Skip to main content
Log in

Carbon Monoxide Stimulates Both Mitophagy And Mitochondrial Biogenesis to Mediate Protection Against Oxidative Stress in Astrocytes

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Astrocytes are key glial cells for the metabolic and functional support of the brain. Mitochondrial quality control (MQC), in particular the balance between mitophagy and mitochondrial biogenesis, is a major event for the maintenance of cellular homeostasis. Carbon monoxide (CO) is an endogenous gasotransmitter that inhibits cell death and inflammation by targeting mitochondria. It is well established that CO promotes cytoprotection by increasing mitochondrial population and metabolism (oxidative phosphorylation). Thus, it is hypothesized that CO-induced cytoprotection may also be mediated by the balance between mitophagy and mitochondrial biogenesis. Herein, the carbon monoxide releasing molecule-A1 (CORM-A1) was used in primary cultures of astrocytes to assess CO role on mitochondrial turnover. PINK1/Parkin-dependent mitophagy was stimulated by CORM-A1 following 1 h of treatment. While at 24 h after treatment, CORM-A1 increased mitochondrial population, which may indicate mitochondrial biogenesis. In fact, mitochondrial biogenesis was confirmed by the enhancement of PGC-1α expression that upregulates several mitochondrial transcription factors. Furthermore, inhibition of mitophagy by knocking down PINK1 expression reverted CO-induced mitochondrial biogenesis, indicating that mitochondrial turnover is dependent on modulation of mitophagy. Finally, CORM-A1 prevented astrocytic cell death induced by oxidative stress in a mitophagy-dependent manner. In fact, whenever PINK1 was knocked down, CORM-A1-induced cytoprotection was lost. In summary, CORM-A1 stimulates mitochondrial turnover, which in turn prevents astrocytic cell death. CO cytoprotection depends on increasing mitochondrial population and on eliminating dysfunctional mitochondria.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Subscribe and save

Springer+ Basic
$34.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or eBook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Data Availability

The datasets generated during and/or analysed during the current study are available from the corresponding author on a reasonable request.

Code Availability

Not applicable

References

  1. Wang Z, Figueiredo-Pereira C, Oudot C et al (2017) Mitochondrion: a common organelle for distinct cell deaths? In: International Review of Cell and Molecular Biology. Elsevier Inc., pp 245–287

  2. Galluzzi L, Kepp O, Kroemer G (2012) Mitochondria: master regulators of danger signalling. Nat Rev Mol Cell Biol 13:780–788

    Article  CAS  Google Scholar 

  3. Youle RJ, Narendra DP (2011) Mechanisms of mitophagy. Nat Rev Mol Cell Biol 12:9–14. https://doi.org/10.1038/nrm3028

    Article  CAS  Google Scholar 

  4. Gomes L, Di BG, Scorrano L (2011) During autophagy mitochondria elongate, are spared from degradation and sustain cell viability. Nat Cell Biol 13:589–598. https://doi.org/10.1038/ncb2220.During

    Article  CAS  Google Scholar 

  5. Pickles S, Vigié P, Youle RJ (2018) Mitophagy and quality control mechanisms in mitochondrial maintenance. Curr Biol 28:R170–R185. https://doi.org/10.1016/j.cub.2018.01.004

    Article  CAS  Google Scholar 

  6. Narendra D, Tanaka A, Suen D-F, Youle RJ (2008) Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol 183:795–803. https://doi.org/10.1083/jcb.200809125

    Article  Google Scholar 

  7. Terešak P, Lapao A, Subic N et al (2022) Regulation of PRKN-independent mitophagy. Autophagy 18:24–39. https://doi.org/10.1080/15548627.2021.1888244

    Article  CAS  Google Scholar 

  8. Narendra DP, Jin SM, Tanaka A et al (2010) PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol 8:e1000298. https://doi.org/10.1371/journal.pbio.1000298

    Article  CAS  Google Scholar 

  9. Fiesel F, Hudec R, Springer W (2016) Non-radioactive in vitro PINK1 kinase assays using ubiquitin or Parkin as substrate. Bio-Protoc 6:1–11. https://doi.org/10.21769/bioprotoc.1946

    Article  CAS  Google Scholar 

  10. Shiba-Fukushima K, Imai Y, Yoshida S et al (2012) PINK1-mediated phosphorylation of the Parkin ubiquitin-like domain primes mitochondrial translocation of Parkin and regulates mitophagy. Sci Rep 2:1002. https://doi.org/10.1038/srep01002

    Article  CAS  Google Scholar 

  11. Andersson U, Scarpulla RC (2001) PGC-1-related coactivator, a novel, serum-inducible coactivator of nuclear respiratory factor 1-dependent transcription in mammalian cells. Mol Cell Biol 21:3738–3749. https://doi.org/10.1128/mcb.21.11.3738-3749.2001

    Article  CAS  Google Scholar 

  12. Suliman HB, Carraway MS, Tatro LG, Piantadosi CA (2007) A new activating role for CO in cardiac mitochondrial biogenesis. J Cell Sci 120:299–308. https://doi.org/10.1242/jcs.03318

    Article  CAS  Google Scholar 

  13. Almeida AS, Queiroga CSF, Sousa MFQ et al (2012) Carbon monoxide modulates apoptosis by reinforcing oxidative metabolism in astrocytes: Role of Bcl-2. J Biol Chem 287:10761–10770. https://doi.org/10.1074/jbc.M111.306738

    Article  CAS  Google Scholar 

  14. Palikaras K, Lionaki E, Tavernarakis N (2018) Mechanisms of mitophagy in cellular homeostasis, physiology and pathology. Nat Cell Biol 20:1013–1022. https://doi.org/10.1038/s41556-018-0176-2

    Article  CAS  Google Scholar 

  15. Palikaras K, Lionaki E, Tavernarakis N (2015) Coordination of mitophagy and mitochondrial biogenesis during ageing in C. elegans. Nature 521:525–528. https://doi.org/10.1038/nature14300

    Article  CAS  Google Scholar 

  16. Motterlini R, Foresti R (2017) Biological signaling by carbon monoxide and carbon monoxide-releasing molecules. Am J Physiol Physiol 312:C302–C313. https://doi.org/10.1152/ajpcell.00360.2016

    Article  Google Scholar 

  17. Figueiredo-Pereira C, Dias-Pedroso D, Soares NL, Vieira HLA (2020) CO-mediated cytoprotection is dependent on cell metabolism modulation. Redox Biol 32:101470

    Article  CAS  Google Scholar 

  18. Queiroga CSF, Vercelli A, Vieira HLA (2015) Carbon monoxide and the CNS: challenges and achievements. Br J Pharmacol 172:1533–1545. https://doi.org/10.1111/bph.12729

    Article  CAS  Google Scholar 

  19. Basuroy S, Leffler CW, Parfenova H (2013) CORM-A1 prevents blood-brain barrier dysfunction caused by ionotropic glutamate receptor-mediated endothelial oxidative stress and apoptosis. AJP Cell Physiol 304:C1105–C1115. https://doi.org/10.1152/ajpcell.00023.2013

    Article  CAS  Google Scholar 

  20. Oliveira SR, Figueiredo-Pereira C, Duarte CB, Vieira HLA (2019) P2X7 receptors mediate CO-induced alterations in gene expression in cultured cortical astrocytes—transcriptomic study. Mol Neurobiol 56:3159–3174. https://doi.org/10.1007/s12035-018-1302-7

    Article  CAS  Google Scholar 

  21. Motterlini R, Sawle P, Hammad J et al (2005) CORM-A1: a new pharmacologically active carbon monoxide-releasing molecule. Faseb J 19:284–286. https://doi.org/10.1096/fj.04-2169fje

    Article  CAS  Google Scholar 

  22. Almeida AS, Soares NL, Sequeira CO et al (2018) Improvement of neuronal differentiation by carbon monoxide: Role of pentose phosphate pathway. Redox Biol 17:338–347. https://doi.org/10.1016/j.redox.2018.05.004

    Article  CAS  Google Scholar 

  23. Almeida AS, Soares NL, Vieira M et al (2016) Carbon monoxide releasing molecule-A1 (CORM-A1) improves neurogenesis: Increase of neuronal differentiation yield by preventing cell death. PLoS One 11:AR. https://doi.org/10.1371/journal.pone.0154781

  24. Verma A, Hirsch DJ, Glatt CE et al (1993) Carbon monoxide: a putative neural messenger. Science (80- ) 259:381 LP–384. https://doi.org/10.1126/science.7678352

  25. Leffler CW, Parfenova H, Jaggar JH (2011) Carbon monoxide as an endogenous vascular modulator. Am J Physiol - Hear Circ Physiol 301:H1–H11. https://doi.org/10.1152/ajpheart.00230.2011

    Article  CAS  Google Scholar 

  26. Vieira HLA, Queiroga CSF, Alves PM (2008) Pre-conditioning induced by carbon monoxide provides neuronal protection against apoptosis. J Neurochem 107:375–384. https://doi.org/10.1111/j.1471-4159.2008.05610.x

    Article  CAS  Google Scholar 

  27. Schallner N, Romão CC, Biermann J et al (2013) Carbon monoxide abrogates ischemic insult to neuronal cells via the soluble guanylate cyclase-cGMP pathway. PLoS One 8. https://doi.org/10.1371/journal.pone.0060672

  28. Queiroga CSF, Alves RMA, Conde SV et al (2016) Paracrine effect of carbon monoxide – astrocytes promote neuroprotection through purinergic signaling in mice. J Cell Sci 129:3178–3188. https://doi.org/10.1242/jcs.187260

    Article  CAS  Google Scholar 

  29. Queiroga CSF, Almeida AS, Martel C et al (2010) Glutathionylation of adenine nucleotide translocase induced by carbon monoxide prevents mitochondrial membrane permeabilization and apoptosis. J Biol Chem 285:17077–17088. https://doi.org/10.1074/jbc.M109.065052

    Article  CAS  Google Scholar 

  30. Bani-Hani MG, Greenstein D, Mann BE et al (2006) A carbon monoxide-releasing molecule (CORM-3) attenuates lipopolvsaccharide- And interferon-γ-induced inflammation in microglia. Pharmacol Rep 58:132–144

    Google Scholar 

  31. Wilson JLJL, Bouillaud F, Almeida ASAS et al (2017) Carbon monoxide reverses the metabolic adaptation of microglia cells to an inflammatory stimulus. Free Radic Biol Med 104:311–323. https://doi.org/10.1016/j.freeradbiomed.2017.01.022

    Article  CAS  Google Scholar 

  32. Soares NL, Paiva I, Bravo J et al (2022) Carbon monoxide modulation of microglia-neuron communication: anti-neuroinflammatory and neurotrophic role. Mol Neurobiol 59:872–889. https://doi.org/10.1007/s12035-021-02643-z

    Article  CAS  Google Scholar 

  33. Dias-Pedroso D, Ramalho JS, Sardão VA et al (2022) Carbon monoxide-neuroglobin axis targeting metabolism against inflammation in BV-2 microglial cells. Mol Neurobiol 59:916–931. https://doi.org/10.1007/s12035-021-02630-4

    Article  CAS  Google Scholar 

  34. Wang J, Zhang D, Fu X et al (2018) Carbon monoxide-releasing molecule-3 protects against ischemic stroke by suppressing neuroinflammation and alleviating blood-brain barrier disruption. J Neuroinflammation 15:188. https://doi.org/10.1186/s12974-018-1226-1

    Article  CAS  Google Scholar 

  35. Queiroga CSF, Tomasi S, Widerøe M et al (2012) Preconditioning triggered by carbon monoxide (CO) provides neuronal protection following perinatal hypoxia-ischemia. PLoS One 7. https://doi.org/10.1371/journal.pone.0042632

  36. Yabluchanskiy A, Sawle P, Homer-Vanniasinkam S et al (2012) CORM-3, a carbon monoxide-releasing molecule, alters the inflammatory response and reduces brain damage in a rat model of hemorrhagic stroke*. Crit Care Med 40:544–552

    Article  CAS  Google Scholar 

  37. Lee S, Lee S-J, Coronata AA et al (2014) Carbon monoxide confers protection in sepsis by enhancing Beclin 1-dependent autophagy and phagocytosis. Antioxid Redox Signal 20:432–442. https://doi.org/10.1089/ars.2013.5368

    Article  CAS  Google Scholar 

  38. Kim D-SS, Song L, Wang J et al (2018) Carbon monoxide inhibits islet apoptosis via induction of autophagy. Antioxid Redox Signal 28:1309–1322. https://doi.org/10.1089/ars.2016.6979

    Article  CAS  Google Scholar 

  39. Figueiredo-Pereira C, Menezes R, Ferreira S et al (2019) Carbon monoxide released by CORM-A1 prevents yeast cell death via autophagy stimulation. FEMS Yeast Res 19. https://doi.org/10.1093/femsyr/foz051

  40. Lee S-J, Ryter SW, Xu J-F et al (2011) Carbon monoxide activates autophagy via mitochondrial reactive oxygen species formation. Am J Respir Cell Mol Biol 45:867–873

    Article  CAS  Google Scholar 

  41. McWilliams TG, Prescott AR, Allen GFG et al (2016) mito-QC illuminates mitophagy and mitochondrial architecture in vivo. J Cell Biol 214:333–345. https://doi.org/10.1083/jcb.201603039

    Article  CAS  Google Scholar 

  42. Sá Santos S, Fonseca LL, Monteiro MAR et al (2005) Culturing primary brain astrocytes under a fully controlled environment in a novel bioreactor. J Neurosci Res 79:26–32. https://doi.org/10.1002/jnr.20279

    Article  CAS  Google Scholar 

  43. Mauro-Lizcano M, Esteban-Martínez L, Seco E et al (2015) New method to assess mitophagy flux by flow cytometry. Autophagy 11:833–843. https://doi.org/10.1080/15548627.2015.1034403

    Article  CAS  Google Scholar 

  44. Hull TD, Boddu R, Guo L et al (2016) Heme oxygenase-1 regulates mitochondrial quality control in the heart. JCI Insight 1. https://doi.org/10.1172/jci.insight.85817

  45. Allaman I, Be M, Magistretti PJ et al (2011) Astrocyte-neuron metabolic relationships: for better and for worse. Trends Neurosci 34:76–87. https://doi.org/10.1016/j.tins.2010.12.001

    Article  CAS  Google Scholar 

  46. Iadecola C (2017) The neurovascular unit coming of age: a journey through neurovascular coupling in health and disease. Neuron 96:17–42

    Article  CAS  Google Scholar 

  47. Ivankovic D, Chau K, Schapira AHV, Gegg ME (2016) Mitochondrial and lysosomal biogenesis are activated following <scp>PINK</scp> 1/parkin-mediated mitophagy. J Neurochem 136:388–402. https://doi.org/10.1111/jnc.13412

    Article  CAS  Google Scholar 

  48. Kuroda Y, Mitsui T, Kunishige M et al (2006) Parkin enhances mitochondrial biogenesis in proliferating cells. Hum Mol Genet 15:883–895. https://doi.org/10.1093/hmg/ddl006

    Article  CAS  Google Scholar 

  49. Peng K, Xiao J, Yang L et al (2019) Mutual antagonism of PINK1/Parkin and PGC-1α contributes to maintenance of mitochondrial homeostasis in rotenone-induced neurotoxicity. Neurotox Res 35:331–343. https://doi.org/10.1007/s12640-018-9957-4

    Article  CAS  Google Scholar 

  50. Nishida T, Yamada Y (2016) SUMOylation of the KRAB zinc-finger transcription factor PARIS/ZNF746 regulates its transcriptional activity. Biochem Biophys Res Commun 473:1261–1267. https://doi.org/10.1016/j.bbrc.2016.04.051

    Article  CAS  Google Scholar 

  51. Lupo A, Cesaro E, Montano G et al (2013) KRAB-zinc finger proteins : a repressor family displaying multiple biological functions. 268–278

  52. Zhang Y, Sauler M, Shinn AS et al (2014) Endothelial PINK1 mediates the protective effects of NLRP3 deficiency during lethal oxidant injury. J Immunol 192:5296–5304. https://doi.org/10.4049/jimmunol.1400653

    Article  CAS  Google Scholar 

  53. Suliman HB, Carraway MS, Ali AS et al (2007) The CO / HO system reverses inhibition of mitochondrial biogenesis and prevents murine doxorubicin cardiomyopathy. J Clin Invest 117:3730–3741. https://doi.org/10.1172/JCI32967.3730

    Article  CAS  Google Scholar 

  54. Choi YK, Park JH, Baek Y-Y et al (2016) Carbon monoxide stimulates astrocytic mitochondrial biogenesis via L-type Ca 2+ channel-mediated PGC-1α/ERRα activation. Biochem Biophys Res Commun 479:297–304. https://doi.org/10.1016/j.bbrc.2016.09.063

    Article  CAS  Google Scholar 

  55. Kim HJ, Joe Y, Rah S-Y et al (2018) Carbon monoxide-induced TFEB nuclear translocation enhances mitophagy/mitochondrial biogenesis in hepatocytes and ameliorates inflammatory liver injury. Cell Death Dis 9:1060. https://doi.org/10.1038/s41419-018-1112-x

    Article  CAS  Google Scholar 

  56. Taillé C, El-Benna J, Lanone S et al (2005) Mitochondrial respiratory chain and NAD(P)H oxidase are targets for the antiproliferative effect of carbon monoxide in human airway smooth muscle. J Biol Chem 280:25350–25360. https://doi.org/10.1074/jbc.M503512200

    Article  CAS  Google Scholar 

  57. Zuckerbraun BS, Chin BY, Bilban M et al (2007) Carbon monoxide signals via inhibition of cytochrome c oxidase and generation of mitochondrial reactive oxygen species. FASEB J 21:1099–1106. https://doi.org/10.1096/fj.06-6644com

    Article  CAS  Google Scholar 

  58. Wegiel B, Gallo D, Csizmadia E et al (2013) Carbon monoxide expedites metabolic exhaustion to inhibit tumor growth. Cancer Res 73:7009–7021. https://doi.org/10.1158/0008-5472.CAN-13-1075

    Article  CAS  Google Scholar 

  59. Almeida AS, Sonnewald U, Alves PM, Vieira HLA (2016) Carbon monoxide improves neuronal differentiation and yield by increasing the functioning and number of mitochondria. J Neurochem 138:423–435. https://doi.org/10.1111/jnc.13653

    Article  CAS  Google Scholar 

  60. Upadhyay KK, Jadeja RN, Vyas HS et al (2020) Carbon monoxide releasing molecule-A1 improves nonalcoholic steatohepatitis via Nrf2 activation mediated improvement in oxidative stress and mitochondrial function. Redox Biol 28:101314. https://doi.org/10.1016/j.redox.2019.101314

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Ian Ganley, University of Dundee, for kindly providing the MitoQC mice. We also thank Flow Cytometry Facility of NMS-UNL, as well as UCIBIO’s BioLabs, NOVA-FCT-UNL.

Funding

The funding agency that supported the work is “Fundação para a Ciência e Tecnologia” (FCT) with 4 projects: Applied Molecular Biosciences Unit-UCIBIO (UID/Multi/04378/2020), iNOVA4Health - Programme in Translational Medicine (UID/Multi/04462/2013), LA/P/0140/2020 of the Associate Laboratory Institute for Health and Bioeconomy and PTDC/MEC-NEU/28750/2017 and the PhD scholarship for CFP with reference PD/BD/106057/2015.

Author information

Authors and Affiliations

Authors

Contributions

CFP designed the study, conducted the experiments, helped with data analysis and partially wrote the manuscript, and approved the final version of the manuscript. BVZ designed the study, conducted the experiments, helped with data analysis, and approved the final version of the manuscript. PCC conducted some experiments, helped with data analysis, and approved the final version of the manuscript. DT conducted some experiments and approved the final version of the manuscript. IRP helped with mitoQC model and approved the final version of the manuscript. PB designed the study, helped with data analysis, and approved the final version of this manuscript. PJO helped with data analysis and approved the final version of the manuscript. HLAV designed the study, helped with data analysis and evaluation, wrote the manuscript, and approved the final version of this manuscript.

Corresponding author

Correspondence to Helena L. A. Vieira.

Ethics declarations

Ethics Approval

Not applicable

Consent to Participate

Not applicable

Consent for Publication

Not applicable

Conflict of Interest

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary Figure 1

Purity of primary culture of astrocytes. Representative picture of immunofluorescent microscopy using anti-GFAP to verify the purity level of astrocytic primary cell culture. (PNG 953 kb)

High resolution image (TIFF 2909 kb)

Supplementary Figure 2

Control of knocking down Pink1 by siRNA transfection. Primary culture of astrocytes were transfected with Pink1 siRNA for 24 or 48h with 10 or 15 pmol of siRNA per well (24 well plates). Pink1 expression was assessed by western blot analysis. One representative membrane of 24h of transfection is presented and the respective quantification of four biological replicates and data were analysed with the Mann-Whitney test, * p<0.05 compared to control. (PNG 170 kb)

High resolution image (TIFF 4207 kb)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Figueiredo-Pereira, C., Villarejo-Zori, B., Cipriano, P.C. et al. Carbon Monoxide Stimulates Both Mitophagy And Mitochondrial Biogenesis to Mediate Protection Against Oxidative Stress in Astrocytes. Mol Neurobiol 60, 851–863 (2023). https://doi.org/10.1007/s12035-022-03108-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-022-03108-7

Keywords

Navigation