Next Article in Journal
Mesenchymal Stem Cell Sheet Centrifuge-Assisted Layering Augments Pro-Regenerative Cytokine Production
Previous Article in Journal
H2A-H2B Histone Dimer Plasticity and Its Functional Implications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Nitric Oxide Trickle Drives Heme into Hemoglobin and Muscle Myoglobin

Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH 44195, USA
*
Author to whom correspondence should be addressed.
Cells 2022, 11(18), 2838; https://doi.org/10.3390/cells11182838
Submission received: 31 August 2022 / Accepted: 2 September 2022 / Published: 12 September 2022
(This article belongs to the Section Cell Signaling)

Abstract

:
Ever since the days of NO being proclaimed as the “molecule of the year”, the molecular effects of this miracle gas on the globins have remained elusive. While its vasodilatory role in the cardiopulmonary system and the vasculature is well recognized, the molecular underpinnings of the NO–globin axis are incompletely understood. We show, by transwell co-culture of nitric oxide (NO) generating, HEK eNOS/nNOS cells, and K562 erythroid or C2C12 muscle myoblasts, that low doses of NO can effectively insert heme into hemoglobin (Hb) and myoglobin (Mb), making NO not only a vasodilator, but also a globin heme trigger. We found this process to be dependent on the NO flux, occurring at low NO doses and fading at higher doses. This NO-triggered heme insertion occurred into Hb in just 30 min in K562 cells and into muscle Mb in C2C12 myoblasts between 30 min and 1 h, suggesting that the classical effect of NO on upregulation of globin (Hb or Mb) is just not transcriptional, but may involve sufficient translational events where NO can cause heme-downloading into the apo-globins (Hb/Mb). This effect of NO is unexpected and highlights its significance in maintaining globins in its heme-containing holo-form, where such heme insertions might be required in the circulating blood or in the muscle cells to perform spontaneous functions.

1. Introduction

Nitric oxide (NO) is a signal molecule and, amidst other functions, it plays a critical role in the regulation of vascular tone [1,2,3,4]. NO, generated by NOS enzymes [5,6,7,8] or produced by noncanonical means [9], is a central component of the NO-sGC-cGMP signal pathway and works by activating the soluble guanylate cyclase (sGC), an obligate heterodimer of α, β-subunits (sGC-αβ) [10,11,12]. cGMP produced by NO activation of sGC is known to transcriptionally activate fetal Hb (γ) gene expression and may also occur in primary human erythroblasts [13,14]. Likewise, NO is also known to transcriptionally stimulate myoglobin (Mb) gene expression [15]. While the heme prosthetic group is an essential component of these globins and is needed for globin functionality, our studies with globin heme maturations [16,17] have revealed that, in cells, there is a substantial amount of the heme-free globin or apo-globin (apo-Hb/apo-Mb), and this was previously unappreciated. Moreover, NO can directly cause heme insertion into the sGCβ subunit to cause its heterodimerization and subsequent activation [18,19].
In light of these finding, we speculated that the NO-driven globin synthesis and subsequent heme insertion/maturation can also have a translational component, wherein heme insertion into a pre-existing globin maybe possible. We thus studied the NO-driven globin heme insertion/maturation using transwell co-cultures of apical NO-producing cells and basal Hb/Mb-expressing cells. We document here that NO generated from both eNOS or nNOS can drive heme insertion into pre-existing fetal (K562) hemoglobin (Hbγ). A similar effect occurs in C2C12 muscle myoblasts, where the NO generated from nNOS triggers heme into muscle cells, and this may corroborate with physiological events ocurring at sites of spontaneous activity.

2. Material and Methods

2.1. Reagents

All chemicals were purchased from Sigma (St. Louis, MO, USA) and Fischer chemicals (Fair Lawn, NJ, USA). NOS inhibitors 1400 W, L-NMMA and NO scavenger, and Carboxy-PTIO were obtained from Sigma. Transwell inserts were purchased from Cell Treat Scientific Products (Pepperell, MA, USA). Stable cell lines of HEK293 expressing eNOS or nNOS were obtained from Dr. Dennis Stuehr’s lab, and these were originally gifted by Prof. Solomon Synder (Johns Hopkins, Baltimore, MD, USA). C2C12 and K562 cells were purchased from American Type Culture Collection (ATCC; Manassas, VA, USA). Antibodies specific to Mb, Hbα, or Hbγ were obtained from Santa Cruz BioTech (Dallas, TX, USA), while eNOS and nNOS antibodies were purchased from Cell Signaling Tech (Danvers, MA, USA).

2.2. Transwell Co-Culture and Growth/Induction of Cells

All cell lines were grown and harvested as previously described [17,20]. HEK cells expressing stable lines of eNOS or nNOS were first grown to confluency (50–60%); treated for 6 h with −/+ NOS inhibitors (20 µM of 1400 W, 10 µM of L-NMMA) −/+ NO scavenger, or Carboxy-PTIO (100 µΜ for a total time of 1.5 to 2 h, including 30 min incubation in co-culture with K562 cells); and then activated with Ca ionophore (10 µM) for an additional 30 min to induce nitric oxide (NO) generation. These cells were then co-cultured with basal K562/C2C12 cells for various lengths of time between 0 and 6 h (Figure 1). The cells were then harvested at various time points and the generated supernatants were assayed by Western blots for protein expression, globin heme insertion was analyzed by heme-stains, and NO generation (as nitrite) from NOSs was estimated by an ozone-based chemiluminescent assay [21]. Band intensities on Western blots or heme-stains were quantified using Image J quantification software (NIH).

2.3. Statistics

Statistics was performed on experiments that were repeated three times. Values were depicted as mean of n = 3 measures, ±SD. * p < 0.05 was considered significant, by one-way ANOVA.

3. Results

In order to determine the effect of NOS-generated NO on globin heme insertion, we first cultured HEK cells stably expressing eNOS or nNOS as apical cells in transwell inserts, while cultures of K562 were maintained in parallel. The corresponding NOS enzymes in the two stable lines were activated for NO generation by Ca ionophore for 30 min before these inserts were incubated by placing on top of K562 cells as transwell co-cultures (Figure 1 and Figure 2). Control experiments where the NOS lines were treated with NOS inhibitors 1400W or L-NMMA or with NO scavenger and Carboxy-PTIO (Figure S1) were performed in parallel [22]. These co-cultures were maintained for various lengths of time between 0 and 6 h, with both the apical and basal cells being harvested at specific time points and cell supernatants generated. As depicted in Figure 2, NO generated from both eNOS or nNOS caused a distinct heme insertion into Hbαγ in K562 cells (Figure 2A,B). There was an increase in the protein levels of Hbα/γ from 0 to 0.5 h of incubation with the activated apical NOS cells, but a still higher increase in the heme-levels of Hbαγ in the same window (a 1.1-fold Hbαγ protein increase compared with a 5.5-fold increase in Hbαγ heme levels caused by 30 min incubation of activated eNOS, and a 1.1-fold increase in Hbαγ protein compared with a nearly 7-fold increase in Hbαγ heme levels under a similar window of nNOS incubation, Figure 2B, lower panel), which suggests that heme-insertion occurred into the pre-existing apo-Hb. Control cultures of NOS lines that received NOS inhibitors (1400 W or L-NMMA) or those treated with Carboxy-PTIO (Figure S1) did not show such heme insertion into the Hbαγ, thereby proving that NO generated from the NOSs was the causative factor (Figure 2C,D and Figure S1). This NO-promoted heme insertion gradually decreased as NO levels accumulated from 0 to 6 h (Figure 2A,B), suggesting that such heme insertion was most effective at low NO doses (Figure 2C,D). We, however, evidenced some lowering in NOS protein levels at the 6 h period, which may have been because stable lines in G418 free media, when co-cultured for a long time, may cause lowering of NOS protein expression, due to deactivation of the plasmid DNA (Figure 2A). These data suggest that NO generated from NOS enzymes can cause a quick heme insertion into the pre-existing apo-Hb present in K562 cells [17]. We performed similar experiments with a combination of HEK nNOS and mouse C2C12 myoblasts, where Ca ionophore activated nNOS stables, meaning NO was incubated with C2C12 myoblasts, and heme insertion into the Mb was studied within a 0–6 h window. As depicted in Figure 3, we witnessed a similar NO-promoted heme insertion into muscle Mb, which was at a maximum at 1 h, before fading at higher NO doses. Again, in the C2C12 cells, a great majority of Mb is in the heme-free or apo state [16] and heme insertion into apo-Mb readily occurs at low NO doses. Together, our results confirm that low NO doses cause a direct heme insertion into the apo-globins, and this insertion primarily seems to occur post-translationally as there is little increase in the globin protein levels.

4. Discussion

While NO has long been postulated to bring about better oxygenation, the one explicit way by which this can happen is by increasing the heme-containing Hb levels in the blood, such that it can bind and deliver more oxygen [23]. The implications of these findings suggest that, in the vasculature, the vasodilatory effect of NO [23,24] can also be coupled with an increase in blood Hb levels. The latter effect is a novel outcome of our current findings and may happen simultaneously with vasodilation. Our data also suggest that NO can increase globin gene synthesis at the transcriptional level (Figure 2A,B), and this has been implicitly depicted in earlier studies [14], while post-translationally, it can cause heme insertion into the apo-globins, and thus can be implicated to be working at both levels. While the observed effects of NO are caused by low NO levels, which cause heme insertion into the globins, high NO levels actually inhibit heme insertion into Hb, as we determined earlier. Here, the optimum NO donor concentrations (from NOC-18) were close to 125 µM, which caused inhibition of Hb heme [25]. Previous studies also found that NO generated from iNOS inhibits its own heme insertion and subsequent dimerization, when NO accumulates post 12 h of iNOS induction in mouse macrophages (RAW 264.7), but not in the presence of NOS inhibitor, L-NAME [26]. Moreover, in our current study, the heme insertion we observe into Hb or Mb typically occurs in the low NO range (0–4 µM, as depicted from nitrite values, Figure 2C and Figure 3C), while higher nitrite values of >8 µM result in lowered heme insertion. Thus, it can be inferred that NO plays a dual role of promoting heme insertion at lower doses while impeding heme insertion at higher doses (Figure 2 and Figure 3).
Mechanistically, we do not understand how such NO-mediated globin heme insertion can occur, but may require the GAPDH-hsp90 nexus as these are proteins needed for the transport and delivery of heme into the globins [27]; at this stage, however, it would be rather speculative to suggest this, and further work is needed. There can be many scenarios where such NO-mediated globin heme insertions can occur. The most common of them can be in pathologies including tumors where there is inflammation and NOS enzymes are upregulated [28,29]. Here, NO-mediated Hb/Mb heme insertion would be a possible outcome to heme-mature these globins, such that the harmful oxidant effect can be countered with heme-containing globins [15,30]. Other scenarios may include the formation of a neuromuscular junction, where neuronal NO plays a critical role [31], and here, the resident Mb heme-maturation can be significant to improve the muscle tone. NO-mediated buildup of muscle tone during physical exercise may also involve the NO-mediated Mb heme insertion process. Moreover, during early developmental stages, the NO-mediated globin heme insertion can be a critical event [32,33]. Lastly, when NO is used as a inhalation therapy [34], such NO-mediated processes are likely to be in play, where it can increase insertions of globin heme. However, at this stage, it is difficult to corroborate these findings with actual physiological events, but there can be many such instances where these are likely to happen and our current findings, which are thought provoking, can allow further investigations into these critical processes.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/cells11182838/s1, Figure S1. NO induces heme-insertion into fetal Hb and this heme-insertion is inhibited in the presence of NO scavenger, Carboxy-PTIO; Figure S2. Full gels of western blots as depicted in Figure 2A. Molecular weight markers (KDa) are depicted at the left of the gels; Figure S3. Full gels of western blots as depicted in Figure 2A; Figure S4. Full gels of western blots as depicted in Figure 3A,B; Figure S5. Full gels of western blots as depicted in Figure S1B.

Author Contributions

A.G., M.P.S. and B.T. designed the experiments. A.G., M.P.S. and B.T. performed all transwell co-culture studies and analyzed all the data. A.G. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by National Institute of Health Grants R56HL139564 and R01HL150049 (A.G.)

Institutional Review Board Statement

No human or animal studies were performed in this study.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chen, K.; Pittman, R.N.; Popel, A.S. Nitric oxide in the vasculature: Where does it come from and where does it go? A quantitative perspective. Antioxid. Redox Signal. 2008, 10, 1185–1198. [Google Scholar] [CrossRef] [PubMed]
  2. Furchgott, R.F.; Zawadzki, J.V. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature 1980, 288, 373–376. [Google Scholar] [CrossRef] [PubMed]
  3. Palmer, R.M.; Ferrige, A.G.; Moncada, S. Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature 1987, 327, 524–526. [Google Scholar] [CrossRef] [PubMed]
  4. Jin, R.C.; Loscalzo, J. Vascular Nitric Oxide: Formation and Function. J. Blood Med. 2010, 2010, 147–162. [Google Scholar] [CrossRef] [PubMed]
  5. Murad, F. Shattuck Lecture. Nitric oxide and cyclic GMP in cell signaling and drug development. N. Engl. J. Med. 2006, 355, 2003–2011. [Google Scholar] [CrossRef]
  6. Stuehr, D.J. Mammalian nitric oxide synthases. Biochim. Biophys. Acta 1999, 1411, 217–230. [Google Scholar] [CrossRef]
  7. Stuehr, D.J.; Haque, M.M. Nitric oxide synthase enzymology in the 20 years after the Nobel Prize. Br. J. Pharmacol. 2019, 176, 177–188. [Google Scholar] [CrossRef]
  8. Marletta, M.A. Nitric oxide synthase: Aspects concerning structure and catalysis. Cell 1994, 78, 927–930. [Google Scholar] [CrossRef]
  9. Kapil, V.; Khambata, R.S.; Jones, D.A.; Rathod, K.; Primus, C.; Massimo, G.; Fukuto, J.M.; Ahluwalia, A. The Noncanonical Pathway for In Vivo Nitric Oxide Generation: The Nitrate-Nitrite-Nitric Oxide Pathway. Pharmacol. Rev. 2020, 72, 692–766. [Google Scholar] [CrossRef]
  10. Bryan, N.S.; Bian, K.; Murad, F. Discovery of the nitric oxide signaling pathway and targets for drug development. Front. Biosci. 2009, 14, 1–18. [Google Scholar] [CrossRef] [Green Version]
  11. Derbyshire, E.R.; Marletta, M.A. Biochemistry of soluble guanylate cyclase. In Handbook of Experimental Pharmacology; Springer: Cham, Switzerland, 2009; pp. 17–31. [Google Scholar] [CrossRef]
  12. Derbyshire, E.R.; Marletta, M.A. Structure and regulation of soluble guanylate cyclase. Annu. Rev. Biochem. 2012, 81, 533–559. [Google Scholar] [CrossRef] [PubMed]
  13. Cokic, V.P.; Schechter, A.N. Effects of nitric oxide on red blood cell development and phenotype. Curr. Top. Dev. Biol. 2008, 82, 169–215. [Google Scholar] [CrossRef] [PubMed]
  14. Cokic, V.P.; Smith, R.D.; Beleslin-Cokic, B.B.; Njoroge, J.M.; Miller, J.L.; Gladwin, M.T.; Schechter, A.N. Hydroxyurea induces fetal hemoglobin by the nitric oxide-dependent activation of soluble guanylyl cyclase. J. Clin. Invest. 2003, 111, 231–239. [Google Scholar] [CrossRef] [PubMed]
  15. Flonta, S.E.; Arena, S.; Pisacane, A.; Michieli, P.; Bardelli, A. Expression and functional regulation of myoglobin in epithelial cancers. Am. J. Pathol. 2009, 175, 201–206. [Google Scholar] [CrossRef] [PubMed]
  16. Ghosh, A.; Dai, Y.; Biswas, P.; Stuehr, D.J. Myoglobin maturation is driven by the hsp90 chaperone machinery and by soluble guanylyl cyclase. FASEB J. 2019, 33, 9885–9896. [Google Scholar] [CrossRef] [PubMed]
  17. Ghosh, A.; Garee, G.; Sweeny, E.A.; Nakamura, Y.; Stuehr, D.J. Hsp90 chaperones hemoglobin maturation in erythroid and nonerythroid cells. Proc. Natl. Acad. Sci. USA 2018, 115, E1117–E1126. [Google Scholar] [CrossRef]
  18. Dai, Y.; Faul, E.M.; Ghosh, A.; Stuehr, D.J. NO rapidly mobilizes cellular heme to trigger assembly of its own receptor. Proc. Natl. Acad. Sci. USA 2022, 119, e2115774119. [Google Scholar] [CrossRef]
  19. Ghosh, A.; Stasch, J.P.; Papapetropoulos, A.; Stuehr, D.J. Nitric oxide and heat shock protein 90 activate soluble guanylate cyclase by driving rapid change in its subunit interactions and heme content. J. Biol. Chem. 2014, 289, 15259–15271. [Google Scholar] [CrossRef]
  20. Ghosh, A.; Chawla-Sarkar, M.; Stuehr, D.J. Hsp90 interacts with inducible NO synthase client protein in its heme-free state and then drives heme insertion by an ATP-dependent process. FASEB J. 2011, 25, 2049–2060. [Google Scholar] [CrossRef] [Green Version]
  21. Hausladen, A.; Rafikov, R.; Angelo, M.; Singel, D.J.; Nudler, E.; Stamler, J.S. Assessment of nitric oxide signals by triiodide chemiluminescence. Proc. Natl. Acad. Sci. USA 2007, 104, 2157–2162. [Google Scholar] [CrossRef]
  22. Chowdhury, A.A.; Chaudhuri, J.; Biswas, N.; Manna, A.; Chatterjee, S.; Mahato, S.K.; Chaudhuri, U.; Jaisankar, P.; Bandyopadhyay, S. Synergistic apoptosis of CML cells by buthionine sulfoximine and hydroxychavicol correlates with activation of AIF and GSH-ROS-JNK-ERK-iNOS pathway. PLoS ONE 2013, 8, e73672. [Google Scholar] [CrossRef] [PubMed]
  23. Helms, C.C.; Gladwin, M.T.; Kim-Shapiro, D.B. Erythrocytes and Vascular Function: Oxygen and Nitric Oxide. Front. Physiol. 2018, 9, 125. [Google Scholar] [CrossRef]
  24. Zhao, Y.; Vanhoutte, P.M.; Leung, S.W. Vascular nitric oxide: Beyond eNOS. J. Pharmacol. Sci. 2015, 129, 83–94. [Google Scholar] [CrossRef]
  25. Waheed, S.M.; Ghosh, A.; Chakravarti, R.; Biswas, A.; Haque, M.M.; Panda, K.; Stuehr, D.J. Nitric oxide blocks cellular heme insertion into a broad range of heme proteins. Free Radic. Biol. Med. 2010, 48, 1548–1558. [Google Scholar] [CrossRef] [PubMed]
  26. Albakri, Q.A.; Stuehr, D.J. Intracellular assembly of inducible NO synthase is limited by nitric oxide-mediated changes in heme insertion and availability. J. Biol. Chem. 1996, 271, 5414–5421. [Google Scholar] [CrossRef] [PubMed]
  27. Tupta, B.; Stuehr, E.; Sumi, M.P.; Sweeny, E.A.; Smith, B.; Stuehr, D.J.; Ghosh, A. GAPDH is involved in the heme-maturation of myoglobin and hemoglobin. FASEB J. 2022, 36, e22099. [Google Scholar] [CrossRef]
  28. Thomas, D.D.; Wink, D.A. NOS2 as an Emergent Player in Progression of Cancer. Antioxid. Redox Signal. 2017, 26, 963–965. [Google Scholar] [CrossRef]
  29. Cobbs, C.S.; Brenman, J.E.; Aldape, K.D.; Bredt, D.S.; Israel, M.A. Expression of nitric oxide synthase in human central nervous system tumors. Cancer Res. 1995, 55, 727–730. [Google Scholar]
  30. Maman, S.; Sagi-Assif, O.; Yuan, W.; Ginat, R.; Meshel, T.; Zubrilov, I.; Keisari, Y.; Lu, W.; Lu, W.; Witz, I.P. The Beta Subunit of Hemoglobin (HBB2/HBB) Suppresses Neuroblastoma Growth and Metastasis. Cancer Res. 2017, 77, 14–26. [Google Scholar] [CrossRef]
  31. Godfrey, E.W.; Schwarte, R.C. The role of nitric oxide signaling in the formation of the neuromuscular junction. J. Neurocytol. 2003, 32, 591–602. [Google Scholar] [CrossRef]
  32. Ikuta, T.; Sellak, H.; Odo, N.; Adekile, A.D.; Gaensler, K.M. Nitric Oxide-cGMP Signaling Stimulates Erythropoiesis through Multiple Lineage-Specific Transcription Factors: Clinical Implications and a Novel Target for Erythropoiesis. PLoS ONE 2016, 11, e0144561. [Google Scholar] [CrossRef]
  33. De Palma, C.; Clementi, E. Nitric oxide in myogenesis and therapeutic muscle repair. Mol. Neurobiol. 2012, 46, 682–692. [Google Scholar] [CrossRef] [PubMed]
  34. Bloch, K.D.; Ichinose, F.; Roberts, J.D., Jr.; Zapol, W.M. Inhaled NO as a therapeutic agent. Cardiovasc. Res. 2007, 75, 339–348. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Representation of a transwell co-culture. HEK cells expressing stable lines of eNOS or nNOS were activated with Ca ionophore to induce nitric oxide (NO) generation, −/+ NOS inhibitors (1400W/L-NMMA), and then co-cultured with basal K562/C2C12 cells for various lengths of time between 0 and 6 h. Expressions of the globins (Hb/Mb) were assayed by Western blots, globin heme insertion was analyzed by heme-stains, and NO generation (as nitrite) from NOSs was estimated by an ozone-based chemiluminescent assay.
Figure 1. Representation of a transwell co-culture. HEK cells expressing stable lines of eNOS or nNOS were activated with Ca ionophore to induce nitric oxide (NO) generation, −/+ NOS inhibitors (1400W/L-NMMA), and then co-cultured with basal K562/C2C12 cells for various lengths of time between 0 and 6 h. Expressions of the globins (Hb/Mb) were assayed by Western blots, globin heme insertion was analyzed by heme-stains, and NO generation (as nitrite) from NOSs was estimated by an ozone-based chemiluminescent assay.
Cells 11 02838 g001
Figure 2. NO induces heme-insertion into fetal Hb. Stable lines of HEK cells expressing eNOS/nNOS were cultured and activated with Ca ionophore for 30 min, −/+ NOS inhibitors (1400 W/L-NMMA) before being co-cultured with K562 cells in a transwell for various lengths of time between 0 and 6 h. The cultures were then harvested and the generated supernatants were assayed for protein expression by Western blots, Hb heme by heme-stain, and eNOS/nNOS-generated NO (as nitrite) by an ozone-based chemiluminescent assay. Panel (A) Protein expression of eNOS/nNOS, Hbα/γ, and Hb heme-stains, as indicated. Panel (B) Corresponding densitometries of NOSs and Hb protein or heme levels, as depicted in panel A. Panel (C) NO estimation as nitrite by a chemiluminescent assay. Panel (D) Left, representative heme-stains of Hb, as indicated. Right, mean densitometries of heme-stains from three independent experiments. Values depicted are mean n = 3, ±SD. * p < 0.05, by one-way ANOVA. Molecular weight markers (KDa) are depicted at the left of gel bands throughout the figure legends.
Figure 2. NO induces heme-insertion into fetal Hb. Stable lines of HEK cells expressing eNOS/nNOS were cultured and activated with Ca ionophore for 30 min, −/+ NOS inhibitors (1400 W/L-NMMA) before being co-cultured with K562 cells in a transwell for various lengths of time between 0 and 6 h. The cultures were then harvested and the generated supernatants were assayed for protein expression by Western blots, Hb heme by heme-stain, and eNOS/nNOS-generated NO (as nitrite) by an ozone-based chemiluminescent assay. Panel (A) Protein expression of eNOS/nNOS, Hbα/γ, and Hb heme-stains, as indicated. Panel (B) Corresponding densitometries of NOSs and Hb protein or heme levels, as depicted in panel A. Panel (C) NO estimation as nitrite by a chemiluminescent assay. Panel (D) Left, representative heme-stains of Hb, as indicated. Right, mean densitometries of heme-stains from three independent experiments. Values depicted are mean n = 3, ±SD. * p < 0.05, by one-way ANOVA. Molecular weight markers (KDa) are depicted at the left of gel bands throughout the figure legends.
Cells 11 02838 g002
Figure 3. NO induces heme insertion into muscle Mb. Stable lines of HEK cells expressing nNOS were cultured and activated with Ca ionophore for 30 min, −/+ NOS inhibitors (1400 W/L-NMMA) before being co-cultured with C2C12 muscle myoblasts in a transwell for various lengths of time between 0 and 6 h. The harvested cultures were assayed similarly as described in Figure 2. Panel (A,B) Protein expression of nNOS and Mb, as indicated. Panel (C) Generated NO estimated as nitrite by a chemiluminescent assay. Panel (D) Upper, representative heme-stains of Mb, as indicated. Lower, mean densitometries of heme-stains from three independent experiments. Values depicted are mean n = 3, ±SD. * p < 0.05, by one-way ANOVA.
Figure 3. NO induces heme insertion into muscle Mb. Stable lines of HEK cells expressing nNOS were cultured and activated with Ca ionophore for 30 min, −/+ NOS inhibitors (1400 W/L-NMMA) before being co-cultured with C2C12 muscle myoblasts in a transwell for various lengths of time between 0 and 6 h. The harvested cultures were assayed similarly as described in Figure 2. Panel (A,B) Protein expression of nNOS and Mb, as indicated. Panel (C) Generated NO estimated as nitrite by a chemiluminescent assay. Panel (D) Upper, representative heme-stains of Mb, as indicated. Lower, mean densitometries of heme-stains from three independent experiments. Values depicted are mean n = 3, ±SD. * p < 0.05, by one-way ANOVA.
Cells 11 02838 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sumi, M.P.; Tupta, B.; Ghosh, A. Nitric Oxide Trickle Drives Heme into Hemoglobin and Muscle Myoglobin. Cells 2022, 11, 2838. https://doi.org/10.3390/cells11182838

AMA Style

Sumi MP, Tupta B, Ghosh A. Nitric Oxide Trickle Drives Heme into Hemoglobin and Muscle Myoglobin. Cells. 2022; 11(18):2838. https://doi.org/10.3390/cells11182838

Chicago/Turabian Style

Sumi, Mamta P., Blair Tupta, and Arnab Ghosh. 2022. "Nitric Oxide Trickle Drives Heme into Hemoglobin and Muscle Myoglobin" Cells 11, no. 18: 2838. https://doi.org/10.3390/cells11182838

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop