G Model

ARTICLE IN PRESS

BC 4551 1–6

The International Journal of Biochemistry & Cell Biology xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

The International Journal of Biochemistry & Cell Biology journal homepage: www.elsevier.com/locate/biocel

Organelles in focus

1

The complex crosstalk between mitochondria and the nucleus: What goes in between?夽

2

3

4 5 6

Q1

Umut Cagina , José Antonio Enriqueza,b,∗ a b

Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias Universidad de Zaragoza, Zaragoza, Spain

7

8 22

a r t i c l e

i n f o

a b s t r a c t

9 10 11 12 13 14

Article history: Received 12 December 2014 Received in revised form 21 January 2015 Accepted 29 January 2015 Available online xxx

15 16 17 18 19 20 21

Keywords: Mitochondria Oxidative phosphorylation Mitochondrial capacity Mitochondrial diseases Retrograde signaling

Mitochondria are critical metabolic hubs in which catabolic and anabolic cellular processes converge and are integrated. To perform their function, mitochondria also need to respond to signals that monitor their function and send continuous feedback to the nucleus and other organelles to trigger the required expression programs (for example, stabilization of hypoxia-inducible factor 1 − ˛). Unsurprisingly, mitochondrial dysfunction results in wide range of disorders. Understanding how cells adapt to changes in mitochondrial function is critical for the evaluation of mitochondrial disorders and the development of potential treatments. Each type of mitochondrial dysfunction results in a unique transcriptional response. Here we review the role of nuclear-encoded factors in the response to changes in mitochondrial function and discuss their relevance to metabolic homeostasis, outlining the diverse and complex ways in which nuclei adapt to maintain mitochondrial homeostasis. This article is part of a Directed Issue entitled: Mitochondrial Diseases. © 2015 Published by Elsevier Ltd.

1. Introduction Organelle facts: - Mitochondrial dysfunction activates regulatory pathways that induce changes in nuclear gene expression able to monitor the organelle status. - Mitochondrial function is connected to cell signaling pathways through changes in redox and phosphate pairs (NAD/NADH; CoQH2/CoQ and ATP/ADP vs. AMP), critical metabolite concentrations (succinate, aK-glutarate, AcetylCoA, etc.), and reactive oxygen species (ROS) production. - Variations in mitochondrial status modulate specific transcriptional programs. - The process that transduces the information required for the cell to adapt to a mitochondrial challenge is called “retrograde signaling”.

夽 This article is part of a Directed Issue entitled: Mitochondrial Diseases. Q2 ∗ Corresponding author at: Centro Nacional de Investigaciones Cardiovasculares Carlos III, Departamento de Desarrollo y Reparación Cardiovascular, Melchor Fernandez Almabro 3, 28029 Madrid, Spain. Tel.: +34 914531200; fax: +34 914531240. E-mail address: [email protected] (J.A. Enriquez).

23

Q3

Mitochondria are well known for their role of producing energy by oxidative phosphorylation, but these organelles also make critical contributions to other areas of cell metabolism, information flux (calcium, ROS, etc.) and cell-death cascades (apoptosis). The first clinical report of a genuine mitochondrial dysfunction was published by Rolf Luft in 1959 (Ernster et al., 1959). Since then, a number of diseases have been linked to mitochondrial dysfunction, and current figures show that 1 in every 5000 individuals are affected by a mitochondrial disorder (Pfeffer et al., 2012). The wide range of pathologies caused by mitochondrial dysfunction include lactic acidosis, skeletal myopathy, deafness, neurodegenerative diseases, muscular disorders, cardiomyopathy, diabetes and cancer (Vafai and Mootha, 2012). The varied symptoms and the involvement of multiple tissues make mitochondrial disorders hard to diagnose and treat, and diagnosis is best achieved by identifying the underlying genetic alteration. Mitochondrial dysfunction can alter cell signaling because of its primary role in synthesizing critical metabolites (NADH/NAD+ , ATP/ADP, ATP/AMP, succinate/a-ketoglutarate, etc.). Mitochondrial dysfunction can thus affect sirtuin-mediated signaling (NADH/NAD+ ), AMPK signaling (ATP/AMP), mTOR signaling (ATP/ADP) or Hif1a signaling (succinate/a-ketoglutarate), and can

http://dx.doi.org/10.1016/j.biocel.2015.01.026 1357-2725/© 2015 Published by Elsevier Ltd.

Please cite this article in press as: Cagin U, Enriquez JA. The complex crosstalk between mitochondria and the nucleus: What goes in between? Int J Biochem Cell Biol (2015), http://dx.doi.org/10.1016/j.biocel.2015.01.026

24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45

G Model BC 4551 1–6

U. Cagin, J.A. Enriquez / The International Journal of Biochemistry & Cell Biology xxx (2015) xxx–xxx

2 46 47 48 49 50 51

52

ARTICLE IN PRESS

also impact signaling mediated by reactive oxygen species (ROS) or Ca2+ (Jones et al., 2012). Depending on the cell type and type of mitochondrial dysfunction, one or several of these paths might be especially affected. Understanding the detailed signaling response to mitochondrial dysfunctions will help to generate a better understanding of the complex puzzle of mitochondrial disorders. 2. Organelle function

103

In addition to these signaling molecules, which monitor cell metabolic status and thus mitochondrial performance, information about mitochondrial quality is also transduced to the cell via specific mitochondria-derived pathways, among which the mitochondrial unfolding protein response (mtUPR) is gaining much attention (Mottis et al., 2014). It has also been suggested that mitochondria-to-nucleus retrograde signaling can be envisioned as a complementary mechanism to these mitochondrial quality control systems (Jazwinski, 2013). Changes in nuclear gene expression to maintain mitochondrial homeostasis are not limited to nuclear-encoded mitochondrial proteins. The retrograde response also changes the global nuclear gene expression pattern. Identifying the components of these changes was the first question to be addressed by research into mitochondria-nuclear interactions in the late 1980s. Communication between the mitochondrial and nuclear genomes was first postulated in 1987 in yeast (Parikh et al., 1987). The first factors implicated in retrograde signaling (including COX VI, CIT2 and RTG proteins) were also identified in yeast (Butow et al., 1988; Jia et al., 1997; Liao et al., 1991; Liu and Butow, 2006; Rothermel et al., 1997) (Table 1). Further analysis of Rtg proteins (Rtg1–3) showed that these proteins are normally present in the cytoplasm and translocate to nucleus upon retrograde signaling (Sekito et al., 2000). Subsequent studies in yeast also characterized more factors and also signaling pathways such as RAS2 and the TOR signaling pathway (Butow and Avadhani, 2004; Kirchman et al., 1999; Liu and Butow, 2006) (Table 1). The TOR pathway has also being linked to Rtg proteins in the context of retrograde signaling (Breitkreutz et al., 2010; Dilova et al., 2004; Dilova et al., 2002; Giannattasio et al., 2005; Komeili et al., 2000). Furthermore, yeast studies have shown that dysfunctional mitochondria inhibit TORC1 (Kawai et al., 2011). These studies were followed by others that examined retrograde signaling in more depth and also in other cell types and organisms. Understanding the conservation of the retrograde response between species is very important and therefore studies different organisms are important, especially regarding the differences in mitochondrial homeostasis between yeast and animal cells, particularly during mitochondrial biogenesis (Heddi et al., 1993; Jazwinski, 2013; Poyton and McEwen, 1996). In mammalian cells, the first proteins implicated in retrograde signaling (Cox-Va, b-actin, myc, GAPDH, EF-1) were characterized after the studies carried out in yeast (Marusich et al., 1997; Wang and Morais, 1997) (Table 1). Several studies subsequently revealed the diverse nature of mammalian retrograde signaling, including activation of NFAT, ATF2, NFKb, MAPK, PKC, Egr-1 and CEBP, and CamKIV-mediated activation of CREB (Amuthan et al., 2002; Arnould et al., 2002; Biswas et al., 1999, 2003; Butow and Avadhani, 2004). It should be noted that NFKb has roles in mitochondrial biogenesis and the response to ROS (Srinivasan et al., 2010).

104

2.1. Conservation and diversity in retrograde signaling

53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102

105 106

Several microarray analyses in yeast showed that mitochondriato-nucleus communication is not limited to a handful of factors

but is instead a global response alters the expression levels of different sets of genes depending on the type of mitochondrial dysfunction (Epstein et al., 2001; McCammon et al., 2003; Traven et al., 2001). Consistent with the yeast studies, the mTOR pathway has been implicated in retrograde signaling in mammalian cells, confirming the conserved nature of retrograde signaling between species (Komeili et al., 2000; Laplante and Sabatini, 2012). Although the retrograde signaling response has yet to be resolved in full, nuclear response elements commonly involved in mitochondrial dysfunction along with more specific ones have been identified. Genome-wide microarray transcriptome studies have been especially important in identifying genes that are differentially expressed upon mitochondrial dysfunction. These studies had performed using different cell lines; one of the earliest studies used a mtDNA depleted human breast cancer cell line (Delsite et al., 2002). However, there is no consensus in the published data. The type of mitochondrial dysfunction seems to have a strong influence on the observed pattern of nuclear gene expression. This has been shown by comparing the expression profiles of Rho0 (mtDNA-depleted) cells and cells carrying the A3243G mutation, which suggests partially distinct multiple pathways of retrograde signaling (Jahangir Tafrechi et al., 2005). Furthermore, RT-PCR and GeneChip microarray analyses consistently show non-identical gene expression patterns in different Rho0 cells lines (Miceli and Jazwinski, 2005; Mineri et al., 2009). 2.2. Key players in the transcriptional response Nonetheless, some signaling pathways are repeatedly reported to be involved in retrograde signaling: mTOR, NFKB, c-JUN/JNK and RXRA, NFATs, PKC, CamK IV, PI3K/AKT, Sirt1, No/CO (Jones et al., 2012). The importance of these pathways is further supported by the implication of defined transcription factors such as MYC, CREB, CEBP, FOXO-1, Egr-1, NRF-1, TFAM, ATF2, ctBP1, and PGC-1a/PRC (Jones et al., 2012).

107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131

132

133 134 135 136 137 138 139

3. Cell physiology

140

3.1. Implications for metabolism

141

Cells affected by mitochondrial dysfunction have to adapt to the sub-optimal energy metabolism. The most common adaptive response to decreased mitochondrial ATP production is increased glycolytic flux to lactate (von Kleist-Retzow et al., 2007). In yeast, the metabolic adaptation to an incomplete TCA cycle requires production of metabolites such as glutamate (Butow and Avadhani, 2004). Therefore cell survival in these circumstances can be helped by the use of alternative and more varied carbon sources. Common mechanisms observed in yeast are increased glyoxylate cycle activity and fatty acid ␤-oxidation (Jazwinski, 2013), mechanisms also triggered by nutrient deprivation (Wang et al., 2010). In addition, microarray studies of mtDNA-depleted cells revealed that TCA cycle and glyoxylate cycle genes are controlled similarly (Epstein et al., 2001). These metabolic changes in the switch from OXPHOS to glycolysis were corroborated by transcriptomic analysis in Drosophila melanogaster OXPHOS mutants (Freije et al., 2012). However, cells cannot depend solely on glyoxylate cycle for survival, and must therefore activate additional mechanisms. 3.2. Mitochondrial biogenesis, capacity and clearance The purpose of nuclear–mitochondrial cross-talk is to achieve a fine-tuned and functional mitochondrial population. The cell should have the required amount of healthy mitochondria and clear any unhealthy organelles. Mitochondrial quality control is the term usually used to describe the set of events and processes geared

Please cite this article in press as: Cagin U, Enriquez JA. The complex crosstalk between mitochondria and the nucleus: What goes in between? Int J Biochem Cell Biol (2015), http://dx.doi.org/10.1016/j.biocel.2015.01.026

142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159

160

161 162 163 164 165

G Model

ARTICLE IN PRESS

BC 4551 1–6

U. Cagin, J.A. Enriquez / The International Journal of Biochemistry & Cell Biology xxx (2015) xxx–xxx

3

Table 1 Historical outline of retrograde signaling in yeast and mammals.

Yeast

Mammalian

Significance

Genes & pathways identified

References

Discovery of first factors

RTGs, CIT1, CIT2, COX VI

First signaling pathways

RAS2, TOR

Parikh et al. (1987), Butow et al. (1988), Liao et al. (1991), Rothermel et al. (1997), Sekito et al. (2000) Kirchman et al. (1999), Komeili et al. (2000), Dilova et al. (2002), Butow and Avadhani (2004) Epstein et al. (2001), Traven et al. (2001), McCammon et al. (2003) Marusich et al. (1997), Wang and Morais (1997)

First transciptome studies Discovery of first factors First signaling pathways

␤-Actin, EF-1, myc, Cox-Va, SD Calcium signaling, CREB signaling, NFK␤

First transciptome studies

166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216

to achieving optimum mitochondrial homeostasis. The four main events in this process are biogenesis, mitophagy, fusion/fission and movement (MacAskill and Kittler, 2010; Scarpulla, 2011; Youle and Narendra, 2011; Chan, 2012). Mitochondrial biogenesis and fission have been shown to determine the capacity of cells to adapt to respiratory chain dysfunction, and nitric oxide has also been shown to be important for retrograde signaling (Benard et al., 2013). In addition, a major role in the clearance of accumulated proteins from mitochondria is played by the mtUPR (Martinus et al., 1996). The mtUPR is activated in Caenorhabditis elegans upon OXPHOS dysfunction (Haynes and Ron, 2010). Several transcription factors participate in mtUPR activation, including CHOP and CEBP, overlapping TFs associated with retrograde signaling (Horibe and Hoogenraad, 2007). Adjustments to mitochondrial biogenesis and capacity are also triggered by other kinds of cellular stress and upon changes in the metabolic environment. Mitochondrial biogenesis is regulated by nuclear-encoded proteins, transcription factors and co-activators (Scarpulla, 2006, 2011), but also maintains a degree of autonomy (Enriquez et al., 1999). The transcription factors NRF1, NRF2, ERR␣, CREB, and YY1 can regulate the expression of nDNA and, indirectly, mtDNA encoded mitochondrial genes, resulting in adjustments to mitochondrial function and oxidative capacity (Basu et al., 1997; Scarpulla, 2011; Schreiber et al., 2004; Vercauteren et al., 2006). In addition to TFs, an even more important role in mitochondrial biogenesis is played by transcriptional co-activators. The ‘master regulator’ of mitochondrial biogenesis is the widely studied transcriptional co-activator PGC1␣ (Lin et al., 2005; Scarpulla, 2012; Wenz, 2011). The known downstream TF targets of PGC1␣ are PPARs (regulators of fatty acid oxidation), NRF1 and ERR␣ (regulators of mitochondrial biogenesis) (Wenz, 2013). PGC1␣ has two homologues, PGC1B and PRC (PGC1 related co-activator), which have partially overlapping functions (Gleyzer and Scarpulla, 2011, 2013; Scarpulla, 2011). The three PGC factors are induced or activated in response to a variety of cellular stresses. PGC1␣ expression is regulated by other factors, including YY1, mTOR, CREB, PPARs, and p53 (Cunningham et al., 2007; Sahin et al., 2011; Scarpulla, 2008a,b). PGC1␣ activity can also be adjusted by post-translational modification by Akt, p38 MAPK, AMPK and Sirt1 (Canto and Auwerx, 2009; Fan et al., 2004). The three main metabolic changes that regulate PGC1␣ are caloric restriction, ROS and hypoxia (reviewed in detail in Wenz, 2013), with all three cases affecting mitochondrial performance. Regulation of mtDNA gene expression requires proteins encoded by nDNA. Nuclear-encoded TFs needed for mitochondrial transcription are POLRMT, TFB2M, TFAM and MTERFs (Arnold et al., 2012; Peralta et al., 2012). Mitochondrial transcripts are translated in the specific mitoribosomes (see Rorbach and Minczuk, 2012). Mammalian mtDNA encodes for only 13 proteins, all of which are subunits of OXPHOS complexes. All other OXPHOS protein subunit are encoded by nDNA. The co-assembly of OXPHOS subunits into

Biswas et al. (1999, 2003), Amuthan et al. (2001, 2002), Arnould et al. (2002) Delsite et al. (2002), Jahangir Tafrechi et al. (2005), Miceli and Jazwinski (2005)

complexes and supercomplexes is therefore a very intricate process that requires the action of dedicated chaperones (Ghezzi and Zeviani, 2012; Lapuente-Brun et al., 2013; McKenzie et al., 2011; Q4 Mick et al., 2010) (Fig. 1). 4. Organelle pathology The link between mitochondrial dysfunction and disease has led to intense research into the role of mito-nuclear signaling to develop potential treatments (Cerutti et al., 2014; Khan et al., 2014; Pirinen et al., 2014). A recent comparative analysis of tranciptomic data from different species (human, fly, mouse and worm) showed that mitochondrial dysfunction involves the dysregulation of conserved genes involved in the retrograde signaling response (Zhang et al., 2014). Two major human health problems that appear to be tightly linked to alterations in the mito-nuclear signaling are aging-related diseases and cancer (Reeve et al., 2008; Swerdlow, 2009). 4.1. Aging Lifespan studies in C. elegans suggest that mitochondrial-nuclear crosstalk is an important determiner of lifespan, with lifespan extended by increased glyoxylate cycle activity and knock-down of certain respiratory genes (Cristina et al., 2009; Lee et al., 2010; Vanfleteren and De Vreese, 1995). ROS, HIF1-␣ and mtUPR are implicated in these processes and also in the development of neurodegenerative disorders (Durieux et al., 2011; Houtkooper et al., 2013; Lee et al., 2010; Rugarli and Langer, 2012). Lifespan extension phenotypes have also been observed in D. melanogaster with respiratory chain dysfunction (Copeland et al., 2009; Liu et al., 2011), and the role of the mtUPR response has also been demonstrated in mice (Houtkooper et al., 2013). Loss of mitochondrial homeostasis is also linked to neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease (Lin et al., 2002; Simon et al., 2004). 4.2. Cancer A link between mitochondria and cancer has been proposed repeatedly. However, the contribution of mitochondrial dysfunction to invasiveness and transformation remains unresolved (Amuthan et al., 2001, 2002; Guha et al., 2007). Some studies suggest that the metastatic phenotype is affected by mtDNA mutations that lead to depletion of mtDNA as a consequence of increased ROS signaling (Ishikawa et al., 2008; Kulawiec et al., 2009). Retrograde signaling involving both well functioning and dysfunctional mitochondria, has been implicated in cancer. More recent work showed that a metastatic phenotype can result from overloading of the electron transport chain due to excess superoxide production (Porporato et al., 2014). Furthermore, PGC1␣ may also contribute to cancer progression (Jones et al., 2012). In some tumors linked

Please cite this article in press as: Cagin U, Enriquez JA. The complex crosstalk between mitochondria and the nucleus: What goes in between? Int J Biochem Cell Biol (2015), http://dx.doi.org/10.1016/j.biocel.2015.01.026

217 218 219 220

221

222 223 224 225 226 227 228 229 230 231 232

233

234 235 236 237 238 239 240 241 242 243 244 245 246 247

248

249 250 251 252 253 254 255 256 257 258 259 260 261

G Model BC 4551 1–6

ARTICLE IN PRESS U. Cagin, J.A. Enriquez / The International Journal of Biochemistry & Cell Biology xxx (2015) xxx–xxx

4

Fig. 1. Overview of retrograde signaling. There are at least four types of mitochondrial dysfunction, which lead to the generation of at least five types of signal: ATP/AMP, NADH/NAD+ , ROS, cytosolic Ca2+ , reduction in membrane potential ( ). Activation of signaling (red arrows) triggers changes in nuclear gene expression that promote diverse responses represented by purple arrows

262 263 264

265

266 267 268 269 270 271 272 273

274 Q5

275

276

to mtDNA mutations and associated mitochondrial defects, mitochondrial biogenesis is upregulated in an apparently compensatory mechanism (Gasparre et al., 2011)). 5. Future outlook A comprehensive knowledge of the interconnected roles of mitochondrial quality control and mito-nuclear signaling will provide the key to a better understanding of human health disorders in which mitochondrial dysfunction plays role, from mitochondrial diseases to metabolic syndrome and degenerative diseases. This knowledge can be expected to pave the way to new treatments to achieve better quality of life for people affected by these conditions. Uncited references Laplante, 2012. Acknowledgements

We thank Dr. Concepción Jimenez for technical assistance. 278 Q6 This study was supported by grants from the Ministerio de 279 Economía y Competitividad (SAF2012-1207 & CSD2007-00020), 280 the Comunidad de Madrid (CAM/API1009), the EU (Mitochondrial 281 European Educational Training, MEET: European Commission Sev282 enth Framework Programme, FP7-PEOPLE-2012-ITN MARIE CURIE, 283 grant agreement No. 317433) and the Instituto de Salud Carlos III 284 (FIS grant PI11-00078). The CNIC is supported by the Ministerio de 285 Q7 Economía y Competitividad and the Pro-CNIC Foundation. 277

286

287 288 289 290

References 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;21:7839–49.

Amuthan G, Biswas G, Zhang SY, Klein-Szanto A, Vijayasarathy C, Avadhani NG. Mitochondria-to-nucleus stress signaling induces phenotypic changes, tumor progression and cell invasion. EMBO J 2001;20:1910–20. Arnold JJ, Smidansky ED, Moustafa IM, Cameron CE. Human mitochondrial RNA polymerase: structure-function, mechanism and inhibition. Biochim Biophys Acta 2012;1819:948–60. Arnould T, Vankoningsloo S, Renard P, Houbion A, Ninane N, Demazy C, et al. CREB activation induced by mitochondrial dysfunction is a new signaling pathway that impairs cell proliferation. EMBO J 2002;21:53–63. Basu A, Lenka N, Mullick J, Avadhani NG. Regulation of murine cytochrome oxidase Vb gene expression in different tissues and during myogenesis. Role of a YY-1 factor-binding negative enhancer. J Biol Chem 1997;272:5899–908. Benard G, Trian T, Bellance N, Berger P, Lavie J, Espil-Taris C, et al. Adaptative capacity of mitochondrial biogenesis and of mitochondrial dynamics in response to pathogenic respiratory chain dysfunction. Antioxid Redox Signal 2013;19:350–65. Biswas G, Adebanjo OA, Freedman BD, Anandatheerthavarada HK, Vijayasarathy C, Zaidi M, 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 J 1999;18:522–33. Biswas G, Anandatheerthavarada HK, Zaidi M, Avadhani NG. Mitochondria to nucleus stress signaling: a distinctive mechanism of NFkappaB/Rel activation through calcineurin-mediated inactivation of IkappaBbeta. J Cell Biol 2003;161:507–19. Breitkreutz A, Choi H, Sharom JR, Boucher L, Neduva V, Larsen B, et al. A global protein kinase and phosphatase interaction network in yeast. Science 2010;328:1043–6. Butow RA, Avadhani NG. Mitochondrial signaling: the retrograde response. Mol Cell 2004;14:1–15. Butow RA, Docherty R, Parikh VS. A path from mitochondria to the yeast nucleus. Philos Trans R Soc Lond B: Biol Sci 1988;319:127–33. Canto C, Auwerx J. PGC-1alpha, SIRT1 and AMPK, an energy sensing network that controls energy expenditure. Curr Opin Lipidol 2009;20:98–105. Cerutti R, Pirinen E, Lamperti C, Marchet S, Sauve AA, Li W, et al. NAD(+)-dependent activation of Sirt1 corrects the phenotype in a mouse model of mitochondrial disease. Cell Metab 2014;19:1042–9. Chan DC. Fusion and fission: interlinked processes critical for mitochondrial health. Annu Rev Genet 2012;46:265–87. Copeland JM, Cho J, Lo T Jr, Hur JH, Bahadorani S, Arabyan T, et al. Extension of Drosophila life span by RNAi of the mitochondrial respiratory chain. Curr Biol: CB 2009;19:1591–8. Cristina D, Cary M, Lunceford A, Clarke C, Kenyon C. A regulated response to impaired respiration slows behavioral rates and increases lifespan in Caenorhabditis elegans. PLoS Genet 2009;5:e1000450. Cunningham JT, Rodgers JT, Arlow DH, Vazquez F, Mootha VK, Puigserver P. mTOR controls mitochondrial oxidative function through a YY1-PGC-1alpha transcriptional complex. Nature 2007;450:736–40. Delsite R, Kachhap S, Anbazhagan R, Gabrielson E, Singh KK. Nuclear genes involved in mitochondria-to-nucleus communication in breast cancer cells. Mol Cancer 2002;1:6.

Please cite this article in press as: Cagin U, Enriquez JA. The complex crosstalk between mitochondria and the nucleus: What goes in between? Int J Biochem Cell Biol (2015), http://dx.doi.org/10.1016/j.biocel.2015.01.026

291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339

G Model BC 4551 1–6

ARTICLE IN PRESS U. Cagin, J.A. Enriquez / The International Journal of Biochemistry & Cell Biology xxx (2015) xxx–xxx

340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425

Dilova I, Aronova S, Chen JC, Powers T. Tor signaling and nutrient-based signals converge on Mks1p phosphorylation to regulate expression of Rtg1.Rtg3pdependent target genes. J Biol Chem 2004;279:46527–35. Dilova I, Chen CY, Powers T. Mks1 in concert with TOR signaling negatively regulates RTG target gene expression in S. cerevisiae. Curr Biol: CB 2002;12:389–95. Durieux J, Wolff S, Dillin A. The cell-non-autonomous nature of electron transport chain-mediated longevity. Cell 2011;144:79–91. Enriquez JA, Fernandez-Silva P, Montoya J. Autonomous regulation in mammalian mitochondrial DNA transcription. Biol Chem 1999;380:737–47. Epstein CB, Waddle JA, Hale Wt Dave V, Thornton J, Macatee TL, et al. Genome-wide responses to mitochondrial dysfunction. Mol Biol Cell 2001;12:297–308. Ernster L, Ikkos D, Luft R. Enzymic activities of human skeletal muscle mitochondria: a tool in clinical metabolic research. Nature 1959;184:1851–4. Fan M, Rhee J, St-Pierre J, Handschin C, Puigserver P, Lin J, et al. Suppression of mitochondrial respiration through recruitment of p160 myb binding protein to PGC-1alpha: modulation by p38 MAPK. Genes Dev 2004;18:278–89. Freije WA, Mandal S, Banerjee U. Expression profiling of attenuated mitochondrial function identifies retrograde signals in Drosophila. G3 2012;2: 843–51. Gasparre G, Romeo G, Rugolo M, Porcelli AM. Learning from oncocytic tumors: why choose inefficient mitochondria. Biochim Biophys Acta 2011;1807:633–42. Ghezzi D, Zeviani M. Assembly factors of human mitochondrial respiratory chain complexes: physiology and pathophysiology. Adv Exp Med Biol 2012;748:65–106. Giannattasio S, Liu Z, Thornton J, Butow RA. Retrograde response to mitochondrial dysfunction is separable from TOR1/2 regulation of retrograde gene expression. J Biol Chem 2005;280:42528–35. Gleyzer N, Scarpulla RC. PGC-1-related coactivator (PRC), a sensor of metabolic stress, orchestrates a redox-sensitive program of inflammatory gene expression. J Biol Chem 2011;286:39715–25. Gleyzer N, Scarpulla RC. Activation of a PGC-1-related coactivator (PRC)-dependent inflammatory stress program linked to apoptosis and premature senescence. J Biol Chem 2013;288:8004–15. Guha M, Srinivasan S, Biswas G, Avadhani NG. Activation of a novel calcineurinmediated insulin-like growth factor-1 receptor pathway, altered metabolism, and tumor cell invasion in cells subjected to mitochondrial respiratory stress. J Biol Chem 2007;282:14536–46. Haynes CM, Ron D. The mitochondrial UPR-protecting organelle protein homeostasis. J Cell Sci 2010;123:3849–55. Heddi A, Lestienne P, Wallace DC, Stepien G. Mitochondrial DNA expression in mitochondrial myopathies and coordinated expression of nuclear genes involved in ATP production. J Biol Chem 1993;268:12156–63. Horibe T, Hoogenraad NJ. The chop gene contains an element for the positive regulation of the mitochondrial unfolded protein response. PLoS ONE 2007;2:e835. Houtkooper RH, Mouchiroud L, Ryu D, Moullan N, Katsyuba E, Knott G, et al. Mitonuclear protein imbalance as a conserved longevity mechanism. Nature 2013;497:451–7. Ishikawa K, Hashizume O, Koshikawa N, Fukuda S, Nakada K, Takenaga K, et al. Enhanced glycolysis induced by mtDNA mutations does not regulate metastasis. FEBS Lett 2008;582:3525–30. Jahangir Tafrechi RS, Svensson PJ, Janssen GM, Szuhai K, Maassen JA, Raap AK. Distinct nuclear gene expression profiles in cells with mtDNA depletion and homoplasmic A3243G mutation. Mutat Res 2005;578:43–52. Jazwinski SM. The retrograde response: when mitochondrial quality control is not enough. Biochim Biophys Acta 2013;1833:400–9. Jia Y, Rothermel B, Thornton J, Butow RA. A basic helix-loop-helix-leucine zipper transcription complex in yeast functions in a signaling pathway from mitochondria to the nucleus. Mol Cell Biol 1997;17:1110–7. Jones AW, Yao Z, Vicencio JM, Karkucinska-Wieckowska A, Szabadkai G. PGC-1 family coactivators and cell fate: roles in cancer, neurodegeneration, cardiovascular disease and retrograde mitochondria-nucleus signalling. Mitochondrion 2012;12:86–99. Kawai S, Urban J, Piccolis M, Panchaud N, De Virgilio C, Loewith R. Mitochondrial genomic dysfunction causes dephosphorylation of Sch9 in the yeast Saccharomyces cerevisiae. Eukaryot Cell 2011;10:1367–9. Khan NA, Auranen M, Paetau I, Pirinen E, Euro L, Forsstrom S, et al. Effective treatment of mitochondrial myopathy by nicotinamide riboside, a vitamin B3. EMBO Mol Med 2014;6:721–31. Kirchman PA, Kim S, Lai CY, Jazwinski SM. Interorganelle signaling is a determinant of longevity in Saccharomyces cerevisiae. Genetics 1999;152:179–90. Komeili A, Wedaman KP, O’Shea EK, Powers T. Mechanism of metabolic control. Target of rapamycin signaling links nitrogen quality to the activity of the Rtg1 and Rtg3 transcription factors. J Cell Biol 2000;151:863–78. Kulawiec M, Owens KM, Singh KK. Cancer cell mitochondria confer apoptosis resistance and promote metastasis. Cancer Biol Ther 2009;8:1378–85. Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell 2012;149:274–93. Lapuente-Brun E, Moreno-Loshuertos R, Acin-Perez R, Latorre-Pellicer A, Colas C, Balsa E, et al. Supercomplex assembly determines electron flux in the mitochondrial electron transport chain. Science 2013;340:1567–70. Lee SJ, Hwang AB, Kenyon C. Inhibition of respiration extends C. elegans life span via reactive oxygen species that increase HIF-1 activity. Curr Biol: CB 2010;20:2131–6. Liao XS, Small WC, Srere PA, Butow RA. Intramitochondrial functions regulate nonmitochondrial citrate synthase (CIT2) expression in Saccharomyces cerevisiae. Mol Cell Biol 1991;11:38–46.

5

Lin J, Handschin C, Spiegelman BM. Metabolic control through the PGC-1 family of transcription coactivators. Cell Metab 2005;1:361–70. Lin MT, Simon DK, Ahn CH, Kim LM, Beal MF. High aggregate burden of somatic mtDNA point mutations in aging and Alzheimer’s disease brain. Hum Mol Genet 2002;11:133–45. Liu J, Wu Q, He D, Ma T, Du L, Dui W, et al. Drosophila sbo regulates lifespan through its function in the synthesis of coenzyme Q in vivo. J Genet Genomics 2011;38:225–34. Liu Z, Butow RA. Mitochondrial retrograde signaling. Annu Rev Genet 2006;40:159–85. MacAskill AF, Kittler JT. Control of mitochondrial transport and localization in neurons. Trends Cell Biol 2010;20:102–12. Martinus RD, Garth GP, Webster TL, Cartwright P, Naylor DJ, Hoj PB, et al. Selective induction of mitochondrial chaperones in response to loss of the mitochondrial genome. Eur J Biochem 1996;240:98–103, FEBS. Marusich MF, Robinson BH, Taanman JW, Kim SJ, Schillace R, Smith JL, et al. Expression of mtDNA and nDNA encoded respiratory chain proteins in chemically and genetically-derived Rho0 human fibroblasts: a comparison of subunit proteins in normal fibroblasts treated with ethidium bromide and fibroblasts from a patient with mtDNA depletion syndrome. Biochim Biophys Acta 1997;1362:145–59. McCammon MT, Epstein CB, Przybyla-Zawislak B, McAlister-Henn L, Butow RA. Global transcription analysis of Krebs tricarboxylic acid cycle mutants reveals an alternating pattern of gene expression and effects on hypoxic and oxidative genes. Mol Biol Cell 2003;14:958–72. McKenzie M, Tucker EJ, Compton AG, Lazarou M, George C, Thorburn DR, et al. Mutations in the gene encoding C8orf38 block complex I assembly by inhibiting production of the mitochondria-encoded subunit ND1. J Mol Biol 2011;414:413–26. Miceli MV, Jazwinski SM. Nuclear gene expression changes due to mitochondrial dysfunction in ARPE-19 cells: implications for age-related macular degeneration. Investig Ophthalmol Vis Sci 2005;46:1765–73. Mick DU, Vukotic M, Piechura H, Meyer HE, Warscheid B, Deckers M, et al. Coa3 and Cox14 are essential for negative feedback regulation of COX1 translation in mitochondria. J Cell Biol 2010;191:141–54. Mineri R, Pavelka N, Fernandez-Vizarra E, Ricciardi-Castagnoli P, Zeviani M, Tiranti V. How do human cells react to the absence of mitochondrial DNA? PLoS ONE 2009;4:e5713. Mottis A, Jovaisaite V, Auwerx J. The mitochondrial unfolded protein response in mammalian physiology. Mamm Genome: Off J Int Mamm Genome Soc 2014;25:424–33. Parikh VS, Morgan MM, Scott R, Clements LS, Butow RA. The mitochondrial genotype can influence nuclear gene expression in yeast. Science 1987;235:576–80. Peralta S, Wang X, Moraes CT. Mitochondrial transcription: lessons from mouse models. Biochim Biophys Acta 2012;1819:961–9. Pfeffer G, Majamaa K, Turnbull DM, Thorburn D, Chinnery PF. Treatment for mitochondrial disorders. Cochrane Database Syst Rev 2012;4:CD004426. Pirinen E, Canto C, Jo YS, Morato L, Zhang H, Menzies KJ, et al. Pharmacological Inhibition of poly(ADP-ribose) polymerases improves fitness and mitochondrial function in skeletal muscle. Cell Metab 2014;19:1034–41. Porporato PE, Payen VL, Perez-Escuredo J, De Saedeleer CJ, Danhier P, Copetti T, et al. A mitochondrial switch promotes tumor metastasis. Cell Rep 2014;8:754–66. Poyton RO, McEwen JE. Crosstalk between nuclear and mitochondrial genomes. Annu Rev Biochem 1996;65:563–607. Reeve AK, Krishnan KJ, Turnbull DM. Age related mitochondrial degenerative disorders in humans. Biotechnol J 2008;3:750–6. Rorbach J, Minczuk M. The post-transcriptional life of mammalian mitochondrial RNA. Biochem J 2012;444:357–73. Rothermel BA, Thornton JL, Butow RA. Rtg3p, a basic helix-loop-helix/leucine zipper protein that functions in mitochondrial-induced changes in gene expression, contains independent activation domains. J Biol Chem 1997;272:19801–7. Rugarli EI, Langer T. Mitochondrial quality control: a matter of life and death for neurons. EMBO J 2012;31:1336–49. Sahin E, Colla S, Liesa M, Moslehi J, Muller FL, Guo M, et al. Telomere dysfunction induces metabolic and mitochondrial compromise. Nature 2011;470: 359–65. Scarpulla RC. Nuclear control of respiratory gene expression in mammalian cells. J Cell Biochem 2006;97:673–83. Scarpulla RC. Nuclear control of respiratory chain expression by nuclear respiratory factors and PGC-1-related coactivator. Ann N Y Acad Sci 2008a;1147:321–34. Scarpulla RC. Transcriptional paradigms in mammalian mitochondrial biogenesis and function. Physiol Rev 2008b;88:611–38. Scarpulla RC. Metabolic control of mitochondrial biogenesis through the PGC-1 family regulatory network. Biochim Biophys Acta 2011;1813:1269–78. Scarpulla RC. Nucleus-encoded regulators of mitochondrial function: integration of respiratory chain expression, nutrient sensing and metabolic stress. Biochim Biophys Acta 2012;1819:1088–97. Schreiber SN, Emter R, Hock MB, Knutti D, Cardenas J, Podvinec M, et al. The estrogen-related receptor alpha (ERRalpha) functions in PPARgamma coactivator 1alpha (PGC-1alpha)-induced mitochondrial biogenesis. Proc Natl Acad Sci USA 2004;101:6472–7. Sekito T, Thornton J, Butow RA. Mitochondria-to-nuclear signaling is regulated by the subcellular localization of the transcription factors Rtg1p and Rtg3p. Mol Biol Cell 2000;11:2103–15. Simon DK, Lin MT, Zheng L, Liu GJ, Ahn CH, Kim LM, et al. Somatic mitochondrial DNA mutations in cortex and substantia nigra in aging and Parkinson’s disease. Neurobiol Aging 2004;25:71–81.

Please cite this article in press as: Cagin U, Enriquez JA. The complex crosstalk between mitochondria and the nucleus: What goes in between? Int J Biochem Cell Biol (2015), http://dx.doi.org/10.1016/j.biocel.2015.01.026

426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511

G Model BC 4551 1–6 6 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527

ARTICLE IN PRESS U. Cagin, J.A. Enriquez / The International Journal of Biochemistry & Cell Biology xxx (2015) xxx–xxx

Srinivasan V, Kriete A, Sacan A, Jazwinski SM. Comparing the yeast retrograde response and NF-kappaB stress responses: implications for aging. Aging Cell 2010;9:933–41. Swerdlow RH. The neurodegenerative mitochondriopathies. J Alzheimers Dis: JAD 2009;17:737–51. Traven A, Wong JM, Xu D, Sopta M, Ingles CJ. Interorganellar communication. Altered nuclear gene expression profiles in a yeast mitochondrial DNA mutant. J Biol Chem 2001;276:4020–7. Vafai SB, Mootha VK. Mitochondrial disorders as windows into an ancient organelle. Nature 2012;491:374–83. Vanfleteren JR, De Vreese A. The gerontogenes age-1 and daf-2 determine metabolic rate potential in aging Caenorhabditis elegans. FASEB J: Off Publ Fed Am Soc Exp Biol 1995;9:1355–61. Vercauteren K, Pasko RA, Gleyzer N, Marino VM, Scarpulla RC. PGC-1-related coactivator: immediate early expression and characterization of a CREB/NRF-1 binding domain associated with cytochrome c promoter occupancy and respiratory growth. Mol Cell Biol 2006;26:7409–19.

von Kleist-Retzow JC, Hornig-Do HT, Schauen M, Eckertz S, Dinh TA, Stassen F, et al. Impaired mitochondrial Ca2+ homeostasis in respiratory chain-deficient cells but efficient compensation of energetic disadvantage by enhanced anaerobic glycolysis due to low ATP steady state levels. Exp Cell Res 2007;313:3076–89. Wang H, Morais R. Up-regulation of nuclear genes in response to inhibition of mitochondrial DNA expression in chicken cells. Biochim Biophys Acta 1997;1352:325–34. Wang J, Jiang JC, Jazwinski SM. Gene regulatory changes in yeast during life extension by nutrient limitation. Exp Gerontol 2010;45:621–31. Wenz T. Mitochondria and PGC-1alpha in aging and age-associated diseases. J Aging Res 2011;2011:810619. Wenz T. Regulation of mitochondrial biogenesis and PGC-1alpha under cellular stress. Mitochondrion 2013;13:134–42. Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol 2011;12:9–14. Zhang Z, Hailat Z, Falk MJ, Chen XW. Integrative analysis of independent transcriptome data for rare diseases. Methods 2014;69:315–25.

Please cite this article in press as: Cagin U, Enriquez JA. The complex crosstalk between mitochondria and the nucleus: What goes in between? Int J Biochem Cell Biol (2015), http://dx.doi.org/10.1016/j.biocel.2015.01.026

528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544

The complex crosstalk between mitochondria and the nucleus: What goes in between?

Mitochondria are critical metabolic hubs in which catabolic and anabolic cellular processes converge and are integrated. To perform their function, mi...
699KB Sizes 4 Downloads 7 Views