Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Int J Mol Sci ; 25(8)2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38673910

ABSTRACT

Endothelial cell (EC) injury is a crucial contributor to the progression of diabetic kidney disease (DKD), but the specific EC populations and mechanisms involved remain elusive. Kidney ECs (n = 5464) were collected at three timepoints from diabetic BTBRob/ob mice and non-diabetic littermates. Their heterogeneity, transcriptional changes, and alternative splicing during DKD progression were mapped using SmartSeq2 single-cell RNA sequencing (scRNAseq) and elucidated through pathway, network, and gene ontology enrichment analyses. We identified 13 distinct transcriptional EC phenotypes corresponding to different kidney vessel subtypes, confirmed through in situ hybridization and immunofluorescence. EC subtypes along nephrons displayed extensive zonation related to their functions. Differential gene expression analyses in peritubular and glomerular ECs in DKD underlined the regulation of DKD-relevant pathways including EIF2 signaling, oxidative phosphorylation, and IGF1 signaling. Importantly, this revealed the differential alteration of these pathways between the two EC subtypes and changes during disease progression. Furthermore, glomerular and peritubular ECs also displayed aberrant and dynamic alterations in alternative splicing (AS), which is strongly associated with DNA repair. Strikingly, genes displaying differential transcription or alternative splicing participate in divergent biological processes. Our study reveals the spatiotemporal regulation of gene transcription and AS linked to DKD progression, providing insight into pathomechanisms and clues to novel therapeutic targets for DKD treatment.


Subject(s)
Alternative Splicing , Diabetic Nephropathies , Endothelial Cells , Single-Cell Analysis , Transcriptome , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Mice , Single-Cell Analysis/methods , Endothelial Cells/metabolism , Endothelial Cells/pathology , Kidney/metabolism , Kidney/pathology , Gene Expression Regulation , Transcription, Genetic , Gene Expression Profiling/methods , Male
2.
Biomedicines ; 10(6)2022 Jun 06.
Article in English | MEDLINE | ID: mdl-35740360

ABSTRACT

Hypercoagulation and endothelial dysfunction play central roles in severe forms of COVID-19 infections, but the molecular mechanisms involved are unclear. Increased plasma levels of the inflammatory cytokine and TIE2 receptor antagonist Angiopoietin-2 were reported in severely ill COVID-19 patients. In vitro experiments suggest that Angiopoietin-2 bind and inhibits thrombomodulin. Thrombomodulin is expressed on the luminal surface of endothelial cells where it is an important member of the intrinsic anticoagulant pathway through activation of protein C. Using clinical data, mouse models, and in vitro assays, we tested if Angiopoietin-2 plays a causal role in COVID-19-associated hypercoagulation through direct inhibition of thrombin/thrombomodulin-mediated physiological anticoagulation. Angiopoietin-2 was measured in 61 patients at admission, and after 10 days in the 40 patients remaining in the ICU. We found that Angiopoietin-2 levels were increased in COVID-19 patients in correlation with disease severity, hypercoagulation, and mortality. In support of a direct effect of Angiopoietin-2 on coagulation, we found that injected Angiopoietin-2 in mice associated to thrombomodulin and resulted in a shortened tail bleeding time, decreased circulating levels of activated protein C, and increased plasma thrombin/antithrombin complexes. Conversely, bleeding time was increased in endothelial-specific Angiopoietin-2 knockout mice, while knockout of Tie2 had no effect on tail bleeding. Using in vitro assays, we found that Angiopoietin-2 inhibited thrombomodulin-mediated anticoagulation and protein C activation in human donor plasma. Our data suggest a novel in vivo mechanism for Angiopoietin-2 in COVID-19-associated hypercoagulation, implicating that Angiopoietin-2 inhibitors may be effective in the treatment of hypercoagulation in severe COVID-19 infection.

3.
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
4.
Circ Res ; 128(4): e46-e62, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33375813

ABSTRACT

RATIONALE: Pericytes are capillary mural cells playing a role in stabilizing newly formed blood vessels during development and tissue repair. Loss of pericytes has been described in several brain disorders, and genetically induced pericyte deficiency in the brain leads to increased macromolecular leakage across the blood-brain barrier (BBB). However, the molecular details of the endothelial response to pericyte deficiency remain elusive. OBJECTIVE: To map the transcriptional changes in brain endothelial cells resulting from lack of pericyte contact at single-cell level and to correlate them with regional heterogeneities in BBB function and vascular phenotype. METHODS AND RESULTS: We reveal transcriptional, morphological, and functional consequences of pericyte absence for brain endothelial cells using a combination of methodologies, including single-cell RNA sequencing, tracer analyses, and immunofluorescent detection of protein expression in pericyte-deficient adult Pdgfbret/ret mice. We find that endothelial cells without pericyte contact retain a general BBB-specific gene expression profile, however, they acquire a venous-shifted molecular pattern and become transformed regarding the expression of numerous growth factors and regulatory proteins. Adult Pdgfbret/ret brains display ongoing angiogenic sprouting without concomitant cell proliferation providing unique insights into the endothelial tip cell transcriptome. We also reveal heterogeneous modes of pericyte-deficient BBB impairment, where hotspot leakage sites display arteriolar-shifted identity and pinpoint putative BBB regulators. By testing the causal involvement of some of these using reverse genetics, we uncover a reinforcing role for angiopoietin 2 at the BBB. CONCLUSIONS: By elucidating the complexity of endothelial response to pericyte deficiency at cellular resolution, our study provides insight into the importance of brain pericytes for endothelial arterio-venous zonation, angiogenic quiescence, and a limited set of BBB functions. The BBB-reinforcing role of ANGPT2 (angiopoietin 2) is paradoxical given its wider role as TIE2 (TEK receptor tyrosine kinase) receptor antagonist and may suggest a unique and context-dependent function of ANGPT2 in the brain.


Subject(s)
Blood-Brain Barrier/metabolism , Pericytes/cytology , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/pathology , Cell Proliferation , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/physiology , Lymphokines/deficiency , Lymphokines/genetics , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic , Pericytes/metabolism , Pericytes/pathology , Platelet-Derived Growth Factor/deficiency , Platelet-Derived Growth Factor/genetics , Single-Cell Analysis , Transcriptome
5.
PLoS One ; 13(1): e0189433, 2018.
Article in English | MEDLINE | ID: mdl-29293543

ABSTRACT

Presence of tubulointerstitial fibrosis is predictive of progressive decline in kidney function, independent of its underlying cause. Injury to the renal microvasculature is a major factor in the progression of fibrosis and identification of factors that regulate endothelium in fibrosis is desirable as they might be candidate targets for treatment of kidney diseases. The current study investigates how loss of Angipoietin-1 (Angpt1), a ligand for endothelial tyrosine-kinase receptor Tek (also called Tie2), affects tubulointerstitial fibrosis and renal microvasculature. Inducible Angpt1 knockout mice were subjected to unilateral ureteral obstruction (UUO) to induce fibrosis, and kidneys were collected at different time points up to 10 days after obstruction. Staining for aSMA showed that Angpt1 deficient kidneys had significantly more fibrosis compared to wildtype mice 3, 6, and 10 days after UUO. Further investigation 3 days after UUO showed a significant increase of Col1a1 and vimentin in Angpt1 deficient mice, as well as increased gene expression of Tgfb1, Col1a1, Fn1, and CD44. Kidney injury molecule 1 (Kim1/Havcr1) was significantly more increased in Angpt1 deficient mice 1 and 3 days after UUO, suggesting a more severe injury early in the fibrotic process in Angpt1 deficient mice. Staining for endomucin showed that capillary rarefaction was evident 3 days after UUO and Angpt1 deficient mice had significantly less capillaries 6 and 10 days after UUO compared to UUO kidneys in wildtype mice. RNA sequencing revealed downregulation of several markers for endothelial cells 3 days after UUO, and that Angpt1 deficient mice had a further downregulation of Emcn, Plvap, Pecam1, Erg, and Tek. Our results suggest that loss of Angpt1 is central in capillary rarefaction and fibrogenesis and propose that manipulations to maintain Angpt1 levels may slow down fibrosis progression.


Subject(s)
Angiopoietin-1/physiology , Capillaries/physiopathology , Kidney/blood supply , Nephritis, Interstitial/genetics , Angiopoietin-1/genetics , Animals , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction
6.
BMC Cancer ; 17(1): 539, 2017 Aug 11.
Article in English | MEDLINE | ID: mdl-28800750

ABSTRACT

BACKGROUND: Angipoietin-1 activation of the tyrosine kinase receptor Tek expressed mainly on endothelial cells leads to survival and stabilization of endothelial cells. Studies have shown that Angiopoietin-1 counteracts permeability induced by a number of stimuli. Here, we test the hypothesis that loss of Angiopoietin-1/Tek signaling in the vasculature would increase metastasis. METHODS: Angiopoietin-1 was deleted in mice just before birth using floxed Angiopoietin-1 and Tek mice crossed to doxycycline-inducible bitransgenic ROSA-rtTA/tetO-Cre mice. By crossing Angiopoietin-1 knockout mice to the MMTV-PyMT autochthonous mouse breast cancer model, we investigated primary tumor growth and metastasis to the lung. Furthermore, we utilized B16F10 melanoma cells subcutaneous and experimental lung metastasis models in Angiopoietin-1 and Tek knockout mice. RESULTS: We found that primary tumor growth in MMTV-PyMT mice was unaffected, while metastasis to the lung was significantly increased in Angiopoietin-1 knockout MMTV-PyMT mice. In addition, angiopoietin-1 deficient mice exhibited a significant increase in lung metastasis of B16F10 melanoma cells, compared to wild type mice 3 weeks after injection. Additional experiments showed that this was likely an early event due to increased attachment or extravasation of tumor cells, since seeding of tumor cells was significantly increased 4 and 24 h post tail vein injection. Finally, using inducible Tek knockout mice, we showed a significant increase in tumor cell seeding to the lung, suggesting that Angiopoietin-1/Tek signaling is important for vascular integrity to limit metastasis. CONCLUSIONS: This study show that loss of the Angiopoietin-1/Tek vascular growth factor system leads to increased metastasis without affecting primary tumor growth.


Subject(s)
Angiopoietin-1/genetics , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Melanoma/pathology , Signal Transduction , Angiopoietin-1/metabolism , Animals , Female , Gene Expression , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Knockout , Neoplasm Metastasis/genetics
7.
Eur J Pharm Biopharm ; 114: 186-193, 2017 May.
Article in English | MEDLINE | ID: mdl-28161551

ABSTRACT

BACKGROUND: Docetaxel is a cytostatic agent approved for treatment of non-small cell lung cancer as well as other cancers. Although docetaxel is an effective cytostatic agent, its effectiveness in clinical practice is associated with a variety of acute and long term side-effects. To overcome systemic side-effects, a slow release formulation based on calcium sulfate with docetaxel for intra-tumoral administration was developed. METHODS: Two formulations with the calcium sulfate NanoZolid technology were generated with a twofold difference in docetaxel drug load. The formulations were injected intra-tumorally as a paste which solidified within the tumor. The effects of the two intra-tumoral injection formulations were tested in female mice (n=60) inoculated with subcutaneous Lewis lung carcinoma cells. The two formulations were compared to systemic intraperitoneal injection of docetaxel and a placebo formulation without docetaxel. Tumor volumes were measured and systemic side-effects were evaluated using body weight and cell counts from whole blood as well as plasma concentrations. RESULTS: Both docetaxel formulations showed a significantly higher antitumor efficacy compared to placebo, which was comparable to that of systemic administration of docetaxel. Moreover, the intra-tumoral formulations with docetaxel showed reduced systemic toxicity compared to systemic treatment, including less weight loss and no decrease in blood cell counts. CONCLUSIONS: The results suggest that intra-tumoral slow release calcium sulfate based formulations with docetaxel can be an alternative strategy as an efficient local antitumoral treatment with reduced systemic toxicity.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Calcium Sulfate/chemistry , Taxoids/administration & dosage , Taxoids/pharmacology , Animals , Antineoplastic Agents, Phytogenic/toxicity , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/pathology , Docetaxel , Drug Compounding , Excipients , Female , Humans , Injections, Intralesional , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Taxoids/toxicity
8.
Annu Rev Physiol ; 78: 437-61, 2016.
Article in English | MEDLINE | ID: mdl-26863327

ABSTRACT

The glomerulus is a highly specialized microvascular bed that filters blood to form primary urinary filtrate. It contains four cell types: fenestrated endothelial cells, specialized vascular support cells termed podocytes, perivascular mesangial cells, and parietal epithelial cells. Glomerular cell-cell communication is critical for the development and maintenance of the glomerular filtration barrier. VEGF, ANGPT, EGF, SEMA3A, TGF-ß, and CXCL12 signal in paracrine fashions between the podocytes, endothelium, and mesangium associated with the glomerular capillary bed to maintain filtration barrier function. In this review, we summarize the current understanding of these signaling pathways in the development and maintenance of the glomerulus and the progression of disease.


Subject(s)
Capillaries/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Diseases/metabolism , Kidney Glomerulus/metabolism , Animals , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Humans , Signal Transduction/physiology
9.
J Clin Invest ; 124(10): 4320-4, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25202984

ABSTRACT

Glaucoma is a leading cause of blindness, afflicting more than 60 million people worldwide. Increased intraocular pressure (IOP) due to impaired aqueous humor drainage is a major risk factor for the development of glaucoma. Here, we demonstrated that genetic disruption of the angiopoietin/TIE2 (ANGPT/TIE2) signaling pathway results in high IOP, buphthalmos, and classic features of glaucoma, including retinal ganglion degeneration and vision loss. Eyes from mice with induced deletion of Angpt1 and Angpt2 (A1A2Flox(WB) mice) lacked drainage pathways in the corneal limbus, including Schlemm's canal and lymphatic capillaries, which share expression of the PROX1, VEGFR3, and FOXC family of transcription factors. VEGFR3 and FOXCs have been linked to lymphatic disorders in patients, and FOXC1 has been linked to glaucoma. In contrast to blood endothelium, in which ANGPT2 is an antagonist of ANGPT1, we have shown that both ligands cooperate to regulate TIE2 in the lymphatic network of the eye. While A1A2Flox(WB) mice developed high IOP and glaucoma, expression of ANGPT1 or ANGPT2 alone was sufficient for ocular drainage. Furthermore, we demonstrated that loss of FOXC2 from lymphatics results in TIE2 downregulation, suggesting a mechanism for ocular defects in patients with FOXC mutations. These data reveal a pathogenetic and molecular basis for glaucoma and demonstrate the importance of angiopoietin ligand cooperation in the lymphatic endothelium.


Subject(s)
Angiopoietin-1/genetics , Glaucoma/pathology , Ocular Hypertension/pathology , Receptor, TIE-2/genetics , Angiopoietin-2/genetics , Animals , Aqueous Humor , Cell Separation , Disease Models, Animal , Down-Regulation , Flow Cytometry , Forkhead Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Intraocular Pressure , Ligands , Lymphatic System/pathology , Mice , Mice, Knockout , Mutation , Trabecular Meshwork/metabolism , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism
10.
J Am Soc Nephrol ; 25(11): 2459-70, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24904088

ABSTRACT

Podocytes are terminally differentiated cells with an elaborate cytoskeleton and are critical components of the glomerular barrier. We identified a bHLH transcription factor, Tcf21, that is highly expressed in developing and mature podocytes. Because conventional Tcf21 knockout mice die in the perinatal period with major cardiopulmonary defects, we generated a conditional Tcf21 knockout mouse to explore the role of this transcription factor in podocytes in vivo. Tcf21 was deleted from podocytes and podocyte progenitors using podocin-cre (podTcf21) and wnt4-cre (wnt4creTcf21) driver strains, respectively. Loss of Tcf21 from capillary-loop stage podocytes (podTcf21) results in simplified glomeruli with a decreased number of endothelial and mesangial cells. By 5 weeks of age, 40% of podTcf21 mice develop massive proteinuria and lesions similar to FSGS. Notably, the remaining 60% of mice do not develop proteinuria even when aged to 8 months. By contrast, earlier deletion of Tcf21 from podocyte precursors (wnt4creTcf21) results in a profound developmental arrest of podocyte differentiation and renal failure in 100% of mice during the perinatal period. Taken together, our results demonstrate a critical role for Tcf21 in the differentiation and maintenance of podocytes. Identification of direct targets of this transcription factor may provide new therapeutic avenues for proteinuric renal disease, including FSGS.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Diabetes Mellitus, Experimental/physiopathology , Glomerulosclerosis, Focal Segmental/physiopathology , Podocytes/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Cell Line , Cellular Senescence/physiology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Kidney Glomerulus/embryology , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Lac Operon , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Phenotype , Podocytes/pathology , Proteinuria/genetics , Proteinuria/pathology , Proteinuria/physiopathology
11.
Am J Physiol Renal Physiol ; 304(1): F120-6, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23054256

ABSTRACT

Deletion of the von Hippel-Lindau tumor suppressor (Vhl) gene from renal podocytes of mice (podVhl KO) leads to rapidly progressive glomerulonephritis (RPGN), a clinical syndrome characterized by rapid loss of renal function and crescents on renal biopsy. Genomic profiling of glomeruli isolated from podVhl knockout (KO) mice and from patients with RPGN identified a fingerprint of genes regulated by hypoxia-inducible factors (HIF), important substrates of the product of the VHL gene. Here, we show that stabilization of Hifs in podocytes is both required and sufficient for the glomerular phenotype observed in podVhl KO mice. Genetic deletion of the obligate dimerization partner Arnt/Hif1b that is essential for Hif transcriptional function rescues the phenotype. Conversely, stabilization of HIF2A alone in podocytes results in crescentic glomerular disease. Together, our results show that the Hif pathway and Hif2a in particular are key players in maintenance of the glomerular barrier.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Glomerulonephritis/pathology , Kidney Glomerulus/metabolism , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/physiology , Glomerulonephritis/genetics , Humans , Kidney Glomerulus/pathology , Mice , Mice, Knockout , Podocytes/physiology , Von Hippel-Lindau Tumor Suppressor Protein/genetics
12.
PLoS One ; 7(11): e50996, 2012.
Article in English | MEDLINE | ID: mdl-23226445

ABSTRACT

The kidney filtration barrier is formed by the combination of endothelial cells, basement membrane and epithelial cells called podocytes. These specialized actin-rich cells form long and dynamic protrusions, the foot processes, which surround glomerular capillaries and are connected by specialized intercellular junctions, the slit diaphragms. Failure to maintain the filtration barrier leads to massive proteinuria and nephrosis. A number of proteins reside in the slit diaphragm, notably the transmembrane proteins Nephrin and Neph1, which are both able to act as tyrosine phosphorylated scaffolds that recruit cytoplasmic effectors to initiate downstream signaling. While association between tyrosine-phosphorylated Neph1 and the SH2/SH3 adaptor Grb2 was shown in vitro to be sufficient to induce actin polymerization, in vivo evidence supporting this finding is still lacking. To test this hypothesis, we generated two independent mouse lines bearing a podocyte-specific constitutive inactivation of the Grb2 locus. Surprisingly, we show that mice lacking Grb2 in podocytes display normal renal ultra-structure and function, thus demonstrating that Grb2 is not required for the establishment of the glomerular filtration barrier in vivo. Moreover, our data indicate that Grb2 is not required to restore podocyte function following kidney injury. Therefore, although in vitro experiments suggested that Grb2 is important for the regulation of actin dynamics, our data clearly shows that its function is not essential in podocytes in vivo, thus suggesting that Grb2 rather plays a secondary role in this process.


Subject(s)
GRB2 Adaptor Protein/metabolism , Glomerular Filtration Barrier/metabolism , Animals , Crosses, Genetic , Female , Gene Silencing , Genotype , Glomerular Filtration Barrier/pathology , Glomerular Filtration Barrier/physiopathology , Glomerular Filtration Barrier/ultrastructure , Integrases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Organ Specificity , Podocytes/metabolism , Podocytes/pathology , Podocytes/ultrastructure
13.
Diabetes ; 61(11): 2958-66, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23093658

ABSTRACT

Vascular endothelial growth factor A (VEGFA) expression is increased in glomeruli in the context of diabetes. Here, we tested the hypothesis that this upregulation of VEGFA protects the glomerular microvasculature in diabetes and that therefore inhibition of VEGFA will accelerate nephropathy. To determine the role of glomerular Vegfa in the development and progression of diabetic nephropathy, we used an inducible Cre-loxP gene-targeting system that enabled genetic deletion of Vegfa selectively from glomerular podocytes of wild-type or diabetic mice. Type 1 diabetes was induced in mice using streptozotocin (STZ). We then assessed the extent of glomerular dysfunction by measuring proteinuria, glomerular pathology, and glomerular cell apoptosis. Vegfa expression increased in podocytes in the STZ model of diabetes. After 7 weeks of diabetes, diabetic mice lacking Vegfa in podocytes exhibited significantly greater proteinuria with profound glomerular scarring and increased apoptosis compared with control mice with diabetes or Vegfa deletion without diabetes. Reduced local production of glomerular Vegfa in a mouse model of type 1 diabetes promotes endothelial injury accelerating the progression of glomerular injury. These results suggest that upregulation of VEGFA in diabetic kidneys protects the microvasculature from injury and that reduction of VEGFA in diabetes may be harmful.


Subject(s)
Diabetic Nephropathies/metabolism , Kidney Glomerulus/blood supply , Kidney Glomerulus/metabolism , Microvessels/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis , Cells, Cultured , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/complications , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Diabetic Nephropathies/urine , Disease Progression , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Hyperglycemia/etiology , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Microvessels/pathology , Podocytes/metabolism , Podocytes/pathology , Proteinuria/etiology , RNA, Messenger/metabolism , Random Allocation , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics
14.
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
15.
Dev Biol ; 367(1): 40-54, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22546694

ABSTRACT

There is a reciprocal interaction between pancreatic islet cells and vascular endothelial cells (EC) in which EC-derived signals promote islet cell differentiation and islet development while islet cell-derived angiogenic factors promote EC recruitment and extensive islet vascularization. To examine the role of angiogenic factors in the coordinated development of islets and their associated vessels, we used a "tet-on" inducible system (mice expressing rat insulin promoter-reverse tetracycline activator transgene and a tet-operon-angiogenic factor transgene) to increase the ß cell production of vascular endothelial growth factor-A (VEGF-A), angiopoietin-1 (Ang1), or angiopoietin-2 (Ang2) during islet cell differentiation and islet development. In VEGF-A overexpressing embryos, ECs began to accumulate around epithelial tubes residing in the central region of the developing pancreas (associated with endocrine cells) as early as embryonic day 12.5 (E12.5) and increased dramatically by E16.5. While α and ß cells formed islet cell clusters in control embryos at E16.5, the increased EC population perturbed endocrine cell differentiation and islet cell clustering in VEGF-A overexpressing embryos. With continued overexpression of VEGF-A, α and ß cells became scattered, remained adjacent to ductal structures, and never coalesced into islets, resulting in a reduction in ß cell proliferation and ß cell mass at postnatal day 1. A similar impact on islet morphology was observed when VEGF-A was overexpressed in ß cells during the postnatal period. In contrast, increased expression of Ang1 or Ang2 in ß cells in developing or adult islets did not alter islet differentiation, development, or morphology, but altered islet EC ultrastructure. These data indicate that (1) increased EC number does not promote, but actually impairs ß cell proliferation and islet formation; (2) the level of VEGF-A production by islet endocrine cells is critical for islet vascularization during development and postnatally; (3) angiopoietin-Tie2 signaling in endothelial cells does not have a crucial role in the development or maintenance of islet vascularization.


Subject(s)
Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Vascular Endothelial Growth Factor A/metabolism , Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Animals , Cell Count , Endothelial Cells/metabolism , Islets of Langerhans/blood supply , Islets of Langerhans/metabolism , Mice
16.
J Clin Invest ; 121(6): 2278-89, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21606590

ABSTRACT

Angiopoietin-1/Tek signaling is a critical regulator of blood vessel development, with conventional knockout of angiopoietin-1 or Tek in mice being embryonically lethal due to vascular defects. In addition, angiopoietin-1 is thought to be required for the stability of mature vessels. Using a Cre-Lox conditional gene targeting approach, we have studied the role of angiopoietin-1 in embryonic and adult vasculature. We report here that angiopoietin-1 is critical for regulating both the number and diameter of developing vessels but is not required for pericyte recruitment. Cardiac-specific knockout of angiopoietin-1 reproduced the phenotype of the conventional knockout, demonstrating that the early vascular abnormalities arise from flow-dependent defects. Strikingly, deletion in the entire embryo after day E13.5 produced no immediate vascular phenotype. However, when combined with injury or microvascular stress, angiopoietin-1 deficiency resulted in profound organ damage, accelerated angiogenesis, and fibrosis. These findings redefine our understanding of the biological roles of angiopoietin-1: it is dispensable in quiescent vessels but has a powerful ability to modulate the vascular response after injury.


Subject(s)
Angiopoietin-1/physiology , Blood Vessels/embryology , Blood Vessels/injuries , Neovascularization, Physiologic/physiology , Wound Healing/physiology , Angiopoietin-1/deficiency , Angiopoietin-1/genetics , Animals , Blood Vessels/cytology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/physiopathology , Fetal Heart/growth & development , Fetal Heart/pathology , Gene Expression Regulation, Developmental , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/pathology , Liver/blood supply , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Neovascularization, Pathologic/embryology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/physiopathology , Pericytes/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Receptor, TIE-1/physiology , Receptor, TIE-2 , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology
17.
Curr Opin Nephrol Hypertens ; 20(3): 258-62, 2011 May.
Article in English | MEDLINE | ID: mdl-21455064

ABSTRACT

PURPOSE OF REVIEW: This review discusses the recent evidence that intrinsic glomerular cells including podocytes, parietal epithelial cells and progenitor cells within Bowman's capsule contribute to cellular crescents. RECENT FINDINGS: Using a variety of newer molecular markers and lineage tracing experiments, investigators have clearly demonstrated that glomerular cells play a key role in the development and progression of cellular crescents in experimental and human disease. SUMMARY: Crescentic glomerulonephritis is associated with significant morbidity and mortality. Current therapies target the immune system. The recent finding that nonimmune cells also play a role in crescent formation highlights the need to identify alternate and complimentary therapeutic strategies.


Subject(s)
Bowman Capsule/pathology , Glomerulonephritis/etiology , Podocytes/physiology , Animals , Glomerulonephritis/pathology , Humans , Intermediate Filament Proteins/physiology , Mice , Nerve Tissue Proteins/physiology , Nestin , Stem Cells/physiology
18.
Cell Metab ; 12(4): 329-340, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20889126

ABSTRACT

Diabetic nephropathy (DN) is the leading cause of renal failure in the world. It is characterized by albuminuria and abnormal glomerular function and is considered a hyperglycemic "microvascular" complication of diabetes, implying a primary defect in the endothelium. However, we have previously shown that human podocytes have robust responses to insulin. To determine whether insulin signaling in podocytes affects glomerular function in vivo, we generated mice with specific deletion of the insulin receptor from their podocytes. These animals develop significant albuminuria together with histological features that recapitulate DN, but in a normoglycemic environment. Examination of "normal" insulin-responsive podocytes in vivo and in vitro demonstrates that insulin signals through the MAPK and PI3K pathways via the insulin receptor and directly remodels the actin cytoskeleton of this cell. Collectively, this work reveals the critical importance of podocyte insulin sensitivity for kidney function.


Subject(s)
Insulin/physiology , Kidney/physiology , Podocytes/physiology , Animals , Diabetic Nephropathies , Kidney Glomerulus/cytology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction/physiology
19.
Curr Opin Nephrol Hypertens ; 18(4): 331-5, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19458528

ABSTRACT

PURPOSE OF REVIEW: In 2008, more than 376 papers were published on the glomerular barrier. Most of them dealt with the podocyte and its role in kidney disease. RECENT FINDINGS: There is new information on signaling pathways that are utilized in podocytes during proteinuria. Interestingly, the glomerular endothelium, with its fenestrae and glycocalyx, seems to be important for the maintenance of an intact glomerular barrier. All new advances at the molecular level are compatible with a highly size and charge-selective glomerular membrane and refute the concept of a 'leaky' glomerular barrier with tubular retrieval of intact albumin. Still, the hypothesis has its advocates, keeping a stimulating 'charge debate' alive. SUMMARY: Glomerular diseases account for 90% of chronic kidney disease requiring dialysis and transplantation at an annual cost of $20 billion in the USA. In clinical practice, we lack specific treatment of these diseases, giving us plenty of room for improvement. Future research should be directed toward deeper understanding of the signaling pathways involved in different conditions of proteinuria, the cross-talk between cell types in the glomerulus, and the identification of novel targets for treatment of acquired kidney disease.


Subject(s)
Kidney Glomerulus/metabolism , Animals , Endothelial Cells/physiology , Humans , Permeability , Podocytes/physiology , Signal Transduction , Vascular Endothelial Growth Factor A/physiology
20.
J Clin Invest ; 119(4): 943-53, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19287096

ABSTRACT

Studies in humans and animal models indicate a key contribution of angiotensin II to the pathogenesis of glomerular diseases. To examine the role of type 1 angiotensin (AT1) receptors in glomerular inflammation associated with autoimmune disease, we generated MRL-Faslpr/lpr (lpr) mice lacking the major murine type 1 angiotensin receptor (AT1A); lpr mice develop a generalized autoimmune disease with glomerulonephritis that resembles SLE. Surprisingly, AT1A deficiency was not protective against disease but instead substantially accelerated mortality, proteinuria, and kidney pathology. Increased disease severity was not a direct effect of immune cells, since transplantation of AT1A-deficient bone marrow did not affect survival. Moreover, autoimmune injury in extrarenal tissues, including skin, heart, and joints, was unaffected by AT1A deficiency. In murine systems, there is a second type 1 angiotensin receptor isoform, AT1B, and its expression is especially prominent in the renal glomerulus within podocytes. Further, expression of renin was enhanced in kidneys of AT1A-deficient lpr mice, and they showed evidence of exaggerated AT1B receptor activation, including substantially increased podocyte injury and expression of inflammatory mediators. Administration of losartan, which blocks all type 1 angiotensin receptors, reduced markers of kidney disease, including proteinuria, glomerular pathology, and cytokine mRNA expression. Since AT1A-deficient lpr mice had low blood pressure, these findings suggest that activation of type 1 angiotensin receptors in the glomerulus is sufficient to accelerate renal injury and inflammation in the absence of hypertension.


Subject(s)
Autoimmune Diseases/etiology , Nephritis/etiology , Receptor, Angiotensin, Type 1/physiology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/physiopathology , Chemokines/genetics , Cytokines/genetics , Female , Kidney/injuries , Kidney/pathology , Kidney/physiopathology , Male , Mice , Mice, Inbred MRL lpr , Mice, Knockout , Nephritis/immunology , Nephritis/pathology , Nephritis/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Angiotensin, Type 1/deficiency , Receptor, Angiotensin, Type 1/genetics , Renin-Angiotensin System/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...