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
. 2020 Nov 1:104-105:36-43.
doi: 10.1016/j.niox.2020.08.005. Epub 2020 Sep 4.

Myoglobin promotes nitrite-dependent mitochondrial S-nitrosation to mediate cytoprotection after hypoxia/reoxygenation

Affiliations

Myoglobin promotes nitrite-dependent mitochondrial S-nitrosation to mediate cytoprotection after hypoxia/reoxygenation

Kelly Quesnelle et al. Nitric Oxide. .

Abstract

It is well established that myoglobin supports mitochondrial respiration through the storage and transport of oxygen as well as through the scavenging of nitric oxide. However, during ischemia/reperfusion (I/R), myoglobin and mitochondria both propagate myocardial injury through the production of oxidants. Nitrite, an endogenous signaling molecule and dietary constituent, mediates potent cardioprotection after I/R and this effect relies on its interaction with both myoglobin and mitochondria. While independent mechanistic studies have demonstrated that nitrite-mediated cardioprotection requires the presence of myoglobin and the post-translational S-nitrosation of critical cysteine residues on mitochondrial complex I, it is unclear whether myoglobin directly catalyzes the S-nitrosation of complex I or whether mitochondrial-dependent nitrite reductase activity contributes to S-nitrosation. Herein, using purified myoglobin and isolated mitochondria, we characterize and directly compare the nitrite reductase activities of mitochondria and myoglobin and assess their contribution to mitochondrial S-nitrosation. We demonstrate that myoglobin is a significantly more efficient nitrite reductase than isolated mitochondria. Further, deoxygenated myoglobin catalyzes the nitrite-dependent S-nitrosation of mitochondrial proteins. This reaction is enhanced in the presence of oxidized (Fe3+) myoglobin and not significantly affected by inhibitors of mitochondrial respiration. Using a Chinese Hamster Ovary cell model stably transfected with human myoglobin, we show that both myoglobin and mitochondrial complex I expression are required for nitrite-dependent attenuation of cell death after anoxia/reoxygenation. These data expand the understanding of myoglobin's role both as a nitrite reductase to a mediator of S-nitrosation and as a regulator of mitochondrial function, and have implications for nitrite-mediated cardioprotection after I/R.

Keywords: Complex I; Ischemia; Mitochondria; Myoglobin; Nitrite; S-nitrosothiol.

PubMed Disclaimer

Figures

Figure 1:
Figure 1:. Myoglobin is a more efficient nitrite reductase than mitochondria that promotes SNO formation.
(A) The rate of NO generated in the headspace of the reaction of anoxic mitochondria (0–20mg/ml) with nitrite (1mM). (B) NO generated in the headspace of the reaction of ferrous (closed dot), 25% oxidized (triangles) or 100% oxidized (squares) myoglobin (0–100μM) with nitrite (1mM). (C) The rate of NO generation by isolated mitochondria (2mg/ml) or ferrous myoglobin (25 μM) in the presence of nitrite (0–1mM) in anoxia. (D) Total mitochondrial SNO generated after 10 minutes of incubation of nitrite (0–100 μM) with mitochondria alone (2mg/ml; open squares) or mitochondria with ferrous myoglobin (25 μM; closed circles) in anoxia. (E) The concentration of mitochondrial SNO generated after incubation of 100% ferrous (closed bars) or 25% metmyoglobin (open bars) with nitrite (25 μM). *p<0.01 and #p<0.05 versus mitochondria alone. *p<0.01 and #p<0.05 versus ferrous myoglobin for panels C and F. Data are means ± SEM. n=5 for all panels.
Figure 2:
Figure 2:. Myoglobin enhances cellular NO generation and S-nitrosation.
(A) Representative Western blot demonstrating myoglobin expression in CHO-Mb cells and lack of expression in CHO-R cells. Western blot shows MYC tag on myoglobin and α-tubulin control in each cell type. (B) The difference spectra between CHO-R and CHO-Mb cells. (C) Representative chemiluminescence trace showing NO generation by CHO-R (gray) and CHO-Mb (black) cells. (D) Quantitation of NO generation rate from traces like those shown in (C) of untreated (control) CHO-R (open bars) and CHO-Mb (filled bars) cells and those treated with potassium ferricyanide (100μM) or potassium cyanide (25μM) and Myxathiazol (20μM). (E) S-nitrosothiol concentration measured in the whole CHO-R or CHO-Mb cell or mitochondria isolated from those cells after anoxic treatment with nitrite (25μM) for 10 min. *p<0.01. Data are means ± SEM. n=5 for panels D and E.
Figure 3:
Figure 3:. Myoglobin expression enhances nitrite-dependent attenuation of cell death after hypoxia/reoxygenation.
(A) Cell death (percent of lactate dehydrogenase release) in CHO-R (open circles) and CHO-Mb (filled squares) cells after hypoxia/reoxygenation with nitrite treatment (0–100μM). (B) Mitochondrial complex I activity in CHO-R (open bars) and CHO-Mb (filled bars) cells in normoxia (control), after nitrite (25μM) treatment in normoxia (Nitrite), after hypoxia/reoxygenation (H/R), or after hypoxia/reoxygenation in the presence of nitrite (25μM; H/R + Nitrite). (C) Hydrogen peroxide production by CHO-R (open bars) and CHO-Mb (filled bars) cells in normoxia (Control), after hypoxia/reoxygenation (H/R), or after hypoxia/reoxygenation in the presence of nitrite (25μM; H/R + Nitrite). (D) S-nitrosothiol levels of mitochondria isolated from CHO-R and CHO-Mb cells in normoxia (control) after hypoxia/reoxygenation (H/R), or hypoxia/reoxygenation in the presence of nitrite either 10 minutes (10m) or 2 hours (2h) after reoxygenation. *p<0.01; #p<0.05. Data are means ± SEM. n=5 for all panels.
Figure 4:
Figure 4:. Complex I is required for nitrite and myoglobin dependent protection from hypoxia/reoxygenation.
(A) Representative Western blot and quantification of complex I assembly factor NDUFAF1 protein expression following 72h of incubation with scrambled (scrm) or siRNA targeted to NDUFAF1. (B) Complex 1 activity (rate of rotenone-sensitive NADH consumption) following NDUFAF1 knockdown, normalized as a percentage of cells treated with the scramble control (scrm). (C) NO production by CHO-R and CHO-Mb cells treated with scrambled (open bar) or siRNA to NDUFAF1 in the presence of nitrite (1mM) in anoxia. (D) Quantification of cell death (LDH release) as a percent of normoxic cells in CHO-Mb with NDUFAF1 (scrm) or after siRNA to NDUFAF1 (siRNA) after H/R in the presence or absence (ctrl) of 20uM nitrite. *p<0.01; #p<0.05. Data are means ± SEM. n>3 for all panels.
Figure 5:
Figure 5:. The reaction between nitrite and myoglobin S-nitrosates mitochondrial complex I to mediate cytoprotection.
During hypoxia, nitrite in the cell is reduced to NO to some extent by mitochondrial complex IV (gray dotted arrow), but with greater efficiency by myoglobin (black arrow). This reaction between nitrite and myoglobin results in the S-nitrosation (SNO) of mitochondrial complex I, which inhibits complex I activity and results in decreased mitochondrial ROS production, leading to cytoprotection of cardiomyocytes.

Similar articles

Cited by

References

    1. Hendgen-Cotta UB, Esfeld S, Coman C, Ahrends R, Klein-Hitpass L, Flogel U, Rassaf T, Totzeck M, A novel physiological role for cardiac myoglobin in lipid metabolism, Sci Rep 7 (2017) 43219. - PMC - PubMed
    1. Brunori M, Nitric oxide, cytochrome-c oxidase and myoglobin, Trends Biochem Sci 26 (2001) 21–3. - PubMed
    1. Kamga C, Krishnamurthy S, Shiva S, Myoglobin and mitochondria: a relationship bound by oxygen and nitric oxide, Nitric Oxide 26 (2012) 251–8. - PMC - PubMed
    1. Wittenberg JB, Wittenberg BA, Myoglobin-enhanced oxygen delivery to isolated cardiac mitochondria, J Exp Biol 210 (2007) 2082–90. - PubMed
    1. Garry DJ, Kanatous SB, Mammen PP, Emerging roles for myoglobin in the heart, Trends Cardiovasc Med 13 (2003) 111–6. - PubMed

Publication types

LinkOut - more resources

-