Mitochondrial signaling: forwards, backwards, and in between

Sean P Whelan, Brian S Zuckerbraun, Sean P Whelan, Brian S Zuckerbraun

Abstract

Mitochondria are semiautonomous organelles that are a defining characteristic of almost all eukaryotic cells. They are vital for energy production, but increasing evidence shows that they play important roles in a wide range of cellular signaling and homeostasis. Our understanding of nuclear control of mitochondrial function has expanded over the past half century with the discovery of multiple transcription factors and cofactors governing mitochondrial biogenesis. More recently, nuclear changes in response to mitochondrial messaging have led to characterization of retrograde mitochondrial signaling, in which mitochondria have the ability to alter nuclear gene expression. Mitochondria are also integral to other components of stress response or quality control including ROS signaling, unfolded protein response, mitochondrial autophagy, and biogenesis. These avenues of mitochondrial signaling are discussed in this review.

Figures

Figure 1
Figure 1
Diagrammatic summary of the nuclear control of mitochondrial functions by NRF-1 and NRF-2 (GABP). NRFs contribute both directly and indirectly to the expression of many genes required for the maintenance and function of the mitochondrial respiratory apparatus. NRFs act on genes encoding cytochrome c, the majority of nuclear subunits of respiratory complexes I–V, and the rate-limiting heme biosynthetic enzyme 5-aminolevulinate synthase. In addition, NRFs promote the expression of key components of the mitochondrial transcription and translation machinery that are necessary for the production of respiratory subunits encoded by mtDNA. These include Tfam, TFB1M, and TFB2M as well as a number of mitochondrial ribosomal proteins and tRNA synthetases. Recent findings suggest that NRFs are also involved in the expression of key components of the protein import and assembly machinery. Adapted with permission from [2].
Figure 2
Figure 2
Positive and negative regulators of the retrograde pathway. The retrograde pathway is constitutively inhibited by Mks1p as well as TOR1/2p/Lst8p which hyperphosphorylates (P) the Rtg1/3p heterodimer. Bmh1/2p stabilizes the phosphorylated Mks1p contributing to its activity and preventing its degradation. Mitochondrial stress activates Rtg2p which dephosphorylates Mks1p. Mks1p then dissociates from Bmh1/2p and is degraded by Grr1p. Rtg2p also inhibits the inhibitory factor Lst8p. Additionally, Lst8p is part of the TOR1/2p complex and is also controlled by canonical regulators of TOR. The disinhibition of the Rtg1/3p heterodimer allows dephosphorylation and translocation to the nucleus where it activates the RTG genes. The prototypical RTG gene CIT2 encodes peroxisomal citrate synthase (CIT2) which converts Acetyl-CoA and oxaloacetic acid (OAA) to citrate. This contributes nitrogen to the TCA cycle in order to maintain an adequate supply of α-ketoglutarate. Ultimately, this leads to production of glutamate which is the ultimate source of biosynthetic reactions in yeast. The plasma membrane amino acid sensor SPS inhibits Rtg2p in a negative feedback mechanism in the presence of excess glutamate.
Figure 3
Figure 3
Hypothetical model of the C. elegans UPRmt pathway. Protein conformational stress in the mitochondrial matrix triggers CLPP-1 proteolysis of an unknown substrate, producing a stress signal (blue line). The stress signal is conveyed to the cytoplasm and induces nuclear translocation and complex formation of UBL-5 and DVE-1, as well as binding of DVE-1 to the promoter of the chaperone target gene, HSP-60. This stress-signaling pathway results in the induction of mitochondrial chaperone genes, HSP-60 and HSP-6. ubl-5 expression is also upregulated, which in turn amplifies the UPRmt signal (green-dotted line). HSP-60 and HSP-6 are imported into the mitochondria, where they help to restore protein homeostasis by refolding rogue proteins. Adapted with permission from [51].

References

    1. Ernster L, Schatz G. Mitochondria: a historical review. Journal of Cell Biology. 1981;91(3, part 2):227s–255s.
    1. Scarpulla RC. Transcriptional paradigms in mammalian mitochondrial biogenesis and function. Physiological Reviews. 2008;88(2):611–638.
    1. Parikh VS, Morgan MM, Scott R, Clements LS, Butow RA. The mitochondrial genotype can influence nuclear gene expression in yeast. Science. 1987;235(4788):576–580.
    1. Pejznochova M, Tesarova M, Hansikova H, et al. Mitochondrial DNA content and expression of genes involved in mtDNA transcription, regulation and maintenance during human fetal development. Mitochondrion. 2010;10(4):321–329.
    1. Hsieh CJ, Weng SW, Liou CW, et al. Tissue-specific differences in mitochondrial DNA content in type 2 diabetes. Diabetes Research and Clinical Practice. 2011;92(1):106–110.
    1. Wallace DC. Mitochondria, bioenergetics, and the epigenome in eukaryotic and human evolution. Cold Spring Harbor Symposia on Quantitative Biology. 2009;74:383–393.
    1. Scarpulla RC. Nuclear activators and coactivators in mammalian mitochondrial biogenesis. Biochimica et Biophysica Acta. 2002;1576(1-2):1–14.
    1. Fisher RP, Clayton DA. A transcription factor required for promoter recognition by human mitochondrial RNA polymerase. Accurate initiation at the heavy- and light-strand promoters dissected and reconstituted in vitro. The Journal of Biological Chemistry. 1985;260(20):11330–11338.
    1. Fisher RP, Clayton DA. Purification and characterization of human mitochondrial transcription factor 1. Molecular and Cellular Biology. 1988;8(8):3496–3509.
    1. Amiott EA, Jaehning JA. Mitochondrial transcription is regulated via an ATP “sensing” mechanism that couples RNA abundance to respiration. Molecular Cell. 2006;22(3):329–338.
    1. Tracy RL, Stern DB. Mitochondrial transcription initiation: promotor structures and RNA polymerases. Current Genetics. 1995;28(3):205–216.
    1. Tiranti V, Savoia A, Forti F, et al. Identification of the gene encoding the human mitochondrial RNA polymerase (h-mtRPOL) by cyberscreening of the expressed sequence tags database. Human Molecular Genetics. 1997;6(4):615–625.
    1. Evans MJ, Scarpulla RC. Both upstream and intron sequence elements are required for elevated expression of the rat somatic cytochrome c gene in COS-1 cells. Molecular and Cellular Biology. 1988;8(1):35–41.
    1. Evans MJ, Scarpulla RC. Interaction of nuclear factors with multiple sites in the somatic cytochrome c promoter. Characterization of upstream NRF-1, ATF, and intron Sp1 recognition sequences. The Journal of Biological Chemistry. 1989;264(24):14361–14368.
    1. Scarpulla RC. Transcriptional activators and coactivators in the nuclear control of mitochondrial function in mammalian cells. Gene. 2002;286(1):81–89.
    1. Evans MJ, Scarpulla RC. NRF-1: a trans-activator of nuclear-encoded respiratory genes in animal cells. Genes and Development. 1990;4(6):1023–1034.
    1. Zaid A, Li R, Luciakova K, Barath P, Nery S, Nelson BD. On the role of the general transcription factor Sp1 in the activation and repression of diverse mammalian oxidative phosphorylation genes. Journal of Bioenergetics and Biomembranes. 1999;31(2):129–135.
    1. Spiegelman BM. Transcriptional control of energy homeostasis through the PGC1 coactivators. Novartis Foundation Symposium. 2007;286:196–203.
    1. Meirhaeghe A, Crowley V, Lenaghan C, et al. Characterization of the human, mouse and rat PGC1β (peroxisomeproliferator-activated receptor-γ co-activator 1β) gene in vitro and in vivo. Biochemical Journal. 2003;373(1):155–165.
    1. Biswas G, Adebanjo OA, Freedman BD, et al. Retrograde Ca2+ signaling in C2C12 skeletal myocytes in response to mitochondrial genetic and metabolic stress: a novel mode of inter-organelle crosstalk. EMBO Journal. 1999;18(3):522–533.
    1. Schwarzlander M, Konig AC, Sweetlove LJ, Finkemeier I. The impact of impaired mitochondrial function on retrograde signalling: a meta-analysis of transcriptomic responses. Journal of Experimental Botany. 2012;(4):1735–1750.
    1. Liao X, Small WC, Srere PA, Butow RA. Intramitochondrial functions regulate nonmitochondrial citrate synthase (CIT2) expression in Saccharomyces cerevisiae . Molecular and Cellular Biology. 1991;11(1):38–46.
    1. Liao X, Butow RA. RTG1 and RTG2: two yeast genes required for a novel path of communication from mitochondria to the nucleus. Cell. 1993;72(1):61–71.
    1. Liu Z, Butow RA. A transcriptional switch in the expression of yeast tricarboxylic acid cycle genes in response to a reduction or loss of respiratory function. Molecular and Cellular Biology. 1999;19(10):6720–6728.
    1. Epstein CB, Waddle JA, Hale W, et al. Genome-wide responses to mitochondrial dysfunction. Molecular Biology of the Cell. 2001;12(2):297–308.
    1. Freije WA, Mandal S, Banerjee U. Expression profiling of attenuated mitochondrial function identifies retrograde signals in Drosophila. G3. 2012;2(8):843–851.
    1. Liu Z, Spírek M, Thornton J, Butow RA. A novel degron-mediated degradation of the RTG pathway regulator, Mks1p, by SCF. Molecular Biology of the Cell. 2005;16(10):4893–4904.
    1. Liu Z, Butow RA. Mitochondrial retrograde signaling. Annual Review of Genetics. 2006;40:159–185.
    1. Dilova I, Chen CY, Powers T. Mks1 in concert with TOR signaling negatively regulates RTG target gene expression in S. cerevisiae . Current Biology. 2002;12(5):389–395.
    1. Liu Z, Sekito T, Špírek M, Thornton J, Butow RA. Retrograde signaling is regulated by the dynamic interaction between Rtg2p and Mks1p. Molecular Cell. 2003;12(2):401–411.
    1. Owusu-Ansah E, Yavari A, Mandal S, Banerjee U. Distinct mitochondrial retrograde signals control the G1-S cell cycle checkpoint. Nature Genetics. 2008;40(3):356–361.
    1. Berridge MJ, Bootman MD, Lipp P. Calcium—a life and death signal. Nature. 1998;395(6703):645–648.
    1. Carafoli E. The interplay of mitochondria with calcium: an historical appraisal. Cell Calcium. 2012;52(1):1–8.
    1. Nicholls DG. Mitochondria and calcium signaling. Cell Calcium. 2005;38(3-4):311–317.
    1. Bayir H, Kagan VE. Bench-to-bedside review: mitochondrial injury, oxidative stress and apoptosis—there is nothing more practical than a good theory. Critical Care. 2008;12(1, article 206)
    1. Butow RA, Avadhani NG. Mitochondrial signaling: the retrograde response. Molecular Cell. 2004;14(1):1–15.
    1. Amuthan G, Biswas G, Ananadatheerthavarada HK, Vijayasarathy C, Shephard HM, Avadhani NG. Mitochondrial stress-induced calcium signaling, phenotypic changes and invasive behavior in human lung carcinoma A549 cells. Oncogene. 2002;21(51):7839–7849.
    1. Arnould T, Vankoningsloo S, Renard P, et al. Creb activation induced by mitochondrial dysfunction is a new signaling pathway that impairs cell proliferation. EMBO Journal. 2002;21(1-2):53–63.
    1. Vankoningsloo S, de Pauw A, Houbion A, et al. CREB activation induced by mitochondrial dysfunction triggers triglyceride accumulation in 3T3-L1 preadipocytes. Journal of Cell Science. 2006;119(part 7):1266–1282.
    1. Biswas G, Anandatheerthavarada HK, Zaidi M, Avadhani NG. Mitochondria to nucleus stress signaling: a distinctive mechanism of NFκB/Rel activation through calcineurin-mediated inactivation of IκBβ . Journal of Cell Biology. 2003;161(3):507–519.
    1. Srinivasan V, Kriete A, Sacan A, Jazwinski SM. Comparing the yeast retrograde response and NF-κB stress responses: implications for aging. Aging Cell. 2010;9(6):933–941.
    1. Le Bras M, Clément MV, Pervaiz S, Brenner C. Reactive oxygen species and the mitochondrial signaling pathway of cell death. Histology and Histopathology. 2005;20(1):205–219.
    1. Itoh K, Wakabayashi N, Katoh Y, et al. Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes and Development. 1999;13(1):76–86.
    1. Formentini L, Sanchez-Arago M, Sanchez-Cenizo L, Cuezva JM. The mitochondrial ATPase inhibitory factor 1 triggers a ROS-mediated retrograde prosurvival and proliferative response. Molecular Cell. 2012;45(6):731–742.
    1. Park JS, Sharma LK, Li H, et al. A heteroplasmic, not homoplasmic, mitochondrial DNA mutation promotes tumorigenesis via alteration in reactive oxygen species generation and apoptosis. Human Molecular Genetics. 2009;18(9):1578–1589.
    1. Lee HC, Chang CM, Chi CW. Somatic mutations of mitochondrial DNA in aging and cancer progression. Ageing Research Reviews. 2010;9(supplement 1):S47–S58.
    1. Correia RL, Oba-Shinjo SM, Uno M, Huang N, Marie SKN. Mitochondrial DNA depletion and its correlation with TFAM, TFB1M, TFB2M and POLG in human diffusely infiltrating astrocytomas. Mitochondrion. 2011;11(1):48–53.
    1. Wallace DC. Mitochondria and cancer. Nature Reviews Cancer. 2012;12(10):685–698.
    1. Pellegrino MW, Nargund AM, Haynes CM. Signaling the mitochondrial unfolded protein response. Biochimica et Biophysica Acta. 2013;1833(2):410–416.
    1. Haynes CM, Petrova K, Benedetti C, Yang Y, Ron D. ClpP mediates activation of a mitochondrial unfolded protein response in C. elegans . Developmental Cell. 2007;13(4):467–480.
    1. Broadley SA, Hartl FU. Mitochondrial stress signaling: a pathway unfolds. Trends in Cell Biology. 2008;18(1):1–4.
    1. Aldridge JE, Horibe T, Hoogenraad NJ. Discovery of genes activated by the mitochondrial unfolded protein response (mtUPR) and cognate promoter elements. PLoS ONE. 2007;2(9, article e874)
    1. Horibe T, Hoogenraad NJ. The Chop gene contains an element for the positive regulation of the mitochondrial unfolded protein response. PLoS ONE. 2007;2(9, article e835)
    1. Siegelin MD, Dohi T, Raskett CM, et al. Exploiting the mitochondrial unfolded protein response for cancer therapy in mice and human cells. Journal of Clinical Investigation. 2011;121(4):1349–1360.
    1. Papa L, Germain D. Estrogen receptor mediates a distinct mitochondrial unfolded protein response. Journal of Cell Science. 2011;124(9):1396–1402.
    1. Bernales S, Soto MM, McCullagh E. Unfolded protein stress in the endoplasmic reticulum and mitochondria: a role in neurodegeneration. Frontiers in Aging Neuroscience. 2012;4(article 5)
    1. Witte ME, Nijland PG, Drexhage JA, et al. Reduced expression of PGC-1α partly underlies mitochondrial changes and correlates with neuronal loss in multiple sclerosis cortex. Acta Neuropathologica. 2013;125(2, article 231)
    1. Zorov DB, Juhaszova M, Sollott SJ. Mitochondrial ROS-induced ROS release: an update and review. Biochimica et Biophysica Acta. 2006;1757(5-6):509–517.
    1. Zorov DB, Filburn CR, Klotz LO, Zweier JL, Sollott SJ. Reactive oxygen species (ROS)-induced ROS release: a new phenomenon accompanying induction of the mitochondrial permeability transition in cardiac myocytes. Journal of Experimental Medicine. 2000;192(7):1001–1014.
    1. Zhou L, Aon MA, Almas T, Cortassa S, Winslow RL, O’Rourke B. A reaction-diffusion model of ROS-induced ROS release in a mitochondrial network. PLoS Computational Biology. 2010;6(1)e1000657
    1. Park J, Choi C. Contribution of mitochondrial network dynamics to intracellular ROS signaling. Communicative and Integrative Biology. 2012;5(1):81–83.
    1. Zinkevich NS, Gutterman DD. ROS-induced ROS release in vascular biology: redox-redox signaling. The American Journal of Physiology. 2011;301(3):H647–H653.
    1. Huang J, Lam GY, Brumell JH. Autophagy signaling through reactive oxygen species. Antioxidants and Redox Signaling. 2011;14(11):2215–2231.
    1. Kadandale P, Kiger AA. Role of selective autophagy in cellular remodeling: “self-eating” into shape. Autophagy. 2010;6(8):1194–1195.
    1. Wang Y, Nartiss Y, Steipe B, McQuibban GA, Kim PK. ROS-induced mitochondrial depolarization initiates PARK2/PARKIN-dependent mitochondrial degradation by autophagy. Autophagy. 2012;8(10):1462–1476.
    1. Chen X, Li M, Chen D, et al. Autophagy induced by calcium phosphate precipitates involves endoplasmic reticulum membranes in autophagosome biogenesis. PLoS ONE. 2012;7(12)e52347
    1. Arnoult D, Rismanchi N, Grodet A, et al. Bax/bak-dependent release of DDP/TIMM8a promotes Drp1-mediated mitochondrial fission and mitoptosis during programmed cell death. Current Biology. 2005;15(23):2112–2118.
    1. Lyamzaev KG, Nepryakhina OK, Saprunova VB, et al. Novel mechanism of elimination of malfunctioning mitochondria (mitoptosis): formation of mitoptotic bodies and extrusion of mitochondrial material from the cell. Biochimica et Biophysica Acta. 2008;1777(7-8):817–825.
    1. Chen SD, Yang DI, Lin TK, Shaw FZ, Liou CW, Chuang YC. Roles of oxidative stress, apoptosis, PGC-1α and mitochondrial biogenesis in cerebral ischemia. International Journal of Molecular Sciences. 2011;12(10):7199–7215.
    1. Gleyzer N, Vercauteren K, Scarpulla RC. Control of mitochondrial transcription specificity factors (TFB1M and TFB2M) by nuclear respiratory factors (NRF-1 and NRF-2) and PGC-1 family coactivators. Molecular and Cellular Biology. 2005;25(4):1354–1366.
    1. Jahnke VE, Sabido O, Freyssenet D. Control of mitochondrial biogenesis, ROS level, and cytosolic Ca2+ concentration during the cell cycle and the onset of differentiation in L6E9 myoblasts. The American Journal of Physiology. 2009;296(5):C1185–C1194.
    1. Underhill DM, Ozinsky A. Phagocytosis of microbes: complexity in action. Annual Review of Immunology. 2002;20:825–852.
    1. West AP, Brodsky IE, Rahner C, et al. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature. 2011;472(7344):476–480.
    1. Harman D. Aging: a theory based on free radical and radiation chemistry. Journal of Gerontology. 1956;11(3):298–300.
    1. Barja G, Herrero A. Oxidative damage to mitochondrial DNA is inversely related to maximum life span in the heart and brain of mammals. FASEB Journal. 2000;14(2):312–318.
    1. Severin FF, Severina II, Antonenko YN, et al. Penetrating cation/fatty acid anion pair as a mitochondria-targeted protonophore. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(2):663–668.
    1. Skulachev VP, Anisimov VN, Antonenko YN, et al. An attempt to prevent senescence: a mitochondrial approach. Biochimica et Biophysica Acta. 2009;1787(5):437–461.
    1. Smith RA, Porteous CM, Coulter CV, Murphy MP. Selective targeting of an antioxidant to mitochondria. European Journal of Biochemistry. 1999;263(3):709–716.
    1. James AM, Cochemé HM, Smith RAJ, Murphy MP. Interactions of mitochondria-targeted and untargeted ubiquinones with the mitochondrial respiratory chain and reactive oxygen species: implications for the use of exogenous ubiquinones as therapies and experimental tools. The Journal of Biological Chemistry. 2005;280(22):21295–21312.
    1. Murphy MP, Smith RA. Targeting antioxidants to mitochondria by conjugation to lipophilic cations. Annual Review of Pharmacology and Toxicology. 2007;47:629–656.
    1. Skulachev VP, Antonenko YN, Cherepanov DA, et al. Prevention of cardiolipin oxidation and fatty acid cycling as two antioxidant mechanisms of cationic derivatives of plastoquinone (SkQs) Biochimica et Biophysica Acta. 2010;1797(6-7):878–889.
    1. Jazwinski SM. The retrograde response: when mitochondrial quality control is not enough. Biochimica et Biophysica Acta. 2013;1833(2):400–409.
    1. Sinclair DA, Guarente L. Extrachromosomal rDNA circles—a cause of aging in yeast. Cell. 1997;91(7):1033–1042.
    1. Jazwinski SM. The genetics of aging in the yeast Saccharomyces cerevisiae . Genetica. 1993;91(1–3):35–51.
    1. Jazwinski SM. The retrograde response links metabolism with stress responses, chromatin-dependent gene activation, and genome stability in yeast aging. Gene. 2005;354(1-2):22–27.

Source: PubMed

Подписаться