Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Nat Genet ; 54(11): 1690-1701, 2022 11.
Article in English | MEDLINE | ID: mdl-36303074

ABSTRACT

Adult kidney organoids have been described as strictly tubular epithelia and termed tubuloids. While the cellular origin of tubuloids has remained elusive, here we report that they originate from a distinct CD24+ epithelial subpopulation. Long-term-cultured CD24+ cell-derived tubuloids represent a functional human kidney tubule. We show that kidney tubuloids can be used to model the most common inherited kidney disease, namely autosomal dominant polycystic kidney disease (ADPKD), reconstituting the phenotypic hallmark of this disease with cyst formation. Single-cell RNA sequencing of CRISPR-Cas9 gene-edited PKD1- and PKD2-knockout tubuloids and human ADPKD and control tissue shows similarities in upregulation of disease-driving genes. Furthermore, in a proof of concept, we demonstrate that tolvaptan, the only approved drug for ADPKD, has a significant effect on cyst size in tubuloids but no effect on a pluripotent stem cell-derived model. Thus, tubuloids are derived from a tubular epithelial subpopulation and represent an advanced system for ADPKD disease modeling.


Subject(s)
Cysts , Polycystic Kidney, Autosomal Dominant , Adult , Humans , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics , Organoids , Kidney , CD24 Antigen/genetics
2.
JCI Insight ; 4(18)2019 09 19.
Article in English | MEDLINE | ID: mdl-31534053

ABSTRACT

The cellular origins of glomerulosclerosis involve activation of parietal epithelial cells (PECs) and progressive podocyte depletion. While mammalian target of rapamycin-mediated (mTOR-mediated) podocyte hypertrophy is recognized as an important signaling pathway in the context of glomerular disease, the role of podocyte hypertrophy as a compensatory mechanism preventing PEC activation and glomerulosclerosis remains poorly understood. In this study, we show that glomerular mTOR and PEC activation-related genes were both upregulated and intercorrelated in biopsies from patients with focal segmental glomerulosclerosis (FSGS) and diabetic nephropathy, suggesting both compensatory and pathological roles. Advanced morphometric analyses in murine and human tissues identified podocyte hypertrophy as a compensatory mechanism aiming to regulate glomerular functional integrity in response to somatic growth, podocyte depletion, and even glomerulosclerosis - all of this in the absence of detectable podocyte regeneration. In mice, pharmacological inhibition of mTOR signaling during acute podocyte loss impaired hypertrophy of remaining podocytes, resulting in unexpected albuminuria, PEC activation, and glomerulosclerosis. Exacerbated and persistent podocyte hypertrophy enabled a vicious cycle of podocyte loss and PEC activation, suggesting a limit to its beneficial effects. In summary, our data highlight a critical protective role of mTOR-mediated podocyte hypertrophy following podocyte loss in order to preserve glomerular integrity, preventing PEC activation and glomerulosclerosis.


Subject(s)
Albuminuria/chemically induced , Diabetic Nephropathies/pathology , Everolimus/adverse effects , Glomerulosclerosis, Focal Segmental/pathology , TOR Serine-Threonine Kinases/metabolism , Aged , Aged, 80 and over , Animals , Biopsy , Cells, Cultured , Child, Preschool , Datasets as Topic , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/drug therapy , Epithelial Cells/pathology , Everolimus/administration & dosage , Female , Gene Expression Profiling , Humans , Hypertrophy/drug therapy , Hypertrophy/pathology , Infant , Male , Mice , Mice, Knockout , Middle Aged , Podocytes , Primary Cell Culture , Regeneration , Signal Transduction/drug effects , Signal Transduction/genetics , Streptozocin/toxicity , TOR Serine-Threonine Kinases/analysis , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism , Up-Regulation , Young Adult
3.
Kidney Int ; 95(5): 1103-1119, 2019 05.
Article in English | MEDLINE | ID: mdl-30827511

ABSTRACT

Platelet-derived growth factors (PDGF) have been implicated in kidney disease progression. We previously found that PDGF-C is upregulated at sites of renal fibrosis and that antagonism of PDGF-C reduces fibrosis in the unilateral ureteral obstruction model. We studied the role of PDGF-C in collagen 4A3-/- ("Alport") mice, a model of progressive renal fibrosis with greater relevance to human kidney disease. Alport mice were crossbred with PDGF-C-/- mice or administered a neutralizing PDGF-C antibody. Both PDGF-C deficiency and neutralization reduced serum creatinine and blood urea nitrogen levels and mitigated glomerular injury, renal fibrosis, and renal inflammation. Unexpectedly, systolic blood pressure was also reduced in both Alport and wild-type mice treated with a neutralizing PDGF-C antibody. Neutralization of PDGF-C reduced arterial wall thickness in the renal cortex of Alport mice. Aortic rings isolated from anti-PDGF-C-treated wildtype mice exhibited reduced tension and faster relaxation than those of untreated mice. In vitro, PDGF-C upregulated angiotensinogen in aortic tissue and in primary hepatocytes and induced nuclear factor κB (NFκB)/p65-binding to the angiotensinogen promoter in hepatocytes. Neutralization of PDGF-C suppressed transcript expression of angiotensinogen in Alport mice and angiotensin II receptor type 1 in Alport and wildtype mice. Finally, administration of neutralizing PDGF-C antibodies ameliorated angiotensin II-induced hypertension in healthy mice. Thus, in addition to its key role in mediating renal fibrosis, we identified PDGF-C as a mediator of hypertension via effects on renal vasculature and on the renin-angiotensin system. The contribution to both renal fibrosis and hypertension render PDGF-C an attractive target in progressive kidney disease.


Subject(s)
Hypertension/pathology , Kidney/pathology , Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Angiotensinogen/metabolism , Animals , Blood Pressure/genetics , Cells, Cultured , Collagen Type IV/genetics , Disease Models, Animal , Fibrosis , Hepatocytes , Humans , Hypertension/etiology , Hypertension/genetics , Lymphokines/antagonists & inhibitors , Male , Mice , Mice, Knockout , Platelet-Derived Growth Factor/antagonists & inhibitors , Primary Cell Culture , Up-Regulation , Ureter/surgery
4.
J Histochem Cytochem ; 66(1): 7-22, 2018 01.
Article in English | MEDLINE | ID: mdl-29077526

ABSTRACT

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


Subject(s)
Glomerular Mesangium/growth & development , Glomerular Mesangium/pathology , Glomerulonephritis/pathology , Receptors, CCR7/analysis , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Gene Deletion , Gene Expression Regulation, Developmental , Glomerular Mesangium/cytology , Glomerular Mesangium/ultrastructure , Glomerulonephritis/genetics , Kidney/cytology , Kidney/growth & development , Kidney/pathology , Kidney/ultrastructure , Male , Mice, Inbred C57BL , Rats, Wistar , Receptors, CCR7/genetics
5.
J Am Soc Nephrol ; 28(5): 1408-1420, 2017 May.
Article in English | MEDLINE | ID: mdl-27895155

ABSTRACT

For several decades, glucocorticoids have been used empirically to treat rapid progressive GN. It is commonly assumed that glucocorticoids act primarily by dampening the immune response, but the mechanisms remain incompletely understood. In this study, we inactivated the glucocorticoid receptor (GR) specifically in kidney epithelial cells using Pax8-Cre/GRfl/fl mice. Pax8-Cre/GRfl/fl mice did not exhibit an overt spontaneous phenotype. In mice treated with nephrotoxic serum to induce crescentic nephritis (rapidly progressive GN), this genetic inactivation of the GR in kidney epithelial cells exerted renal benefits, including inhibition of albuminuria and cellular crescent formation, similar to the renal benefits observed with high-dose prednisolone in control mice. However, genetic inactivation of the GR in kidney epithelial cells did not induce the immunosuppressive effects observed with prednisolone. In vitro, prednisolone and the pharmacologic GR antagonist mifepristone each acted directly on primary cultures of parietal epithelial cells, inhibiting cellular outgrowth and proliferation. In wild-type mice, pharmacologic treatment with the GR antagonist mifepristone also attenuated disease as effectively as high-dose prednisolone without the systemic immunosuppressive effects. Collectively, these data show that glucocorticoids act directly on activated glomerular parietal epithelial cells in crescentic nephritis. Furthermore, we identified a novel therapeutic approach in crescentic nephritis, that of glucocorticoid antagonism, which was at least as effective as high-dose prednisolone with potentially fewer adverse effects.


Subject(s)
Glomerulonephritis/drug therapy , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Animals , Epithelium , Kidney Glomerulus/drug effects , Kidney Glomerulus/physiopathology , Mice , Prednisolone , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/physiology
6.
J Am Soc Nephrol ; 27(12): 3678-3689, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27151923

ABSTRACT

DNA binding protein A (DbpA) is a member of the human cold shock domain-containing protein superfamily, with known functions in cell proliferation, differentiation, and stress responses. DbpA mediates tight junction-associated activities in tubular epithelial cells, but the function of DbpA in mesangial cells is unknown. Here, we found DbpA protein expression restricted to vascular smooth muscle cells in healthy human kidney tissue but profound induction of DbpA protein expression within the glomerular mesangial compartment in mesangioproliferative nephritis. In vitro, depletion or overexpression of DbpA using lentiviral constructs led to inhibition or promotion, respectively, of mesangial cell proliferation. Because platelet-derived growth factor B (PDGF-B) signaling has a pivotal role in mesangial cell proliferation, we examined the regulatory effect of PDGF-B on DbpA. In vitro studies of human and rat mesangial cells confirmed a stimulatory effect of PDGF-B on DbpA transcript numbers and protein levels. Additional in vivo investigations showed DbpA upregulation in experimental rat anti-Thy1.1 nephritis and murine mesangioproliferative nephritis models. To interfere with PDGF-B signaling, we injected nephritic rats with PDGF-B neutralizing aptamers or the MEK/ERK inhibitor U0126. Both interventions markedly decreased DbpA protein expression. Conversely, continuous PDGF-B infusion in healthy rats induced DbpA expression predominantly within the mesangial compartment. Taken together, these results indicate that DbpA is a novel target of PDGF-B signaling and a key mediator of mesangial cell proliferation.


Subject(s)
Cold Shock Proteins and Peptides/physiology , DNA-Binding Proteins/physiology , Glomerular Mesangium/pathology , Glomerular Mesangium/physiology , Glomerulonephritis/etiology , Mesangial Cells/pathology , Animals , Cell Proliferation , Cells, Cultured , Humans , Lupus Nephritis/etiology , Rats
7.
Biochem Biophys Res Commun ; 473(4): 1177-1184, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27074579

ABSTRACT

Mucociliary clearance requires the distinct orientation and coordinated movement of airway cilia, which is established through planar cell polarity signaling (PCP). The atypical cadherin Dachsous1 (Dchs1) is a transmembrane protein that regulates PCP in D. melanogaster. However, little is known about Dchs1 expression and its potential role in PCP in mammalian adult tissues. Here, we show that Dchs1 is ubiquitously expressed in mouse embryos, but exhibits a highly restricted expression to lung tissues in the adult stage. Strikingly, human Dchs1 localized exclusively to the base of the ciliary apparatus in cultured human respiratory epithelial cells with differentiated motile 9 + 2 cilia. This localization could be functionally important as we observed aberrant DCHS1 mRNA expression in human non-small cell lung cancer tissue. In sum, we establish Dchs1 as a component of the membrane domain surrounding the ciliary base. This suggests a specific role of Dchs1 in PCP-dependent organization of ciliary function and a possible role in lung disease.


Subject(s)
Aging/metabolism , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cilia/metabolism , Lung Neoplasms/metabolism , Respiratory Mucosa/metabolism , Aging/pathology , Animals , Cadherin Related Proteins , Carcinoma, Non-Small-Cell Lung/pathology , Cilia/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation, Developmental , Humans , Lung Neoplasms/pathology , Mice , Respiratory Mucosa/pathology , Subcellular Fractions/metabolism , Subcellular Fractions/pathology , Tissue Distribution
8.
J Am Soc Nephrol ; 27(1): 132-42, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26041841

ABSTRACT

The role of IL-6 signaling in renal diseases remains controversial, with data describing both anti-inflammatory and proinflammatory effects. IL-6 can act via classic signaling, engaging its two membrane receptors gp130 and IL-6 receptor (IL-6R). Alternatively, IL-6 trans-signaling requires soluble IL-6R (sIL-6R) to act on IL-6R-negative cells that express gp130. Here, we characterize the role of both pathways in crescentic nephritis. Patients with crescentic nephritis had significantly elevated levels of IL-6 in both serum and urine. Similarly, nephrotoxic serum-induced nephritis (NTN) in BALB/c mice was associated with elevated serum IL-6 levels. Levels of serum sIL-6R and renal downstream signals of IL-6 (phosphorylated signal transducer and activator of transcription 3, suppressor of cytokine signaling 3) increased over time in this model. Simultaneous inhibition of both IL-6 signaling pathways using anti-IL-6 antibody did not have a significant impact on NTN severity. In contrast, specific inhibition of trans-signaling using recombinant sgp130Fc resulted in milder disease. Vice versa, specific activation of trans-signaling using a recombinant IL-6-sIL-6R fusion molecule (Hyper-IL-6) significantly aggravated NTN and led to increased systolic BP in NTN mice. This correlated with increased renal mRNA synthesis of the Th17 cell cytokine IL-17A and decreased synthesis of resistin-like alpha (RELMalpha)-encoding mRNA, a surrogate marker of lesion-mitigating M2 macrophage subtypes. Collectively, our data suggest a central role for IL-6 trans-signaling in crescentic nephritis and offer options for more effective and specific therapeutic interventions in the IL-6 system.


Subject(s)
Glomerulonephritis/etiology , Interleukin-6/physiology , Animals , Humans , Male , Mice , Mice, Inbred BALB C , Signal Transduction
9.
Sci Rep ; 5: 14685, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26423228

ABSTRACT

Anti-cytokine therapies have substantially improved the treatment of inflammatory and autoimmune diseases. Cytokine-targeting drugs are usually biologics such as antibodies or other engineered proteins. Production of biologics, however, is complex and intricate and therefore expensive which might limit therapeutic application. To overcome this limitation we developed a strategy that involves the design of an optimized, monogenic cytokine inhibitor and the protein producing capacity of the host. Here, we engineered and characterized a receptor fusion protein, mIL-6-RFP-Fc, for the inhibition of interleukin-6 (IL-6), a well-established target in anti-cytokine therapy. Upon application in mice mIL-6-RFP-Fc inhibited IL-6-induced activation of the transcription factor STAT3 and ERK1/2 kinases in liver and kidney. mIL-6-RFP-Fc is encoded by a single gene and therefore most relevant for gene transfer approaches. Gene transfer through hydrodynamic plasmid delivery in mice resulted in hepatic production and secretion of mIL-6-RFP-Fc into the blood in considerable amounts, blocked hepatic acute phase protein synthesis and improved kidney function in an ischemia and reperfusion injury model. Our study establishes receptor fusion proteins as promising agents in anti-cytokine therapies through gene therapeutic approaches for future targeted and cost-effective treatments. The strategy described here is applicable for many cytokines involved in inflammatory and other diseases.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Interleukin-6/antagonists & inhibitors , Reperfusion Injury/drug therapy , Animals , Anti-Inflammatory Agents/pharmacokinetics , Cytokine Receptor gp130/genetics , Drug Evaluation, Preclinical , Gene Expression , HEK293 Cells , Hep G2 Cells , Humans , Immunoglobulin Fc Fragments/genetics , Interleukin-6/immunology , Kidney/blood supply , Kidney/drug effects , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Plasmids/genetics , Protein Interaction Domains and Motifs , Receptors, Interleukin-6/genetics , Recombinant Fusion Proteins/genetics , Transfection
10.
Am J Pathol ; 185(8): 2132-42, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26216283

ABSTRACT

We have identified platelet-derived growth factor (PDGF)-CC as a potent profibrotic mediator in kidney fibrosis and pro-angiogenic mediator in glomeruli. Because renal fibrosis is associated with progressive capillary rarefaction, we asked whether PDGF-CC neutralization in fibrosis might have detrimental anti-angiogenic effects leading to aggravated peritubular capillary loss. We analyzed capillary rarefaction in mice with and without PDGF-CC neutralization (using genetically deficient mice and neutralizing antibodies), in three different models of renal interstitial fibrosis, unilateral ureteral obstruction, unilateral ischemia-reperfusion, Col4a3-deficient (Alport) mice, and healthy animals. Independent of the effect of PDGF-CC neutralization on renal fibrosis, we found no difference in capillary rarefaction between PDGF-CC-neutralized mice and mice with intact PDGF-CC. We also found no differences in microvascular leakage (determined by extravasation of Evans Blue Dye) and in renal relative blood volume quantified using in vivo microcomputed tomography. PDGF-CC neutralization had no effects on renal microvasculature in healthy animals. Capillary endothelium did not express PDGF receptor-α, suggesting that potential PDGF-CC effects would have to be indirect. PDGF-CC neutralization or deficiency was not associated with preservation or accelerated loss of peritubular capillaries, suggesting no significant pro-angiogenic effects of PDGF-CC during renal fibrosis. From a clinical perspective, the profibrotic effects of PDGF-CC outweigh the pro-angiogenic effects and, thus, do not limit a potential therapeutic use of PDGF-CC inhibition in renal fibrosis.


Subject(s)
Capillaries/metabolism , Kidney Diseases/metabolism , Kidney/metabolism , Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Animals , Capillaries/pathology , Disease Models, Animal , Fibrosis/metabolism , Fibrosis/pathology , Kidney/pathology , Kidney Diseases/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lymphokines/genetics , Mice , Mice, Knockout , Platelet-Derived Growth Factor/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
11.
PLoS One ; 9(10): e109631, 2014.
Article in English | MEDLINE | ID: mdl-25310591

ABSTRACT

BACKGROUND: In clinical practice, there is a lack of markers for the non-invasive diagnosis and follow-up of kidney disease. Exosomes are membrane vesicles, which are secreted from their cells of origin into surrounding body fluids and contain proteins and mRNA which are protected from digestive enzymes by a cell membrane. METHODS: Toxic podocyte damage was induced by puromycin aminonucleoside in rats (PAN). Urinary exosomes were isolated by ultracentrifugation at different time points during the disease. Exosomal mRNA was isolated, amplified, and the mRNA species were globally assessed by gene array analysis. Tissue-specific gene and protein expression was assessed by RT-qPCR analysis and immunohistochemistry. RESULTS: Gene array analysis of mRNA isolated from urinary exosomes revealed cystatin C mRNA as one of the most highly regulated genes. Its gene expression increased 7.5-fold by day 5 and remained high with a 1.9-fold increase until day 10. This was paralleled by a 2-fold increase in cystatin C mRNA expression in the renal cortex. Protein expression in the kidneys also dramatically increased with de novo expression of cystatin C in glomerular podocytes in parts of the proximal tubule and the renal medulla. Urinary excretion of cystatin C increased approximately 2-fold. CONCLUSION: In this proof-of-concept study, we could demonstrate that changes in urinary exosomal cystatin C mRNA expression are representative of changes in renal mRNA and protein expression. Because cells lining the urinary tract produce urinary exosomal cystatin C mRNA, it might be a more specific marker of renal damage than glomerular-filtered free cystatin C.


Subject(s)
Cystatin C/urine , Exosomes/drug effects , Gene Expression/drug effects , Kidney/drug effects , Podocytes/drug effects , Puromycin Aminonucleoside/toxicity , Animals , Biomarkers , Cystatin C/genetics , Exosomes/genetics , Kidney/metabolism , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Male , Podocytes/metabolism , RNA, Messenger/urine , Rats , Rats, Sprague-Dawley , Up-Regulation
12.
Am J Physiol Renal Physiol ; 307(3): F346-55, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24899055

ABSTRACT

Renal inflammation, in particular glomerular, is often characterized by increased IL-6 levels. The in vivo relevance of IL-6 signaling in glomerular podocytes, which play central roles in most glomerular diseases, is unknown. Here, we show that in normal mice, podocytes express gp130, the common signal-transducing receptor subunit of the IL-6 family of cytokines. Following systemic IL-6 or LPS injection in mice, podocyte IL-6 signaling was evidenced by downstream STAT3 phosphorylation. Next, we generated mice deficient for gp130 in podocytes. Expectedly, these mice exhibited abrogated IL-6 downstream signaling in podocytes. At the age of 40 wk, they did not show spontaneous renal pathology or abnormal renal function. The mice were then challenged using two LPS injury models as well as nephrotoxic serum to induce crescentic nephritis. Under all conditions, circulating IL-6 levels increased markedly and the mice developed the pathological hallmarks of the corresponding injury models such as proteinuria and development of glomerular crescents, respectively. However, despite the capacity of normal podocytes to transduce IL-6 family signals downstream, there were no significant differences between mice bearing the podocyte-specific gp130 deletion and their control littermates in any of these models. In conclusion, under the different conditions tested, gp130 signaling was not a critical component of the (patho-)biology of the podocyte in vivo.


Subject(s)
Glycoproteins/metabolism , Interleukin-6/metabolism , Podocytes/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Disease Models, Animal , Female , Gene Deletion , Glycoproteins/genetics , Interleukin-6/genetics , Lipopolysaccharides/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nephritis/chemically induced , Nephritis/metabolism , Nephritis/pathology , Phosphorylation , Podocytes/pathology , STAT3 Transcription Factor/metabolism
13.
PLoS One ; 9(3): e92115, 2014.
Article in English | MEDLINE | ID: mdl-24667162

ABSTRACT

Mesenchymal stem cell (MSC) transplantation has the potential for organ repair. Nevertheless, some factors might lessen the regenerative potential of MSCs, e.g. donor age or systemic disease. It is thus important to carefully assess the patient's suitability for autologous MSC transplantation. Here we investigated the effects of chronic kidney disease (CKD) on MSC function. We isolated bone marrow MSCs from remnant kidney rats (RK) with CKD (CKD-RK-MSC) and found signs of premature senescence: spontaneous adipogenesis, reduced proliferation capacity, active senescence-associated-ß-galactosidase, accumulation of actin and a modulated secretion profile. The functionality of CKD-RK-MSCs in vivo was tested in rats with acute anti-Thy1.1-nephritis, where healthy MSCs have been shown to be beneficial. Rats received healthy MSCs, CKD-RK-MSC or medium by injection into the left renal artery. Kidneys receiving healthy MSCs exhibited accelerated healing of glomerular lesions, whereas CKD-RK-MSC or medium exerted no benefit. The negative influence of advanced CKD/uremia on MSCs was confirmed in a second model of CKD, adenine nephropathy (AD). MSCs from rats with adenine nephropathy (CKD-AD-MSC) also exhibited cellular modifications and functional deficits in vivo. We conclude that CKD leads to a sustained loss of in vitro and in vivo functionality in MSCs, possibly due to premature cellular senescence. Considering autologous MSC therapy in human renal disease, studies identifying uremia-associated mechanisms that account for altered MSC function are urgently needed.


Subject(s)
Cellular Senescence/physiology , Kidney Glomerulus/pathology , Mesenchymal Stem Cells/cytology , Nephritis/pathology , Regeneration/physiology , Renal Insufficiency, Chronic/pathology , Animals , Blotting, Western , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Fibroblasts/cytology , Humans , Immunoenzyme Techniques , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mesenchymal Stem Cell Transplantation , Nephritis/immunology , Nephritis/therapy , RNA, Messenger/genetics , Rats , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction , Renal Insufficiency, Chronic/therapy , Reverse Transcriptase Polymerase Chain Reaction , Thy-1 Antigens/immunology , Tissue Donors
14.
Kidney Int Suppl (2011) ; 4(1): 65-69, 2014 Nov.
Article in English | MEDLINE | ID: mdl-26312152

ABSTRACT

Renal fibrosis is the hallmark of chronic kidney disease progression and is characterized by an exaggerated wound-healing process with the production of renal scar tissue. It comprises both the glomerular and the tubulointerstitial compartments. Among the factors that contribute to kidney fibrosis, the members of the platelet-derived growth factor (PDGF) family are among the best characterized ones. They appear to be the key factors in driving renal fibrosis, independent of the underlying kidney disease. The PDGF family consists of four isoforms (PDGF-A, -B, -C, and -D) and two receptor chains (PDGFR-α and -ß), which are constitutively or inducibly expressed in most renal cells. These components have an irreplaceable role in kidney development by recruitment of mesenchymal cells to the glomerular and tubulointerstitial compartments. They further regulate multiple pathophysiologic processes including cell proliferation, cell migration, expression and accumulation of extracellular matrix, production and secretion of pro- and anti-inflammatory mediators, vascular permeability, and hemodynamics. This review provides a brief update on the role of different PDGF isoforms in the development of glomerulosclerosis and tubulointerstitial fibrosis, newly identified endogeneous PDGF antagonists, and resulting potential therapies.

15.
Cell Commun Signal ; 11: 63, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-24103640

ABSTRACT

BACKGROUND: The Y-box protein-1 (YB-1) fulfills pleiotropic functions relating to gene transcription, mRNA processing, and translation. It remains elusive how YB-1 shuttling into the nuclear and cytoplasmic compartments is regulated and whether limited proteolysis by the 20S proteasome releases fragments with distinct function(s) and subcellular distribution(s). RESULTS: To address these questions, mapping of domains responsible for subcellular targeting was performed. Three nuclear localization signals (NLS) were identified. NLS-1 (aa 149-156) and NLS-2 (aa 185-194) correspond to residues with unknown function(s), whereas NLS-3 (aa 276-292) matches with a designated multimerization domain. Nuclear export signal(s) were not identified. Endoproteolytic processing by the 20S proteasome before glycine 220 releases a carboxy-terminal fragment (CTF), which localized to the nucleus, indicating that NLS-3 is operative. Genotoxic stress induced proteolytic cleavage and nuclear translocation of the CTF. Co-expression of the CTF and full-length YB-1 resulted in an abrogated transcriptional activation of the MMP-2 promoter, indicating an autoregulatory inhibitory loop, whereas it fulfilled similar trans-repressive effects on the collagen type I promoter. CONCLUSION: Compartmentalization of YB-1 protein derivatives is controlled by distinct NLS, one of which targets a proteolytic cleavage product to the nucleus. We propose a model for an autoregulatory negative feedback loop that halts unlimited transcriptional activation.


Subject(s)
Y-Box-Binding Protein 1/metabolism , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Humans , Mesangial Cells/metabolism , Nuclear Export Signals , Nuclear Localization Signals/metabolism , Protein Structure, Tertiary , Proteolysis , Rats , Transcription, Genetic , Y-Box-Binding Protein 1/chemistry
16.
J Am Soc Nephrol ; 24(12): 1966-80, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23970123

ABSTRACT

Under physiologic conditions, significant amounts of plasma protein pass the renal filter and are reabsorbed by proximal tubular cells, but it is not clear whether the endocytosed protein, particularly albumin, is degraded in lysosomes or returned to the circulatory system intact. To resolve this question, a transgenic mouse with podocyte-specific expression of doxycycline-inducible tagged murine albumin was developed. To assess potential glomerular backfiltration, two types of albumin with different charges were expressed. On administration of doxycycline, podocytes expressed either of the two types of transgenic albumin, which were secreted into the primary filtrate and reabsorbed by proximal tubular cells, resulting in serum accumulation. Renal transplantation experiments confirmed that extrarenal transcription of transgenic albumin was unlikely to account for these results. Genetic deletion of the neonatal Fc receptor (FcRn), which rescues albumin and IgG from lysosomal degradation, abolished transcytosis of both types of transgenic albumin and IgG in proximal tubular cells. In summary, we provide evidence of a transcytosis within the kidney tubular system that protects albumin and IgG from lysosomal degradation, allowing these proteins to be recycled intact.


Subject(s)
Albuminuria/metabolism , Kidney Tubules, Proximal/metabolism , Models, Biological , Serum Albumin/metabolism , Transcytosis/physiology , Animals , Anti-Bacterial Agents/pharmacology , Doxycycline/pharmacology , Endocytosis/physiology , Gene Expression/drug effects , Humans , Immunoglobulin G/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Kidney Transplantation , Lysosomes/metabolism , Membrane Proteins/genetics , Mice , Mice, Transgenic , Podocytes/metabolism , Protein Structure, Tertiary , Rats , Rats, Transgenic , Serum Albumin/chemistry , Serum Albumin/genetics
17.
J Immunol ; 191(5): 2604-13, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23872051

ABSTRACT

In vitro studies identified Y-box-binding protein (YB)-1 as a key regulator of inflammatory mediators. In this study, we observed increased levels of secreted YB-1 in sera from sepsis patients. This led us to investigate the in vivo role of YB-1 in murine models of acute peritonitis following LPS injection, in sterile renal inflammation following unilateral ureteral obstruction, and in experimental pyelonephritis. LPS injection enhanced de novo secretion of YB-1 into the urine and the peritoneal fluid of LPS-treated mice. Furthermore, we could demonstrate a significant, transient upregulation and posttranslational modification (phosphorylation at serine 102) of YB-1 in renal and inflammatory cells. Increased renal cytoplasmic YB-1 amounts conferred enhanced expression of proinflammatory chemokines CCL2 and CCL5. Along these lines, heterozygous YB-1 knockout mice (YB-1(+/d)) that display 50% reduced YB-1 levels developed significantly lower responses to both LPS and sterile inflammation induced by unilateral ureteral obstruction. This included diminished immune cell numbers due to impaired migration propensities and reduced chemokine expression. YB-1(+/d) mice were protected from LPS-associated mortality (20% mortality on day 3 versus 80% in wild-type controls); however, immunosuppression in YB-1(+/d) animals resulted in 50% mortality. In conclusion, our findings identify YB-1 as a major, nonredundant mediator in both systemic and local inflammatory responses.


Subject(s)
Inflammation/immunology , Sepsis/immunology , Transcription Factors/immunology , Animals , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Immunoprecipitation , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nephritis/immunology , Nephritis/metabolism , Peritonitis/immunology , Peritonitis/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/metabolism , Transcription Factors/metabolism
18.
Nephrol Dial Transplant ; 28(4): 889-900, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23229927

ABSTRACT

BACKGROUND: Platelet-derived growth factors (PDGF)-AA and -CC mediate renal fibroblast proliferation and/or renal fibrosis. Whereas PDGF-CC binds to both the PDGF receptors (PDGFRs)-αα- and -αß, PDGF-AA binds more selectively to the αα-receptor, suggesting potential differences in the biological activities. METHODS: We compared signal transduction, gene expression as well as changes in the proteome induced by PDGF-AA and -CC in rat renal fibroblasts, which express both PDGFR subunits. The growth factor concentrations used were chosen based on their equipotency in inducing rat renal fibroblast proliferation. RESULTS: Both PDGF-AA and PDGF-CC induced phosphorylation and activation of extracellular signal-regulated kinase 1 (ERK1) and ERK2. Renal fibroblast proliferation induced by either PDGF-AA or -CC could be blocked by signal transduction inhibitors of the mitogen-activated protein kinase (MAPK)-, Janus-kinase (JAK)/signal transducers and activators of transcription (STAT) and phosphatidyl-inositol-3-kinase (PI3K) pathway, pointing to the involvement of all the three pathways. However, quantitative differences between both the stimulations were minor. Additive or synergistic effects by stimulating simultaneously with PDGF-AA and -CC were not observed. Using a proteomic approach we found eleven differentially expressed proteins, which were quantitatively altered after treatment with either PDGF-AA or PDGF-CC. The regulation of calreticulin and inorganic pyrophosphatase 1 could be verified by western blotting. CONCLUSIONS: PDGF-AA and -CC exhibit almost identical biological effects on signal transduction and proteome in cultured renal fibroblasts, suggesting that the ligands exert their activity essentially through the commonly bound PDGFR-αα. Nonetheless, two differentially expressed proteins were identified which might be involved in the development of renal failure.


Subject(s)
Fibroblasts/metabolism , Kidney/metabolism , Lymphokines/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Platelet-Derived Growth Factor/metabolism , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , Electrophoretic Mobility Shift Assay , Fibroblasts/cytology , Kidney/cytology , Lymphokines/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Phosphorylation , Platelet-Derived Growth Factor/genetics , Proteomics , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
Am J Pathol ; 182(1): 107-17, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23141925

ABSTRACT

Platelet-derived growth factors (PDGF) are key mediators of organ fibrosis. We investigated whether PDGF-C(-/-) mice or mice treated with neutralizing PDGF-C antibodies are protected from bile duct ligation-induced liver fibrosis, and we compared the effects with those of PDGF-C deficiency or neutralization on kidney fibrosis induced by unilateral ureteral obstruction. Unexpectedly, and in contrast to kidney fibrosis, PDGF-C deficiency or antagonism did not protect from liver fibrosis or functional liver impairment. Furthermore, the hepatic infiltration of monocytes/macrophages/dendritic cells and chemokine mRNA expression (CC chemokine ligand [CCL]5, CCL2, and CC chemokine receptor 2 [CCR2]) remained unchanged. Transcript expression of PDGF ligands increased in both liver and kidney fibrosis and was not affected by neutralization of PDGF-C. In kidney fibrosis, PDGF-C deficiency or antagonism led to reduced expression and signaling of PDGF-receptor (R)-α- and PDGFR-ß-chains. In contrast, in liver fibrosis there was either no difference (PDGF-C(-/-) mice) or even an upregulation of PDGFR-ß and signaling (anti-PDGF-C group). Finally, in vitro studies in portal myofibroblasts pointed to a predominant role of PDGF-B and PDGF-D signaling in liver fibrosis. In conclusion, our study revealed significant differences between kidney and liver fibrosis in that PDGF-C mediates kidney fibrosis, whereas antagonism of PDGF-C in liver fibrosis appears to be counteracted by significant upregulation and increased PDGFR-ß signaling. PDGF-C antagonism, therefore, may not be effective to treat liver fibrosis.


Subject(s)
Kidney/pathology , Liver Cirrhosis/metabolism , Lymphokines/physiology , Platelet-Derived Growth Factor/physiology , Receptors, Platelet-Derived Growth Factor/physiology , Animals , Cells, Cultured , Disease Models, Animal , Fibrosis , Liver Cirrhosis/etiology , Liver Cirrhosis/prevention & control , Lymphokines/antagonists & inhibitors , Lymphokines/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/metabolism , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/deficiency , Rats , Rats, Sprague-Dawley , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction/physiology , Up-Regulation/physiology , Ureteral Obstruction/complications
20.
Kidney Int ; 83(2): 251-63, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23254899

ABSTRACT

Growth arrest-specific protein-1 (GAS1) is a GPI-anchored protein which is highly expressed in embryonic mouse fibroblasts and inhibits their proliferation. Glomerular mesangial cells release soluble GAS1 protein into the supernatant in vitro. Growth arrest led to GAS1 overexpression and increased release. Secretion involved disintegrin and metalloproteinase 10 and 17 as signified by inhibition experiments. Recombinant soluble GAS1 protein inhibited the proliferation of mesangial cells. Conversely, the induction of mesangial cell proliferation by PDGF-BB or -DD led to downregulation of GAS1 mRNA. Specific ligands of the PDGF α-receptor, PDGF-AA and -CC, had no effect. The GAS1 protein was localized in podocytes in kidneys from healthy rats. During the time course of mesangioproliferative glomerulonephritis in anti-Thy1.1-treated rats, glomerular GAS1 expression decreased prior to the onset of mesangial cell proliferation and increased at later stages during glomerular recovery. Finally, a plasmid expressing soluble GAS1 fused to an Fc fragment was systemically overexpressed in rats with mesangioproliferative glomerulonephritis. This ameliorated renal damage was indicated by decreased albuminuria and serum creatinine. Gas1/Fc-transfected rats also exhibited a reduction of the glomerular mesangial cell activation and proliferation. Thus, GAS1 is a novel endogenous inhibitor of glomerular mesangial cell proliferation and may be a novel therapeutic target in mesangioproliferative glomerular diseases.


Subject(s)
Cell Cycle Proteins/physiology , Mesangial Cells/physiology , ADAM Proteins/physiology , ADAM10 Protein , ADAM17 Protein , Amyloid Precursor Protein Secretases/physiology , Animals , Becaplermin , Cell Cycle Proteins/genetics , Cell Proliferation , Cells, Cultured , GPI-Linked Proteins/genetics , GPI-Linked Proteins/physiology , Humans , Isoantibodies/pharmacology , Lymphokines/pharmacology , Membrane Proteins/physiology , Mice , Oligonucleotide Array Sequence Analysis , Platelet-Derived Growth Factor/pharmacology , Podocytes/metabolism , Proto-Oncogene Proteins c-sis/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...