Skip to main content

Beyond energy provider: multifunction of lipid droplets in embryonic development

Abstract

Since the discovery, lipid droplets (LDs) have been recognized to be sites of cellular energy reserves, providing energy when necessary to sustain cellular life activities. Many studies have reported large numbers of LDs in eggs and early embryos from insects to mammals. The questions of how LDs are formed, what role they play, and what their significance is for embryonic development have been attracting the attention of researchers. Studies in recent years have revealed that in addition to providing energy for embryonic development, LDs in eggs and embryos also function to resist lipotoxicity, resist oxidative stress, inhibit bacterial infection, and provide lipid and membrane components for embryonic development. Removal of LDs from fertilized eggs or early embryos artificially leads to embryonic developmental arrest and defects. This paper reviews recent studies to explain the role and effect mechanisms of LDs in the embryonic development of several species and the genes involved in the regulation. The review contributes to understanding the embryonic development mechanism and provides new insight for the diagnosis and treatment of diseases related to embryonic developmental abnormalities.

Introduction

Lipid droplets (LDs) are highly dynamic subcellular organelles that play an important role in lipid storage, metabolic homeostasis control, and the maintenance of dynamic stability in the intracellular environment [1]. They possess an outer monolayer consisting of phospholipids and proteins, encasing a neutral lipid core, such as triacylglycerols (TAGs), which is hydrophobic. Highly dynamic organelles exhibit significant morphological variations among different cells or at different metabolic levels. During cellular starvation, TAG lipase in the cytoplasm hydrolyzes LDs, a process called lipolysis. The resulting decomposed products then generate the energy required by the cell through the fatty acids (FAs) beta-oxidation of mitochondria [2].

Autophagy is an intracellular degradation process in eukaryotic cells that transports cytoplasmic components to lysosomes or vesicles for degradation or recycling. Furthermore, autophagy-mediated degradation of LDs, known as lipophagy, has been observed sequentially in mammals [3] and yeast [4], and it may also occur in plants [5]. Studies in yeast and animals have revealed that lipophagy serves various functions, including intracellular environmental homeostasis, energy production, and stress mitigation [2, 6,7,8,9,10,11,12].

Intracellular deposition of free fatty acids (FFAs) can have toxic effects, leading to impaired hepatocyte function and apoptosis [13,14,15]. When FFAs levels exceed the cellular metabolic capacity, resulting in an excess of FFAs, lipids are deposited intracellularly, releasing inflammatory factors that initiate apoptotic signals and sensitize cells to inflammation and injury [16]. This damage eventually leads to cell damage or apoptosis, a condition known as lipotoxicity [17]. Lipids that may be toxic to cells, such as cholesterol and FFAs, are sequestered within LDs [18]. Maternal nutritional overload can lead to lipotoxicity and oxidative stress. Excessive intake of nutrients such as fat and sugar will increase the risk of fetal fat deposition, fetal obesity, and a lipid metabolism disorder. These metabolites further trigger oxidative stress and a series of harmful reactions. Additionally, maternal nutritional overload may cause abnormal changes in the placenta, increasing fetal exposure to harmful substances and the potential for oxidative stress. Furthermore, nutritional overload can affect the normal function of the maternal immune system, inducing obesity-related inflammation and immune response, thereby exacerbating fetal oxidative stress and lipotoxicity [19]. During embryonic development, maternal nutritional overload can result in lipotoxicity, leading to oxidative stress and a decrease in the quality of embryonic development. However, the anti-stress ability of LDs stabilizes the cellular environment and significantly reduces the potential for cellular lipotoxicity, thereby ensuring normal cellular function.

LDs can be generated through various mechanisms and are dynamically connected to other organelles, facilitating the exchange of lipids and proteins [1, 20,21,22,23,24,25]. LDs serve as storage sites for diverse lipids that can act as signaling molecules or be converted into signaling molecules upon release. FFAs released from LDs can directly bind to cell surface receptors, triggering intracellular signal transduction pathways. These pathways involve peroxisome proliferator-activated receptors (PPARs), sterol regulatory element-binding proteins (SREBPs), and nuclear factor kappa-B transcription factors [26,27,28]. LDs actively store biologically active signaling molecules internally and influence signaling pathways by regulating their release and production of signaling molecules [28, 29]. Changes in intracellular gene expression during embryonic development suggest that genes play a regulatory role in embryogenesis and enable LDs to interact with other organelles.

In this review, we provide a comprehensive overview of current research in the field of LDs in embryos. We focus on elucidating the functional roles of LDs during different stages of embryonic development and explore the gene targets associated with LDs. Additionally, we discuss how understanding LD function and gene targets can contribute to advancements in vitro embryo culture and human in vitro fertilization (IVF) technology, providing new insights and ideas.

The role of LDs in embryonic development

The distributions of LDs in embryonic development

LDs are widely distributed in the reproductive system, and their abundance and distribution can be observed during embryonic development. Bik et al. employed near-infrared, mid-infrared, and Raman imaging techniques to investigate the distribution of LDs and other substances in fertilized Medaka Fish eggs [30]. Electron microscopy analysis of brown adipocytes in embryos revealed a potential association between LDs and glycogen [31]. Gupta et al. fixed zebrafish embryos at the cleavage stage using a 4% paraformaldehyde fixative and performed total internal reflection fluorescence (TIRF) imaging. Nile red-stained LDs were observed to accumulate at the cleavage groove using a 100×TIRF lens in an Olympus TIRF microscope. Similarly, Yang et al. employed the same approach to examine LDs in mouse Cumulus–oocyte complexes [32, 33].

Hallberg’s laser scanning microscopy observations showed significant changes in LipidTOX™ stained LD distribution during the cleavage of experimentally treated bovine oocytes [34]. To determine the nature of pro-Sudanese LDs in sheep blastocysts, this study used acetone to distinguish polar lipids from neutral lipids, which were soluble in acetone, and the complete absence of Sudan Black B staining was observed in light microscopy as LDs containing neutral lipids [35]. Tao et al. incubated the porcine embryo samples for 1 h and observed them under a confocal microscope and observed not only the distribution of LDs at the cleavage stage but likewise the number of LDs at the blastocyst stage [36].

LDs are present not only during the process of embryonic development but also in germ cells. Transmission electron microscopy observation of semi-thin sections of in vitro mature porcine oocytes subjected to electrical stimulation revealed a more uniform distribution of LDs in oocytes developing in vivo compared to those developed in vitro [37]. Microscopic examination of quail, duck, and turkey oocytes after induction of lipogenic differentiation also revealed the presence of LDs [38]. These observed results that LDs are pervasive throughout the development of life, not only in the oocytes but also in all stages of embryonic development, and may play a potential role in embryonic development.

Multifunction of LDs in embryo

LDs may have multiple roles in the reproductive system, including energy storage and metabolism, lipid membrane conversion, signaling, mitigation of cellular stress, and temporary storage of proteins.

Within cells, FAs exhibit various destinies. In addition to their roles in membrane assimilation, lipid reserve storage, and as signaling molecules in lipid pathways, FAs can undergo oxidation, releasing energy and producing carbon dioxide and water as byproducts. Embryonic cells are adaptive and require appropriate amounts of FAs. As an indirect repository of intracellular FAs, LDs play an irreplaceable role in maintaining energy metabolic homeostasis. Cells use two main mechanisms to mobilize FAs during nutrient stress. One mechanism is autophagic digestion via membrane-bound organelles (i.e., the endoplasmic reticulum) or LDs [39,40,41,42]. This involves autophagosomes engulfing organelles/LDs and fusing with lysosomes, where hydrolases digest the organelles/LDs and release FFAs that rapidly enter the cytoplasm [3]. The second mechanism for the mobilization of FAs during starvation is through the lipolytic consumption of LDs. In this process, cytoplasmic neutral lipase directly hydrolyzes TAG on the LD surface. Mitochondria are the main site of β-oxidation, and during nutrient stress, FAs are catabolized by enzymes to maintain energy levels in embryonic cells.

The morphology and number of cytoplasmic LDs change during the maturation and fertilization of porcine oocytes [43]. There are significant differences in LDs in embryos before in vivo and in vitro implantation, which may be related to the energy requirements during porcine embryo development [37]. An LD was formed during the transformation of mouse embryonic stem cells (ESCs) to 2-cell stage embryo-like cells (2CLCs). Intriguingly, the glycolytic capacity and respiratory activity of 2CLCs are weaker than those of ESCs, and it is reasonable to assume that the ATP levels of 2CLCs are lower than those of ESCs. However, the difference in ATP levels between 2CLC and ESCs is not significant, indicating the potential to utilize a unique energy metabolic pathway [44].

Two degradation systems, the ubiquitin-proteasome pathway, and autophagy are required in early embryonic development to degrade maternal proteins and lipids into nutrients and raw materials for embryonic development. LDs are also required for normal embryonic development [45]. Using the autophagic degradation system expressing P62 as LD autophagic cargo to perform forced lipophagy is an intriguing approach. In this process, LDs aggregate and translocate to the cell periphery, leading to reduced viability of mouse embryos due to excessive energy depletion [46]. The zebrafish oocyte-to-embryo transition necessitates an additional ATP pulse to maintain the dynamic homeostasis of the embryo. Interestingly, instead of consuming the maternally provided yolk-FFAs pool and yolk-FACoA pool during this pulse preparation, LD-mediated lipolysis is utilized to provide the energy required to reach the pulse. This demonstrates the important role of LDs in supplying energy during early zebrafish embryonic development [47].

Lipophagy does not necessarily directly convert to ATP for cellular energy supply. As the embryo develops, there is a decrease in lipids such as phospholipids and TAGs in the egg, while the opposite trend is observed in the glycogen near-infrared spectrum, suggesting that lipid consumption accompanies carbohydrate production [30]. Due to the biogenesis of LDs by glycogen during embryonic development, newborn rodents are filled with LDs after the first cold exposure [31]. These studies indicate that the interconversion of LDs and other nutrients during embryonic development ensures normal development (Fig. 1). LDs can serve as a source of membrane precursors for blastocyst cellularization (Fig. 2). Additionally, during differentiation, the plasma membrane invaginates and fuses into the nuclear membrane of the nucleus of the embryonic peripheral syncytium [48], with LDs playing a significant role in the composition of membrane components during cell differentiation.

Fig. 1
figure 1

Intra-embryonic LD-glycogen conversion: FA, a TAG breakdown product within LDs, will be involved in gluconeogenesis to generate glycogen, which is biogenic to LDs during embryonic development. Green I: Gluconeogenesis. Red II: Biogenesis of LDs from glycogen. Abbreviations: LD, lipid droplet; FA, fatty acid; TAG, triacylglycerol; Glu, glucose; GLUT, glucose transporter; Gn, glycogen; ER, endoplasmic reticulum; TCA, tricarboxylic acid cycle; β-Ox, β-oxidation; ATP, adenosine triphosphate LIP1; lipase 1

Fig. 2
figure 2

LD membranes are the source of membrane precursors for blastocyst cellularization. LD membranes gradually invaginate to become membrane precursors for the plasma membrane and other membrane structures during blastocyst cellularization division

LDs also play a role in alleviating cellular stress during embryonic development. Overfeeding during mare gestation did not affect the accumulation of LDs in the blastocyst during the first seven days, indicating that the physical condition of the mother does not immediately impact embryo development [49]. This is attributed to the stress-relieving effect of LDs. However, the ability of LDs to alleviate stress is also limited. Compared to control mice, mice fed high-fat diets showed significantly increased lipid accumulation in oocytes and enhanced endoplasmic reticulum stress, resulting in low fertilization and blastocyst rates and reduced embryonic developmental potential. Maternal hyperthermia during critical stages of embryonic development can lead to the accumulation of LDs in the trophectoderm, which may result in malformation or developmental delay in rat fetuses [50, 51].

Enhanced lipophagy of LDs in mouse cervical tissue may be controlled by progesterone. Lipolytic enzyme levels and LDs lipolysis in mid-pregnancy are closely related, suggesting that energy supply or hormonal facilitation is required to maintain cervical closure. Additionally, in a biotin-deficient environment, biotin, an essential vitamin for lipid and protein synthesis, cannot recruit TAGs into LDs [52]. The formation of LDs mitigates the extent of cell damage since excessive deposition of FFAs inside cells can cause lipotoxicity.

What would LDs do if embryos were in an environment filled with bacterial contamination? Bacterial contamination poses an extremely serious hazard to embryonic development. When simulating a bacterial infection environment, LDs in Drosophila play a key role in embryonic development. Not only do LD interconvert with glycogen in embryonic cells and provide energy for embryonic development through lipophagy, but their membrane components also act as membrane precursors for syncytium division. Additionally, LDs can temporarily store maternally supplied proteins and nutrients, preventing the degradation of excess histones before entering the nucleus and avoiding the onset of lipotoxicity. They can also load histones onto the LD, enhancing the defense against bacterial infestation [53]. Furthermore, a significant increase in the size of LDs was observed in a sample of female dogs with pus accumulation in the uterus [54].

Intriguingly, maternal proteins are provided to the embryo, but some proteins are not immediately used during embryonic development, and LDs temporarily isolate proteins provided by the mother or proteins that are not immediately bound until the embryonic cells need them to supply effective proteins (Fig. 3) [55]. This suggests that LDs are not only good stress relievers and disease fighters for cells but also protein reservoirs that temporarily bind free proteins.

Fig. 3
figure 3

LDs are temporary reservoirs of free proteins. The proteins provided by the mother do not function immediately in the embryonic cells, and the LDs bind to them, thus avoiding the degradation of histones by intracellular enzymes. When these proteins are needed after embryonic cell division, the LDs release them

The importance of LDs for embryonic development

In animal production and laboratory animal research, energy and nutrients are critical during embryonic development. Cumulus–oocyte complexes have a potential role in regulating TAG metabolism and β-oxidation processes produced by lipophagy, and elucidating this regulatory role could develop the potential for oocyte development in domestic animals [56]. Furthermore, extensive studies of embryonic in vitro culture techniques in animals have found that the presence of serum increases the abundance of neutral LDs, but the accumulation is heterogeneous. The reason for the uneven distribution is the result of the unique adaptation of LDs [35], as certain regions of the cell are more in need of LD participation, leading to a greater number of LDs in these regions.

The size and number of intra-embryonic LDs in B. indicus and B. taurus at the mulberry blastocyst stage affect the outcome of embryos after cryopreservation. The reason is that freezing is the denaturation of structural proteins on the LD surface, leading to the rupture of LDs, the outflow of their contents, and an increase in saturated fatty acids (SFAs) in the cytoplasm, causing lipotoxicity [57]. This finding provides a new strategy for the cryopreservation of embryos, exploring whether controlled lipolysis of LDs prior to preservation can increase embryo viability. Another approach is to load LD with specific proteins that are resistant to cold stress, aiming to prevent LD rupture due to low temperature and improve the pregnancy rate. This idea also opens up new possibilities for the preservation of blastocyst stage embryos.

For human reproduction, maternal rats with obesity and alcohol consumption have a higher incidence of congenital heart disease in their offspring than normal individuals due to the abnormal formation of LDs as a result of abnormal placental lipids [58]. Lipophagy in patients with advanced ovarian cancer is non-regulatory, and cancer cells enhance lipophagy to provide energy for their proliferation, and inhibition of lipophagy can effectively inhibit cancer progression [59]. These studies show that the health status of the mother has an impact on the development of the offspring, which sheds light on the implications for human diet and disease during pregnancy preparation, and even the role of LD as a temporary repository of proteins to specifically bind anti-cancer proteins and thus help in cancer treatment, which is not only a potential way to save mothers with cancer from reproducing offspring properly but also a new way of thinking about the fight against cancer.

In vitro embryo culture technology plays an important role in livestock production, and the quality of embryo development affects productivity. Treatment of in vitro embryos with appropriate concentrations of melatonin has been shown to increase the number of LDs while decreasing their size, improving oocyte quality and providing necessary energy for embryonic development. Feeding fish oil to broiler females during gestation can reduce fat deposition in the offspring and reduce the potential risk of obesity in the offspring [60]. The effect of adding a synthetic serum to cultured embryos was similar to that of adding fetal calf serum [61], and the serum did not cause lipid accumulation or organelle damage to bovine embryonic cells [62], reducing the concentration of fetal bovine serum to 5% for culturing in vitro embryonic cells is an appropriate concentration [63]. Supplementation with all-trans retinoic acid [64], L-carnitine [65], melatonin [66], and dehydroepiandrosterone [67] in serum can improve in vitro embryo culturing, which provides a method for the livestock industry including other fields that require embryo culturing in vitro, and hopefully, more serum additives will enhance embryo In vitro culture viability. However, human in vitro embryo culture is still in the IVF period, and there are many difficulties in the late in vitro culture technology. Experimental findings obtained from animal studies may help address the challenges of in vitro culture of human embryos, representing a significant potential breakthrough.

LDs-related genes and regulatory functions in embryonic development

Signal pathways in embryonic development

Regulation of the AMPK pathway is essential for energy metabolism during embryonic development. AMPK is a heterotrimeric complex consisting of three subunits, which β and γ as catalytic subunits and α is a regulatory subunit [68]. AMPK activity can be activated by kinases and pharmacological activators, leading to the activation of AMPK subunits [69]. The intracellular AMP/ATP ratio influences the activity of AMPK. When AMP/ATP levels are elevated, AMPK is activated, inhibiting lipid synthesis and promoting FA oxidation [70, 71]. AMPK acts as a stress response in conditions of energy deficiency and oxidative stress. It phosphorylates target proteins to regulate lipid metabolism [72]. Acetyl coenzyme A carboxylase (ACC) is a key enzyme involved in the synthesis of long-chain FAs, specifically encoded by acetyl coenzyme A carboxylase 1 [73]. During oocyte development, the ACC gene promotes the potential for oocytes to differentiate into adipocytes [66]. Activated AMPK phosphorylates and inactivates ACC, reducing FA synthesis through the AMPK/ACC pathway. This promotes the expression of carnitine palmitoyltransferase 1(CPT1) and increases FA oxidation. The ACC gene plays a significant role in LD formation, and its reduced activity affects the TAG within LDs, thus impacting energy storage and metabolism [74, 75]. Expression of ACC and CPT1 genes provides the energetic foundation for subsequent embryonic developmental processes. During development, if there is nutrient overload, embryonic cells are regulated through the PI3K/AKT pathway. The production of 3’-phosphorylated phosphatidylinositol activates the PI3K/AKT pathway, which may regulate the expression of certain lipid genes during embryonic development [76].

Fatty acid synthase (FASN) is a key enzyme involved in the de novo synthesis of FAs. It catalyzes the production of palmitate and 16-carbon long FAs from acetyl coenzyme A and malonate coenzyme A [77]. SREBP1C activates FASN by binding to its promoter region, with contains sterol regulatory elements [78]. Studies have shown that PI3K/AKT signaling pathway influences the expression of SREBP1C/FASN, regulating the conversion of glycolipids in cells in response to sugar concentration [79]. This process may involve the biogenic effects of glycogen on LDs, leading to increased FASN levels and promoting LD synthesis. Consequently, energy is converted into lipids for storage in LDs, helping to regulate nutrient levels during embryonic development.

The Wnt pathway plays a crucial role in regulating stem cell pluripotency and determining cell fate during embryonic development. It also controls the formation of the embryonic axis, axons, organs, and other important processes during embryonic development. The components of the Wnt pathway include Wnt ligands, G protein-coupled transmembrane frizzled receptors, and low-density lipoprotein-related receptor co-receptors [80]. Monounsaturated fatty acids (MUFAs) activate the Wnt/β-catenin pathway, transmitting signals to the nucleus [81], A similar process may occur during embryonic development. Three distinct isoforms of SREBPs, namely SREBP1a, SREBP1c, and SREBP2, are expressed in various human tissues. These isoforms are encoded by separate genes [82]. The SREBP1c isoform primarily governs FA synthesis, while SREBP2 regulates genes involved in cholesterol biosynthesis and embryonic development. Interestingly, the SREBP1a isoform is involved in both lipogenic pathways [83,84,85]. Depletion of SREBP1 leads to decreased levels of unsaturated lipids and triggers apoptotic cell death when cells have limited access to exogenous lipids. Activation of the Wnt/β-catenin pathway induces SREBP-1c to activate genes necessary for FA and triacylglycerol synthesis, such as stearoyl-CoA desaturase 1 (SCD1).

SCD1 is an integral protein located in the endoplasmic reticulum membrane. It catalyzes the synthesis of polyunsaturated FAs, such as oleic acid (C18:1) and palmitoleic acid (C16:1), from FAs like palmitic acid (C16:0) and stearic acid (C18:0). This enzymatic activity introduces cis double bonds between carbons 9 and 10 of the stearic acid and palmitic acid. Oleic acid, an unsaturated FA, is a significant byproduct of SCD1 activity. Intriguingly, the extent of exogenous oleic acid supplementation positively affects LD formation in embryos. In contrast, the presence of saturated FAs has detrimental effects on both embryo development and LD formation [86]. This highlights the critical role of SCD1 in early embryonic development and emphasizes its importance in this intricate process.

Genes regulating LDs formation in the embryo

LDs in the embryo require proper gene expression regulation to function. Perilipin, a core LD-associated protein, is well-known for its important role in lipid metabolism. Recent studies have also identified other crucial genes involved in lipid metabolism. Among these genes, PPARs are members of the intranuclear receptor transcription factor superfamily that regulate the expression of target genes. PPARα, in particular, is a significant nuclear receptor that controls the expression of the CPT1 gene, which is involved in FA catabolism [87]. The activity of PPARγ affects the formation of LDs in adipocytes [88]. Long-chain lipid CoA synthase is a key player in body lipid metabolism and is associated with various diseases. Specifically, long-chain acyl-CoA synthetases 1 (ACSL1) is a target gene of PPARα and co-regulates lipid metabolism in the body. SCD1 is a rate-limiting enzyme that converts SFA to MUFA and plays a role in LD formation through phospholipid formation.

DGAT1, CD36, or NR1H3 have been identified as markers associated with lipids in porcine and bovine blastocysts. These genes are involved in LD synthesis [89]. Intriguingly, blocking the very long chain fatty acid enzyme 5(ELOVL5) gene reduced the expression of related lipids and promoted intracytoplasmic LD deposition in blastocysts. However, this blockade did not affect embryo development or blastocyst cell number [90], possibly due to compensatory effects of other ELOVL family genes on lipid metabolism [91]. In nematode embryos, the SEIPIN-1 pair controls LD size and lipid homeostasis. Mutations in SEIP-1 lead to dysregulation of the lipid-permeable membrane in the innermost layer of the embryonic eggshell, resulting in embryonic death. However, supplementation with polyunsaturated FAs can resolve this issue [92]. Other genes associated with LD function in the embryo include GPI [93], LSD [94], Myosin family [32], and other genes regulated.

LD-related genes contribute to embryonic development

Diacylglycerol Acyltransferase (DGAT) is the acyl-coenzyme A required to catalyze the final step of TAG synthesis. The genes encoding two DGAT enzymes, DGAT1 and DGAT2, were discovered long ago [95]. Changes in DGAT2 expression during oocyte formation suggest increased lipid synthesis in oocytes [63]. TAG stored in LDs serves as backup energy for the first embryonic cleavage [96]. The expression of DGAT2 and ACC genes provides the energy basis for subsequent embryonic development.

Myosin-1(Myo1) is a motor protein involved in early embryonic development. It binds to other motor proteins and is recruited to specific binding sites involved in membrane invagination and rupture [97]. Intriguingly, zebrafish oval spheres contain dynamic LDs, and embryos with suppressed Myo1 show an accumulation of LD in the sulcus line [32]. Myo1 is involved in maintaining the oval sulcus and driving the movement of LDs. ACSLs modify FFAs by catalyzing the formation of Acyl-CoA and activating them. TriacsinC treatment of non-defatted embryos leads to substantial LD degradation. Other drugs that promote lipase activity do not reduce LD intensity, but ACSL activity reduction leads to embryonic developmental defects [98]. These results suggest that ACSL activity is crucial for the synthesis and maintenance of LDs and is a key factor in LD biogenesis.

SEIPIN is an evolutionarily conserved protein encoded by the Berardinelli-Seip congenital lipodystrophy 2 gene. It is localized to the endoplasmic reticulum [99, 100] and plays a key role in biogenesis. During Caenorhabditis elegans embryo development, SEIPIN1 is involved in forming the permeability barrier, which protects the embryo from toxic molecules and damage. SEIPIN1-deficient Caenorhabditis elegans mutants disrupt the dynamic balance of FFAs in embryos, leading to embryonic death [92]. This suggests that SEIPIN1 plays a critical role in FFA storage during embryonic development. Genes associated with LDs regulate the internal environment of embryonic development during the various processes of embryonic development, ensuring proper embryonic development and positioning of embryo culture at the genetic level (Fig. 4).

Fig. 4
figure 4

Signal pathways associated with LDs at various stages of embryonic development. In the oocyte, the ACACA gene encodes acetyl coenzyme A carboxylase 1 by the AMPK pathway, which then enters the cytoplasm and converts Acetyl-CoA to Malonyl-CoA. DGAT synthesizes TAGs to encapsulate LDs. During the cleavage stage, Myosin1 drives LDs to the cleavage groove and maintains the formation of the cleavage groove by Wnt/β-catenin. SCD1 catalyzes the conversion of SFA to MUFA. During the mulberry embryo period, ACSL converts long-chain FAs to acyl-CoA by PI3K/AKT pathway. SEIPIN is an important gene for LD biogenesis throughout embryonic development. These genes may function at other stages of the embryo, and the timeline is based on available studies

LD-related genes can be potential genetic targets during embryonic development

Whether for improving embryonic development in vivo or applying it to in vitro embryo culture techniques, regulating the expression of the aforementioned genes may yield the desired results. PPARs are important nuclear receptors that regulate FA metabolism in the body. By controlling the upstream control genes of PPARs, lipid metabolism can be improved in embryos with severe fat deposition, creating an ideal cellular environment for embryonic development. PPARs can play a key role in regulating the overall development of embryonic tissues. CPT1 and ASCL1 are target genes of PPARα, and the expression status of these pathways directly impacts lipid metabolism in the embryo. DGAT and ACC are key enzymes involved in the synthesis of long-chain FAs and are closely related to LD production. Interestingly, Myo1 is a key gene in embryonic cell division [101]. Exploring the effect of Myosin gene family expression on the rate of embryonic cellularization would be an interesting direction to investigate, as it may facilitate the process of embryonic development.

In conclusion, these genes associated with LDs will be favorable targets for studying the functional expression of LDs in the embryo and exploring their regulation will be of great significance for embryo development and culture techniques.

Conclusion and prospect

The current understanding of the involvement of LDs in embryonic development remains in its nascent stages despite ongoing attempts to explicate their role. The extant literature on LDs and their associated genes has primarily concentrated on their functions in the liver, adipose tissue, and macrophages. This review seeks to address this gap in knowledge by elucidating the function of LDs and related genes in the context of embryonic development.

Due to the biological intricacies of embryonic development, the lipid composition is a crucial aspect that needs to be considered. Further investigations into the role of LDs in the embryo can provide additional insights into their importance. A deeper understanding of the interconversion of LDs with other nutrients, such as proteins, is necessary to comprehend the influence of LDs on embryonic development. Additionally, it is pertinent to investigate the process of embryonic cellularization, which involves the formation of membrane precursors from LDs, and the storage and release of proteins on these precursors. The mechanisms of protein storage and release, as well as the regulation of DGAT during embryonic development, also warrant further exploration. Furthermore, it is essential to investigate whether modulating Myo1 protein can accelerate LDs’ movement along microfilaments to the cleavage groove and promote the cleavage process. Additionally, studying the impact of serum concentration on LD levels in cells through in vitro embryo culture techniques is also necessary. By addressing these questions, we can overcome the challenges posed by in vitro embryo culture technology in animal production and enhance production efficiency. Additionally, addressing these queries may result in breakthroughs in human IVF technology and may extend the duration of in vitro culture. Furthermore, resolving these queries can provide therapeutic targets for the treatment of embryos with maternal obesity and other inflammatory diseases.

Data Availability

Not applicable.

Abbreviations

LDs:

Lipid droplets

TAG:

Triacylglycerol

FAs:

Fatty acids

FFAs:

Free fatty acids

PPARs:

Peroxisome proliferator-activated receptors

SREBPs:

Sterol regulatory element-binding proteins

IVF:

In vitro fertilization

TIRF:

Total internal reflection fluorescence

ESCs:

Embryonic stem cells

2CLCs:

2-cell stage embryo-like cells

ACC:

Acetyl coenzyme A carboxylase

CTP1:

Carnitine palmitoyltransferase 1

FASN:

Fatty acid synthase

MUFAs:

Monounsaturated fatty acids

SCD1:

Stearoyl-CoA desaturase 1

ACSLs:

Long-chain acyl-CoA synthetases

ACSL1:

Long-chain acyl-CoA synthetase 1

DGAT:

Diacylglycerol Acyltransferase

Myo1:

Myosin-1

References

  1. Olzmann JA, Carvalho P. Dynamics and functions of lipid droplets. Nat Rev Mol Cell Biol. 2019;20(3):137–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Schepers J, Behl C. Lipid droplets and autophagy-links and regulations from yeast to humans. J Cell Biochem. 2021;122(6):602–11.

    Article  CAS  PubMed  Google Scholar 

  3. Singh R, Kaushik S, Wang Y, Xiang Y, Novak I, Komatsu M, Tanaka K, Cuervo AM, Czaja MJ. Autophagy regulates lipid metabolism. Nature. 2009;458(7242):1131–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. van Zutphen T, Todde V, de Boer R, Kreim M, Hofbauer HF, Wolinski H, Veenhuis M, van der Klei IJ, Kohlwein SD. Lipid droplet autophagy in the yeast Saccharomyces cerevisiae. Mol Biol Cell. 2014;25(2):290–301.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Kurusu T, Koyano T, Hanamata S, Kubo T, Noguchi Y, Yagi C, Nagata N, Yamamoto T, Ohnishi T, Okazaki Y, et al. OsATG7 is required for autophagy-dependent lipid metabolism in rice postmeiotic anther development. Autophagy. 2014;10(5):878–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ogasawara Y, Tsuji T, Fujimoto T. Multifarious roles of lipid droplets in autophagy - target, product, and what else? Semin Cell Dev Biol. 2020;108:47–54.

    Article  CAS  PubMed  Google Scholar 

  7. Filali-Mouncef Y, Hunter C, Roccio F, Zagkou S, Dupont N, Primard C, Proikas-Cezanne T, Reggiori F. The menage a trois of autophagy, lipid droplets and liver disease. Autophagy. 2022;18(1):50–72.

    Article  CAS  PubMed  Google Scholar 

  8. Khawar MB, Abbasi MH, Rafiq M, Naz N, Mehmood R, Sheikh N. A Decade of Mighty Lipophagy: What We Know and What Facts We Need to Know? Oxid Med Cell Longev 2021, 2021:5539161.

  9. Elander PH, Minina EA, Bozhkov PV. Autophagy in turnover of lipid stores: trans-kingdom comparison. J Exp Bot. 2018;69(6):1301–11.

    Article  CAS  PubMed  Google Scholar 

  10. Wang CW. Lipid droplets, lipophagy, and beyond. Biochim Biophys Acta. 2016;1861(8 Pt B):793–805.

    Article  CAS  PubMed  Google Scholar 

  11. Jaishy B, Abel ED. Lipids, lysosomes, and autophagy. J Lipid Res. 2016;57(9):1619–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zechner R, Madeo F, Kratky D. Cytosolic lipolysis and lipophagy: two sides of the same coin. Nat Rev Mol Cell Biol. 2017;18(11):671–84.

    Article  CAS  PubMed  Google Scholar 

  13. Wang L, Liu Y, Zhang X, Ye Y, Xiong X, Zhang S, Gu L, Jian Z, Wang H. Endoplasmic reticulum stress and the unfolded protein response in cerebral Ischemia/Reperfusion Injury. Front Cell Neurosci. 2022;16:864426.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Weiss TS, Lupke M, Ibrahim S, Buechler C, Lorenz J, Ruemmele P, Hofmann U, Melter M, Dayoub R. Attenuated lipotoxicity and apoptosis is linked to exogenous and endogenous augmenter of liver regeneration by different pathways. PLoS ONE. 2017;12(9):e0184282.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Bellanti F, Mitarotonda D, Tamborra R, Blonda M, Iannelli G, Petrella A, Sanginario V, Iuliano L, Vendemiale G, Serviddio G. Oxysterols induce mitochondrial impairment and hepatocellular toxicity in non-alcoholic fatty liver disease. Free Radic Biol Med. 2014;75(Suppl 1):16–7.

    Article  Google Scholar 

  16. de Andrade Melo-Sterza F, Poehland R. Lipid Metabolism in Bovine Oocytes and Early Embryos under In Vivo, In Vitro, and Stress Conditions. Int J Mol Sci 2021, 22(7).

  17. Kusminski CM, Shetty S, Orci L, Unger RH, Scherer PE. Diabetes and apoptosis: lipotoxicity. Apoptosis. 2009;14(12):1484–95.

    Article  CAS  PubMed  Google Scholar 

  18. Csaky Z, Garaiova M, Kodedova M, Valachovic M, Sychrova H, Hapala I. Squalene lipotoxicity in a lipid droplet-less yeast mutant is linked to plasma membrane dysfunction. Yeast. 2020;37(1):45–62.

    Article  CAS  PubMed  Google Scholar 

  19. Rao A, Satheesh A, Nayak G, Poojary P, Kumari S, Kalthur S, Mutalik S, Adiga S, Kalthur G. High-fat diet leads to elevated lipid accumulation and endoplasmic reticulum stress in oocytes, causing poor embryo development. Reprod Fertil Dev. 2020;32(14):1169–79.

    Article  CAS  PubMed  Google Scholar 

  20. Cohen S. Lipid droplets as organelles. Int Rev Cell Mol Biol. 2018;337:83–110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Herms A, Bosch M, Reddy BJ, Schieber NL, Fajardo A, Ruperez C, Fernandez-Vidal A, Ferguson C, Rentero C, Tebar F, et al. AMPK activation promotes lipid droplet dispersion on detyrosinated microtubules to increase mitochondrial fatty acid oxidation. Nat Commun. 2015;6:7176.

    Article  PubMed  Google Scholar 

  22. Nguyen TB, Louie SM, Daniele JR, Tran Q, Dillin A, Zoncu R, Nomura DK, Olzmann JA. DGAT1-Dependent lipid Droplet Biogenesis protects mitochondrial function during Starvation-Induced Autophagy. Dev Cell. 2017;42(1):9–21e25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Rambold AS, Cohen S, Lippincott-Schwartz J. Fatty acid trafficking in starved cells: regulation by lipid droplet lipolysis, autophagy, and mitochondrial fusion dynamics. Dev Cell. 2015;32(6):678–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Thiam AR, Beller M. The why, when and how of lipid droplet diversity. J Cell Sci. 2017;130(2):315–24.

    CAS  PubMed  Google Scholar 

  25. Valm AM, Cohen S, Legant WR, Melunis J, Hershberg U, Wait E, Cohen AR, Davidson MW, Betzig E, Lippincott-Schwartz J. Applying systems-level spectral imaging and analysis to reveal the organelle interactome. Nature. 2017;546(7656):162–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Papackova Z, Cahova M. Fatty acid signaling: the new function of intracellular lipases. Int J Mol Sci. 2015;16(2):3831–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Schmitz G, Ecker J. The opposing effects of n-3 and n-6 fatty acids. Prog Lipid Res. 2008;47(2):147–55.

    Article  CAS  PubMed  Google Scholar 

  28. Zechner R, Zimmermann R, Eichmann TO, Kohlwein SD, Haemmerle G, Lass A, Madeo F. FAT SIGNALS–lipases and lipolysis in lipid metabolism and signaling. Cell Metab. 2012;15(3):279–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Coleman RA, Mashek DG. Mammalian triacylglycerol metabolism: synthesis, lipolysis, and signaling. Chem Rev. 2011;111(10):6359–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bik E, Ishigaki M, Blat A, Jasztal A, Ozaki Y, Malek K, Baranska M. Lipid droplet composition varies based on Medaka Fish Eggs Development as revealed by NIR-, MIR-, and Raman Imaging. Molecules 2020, 25(4).

  31. Mayeuf-Louchart A. Uncovering the role of glycogen in Brown Adipose tissue. Pharm Res. 2021;38(1):9–14.

    Article  CAS  PubMed  Google Scholar 

  32. Gupta P, Martin R, Knolker HJ, Nihalani D, Kumar Sinha D. Myosin-1 inhibition by PClP affects membrane shape, cortical actin distribution and lipid droplet dynamics in early zebrafish embryos. PLoS ONE. 2017;12(7):e0180301.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Yang X, Dunning KR, Wu LL, Hickey TE, Norman RJ, Russell DL, Liang X, Robker RL. Identification of perilipin-2 as a lipid droplet protein regulated in oocytes during maturation. Reprod Fertil Dev. 2010;22(8):1262–71.

    Article  CAS  PubMed  Google Scholar 

  34. Hallberg I, Kjellgren J, Persson S, Orn S, Sjunnesson Y. Perfluorononanoic acid (PFNA) alters lipid accumulation in bovine blastocysts after oocyte exposure during in vitro maturation. Reprod Toxicol. 2019;84:1–8.

    Article  CAS  PubMed  Google Scholar 

  35. Reis A, McCallum GJ, McEvoy TG. Accumulation and distribution of neutral lipid droplets is non-uniform in ovine blastocysts produced in vitro in either the presence or absence of serum. Reprod Fertil Dev. 2005;17(8):815–23.

    Article  CAS  PubMed  Google Scholar 

  36. Tao R, Bi J, Zhu F, Wang X, Jia C, Xu H, He X, Li J. Division behaviours and their effects on pre-implantation development of pig embryos. Reprod Domest Anim. 2022;57(9):1016–28.

    Article  CAS  PubMed  Google Scholar 

  37. Kikuchi K, Ekwall H, Tienthai P, Kawai Y, Noguchi J, Kaneko H, Rodriguez-Martinez H. Morphological features of lipid droplet transition during porcine oocyte fertilisation and early embryonic development to blastocyst in vivo and in vitro. Zygote. 2002;10(4):355–66.

    Article  PubMed  Google Scholar 

  38. Lee J, Kim DH, Suh Y, Lee K. Research note: potential usage of DF-1 cell line as a new cell model for avian adipogenesis. Poult Sci. 2021;100(5):101057.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Axe EL, Walker SA, Manifava M, Chandra P, Roderick HL, Habermann A, Griffiths G, Ktistakis NT. Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum. J Cell Biol. 2008;182(4):685–701.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hayashi-Nishino M, Fujita N, Noda T, Yamaguchi A, Yoshimori T, Yamamoto A. A subdomain of the endoplasmic reticulum forms a cradle for autophagosome formation. Nat Cell Biol. 2009;11(12):1433–7.

    Article  CAS  PubMed  Google Scholar 

  41. Kristensen AR, Schandorff S, Hoyer-Hansen M, Nielsen MO, Jaattela M, Dengjel J, Andersen JS. Ordered organelle degradation during starvation-induced autophagy. Mol Cell Proteomics. 2008;7(12):2419–28.

    Article  CAS  PubMed  Google Scholar 

  42. Yla-Anttila P, Vihinen H, Jokitalo E, Eskelinen EL. 3D tomography reveals connections between the phagophore and endoplasmic reticulum. Autophagy. 2009;5(8):1180–5.

    Article  PubMed  Google Scholar 

  43. Ibayashi M, Aizawa R, Mitsui J, Tsukamoto S. Homeostatic regulation of lipid droplet content in mammalian oocytes and embryos. Reproduction. 2021;162(6):R99–R109.

    Article  CAS  PubMed  Google Scholar 

  44. Furuta A, Nakamura T. Lipid droplets are formed in 2-cell-like cells. J Reprod Dev. 2021;67(2):79–81.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Tsukamoto S, Tatsumi T. Degradation of maternal factors during preimplantation embryonic development. J Reprod Dev. 2018;64(3):217–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Tatsumi T, Takayama K, Ishii S, Yamamoto A, Hara T, Minami N, Miyasaka N, Kubota T, Matsuura A, Itakura E et al. Forced lipophagy reveals that lipid droplets are required for early embryonic development in mouse. Development 2018, 145(4).

  47. Dutta A, Sinha DK. Zebrafish lipid droplets regulate embryonic ATP homeostasis to power early development. Open Biol 2017, 7(7).

  48. Chen L, Dumelie JG, Li X, Cheng MH, Yang Z, Laver JD, Siddiqui NU, Westwood JT, Morris Q, Lipshitz HD, et al. Global regulation of mRNA translation and stability in the early Drosophila embryo by the Smaug RNA-binding protein. Genome Biol. 2014;15(1):R4.

    Article  PubMed  PubMed Central  Google Scholar 

  49. NMM DF, Gibson CME, van Doorn DA, Roelfsema E, de Ruijter-Villani M, Stout TAE. Effect of Overfeeding Shetland Pony Mares on embryonic glucose and lipid Accumulation, and expression of imprinted genes. Anim (Basel) 2021, 11(9).

  50. Rao A, Satheesh A, Nayak G, Poojary PS, Kumari S, Kalthur SG, Mutalik S, Adiga SK, Kalthur G. High-fat diet leads to elevated lipid accumulation and endoplasmic reticulum stress in oocytes, causing poor embryo development. Reprod Fertil Dev. 2020;32(14):1169–79.

    Article  CAS  PubMed  Google Scholar 

  51. Padmanabhan R, Al-Menhali NM, Ahmed I, Kataya HH, Ayoub MA. Histological, histochemical and electron microscopic changes of the placenta induced by maternal exposure to hyperthermia in the rat. Int J Hyperthermia. 2005;21(1):29–44.

    Article  CAS  PubMed  Google Scholar 

  52. Tao L, Zhang H, Wang H, Li L, Huang L, Su F, Yuan X, Luo M, Ge L. Characteristics of lipid droplets and the expression of proteins involved in lipolysis in the murine cervix during mid-pregnancy. Reprod Fertil Dev. 2020;32(11):967–75.

    Article  CAS  PubMed  Google Scholar 

  53. Anand P, Cermelli S, Li Z, Kassan A, Bosch M, Sigua R, Huang L, Ouellette AJ, Pol A, Welte MA, et al. A novel role for lipid droplets in the organismal antibacterial response. Elife. 2012;1:e00003.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Leitner N, Hlavaty J, Heider S, Ertl R, Gabriel C, Walter I. Lipid droplet dynamics in healthy and pyometra-affected canine endometrium. BMC Vet Res. 2022;18(1):221.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Cermelli S, Guo Y, Gross SP, Welte MA. The lipid-droplet proteome reveals that droplets are a protein-storage depot. Curr Biol. 2006;16(18):1783–95.

    Article  CAS  PubMed  Google Scholar 

  56. Dunning KR, Russell DL, Robker RL. Lipids and oocyte developmental competence: the role of fatty acids and beta-oxidation. Reproduction. 2014;148(1):R15–27.

    Article  CAS  PubMed  Google Scholar 

  57. Lopez-Damian EP, Jimenez-Medina JA, Lammoglia MA, Pimentel JA, Agredano-Moreno LT, Wood C, Galina CS, Fiordelisio T. Lipid droplets in clusters negatively affect Bos indicus embryos during cryopreservation. Anat Histol Embryol. 2018;47(5):435–43.

    Article  PubMed  Google Scholar 

  58. Linask KK, Han M. Acute alcohol exposure during mouse gastrulation alters lipid metabolism in placental and heart development: Folate prevention. Birth Defects Res A Clin Mol Teratol. 2016;106(9):749–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ray U, Roy D, Jin L, Thirusangu P, Staub J, Xiao Y, Kalogera E, Wahner Hendrickson AE, Cullen GD, Goergen K, et al. Group III phospholipase A2 downregulation attenuated survival and metastasis in ovarian cancer and promotes chemo-sensitization. J Exp Clin Cancer Res. 2021;40(1):182.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Beckford RC, Howard SJ, Das S, Farmer AT, Campagna SR, Yu J, Hettich RL, Wilson JL, Voy BH. Maternal consumption of fish oil programs reduced adiposity in broiler chicks. Sci Rep. 2017;7(1):13129.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Ordonez-Leon EA, Merchant H, Medrano A, Kjelland M, Romo S. Lipid droplet analysis using in vitro bovine oocytes and embryos. Reprod Domest Anim. 2014;49(2):306–14.

    Article  CAS  PubMed  Google Scholar 

  62. Crocco MC, Kelmansky DM, Mariano MI. Does serum cause lipid-droplet accumulation in bovine embryos produced in vitro, during developmental days 1 to 4? J Assist Reprod Genet. 2013;30(10):1377–88.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Del Collado M, Saraiva NZ, Lopes FL, Gaspar RC, Padilha LC, Costa RR, Rossi GF, Vantini R, Garcia JM. Influence of bovine serum albumin and fetal bovine serum supplementation during in vitro maturation on lipid and mitochondrial behaviour in oocytes and lipid accumulation in bovine embryos. Reprod Fertil Dev 2015.

  64. Kim DH, Lee J, Suh Y, Cressman M, Lee K. Research note: all-trans retinoic acids induce adipogenic differentiation of chicken embryonic fibroblasts and preadipocytes. Poult Sci. 2020;99(12):7142–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Verma M, Pandey S, Bhat IA, Mukesh B, Anand J, Chandra V, Sharma GT. Impact of l-carnitine on lipid content and post thaw survivability of buffalo embryos produced in vitro. Cryobiology. 2018;82:99–105.

    Article  CAS  PubMed  Google Scholar 

  66. Jin JX, Lee S, Taweechaipaisankul A, Kim GA, Lee BC. Melatonin regulates lipid metabolism in porcine oocytes. J Pineal Res 2017, 62(2).

  67. Li LL, Wang D, Ge CY, Yu L, Zhao JL, Ma HT. Dehydroepiandrosterone reduced lipid droplet accumulation via inhibiting cell proliferation and improving mitochondrial function in primary chicken hepatocytes. Physiol Res. 2018;67(3):443–56.

    Article  CAS  PubMed  Google Scholar 

  68. Willows R, Navaratnam N, Lima A, Read J, Carling D. Effect of different gamma-subunit isoforms on the regulation of AMPK. Biochem J. 2017;474(10):1741–54.

    Article  CAS  PubMed  Google Scholar 

  69. Gai H, Zhou F, Zhang Y, Ai J, Zhan J, You Y, Huang W. Coniferaldehyde ameliorates the lipid and glucose metabolism in palmitic acid-induced HepG2 cells via the LKB1/AMPK signaling pathway. J Food Sci. 2020;85(11):4050–60.

    Article  CAS  PubMed  Google Scholar 

  70. Grahame Hardie D. Regulation of AMP-activated protein kinase by natural and synthetic activators. Acta Pharm Sin B. 2016;6(1):1–19.

    Article  PubMed  Google Scholar 

  71. Ponnusamy L, Natarajan SR, Thangaraj K, Manoharan R. Therapeutic aspects of AMPK in breast cancer: Progress, challenges, and future directions. Biochim Biophys Acta Rev Cancer. 2020;1874(1):188379.

    Article  CAS  PubMed  Google Scholar 

  72. Hardie DG. AMPK: positive and negative regulation, and its role in whole-body energy homeostasis. Curr Opin Cell Biol. 2015;33:1–7.

    Article  CAS  PubMed  Google Scholar 

  73. Gao X, Lin SH, Ren F, Li JT, Chen JJ, Yao CB, Yang HB, Jiang SX, Yan GQ, Wang D, et al. Acetate functions as an epigenetic metabolite to promote lipid synthesis under hypoxia. Nat Commun. 2016;7:11960.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Li D, Liu F, Wang X, Li X. Apple Polyphenol Extract alleviates High-Fat-Diet-Induced hepatic steatosis in male C57BL/6 mice by targeting LKB1/AMPK pathway. J Agric Food Chem. 2019;67(44):12208–18.

    Article  CAS  PubMed  Google Scholar 

  75. Smith BK, Marcinko K, Desjardins EM, Lally JS, Ford RJ, Steinberg GR. Treatment of nonalcoholic fatty liver disease: role of AMPK. Am J Physiol Endocrinol Metab. 2016;311(4):E730–40.

    Article  PubMed  Google Scholar 

  76. Fei Z, Bera TK, Liu X, Xiang L, Pastan I. Ankrd26 gene disruption enhances adipogenesis of mouse embryonic fibroblasts. J Biol Chem. 2011;286(31):27761–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Zhou W, Tu Y, Simpson PJ, Kuhajda FP. Malonyl-CoA decarboxylase inhibition is selectively cytotoxic to human breast cancer cells. Oncogene. 2009;28(33):2979–87.

    Article  CAS  PubMed  Google Scholar 

  78. Zhao X, Feng D, Wang Q, Abdulla A, Xie XJ, Zhou J, Sun Y, Yang ES, Liu LP, Vaitheesvaran B, et al. Regulation of lipogenesis by cyclin-dependent kinase 8-mediated control of SREBP-1. J Clin Invest. 2012;122(7):2417–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Zhao J, Zhang X, Gao T, Wang S, Hou Y, Yuan P, Yang Y, Yang T, Xing J, Li J, et al. SIK2 enhances synthesis of fatty acid and cholesterol in ovarian cancer cells and tumor growth through PI3K/Akt signaling pathway. Cell Death Dis. 2020;11(1):25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Tepekoy F, Akkoyunlu G, Demir R. The role of wnt signaling members in the uterus and embryo during pre-implantation and implantation. J Assist Reprod Genet. 2015;32(3):337–46.

    Article  PubMed  Google Scholar 

  81. Luo Y, Huang S, Wei J, Zhou H, Wang W, Yang J, Deng Q, Wang H, Fu Z. Long noncoding RNA LINC01606 protects colon cancer cells from ferroptotic cell death and promotes stemness by SCD1-Wnt/beta-catenin-TFE3 feedback loop signalling. Clin Transl Med. 2022;12(4):e752.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Shimomura I, Shimano H, Horton JD, Goldstein JL, Brown MS. Differential expression of exons 1a and 1c in mRNAs for sterol regulatory element binding protein-1 in human and mouse organs and cultured cells. J Clin Invest. 1997;99(5):838–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Vergnes L, Chin RG, de Aguiar Vallim T, Fong LG, Osborne TF, Young SG, Reue K. SREBP-2-deficient and hypomorphic mice reveal roles for SREBP-2 in embryonic development and SREBP-1c expression. J Lipid Res. 2016;57(3):410–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Song Z, Xiaoli AM, Yang F. Regulation and Metabolic Significance of De Novo Lipogenesis in Adipose Tissues. Nutrients 2018, 10(10).

  85. Eberle D, Hegarty B, Bossard P, Ferre P, Foufelle F. SREBP transcription factors: master regulators of lipid homeostasis. Biochimie. 2004;86(11):839–48.

    Article  CAS  PubMed  Google Scholar 

  86. Aardema H, Vos PL, Lolicato F, Roelen BA, Knijn HM, Vaandrager AB, Helms JB, Gadella BM. Oleic acid prevents detrimental effects of saturated fatty acids on bovine oocyte developmental competence. Biol Reprod. 2011;85(1):62–9.

    Article  CAS  PubMed  Google Scholar 

  87. Li S, Yang Z, Zhang H, Peng M, Ma H. Potential role of ALDH3A2 on the lipid and glucose metabolism regulated by (-)-hydroxycitric acid in chicken embryos. Anim Sci J. 2019;90(8):961–76.

    Article  CAS  PubMed  Google Scholar 

  88. Ishibashi K, Takeda Y, Nakatani E, Sugawara K, Imai R, Sekiguchi M, Takahama R, Ohkura N, Atsumi GI. Activation of PPARgamma at an early stage of differentiation enhances adipocyte differentiation of MEFs derived from type II Diabetic TSOD mice and alters lipid droplet morphology. Biol Pharm Bull. 2017;40(6):852–9.

    Article  CAS  PubMed  Google Scholar 

  89. Kajdasz A, Warzych E, Derebecka N, Madeja ZE, Lechniak D, Wesoly J, Pawlak P. Lipid Stores and Lipid Metabolism Associated Gene Expression in Porcine and Bovine Parthenogenetic Embryos Revealed by Fluorescent Staining and RNA-seq. Int J Mol Sci 2020, 21(18).

  90. Lanzarini F, Pereira FA, Camargo J, Oliveira AM, Belaz KRA, Melendez-Perez JJ, Eberlin MN, Brum MCS, Mesquita FS, Sudano MJ. ELOVL5 participates in embryonic lipid determination of Cellular membranes and cytoplasmic droplets. Int J Mol Sci 2021, 22(3).

  91. Sun SX, Ren TY, Li X, Cao XJ, Gao J. Polyunsaturated fatty acids synthesized by freshwater fish: a new insight to the roles of elovl2 and elovl5 in vivo. Biochem Bioph Res Co. 2020;532(3):414–9.

    Article  CAS  Google Scholar 

  92. Bai X, Huang LJ, Chen SW, Nebenfuehr B, Wysolmerski B, Wu JC, Olson SK, Golden A, Wang CW. Loss of the seipin gene perturbs eggshell formation in Caenorhabditis elegans. Development 2020, 147(20).

  93. Li S, Yang Z, Zhang H, Peng M, Ma H. (-)-Hydroxycitric Acid Influenced Fat Metabolism via modulating of glucose-6-phosphate isomerase expression in Chicken embryos. J Agric Food Chem. 2019;67(26):7336–47.

    Article  CAS  PubMed  Google Scholar 

  94. Fauny JD, Silber J, Zider A. Drosophila lipid Storage Droplet 2 gene (Lsd-2) is expressed and controls lipid storage in wing imaginal discs. Dev Dyn. 2005;232(3):725–32.

    Article  CAS  PubMed  Google Scholar 

  95. Yen CL, Stone SJ, Koliwad S, Harris C, Farese RV Jr. Thematic review series: glycerolipids. DGAT enzymes and triacylglycerol biosynthesis. J Lipid Res. 2008;49(11):2283–301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Uzbekova S, Elis S, Teixeira-Gomes AP, Desmarchais A, Maillard V, Labas V. MALDI Mass Spectrometry imaging of lipids and gene expression reveals differences in fatty acid metabolism between follicular compartments in Porcine Ovaries. Biology (Basel). 2015;4(1):216–36.

    CAS  PubMed  Google Scholar 

  97. Idrissi FZ, Grotsch H, Fernandez-Golbano IM, Presciatto-Baschong C, Riezman H, Geli MI. Distinct acto/myosin-I structures associate with endocytic profiles at the plasma membrane. J Cell Biol. 2008;180(6):1219–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Aizawa R, Ibayashi M, Tatsumi T, Yamamoto A, Kokubo T, Miyasaka N, Sato K, Ikeda S, Minami N, Tsukamoto S. Synthesis and maintenance of lipid droplets are essential for mouse preimplantation embryonic development. Development 2019, 146(22).

  99. Magre J, Delepine M, Khallouf E, Gedde-Dahl T Jr, Van Maldergem L, Sobel E, Papp J, Meier M, Megarbane A, Bachy A, et al. Identification of the gene altered in Berardinelli-Seip congenital lipodystrophy on chromosome 11q13. Nat Genet. 2001;28(4):365–70.

    Article  CAS  PubMed  Google Scholar 

  100. Windpassinger C, Auer-Grumbach M, Irobi J, Patel H, Petek E, Horl G, Malli R, Reed JA, Dierick I, Verpoorten N, et al. Heterozygous missense mutations in BSCL2 are associated with distal hereditary motor neuropathy and silver syndrome. Nat Genet. 2004;36(3):271–6.

    Article  CAS  PubMed  Google Scholar 

  101. Deneke VE, Puliafito A, Krueger D, Narla AV, De Simone A, Primo L, Vergassola M, De Renzis S, Di Talia S. Self-Organized Nuclear Positioning synchronizes the cell cycle in Drosophila embryos. Cell. 2019;177(4):925–941e917.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Key Research and Development Program of China (No. 2021YFF1000601), the National Natural Science Foundation of China (No. 32172700), the Joint Funds of the National Natural Science Foundation of China (No. U20A2052), the Fundamental Research Funds for the Central Universities(No.2662022DKPY002).

Author information

Authors and Affiliations

Authors

Contributions

T.L. wrote this review and contributed to the figures. Y.J. provided the idea. J.W. and Z.R. supervised the study. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Zhuqing Ren.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no conflict of interest.

Additional information

Publisher’s Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, T., Jin, Y., Wu, J. et al. Beyond energy provider: multifunction of lipid droplets in embryonic development. Biol Res 56, 38 (2023). https://doi.org/10.1186/s40659-023-00449-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40659-023-00449-y

Keywords