Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Case Rep Nephrol Dial ; 12(1): 63-72, 2022.
Article in English | MEDLINE | ID: mdl-35702378

ABSTRACT

Immunotactoid glomerulopathy (ITG) occurs infrequently and is characterized by organized IgG containing deposits. It most usually manifests as a concomitant disease of a broad spectrum of oncologic entities. We here present an exceptional case of ITG without glomerular light chain restriction secondary to a IgM kappa type monoclonal gammopathy of undetermined significance. Due to nephrotic syndrome and deterioration of kidney function a rituximab monotherapy was initiated without targeting the plasmacellular augmentation, which was confirmed as the underlying process. The treatment led to a long-term improvement of proteinuria and stabilization of glomerular filtration rate. Its therapeutic effect has to be attributed to immunomodulatory capacities and targeting of podocytes rather than to be interpreted as directed against a bone marrow or glomerular clone. We conclude that rituximab therapy may be a valuable part of the therapeutic options in ITG irrespective of the underlying oncologic entity.

2.
J Clin Med ; 11(3)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35160109

ABSTRACT

Kidney transplant (KTx) recipients are a high-risk population for osteoporotic fractures. We herein aim to identify the role of pre-transplant parathyroidectomy (PTX) and other modifiable factors associated with osteoporotic fractures in KTx recipients. We conducted a retrospective study involving 711 adult patients (4608 patient-years) who were transplanted at our center between January 2007 and June 2015. Clinical data were extracted from patients' electronic medical records. Different laboratory and clinical parameters for mineral bone disease (MBD) and osteoporosis, including medication, were evaluated. We chose fracture events unrelated to malignancies or adequate trauma as the primary endpoint. Osteoporotic fractures occurred in 47 (6.6%) patients (median 36.7 months, IQR 45.9) after KTx (fracture incidence of 10 per 1000 person-years). Prior to KTx, subtotal PTX was performed in 116 patients (16.3%, median time 4.2 years before KTx, IQR 5.0). Of the patients with fracture (n = 47), only one (2.2%) patient had previously undergone PTX. After adjusting for the known fracture risk factors MBD and osteoporosis, PTX remained a protective factor against fractures (HR 0.134, CI 0.018-0.991, p = 0.049). We observed a reduced risk for pathological fractures in KTx patients who underwent PTX, independent from elevated parathyroid hormone at the time of KTx or afterwards.

3.
Cell Death Dis ; 12(1): 117, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33483469

ABSTRACT

The WW-and-C2-domain-containing (WWC) protein family is involved in the regulation of cell differentiation, cell proliferation, and organ growth control. As upstream components of the Hippo signaling pathway, WWC proteins activate the Large tumor suppressor (LATS) kinase that in turn phosphorylates Yes-associated protein (YAP) and its paralog Transcriptional coactivator-with-PDZ-binding motif (TAZ) preventing their nuclear import and transcriptional activity. Inhibition of WWC expression leads to downregulation of the Hippo pathway, increased expression of YAP/TAZ target genes and enhanced organ growth. In mice, a ubiquitous Wwc1 knockout (KO) induces a mild neurological phenotype with no impact on embryogenesis or organ growth. In contrast, we could show here that ubiquitous deletion of Wwc2 in mice leads to early embryonic lethality. Wwc2 KO embryos display growth retardation, a disturbed placenta development, impaired vascularization, and finally embryonic death. A whole-transcriptome analysis of embryos lacking Wwc2 revealed a massive deregulation of gene expression with impact on cell fate determination, cell metabolism, and angiogenesis. Consequently, a perinatal, endothelial-specific Wwc2 KO in mice led to disturbed vessel formation and vascular hypersprouting in the retina. In summary, our data elucidate a novel role for Wwc2 as a key regulator in early embryonic development and sprouting angiogenesis in mice.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Embryonic Development/physiology , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Differentiation/physiology , Female , Hippo Signaling Pathway , Male , Mice , Mice, Knockout , Neovascularization, Physiologic/physiology , Signal Transduction
4.
Thromb Haemost ; 119(11): 1827-1838, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31493777

ABSTRACT

The endothelial glycocalyx (eGC), a carbohydrate-rich layer lining the luminal surface of the endothelium, provides a first vasoprotective barrier against vascular leakage in sepsis. We hypothesized that angiopoietin-2 (Angpt-2), antagonist of the endothelium-stabilizing receptor Tie2, induces a rapid loss of the eGC in human sepsis. Using intravital microscopy, we measured the perfused boundary region (PBR), an inverse parameter of eGC dimensions in sublingual microvessels, in patients with sepsis and age-matched nonseptic subjects. Median PBR values were significantly higher in patients compared with controls and correlated with serum Angpt-2 levels. To transfer and further explore these findings in a cell culture system, we exposed endothelial cells (ECs) to serum (5%) from a subgroup of septic patients and nonseptic controls. Confocal and atomic force microscopy revealed that sepsis serum, but not control serum, induced thinning of the eGC on human ECs in vitro, which correlated with paired PBR values obtained in vivo (r = 0.96, p < 0.01). Inhibition of Angpt-2 or Tie2 activation completely abolished eGC damage. Mechanistically, sepsis-induced eGC breakdown required the loss of its main constituent heparan sulfate; a result of heparan sulfate-specific enzyme heparanase, which was suppressed by Tie2 activation. Finally, Tie2 activation, but not Angpt-2 inhibition, initiated after septic or enzymatic damage provoked rapid refurbishment of the eGC. Our data indicate that eGC breakdown in human sepsis is mediated via Tie2 deactivation by Angpt-2. Activation of Tie2 seems to accelerate recovery of the eGC and might hold promise as a therapeutic target in human sepsis.


Subject(s)
Endothelial Cells/metabolism , Glycocalyx/metabolism , Receptor, TIE-2/metabolism , Sepsis/metabolism , Adult , Aged , Angiopoietin-2/blood , Case-Control Studies , Cell Line , Endothelial Cells/pathology , Female , Glucuronidase/metabolism , Glycocalyx/pathology , Heparitin Sulfate/metabolism , Humans , Intravital Microscopy , Male , Microscopy, Atomic Force , Middle Aged , Phosphorylation , Prospective Studies , Sepsis/blood , Sepsis/pathology , Signal Transduction
6.
Cell Death Dis ; 9(9): 850, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30154411

ABSTRACT

Podocytes are crucial for the establishment of the blood-urine filtration barrier in the glomeruli of the kidney. These cells are mainly affected during glomerulopathies causing proteinuria and kidney function impairment. Ongoing podocyte injury leads to podocyte loss, finally followed by end-stage kidney disease. Podocytes display a predominant nuclear localization of YAP (Yes-associated protein), one effector protein of the Hippo pathway, which regulates the balance between proliferation, differentiation, and apoptosis in cells. Nuclear active YAP seems to be critical for podocyte survival in vivo and in vitro. We can show here that different treatments leading to sequestration of YAP into the cytoplasm in podocytes, like decreased rigidity of the substrate, incubation with dasatinib, or overexpression of Hippo pathway members result in the induction of apoptosis. A RNA sequencing analysis of large tumor suppressor kinase 2 (LATS2) overexpressing podocytes confirmed a significant upregulation of apoptotic genes. The downregulation of Hippo pathway components suggests a feedback mechanism in podocytes. Noteworthy was the regulation of genes involved in cell-cell junction, the composition of the extracellular space, and cell migration. This suggests an influence of Hippo pathway activity on podocyte integrity. As focal segmental glomerulopathy (FSGS) goes along with an activation of the Hippo pathway in podocytes, a comparison of our data with two independent studies of transcriptional regulation in human FSGS glomeruli obtained from the Nephroseq database was performed. This comparison affirmed a multitude of consistent transcriptional changes concerning the regulation of genes influencing apoptosis and the Hippo signaling pathway as well as cell junction formation and cell migration. The link between Hippo pathway activation in podocytes and the regulation of junction and migration processes in vivo might be a fundamental mechanism of glomerular sclerosis and loss of renal function.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Nucleus/metabolism , Phosphoproteins/metabolism , Podocytes/metabolism , Protein Transport/physiology , Apoptosis/physiology , Cell Line , Cell Movement/physiology , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Kidney/metabolism , Renal Insufficiency/metabolism , Signal Transduction/physiology , Transcription Factors , Transcription, Genetic/physiology , Tumor Suppressor Proteins/metabolism , Up-Regulation/physiology , YAP-Signaling Proteins
7.
Hepatology ; 67(4): 1546-1559, 2018 04.
Article in English | MEDLINE | ID: mdl-29116649

ABSTRACT

The Hippo pathway regulates cell differentiation, proliferation, and apoptosis. Upon activation, it inhibits the import of the transcriptional coactivator yes-associated protein (YAP) into the nucleus, thus suppressing transcription of pro-proliferative genes. Hence, dynamic and precise control of the Hippo pathway is crucial for organ size control and the prevention of tumor formation. Hippo signaling is controlled by a growing number of upstream regulators, including WW and C2 domain-containing (WWC) proteins, which trigger a serine/threonine kinase pathway. One component of this is the large tumor suppressor (LATS) kinase, which phosphorylates YAP, trapping it in the cytoplasm. WWC proteins have been shown to interact with LATS in vitro and stimulate its kinase activity, thus directly promoting cytoplasmic accumulation of phosphorylated YAP. However, the function of the WWC proteins in the regulation of cell proliferation, organ size control, and tumor prevention in vivo has not yet been determined. Here, we show that loss of hepatic WWC expression in mice leads to tissue overgrowth, inflammation, fibrosis, and formation of liver carcinoma. WWC-deficient mouse livers display reduced LATS activity, increased YAP-mediated gene transcription, and enhanced proliferation of hepatic progenitor cells. In addition, loss of WWC expression in the liver accelerates the turnover of angiomotin proteins, which act as negative regulators of YAP activity. CONCLUSION: Our data define an essential in vivo function for WWC proteins as regulators of canonical and noncanonical Hippo signaling in hepatic cell growth and liver tumorigenesis. Thus, expression of WWC proteins may serve as novel prognostic factors in human liver carcinoma. (Hepatology 2018;67:1546-1559).


Subject(s)
Carcinogenesis/metabolism , Hepatocytes/metabolism , Liver Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Fluorescent Antibody Technique , Genotyping Techniques , Hippo Signaling Pathway , Immunohistochemistry , In Situ Hybridization , Liver/pathology , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Two-Hybrid System Techniques
8.
J Am Soc Nephrol ; 28(7): 2093-2107, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28154200

ABSTRACT

The nephron is the basic physiologic subunit of the mammalian kidney and is made up of several apicobasally polarized epithelial cell types. The process of apicobasal polarization in animal cells is controlled by the evolutionarily conserved Crumbs (CRB), Partitioning-defective, and Scribble protein complexes. Here, we investigated the role of protein associated with LIN-7 1 (Pals1, also known as Mpp5), a core component of the apical membrane-determining CRB complex in the nephron. Pals1 interacting proteins, including Crb3 and Wwtr1/Taz, have been linked to renal cyst formation in mice before. Immunohistologic analysis revealed Pals1 expression in renal tubular cells and podocytes of human kidneys. Mice lacking one Pals1 allele (functionally haploid for Pals1) in nephrons developed a fully penetrant phenotype, characterized by cyst formation and severe defects in renal barrier function, which led to death within 6-8 weeks. In Drosophila nephrocytes, deficiency of the Pals1 ortholog caused alterations in slit-diaphragm-like structures. Additional studies in epithelial cell culture models revealed that Pals1 functions as a dose-dependent upstream regulator of the crosstalk between Hippo- and TGF-ß-mediated signaling. Furthermore, Pals1 haploinsufficiency in mouse kidneys associated with the upregulation of Hippo pathway target genes and marker genes of TGF-ß signaling, including biomarkers of renal diseases. These findings support a link between apical polarity proteins and renal diseases, especially renal cyst diseases. Further investigation of the Pals1-linked networks is required to decipher the mechanisms underlying the pathogenesis of these diseases.


Subject(s)
Haploinsufficiency , Kidney Diseases, Cystic/genetics , Membrane Proteins/genetics , Nucleoside-Phosphate Kinase/genetics , Proteinuria/genetics , Animals , Drosophila , Female , Male , Mice
9.
Mol Biol Evol ; 31(7): 1710-23, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24682284

ABSTRACT

The scaffolding protein KIBRA (also called WWC1) is involved in the regulation of important intracellular transport processes and the establishment of cell polarity. Furthermore, KIBRA/WWC1 is an upstream regulator of the Hippo signaling pathway that controls cell proliferation and organ size in animals. KIBRA/WWC1 represents only one member of the WWC protein family that also includes the highly similar proteins WWC2 and WWC3. Although the function of KIBRA/WWC1 was studied intensively in cells and animal models, the importance of WWC2 and WWC3 was not yet elucidated. Here, we describe evolutionary, molecular, and functional aspects of the WWC family. We show that the WWC genes arose in the ancestor of bilateral animals (clades such as insects and vertebrates) from a single founder gene most similar to the present KIBRA/WWC1-like sequence of Drosophila. This situation was still maintained until the common ancestor of lancelet and vertebrates. In fish, a progenitor-like sequence of mammalian KIBRA/WWC1 and WWC2 is expressed together with WWC3. Finally, in all tetrapods, the three family members, KIBRA/WWC1, WWC2, and WWC3, are found, except for a large genomic deletion including WWC3 in Mus musculus. At the molecular level, the highly conserved WWC proteins share a similar primary structure, the ability to form homo- and heterodimers and the interaction with a common set of binding proteins. Furthermore, all WWC proteins negatively regulate cell proliferation and organ growth due to a suppression of the transcriptional activity of YAP, the major effector of the Hippo pathway.


Subject(s)
Carrier Proteins/genetics , Phosphoproteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Carrier Proteins/metabolism , Cell Proliferation , Evolution, Molecular , HEK293 Cells , Humans , Multigene Family , Organ Specificity , Phosphoproteins/metabolism , Phylogeny , Sequence Deletion , Signal Transduction , Tumor Suppressor Proteins/metabolism
10.
Cell Signal ; 26(7): 1392-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24642126

ABSTRACT

In mammals, the KIBRA locus has been associated with memory performance and cognition by genome-wide single nucleotide polymorphism screening. Genetic studies in Drosophila and human cells have identified KIBRA as a novel regulator of the Hippo signaling pathway, which plays a critical role in human tumorigenesis. Recent studies also indicated that KIBRA is involved in other physiological processes including cell polarity, membrane/vesicular trafficking, mitosis and cell migration. At the biochemical level, KIBRA protein is highly phosphorylated by various kinases in epithelial cells. Here, we discuss the updates concerning the function and regulation of KIBRA in the brain and beyond.


Subject(s)
Brain/metabolism , Carcinogenesis/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kidney/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Amino Acid Sequence , Animals , Cell Movement/genetics , Cell Polarity/genetics , Cognition/physiology , Drosophila , Hippo Signaling Pathway , Humans , Memory/physiology , Mice , Mitosis/genetics , Molecular Sequence Data , Protein Serine-Threonine Kinases/genetics , Protein Transport/genetics
11.
J Neurochem ; 128(5): 686-700, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24117625

ABSTRACT

The WWC1 gene has been genetically associated with human episodic memory performance, and its product KIdney/BRAin protein (KIBRA) has been shown to interact with the atypical protein kinase protein kinase M ζ (PKMζ). Although recently challenged, PKMζ remains a candidate postsynaptic regulator of memory maintenance. Here, we show that PKMζ is subject to rapid proteasomal degradation and that KIBRA is both necessary and sufficient to counteract this process, thus stabilizing the kinase and maintaining its function for a prolonged time. We define the binding sequence on KIBRA, a short amino acid motif near the C-terminus. Both hippocampal knock-down of KIBRA in rats and KIBRA knock-out in mice result in decreased learning and memory performance in spatial memory tasks supporting the notion that KIBRA is a player in episodic memory. Interestingly, decreased memory performance is accompanied by decreased PKMζ protein levels. We speculate that the stabilization of synaptic PKMζ protein levels by KIBRA may be one mechanism by which KIBRA acts in memory maintenance. KIBRA/WWC1 has been genetically associated with human episodic memory. KIBRA has been shown to be post-synaptically localized, but its function remained obscure. Here, we show that KIBRA shields PKMζ, a kinase previously linked to memory maintenance, from proteasomal degradation via direct interaction. KIBRA levels in the rodent hippocampus correlate closely both to spatial memory performance in rodents and to PKMζ levels. Our findings support a role for KIBRA in memory, and unveil a novel function for this protein.


Subject(s)
Carrier Proteins/physiology , Co-Repressor Proteins/physiology , Learning/physiology , Memory/physiology , Protein Kinase C/physiology , Amino Acid Sequence , Animals , Avoidance Learning/physiology , Behavior, Animal/physiology , Blotting, Western , Carrier Proteins/metabolism , Co-Repressor Proteins/metabolism , Dependovirus/genetics , Genetic Complementation Test , Hippocampus/metabolism , Hippocampus/physiology , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Molecular Sequence Data , Phosphoproteins , Polymerase Chain Reaction , Protein Binding , Protein Kinase C/metabolism , Rats , Rats, Wistar , Stereotaxic Techniques
12.
Cell Signal ; 26(2): 343-51, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24269383

ABSTRACT

In mammals, KIBRA is defined as a memory performance-associated protein. The physiological function and regulation of KIBRA in non-neuronal cells are much less understood. Recent studies have identified KIBRA as a novel regulator of the Hippo signaling pathway, which plays a critical role in tumorigenesis by inhibiting cell proliferation and promoting apoptosis. We recently reported that KIBRA is phosphorylated by the mitotic kinases Aurora and cyclin-dependent kinase 1 during mitosis. In this current study, we show that KIBRA is also phosphorylated by the ERK (extracellular signal-regulated kinases)-RSK (p90 ribosomal S6 kinases) cascade. We demonstrated that ERK1/2 phosphorylate KIBRA at Ser(548) in cells as well as in vitro. Moreover, we found that RSK1/2 specifically phosphorylates KIBRA at two highly conserved sites (Thr(929) and Ser(947)) in vitro and in cells. RSK-mediated phosphorylation is required for KIBRA binding to RSK1, but not RSK2. Surprisingly, KIBRA knockdown impaired cell migration and proliferation in breast cancer cells. By using inducible-expression cell lines, we further show that phospho-regulation of KIBRA by ERK1/2 and RSK1/2 is required for proper cell proliferation and RSK-mediated phosphorylation also modulates KIBRA's migratory activity in MDA-MB-231 breast cancer cells. Our findings uncover unexpected results and a new mechanism through which KIBRA regulates cell migration and proliferation.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphoproteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Benzamides/pharmacology , Butadienes/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Enzyme Inhibitors/pharmacology , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , MCF-7 Cells , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Molecular Sequence Data , Nitriles/pharmacology , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphorylation/drug effects , RNA, Small Interfering/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/chemistry , Signal Transduction/drug effects
13.
Semin Nephrol ; 32(4): 377-84, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22958492

ABSTRACT

The renin-angiotensin-aldosterone system (RAAS) plays a critical role in kidney function and its inhibition reduces proteinuria and preserves kidney function in patients with chronic kidney disease. Recent studies have shown that podocytes generate many components of the RAAS and they express receptors of RAAS, including angiotensin II, mineralocorticoid, and prorenin receptors. Crucial functions of podocytes, such as contraction, apoptosis, autophagocytosis, and cytoskeletal organization, have been shown to be regulated by the angiotensin II type 1 receptors. An activation of the glomerular RAAS and protection from podocyte injury by RAAS inhibitors have been shown in many glomerular diseases. Exploring the interaction between the local RAAS and the signaling involved in RAAS activation in podocytes will lead to new therapeutic strategies of podocyte protection.


Subject(s)
Angiotensin II/metabolism , Podocytes/metabolism , Receptors, Angiotensin/metabolism , Renin-Angiotensin System/physiology , Aldosterone/metabolism , Animals , Humans , Podocytes/physiology , Receptors, Cell Surface/metabolism , Signal Transduction , Prorenin Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...