You are on page 1of 11

Biology of glomerular podocytes

Author:
Pierre Ronco, MD, PhD
Section Editors:
Richard J Glassock, MD, MACP
Brad H Rovin, MD
Deputy Editor:
John P Forman, MD, MSc

Contributor Disclosures

All topics are updated as new evidence becomes available and our peer review
process is complete.
Literature review current through: Sep 2016. | This topic last updated: Jul 12,
2013.

INTRODUCTION — The healthy kidney filters metabolic byproducts into the urine but
prevents the passage of albumin and other larger essential molecules. This selective
filtration occurs across the glomerular capillary wall:

●Under normal circumstances, the glomerular capillary wall is extremely


permeable to water and small solutes, but negligibly to albumin or other proteins
of equivalent molecular weight or larger.
●Defects in the glomerular capillary wall result in increased permeability to
albumin and proteins of similar size or even larger, causing proteinuria.
●Electrical potential differences generated by transglomerular flow may modulate
the flux of anionic (charged) albumin across the glomerular capillary wall [1].

The traditional view that the glomerular capillary wall hinders the transit of protein is
largely based upon micropuncture studies that demonstrated very low concentrations
of albumin in Bowman's space in non-nephrotic animals [2,3]. This view has been
challenged by newer data that have demonstrated by intravital 2-photon microscopy,
much higher concentrations of albumin in Bowman's space than were reported
previously [4]. Given the novelty of the technique of intravital 2-photon microscopy,
these observations need to be validated in other systems and species, especially since
they substantially alter our understanding of the pathogenesis of proteinuria [5-7].

It is not known, however, why the filter does not routinely clog with large proteins that
enter the glomerular basement membrane (GBM). It has been hypothesized that
proteins cross the GBM mainly by diffusion rather than by liquid flow, whereas water
crosses entirely by flow [8]. An active transport that removes immunoglobulins that
accumulate at the filtration barrier has been identified [9]. This transport involves the
neonatal Fc receptor (FcRn). In a murine model, IgG accumulated in the GBM of FcRn-
deficient mice as they aged, and tracer studies showed delayed clearance of IgG from
the kidneys of FcRn-deficient mice [9]. Genetic or acquired impairment of the clearance
machinery may be a common mechanism promoting glomerular diseases.

Over the last decade, studies among patients with hereditary proteinuric syndromes
have markedly advanced our knowledge of the structure and composition of the
glomerular capillary wall and the changes in its composition that lead to proteinuria.
However, the changes in the glomerular capillary wall that underlie proteinuria in
hereditary diseases do not necessarily account for the proteinuria that accompanies
acquired causes of nephrotic syndrome, which are much more common. Thus, the
underlying molecular mechanisms of acquired proteinuric diseases remain less well
characterized.

It is increasingly appreciated that defects in podocyte structure and function result in


increased glomerular permeability. This topic review will focus primarily on the biology
of glomerular podocytes and defects in podocyte structure that appear to underlie
many proteinuric renal diseases.

Reviews of clinical proteinuria, including causes, manifestations, and treatment of


specific glomerular disease, are presented separately. (See "Epidemiology,
classification, and pathogenesis of focal segmental glomerulosclerosis" and "Treatment
of primary focal segmental glomerulosclerosis".)

GLOMERULAR CAPILLARY WALL — To better appreciate the biology of glomerular


podocytes, it is important to understand the structural and functional anatomy of the
glomerular capillary wall.

The glomerular capillary wall, through which the filtrate must pass, consists of the
following three layers:

●Fenestrated capillary endothelium, extensively coated with a layer of polyanionic


glycosoaminoglycans and glycoproteins.
●Glomerular basement membrane (GBM), containing heparin sulfate and other
anionic glycosoaminoglycans.
●Podocytes (or epithelial cells), which are attached to the GBM by discrete foot
processes. The pores between the foot processes (slit pores) are closed by a thin
membrane called the slit diaphragm, which functions as a modified adherens
junction and may also be permeated by anatomical pores [10].

Defects in any of the three components of the glomerular capillary wall can lead to
proteinuria (picture 1) [11-13]. In addition, cross-talk between podocytes and
endothelial and mesangial cells are key to the maintenance of glomerular capillary wall
function. As examples, the production of vascular endothelial growth factor (VEGF) by
podocytes is necessary for the integrity of the glomerular endothelium [14], and the
upregulation and secretion of the podocyte protein, angiopoietin-like-4 (Angptl4) into
the glomerular capillary wall causes marked proteinuria in experimental models of
nephrotic syndrome [15].

PODOCYTES — Podocytes are terminally differentiated epithelial cells that have large
cell bodies and long primary or major processes. The primary processes attach to the
underlying GBM via multiple foot processes. Adhesion molecules, such as alpha3beta1
integrin complex and dystroglycan (which are present on the basal membrane of foot
processes), attach the podocyte to the GBM [16].
Slit diaphragm — The interdigitating foot processes of adjacent podocytes are joined
laterally by slit diaphragms that bridge the intervening filtration slits. The following
proteins have been found to comprise the slit diaphragm [17]:

●Nephrin [18]
●Neph1 and Neph2 [19-22]
●FAT1 and FAT 2 [23]
●Podocin [24]
●Transient receptor potential cation channel 6 (TRPC6) [25,26].
●Tight junction proteins, including junctional adhesion molecule A, occludin and
cingulin [27]

Slit diaphragms interact with the actin cytoskeleton of podocyte foot processes via
linker proteins. These include CD2-associated protein (CD2AP) [28-30], Nck [31,32],
zona occludens-1, and the catenins. Selective deletion of Nck expression in podocytes
of adult mice rapidly leads to proteinuria, glomerulosclerosis, and altered morphology
of foot processes [33]. Mutations of some of the genes that encode slit diaphragm
proteins cause rearrangement of the actin cytoskeleton, which results in foot process
effacement and proteinuria [33].

Nephrin and Neph1 also interact directly with the Par3-aPKC protein complex at the
podocyte intercellular junction, suggesting that this complex plays a key role in the
establishment and maintenance of podocyte polarity as it does in other polarized
epithelia and neurons [34-36].

Reviews of the evidence showing that abnormalities of these proteins are causative in
some congenital disorders are presented separately. (See "Congenital and infantile
nephrotic syndrome" and "Epidemiology, classification, and pathogenesis of focal
segmental glomerulosclerosis".)

Actin cytoskeleton — Podocyte architecture is maintained by a contractile apparatus


composed of microfilaments comprising actin, type I and type II myosins, alpha-actinin,
talin, paxillin, and vinculin, and palladin [37,38]. This cytoskeleton helps to support the
glomerular capillary wall, which is subject to the high hydrostatic pressure necessary
for glomerular filtration and plays a key role in the foot process architecture (figure 1).
The actin cytoskeleton also contributes to the highly motile character of these
specialized cells.

Immunofluorescence and confocal microscopy have demonstrated two distinct actin


cytoskeletal networks present in the foot processes [39]:

●Dense actin bundles are present above the level of the slit diaphragm and
extend parallel to the longitudinal axis.
●A cortical actin network extends just below the plasma membrane of the foot
processes.
Different actin-binding proteins are associated with each of these networks. Thus,
whereas alpha-actinin and synaptopodin are associated with the actin bundles above
the slit diaphragm, the cortical actin network co-localizes with cortactin [39].

Foot process effacement — Flattening of foot processes or foot process effacement


is a characteristic histologic finding associated with most primary and secondary
glomerular diseases. Rearrangement of the actin cytoskeleton is necessary for foot
process effacement to occur and stabilization of the actin cytoskeleton decreases
proteinuria. The actin cytoskeleton is a fluid structure that can be quickly and reversibly
reorganized. It is a direct target of cyclosporine A, which is an effective treatment for
reducing nephrotic range proteinuria among patients with minimal change disease [40].
The effect of cyclosporine A on the cytoskeleton is independent of (though does not
exclude) its effect on T cell dysfunction [40].

Actin is normally organized into coordinated stress fibers in mature foot processes.
Podocyte injury causes the rearrangement of actin to a dense network [16].

The cytoskeleton can be altered by at least four different mechanisms:

●Direct injury of podocytes can occur by systemic or locally produced toxins (ie,
reactive oxygen species), viral infection, drugs (pamidronate, interferon) or local
activation of the renin angiotensin system [41].
●Abnormalities of cytoskeletal structural proteins can adversely affect cytoskeletal
dynamics. An example is mutations of alpha-actinin-4, which cause hereditary
focal segmental glomerular sclerosis [42]. This abnormality increases the affinity
of alpha-actinin-4 for actin, which may alter cytoskeletal fluidity.
(See "Epidemiology, classification, and pathogenesis of focal segmental
glomerulosclerosis".)

The disruption of myosin 1e in mice promotes podocyte injury with foot process
effacement [43]. Genetic variations in the MYH9 locus are associated with
progressive nondiabetic proteinuric kidney disease in African Americans [44,45].
MYH9 encodes nonmuscle myosin heavy chain type II isoform A, expressed by
podocytes and other cells in the kidney. (See "Epidemiology, classification, and
pathogenesis of focal segmental glomerulosclerosis", section on 'FSGS in African
Americans'.)
●Injury to the slit diaphragm, arising from congenital or acquired disorders, can
initiate abnormal actin and nephrin signaling, resulting in cytoskeletal
reorganization [11,16-18,21,22,46].
●Changes in the structure of the GBM may lead to cytoskeletal derangements, as
observed in laminin beta 2 deficient mice, in which proteinuria precedes foot
process effacement [47].

Direct podocyte injury — Podocytes may be direct targets of injury in acquired


proteinuric diseases [48]. Injury may be immunologic or non-immunologic. An example
of an immunologic cause of podocyte injury is the effect of interleukin-13 (IL-13)
[49,50]. Accumulating data have suggested that IL-13 is a permeability factor that
causes minimal change disease. (See "Etiology, clinical features, and diagnosis of
minimal change disease in adults".) Isolated human and rat glomeruli and cultured
human and rat podocytes express receptors for IL-13, and the exposure of cells to IL-
13 initiates intracellular signaling, changes in transepithelial resistance, and changes in
protein trafficking [49,50]. These data suggest that a direct effect of IL-13 on podocytes
may underlie minimal change disease.

Non-immunologic causes of podocyte injury include viral infection (specifically HIV),


drug toxicity (eg, pamidronate), local activation of the renin-angiotensin system (RAS),
and reactive oxygen species (ROS).

HIV nephropathy is characterized by collapsing focal segmental glomerulosclerosis.


(See "HIV-associated nephropathy (HIVAN)".)

Clinical studies and animal models have shown that the podocyte is directly infected by
HIV genes and may provide a reservoir for replicating virus [51,52]. Exogenous
expression of HIV genes, Nef and Vpr, causes dedifferentiation, proliferation and loss
of contact inhibition, and apoptosis of podocytes [51-58]. Some believe that parvo B19
virus may have similar toxic effects as HIV on podocyte [48].

Inhibition of systemic RAS limits the progression of chronic kidney disease in the
setting of proteinuria [59]. This is due, in part, to a reduction of transglomerular
pressure. However, locally produced angiotensin may also have direct toxic effects on
the podocyte. Angiotensin receptors -1 and -2 are expressed by podocytes [60].
Mechanical strain of cultured podocytes increases expression of the angiotensin
receptor-1 as well as angiotensin-II and enhances podocyte apoptosis, which is
abrogated by angiotensin receptor blockade [41]. Furthermore, a transgenic rat that
overexpresses the angiotensin receptor-1, specifically in podocytes, is characterized by
proteinuria and structural changes in the podocyte that include effacement and
detachment [61].

Reactive oxygen species are produced in a number of conditions including puromycin


aminonucleoside nephrosis [62], complement activation, and metabolic disorders such
as obesity. In obese patients that are prone to develop focal segmental
glomerulosclerosis, albuminuria is negatively correlated with plasma adiponectin, while
adiponectin knock-out mice exhibit albuminuria and effacement of podocyte foot
processes. The protective effect of adiponectin seems to occur through activation of
adiponectin receptors expressed on the podocyte membrane and reduction of oxidative
stress [63].

A number of signaling pathways may lead to podocyte injury and proteinuria. In human
proteinuric kidney diseases such as diabetic nephropathy and focal segmental
glomerulosclerosis, upregulation of Wnt1 and active beta-catenin is observed in
podocytes, while podocyte-specific knock-out of beta-catenin protects against
development of albuminuria [64]. Podocytes possess the complete machinery for
glutamatergic signaling, and derangements in that signaling may lead to proteinuric
kidney diseases [65]. The Notch pathway seems to be involved in the development of
proteinuria because Notch intracellular domain is detected in injured podocytes and
genetic inactivation or pharmacologic inhibition of Notch ameliorates proteinuria and
podocyte damage [66].

Podocyte-specific deletion of Dicer, an enzyme that generates microRNA (small,


noncoding RNA that function as important regulators of gene expression) alters
cytoskeletal dynamics, and induces proteinuria and glomerulosclerosis [67,68].

Podocyte depletion — A decrease in podocyte number resulting from apoptosis or


detachment from the GBM may contribute to proteinuria and progressive
glomerulosclerosis [69-72]. In many glomerular disorders that are characterized by
hypertrophy, the volume of the glomerular tuft increases, despite the decrease in
podocyte number [71]. The resulting decrease in podocyte density causes areas of
denuded GBM, which are foci for adhesions to parietal epithelial cells and eventual
crescent formation [70].

Cytoskeletal rearrangement — The mechanism by which podocyte injury or


mutations in genes for slit diaphragm proteins initiate rearrangement of the
cytoskeleton is unclear. Similar intracellular events, such as phosphorylation of
constituent proteins, appear to occur during development of the foot processes and
after podocyte injury. During podocyte differentiation, for example, nephrin is transiently
phosphorylated on multiple tyrosine residues by the Src family kinase, Fyn [31].
Phosphorylation of nephrin allows its interaction with Nck, which is required for actin
reorganization [73]. Decreased nephrin phosphorylation may occur following podocyte
injury [31-33].

Another possible mechanism of cytoskeletal rearrangement is via the degradation of


dynamin, a GTPase protein that is generally considered to have a key role in deforming
cellular membranes. Dynamin associates with actin, possibly via the actin-binding
protein, cortactin [74,75].

Cleavage of dynamin by cathepsin-L may initiate reorganization of the cytoskeleton,


resulting in clinical disease. Support for this is provided by findings in animal models
and some human glomerular diseases [76,77] (figure 2):

●Levels of cathepsin-L were found to be increased in a murine model of minimal


change disease [76]. This increase has also been observed in patients with
membranous nephropathy, focal segmental glomerulosclerosis, and diabetic
nephropathy, although significant increases were not found in those with other
proteinuric kidney diseases [77].
●Proteinuria and foot process effacement induced by lipopolysaccharide are
abrogated in cathepsin-L-knockout mice [76].
●Whereas the expression of a cathepsin-L-degraded dynamin fragment causes
foot process effacement and proteinuria, the expression of cathepsin-L-resistant
dynamin mutants decreases proteinuria

Cytoskeletal rearrangement may also be induced by the urokinase receptor (uPAR) via
its activation of the vitronectin receptor, alphavbeta3 integrin [78]. The absence of
uPAR, beta3 integrin or the alphavbeta3 integrin ligand, vitronectin, protects mice from
developing lipopolysaccharide-induced proteinuria. This protection is abolished by
exogenous expression of either uPAR or a constitutively active beta3 integrin [78].
uPAR may be required to activate alphavbeta3 integrin in podocytes promoting cell
motility and activation of small GTPases.

Proteinuria as a toxin — Although massive proteinuria may occur in the absence of


obvious structural change in the slit diaphragm and foot processes, sustained
proteinuria event is invariably eventually associated with foot process effacement
[47,79-82]. The mechanism underlying this phenomenon is unclear. One possibility is
that exposure of podocytes to an increased concentration of albumin and IgG is
sufficient to induce changes in the cytoskeleton [83].

SUMMARY

●The glomerular capillary wall consists of the fenestrated capillary endothelium,


glomerular basement membrane (GBM), and glomerular podocytes (or epithelial
cells). Defects in any of the three components of the glomerular capillary wall can
lead to proteinuria. (See 'Glomerular capillary wall' above.)
●Glomerular podocytes are terminally differentiated epithelial cells that have large
cell bodies and long primary or major processes. The primary processes attach to
the underlying GBM via multiple foot processes. (See 'Podocytes' above.)
●The interdigitating foot processes of adjacent podocytes are joined laterally by
slit diaphragms that bridge the intervening filtration slits. A number of proteins
have been found to comprise the slit diaphragm; these include nephrin, neph1,
neph2, FAT1, FAT 2, podocin, TRPC6 and tight junction proteins. (See 'Slit
diaphragm' above.)
●Foot process effacement of glomerular podocytes is a characteristic histologic
finding associated with glomerular diseases affecting the podocyte.
Rearrangement of the actin cytoskeleton is necessary for foot process effacement
to occur. (See 'Foot process effacement'above.)
●Possible molecular mechanisms underlying cytoskeletal rearrangement
include phosphorylation/dephosphorylation of nephrin and cleavage of dynamin (a
GTPase protein) by cathepsin-L. Activation of the urokinase receptor (uPAR) and
initiation of beta3 integrin signaling may also be involved (see 'Cytoskeletal
rearrangement' above).
Use of UpToDate is subject to the Subscription and License Agreement.

REFERENCES

1. Hausmann R, Kuppe C, Egger H, et al. Electrical forces determine glomerular


permeability. J Am Soc Nephrol 2010; 21:2053.
2. Oken DE, Flamenbaum W. Micropuncture studies of proximal tubule albumin
concentrations in normal and nephrotic rats. J Clin Invest 1971; 50:1498.
3. Tojo A, Endou H. Intrarenal handling of proteins in rats using fractional micropuncture
technique. Am J Physiol 1992; 263:F601.
4. Russo LM, Sandoval RM, McKee M, et al. The normal kidney filters nephrotic levels of
albumin retrieved by proximal tubule cells: retrieval is disrupted in nephrotic states.
Kidney Int 2007; 71:504.
5. Gekle M. Renal albumin handling: a look at the dark side of the filter. Kidney Int 2007;
71:479.
6. Birn H, Christensen EI. Renal albumin absorption in physiology and pathology. Kidney
Int 2006; 69:440.
7. Comper WD, Haraldsson B, Deen WM. Resolved: normal glomeruli filter nephrotic
levels of albumin. J Am Soc Nephrol 2008; 19:427.
8. Smithies O. Why the kidney glomerulus does not clog: a gel permeation/diffusion
hypothesis of renal function. Proc Natl Acad Sci U S A 2003; 100:4108.
9. Akilesh S, Huber TB, Wu H, et al. Podocytes use FcRn to clear IgG from the
glomerular basement membrane. Proc Natl Acad Sci U S A 2008; 105:967.
10. Gagliardini E, Conti S, Benigni A, et al. Imaging of the porous ultrastructure of the
glomerular epithelial filtration slit. J Am Soc Nephrol 2010; 21:2081.
11. Kalluri R. Proteinuria with and without renal glomerular podocyte effacement. J Am Soc
Nephrol 2006; 17:2383.
12. Singh A, Satchell SC, Neal CR, et al. Glomerular endothelial glycocalyx constitutes a
barrier to protein permeability. J Am Soc Nephrol 2007; 18:2885.
13. Jefferson JA, Shankland SJ, Pichler RH. Proteinuria in diabetic kidney disease: a
mechanistic viewpoint. Kidney Int 2008; 74:22.
14. Eremina V, Jefferson JA, Kowalewska J, et al. VEGF inhibition and renal thrombotic
microangiopathy. N Engl J Med 2008; 358:1129.
15. Clement LC, Avila-Casado C, Macé C, et al. Podocyte-secreted angiopoietin-like-4
mediates proteinuria in glucocorticoid-sensitive nephrotic syndrome. Nat Med 2011;
17:117.
16. Kerjaschki D. Caught flat-footed: podocyte damage and the molecular bases of focal
glomerulosclerosis. J Clin Invest 2001; 108:1583.
17. Tryggvason K, Patrakka J, Wartiovaara J. Hereditary proteinuria syndromes and
mechanisms of proteinuria. N Engl J Med 2006; 354:1387.
18. Kestilä M, Lenkkeri U, Männikkö M, et al. Positionally cloned gene for a novel
glomerular protein--nephrin--is mutated in congenital nephrotic syndrome. Mol Cell
1998; 1:575.
19. Donoviel DB, Freed DD, Vogel H, et al. Proteinuria and perinatal lethality in mice
lacking NEPH1, a novel protein with homology to NEPHRIN. Mol Cell Biol 2001;
21:4829.
20. Sellin L, Huber TB, Gerke P, et al. NEPH1 defines a novel family of podocin interacting
proteins. FASEB J 2003; 17:115.
21. Ihalmo P, Palmén T, Ahola H, et al. Filtrin is a novel member of nephrin-like proteins.
Biochem Biophys Res Commun 2003; 300:364.
22. Barletta GM, Kovari IA, Verma RK, et al. Nephrin and Neph1 co-localize at the
podocyte foot process intercellular junction and form cis hetero-oligomers. J Biol Chem
2003; 278:19266.
23. Ciani L, Patel A, Allen ND, ffrench-Constant C. Mice lacking the giant protocadherin
mFAT1 exhibit renal slit junction abnormalities and a partially penetrant cyclopia and
anophthalmia phenotype. Mol Cell Biol 2003; 23:3575.
24. Boute N, Gribouval O, Roselli S, et al. NPHS2, encoding the glomerular protein
podocin, is mutated in autosomal recessive steroid-resistant nephrotic syndrome. Nat
Genet 2000; 24:349.
25. Reiser J, Polu KR, Möller CC, et al. TRPC6 is a glomerular slit diaphragm-associated
channel required for normal renal function. Nat Genet 2005; 37:739.
26. Winn MP, Conlon PJ, Lynn KL, et al. A mutation in the TRPC6 cation channel causes
familial focal segmental glomerulosclerosis. Science 2005; 308:1801.
27. Fukasawa H, Bornheimer S, Kudlicka K, Farquhar MG. Slit diaphragms contain tight
junction proteins. J Am Soc Nephrol 2009; 20:1491.
28. Schwarz K, Simons M, Reiser J, et al. Podocin, a raft-associated component of the
glomerular slit diaphragm, interacts with CD2AP and nephrin. J Clin Invest 2001;
108:1621.
29. Huber TB, Kottgen M, Schilling B, et al. Interaction with podocin facilitates nephrin
signaling. J Biol Chem 2001; 276:41543.
30. Huber TB, Schermer B, Müller RU, et al. Podocin and MEC-2 bind cholesterol to
regulate the activity of associated ion channels. Proc Natl Acad Sci U S A 2006;
103:17079.
31. Verma R, Kovari I, Soofi A, et al. Nephrin ectodomain engagement results in Src
kinase activation, nephrin phosphorylation, Nck recruitment, and actin polymerization. J
Clin Invest 2006; 116:1346.
32. Uchida K, Suzuki K, Iwamoto M, et al. Decreased tyrosine phosphorylation of nephrin
in rat and human nephrosis. Kidney Int 2008; 73:926.
33. Jones N, New LA, Fortino MA, et al. Nck proteins maintain the adult glomerular
filtration barrier. J Am Soc Nephrol 2009; 20:1533.
34. Hartleben B, Schweizer H, Lübben P, et al. Neph-Nephrin proteins bind the Par3-Par6-
atypical protein kinase C (aPKC) complex to regulate podocyte cell polarity. J Biol
Chem 2008; 283:23033.
35. Huber TB, Hartleben B, Winkelmann K, et al. Loss of podocyte aPKClambda/iota
causes polarity defects and nephrotic syndrome. J Am Soc Nephrol 2009; 20:798.
36. Hirose T, Satoh D, Kurihara H, et al. An essential role of the universal polarity protein,
aPKClambda, on the maintenance of podocyte slit diaphragms. PLoS One 2009;
4:e4194.
37. Faul C, Asanuma K, Yanagida-Asanuma E, et al. Actin up: regulation of podocyte
structure and function by components of the actin cytoskeleton. Trends Cell Biol 2007;
17:428.
38. Endlich N, Schordan E, Cohen CD, et al. Palladin is a dynamic actin-associated protein
in podocytes. Kidney Int 2009; 75:214.
39. Ichimura K, Kurihara H, Sakai T. Actin filament organization of foot processes in rat
podocytes. J Histochem Cytochem 2003; 51:1589.
40. Faul C, Donnelly M, Merscher-Gomez S, et al. The actin cytoskeleton of kidney
podocytes is a direct target of the antiproteinuric effect of cyclosporine A. Nat Med
2008; 14:931.
41. Durvasula RV, Petermann AT, Hiromura K, et al. Activation of a local tissue
angiotensin system in podocytes by mechanical strain. Kidney Int 2004; 65:30.
42. Michaud JL, Chaisson KM, Parks RJ, Kennedy CR. FSGS-associated alpha-actinin-4
(K256E) impairs cytoskeletal dynamics in podocytes. Kidney Int 2006; 70:1054.
43. Krendel M, Kim SV, Willinger T, et al. Disruption of Myosin 1e promotes podocyte
injury. J Am Soc Nephrol 2009; 20:86.
44. Kao WH, Klag MJ, Meoni LA, et al. MYH9 is associated with nondiabetic end-stage
renal disease in African Americans. Nat Genet 2008; 40:1185.
45. Kopp JB, Smith MW, Nelson GW, et al. MYH9 is a major-effect risk gene for focal
segmental glomerulosclerosis. Nat Genet 2008; 40:1175.
46. Hussain S, Romio L, Saleem M, et al. Nephrin deficiency activates NF-kappaB and
promotes glomerular injury. J Am Soc Nephrol 2009; 20:1733.
47. Jarad G, Cunningham J, Shaw AS, Miner JH. Proteinuria precedes podocyte
abnormalities inLamb2-/- mice, implicating the glomerular basement membrane as an
albumin barrier. J Clin Invest 2006; 116:2272.
48. Shankland SJ. The podocyte's response to injury: role in proteinuria and
glomerulosclerosis. Kidney Int 2006; 69:2131.
49. Van Den Berg JG, Aten J, Chand MA, et al. Interleukin-4 and interleukin-13 act on
glomerular visceral epithelial cells. J Am Soc Nephrol 2000; 11:413.
50. Van Den Berg JG, Aten J, Annink C, et al. Interleukin-4 and -13 promote basolateral
secretion of H(+) and cathepsin L by glomerular epithelial cells. Am J Physiol Renal
Physiol 2002; 282:F26.
51. Bruggeman LA, Ross MD, Tanji N, et al. Renal epithelium is a previously unrecognized
site of HIV-1 infection. J Am Soc Nephrol 2000; 11:2079.
52. Winston JA, Bruggeman LA, Ross MD, et al. Nephropathy and establishment of a renal
reservoir of HIV type 1 during primary infection. N Engl J Med 2001; 344:1979.
53. Sunamoto M, Husain M, He JC, et al. Critical role for Nef in HIV-1-induced podocyte
dedifferentiation. Kidney Int 2003; 64:1695.
54. He JC, Husain M, Sunamoto M, et al. Nef stimulates proliferation of glomerular
podocytes through activation of Src-dependent Stat3 and MAPK1,2 pathways. J Clin
Invest 2004; 114:643.
55. Husain M, D'Agati VD, He JC, et al. HIV-1 Nef induces dedifferentiation of podocytes in
vivo: a characteristic feature of HIVAN. AIDS 2005; 19:1975.
56. Zhong J, Zuo Y, Ma J, et al. Expression of HIV-1 genes in podocytes alone can lead to
the full spectrum of HIV-1-associated nephropathy. Kidney Int 2005; 68:1048.
57. Zuo Y, Matsusaka T, Zhong J, et al. HIV-1 genes vpr and nef synergistically damage
podocytes, leading to glomerulosclerosis. J Am Soc Nephrol 2006; 17:2832.
58. Shah SN, He CJ, Klotman P. Update on HIV-associated nephropathy. Curr Opin
Nephrol Hypertens 2006; 15:450.
59. Remuzzi G, Ruggenenti P, Perico N. Chronic renal diseases: renoprotective benefits of
renin-angiotensin system inhibition. Ann Intern Med 2002; 136:604.
60. Sharma M, Sharma R, Greene AS, et al. Documentation of angiotensin II receptors in
glomerular epithelial cells. Am J Physiol 1998; 274:F623.
61. Hoffmann S, Podlich D, Hähnel B, et al. Angiotensin II type 1 receptor overexpression
in podocytes induces glomerulosclerosis in transgenic rats. J Am Soc Nephrol 2004;
15:1475.
62. Mayrhofer C, Krieger S, Huttary N, et al. Alterations in fatty acid utilization and an
impaired antioxidant defense mechanism are early events in podocyte injury: a
proteomic analysis. Am J Pathol 2009; 174:1191.
63. Sharma K, Ramachandrarao S, Qiu G, et al. Adiponectin regulates albuminuria and
podocyte function in mice. J Clin Invest 2008; 118:1645.
64. Dai, C, JASN 2009, doi: 10:10. 1681/ASN.20090 10019.
65. Giardino, L, JASN 2009, doi: 10.1681/ASN.200812 1286.
66. Niranjan T, Bielesz B, Gruenwald A, et al. The Notch pathway in podocytes plays a role
in the development of glomerular disease. Nat Med 2008; 14:290.
67. Harvey SJ, Jarad G, Cunningham J, et al. Podocyte-specific deletion of dicer alters
cytoskeletal dynamics and causes glomerular disease. J Am Soc Nephrol 2008;
19:2150.
68. Shi S, Yu L, Chiu C, et al. Podocyte-selective deletion of dicer induces proteinuria and
glomerulosclerosis. J Am Soc Nephrol 2008; 19:2159.
69. Kim YH, Goyal M, Kurnit D, et al. Podocyte depletion and glomerulosclerosis have a
direct relationship in the PAN-treated rat. Kidney Int 2001; 60:957.
70. Kriz W, LeHir M. Pathways to nephron loss starting from glomerular diseases-insights
from animal models. Kidney Int 2005; 67:404.
71. Pagtalunan ME, Miller PL, Jumping-Eagle S, et al. Podocyte loss and progressive
glomerular injury in type II diabetes. J Clin Invest 1997; 99:342.
72. Lemley KV, Lafayette RA, Safai M, et al. Podocytopenia and disease severity in IgA
nephropathy. Kidney Int 2002; 61:1475.
73. Jones N, Blasutig IM, Eremina V, et al. Nck adaptor proteins link nephrin to the actin
cytoskeleton of kidney podocytes. Nature 2006; 440:818.
74. Schafer DA. Regulating actin dynamics at membranes: a focus on dynamin. Traffic
2004; 5:463.
75. Merrifield CJ, Feldman ME, Wan L, Almers W. Imaging actin and dynamin recruitment
during invagination of single clathrin-coated pits. Nat Cell Biol 2002; 4:691.
76. Reiser J, Oh J, Shirato I, et al. Podocyte migration during nephrotic syndrome requires
a coordinated interplay between cathepsin L and alpha3 integrin. J Biol Chem 2004;
279:34827.
77. Sever S, Altintas MM, Nankoe SR, et al. Proteolytic processing of dynamin by
cytoplasmic cathepsin L is a mechanism for proteinuric kidney disease. J Clin Invest
2007; 117:2095.
78. Wei C, Möller CC, Altintas MM, et al. Modification of kidney barrier function by the
urokinase receptor. Nat Med 2008; 14:55.
79. Topham PS, Kawachi H, Haydar SA, et al. Nephritogenic mAb 5-1-6 is directed at the
extracellular domain of rat nephrin. J Clin Invest 1999; 104:1559.
80. PIRANI CL, POLLAK VE, LANNIGAN R, FOLLI G. THE RENAL GLOMERULAR
LESIONS OF PRE-ECLAMPSIA: ELECTRON MICROSCOPIC STUDIES. Am J Obstet
Gynecol 1963; 87:1047.
81. Good KS, O'Brien K, Schulman G, et al. Unexplained nephrotic-range proteinuria in a
38-year-old man: a case of "no change disease". Am J Kidney Dis 2004; 43:933.
82. Branten AJ, van den Born J, Jansen JL, et al. Familial nephropathy differing from
minimal change nephropathy and focal glomerulosclerosis. Kidney Int 2001; 59:693.
83. Morigi M, Buelli S, Angioletti S, et al. In response to protein load podocytes reorganize
cytoskeleton and modulate endothelin-1 gene: implication for permselective
dysfunction of chronic nephropathies. Am J Pathol 2005; 166:1309.

Topic 3091 Version 12.0

You might also like