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 24;9(1):127.
doi: 10.1038/s41392-024-01831-2.

DEAD-box helicase 17 (DDX17) protects cardiac function by promoting mitochondrial homeostasis in heart failure

Affiliations

DEAD-box helicase 17 (DDX17) protects cardiac function by promoting mitochondrial homeostasis in heart failure

Mingjing Yan et al. Signal Transduct Target Ther. .

Abstract

DEAD-box helicase 17 (DDX17) is a typical member of the DEAD-box family with transcriptional cofactor activity. Although DDX17 is abundantly expressed in the myocardium, its role in heart is not fully understood. We generated cardiomyocyte-specific Ddx17-knockout mice (Ddx17-cKO), cardiomyocyte-specific Ddx17 transgenic mice (Ddx17-Tg), and various models of cardiomyocyte injury and heart failure (HF). DDX17 is downregulated in the myocardium of mouse models of heart failure and cardiomyocyte injury. Cardiomyocyte-specific knockout of Ddx17 promotes autophagic flux blockage and cardiomyocyte apoptosis, leading to progressive cardiac dysfunction, maladaptive remodeling and progression to heart failure. Restoration of DDX17 expression in cardiomyocytes protects cardiac function under pathological conditions. Further studies showed that DDX17 can bind to the transcriptional repressor B-cell lymphoma 6 (BCL6) and inhibit the expression of dynamin-related protein 1 (DRP1). When DDX17 expression is reduced, transcriptional repression of BCL6 is attenuated, leading to increased DRP1 expression and mitochondrial fission, which in turn leads to impaired mitochondrial homeostasis and heart failure. We also investigated the correlation of DDX17 expression with cardiac function and DRP1 expression in myocardial biopsy samples from patients with heart failure. These findings suggest that DDX17 protects cardiac function by promoting mitochondrial homeostasis through the BCL6-DRP1 pathway in heart failure.

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Heart failure and cardiomyocyte injury caused by various pathological factors lead to decreased DDX17 expression. a, b The average data of mouse cardiac function as shown by the left ventricular EF and FS of the sham and TAC-induced chronic heart failure mice (n = 6). ce Ddx17 mRNA and protein expression in the myocardium from the sham or TAC-induced chronic heart failure mouse models (n = 6). f Representative images of hearts showing the cardiac morphology of the control (Con) and Dox-treated (Dox) mice (n = 6); scale bar, 2 mm. g Representative images of H&E-stained heart sections from the Con and Dox-treated mice (n = 6); scale bar, 2 mm. h, i Mouse cardiac function as shown by the left ventricular EF and FS of Con and Dox-treated mice (n = 6). jl Ddx17 mRNA (n = 6) and protein (n = 6) expression in Con and Dox-treated mouse hearts. m, n DDX17 expression in NMVMs treated with different injury factors: serum-free medium (SF), hypoxia for 16 h (HP), 2 μg/mL TNF-α, 100 μM H2O2, and 0.5 μM Dox (n = 3). o, p DDX17 protein expression in NMVMs treated with normoxia or hypoxia for 16 h (n = 5). q, r DDX17 protein levels and the average data of NMVMs treated with different concentrations of H2O2 for 24 h (n = 3). s, t DDX17 protein levels and the average data of NMVMs treated with 0.1, 0.5, and 1 μM Dox for 24 h (n = 3). *P < 0.05, **P < 0.01, and ***P < 0.001
Fig. 2
Fig. 2
Cardiomyocyte-specific Ddx17 knockout leads to reduced cardiac function and exacerbates Dox-induced heart failure in mice. a, b Western blot and the average data of DDX17 in isolated NMVMs of control (Con) and Ddx17-cKO mice (n = 6). c Western blot of DDX17 levels in heart, skeletal muscle, liver and kidney of Con and Ddx17-cKO mice (n = 3). d Mice were injected intraperitoneally with saline or 7.5 mg/kg Dox 3 times every other day. Survival curve of the mice in the four groups (n = 18). e Body weight of mice in the control + saline (Con + Saline), Ddx17-cKO + saline (Ddx17-cKO + Saline), control + doxorubicin (Con + Dox) and Ddx17-cKO + doxorubicin (Ddx17-cKO + Dox) groups (n = 5). f Representative images of the hearts from the mice in Con + Saline, Ddx17-cKO + Saline, Con + Dox, and Ddx17-cKO + Dox groups, the scale bar represents 2 mm. g Representative H&E-stained heart sections from mice in the four groups (n = 5); scale bar, 50 μm. h, i Representative images of Sirius Red-stained hearts from mice in the four groups and semiquantitative analysis of the fibrosis area ratio (n = 5); scale bar, 200 μm. jl Representative images of echocardiography of mouse hearts and the average data of cardiac function of left ventricular EF (k) and FS (l) in the four groups; n = 10 for each group. m TUNEL staining quantification results of myocardial tissue in the four groups; n = 5 for each group. n, o Mouse serum LDH (n) and CK-MB (o) levels in the four groups (n = 6). *P < 0.05, **P < 0.01, and ***P < 0.001
Fig. 3
Fig. 3
Overexpression of Ddx17 in cardiomyocytes attenuates myocardial injury and improves cardiac function under pathological conditions. a Western blot of DDX17 protein expression from control (Con) and two cardiac-specific Ddx17-overexpressing mouse lines (Ddx17-Tg-H and Ddx17-Tg) (n = 5). b DDX17 expression in heart, skeletal muscle, liver and kidney of control (Con) and Ddx17-Tg mice (n = 3). c Doxorubicin (7.5 mg/kg) or an equivalent volume of saline was administered to mice by intraperitoneal injection every other day for a total of three injections in the control (Con) and Ddx17-Tg mice. Survival curves of mice in the control + saline (Con + Saline), Ddx17-transgene + saline (Ddx17-Tg + Saline), control + doxorubicin (Con + Dox), and Ddx17-transgene + doxorubicin (Ddx17-Tg + Dox) groups (n = 18). d Body weight of the mice in the Con + Saline, Ddx17-Tg + Saline, Con + Dox, and Ddx17-Tg + Dox groups (n = 6). eg Representative images of echocardiography of mouse heart and the average data of cardiac function of left ventricle EF (f) and FS (g) in the four groups (n = 11). h, i Representative images of heart morphologies and H&E-stained heart sections in the four groups (n = 6). jk Representative images of Sirius Red-stained mouse heart sections and quantification of myocardial fibrosis area ratio in the four groups (n = 5); scale bar, 200 μm. l TUNEL staining of myocardial tissue in Con + Saline, Ddx17-Tg + Saline, Con + Dox, and Ddx17-Tg + Dox, (n = 5); scale bar, 50 μm. m Quantification of TUNEL staining of myocardial tissue in the four groups (n = 5). *P < 0.05, **P < 0.01, and ***P < 0.001
Fig. 4
Fig. 4
DDX17 plays an important role in maintaining mitochondrial morphology and function in cardiomyocytes. a Transmission electron microscopy (TEM) of LVs from control (Con) and Ddx17-cardiomyocyte-specific knockout (cKO) mice (scale bars: low-1 μm, medium-500 nm, high-200 nm) (n = 5). b, c Statistical analysis of mitochondrial length and mitochondrial area in control (Con) and Ddx17-cKO mice (n = 48). d, e Mitochondrial membrane potential (ΔΨm) analyzed by JC-1 red/green fluorescence intensity in control (Con) and Ddx17-overexpressing (Ddx17-OE) HL-1 cells treated with normoxic and hypoxic conditions (n = 3); scale bar: 50 μm. f, g Mitochondrial permeability transition pore (mPTP) analyzed by calcein-AM fluorescence intensity in control (Con) and Ddx17-overexpressing (Ddx17-OE) HL-1 cells (n = 4); scale bar: 90 μm. h, i NMVMs were infected with Ddx17-overexpressing adenovirus (Ddx17-OE) or its control (Con) for 24 h and then treated with PBS or Dox for 24 h. Mitochondria of cardiomyocytes were stained with Mito-Tracker Red and nuclei were stained with DAPI, and the rate of mitochondrial fission was analyzed by confocal microscopy (n = 4); scale bar corresponds to 20 μm. j Cellular ATP concentration in the NMVMs of control (Con) and Ddx17-cKO mice treated with PBS or Dox (n = 4). k Cellular ATP concentration in the NMVMs of control (Con) and Ddx17-Tg mice treated with PBS or Dox (n = 4). l, m 8-OHdG and MDA levels in NMVMs from each group (n = 3). nr HL-1 cardiomyocytes were infected with Ddx17 overexpressing adenovirus (Ddx17-OE) or its control (Con) for 24 h and then treated with PBS or 0.5 μM Dox for 24 h. Based on the measured mitochondrial OCR of HL-l cells in response to 1 μM oligomycin, 1 μM FCCP and 0.5 μM rotenone/antimycin A, the basal respiration, maximal respiration, ATP production and spare respiratory capacity were measured using a Seahorse flux analyser (n = 4). s Western blot of DRP1, MFN1 and MFN2 in the left ventricle of mice in the control + saline (Con + Saline), Ddx17-cKO + saline (Ddx17-cKO + Saline), control + doxorubicin (Con + Dox) and Ddx17-cKO + doxorubicin (Ddx17-cKO + Dox) groups (n = 6). t Western blot of DRP1, MFN1 and MFN2 in LVs from mice in the control + saline (Con + Saline), Ddx17-transgene + saline (Ddx17-Tg + Saline), control + doxorubicin (Con + Dox) and Ddx17-transgene+doxorubicin (Ddx17-Tg + Dox) groups (n = 6). *P < 0.05, **P < 0.01, and ***P < 0.001
Fig. 5
Fig. 5
DDX17 coordinates with BCL6 in the transcriptional repression of the Drp1 gene in cardiomyocytes. a, b GO terms responding to promoters with higher (a) and lower (b) methylation levels in NMVMs from Ddx17-Tg mice compared to controls (Con) (n = 3). Representative genes are indicated below. c Motif enrichment analysis of proximal NDRs in Ddx17-Tg and control (Con) NMVMs (n = 3). d, e Co-IP experiments were performed with DDX17 (d) and BCL6 (e) antibodies to analyze the interaction of DDX17 and BCL6 in control (Con) and Ddx17-cKO NMVMs (n = 4). f Immunofluorescence staining of DDX17 (red), BCL6 (green) and nuclei (DAPI, blue) in cultured wild-type NMVMs; scale bar, 20 μm (n = 3). g To investigate the regulatory effects of DDX17 and BCL6 overexpression on Drp1 promoter activity, HEK293A cells were transfected with Ddx17 (pcDNA-Ddx17) and/or Bcl6 (pcDNA-Bcl6) expression plasmids and simultaneously cotransfected with the Drp1 0.8-kb wild-type promoter (pGL3-Drp1-WT) luciferase reporter plasmid, and Drp1 promoter activity was analyzed by luciferase assay (n = 3). h HEK293A cells were transfected with Ddx17 (pcDNA-Ddx17) and/or Bcl6 (pcDNA-Bcl6) expression plasmids and simultaneously cotransfected with the 0.8 kb Drp1 promoter mutation plasmid (pGL3-Drp1-MUT) with mutated BCL6 binding sites using the pGL3-basic plasmid (n = 3). i HEK293A cells were cotransfected with the Drp1 promoter plasmid (pGL3-Drp1-WT) and different concentrations of the Bcl6 expression plasmid (pcDNA-Bcl6) to detect Drp1 promoter activity (n = 3). j ChIP analysis of NMVMs revealed the recruitment of BCL6 to regions containing BCL6 binding sites within the promoter region of Drp1 by quantitative real-time PCR (n = 3). kn HL-1 cells were transfected with Ddx17 siRNA and/or Drp1 siRNA for 24 h and then treated with doxorubicin for 24 h. Expression of DDX17, DRP1 and c-CASP-3 was analyzed by western blot, and GAPDH was used as a protein loading control (n = 3). In all co-transfection experiments, pcDNA3.1 was used as the equilibrium plasmid in the different transfection mixtures to balance the total amount of DNA, and NCi was used as the equilibrium RNA in the different transfection mixtures to balance the total amount of RNA (n = 3). oq HL-1 cardiomyocytes were transfected with NCi (Con) or Ddx17 siRNA (Ddx17-KD) for 24 h and then treated with PBS or 0.5 μM Dox for 24 h. Cardiomyocyte mitochondria and cytoplasm were isolated and the expression of mitochondrial DRP1 (Mito-DRP1) and cytoplasmic DRP1 (Cyto-DRP1) was analyzed by western blot. COX IV and β-tubulin were used as protein loading controls for mitochondria and cytoplasm, respectively (n = 4). r HL-1 cells were transfected with NCi (Con) and Ddx17 siRNA (Ddx17-KD) for 24 h, then treated with PBS or 0.5 μM Dox for 24 h and divided into Con, Ddx17-KD, Con + Dox and Ddx17-KD + Dox groups. Cardiomyocyte mitochondria and cytoplasm were extracted separately using a mitochondrial isolation kit. Cytochrome c levels in mitochondria and cytoplasm were analyzed by western blot. COX IV was used as a protein loading control for mitochondria and β-tubulin as a protein loading control for cytoplasmic proteins (n = 4). *P < 0.05, **P < 0.01, and ***P < 0.001
Fig. 6
Fig. 6
DDX17 expression levels are positively correlated with cardiac function in patients with different stages of heart failure. a H&E staining of myocardial tissue from the control (Con) and heart failure (HF) patients (n = 3); scale bar, 100 μm. b Wheat germ agglutinin staining of tissue samples isolated from Con and HF patients (n = 3); scale bar, 100 μm. c Transmission electron microscopy images of myocardial tissue from Con and HF patients (n = 3); scale bar, 2 μm. d Correlation of the DDX17 mRNA levels with left ventricular EF in myocardial tissue from heart failure patients. e Correlation of DRP1 mRNA levels with left ventricular EF in myocardial tissue from heart failure patients. f Correlation of DDX17 with DRP1 mRNA levels in the myocardium of heart failure patients. g Graphic summary of DDX17 protecting cardiac function by promoting mitochondrial homeostasis through the BCL6-DRP1 pathway in heart failure

Similar articles

Cited by

References

    1. Truby LK, Rogers JG. Advanced heart failure: epidemiology, diagnosis, and therapeutic approaches. JACC Heart Fail. 2020;8:523–536. doi: 10.1016/j.jchf.2020.01.014. - DOI - PubMed
    1. Guo J, et al. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther. 2022;7:391. doi: 10.1038/s41392-022-01251-0. - DOI - PMC - PubMed
    1. Xu K, et al. DDX5 and DDX17-multifaceted proteins in the regulation of tumorigenesis and tumor progression. Front Oncol. 2022;12:943032. doi: 10.3389/fonc.2022.943032. - DOI - PMC - PubMed
    1. Fuller-Pace FV. DEAD box RNA helicase functions in cancer. RNA Biol. 2013;10:121–132. doi: 10.4161/rna.23312. - DOI - PMC - PubMed
    1. Wu KJ. The role of miRNA biogenesis and DDX17 in tumorigenesis and cancer stemness. Biomed. J. 2020;43:107–114. doi: 10.1016/j.bj.2020.03.001. - DOI - PMC - PubMed

MeSH terms