Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-r5zm4 Total loading time: 0 Render date: 2024-07-08T02:20:32.136Z Has data issue: false hasContentIssue false

17 - Metabolism of the follicle and oocyte in vivo and in vitro

from Section 3 - Developmental biology

Published online by Cambridge University Press:  05 October 2013

Helen M. Picton
Affiliation:
Division of Reproduction and Early Development, Leeds Institute of Genetics, Health and Therapeutics, University of Leeds, Leeds, UK
Karen E. Hemmings
Affiliation:
Division of Reproduction and Early Development, Leeds Institute of Genetics, Health and Therapeutics, University of Leeds, Leeds, UK
Alan Trounson
Affiliation:
California Institute for Regenerative Medicine
Roger Gosden
Affiliation:
Center for Reproductive Medicine and Infertility, Cornell University, New York
Ursula Eichenlaub-Ritter
Affiliation:
Universität Bielefeld, Germany
Get access

Summary

Introduction

Oocyte metabolism reflects other aspects of the unique biology of this important cell type. The protracted process of mammalian oogenesis exacts a huge metabolic toll on the presumptive gamete. To ensure that the nutritional needs of oocytes are met oogenesis occurs in concert with folliculogenesis. Folliculogenesis is a lengthy process beginning with a primordial oocyte surrounded by a small number of flattened pregranulosa cells and ending with the ovulation of a fully grown, metaphase II oocyte, some weeks or months later. Throughout their development, oocytes and follicle cells are physically and metabolically linked via a complex network of homologous and heterologous gap junctions [1]. Metabolic coupling of oocytes and somatic cells facilitates the transfer of molecules of <1kDa, including ions, amino acids, pyruvate and glucose, molecules such as adenosine triphosphate (ATP) [2], and other signaling molecules and meiosis-arresting signals from the somatic compartment of the follicle to the oocyte and vice versa to provide the physiological basis for oocyte and follicle development [3]. While the metabolic cooperativity between oocytes and their companion granulosa cells is dynamic, discrete differences exist between the nutritional needs of oocytes and somatic granulosa cells and throughout their development oocytes are exposed to a changing nutritional environment as the follicular cells undergo proliferation, antral cavity formation, differentiation, and ovulation. In turn, oocytes have been shown to regulate apoptosis and cholesterol biosynthesis and metabolism by the follicular cells and so impact on follicular development [4].

Type
Chapter
Information
Biology and Pathology of the Oocyte
Role in Fertility, Medicine and Nuclear Reprograming
, pp. 200 - 211
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Kidder, GM, Vanderhyden, BC. Bidirectional communication between oocytes and follicle cells: ensuring oocyte developmental competence. Can J Physiol Pharmacol 2010; 88: 399–413.CrossRefGoogle ScholarPubMed
Heller, DT, Schultz, RM. Ribonucleoside metabolism by mouse oocytes: metabolic cooperativity between the fully grown oocyte and cumulus cells. J Exp Zool 1980; 214: 355–64.CrossRefGoogle ScholarPubMed
Wang, Q, Chi, MM, Schedl, T, et al. An intracellular pathway for glucose transport into mouse oocytes. Am J Physiol Endocrinol Metab 2012; 302: E1511–18.CrossRefGoogle ScholarPubMed
Su, YQ, Suguira, K, Eppig, JJ. Mouse oocytes control granulosa cell development and function: paracrine regulation of cumulus cell metabolism. Semin Reprod Med 2009; 27: 32–42.CrossRefGoogle ScholarPubMed
Homa, STRacowsky, C, McGaughley, RW. Lipid analysis of immature pig oocytes. J Reprod Fertil 1986; 77: 425–34.CrossRefGoogle ScholarPubMed
McEvoy, TG, Coull, GD, Broadbent, PJ, et al. Fatty acid composition of lipids in immature cattle, pig and sheep oocytes with intact zona pellucida. J Reprod Fertil 2000; 118: 163–70.CrossRefGoogle ScholarPubMed
Ferguson, EM, Leese, HJ. A potential role for triglyceride as an energy source during bovine oocyte maturation and early embryo development. Mol Reprod Dev 2006; 73: 1195–201.CrossRefGoogle ScholarPubMed
Sturmey, RG, Reis, A, Leese, HJ, et al. Role of fatty acids in energy provision during oocyte maturation and early embryo development. Reprod Domest Anim 2009; 44 Suppl 3: 50–8.CrossRefGoogle ScholarPubMed
Hemmings, KE, Leese, HJ, Picton, HM. Amino acid turnover by bovine oocytes provides an index of oocyte developmental competence in vitro. Biol Reprod 2012; 86: 165, 1–12.CrossRefGoogle ScholarPubMed
Downs, SM, Utecht, AM. Metabolism of radio labelled glucose by mouse oocytes and oocyte cumulus cell complexes. Biol Reprod 1999; 60: 1446–52.CrossRefGoogle Scholar
Harris, SE, Adriens, I, Leese, HJ, et al. Carbohydrate metabolism by murine ovarian follicles and oocytes grown in vitro. Reproduction 2007; 134: 415–24.CrossRefGoogle ScholarPubMed
Harris, SE, Leese, HJ, Gosden, RG, et al. Pyruvate and oxygen consumption throughout the growth and development of murine oocytes. Mol Reprod Dev 2009; 76: 231–8.CrossRefGoogle ScholarPubMed
Sutton, ML, Gilchrist, RB, Thompson, JG. Effects of in vivo and in vitro environments on the metabolism of the cumulus-oocyte complex and its influence on oocyte developmental capacity. Hum Reprod Update 2003; 9: 35–48.CrossRefGoogle Scholar
Sutton-McDowall, ML, Gilchrist, RB, Thompson, JG. The pivotal role of glucose metabolism in determining oocyte developmental competence. Reproduction 2010; 139: 685–95.CrossRefGoogle ScholarPubMed
Wood, IS, Trayhurn, P. Glucose transporters (GLUT and SGLT): expanded families of sugar transport proteins. Br J Nutr 2003; 89: 3–9.CrossRefGoogle ScholarPubMed
Purcell, SH, Moley, KH. Glucose transporters in gametes and preimplantation embryos. Trends Endocrinol Metab 2009; 20: 483–9.CrossRefGoogle ScholarPubMed
Dan-Goor, M, Sasson, S, Davarashvili, A. Expression of glucose transporter and glucose uptake in human oocytes and preimplantation embryos. Hum Reprod 1997; 12: 2508–10.CrossRefGoogle ScholarPubMed
Augustin, R, Pocar, P, Navarrete-Santos, A, et al. Glucose transporter expression is developmentally regulated in in vitro derived bovine preimplantation embryos. Mol Reprod Dev 2001; 60: 370–6.CrossRefGoogle ScholarPubMed
Zheng, P, Vasenna, R, Latham, KE. Effects of in vitro oocyte maturation and embryo culture on the expression of glucose transporters, glucose metabolism and insulin signalling in rhesus monkey oocytes and preimplantation embryos. Mol Hum Reprod 2007; 13: 361–71.CrossRefGoogle Scholar
Nishimoto, H, Matsutani, R, Yamamoto, S, et al. Gene expression of glucose transporter (GLUT) 1, 3 and 4 in bovine follicle and corpus luteum. J Endocrinol 2006; 188: 111–19.CrossRefGoogle ScholarPubMed
Seamark, RF, Amoto, F, Hendrickson, S, et al. Oxygen uptake, glucose utilization, lactate release and adenine nucleotide content of sheep ovarian follicles in culture: effect of human chorionic goadotrophin. Aust J Biol Sci 1976; 29: 557–63.CrossRefGoogle Scholar
Boland, NJ, Humpherson, PG, Leese, HJ, et al. The effect of glucose metabolism on murine follicle development and steroidogenesis in vitro. Hum Reprod 1994; 9: 617–23.CrossRefGoogle ScholarPubMed
Benomar, Y, Naour, N, Aubourg, A, et al. Insulin and leptin induce Glut4 plasma membrane translocation and glucose uptake in a human neuronal cell line by a phosphatidylinositol 3-kinase- dependent mechanism. Endocrinology 2006; 147: 2550–6.CrossRefGoogle Scholar
Brinster, RL, Harstad, H. Energy metabolism in primordial germ cells of the mouse. Exp Cell Res 1997; 109: 111–17.CrossRefGoogle Scholar
Johnson, NT, Freeman, EA, Gardner, DK, et al. Oxidative metabolism of pyruvate is required for meiotic maturation of murine oocytes in vivo. Biol Reprod 2007; 77: 2–8.CrossRefGoogle ScholarPubMed
Duran, HE, Simsek-Duran, F, Oehninger, SC, et al. The association of reproductive senescence with mitochondrial quantity, function, and DNA integrity in human oocytes at different stages of maturation. Fertil Steril 2011; 96: 384–8.CrossRefGoogle ScholarPubMed
Krisher, RL, Brad, AM, Herrick, JR, et al. A comparative analysis of metabolism and viability in porcine oocytes during in vitro maturation. Anim Reprod Sci 2007; 98: 72–96.CrossRefGoogle ScholarPubMed
Van Blerkom, J, Davis, PW, Lee, J. ATP content of human oocytes and developmental potential and outcome after in vitro fertilization and embryo transfer. Hum Reprod 1995; 10: 415–24.CrossRefGoogle ScholarPubMed
Hardy, K, Hooper, MA, Handyside, AH, et al. Non-invasive measurement of glucose and pyruvate uptake by individual human oocytes and preimplantation embryos. Hum Reprod 1989; 4: 188–91.CrossRefGoogle ScholarPubMed
Harris, SE, Maruthini, D, Tang, T, et al. Metabolism and karyotype analysis of oocytes from patients with PCOS. Hum Reprod 2010; 25: 2305–15.CrossRefGoogle Scholar
Tsutsumi, O, Yano, T, Satoh, K, et al. Studies of hexose kinase activities in human and mouse oocytes. Am J Obstet Gynecol 1990; 162: 1301–4.CrossRefGoogle Scholar
Mangia, F, Epstein, CJ. Biochemical studies of growing mouse oocytes: preparation of oocytes and analysis of glucose-6-phosphate dehydrogenase and lactate dehydrogenase activities. Dev Biol 1975; 45: 211–20.CrossRefGoogle ScholarPubMed
Biggers, JD, Whittingham, DG, Donahue, RP. The pattern of energy metabolism in the mouse oocyte and zygote. Zoology 1967; 58: 560–7.Google ScholarPubMed
Urner, F, Sakkas, D. A possible role for the pentose phosphate pathway of spermatogenesis in gamete fusion in the mouse. Biol Reprod 1999; 60: 733–9.CrossRefGoogle Scholar
Reiger, D, Luskutoff, N. Changes in the metabolism of glucose, pyruvate, glutamine and glycine during maturation of cattle oocytes in vitro. J Reprod Fertil 1994; 100: 257–62.CrossRefGoogle Scholar
Cetica, P, Pintos, L, Dalvit, G, Beconi, M.Activity of key enzymes involved in glucose and triglyceride metabolism during bovine oocyte maturation in vitro. Reproduction 2002; 124: 675–81.CrossRefGoogle ScholarPubMed
Wilding, M, Dale, B, Marino, M, et al. Mitochondrial aggregation patterns and activity in human oocytes and preimplantation embryos. Hum Reprod 2001; 16: 909–17.CrossRefGoogle ScholarPubMed
Almeida, Santos T, El Shourbagy, S, St John, JC. Mitochondrial content reflects oocytes variability and fertilisation outcome. Fertil Steril 2005; 85: 584–91.Google Scholar
Van Blerkom, J. Mitochondria in human oogenesis and preimplantation embryogenesis: engines of metabolism, ionic regulation and developmental competence. Reproduction 2004; 128: 269–80.CrossRefGoogle ScholarPubMed
O’Donnel-Tormey, J, Nathan, CF, Lanks, K, et al. Secretion of pyruvate. An antioxidant defence of mammalian cells. J Exp Med 1987; 165: 500–14.CrossRefGoogle Scholar
Downs, SM, Mosey, JL, Klinger, J. Fatty acid oxidation and meiotic resumption in mouse oocytes. Mol Reprod Dev 2009; 76: 844–53.CrossRefGoogle ScholarPubMed
Dunning, JR, Cashman, K, Russell, DL, et al. Beta-oxidation is essential for mouse oocyte developmental competence and early embryo development. Biol Reprod 2010; 83: 909–18.CrossRefGoogle ScholarPubMed
Somfai, T, Kaneda, M, Akagi, S, et al. Enhancement of lipid metabolism with L-carnitine during in vitro maturation improves nuclear maturation and cleavage ability of follicular porcine oocytes. Reprod Fertil Dev 2011: 23: 912–20.CrossRefGoogle ScholarPubMed
Sutton-McDowell, ML, Feil, D, Robker, RL, et al. Utilisation of endogenous fatty acid stores for energy production in bovine preimplantation embryos. Theriogenology 2012; 77: 1632–41.CrossRefGoogle Scholar
Leese, HJ. Metabolism of the preimplantation mammalian embryo. Oxf Rev Reprod Biol 1991; 13: 35–72.Google ScholarPubMed
Pelland, AMD, Corbett, HE, Baltz, JM. Amino acid transport mechanisms in mouse oocytes during growth and meiotic maturation. Biol Reprod 2009; 81: 1041–54.CrossRefGoogle ScholarPubMed
Van Winkle, LJ. Amino acid transport regulation and early embryo development. Biol Reprod 2001; 64: 1–12.CrossRefGoogle ScholarPubMed
Colonna, R, Mangia, F.Mechanisms of amino acid uptake in cumulus enclosed mouse oocytes. Biol Reprod 1983; 28: 797–803.CrossRefGoogle ScholarPubMed
Cetica, P, Pintos, L, Dalvit, G, Beconi, M. Involvement of enzymes of amino acid metabolism and tricarboxylic acid cycle in bovine oocyte maturation in vitro. Reproduction 2003; 126: 753–63.CrossRefGoogle ScholarPubMed
Chand, AR, Legge, M.Amino acid transport system L activity in developing mouse ovarian follicles. Hum Reprod 2011; 26: 3102–8.CrossRefGoogle ScholarPubMed
Colonna, R, Cecconi, S, Buccione, R, Mangia, F. Amino acid transport systems in growing mouse oocytes. Cell Biol Int Rep 1983; 7: 1007–15.CrossRefGoogle ScholarPubMed
Watson, AJ, Pape, C, Emanuel, JR, Levenson, R, Kidder, GM. Expression of Na, K-ATPase alpha subunit and beta subunit genes during preimplantation development of the mouse. Dev Genet 1990; 11: 41–8.CrossRefGoogle ScholarPubMed
Eppig, JJ, Pendola, FL, Wigglesworth, K, Pendola, JK. Mouse oocytes regulate metabolic cooperativity between granulosa cells and oocytes: amino acid transport. Biol Reprod 2005; 73: 351–7.CrossRefGoogle ScholarPubMed
Choi, HW, Shin, MR, Lee, HS, et al. Differential expression of amino acids transporter genes in mouse blastocysts and embryonic stem cells. Fertil Steril 2005; 84: S393–4.CrossRefGoogle Scholar
Bermudez, M, Wells, D, Malter, H, et al. Expression profiles of individual human oocytes using microarray technology. Reprod Biomed Online 2004; 8: 325–37.CrossRefGoogle ScholarPubMed
Closs, EI, Simon, A, Vekony, N, Rotmann, A.Plasma membrane transporters for arginine. J Nutr 2004; 134: 2752S–9S.CrossRefGoogle Scholar
Verrey, F, Meier, C, Rossier, G, Kuhn, LC. Glycoprotein-associated amino acid exchangers: broadening the range of transport specificity. Pflugers Arch 2000; 440: 503–12.CrossRefGoogle ScholarPubMed
Picton, HM. Activation of follicle development: the primordial follicle, its activation and possible use as source of female genetic material. Theriogenology 2001; 55: 1193–210.CrossRefGoogle Scholar
Harris, SE, Picton, HM. Metabolism of follicles and oocytes during growth and maturation. In: Tan, SL, Chian, RC, Buckett, W, eds. In-Vitro Maturation of Human Oocytes: Basic Science to Clinical Application. Oxon: Informa Health. 2007; 15–36.Google Scholar
Eppig, J.Analysis of mouse oogenesis in vitro. Oocyte isolation and the utilization of exogenous energy sources by growing oocytes. J Exp Zool 1976; 198: 375–82.CrossRefGoogle ScholarPubMed
Barbehenn, EK, Wales, RG, Lowry, OH. The explanation for the blockade of glycolysis in early mouse embryos. Proc Natl Acad Sci USA 1974; 71: 1056–60.CrossRefGoogle ScholarPubMed
Barbehenn, EK, Wales, RG, Lowry, OH. Measurement of metabolites in single preimplantation embryos; a new means to study metabolic control in early embryos. J Embryol Exp Morphol 1978; 43: 29–46.Google ScholarPubMed
Butcher, L, Coates, A, Martin, KL, et al. Metabolism of pyruvate by the early human embryo. Biol Reprod 1998; 58: 1054–6.CrossRefGoogle ScholarPubMed
Gougeon, A.Regulation of ovarian follicular development in primates – facts and hypotheses. Endocr Rev 1996; 17: 121–55.CrossRefGoogle ScholarPubMed
Dumollard, R, Duchen, M, Carroll, J. The role of mitochondrial function in the oocyte and embryo. Curr Top Dev Biol 2007; 77: 21–49.CrossRefGoogle ScholarPubMed
Conti, M, Hsieh, M, Zamah, AM, et al. Novel signaling mechanisms in the ovary during oocyte maturation and ovulation. Mol Cell Endocrinol 2012; 356: 65–73.CrossRefGoogle ScholarPubMed
Gosden, RG, Byatt-Smith, JG. Oxygen concentration gradient across the ovarian follicular epithelium: model, predictions and implications. Hum Reprod 1986; 1: 65–8.CrossRefGoogle ScholarPubMed
Sugiura, K, Pendola, FL, Eppig, JJ. Oocyte control of metabolic cooperativity between oocytes and companion granulosa cells: energy metabolism. Dev Biol 2005; 279: 20–30.CrossRefGoogle ScholarPubMed
Gilchrist, RB, Lane, M, Thompson, JG. Oocyte secreted factors: regulators of cumulus function and oocyte quality. Hum Reprod Update 2008; 14: 159–77.CrossRefGoogle ScholarPubMed
Harris, SE, Gopichandran, N, Picton, HM, et al. Nutrient concentrations in murine follicle, oviduct and uterine fluids. Theriogenology 2005; 64: 992–1006.CrossRefGoogle Scholar
Orsi, NM, Gopichandran, N, Leese, HJ, et al. Fluctuations in bovine ovarian follicular fluid composition throughout the oestrous cycle: a comparison with plasma and a TCM-199-based maturation medium. Reproduction 2005; 192: 219–28.CrossRefGoogle Scholar
Leese, HJ, Lenton, EA. Glucose and lactate in human follicular fluid: concentration and inter relationships. Hum Reprod 1990; 5: 915–19.CrossRefGoogle Scholar
Hillier, SG, Purohit, A, Reichert, LEControl of granulosa cell lactate production by follicle-stimulating hormone and androgen. Endocrinology 1985; 116:1163–7.CrossRefGoogle ScholarPubMed
Downs, SM, Humpherson, PG, Martin, KL, et al. Glucose utilization during gonadotropin-induced meiotic maturation in cumulus cell-enclosed mouse oocytes. Mol Reprod Dev 1996; 44: 121–31.3.0.CO;2-7>CrossRefGoogle ScholarPubMed
Lin, Y, Fridstrom, M, Hillensjo, T. Insulin stimulation of lactate accumulation in isolated human granulosa-luteal cells: a comparison between normal and polycystic ovaries. Hum Reprod 1997; 12: 2469–72.CrossRefGoogle ScholarPubMed
Nilsson, L. Acute effects of gonadotrophins and prostaglandins on the metabolism of isolated ovarian follicles from PMSG-treated immature rats. Acta Endocrinol (Copenh) 1974; 77: 540–58.Google Scholar
Hillensjo, T. Oocyte maturation and glycolysis in isolated pre-ovulatory follicles of PMS-injected immature rats. Acta Endocrinol (Copenh) 1976; 82: 809–30.Google ScholarPubMed
Roberts, R, Stark, J, Iatropoulou A, et al. Energy substrate metabolism of mouse cumulus-oocyte complexes: response to follicle-stimulating hormone is mediated by the phosphatidylinositol 3-kinase pathway and is associated with oocyte maturation. Biol Reprod 2004; 71: 199–209.CrossRefGoogle ScholarPubMed
Roy, SK, Terada, DM. Activities of glucose metabolic enzymes in human preantral follicles: in vitro modulation by follicle-stimulating hormone, luteinizing hormone, epidermal growth factor, insulin-like growth factor I, and transforming growth factor beta1. Biol Reprod 1999; 60: 763–8.CrossRefGoogle ScholarPubMed
Brogan, RS, MacGibeny, M, Mix, S, et al. Dynamics of intra-follicular glucose during luteinization of macaque ovarian follicles. Mol Cell Endocrinol 2011; 332: 189–95.CrossRefGoogle ScholarPubMed
Sutton-McDowall, ML, Mottershead, DG, Gardner, DK, et al. Metabolic differences in bovine cumulus oocyte complexes matured in vitro in the presence or absence of follicle stimulating hormone and bone morphogenetic protein 15. Biol Reprod. 2012; 87: 1–8.CrossRefGoogle ScholarPubMed
Downs, SM, Mastropolo, AM. Culture conditions affect meiotic regulation in cumulus cell-enclosed mouse oocytes. Mol Reprod Dev 1997; 46: 551–66.3.0.CO;2-Z>CrossRefGoogle ScholarPubMed
Shim, C, Lee, DK, Lee, CC, et al. Inhibitory effect of purines in meiotic resumption of denuded mouse oocytes. Mol Reprod Dev 1992; 314: 280–6.CrossRefGoogle Scholar
Downs, SM. Adenosine blocks hormone-induced meiotic maturation by suppressing purine de novo synthesis. Mol Reprod Dev 2000; 56: 172–9.3.0.CO;2-2>CrossRefGoogle ScholarPubMed
Downs, SM, Verhoeven, A.Glutamine and the maintenance of meiotic arrest in mouse oocytes: influences of culture medium, glucose and cumulus cells. Mol Reprod Dev 2003; 66: 90–7.CrossRefGoogle Scholar
Billig, H, Magnusson, C.Gonadotropin-induced inhibition of oxygen consumption in rat oocyte-cumulus complexes: relief by adenosine. Biol Reprod 1985; 33: 890–8.CrossRefGoogle ScholarPubMed
Downs, SM, Houghton, FD, Humpherson, PG, et al. Substrate utilization and maturation of cumulus cell-enclosed mouse oocytes: evidence that pyruvate oxidation does not mediate meiotic induction. J Reprod Fertil 1997; 110: 1–10.CrossRefGoogle Scholar
Colton, SA, Humpherson, PG, Leese, HJ, et al. Physiological changes in oocyte-cumulus cell complexes from diabetic mice that potentially influence meiotic regulation. Biol Reprod 2003; 69: 761–70.CrossRefGoogle ScholarPubMed
Colton, SA, Downs, SM. Potential role for the sorbitol pathway in the meiotic dysfunction exhibited by oocytes from diabetic mice. J Exp Zool A Comp Exp Biol 2004; 301: 439–48.CrossRefGoogle ScholarPubMed
Wang, Q, Ratchford, AM, Chi, MM, et al. Maternal diabetes causes mitochondrial dysfunction and meiotic defects in murine oocytes. Mol Endocrinol 2009; 23: 1603–12.CrossRefGoogle ScholarPubMed
Landau, S, Braw-Tal, R, Kaim, M, et al. Preovulatory follicular status and diet affect the insulin and glucose content of follicles in high-yielding dairy cows. Anim Reprod Sci 2000; 64: 181–97.CrossRefGoogle ScholarPubMed
Leese, HJ. Metabolism of the preimplantation embryo: 40 years on. Reproduction 2012; 143: 417–27.CrossRefGoogle Scholar
Picton, HM, Harris, SE, Muruvi, W, Chambers, EL. The in vitro growth and maturation of follicles. Reproduction 2008; 136: 703–15.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×