Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Transpl Infect Dis ; 26(1): e14233, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38180168

ABSTRACT

BACKGROUND: Cytomegalovirus (CMV) infections are a common complication after kidney transplantation (KTx) and negatively affecting patient outcome. Valganciclovir (VGC) prophylaxis is often limited by drug-induced side effects and dose reduction due to decline in kidney function. METHOD: In the present study, episodes of CMV viremia in the first year after KTx in a cohort of 316 recipients were analyzed retrospectively to identify risk factors linked to persistent infections. RESULTS: In the studied cohort, 18.7% of patients showed a high-risk (HR) constellation (D+/R-) for CMV infections. CMV viremia affected 22% of our cohort, with HR patients being the most affected cohort (44.1%). Within this group, most viremic events (65.3%) occurred while patients were still on prophylactic therapy, showing significantly higher viral loads and a longer duration compared to seropositive recipients. CONCLUSION: The analysis at hand revealed that detection of viremia under ongoing antiviral prophylaxis bears an increased risk for sustained viral replication and antiviral drug resistance in HR patients. We identified low estimated glomerular filtration rate (eGFR) and lower dose VGC prophylaxis post-KTx as a risk factor for breakthrough infections in HR patients in our single center cohort. These patients might benefit from a closer CMV monitoring or novel prophylactic agents as letermovir.


Subject(s)
Cytomegalovirus Infections , Kidney Transplantation , Humans , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Cytomegalovirus , Kidney Transplantation/adverse effects , Retrospective Studies , Viremia/drug therapy , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/epidemiology , Cytomegalovirus Infections/prevention & control , Valganciclovir/therapeutic use , Transplant Recipients , Ganciclovir/therapeutic use , Ganciclovir/pharmacology
2.
J Thromb Haemost ; 21(3): 559-572, 2023 03.
Article in English | MEDLINE | ID: mdl-36696206

ABSTRACT

BACKGROUND: The von Willebrand factor-directed nanobody caplacizumab has greatly changed the treatment of immune thrombotic thrombocytopenic purpura (iTTP) in recent years. Data from randomized controlled trials established efficacy and safety. OBJECTIVES: This study aims to address open questions regarding patient selection, tailoring of therapy duration, obstacles in prescribing caplacizumab in iTTP, effect on adjunct treatment, and outcomes in the real-world setting. METHODS: We report retrospective, observational cohorts of 113 iTTP episodes treated with caplacizumab and 119 historical control episodes treated without caplacizumab. We aggregated data from the caplacizumab phase II/III trials and real-world data from France, the United Kingdom, Germany, and Austria (846 episodes, 396 treated with caplacizumab, and 450 historical controls). RESULTS: Caplacizumab was efficacious in iTTP, independent of the timing of therapy initiation, but curtailed the time of active iTTP only when used in the first-line therapy within 72 hours after diagnosis and until at least partial ADAMTS13-activity remission. Aggregated data from multiple study populations showed that caplacizumab use resulted in significant absolute risk reduction of 2.87% for iTTP-related mortality (number needed to treat 35) and a relative risk reduction of 59%. CONCLUSION: Caplacizumab should be used in first line and until ADAMTS13-remission, lowers iTTP-related mortality and refractoriness, and decreases the number of daily plasma exchange and hospital stay. This trial is registered at www. CLINICALTRIALS: gov as #NCT04985318.


Subject(s)
Purpura, Thrombocytopenic, Idiopathic , Purpura, Thrombotic Thrombocytopenic , Single-Domain Antibodies , Thrombosis , Humans , Retrospective Studies , Treatment Outcome , ADAMTS13 Protein
4.
Microorganisms ; 9(8)2021 Aug 04.
Article in English | MEDLINE | ID: mdl-34442744

ABSTRACT

The treatment options for cytomegalovirus (CMV) infections in immunosuppressed patients are limited, mainly consisting of (val-)ganciclovir (VGC/GCV) as the first-line treatment. We report on three transplant recipients, one stem cell transplant (allo-HSCT) patient and two kidney transplant (KTx) recipients, with prolonged CMV viremia treated with a combined therapy based on letermovir (LMV), CMV-specific intravenous immunoglobulins (IVIg), and VGC/GCV, which led to the sustained control of CMV viremia in all patients.

5.
Dtsch Med Wochenschr ; 146(13-14): 915-917, 2021 Jul.
Article in German | MEDLINE | ID: mdl-34256408

ABSTRACT

Increasing insight into the clinical phenotype and mechanisms of SARS-CoV-2 infections and COVID-19 has identified damage of the kidneys as a key player in the course of the disease. This manuscript updates our previous summary with current knowledge on kidney involvement in COVID-19 and chronic kidney disease as a risk factor for severe COVID-19, as well as recommendations regarding SARS-CoV-2 vaccination for patients suffering from chronic kidney disease and after organ transplantation, respectively. populations, SARS-CoV-2 vaccination is strongly recommended for all patients suffering from chronic kidney disease and after kidney transplantation.


Subject(s)
COVID-19 Vaccines/administration & dosage , COVID-19 , Organ Transplantation , Renal Insufficiency, Chronic/complications , SARS-CoV-2/immunology , COVID-19/complications , COVID-19/prevention & control , Humans , Organ Transplantation/adverse effects , Risk Factors
6.
Cells ; 10(3)2021 03 20.
Article in English | MEDLINE | ID: mdl-33804736

ABSTRACT

Diseases of the renal filtration unit-the glomerulus-are the most common cause of chronic kidney disease. Podocytes are the pivotal cell type for the function of this filter and focal-segmental glomerulosclerosis (FSGS) is a classic example of a podocytopathy leading to proteinuria and glomerular scarring. Currently, no targeted treatment of FSGS is available. This lack of therapeutic strategies is explained by a limited understanding of the defects in podocyte cell biology leading to FSGS. To date, most studies in the field have focused on protein-coding genes and their gene products. However, more than 80% of all transcripts produced by mammalian cells are actually non-coding. Here, long non-coding RNAs (lncRNAs) are a relatively novel class of transcripts and have not been systematically studied in FSGS to date. The appropriate tools to facilitate lncRNA research for the renal scientific community are urgently required due to a row of challenges compared to classical analysis pipelines optimized for coding RNA expression analysis. Here, we present the bioinformatic pipeline CALINCA as a solution for this problem. CALINCA automatically analyzes datasets from murine FSGS models and quantifies both annotated and de novo assembled lncRNAs. In addition, the tool provides in-depth information on podocyte specificity of these lncRNAs, as well as evolutionary conservation and expression in human datasets making this pipeline a crucial basis to lncRNA studies in FSGS.


Subject(s)
Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Podocytes/metabolism , Podocytes/pathology , RNA, Long Noncoding/metabolism , Software , Animals , Disease Models, Animal , Gene Expression Regulation , Humans , Male , Mice, Inbred BALB C , RNA, Long Noncoding/genetics , Reproducibility of Results
7.
Immun Inflamm Dis ; 9(2): 513-520, 2021 06.
Article in English | MEDLINE | ID: mdl-33559399

ABSTRACT

BACKGROUND: Recently, chronic hepatitis E virus (HEV) infections gained increasing attention as a possible cause for elevated liver enzymes of unknown origin and liver cirrhosis in solid organ transplant recipients. Reduction of immunosuppressive therapy and/or use of antiviral drug ribavirin have been established as possible treatment strategies. METHODS: The efficacy of dose reduction of mycophenolic acid (MPA) and ribavirin therapy was retrospectively analyzed in eight renal transplant patients of our outpatient clinic who were diagnosed with HEV infection by detection of specific antibodies (immunoglobulin M and immunoglobulin G) and/or positive RNA in blood and stool. In four patients serial HEV viral loads in blood were measured. RESULTS: Only one patient reached HEV clearance after reduction of immunosuppressive therapy (predominantly reduction of MPA daily dose) alone, whereas six patients were treated with ribavirin after reduction of immunosuppressive therapy due to persistent virus replication. Four of six patients reached HEV clearance after 3 months of ribavirin therapy. HEV clearance was observed after 34-42 days. Two patients, both treated with rituximab within the last 12 months before diagnosis of HEV infection, needed prolonged ribavirin therapy due to persistent viral replication. CONCLUSION: Reduction of daily dose of MPA therapy alone in transplant patients with chronic HEV infection may not be sufficient to control viral replication. HEV clearance under ribavirin therapy shows interindividual variability. Therefore, serial viral monitoring may be useful to personalize treatment duration. Rituximab therapy is a risk factor for complicated-to-treat chronic HEV infection.


Subject(s)
Hepatitis E virus , Hepatitis E , Kidney Transplantation , Hepatitis E/diagnosis , Hepatitis E/drug therapy , Hepatitis E virus/genetics , Humans , Kidney Transplantation/adverse effects , RNA, Viral , Retrospective Studies
8.
Transplant Proc ; 53(3): 786-792, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32981693

ABSTRACT

BACKGROUND: Health-related quality of life (HRQL), fatigue, anxiety, and depression are crucial for the living kidney donor (LKD). Follow-up data for HRQL of LKDs comparing surgical techniques, especially regarding hand-assisted retroperitoneoscopic donor nephrectomy (HARP), are sparse. The aim of this study was to evaluate the influence of abdominal wall trauma minimized by HARP in comparison to open anterior approach donor nephrectomy (AA) on HRQL and additional psychosocial aspects of LKDs during the long-term follow-up. MATERIAL AND METHODS: This is a cross-sectional study comparing psychosocial aspects of LKD between HARP and AA. RESULTS: This study included 100 LKDs (68 HARP, 28 AA, and 4 were excluded secondary to incomplete data). The time to follow-up was 22.6 ± 11.7 (HARP) vs 58.7 ± 13.9 (AA) months (P < .005). Complications ≥3a° due to Clavien-Dindo classification was 0% in both groups. There were higher scores in all physical aspects for HARP donors vs AA donors at that time (physical function: 89.8 ± 14.6 vs 80.0 ± 19.9, P = .008, and the physical component score: 53.9 ± 7.6 vs 48.6 ± 8.5, P = .006). One year later (follow-up time + 12 months), HRQL for HARP donors was still higher. Mental items showed no significant differences. HARP donors showed better physical scores compared to the age-matched nondonor population (AA donors had lower scores). Neither the Multidimensional Fatigue Inventory-20 (MFI-20) or the Hospital Anxiety and Depression Scale (HADS) showed any differences between the 2 groups. Fatigue scores were higher for HARP and for AA compared to the age-matched population. CONCLUSIONS: LKDs undergoing HARP showed better physical performance as part of HRQL in the long-term follow-up.


Subject(s)
Hand-Assisted Laparoscopy/methods , Kidney Transplantation , Nephrectomy/methods , Retroperitoneal Space/surgery , Tissue and Organ Harvesting/methods , Adult , Cross-Sectional Studies , Female , Humans , Kidney/surgery , Living Donors , Male , Middle Aged , Outcome Assessment, Health Care , Physical Functional Performance , Postoperative Period , Quality of Life , Time
9.
Dtsch Med Wochenschr ; 145(15): 1068-1073, 2020 Jul.
Article in German | MEDLINE | ID: mdl-32731281

ABSTRACT

Increasing insight into the clinical phenotype and mechanisms of SARS-CoV-2 infections and COVID-19 has identified damage of the kidneys as a key player in the course of the disease. This manuscript summarizes the current knowledge on direct viral infection of kidney tissue, proteinuria and acute kidney injury in COVID-19, and management of patients on chronic dialysis as well as after kidney transplantation. Direct infection of podocytes and proximal tubular cells by SARS-CoV-2 has been confirmed and results in proteinuria and hematuria at an early stage of COVID-19. In this context, any kidney affection is a predictor of worse outcomes among COVID-19 patients irrespective of the initial presentation and increases the risk of acute kidney injury. Specific therapies for kidney damage and acute kidney injury within COVID-19 that could be generally recommended are currently lacking. Patients on chronic hemodialysis in particular are at risk for contracting SARS-CoV-2 infections as indicated by outbreaks and super-spreading events in hemodialysis facilities. Immunosuppressive therapy after kidney transplantation needs to be adapted upon diagnosis of COVID-19 depending on the severity of the initial presentation.


Subject(s)
Acute Kidney Injury , Coronavirus Infections , Pandemics , Pneumonia, Viral , Acute Kidney Injury/physiopathology , Acute Kidney Injury/virology , Betacoronavirus , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/physiopathology , Hematuria , Humans , Kidney/physiopathology , Kidney/virology , Pneumonia, Viral/complications , Pneumonia, Viral/physiopathology , Proteinuria , Renal Dialysis , Renal Insufficiency, Chronic/therapy , Risk Factors , SARS-CoV-2
10.
J Am Soc Nephrol ; 29(8): 2060-2068, 2018 08.
Article in English | MEDLINE | ID: mdl-29794128

ABSTRACT

Background Three different cell types constitute the glomerular filter: mesangial cells, endothelial cells, and podocytes. However, to what extent cellular heterogeneity exists within healthy glomerular cell populations remains unknown.Methods We used nanodroplet-based highly parallel transcriptional profiling to characterize the cellular content of purified wild-type mouse glomeruli.Results Unsupervised clustering of nearly 13,000 single-cell transcriptomes identified the three known glomerular cell types. We provide a comprehensive online atlas of gene expression in glomerular cells that can be queried and visualized using an interactive and freely available database. Novel marker genes for all glomerular cell types were identified and supported by immunohistochemistry images obtained from the Human Protein Atlas. Subclustering of endothelial cells revealed a subset of endothelium that expressed marker genes related to endothelial proliferation. By comparison, the podocyte population appeared more homogeneous but contained three smaller, previously unknown subpopulations.Conclusions Our study comprehensively characterized gene expression in individual glomerular cells and sets the stage for the dissection of glomerular function at the single-cell level in health and disease.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Profiling , Kidney Glomerulus/physiology , Mesangial Cells/metabolism , Podocytes/metabolism , Sequence Analysis, RNA , Animals , Cells, Cultured , Gene Expression Regulation , Kidney Glomerulus/cytology , Male , Mice , Mice, Inbred Strains , Reference Values
11.
FASEB J ; 32(9): 4696-4713, 2018 09.
Article in English | MEDLINE | ID: mdl-29694247

ABSTRACT

Podocytes are highly specialized, epithelial, postmitotic cells, which maintain the renal filtration barrier. When adapting to considerable metabolic and mechanical stress, podocytes need to accurately maintain their proteome. Immortalized podocyte cell lines are a widely used model for studying podocyte biology in health and disease in vitro. In this study, we performed a comprehensive proteomic analysis of the cultured human podocyte proteome in both proliferative and differentiated conditions at a depth of >7000 proteins. Similar to mouse podocytes, human podocyte differentiation involved a shift in proteostasis: undifferentiated podocytes have high expression of proteasomal proteins, whereas differentiated podocytes have high expression of lysosomal proteins. Additional analyses with pulsed stable-isotope labeling by amino acids in cell culture and protein degradation assays determined protein dynamics and half-lives. These studies unraveled a globally increased stability of proteins in differentiated podocytes. Mitochondrial, cytoskeletal, and membrane proteins were stabilized, particularly in differentiated podocytes. Importantly, protein half-lives strongly contributed to protein abundance in each state. These data suggest that regulation of protein turnover of particular cellular functions determines podocyte differentiation, a paradigm involving mitophagy and, potentially, of importance in conditions of increased podocyte stress and damage.-Schroeter, C. B., Koehler, S., Kann, M., Schermer, B., Benzing, T., Brinkkoetter, P. T., Rinschen, M. M. Protein half-life determines expression of proteostatic networks in podocyte differentiation.


Subject(s)
Cell Differentiation/physiology , Organogenesis/physiology , Podocytes/metabolism , Proteins/metabolism , Cell Line , Cells, Cultured , Cytoplasm/metabolism , Cytoskeleton/metabolism , Humans , Membrane Proteins/metabolism , Proteomics/methods
12.
Kidney Int ; 93(6): 1308-1319, 2018 06.
Article in English | MEDLINE | ID: mdl-29530281

ABSTRACT

In diseases of many parenchymatous organs, heterogeneous deterioration of individual functional units determines the clinical prognosis. However, the molecular characterization at the level of such individual subunits remains a technological challenge that needs to be addressed in order to better understand pathological mechanisms. Proteinuric glomerular kidney diseases are frequent and assorted diseases affecting a fraction of glomeruli and their draining tubules to variable extents, and for which no specific treatment exists. Here, we developed and applied a mass spectrometry-based methodology to investigate heterogeneity of proteomes from individually isolated nephron segments from mice with proteinuric kidney disease. In single glomeruli from two different mouse models of sclerotic glomerular disease, we identified a coherent protein expression module consisting of extracellular matrix protein deposition (reflecting glomerular sclerosis), glomerular albumin (reflecting proteinuria) and LAMP1, a lysosomal protein. This module was associated with a loss of podocyte marker proteins while genetic ablation of LAMP1-correlated lysosomal proteases could ameliorate glomerular damage in vivo. Furthermore, proteomic analyses of individual glomeruli from patients with genetic sclerotic and non-sclerotic proteinuric diseases revealed increased abundance of lysosomal proteins, in combination with a decreased abundance of mutated gene products. Thus, altered protein homeostasis (proteostasis) is a conserved key mechanism in proteinuric kidney diseases. Moreover, our technology can capture intra-individual variability in diseases of the kidney and other tissues at a sub-biopsy scale.


Subject(s)
Glomerulonephritis/metabolism , Nephrons/metabolism , Proteinuria/metabolism , Proteome , Proteomics/methods , Tandem Mass Spectrometry , Animals , Biological Variation, Individual , Biomarkers/metabolism , Disease Models, Animal , Extracellular Matrix Proteins/metabolism , Glomerulonephritis/genetics , Glomerulonephritis/pathology , Glomerulonephritis/physiopathology , Humans , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Male , Mice , Mice, Knockout , Nephrons/pathology , Nephrons/physiopathology , Nephrotic Syndrome/genetics , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Nephrotic Syndrome/physiopathology , Podocytes/metabolism , Podocytes/pathology , Proteinuria/genetics , Proteinuria/pathology , Proteinuria/physiopathology , Proteostasis , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reproducibility of Results , Serum Albumin/metabolism , WT1 Proteins
13.
J Am Soc Nephrol ; 28(10): 2867-2878, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28724775

ABSTRACT

Regulated intracellular proteostasis, controlled in part by proteolysis, is essential in maintaining the integrity of podocytes and the glomerular filtration barrier of the kidney. We applied a novel proteomics technology that enables proteome-wide identification, mapping, and quantification of protein N-termini to comprehensively characterize cleaved podocyte proteins in the glomerulus in vivo We found evidence that defined proteolytic cleavage results in various proteoforms of important podocyte proteins, including those of podocin, nephrin, neph1, α-actinin-4, and vimentin. Quantitative mapping of N-termini demonstrated perturbation of protease action during podocyte injury in vitro, including diminished proteolysis of α-actinin-4. Differentially regulated protease substrates comprised cytoskeletal proteins as well as intermediate filaments. Determination of preferential protease motifs during podocyte damage indicated activation of caspase proteases and inhibition of arginine-specific proteases. Several proteolytic processes were clearly site-specific, were conserved across species, and could be confirmed by differential migration behavior of protein fragments in gel electrophoresis. Some of the proteolytic changes discovered in vitro also occurred in two in vivo models of podocyte damage (WT1 heterozygous knockout mice and puromycin aminonucleoside-treated rats). Thus, we provide direct and systems-level evidence that the slit diaphragm and podocyte cytoskeleton are regulated targets of proteolytic modification, which is altered upon podocyte damage.


Subject(s)
Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Kidney Diseases/metabolism , Podocytes/metabolism , Proteolysis , Animals , Cells, Cultured , Humans , Male , Mice, Knockout , Proteome , Proteomics/methods , Rats
14.
Sci Signal ; 10(474)2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28400537

ABSTRACT

Podocytes are terminally differentiated cells of the kidney filtration barrier. They are subjected to physiological filtration pressure and considerable mechanical strain, which can be further increased in various kidney diseases. When injury causes cytoskeletal reorganization and morphological alterations of these cells, the filtration barrier may become compromised and allow proteins to leak into the urine (a condition called proteinuria). Using time-resolved proteomics, we showed that podocyte injury stimulated the activity of the transcriptional coactivator YAP and the expression of YAP target genes in a rat model of glomerular disease before the development of proteinuria. Although the activities of YAP and its ortholog TAZ are activated by mechanical stress in most cell types, injury reduced YAP and TAZ activity in cultured human and mouse podocyte cell lines grown on stiff substrates. Culturing these cells on soft matrix or inhibiting stress fiber formation recapitulated the damage-induced YAP up-regulation observed in vivo, indicating a mechanotransduction-dependent mechanism of YAP activation in podocytes. YAP overexpression in cultured podocytes increased the abundance of extracellular matrix-related proteins that can contribute to fibrosis. YAP activity was increased in mouse models of diabetic nephropathy, and the YAP target CTGF was highly expressed in renal biopsies from glomerular disease patients. Although overexpression of human YAP in mice induced mild proteinuria, pharmacological inhibition of the interaction between YAP and its partner TEAD in rats ameliorated glomerular disease and reduced damage-induced mechanosignaling in the glomeruli. Thus, perturbation of YAP-dependent mechanosignaling is a potential therapeutic target for treating some glomerular diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Mechanotransduction, Cellular , Phosphoproteins/metabolism , Podocytes/metabolism , Transcription Factors/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Fluorescent Antibody Technique , HEK293 Cells , Humans , Kidney Glomerulus/metabolism , Male , Mice , Phosphoproteins/genetics , Podocytes/cytology , Podocytes/drug effects , Proteinuria/genetics , Proteinuria/metabolism , Proteomics , Puromycin Aminonucleoside/pharmacology , Rats , Stress, Mechanical , Transcription Factors/genetics , YAP-Signaling Proteins
15.
Am J Physiol Cell Physiol ; 311(3): C404-17, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27357545

ABSTRACT

The renal filtration barrier is maintained by the renal podocyte, an epithelial postmitotic cell. Immortalized mouse podocyte cell lines-both in the differentiated and undifferentiated state-are widely utilized tools to estimate podocyte injury and cytoskeletal rearrangement processes in vitro. Here, we mapped the cultured podocyte proteome at a depth of more than 8,800 proteins and quantified 7,240 proteins. Copy numbers of proteins mutated in forms of hereditary nephrotic syndrome or focal segmental glomerulosclerosis (FSGS) were assessed. We found that cultured podocytes express abundant copy numbers of endogenous receptors, such as tyrosine kinase membrane receptors, the G protein-coupled receptor (GPCR), NPR3 (ANP receptor), and several poorly characterized GPCRs. The data set was correlated with deep mapping mRNA sequencing ("mRNAseq") data from the native mouse podocyte, the native mouse podocyte proteome and staining intensities from the human protein atlas. The generated data set was similar to these previously published resources, but several native and high-abundant podocyte-specific proteins were not identified in the data set. Notably, this data set detected general perturbations in proteostatic mechanisms as a dominant alteration during podocyte differentiation, with high proteasome activity in the undifferentiated state and markedly increased expression of lysosomal proteins in the differentiated state. Phosphoproteomics analysis of mouse podocytes at a resolution of more than 3,000 sites suggested a preference of phosphorylation of actin filament-associated proteins in the differentiated state. The data set obtained here provides a resource and provides the means for deep mapping of the native podocyte proteome and phosphoproteome in a similar manner.


Subject(s)
Cell Differentiation/physiology , Podocytes/metabolism , Podocytes/physiology , Proteome/metabolism , Animals , Cell Line , Glomerulosclerosis, Focal Segmental/metabolism , Humans , Kidney/metabolism , Kidney/physiology , Mice , Phosphorylation/physiology , Proteins/metabolism
16.
Nat Genet ; 47(6): 647-53, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25961942

ABSTRACT

Cardiovascular disease is the most common cause of death worldwide, and hypertension is the major risk factor. Mendelian hypertension elucidates mechanisms of blood pressure regulation. Here we report six missense mutations in PDE3A (encoding phosphodiesterase 3A) in six unrelated families with mendelian hypertension and brachydactyly type E (HTNB). The syndrome features brachydactyly type E (BDE), severe salt-independent but age-dependent hypertension, an increased fibroblast growth rate, neurovascular contact at the rostral-ventrolateral medulla, altered baroreflex blood pressure regulation and death from stroke before age 50 years when untreated. In vitro analyses of mesenchymal stem cell-derived vascular smooth muscle cells (VSMCs) and chondrocytes provided insights into molecular pathogenesis. The mutations increased protein kinase A-mediated PDE3A phosphorylation and resulted in gain of function, with increased cAMP-hydrolytic activity and enhanced cell proliferation. Levels of phosphorylated VASP were diminished, and PTHrP levels were dysregulated. We suggest that the identified PDE3A mutations cause the syndrome. VSMC-expressed PDE3A deserves scrutiny as a therapeutic target for the treatment of hypertension.


Subject(s)
Brachydactyly/genetics , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Hypertension/congenital , Adolescent , Adult , Amino Acid Sequence , Animals , Base Sequence , Case-Control Studies , Cell Differentiation , Child , Female , Genetic Association Studies , HeLa Cells , Humans , Hypertension/genetics , Kinetics , Male , Mesenchymal Stem Cells/physiology , Mice , Middle Aged , Molecular Sequence Data , Mutation, Missense , Myocytes, Smooth Muscle/physiology , Pedigree
17.
Development ; 142(7): 1254-66, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25804736

ABSTRACT

Development of the metanephric kidney depends on tightly regulated interplay between self-renewal and differentiation of a nephron progenitor cell (NPC) pool. Several key factors required for the survival of NPCs have been identified, including fibroblast growth factor (FGF) signaling and the transcription factor Wilms' tumor suppressor 1 (WT1). Here, we present evidence that WT1 modulates FGF signaling by activating the expression of growth arrest-specific 1 (Gas1), a novel WT1 target gene and novel modulator of FGF signaling. We show that WT1 directly binds to a conserved DNA binding motif within the Gas1 promoter and activates Gas1 mRNA transcription in NPCs. We confirm that WT1 is required for Gas1 expression in kidneys in vivo. Loss of function of GAS1 in vivo results in hypoplastic kidneys with reduced nephron mass due to premature depletion of NPCs. Although kidney development in Gas1 knockout mice progresses normally until E15.5, NPCs show decreased rates of proliferation at this stage and are depleted as of E17.5. Lastly, we show that Gas1 is selectively required for FGF-stimulated AKT signaling in vitro. In summary, our data suggest a model in which WT1 modulates receptor tyrosine kinase signaling in NPCs by directing the expression of Gas1.


Subject(s)
Cell Cycle Proteins/metabolism , Fibroblast Growth Factors/metabolism , Nephrons/metabolism , Signal Transduction , Stem Cells/metabolism , WT1 Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Proliferation , DNA/genetics , Enzyme Activation/drug effects , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice, Knockout , Models, Animal , Nephrons/abnormalities , Nephrons/embryology , Nephrons/pathology , Organ Culture Techniques , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-ret/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
EMBO Mol Med ; 7(3): 275-87, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25643582

ABSTRACT

Mitochondrial dysfunction and alterations in energy metabolism have been implicated in a variety of human diseases. Mitochondrial fusion is essential for maintenance of mitochondrial function and requires the prohibitin ring complex subunit prohibitin-2 (PHB2) at the mitochondrial inner membrane. Here, we provide a link between PHB2 deficiency and hyperactive insulin/IGF-1 signaling. Deletion of PHB2 in podocytes of mice, terminally differentiated cells at the kidney filtration barrier, caused progressive proteinuria, kidney failure, and death of the animals and resulted in hyperphosphorylation of S6 ribosomal protein (S6RP), a known mediator of the mTOR signaling pathway. Inhibition of the insulin/IGF-1 signaling system through genetic deletion of the insulin receptor alone or in combination with the IGF-1 receptor or treatment with rapamycin prevented hyperphosphorylation of S6RP without affecting the mitochondrial structural defect, alleviated renal disease, and delayed the onset of kidney failure in PHB2-deficient animals. Evidently, perturbation of insulin/IGF-1 receptor signaling contributes to tissue damage in mitochondrial disease, which may allow therapeutic intervention against a wide spectrum of diseases.


Subject(s)
Insulin/metabolism , Mitochondria/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Renal Insufficiency , Signal Transduction , Animals , Gene Deletion , Mice, Inbred C57BL , Phosphorylation , Prohibitins , Protein Processing, Post-Translational , Receptor, Insulin/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ribosomal Protein S6/metabolism
19.
J Am Soc Nephrol ; 26(9): 2097-104, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25636411

ABSTRACT

The transcription factor Wilms' tumor suppressor 1 (WT1) is key to podocyte development and viability; however, WT1 transcriptional networks in podocytes remain elusive. We provide a comprehensive analysis of the genome-wide WT1 transcriptional network in podocytes in vivo using chromatin immunoprecipitation followed by sequencing (ChIPseq) and RNA sequencing techniques. Our data show a specific role for WT1 in regulating the podocyte-specific transcriptome through binding to both promoters and enhancers of target genes. Furthermore, we inferred a podocyte transcription factor network consisting of WT1, LMX1B, TCF21, Fox-class and TEAD family transcription factors, and MAFB that uses tissue-specific enhancers to control podocyte gene expression. In addition to previously described WT1-dependent target genes, ChIPseq identified novel WT1-dependent signaling systems. These targets included components of the Hippo signaling system, underscoring the power of genome-wide transcriptional-network analyses. Together, our data elucidate a comprehensive gene regulatory network in podocytes suggesting that WT1 gene regulatory function and podocyte cell-type specification can best be understood in the context of transcription factor-regulatory element network interplay.


Subject(s)
Gene Expression Regulation , Podocytes , Repressor Proteins/genetics , Signal Transduction/genetics , Transcriptome , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Chromatin Immunoprecipitation , Forkhead Transcription Factors/genetics , Genomics , Hippo Signaling Pathway , LIM-Homeodomain Proteins/genetics , MafB Transcription Factor/genetics , Mice , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sequence Analysis, DNA , Sequence Analysis, RNA , Transcription Factors/genetics , WT1 Proteins
20.
Dev Dyn ; 242(6): 790-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23559562

ABSTRACT

BACKGROUND: The DNA-binding transcription factor Wilms' Tumor Suppressor-1 (WT1) plays an essential role in nephron progenitor differentiation during renal development. We previously used Wt1 chromatin-immunoprecipitation coupled to microarray (ChIP-chip) to identify novel Wt1 target genes that may regulate nephrogenesis in vivo. We discovered that all three members of the SoxC subfamily, namely, Sox4, Sox11, and Sox12, are bound by Wt1 in mouse embryonic kidneys in vivo. SoxC genes play master roles in determining neuronal and mesenchymal progenitor cell fate in a multitude of developmental processes, but their function in the developing kidney is largely unknown. RESULTS: Here we show that all three SoxC genes are expressed in the nephrogenic lineages during renal development. Conditional ablation of Sox4 in nephron progenitors and their cellular descendants (Sox4(nephron-) mice) results in a significant reduction in nephron endowment. By postnatal day (P)7, Sox4(nephron-) renal corpuscles exhibit reduced numbers of Wt1+ podocytes together with loss of expression of the slit diaphragm protein nephrin. Sox4(nephron-) mice develop early-onset proteinacious glomerular injury within 2 weeks of birth progressing to end-stage renal failure within 5-9 months. CONCLUSIONS: Collectively, our results demonstrate an essential requirement of Sox4 for normal renal development in vivo.


Subject(s)
Gene Expression Regulation, Developmental , Kidney/embryology , SOXC Transcription Factors/metabolism , Alleles , Animals , Cell Lineage , Chromatin Immunoprecipitation , In Situ Hybridization , Kidney Glomerulus/metabolism , Mice , Microscopy, Electron, Transmission , Nephrons/metabolism , Oligonucleotide Array Sequence Analysis , Renal Insufficiency/genetics , Stem Cells/cytology , Time Factors , WT1 Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...