Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 17(12): 1361-1372, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27798618

ABSTRACT

Hemolysis drives susceptibility to bacterial infections and predicts poor outcome from sepsis. These detrimental effects are commonly considered to be a consequence of heme-iron serving as a nutrient for bacteria. We employed a Gram-negative sepsis model and found that elevated heme levels impaired the control of bacterial proliferation independently of heme-iron acquisition by pathogens. Heme strongly inhibited phagocytosis and the migration of human and mouse phagocytes by disrupting actin cytoskeletal dynamics via activation of the GTP-binding Rho family protein Cdc42 by the guanine nucleotide exchange factor DOCK8. A chemical screening approach revealed that quinine effectively prevented heme effects on the cytoskeleton, restored phagocytosis and improved survival in sepsis. These mechanistic insights provide potential therapeutic targets for patients with sepsis or hemolytic disorders.


Subject(s)
Gram-Negative Bacterial Infections/immunology , Guanine Nucleotide Exchange Factors/metabolism , Heme/metabolism , Hemolysis/immunology , Macrophages/immunology , Phagocytosis , Sepsis/immunology , Animals , Anti-Bacterial Agents/therapeutic use , Cytoskeleton/metabolism , Female , Gram-Negative Bacterial Infections/drug therapy , Guanine Nucleotide Exchange Factors/genetics , Heme Oxygenase-1/genetics , Hemolysis/drug effects , Humans , Immune Evasion , Macrophages/drug effects , Macrophages/microbiology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/drug effects , Quinine/therapeutic use , RAW 264.7 Cells , Sepsis/drug therapy , cdc42 GTP-Binding Protein/metabolism
2.
Cell ; 151(2): 384-99, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23063127

ABSTRACT

Vascular endothelial growth factor and its receptors, FLK1/KDR and FLT1, are key regulators of angiogenesis. Unlike FLK1/KDR, the role of FLT1 has remained elusive. FLT1 is produced as soluble (sFLT1) and full-length isoforms. Here, we show that pericytes from multiple tissues produce sFLT1. To define the biologic role of sFLT1, we chose the glomerular microvasculature as a model system. Deletion of Flt1 from specialized glomerular pericytes, known as podocytes, causes reorganization of their cytoskeleton with massive proteinuria and kidney failure, characteristic features of nephrotic syndrome in humans. The kinase-deficient allele of Flt1 rescues this phenotype, demonstrating dispensability of the full-length isoform. Using cell imaging, proteomics, and lipidomics, we show that sFLT1 binds to the glycosphingolipid GM3 in lipid rafts on the surface of podocytes, promoting adhesion and rapid actin reorganization. sFLT1 also regulates pericyte function in vessels outside of the kidney. Our findings demonstrate an autocrine function for sFLT1 to control pericyte behavior.


Subject(s)
Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Podocytes/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Gangliosides/metabolism , Humans , In Vitro Techniques , Lipid Metabolism , Lipids/chemistry , Mice , Mice, Transgenic , Pericytes/metabolism , Proteinuria/metabolism , Signal Transduction , Syndecans/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics
3.
Nature ; 565(7740): 505-510, 2019 01.
Article in English | MEDLINE | ID: mdl-30651639

ABSTRACT

The increasing prevalence of diabetes has resulted in a global epidemic1. Diabetes is a major cause of blindness, kidney failure, heart attacks, stroke and amputation of lower limbs. These are often caused by changes in blood vessels, such as the expansion of the basement membrane and a loss of vascular cells2-4. Diabetes also impairs the functions of endothelial cells5 and disturbs the communication between endothelial cells and pericytes6. How dysfunction of endothelial cells and/or pericytes leads to diabetic vasculopathy remains largely unknown. Here we report the development of self-organizing three-dimensional human blood vessel organoids from pluripotent stem cells. These human blood vessel organoids contain endothelial cells and pericytes that self-assemble into capillary networks that are enveloped by a basement membrane. Human blood vessel organoids transplanted into mice form a stable, perfused vascular tree, including arteries, arterioles and venules. Exposure of blood vessel organoids to hyperglycaemia and inflammatory cytokines in vitro induces thickening of the vascular basement membrane. Human blood vessels, exposed in vivo to a diabetic milieu in mice, also mimic the microvascular changes found in patients with diabetes. DLL4 and NOTCH3 were identified as key drivers of diabetic vasculopathy in human blood vessels. Therefore, organoids derived from human stem cells faithfully recapitulate the structure and function of human blood vessels and are amenable systems for modelling and identifying the regulators of diabetic vasculopathy, a disease that affects hundreds of millions of patients worldwide.


Subject(s)
Basement Membrane/pathology , Blood Vessels/pathology , Diabetic Angiopathies/pathology , Models, Biological , Organoids/pathology , Organoids/transplantation , Adaptor Proteins, Signal Transducing , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Arteries/cytology , Arteries/drug effects , Arterioles/cytology , Arterioles/drug effects , Basement Membrane/cytology , Basement Membrane/drug effects , Blood Vessels/cytology , Blood Vessels/drug effects , Blood Vessels/growth & development , Calcium-Binding Proteins , Diabetic Angiopathies/enzymology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Hyperglycemia/complications , In Vitro Techniques , Inflammation Mediators/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Organoids/cytology , Organoids/drug effects , Pericytes/cytology , Pericytes/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Receptor, Notch3/metabolism , Signal Transduction , Venules/cytology , Venules/drug effects
4.
FASEB J ; 34(4): 5453-5464, 2020 04.
Article in English | MEDLINE | ID: mdl-32086849

ABSTRACT

The foot processes of podocytes exhibit a dynamic actin cytoskeleton, which maintains their complex cell structure and antagonizes the elastic forces of the glomerular capillary. Interdigitating secondary foot processes form a highly selective filter for proteins in the kidney, the slit membrane. Knockdown of slit membrane components such as Nephrin or Neph1 and cytoskeletal adaptor proteins such as CD2AP in mice leads to breakdown of the filtration barrier with foot process effacement, proteinuria, and early death of the mice. Less is known about the crosstalk between the slit membrane-associated proteins and cytoskeletal components inside the podocyte foot processes. Our study shows that LASP-1, an actin-binding protein, is highly expressed in podocytes. Electron microscopy studies demonstrate that LASP-1 is found at the slit membrane suggesting a role in anchoring slit membrane components to the actin cytoskeleton. Live cell imaging experiments with transfected podocytes reveal that LASP-1 is either part of a highly dynamic granular complex or a static, actin cytoskeleton-bound protein. We identify CD2AP as a novel LASP-1 binding partner that regulates its association with the actin cytoskeleton. Activation of the renin-angiotensin-aldosterone system, which is crucial for podocyte function, leads to phosphorylation and altered localization of LASP-1. In vivo studies using the Drosophila nephrocyte model indicate that Lasp is necessary for the slit membrane integrity and functional filtration.


Subject(s)
Actin Cytoskeleton/physiology , Cell Membrane/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Kidney/physiology , Microfilament Proteins/metabolism , Podocytes/physiology , Animals , Drosophila Proteins/genetics , Microfilament Proteins/genetics , Phosphorylation
5.
Kidney Int ; 97(1): 89-94, 2020 01.
Article in English | MEDLINE | ID: mdl-31718844

ABSTRACT

Renal allograft rejection can be prevented by immunological tolerance, which may be associated with de novo formed lymphatic vessels in the donor kidney after transplantation in man. A suitable mouse model of renal allograft rejection in which lymphangiogenesis can be deliberately induced in the graft is critical for elucidating the mechanisms responsible for the association between attenuated transplant rejection and abundance of lymphatic vessels. Here we describe the development of a novel mouse model of rapid renal transplant rejection in which transgenic induction of lymphangiogenesis in the immune-incompatible graft greatly extends its survival time. Thus, our novel approach may facilitate exploitation of lymphangiogenesis in the grafted organ.


Subject(s)
Graft Rejection/prevention & control , Graft Survival/immunology , Kidney Diseases/surgery , Kidney Transplantation/adverse effects , Lymphangiogenesis/immunology , Allografts/immunology , Allografts/pathology , Animals , Disease Models, Animal , Female , Gene Knock-In Techniques , Graft Rejection/immunology , Graft Rejection/pathology , Humans , Kidney/immunology , Kidney/pathology , Longevity/immunology , Lymphatic Vessels/immunology , Lymphatic Vessels/pathology , Male , Mice , Mice, Transgenic , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism
6.
Gerontology ; 66(5): 419-426, 2020.
Article in English | MEDLINE | ID: mdl-32580201

ABSTRACT

The lymphatic system is responsible for fluid drainage from almost every organ in the body. It sustains tissue homeostasis and is also a central part of the immune system. With the discovery of cell-specific markers and transgenic mouse models, it has become possible to gain some insight into the developmental and functional roles of lymphatic endothelial cells (LECs). Only recently, a more direct regulatory role has been assigned to LECs in their functions in immunity responses and chronic diseases. Here, we discuss the changes occurring in aged lymphatic system and the role of lymphatic capillaries in some age-related diseases and experimental animal models.


Subject(s)
Aging/immunology , Lymphatic Vessels/immunology , Animals , Endothelial Cells/immunology , Humans , Mice
7.
Clin Otolaryngol ; 43(5): 1335-1344, 2018 10.
Article in English | MEDLINE | ID: mdl-29992788

ABSTRACT

BACKGROUND: The purpose of this study was to determine whether the expression of 15-lipoxygenase-1 (ALOX15) in primary tumour specimens predicts lymph node metastasis and subsequently clinical outcome in Merkel cell carcinoma (MCC) patients. METHODS: A retrospective medical chart review of 33 patients was performed between 1994 and 2014. Eleven out of 33 (33%) Patients with primary MCC stages I and II were categorised as group I. Twenty two out of 33 (67%) Patients with regional lymph node metastases and/or distant metastases were defined as group II. All available tumour samples were immunostained for ALOX15, Podoplanin and MCPyV large T-protein antibody. RESULTS: ALOX15 expression was observed in 19/23 (83%) primary tumour samples and in all lymph node metastasis. Primary tumours in patients with stage III and IV disease showed a higher expression rate of ALOX15 compared to patients with early stage disease (11/12 (92%) and 8/11 (73%), respectively). In group I, five patients (45%) were MCPyV positive, whereas in group II, 15 patients (68%) were MCPyV positive. The median lymphatic vessel density in ALOX15 negative group I primary tumour samples was lower compared to the median lymphatic vessel density in ALOX15 positive group I primary tumour probes (2.7 range, 1-4.3 vs 4.7 range, 4.0-7.3). Furthermore, all 17 samples of MCC metastases showed ALOX15 expression with a median lymphatic vessel density (not lymph node metastases) of 5.3 (range 2.0-7.3). CONCLUSION: In the current study, we were able to show ALOX15 expression in the primary MCC sample and the metastasis sample. Based on the findings of the current study, expression rate of ALOX15 in primary MCC and metastases is possibly linked to an increased lymphatic vessel density.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Carcinoma, Merkel Cell/metabolism , Carcinoma, Merkel Cell/secondary , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Aged , Aged, 80 and over , Biomarkers/metabolism , Carcinoma, Merkel Cell/mortality , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Predictive Value of Tests , Retrospective Studies , Skin Neoplasms/mortality , Survival Rate
8.
Am J Pathol ; 186(5): 1128-39, 2016 05.
Article in English | MEDLINE | ID: mdl-27105734

ABSTRACT

Mitochondrial fusion is essential for maintenance of mitochondrial function and requires the prohibitin ring complex subunit prohibitin-2 (PHB2) at the mitochondrial inner membrane. Loss of the stomatin/PHB/flotillin/HflK/C (SPFH) domain containing protein PHB2 causes mitochondrial dysfunction and defective mitochondria-mediated signaling, which is implicated in a variety of human diseases, including progressive renal disease. Here, we provide evidence of additional, extra-mitochondrial functions of this membrane-anchored protein. Immunofluorescence and immunogold labeling detected PHB2 at mitochondrial membranes and at the slit diaphragm, a specialized cell junction at the filtration slit of glomerular podocytes. PHB2 coprecipitated with podocin, another SPFH domain-containing protein, essential for the assembly of the slit diaphragm protein-lipid supercomplex. Consistent with an evolutionarily conserved extra-mitochondrial function, the ortholog of PHB2 in Caenorhabditis elegans was also not restricted to mitochondria but colocalized with the mechanosensory complex that requires the podocin ortholog MEC2 for assembly. Knockdown of phb-2 partially phenocopied loss of mec-2 in touch neurons of the nematode, resulting in impaired gentle touch sensitivity. Collectively, these data indicate that, besides its established role in mitochondria, PHB2 may have an additional function in conserved protein-lipid complexes at the plasma membrane.


Subject(s)
Mitochondria/physiology , Podocytes/physiology , Repressor Proteins/deficiency , Animals , Caenorhabditis elegans Proteins , Cells, Cultured , HEK293 Cells , Humans , Intercellular Junctions/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Kidney/physiology , Mechanoreceptors/physiology , Mechanotransduction, Cellular/physiology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Mitochondria/ultrastructure , Mitochondrial Diseases/etiology , Mitochondrial Diseases/physiopathology , Mitochondrial Membranes/physiology , Mitochondrial Membranes/ultrastructure , Podocytes/ultrastructure , Prohibitins , Proteinuria/etiology , Proteinuria/physiopathology , Touch/physiology
9.
Proc Natl Acad Sci U S A ; 111(27): E2817-26, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-24958889

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) is a key regulator of cell metabolism and autophagy. Despite widespread clinical use of mTORC1 inhibitors, the role of mTORC1 in renal tubular function and kidney homeostasis remains elusive. By using constitutive and inducible deletion of conditional Raptor alleles in renal tubular epithelial cells, we discovered that mTORC1 deficiency caused a marked concentrating defect, loss of tubular cells, and slowly progressive renal fibrosis. Transcriptional profiling revealed that mTORC1 maintains renal tubular homeostasis by controlling mitochondrial metabolism and biogenesis as well as transcellular transport processes involved in countercurrent multiplication and urine concentration. Although mTORC2 partially compensated for the loss of mTORC1, exposure to ischemia and reperfusion injury exaggerated the tubular damage in mTORC1-deficient mice and caused pronounced apoptosis, diminished proliferation rates, and delayed recovery. These findings identify mTORC1 as an important regulator of tubular energy metabolism and as a crucial component of ischemic stress responses.


Subject(s)
Homeostasis/physiology , Ischemia/physiopathology , Kidney Tubules/physiology , Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Blotting, Western , Kidney Tubules/blood supply , Magnetic Resonance Imaging , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/genetics , Polyuria/genetics , TOR Serine-Threonine Kinases/genetics , Transcription, Genetic
10.
J Am Soc Nephrol ; 27(11): 3266-3270, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27324941

ABSTRACT

This overview summarizes selected major developments over the last 40 years in understanding podocyte biology and its involvement in glomerular disease subjectively from my perspective. Serendipity has played a major role in my contributions to investigative nephrology that range from basic mechanisms of immune deposit formation in experimental membranous nephropathy to the role of a microRNA in FSGS. This review emphasizes the importance of continuous reality checks of experimental results obtained in vitro or with genetically modified animals with human disease.


Subject(s)
Awards and Prizes , Podocytes/physiology , Animals , Glomerulonephritis, Membranous/etiology , Humans , MicroRNAs/physiology
11.
J Am Soc Nephrol ; 27(4): 1042-54, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26260163

ABSTRACT

Hereditary angiopathy, nephropathy, aneurysms, and muscle cramps (HANAC) syndrome is an autosomal dominant syndrome caused by mutations in COL4A1 that encodes the α1 chain of collagen IV, a major component of basement membranes. Patients present with cerebral small vessel disease, retinal tortuosity, muscle cramps, and kidney disease consisting of multiple renal cysts, chronic kidney failure, and sometimes hematuria. Mutations producing HANAC syndrome localize within the integrin binding site containing CB3[IV] fragment of the COL4A1 protein. To investigate the pathophysiology of HANAC syndrome, we generated mice harboring the Col4a1 p.Gly498Val mutation identified in a family with the syndrome. Col4a1 G498V mutation resulted in delayed glomerulogenesis and podocyte differentiation without reduction of nephron number, causing albuminuria and hematuria in newborns. The glomerular defects resolved within the first month, but glomerular cysts developed in 3-month-old mutant mice. Abnormal structure of Bowman's capsule was associated with metalloproteinase induction and activation of the glomerular parietal epithelial cells that abnormally expressed CD44,α-SMA, ILK, and DDR1. Inflammatory infiltrates were observed around glomeruli and arterioles. Homozygous Col4a1 G498V mutant mice additionally showed dysmorphic papillae and urinary concentration defects. These results reveal a developmental role for the α1α1α2 collagen IV molecule in the embryonic glomerular basement membrane, affecting podocyte differentiation. The observed association between molecular alteration of the collagenous network in Bowman's capsule of the mature kidney and activation of parietal epithelial cells, matrix remodeling, and inflammation may account for glomerular cyst development and CKD in patients with COL4A1-related disorders.


Subject(s)
Collagen Type IV/genetics , Kidney Diseases, Cystic/etiology , Muscle Cramp/complications , Muscle Cramp/genetics , Mutation , Raynaud Disease/complications , Raynaud Disease/genetics , Age Factors , Animals , Animals, Newborn , Kidney Diseases, Cystic/metabolism , Kidney Glomerulus/metabolism , Mice , Muscle Cramp/metabolism , Muscle Cramp/physiopathology , Permeability , Raynaud Disease/metabolism , Raynaud Disease/physiopathology
12.
Am J Physiol Renal Physiol ; 311(1): F112-9, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27122542

ABSTRACT

Polarity signaling through the atypical PKC (aPKC)-Par polarity complex is essential for the development and maintenance of the podocyte architecture and the function of the glomerular filtration barrier of the kidney. To study the contribution of Par3A in this complex, we generated a novel Pard3 podocyte-specific knockout mouse model by targeting exon 6 of the Pard3 gene. Genetic deletion of Pard3a did not impair renal function, neither at birth nor later in life. Even challenging the animals did not result in glomerular disease. Despite its well-established role in aPKC-mediated signaling, Par3A appears to be dispensable for the function of the glomerular filtration barrier. Moreover, its homolog Pard3b, and not Pard3a, is the dominant Par3 gene expressed in podocytes and found at the basis of the slit diaphragm, where it partially colocalizes with podocin. In conclusion, Par3A function is either dispensable for slit diaphragm integrity, or compensatory mechanisms and a high redundancy of the different polarity proteins, including Par3B, Lgl, or PALS1, maintain the function of the glomerular filtration barrier, even in the absence of Par3A.


Subject(s)
Cell Adhesion Molecules/metabolism , Glomerular Filtration Barrier/physiology , Kidney/physiology , Adaptor Proteins, Signal Transducing , Animals , Cell Adhesion Molecules/genetics , Cell Cycle Proteins , Cells, Cultured , Female , Kidney/pathology , Lipopolysaccharides/toxicity , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nucleoside-Phosphate Kinase/genetics , Nucleoside-Phosphate Kinase/metabolism , Podocytes/drug effects , Podocytes/metabolism , Podocytes/pathology , Primary Cell Culture , Serum Albumin, Bovine/toxicity
13.
Am J Physiol Heart Circ Physiol ; 311(4): H981-H990, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27496878

ABSTRACT

Chronic allograft fibrosis is the major cause of graft loss in kidney transplantation. Progression can only be reduced by inhibition of the renin-angiotensin system (RAS). We tested the hypothesis that the protection provided by angiotensin-converting enzyme (ACE) inhibition also decreases capillary rarefaction, lymphangiogenesis, and podocyte injury in allograft fibrosis. Fisher kidneys were transplanted into bilaterally nephrectomized Lewis rats treated with enalapril (60 mg/kg per day) (ACE inhibitor, ACEi) or vehicle. Proteinuria, blood urea nitrogen, and plasma creatinine were regularly assessed, and grafts were harvested for morphological and immunohistological analysis at various times up to 32 wk. In the vehicle group, many new lymphatic capillaries and severe and diffuse mononuclear infiltration of allografts were observed already 1 wk after transplantation. Lymphangiogenesis increased until week 4, by which time inflammatory infiltration became focal. Lymphatic capillaries were often located at sites of inflammation. Progressive interstitial fibrosis, glomerulosclerosis, capillary rarefaction, and proteinuria appeared later, at weeks 4-12 The number of lymphatic capillary cross sections strongly correlated with the interstitial fibrosis score. Podoplanin immunostaining, a marker of healthy podocytes, disappeared from inflamed or sclerotic glomerular areas. ACEi protected from lymphangiogenesis and associated inflammation, preserved glomerular podoplanin protein expression, and reduced glomerulosclerosis, proteinuria, tubulointerstitial fibrosis, and blood capillary rarefaction at 32 wk. In conclusion, ACEi considerably decreased and/or delayed both glomerulosclerosis and tubulointerstitial injury. Prevention of glomerular podoplanin loss and proteinuria could be attributed to the known intraglomerular pressure-lowering effects of ACEi. Reduction of lymphangiogenesis could contribute to amelioration of tubulointerstitial fibrosis and inflammatory infiltration after ACEi.


Subject(s)
Allografts/drug effects , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Capillaries/drug effects , Enalapril/pharmacology , Kidney Transplantation , Kidney/drug effects , Lymphangiogenesis/drug effects , Lymphatic Vessels/drug effects , Allografts/pathology , Animals , Capillaries/pathology , Fibrosis , Kidney/pathology , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Lymphatic Vessels/pathology , Male , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/metabolism , Podocytes/drug effects , Podocytes/metabolism , Proteinuria , Rats , Rats, Inbred F344 , Rats, Inbred Lew
14.
J Am Soc Nephrol ; 26(6): 1389-401, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25270065

ABSTRACT

Parietal epithelial cells have been identified as potential progenitor cells in glomerular regeneration, but the molecular mechanisms underlying this process are not fully defined. Here, we established an immortalized polyclonal human parietal epithelial cell (hPEC) line from naive human Bowman's capsule cells isolated by mechanical microdissection. These hPECs expressed high levels of PEC-specific proteins and microRNA-193a (miR-193a), a suppressor of podocyte differentiation through downregulation of Wilms' tumor 1 in mice. We then investigated the function of miR-193a in the establishment of podocyte and PEC identity and determined whether inhibition of miR-193a influences the behavior of PECs in glomerular disease. After stable knockdown of miR-193a, hPECs adopted a podocyte-like morphology and marker expression, with decreased expression levels of PEC markers. In mice, inhibition of miR-193a by complementary locked nucleic acids resulted in an upregulation of the podocyte proteins synaptopodin and Wilms' tumor 1. Conversely, overexpression of miR-193a in vivo resulted in the upregulation of PEC markers and the loss of podocyte markers in isolated glomeruli. Inhibition of miR-193a in a mouse model of nephrotoxic nephritis resulted in reduced crescent formation and decreased proteinuria. Together, these results show the establishment of a human PEC line and suggest that miR-193a functions as a master switch, such that glomerular epithelial cells with high levels of miR-193a adopt a PEC phenotype and cells with low levels of miR-193a adopt a podocyte phenotype. miR-193a-mediated maintenance of PECs in an undifferentiated reactive state might be a prerequisite for PEC proliferation and migration in crescent formation.


Subject(s)
Cell Transdifferentiation/genetics , Gene Expression Regulation , Glomerulosclerosis, Focal Segmental/genetics , MicroRNAs/genetics , Podocytes/metabolism , Animals , Bowman Capsule/cytology , Cells, Cultured , Disease Models, Animal , Epithelial Cells/metabolism , Fluorescent Antibody Technique , Glomerulosclerosis, Focal Segmental/physiopathology , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/physiopathology , Mice , Mice, Transgenic , Phenotype , Polymerase Chain Reaction/methods , Random Allocation , Statistics, Nonparametric
15.
J Biol Chem ; 289(25): 17454-67, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24817115

ABSTRACT

Podocytes are highly differentiated cells and critical elements for the filtration barrier of the kidney. Loss of their foot process (FP) architecture (FP effacement) results in urinary protein loss. Here we show a novel role for the neutral amino acid glutamine in structural and functional regulation of the kidney filtration barrier. Metabolic flux analysis of cultured podocytes using genetic, toxic, and immunologic injury models identified increased glutamine utilization pathways. We show that glutamine uptake is increased in diseased podocytes to couple nutrient support to increased demand during the disease state of FP effacement. This feature can be utilized to transport increased amounts of glutamine into damaged podocytes. The availability of glutamine determines the regulation of podocyte intracellular pH (pHi). Podocyte alkalinization reduces cytosolic cathepsin L protease activity and protects the podocyte cytoskeleton. Podocyte glutamine supplementation reduces proteinuria in LPS-treated mice, whereas acidification increases glomerular injury. In summary, our data provide a metabolic opportunity to combat urinary protein loss through modulation of podocyte amino acid utilization and pHi.


Subject(s)
Podocytes/metabolism , Proteinuria/metabolism , Animals , Biological Transport, Active/genetics , Biological Transport, Active/immunology , Cells, Cultured , Cytoskeleton/genetics , Cytoskeleton/immunology , Cytoskeleton/metabolism , Cytoskeleton/pathology , Hydrogen-Ion Concentration , Mice , Mice, Knockout , Podocytes/immunology , Podocytes/pathology , Proteinuria/genetics , Proteinuria/immunology , Proteinuria/pathology
16.
Am J Pathol ; 184(6): 1727-39, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24726496

ABSTRACT

Diabetic nephropathy is a complication of diabetes and a major cause of end-stage renal disease. To characterize the early pathophysiological mechanisms leading to glomerular podocyte injury in diabetic nephropathy, we performed quantitative proteomic profiling of glomeruli isolated from rats with streptozotocin-induced diabetes and controls. Fluorescence-based two-dimensional difference gel electrophoresis, coupled with mass spectrometry, identified 29 differentially expressed spots, including actin-binding protein ezrin and its interaction partner, NHERF2, which were down-regulated in the streptozotocin group. Knockdown of ezrin by siRNA in cultured podocytes increased glucose uptake compared with control siRNA-transfected cells, apparently by increasing translocation of glucose transporter GLUT1 to the plasma membrane. Knockdown of ezrin also induced actin remodeling under basal conditions, but reduced insulin-stimulated actin reorganization. Ezrin-dependent actin remodeling involved cofilin-1 that is essential for the turnover and reorganization of actin filaments. Phosphorylated, inactive cofilin-1 was up-regulated in diabetic glomeruli, suggesting altered actin dynamics. Furthermore, IHC analysis revealed reduced expression of ezrin in the podocytes of patients with diabetes. Our findings suggest that ezrin may play a role in the development of the renal complication in diabetes by regulating transport of glucose and organization of the actin cytoskeleton in podocytes.


Subject(s)
Actin Cytoskeleton/metabolism , Cytoskeletal Proteins/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Glucose Transporter Type 1/metabolism , Glucose/metabolism , Podocytes/metabolism , Actin Cytoskeleton/pathology , Actins/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/pathology , Down-Regulation , Gene Knockdown Techniques , Male , Rats , Rats, Sprague-Dawley
17.
Nat Genet ; 38(12): 1397-405, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17086182

ABSTRACT

Nephrotic syndrome, a malfunction of the kidney glomerular filter, leads to proteinuria, edema and, in steroid-resistant nephrotic syndrome, end-stage kidney disease. Using positional cloning, we identified mutations in the phospholipase C epsilon gene (PLCE1) as causing early-onset nephrotic syndrome with end-stage kidney disease. Kidney histology of affected individuals showed diffuse mesangial sclerosis (DMS). Using immunofluorescence, we found PLCepsilon1 expression in developing and mature glomerular podocytes and showed that DMS represents an arrest of normal glomerular development. We identified IQ motif-containing GTPase-activating protein 1 as a new interaction partner of PLCepsilon1. Two siblings with a missense mutation in an exon encoding the PLCepsilon1 catalytic domain showed histology characteristic of focal segmental glomerulosclerosis. Notably, two other affected individuals responded to therapy, making this the first report of a molecular cause of nephrotic syndrome that may resolve after therapy. These findings, together with the zebrafish model of human nephrotic syndrome generated by plce1 knockdown, open new inroads into pathophysiology and treatment mechanisms of nephrotic syndrome.


Subject(s)
Mutation , Nephrotic Syndrome/enzymology , Nephrotic Syndrome/genetics , Type C Phospholipases/genetics , Animals , Child , Child, Preschool , Cloning, Molecular , Disease Models, Animal , Female , Gene Targeting , Genes, Recessive , Homozygote , Humans , Infant , Kidney/enzymology , Kidney/pathology , Male , Models, Genetic , Mutation, Missense , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/pathology , Phosphoinositide Phospholipase C , Rats , Sequence Deletion , Zebrafish/genetics
18.
J Biol Chem ; 288(51): 36598-609, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24194522

ABSTRACT

Gain-of-function mutations in the calcium channel TRPC6 lead to autosomal dominant focal segmental glomerulosclerosis and podocyte expression of TRPC6 is increased in some acquired human glomerular diseases, particularly in membranous nephropathy. These observations led to the hypothesis that TRPC6 overactivation is deleterious to podocytes through pathological calcium signaling, both in genetic and acquired diseases. Here, we show that the effects of TRPC6 on podocyte function are context-dependent. Overexpression of TRPC6 alone did not directly affect podocyte morphology and cytoskeletal structure. Unexpectedly, however, overexpression of TRPC6 protected podocytes from complement-mediated injury, whereas genetic or pharmacological TRPC6 inactivation increased podocyte susceptibility to complement. Mechanistically, this effect was mediated by Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) activation. Podocyte-specific TRPC6 transgenic mice showed stronger CaMKII activation, reduced podocyte foot process effacement and reduced levels of proteinuria during nephrotoxic serum nephritis, whereas TRPC6 null mice exhibited reduced CaMKII activation and higher levels of proteinuria compared with wild type littermates. Human membranous nephropathy biopsy samples showed podocyte staining for active CaMKII, which correlated with the degree of TRPC6 expression. Together, these data suggest a dual and context dependent role of TRPC6 in podocytes where acute activation protects from complement-mediated damage, but chronic overactivation leads to focal segmental glomerulosclerosis.


Subject(s)
Complement System Proteins/metabolism , Glomerulosclerosis, Focal Segmental/metabolism , Podocytes/metabolism , TRPC Cation Channels/metabolism , Animals , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Enzyme Activation , Glomerulonephritis, Membranous/metabolism , Glomerulonephritis, Membranous/pathology , Glomerulosclerosis, Focal Segmental/pathology , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Podocytes/pathology , Proteinuria/metabolism , TRPC Cation Channels/genetics , TRPC6 Cation Channel
19.
PLoS Pathog ; 8(7): e1002811, 2012.
Article in English | MEDLINE | ID: mdl-22911155

ABSTRACT

Invasive fungal infections by Candida albicans (Ca) are a frequent cause of lethal sepsis in intensive care unit patients. While a contribution of type I interferons (IFNs-I) in fungal sepsis remains unknown, these immunostimulatory cytokines mediate the lethal effects of endotoxemia and bacterial sepsis. Using a mouse model lacking a functional IFN-I receptor (Ifnar1⁻/⁻), we demonstrate a remarkable protection against invasive Ca infections. We discover a mechanism whereby IFN-I signaling controls the recruitment of inflammatory myeloid cells, including Ly6C(hi) monocytes and neutrophils, to infected kidneys by driving expression of the chemokines CCL2 and KC. Within kidneys, monocytes differentiate into inflammatory DCs but fail to functionally mature in Ifnar1⁻/⁻ mice, as demonstrated by the impaired upregulation of the key activation markers PDCA1 and iNOS. The increased activity of inflammatory monocytes and neutrophils results in hyper-inflammation and lethal kidney pathology. Pharmacological diminution of monocytes and neutrophils by treating mice with pioglitazone, a synthetic agonist of the nuclear receptor peroxisome proliferator-activated receptor-γ (PPAR-γ), strongly reduces renal immunopathology during Ca infection and improves mouse survival. Taken together, our data connect for the first time the sepsis-promoting functions of IFNs-I to the CCL2-mediated recruitment and the activation of inflammatory monocytes/DCs with high host-destructing potency. Moreover, our data demonstrate a therapeutic relevance of PPAR-γ agonists for microbial infectious diseases where inflammatory myeloid cells may contribute to fatal tissue damage.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Interferon Type I/metabolism , Monocytes/immunology , Neutrophils/immunology , Animals , Antigens, CD/biosynthesis , Antigens, Ly/biosynthesis , Candidemia/mortality , Candidiasis/pathology , Chemokine CCL2/biosynthesis , Chemokine CXCL1/biosynthesis , Dendritic Cells/immunology , Inflammation/drug therapy , Inflammation/immunology , Kidney/immunology , Kidney/microbiology , Male , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Neutrophils/drug effects , Nitric Oxide Synthase Type II/biosynthesis , PPAR gamma/agonists , Pioglitazone , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Signal Transduction/genetics , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use
20.
Cancer Cell ; 9(4): 261-72, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16616332

ABSTRACT

The expression of podoplanin, a small mucin-like protein, is upregulated in the invasive front of a number of human carcinomas. We have investigated podoplanin function in cultured human breast cancer cells, in a mouse model of pancreatic beta cell carcinogenesis, and in human cancer biopsies. Our results indicate that podoplanin promotes tumor cell invasion in vitro and in vivo. Notably, the expression and subcellular localization of epithelial markers are unaltered, and mesenchymal markers are not induced in invasive podoplanin-expressing tumor cells. Rather, podoplanin induces collective cell migration by filopodia formation via the downregulation of the activities of small Rho family GTPases. In conclusion, podoplanin induces an alternative pathway of tumor cell invasion in the absence of epithelial-mesenchymal transition (EMT).


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Epithelial Cells/pathology , Membrane Glycoproteins/metabolism , Mesoderm/pathology , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , DNA-Binding Proteins/metabolism , Disease Progression , Down-Regulation , Epithelial Cells/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Leukemia, B-Cell/metabolism , Leukemia, B-Cell/pathology , Mesoderm/metabolism , Mice , Mice, Transgenic , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Organic Cation Transport Proteins/metabolism , Pseudopodia/metabolism , Transcription Factors/metabolism , cdc42 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL