• No results found

Glycogen Synthase Kinase 3β Dictates Podocyte Motility and Focal Adhesion Turnover by Modulating Paxillin Activity Implications for the Protective Effect of Low-Dose Lithium in Podocytopathy

N/A
N/A
Protected

Academic year: 2021

Share "Glycogen Synthase Kinase 3β Dictates Podocyte Motility and Focal Adhesion Turnover by Modulating Paxillin Activity Implications for the Protective Effect of Low-Dose Lithium in Podocytopathy"

Copied!
15
0
0

Loading.... (view fulltext now)

Full text

(1)

EPITHELIAL AND MESENCHYMAL CELL BIOLOGY

Glycogen Synthase Kinase 3

b Dictates Podocyte Motility

and Focal Adhesion Turnover by Modulating Paxillin

Activity

Implications for the Protective Effect of Low-Dose Lithium in

Podocytopathy

Weiwei Xu,*yYan Ge,yZhihong Liu,* and Rujun Gongy

From the National Clinical Research Center of Kidney Disease,* Jinling Hospital, Nanjing University School of Medicine, Nanjing, China; and the Division of Kidney Disease and Hypertension,yDepartment of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, Rhode Island

Accepted for publication June 10, 2014.

Address correspondence to Rujun Gong, M.D., Ph.D., Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, 593 Eddy St., Providence, RI 02903. E-mail:

[email protected].

Aberrant focal adhesion turnover is centrally involved in podocyte actin cytoskeleton disorganization and foot process effacement. The structural and dynamic integrity of focal adhesions is orchestrated by multiple cell signaling molecules, including glycogen synthase kinase 3b (GSK3b), a multitasking kinase lately identified as a mediator of kidney injury. However, the role of GSK3b in podocytopathy remains obscure. In doxorubicin (Adriamycin)-injured podocytes, lithium, a GSK3b inhibitor and neuroprotective mood stabilizer, obliterated the accelerated focal adhesion turnover, rectified podocyte hypermotility, and restored actin cytoskeleton integrity. Mechanistically, lithium counteracted the doxorubicin-elicited GSK3b overactivity and the hyperphosphorylation and overactivation of paxillin, a focal adhesioneassociated adaptor protein.Moreover, forced expression of a dominant negative kinase dead mutant of GSK3b highly mimicked, whereas ectopic expression of a constitutively active GSK3b mutant abolished, the effect of lithium in doxorubicin-injured podocytes, suggesting that the effect of lithium is mediated, at least in part, through inhibition of GSK3b. Furthermore, paxillin interacted with GSK3b and served as its substrate. In mice with doxorubicin ne-phropathy, a single low dose of lithium ameliorated proteinuria and glomerulosclerosis. Consistently, lithium therapy abrogated GSK3b overactivity, blunted paxillin hyperphosphorylation, and reinstated actin cyto-skeleton integrity in glomeruli associated with an early attenuation of podocyte foot process effacement. Thus, GSK3b-modulated focal adhesion dynamics might serve as a novel therapeutic target for podocytop-athy. (Am J Pathol 2014, 184: 2742e2756;http://dx.doi.org/10.1016/j.ajpath.2014.06.027)

Glomerular visceral epithelial cells or podocytes are a core structural constituent of the glomerularfiltration barrier, with elaborate interdigitating foot processes that envelop the capillaries of the glomeruli in the kidney, control glomerular permselectivity, and prevent protein in the bloodstream from leaking into the urine.1e4Converging evidence suggests that the podocyticfilter barrier is not static but a highly dynamic structure that is regulated via the motility of podocyte foot processes.5e7 The molecular basis of foot process motility lies in the constant dynamics of the molecular machinery that sustains the foot process architecture.5e7 Actin is the

principal component of the cytoskeletal machinery of foot processes and forms a subcortical network of branched fila-ments as well as bundled filaments that run longitudinally through the processes with contractility.8Actin exists in foot processes in a state of dynamic equilibrium between assem-bly and disassemassem-bly, which is important for maintaining the

Supported in part by NIH grant R01DK092485 (R.G.), the China 973 program 2012CB517600 (Z.L.), and the International Society of Nephrology Sister Renal Center Trio Program.

Disclosures: None declared.

(2)

homeostasis of the glomerularfiltration barrier. In response to various pathogenic mediators, including oxidative stress, circulating permeability factors, and nephrotoxins such as doxorubicin (Adriamycin), the parallel actin bundles depo-lymerize, resulting in foot process effacement, a pathologic hallmark of podocyte injury and dysfunction.9e13

Focal adhesions (FAs), by which cells are anchored to the extracellular matrix, are a crucial determinant of actin cyto-skeleton integrity and cell motility.14,15Molecules from FA structures connect the extracellular matrix to bundles of actin filaments, enabling the growing actin network to push the plasma membrane and the contractile stressfibers to pull the cell body, corresponding to protrusive and retractive activ-ities.14,16 Dynamic turnover of FAs is indispensable for constant motility and reorganization of cell edges that man-ifest as boundary curvature waves.17 A cycle of cellular boundary motion commences with the formation of nascent adhesions, which initiate actin assembly and, thus, allow the growing actin network to push the cell protrusion forward. The nascent adhesion or focal complex, a precursor of the FA, is smaller in size, with weaker adhesive force and rapid turnover.18,19 Subsequently, nascent adhesions will either disassemble rapidly or mature to be FAs. The FAs usually contain multiple structural and regulatory molecules, among which paxillin acts as a pivotal adaptor protein to provide docking sites for cytoskeletons and to recruit FA regulators that control actin dynamics and FA stability.20The rate of FA turnover determines cell motility and governs the podocyte foot process dynamics. Consistently, targeted manipulation of FA turnover in podocytes by enhancing or intercepting the activity of FA regulatory molecules incurred foot process effacement and podocyte dysfunction.21e23

Glycogen synthase kinase 3b (GSK3b), a well-conserved and ubiquitously expressed serine/threonine protein kinase, plays a key role in the regulation of cytoskeleton organization and cellular motility.24,25 Indeed, inhibition of GSK3b has been found to reduce cell motility in multiple cells, including vascular smooth muscle cells,26glioma cells,27gastric cancer cells,28 and renal tubular epithelial cells.29 In the kidney, GSK3b has lately been implicated in acute kidney injury and repair.30However, its role in podocyte injury and foot process cytoskeletal disarrangement remains unknown. This study examined the role of GSK3b in a model of hypermotility-associated podocytopathy induced by doxo-rubicin injury in vivo and in vitro. The potential intervention effect of GSK3b blockade by a single low dose of lithium, a selective GSK3b inhibitor and US Food and Drug Admin-istrationeapproved mood stabilizer, on podocyte motility and dysfunction was accordingly delineated.

Materials and Methods

Cell Culture and Transient Transfection

Conditionally immortalized mouse podocytes in culture were a gift from Dr. Stuart Shankland (University of Washington,

Seattle, WA).31 The cells between passages 21 to 25 were used. Podocytes were cultured in RPMI 1640 medium (Life Technologies, Grand Island, NY) supplemented with 10% fetal bovine serum (Life Technologies), 0.075% sodium bi-carbonate (Sigma-Aldrich, St. Louis, MO), 0.075% sodium pyruvate (Sigma-Aldrich), 100 U/mL of penicillin, and 100 mg/mL of streptomycin (Life Technologies) in a humidified incubator with 5% CO2. The cells were cultured at 33C with

50 U/mL of recombinant mouse interferon-g (Millipore, Billerica, MA) on collagen-coated Petri dishes. The cells were transferred to 37C incubators without interferon-g to induce differentiation, which took 14 days, and then were treated with doxorubicin (0.25 mg/mL; Sigma-Aldrich) and/or lithium chloride (10 mmol/L; Sigma-Aldrich). Podocytes were cultured under permissive conditions (33C) and were pre-pared for immunoblot analysis after sodium chloride (10 mmol/L) treatment for 24 hours. Subcultures of the immor-talized mouse podocytes were maintained under nonpermissive conditions (37C) to induce differentiation for 14 days and then were treated with lithium chloride (10 or 20 mmol/L) for 24 or 48 hours. As control, cells were treated with sodium chloride (10 mmol/L) for 24 hours. Cells were subsequently collected and prepared for Western blot analysis and immunohisto-chemical staining. The expression vectors encoding the constitutively active GSK3b mutant (S9A-GSK3b-HA/ pcDNA3), kinase-dead GSK3b mutant (KD-GSK3b-HA/ pcDNA3), and wild-type GSK3b (WT-GSK3b-HA/pcDNA3) were a gift from Dr. Gail V.W. Johnson (University of Alabama at Birmingham, Birmingham, AL).32 The vector encoding greenfluorescent proteinepaxillin was a gift from Dr. Luc Sabourin (Ottawa Hospital Research Institute, Ottawa, ON, Canada).33Podocytes were transfected by using Lipofectamine 2000 reagent (Life Technologies) as previously described.34

Cell Migration Assay

Confluent monolayers of differentiated podocytes were scraped with a 10-mL pipette after different treatments. Images of the same area were acquired at indicated time points using an inverted microscope and were analyzed using the ImageJ version 1.48 (NIH, Bethesda, MD) image processing program. The percentage of cell migration area was calculated as

ðArea0 hour Areaindicated timeÞ=Area0 hour ð1Þ

Time-Lapse Fluorescence Microscopy

Podocytes transected with green fluorescent proteine paxillin were subjected to different treatments and placed in a heating chamber (37C) on the stage of a time-lapse fluorescence microscope (Axiovert; Zeiss, Cologne, Ger-many). Images were taken at 2-minute intervals. The FA turnover rate was calculated using the Focal Adhesion Analysis Server web tool (http://faas.bme.unc.edu, last accessed October 7, 2013), as described previously.35

(3)

Western Immunoblot Analysis

Cultured cells were lyzed and animal tissues homogenized in radioimmunoprecipitation assay buffer supplemented with protease inhibitors and samples were processed for immu-noblot analysis. The antibodies against paxillin, GSK3b, p-GSK3b (S9), synaptopodin, and glyceraldehyde-3-phosphate dehydrogenase were purchased from Santa Cruz Biotechnology (Santa Cruz, CA), and those against paxillin, phosphorylated paxillin (S126), and nephrin were acquired from Cell Signaling Technology Inc. (Danvers, MA) and Pro-gen Biotechnik GmbH (Heidelberg, Germany), respectively.

Animal Experiment Design

Animal studies were approved by the Rhode Island Hospital Animal Care and Use Committee, and they conform to the US Department of Agriculture regulations and the NIH’s Guide for the Care and Use of Laboratory Animals.36Male BALB/c mice weighing 20 to 25 g and aged 8 weeks were randomly assigned to the following treatments. A single dose of lithium chloride (40 mg/kg) or an equal molar amount (1 mEq/kg) of sodium chloride as saline was given via i.p. in-jection on day 0. Doxorubicin (10 mg/kg) or an equal volume of vehicle was given as a tail vein injection 6 hours later. Groups control, LiCl, NaClþ ADR, and LiCl þ ADR refer to sodium chlorideþ vehicle, lithium chloride þ vehicle, sodium chloride þ doxorubicin, and lithium chloride þ doxorubicin treatments, respectively. Spot urine was col-lected on postinjury days 0, 1, 3, 5, 7, 10, and 14. Mice were sacrificed on days 3, 7, and 14. Six mice were randomly assigned to each group for each observed time point.

Urine Analyses

To discern the protein compositions in urine, equal amounts of urine samples were subjected to SDS-PAGE followed by Coomassie Blue (Sigma-Aldrich) staining. Urine albumin concentration was measured using a mouse albumin enzyme-linked immunosorbent assay quantitation kit (Bethyl Laboratories Inc., Montgomery, TX). Urine creati-nine concentration was measured by a creaticreati-nine assay kit (BioAssay Systems, Hayward, CA).

Morphologic Studies

Formalin-fixed kidneys were embedded in paraffin and pre-pared sections (3-mm thick). For general histologic analysis, sections were processed for periodic acideSchiff staining. The morphologic features of all the sections were assessed by a single observer (W.X.) in a blinded manner. A semi-quantitative glomerulosclerosis score was used to evaluate the degree of glomerulosclerosis. The severity of sclerosis for each glomerulus was graded from 0 to 4 as follows: 0 repre-sents no lesions; 1, sclerosis of<25% of the glomerulus; and 2, 3, and 4, sclerosis of 25% to 50%, >50% to 75%, and

>75% of the glomerulus, respectively. A whole-kidney average glomerulosclerosis score was obtained by aver-aging scores from all glomeruli on one section.

Immuno

fluorescence Staining

Podocytes or cryosections of kidneys were fixed with 4% paraformaldehyde (Sigma-Aldrich), permeabilized, and stained with primary antibodies against paxillin, GSK3b, and synaptopodin, followed by Alexa fluorophoreeconjugated secondary antibody staining (Life Technologies). Filamen-tous actin (F-actin) was stained by rhodamine phalloidin (Cytoskeleton Inc., Denver, CO). Finally, cells were coun-terstained with DAPI, mounted with Vectashield mounting medium (Vector Laboratories, Burlingame, CA), and visu-alized using afluorescence microscope. As a negative con-trol, the primary antibody was replaced by preimmune serum from the same species. The confocal images were acquired using an LSM 710 Meta confocal microscope (Zeiss). For dual-color staining, images were acquired sequentially to avoid dye interference. ImageJ software was used for post-processing of the images, eg, scaling, merging, and co-localization analysis.

Glomerular Isolation

Mice were anesthetized and perfused by infusing the abdom-inal artery with 5 mL of phosphate-buffered saline containing 8 107Dynabeads M-450 beads (Dynal Biotech ASA, Oslo, Norway). After perfusion, the kidneys were removed, minced into 1-mm3pieces, and digested in collagenase A, and the glomeruli-containing Dynabeads were collected using a magnetic particle concentrator as described previously.37

Transmission Electron Microscopy

For transmission electron microscopy, kidney cortical tis-sues were cut into small pieces (1 mm3),fixed with 2.5% glutaraldehyde, and embedded in Epon 812 (Polysciences Inc., Warrington, PA). Conventional electron micrographs were obtained using an EM-10 microscope (Zeiss) operated at 60 kV. The podocyte foot process density was estimated by dividing the total length of glomerular basement mem-brane by the total number of foot processes present in each micrograph.

Statistical Analysis

For immunoblot analysis, bands were scanned and the inte-grated pixel density was determined using a densitometer and the ImageJ analysis program. All in vitro studies and immunoblot analyses were performed with triplicate samples and were repeated three to six times. All the data are expressed as means SD or as otherwise indicated. Statis-tical analysis of the data from multiple groups was performed by analysis of variance followed by Student-NewmaneKeuls

(4)

tests. Data from two groups were compared by Student’s t-test or Wilcoxon rank sum test. Linear regression analysis was applied to examine possible relationships between two parameters. P< 0.05 was considered significant.

Results

Lithium Abrogates Podocyte Hypermotility Induced by

Doxorubicin Stimulation

The conditionally immortalized differentiated murine podo-cytes in culture exhibited typical arborized morphologic features and were characterized as expressing multiple podocyte markers (Supplemental Figure S1). Evidence sug-gests that podocytes are motile cells with considerable constitutive motility.38 Indeed, as revealed by a traditional cell migration assay for assessing cellular motility, podocytes under basal conditions possessed a substantial migratory ca-pacity that lessened the distances between the leading edges of the migrating podocyte sheets. Cells were pretreated with lithium or sodium for 6 hours, followed by doxorubicin injury or vehicle treatment. The basal migrating activity of podocytes was relatively reduced by lithium treatment alone but was unaffected by sodium treatment. In contrast, doxorubicin-injured podocytes demonstrated strikingly accelerated closure of the gap between the invading fronts of the cells, suggesting enhanced podocyte motility. This effect of doxo-rubicin was markedly abrogated by lithium treatment (Figure 1A). These morphologic findings were further corroborated by quantitative measurements of cell migration area (Figure 1B). In addition to rectifying hypermotility, lithium also seemed to elevate the expression of podocyte markers, such as nephrin, in the conditionally immortalized differentiated murine podocytes (Supplemental Figure S1).

Lithium Preserves FA and Actin Cytoskeleton Integrity

in Doxorubicin-Injured Podocytes

FA turnover is a prerequisite for cell spreading and migration; accordingly, FA dynamics has been implicated in the control of cellular motility.15 To understand whether the effects of doxorubicin and lithium on podocyte motility were associated with alterations in FA and the ensuing changes in actin cyto-skeleton, podocytes were subjected to phalloidin labeling for F-actin and to immunofluorescence staining for paxillin, a core structural component of FA. Differentiated podocytes either under normal conditions or after vehicle treatment demon-strated abundant FAs that were located at the cell edges, accompanied by intense phalloidin-labeled ventral stressfibers that were anchored to FAs at both ends (Figure 2A). The nuclear staining of paxillin was seemingly nonspecific because it was also probed in the negative control staining, where the primary antibody was replaced with preimmune IgG (Figure 2A). Lithium treatment alone slightly reduced the number of FAs but barely affected the size of FAs, suggesting a stabilized FA. Correspondingly, lithium aloneetreated podocytes exhibited a stretched cellular shape and a ventral stressfiber with long paralleled cortical stress fibers as major F-actin that were indistinguishable from the morphologic fea-tures of control podocytes. In contrast, in doxorubicin-injured podocytes, the number of FAs was substantially increased, Figure 1 Lithium treatment abrogates Adriamycin (ADR;

doxorubicin)-elicited podocyte hypermotility as assessed by cell migration assay. A: Differentiated podocytes were stimulated with 0.25 mg/mL of ADR or an equal volume of vehicle 6 hours after pretreatment with 10 mmol/L lithium chloride (LiCl) or 10 mmol/L sodium chloride (NaCl), and subsequently scratch was processed using a 10-mL pipette. Control cells were treated with NaCl and vehicle. The observation was made immediately after scratch (0:00 hour) and at 24:00 hours. B: Quantification of the cell migration area by computerized morphometric analysis. Data are given as means SD. n Z 20 areas from three independent experiments. *P< 0.05 versus all other groups;yP< 0.05 versus sodium treatment alone. Original magnification, 200.

(5)

and FAs dramatically shrunk to the size that was likely tantamount to that of focal complexes or nascent adhesions, denoting a more dynamic FA. In agreement, doxorubicin-injured podocytes exhibited an asterlike cell shape and actin cytoskeleton disruption that manifested as increased expres-sion of corticalfilaments, diminished ventral stress fibers, more transverse arcs, and sporadic short dorsal stress fi-bers that were connected to the FA only at one end. This cytopathic effect is reminiscent of the actin changes that are observed in foot process effacement in vivo in doxo-rubicin podocytopathy.1e4 Lithium treatment strikingly prevented the doxorubicin-induced increase in FA numbers and shrinkage in FA sizes, retained stressfibers, and largely preserved actin cytoskeleton integrity. These morphologic findings were subsequently corroborated by

morphometric measurements of FA sizes and absolute FA counts (Figure 2, B and C).

Lithium Normalizes the Doxorubicin-Accelerated

Dynamics of FA Turnover in Podocytes

Fixed podocytes provide only a brief snapshot of FA expression. By observing alive podocytes, however, addi-tional insights into FA dynamics could be gained to validate or complement the findings from fixed cells. To further define the functional impact of lithium- and doxorubicin-regulated FA numbers and sizes on FA dynamics, live imaging of podocytes was performed. Green fluorescent proteineconjugated paxillin was ectopically expressed in podocytes by transient transfection so that turnover of FAs Figure 2 Lithium preserves FAs and actin cytoskeleton integrity in Adriamycin (ADR; doxorubicin)-injured podocytes. Differentiated podocytes were stimulated with 0.25 mg/mL of ADR or an equal volume of vehicle 6 hours after pretreatment with 10 mmol/L lithium chloride (LiCl) or 10 mmol/L sodium chloride (NaCl). Eight hours later, cells werefixed and subjected to double staining for cytoskeletal F-actin with rhodamine phalloidin and paxillin, an FA marker. A: Control podocytes displayed evident FAs located at the cell edges associated with intense phalloidin-labeled ventral stressfibers anchored to FAs at both ends. Lithium aloneetreated podocytes demonstrated slightly fewer FAs, a more stretched cellular shape, and ventral stress fibers with long paralleled cortical stressfibers similar to the morphologic features of control podocytes. In contrast, ADR-injured podocytes had an increased number of small FAs and displayed an asterlike cell shape as well as actin cytoskeleton disruption that manifested as increased expression of cortical actinfilaments, drastically diminished ventral stressfibers, more transverse arcs, and sporadic short dorsal stress fibers connected to the FA at only one end. Lithium treatment prevented the effect of ADR, restored the number and size of FAs, retained stressfibers, and largely preserveed actin cystoskeleton integrity in podocytes. The staining of paxillin in the nucleus was nonspecific because it was also noted in negative control, where the antipaxillin primary antibody was replaced with preimmune IgG. Boxed regions in the paxillin images are shown at higher magnification. B: Computerized morphometric quantification of FA size. ADR reduced the average size of FAs from approximately 3 mm2to<1 mm2, and this effect was obliterated by lithium pretreatment. C: Quantification of the number of FAs per podocyte. Lithium treatment alone reduced the number of FAs from 137 to approximately 71 per cell, whereas ADR increased FA number to approximately 275 per cell, and this effect was abrogated by lithium pretreatment. Data are given as means SD. n Z 30 cells from three independent experiments. *P < 0.05 versus control;yP< 0.05 versus LiCl þ ADR. Scale bar Z 10 mm (A). Original magnification: 800 (boxed regions in A).

(6)

Figure 3 Lithium corrects the Adriamycin (ADR; doxorubicin)-accelerated dynamics of FA turnover in podocytes. A: Differentiated podocytes were transiently transfected with a vector encoding greenfluorescent protein (GFP)epaxillin and were subjected to time-lapse microscopy for 1 hour, followed by fluorescence immunocytochemical staining for synaptopodin. Representative fluores-cent micrographs of synaptopodin staining showed that podocytes retain the podocyte marker protein synaptopodin. B: Podocytes transfected with GFP-paxillin were injured with ADR for 8 hours after 10 mmol/L lithium chloride (LiCl) or 10 mmol/L sodium chloride (NaCl) treatment for 6 hours. Subse-quently, live podocytes were subjected to time-lapsefluorescence microscopy for 1 hour with 2 minutes between microscopic image frames. The Detail column represents a series of time-lapse microscopic image frames aligned to show the temporal evolution of individual FAs from assembly to disassembly in differently treated podocytes. Because image frames were captured at afixed rate (0.5 frames per minute), the number of image frames showing the temporal evolution of an indi-vidual FA accordingly correlated the FA dynamics. Thus, hypodynamics and hyperdynamics of FA turnover were indicated by more and less frames, respectively. ADR-treated podocytes shrank rapidly, as shown by the whole cell image and exhibit an accelerated FA turnover (Detail column). This effect was abrogated by lithium pretreatment. Boxed regions focal adhesions, whose turnover is shown in Detail column. C: Quantification of FA assembly rates. Lithium treatment alone slightly reduced the assembly rate. ADR drastically increased the FA as-sembly rate, which was significantly obliterated by lithium pretreatment. D: Quantification of FA disassembly rates. ADR markedly increased the FA disassembly rate, and lithium pretreatment pre-vented the effect. Horizontal bars indicate the median values and the top and bottom lines of the boxes indicate the 3rd and 1st quartile, respectively (C and D). Data are given as medians ranges (C and D). nZ 30 cells from six experiments (C and D). *P< 0.05 versus all other groups by Wilcoxon rank sum test (C and D). Scale barZ 10 mm (A and B).

(7)

could be visualized and documented by time-lapse fluores-cence microscopy. After transfection and time-lapse mi-croscopy, control cells retained podocyte morphology and evidently expressed typical podocyte marker molecules, including synaptopodin (Figure 3A), suggesting that podo-cytes were maintained in a healthy state. Consistent with a constitutive motility, basal FA dynamics were evidently noted in podocytes under normal conditions and were barely affected by vehicle and sodium treatment (Figure 3B). Doxorubicin injury substantially accelerated FA turnover, as reflected by a reduced number of microscopic image frames showing the temporal evolution of an individual FA (Figure 3B). In contrast, lithium treatment resulted in more stable FA dynamics and prominently counteracted the effect of doxorubicin in the observed podocytes. Computerized morphometric analysis of FA turnover rates revealed that both assembly and disassembly rates of FA were signi fi-cantly elevated in doxorubicin-injured cells. Concomitant lithium treatment largely prevented the effect of doxorubicin and normalized the FA assembly and disassembly rates to the levels of normal podocytes (Figure 3, C and D).

Lithium Obliterates Doxorubicin-Elicited GSK3b

Overactivity and Paxillin Hyperphosphorylation

Next we tested whether lithium counteracted the effect of doxorubicin on FA turnover through a direct action on FA

molecules, such as paxillin. Differentiated podocytes were pretreated with lithium or sodium for 6 hours, followed by doxorubicin injury or vehicle treatment. Inhibitory phos-phorylation of GSK3b was evidently increased by lithium, in agreement with the role of lithium as a specific inhibitor of GSK3b (Figure 4A). Doxorubicin injury prominently diminished GSK3b phosphorylation at all observed time points, denoting GSK3b overactivity. This effect was largely abrogated by lithium treatment. Paxillin phosphorylation, on the contrary, exhibited opposing tendencies in response to doxorubicin injury or lithium treatment: doxorubicin enhanced whereas lithium abolished paxillin phosphoryla-tion. Densitometric analysis of immunblots confirmed these findings and revealed an inverse correlation between the changes in GSK3b phosphorylation and the changes in paxillin phosphorylation (Figure 4, B and C).

GSK3b Is Necessary and Suf

ficient for Paxillin

Overactivation, FA Instability, and Podocyte

Hypermotility

To examine a possible causal relationship between GSK3b and paxillin phosphorylation and activation as well as the ensuing changes in FA dynamics and podocyte motility, the activity of GSK3b was selectively manipulated by forced expression of vectors encoding the hemagglutinin-conjugated wild-type GSK3b, a dominant negative kinase Figure 4 Inhibitory phosphorylation of GSK3b is negatively associated with paxillin phosphorylation and activation in podocytes. A: Cell lysates were prepared from treated differentiated podocytes at the indicated time points after Adriamycin (ADR; doxorubicin) injury and were subjected to Western blot analysis. ADR injury reduced inhibitory phosphorylation of GSK3b but enhanced paxillin phosphorylation, whereas lithium chloride (LiCl), as a selective inhibitor of GSK3b, counteracted this effect, potentiated GSK3b phosphorylation, and diminished paxillin phosphorylation at different time points. B: Densitometric analysis of immunoblots quantified the relative levels of phosphorylated paxillin/total paxillin ratios and phosphorylated GSK3b/total GSK3b ratios at different time points. C: Linear regression analysis showed a negative correlation between inhibitory phosphorylation of GSK3b and paxillin phosphorylation and activation in podocytes. The correlation coefficient r was 0.7739. White, gray, and black colors represent 2, 8, and 24 hours, respectively. Data are given as means  SD. nZ 6 separate experiments (B); n Z 6 representative experiments (C). *P < 0.05 versus control;yP< 0.05 versus LiCl þ ADR.

(8)

dead mutant (KD) or a constitutively active mutant (S9A) of GSK3b. Immunofluorescence staining for hemagglutinin revealed a satisfactory transfection efficiency (>70%). Forced expression of KD reduced paxillin phosphorylation (Figure 5, A and B) and diminished the doxorubicin-elicited podocyte injury, marked by an increased number and reduced size of FAs as well as actin cytoskeleton disorga-nization that manifested as increased expression of cortical filaments and diminished stress fibers, reminiscent of the

protective effect of lithium (Figure 5, CeE). In contrast, ectopic expression of S9A prominently elicited hyper-phosphorylation and overactivation of paxillin (Figure 5, A and B) under basal conditions, associated with an increased number and reduced size of FAs as well as disruption of actin cytoskeleton integrity, mimicking the effect of doxorubicin (Figure 5, CeE). Moreover, on doxorubicin injury, the pro-tective effect of lithium on FAs and actin cytoskeleton was largely abolished in cells expressing S9A (Figure 5, CeE), Figure 5 GSK3b regulates paxillin phosphory-lation in podocytes and determines the ensuing FA dynamics. A: Differentiated podocytes were trans-fected with vectors encoding the hemagglutinin (HA)-conjugated wild-type (WT) GSK3b or a domi-nant negative KD or a constitutively active (S9A) mutant of GSK3b. Cells were harvested 48 hours after transfection, and cell lysates were subjected to immunoblot analysis for phosphorylated (p) paxillin, total paxillin, HA, and glyceraldehyde-3-phosphate dehydrogenase (GAPDH). B: Densito-metric analysis of immunoblots quantified the relative levels of p-paxillin/total paxillin ratios. Cells were treated with 10 mmol/L lithium chloride (LiCl) or 10 mmol/L sodium chloride (NaCl) for 6 hours before injury with vehicle or 0.25 mg/mL of Adriamycin (ADR; doxorubicin). C: Podocytes were fixed 8 hours after injury and were subjected to phalloidin labeling of F-actin (red) and immuno-fluorescence staining for paxillin (green). Forced expression of KD diminished the ADR-elicited podocyte injury, marked by an increased number and reduced size of FAs as well as actin cytoskeleton disorganization that manifests as increased expression of cortical filaments and diminished stressfibers, reminiscent of the protective effect of lithium. In contrast, ectopic expression of S9A prominently increased the number and reduced the size of FAs and disrupted actin cytoskeleton integrity under basal conditions, mimicking the effect of ADR injury. On ADR injury, the protective effect of lithium on FAs and actin cytoskeleton was largely abolished in cells expressing S9A. Arrow-heads indicate representative FAs. D: FA number quantification by computerized morphometric analysis. E: Computerized morphometric analysis of the size of FAs. F: Number and size quantification of FAs in ADR-injured podocytes after lithium treat-ment (Figure 2A) or in ADR-injured KD-expressing podocytes (Figure 5C) by computerized morpho-metric analysis; not statistically significant be-tween the two groups. Data are given as means SD. nZ 6 representative experiments (B); n Z 25 cells, three independent experiments (DeF). *P< 0.05 versus all other groups (B) or versus vehicle treated WT-expressing cells (D and E);

yP< 0.05 versus ADR-injured WT-expressing cells

(D and E),zP< 0.05 versus ADR-injured and LiCl-treated WT-expressing cells (D and E). Scale barZ 5 mm (C).

(9)

suggesting that inhibitory phosphorylation of GSK3b is, at least in part, responsible for the protection conferred by lithium. Consistently, on doxorubicin injury, lithium-treated podocytes and KD-overexpressing podocytes displayed comparable numbers and sizes of FAs (Figure 5F). Consistent with the role of FA and actin cytoskeleton in cellular motility, forced expression of S9A reinforced, whereas ectopic expression of KD mitigated, the doxorubicin-accelerated closure of the gap between the leading edges of the

migrating podocyte sheets as assessed by the cell migration assay, thus inferring enhanced and impeded podocyte motility, respectively (Figure 6, AeD). Moreover, the suppressive ef-fect of lithium on doxorubicin-elicited cell migration was substantially abrogated in S9A-overexpressing podocytes injured with doxorubicin (Figure 6, B and D), again suggesting that GSK3b inhibition is an indispensable and key mechanism accounting for the effect of lithium on podocyte motility. Consistently, lithium-treated podocytes and KD-overexpressing Figure 6 Modulation of GSK3b activity affects podocyte motility. Differentiated podocytes were transfected with vectors encoding the hemagglutinin (HA)-conjugated wild-type (WT), a dominant negative KD, or a constitutively active (S9A) mutant of GSK3b and then were treated with 10 mmol/L lithium chloride (LiCl) or 10 mmol/L sodium chloride (NaCl) for 6 hours before injury with vehicle or 0.25 mg/mL of Adriamycin (ADR; doxorubicin). Cells were then subjected to cell migration assay for the indicated time. Cell migration assay of podocytes transfected with WT or KD (A) or with WT or S9A (B) after the indicated treatments. Quantification of the cell migration area of podocytes transfected with WT or KD (C) or with WT or S9A (D) after the indicated treatments by computerized morphometric analysis. E: Quantification of the cell migration assay of ADR-injured podocytes after lithium treatment (Figure 1A) or ADR-injured KD-expressing podocytes from A by computerized morphometric analysis; not statistically significant between the two groups. Data are given as means  SD. n Z 20 areas from three independent experiments (C and E); nZ 6 areas, three independent experiments (D). *P < 0.05 versus vehicle-treated WT-expressing cells (C and D);

(10)

podocytes displayed comparable migration activity on doxoru-bicin injury (Figure 6E).

Paxillin Co-Localizes and Physically Interacts with

GSK3b as Its Putative Substrate in Podocytes

To further decipher the mechanistic essence of the GSK3b-mediated regulation of paxillin phosphorylation and acti-vation, the subcellular physical association between GSK3b and paxillin was examined by dual-color fluorescence immunocytochemical staining. A high-powered view of normal podocytes by confocal fluorescence microscopy revealed a close spatial association and co-localization be-tween a discrete pool of GSK3b and paxillin in the xy and z planes (Figure 7A). To validate this morphologic observa-tion, immunoprecipitation was performed and demonstrated that GSK3b evidently coprecipitated with paxillin in lysates of cultured podocytes and in homogenates of glomeruli isolated from murine kidneys, suggesting that GSK3b physically interacts with paxillin in podocytes in vivo and in vitro (Figure 7B). To further define the mechanism by which GSK3b regulates paxillin phosphorylation, the amino acid sequences of paxillin (UniProtKB/Swiss-Prot accession number Q8VI36.1) were subjected to computational active site analysis (http://scansite.mit.edu/motifscan_seq.phtml, last accessed December 28, 2013) for putative consensus phosphorylation motifs for GSK3b. In silico analysis deduced that residues S126, S226, S328, and S336 of paxillin reside in the consensus motifs for phosphorylation by GSK3b, with prediction scores higher than 0.5 denoting

high-confidence matches to GSK3b phosphorylation motifs (Figure 7, C and D). Collectively, these data suggest that paxillin is a putative cognate substrate for GSK3b.

A Single Low Dose of Lithium Ameliorates Podocyte

Foot Process Effacement, Attenuates Proteinuria, and

Improves Glomerulosclerosis in Experimental

Doxorubicin Nephropathy

To further explore whether the GSK3b-regulated FA dy-namics are involved in podocytopathy in vivo and also to assess the possible effect of therapeutic targeting of GSK3b, we used the mouse model of doxorubicin nephropathy, which is accounted for, in part, by podocyte hypermotility and re-capitulates key features of podocytopathy and focal and segmental glomerulosclerosis in humans, including podocyte foot process effacement, massive proteinuria, and progressive glomerulosclerosis.39,40Mice were injured with an intravenous injection of 10 mg/kg doxorubicin 6 hours after an i.p. injection of a low dose of 40 mg/kg of lithium chloride or an equal molar amount (1 mEq/kg) of sodium chloride saline. Doxorubicin injury elicited heavy proteinuria that peaked on days 5 and 7 and then partially receded on day 14 as determined by urine electrophoresis and urine albumin/creatinine ratios (Figure 8, A and B) and was associated with progressive glomerulosclerosis on periodic acideSchiff staining and with extensive foot pro-cess effacement on electron microscopy (Figure 8, C and D). Lithium therapy considerably attenuated proteinuria, amelio-rated glomerulosclerosis, and substantially improved foot Figure 7 Paxillin interacts with GSK3b as its putative substrate in podocytes. A: Differentiated podocytes werefixed and subjected to dual-color immu-nocytochemical staining for GSK3b (green) and paxillin (red). A high-powered view of normal podocytes by confocalfluorescence microscopy revealed a close spatial association and co-localization (arrowheads) between a discrete pool of GSK3b and paxillin in the xy and z planes. B: Lysates of cultured podocytes and homogenates of glomeruli isolated from normal murine kidneys by the magnetic beadsebased approach were subjected to immunoprecipitation (IP) assay by using an anti-GSK3b antibody or the preimmune IgG. Subsequently, immunoprecipitates were processed for immunoblot analysis for paxillin. Arrow indicates the band for paxillin. C: In silico analysis demonstrated that amino acid residues S126, S226, S328, and S336 of paxillin reside in the consensus motifs for phos-phorylation by GSK3b, denoting paxillin as a cognate substrate for GSK3b. D: Characteristics of consensus GSK3b phosphos-phorylation motifs, including the predicted phosphorylation sites, prediction confidence scores, and sequences in paxillin, as estimated by in silico analysis. Scale bar Z 5 mm (A). IB, immunoblot.

(11)

Figure 8 Lithium attenuates podocyte effacement and ameliorates proteinuria and progressive glomerulosclerosis in experimental Adriamycin (ADR; doxorubicin) nephropathy. A: Mice were injured with ADR or an equal volume of vehicle 6 hours after a single i.p. injection of 40 mg/kg of lithium chloride (LiCl) or an equal molar amount (1 mEq/kg) of sodium chloride as saline. Urine was collected at the indicated time points and was subjected to SDS-PAGE and staining with Coomassie Brilliant Blue. Bovine serum albumin (BSA), 5, 10, 20, and 40 mg, served as standard control. Urine samples (1.5 mL) collected on the indicated postinjury days from each group were loaded. B: Quantification of urine albumin levels adjusted with urine creatinine concentrations. C: Repre-sentative micrographs demonstrated periodic acideSchiff staining mouse kidneys. ADR-induced injury was featured by glomerular matrix accumulation and protein casts. D: Electron microscopy of kidney specimens procured from animals on day 7. Podocyte injury featured by extensive foot process effacement is evident in ADR-treated kidney, and lithium therapy significantly attenuates this lesion. E: Morphometric analysis of glomerulosclerosis scores on kidney sections prepared on day 14. F: Absolute count of the number of foot processes per unit length of glomerular basement membrane (GBM) on electron mi-crographs of kidney specimens. Data are given as means SD. n Z 6 (B, E, and F). *P < 0.05 versus control group (B, E, and F);yP< 0.05 versus LiCl þ ADR (B, E, and F). Scale bars: 20 mm (C); 2 mm (D).

(12)

process effacement in doxorubicin-injured mice, consistent with a podocyte protective and antiproteinuric effect. Control mice were treated with sodium or lithium 6 hours before vehicle injection, and no noticeable changes in proteinuria or renal histologic features were noted. These morphologic find-ings were further corroborated by the semiquantitative morphometric measurements of glomerulosclerosis scores and the absolute count of the number of foot processes per unit length of glomerular basement membrane (Figure 8, E and F).

Lithium Mitigates GSK3b Overactivity, Prevents

Paxillin Activation, and Reinstates Actin Cytoskeleton

Integrity in Doxorubicin-Injured Glomeruli

To examine the molecular changes associated with the lithium-induced remission of proteinuria, glomeruli were isolated from kidneys by the magnetic beadsebased approach and were homogenized for immunoblot analysis. Lithium treatment substantially enhanced the inhibitory phosphory-lation of GSK3b, suggestive of repressed GSK3b activity, concomitant with diminished phosphorylation of paxillin (Figure 9, A and B). In contrast, doxorubicin injury signi fi-cantly enhanced the activity of GSK3b, as marked by the reduced inhibitory phosphorylation of GSK3b associated with accentuated phosphorylation of paxillin on all observed time points. This effect was largely abolished by lithium treatment. To examine the effect of lithium on the ensuing actin cytoskeleton organization, kidney specimens procured

from animals on day 14 were subjected to phalloidin labeling for F-actin and to immunofluorescence staining for synapto-podin, a podocyte marker (Figure 9C). Confocalfluorescence microscopy demonstrated that intense F-actin was found to locate extensively to all over the glomerular tufts in normal kidneys. Podocyte expression of F-actin was highlighted by co-localization of F-actin signals (red) with synaptopodin staining (green). Lithium treatment alone barely affected either F-actin expression or synaptopodin expression in glomeruli and podocytes. In contrast, doxorubicin induced prominent podocyte injury, as evidenced by reduced syn-aptopodin expression, and also diminished F-actin expression in the periphery of glomerular tufts, consistent with podocyte localization. In agreement, the co-localization of F-actin with synaptopodin was considerably lessened in doxorubicin-injured kidneys, suggestive of a disorganized actin cytoskel-etal network in the remnant intact podocytes. Lithium therapy largely abrogated this injurious effect of doxorubicin, pre-vented the reduction in synaptopodin expression, and rein-stated F-actin and synaptopodin coexpression in podocytes (Figure 9D).

Discussion

A growing body of evidence indicates that podocytes are motile cells and that podocyte foot process motility is vital for maintaining the structural and functional homeostasis of the glomerularfiltration barrier.1,5,6FAs, by which cells are Figure 9 Lithium counteracts the Adriamycin (ADR; doxorubicin)-induced GSK3b overactivity and paxillin hyperphosphorylation in glomeruli and re-instates actin cytoskeleton integrity in glomerular podocytes. A: Glomeruli were isolated from kidneys from differently treated animals by the magnetic beadsebased approach and were homogenized for immunoblot analysis for phosphorylated GSK3b, phosphorylated paxillin, total GSK3b, total paxillin, and glyceraldehyde-3-phosphate dehydrogenase (GAPDH). B: Densitometric Western blot analysis estimates the relative levels of phosphorylated GSK3b/total GSK3b ratios and phosphorylated pax-illin/total paxillin ratios in isolated glomeruli from different groups. C: Frozen kidney sections procured on day 14 were subjected to phalloidin labeling of F-actin (red) as well as immunofluorescence staining for synaptopodin (green), a podocyte marker. Confocal microscopy images. ADR injury not only reduces synaptopodin expression but also di-minishes the integrated pixel density of the merged areas (yellow), where F-actin co-localizes with syn-aptopodin, suggesting a disorganized actin cyto-skeletal network in the remnant intact podocytes. Computerized morphometric analysis of the ratios of integrated pixel densities between yellow signal to green signal in immunofluorescence micrographs obtained in C and D. Data are given as means SD. nZ 6 (B and D). *P < 0.05 versus control group (B) or versus all other groups (D);yP< 0.05 versus LiCl þ ADR (B). Scale bar Z 20 mm (C).

(13)

anchored to the extracellular matrix, are a major determinant of actin cytoskeleton dynamics and cellular motility.15Thus, high FA turnover is associated with high motility of cells.20 To our knowledge, the present study is thefirst attempt to explore the GSK3b-controlled FA turnover and the ensuing effects on actin cytoskeleton organization and cell motility in podocytes (Figure 10). We demonstrated that lithium, a selective GSK3b inhibitor, protected podocytes from doxo-rubicin injury in vitro and in vivo. Although a variety of other mechanisms might also contribute, it seems that lithium conferred this podocyte protective action, at least in part, by counteracting the doxorubicin-elicited GSK3b overactivity; intercepting the GSK3b-directed hyperphosphorylation and overactivation of paxillin, a core structural component of FAs; and subsequently impeding FA turnover and overriding podocyte hypermotility (Figure 10).

GSK3b situates at the nexus of multiple crucial cell signaling pathways and is centrally involved in the pathogenesis of dis-ease in multifaceted organ systems, including the kidney.41As a redox-sensitive signaling transducer, the activity of GSK3b could be substantially enhanced on oxidative stress induced by a multitude of podocyte-injurious mediators, such as doxoru-bicin and chronic kidney injuries.42We recently uncovered that expression of GSK3b is aberrantly up-regulated in diseased human kidneys in tubules and glomeruli.43Similarly, Waters and Koziell44also noted up-regulation of GSK3b in human podocytes in association with specific NPHS1 mutations. Consistently, gene-targeted knock-in mice with mutated uninhibitable GSK3 developed albuminuria and podocyte injury, suggesting a detrimental role of GSK3 in podocyte injury.45In contrast, studies exploiting selective small mole-cule inhibitors of GSK3b reached conflicting conclusions. For example, inhibition of GSK3b by the selective small molecule inhibitor 6-bromoindirubin-30-oxime (BIO) at a low dose dramatically normalized proteinuria and attenuated histologic injury of glomeruli in rat models of diabetic nephropathy, although hyperglycemia was not corrected, implying direct antiproteinuric and renoprotective action.46However, Matsui et al47found that high-dose BIO exacerbated proteinuria and loss of glomerular nephrin in puromycin-injured rats. Another study by Dai et al48reported that a transient and low level of proteinuria followed by a rapid spontaneous remission was provoked by an ultrahigh dose of lithium chloride (16 mmol/ kg), which is almost two times the median lethal dose of lithium chloride in mice. In contrast, we demonstrated that low-dose lithium conferred prominent protection against podocyte injury. Of note, as typical chemical inhibitors of kinases, GSK3b blockers, including lithium, BIO, and 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione, if used at high doses, could have nonselective off-target effects and could induce cytotoxicity and even lethality.49Thus, the most likely expla-nation for these conflicting findings might be the difference in the doses of GSK3b inhibitor. Collectively, accumulating ev-idence indicates that GSK3b promotes podocyte injury and proteinuria, and inhibition of GSK3b by low-dose inhibitors might be beneficial for podocytopathy.

Lithium, a selective inhibitor of GSK3b, has been commonly and safely used for the past 50 years as a US Food and Drug Administrationeapproved first-line drug to treat bipolar affective disorders.50,51 Recent evidence revealed that blockade of GSK3b by lithium reduces cellular motility in a variety of cells, including vascular smooth muscle cells,26 glioma cells,27 gastric cancer cells,28 and airway epithelial cells,52suggesting that lithium might be a choice of therapy for diseases associated with cellular hypermotility, Figure 10 Schematic diagram detailing the mechanism of the GSK3b-governed FA dynamics in the pathogenesis of podocytopathy. GSK3b plays a key role in the regulation of FA turnover and podocyte motility by directing paxillin phosphorylation and activation and subsequently controlling actin cytoskeleton dynamics. As a redox-sensitive signaling transducer, the ac-tivity of GSK3b could be reinforced on oxidative stress induced by a variety of podocyte-injurious mediators, including Adriamycin (doxorubicin). GSK3b overactivity will elicit paxillin hyperphosphorylation and overactivation and thus cause the ensuing actin cytoskeleton disorganization and podocyte hypermotility, ultimately resulting in podocyte foot process effacement, massive proteinuria, and progressive glomerulosclerosis. GSK3b is a drug-gable target that could be blocked by lithium, a selective inhibitor of GSK3b and US Food and Drug Administrationeapproved mood stabilizer that has been safely used for>50 years as a first-line therapy for affective psychiatric disorders. Lithium treatment could override the Adriamycin-elicited GSK3b overactivity, counteract paxillin hyperphosphorylation and overactivation, and thereby obliterate podocyte hypermotility and reinstate actin cyto-skeleton integrity. Consequently, lithium therapy could ameliorate the Adriamycin-induced podocyte foot process effacement, induce proteinuria remission, and improve glomerulosclerosis.

(14)

such as malignant tumor metastasis. Podocyte hypermotility is a central pathogenic mechanism accounting for nephrotic glomerulopathy induced by a variety of mediators, including soluble urokinase-type plasminogen activator receptor,12 proteases, and nephrotoxins such as doxorubicin.9,10 Correction of podocyte hypermotility via therapeutic target-ing of FA dynamics, a prerequisite of cell migration and motility, has been shown to successfully override podocyte injuries induced by podocytopathic mediators and to improve the podocyte structure and function.23In this study, lithium, through stabilizing FA dynamics, also counteracted doxorubicin-elicited podocyte hypermotility and consis-tently resulted in a podocyte-protective and antiproteinuric effect in doxorubicin nephropathy. Apparently, this study might have an immediate implication for clinical translation into prophylactic treatment for recurrent focal and segmental glomerulosclerosis in kidney transplant patients, which has been attributed to a rapid podocytic injury associated with podocyte hypermotility caused by circulating permeability factors, such as soluble urokinase-type plasminogen acti-vator receptor.12,53

Of note, a basal level of podocyte motility is essential for sustaining the glomerular filtration barrier homeostasis. Podocyte motility that is too low secondary to genetic de-fects of cytoskeleton structural or regulatory molecules has been associated with cytopathic changes in podocytes that ultimately also result in focal and segmental glomerulo-sclerosis.1 Therefore, provided the observation that the lithium represses FA turnover and podocyte motility in normal podocytes, one of the conceivable concerns would be the potential podocytopathic effect of lithium therapy. Indeed, long-term lithium therapy primarily for psychiatric disorders has been complicated by some renal adverse ef-fects, such as nephrotic syndrome, glomerular disease, and interstitial nephritis, as reported by Markowitz et al54 in a case series report. However, according to a large-scale epidemiology study,55the incidence of chronic kidney dis-ease in lithium-treated patients is actually comparable with that in the general population, suggesting that the lithium-associated renal adverse effects are uncommon. Further-more, patients with lithium-associated kidney diseases usually have received lithium therapy at the psychiatric high dose for a long time (usually >10 years). In the present study, the single dose of lithium used (40 mg/kg) is much lower than the standard dose of lithium that has been safely and routinely used for neurobiology research (120 mg/kg) in rodents, and no detectable changes in glomerular histologic features or function were observed in control mice, sug-gesting that low-dose lithium might be protective for podocyte injury. Therefore, it seems that short-term use of low-dose lithium is safe in humans and might be a prom-ising approach for preventing podocytopathies.

In summary, GSK3b plays an important role in the regula-tion of FA turnover and podocyte motility by directing paxillin phosphorylation and activation and subsequently controlling actin cytoskeleton dynamics. Lithium, an inhibitor of GSK3b,

attenuated the doxorubicin-elicited paxillin phosphorylation and rapid FA turnover, reinstated actin cytoskeleton integrity, and overrode podocyte hypermotility. In experimental doxo-rubicin nephropathy, a single low dose of lithium effectively suppressed the overactivity of GSK3b and paxillin, recovered actin cytoskeleton in glomerular podocytes, prevented podo-cyte foot process effacement, and attenuated proteinuria (Figure 10). Collectively, this study suggests that the GSK3b-governed FA dynamics might serve as a novel therapeutic target for podocytopathy.

Supplemental Data

Supplemental material for this article can be found at http://dx.doi.org/10.1016/j.ajpath.2014.06.027.

References

1. Welsh GI, Saleem MA: The podocyte cytoskeleton: key to a functioning glomerulus in health and disease. Nat Rev Nephrol 2012, 8:14e21

2. Mundel P, Reiser J: Proteinuria: an enzymatic disease of the podo-cyte? Kidney Int 2010, 77:571e580

3. Mundel P, Shankland SJ: Podocyte biology and response to injury. J Am Soc Nephrol 2002, 13:3005e3015

4. Pavenstadt H, Kriz W, Kretzler M: Cell biology of the glomerular podocyte. Physiol Rev 2003, 83:253e307

5. Peti-Peterdi J, Sipos A: A high-powered view of thefiltration barrier. J Am Soc Nephrol 2010, 21:1835e1841

6. Kistler AD, Altintas MM, Reiser J: Podocyte GTPases regulate kid-neyfilter dynamics. Kidney Int 2012, 81:1053e1055

7. Wang L, Ellis MJ, Gomez JA, Eisner W, Fennell W, Howell DN, Ruiz P, Fields TA, Spurney RF: Mechanisms of the proteinuria induced by Rho GTPases. Kidney Int 2012, 81:1075e1085

8. Faul C, Asanuma K, Yanagida-Asanuma E, Kim K, Mundel P: Actin up: regulation of podocyte structure and function by components of the actin cytoskeleton. Trends Cell Biol 2007, 17:428e437

9. Koshikawa M, Mukoyama M, Mori K, Suganami T, Sawai K, Yoshioka T, Nagae T, Yokoi H, Kawachi H, Shimizu F, Sugawara A, Nakao K: Role of p38 mitogen-activated protein kinase activation in podocyte injury and proteinuria in experimental nephrotic syndrome. J Am Soc Nephrol 2005, 16:2690e2701

10. Liu H, Gao X, Xu H, Feng C, Kuang X, Li Z, Zha X: Alpha-Actinin-4 is involved in the process by which dexamethasone protects actin cytoskeleton stabilization from adriamycin-induced podocyte injury. Nephrology (Carlton) 2012, 17:669e675

11. Lennon R, Singh A, Welsh GI, Coward RJ, Satchell S, Ni L, Mathieson PW, Bakker WW, Saleem MA: Hemopexin induces nephrin-dependent reorganization of the actin cytoskeleton in podo-cytes. J Am Soc Nephrol 2008, 19:2140e2149

12. Wei C, El Hindi S, Li J, Fornoni A, Goes N, Sageshima J, Maiguel D, Karumanchi SA, Yap HK, Saleem M, Zhang Q, Nikolic B, Chaudhuri A, Daftarian P, Salido E, Torres A, Salifu M, Sarwal MM, Schaefer F, Morath C, Schwenger V, Zeier M, Gupta V, Roth D, Rastaldi MP, Burke G, Ruiz P, Reiser J: Circulating urokinase re-ceptor as a cause of focal segmental glomerulosclerosis. Nat Med 2011, 17:952e960

13. Koukouritaki SB, Tamizuddin A, Lianos EA: Enhanced expression of the cytoskeletal-associated protein, paxillin, in experimental nephrotic syndrome. J Investig Med 1998, 46:284e289

14. Carragher NO, Frame MC: Focal adhesion and actin dynamics: a place where kinases and proteases meet to promote invasion. Trends Cell Biol 2004, 14:241e249

(15)

15. Lauffenburger DA, Horwitz AF: Cell migration: a physically inte-grated molecular process. Cell 1996, 84:359e369

16. Gardel ML, Schneider IC, Aratyn-Schaus Y, Waterman CM: Me-chanical integration of actin and adhesion dynamics in cell migration. Annu Rev Cell Dev Biol 2010, 26:315e333

17. Wehrle-Haller B: Structure and function of focal adhesions. Curr Opin Cell Biol 2012, 24:116e124

18. Wehrle-Haller B: Assembly and disassembly of cell matrix adhesions. Curr Opin Cell Biol 2012, 24:569e581

19. Worth DC, Parsons M: Adhesion dynamics: mechanisms and mea-surements. Int J Biochem Cell Biol 2008, 40:2397e2409

20. Abou Zeid N, Valles AM, Boyer B: Serine phosphorylation regulates paxillin turnover during cell migration. Cell Commun Signal 2006, 4:8

21. Dai C, Stolz DB, Bastacky SI, St-Arnaud R, Wu C, Dedhar S, Liu Y: Essential role of integrin-linked kinase in podocyte biology: bridging the integrin and slit diaphragm signaling. J Am Soc Nephrol 2006, 17:2164e2175

22. Topham PS, Haydar SA, Kuphal R, Lightfoot JD, Salant DJ: Com-plement-mediated injury reversibly disrupts glomerular epithelial cell actin microfilaments and focal adhesions. Kidney Int 1999, 55: 1763e1775

23. Ma H, Togawa A, Soda K, Zhang J, Lee S, Ma M, Yu Z, Ardito T, Czyzyk J, Diggs L, Joly D, Hatakeyama S, Kawahara E, Holzman L, Guan JL, Ishibe S: Inhibition of podocyte FAK protects against pro-teinuria and foot process effacement. J Am Soc Nephrol 2010, 21: 1145e1156

24. Sun T, Rodriguez M, Kim L: Glycogen synthase kinase 3 in the world of cell migration. Dev Growth Differ 2009, 51:735e742

25. Wu X, Shen QT, Oristian DS, Lu CP, Zheng Q, Wang HW, Fuchs E: Skin stem cells orchestrate directional migration by regulating microtubule-ACF7 connections through GSK3beta. Cell 2011, 144: 341e352

26. Wang Z, Zhang X, Chen S, Wang D, Wu J, Liang T, Liu C: Lithium chloride inhibits vascular smooth muscle cell proliferation and migration and alleviates injury-induced neointimal hyperplasia via induction of PGC-1alpha. PLoS One 2013, 8:e55471

27. Nowicki MO, Dmitrieva N, Stein AM, Cutter JL, Godlewski J, Saeki Y, Nita M, Berens ME, Sander LM, Newton HB, Chiocca EA, Lawler S: Lithium inhibits invasion of glioma cells; possible involvement of glycogen synthase kinase-3. Neuro Oncol 2008, 10:690e699

28. Ryu YK, Lee YS, Lee GH, Song KS, Kim YS, Moon EY: Regu-lation of glycogen synthase kinase-3 by thymosin beta-4 is asso-ciated with gastric cancer cell migration. Int J Cancer 2012, 131: 2067e2077

29. Peng J, Ramesh G, Sun L, Dong Z: Impaired wound healing in hypoxic renal tubular cells: roles of hypoxia-inducible factor-1 and glycogen synthase kinase 3beta/beta-catenin signaling. J Pharmacol Exp Ther 2012, 340:176e184

30. Wang Z, Ge Y, Bao H, Dworkin L, Peng A, Gong R: Redox-sensitive glycogen synthase kinase 3beta-directed control of mitochondrial permeability transition: rheostatic regulation of acute kidney injury. Free Radic Biol Med 2013, 65C:849e858

31. Shankland SJ, Pippin JW, Reiser J, Mundel P: Podocytes in culture: past, present, and future. Kidney Int 2007, 72:26e36

32. Cho JH, Johnson GV: Primed phosphorylation of tau at Thr231 by glycogen synthase kinase 3beta (GSK3beta) plays a critical role in regulating tau’s ability to bind and stabilize microtubules. J Neuro-chem 2004, 88:349e358

33. Quizi JL, Baron K, Al-Zahrani KN, O’Reilly P, Sriram RK, Conway J, Laurin AA, Sabourin LA: SLK-mediated phosphorylation of paxillin is required for focal adhesion turnover and cell migration. Oncogene 2013, 32:4656e4663

34. Gong R, Rifai A, Ge Y, Chen S, Dworkin LD: Hepatocyte growth factor suppresses proinflammatory NFkappaB activation through GSK3beta inactivation in renal tubular epithelial cells. J Biol Chem 2008, 283:7401e7410

35. Matthew E, Berginski SMG: The Focal Adhesion Analysis Server: a web tool for analyzing focal adhesion dynamics. F1000Res 2013, 2:68

36. Committee for the Update of the Guide for the Care and Use of Laboratory Animals; National Research Council: Guide for the Care and Use of Laboratory Animals: Eighth Edition. Washington, DC, National Academies Press, 2011

37. Takemoto M, Asker N, Gerhardt H, Lundkvist A, Johansson BR, Saito Y, Betsholtz C: A new method for large scale isolation of kidney glomeruli from mice. Am J Pathol 2002, 161:799e805

38. Endlich N, Simon O, Gopferich A, Wegner H, Moeller MJ, Rumpel E, Kotb AM, Endlich K: Two-photon microscopy reveals stationary podocytes in living zebrafish larvae. J Am Soc Nephrol 2014, 25:681e686

39. Lee VW, Harris DC: Adriamycin nephropathy: a model of focal segmental glomerulosclerosis. Nephrology (Carlton) 2011, 16: 30e38

40. Wang Y, Wang YP, Tay YC, Harris DC: Progressive adriamycin nephropathy in mice: sequence of histologic and immunohisto-chemical events. Kidney Int 2000, 58:1797e1804

41. Wang Z, Havasi A, Gall J, Bonegio R, Li Z, Mao H, Schwartz JH, Borkan SC: GSK3beta promotes apoptosis after renal ischemic injury. J Am Soc Nephrol 2010, 21:284e294

42. Wang SH, Shih YL, Kuo TC, Ko WC, Shih CM: Cadmium toxicity toward autophagy through ROS-activated GSK-3beta in mesangial cells. Toxicol Sci 2009, 108:124e131

43. Gong R, Ge Y, Chen S, Liang E, Esparza A, Sabo E, Yango A, Gohh R, Rifai A, Dworkin LD: Glycogen synthase kinase 3beta: a novel marker and modulator of inflammatory injury in chronic renal allograft disease. Am J Transplant 2008, 8:1852e1863

44. Waters A, Koziell A: Activation of canonical Wnt signaling meets with podocytopathy. J Am Soc Nephrol 2009, 20:1864e1866

45. Boini KM, Amann K, Kempe D, Alessi DR, Lang F: Proteinuria in mice expressing PKB/SGK-resistant GSK3. Am J Physiol Renal Physiol 2009, 296:F153eF159

46. Lin CL, Wang JY, Huang YT, Kuo YH, Surendran K, Wang FS: Wnt/beta-catenin signaling modulates survival of high glucose-stressed mesangial cells. J Am Soc Nephrol 2006, 17:2812e2820

47. Matsui I, Ito T, Kurihara H, Imai E, Ogihara T, Hori M: Snail, a transcriptional regulator, represses nephrin expression in glomerular epithelial cells of nephrotic rats. Lab Invest 2007, 87:273e283

48. Dai C, Stolz DB, Kiss LP, Monga SP, Holzman LB, Liu Y: Wnt/beta-catenin signaling promotes podocyte dysfunction and albuminuria. J Am Soc Nephrol 2009, 20:1997e2008

49. Cohen P, Goedert M: GSK3 inhibitors: development and therapeutic potential. Nat Rev Drug Discov 2004, 3:479e487

50. Marmol F: Lithium: bipolar disorder and neurodegenerative dis-eases: possible cellular mechanisms of the therapeutic effects of lithium. Prog Neuropsychopharmacol Biol Psychiatry 2008, 32: 1761e1771

51. Yang ES, Wang H, Jiang G, Nowsheen S, Fu A, Hallahan DE, Xia F: Lithium-mediated protection of hippocampal cells involves enhancement of DNA-PK-dependent repair in mice. J Clin Invest 2009, 119:1124e1135

52. Wang WC, Kuo CY, Tzang BS, Chen HM, Kao SH: IL-6 augmented motility of airway epithelial cell BEAS-2B via Akt/GSK-3beta signaling pathway. J Cell Biochem 2012, 113:3567e3575

53. Morath C, Wei C, Macher-Goeppinger S, Schwenger V, Zeier M, Reiser J: Management of severe recurrent focal segmental glomer-ulosclerosis through circulating soluble urokinase receptor modi fica-tion. Am J Ther 2013, 20:226e229

54. Markowitz GS, Radhakrishnan J, Kambham N, Valeri AM, Hines WH, D’Agati VD: Lithium nephrotoxicity: a progressive combined glomerular and tubulointerstitial nephropathy. J Am Soc Nephrol 2000, 11:1439e1448

55. Bendz H, Schon S, Attman PO, Aurell M: Renal failure occurs in chronic lithium treatment but is uncommon. Kidney Int 2010, 77: 219e224

References

Related documents

perceived facilitators and barriers to implementing one or more of the following telemedicine modalities: telephone-based care, web-based screening, web-based

103 In the next year, the same Congress passed HIPAA, another provisions addressing the misuse of medical information protecting against the “discrimination

An extensive drill-hole measurement campaign was carried out in November and December 2011 collecting information on freeboard, snow depth and snow density, sea ice thick- ness

The hydrodynamic mechanism which is responsible for the link between the run-back flow pulsations and the impinging gas jet oscillation is well captured by the first five POD modes

Keywords: algebraic attacks, block ciphers, keystream generators, low-degree equa- tions, annihilators, algorithms, bounds..

gested that the United States should look at some variations of a free trade agreement to see if there are not ways of applying this approach to trade negotiations