22
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Editorial: Podocyte Pathology and Nephropathy

      editorial

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Podocytes, glomerular visceral cells of epithelial origin, are the direct target of both immune and non-immune forms of injury in many diverse glomerular diseases. They do not typically proliferate in the mature kidney, which means that damaged cells are not replaced by new ones. It is now generally accepted that dysfunction or loss of these cells underlies progression of almost all glomerulopathies. Progressing glomerular diseases, of which diabetic nephropathy (DN) is the most common, are the most frequent cause of end-stage renal disease. Therefore, identification of the pathways that lead to podocyte injury is essential for developing more effective, targeted therapies. The articles presented in this research topic give a comprehensive overview of recent discoveries into the mechanisms of podocyte impairment and discuss the possibilities of podocyte-targeted therapies. Each podocyte contacts its neighboring cells via a slit diaphragm (SD) connecting their interdigitating foot processes (FPs). Via transmembrane adhesion receptors in the basolateral domain of the FP, the podocytes interact with the glomerular basement membrane (GBM). Both these forms of adhesions are crucial for maintaining podocyte structure, which in turn determines the integrity and permeability of the glomerular filtration barrier. As highlighted by Lennon et al. (1), the cell–cell and cell–matrix junctions drive the coordinated response of podocytes to environmental cues in order to regulate glomerular filtration. Recent findings indicate that there are two distinct FP types, anchoring FPs (AFPs) and ordinary FPs (OFPs), both of which are involved in the regulation of fluid outflow from the subpodocyte space (2). The components of SD and adhesion complexes in FPs transduce signals from outside of the podocyte to the actin cytoskeleton inside the cell. Up to now, almost 100 actin associated proteins have been discovered in mammalian podocytes, with distribution specific for apical, SD, and basal domains of the FP membrane (2). Dysregulation of signaling is likely to lead to actin reorganization and podocyte foot process effacement, which is typically observed in proteinuric diseases. In contrast to traditional interpretations of this loss of shape as a pathological derangement, Kriz et al. (3) indicate that adhesions are reinforced in effaced FPs and therefore it seems to be a protective mechanism against detachment. Alterations in podocyte phenotype and structure are particularly prominent when proteinuria reaches nephrotic range. It is accepted that loss of the specialized podocyte morphology is associated with transition from epithelial to a more mesenchymal phenotype irrespective of the underlying causes that include both genetic defects and mediators from the microenvironment. However, May et al. (4) note that podocytes display partial features of both mesenchymal and epithelial cells. Therefore, dependent on the clinical conditions, dedifferentiation in disease could result in regression to either of these states. Upon treatment, these changes are reversible only if the insult is not very severe, as for example in minimal change disease (MCD). In focal segmental glomerulosclerosis (FSGS), phenotypic dedifferentiation of podocytes is not only irreversible but progressive. Using a rat model, Kriz et al. (3) have performed a detailed structural study demonstrating how podocytes reinforce attachment to the GBM and how they detach. It appears that if the protective mechanisms fail, viable podocytes, mostly in clusters, detach from the GBM. Some of them may reach the renal pelvis as living cells, while other may develop contacts to the parietal epithelium, forming crescents that connect glomerular capillaries with the Bowman’s capsule. Podocyte depletion represents one of the earliest cellular lesions affecting the diabetic kidney, and decreased number of podocytes in glomeruli is the strongest predictor of progression of both type 1 and 2 DN. Activation of protein kinase C (PKC) seems to play a critical role in pathogenesis of DN. Teng et al. (5) point out that conventional as well as atypical forms of PKC, which play a pivotal role in the regulation of podocyte physiology, may be a destructive factor when hyperactivated in disease conditions. PKC activation results in downregulation of podocyte and SD structural proteins such as P-cadherin or β-catenin, which may contribute to the disruption of podocyte integrity. In DN, the PKC isoforms may also mediate the high glucose-induced overproduction of VEGF and increased TGFβ signaling in podocytes, with subsequent impairment of the glomerular filtration barrier. Recent findings reveal that abnormal intracellular accumulation of sphingolipids modulates podocyte functions in glomerular disorders of both genetic and non-genetic origin. Based on their experimental results, Merscher and Fornoni (6) report that in FSGS, suPAR-dependent αVβ3 integrin activation decreased expression of sphingomyelin-like phosphodiesterase 3b (SMPDL3b) resulting in increased accumulation of sphingomyelin, which is associated with remodeling of the podocyte actin cytoskeleton, loss of stress fibers, and a shift from a migratory to an apoptotic phenotype. In contrast, in the diabetic kidney SMPDL3b expression was elevated, nonetheless rendering podocytes more susceptible to apoptosis. These observations indicate that podocyte responses to sphingolipids are complex and require additional research. Hereditary, but also sporadic, nephrotic syndrome (NS) is frequently associated with mutations in podocyte genes encoding functional and structural proteins. However, currently known mutations explain <40% of NS cases (7). Moreover, immunosuppression appears to be effective in about 8–10% of genetic disorders. Recent advances in our knowledge about the podocyte transcriptosome and proteasome have led to identification and characterization of novel disease-causing variants and disease-modifying genes. At present, almost 50 podocyte genes directly associated with human NS have been identified. It has also become apparent that monogenic defects correlating with some morphological changes in podocytes do not explain completely the pathogenesis of congenital podocytopathies. An understanding of the mechanisms of podocyte impairment will allow for the design of targeted therapeutic approaches that may prevent deterioration of glomerular function, e.g., in allograft recipients. The frequency of recurrent as well as newly developed post-transplant kidney diseases, such as DN, is relatively high and now it is clear that podocytes are the initial site of injury. Detailed pathogenesis of post-transplant diabetes is not known; however, numerous risk factors have been identified (8). Although direct treatment of genetic disorders still remains a question for the future, certain biochemical pathways in podocytes already seem to be a promising target for current therapies. For example, it has been shown recently that SMPDL3b in podocyte lipid rafts is a direct target for rituximab, which prevents downregulation of this protein and podocyte injury (7). Furthermore, calcineurin inhibitors (CNIs) that, together with glucocorticoids and mTOR inhibitors, have been traditionally used as anti-inflammatory agents have been shown to also act directly on podocytes (9). Diverse actions of CNIs on podocytes include stabilizing the actin cytoskeleton and inhibiting NFAT-dependent podocyte apoptosis. Other immunosuppressive drugs, such as abatacept or belatacept, also directly target the podocytes, suggesting this is an exciting area for future research (8). Early, sensitive, and specific diagnosis of ongoing podocyte injury is still lacking. Microalbuminuria, which results from podocyte dysfunction, has been accepted as the earliest marker of DN. However, patients presenting with microalbuminuria already show advanced damage of the glomerular filtration barrier (10), while nephrinuria may precede this manifestation (11). It has been shown recently that urinary podocyte damage biomarkers correlate with cystatin C and eGFR even in normoalbuminuric patients with type 2 diabetes (11). Yet, due to technical problems, such as low quantities of podocyte proteins or their proteolytic digestion, using free urinary podocyte proteins as diagnostic markers appears to be questionable. Musante et al. (12) describe a promising new method in which exosomes carrying specific podocyte biomarkers are isolated from urine. In one pilot study, the levels of urinary exosomal mRNA for cystatin C showed a marked upregulation after the induction of podocyte damage in the puromycin aminonucleoside nephrosis (PAN) rat model (13). However, this was accompanied by the de novo expression of cystatin C not only in podocytes but also in tubular epithelial cells. On the other hand, in an experimental model of DN, which is associated with podocyte injury, cystatin C immunoreactivity in tubules was not changed (14). So far, there are only few studies referring to the urinary exosomal mRNA, and data concerning levels and cellular origin of cystatin C mRNA in urine are still missing. Nevertheless, correlation between cystatin C and podocyte-specific genes in urinary exosomes could probably allow for early specific diagnosis of podocyte damage prior to the symptomatic podocytopathy. Conflict of Interest Statement The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

          Related collections

          Most cited references11

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          The Importance of Podocyte Adhesion for a Healthy Glomerulus

          Podocytes are specialized epithelial cells that cover the outer surfaces of glomerular capillaries. Unique cell junctions, known as slit diaphragms, which feature nephrin and Neph family proteins in addition to components of adherens, tight, and gap junctions, connect adjacent podocyte foot processes. Single gene disorders affecting the slit diaphragm result in nephrotic syndrome in humans, characterized by massive loss of protein across the capillary wall. In addition to specialized cell junctions, interconnecting podocytes also adhere to the glomerular basement membrane (GBM) of the capillary wall. The GBM is a dense network of secreted, extracellular matrix (ECM) components and contains tissue-restricted isoforms of collagen IV and laminin in addition to other structural proteins and ECM regulators such as proteases and growth factors. The specialized niche of the GBM provides a scaffold for endothelial cells and podocytes to support their unique functions and human genetic mutations in GBM components lead to renal failure, thus highlighting the importance of cell–matrix interactions in the glomerulus. Cells adhere to ECM via adhesion receptors, including integrins, syndecans, and dystroglycan and in particular the integrin heterodimer α3β1 is required to maintain barrier integrity. Therefore, the sophisticated function of glomerular filtration relies on podocyte adhesion both at cell junctions and at the interface with the ECM. In health, the podocyte coordinates signals from cell junctions and cell–matrix interactions, in response to environmental cues in order to regulate filtration and as our understanding of mechanisms that control cell adhesion in the glomerulus develops, then insight into the effects of disease will improve. The ultimate goal will be to develop targeted therapies to prevent or repair defects in the filtration barrier and to restore glomerular function.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Use and Isolation of Urinary Exosomes as Biomarkers for Diabetic Nephropathy

            Diabetes represents a major threat to public health and the number of patients is increasing alarmingly in the global scale. Particularly, the diabetic kidney disease (nephropathy, DN) together with its cardiovascular complications cause immense human suffering, highly increased risk of premature deaths, and lead to huge societal costs. DN is first detected when protein appears in urine (microalbuminuria). As in other persisting proteinuric diseases (like vasculitis) it heralds irreversible damage of kidney functions up to non-functional (end-stage) kidney and ultimately calls for kidney replacement therapy (dialysis or kidney transplantation). While remarkable progress has been made in understanding the genetic and molecular factors associating with chronic kidney diseases, breakthroughs are still missing to provide comprehensive understanding of events and mechanisms associated. Non-invasive diagnostic tools for early diagnostics of kidney damage are badly needed. Exosomes – small vesicular structures present in urine are released by all cell types along kidney structures to present with distinct surface assembly. Furthermore, exosomes carry a load of special proteins and nucleic acids. This “cargo” faithfully reflects the physiological state of their respective cells of origin and appears to serve as a new pathway for downstream signaling to target cells. Accordingly, exosome vesicles are emerging as a valuable source for disease stage-specific information and as fingerprints of disease progression. Unfortunately, technical issues of exosome isolation are challenging and, thus, their full potential remains untapped. Here, we review the molecular basis of exosome secretion as well as their use to reveal events along the nephron. In addition to novel molecular information, the new methods provide the needed accurate, personalized, non-invasive, and inexpensive future diagnostics.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Podocyte Dedifferentiation: A Specialized Process for a Specialized Cell

              The podocyte is one of the two cell types that contribute to the formation of the glomerular filtration barrier (GFB). It is a highly specialized cell with a unique structure. The key feature of the podocyte is its foot processes that regularly interdigitate. A structure known as the slit diaphragm can be found bridging the interdigitations. This molecular sieve comprises the final layer of the GFB. It is well accepted that the podocyte is the target cell in the pathogenesis of nephrotic syndrome. In nephrotic syndrome, the GFB no longer restricts the passage of macromolecules and protein is lost into the urine. A number of phenotypic and morphological changes are seen in the diseased podocyte and in the literature these have been described as an epithelial–mesenchymal transition (EMT). However, there is a growing appreciation that this term does not accurately describe the changes that are seen. Definitions of type-2 EMT are based on typical epithelial cells. While the podocyte is known as a visceral epithelial cell, it is not a typical epithelial cell. Moreover, podocytes have several features that are more consistent with mesenchymal cells. Therefore, we suggest that the term podocyte disease transformation is more appropriate.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Endocrinol (Lausanne)
                Front Endocrinol (Lausanne)
                Front. Endocrinol.
                Frontiers in Endocrinology
                Frontiers Media S.A.
                1664-2392
                17 September 2015
                2015
                : 6
                : 145
                Affiliations
                [1] 1Department of Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk , Gdansk, Poland
                [2] 2Laboratory of Molecular and Cellular Nephrology Gdansk, Miroslaw Mossakowski Medical Research Center of the Polish Academy of Sciences , Warsaw, Poland
                [3] 3Academic Renal Unit, University of Bristol , Bristol, UK
                [4] 4Department of Clinical Chemistry, Medical University of Gdansk , Gdansk, Poland
                Author notes

                Edited by: Aaron Vinik, Eastern Virginia Medical School, USA

                Reviewed by: Ramin Tolouian, Eastern Virginia Medical School, USA

                *Correspondence: Barbara Lewko, blew@ 123456gumed.edu.pl

                Specialty section: This article was submitted to Diabetes, a section of the journal Frontiers in Endocrinology

                Article
                10.3389/fendo.2015.00145
                4585015
                37b9a8b7-e8e5-4a9e-9caa-9e7a8c2a23fb
                Copyright © 2015 Lewko, Welsh and Jankowski.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 23 June 2015
                : 02 September 2015
                Page count
                Figures: 0, Tables: 0, Equations: 0, References: 14, Pages: 3, Words: 1957
                Categories
                Endocrinology
                Editorial

                Endocrinology & Diabetes
                podocytes,diabetes,kidney disease,proteinuria,foot processes,exosomes
                Endocrinology & Diabetes
                podocytes, diabetes, kidney disease, proteinuria, foot processes, exosomes

                Comments

                Comment on this article