Accessibility / Report Error

Maternal transmission of mitochondrial diseases

Abstract

Given the major role of the mitochondrion in cellular homeostasis, dysfunctions of this organelle may lead to several common diseases in humans. Among these, maternal diseases linked to mitochondrial DNA (mtDNA) mutations are of special interest due to the unclear pattern of mitochondrial inheritance. Multiple copies of mtDNA are present in a cell, each encoding for 37 genes essential for mitochondrial function. In cases of mtDNA mutations, mitochondrial malfunctioning relies on mutation load, as mutant and wild-type molecules may co-exist within the cell. Since the mutation load associated with disease manifestation varies for different mutations and tissues, it is hard to predict the progeny phenotype based on mutation load in the progenitor. In addition, poorly understood mechanisms act in the female germline to prevent the accumulation of deleterious mtDNA in the following generations. In this review, we outline basic aspects of mitochondrial inheritance in mammals and how they may lead to maternally-inherited diseases. Furthermore, we discuss potential therapeutic strategies for these diseases, which may be used in the future to prevent their transmission.

Keywords:
Oocyte; germline; mitochondrial dynamics; mtDNA; metabolism

Introduction

The mitochondrion gained its deserved reputation in cell biology due to its role as the cellular powerhouse, with most of the adenosine triphosphate (ATP) in eukaryotic cells being supplied by this organelle (Wallace, 2013Wallace DC (2013) Bioenergetics in human evolution and disease: implications for the origins of biological complexity and the missing genetic variation of common diseases. Philos Trans R Soc Lond B Biol Sci 368:20120267.). However, mitochondria play several functions in the cell that far exceed the role in ATP generation. These are linked with buffering of Ca+2 levels, innate immunity, apoptosis and biogenesis of iron-sulfur clusters (Yasukawa et al., 2009Yasukawa K, Oshiumi H, Takeda M, Ishihara N, Yanagi Y, Seya T, Kawabata S and Koshiba T (2009) Mitofusin 2 inhibits mitochondrial antiviral signaling. Sci Signal 2:ra47.; Naon and Scorrano 2014Naon D and Scorrano L (2014) At the right distance: ER-mitochondria juxtaposition in cell life and death. Biochim Biophys Acta 1843:2184–2194.; Stehling et al., 2014Stehling O, Wilbrecht C and Lill R (2014) Mitochondrial iron-sulfur protein biogenesis and human disease. Biochimie 100:61-77.). Moreover, mitochondria closely interact with other organelles such as the endoplasmic reticulum (ER) and regulate several pathways in the cell (de Brito and Scorrano, 2009de Brito OM and Scorrano L (2009) Mitofusin-2 regulates mitochondrial and endoplasmic reticulum morphology and tethering: The role of Ras. Mitochondrion 9:222-226.; Betz et al., 2013Betz C, Stracka D, Prescianotto-baschong C, Frieden M and Demaurex N (2013) mTOR complex 2-Akt signaling at mitochondria-associated endoplasmic reticulum membranes (MAM) regulates mitochondrial physiology. Proc Nat Acad Sci U S A 110:12526-12534.; Chen et al., 2014Chen KH, Dasgupta A, Ding J, Indig FE, Ghosh P and Longo DL (2014) Role of mitofusin 2 (Mfn2) in controlling cellular proliferation. FASEB J 28:382-394.; Carreras-Sureda et al., 2017Carreras-Sureda A, Pihán P and Hetz C (2017) The unfolded protein response: At the intersection between endoplasmic reticulum function and mitochondrial bioenergetics. Front Oncol 7:1-7.; Xu et al., 2017Xu K, Chen G, Li X, Wu X, Chang Z, Xu J, Zhu Y, Yin P, Liang X and Dong L (2017) MFN2 suppresses cancer progression through inhibition of mTORC2/Akt signaling. Sci Rep 7:41718.). As a result, perturbations in mitochondrial function may dramatically disturb cellular homeostasis, resulting in several common diseases in humans (Bach et al., 2003Bach D, Pich S, Soriano FX, Vega N, Baumgartner B, Oriola J, Daugaard JR, Lloberas J, Camps M, Zierath JR et al. (2003) Mitofusin-2 determines mitochondrial network architecture and mitochondrial metabolism: A novel regulatory mechanism altered in obesity. J Biol Chem 278:17190-17197.; Chen et al., 2007Chen H, McCaffery JM and Chan DC (2007) Mitochondrial fusion protects against neurodegeneration in the cerebellum. Cell 130:548-62., 2010Chen H, Vermulst M, Wang YE, Chomyn A, Prolla TA, McCaffery JM and Chan DC (2010) Mitochondrial fusion is required for mtDNA stability in skeletal muscle and tolerance of mtDNA mutations. Cell 141:280-9.; Schaefer et al., 2008Schaefer AM, McFarland R, Blakely EL, He L, Whittaker RG, Taylor RW, Chinnery PF and Turnbull DM (2008) Prevalence of mitochondrial DNA disease in adults. Ann Neurol 63:35-9.; Misko et al., 2012Misko AL, Sasaki Y, Tuck E, Milbrandt J and Baloh RH (2012) Mitofusin2 mutations disrupt axonal mitochondrial positioning and promote axon degeneration. J Neurosci 32:4145–4155.; Schon et al., 2012Schon EA, DiMauro S and Hirano M (2012) Human mitochondrial DNA: Roles of inherited and somatic mutations. Nat Rev Genet 13:878-90.; Sebastian et al., 2012Sebastian D, Hernandez-Alvarez MI, Segales J, Sorianello E, Munoz JP, Sala D, Waget A, Liesa M, Paz JC, Gopalacharyulu P et al. (2012) Mitofusin 2 (Mfn2) links mitochondrial and endoplasmic reticulum function with insulin signaling and is essential for normal glucose homeostasis. Proc Natl Acad Sci U S A 109:5523-5528.; Eschbach et al., 2013Eschbach J, Sinniger J, Bouitbir J, Fergani A, Schlagowski A-I, Zoll J, Geny B, René F, Larmet Y, Marion V et al. (2013) Dynein mutations associated with hereditary motor neuropathies impair mitochondrial morphology and function with age. Neurobiol Dis 58:220-30.; Payne et al., 2013Payne BAI, Wilson IJ, Yu-Wai-Man P, Coxhead J, Deehan D, Horvath R, Taylor RW, Samuels DC, Santibanez-Koref M and Chinnery PF (2013) Universal heteroplasmy of human mitochondrial DNA. Hum Mol Genet 22:384–390.; Schneeberger et al., 2013Schneeberger M, Dietrich MO, Sebastián D, Imbernón M, Castaño C, Garcia A, Esteban Y, Gonzalez-Franquesa A, Rodríguez IC, Bortolozzi A et al. (2013) Mitofusin 2 in POMC neurons connects ER stress with leptin resistance and energy imbalance. Cell 155:172-187.; Pareyson et al., 2015Pareyson D, Saveri P, Sagnelli A and Piscosquito G (2015) Mitochondrial dynamics and inherited peripheral nerve diseases. Neurosci Lett 596:66–77.; Ramírez et al., 2017Ramírez S, Gómez-Valadés AG, Schneeberger M, Varela L, Haddad-Tóvolli R, Altirriba J, Noguera E, Drougard A, Flores-Martínez Á, Imbernón M et al. (2017) Mitochondrial dynamics mediated by Mitofusin 1 is required for POMC neuron glucose-sensing and insulin release control. Cell Metab 25:1390-1399.e6.).

Amongst mitochondria-associated diseases, those primarily linked to mitochondrial DNA (mtDNA) mutations have been a topic of great interest given their severe outcome and unclear pattern of inheritance (Craven et al., 2017Craven L, Alston CL, Taylor RW and Turnbull DM (2017) Recent advances in mitochondrial disease. Annu Rev Genomics Hum Genet 18:257-275.). However, mtDNA mutations can also associate with nuclear mutations, leading to common diseases in humans such as cancer, diabetes, Alzheimer, and Parkinson (Wallace 2011Wallace DC (2011) Bioenergetic origins of complexity and disease. Cold Spring Harb Symp Quantit Biol 76:1-16.; Schon et al., 2012Schon EA, DiMauro S and Hirano M (2012) Human mitochondrial DNA: Roles of inherited and somatic mutations. Nat Rev Genet 13:878-90.; Stewart and Chinnery 2015Stewart JB and Chinnery PF (2015) The dynamics of mitochondrial DNA heteroplasmy: Implications for human health and disease. Nat Rev Genet 16:530-42.). Thereby, recent findings have associated obesity with mitochondrial dysfunction in oocytes and increased risk of metabolic diseases in offspring (Wu et al., 2015Wu LL, Russell DL, Wong SL, Chen M, Tsai TS, St John JC, Norman RJ, Febbraio MA, Carroll J and Robker RL (2015) Mitochondrial dysfunction in oocytes of obese mothers: transmission to offspring and reversal by pharmacological endoplasmic reticulum stress inhibitors. Development 142:681-691.; Saben et al., 2016Saben JL, Boudoures AL, Asghar Z, Cusumano A, Scheaffer S, Moley KH, Saben JL, Boudoures AL, Asghar Z, Thompson A et al. (2016) Mitochondrial dysfunction via germline changes across three generations maternal metabolic syndrome programs mitochondrial dysfunction via germline changes across three generations. Cell Rep 16:1-8.). In mammals, mitochondria are uniparentally transmitted by females (Sutovsky et al., 1999Sutovsky P, Moreno RD, Ramalho-Santos J, Dominko T, Simerly C and Schatten G (1999) Ubiquitin tag for sperm mitochondria. Nature 402:371-372.). Thus, maternal mitochondria are replicated during early embryogenesis to colonize somatic and germline tissues (St John, 2019St John JC (2019) Mitochondria and female germline stem cells - a mitochondrial DNA perspective. Cells 8:852.). As a result, mitochondrial abnormalities present in oocytes can be perpetuated and lead to disease in offspring (Payne et al., 2013Payne BAI, Wilson IJ, Yu-Wai-Man P, Coxhead J, Deehan D, Horvath R, Taylor RW, Samuels DC, Santibanez-Koref M and Chinnery PF (2013) Universal heteroplasmy of human mitochondrial DNA. Hum Mol Genet 22:384–390.; Saben et al., 2016Saben JL, Boudoures AL, Asghar Z, Cusumano A, Scheaffer S, Moley KH, Saben JL, Boudoures AL, Asghar Z, Thompson A et al. (2016) Mitochondrial dysfunction via germline changes across three generations maternal metabolic syndrome programs mitochondrial dysfunction via germline changes across three generations. Cell Rep 16:1-8.; Craven et al., 2017Craven L, Alston CL, Taylor RW and Turnbull DM (2017) Recent advances in mitochondrial disease. Annu Rev Genomics Hum Genet 18:257-275.; Wei et al., 2019Wei W, Tuna S, Keogh MJ, Smith KR, Aitman TJ, Beales PL, Bennett DL, Gale DP, Bitner-Glindzicz MAK, Black GC et al. (2019) Germline selection shapes human mitochondrial DNA diversity. Science 364:eaau6520.). In this review, we outline basic aspects of mitochondrial transmission in mammalian germline and how they may lead to maternally inherited diseases. Furthermore, we discuss potential therapeutic strategies for these diseases, which may be used in the future to prevent their transmission.

Basic aspects of mitochondria

Mitochondria are double-membrane organelles with two distinct compartments, the inter-membrane space and the matrix. Most enzymes taking part in oxidative phosphorylation of energetic molecules (i.e., sugars, fats and proteins), including those of the Krebs cycle, are located in the mitochondrial matrix. The energy extracted from these molecules is then used by three (I, III and IV) out of four complexes imbedded in the inner mitochondrial membrane to pump H+ from the matrix to the inter-membrane space. This creates a difference in electric potential (the mitochondrial membrane potential – ΔΨm). In turn, a fifth complex (V) phosphorylates ADP into ATP using the electrochemical energy derived from the H+ return to the matrix.

Mitochondria harbor their own genome, the mtDNA, which in mammals is ~16.5-kb long and encodes for 13 mRNAs, 2 rRNAs, and 22 tRNAs. These genes are essential for ATP synthesis in mitochondria as the 13 mtDNA-encoded proteins play key roles in complexes I, III, IV, and V of the electron transport chain. However, nearly 1,200 different proteins are present in mitochondria (i.e., complexes I to V are composed of ~80 proteins), most of which are encoded in the nucleus, translated in the cytoplasm and imported by mitochondria. Proteins regulating mtDNA replication, transcription and repair are similarly derived from the nucleus. Therefore, although mtDNA-encoded proteins are essential for ATP production in mitochondria, the nucleus exerts a broader role in regulating mitochondrial function (Garesse and Vallejo, 2001Garesse R and Vallejo CG (2001) Animal mitochondrial biogenesis and function: A regulatory cross-talk between two genomes. Gene 263:1-16.; Scarpulla, 2002Scarpulla RC (2002) Transcriptional activators and coactivators in the nuclear control of mitochondrial function in mammalian cells. Gene 286:81-9.; Battersby et al., 2003Battersby BJ, Loredo-Osti JC and Shoubridge EA (2003) Nuclear genetic control of mitochondrial DNA segregation. Nat Genet 33:183-186.).

Hundreds to thousands of mitochondria are present in each cell (Wassarman and Josefowicz, 1978Wassarman PM and Josefowicz WJ (1978) Oocyte development in the mouse: An ultrastructural comparison of oocytes isolated at various stages of growth and meiotic competence. J Morphol 156:209-235.; Jansen and De Boer, 1998Jansen RPS and De Boer K (1998) The bottleneck: Mitochondrial imperatives in oogenesis and ovarian follicular fate. Mol Cellular Endocrinol 145:81–88.; Motta et al., 2000Motta PM, Nottola SA, Makabe S and Heyn R (2000) Mitochondrial morphology in human fetal and adult female germ cells. Hum Reprod 15 Suppl 2:129–147.). These are, albeit, not isolated from each other. Actually, through repeated cycles of fusion and fission, mitochondria exchange membranes, solutes, metabolites, proteins, RNAs, and mtDNAs, resulting in electrically coupled organelles. The balance of fusion to fission also regulates mitochondrial number, morphology, transport, function, and turnover, which is collectively known as mitochondrial dynamics (Mishra and Chan, 2014Mishra P and Chan DC (2014) Mitochondrial dynamics and inheritance during cell division, development and disease. Nat Rev Mol Cell Biol 15:634–646.). Both, fusion- and fission-deficient cells exhibit mitochondrial heterogeneity and dysfunction (Eura et al., 2003Eura Y, Ishihara N, Yokota S and Mihara K (2003) Two mitofusin proteins, mammalian homologues of FZO, with distinct functions are both required for mitochondrial fusion. J Biochem 134:333-344.; Chen et al., 2003Chen H, Detmer SA, Ewald AJ, Griffin EE, Fraser SE and Chan DC (2003) Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J Cell Biol 160:189-200., 2005Chen H, Chomyn A and Chan DC (2005) Disruption of fusion results in mitochondrial heterogeneity and dysfunction. J Biol Chem 280:26185-92.; Ishihara et al., 2009Ishihara N, Nomura M, Jofuku A, Kato H, Suzuki SO, Masuda K, Otera H, Nakanishi Y, Nonaka I, Goto Y-I et al. (2009) Mitochondrial fission factor Drp1 is essential for embryonic development and synapse formation in mice. Nat Cell Biol 11:958–66.; Udagawa et al., 2014Udagawa O, Ishihara T, Maeda M, Matsunaga Y, Tsukamoto S, Kawano N, Miyado K, Shitara H, Yokota S, Nomura M et al. (2014) Mitochondrial Fission Factor Drp1 maintains oocyte quality via dynamic rearrangement of multiple organelles. Curr Biol 24:2451-2458.; Wakai et al., 2014Wakai T, Harada Y, Miyado K and Kono T (2014) Mitochondrial dynamics controlled by mitofusins define organelle positioning and movement during mouse oocyte maturation. Mol Hum Reprod 20:1090-1100.), supporting the importance of these events to mitochondrial health. In keeping with this, fragmentation of the mitochondrial network has been associated with a low bioenergetic state (i.e., in oocytes), while its elongation implies a high bioenergetic yielding, such as that of liver, muscle, and brain (Bach et al., 2003Bach D, Pich S, Soriano FX, Vega N, Baumgartner B, Oriola J, Daugaard JR, Lloberas J, Camps M, Zierath JR et al. (2003) Mitofusin-2 determines mitochondrial network architecture and mitochondrial metabolism: A novel regulatory mechanism altered in obesity. J Biol Chem 278:17190-17197.; Zorzano et al., 2015Zorzano A, Hernández-Alvarez MI, Sebastián D and Muñoz JP (2015) Mitofusin 2 as a driver that controls energy metabolism and insulin signaling. Antioxid Redox Signal 22:1020-31.; Schrepfer and Scorrano 2016Schrepfer E and Scorrano L (2016) Mitofusins, from mitochondria to metabolism. Mol Cell 61:683-694.).

Several proteins regulate mitochondrial fission, with the Dynamin-related protein 1 (DRP1) being the best characterized (Ishihara et al., 2009Ishihara N, Nomura M, Jofuku A, Kato H, Suzuki SO, Masuda K, Otera H, Nakanishi Y, Nonaka I, Goto Y-I et al. (2009) Mitochondrial fission factor Drp1 is essential for embryonic development and synapse formation in mice. Nat Cell Biol 11:958–66.). DRP1 is a cytosolic protein that is recruited to mitochondria by multiple receptors, including mitochondrial fission factor (MMF), mitochondrial dynamic proteins of 49 kDa (MID49) and 51 kDa (MID51), and fission 1 (FIS1) (Mishra and Chan, 2014Mishra P and Chan DC (2014) Mitochondrial dynamics and inheritance during cell division, development and disease. Nat Rev Mol Cell Biol 15:634–646.; Schrepfer and Scorrano 2016Schrepfer E and Scorrano L (2016) Mitofusins, from mitochondria to metabolism. Mol Cell 61:683-694.). In turn, the optic atrophy 1 (OPA1) regulates inner membrane fusion and cristae remodeling (Olichon et al., 2002Olichon A, Emorine LJ, Descoins E, Pelloquin L, Brichese L, Gas N, Guillou E, Delettre C, Valette A, Hamel CP et al. (2002) The human dynamin-related protein OPA1 is anchored to the mitochondrial inner membrane facing the inter-membrane space. FEBS Lett 523:171–176., 2003Olichon A, Baricault L, Gas N, Guillou E, Valette A, Belenguer P and Lenaers G (2003) Loss of OPA1 perturbates the mitochondrial inner membrane structure and integrity, leading to cytochrome c release and apoptosis. J Biol Chem 278:7743–7746.; Cipolat et al., 2004Cipolat S, de Brito OM, Dal Zilio B and Scorrano L (2004) OPA1 requires mitofusin 1 to promote mitochondrial fusion. Proc Nat Acad Sci U S A 101:15927-15932.; Griparic et al., 2004Griparic L, Van Der Wel NN, Orozco IJ, Peters PJ and Van Der Bliek AM (2004) Loss of the intermembrane space protein Mgm1/OPA1 induces swelling and localized constrictions along the lengths of mitochondria. J Biol Chem 279:18792-18798.; Pernas and Scorrano 2016Pernas L and Scorrano L (2016) Mito-morphosis: Mitochondrial fusion, fission, and cristae remodeling as key mediators of cellular function. Annu Rev Physiol 78:505–531.), whereas mitofusins 1 (MFN1) and 2 (MFN2) regulate outer membrane fusion (Chen et al., 2003Chen H, Detmer SA, Ewald AJ, Griffin EE, Fraser SE and Chan DC (2003) Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J Cell Biol 160:189-200., 2005Chen H, Chomyn A and Chan DC (2005) Disruption of fusion results in mitochondrial heterogeneity and dysfunction. J Biol Chem 280:26185-92., 2007Chen H, McCaffery JM and Chan DC (2007) Mitochondrial fusion protects against neurodegeneration in the cerebellum. Cell 130:548-62., 2010Chen H, Vermulst M, Wang YE, Chomyn A, Prolla TA, McCaffery JM and Chan DC (2010) Mitochondrial fusion is required for mtDNA stability in skeletal muscle and tolerance of mtDNA mutations. Cell 141:280-9.; Ishihara et al., 2004Ishihara N, Eura Y and Mihara K (2004) Mitofusin 1 and 2 play distinct roles in mitochondrial fusion reactions via GTPase activity. J Cell Sci 117:6535–6546.; Schrepfer and Scorrano, 2016Schrepfer E and Scorrano L (2016) Mitofusins, from mitochondria to metabolism. Mol Cell 61:683-694.). Mitochondrial fusion is initiated by homo and heterotypic interaction of MFN1 and MFN2 from two adjacent organelles (Ishihara et al., 2004Ishihara N, Eura Y and Mihara K (2004) Mitofusin 1 and 2 play distinct roles in mitochondrial fusion reactions via GTPase activity. J Cell Sci 117:6535–6546.; Schrepfer and Scorrano, 2016Schrepfer E and Scorrano L (2016) Mitofusins, from mitochondria to metabolism. Mol Cell 61:683-694.). Given that MFN2 is present on the ER membrane, it also regulates ER-mitochondria tethering (de Brito and Scorrano, 2008de Brito OM and Scorrano L (2008) Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature 456:605-610.). This connection, known as ER mitochondria-associated membranes (MAMs), has been shown to play an essential role in the regulation of ER, mitochondrial, and cellular functions (Ngoh et al., 2012Ngoh GA, Papanicolaou KN and Walsh K (2012) Loss of mitofusin 2 promotes endoplasmic reticulum stress. J Biol Chem 287:20321–20332.; Hamasaki et al., 2013Hamasaki M, Furuta N, Matsuda A, Nezu A, Yamamoto A, Fujita N, Oomori H, Noda T, Haraguchi T, Hiraoka Y et al. (2013) Autophagosomes form at ER–mitochondria contact sites. Nature 495:389-393.; Schneeberger et al., 2013Schneeberger M, Dietrich MO, Sebastián D, Imbernón M, Castaño C, Garcia A, Esteban Y, Gonzalez-Franquesa A, Rodríguez IC, Bortolozzi A et al. (2013) Mitofusin 2 in POMC neurons connects ER stress with leptin resistance and energy imbalance. Cell 155:172-187.; Muñoz et al., 2014; Carreras-Sureda et al., 2017Carreras-Sureda A, Pihán P and Hetz C (2017) The unfolded protein response: At the intersection between endoplasmic reticulum function and mitochondrial bioenergetics. Front Oncol 7:1-7.; Pathak and Trebak, 2018Pathak T and Trebak M (2018) Mitochondrial Ca2+ signaling. Pharmacol Ther 192:112–123.). MFN2 downregulation is associated with decreased expression of subunits of the Krebs cycle and electron transport chain, reduced oxygen consumption, lower ΔΨm, and increased reactive oxygen species (ROS) (Santel and Fuller, 2001Santel A and Fuller MT (2001) Control of mitochondrial morphology by a human mitofusin. J Cell Sci 114:867-874.; Yasukawa et al., 2009Yasukawa K, Oshiumi H, Takeda M, Ishihara N, Yanagi Y, Seya T, Kawabata S and Koshiba T (2009) Mitofusin 2 inhibits mitochondrial antiviral signaling. Sci Signal 2:ra47.; Ngoh et al., 2012Ngoh GA, Papanicolaou KN and Walsh K (2012) Loss of mitofusin 2 promotes endoplasmic reticulum stress. J Biol Chem 287:20321–20332.; Wakai et al., 2014Wakai T, Harada Y, Miyado K and Kono T (2014) Mitochondrial dynamics controlled by mitofusins define organelle positioning and movement during mouse oocyte maturation. Mol Hum Reprod 20:1090-1100.; Filadi et al., 2015Filadi R, Greotti E, Turacchio G, Luini A, Pozzan T and Pizzo P (2015) Mitofusin 2 ablation increases endoplasmic reticulum-mitochondria coupling. Proc Nat Acad Sci U S A 112:E2174-81.; Schrepfer and Scorrano, 2016Schrepfer E and Scorrano L (2016) Mitofusins, from mitochondria to metabolism. Mol Cell 61:683-694.). These effects of MFN2 seem to be more evident in muscle, liver and hypothalamic neurons, tissues in which expression of MFN2 is enhanced (Chen et al., 2007Chen H, McCaffery JM and Chan DC (2007) Mitochondrial fusion protects against neurodegeneration in the cerebellum. Cell 130:548-62.; Chen et al., 2010Chen H, Vermulst M, Wang YE, Chomyn A, Prolla TA, McCaffery JM and Chan DC (2010) Mitochondrial fusion is required for mtDNA stability in skeletal muscle and tolerance of mtDNA mutations. Cell 141:280-9.; Schneeberger et al., 2013Schneeberger M, Dietrich MO, Sebastián D, Imbernón M, Castaño C, Garcia A, Esteban Y, Gonzalez-Franquesa A, Rodríguez IC, Bortolozzi A et al. (2013) Mitofusin 2 in POMC neurons connects ER stress with leptin resistance and energy imbalance. Cell 155:172-187.; Schrepfer and Scorrano 2016Schrepfer E and Scorrano L (2016) Mitofusins, from mitochondria to metabolism. Mol Cell 61:683-694.). MFN2 expression has also been inversely linked with ER stress, insulin signaling and diabetes (Bach et al., 2003Bach D, Pich S, Soriano FX, Vega N, Baumgartner B, Oriola J, Daugaard JR, Lloberas J, Camps M, Zierath JR et al. (2003) Mitofusin-2 determines mitochondrial network architecture and mitochondrial metabolism: A novel regulatory mechanism altered in obesity. J Biol Chem 278:17190-17197.; Mingrone et al., 2005Mingrone G, Manco M, Calvani M, Castagneto M, Naon D and Zorzano A (2005) Could the low level of expression of the gene encoding skeletal muscle mitofusin-2 account for the metabolic inflexibility of obesity? Diabetologia 48:2108–2114.; Sebastian et al., 2012Sebastian D, Hernandez-Alvarez MI, Segales J, Sorianello E, Munoz JP, Sala D, Waget A, Liesa M, Paz JC, Gopalacharyulu P et al. (2012) Mitofusin 2 (Mfn2) links mitochondrial and endoplasmic reticulum function with insulin signaling and is essential for normal glucose homeostasis. Proc Natl Acad Sci U S A 109:5523-5528.; Schneeberger et al., 2013Schneeberger M, Dietrich MO, Sebastián D, Imbernón M, Castaño C, Garcia A, Esteban Y, Gonzalez-Franquesa A, Rodríguez IC, Bortolozzi A et al. (2013) Mitofusin 2 in POMC neurons connects ER stress with leptin resistance and energy imbalance. Cell 155:172-187.; Zorzano et al., 2015Zorzano A, Hernández-Alvarez MI, Sebastián D and Muñoz JP (2015) Mitofusin 2 as a driver that controls energy metabolism and insulin signaling. Antioxid Redox Signal 22:1020-31.; Sarparanta et al., 2017Sarparanta J, García-Macia M and Singh R (2017) Autophagy and mitochondria in obesity and type 2 diabetes. Curr Diabetes Rev 13:352-369.).Muñoz JP, Ivanova S, Sánchez-Wandelmer J, Martínez-Cristóbal P, Noguera E, Sancho A, Díaz-Ramos A, Hernández-Alvarez MI, Sebastián D, Mauvezin C et al. (2013) Mfn2 modulates the UPR and mitochondrial function via repression of PERK. EMBO J 32:2348-2361.

Mitochondria in female germ cells

The earliest stages of embryogenesis are characterized by rapid cell division (i.e., cleavage) that gives rise to blastocysts. During these stages, the embryo relies on maternal factors inherited from the oocyte (i.e., mRNAs, proteins and mitochondria), as the embryonic genome is transcriptionally inactive. Also, in agreement with the “embryo silent” hypothesis, mitochondria show low activity during these stages to protect embryonic cells from oxidative damage (Leese, 2012Leese HJ (2012) Metabolism of the preimplantation embryo: 40 Years on. Reproduction 143:417–427.). At the blastocyst stage, increased protein synthesis and blastocoel expansion is accompanied by upregulation of mitochondrial activity in cells that give rise to extraembryonic tissues (i.e., the trophectoderm) (Trimarchi et al., 2000Trimarchi JR, Liu L, Porterfield DM, Smith PJ and Keefe DL (2000) Oxidative phosphorylation-dependent and -independent oxygen consumption by individual preimplantation mouse embryos. Biol Reprod 62:1866-74.; May-Panloup et al., 2005May-Panloup P, Vignon X, Chrétien MF, Heyman Y, Tamassia M, Malthièry Y and Reynier P (2005) Increase of mitochondrial DNA content and transcripts in early bovine embryogenesis associated with upregulation of mtTFA and NRF1 transcription factors. Reprod Biol Endocrinol 3:65.; Hashimoto et al., 2017Hashimoto S, Morimoto N, Yamanaka M, Matsumoto H, Yamochi T, Goto H, Inoue M, Nakaoka Y, Shibahara H and Morimoto Y (2017) Quantitative and qualitative changes of mitochondria in human preimplantation embryos. J Assist Reprod Genet 34:573-580.; St John, 2019St John JC (2019) Mitochondria and female germline stem cells - a mitochondrial DNA perspective. Cells 8:852.). Activation of mitochondrial function is postponed, however, in the inner cell mass that originates the embryo proper. Mitochondrial architecture and function seem to remain underdeveloped in cells committed with germline specification, and mtDNA replication is only resumed with primordial germ cell (PGC) differentiation (Wassarman and Josefowicz, 1978Wassarman PM and Josefowicz WJ (1978) Oocyte development in the mouse: An ultrastructural comparison of oocytes isolated at various stages of growth and meiotic competence. J Morphol 156:209-235.; Motta et al., 2000Motta PM, Nottola SA, Makabe S and Heyn R (2000) Mitochondrial morphology in human fetal and adult female germ cells. Hum Reprod 15 Suppl 2:129–147.; Cree et al., 2008Cree LM, Samuels DC, Sousa Lopes SC, Rajasimha HK, Wonnapinij P, Mann JR, Dahl H-HM and Chinnery PF (2008) A reduction of mitochondrial DNA molecules during embryogenesis explains the rapid segregation of genotypes. Nat Genet 40:249-54.; Wai et al., 2008Wai T, Teoli D and Shoubridge EA (2008) The mitochondrial DNA genetic bottleneck results from replication of a subpopulation of genomes. Nat Genet 40:1484-1488.; St John et al., 2010St John JC, Facucho-Oliveira J, Jiang Y, Kelly R and Salah R (2010) Mitochondrial DNA transmission, replication and inheritance: A journey from the gamete through the embryo and into offspring and embryonic stem cells. Hum Reprod Update 16:488-509.; Floros et al., 2018Floros VI, Pyle A, Dietmann S, Wei W, Tang WCW, Irie N, Payne B, Capalbo A, Noli L, Coxhead J et al. (2018) Segregation of mitochondrial DNA heteroplasmy through a developmental genetic bottleneck in human embryos. Nat Cell Biol 20:144–151.; Chiaratti et al., 2018Chiaratti MR, Garcia BM, Carvalho KF, Machado TS, Ribeiro FKDS and Macabelli CH (2018) The role of mitochondria in the female germline: Implications to fertility and inheritance of mitochondrial diseases. Cell Biol Int 42:1-39.; St John, 2019St John JC (2019) Mitochondria and female germline stem cells - a mitochondrial DNA perspective. Cells 8:852.).

Among the hundreds of cells in the developing fetus, PGCs originate from a few dozen located at the basis of allantois. Yet, after migration to the genital ridge, PGCs proliferate quickly to generate in females millions of oogonia (Leitch et al., 2013Leitch HG, Tang WWC and Surani MA (2013) Primordial germ-cell development and epigenetic reprogramming in mammals. Curr Top Dev Biol 104:149–187.). After entering meiosis, these primary oocytes receive a cover layer of somatic pre-granulosa cells, giving rise to primordial follicles still during fetal life. These follicles constitute the ovarian reserve that females carry throughout their reproductive life (Oktem and Urman, 2010Oktem O and Urman B (2010) Understanding follicle growth in vivo. Hum Reprod 25:2944–2954.). After puberty, the ovary provides an adequate environment for follicle growth and maturation (Clarke, 2017Clarke HJ (2017) Regulation of germ cell development by intercellular signaling in the mammalian ovarian follicle. Wiley Interdiscip Rev Dev Biol 7:e294.). During this period, the oocyte stockpiles several molecules that are required later during embryogenesis. This includes a ~1,000-fold increase in mitochondria (Jansen and De Boer, 1998Jansen RPS and De Boer K (1998) The bottleneck: Mitochondrial imperatives in oogenesis and ovarian follicular fate. Mol Cellular Endocrinol 145:81–88.; Cree et al., 2008Cree LM, Samuels DC, Sousa Lopes SC, Rajasimha HK, Wonnapinij P, Mann JR, Dahl H-HM and Chinnery PF (2008) A reduction of mitochondrial DNA molecules during embryogenesis explains the rapid segregation of genotypes. Nat Genet 40:249-54.; Wai et al., 2008Wai T, Teoli D and Shoubridge EA (2008) The mitochondrial DNA genetic bottleneck results from replication of a subpopulation of genomes. Nat Genet 40:1484-1488.; St John, 2019St John JC (2019) Mitochondria and female germline stem cells - a mitochondrial DNA perspective. Cells 8:852.), which accounts for the largest mitochondrial content amongst all cells in mammals. In spite of this, mitochondria display several characteristics that suggest they are immature and low functional in oocytes (Arhin et al., 2018Arhin SK, Lu J, Xi H and Jin X (2018) Energy requirements in mammalian oogenesis. Cell Mol Biol 64:12-19.). In fact, oocytes lacking the pyruvate dehydrogenase E1 alpha 1 (PDHA1), a key gene required for mitochondrial activity, successfully develop during most part of oogenesis and are ovulated (Johnson et al., 2007Johnson MT, Freeman EA, Gardner DK and Hunt PA (2007) Oxidative metabolism of pyruvate is required for meiotic maturation of murine oocytes in vivo. Biol Reprod 77:2–8.). Thus, although mitochondria do play an essential role during the final steps of oocyte development, the “embryo silent” hypothesis likely extends to oogenesis too (Arhin et al., 2018Arhin SK, Lu J, Xi H and Jin X (2018) Energy requirements in mammalian oogenesis. Cell Mol Biol 64:12-19.). Accordingly, somatic cells surrounding the oocyte (i.e., cumulus cells) provide the oocyte with several energetic molecules, including amino acids, cholesterol, pyruvate, AMP, and ATP (Su et al., 2007Su YQ, Sugiura K, Wigglesworth K, O’Brien MJ, Affourtit JP, Pangas SA, Matzuk MM and Eppig JJ (2007) Oocyte regulation of metabolic cooperativity between mouse cumulus cells and oocytes: BMP15 and GDF9 control cholesterol biosynthesis in cumulus cells. Development 135:111-121., 2009Su YQ, Sugiura K and Eppig JJ (2009) Mouse oocyte control of granulosa cell development and function: Paracrine regulation of cumulus cell metabolism. Semin Reprod Med 27:32-42.; Sugiura et al., 2007Sugiura K, Su YQ, Diaz FJ, Pangas SA, Sharma S, Wigglesworth K, O’Brien MJ, Matzuk MM, Shimasaki S and Eppig JJ (2007) Oocyte-derived BMP15 and FGFs cooperate to promote glycolysis in cumulus cells. Development 134:2593-2603.). Moreover, the adenosine salvage pathway seems to be a key source of ATP, giving it can be generated from abundant amounts of cyclic AMP (cAMP) present in oocytes (Scantland et al., 2014Scantland S, Tessaro I, Macabelli CH, Macaulay AD, Cagnone G, Fournier É, Luciano AM and Robert C (2014) The adenosine salvage pathway as an alternative to mitochondrial production of ATP in maturing mammalian oocytes. Biol Reprod 91:1-11.).

If mitochondria are not highly active in oocytes, why are they present in massive amounts before fertilization? This can be, at least, partially explained by downregulation of mitochondrial biogenesis during early embryogenesis; mitochondria are segregated among hundreds of embryonic cells without any increase in number up to the time of embryo implantation (Pikó and Taylor, 1987Pikó L and Taylor KD (1987) Amounts of mitochondrial DNA and abundance of some mitochondrial gene transcripts in early mouse embryos. Dev Biol 123:364–374.; Thundathil et al., 2005Thundathil J, Filion F and Smith LC (2005) Molecular control of mitochondrial function in preimplantation mouse embryos. Mol Reprod Dev 71:405-13.; Cree et al., 2008Cree LM, Samuels DC, Sousa Lopes SC, Rajasimha HK, Wonnapinij P, Mann JR, Dahl H-HM and Chinnery PF (2008) A reduction of mitochondrial DNA molecules during embryogenesis explains the rapid segregation of genotypes. Nat Genet 40:249-54.; Wai et al., 2008Wai T, Teoli D and Shoubridge EA (2008) The mitochondrial DNA genetic bottleneck results from replication of a subpopulation of genomes. Nat Genet 40:1484-1488.; St John, 2019St John JC (2019) Mitochondria and female germline stem cells - a mitochondrial DNA perspective. Cells 8:852.). Therefore, a threshold number of mitochondria is necessary in oocytes to assure that every embryonic cell will inherit a minimum complement of mitochondria (Chiaratti and Meirelles, 2010Chiaratti MR and Meirelles FV (2010) Mitochondrial DNA copy number, a marker of viability for oocytes. Biol Reprod 83:1-2.; Wai et al., 2010Wai T, Ao A, Zhang X, Cyr D, Dufort D and Shoubridge EA (2010) The role of mitochondrial DNA copy number in mammalian fertility. Biol Reprod 83:52-62.). In keeping with this idea, extensive fragmentation of the mitochondrial network in oocytes allows for efficient segregation of mitochondria during early embryogenesis (Ashley et al., 1989Ashley MV, Laipis PJ and Hauswirth WW (1989) Rapid segregation of heteroplasmic bovine mitochondria. Nucleic Acids Res 17:7325-7331.; Cree et al., 2008Cree LM, Samuels DC, Sousa Lopes SC, Rajasimha HK, Wonnapinij P, Mann JR, Dahl H-HM and Chinnery PF (2008) A reduction of mitochondrial DNA molecules during embryogenesis explains the rapid segregation of genotypes. Nat Genet 40:249-54.; Ferreira et al., 2010Ferreira CR, Burgstaller JP, Perecin F, Garcia JM, Chiaratti MR, Méo SC, Müller M, Smith LC, Meirelles FV and Steinborn R (2010) Pronounced segregation of donor mitochondria introduced by bovine ooplasmic transfer to the female germ-line. Biol Reprod 82:563-71.; Lee et al., 2012bLee H, Ma H, Juanes R, Tachibana M, Sparman M, Woodward J, Ramsey C, Xy J, Kand EJ, Amato P et al. (2012b) Rapid mitochondrial DNA segregation in primate preimplantation embryos precedes somatic and germline bottleneck. Cell Rep 1:506–515.). Upregulation of pro-fission proteins (i.e., DRP1) and downregulation of MFN2 likely supports mitochondrial fragmentation during oogenesis (Udagawa et al., 2014Udagawa O, Ishihara T, Maeda M, Matsunaga Y, Tsukamoto S, Kawano N, Miyado K, Shitara H, Yokota S, Nomura M et al. (2014) Mitochondrial Fission Factor Drp1 maintains oocyte quality via dynamic rearrangement of multiple organelles. Curr Biol 24:2451-2458.; Machado et al., 2018Machado TS, Carvalho KF, Garcia BM, Zangirolamo AF, Macabelli CH, Sugiyama FHC, Grejo MP, Augusto Neto JD, Ribeiro FKS, Sarapiao FD et al. (2018) Mitofusin 1 is required for the oocyte-granulosa cell communication that regulates oogenesis. bioRxiv 10.1101/498642.; Hou et al., 2019Hou X, Zhu S, Zhang H, Li C, Qiu D, Ge J, Guo X and Wang Q (2019) Mitofusin1 in oocyte is essential for female fertility. Redox Biol 21:101110.; Zhang et al., 2019bZhang M, Bener MB, Jiang Z, Wang T, Esencan E, Scott R, Horvath T and Seli E (2019b) Mitofusin 2 plays a role in oocyte and follicle development, and is required to maintain ovarian follicular reserve during reproductive aging. Aging 11:3919-3938.). However, oocytes do retain fusion competence, as loss of DRP1 leads to mitochondrial elongation (Udagawa et al., 2014Udagawa O, Ishihara T, Maeda M, Matsunaga Y, Tsukamoto S, Kawano N, Miyado K, Shitara H, Yokota S, Nomura M et al. (2014) Mitochondrial Fission Factor Drp1 maintains oocyte quality via dynamic rearrangement of multiple organelles. Curr Biol 24:2451-2458.). Moreover, MFN1 is required for oocyte growth and ovulation; MFN1 loss impairs oocyte-somatic cell communication, disrupting folliculogenesis (Machado et al., 2018Machado TS, Carvalho KF, Garcia BM, Zangirolamo AF, Macabelli CH, Sugiyama FHC, Grejo MP, Augusto Neto JD, Ribeiro FKS, Sarapiao FD et al. (2018) Mitofusin 1 is required for the oocyte-granulosa cell communication that regulates oogenesis. bioRxiv 10.1101/498642.; Hou et al., 2019Hou X, Zhu S, Zhang H, Li C, Qiu D, Ge J, Guo X and Wang Q (2019) Mitofusin1 in oocyte is essential for female fertility. Redox Biol 21:101110.; Zhang et al., 2019aZhang M, Bener MB, Jiang Z, Wang T, Esencan E, Scott III R, Horvath T and Seli E (2019a) Mitofusin 1 is required for female fertility and to maintain ovarian follicular reserve. Cell Death Dis 10:560.,bZhang M, Bener MB, Jiang Z, Wang T, Esencan E, Scott R, Horvath T and Seli E (2019b) Mitofusin 2 plays a role in oocyte and follicle development, and is required to maintain ovarian follicular reserve during reproductive aging. Aging 11:3919-3938.).

Mitochondrial diseases originated from mtDNA mutations

Diseases caused by mutations in mtDNA are mostly severe and affect ~1 in 4,300 people all over the world (Schaefer et al., 2008Schaefer AM, McFarland R, Blakely EL, He L, Whittaker RG, Taylor RW, Chinnery PF and Turnbull DM (2008) Prevalence of mitochondrial DNA disease in adults. Ann Neurol 63:35-9.). In addition, almost every person (including healthy people) carries very low levels of mutant mtDNA (Payne et al., 2013Payne BAI, Wilson IJ, Yu-Wai-Man P, Coxhead J, Deehan D, Horvath R, Taylor RW, Samuels DC, Santibanez-Koref M and Chinnery PF (2013) Universal heteroplasmy of human mitochondrial DNA. Hum Mol Genet 22:384–390.) that may be passed down to following generations and associate with late-onset diseases, such as Parkinson disease, Alzheimer disease, and common cancers (Poulton et al., 2010Poulton J, Chiaratti MR, Meirelles FV, Kennedy S, Wells D and Holt IJ (2010) Transmission of mitochondrial DNA diseases and ways to prevent them. PLoS Genet 6:e1001066.;Wallace, 2011Wallace DC (2011) Bioenergetic origins of complexity and disease. Cold Spring Harb Symp Quantit Biol 76:1-16.; Schon et al., 2012Schon EA, DiMauro S and Hirano M (2012) Human mitochondrial DNA: Roles of inherited and somatic mutations. Nat Rev Genet 13:878-90.; Gorman et al., 2015Gorman GS, Schaefer AM, Ng Y, Gomez N, Blakely EL, Alston CL, Feeney C, Horvath R, Yu-Wai-Man P, Chinnery PF et al. (2015) Prevalence of nuclear and mitochondrial DNA mutations related to adult mitochondrial disease. Ann Neurol 77:753-759.; Stewart and Chinnery, 2015Stewart JB and Chinnery PF (2015) The dynamics of mitochondrial DNA heteroplasmy: Implications for human health and disease. Nat Rev Genet 16:530-42.). With rare exceptions (Luo et al., 2018Luo S, Valencia CA, Zhang J, Lee NC, Slone J, Gui B, Wang X, Li Z, Dell S, Brown J et al. (2018) Biparental inheritance of mitochondrial DNA in humans. Proc Natl Acad Sci U S A 115:13039–13044.), mitochondria are inherited exclusively from the mother (Wallace and Chalkia, 2013Wallace DC and Chalkia D (2013) Mitochondrial DNA genetics and the heteroplasmy conundrum in evolution and disease. Cold Spring Harb Perspect Biol 5:a021220.). This uniparental pattern of inheritance is explained by the presence of several thousand mitochondria in the ovulated oocyte, against only dozens in the sperm (Wai et al., 2010Wai T, Ao A, Zhang X, Cyr D, Dufort D and Shoubridge EA (2010) The role of mitochondrial DNA copy number in mammalian fertility. Biol Reprod 83:52-62.). Additionally, the early embryo actively eliminates paternal mitochondria introduced into the oocyte during fertilization (Sutovsky et al., 1999Sutovsky P, Moreno RD, Ramalho-Santos J, Dominko T, Simerly C and Schatten G (1999) Ubiquitin tag for sperm mitochondria. Nature 402:371-372.; Rojansky et al., 2016Rojansky R, Cha MY and Chan DC (2016) Elimination of paternal mitochondria in mouse embryos occurs through autophagic degradation dependent on PARKIN and MUL1. eLife 5:1–18.). Although it is not clear why sperm mitochondria are excluded from the developing embryo, their elimination is in agreement with the “embryo silent” hypothesis, as sperm mitochondria are elongated, contain well-developed cristae and are highly active (Sutovsky et al., 1999Sutovsky P, Moreno RD, Ramalho-Santos J, Dominko T, Simerly C and Schatten G (1999) Ubiquitin tag for sperm mitochondria. Nature 402:371-372.; Ford, 2004Ford WCL (2004) Regulation of sperm function by reactive oxygen species. Hum Reprod Update 10:387-399.; Ruiz-Pesini et al., 2007Ruiz-Pesini E, Díez-Sánchez C, López-Pérez MJ and Enríquez JA (2007) The role of the mitochondrion in sperm function: Is there a place for oxidative phosphorylation or is this a purely glycolytic process? Curr Top Dev Biol 77:3-19.; Wai et al., 2010Wai T, Ao A, Zhang X, Cyr D, Dufort D and Shoubridge EA (2010) The role of mitochondrial DNA copy number in mammalian fertility. Biol Reprod 83:52-62.; Rojansky et al., 2016Rojansky R, Cha MY and Chan DC (2016) Elimination of paternal mitochondria in mouse embryos occurs through autophagic degradation dependent on PARKIN and MUL1. eLife 5:1–18.).

Mutations in mtDNA are much more frequent than in the nuclear DNA (Johnson and Johnson, 2001Johnson AA and Johnson KA (2001) Exonuclease proofreading by human mitochondrial DNA polymerase. J Biol Chem 276:38097–107.), which was initially thought to be explained by mtDNA proximity to ROS generation sites; the mitochondrial genome is attached to the inner mitochondrial membrane, close to complexes involved with the electron transport chain (Wallace, 2005Wallace DC (2005) A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine. Annu Rev Genet 39:359-407.). However, there is now data supporting that most mtDNA mutations originate from replication errors of the mtDNA polymerase (Kauppila et al., 2017Kauppila TES, Kauppila JHK and Larsson NG (2017) Mammalian mitochondria and aging: An update. Cell Metab 25:57–71.). In humans, mice, and flies, for instance, transition mutations, which are indicative of replication errors, are more common than transversions, which often result from oxidative damage (Tomas, 1993Tomas L (1993) Instability and decay of the primary structure of DNA. Nature 362:709-715.; Zheng et al., 2006Zheng W, Khrapko K, Coller HA, Thilly WG and Copeland WC (2006) Origins of human mitochondrial point mutations as DNA polymerase γ-mediated errors. Mut Res 599:11-20.; Kennedy et al., 2013Kennedy SR, Salk JJ, Schmitt MW and Loeb LA (2013) Ultra-sensitive sequencing reveals an age-related increase in somatic mitochondrial mutations that are inconsistent with oxidative damage. PLoS Genet 9:e1003794.; Itsara et al., 2014Itsara LS, Kennedy SR, Fox EJ, Yu S, Hewitt JJ, Sanchez-Contreras M, Cardozo-Pelaez F and Pallanck LJ (2014) Oxidative stress is not a major contributor to somatic mitochondrial DNA mutations. PLoS Genet 10:1003974.). In fact, the machinery of DNA repair in the mitochondrion does not seem to be as effective as in the nucleus (Vermulst et al., 2008Vermulst M, Wanagat J, Kujoth GC, Bielas JH, Rabinovitch PS, Prolla TA and Loeb LA (2008) DNA deletions and clonal mutations drive premature aging in mitochondrial mutator mice. Nat Genet 40:392-394.; Maynard et al., 2009Maynard S, Schurman SH, Harboe C, de Souza-Pinto NC and Bohr VA (2009) Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis 30:2–10.; Kazak et al., 2012Kazak L, Reyes A and Holt IJ (2012) Minimizing the damage: Repair pathways keep mitochondrial DNA intact. Nat Rev Mol Cell Biol 13:659–671.; Muftuoglu et al., 2014Muftuoglu M, Mori MP and de Souza-Pinto NC (2014) Formation and repair of oxidative damage in the mitochondrial DNA. Mitochondrion 17:164-181.). Thus, intense replication of mtDNA during oogenesis makes it prone to replication errors (Wai et al., 2008Wai T, Teoli D and Shoubridge EA (2008) The mitochondrial DNA genetic bottleneck results from replication of a subpopulation of genomes. Nat Genet 40:1484-1488.; Mahrous et al., 2012Mahrous E, Yang Q and Clarke HJ (2012) Regulation of mitochondrial DNA accumulation during oocyte growth and meiotic maturation in the mouse. Reproduction 144:177–185.; Wei et al., 2019Wei W, Tuna S, Keogh MJ, Smith KR, Aitman TJ, Beales PL, Bennett DL, Gale DP, Bitner-Glindzicz MAK, Black GC et al. (2019) Germline selection shapes human mitochondrial DNA diversity. Science 364:eaau6520.).

The existence of a DNA repair machinery inside mitochondria is well established, but not fully characterized (Scheibye-Knudsen et al., 2015Scheibye-Knudsen M, Fang EF, Croteau DL, Wilson DM and Bohr VA (2015) Protecting the mitochondrial powerhouse. Trends Cell Biol 25:158-170.). Most genes encoding for factors involved in this machinery are shared with the nucleus; alternative variants of these genes allow for the protein to be targeted either to the nucleus or the mitochondrion (Muftuoglu et al., 2014Muftuoglu M, Mori MP and de Souza-Pinto NC (2014) Formation and repair of oxidative damage in the mitochondrial DNA. Mitochondrion 17:164-181.; Scheibye-Knudsen et al., 2015Scheibye-Knudsen M, Fang EF, Croteau DL, Wilson DM and Bohr VA (2015) Protecting the mitochondrial powerhouse. Trends Cell Biol 25:158-170.). The best-known pathway of DNA repair in mitochondria is base excision repair (BER). Yet, several other enzymes involved with mismatch repair (MMR), non-homologous end joining (NHEJ), and direct repair have been reported in mitochondria (Maynard et al., 2009Maynard S, Schurman SH, Harboe C, de Souza-Pinto NC and Bohr VA (2009) Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis 30:2–10., 2010Maynard S, de Souza-Pinto NC, Scheibye-Knudsen M and Bohr VA (2010) Mitochondrial base excision repair assays. Methods 51:416–25.; Ruhanen et al., 2010Ruhanen H, Borrie S, Szabadkai G, Tyynismaa H, Jones AWE, Kang D, Taanman JW and Yasukawa T (2010) Mitochondrial single-stranded DNA binding protein is required for maintenance of mitochondrial DNA and 7S DNA but is not required for mitochondrial nucleoid organisation. Biochim Biophys Acta 1803:931–939.; Halsne et al., 2012Halsne R, Esbensen Y, Wang W, Scheffler K, Suganthan R, Bjørås M and Eide L (2012) Lack of the DNA glycosylases MYH and OGG1 in the cancer prone double mutant mouse does not increase mitochondrial DNA mutagenesis. DNA Repair 11:278-285.; Kazak et al., 2012Kazak L, Reyes A and Holt IJ (2012) Minimizing the damage: Repair pathways keep mitochondrial DNA intact. Nat Rev Mol Cell Biol 13:659–671.; Sharma et al., 2014Sharma NK, Lebedeva M, Thomas T, Kovalenko OA, Stumpf JD, Shadel GS and Santos JH (2014) Intrinsic mitochondrial DNA repair defects in Ataxia Telangiectasia. DNA Repair 13:22-31.; Scheibye-Knudsen et al., 2015Scheibye-Knudsen M, Fang EF, Croteau DL, Wilson DM and Bohr VA (2015) Protecting the mitochondrial powerhouse. Trends Cell Biol 25:158-170.). Moreover, although homologous recombination (HR) has not been proved to contribute with mtDNA repair (Kazak et al., 2012Kazak L, Reyes A and Holt IJ (2012) Minimizing the damage: Repair pathways keep mitochondrial DNA intact. Nat Rev Mol Cell Biol 13:659–671.; Hagström et al., 2014Hagström E, Freyer C, Battersby BJ, Stewart JB and Larsson NG (2014) No recombination of mtDNA after heteroplasmy for 50 generations in the mouse maternal germline. Nucleic Acids Res 42:1111-1116.; Scheibye-Knudsen et al., 2015Scheibye-Knudsen M, Fang EF, Croteau DL, Wilson DM and Bohr VA (2015) Protecting the mitochondrial powerhouse. Trends Cell Biol 25:158-170.), mitochondria do import RAD51, one of the most prominent enzymes of HR (Sage et al., 2010Sage JM, Gildemeister OS and Knight KL (2010) Discovery of a novel function for human RadJ Biol Chem 285:18984–18990.; Chen, 2013Chen XJ (2013) Mechanism of homologous recombination and implications for aging-related deletions in mitochondrial DNA. Microbiol Mol Biol Rev 77:476–496.). RAD51 has also been linked with mtDNA synthesis under replicative stress (Sage and Knight, 2013Sage JM and Knight KL (2013) Human Rad51 promotes mitochondrial DNA synthesis under conditions of increased replication stress. Mitochondrion 13:350-356.), and in oocytes RAD51 is required for mitochondrial function (Kim et al., 2016Kim KH, Park JH, Kim EY, Ko JJ, Park KS and Lee KA (2016) The role of Rad51 in safeguarding mitochondrial activity during the meiotic cell cycle in mammalian oocytes. Sci Rep 6:34110.).

Given that most cells contain several mtDNA molecules, a de novo mutation creates a condition termed heteroplasmy, characterized by the co-existence of two or more mtDNA genotypes (i.e., wild-type and mutant mtDNAs) within the same cell or organelle. Heteroplasmy commonly protects the cell, as most mtDNA mutations are recessive. Unless the mutation level exceeds a critical threshold necessary to cause a biochemical defect (i.e., above 60-90%), the mutation effect will be masked by wild-type molecules (Schon et al., 2012Schon EA, DiMauro S and Hirano M (2012) Human mitochondrial DNA: Roles of inherited and somatic mutations. Nat Rev Genet 13:878-90.; Aanen et al., 2014Aanen DK, Spelbrink JN and Beekman M (2014) What cost mitochondria? The maintenance of functional mitochondrial DNA within and across generations. Philos Trans R Soc Lond B Biol Sci 369:20130438.; Haig, 2016Haig D (2016) Intracellular evolution of mitochondrial DNA (mtDNA) and the tragedy of the cytoplasmic commons. BioEssays 38:549-555.). In addition, a mechanism known as the mitochondrial genetic bottleneck (Hauswirth and Laipis, 1982Hauswirth WW and Laipis PJ (1982) Mitochondrial DNA polymorphism in a maternal lineage of Holstein cows. Proc Natl Acad Sci U S A 79:4686-4690.; Olivo et al., 1983Olivo PD, Van de Walle MJ, Laipis PJ and Hauswirth WW (1983) Nucleotide sequence evidence for rapid genotypic shifts in the bovine mitochondrial DNA D-loop. Nature 306:400–402.; Hauswirth et al., 1984Hauswirth WW, Van de Walle MJ, Laipis PJ and Olivo PD (1984) Heterogeneous mitochondrial DNA D-loop sequences in bovine tissue. Cell 37:1001–1007.; Jenuth et al., 1996Jenuth J, Peterson A, Fu K and Shoubridge E (1996) Random genetic drift in the female germline explains the rapid segregation of mammalian mitochondrial DNA. Nat Genet 14:146–151.; Burgstaller et al., 2018Burgstaller JP, Kolbe T, Havlicek V, Hembach S, Poulton J, Piálek J, Steinborn R, Rülicke T, Brem G, Jones NS et al. (2018) Large-scale genetic analysis reveals mammalian mtDNA heteroplasmy dynamics and variance increase through lifetimes and generations. Nat Commun 9:1-12.) acts in the germline to rapidly re-establish homoplasmy (i.e., the presence of a single mtDNA genotype). This mechanism is based on the absence of mtDNA replication during early embryogenesis, which forces wild-type and mutant mtDNAs to segregate. Also, few cells among the hundreds present in the embryo differentiate into PGCs, resulting in a sampling effect that efficiently selects one mtDNA genotype to populate the following generation (Stewart and Chinnery, 2015Stewart JB and Chinnery PF (2015) The dynamics of mitochondrial DNA heteroplasmy: Implications for human health and disease. Nat Rev Genet 16:530-42.; Burgstaller et al., 2018Burgstaller JP, Kolbe T, Havlicek V, Hembach S, Poulton J, Piálek J, Steinborn R, Rülicke T, Brem G, Jones NS et al. (2018) Large-scale genetic analysis reveals mammalian mtDNA heteroplasmy dynamics and variance increase through lifetimes and generations. Nat Commun 9:1-12.). However, the selected genotype can be either wild-type or mutant, generating genetic variability to be put to test at the cellular, organismal, or population level (Figure 1).

Figure 1
Mitochondrial kinetics in the female germline. Throughout germline development, the number of mitochondrial DNA (mtDNA) molecules per cell varies from 105 - 106 in mature oocytes (before fertilization), 102 - 103 in primordial germ cells (PGCs) and 105 - 106 back to mature oocytes. This variation in copy number accounts for the mitochondrial genetic bottleneck, which forces segregation of mtDNA molecules. In line with this, the mitochondrial network is fragmented in oocytes, allowing efficient partitioning of mitochondria among hundreds of cells until embryonic implantation. In addition, only few cells in the embryo differentiate into PGCs, supporting a sampling effect towards selection of a single mtDNA genotype to populate the future oocyte.

Mutations in mtDNA may vary considering their effect on mitochondrial function from neutral to deleterious. Among deleterious mutations, those affecting tRNA are the most frequent in humans. This is counter-intuitive though, as tRNA genes account for only 10% of the total coding capacity of mtDNA (Schon et al., 2012Schon EA, DiMauro S and Hirano M (2012) Human mitochondrial DNA: Roles of inherited and somatic mutations. Nat Rev Genet 13:878-90.). However, in comparison with protein-coding genes, tRNA mutations are considered to be less severe, as higher levels (above 90%) are required to cause a biochemical defect (Yoneda et al., 1995Yoneda M, Miyatake T and Attardi G (1995) Heteroplasmic mitochondrial tRNA(Lys) mutation and its complementation in MERRF patient-derived mitochondrial transformants. Muscle Nerve Suppl 3:S95-101.). This finding is in agreement with several works that have provided evidence in support of purifying selection acting in germ cells against deleterious mtDNA mutations (Rand, 2008Rand DM (2008) Mitigating mutational meltdown in mammalian mitochondria. PLoS Biol 6:e35.) (Figure 2). For instance, Stewart and colleagues have shown that mice with a burden of mtDNA mutations are less likely to transmit to offspring non-synonymous changes in protein-coding genes (Stewart et al., 2008Stewart JB, Freyer C, Elson JL, Wredenberg A, Cansu Z, Trifunovic A and Larsson NG (2008) Strong purifying selection in transmission of mammalian mitochondrial DNA. PLoS Biol 6:e10.). In contrast, synonymous substitutions in protein-coding genes and mutations in tRNAs and rRNAs were present at higher levels (Stewart et al., 2008Stewart JB, Freyer C, Elson JL, Wredenberg A, Cansu Z, Trifunovic A and Larsson NG (2008) Strong purifying selection in transmission of mammalian mitochondrial DNA. PLoS Biol 6:e10.). Similar observations have been reported for flies, mice, and humans (Sato et al., 2007Sato A, Nakada K, Shitara H, Kasahara A, Yonekawa H and Hayashi JI (2007) Deletion-mutant mtDNA increases in somatic tissues but decreases in female germ cells with age. Genetics 177:2031-2037.; Fan et al., 2008Fan W, Waymire KG, Narula N, Li P, Rocher C, Coskun PE, Vannan MA, Narula J, Macgregor GR and Wallace DC (2008) A mouse model of mitochondrial disease reveals germline selection against severe mtDNA mutations. Science 319:958-62.; Freyer et al., 2012Freyer C, Cree LM, Mourier A, Stewart JB, Koolmeister C, Milenkovic D, Wai T, Floros VI, Hagström E, Chatzidaki EE et al. (2012) Variation in germline mtDNA heteroplasmy is determined prenatally but modified during subsequent transmission. Nat Genet 44:1282-1285.; Sharpley et al., 2012Sharpley MS, Marciniak C, Eckel-Mahan K, McManus M, Crimi M, Waymire K, Lin CS, Masubuchi S, Friend N, Koike M et al. (2012) Heteroplasmy of mouse mtDNA is genetically unstable and results in altered behavior and cognition. Cell 151:333-43.; Hill et al., 2014Hill JH, Chen Z and Xu H (2014) Selective propagation of functional mitochondrial DNA during oogenesis restricts the transmission of a deleterious mitochondrial variant. Nat Genet 46:389–92.; Ma et al., 2014Ma H, Xu H and O’Farrell PH (2014) Transmission of mitochondrial mutations and action of purifying selection in Drosophila melanogaster. Nat Genet 46:393–7.; Li et al., 2016Li M, Rothwell R, Vermaat M, Wachsmuth M, Schröder R, Laros JFJ, van Oven M, de Bakker PIW, Bovenberg JA, van Duijn CM et al. (2016) Transmission of human mtDNA heteroplasmy in the genome of the Netherlands families: Support for a variable-size bottleneck. Genome Res 26:417–26.; Floros et al., 2018Floros VI, Pyle A, Dietmann S, Wei W, Tang WCW, Irie N, Payne B, Capalbo A, Noli L, Coxhead J et al. (2018) Segregation of mitochondrial DNA heteroplasmy through a developmental genetic bottleneck in human embryos. Nat Cell Biol 20:144–151.; Wei et al., 2019Wei W, Tuna S, Keogh MJ, Smith KR, Aitman TJ, Beales PL, Bennett DL, Gale DP, Bitner-Glindzicz MAK, Black GC et al. (2019) Germline selection shapes human mitochondrial DNA diversity. Science 364:eaau6520.), suggesting a conserved mechanism of purifying selection was established early during evolution. Accordingly, Lieber et al. (2019)Lieber T, Jeedigunta SP, Palozzi JM, Lehmann R and Hurd TR (2019) Mitochondrial fragmentation drives selective removal of deleterious mtDNA in the germline. Nature 570:380–384. recently reported that mitochondrial fragmentation is required to drive selective removal of deleterious mtDNA during early oogenesis in Drosophila. Fragmentation likely enhances association between mitochondrial genotype and phenotype, favoring one genotype over another (Aanen et al., 2014Aanen DK, Spelbrink JN and Beekman M (2014) What cost mitochondria? The maintenance of functional mitochondrial DNA within and across generations. Philos Trans R Soc Lond B Biol Sci 369:20130438.; Haig, 2016Haig D (2016) Intracellular evolution of mitochondrial DNA (mtDNA) and the tragedy of the cytoplasmic commons. BioEssays 38:549-555.). Nonetheless, at least in Drosophila, this mechanism does not rely on autophagic elimination of mutant mtDNA. Instead, mitophagic proteins enable preferential replication of wild-type mtDNA to outcompete their mutant counterparts (Hill et al., 2014Hill JH, Chen Z and Xu H (2014) Selective propagation of functional mitochondrial DNA during oogenesis restricts the transmission of a deleterious mitochondrial variant. Nat Genet 46:389–92.; Ma et al., 2014Ma H, Xu H and O’Farrell PH (2014) Transmission of mitochondrial mutations and action of purifying selection in Drosophila melanogaster. Nat Genet 46:393–7.; Lieber et al., 2019Lieber T, Jeedigunta SP, Palozzi JM, Lehmann R and Hurd TR (2019) Mitochondrial fragmentation drives selective removal of deleterious mtDNA in the germline. Nature 570:380–384.).

Figure 2
Mitochondrial DNA inheritance in somatic and germ cells. Different mitochondria in a single somatic cell (A) are interconnected by constant events of fusion and fission, allowing them to share membranes, solutes, metabolites, proteins, RNAs and DNA (mitochondrial DNA – mtDNA). Hence, when a mutation in mtDNA arises, it can rapidly spread throughout the mitochondrial network. In this case, mutant (red circles) and wild-type (green circles) mtDNAs may co-exist, which is known as heteroplasmy. In comparison, homoplasmic mitochondria contain a single mtDNA genotype, either mutant or wild-type. Unless the mutation level exceeds a critical threshold necessary to cause a biochemical defect (i.e., above 60-90%; red mitochondria), the mutation effect will be masked by wild-type molecules (green mitochondria with both mutant and wild-type mtDNA). In germ cells (B), downregulation of fusion likely minimizes heteroplasmy within mitochondria, enhancing selection at the organellar level (i.e, stronger association between mitochondrial genotype and phenotype). In addition, decreased fusion leads to mitochondrial fragmentation, enhancing mtDNA segregation among embryonic cells. Hence, decreased levels of mtDNA in primordial germ cells (PGCs) makes possible selection at the cellular level (i.e., stronger association between mitochondrial genotype and cellular phenotype). Thus, as a result of selection against deleterious mutations, mature oocyte from the next generation may contain lower levels of mutant mtDNA.

In spite of the mounting evidence in support of a filter against mutant mtDNA in the female germline, this is not a resolved issue. Actually, there are conflicting data arguing against this filter, which has been generating much debate over the topic (Burr et al., 2018Burr SP, Pezet M and Chinnery PF (2018) Mitochondrial DNA heteroplasmy and purifying selection in the mammalian female germ line. Dev Growth Differ 60:21-32.). Other questions involving the issue are: i) why would purifying selection be restricted to germline? ii) can one manipulate selection to avoid the accumulation of mutant mtDNA in somatic tissues? Whilst these questions remain unresolved, it is very likely that the purifying selection behaves differently for different mtDNA mutations and different nuclear genetic backgrounds.

Transmission of metabolic diseases linked to mitochondria dysfunction

Obesity and type II diabetes are currently recognized as the most endemic diseases in the human population. The frequency of these syndromes is increasing over the years; currently, nearly half of worldwide population suffers from obesity (Barnett, 2019Barnett R (2019) Type 2 diabetes. Lancet 394:557.; Blüher, 2019Blüher M (2019) Obesity: Global epidemiology and pathogenesis. Nat Rev Endocrinol 15:288-298.). Obesity and type II diabetes share similar metabolic alterations and are believed to be highly correlated (Volaco et al., 2018Volaco A, Cavalcanti AM, Filho RP and Precoma DB (2018) Socioeconomic status: The missing link between obesity and diabetes mellitus? Curr Diabetes Rev 14:321-326.). Transmission of these diseases to the following generations can occur through both parents, yet the maternal contribution has been shown to be larger (Shankar et al., 2008Shankar K, Harrell A, Liu X, Gilchrist JM, Ronis MJJ and Badger TM (2008) Maternal obesity at conception programs obesity in the offspring. Am J Physiol Regul Integr Comp Physiol 294:528-538.; Jungheim et al., 2010Jungheim ES, Schoeller EL, Marquard KL, Louden ED, Schaffer JE and Moley KH (2010) Diet-induced obesity model: Abnormal oocytes and persistent growth abnormalities in the offspring. Endocrinology 151:4039–4046.; Rattanatray et al., 2010Rattanatray L, MacLaughlin SM, Kleemann DO, Walker SK, Muhlhausler BS and McMillen IC (2010) Impact of maternal periconceptional overnutrition on fat mass and expression of adipogenic and lipogenic genes in visceral and subcutaneous fat depots in the postnatal lamb. Endocrinology 151:5195–5205.; Ruager-Martin et al., 2010Ruager-Martin R, Hyde MJ and Modi N (2010) Maternal obesity and infant outcomes. Early Hum Dev 86:715–722.; Luzzo et al., 2012Luzzo KM, Wang Q, Purcell SH, Chi M, Jimenez PT, Grindler N, Schedl T and Moley KH (2012) High fat diet induced developmental defects in the mouse: Oocyte meiotic aneuploidy and fetal growth retardation/brain defects. PLoS One 7:e0049217.). In humans, for instance, offspring body mass index (BMI) correlated through three generations with maternal but not paternal BMI (Murrin et al., 2012Murrin CM, Kelly GE, Tremblay RE and Kelleher CC (2012) Body mass index and height over three generations: Evidence from the Lifeways cross-generational cohort study. BMC Public Health 12:81.). Likewise, maternal overnutrition in mice leads to offspring that are glucose intolerant and present increased cholesterol and body fat (Jungheim et al., 2010Jungheim ES, Schoeller EL, Marquard KL, Louden ED, Schaffer JE and Moley KH (2010) Diet-induced obesity model: Abnormal oocytes and persistent growth abnormalities in the offspring. Endocrinology 151:4039–4046.). These alterations can last up to the third generation, even when pups are fed a regular diet (Saben et al., 2016Saben JL, Boudoures AL, Asghar Z, Cusumano A, Scheaffer S, Moley KH, Saben JL, Boudoures AL, Asghar Z, Thompson A et al. (2016) Mitochondrial dysfunction via germline changes across three generations maternal metabolic syndrome programs mitochondrial dysfunction via germline changes across three generations. Cell Rep 16:1-8.). Although epigenetic alterations in the nucleus play a major role in the regulation of these effects (Agarwal et al., 2018Agarwal P, Morriseau TS, Kereliuk SM, Doucette CA, Wicklow BA and Dolinsky VW (2018) Maternal obesity, diabetes during pregnancy and epigenetic mechanisms that influence the developmental origins of cardiometabolic disease in the offspring. Crit Rev Clin Lab Sci 55:71-101.; Wang et al., 2018Wang Q, Tang SB, Song XB, Deng TF, Zhang TT, Yin S, Luo SM, Shen W, Zhang CL and Ge ZJ (2018) High-glucose concentrations change DNA methylation levels in human IVM oocytes. Hum Reprod 33:474-481.), other maternal factors have also been taken into account (Wu et al., 2015Wu LL, Russell DL, Wong SL, Chen M, Tsai TS, St John JC, Norman RJ, Febbraio MA, Carroll J and Robker RL (2015) Mitochondrial dysfunction in oocytes of obese mothers: transmission to offspring and reversal by pharmacological endoplasmic reticulum stress inhibitors. Development 142:681-691.; Saben et al., 2016Saben JL, Boudoures AL, Asghar Z, Cusumano A, Scheaffer S, Moley KH, Saben JL, Boudoures AL, Asghar Z, Thompson A et al. (2016) Mitochondrial dysfunction via germline changes across three generations maternal metabolic syndrome programs mitochondrial dysfunction via germline changes across three generations. Cell Rep 16:1-8.).

Among the factors that contribute with maternal transmission of metabolic diseases, mitochondria are a main candidate giving their maternal-exclusive inheritance. In fact, mitochondrial defects in somatic tissues have been associated with obesity, diabetes and cardiovascular disease (Silva et al., 2000Silva JP, Köhler M, Graff C, Oldfors A, Magnuson MA, Berggren PO and Larsson NG (2000) Impaired insulin secretion and β-cell loss in tissue-specific knockout mice with mitochondrial diabetes. Nat Genet 26:336-340.; Sarparanta et al., 2017Sarparanta J, García-Macia M and Singh R (2017) Autophagy and mitochondria in obesity and type 2 diabetes. Curr Diabetes Rev 13:352-369.; Ferey et al., 2019Ferey JLA, Boudoures AL, Reid M, Drury A, Scheaffer S, Modi Z, Kovacs A, Pietka T, DeBosch BJ, Thompson MD et al. (2019) A maternal high-fat, high-sucrose diet induces transgenerational cardiac mitochondrial dysfunction independently of maternal mitochondrial inheritance. Am J Physiol Heart Circ Physiol 316:H1202–H1210.). For instance, mtDNA mutations impacting mitochondrial function and ATP production link with abnormal insulin release and β-cell development, insulin resistance, and diabetes (Poulton et al., 1998Poulton J, Scott Brown M, Cooper A, Marchington DR and Phillips DIW (1998) A common mitochondrial DNA variant is associated with insulin resistance in adult life. Diabetologia 41:54–58.; Silva et al., 2000Silva JP, Köhler M, Graff C, Oldfors A, Magnuson MA, Berggren PO and Larsson NG (2000) Impaired insulin secretion and β-cell loss in tissue-specific knockout mice with mitochondrial diabetes. Nat Genet 26:336-340.; Kaufman et al., 2015Kaufman BA, Li C and Soleimanpour SA (2015) Mitochondrial regulation of β-cell function: Maintaining the momentum for insulin release. Mol Aspects Med 42:91–104.). In this context, Tanaka et al. (2002)Tanaka M, Fuku N, Takeyasu T, Guo LJ, Hirose R, Kurata M, Borgeld HJW, Yamada Y, Maruyama W, Arai Y et al. (2002) Golden mean to longevity: Rareness of mitochondrial cytochrome b variants in centenarians but not in patients with Parkinson’s disease. J Neurosci Res 70:347-355. demonstrated that single nucleotide polymorphisms in mtDNA (mtSNPs) may result in decreased energy expenditure, leading to obesity. Moreover, several studies have associated mtSNPs with type II diabetes and obesity (Rivera et al., 1999Rivera MA, Pérusse L, Gagnon J, Dionne FT, Leon AS, Rao DC, Skinner JS, Wilmore JH, Sjöström L and Bouchard C (1999) A mitochondrial DNA D-loop polymorphism and obesity in three cohorts of women. Int J Obes Relat Metab Disord 23:666–668.; Fuku et al., 2002Fuku N, Oshida Y, Takeyasu T, Guo LJ, Sato Y, Fuku N, Oshida Y, Takeyasu T, Guo LJ, Sato Y et al. (2002) Mitochondrial ATPase subunit 6 and cytochrome b gene polymorphisms in young obese adults. Biochem Biophys Res Commun 290:1199–1205.; Okura et al., 2003Okura T, Koda M, Ando F, Niino N, Tanaka M and Shimokata H (2003) Association of the mitochondrial DNA 15497G/A polymorphism with obesity in a middle-aged and elderly Japanese population. Hum Genet 113:432–6.; Guo et al., 2005Guo LJ, Oshida Y, Fuku N, Takeyasu T, Fujita Y, Kurata M, Sato Y, Ito M and Tanaka M (2005) Mitochondrial genome polymorphisms associated with type-2 diabetes or obesity. Mitochondrion 5:15-33.). These mtSNPs can be located in genes coding for rRNAs, tRNAs, mRNAs (i.e., MT-CYB or MT-ATP6), and even in the non-coding region of mtDNA, the D-loop. Similarly, it was recently described that several mtDNA mutations in tRNAs lead to polycystic ovarian syndrome and metabolic alterations (Ding et al., 2018Ding Y, Xia BH, Zhang CJ and Zhuo GC (2018) Mitochondrial tRNALeu(UUR) C3275T, tRNAGln T4363C and tRNALys A8343G mutations may be associated with PCOS and metabolic syndrome. Gene 642:299–306.), both closely related to type II diabetes and obesity. Altogether, these findings provide evidence that mtDNA mutations may underpin maternal transmission of metabolic diseases.

Apart from mtDNA mutations, mitochondrial damage in oocytes has also been linked with increased risk of metabolic diseases in offspring. Obesity leads to increased lipid content in the follicular fluid, cumulus cells, and oocytes, which in turn damage organelles such as mitochondria and the ER (Wang et al., 2009Wang Q, Ratchford AM, Chi MM, Schoeller E, Frolova A, Schedl T and Moley KH (2009) Maternal diabetes causes mitochondrial dysfunction and meiotic defects in murine oocytes. Mol Endocrinol 23:1603-1612.; Wu et al., 2010Wu LL, Dunning KR, Yang X, Russell DL, Lane M, Norman RJ and Robker RL (2010) High-fat diet causes lipotoxicity responses in cumulus - oocyte complexes and decreased fertilization rates. Endocrinology 151:5438-5445.; Fullston et al., 2015Fullston T, Shehadeh H, Sandeman LY, Kang WX, Wu LL, Robker RL, McPherson NO and Lane M (2015) Female offspring sired by diet induced obese male mice display impaired blastocyst development with molecular alterations to their ovaries, oocytes and cumulus cells. J Assist Reprod Genet 32:725-735.; Ruebel et al., 2017Ruebel ML, Cotter M, Sims CR, Moutos DM, Badger TM, Cleves MA, Shankar K and Andres A (2017) Obesity modulates inflammation and lipidmetabolism oocyte gene expression: A single-cell transcriptome perspective. J Clin Endocrinol Metab 102:2029–2038.). Impaired ER function can lead to activation of the unfolded protein response (UPR) and Ca+2 release, further disrupting mitochondrial function (i.e., decreased Δψm and increased ROS) and oocyte homeostasis (Wu et al., 2010Wu LL, Dunning KR, Yang X, Russell DL, Lane M, Norman RJ and Robker RL (2010) High-fat diet causes lipotoxicity responses in cumulus - oocyte complexes and decreased fertilization rates. Endocrinology 151:5438-5445., 2015Wu LL, Russell DL, Wong SL, Chen M, Tsai TS, St John JC, Norman RJ, Febbraio MA, Carroll J and Robker RL (2015) Mitochondrial dysfunction in oocytes of obese mothers: transmission to offspring and reversal by pharmacological endoplasmic reticulum stress inhibitors. Development 142:681-691.; Luzzo et al., 2012Luzzo KM, Wang Q, Purcell SH, Chi M, Jimenez PT, Grindler N, Schedl T and Moley KH (2012) High fat diet induced developmental defects in the mouse: Oocyte meiotic aneuploidy and fetal growth retardation/brain defects. PLoS One 7:e0049217.; Hou et al., 2016Hou YJ, Zhu CC, Duan X, Liu HL, Wang Q and Sun SC (2016) Both diet and gene mutation induced obesity affect oocyte quality in mice. Sci Rep 6:1–10.). Besides impacting oocyte competence and fertility (Wu et al., 2015Wu LL, Russell DL, Wong SL, Chen M, Tsai TS, St John JC, Norman RJ, Febbraio MA, Carroll J and Robker RL (2015) Mitochondrial dysfunction in oocytes of obese mothers: transmission to offspring and reversal by pharmacological endoplasmic reticulum stress inhibitors. Development 142:681-691.; Pasquariello et al., 2019Pasquariello R, Ermisch AF, Silva E, McCormick S, Logsdon D, Barfield JP, Schoolcraft WB and Krisher RL (2019) Alterations in oocyte mitochondrial number and function are related to spindle defects and occur with maternal aging in mice and humans. Biol Reprod 100:971–981.), these mitochondrial abnormalities can be passed down to the following generations, increasing their risk to develop metabolic diseases (Saben et al., 2016Saben JL, Boudoures AL, Asghar Z, Cusumano A, Scheaffer S, Moley KH, Saben JL, Boudoures AL, Asghar Z, Thompson A et al. (2016) Mitochondrial dysfunction via germline changes across three generations maternal metabolic syndrome programs mitochondrial dysfunction via germline changes across three generations. Cell Rep 16:1-8.). Hence, mice born to pregnant females under a high-fat/high-sucrose diet have impaired peripheral insulin signaling which associates with abnormal mitochondrial function and dynamics in skeletal muscle up to the third generation (Saben et al., 2016Saben JL, Boudoures AL, Asghar Z, Cusumano A, Scheaffer S, Moley KH, Saben JL, Boudoures AL, Asghar Z, Thompson A et al. (2016) Mitochondrial dysfunction via germline changes across three generations maternal metabolic syndrome programs mitochondrial dysfunction via germline changes across three generations. Cell Rep 16:1-8.). Similar mitochondrial abnormalities were present in oocytes from the first and second generations, even though these were fed a regular diet (Saben et al., 2016Saben JL, Boudoures AL, Asghar Z, Cusumano A, Scheaffer S, Moley KH, Saben JL, Boudoures AL, Asghar Z, Thompson A et al. (2016) Mitochondrial dysfunction via germline changes across three generations maternal metabolic syndrome programs mitochondrial dysfunction via germline changes across three generations. Cell Rep 16:1-8.). Therefore, apart from epigenetic alterations in the nucleus, mitochondria also contribute with the metabolic programing resulting from maternal overnutrition. Given that epigenetic marks in mtDNA regulate expression of this genome (Kobayashi et al., 2012Kobayashi H, Sakurai T, Imai M, Takahashi N, Fukuda A, Yayoi O, Sato S, Nakabayashi K, Hata K, Sotomaru Y et al. (2012) Contribution of intragenic DNA methylation in mouse gametic DNA methylomes to establish Oocyte-specific heritable marks. PLoS Genet 8:e1002440.; Sun et al., 2013Sun Z, Terragni J, Borgaro JG, Liu Y, Yu L, Guan S, Wang H, Sun D, Cheng X, Zhu Z et al. (2013) High-resolution enzymatic mapping of genomic 5-Hydroxymethylcytosine in mouse embryonic stem cells. Cell Rep 3:567-576.; Sirard, 2019Sirard MA (2019) Distribution and dynamics of mitochondrial DNA methylation in oocytes, embryos and granulosa cells. Sci Rep 9:11937.), it remains to be investigated whether these can also explain maternal transmission of metabolic diseases.

Treatment options for preventing mitochondrial disease transmission

Due to the poor understanding of the mechanisms regulating transmission of mitochondria-related diseases, there are few treatment options available to prevent their inheritance to the following generations (Craven et al., 2017Craven L, Alston CL, Taylor RW and Turnbull DM (2017) Recent advances in mitochondrial disease. Annu Rev Genomics Hum Genet 18:257-275.). With respect to non-genetic alterations in mitochondria, the oocyte might benefit from treatments performed before fertilization, during the in vitro maturation. The idea is to expose the oocyte for a period of ~24 h to drugs such as L-carnitine, rosiglitazone, salubrinal, or BGP-15, which potentially enhance mitochondria activity, decrease lipid content, and mitigate ER stress. In fact, treatments involving one or more of these drugs have been shown to mitigate the defects in the oocyte and the next generation (Wu et al., 2010Wu LL, Dunning KR, Yang X, Russell DL, Lane M, Norman RJ and Robker RL (2010) High-fat diet causes lipotoxicity responses in cumulus - oocyte complexes and decreased fertilization rates. Endocrinology 151:5438-5445., 2015Wu LL, Russell DL, Wong SL, Chen M, Tsai TS, St John JC, Norman RJ, Febbraio MA, Carroll J and Robker RL (2015) Mitochondrial dysfunction in oocytes of obese mothers: transmission to offspring and reversal by pharmacological endoplasmic reticulum stress inhibitors. Development 142:681-691.; Dunning and Robker, 2012Dunning KR and Robker RL (2012) Promoting lipid utilization with l-carnitine to improve oocyte quality. Anim Reprod Sci 134:69-75.; Liang et al., 2017Liang LF, Qi ST, Xian YX, Huang L, Sun XF and Wang WH (2017) Protective effect of antioxidants on the pre-maturation aging of mouse oocytes. Sci Rep 7:1434.). However, a major challenge in making these treatments available is to overcome the side effects of in vitro maturation (Lonergan and Fair, 2016Lonergan P and Fair T (2016) Maturation of oocytes in vitro. Ann Rev Anim Biosci 4:255–268.; Yang and Chian, 2017Yang ZY and Chian RC (2017) Development of invitro maturation techniques for clinical applications. Fertil Steril 108:577-584.). Given this is a critical period of oocyte development, which encompasses meiotic resumption from prophase I (dictyate) to metaphase II, any perturbation in oocyte homeostasis may lead to mis-segregation of chromosomes and aneuploidy (Greaney et al., 2017Greaney J, Wei Z and Homer H (2017) Regulation of chromosome segregation in oocytes and the cellular basis for female meiotic errors. Hum Reprod Update 10.1093/humupd/dmx035.; Danadova et al., 2017Danadova J, Matijescukova N, Danylevska AMG and Anger M (2017) Increased frequency of chromosome congression defects and aneuploidy in mouse oocytes cultured at lower temperature. Reprod Fertil Dev 29:968.). In addition, in vitro maturation on its own leads to metabolic alterations that mimic those of oocytes from obese donors (i.e., mitochondrial dysfunction and increased lipid content), potentially impacting the next generation (Farin et al., 2006Farin PW, Piedrahita JA and Farin CE (2006) Errors in development of fetuses and placentas from in vitro-produced bovine embryos. Theriogenology 65:178–91.; Li et al., 2014Li H, Jia GH, Lu XL, Zhang G, Tian KY, Li JT and Zhang JM (2014) In vitro maturation of oocytes is not a risk factor for adult metabolic syndrome of mouse offspring. Eur J Obstet Gynecol Reprod Biol 174:96–99.; del Collado et al., 2017adel Collado M, da Silveira JC, Oliveira MLF, Alves BMSM, Simas RC, Godoy AT, Coelho MB, Marques LA, Carriero MM, Nogueira MFG et al. (2017a) In vitro maturation impacts cumulus–oocyte complex metabolism and stress in cattle. Reproduction 154:881–893.; del Collado et al., 2017b; Wang et al., 2018del Collado M, da Silveira JC, Sangalli JR, Andrade GM, Sousa LRDS, Silva LA, Meirelles FV and Perecin F (2017b) Fatty acid binding protein 3 and transzonal projections are involved in lipid accumulation during in vitro maturation of bovine oocytes. Sci Rep 7:2645.). Thus, these alternatives are not currently available in humans.

An alternative option to treat oocytes harboring mitochondria abnormalities, particularly those caused by mtDNA mutations, is known as mitochondrial replacement therapy (MRT; Figure 3). This method involves replacement of abnormal mitochondria in the oocyte by functional ones provided by a donated oocyte (Wolf et al., 2017Wolf DP, Hayama T and Mitalipov S (2017) Mitochondrial genome inheritance and replacement in the human germline. EMBO J 36:2177-2181.). More specifically, ovulated oocytes at the metaphase-II stage are collected from both the patient and a “healthy” donor not containing mitochondrial abnormalities. With the aid of a micromanipulation set, the spindle from the donated oocyte is replaced by the patient’s spindle. The resulting oocyte containing the patient’s spindle and donated mitochondria is then fertilized to allow development to term. Provided that the large majority of mitochondria is replaced by donated ones, MRT has virtually the potential to prevent transmission of mitochondrial diseases. Yet, ~1% of mitochondria from the patient’s oocyte are transferred along with the spindle. This level can be even higher (up to 4%) when pronuclear zygotes are used instead of metaphase-II oocytes, which can lead in ~15% of cases to a reversal back to the patient’s mtDNA (Hyslop et al., 2016Hyslop LA, Blakeley P, Craven L, Richardson J, Fogarty NME, Fragouli E, Lamb M, Wamaitha SE, Prathalingam N, Zhang Q et al. (2016) Towards clinical application of pronuclear transfer to prevent mitochondrial DNA disease. Nature 534:383–386.; Kang et al., 2016Kang E, Wu J, Gutierrez NM, Koski A, Tippner-Hedges R, Agaronyan K, Platero-Luengo A, Martinez-Redondo P, Ma H, Lee Y et al. (2016) Mitochondrial replacement in human oocytes carrying pathogenic mitochondrial DNA mutations. Nature 540:270–275.). Although hard to explain, rapid mtDNA segregation and bottleneck during preimplantation development might account for these quick shifts in mtDNA genotype (Lee et al., 2012aLee HS, Ma H, Juanes RC, Tachibana M, Sparman M, Woodward J, Ramsey C, Xu J, Kang EJ, Amato P et al. (2012a) Rapid mitochondrial DNA segregation in primate preimplantation embryos precedes somatic and germline bottleneck. Cell Rep 1:506–15.; Freyer et al., 2012Freyer C, Cree LM, Mourier A, Stewart JB, Koolmeister C, Milenkovic D, Wai T, Floros VI, Hagström E, Chatzidaki EE et al. (2012) Variation in germline mtDNA heteroplasmy is determined prenatally but modified during subsequent transmission. Nat Genet 44:1282-1285.). Alternatively, it has been proposed that a specific population of mtDNA is tagged in oocytes (i.e., from spindle-surrounding mitochondria) for replication during early development (Wolf et al., 2017Wolf DP, Hayama T and Mitalipov S (2017) Mitochondrial genome inheritance and replacement in the human germline. EMBO J 36:2177-2181.). No matter the mechanism underlying these unexpected results, they highlight the need for careful studies before the clinical practice of MRT (Wolf et al., 2017Wolf DP, Hayama T and Mitalipov S (2017) Mitochondrial genome inheritance and replacement in the human germline. EMBO J 36:2177-2181.; Craven et al., 2018Craven L, Murphy J, Turnbull DM, Taylor RW, Gorman GS and McFarland R (2018) Scientific and ethical issues in mitochondrial donation. New Bioeth 24:57-73.).

Figure 3
New technologies for preventing inheritance of mitochondrial diseases. The mitochondrial replacement therapy (MRT; A) proposes the replacement of a patient’s mitochondria in oocytes by donor mitochondria. Towards that, mature oocytes arrested at the metaphase-II stage are collected from the patient and a donor. While the patient’s oocytes are supposed to contain mutant (red) mitochondrial DNA (mtDNA), donor oocytes should contain only wild-type (green) mtDNA. Next, the spindle is removed from the patient’s oocyte (donor karyoplast) to be injected into the donor oocyte from which the spindle was previously removed (donor cytoplast). Fertilization of the reconstructed oocyte should lead to a blastocyst, which can be used for embryonic stem cell (ESC) derivation. Although MRT allows transplantation of karyoplast with minimal carryover (~1%) of mutant mtDNA, recent data have provided evidence of a reversal in ESCs back to 100% mutant mtDNA (Hyslop et al., 2016Hyslop LA, Blakeley P, Craven L, Richardson J, Fogarty NME, Fragouli E, Lamb M, Wamaitha SE, Prathalingam N, Zhang Q et al. (2016) Towards clinical application of pronuclear transfer to prevent mitochondrial DNA disease. Nature 534:383–386.; Kang et al., 2016Kang E, Wu J, Gutierrez NM, Koski A, Tippner-Hedges R, Agaronyan K, Platero-Luengo A, Martinez-Redondo P, Ma H, Lee Y et al. (2016) Mitochondrial replacement in human oocytes carrying pathogenic mitochondrial DNA mutations. Nature 540:270–275.). An alternative strategy to MRT is the nuclease-mediated elimination of mutant mtDNA (B), which relies on the use of mitochondrial-targeted restriction endonucleases (mito-TALENs). These nucleases are designed to selectively cut mutant mtDNA, but not wild-type molecules. However, ~10% of targeted molecules were shown to be left uncut in newborns after use of mito-TALENs (Reddy et al., 2015Reddy P, Ocampo A, Suzuki K, Luo J, Bacman SR, Williams SL, Sugawara A, Okamura D, Tsunekawa Y, Wu J et al. (2015) Selective elimination of mitochondrial mutations in the germline by genome editing. Cell 161:459).

With the advances in genome editing technologies, another potential strategy to prevent transmission of mitochondrial abnormalities is the targeted elimination of mutant mtDNA in oocytes or early embryos (Figure 3). As a proof of concept, Reddy et al. (2015)Reddy P, Ocampo A, Suzuki K, Luo J, Bacman SR, Williams SL, Sugawara A, Okamura D, Tsunekawa Y, Wu J et al. (2015) Selective elimination of mitochondrial mutations in the germline by genome editing. Cell 161:459 used mitochondrial-targeted restriction endonucleases (mito-TALENs) to selectively eliminate mutant mtDNA in mice and humans. Although this strategy proved efficient, ~10% of targeted molecules (i.e., mutant mtDNA) were left in oocytes, embryos and offspring produced after the use of mito-TALENs. Moreover, given that the mtDNA is not replicated during early embryogenesis (Pikó and Taylor, 1987Pikó L and Taylor KD (1987) Amounts of mitochondrial DNA and abundance of some mitochondrial gene transcripts in early mouse embryos. Dev Biol 123:364–374.; Thundathil et al., 2005Thundathil J, Filion F and Smith LC (2005) Molecular control of mitochondrial function in preimplantation mouse embryos. Mol Reprod Dev 71:405-13.; Cree et al., 2008Cree LM, Samuels DC, Sousa Lopes SC, Rajasimha HK, Wonnapinij P, Mann JR, Dahl H-HM and Chinnery PF (2008) A reduction of mitochondrial DNA molecules during embryogenesis explains the rapid segregation of genotypes. Nat Genet 40:249-54.), the use of mito-TALENs resulted in mtDNA-depleted embryos (Reddy et al., 2015Reddy P, Ocampo A, Suzuki K, Luo J, Bacman SR, Williams SL, Sugawara A, Okamura D, Tsunekawa Y, Wu J et al. (2015) Selective elimination of mitochondrial mutations in the germline by genome editing. Cell 161:459). Although in the newborns the content of mtDNA was normal (Reddy et al., 2015Reddy P, Ocampo A, Suzuki K, Luo J, Bacman SR, Williams SL, Sugawara A, Okamura D, Tsunekawa Y, Wu J et al. (2015) Selective elimination of mitochondrial mutations in the germline by genome editing. Cell 161:459), the lower levels of mtDNA (and likely of mitochondria too) in oocytes and embryos could lead to poorer developmental rates (Wai et al., 2010Wai T, Ao A, Zhang X, Cyr D, Dufort D and Shoubridge EA (2010) The role of mitochondrial DNA copy number in mammalian fertility. Biol Reprod 83:52-62.). Based on these uncertainties, mito-TALENs are not currently taken as a viable alternative to prevent transmission of mtDNA-linked diseases (Wolf et al., 2017Wolf DP, Hayama T and Mitalipov S (2017) Mitochondrial genome inheritance and replacement in the human germline. EMBO J 36:2177-2181.).

Final considerations

Mitochondrial abnormalities have been linked with maternal transmission of important diseases in humans. Among these, mtDNA mutations in oocytes can be transmitted to the following generations and cause severe diseases. In addition, maternal obesity damages mitochondria in oocytes, leading to poor fertility and increased risk of metabolic diseases in offspring. Understanding how mitochondrial abnormalities are established and transmitted are of fundamental importance to mitigate their incidence in the human population. Moreover, treatment options involving manipulation of oocytes and early embryos are currently under consideration and may become available in the future to prevent transmission of mitochondria-associated diseases.

Conflict of Interests

The authors declare that there is no conflict of interest that could be perceived as prejudicial to the impartiality of the reported research.

Author contributions

MRC and MDC conceived the study. MRC and MDC reviewed previous publications. MRC, CHM, JDAN, MPG, AKP, FP and MDC wrote the manuscript. All authors read and approved the final version.

Acknowledgments

We would like to thank the São Paulo Research Foundation (FAPESP – grant # 2016/07868-4, 2017/05899-2, 2017/19825-0, 2017/25916-9 and 2018/13155-6) and the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior – Brazil (CAPES – finance code 001).

References

  • Aanen DK, Spelbrink JN and Beekman M (2014) What cost mitochondria? The maintenance of functional mitochondrial DNA within and across generations. Philos Trans R Soc Lond B Biol Sci 369:20130438.
  • Agarwal P, Morriseau TS, Kereliuk SM, Doucette CA, Wicklow BA and Dolinsky VW (2018) Maternal obesity, diabetes during pregnancy and epigenetic mechanisms that influence the developmental origins of cardiometabolic disease in the offspring. Crit Rev Clin Lab Sci 55:71-101.
  • Arhin SK, Lu J, Xi H and Jin X (2018) Energy requirements in mammalian oogenesis. Cell Mol Biol 64:12-19.
  • Ashley MV, Laipis PJ and Hauswirth WW (1989) Rapid segregation of heteroplasmic bovine mitochondria. Nucleic Acids Res 17:7325-7331.
  • Bach D, Pich S, Soriano FX, Vega N, Baumgartner B, Oriola J, Daugaard JR, Lloberas J, Camps M, Zierath JR et al. (2003) Mitofusin-2 determines mitochondrial network architecture and mitochondrial metabolism: A novel regulatory mechanism altered in obesity. J Biol Chem 278:17190-17197.
  • Barnett R (2019) Type 2 diabetes. Lancet 394:557.
  • Battersby BJ, Loredo-Osti JC and Shoubridge EA (2003) Nuclear genetic control of mitochondrial DNA segregation. Nat Genet 33:183-186.
  • Betz C, Stracka D, Prescianotto-baschong C, Frieden M and Demaurex N (2013) mTOR complex 2-Akt signaling at mitochondria-associated endoplasmic reticulum membranes (MAM) regulates mitochondrial physiology. Proc Nat Acad Sci U S A 110:12526-12534.
  • Blüher M (2019) Obesity: Global epidemiology and pathogenesis. Nat Rev Endocrinol 15:288-298.
  • Burgstaller JP, Kolbe T, Havlicek V, Hembach S, Poulton J, Piálek J, Steinborn R, Rülicke T, Brem G, Jones NS et al. (2018) Large-scale genetic analysis reveals mammalian mtDNA heteroplasmy dynamics and variance increase through lifetimes and generations. Nat Commun 9:1-12.
  • Burr SP, Pezet M and Chinnery PF (2018) Mitochondrial DNA heteroplasmy and purifying selection in the mammalian female germ line. Dev Growth Differ 60:21-32.
  • Carreras-Sureda A, Pihán P and Hetz C (2017) The unfolded protein response: At the intersection between endoplasmic reticulum function and mitochondrial bioenergetics. Front Oncol 7:1-7.
  • Chen H, Detmer SA, Ewald AJ, Griffin EE, Fraser SE and Chan DC (2003) Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J Cell Biol 160:189-200.
  • Chen H, Chomyn A and Chan DC (2005) Disruption of fusion results in mitochondrial heterogeneity and dysfunction. J Biol Chem 280:26185-92.
  • Chen H, McCaffery JM and Chan DC (2007) Mitochondrial fusion protects against neurodegeneration in the cerebellum. Cell 130:548-62.
  • Chen H, Vermulst M, Wang YE, Chomyn A, Prolla TA, McCaffery JM and Chan DC (2010) Mitochondrial fusion is required for mtDNA stability in skeletal muscle and tolerance of mtDNA mutations. Cell 141:280-9.
  • Chen KH, Dasgupta A, Ding J, Indig FE, Ghosh P and Longo DL (2014) Role of mitofusin 2 (Mfn2) in controlling cellular proliferation. FASEB J 28:382-394.
  • Chen XJ (2013) Mechanism of homologous recombination and implications for aging-related deletions in mitochondrial DNA. Microbiol Mol Biol Rev 77:476–496.
  • Chiaratti MR and Meirelles FV (2010) Mitochondrial DNA copy number, a marker of viability for oocytes. Biol Reprod 83:1-2.
  • Chiaratti MR, Garcia BM, Carvalho KF, Machado TS, Ribeiro FKDS and Macabelli CH (2018) The role of mitochondria in the female germline: Implications to fertility and inheritance of mitochondrial diseases. Cell Biol Int 42:1-39.
  • Cipolat S, de Brito OM, Dal Zilio B and Scorrano L (2004) OPA1 requires mitofusin 1 to promote mitochondrial fusion. Proc Nat Acad Sci U S A 101:15927-15932.
  • Clarke HJ (2017) Regulation of germ cell development by intercellular signaling in the mammalian ovarian follicle. Wiley Interdiscip Rev Dev Biol 7:e294.
  • Craven L, Alston CL, Taylor RW and Turnbull DM (2017) Recent advances in mitochondrial disease. Annu Rev Genomics Hum Genet 18:257-275.
  • Craven L, Murphy J, Turnbull DM, Taylor RW, Gorman GS and McFarland R (2018) Scientific and ethical issues in mitochondrial donation. New Bioeth 24:57-73.
  • Cree LM, Samuels DC, Sousa Lopes SC, Rajasimha HK, Wonnapinij P, Mann JR, Dahl H-HM and Chinnery PF (2008) A reduction of mitochondrial DNA molecules during embryogenesis explains the rapid segregation of genotypes. Nat Genet 40:249-54.
  • Danadova J, Matijescukova N, Danylevska AMG and Anger M (2017) Increased frequency of chromosome congression defects and aneuploidy in mouse oocytes cultured at lower temperature. Reprod Fertil Dev 29:968.
  • de Brito OM and Scorrano L (2008) Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature 456:605-610.
  • de Brito OM and Scorrano L (2009) Mitofusin-2 regulates mitochondrial and endoplasmic reticulum morphology and tethering: The role of Ras. Mitochondrion 9:222-226.
  • del Collado M, da Silveira JC, Oliveira MLF, Alves BMSM, Simas RC, Godoy AT, Coelho MB, Marques LA, Carriero MM, Nogueira MFG et al. (2017a) In vitro maturation impacts cumulus–oocyte complex metabolism and stress in cattle. Reproduction 154:881–893.
  • del Collado M, da Silveira JC, Sangalli JR, Andrade GM, Sousa LRDS, Silva LA, Meirelles FV and Perecin F (2017b) Fatty acid binding protein 3 and transzonal projections are involved in lipid accumulation during in vitro maturation of bovine oocytes. Sci Rep 7:2645.
  • Ding Y, Xia BH, Zhang CJ and Zhuo GC (2018) Mitochondrial tRNALeu(UUR) C3275T, tRNAGln T4363C and tRNALys A8343G mutations may be associated with PCOS and metabolic syndrome. Gene 642:299–306.
  • Dunning KR and Robker RL (2012) Promoting lipid utilization with l-carnitine to improve oocyte quality. Anim Reprod Sci 134:69-75.
  • Eschbach J, Sinniger J, Bouitbir J, Fergani A, Schlagowski A-I, Zoll J, Geny B, René F, Larmet Y, Marion V et al. (2013) Dynein mutations associated with hereditary motor neuropathies impair mitochondrial morphology and function with age. Neurobiol Dis 58:220-30.
  • Eura Y, Ishihara N, Yokota S and Mihara K (2003) Two mitofusin proteins, mammalian homologues of FZO, with distinct functions are both required for mitochondrial fusion. J Biochem 134:333-344.
  • Fan W, Waymire KG, Narula N, Li P, Rocher C, Coskun PE, Vannan MA, Narula J, Macgregor GR and Wallace DC (2008) A mouse model of mitochondrial disease reveals germline selection against severe mtDNA mutations. Science 319:958-62.
  • Farin PW, Piedrahita JA and Farin CE (2006) Errors in development of fetuses and placentas from in vitro-produced bovine embryos. Theriogenology 65:178–91.
  • Ferey JLA, Boudoures AL, Reid M, Drury A, Scheaffer S, Modi Z, Kovacs A, Pietka T, DeBosch BJ, Thompson MD et al. (2019) A maternal high-fat, high-sucrose diet induces transgenerational cardiac mitochondrial dysfunction independently of maternal mitochondrial inheritance. Am J Physiol Heart Circ Physiol 316:H1202–H1210.
  • Ferreira CR, Burgstaller JP, Perecin F, Garcia JM, Chiaratti MR, Méo SC, Müller M, Smith LC, Meirelles FV and Steinborn R (2010) Pronounced segregation of donor mitochondria introduced by bovine ooplasmic transfer to the female germ-line. Biol Reprod 82:563-71.
  • Filadi R, Greotti E, Turacchio G, Luini A, Pozzan T and Pizzo P (2015) Mitofusin 2 ablation increases endoplasmic reticulum-mitochondria coupling. Proc Nat Acad Sci U S A 112:E2174-81.
  • Floros VI, Pyle A, Dietmann S, Wei W, Tang WCW, Irie N, Payne B, Capalbo A, Noli L, Coxhead J et al. (2018) Segregation of mitochondrial DNA heteroplasmy through a developmental genetic bottleneck in human embryos. Nat Cell Biol 20:144–151.
  • Ford WCL (2004) Regulation of sperm function by reactive oxygen species. Hum Reprod Update 10:387-399.
  • Freyer C, Cree LM, Mourier A, Stewart JB, Koolmeister C, Milenkovic D, Wai T, Floros VI, Hagström E, Chatzidaki EE et al. (2012) Variation in germline mtDNA heteroplasmy is determined prenatally but modified during subsequent transmission. Nat Genet 44:1282-1285.
  • Fuku N, Oshida Y, Takeyasu T, Guo LJ, Sato Y, Fuku N, Oshida Y, Takeyasu T, Guo LJ, Sato Y et al. (2002) Mitochondrial ATPase subunit 6 and cytochrome b gene polymorphisms in young obese adults. Biochem Biophys Res Commun 290:1199–1205.
  • Fullston T, Shehadeh H, Sandeman LY, Kang WX, Wu LL, Robker RL, McPherson NO and Lane M (2015) Female offspring sired by diet induced obese male mice display impaired blastocyst development with molecular alterations to their ovaries, oocytes and cumulus cells. J Assist Reprod Genet 32:725-735.
  • Garesse R and Vallejo CG (2001) Animal mitochondrial biogenesis and function: A regulatory cross-talk between two genomes. Gene 263:1-16.
  • Gorman GS, Schaefer AM, Ng Y, Gomez N, Blakely EL, Alston CL, Feeney C, Horvath R, Yu-Wai-Man P, Chinnery PF et al. (2015) Prevalence of nuclear and mitochondrial DNA mutations related to adult mitochondrial disease. Ann Neurol 77:753-759.
  • Greaney J, Wei Z and Homer H (2017) Regulation of chromosome segregation in oocytes and the cellular basis for female meiotic errors. Hum Reprod Update 10.1093/humupd/dmx035.
  • Griparic L, Van Der Wel NN, Orozco IJ, Peters PJ and Van Der Bliek AM (2004) Loss of the intermembrane space protein Mgm1/OPA1 induces swelling and localized constrictions along the lengths of mitochondria. J Biol Chem 279:18792-18798.
  • Guo LJ, Oshida Y, Fuku N, Takeyasu T, Fujita Y, Kurata M, Sato Y, Ito M and Tanaka M (2005) Mitochondrial genome polymorphisms associated with type-2 diabetes or obesity. Mitochondrion 5:15-33.
  • Hagström E, Freyer C, Battersby BJ, Stewart JB and Larsson NG (2014) No recombination of mtDNA after heteroplasmy for 50 generations in the mouse maternal germline. Nucleic Acids Res 42:1111-1116.
  • Haig D (2016) Intracellular evolution of mitochondrial DNA (mtDNA) and the tragedy of the cytoplasmic commons. BioEssays 38:549-555.
  • Halsne R, Esbensen Y, Wang W, Scheffler K, Suganthan R, Bjørås M and Eide L (2012) Lack of the DNA glycosylases MYH and OGG1 in the cancer prone double mutant mouse does not increase mitochondrial DNA mutagenesis. DNA Repair 11:278-285.
  • Hamasaki M, Furuta N, Matsuda A, Nezu A, Yamamoto A, Fujita N, Oomori H, Noda T, Haraguchi T, Hiraoka Y et al. (2013) Autophagosomes form at ER–mitochondria contact sites. Nature 495:389-393.
  • Hashimoto S, Morimoto N, Yamanaka M, Matsumoto H, Yamochi T, Goto H, Inoue M, Nakaoka Y, Shibahara H and Morimoto Y (2017) Quantitative and qualitative changes of mitochondria in human preimplantation embryos. J Assist Reprod Genet 34:573-580.
  • Hauswirth WW and Laipis PJ (1982) Mitochondrial DNA polymorphism in a maternal lineage of Holstein cows. Proc Natl Acad Sci U S A 79:4686-4690.
  • Hauswirth WW, Van de Walle MJ, Laipis PJ and Olivo PD (1984) Heterogeneous mitochondrial DNA D-loop sequences in bovine tissue. Cell 37:1001–1007.
  • Hill JH, Chen Z and Xu H (2014) Selective propagation of functional mitochondrial DNA during oogenesis restricts the transmission of a deleterious mitochondrial variant. Nat Genet 46:389–92.
  • Hou X, Zhu S, Zhang H, Li C, Qiu D, Ge J, Guo X and Wang Q (2019) Mitofusin1 in oocyte is essential for female fertility. Redox Biol 21:101110.
  • Hou YJ, Zhu CC, Duan X, Liu HL, Wang Q and Sun SC (2016) Both diet and gene mutation induced obesity affect oocyte quality in mice. Sci Rep 6:1–10.
  • Hyslop LA, Blakeley P, Craven L, Richardson J, Fogarty NME, Fragouli E, Lamb M, Wamaitha SE, Prathalingam N, Zhang Q et al. (2016) Towards clinical application of pronuclear transfer to prevent mitochondrial DNA disease. Nature 534:383–386.
  • Ishihara N, Eura Y and Mihara K (2004) Mitofusin 1 and 2 play distinct roles in mitochondrial fusion reactions via GTPase activity. J Cell Sci 117:6535–6546.
  • Ishihara N, Nomura M, Jofuku A, Kato H, Suzuki SO, Masuda K, Otera H, Nakanishi Y, Nonaka I, Goto Y-I et al. (2009) Mitochondrial fission factor Drp1 is essential for embryonic development and synapse formation in mice. Nat Cell Biol 11:958–66.
  • Itsara LS, Kennedy SR, Fox EJ, Yu S, Hewitt JJ, Sanchez-Contreras M, Cardozo-Pelaez F and Pallanck LJ (2014) Oxidative stress is not a major contributor to somatic mitochondrial DNA mutations. PLoS Genet 10:1003974.
  • Jansen RPS and De Boer K (1998) The bottleneck: Mitochondrial imperatives in oogenesis and ovarian follicular fate. Mol Cellular Endocrinol 145:81–88.
  • Jenuth J, Peterson A, Fu K and Shoubridge E (1996) Random genetic drift in the female germline explains the rapid segregation of mammalian mitochondrial DNA. Nat Genet 14:146–151.
  • Johnson AA and Johnson KA (2001) Exonuclease proofreading by human mitochondrial DNA polymerase. J Biol Chem 276:38097–107.
  • Johnson MT, Freeman EA, Gardner DK and Hunt PA (2007) Oxidative metabolism of pyruvate is required for meiotic maturation of murine oocytes in vivo. Biol Reprod 77:2–8.
  • Jungheim ES, Schoeller EL, Marquard KL, Louden ED, Schaffer JE and Moley KH (2010) Diet-induced obesity model: Abnormal oocytes and persistent growth abnormalities in the offspring. Endocrinology 151:4039–4046.
  • Kang E, Wu J, Gutierrez NM, Koski A, Tippner-Hedges R, Agaronyan K, Platero-Luengo A, Martinez-Redondo P, Ma H, Lee Y et al. (2016) Mitochondrial replacement in human oocytes carrying pathogenic mitochondrial DNA mutations. Nature 540:270–275.
  • Kaufman BA, Li C and Soleimanpour SA (2015) Mitochondrial regulation of β-cell function: Maintaining the momentum for insulin release. Mol Aspects Med 42:91–104.
  • Kauppila TES, Kauppila JHK and Larsson NG (2017) Mammalian mitochondria and aging: An update. Cell Metab 25:57–71.
  • Kazak L, Reyes A and Holt IJ (2012) Minimizing the damage: Repair pathways keep mitochondrial DNA intact. Nat Rev Mol Cell Biol 13:659–671.
  • Kennedy SR, Salk JJ, Schmitt MW and Loeb LA (2013) Ultra-sensitive sequencing reveals an age-related increase in somatic mitochondrial mutations that are inconsistent with oxidative damage. PLoS Genet 9:e1003794.
  • Kim KH, Park JH, Kim EY, Ko JJ, Park KS and Lee KA (2016) The role of Rad51 in safeguarding mitochondrial activity during the meiotic cell cycle in mammalian oocytes. Sci Rep 6:34110.
  • Kobayashi H, Sakurai T, Imai M, Takahashi N, Fukuda A, Yayoi O, Sato S, Nakabayashi K, Hata K, Sotomaru Y et al. (2012) Contribution of intragenic DNA methylation in mouse gametic DNA methylomes to establish Oocyte-specific heritable marks. PLoS Genet 8:e1002440.
  • Lee HS, Ma H, Juanes RC, Tachibana M, Sparman M, Woodward J, Ramsey C, Xu J, Kang EJ, Amato P et al. (2012a) Rapid mitochondrial DNA segregation in primate preimplantation embryos precedes somatic and germline bottleneck. Cell Rep 1:506–15.
  • Lee H, Ma H, Juanes R, Tachibana M, Sparman M, Woodward J, Ramsey C, Xy J, Kand EJ, Amato P et al. (2012b) Rapid mitochondrial DNA segregation in primate preimplantation embryos precedes somatic and germline bottleneck. Cell Rep 1:506–515.
  • Leese HJ (2012) Metabolism of the preimplantation embryo: 40 Years on. Reproduction 143:417–427.
  • Leitch HG, Tang WWC and Surani MA (2013) Primordial germ-cell development and epigenetic reprogramming in mammals. Curr Top Dev Biol 104:149–187.
  • Li H, Jia GH, Lu XL, Zhang G, Tian KY, Li JT and Zhang JM (2014) In vitro maturation of oocytes is not a risk factor for adult metabolic syndrome of mouse offspring. Eur J Obstet Gynecol Reprod Biol 174:96–99.
  • Li M, Rothwell R, Vermaat M, Wachsmuth M, Schröder R, Laros JFJ, van Oven M, de Bakker PIW, Bovenberg JA, van Duijn CM et al. (2016) Transmission of human mtDNA heteroplasmy in the genome of the Netherlands families: Support for a variable-size bottleneck. Genome Res 26:417–26.
  • Liang LF, Qi ST, Xian YX, Huang L, Sun XF and Wang WH (2017) Protective effect of antioxidants on the pre-maturation aging of mouse oocytes. Sci Rep 7:1434.
  • Lieber T, Jeedigunta SP, Palozzi JM, Lehmann R and Hurd TR (2019) Mitochondrial fragmentation drives selective removal of deleterious mtDNA in the germline. Nature 570:380–384.
  • Lonergan P and Fair T (2016) Maturation of oocytes in vitro Ann Rev Anim Biosci 4:255–268.
  • Luo S, Valencia CA, Zhang J, Lee NC, Slone J, Gui B, Wang X, Li Z, Dell S, Brown J et al. (2018) Biparental inheritance of mitochondrial DNA in humans. Proc Natl Acad Sci U S A 115:13039–13044.
  • Luzzo KM, Wang Q, Purcell SH, Chi M, Jimenez PT, Grindler N, Schedl T and Moley KH (2012) High fat diet induced developmental defects in the mouse: Oocyte meiotic aneuploidy and fetal growth retardation/brain defects. PLoS One 7:e0049217.
  • Ma H, Xu H and O’Farrell PH (2014) Transmission of mitochondrial mutations and action of purifying selection in Drosophila melanogaster. Nat Genet 46:393–7.
  • Machado TS, Carvalho KF, Garcia BM, Zangirolamo AF, Macabelli CH, Sugiyama FHC, Grejo MP, Augusto Neto JD, Ribeiro FKS, Sarapiao FD et al. (2018) Mitofusin 1 is required for the oocyte-granulosa cell communication that regulates oogenesis. bioRxiv 10.1101/498642.
  • Mahrous E, Yang Q and Clarke HJ (2012) Regulation of mitochondrial DNA accumulation during oocyte growth and meiotic maturation in the mouse. Reproduction 144:177–185.
  • May-Panloup P, Vignon X, Chrétien MF, Heyman Y, Tamassia M, Malthièry Y and Reynier P (2005) Increase of mitochondrial DNA content and transcripts in early bovine embryogenesis associated with upregulation of mtTFA and NRF1 transcription factors. Reprod Biol Endocrinol 3:65.
  • Maynard S, de Souza-Pinto NC, Scheibye-Knudsen M and Bohr VA (2010) Mitochondrial base excision repair assays. Methods 51:416–25.
  • Maynard S, Schurman SH, Harboe C, de Souza-Pinto NC and Bohr VA (2009) Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis 30:2–10.
  • Mingrone G, Manco M, Calvani M, Castagneto M, Naon D and Zorzano A (2005) Could the low level of expression of the gene encoding skeletal muscle mitofusin-2 account for the metabolic inflexibility of obesity? Diabetologia 48:2108–2114.
  • Mishra P and Chan DC (2014) Mitochondrial dynamics and inheritance during cell division, development and disease. Nat Rev Mol Cell Biol 15:634–646.
  • Misko AL, Sasaki Y, Tuck E, Milbrandt J and Baloh RH (2012) Mitofusin2 mutations disrupt axonal mitochondrial positioning and promote axon degeneration. J Neurosci 32:4145–4155.
  • Motta PM, Nottola SA, Makabe S and Heyn R (2000) Mitochondrial morphology in human fetal and adult female germ cells. Hum Reprod 15 Suppl 2:129–147.
  • Muftuoglu M, Mori MP and de Souza-Pinto NC (2014) Formation and repair of oxidative damage in the mitochondrial DNA. Mitochondrion 17:164-181.
  • Muñoz JP, Ivanova S, Sánchez-Wandelmer J, Martínez-Cristóbal P, Noguera E, Sancho A, Díaz-Ramos A, Hernández-Alvarez MI, Sebastián D, Mauvezin C et al. (2013) Mfn2 modulates the UPR and mitochondrial function via repression of PERK. EMBO J 32:2348-2361.
  • Murrin CM, Kelly GE, Tremblay RE and Kelleher CC (2012) Body mass index and height over three generations: Evidence from the Lifeways cross-generational cohort study. BMC Public Health 12:81.
  • Naon D and Scorrano L (2014) At the right distance: ER-mitochondria juxtaposition in cell life and death. Biochim Biophys Acta 1843:2184–2194.
  • Ngoh GA, Papanicolaou KN and Walsh K (2012) Loss of mitofusin 2 promotes endoplasmic reticulum stress. J Biol Chem 287:20321–20332.
  • Oktem O and Urman B (2010) Understanding follicle growth in vivo Hum Reprod 25:2944–2954.
  • Okura T, Koda M, Ando F, Niino N, Tanaka M and Shimokata H (2003) Association of the mitochondrial DNA 15497G/A polymorphism with obesity in a middle-aged and elderly Japanese population. Hum Genet 113:432–6.
  • Olichon A, Baricault L, Gas N, Guillou E, Valette A, Belenguer P and Lenaers G (2003) Loss of OPA1 perturbates the mitochondrial inner membrane structure and integrity, leading to cytochrome c release and apoptosis. J Biol Chem 278:7743–7746.
  • Olichon A, Emorine LJ, Descoins E, Pelloquin L, Brichese L, Gas N, Guillou E, Delettre C, Valette A, Hamel CP et al. (2002) The human dynamin-related protein OPA1 is anchored to the mitochondrial inner membrane facing the inter-membrane space. FEBS Lett 523:171–176.
  • Olivo PD, Van de Walle MJ, Laipis PJ and Hauswirth WW (1983) Nucleotide sequence evidence for rapid genotypic shifts in the bovine mitochondrial DNA D-loop. Nature 306:400–402.
  • Pareyson D, Saveri P, Sagnelli A and Piscosquito G (2015) Mitochondrial dynamics and inherited peripheral nerve diseases. Neurosci Lett 596:66–77.
  • Pasquariello R, Ermisch AF, Silva E, McCormick S, Logsdon D, Barfield JP, Schoolcraft WB and Krisher RL (2019) Alterations in oocyte mitochondrial number and function are related to spindle defects and occur with maternal aging in mice and humans. Biol Reprod 100:971–981.
  • Pathak T and Trebak M (2018) Mitochondrial Ca2+ signaling. Pharmacol Ther 192:112–123.
  • Payne BAI, Wilson IJ, Yu-Wai-Man P, Coxhead J, Deehan D, Horvath R, Taylor RW, Samuels DC, Santibanez-Koref M and Chinnery PF (2013) Universal heteroplasmy of human mitochondrial DNA. Hum Mol Genet 22:384–390.
  • Pernas L and Scorrano L (2016) Mito-morphosis: Mitochondrial fusion, fission, and cristae remodeling as key mediators of cellular function. Annu Rev Physiol 78:505–531.
  • Pikó L and Taylor KD (1987) Amounts of mitochondrial DNA and abundance of some mitochondrial gene transcripts in early mouse embryos. Dev Biol 123:364–374.
  • Poulton J, Scott Brown M, Cooper A, Marchington DR and Phillips DIW (1998) A common mitochondrial DNA variant is associated with insulin resistance in adult life. Diabetologia 41:54–58.
  • Poulton J, Chiaratti MR, Meirelles FV, Kennedy S, Wells D and Holt IJ (2010) Transmission of mitochondrial DNA diseases and ways to prevent them. PLoS Genet 6:e1001066.
  • Ramírez S, Gómez-Valadés AG, Schneeberger M, Varela L, Haddad-Tóvolli R, Altirriba J, Noguera E, Drougard A, Flores-Martínez Á, Imbernón M et al. (2017) Mitochondrial dynamics mediated by Mitofusin 1 is required for POMC neuron glucose-sensing and insulin release control. Cell Metab 25:1390-1399.e6.
  • Rand DM (2008) Mitigating mutational meltdown in mammalian mitochondria. PLoS Biol 6:e35.
  • Rattanatray L, MacLaughlin SM, Kleemann DO, Walker SK, Muhlhausler BS and McMillen IC (2010) Impact of maternal periconceptional overnutrition on fat mass and expression of adipogenic and lipogenic genes in visceral and subcutaneous fat depots in the postnatal lamb. Endocrinology 151:5195–5205.
  • Reddy P, Ocampo A, Suzuki K, Luo J, Bacman SR, Williams SL, Sugawara A, Okamura D, Tsunekawa Y, Wu J et al. (2015) Selective elimination of mitochondrial mutations in the germline by genome editing. Cell 161:459
  • Rivera MA, Pérusse L, Gagnon J, Dionne FT, Leon AS, Rao DC, Skinner JS, Wilmore JH, Sjöström L and Bouchard C (1999) A mitochondrial DNA D-loop polymorphism and obesity in three cohorts of women. Int J Obes Relat Metab Disord 23:666–668.
  • Rojansky R, Cha MY and Chan DC (2016) Elimination of paternal mitochondria in mouse embryos occurs through autophagic degradation dependent on PARKIN and MUL1. eLife 5:1–18.
  • Ruager-Martin R, Hyde MJ and Modi N (2010) Maternal obesity and infant outcomes. Early Hum Dev 86:715–722.
  • Ruebel ML, Cotter M, Sims CR, Moutos DM, Badger TM, Cleves MA, Shankar K and Andres A (2017) Obesity modulates inflammation and lipidmetabolism oocyte gene expression: A single-cell transcriptome perspective. J Clin Endocrinol Metab 102:2029–2038.
  • Ruhanen H, Borrie S, Szabadkai G, Tyynismaa H, Jones AWE, Kang D, Taanman JW and Yasukawa T (2010) Mitochondrial single-stranded DNA binding protein is required for maintenance of mitochondrial DNA and 7S DNA but is not required for mitochondrial nucleoid organisation. Biochim Biophys Acta 1803:931–939.
  • Ruiz-Pesini E, Díez-Sánchez C, López-Pérez MJ and Enríquez JA (2007) The role of the mitochondrion in sperm function: Is there a place for oxidative phosphorylation or is this a purely glycolytic process? Curr Top Dev Biol 77:3-19.
  • Saben JL, Boudoures AL, Asghar Z, Cusumano A, Scheaffer S, Moley KH, Saben JL, Boudoures AL, Asghar Z, Thompson A et al. (2016) Mitochondrial dysfunction via germline changes across three generations maternal metabolic syndrome programs mitochondrial dysfunction via germline changes across three generations. Cell Rep 16:1-8.
  • Sage JM, Gildemeister OS and Knight KL (2010) Discovery of a novel function for human RadJ Biol Chem 285:18984–18990.
  • Sage JM and Knight KL (2013) Human Rad51 promotes mitochondrial DNA synthesis under conditions of increased replication stress. Mitochondrion 13:350-356.
  • Santel A and Fuller MT (2001) Control of mitochondrial morphology by a human mitofusin. J Cell Sci 114:867-874.
  • Sarparanta J, García-Macia M and Singh R (2017) Autophagy and mitochondria in obesity and type 2 diabetes. Curr Diabetes Rev 13:352-369.
  • Sato A, Nakada K, Shitara H, Kasahara A, Yonekawa H and Hayashi JI (2007) Deletion-mutant mtDNA increases in somatic tissues but decreases in female germ cells with age. Genetics 177:2031-2037.
  • Scantland S, Tessaro I, Macabelli CH, Macaulay AD, Cagnone G, Fournier É, Luciano AM and Robert C (2014) The adenosine salvage pathway as an alternative to mitochondrial production of ATP in maturing mammalian oocytes. Biol Reprod 91:1-11.
  • Scarpulla RC (2002) Transcriptional activators and coactivators in the nuclear control of mitochondrial function in mammalian cells. Gene 286:81-9.
  • Schaefer AM, McFarland R, Blakely EL, He L, Whittaker RG, Taylor RW, Chinnery PF and Turnbull DM (2008) Prevalence of mitochondrial DNA disease in adults. Ann Neurol 63:35-9.
  • Scheibye-Knudsen M, Fang EF, Croteau DL, Wilson DM and Bohr VA (2015) Protecting the mitochondrial powerhouse. Trends Cell Biol 25:158-170.
  • Schneeberger M, Dietrich MO, Sebastián D, Imbernón M, Castaño C, Garcia A, Esteban Y, Gonzalez-Franquesa A, Rodríguez IC, Bortolozzi A et al. (2013) Mitofusin 2 in POMC neurons connects ER stress with leptin resistance and energy imbalance. Cell 155:172-187.
  • Schon EA, DiMauro S and Hirano M (2012) Human mitochondrial DNA: Roles of inherited and somatic mutations. Nat Rev Genet 13:878-90.
  • Schrepfer E and Scorrano L (2016) Mitofusins, from mitochondria to metabolism. Mol Cell 61:683-694.
  • Sebastian D, Hernandez-Alvarez MI, Segales J, Sorianello E, Munoz JP, Sala D, Waget A, Liesa M, Paz JC, Gopalacharyulu P et al. (2012) Mitofusin 2 (Mfn2) links mitochondrial and endoplasmic reticulum function with insulin signaling and is essential for normal glucose homeostasis. Proc Natl Acad Sci U S A 109:5523-5528.
  • Shankar K, Harrell A, Liu X, Gilchrist JM, Ronis MJJ and Badger TM (2008) Maternal obesity at conception programs obesity in the offspring. Am J Physiol Regul Integr Comp Physiol 294:528-538.
  • Sharma NK, Lebedeva M, Thomas T, Kovalenko OA, Stumpf JD, Shadel GS and Santos JH (2014) Intrinsic mitochondrial DNA repair defects in Ataxia Telangiectasia. DNA Repair 13:22-31.
  • Sharpley MS, Marciniak C, Eckel-Mahan K, McManus M, Crimi M, Waymire K, Lin CS, Masubuchi S, Friend N, Koike M et al. (2012) Heteroplasmy of mouse mtDNA is genetically unstable and results in altered behavior and cognition. Cell 151:333-43.
  • Silva JP, Köhler M, Graff C, Oldfors A, Magnuson MA, Berggren PO and Larsson NG (2000) Impaired insulin secretion and β-cell loss in tissue-specific knockout mice with mitochondrial diabetes. Nat Genet 26:336-340.
  • Sirard MA (2019) Distribution and dynamics of mitochondrial DNA methylation in oocytes, embryos and granulosa cells. Sci Rep 9:11937.
  • St John JC (2019) Mitochondria and female germline stem cells - a mitochondrial DNA perspective. Cells 8:852.
  • St John JC, Facucho-Oliveira J, Jiang Y, Kelly R and Salah R (2010) Mitochondrial DNA transmission, replication and inheritance: A journey from the gamete through the embryo and into offspring and embryonic stem cells. Hum Reprod Update 16:488-509.
  • Stehling O, Wilbrecht C and Lill R (2014) Mitochondrial iron-sulfur protein biogenesis and human disease. Biochimie 100:61-77.
  • Stewart JB and Chinnery PF (2015) The dynamics of mitochondrial DNA heteroplasmy: Implications for human health and disease. Nat Rev Genet 16:530-42.
  • Stewart JB, Freyer C, Elson JL, Wredenberg A, Cansu Z, Trifunovic A and Larsson NG (2008) Strong purifying selection in transmission of mammalian mitochondrial DNA. PLoS Biol 6:e10.
  • Su YQ, Sugiura K and Eppig JJ (2009) Mouse oocyte control of granulosa cell development and function: Paracrine regulation of cumulus cell metabolism. Semin Reprod Med 27:32-42.
  • Su YQ, Sugiura K, Wigglesworth K, O’Brien MJ, Affourtit JP, Pangas SA, Matzuk MM and Eppig JJ (2007) Oocyte regulation of metabolic cooperativity between mouse cumulus cells and oocytes: BMP15 and GDF9 control cholesterol biosynthesis in cumulus cells. Development 135:111-121.
  • Sugiura K, Su YQ, Diaz FJ, Pangas SA, Sharma S, Wigglesworth K, O’Brien MJ, Matzuk MM, Shimasaki S and Eppig JJ (2007) Oocyte-derived BMP15 and FGFs cooperate to promote glycolysis in cumulus cells. Development 134:2593-2603.
  • Sun Z, Terragni J, Borgaro JG, Liu Y, Yu L, Guan S, Wang H, Sun D, Cheng X, Zhu Z et al. (2013) High-resolution enzymatic mapping of genomic 5-Hydroxymethylcytosine in mouse embryonic stem cells. Cell Rep 3:567-576.
  • Sutovsky P, Moreno RD, Ramalho-Santos J, Dominko T, Simerly C and Schatten G (1999) Ubiquitin tag for sperm mitochondria. Nature 402:371-372.
  • Tanaka M, Fuku N, Takeyasu T, Guo LJ, Hirose R, Kurata M, Borgeld HJW, Yamada Y, Maruyama W, Arai Y et al. (2002) Golden mean to longevity: Rareness of mitochondrial cytochrome b variants in centenarians but not in patients with Parkinson’s disease. J Neurosci Res 70:347-355.
  • Thundathil J, Filion F and Smith LC (2005) Molecular control of mitochondrial function in preimplantation mouse embryos. Mol Reprod Dev 71:405-13.
  • Tomas L (1993) Instability and decay of the primary structure of DNA. Nature 362:709-715.
  • Trimarchi JR, Liu L, Porterfield DM, Smith PJ and Keefe DL (2000) Oxidative phosphorylation-dependent and -independent oxygen consumption by individual preimplantation mouse embryos. Biol Reprod 62:1866-74.
  • Udagawa O, Ishihara T, Maeda M, Matsunaga Y, Tsukamoto S, Kawano N, Miyado K, Shitara H, Yokota S, Nomura M et al. (2014) Mitochondrial Fission Factor Drp1 maintains oocyte quality via dynamic rearrangement of multiple organelles. Curr Biol 24:2451-2458.
  • Vermulst M, Wanagat J, Kujoth GC, Bielas JH, Rabinovitch PS, Prolla TA and Loeb LA (2008) DNA deletions and clonal mutations drive premature aging in mitochondrial mutator mice. Nat Genet 40:392-394.
  • Volaco A, Cavalcanti AM, Filho RP and Precoma DB (2018) Socioeconomic status: The missing link between obesity and diabetes mellitus? Curr Diabetes Rev 14:321-326.
  • Wai T, Teoli D and Shoubridge EA (2008) The mitochondrial DNA genetic bottleneck results from replication of a subpopulation of genomes. Nat Genet 40:1484-1488.
  • Wai T, Ao A, Zhang X, Cyr D, Dufort D and Shoubridge EA (2010) The role of mitochondrial DNA copy number in mammalian fertility. Biol Reprod 83:52-62.
  • Wakai T, Harada Y, Miyado K and Kono T (2014) Mitochondrial dynamics controlled by mitofusins define organelle positioning and movement during mouse oocyte maturation. Mol Hum Reprod 20:1090-1100.
  • Wallace DC (2011) Bioenergetic origins of complexity and disease. Cold Spring Harb Symp Quantit Biol 76:1-16.
  • Wallace DC (2005) A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine. Annu Rev Genet 39:359-407.
  • Wallace DC (2013) Bioenergetics in human evolution and disease: implications for the origins of biological complexity and the missing genetic variation of common diseases. Philos Trans R Soc Lond B Biol Sci 368:20120267.
  • Wallace DC and Chalkia D (2013) Mitochondrial DNA genetics and the heteroplasmy conundrum in evolution and disease. Cold Spring Harb Perspect Biol 5:a021220.
  • Wang Q, Ratchford AM, Chi MM, Schoeller E, Frolova A, Schedl T and Moley KH (2009) Maternal diabetes causes mitochondrial dysfunction and meiotic defects in murine oocytes. Mol Endocrinol 23:1603-1612.
  • Wang Q, Tang SB, Song XB, Deng TF, Zhang TT, Yin S, Luo SM, Shen W, Zhang CL and Ge ZJ (2018) High-glucose concentrations change DNA methylation levels in human IVM oocytes. Hum Reprod 33:474-481.
  • Wassarman PM and Josefowicz WJ (1978) Oocyte development in the mouse: An ultrastructural comparison of oocytes isolated at various stages of growth and meiotic competence. J Morphol 156:209-235.
  • Wei W, Tuna S, Keogh MJ, Smith KR, Aitman TJ, Beales PL, Bennett DL, Gale DP, Bitner-Glindzicz MAK, Black GC et al. (2019) Germline selection shapes human mitochondrial DNA diversity. Science 364:eaau6520.
  • Wolf DP, Hayama T and Mitalipov S (2017) Mitochondrial genome inheritance and replacement in the human germline. EMBO J 36:2177-2181.
  • Wu LL, Dunning KR, Yang X, Russell DL, Lane M, Norman RJ and Robker RL (2010) High-fat diet causes lipotoxicity responses in cumulus - oocyte complexes and decreased fertilization rates. Endocrinology 151:5438-5445.
  • Wu LL, Russell DL, Wong SL, Chen M, Tsai TS, St John JC, Norman RJ, Febbraio MA, Carroll J and Robker RL (2015) Mitochondrial dysfunction in oocytes of obese mothers: transmission to offspring and reversal by pharmacological endoplasmic reticulum stress inhibitors. Development 142:681-691.
  • Xu K, Chen G, Li X, Wu X, Chang Z, Xu J, Zhu Y, Yin P, Liang X and Dong L (2017) MFN2 suppresses cancer progression through inhibition of mTORC2/Akt signaling. Sci Rep 7:41718.
  • Yang ZY and Chian RC (2017) Development of invitro maturation techniques for clinical applications. Fertil Steril 108:577-584.
  • Yasukawa K, Oshiumi H, Takeda M, Ishihara N, Yanagi Y, Seya T, Kawabata S and Koshiba T (2009) Mitofusin 2 inhibits mitochondrial antiviral signaling. Sci Signal 2:ra47.
  • Yoneda M, Miyatake T and Attardi G (1995) Heteroplasmic mitochondrial tRNA(Lys) mutation and its complementation in MERRF patient-derived mitochondrial transformants. Muscle Nerve Suppl 3:S95-101.
  • Zhang M, Bener MB, Jiang Z, Wang T, Esencan E, Scott III R, Horvath T and Seli E (2019a) Mitofusin 1 is required for female fertility and to maintain ovarian follicular reserve. Cell Death Dis 10:560.
  • Zhang M, Bener MB, Jiang Z, Wang T, Esencan E, Scott R, Horvath T and Seli E (2019b) Mitofusin 2 plays a role in oocyte and follicle development, and is required to maintain ovarian follicular reserve during reproductive aging. Aging 11:3919-3938.
  • Zheng W, Khrapko K, Coller HA, Thilly WG and Copeland WC (2006) Origins of human mitochondrial point mutations as DNA polymerase γ-mediated errors. Mut Res 599:11-20.
  • Zorzano A, Hernández-Alvarez MI, Sebastián D and Muñoz JP (2015) Mitofusin 2 as a driver that controls energy metabolism and insulin signaling. Antioxid Redox Signal 22:1020-31.
  • Associate Editor: Carlos R. Machado

Publication Dates

  • Publication in this collection
    02 Mar 2020
  • Date of issue
    2020

History

  • Received
    19 Mar 2019
  • Accepted
    01 Nov 2019
Sociedade Brasileira de Genética Rua Cap. Adelmio Norberto da Silva, 736, 14025-670 Ribeirão Preto SP Brazil, Tel.: (55 16) 3911-4130 / Fax.: (55 16) 3621-3552 - Ribeirão Preto - SP - Brazil
E-mail: editor@gmb.org.br