Kidney Fibrosis: Lock out SIRT4

The accumulation of SIRT4 in the nuclei of kidney cells drives kidney fibrosis, so blocking the movement of this protein could be a potential therapeutic strategy against fibrosis.
  1. Kaiqiang Zhao
  2. Zhongjun Zhou  Is a corresponding author
  1. School of Biomedical Sciences, University of Hong Kong, Hong Kong

Chronic kidney disease is estimated to affect over 10% of the global population (Kovesdy, 2022). Its most common feature is kidney fibrosis, which is characterized by the accumulation of extracellular matrix proteins in tissues, causing structural damage that can impair the function of the kidneys (Huang et al., 2023).

Previous work revealed that complex mechanisms and protein mediators drive kidney fibrosis, including the protein TGF-β1, which plays a vital role in promoting the expression of pro-fibrotic genes. TGF-β1 acts through a well-established signaling pathway that eventually activates a set of three transcription factors – Smad2, Smad3 and Smad4 – which collectively move from the cytoplasm to the nucleus where they promote the transcription of pro-fibrotic genes (Meng et al., 2016). The transport of molecules from the cytoplasm to the nucleus occurs continuously in cells throughout the body and must be tightly controlled to prevent aberrant traffic that can drive disease.

However, directly inhibiting TGF-β1 signaling to prevent kidney fibrosis is not an ideal therapeutic strategy due to the potential adverse effects of disrupting other biological processes involving TGF-β1. Therefore, a greater understanding of the mechanisms underlying TGF-β1-induced fibrosis is required to identify alternative approaches. Now, in eLife, Lin Kang, Zhen Liang, Shu Yang and colleagues – including Guangyan Yang, Jiaqing Xiang and Xiaoxiao Yang as joint first authors – report how unusual TGF-β1-driven translocation of the protein Sirtuin 4 into the nucleus contributes to the progression of kidney fibrosis (Yang et al., 2024).

Sirtuin 4 (also known as SIRT4 for short) is normally located in the mitochondria. However, Yang et al. (who are based at the First Affiliated Hospital of Southern University of Science and Technology and Hefei University of Technology) found increased amounts of SIRT4 in the nuclei of kidney cells from patients with chronic kidney disease. Similarly, they also discovered elevated levels of nuclear SIRT4 in the kidneys of mice that had been injured during surgery to induce kidney fibrosis experimentally.

Intriguingly, Yang et al. found that deleting the gene for SIRT4 significantly reduced the extent of kidney fibrosis in mice following an injury; this occurred either globally throughout the body or specifically in the epithelial cells lining tubules in the kidney. Consistent with this finding, overexpressing the gene in the same cells markedly enhanced the extent of kidney fibrosis. Taken together, these results suggest that abnormal nuclear accumulation of SIRT4 contributes to kidney fibrosis.

To investigate the mechanism underlying SIRT4-mediated fibrosis, Yang et al. carried out experiments in human kidney tubule epithelial cells to identify which proteins interact with SIRT4 upon TGF-β1 treatment. Interestingly, SIRT4 interacted with several proteins that localized in the nucleus, including U2AF2 (short for U2 small nuclear RNA auxiliary factor 2). This protein plays an important role in assembling the molecular machinery known as a spliceosome, which removes unnecessary parts of pre-mRNA to form the mature mRNA that is translated into a protein (Zorio and Blumenthal, 1999; Agrawal et al., 2016).

Yang et al. found that TGF-β1 treatment induced SIRT4 to remove acetyl groups from U2AF2. This allowed U2AF2 to splice the pre-mRNA of a pro-fibrotic protein called CCN2 (short for cell communication network 2), causing expression of this protein to increase. Together, these findings identify the SIRT4-U2AF2-CCN2 axis as a novel mechanism of TGF-β1-induced kidney fibrosis (Figure 1).

Nuclear import of SIRT4 regulates the progression of kidney fibrosis.

TGF-β1 increases the expression of genes that promote kidney fibrosis in epithelial cells lining kidney tubules (black outline). First, TGF-β1 triggers the protein SIRT4 (green) to translocate from the mitochondria (top right) to the cytoplasm through BAX/BAK pores (blue rectangles). Simultaneously, TGF-β1 also activates ERK1/2 (blue oval), which phosphorylate SIRT4 once it is in the cytosol (represented by the addition of an orange circle containing the letter P). Phosphorylated SIRT4 then binds to importin α1 (orange oval) and is transported into the nucleus (bottom) where it removes acetyl groups (orange circle containing the letters Ac) from the protein U2AF2 (grey oval). This allows U2AF2 to splice the pre-mRNA of the pro-fibrotic protein CCN2, resulting in more mature mRNA that can be translated into CCN2 and promote the progression of kidney fibrosis. BAX: B-cell lymphoma 2-associated X protein; BAK: B-cell lymphoma 2 antagonist killer 1 pore; ERK1/2: extracellular signal-regulated kinase 1 and 2; U2AF2: U2 small nuclear RNA auxiliary factor 2; CCN2: cell communication network 2.

Yang et al. next set out to explore how SIRT4 translocates from the mitochondria to the nucleus upon TGF-β1 treatment. The experiments showed that TGF-β1-induced release of SIRT4 from mitochondria occurs through pores formed by protein polymers known as BAX and BAK, which are located in the outer membrane of mitochondria (Cosentino and García-Sáez, 2017). Screening using a variety of protein inhibitors demonstrated that in order to translocate, SIRT4 must be phosphorylated by enzymes known as ERK1 and ERK2. This modification allows SIRT4 to bind to a nuclear-transport receptor called importin α1, which moves the protein into the nucleus. Finally, Yang et al. treated mice with anti-SIRT4 antibodies that only inhibit expression of SIRT4 in the nucleus, and found this targeted disruption could sufficiently alleviate surgery-induced kidney fibrosis.

These findings open an exciting avenue for the development of a novel therapeutic strategy against kidney fibrosis: to lock the ‘bad guy’ – SIRT4 – out of the nuclei of kidney cells. It is conceivable that in the future, small molecules that hinder SIRT4 from interacting with ERK1/2 or importin α1 could be screened or synthesized to treat kidney fibrosis. However, nuclear SIRT4 may play beneficial roles in other cells and tissues (Zeng et al., 2018), thus further clinical studies are needed to evaluate the safety of such a strategy.

References

Article and author information

Author details

  1. Kaiqiang Zhao

    Kaiqiang Zhao is in the School of Biomedical Sciences, University of Hong Kong, Hong Kong, Hong Kong

    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-3680-2120
  2. Zhongjun Zhou

    Zhongjun Zhou is in the School of Biomedical Sciences, University of Hong Kong, Hong Kong, Hong Kong

    For correspondence
    zhongjun@hku.hk
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-7092-8128

Publication history

  1. Version of Record published:

Copyright

© 2024, Zhao and Zhou

This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 197
    views
  • 23
    downloads
  • 0
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Kaiqiang Zhao
  2. Zhongjun Zhou
(2024)
Kidney Fibrosis: Lock out SIRT4
eLife 13:e102355.
https://doi.org/10.7554/eLife.102355

Further reading

    1. Cell Biology
    Li Sun, Xuejin Chen ... Quan-wen Jin
    Research Article

    Mitotic anaphase onset is a key cellular process tightly regulated by multiple kinases. The involvement of mitogen-activated protein kinases (MAPKs) in this process has been established in Xenopus egg extracts. However, the detailed regulatory cascade remains elusive, and it is also unknown whether the MAPK-dependent mitotic regulation is evolutionarily conserved in the single-cell eukaryotic organisms such as fission yeast (Schizosaccharomyces pombe). Here, we show that two MAPKs in S. pombe indeed act in concert to restrain anaphase-promoting complex/cyclosome (APC/C) activity upon activation of the spindle assembly checkpoint (SAC). One MAPK, Pmk1, binds to and phosphorylates Slp1Cdc20, the co-activator of APC/C. Phosphorylation of Slp1Cdc20 by Pmk1, but not by Cdk1, promotes its subsequent ubiquitylation and degradation. Intriguingly, Pmk1-mediated phosphorylation event is also required to sustain SAC under environmental stress. Thus, our study establishes a new underlying molecular mechanism of negative regulation of APC/C by MAPK upon stress stimuli, and provides a previously unappreciated framework for regulation of anaphase entry in eukaryotic cells.

    1. Cancer Biology
    2. Cell Biology
    Alexandra Urbancokova, Terezie Hornofova ... Pavla Vasicova
    Research Article

    PML, a multifunctional protein, is crucial for forming PML-nuclear bodies involved in stress responses. Under specific conditions, PML associates with nucleolar caps formed after RNA polymerase I (RNAPI) inhibition, leading to PML-nucleolar associations (PNAs). This study investigates PNAs-inducing stimuli by exposing cells to various genotoxic stresses. We found that the most potent inducers of PNAs introduced topological stress and inhibited RNAPI. Doxorubicin, the most effective compound, induced double-strand breaks (DSBs) in the rDNA locus. PNAs co-localized with damaged rDNA, segregating it from active nucleoli. Cleaving the rDNA locus with I-PpoI confirmed rDNA damage as a genuine stimulus for PNAs. Inhibition of ATM, ATR kinases, and RAD51 reduced I-PpoI-induced PNAs, highlighting the importance of ATM/ATR-dependent nucleolar cap formation and homologous recombination (HR) in their triggering. I-PpoI-induced PNAs co-localized with rDNA DSBs positive for RPA32-pS33 but deficient in RAD51, indicating resected DNA unable to complete HR repair. Our findings suggest that PNAs form in response to persistent rDNA damage within the nucleolar cap, highlighting the interplay between PML/PNAs and rDNA alterations due to topological stress, RNAPI inhibition, and rDNA DSBs destined for HR. Cells with persistent PNAs undergo senescence, suggesting PNAs help avoid rDNA instability, with implications for tumorigenesis and aging.