Abstract
Individuals with congenital renal hypoplasia display a defect in the growth of nephrons during development. Many genes that affect the initial induction of nephrons have been identified, but little is known about the regulation of postinductive stages of kidney development. In the absence of the growth factor bone morphogenic protein 7 (BMP7), kidney development arrests after induction of a small number of nephrons. The role of BMP7 after induction, however, has not been fully investigated. Here, we generated a podocyte-specific conditional knockout of BMP7 (Bmp7flox/flox;Nphs2-Cre+ [BMP7 CKO]) to study the role of podocyte-derived BMP7 in nephron maturation. By postnatal day 4, 65% of BMP7 CKO mice had hypoplastic kidneys, but glomeruli demonstrated normal patterns of laminin and collagen IV subunit expression. Developing proximal tubules, however, were reduced in number and demonstrated impaired cellular proliferation. We examined signaling pathways downstream of BMP7; the level of cortical phosphorylated Smad1, 5, and 8 was unchanged in BMP CKO kidneys, but phosphorylated p38 mitogen-activated protein kinase was significantly decreased. In addition, β-catenin was reduced in BMP7 CKO kidneys, and its localization to intracellular vesicles suggested that it had been targeted for degradation. In summary, these results define a BMP7-mediated regulatory axis between glomeruli and proximal tubules during kidney development.
Renal malformations account for most young children who have long-term renal failure and consequently require dialysis and transplantation.1,2 Among them, congenital renal hypoplasia is characterized by reduced renal mass and nephron number, the primary cause of which has been considered to be a growth defect of nephrons. Many genes that affect the initial induction of nephrons from progenitor cells have been identified, such as WT1,3 Pax2,4 Odd1,5 and Six26; however, we know little about the molecular mechanisms that control the later stages of kidney development, when there is a marked elongation of the tubular portion of each nephron.7
Bone morphogenic protein 7 (BMP7) has been identified as a growth factor important in early stages of kidney development. In its absence, kidney development is arrested after induction of a small number of nephrons8,9; however, the role of BMP7 in subsequent nephron maturation was not fully investigated because of the neonatal lethality of this mouse model. BMP7 is expressed in the ureteric bud and subsequently in the cap mesenchyme and then in the early tubules derived from the mesenchyme.8,10,11 As the nephron develops, BMP7 is expressed in podocytes of the maturing glomeruli.12,13 Because the size of each nephron increases during maturation of the kidney, we hypothesized that podocyte-derived BMP7 might be involved in driving tubular epithelial expansion. Thus, we generated a podocyte-specific conditional knockout mouse model of BMP7 (BMP7 CKO mice) and analyzed postnatal kidney development.
RESULTS
Localization of BMP7 and Its Receptors in Postnatal Kidneys
The major compartments of BMP7 expression are the derivatives of the ureteric bud, early nephrons, and the podocytes of immature glomeruli (Figure 1A). In situ hybridization localization of BMP receptors showed expression mainly in the nephrogenic zone of wild-type newborn kidneys (Figure 1B), consistent with previous reports,14,15 but also demonstrated wide expression pattern throughout the kidneys, including tubules and glomeruli.
Figure 1.
Expression of BMP7 and its receptors in postnatal kidneys. In situ hybridization with DIG-labeled probes. (A, a through f) BMP7 mRNA in control and mutant kidneys. Glomeruli are indicated by arrowheads (a) and dashed circles (c through f). Genotypes are indicated at the top of panels. At P0, BMP7 is expressed in glomeruli (arrowheads or circles) and in ureteric bud derivatives (ub) and renal vesicles (rv) of control BMP7flox/flox mice. No BMP7-expressing immature glomeruli are present in BMP7flox/flox;Nphs2-Cre+ mouse kidneys (b). At P4, BMP7 is expressed in glomeruli and S-shaped tubules (S). S-shaped tubules that are well developed, such as that on the right of Ae, are rarely found in BMP7flox/flox;Nphs2-Cre+ mouse kidneys (Af); BMP7 is also absent in mutant glomeruli (A, d and f; also see Figure 3). (g and h) Lower power view of BMP7 expression at P0 antisense (g) and sense (h). Bar = 200 μm in a; 50 μm in b; 0.5 mm in g. (B) BMP7 receptor expression at P0; antisense and sense probe staining is shown. The receptors are designated above each column. Low-power views (a through d) and high-power views (e through h) are shown. ActRII, activin receptor II; BMPRII, BMP receptor type II. Bar = 0.5 mm in a; 200 μm in e.
Generation of Podocyte-Specific Conditional Knockout Mouse Model of BMP7
Mice with floxed BMP7 exon 1 (BMP7flox/flox) were crossed with mice that express the Cre-recombinase from the NPHS2 gene promoter (see the Concise Methods section for the strain used in this study)16,17 (Supplemental Figure 3), finally obtaining mice homozygous for the BMP7 floxed allele and carrying the NPHS2-Cre transgene. During the course of the genotyping using tail DNA (Supplemental Figure 3), we observed that approximately one third of tail DNA showed evidence of recombination between LoxP sites, even though examination of recombination in the kidney showed the Nphs2-Cre to be podocyte specific (Supplemental Figure 1). When present, the extent of recombination between LoxP sites was variable, suggesting it did not result from germ line recombination; however, the degree of recombination in tail DNA correlated with the most severely affected kidneys. This is consistent with the expectation that kidneys in which there was already one recombined allele would require only a single, instead of two, Nphs2-Cre–mediated recombination events to inactivate BMP7 fully in podocytes and would show more dramatic phenotypes. Because the recombination at LoxP sites did not seem to be an inherited event, the BMP7flox/flox genotype designation is used in this report regardless of whether some degree of Cre-mediated recombination was observed in tail DNA. Importantly, true BMP7 null heterozygotes are reported to have normal kidney development,8,9 such that the phenotype reported here is indeed due to the loss of BMP7 expression by podocytes and is not a BMP7 heterozygous phenotype. Littermates that were homozygous for the floxed BMP7 allele but were negative for NPHS2-Cre (BMP7flox/flox [Nphs2-Cre−]]) were used as controls. Of note, the BMP7 floxed allele used in this study still carries the neomycin resistance gene (neor); however, comparison of BMP7flox/flox mice and wild-type littermates revealed no differences caused by the neor allele by itself. Mice of all genotypes were born in the expected Mendelian frequency.
To evaluate the ability of the NPHS2-Cre recombinase to excise the floxed BMP7 alleles in a podocyte-specific manner in developing kidneys, we examined BMP7 mRNA expression by in situ hybridization in newborn and postnatal day 4(P4) kidneys. In control kidneys, BMP7 expression was observed in podocytes of immature glomeruli (Figure 1A, a, c, and e) in addition to ureteric bud derivatives within the nephrogenic zone; however, in BMP7 CKO kidneys at P0 and P4, the expression was missing specifically in podocytes (Figure 1A), whereas ureteric bud expression was unchanged. A total of 142 (90%) of 158 glomeruli counted from five sections taken from P0 kidneys showed little or no BMP7 expression; among control glomeruli, 97% showed BMP7 expression.
Hypoplasia of BMP7 CKO Kidneys
At P0, both control and BMP7 CKO mice (n = 6 mutants) showed the typical gradient of renal development with normal-appearing nephrogenic zones (data not shown); however, by P4, approximately 65% (19 of 29) of BMP7 CKO mice had hypoplastic kidneys (Figure 2A, b versus a). Among all ages examined between P4 and P10, 40 of 72 BMP7 CKO mice had hypoplastic kidneys ranging from 70 to 20% normal size, compared with five of 77 control mice that had slightly hypoplastic kidneys. In BMP7 CKO kidneys, the nephrogenic zone was largely missing by P4 with only a single layer of maturing nephrons in the most dramatic cases, and the kidneys had irregular surface (Figure 2A, d versus c). Glomeruli appeared normal in mutants at P4 (Figure 2A, f versus e), although, by P7, in the most affected kidneys, we found proteinaceous material in some tubules (data not shown) and observed mild podocyte foot process effacement (Figure 2A, h versus g).
Figure 2.
Histologic analysis of kidneys. (A) P4 histology and P7 electron microscopy. Genotypes are designated at the top of each column. (a and b) Low-power views of BMP7flox/flox (a) and BMP7flox/flox;Nphs2-Cre+ (b) mice. (c and d) Cortex of a and b. Bar = 0.5 mm in a; 100 μm in c. The nephrogenic zone (NZ) is demarcated. G, Mature glomeruli; g, immature glomeruli. Immature glomeruli are not recognizable in BMP7flox/flox;Nphs2-Cre+ kidneys, and the NZ is absent along much of the periphery of the kidney. (e and f) High-power views of glomeruli. (g and h) Electron microscopy of P7 glomeruli; some foot process effacement is evident in BMP7flox/flox;Nphs2-Cre+ (h). Bar = 33 μm in e; 2.5 μm in g. (B) Histology of P10 kidneys. (a and b) Low-power views. (c and d) Medium-power views of cortex. (e and f) High-power view of glomeruli. Designations, bars as in A.
Impaired Nephrogenesis in BMP7 CKO Kidneys
To examine further why fewer nephrons were present in BMP7 CKO kidneys, we examined markers of early nephrogenesis, including BMP7 itself. At P0, BMP7 is mainly found in ureteric bud derivatives and to a lesser extent in the cap mesenchyme (Figure 1Ac). By P4, the ureteric bud derivatives and cap mesenchyme are no longer present, and structures most compatible with comma- and S-shaped bodies are present in the most peripheral zone (Figure 1Ae). The expression of BMP7 seems diminished in S-shaped bodies of mutant kidneys at P4 (Figure 1Af), suggesting that BMP7 expression in the presumptive podocytes is being lost and that this has a marked effect on nephrogenesis.
Consistent with this histology, Wnt418 was also expressed in comma- and S-shaped bodies at P4 in control kidneys (Figure 3C), but analogous structures generally appeared less well developed in BMP7 CKO kidneys (Figure 3D). Wt1, as a marker of glomeruli,3 also seemed to be expressed abnormally in mutant kidneys (Figure 3, E and F), with fewer capillary loop glomeruli present, replaced by abnormal Wt1-expressing structures that probably represented dysplastic glomeruli. Finally, staining with Lotus Tetragonolobus (TG) lectin, to define proximal tubule structure, demonstrated more poorly differentiated structures in the subnephrogenic zone area in BMP7 CKO kidneys as compared with control (Figure 3, G and H). These results indicate a role for BMP7 in the postinductive stage of nephrogenesis; however, at a later stage of development, at P10, mutant kidneys remained hypoplastic, but the histology was relatively normal, suggesting that dysplastic nephrons either achieved some degree of maturation or were not retained after nephrogenesis was complete (Figure 2B).
Figure 3.
Postnatal nephrogenesis. (A and B) Histology of P4 kidneys. S, S-shaped bodies. (B) Abnormal tubular structures are indicated by arrows. (C through H) Expression of markers for nephrogenesis in P4 kidneys. In situ hybridization with DIG-labeled antisense probes for Wnt4 (C and D) and WT1 (E and F). (G and H) Double immunostaining with WT1 (red) and TG lectin (green). Arrows indicate abnormal Wt1-expressing structures. (A, C, E, and G) BMP7flox/flox mice. (B, D, F, and H) BMP7flox/flox;Nphs2-Cre+ mice. Bar = 50 μm in A, C, and G; 100 μm in E.
Glomerular Maturation in BMP7 CKO Kidneys
Although there was a defect in nephron development in BMP7 CKO kidneys, the initial round of nephrons appeared to be structured normally. Glomeruli of BMP7 CKO kidneys seemed to have undergone the usual conversion in laminin and type IV collagen isoforms19 and other markers of podocyte and glomerular differentiation such as podocin, α3β1 integrin, platelet-endothelial cell adhesion molecule (CD31), and desmin were expressed normally in BMP7 CKO glomeruli (Figure 4).
Figure 4.
Glomerular maturation. Genotypes are designated above each column (left, BMP7+/+; right, BMP7flox/flox;Nphs2-Cre+) and the protein stained to the left of each row. (A) Glomerular basement membrane markers. Laminin (Lam) and type IV collagen (Col IV) expression in glomeruli of P4 kidneys. The subunits of laminin, including α2, β1, β2, and γ1, and the subunits of type IV collagen, including α2 and α4, were examined, as indicated at the left. (B) Glomerular differentiation markers. Podocin and α3β1 integrin stain podocytes, platelet-endothelial cell adhesion molecule (PECAM) stains capillary loops, and desmin stains mesangial cells. Magnification, ×60.
Reduced Nephron Size in BMP7 CKO Kidneys
Cebrian et al.7 quantitatively demonstrated in murine kidneys that the number of nephrons increases dramatically from the late embryonic to early postnatal period, in parallel with the growth of a kidney. Because a proximal tubule comprises the largest portion of a single nephron's mass,20 its size should have a significant impact on the size of each nephron as well as the overall size of the kidney. Decreased number of TG lectin–stained proximal tubular sections per WT1-stained glomeruli in BMP7 CKO kidneys demonstrated reduced proximal tubular mass (Figure 5) and, thus, indicated reduction of nephron size.
Figure 5.
Distribution of proximal tubules and glomeruli in P4 kidneys. (A through D) Double immunostaining with WT1 (red) and TG lectin (green). (A and C) BMP7flox/flox mice. (B and D) BMP7flox/flox;Nphs2-Cre+ mice. Bar = 0.5 mm in A; 50 μm in C. (E) Number of proximal tubular slices per glomerulus was determined by counting the numbers of TG lectin–stained proximal tubules and WT1-stained glomeruli in high-power fields. Comparison between kidneys from BMP7flox/flox;Nphs2-Cre− and BMP7flox/flox;Nphs2-Cre+ mice. Six high-power fields in each of three pairs of kidneys were examined. (E) Mean proximal tubule cross-sections per WT1-stained glomeruli for one pair of kidneys; error bars indicate the SE. The other two comparison pairs showed an equivalent difference. All comparisons, P < 0.001. Magnification, ×40.
Decreased Cellular Proliferation of Proximal Tubules in BMP7 CKO Kidneys
Immunohistochemistry to Ki-67, which is a marker for cellular proliferation, demonstrated a significant decrease in Ki-67–positive cells within proximal tubules of mutant compared with control kidneys (Figure 6, A through E). No difference in Ki-67 staining within glomeruli of BMP7 CKO kidneys was observed (Figure 6F), indicating that decreased cellular proliferation was specific to proximal tubules. Terminal deoxynucleotidyl transferase–mediated digoxigenin-deoxyuridine nick-end labeling TUNEL staining did not reveal a significant difference in apoptosis between mutant and control kidneys (data not shown).
Figure 6.
Cell proliferation in P4 kidneys. (A through D) Immunohistochemistry using antibody for Ki-67 (brown). (A and C) BMP7flox/flox mice. (B and D) BMP7flox/flox;Nphs2-Cre+ mice. (E and F) Ki-67–positive cells were counted in high-power fields of proximal tubules (E) and glomeruli (F). Proximal tubules were identified by morphologic criteria, including the presence of the brush border. Comparison between kidneys from BMP7flox/flox;Nphs2-Cre− and BMP7flox/flox;Nphs2-Cre+ mice. Data acquisition was as described in Figure 5. Bar = 100 μm in A; 33 μm in C. Differences were analyzed by t test. P < 0.001. Magnification, ×40.
Canonical Smad Pathway Was not Involved in the BMP7 Effect on Nephron Growth
Because Smad1, 5, and 8 are known to transduce signals upon interaction of BMP7 with its receptors,21–23 we examined the phosphorylation of Smad1, 5, and 8 (Figure 7). Immunohistochemistry demonstrated wide distribution of phosphorylated Smad1-, 5-, and 8-positive (phospho-Smad1, 5, and 8) cells in both control and BMP7 CKO kidneys (Figure 7, A and B). No significant difference in numbers of phospho-Smad1, 5, and 8 cells were observed between control and mutant proximal tubules, however; neither did Western blot of tissue from isolated cortex, from which the medullary papilla had been removed, reveal a difference in the level of phospho-Smad1, 5, and 8 (Figure 7C).
Figure 7.
Phospho-Smad1, 5, and 8 proteins expression. (A and B) Immunohistochemistry using antibody specific for phospho-Smad1, 5, and 8 (brown) in the cortex region of P4 kidneys. (A) BMP7flox/flox mice. (B) BMP7flox/flox;Nphs2-Cre+ mice. Bar = 100 μm. (C) Western blots of kidneys from BMP7flox/flox and BMP7flox/flox;Nphs2-Cre+ mice. The antibody used in the blot is designated to the left of each panel. Seventy-five micrograms of protein extracted from the renal cortex of a single P10 kidney of an individual mouse was loaded in each lane. The amount of phospho-Smad1, 5, and 8 proteins was controlled by the quantity of total Smad1, 5, and 8 and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) proteins.
Increased Inhibitory Smad7 in BMP7 CKO Kidneys
Smad7 has been identified as a member of the inhibitory Smads of the TGF-β superfamily signaling pathways.24 Smad7 may also be involved in noncanonical BMP signaling, as it has been shown to interact with p38 mitogen-activated protein kinase (MAPK) and β-catenin.25,26 To explore the presence of negative regulatory system for canonical Smad pathway, we examined expression of Smad7 by immunohistochemistry (Figure 8). In control kidneys, we noted Smad7 expression in nephrogenic zone, glomeruli, and tubulointerstitial region (Figure 8, A and C), consistent with the findings of previous reports.27,28 In BMP7 CKO kidneys, cytoplasmic expression of Smad7 was increased specifically in proximal tubular epithelium (Figure 8, B and D) that was identified by double-staining with TG lectin (Figure 8, E and F).
Figure 8.
Smad7 protein expression in P4 kidneys. (A through D) Immunohistochemistry using antibody for Smad7 (brown). (A, C, and E) BMP7flox/flox;Nphs2-Cre− mice. (B, D, and F) BMP7flox/flox;Nphs2-Cre+ mice. (A and B) Cortex. (C and D) Proximal tubules. Arrows indicate proximal tubules showing increased Smad7 in D and less staining in C. (E and F) Merged images with immunofluorescence staining for TG lectin (green) to indicate the location of proximal tubules. Bar = 100 μm in A; 50 μm in C and E.
Involvement of Smad-Independent Pathways
BMP also activate Smad-independent intracellular signaling pathways.29 In this study, because BMP7 maintained nephron growth through cellular proliferation of proximal tubular epithelium (Figure 6), we investigated the involvement of other signaling pathways that are known to promote cellular proliferation during organogenesis, such as p38 MAPK pathway and β-catenin–mediated pathways.
Phosphorylated p38 MAPK Localization.
Recently, p38 MAPK was demonstrated to be activated by BMP7 and to promote cellular proliferation in cultured renal collecting duct cells.30 Immunohistochemistry demonstrated wide distribution of phosphorylated p38 MAPK (phospho-p38 MAPK)-positive cells throughout the control kidneys (Figure 9A); however, in BMP7 CKO kidneys, we observed decreased staining specifically in proximal tubules (Figure 9, B and D). Western blot analysis of tissue from isolated cortex demonstrated marked decrease of phospho-p38 MAPK in BMP7 CKO kidneys (Figure 9E), consistent with the immunohistochemistry (Figure 9, B and D, compare with C and E).
Figure 9.
Phospho-p38 MAPK expression. (A through D) Immunohistochemistry using antibody specific for phospho-p38 MAPK (brown) in P4 kidneys. (A and C) BMP7flox/flox mice. (B and D) BMP7flox/flox;Nphs2-Cre+ mice. (A and B) Cortex. (C and D) Proximal tubules. Bar = 100 μm in A; 20 μm in C. (E) Signal bands of immunoblotting. Fifty micrograms of protein extracted from the renal cortex of P10 kidneys was loaded in each lane. Kidneys from BMP7flox/flox and BMP7flox/flox;Nphs2-Cre+ mice were examined. The amount of phospho-p38 MAPK protein was controlled by the quantity of total p38 MAPK and GAPDH proteins.
β-Catenin Localization.
β-Catenin, a major downstream effecter of “canonical” Wnt signals, is known to be involved in regulating the proliferation of epithelial cells during organogenesis.31 Although Wnt signals that specifically affect proximal tubular proliferation are yet to be described, it is also known that BMP signals may affect β-catenin expression during organogenesis.32 In control kidneys, immunofluorescence staining demonstrated basolateral expression in proximal tubules (Figure 10, A and C). In marked contrast, in BMP7 CKO kidneys, β-catenin was highly concentrated within intracellular vesicles of proximal tubular epithelium, suggesting it had been targeted for degradation (Figure 10, B and D). Western blots demonstrated a decreased amount of intact β-catenin in the cortex of BMP7 CKO kidneys (Figure 10E), with a shifted band suggestive of increased phosphorylation, consistent with its being targeted for degradation.
Figure 10.
β-Catenin expression. (A through D) Immunofluorescence staining using antibody specific for β-catenin (red) in P4 kidneys. (A and C) BMP7flox/flox mice. (B and D) BMP7flox/flox;Nphs2-Cre+ mice. Bar = 100 μm in A; 10 μm in C. (E) Signal bands of immunoblotting. Ten micrograms of protein extracted from the renal cortex of P10 kidneys was loaded in each lane. Kidneys from BMP7flox/flox;Nphs2-Cre− and BMP7flox/flox;Nphs2-Cre+ mice were examined. The amount of protein was controlled by the quantity of GAPDH protein.
DISCUSSION
This study was designed to assess the role of podocyte-derived BMP7 in nephron maturation. The original BMP7 knockout mice, developed independently by Dudley et al.8 and by Luo et al.,9 demonstrated an essential role for BMP7 in kidney development. There was a premature loss of the progenitor population, and severely hypoplastic kidneys that led to neonatal lethality were produced. In this study, the use of a conditional allele of BMP7 has allowed us to distinguish the role of BMP7 expressed from specific cell lineages, in this case podocytes, and has allowed the study of postnatal phenotypes that focus more specifically on nephron development. Here we show that podocyte expression of BMP7 has an important role both in early nephron development and in nephron maturation. BMP7 seems to signal through Smad-dependent and -independent pathways and regulate localization of β-catenin in developing tubules.
In the absence of podocyte expression of BMP7, nephron development seemed abnormal. In the most dramatically affected conditional mutant mice, many nephrons induced after the first round were severely dysgenic. This is reminiscent of the original BMP7 mutant mice,8,9 although the conditionally mutant mice developed to a much greater extent than the original null mice. Several explanations—that are not mutually exclusive—can be suggested to account for the difference between the podocyte-specific knockout and the original BMP7 null mice. First, the ureteric bud derivatives and their derivative collecting ducts also express BMP7 (Figure 1A). Consistent with this expression pattern, the development of the collecting system proceeds relatively normally in the conditional mutant mice in contrast to the null mice (Figure 2, A and B). Second, the distinct differences in the extent of development between the earliest nephrons and subsequent rounds suggest either that the earliest nephrons are less dependent on BMP7 in general or that they are less dependent on podocyte-derived BMP7. Because there are not great morphologic differences between early and late nephrons (with the exception of the extension of the loop of Henle into the medulla),20 and, to date, no known molecular genetic differences are known in the regulation of their development,33 it seems unlikely that there is a differing overall requirement for BMP7 among early and late nephron populations. Instead, it is possible that the earliest nephrons have access to other sources of BMP7, possibly from the ureteric bud derivatives, cap mesenchyme, or possibly maternal sources.34 As the nephrogenic zone develops into a more distinct structure, BMP7 expression in the cortex becomes progressively more restricted to ureteric bud derivatives and podocytes (Figure 1Aa). By P4, ureteric bud derivatives have largely disappeared, and podocytes remain the major location of BMP7 expression in the developing cortex (Figure 1Ac). Thus, it is likely that nephron development becomes progressively more dependent on podocyte expression of BMP7 during the course of nephrogenesis. It is also possible that the conditional mutation of the BMP7 locus was more efficient in later rounds of nephrons, although this is not suggested by our in situ analysis of BMP7 expression in conditional mutant mice.
As previously discussed, BMP proteins signal both through Smad-dependent and -independent pathways.29 For example, signaling through p38 MAPK seems to be important in the development of the collecting system30; however, it remains to be determined whether there is a clear distinction between so-called Smad-dependent and -independent pathways. On the one hand, staining for phospho-Smad1, 5, and 8 was not different between control and mutant kidneys. On the other hand, abundant staining and detection of phospho-Smad1, 5, and 8 on Western blots indicated that Smad signaling was indeed in effect in proximal tubules. In addition, increased Smad7 staining in mutant proximal tubules, along with decreased p38 staining, suggested a possible interaction between MAPK signaling and Smad-dependent signals. Supporting this possibility are reports of physical interactions between Smad7 and p38 that may regulate the abundance of p38,25 thereby affecting proliferation. Smad7 has been shown to be induced by stimulation with TGF-β.24 Because Smad7 is an inhibitory Smad, it has been suggested to be part of a negative feedback loop to regulate TGF-β signaling. Our results suggest that in contrast to TGF-β signals, BMP7 signals suppress Smad7 expression. Although it remains unclear whether Smad1, 5, and 8 have a role upstream (or downstream) of Smad7 in driving epithelial proliferation, it is possible that the increased levels of Smad7 provide an indication that epithelial proliferation is regulated by a balance between signals downstream of BMPs and TGF-β.
BMP7 has shown promise as a therapeutic agent to preserve renal epithelia.35–40 This raises the question of whether there is normally ongoing expression of BMP7 in the adult kidney and, if so, what its source is. The podocyte expression of BMP7, as judged by in situ hybridization, seems to diminish in the most mature glomeruli; however it is possible that expression of BMP7 in podocytes continues at a low level throughout adulthood. If this is the case, then it raises the question of whether tubule damage, that may follow glomerular damage, is in part due to a loss of BMP7 expression by the glomerulus. Furthermore, it remains possible that the phenotype we observed is due to secondary effects and that BMP7 is acting completely within the glomerulus. Further studies examining the presence of BMP7 protein will be required to answer this question. In addition, it was previously published that ectopic glomerular expression of Noggin, a BMP antagonist, led to glomerular damage in older mice.13 The full range of BMP expression in the glomerulus is not known, so it cannot be determined whether the Noggin effect relates specifically to BMP7 function. Moreover, our studies have focused on developmental phenotypes, and further study of older mice will be required for full examination of the function of BMP7 in the glomerulus.
CONCISE METHODS
Targeted Inactivation of BMP7 in Podocytes
A new strain of NPHS2-Cre mice was derived in our laboratory using a construct obtained from Dr. Susan Quaggin (Mt. Sinai Hospital, Toronto, Ontario, Canada). Characterization of this strain is shown in Supplemental Figures 1 through 3. NPHS2-cre mice were crossed with BMP7flox/flox, which contain loxP sites upstream and downstream of exon 1 of the BMP7 gene, in two successive rounds of breeding to obtain BMP7flox/flox;Nphs2-Cre+ mice, referred to as BMP7 CKO mice. A detailed report on the construction of this targeting vector and derivation of mutant mice has been submitted (D.G. and A.N.E., in preparation). All procedures with animals were approved by institutional animal care and use committees.
Genotyping
Genotyping information is presented in Supplemental Figure 3.
In Situ Hybridization
Tissue in situ hybridization was performed as described previously.41,42 Riboprobes were obtained or generated from the coding region of mouse: BMP7 (obtained from J.M. Wozney; Genetics Institute, Cambridge, MA), ALK3,43 ALK6,44 ActRII (obtained from V. Rosen, Harvard School of Dental Medicine, Boston, MA), BMPRII (obtained from L. Gamer; Harvard School of Dental Medicine, Boston, MA), Wnt4,18 and WT13 (generated in our laboratory). Sense and antisense probes were synthesized and labeled with digoxigenin-UTP (Roche, Mannheim, Germany).
Immunofluorescence Staining
Immunofluorescence staining was performed as described previously.45,46 Primary antibodies used were as follows: Mouse anti-WT1 (1:100 dilution; Santa Cruz Biotechnology, Santa Cruz, CA), TG lectin (1:100; Vector Laboratories, Burlingame, CA), and mouse anti–β-catenin (1:100; BD Transduction Laboratories, San Jose, CA). Rabbit anti-laminin α2, β1, β2, and γ1 and collagen IVα2 and α4 were used as described previously.19,45 Fluorescence images were captured off a Nikon Eclipse 800 microscope with a Spot 2 cooled color digital camera (Diagnostic Instruments, Sterling Heights, MI) using Spot Software 2.1. Secondary antibody alone was consistently negative on all sections.
Immunohistochemistry
Five-micrometer paraffin sections of 4% paraformaldehyde-fixed kidneys were placed in citrate-buffered solution (pH 6.0) and then boiled for 30 min for antigen retrieval. Endogenous peroxidase was blocked with 3% hydrogen peroxide, and nonspecific binding was blocked with 10% BSA. Primary antibodies used were as follows: Rabbit anti–Ki-67 (1:150; Vector Laboratories); anti–phospho-Smad1, 5, and 8 (1:100; Cell Signaling Technologies, Danvers, MA); anti-Smad7 (1:50; Santa Cruz Biotechnology); and anti–phospho-p38 MAPK (1:50; Cell Signaling Technologies). Diaminobenzidine substrate (Sigma Chemical Co., St. Louis, MO) was used for the color reaction. For Ki-67 staining, sections were counterstained with hematoxylin. Secondary antibody alone was consistently negative on all sections. Proximal tubules were distinguished from distal tubules in the cortex area by their luminal brush borders and relatively large cytoplasmic structures.
Programmed Cell Death Analysis
The analysis was carried out according to the manufacturer's protocol, using Apoptag in situ apoptosis detection kit (Chemicon Int., Temecula, CA).
Immunoblotting
Immunoblotting was performed as described previously.47 Briefly, kidneys were dissected from mice at P10. After dividing the harvested kidneys into cortex and medulla, the cortex was homogenized in 800 μl of RIPA buffer for protein extraction. The following primary antibodies were used: Rabbit anti–phospho-Smad1, 5, and 8; anti-Smad1, 5, and 8 (Santa Cruz Biotechnology); anti–phospho-p38 MAPK and anti-p38 MAPK (Cell Signaling Technologies); mouse anti–β-catenin (BD Transduction Laboratories); and rabbit anti–glyceraldehyde-3-phosphate dehydrogenase (Invitrogen, Carlsbad, CA).
Statistical Analysis
All results are expressed as means ± SEM. Paired evaluations were made for the comparison between control and BMP7 CKO groups. Statistical significance between means was determined with t test. P < 0.001 was considered significant.
DISCLOSURES
None.
Supplementary Material
Acknowledgments
This work was supported by Yoshida Scholarship Foundation Fellowship grant and Japan Society for the Promotion of Science Postdoctoral Fellowship for Research Abroad grant to I.K. and by grants from the National Institute of Diabetes and Digestive and Kidney Diseases to J.K.
Part of this work was presented in abstract form at the annual meeting of the American Society of Nephrology; November 1 through 5, 2007; San Francisco, CA.
We thank Mary Taglienti for assistance with mouse husbandry and Sunny Hartwig, Jacqueline Ho, Valerie Schumacher, and Vicki Rosen for helpful discussions.
Published online ahead of print. Publication date available at www.jasn.org.
Supplemental information for this article is available online at http://www.jasn.org/.
REFERENCES
- 1.Lewis M: Report of the pediatric renal registry. In: The UK Renal Registry: The Second Annual Report, edited by Adaf T, Bristol, Renal Association, 1999, pp 175–187
- 2.Neild G: Congenial abnormalities of the renal tract. In: Comprehensive Clinical Nephrology, edited by Johnson RA, London, Mosby, 2000, pp 9.55.51–59.55.15
- 3.Kreidberg JA, Sariola H, Loring JM, Maeda M, Pelletier J, Housman D, Jaenisch R: WT-1 is required for early kidney development. Cell 74: 679–691, 1993 [DOI] [PubMed] [Google Scholar]
- 4.Torres M, Gomez-Pardo E, Dressler GR, Gruss P: Pax-2 controls multiple steps of urogenital development. Development 121: 4057–4065, 1995 [DOI] [PubMed] [Google Scholar]
- 5.James RG, Kamei CN, Wang Q, Jiang R, Schultheiss TM: Odd-skipped related 1 is required for development of the metanephric kidney and regulates formation and differentiation of kidney precursor cells. Development 133: 2995–3004, 2006 [DOI] [PubMed] [Google Scholar]
- 6.Self M, Lagutin OV, Bowling B, Hendrix J, Cai Y, Dressler GR, Oliver G: Six2 is required for suppression of nephrogenesis and progenitor renewal in the developing kidney. EMBO J 25: 5214–5228, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Cebrian C, Borodo K, Charles N, Herzlinger DA: Morphometric index of the developing murine kidney. Dev Dyn 231: 601–608, 2004 [DOI] [PubMed] [Google Scholar]
- 8.Dudley AT, Lyons KM, Robertson EJ: A requirement for bone morphogenetic protein-7 during development of the mammalian kidney and eye. Genes Dev 9: 2795–2807, 1995 [DOI] [PubMed] [Google Scholar]
- 9.Luo G, Hofmann C, Bronckers AL, Sohocki M, Bradley A, Karsenty G: BMP-7 is an inducer of nephrogenesis, and is also required for eye development and skeletal patterning. Genes Dev 9: 2808–2820, 1995 [DOI] [PubMed] [Google Scholar]
- 10.Dudley AT, Robertson EJ: Overlapping expression domains of bone morphogenetic protein family members potentially account for limited tissue defects in BMP7 deficient embryos. Dev Dyn 208: 349–362, 1997 [DOI] [PubMed] [Google Scholar]
- 11.Lyons KM, Hogan BL, Robertson EJ: Colocalization of BMP 7 and BMP 2 RNAs suggests that these factors cooperatively mediate tissue interactions during murine development. Mech Dev 50: 71–83, 1995 [DOI] [PubMed] [Google Scholar]
- 12.Godin RE, Takaesu NT, Robertson EJ, Dudley AT: Regulation of BMP7 expression during kidney development. Development 125: 3473–3482, 1998 [DOI] [PubMed] [Google Scholar]
- 13.Miyazaki Y, Ueda H, Yokoo T, Utsunomiya Y, Kawamura T, Matsusaka T, Ichikawa I, Hosoya T: Inhibition of endogenous BMP in the glomerulus leads to mesangial matrix expansion. Biochem Biophys Res Commun 340: 681–688, 2006 [DOI] [PubMed] [Google Scholar]
- 14.Bosukonda D, Shih MS, Sampath KT, Vukicevic S: Characterization of receptors for osteogenic protein-1/bone morphogenetic protein-7 (OP-1/BMP-7) in rat kidneys. Kidney Int 58: 1902–1911, 2000 [DOI] [PubMed] [Google Scholar]
- 15.Martinez G, Loveland KL, Clark AT, Dziadek M, Bertram JF: Expression of bone morphogenetic protein receptors in the developing mouse metanephros. Exp Nephrol 9: 372–379, 2001 [DOI] [PubMed] [Google Scholar]
- 16.Moeller MJ, Sanden SK, Soofi A, Wiggins RC, Holzman LB: Podocyte-specific expression of cre recombinase in transgenic mice. Genesis 35: 39–42, 2003 [DOI] [PubMed] [Google Scholar]
- 17.Jones N, Blasutig IM, Eremina V, Ruston JM, Bladt F, Li H, Huang H, Larose L, Li SS, Takano T, Quaggin SE, Pawson T: Nck adaptor proteins link nephrin to the actin cytoskeleton of kidney podocytes. Nature 440: 818–823, 2006 [DOI] [PubMed] [Google Scholar]
- 18.Stark K, Vainio S, Vassileva G, McMahon AP: Epithelial transformation of metanephric mesenchyme in the developing kidney regulated by Wnt-4. Nature 372: 679–683, 1994 [DOI] [PubMed] [Google Scholar]
- 19.Miner JH: Building the glomerulus: A matricentric view. J Am Soc Nephrol 16: 857–861, 2005 [DOI] [PubMed] [Google Scholar]
- 20.Welling LW, Linshaw MA: Structural and functional development of outer versus inner cortical proximal tubules. Pediatr Nephrol 2: 108–114, 1988 [DOI] [PubMed] [Google Scholar]
- 21.Kawabata M, Imamura T, Miyazono K: Signal transduction by bone morphogenetic proteins. Cytokine Growth Factor Rev 9: 49–61, 1998 [DOI] [PubMed] [Google Scholar]
- 22.Macias-Silva M, Hoodless PA, Tang SJ, Buchwald M, Wrana JL: Specific activation of Smad1 signaling pathways by the BMP7 type I receptor, ALK2. J Biol Chem 273: 25628–25636, 1998 [DOI] [PubMed] [Google Scholar]
- 23.Tamaki K, Souchelnytskyi S, Itoh S, Nakao A, Sampath K, Heldin CH, ten Dijke P: Intracellular signaling of osteogenic protein-1 through Smad5 activation. J Cell Physiol 177: 355–363, 1998 [DOI] [PubMed] [Google Scholar]
- 24.Nakao A, Afrakhte M, Moren A, Nakayama T, Christian JL, Heuchel R, Itoh S, Kawabata M, Heldin NE, Heldin CH, ten Dijke P: Identification of Smad7, a TGFbeta-inducible antagonist of TGF-beta signalling. Nature 389: 631–635, 1997 [DOI] [PubMed] [Google Scholar]
- 25.Edlund S, Bu S, Schuster N, Aspenstrom P, Heuchel R, Heldin NE, ten Dijke P, Heldin CH, Landstrom M: Transforming growth factor-beta1 (TGF-beta)-induced apoptosis of prostate cancer cells involves Smad7-dependent activation of p38 by TGF-beta-activated kinase 1 and mitogen-activated protein kinase kinase 3. Mol Biol Cell 14: 529–544, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Han G, Li AG, Liang YY, Owens P, He W, Lu S, Yoshimatsu Y, Wang D, Ten Dijke P, Lin X, Wang XJ: Smad7-induced beta-catenin degradation alters epidermal appendage development. Dev Cell 11: 301–312, 2006 [DOI] [PubMed] [Google Scholar]
- 27.Lan HY, Mu W, Tomita N, Huang XR, Li JH, Zhu HJ, Morishita R, Johnson RJ: Inhibition of renal fibrosis by gene transfer of inducible Smad7 using ultrasound-microbubble system in rat UUO model. J Am Soc Nephrol 14: 1535–1548, 2003 [DOI] [PubMed] [Google Scholar]
- 28.Vrljicak P, Myburgh D, Ryan AK, van Rooijen MA, Mummery CL, Gupta IR: Smad expression during kidney development. Am J Physiol Renal Physiol 286: F625–F633, 2004 [DOI] [PubMed] [Google Scholar]
- 29.Nohe A, Keating E, Knaus P, Petersen NO: Signal transduction of bone morphogenetic protein receptors. Cell Signal 16: 291–299, 2004 [DOI] [PubMed] [Google Scholar]
- 30.Hu MC, Wasserman D, Hartwig S, Rosenblum ND: p38MAPK acts in the BMP7-dependent stimulatory pathway during epithelial cell morphogenesis and is regulated by Smad1. J Biol Chem 279: 12051–12059, 2004 [DOI] [PubMed] [Google Scholar]
- 31.Clevers H: Wnt/beta-catenin signaling in development and disease. Cell 127: 469–480, 2006 [DOI] [PubMed] [Google Scholar]
- 32.He XC, Zhang J, Tong WG, Tawfik O, Ross J, Scoville DH, Tian Q, Zeng X, He X, Wiedemann LM, Mishina Y, Li L: BMP signaling inhibits intestinal stem cell self-renewal through suppression of Wnt-beta-catenin signaling. Nat Genet 36: 1117–1121, 2004 [DOI] [PubMed] [Google Scholar]
- 33.Dressler GR: The cellular basis of kidney development. Annu Rev Cell Dev Biol 22: 509–529, 2006 [DOI] [PubMed] [Google Scholar]
- 34.Borovecki F, Jelic M, Grgurevic L, Sampath KT, Bosukonda D, Vukicevic S: Bone morphogenetic protein-7 from serum of pregnant mice is available to the fetus through placental transfer during early stages of development. Nephron Exp Nephrol 97: e26–32, 2004 [DOI] [PubMed] [Google Scholar]
- 35.Vukicevic S, Basic V, Rogic D, Basic N, Shih MS, Shepard A, Jin D, Dattatreyamurty B, Jones W, Dorai H, Ryan S, Griffiths D, Maliakal J, Jelic M, Pastorcic M, Stavljenic A, Sampath TK: Osteogenic protein-1 (bone morphogenetic protein-7) reduces severity of injury after ischemic acute renal failure in rat. J Clin Invest 102: 202–214, 1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Simon M, Maresh JG, Harris SE, Hernandez JD, Arar M, Olson MS, Abboud HE: Expression of bone morphogenetic protein-7 mRNA in normal and ischemic adult rat kidney. Am J Physiol 276: F382–F389, 1999 [DOI] [PubMed] [Google Scholar]
- 37.Zeisberg M, Hanai J, Sugimoto H, Mammoto T, Charytan D, Strutz F, Kalluri R: BMP-7 counteracts TGF-beta1-induced epithelial-to-mesenchymal transition and reverses chronic renal injury. Nat Med 9: 964–968, 2003 [DOI] [PubMed] [Google Scholar]
- 38.Hruska KA, Guo G, Wozniak M, Martin D, Miller S, Liapis H, Loveday K, Klahr S, Sampath TK, Morrissey J: Osteogenic protein-1 prevents renal fibrogenesis associated with ureteral obstruction. Am J Physiol Renal Physiol 279: F130–F143, 2000 [DOI] [PubMed] [Google Scholar]
- 39.Morrissey J, Hruska K, Guo G, Wang S, Chen Q, Klahr S: Bone morphogenetic protein-7 improves renal fibrosis and accelerates the return of renal function. J Am Soc Nephrol 13[Suppl 1]: S14–S21, 2002 [PubMed] [Google Scholar]
- 40.Wang S, de Caestecker M, Kopp J, Mitu G, Lapage J, Hirschberg R: Renal bone morphogenetic protein-7 protects against diabetic nephropathy. J Am Soc Nephrol 17: 2504–2512, 2006 [DOI] [PubMed] [Google Scholar]
- 41.Gao X, Chen X, Taglienti M, Rumballe B, Little MH, Kreidberg JA: Angioblast-mesenchyme induction of early kidney development is mediated by Wt1 and Vegfa. Development 132: 5437–5449, 2005 [DOI] [PubMed] [Google Scholar]
- 42.Wilkinson DG, Nieto MA: Detection of messenger RNA by in situ hybridization to tissue sections and whole mounts. Methods Enzymol 225: 361–373, 1993 [DOI] [PubMed] [Google Scholar]
- 43.Suzuki A, Thies RS, Yamaji N, Song JJ, Wozney JM, Murakami K, Ueno N: A truncated bone morphogenetic protein receptor affects dorsal-ventral patterning in the early Xenopus embryo. Proc Natl Acad Sci U S A 91: 10255–10259, 1994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Yamaji N, Celeste AJ, Thies RS, Song JJ, Bernier SM, Goltzman D, Lyons KM, Nove J, Rosen V, Wozney JM: A mammalian serine/threonine kinase receptor specifically binds BMP-2 and BMP-4. Biochem Biophys Res Commun 205: 1944–1951, 1994 [DOI] [PubMed] [Google Scholar]
- 45.Miner JH, Li C: Defective glomerulogenesis in the absence of laminin alpha5 demonstrates a developmental role for the kidney glomerular basement membrane. Dev Biol 217: 278–289, 2000 [DOI] [PubMed] [Google Scholar]
- 46.Natoli TA, Liu J, Eremina V, Hodgens K, Li C, Hamano Y, Mundel P, Kalluri R, Miner JH, Quaggin SE, Kreidberg JA: A mutant form of the Wilms’ tumor suppressor gene WT1 observed in Denys-Drash syndrome interferes with glomerular capillary development. J Am Soc Nephrol 13: 2058–2067, 2002 [DOI] [PubMed] [Google Scholar]
- 47.Hartwig S, Hu MC, Cella C, Piscione T, Filmus J, Rosenblum ND: Glypican-3 modulates inhibitory Bmp2-Smad signaling to control renal development in vivo. Mech Dev 122: 928–938, 2005 [DOI] [PubMed] [Google Scholar]
- 48.Novak A, Guo C, Yang W, Nagy A, Lobe CG: Z/EG, a double reporter mouse line that expresses enhanced green fluorescent protein upon Cre-mediated excision. Genesis 28: 147–155, 2000 [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.










