Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2024 May 7;134(13):e165836.
doi: 10.1172/JCI165836.

Fibroblast expression of transmembrane protein smoothened governs microenvironment characteristics after acute kidney injury

Affiliations

Fibroblast expression of transmembrane protein smoothened governs microenvironment characteristics after acute kidney injury

Yuan Gui et al. J Clin Invest. .

Abstract

The smoothened (Smo) receptor facilitates hedgehog signaling between kidney fibroblasts and tubules during acute kidney injury (AKI). Tubule-derived hedgehog is protective in AKI, but the role of fibroblast-selective Smo is unclear. Here, we report that Smo-specific ablation in fibroblasts reduced tubular cell apoptosis and inflammation, enhanced perivascular mesenchymal cell activities, and preserved kidney function after AKI. Global proteomics of these kidneys identified extracellular matrix proteins, and nidogen-1 glycoprotein in particular, as key response markers to AKI. Intriguingly, Smo was bound to nidogen-1 in cells, suggesting that loss of Smo could affect nidogen-1 accessibility. Phosphoproteomics revealed that the 'AKI protector' Wnt signaling pathway was activated in these kidneys. Mechanistically, nidogen-1 interacted with integrin β1 to induce Wnt in tubules to mitigate AKI. Altogether, our results support that fibroblast-selective Smo dictates AKI fate through cell-matrix interactions, including nidogen-1, and offers a robust resource and path to further dissect AKI pathogenesis.

Keywords: Extracellular matrix; Nephrology; Proteomics.

PubMed Disclaimer

Conflict of interest statement

Conflict of interest: The authors have declared that no conflict of interest exists.

Figures

Figure 1
Figure 1. Fibroblast-specific ablation of Smo mitigates ischemic AKI.
(A) Generation of Smo fibroblast-specific deletion mice. (B) Quantitative real-time PCR (qPCR) analysis (n = 4) and Western blot assay showing Smo levels in primary fibroblasts isolated from Gli1-Smo+/+ and Gli1-Smo–/– or Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys. (C) The mouse baseline body weight (BW), kidney-to-body weight (KW/BW) ratios, serum creatinine (Scr), and (D) blood pressure levels (n = 6). At 1 day after IRI, (E) Scr levels in Gli1-Smo+/+ and Gli1-Smo–/– mice (n = 16-17). (F) Periodic Acid–Schiff (PAS) staining showing kidney morphological changes in Gli1-Smo+/+ and Gli1-Smo–/– mice. Blue asterisks indicate injured tubules. Representative micrographs of TUNEL or IHC staining against CD45 and CD3 in Gli1-Smo+/+ and Gli1-Smo–/– kidneys. Scale bar: 25 μm. (G) Western blots assay of NGAL, FasL, and Bad in Gli1-Smo+/+ and Gli1-Smo–/– kidneys (Sham, n = 3; IRI, n = 5). (H) qPCR analysis showing Tnf-α, Mcp1, and Rantes mRNA levels in Gli1-Smo+/+ and Gli1-Smo–/– kidneys (Sham, n = 3; IRI, n = 6). (I) Scr levels in Pdgfr-Smo+/+ and Pdgfr-Smo–/– mice (n = 15-16). (J) Western blots showing NGAL, FasL, and Bad levels in Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys. (K) qPCR analysis showing Tnf-α, Mcp1, and Rantes mRNA levels in Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys (Sham, n = 3; IRI, n = 6). (L) Representative micrographs of PAS, TUNEL, and IHC staining against CD45 in Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys. Scale bar: 25 μm. Arrows indicate positive cells. DAPI is a nuclear counterstain. For all Western blot panels, numbers indicate individual animals in a given group. †P < 0.05 versus sham control, *P < 0.05 versus Gli1-Smo+/+ or Pdgfr-Smo+/+ IRI mice. Dots indicate individual animals in a given group. Graphs are presented as means ± SEM. Differences among groups were analyzed using unpaired t tests or 1-way ANOVA followed by the Student-Newman-Keuls test.
Figure 2
Figure 2. Fibroblast-specific ablation of Smo promotes perivascular mesenchymal cell activation and proliferation after AKI.
At 1 day after IRI, (A) qPCR analyses of Fsp1, vimentin, and αSma mRNA in Gli1-Smo+/+ and Gli1-Smo–/– kidneys (Sham, n = 3; IRI, n = 7-12). (B) qPCR analyses of Fsp1, vimentin, and αSma mRNA in Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys (Sham, n = 3; IRI, n = 6). (C) Western blot assay of Pdgfr-β, vimentin, αSMA, and PCNA proteins in Gli1-Smo+/+ and Gli1-Smo–/– kidneys. (D) Western blots assay of vimentin, αSMA, and PCNA proteins in Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys. (E) Representative micrographs of Pdgfr-β, vimentin, and Ki67 expression in Gli1-Smo+/+ and Gli1-Smo–/– kidneys. Arrows indicate positive cells. Scale bar: 25 μm. (F) Representative micrographs of vimentin and PCNA expression in Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys. Scale bar: 25 μm. Arrows indicate positive cells. DAPI is a nuclear counterstain. For all Western blot panels, numbers indicate individual animals in a given group. †P < 0.05 versus sham control, *P < 0.05 versus Gli1-Smo+/+ or Pdgfr-Smo+/+ IRI mice. Dots indicate individual animals in a given group. Graphs are presented as means ± SEM. Differences among groups were analyzed using 1-way ANOVA, followed by the Student-Newman-Keuls test.
Figure 3
Figure 3. Global proteomics identifies NID1 as a prominent matrix protein in Gli1+ fibroblast-specific Smo deletion kidneys after AKI.
(A) Experimental workflow of the global proteomic analysis. 6 mice in each group were used for mass spectrometry. (B) Principal component analysis of global proteomes from Gli1-Smo+/+ and Gli1-Smo–/– kidneys 1 day after ischemic AKI. (C) Heatmap of ANOVA-significant proteins. Label-free quantitation (LFQ) intensity of represented proteins were z-scored and plotted according to the color bar. 2 clusters of proteins with different patterns of abundance profiles are highlighted in the heatmap. (D) GO cellular compartment terms in each cluster of proteins are plotted with their names and significance. ECM proteins are boxed to indicate the protein group with the largest difference in upregulated proteins. (E) Volcano plot shows the differential proteins between Gli1-Smo+/+ and Gli1-Smo–/– kidneys. Up and down regulated proteins (FC, fold-change) are colored in red and blue, respectively. Yellow dots indicate ECM proteins. (F) Heatmap of differentially expressed ECM proteins. (G and H) Western blots of NID1 protein in Gli1-Smo+/+ and Gli1-Smo–/– or in Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys 1 day after IRI. Numbers indicate individual animals in a given group. (I) Single nucleus RNA-Seq showing NID1 mainly expressed by fibroblasts (Fib) and pericytes (Per) 12 hours after IRI. (Data were extracted from the online database provided by Benjamin Humphrey’s laboratory, http://humphreyslab.com/SingleCell/displaycharts.php) (J) IHC staining showing NID1 protein expression in Gli1-Smo+/+ and Gli1-Smo–/– or in Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys 1 day after IRI. Arrows indicate positive staining. Scale bar: 25 μm. (K) Immunoprecipitation revealing that Smo binds to NID1. NRK-49F cells under CoCl2 stress were immunoprecipitated with Smo or NID1 antibody, followed by immunoblotting with antibody against NID1 (left blot) or Smo (right blot). Differences among groups were analyzed using unpaired t tests.
Figure 4
Figure 4. Phosphoproteomics reveals Wnt pathway activation in fibroblast-specific Smo-deletion kidneys after AKI.
(A) Workflow of phosphoproteomics. (B) Principal component analysis of phosphoproteins from Gli1-Smo+/+ and Gli1-Smo–/– kidneys 1 day after ischemic AKI. (C) Volcano plot of pairwise comparisons (fold-change, FC) between the kidney phosphoproteomes of Gli1-Smo+/+ and Gli1-Smo–/– kidneys 1 day after ischemic AKI. (D) qPCR of Wnt 1, 2, 4, 5A, 5B, 7A, 7B, 9B, 10A, 10B, 11, and 16 mRNA in Gli1-Smo+/+ and Gli1-Smo–/– kidneys 1 day after ischemic AKI. *P < 0.05 (n = 7–12). (E) Western blots of β-catenin, Wnt 1, and Wnt 5A/B proteins in Gli1-Smo+/+ and Gli1-Smo–/– kidneys 1 day after ischemic AKI. Numbers indicate individual animals in a given group. (F) Western blots of β-catenin and Wnt 1 in Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys 1 day after ischemic AKI. Numbers indicate individual animals in a given group. (G) IHC staining showing β-catenin, Wnt1, Wnt 4, and Wnt5A/B in Gli1-Smo+/+ and Gli1-Smo–/– or in Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys 1 day after ischemic AKI. Scale bar: 25 μm. (H) IHC staining showing Wnt1, Wnt 4, and Wnt5A/B expression in kidney biopsy specimens from patients with AKI. Arrows indicate positive staining. Scale bar: 25 μm. Graphs are presented as means ± SEM. Differences between groups were analyzed using unpaired t tests.
Figure 5
Figure 5. Deleting Smo in fibroblasts affects integrins linked to tubular cells after AKI.
(A) Western blot assay showing the expression of integrin receptors linked to tubular cells, including integrin β1, α3, α6, αV, α8, and β6, in Gli1-Smo+/+ and Gli1-Smo–/– kidneys after ischemic AKI at 1 day. Numbers indicate individual animals in a given group. (B and C) Representative Western blot showing the expression of integrin β1 in Gli1-Smo+/+ and Gli1-Smo–/– (B) or in Pdgfr-Smo+/+ and Pdgfr-Smo–/– (C) kidneys at 1 day after ischemic AKI. Numbers indicate individual animals in a given group. Quantitative data are accordingly presented in D and E. Dots indicate individual animals in a given group (Sham, n = 3; IRI, n = 6). †P < 0.05 versus sham control, *P < 0.05 versus Gli1-Smo+/+ or Pdgfr-Smo+/+ IRI mice. (F) IHC staining showing integrin β1 in Gli1-Smo+/+ and Gli1-Smo–/– or in Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys at 1 day after AKI. Scale bar: 25 μm. (G) IHC staining showing NID1 and integrin β1 expression in kidney biopsy specimens from patients with AKI. Arrows indicate positive staining. Scale bar: 25 μm. Graphs are presented as means ± SEM. Differences among groups were analyzed using 1-way ANOVA, followed by the Student-Newman-Keuls test.
Figure 6
Figure 6. Specific deletion of Smo in fibroblasts mitigates AKI induced by cisplatin.
(A) Schematic diagram. i.p., intraperitoneal. At 3 days after cisplatin (Cis) injection, (B) Serum creatinine (Scr) levels in Gli1-Smo+/+ and Gli1-Smo–/– mice (n = 12). (C) Scr levels in Pdgfr-Smo+/+ and Pdgfr-Smo–/– mice (n = 12). (D and E) Western blots assay showing NGAL and Bax protein levels in Gli1-Smo+/+ and Gli1-Smo–/– (D) or in Pdgfr-Smo+/+ and Pdgfr-Smo–/– (E) kidneys. (F) Periodic Acid–Schiff (PAS) staining showing kidney morphological changes in Gli1-Smo+/+ and Gli1-Smo–/– mice. Blue asterisks indicate injured tubules. Representative micrographs of TUNEL assay or IHC staining against CD45 in Gli1-Smo+/+ and Gli1-Smo–/– kidneys. Scale bar: 25 μm. White arrows indicate apoptotic cells adn black arrows indicate CD45+ cells. (G) Representative micrographs of PAS staining, TUNEL assay, and IHC staining against CD45 in Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys. Scale bar: 25 μm. Blue asterisks indicate injured tubules. White and black arrows, respectively, indicate apoptotic cells and CD45+ cells. (H and I) Western blots assay of NID1, Wnt1, and integrin β1 proteins in Gli1-Smo+/+ and Gli1-Smo–/– (H) or in Pdgfr-Smo+/+ and Pdgfr-Smo–/– (I) kidneys. Quantitative data are accordingly presented in J and K (Sham, n = 3; Cis, n = 5). †P < 0.05 versus sham control, *P < 0.05 versus Gli1-Smo+/+ or Pdgfr-Smo+/+ mice. Dots indicate individual animals in a given group. (L) IHC staining showing NID1, Wnt1, and integrin β1 (Intg β1) in Gli1-Smo+/+ and Gli1-Smo–/– or in Pdgfr-Smo+/+ and Pdgfr-Smo–/– kidneys. Scale bar: 25 μm. Arrows indicate positive cells. DAPI is a nuclear counterstain. For all Western blot panels, numbers indicate individual animals in a given group. Graphs are presented as means ± SEM. Differences among groups were analyzed using 1-way ANOVA, followed by the Student-Newman-Keuls test.
Figure 7
Figure 7. NID1 promotes Wnt components expression and reduces tubular cell death in vitro.
(AD) Normal rat kidney fibroblasts (NRK-49F) were transfected with Dicer-substrate Smo siRNA (Smo Dsi) or incubated with Smo inhibitor cyclopamine (CPN, 2.5 μM), then subjected to hypoxic stress (CoCl2, 400 μM) for 24 hours. Compared to control siRNA (NC Dsi) or vehicles, western blots demonstrating that knockdown (A) or inhibition (B) of Smo induced Pdgfr-β, vimentin, αSMA, and PCNA expression, and increased NID1 (C and D). (E) Immunofluorescence staining showing Smo inhibition induced NID1 in fibroblasts under hypoxic stress. Scale bar: 25 μm. Arrows indicate positive staining. (F) Enzyme-linked immunosorbent assay revealed NID1 concentration after incubation with CPN under hypoxic stress (n = 6). (G) Schematic diagram. (H and I) Western blots demonstrating β-catenin, Wnt1, Wnt2, and Wnt5A/B levels after knockdown (H) or inhibition (I) of Smo under hypoxic stress. (J and K) Western blots showing that NID1 recombinant protein (rNID1) elevated β-catenin, Wnt1, Wnt5A/B, and Wnt16 in cultured normal rat kidney proximal tubular cells at different dosages under basal conditions (J) and hypoxic stress (K). (L, M, and O) After stimulation with staurosporine (1 μM) for 3 h, western blots assay showing reduced cleaved-caspase 3 (CCP3) in tubular cells incubated with NID1-enriched CM collected from Smo-knockdown (L) or CPN-treated (M) fibroblasts or directly treated with rNID1 (O). (N and P) Immunofluorescence staining showed fewer CCP3+ cells after treated with NID1-enriched CM (N) or NID1 recombinant protein (P). Quantitative data are presented in Q (n = 3, 4 random images were selected per slide, each dot represents the score of the according image). †P < 0.05 versus control, *P < 0.05 versus vehicle after STS. Scale bar: 25 μm. Cells were costained with DAPI. Arrows indicate positive staining. Graphs are presented as means ± SEM. Differences among groups were analyzed using 1-way ANOVA, followed by the Student-Newman-Keuls test.
Figure 8
Figure 8. NID1-enriched decellularized fibroblast matrix scaffold activates Wnt components and reduces tubular cell death ex vivo.
(A) Experimental design. In step 1, normal rat kidney fibroblasts (NRK-49F) were transfected with Dicer-substrate Smo siRNA (Smo Dsi) or cultured with Smo inhibitor CPN to enrich for NID1. In step 2, scaffolds were isolated, and, in step 3, normal rat kidney proximal tubular cells (NRK-52E) were seeded on top of scaffolds. (B) Western blots assay showing NID1 protein in decellularized fibroblast matrix scaffold. (C and D) Western blots assay showing β-catenin, Wnt2, and Wnt5A/B were activated in NRK-52E cells seeded on NID1-enriched matrix scaffold isolated from Smo-knockdown (C) or CPN-treated (D) fibroblasts under hypoxic stress. (E and F) Western blots assay showing NID1-enriched matrix scaffold isolated from Smo-knockdown (E) or CPN-treated (F) fibroblasts reduced cleaved caspase-3 (CCP3) in the seeded NRK-52E cells. (GI) Western blot assay showing conditioned medium collected from Smo-knockdown fibroblasts (G) or decellularized fibroblast matrix scaffold isolated from Smo-knockdown fibroblasts (H) or NID1 recombinant protein (rNID1) increased integrin β1 in NRK-52E cells under hypoxic stress. (J and K) Under hypoxic stress, knockdown of integrin β1 using Dicer-substrate siRNA (integrin β1 Dsi) repressed β-catenin accumulation after incubation with rNID1 in NRK-52E cells (J) and induced CCP3 (K). (L) Molecular docking analysis showing the binding sites between NID1 and Integrin β1. (M) The strategy of designing a mutant form of NID1. (N and O) Compared with the active form of NID1 human recombinant protein (25 ng/mL), Western blot assay demonstrating that mutant NID1 (25 ng/mL) failed to induce β-catenin expression in NRK-52E cells under hypoxic stress (N) and increased CCP3 after staurosporine (1 μM) stimulation (O). (P) Our model illustrates that loss of fibroblast-selective Smo promotes NID1 to interact with tubular integrin β1 and subsequently activated the Wnt signaling pathway in tubules, forming a favorable microenvironment to protect against AKI.

Similar articles

References

    1. Ronco C, et al. Acute kidney injury. Lancet. 2019;394(10212):1949–1964. doi: 10.1016/S0140-6736(19)32563-2. - DOI - PubMed
    1. Little M, Humphreys B. Regrow or repair: an update on potential regenerative therapies for the kidney. J Am Soc Nephrol. 2021;33(1):15–32. doi: 10.1681/ASN.2021081073. - DOI - PMC - PubMed
    1. Kramann R, et al. Gli1+ pericyte loss induces capillary rarefaction and proximal tubular injury. J Am Soc Nephrol. 2017;28(3):776–784. doi: 10.1681/ASN.2016030297. - DOI - PMC - PubMed
    1. Zhou D, et al. Early activation of fibroblasts is required for kidney repair and regeneration after injury. FASEB J. 2019;33(11):12576–12587. doi: 10.1096/fj.201900651RR. - DOI - PMC - PubMed
    1. Chou YH, et al. Methylation in pericytes after acute injury promotes chronic kidney disease. J Clin Invest. 2020;130(9):4845–4857. doi: 10.1172/JCI135773. - DOI - PMC - PubMed