro, and defined the mechanisms involved. METHODS: Rat VEC in vitro were divided into three groups: (1) sensitized with anti-VEC antibody and exposed to sublytic concentrations of C +/PVG serum (normal complement components); (2) anti-VEC antibody and control C-/PVG serum (C6 deficient); (3) no anti-VEC antibody. DNA synthesis (BrdU staining), mitosis (mitotic figures) and cytokinesis (cell counts) were measured at 24 and 48 hours. To examine the expression of specific S-phase and M-phase cell cycle regulatory proteins and their inhibitors, immunostaining and Western blot analysis was performed for cyclin A, CDK2, p21 and p27, cyclin B and cdc2. RESULTS: In the absence of growth factors, sublytic C5b-9 attack did not increase proliferation. In contrast, sublytic C5b-9 attack (group 1) augmented growth factor induced DNA synthesis by 50% compared to controls (groups 2 and 3; P < 0.001), and was accompanied by increased levels of cyclin A and CDK2, and a decrease in the cyclin kinase inhibitor p27 (but not p21). Sublytic C5b-9 attack reduced the expression of the M phase cell cycle proteins, cyclin B and cdc2, accompanied by reduced mitosis (mitotic figures) and cytokinesis (cell number). CONCLUSIONS: Our results show that the C5b-9 augmented growth factor entry into the S phase in VEC is regulated by changes in specific cell cycle regulatory proteins. However, antibody and complement decreased the M phase cell cycle proteins, and prevented VEC mitosis and cytokinesis, suggesting a delay or arrest at the G2/M phase.


Subject(s)
Complement Membrane Attack Complex/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Kidney Glomerulus/cytology , Animals , Antibodies/pharmacology , Cell Division/drug effects , Cells, Cultured , Complement Membrane Attack Complex/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , DNA/biosynthesis , Epithelial Cells/enzymology , Fibroblast Growth Factor 2/immunology , Fibroblast Growth Factor 2/pharmacology , G2 Phase/drug effects , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Mitogens/metabolism , Mitosis/drug effects , Neutralization Tests , Rats , S Phase/drug effects , Transforming Growth Factor beta/pharmacology
7.
J Clin Invest ; 103(5): 597-604, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10074476

ABSTRACT

Proliferation and apoptosis are increased in many types of inflammatory diseases. A role for the cyclin kinase inhibitor p27(Kip1) (p27) in limiting proliferation has been shown. In this study, we show that p27(-/-) mesangial cells and fibroblasts have strikingly elevated rates of apoptosis, not proliferation, when deprived of growth factors. Apoptosis was rescued by restoration of p27 expression. Cyclin A-cyclin-dependent kinase 2 (CDK2) activity, but not cyclin E-CDK2 activity, was increased in serum-starved p27(-/-) cells, and decreasing CDK2 activity, either pharmacologically (Roscovitine) or by a dominant-negative mutant, inhibited apoptosis. Our results show that a new biological function for the CDK inhibitor p27 is protection of cells from apoptosis by constraining CDK2 activity. These results suggest that CDK inhibitors are necessary for coordinating the cell cycle and cell-death programs so that cell viability is maintained during exit from the cell cycle.


Subject(s)
Apoptosis , CDC2-CDC28 Kinases , Cell Cycle Proteins , Glomerular Mesangium/metabolism , Glomerular Mesangium/pathology , Microtubule-Associated Proteins/metabolism , Tumor Suppressor Proteins , Animals , Apoptosis/genetics , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Enzymologic , Genes, Tumor Suppressor , Mice , Microtubule-Associated Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley
8.
Kidney Int ; 54(2): 416-25, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9690208

ABSTRACT

BACKGROUND: Studies were undertaken to characterize the cellular composition that occurs in glomeruli and the tubulointerstitium of a passive model of complement-independent crescentic nephritis in mice. METHODS: Glomerulonephritis was induced by the injection of antibody to whole rabbit glomeruli, and tissue was examined histologically at 7, 14 and 28 days. RESULTS: Mice developed proteinuria, glomerular crescents, and progressive glomerulosclerosis and tubulointerstitial fibrosis. The majority of the cells within the crescents appeared to be intrinsic ezrin-positive epithelial cells of visceral or parietal origin. Many of the ezrin positive cells were proliferating and expressing the PDGF receptor. Despite expression of the macrophage adhesive protein, osteopontin, the early crescents were devoid of infiltrating macrophages, T cells or myofibroblasts, which could be explained by the finding that the Bowman's capsule remained intact. Tubulointerstitial damage also occurred, and included tubular dilation and atrophy, periglomerular and patchy interstitial infiltration and interstitial fibrosis with increased interstitial deposition of type IV collagen and laminin. Interstitial infiltrating cells included macrophages, CD4+ T lymphocytes, CD8+ T lymphocytes, and activated myofibroblasts. Tubular osteopontin expression was increased in the areas of tubulointerstitial damage and was associated with interstitial macrophage infiltration. CONCLUSIONS: We describe an experimental model of complement-independent murine crescentic nephritis associated with tubulointerstitial injury. Proliferating glomerular epithelial cells are the main cellular components of the crescents in this model.


Subject(s)
Glomerulonephritis/etiology , Kidney Glomerulus/pathology , Kidney Tubules/pathology , Animals , Cell Division , Epithelial Cells/pathology , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Osteopontin , Rabbits , Sheep , Sialoglycoproteins/biosynthesis
9.
J Clin Invest ; 100(10): 2512-20, 1997 Nov 15.
Article in English | MEDLINE | ID: mdl-9366565

ABSTRACT

Glomerular injury is characterized by mesangial cell (MC) proliferation and matrix formation. We sought to determine if reducing the activity of cyclin-dependent kinase 2 (CDK2) with the purine analogue, Roscovitine, decreased MC proliferation in vitro and in vivo. Roscovitine (25 microM) inhibited FCS-induced proliferation (P < 0.0001) in cultured MC. Rats with experimental mesangial proliferative glomerulonephritis (Thy1 model) were divided into two groups. A prevention group received daily intraperitoneal injections of Roscovitine in DMSO (2.8 mg/kg) starting at day 1. A treatment group received daily Roscovitine starting at day 3, when MC proliferation was established. Control Thy1 rats received DMSO alone. MC proliferation (PCNA +/OX7 + double immunostaining) was reduced by > 50% at days 5 and 10 in the Roscovitine prevention group, and at day 5 in the treatment group (P < 0.0001). Early administration of Roscovitine reduced immunostaining for collagen type IV, laminin, and fibronectin at days 5 and 10 (r = 0.984; P < 0.001), which was associated with improved renal function (urinary protein/creatinine, blood urea nitrogen, P < 0.05). We conclude that reducing the activity of CDK2 with Roscovitine in experimental glomerulonephritis decreases cell proliferation and matrix production, resulting in improved renal function, and may be a useful therapeutic intervention in disease characterized by proliferation.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle/drug effects , Cell Nucleus/drug effects , Cyclin-Dependent Kinases/physiology , Glomerular Mesangium/pathology , Glomerulonephritis, Membranoproliferative/pathology , Glomerulonephritis, Membranoproliferative/physiopathology , Protein Serine-Threonine Kinases/physiology , Purines/pharmacology , Animals , Cell Cycle/physiology , Cell Nucleus/ultrastructure , Cells, Cultured , Collagen/biosynthesis , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Fibronectins/biosynthesis , Glomerular Mesangium/drug effects , Glomerular Mesangium/physiopathology , Kidney Function Tests , Laminin/biosynthesis , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rats , Rats, Wistar , Roscovitine
SELECTION OF CITATIONS
SEARCH DETAIL
...