Hostname: page-component-8448b6f56d-m8qmq Total loading time: 0 Render date: 2024-04-24T01:48:26.432Z Has data issue: false hasContentIssue false

Review: Lipid biology in the periparturient dairy cow: contemporary perspectives

Published online by Cambridge University Press:  06 February 2020

J. W. McFadden*
Affiliation:
Department of Animal Science, Cornell University, 48 Judd Falls Rd., Ithaca, 14853, NY, USA
*

Abstract

Coordinated changes in energy metabolism develop to support gestation and lactation in the periparturient dairy cow. Maternal physiology involves the partitioning of nutrients (i.e. glucose, amino acids and fatty acids (FA)) for fetal growth and milk synthesis. However, the inability of the dairy cow to successfully adapt to a productive lactation may trigger metabolic stress characterized by uncontrolled adipose tissue lipolysis and reduced insulin sensitivity. A consequence is lipotoxicity and hepatic triglyceride deposition that favors the development of fatty liver disease (FLD) and ketosis. This review describes contemporary perspectives pertaining to FA surfeit and complex lipid metabolism in the transition dairy cow. The role of saturated and unsaturated FA as bioactive signaling molecules capable of modulating insulin secretion and sensitivity is explored. Moreover, the metabolic fate of FA as influenced by mitochondrial function is considered. This includes the influence of inadequate mitochondrial oxidation on acylcarnitine status and the use of FA for lipid mediator synthesis. Lipid mediators, including the sphingolipid ceramide and diacylglycerol, are evaluated considering their established ability to inhibit insulin signaling and glucose transport in non-ruminant diabetics. The mechanisms of FLD in the transition cow are revisited with attention centered on glycerophospholipid phosphatidylcholine and triglyceride secretion. The relationship between oxidative stress and oxylipids within the context of insulin antagonism, hepatic steatosis and inflammation is also reviewed. Lastly, peripartal hormonal involvement or lack thereof of adipokines (i.e. leptin, adiponectin) and hepatokines (i.e. fibroblast growth factor-21) is described. Similarities and differences in ruminant and non-ruminant physiology are routinely showcased. Unraveling the lipidome of the dairy cow has generated breakthroughs in our understanding of periparturient lipid biology. Therapeutic approaches that target FA and complex lipid metabolism holds promise to enhance cow health, well-being and productive lifespan.

Type
Review Article
Copyright
© The Animal Consortium 2020

Implications

Metabolic stress may compromise the ability of the dairy cow to successfully adapt to a productive lactation and remain fertile and healthy. This review discusses changes in lipid metabolism that define maternal adaptation and metabolic stress in the cow. The narrative describes the roles of fatty acids, mitochondria and acylcarnitines, lipid mediators of insulin antagonism, including ceramide, glycerophospholipids and triglyceride secretion, and oxylipids during oxidative imbalance. The influence of leptin, adiponectin and fibroblast growth factors is considered. Defining the peripartal lipidome is important to refine existing or identify new therapeutic strategies to improve cow health and production efficiency.

Introduction

The dairy cow transitioning from gestation to lactation experiences dynamic changes in energy metabolism that develop as a means to support fetal and neonatal development (Bauman and Currie, Reference Bauman and Currie1980; Roche et al., Reference Roche, Bell, Overton and Loor2013). At the onset of the periparturient period (i.e. late gestation), the utilization of glucose and amino acids to support the growth of the fetus increases. Moreover, the demand for these nutrients as well as fatty acids (FA) and minerals only accelerates as milk production initiates. Although the survival of offspring demands nutrients for reproduction and lactation, the peripartal cow experiences a decline in energy intake immediately prior to parturition. Energy intake also remains inadequate throughout early lactation. The consequence is the cow’s intrinsic reliance on coordinated metabolic adaptations to ensure that nutrient requirements for maintenance, conceptus growth, mammogenesis and lactogenesis are met.

The classic understanding of physiological adaptations that support nutrient partitioning in dairy cattle have been summarized (Baumgard et al., Reference Baumgard, Collier and Bauman2017). In brief, de novo fat synthesis and FA uptake and esterification are blunted in adipose tissue. Whereas, lipolysis is accelerated. Circulating FA are then oxidized in liver and skeletal muscle tissues and incorporated into milk triglycerides in the mammary gland. In muscle, protein synthesis is reduced and amino acid mobilization is enhanced. In turn, amino acids are utilized to support increased rates of gluconeogenesis (i.e. alanine) or milk protein synthesis. Hepatic glycogenolysis and ketogenesis are enhanced. Glucose utilization by skeletal muscle and adipose tissue is reduced to spare glucose for milk lactose synthesis. These metabolic events are controlled in part by a reduction in peripheral insulin sensitivity and responsiveness, as well as reduced pancreatic insulin secretion. Although the mechanisms of reduced insulin-stimulated glucose disposal are not entirely defined in the cow, uncoupling of the somatotropic axis is involved (Lucy et al., Reference Lucy, Jiang and Kobayashi2001, Reference Lucy, Verkerk, Whyte, Macdonald, Burton, Cursons, Roche and Holmes2009). Specifically, plasma somatotropin concentrations increase during early lactation, whereas the concentration of the insulin-sensitizer insulin-like growth factor-1 decline. The cause is likely attributed to a decrease in hepatic growth hormone receptor second messenger signaling in early-lactation dairy cows (Lucy et al., Reference Lucy, Jiang and Kobayashi2001). Nevertheless, low circulating insulin-like growth factor-1 concentrations help reduce glucose transport in skeletal muscle and adipose tissues, and enhance hepatic glucose output (Clemmons, Reference Clemmons2004; Wang et al., Reference Wang, Zhu, Chen, Li, Gao, Li, Zhang, Long and Wang2012). Enhanced adipose tissue responsiveness to catecholamines, and elevations in circulating glucocorticoids and glucagon are also evident in postpartum cows. It is important to emphasize that these adaptations are innate and should not be used to describe metabolic stress if milk production, health or well-being are not compromised. Indeed, cows with high productive efficiency rely heavily on these adaptations. Moreover, one could argue that delayed conception in a cow that maintains high milk production is not due to metabolic stress but simply reflects the metabolic decision of the cow to support one nutrient sink over another.

Metabolic stress is a consequence of inadequate metabolic adaptation (Sordillo and Raphael, Reference Sordillo and Raphael2013). By definition, unresolved metabolic stress triggers metabolic disorders such as subclinical or clinical ketosis, fatty liver disease (FLD) characterized by triglyceride deposition in the absence or presence of inflammation, and hypocalcemia. These disorders are linked to lower milk production, lower conception rates and increased calving intervals, lameness and compromised well-being and productive lifespan in the herd (Mulligan and Doherty, Reference Mulligan and Doherty2008). Implications for metabolic stress include increased replacement rates, inefficient nutrient utilization and economic loss.

This review explores contemporary perspectives in bovine lipid biology defined as the study of lipid metabolism, structure and bioactive functionality and the interactions lipids have with changes in physiology in the dairy cow. The intent is to expand our understanding of nutrient partitioning and metabolic stress in the transition cow. The unique bioactive properties of FA, the physiological relevance of functional mitochondria, emerging insights in sphingolipid and glycerophospholipid biology, the consequences of oxidative imbalance and the involvement of adipokines are examined.

Fatty acids, and insulin secretion and sensitivity

In the transition cow, negative energy balance develops with increased concentrations of palmitic, stearic, oleic and linoleic acids in serum (Rukkwamsuk et al., Reference Rukkwamsuk, Geelen, Kruip and Wensing2000; Douglas et al., Reference Douglas, Rehage, Beaulieu, Bahaa and Drackley2007). These changes reflect the FA profile of adipose tissue. As the proportion of saturated FA increase in plasma, the proportion of long-chain polyunsaturated FA decreases (e.g. arachidonic acid; Douglas et al., Reference Douglas, Rehage, Beaulieu, Bahaa and Drackley2007). In parallel with increased lipolysis, the postpartum liver of the cow accumulates greater amounts of palmitic, oleic and linoleic acids, but not stearic acid (Rukkwamsuk et al., Reference Rukkwamsuk, Geelen, Kruip and Wensing2000). Enrichment of palmitic acid, oleic acid and linoleic acid is observed in hepatic triglycerides and phospholipids, whereas polyunsaturated FA (i.e. arachidonic and eicosapentaenoic acids) depletion is evident (Douglas et al., Reference Douglas, Rehage, Beaulieu, Bahaa and Drackley2007). Although not completely understood, the modulation of plasma FA status has implications for insulin secretion and sensitivity as well as liver health in the dairy cow.

During early lactation, insulin secretion in response to insulinotropic agents (i.e. glucose and propionate) is suppressed (Lomax et al., Reference Lomax, Baird, Mallinson and Symonds1979). This action advantageously removes the inhibitory signal on lipolysis, and thus circulating FA concentrations increase for oxidation and milk fat production. Therefore, it would be counterintuitive for FA to promote insulin secretion and lipogenesis during this stage of lactation. However, this concept deserves consideration especially in the mid- or late-lactation cow that is more responsive to insulinotropic compounds. The reason is that FA promote insulin secretion in non-ruminants (Poitout et al., Reference Poitout, Hagman, Artner, Stein, Harmon and Robertson2006). Evidence also suggests that these responses are uniquely influenced by the type of FA studied. In rats, glucose-stimulated insulin secretion from perfused pancreas is higher in response to co-stimulation with saturated palmitic or stearic acids, relative to oleic or linoleic acids (Stein et al., Reference Stein, Stevenson, Chester, Basit, Daniels, Turley and McGarry1997). The ability of saturated FA supplementation to increase circulating insulin concentrations has been observed in peak-lactation dairy cows (Harvatine and Allen, Reference Harvatine and Allen2006). One notable exception is that chronic excess of palmitic acid reduces insulin synthesis and secretion in human pancreatic islets via mechanisms that may depend on ceramide-mediated inhibition of insulin gene expression as well as reduced β-cell turnover and apoptotic activation (Maedler et al., Reference Maedler, Oberholzer, Bucher, Spinas and Donath2003). Considering that circulating palmitic acid and ceramide is elevated when plasma insulin is low during early lactation, might palmitic acid-induced lipotoxicity and ceramide-dependent mechanisms explain suppressed insulin secretion during early lactation? Although this question remains unanswered, it should be mentioned that monounsaturated FA (i.e. palmitoleic or oleic acids) counteract the harmful effects of palmitic acid on human pancreatic β-cell turnover and function (Maedler et al., Reference Maedler, Oberholzer, Bucher, Spinas and Donath2003). Of potential relevance in the cow, feeding a blend of palmitic and oleic acids increased circulating insulin concentrations in mid-lactation Holstein cows, relative to those fed high palmitic acid or a blend of palmitic and stearic acids (de Souza et al., Reference de Souza, Preseault and Lock2018).

In the cow, the alteration of peripheral insulin sensitivity by FA may influence lipolysis and the mammary utilization of glucose for milk synthesis. Specifically, diet- or lipolytic-derived FA may control insulin signaling, although regulation likely depends on the degree of FA saturation, chain length and concentration, and whether the cow is under homeorhetic control. In non-ruminants, it is hypothesized that saturated FA inhibit insulin sensitivity by promoting lipid mediator synthesis (e.g. ceramide or diacylglycerol (DAG)), inflammation via toll-like receptor-4 signaling, oxidative stress or endoplasmic reticulum stress (Boden, Reference Boden2011). In contrast, n-3 FA such as docosahexaenoic acid (DHA) appear to counteract insulin resistance development by stimulating mitochondrial function, reducing reactive oxygen species (ROS) production and preventing inflammation in non-ruminants, as reviewed by Lepretti et al. (Reference Lepretti, Martucciello, Burgos Aceves, Putti and Lionetti2018). Similar benefits are observed when studying monounsaturated FA (i.e. oleic acid; Palomer et al., Reference Palomer, Pizarro-Delgado, Barroso and Vázquez-Carrera2018). In dairy cattle, an intravenous infusion of saturated tallow caused insulin resistance (Pires et al., Reference Pires, Souza and Grummer2007). Evidence suggests that palmitic acid partitions nutrients to the mammary gland, whereas oleic acid shifts energy away from milk production and toward body fat accretion (de Souza et al., Reference de Souza, Preseault and Lock2018). In addition, abomasal infusion of linseed oil rich in polyunsaturated linolenic acid appeared to enhance the anti-lipolytic effects of insulin in feed-restricted cows (Pires et al., Reference Pires, Pescara, Brickner, Silva del Rio, Cunha and Grummer2008). In growing steers, long-chain eicosapentaenoic and DHA increases insulin sensitivity, relative to animals unsupplemented with n-3 FA (Cartiff et al., Reference Cartiff, Fellner and Eisemann2013). Although a better mechanistic understanding is required, the current literature suggests that saturated FA reduce insulin sensitivity in dairy cattle, whereas unsaturated FA (particularly n-3 FA) enhance insulin action. Future research should determine whether and how saturated and unsaturated FA influence insulin signaling and inflammation in early-lactation cow, and whether these changes are associated with the advancement of FLD, the incidence of metabolic disorders and long-term lactation and reproductive success of the cow.

Acylcarnitines and mitochondrial β-oxidation

A key step in the β-oxidation of FA is the formation of acylcarnitines as a means to shuttle fatty acyl-CoA from the cytosol into the mitochondrion. This process is facilitated by carnitine palmitoyltransferase-1, which is inhibited by malonyl-CoA generated by the lipogenic enzyme acetyl-CoA carboxylase. In the study of obesity and type 2 diabetes, acylcarnitines have received attention as biomarkers of lipid-induced mitochondrial dysfunction (Schooneman et al., Reference Schooneman, Vaz, Houten and Soeters2013). The accumulation of acylcarnitines reflects a reduction in FA oxidation capacity and the partitioning of FA toward the synthesis of triglyceride and lipid mediators of insulin antagonism, including ceramide and diacylglycerol. Types of acylcarnitines include ketone-derived (e.g. the carnitine ester of β-hydroxybutyrate), branched-chain amino acid-derived (e.g. C4-dicarboxylic-carnitine), and acylcarnitines varying in FA chain length (e.g. acetyl-, myristoyl- or palmitoyl-carnitine, respectively). Focus has centered on long-chain acylcarnitines because they are linked to insulin resistance (Schooneman et al., Reference Schooneman, Vaz, Houten and Soeters2013). In the study of Koves et al. (Reference Koves, Ussher, Noland, Slentz, Mosedale, Ilkayeva, Bain, Stevens, Dyck and Newgard2008), chronic high-fat feeding increased postprandial serum total FA as well as C8:1-, C10:3-, C16:0-, C18:0- and C18:1-carnitine concentrations in rats. In the same study, acylcarnitine levels were elevated in gastrocnemius muscles in ad libitum high-fat fed obese Zucker diabetic fatty rats experiencing elevations in FA oxidation to acid-soluble metabolites (a measure of incomplete FA catabolism), relative to lean animals. More recent data suggest that acylcarnitines may directly modulate insulin signaling. In differentiated myotubes of non-ruminant origin, C16:0-carnitine treatment inhibited insulin-stimulated protein kinase B (AKT) activation and glucose uptake (Aguer et al., Reference Aguer, McCoin, Knotts, Thrush, Ono-Moore, McPherson, Dent, Hwang, Adams and Harper2015). In addition, lowering of acetyl-carnitine concentrations by mildronate (an inhibitor of acylcarnitine transferase) prevents palmitate-induced reductions in insulin-stimulated glucose utilization by human primary myotubes derived from lean subjects (Aguer et al., Reference Aguer, McCoin, Knotts, Thrush, Ono-Moore, McPherson, Dent, Hwang, Adams and Harper2015). Interestingly, mildronate also prevented palmitic acid-induced accumulation of ROS, which suggests that acylcarnitines may promote oxidative stress concomitantly with impaired insulin sensitivity.

Acylcarnitine metabolism has also been implicated in the development of FLD. Specifically, hepatic acylcarnitine levels are elevated in humans with non-alcoholic FLD (e.g. C16:0- and C12:0-carnitines in steatosis and steatohepatitis, respectively; Lake et al., Reference Lake, Novak, Shipkova, Aranibar, Robertson, Reily, Lehman-McKeeman, Vaillancourt and Cherrington2015). Albeit de novo lipogenesis plays a role in non-ruminant FLD, mitochondrial dysfunction and FA excess also provides FA substrate for excess triglyceride storage, which favors steatosis. The advancement of hepatic injury in the form of inflammation (steatosis to steatohepatitis) may be provoked by acylcarnitines. In support, C14:0-carnitine supplementation stimulated the expression and secretion of pro-inflammatory tumor necrosis factor-α (TNF-α) from RAW 264.7 macrophage cells (Rutkowsky et al., Reference Rutkowsky, Knotts, Ono-Moore, McCoin, Huang, Schneider, Singh, Adams and Hwang2014). This is of potential significance considering that TNF-α is associated with FLD in humans and dairy cows (Ohtsuka et al., Reference Ohtsuka, Koiwa, Hatsugaya, Kudo, Hoshi, Itoh, Yokota, Okada and Kawamura2001; Marcellini et al., Reference Marcellini, Nobili, Manco and Giannone2007). Interestingly, hepatic branched-chain amino acid concentrations (i.e. leucine, isoleucine and valine) are elevated with amplified acylcarnitine status during advanced FLD (Lake et al., Reference Lake, Novak, Shipkova, Aranibar, Robertson, Reily, Lehman-McKeeman, Vaillancourt and Cherrington2015). Moreover, elevations in branched-chain amino acid concentrations in the presence of high saturated fat intake may overload mitochondrial fuel oxidation via anaplerosis to form incompletely oxidized lipid-derived metabolites such as acylcarnitines and exacerbate insulin resistance (Newgard et al., Reference Newgard, An, Bain, Muehlbauer, Stevens, Lien, Haqq, Shah, Arlotto, Slentz, Rochon, Gallup, Ilkayeva, Wenner, Yancy, Eisenson, Musante, Surwit, Millington, Butler and Svetkey2009). Lastly, although the accrual of hepatic acylcarnitines promotes their accumulation in circulation via mechanisms that may involve organic cation/carnitine transporter-2, it deserves mentioning that the plasma acylcarnitine profile may inadequately reflect hepatic acylcarnitine metabolism in mammals (i.e. plasma levels do not reflect tissue levels; Schooneman et al., Reference Schooneman, Achterkamp, Argmann, Soeters and Houten2014).

In dairy cattle transitioning from gestation to lactation, the capacity to completely oxidize FA to CO2 is lowered, whereas incomplete oxidation to acid-soluble metabolites is elevated (Litherland et al., Reference Litherland, Dann and Drackley2011). In the muscle of transition cow, long-chain acylcarnitines accumulate with short- and medium-chain acylcarnitines during the peripartum (Yang et al., Reference Yang, Sadri, Prehn, Adamski, Rehage, Dänicke, Saremi and Sauerwein2019), suggesting that β-oxidation to acetyl-CoA increased but the tricarboxylic acid cycle was downregulated. In sick transition cows (i.e. those with mastitis, metritis, retained placenta and/or laminitis; n = 6), plasma short-chain propionyl-carnitine concentrations were elevated, relative to six healthy controls (Hailemariam et al., Reference Hailemariam, Mandal, Saleem, Dunn, Wishart and Ametaj2014). In over-conditioned transition cows that mobilize more FA postpartum, plasma C14:0-, C16:0-, C18:0- and C20:0-carnitine levels are elevated, relative to lean cows (Rico et al., Reference Rico, Zang, Haughey, Rius and McFadden2018b). In the same study, plasma total acylcarnitine levels were positively correlated with circulating total FA concentrations, as well as total and C24:0-linked ceramide. Although this study supports that long-chain acylcarnitines were formed and exported out of the mitochondrion, the work did not assess FA oxidation or functionality of the TCA cycle. Regardless, hepatic FA that are not oxidized can serve as substrate for triglyceride esterification and are potentially partitioned toward ceramide synthesis in over-conditioned cows prone to accelerated lipolysis and FLD.

Our understanding of acylcarnitine status within the context of productivity and health requires further study. The reason is because elevations in serum carnitine or long-chain acylcarnitine (e.g. C16:0- or 18:2-carnitine) levels during the transition period or peak lactation have been observed in cows that remain clinically healthy beyond 100 days in milk, relative to those culled beyond peak milk production (Huber et al., Reference Huber, Dänicke, Rehage, Sauerwein, Otto, Rolle-Kampczyk and Von Bergen2016). It remains to be determined whether elevations in acylcarnitines during transition are predictive of metabolic disorders (e.g. ketosis) during early lactation. It is conceivable that fully functional mitochondria that manage FA surfeit during early lactation would protect the cow from lipotoxicity and FLD to enhance performance. In support, dietary carnitine supplementation has been shown to stimulate palmitate β-oxidation and decrease liver triglyceride concentrations in postpartum cows (Carlson et al., Reference Carlson, McFadden, D’Angelo, Woodworth and Drackley2007). Cows with a single nucleotide polymorphism in mitochondrial transcription factor A, an autosomal gene essential for transcription and replication of mitochondrial DNA, are less fertile and more likely to be culled, albeit they produce more milk (Clempson et al., Reference Clempson, Pollott, Brickell, Bourne, Munce and Wathes2011). Moreover, evidence in growing steers suggests that increased mitochondrial function may contribute to enhanced feed efficiency during compensatory growth that follows feed restriction (Connor et al., Reference Connor, Kahl, Elsasser, Parker, Li, Van Tassell, Baldwin and Barao2010).

Lipid mediators, insulin sensitivity and nutrient partitioning

Sphingolipids including ceramides

Sphingolipids are composed of a sphingoid base backbone (i.e. D-erythro-sphingosine) and an FA linked via an amide bond. Types of sphingolipids include ceramide and those with a polar head group, including monohexosylceramide (i.e. glucosyl- or galactosyl-ceramide; GlcCer), lactosylceramide and phosphocholine-containing sphingomyelin. Even more complex in structure are gangliosides, which are glycosphingolipids containing one or more sialic acid residues. Although sphingolipids are diverse in structure, their de novo synthesis begins with ceramide controlled in part by serine palmitoyltransferase and ceramide synthase (CerS). Serine palmitoyltransferase controls the condensation of palmitoyl-CoA and serine to produce 3-ketodihydrosphingosine. Whereas, one of six CerS isoforms attaches a second FA (most often saturated) to sphinganine to synthesize the ceramide precursor dihydroceramide. For instance, CerS6 and CerS2 are involved in the production of the highly abundant C16:0- and C24:0-ceramide in mammals, respectively (Levy and Futerman, Reference Levy and Futerman2010). An alternative pathway that produces ceramide is controlled by sphingomyelinase (SMase; acid or neutral) which hydrolyzes sphingomyelin. Once ceramide is formed, it may be used by glucosylceramide synthase to form GlcCer and eventually gangliosides, converted to sphingomyelin by sphingomyelin synthase, phosphorylated by ceramide kinase, or degraded by ceramidase to form sphingosine and FA (McFadden and Rico, Reference McFadden and Rico2019).

The diversity of sphingolipid structure dictates their equally diverse function. Sphingolipids are structurally important as aggregates within cellular membrane rafts and caveolae that often include caveolin (Liu and Anderson, Reference Liu and Anderson1995). Ceramide, glycosylated ceramide and sphingomyelin are also components of lipoproteins. Specifically, ceramides are primarily enriched in low-density lipoproteins (LDL) as observed in humans (Wiesner et al., Reference Wiesner, Leidl, Boettcher, Schmitz and Liebisch2009) and dairy cows (Davis et al., Reference Davis, Rico, Myers, Coleman, Clapham, Haughey and McFadden2019); however, their incorporation within very-low-density lipoproteins (VLDL) increases with fasting (Wiesner et al., Reference Wiesner, Leidl, Boettcher, Schmitz and Liebisch2009). Bioactive ceramides and glycosylated ceramides behave as second messengers and interact with proteins to modulate cell signaling. From an historical perspective, ceramide is recognized for its role in the downregulation of cell proliferation, induction of cell differentiation and initiation of caspase-mediated programed cell death (Pushkareva et al., Reference Pushkareva, Obeid and Hannun1995). Today, sphingolipids have received attention for their respective role within the progression of type 2 diabetes, non-alcoholic FLD and cardiovascular disease in humans. Emerging evidence also suggests that sphingolipids decrease insulin signaling in dairy cattle, which may accelerate glucose and FA partitioning toward liver and the mammary gland (Rico et al., Reference Rico, Bandaru, Dorskind, Haughey and McFadden2015, Reference Rico, Saed Samii, Mathews, Lovett, Haughey and McFadden2017a and Reference Rico, Myers, Laub, Davis, Zeng and McFadden2018c).

The ability of ceramide to inhibit insulin-stimulated glucose utilization is a feature that defines lipotoxicity and insulin resistance in skeletal muscle and adipose tissue of non-ruminants (Chavez and Summers, Reference Chavez and Summers2012). The mechanisms by which ceramide influences insulin sensitivity are multifaceted. It was first discovered that excess long-chain FA promote lipoapoptosis (i.e. apoptosis caused by FA overaccumulation otherwise known as steatosis) of pancreatic β-cells to contribute to inadequate insulin secretion in Zucker diabetic fatty rats (Shimabukuro et al., Reference Shimabukuro, Higa, Zhou, Wang, Newgard and Unger1998). The apoptotic effect was due, in part, to enhanced de novo ceramide synthesis. Subsequent research demonstrated that ceramide inactivates AKT to downregulate the translocation of glucose transporter-4 to the plasma membrane in myotubes and adipocytes challenged by insulin (Chavez and Summers, Reference Chavez and Summers2012). Although AKT inactivation appears fundamentally involved, ceramide-mediated insulin resistance also appears to include the activation of phosphatase and tensin homolog and protein phosphatase 2A, as well as the caveolin-enriched microdomain-recruitment of protein kinase C-ζ (Chavez and Summers, Reference Chavez and Summers2012). Recent evidence also suggests that extracellular-derived ceramide within LDL may also downregulate insulin signaling in the muscle (Boon et al., Reference Boon, Hoy, Stark, Brown, Meex, Henstridge, Schenk, Meikle, Horowitz, Kingwell, Bruce and Watt2013). Interestingly, pharmacological inhibition of serine palmitoyltransferase or glucosylceramide synthase (de novo ceramide and GlcCer synthesis, respectively) is an effective means to improve insulin sensitivity in the muscle of obese rodents (Aerts et al., Reference Aerts, Ottenhoff, Powlson, Grefhorst, van Eijk, Dubbelhuis, Aten, Kuipers, Serlie and Wennekes2007; Holland et al., Reference Holland, Brozinick, Wang, Hawkins, Sargent, Liu, Narra, Hoehn, Knotts, Siesky, Nelson, Karathanasis, Fontenot, Birnbaum and Summers2007). A similar outcome has been observed in cultured C2C12 myotubes that overexpress acid ceramidase (Chavez et al., Reference Chavez, Holland, Bär, Sandhoff and Summers2005) or in CerS6-deficient mice that exhibit reduced C16:0-ceramide concentrations in white adipose tissue and liver (Turpin et al., Reference Turpin, Nicholls, Willmes, Mourier, Brodesser, Wunderlich, Mauer, Xu, Hammerschmidt and Brönneke2014).

Our understanding of sphingolipid biology in the dairy cow has expanded in recent years. It was first observed that plasma ceramide (e.g. total ceramide, and C18:0-, C20:0-, C22:0- and C24:0-ceramide) and GlcCer (e.g. C16:0- and C18:0-GlcCer) concentrations increase with the transition from gestation to lactation, more so for cows with elevated prepartum body condition and postpartum circulating total FA concentrations (Rico et al., Reference Rico, Bandaru, Dorskind, Haughey and McFadden2015 and Reference Rico, Saed Samii, Mathews, Lovett, Haughey and McFadden2017a). During the transition period, postpartum liver total and C24:0-ceramide concentrations increase progressively in over-conditioned dairy cows with hepatic lipid accumulation, and C16:0-ceramide concentrations increase in skeletal muscle tissue and plasma LDL fractions in all cows independent of prepartum body condition status (Rico et al., Reference Rico, Saed Samii, Mathews, Lovett, Haughey and McFadden2017a; Davis et al., Reference Davis, Rico, Myers, Coleman, Clapham, Haughey and McFadden2019). The enhanced supply of circulating and hepatic ceramide was also observed in non-pregnant and non-lactating Holstein cows intravenously infused a triglyceride emulsion, or feed-restricted to enhance circulating total FA concentrations (Davis et al., Reference Davis, Clegg, Perry and McFadden2017; Rico et al., Reference Rico, Giesy, Haughey, Boisclair and McFadden2018a). In these studies, ceramide supply is positively related to circulating total FA supply, and inversely related to indirect and direct measures of systemic insulin sensitivity in cows (Rico et al., Reference Rico, Bandaru, Dorskind, Haughey and McFadden2015 and Reference Rico, Saed Samii, Mathews, Lovett, Haughey and McFadden2017a; Davis et al., Reference Davis, Clegg, Perry and McFadden2017). These findings suggest that ceramide may antagonize insulin-stimulated glucose uptake. In support, the treatment of primary bovine-differentiated adipocytes with hydrophilic C2:0-ceramide decreased AKT Ser-473 phosphorylation (i.e. activation) and insulin-stimulated 2-deoxy-d-[3H]-glucose uptake (Rico et al., Reference Rico, Myers, Laub, Davis, Zeng and McFadden2018c). While in contrast, the treatment of bovine adipocytes with the serine palmitoyltransferase inhibitor myriocin lowered intracellular ceramide concentrations, and enhanced AKT phosphorylation and 2-deoxy-d-[3H]-glucose uptake in the presence of insulin (Rico et al., Reference Rico, Myers, Laub, Davis, Zeng and McFadden2018c). Collectively, our findings suggest that adipose ceramide accrual may promote lipolysis via the inhibition of insulin signaling. In support, glucose-stimulated reductions in circulating total FA levels are inversely related to plasma ceramide concentrations in lactating cows (Rico et al., Reference Rico, Mathews, Lovett, Haughey and McFadden2016).

It is likely that increases in saturated FA uptake by tissues drives de novo ceramide synthesis in cows. In support, dietary palmitic acid feeding increases plasma and hepatic ceramide levels in mid-lactation cows, relative to no added fat or stearic acid (Rico et al., Reference Rico, Mathews, Lovett, Haughey and McFadden2016 and Reference Rico, Rico, Phipps, Zeng, Corl, Chouinard, Gervais and McFadden2017b). Moreover, treatment of primary bovine neonatal hepatocytes with a de novo synthesis inhibitor prevents intracellular ceramide accumulation in response to palmitic acid incubation (McFadden et al., Reference McFadden, Rico, Erb and White2018). Potentially relevant in the transition cow experiencing FLD, inflammation and ceramide accrual, the induction of sphingomyelin hydrolysis by SMase may also contribute to ceramide synthesis. Several lines of evidence support this hypothesis. First, plasma sphingomyelin levels are lowest at calving (Rico et al., Reference Rico, Saed Samii, Mathews, Lovett, Haughey and McFadden2017a and Reference Rico, Zang, Haughey, Rius and McFadden2018b). Second, pro-inflammatory TNF-α induces acid SMase activation to generate ceramide and inhibit insulin signaling (i.e. insulin receptor substrate-1; Peraldi et al., Reference Peraldi, Hotamisligil, Buurman, White and Spiegelman1996). Third, serum TNF-α activity is negatively correlated with insulin-stimulated glucose disposal in dairy cows with FLD (Ohtsuka et al., Reference Ohtsuka, Koiwa, Hatsugaya, Kudo, Hoshi, Itoh, Yokota, Okada and Kawamura2001). The net contributions of de novo ceramide synthesis and sphingomyelin hydrolysis to ceramide pools within the context of FA supply and inflammation should be further evaluated in the periparturient cow.

Diacylglycerols

It deserves to be stated that the underlying molecular mechanisms of insulin resistance associated with obesity and type 2 diabetes are complex. Case in point, DAG is another lipid mediator and activator of protein kinase C in skeletal muscle and liver (Erion and Shulman, Reference Erion and Shulman2010). In both tissues, DAG accumulation decreases insulin-stimulated insulin receptor substrate tyrosine phosphorylation, and phosphoinositide 3-kinase activation. The result is the downregulation of glucose transporter-4 translocation and glucose uptake in response to insulin in muscle, and the inhibition of glycogen synthesis and induction of gluconeogenesis in liver. The question of whether ceramide or DAG is the leading cause of insulin resistance in diabetics is debated (Petersen and Jurczak, Reference Petersen and Jurczak2016; Summers and Goodpaster, Reference Summers and Goodpaster2016). In the dairy cow, the ability of DAG to modulate insulin signaling has not been tested. However, concurrent with hepatic triglyceride accumulation, liver DAG concentrations increase postpartum, relative to prepartum (Qin et al., Reference Qin, Kokkonen, Salin, Seppänen-Laakso, Taponen, Vanhatalo and Elo2017). In the study of Qin et al. (Reference Qin, Kokkonen, Salin, Seppänen-Laakso, Taponen, Vanhatalo and Elo2017), prepartum overfeeding did not modulate hepatic DAG concentrations. Because evidence does not support the development of insulin resistance in the liver of postpartum cows (Zachut et al., Reference Zachut, Honig, Striem, Zick, Boura-Halfon and Moallem2013), the observed elevation in hepatic DAG concentrations may only represent an intermediate in triglyceride synthesis and not a causal agent of insulin antagonism. Future studies will need to confirm the role of DAG in the liver and muscle of transition cows.

Phosphatidylcholines and hepatic triglyceride disposal

In humans, non-alcoholic FLD is attributed to enhanced de novo lipogenesis involving the induction of sterol regulatory element-binding protein (Kohjima et al., Reference Kohjima, Higuchi, Kato, Kotoh, Yoshimoto, Fujino, Yada, Yada, Harada and Enjoji2008). In contrast, hepatic triglyceride deposition in peripartal cows with FLD is due to enhanced esterification of lipolytic-derived FA. This process is likely exacerbated by the aforementioned inadequate mitochondrial β-oxidation of FA in the liver as well as adipose insulin antagonism, which would enhance FA supply for hepatic uptake. Additionally, the pathophysiology of FLD in cows likely involves a limited capacity to secrete triglyceride within VLDL. Indeed, triglyceride secretion is limited in ruminants, relative to non-ruminants (Pullen et al., Reference Pullen, Liesman and Emery1990), and the rate of hepatic VLDL secretion plateaus as intrahepatic fat content becomes severe in humans with non-alcoholic FLD (Fabbrini et al., Reference Fabbrini, Mohammed, Magkos, Korenblat, Patterson and Klein2008). In transition cows, a downregulation of hepatic apolipoprotein B100 mRNA expression and protein abundance may limit VLDL assembly and export to promote FLD (Bernabucci et al., Reference Bernabucci, Basiricò, Pirazzi, Rueca, Lacetera, Lepri and Nardone2009). An alternative hypothesis focuses on phosphatidylcholine (PC), a glycerophospholipid and principal component of the VLDL monolayer. Studies employing rodent models and choline-deficient diets have demonstrated the essential requirement of PC synthesis for VLDL secretion as a means to prevent steatosis (Fast and Vance, Reference Fast and Vance1995). Although the rapid ruminal degradation of unprotected choline and incorporation of choline into milk phospholipids may partially explain why plasma total choline and PC concentrations are low in postpartum cows (Artegoitia et al., Reference Artegoitia, Middleton, Harte, Campagna and de Veth2014), reduced choline availability has a potential to limit hepatic PC synthesis and VLDL secretion to favor the advancement of steatosis in the ruminant.

Hepatic synthesis of PC involves the cytidine diphosphate (CDP)–choline pathway (i.e. the Kennedy pathway) and the phosphatidylethanolamine N-methyltransferase (PEMT) pathway. Initiating the CDP–choline pathway, choline kinase utilizes free choline and ATP to form the PC-precursor phosphocholine. As observed in rat hepatocytes, choline kinase serves a critical function considering that ~70% of total PC are produced by the CDP–choline pathway (DeLong et al., Reference DeLong, Shen, Thomas and Cui1999). In liver, the PEMT pathway is recognized as a compensatory pathway that relies on coupled folate and transmethylation cycles, which fundamentally define one-carbon metabolism. Methyl donors, including methionine, choline and betaine, and one-carbon donors glycine and serine are utilized to generate S-adenosylmethionine for the transformation of phosphatidylethanolamine to PC by PEMT. Although undefined in cows, the CDP–choline pathway prefers DAG enriched in saturated and monounsaturated FA (i.e. palmitic and oleic acids, respectively). Instead, PEMT prefers phosphatidylethanolamine enriched in long- and very-long-chain polyunsaturated FA, including eicosatetraenoic acid and DHA (DeLong et al., Reference DeLong, Shen, Thomas and Cui1999). Indeed, circulating PC containing DHA has been recognized as a biomarker for PEMT activation in humans (da Costa et al., Reference da Costa, Sanders, Fischer and Zeisel2011). In the transition cow, lipolytic-derived FA (i.e. palmitic and oleic acids) may support CDP–choline pathway activation; however, it is hypothesized that hepatic polyunsaturated FA depletion may prevent PEMT activation (Myers et al., Reference Myers, Rico, Davis, Fontoura, Dineen, Tate and McFadden2019).

Associative relationships between circulating PC and the severity of hepatic steatosis have been repeatedly observed in dairy cows. For example, the levels of serum PC with short-chain FA moieties are low in cows with severe hepatic lipidosis, relative to clinically healthy cows (Imhasly et al., Reference Imhasly, Naegeli, Baumann, von Bergen, Luch, Jungnickel, Potratz and Gerspach2014). Moreover, suppressed hepatic levels of highly unsaturated PC have been observed in Holstein cows with moderate FLD (Saed Samii et al., Reference Saed Samii, Zang, Myers, Grilli and McFadden2018). With the intent to increase hepatic PC synthesis, feeding strategies that emphasize rumen-protected choline or methionine supplementation are routinely studied and applied as a potential means to prevent FLD in transition cows. For example, peripartal dietary rumen-protected choline supplementation has been shown to reduce hepatic triglyceride concentrations in postpartum cows (Zom et al., Reference Zom, Van Baal, Goselink, Bakker, De Veth and Van Vuuren2011). Recent in vitro work has aimed to delineate how choline and methionine are utilized by the CDP–choline and PEMT pathways (Zhou et al., Reference Zhou, Zhou, Batistel, Martinez-Cortés, Pate, Luchini and Loor2018). The potential of methyl donor feeding to enhance hepatic PC synthesis and lipid disposal may also have implications for improving inflammatory and immune status in cows (Zhou et al., Reference Zhou, Bulgari, Vailati-Riboni, Trevisi, Ballou, Cardoso, Luchini and Loor2016). Moving forward, the interplay between dietary FA and methyl donor efficacy to enhance hepatic PC synthesis and VLDL secretion should be examined. In support, Myers et al. (Reference Myers, Rico, Davis, Fontoura, Dineen, Tate and McFadden2019) hypothesize that DHA may selectively induce PEMT activation in cows.

Oxylipids and oxidative stress

Mitochondrial overload of saturated FA triggers the loss of redox homeostasis and accelerates the formation of oxygen radicals (i.e. ROS). This lipotoxic condition called oxidative stress has been implicated in the development of insulin resistance as well as non-alcoholic FLD in non-ruminants (Videla et al., Reference Videla, Rodrigo, Araya and Poniachik2006). As reviewed by Videla et al. (Reference Videla, Rodrigo, Araya and Poniachik2006), persistent ROS production activates serine/threonine kinase signaling cascades that inhibit insulin-stimulated insulin receptor substrate induction. In turn, insulin resistance may enhance intrahepatic lipid content to promote simple steatosis (i.e. non-inflammatory phenotype). However, oxidative stress may also promote Kupffer cell activation to activate redox-sensitive transcription factors, including nuclear factor-κB, and upregulate pro-inflammatory TNF-α. Unfortunately, inflammatory steatohepatitis is also characterized by the upregulation of NADPH oxidase and cytochrome P450 (family 2, subfamily E, polypeptide 1; otherwise known as CYP2E1), which further reduce antioxidant capacity and promote hepatocellular damage. Another consequence of rampant ROS generation includes n-3 long-chain polyunsaturated FA depletion caused by defective FA desaturation and enhanced peroxidation in the liver (Videla et al., Reference Videla, Rodrigo, Araya and Poniachik2004). In addition, polyunsaturated FA subjected to enzymatic (via cyclooxygenase, lipoxygenase and CYP2E1) or non-enzymatic (via ROS) oxidation form oxylipids with diverse inflammatory functions. For instance, oxylipids derived from n-6 arachidonic acid and linoleic acid include pro-inflammatory hydroxyl-octadecadienoic acid and hydroxyl-eicosatetraenoic acid, respectively.

In the transition cow experiencing reduced antioxidant potential (Sordillo and Aitken, Reference Sordillo and Aitken2009), ROS accumulation likely develops in part because of rampant mitochondrial FA oxidation, albeit incomplete breakdown. In turn, oxidative stress may enhance adipose tissue lipolysis to exacerbate oxidant status (Krawczyk et al., Reference Krawczyk, Haller, Ferrante, Zoeller and Corkey2012). One fate of lipolytic-derived unsaturated FA is ROS oxidation, which results in lipid hydroperoxide and isoprostane formation. Fatty acids released by adipose tissue are also used for hepatic β-hydroxybutyrate generation, which may promote hepatocyte apoptosis via ROS-mediated p38 mitogen-activated protein kinase activation (Song et al., Reference Song, Li, Gu, Fu, Peng, Zhao, Zhang, Li, Wang, Li and Liu2016). In addition, lipolytic FA may induce nuclear factor-κB in hepatocytes via ROS-dependent mechanisms that trigger inflammation (Li et al., Reference Li, Huang, Gu, Du, Lei, Yuan, Sun, Wang, Li and Liu2015). These findings suggest that oxidative stress is involved in FLD pathology.

As previously reviewed by Sordillo and Aitken (Reference Sordillo and Aitken2009), the accumulation of ROS or oxylipids likely influences bovine immune responses. For instance, plasma oxylipid levels are correlated with the expression of interleukin-12β and inducible nitric oxide synthase-2 in peripheral blood mononuclear cells derived from healthy transition cows (Raphael et al., Reference Raphael, Halbert, Contreras and Sordillo2014). In endothelial cells, elevations in the arachidonic acid metabolite 15-hydroxy-peroxyeicosatetraenoic acid develops with apoptosis and caspase-3 activation (Sordillo et al., Reference Sordillo, Weaver, Cao, Corl, Sylte and Mullarky2005), and leukocyte recruitment and inflammatory cytokine release (Whatling et al., Reference Whatling, McPheat and Herslof2007). Cytochrome P450- and lipoxygenase-derived oxylipids accumulate in plasma (e.g. 11,12-epoxyoctadecanoic acid) and adipose tissue (e.g. 5-hydroxy-eicosatetraenoic acid) of postpartum cows, respectively, and may act as positive or negative modulators of inflammation and immune cell trafficking during intense lipolysis (Contreras et al., Reference Contreras, Strieder-Barboza, De Souza, Gandy, Mavangira, Lock and Sordillo2017). Inflammatory dysfunction is a key attribute of inflammatory disorders including mastitis and metritis; therefore, the role of oxidized lipids has been considered. For instance, Streptococcus uberis mastitis is characterized by mammary inflammation and the accrual of oxylipids, including hydroxyl-octadecadienoic acid (Ryman et al., Reference Ryman, Pighetti, Lippolis, Gandy, Applegate and Sordillo2015). The amount of dietary antioxidants (i.e. vitamin E and β-carotene) and trace minerals (i.e. copper or zinc) that play a role in antioxidant defense mechanisms, and the type and amount of FA fed to transition cows, may influence oxylipid formation and affiliated health outcomes.

Leptin, adiponectin and fibroblast growth factor-21

Although the role of insulin within the framework of glucose utilization and lipolysis is described, adipokines and hepatokines deserve consideration to understand lipid metabolism in the transition cow. First, the adipokine leptin is produced by adipocytes and mediates its action through the Janus kinase signal transducer and activator of transcription pathway (Yadav et al., Reference Yadav, Kataria, Saini and Yadav2013). The hormone regulates energy metabolism, insulin sensitivity, thyroid hormone secretion, immunity and appetite. In a demonstration of function, insulin resistance and inflammatory non-alcoholic FLD develops in obese leptin-deficient ob/ob mice (Perfield et al., Reference Perfield, Ortinau, Pickering, Ruebel, Meers and Rector2013). Moreover, ob/ob mice experience lipotoxicity including muscle ceramide and DAG accrual (Turpin et al., Reference Turpin, Ryall, Southgate, Darby, Hevener, Febbraio, Kemp, Lynch and Watt2009); however, chronic treatment of ob/ob mice with myriocin decreases circulating ceramides, steatosis and improves glucose homeostasis (Yang et al., Reference Yang, Badeanlou, Bielawski, Roberts, Hannun and Samad2009). In hyperlipidemic animals, leptin acts by preventing the accumulation of ceramide and DAG to protect against insulin resistance (Dube et al., Reference Dube, Bhatt, Dedousis, Bonen and O’Doherty2007). Such results may be explained by a stimulatory effect of leptin on FA breakdown (Shimabukuro et al., Reference Shimabukuro, Koyama, Chen, Wang, Trieu, Lee, Newgard and Unger1997). However, the lipid mediator-lowering effect of leptin is not always observed. In hyperleptinemic lean animals, for example, soleus and superficial vastus muscle concentrations of ceramide are increased, DAG unchanged, and triglyceride decreased (Dube et al., Reference Dube, Bhatt, Dedousis, Bonen and O’Doherty2007). These findings suggest that mechanisms by which leptin modulate insulin action likely vary by adiposity phenotype.

In the late-gestating cow, fatter cows exhibit higher plasma leptin concentrations, relative to lean cows (Kokkonen et al., Reference Kokkonen, Taponen, Anttila, Syrjälä-Qvist, Delavaud, Chilliard, Tuori and Tesfa2005), whereas all cows experience a decline in circulating leptin and white adipose tissue leptin mRNA expression at the onset of lactation (Block et al., Reference Block, Butler, Ehrhardt, Bell, Van Amburgh and Boisclair2001). With this physiology in mind, it can be argued that the observed elevations in muscle FA oxidation occur independent of leptin signaling (Schäff et al., Reference Schäff, Börner, Hacke, Kautzsch, Sauerwein, Spachmann, Schweigel-Röntgen, Hammon and Kuhla2013). In ruminants, leptin may only be responsive during positive energy balance. In support, an intravenous infusion of a triglyceride emulsion increases circulating FA and leptin concentrations during late gestation but not early lactation (Chelikani et al., Reference Chelikani, Keisler and Kennelly2003). The authors suggest that this effect may be due to elevations in insulin-like growth factor-1, which is known to regulate leptin secretion. Although current evidence suggests a limited role of leptin in postpartum dairy cattle, the administration of human leptin to early-lactation cows decreases liver triglyceride content (Ehrhardt et al., Reference Ehrhardt, Foskolos, Giesy, Wesolowski, Krumm, Butler, Quirk, Waldron and Boisclair2016); therefore, strategies that elevate leptin status during the periparturient period may be therapeutic.

Adiponectin is predominantly expressed in adipose tissue, and the expression of adiponectin decreases with increasing adiposity (Yadav et al., Reference Yadav, Kataria, Saini and Yadav2013). This adipokine increases insulin sensitivity, reduces circulating glucose, enhances FA catabolism and prevents inflammation (Yadav et al., Reference Yadav, Kataria, Saini and Yadav2013). The mechanisms of adiponectin action involve the activation of peroxisome proliferator activator receptor-α and AMP-activated protein kinase. In the muscle, downstream outcomes include enhanced FA transport and oxidation, reduced intracellular triglyceride content and enhanced insulin-stimulated translocation of glucose transporters to the plasma membrane.

In multiparous dairy cow transitioning to lactation, circulating adiponectin levels are lowest at parturition (Giesy et al., Reference Giesy, Yoon, Currie, Kim and Boisclair2012). Suppressed adiponectin secretion in the multiparous transition cow could limit FA oxidation and glucose utilization in response to insulin. In support, enhanced plasma adiponectin levels are associated with reduced plasma concentrations of long-chain acylcarnitines (Rodríguez-Gutiérrez et al., Reference Rodríguez-Gutiérrez, Lavalle-González, Martínez-Garza, Landeros-Olvera, López-Alvarenga, Torres-Sepúlveda, González-González, Mancillas-Adame, Salazar-Gonzalez and Villarreal-Pérez2012), and adiponectin decreases ceramide accumulation in obese mice (Holland et al., Reference Holland, Adams, Brozinick, Bui, Miyauchi, Kusminski, Bauer, Wade, Singhal, Cheng, Volk, Kuo, Gordillo, Kharitonenkov and Scherer2013). It deserves to be emphasized that serum adiponectin concentrations appear to increase after parturition in primiparous cows (Urh et al., Reference Urh, Denißen, Harder, Koch, Gerster, Ettle, Kraus, Schmitz, Kuhla, Stamer, Spiekers and Sauerwein2019). It has been hypothesized that greater adiponectin concentrations in primiparous cows may enhance peripheral insulin sensitivity and nutrient partitioning toward growth (Koster et al., Reference De Koster, Urh, Hostens, Van den Broeck, Sauerwein and Opsomer2017; Urh et al., Reference Urh, Denißen, Harder, Koch, Gerster, Ettle, Kraus, Schmitz, Kuhla, Stamer, Spiekers and Sauerwein2019). If this were observed, it can be further hypothesized that circulating acylcarnitines and ceramides would be lower in primiparous cows, relative to multiparous cows.

Studies have also focused on the expression of adiponectin receptor-1 and -2 in bovine tissues during the transition period. Giesy et al. (Reference Giesy, Yoon, Currie, Kim and Boisclair2012) observed elevated mRNA expression of adiponectin receptor-1 and -2 during early lactation in the muscle and liver, respectively, relative to late pregnancy. Similar changes in hepatic adiponectin receptor-2 expression have been confirmed (Saremi et al., Reference Saremi, Winand, Friedrichs, Kinoshita, Rehage, Dänicke, Häussler, Breves, Mielenz and Sauerwein2014). In adipose tissue, Giesy et al. (Reference Giesy, Yoon, Currie, Kim and Boisclair2012) did not observe a change in adiponectin receptor expression, which contrasts with other work demonstrating reduced adiponectin receptor-1 and -2 expression in adipose tissue in early lactation, relative to late gestation (Lemor et al., Reference Lemor, Hosseini, Sauerwein and Mielenz2009; Saremi et al., Reference Saremi, Winand, Friedrichs, Kinoshita, Rehage, Dänicke, Häussler, Breves, Mielenz and Sauerwein2014). Future work should define how changes in adiponectin receptor mRNA expression influence adiponectin signaling.

Fibroblast growth factor-21 (FGF21) lowers blood glucose, insulin and triglyceride concentrations, and reduces body weight in non-ruminants (Holland et al., Reference Holland, Adams, Brozinick, Bui, Miyauchi, Kusminski, Bauer, Wade, Singhal, Cheng, Volk, Kuo, Gordillo, Kharitonenkov and Scherer2013). Interestingly, FGF21 stimulates adiponectin secretion, and adiponectin mediates FGF21-induced increases in energy expenditure (Holland et al., Reference Holland, Adams, Brozinick, Bui, Miyauchi, Kusminski, Bauer, Wade, Singhal, Cheng, Volk, Kuo, Gordillo, Kharitonenkov and Scherer2013). In peripartal cow, plasma FGF21 concentrations peak at parturition. Moreover, plasma FGF21 levels and hepatic FGF21 mRNA are increased by feed restriction in non-pregnant, late-lactating dairy cows (Schoenberg et al., Reference Schoenberg, Waldron, Giesy, Boisclair, Harvatine, Kharitonenkov and Cheng2011); however, elevations in circulating FGF21 does not appear to prevent FA-induced increases in liver triglyceride in non-pregnant, non-lactating dairy cows (Caixeta et al., Reference Caixeta, Giesy, Krumm, Perfield, Butterfield, Schoenberg, Beitz and Boisclair2017). Because plasma FGF21 and ceramide concentrations are elevated and adiponectin secretion is suppressed in transition cows, it would appear that the FGF21–adiponectin–ceramide axis observed in non-ruminants (Holland et al., Reference Holland, Adams, Brozinick, Bui, Miyauchi, Kusminski, Bauer, Wade, Singhal, Cheng, Volk, Kuo, Gordillo, Kharitonenkov and Scherer2013) is not intact in peripartal ruminants but may explain metabolic dysfunction (i.e. FLD and impaired insulin antagonism).

Conclusion

The periparturient cow relies on well-described biochemical and hormonal changes in nutrient metabolism to meet metabolic demands; however, the advent of lipidomics technologies has contemporized our understanding of lipid biology and lipotoxicity during the peripartum period. Today, FA are recognized as bioactive signaling molecules that uniquely influence nutrient utilization and production. The same certainly applies to many of tens of thousands of complex lipids that constitute the bovine lipidome. Therefore, we should be careful not to generalize acylcarnitines, ceramides, diacylglycerols, phosphatidylcholines and oxylipids. Rather, the mechanistic role of these lipids at the species level should be elucidated in the transition cow. Then, if deemed appropriate, they should be targeted to advantageously control nutrient partitioning for therapeutic gain. Such insight has potential to result in the development of optimized dairy cattle nutrition strategies designed to modulate cow metabolism and productive lifespan.

Acknowledgements

The author thanks the organizers of the International Symposium on Ruminant Physiology 2019 for inviting to write this review.

J. W. McFadden 0000-0002-6655-700X

Declaration of interest

The authors declare that there is no conflict of interest for this manuscript.

Ethics statement

None.

Software and data repository resources

None of the data were deposited in an official repository.

References

Aerts, JM, Ottenhoff, R, Powlson, AS, Grefhorst, A, van Eijk, M, Dubbelhuis, PF, Aten, J, Kuipers, F, Serlie, MJ and Wennekes, T 2007. Pharmacological inhibition of glucosylceramide synthase enhances insulin sensitivity. Diabetes 56, 13411349.CrossRefGoogle ScholarPubMed
Aguer, C, McCoin, CS, Knotts, TA, Thrush, AB, Ono-Moore, K, McPherson, R, Dent, R, Hwang, DH, Adams, SH and Harper, ME 2015. Acylcarnitines: potential implications for skeletal muscle insulin resistance. FASEB Journal 29, 336345.CrossRefGoogle ScholarPubMed
Artegoitia, VM, Middleton, JL, Harte, FM, Campagna, SR and de Veth, MJ 2014. Choline and choline metabolite patterns and associations in blood and milk during lactation in dairy cows. PLoS ONE 9, e103412.CrossRefGoogle ScholarPubMed
Bauman, DE and Currie, WB 1980. Partitioning of nutrients during pregnancy and lactation: a review of mechanisms involving homeostasis and homeorhesis. Journal of Dairy Science 63, 15141529.CrossRefGoogle ScholarPubMed
Baumgard, L, Collier, R and Bauman, D 2017. A 100-year review: regulation of nutrient partitioning to support lactation. Journal of Dairy Science 100, 1035310366.CrossRefGoogle ScholarPubMed
Bernabucci, U, Basiricò, L, Pirazzi, D, Rueca, F, Lacetera, N, Lepri, E and Nardone, A 2009. Liver apolipoprotein B 100 expression and secretion are down-regulated early postpartum in dairy cows. Livestock Science 125, 169176.CrossRefGoogle Scholar
Block, S, Butler, W, Ehrhardt, R, Bell, A, Van Amburgh, M and Boisclair, Y 2001. Decreased concentration of plasma leptin in periparturient dairy cows is caused by negative energy balance. Journal of Endocrinology 171, 339348.CrossRefGoogle ScholarPubMed
Boden, G 2011. Obesity, insulin resistance and free fatty acids. Current Opinion in Endocrinology, Diabetes, and Obesity 18, 139143.CrossRefGoogle ScholarPubMed
Boon, J, Hoy, AJ, Stark, R, Brown, RD, Meex, RC, Henstridge, DC, Schenk, S, Meikle, PJ, Horowitz, JF, Kingwell, BA, Bruce, CR and Watt, MJ 2013. Ceramides contained in LDL are elevated in type 2 diabetes and promote inflammation and skeletal muscle insulin resistance. Diabetes 62, 401410.CrossRefGoogle ScholarPubMed
Caixeta, LS, Giesy, SL, Krumm, CS, Perfield, JW, Butterfield, A, Schoenberg, KM, Beitz, DC and Boisclair, YR 2017. Effect of circulating glucagon and free fatty acids on hepatic FGF21 production in dairy cows. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology 313, R526R534.CrossRefGoogle ScholarPubMed
Carlson, DB, McFadden, JW, D’Angelo, A, Woodworth, JC and Drackley, JK 2007. Dietary l-carnitine affects periparturient nutrient metabolism and lactation in multiparous cows. Journal of Dairy Science 90, 34223441.CrossRefGoogle ScholarPubMed
Cartiff, SE, Fellner, V and Eisemann, JH 2013. Eicosapentaenoic and docosahexaenoic acids increase insulin sensitivity in growing steers. Journal of Animal Science 91, 23322342.CrossRefGoogle ScholarPubMed
Chavez, JA, Holland, WL, Bär, J, Sandhoff, K and Summers, SA 2005. Acid ceramidase overexpression prevents the inhibitory effects of saturated fatty acids on insulin signaling. Journal of Biological Chemistry 280, 2014820153.CrossRefGoogle ScholarPubMed
Chavez, JA and Summers, SA 2012. A ceramide-centric view of insulin resistance. Cell Metabolism 15, 585594.CrossRefGoogle ScholarPubMed
Chelikani, PK, Keisler, DH and Kennelly, JJ 2003. Response of plasma leptin concentration to jugular infusion of glucose or lipid is dependent on the stage of lactation of Holstein cows. Journal of Nutrition 133, 41634171.CrossRefGoogle ScholarPubMed
Clemmons, DR 2004. Role of insulin-like growth factor in maintaining normal glucose homeostasis. Hormone Research 62, 7782.CrossRefGoogle ScholarPubMed
Clempson, A, Pollott, G, Brickell, J, Bourne, N, Munce, N and Wathes, D 2011. Polymorphisms in the autosomal genes for mitochondrial function TFAM and UCP2 are associated with performance and longevity in dairy cows. Animal 5, 13351343.CrossRefGoogle ScholarPubMed
Connor, EE, Kahl, S, Elsasser, TH, Parker, JS, Li, RW, Van Tassell, CP, Baldwin, RL and Barao, SM 2010. Enhanced mitochondrial complex gene function and reduced liver size may mediate improved feed efficiency of beef cattle during compensatory growth. Functional and Integrative Genomics 10, 3951.CrossRefGoogle ScholarPubMed
Contreras, GA, Strieder-Barboza, C, De Souza, J, Gandy, J, Mavangira, V, Lock, AL and Sordillo, LM 2017. Periparturient lipolysis and oxylipid biosynthesis in bovine adipose tissues. PloS ONE 12, e0188621.CrossRefGoogle ScholarPubMed
da Costa, K-A, Sanders, LM, Fischer, LM and Zeisel, SH 2011. Docosahexaenoic acid in plasma phosphatidylcholine may be a potential marker for in vivo phosphatidylethanolamine N-methyltransferase activity in humans. American Journal of Clinical Nutrition 93, 968974.CrossRefGoogle ScholarPubMed
Davis, AN, Clegg, JL, Perry, CA and McFadden, JW 2017. Nutrient restriction increases circulating and hepatic ceramide in dairy cows displaying impaired insulin tolerance. Lipids 52, 110.CrossRefGoogle ScholarPubMed
Davis, AN, Rico, JE, Myers, WA, Coleman, ME, Clapham, ME, Haughey, NJ and McFadden, JW 2019. Circulating low-density lipoprotein ceramide concentrations increase in Holstein dairy cows transitioning from gestation to lactation. Journal of Dairy Science 102, 56345646.CrossRefGoogle ScholarPubMed
De Koster, J, Urh, C, Hostens, M, Van den Broeck, W, Sauerwein, H and Opsomer, G 2017. Relationship between serum adiponectin concentration, body condition score, and peripheral tissue insulin response of dairy cows during the dry period. Domestic Animal Endocrinology 59, 100104.CrossRefGoogle ScholarPubMed
DeLong, CJ, Shen, YJ, Thomas, MJ and Cui, Z 1999. Molecular distinction of phosphatidylcholine synthesis between the CDP-choline pathway and phosphatidylethanolamine methylation pathway. Journal of Biological Chemistry 274, 2968329688.CrossRefGoogle ScholarPubMed
de Souza, J, Preseault, CL and Lock, AL 2018. Altering the ratio of dietary palmitic, stearic, and oleic acids in diets with or without whole cottonseed affects nutrient digestibility, energy partitioning, and production responses of dairy cows. Journal of Dairy Science 101, 172185.CrossRefGoogle ScholarPubMed
Douglas, GN, Rehage, J, Beaulieu, AD, Bahaa, AO and Drackley, JK 2007. Prepartum nutrition alters fatty acid composition in plasma, adipose tissue, and liver lipids of periparturient dairy cows. Journal of Dairy Science 90, 29412959.CrossRefGoogle ScholarPubMed
Dube, JJ, Bhatt, BA, Dedousis, N, Bonen, A and O’Doherty, RM 2007. Leptin, skeletal muscle lipids and lipid-induced insulin resistance. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology 293, R642650.CrossRefGoogle ScholarPubMed
Ehrhardt, RA, Foskolos, A, Giesy, SL, Wesolowski, SR, Krumm, CS, Butler, WR, Quirk, SM, Waldron, MR and Boisclair, YR 2016. Increased plasma leptin attenuates adaptive metabolism in early lactating dairy cows. Journal of Endocrinology 229, 145157.CrossRefGoogle ScholarPubMed
Erion, DM and Shulman, GI 2010. Diacylglycerol-mediated insulin resistance. Nature Medicine 16, 400402.CrossRefGoogle ScholarPubMed
Fabbrini, E, Mohammed, BS, Magkos, F, Korenblat, KM, Patterson, BW and Klein, S 2008. Alterations in adipose tissue and hepatic lipid kinetics in obese men and women with nonalcoholic fatty liver disease. Gastroenterology 134, 424431.CrossRefGoogle ScholarPubMed
Fast, DG and Vance, DE 1995. Nascent VLDL phospholipid composition is altered when phosphatidylcholine biosynthesis is inhibited: evidence for a novel mechanism that regulates VLDL secretion. Biochimica et Biophysica Acta (BBA)-Lipids and Lipid Metabolism 1258, 159168.CrossRefGoogle ScholarPubMed
Giesy, SL, Yoon, B, Currie, WB, Kim, JW and Boisclair, YR 2012. Adiponectin deficit during the precarious glucose economy of early lactation in dairy cows. Endocrinology 153, 58345844.CrossRefGoogle ScholarPubMed
Hailemariam, D, Mandal, R, Saleem, F, Dunn, S, Wishart, D and Ametaj, B 2014. Identification of predictive biomarkers of disease state in transition dairy cows. Journal of Dairy Science 97, 26802693.CrossRefGoogle Scholar
Harvatine, KJ and Allen, MS 2006. Effects of fatty acid supplements on milk yield and energy balance of lactating dairy cows. Journal of Dairy Science 89, 10811091.CrossRefGoogle ScholarPubMed
Holland, WL, Adams, AC, Brozinick, JT, Bui, HH, Miyauchi, Y, Kusminski, CM, Bauer, SM, Wade, M, Singhal, E, Cheng, CC, Volk, K, Kuo, M-S, Gordillo, R, Kharitonenkov, A and Scherer, PE 2013. An FGF21-adiponectin-ceramide axis controls energy expenditure and insulin action in mice. Cell Metabolism 17, 790797.CrossRefGoogle ScholarPubMed
Holland, WL, Brozinick, JT, Wang, L-P, Hawkins, ED, Sargent, KM, Liu, Y, Narra, K, Hoehn, KL, Knotts, TA, Siesky, A, Nelson, DH, Karathanasis, SK, Fontenot, GK, Birnbaum, MJ and Summers, SA 2007. Inhibition of ceramide synthesis ameliorates glucocorticoid-, saturated-fat-, and obesity-induced insulin resistance. Cell Metabolism 5, 167179.CrossRefGoogle ScholarPubMed
Huber, K, Dänicke, S, Rehage, J, Sauerwein, H, Otto, W, Rolle-Kampczyk, U and Von Bergen, M 2016. Metabotypes with properly functioning mitochondria and anti-inflammation predict extended productive life span in dairy cows. Scientific Reports 6, 24642.CrossRefGoogle ScholarPubMed
Imhasly, S, Naegeli, H, Baumann, S, von Bergen, M, Luch, A, Jungnickel, H, Potratz, S and Gerspach, C 2014. Metabolomic biomarkers correlating with hepatic lipidosis in dairy cows. BMC Veterinary Research 10, 122.CrossRefGoogle ScholarPubMed
Kohjima, M, Higuchi, N, Kato, M, Kotoh, K, Yoshimoto, T, Fujino, T, Yada, M, Yada, R, Harada, N and Enjoji, M 2008. SREBP-1c, regulated by the insulin and AMPK signaling pathways, plays a role in nonalcoholic fatty liver disease. International Journal of Molecular Medicine 21, 507511.Google Scholar
Kokkonen, T, Taponen, J, Anttila, T, Syrjälä-Qvist, L, Delavaud, C, Chilliard, Y, Tuori, M and Tesfa, AT 2005. Effect of body fatness and glucogenic supplement on lipid and protein mobilization and plasma leptin in dairy cows. Journal of Dairy Science 88, 11271141.CrossRefGoogle ScholarPubMed
Koves, TR, Ussher, JR, Noland, RC, Slentz, D, Mosedale, M, Ilkayeva, O, Bain, J, Stevens, R, Dyck, JR and Newgard, CB 2008. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metabolism 7, 4556.CrossRefGoogle ScholarPubMed
Krawczyk, SA, Haller, JF, Ferrante, T, Zoeller, RA and Corkey, BE 2012. Reactive oxygen species facilitate translocation of hormone sensitive lipase to the lipid droplet during lipolysis in human differentiated adipocytes. PloS ONE 7, e34904.CrossRefGoogle ScholarPubMed
Lake, AD, Novak, P, Shipkova, P, Aranibar, N, Robertson, DG, Reily, MD, Lehman-McKeeman, LD, Vaillancourt, RR and Cherrington, NJ 2015. Branched chain amino acid metabolism profiles in progressive human nonalcoholic fatty liver disease. Amino Acids 47, 603615.CrossRefGoogle ScholarPubMed
Lemor, A, Hosseini, A, Sauerwein, H and Mielenz, M 2009. Transtiion period-related changes in the abundance of the mRNAs of adiponectin and its receptors, of visfatin, and of fatty acid binding receptors in adipose tissue of high-yielding dairy cows. Domestic Animal Endocrinology 37, 3744.CrossRefGoogle ScholarPubMed
Lepretti, M, Martucciello, S, Burgos Aceves, M, Putti, R and Lionetti, L 2018. Omega-3 fatty acids and insulin resistance: focus on the regulation of mitochondria and endoplasmic reticulum stress. Nutrients 10, 350.CrossRefGoogle ScholarPubMed
Levy, M and Futerman, AH 2010. Mammalian ceramide synthases. IUBMB Life 62, 347356.Google ScholarPubMed
Li, X, Huang, W, Gu, J, Du, X, Lei, L, Yuan, X, Sun, G, Wang, Z, Li, X and Liu, G 2015. SREBP-1c overactivates ROS-mediated hepatic NF-κB inflammatory pathway in dairy cows with fatty liver. Cellular Signalling 27, 20992109.CrossRefGoogle ScholarPubMed
Litherland, N, Dann, H and Drackley, J 2011. Prepartum nutrient intake alters palmitate metabolism by liver slices from peripartal dairy cows. Journal of Dairy Science 94, 19281940.CrossRefGoogle ScholarPubMed
Liu, P and Anderson, RG 1995. Compartmentalized production of ceramide at the cell surface. Journal of Biological Chemistry 270, 2717927185.CrossRefGoogle ScholarPubMed
Lomax, MA, Baird, GD, Mallinson, CB and Symonds, H 1979. Differences between lactating and non-lactating dairy cows in concentration and secretion rate of insulin. Biochemical Journal 180, 281289.CrossRefGoogle ScholarPubMed
Lucy, MC, Jiang, H and Kobayashi, Y 2001. Changes in the somatotophic axis associated with the initiation of lactation. Journal of Dairy Science 84, E113E119.CrossRefGoogle Scholar
Lucy, MC, Verkerk, GA, Whyte, BE, Macdonald, KA, Burton, L, Cursons, RT, Roche, JR and Holmes, CW 2009. Somatotropic axis components and nutrient partitioning in genetically diverse dairy cows managed under different feed allowances in a pasture system. Journal of Dairy Science 92, 526539.CrossRefGoogle Scholar
Maedler, K, Oberholzer, J, Bucher, P, Spinas, GA and Donath, MY 2003. Monounsaturated fatty acids prevent the deleterious effects of palmitate and high glucose on human pancreatic β-cell turnover and function. Diabetes 52, 726733.CrossRefGoogle ScholarPubMed
Marcellini, M, Nobili, V, Manco, M and Giannone, G 2007. Correlation of Serum TNF-α Levels and histologic liver injury scores in pediatric nonalcoholic fatty liver disease. American Journal of Clinical Pathology 127, 954960.Google Scholar
McFadden, JW and Rico, JE 2019. Invited review: sphingolipid biology in the dairy cow: the emerging role of ceramide. Journal of Dairy Science 102, 76197639.CrossRefGoogle ScholarPubMed
McFadden, JW, Rico, JE, Erb, SJ and White, HM 2018. Inhibition of serine palmitoyltransferase prevents palmitic acid-induced ceramide synthesis in bovine primary hepatocytes. Journal of Dairy Science 101 (E-suppl. 2), 105.Google Scholar
Mulligan, FJ and Doherty, ML 2008. Production diseases of the transition cow. The Veterinary Journal 176, 39.CrossRefGoogle ScholarPubMed
Myers, WA, Rico, JE, Davis, AN, Fontoura, ABP, Dineen, MJ, Tate, BN and McFadden, JW 2019. Effects of abomasal infusions of fatty acids and one-carbon donors on hepatic ceramide and phosphatidylcholine levels in lactating Holstein dairy cows. Journal of Dairy Science 102, 70877101.CrossRefGoogle Scholar
Newgard, CB, An, J, Bain, JR, Muehlbauer, MJ, Stevens, RD, Lien, LF, Haqq, AM, Shah, SH, Arlotto, M, Slentz, CA, Rochon, J, Gallup, D, Ilkayeva, O, Wenner, BR, Yancy, WS, Eisenson, H, Musante, G, Surwit, RS, Millington, DS, Butler, MD and Svetkey, LP 2009. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metabolism 9, 311326.CrossRefGoogle ScholarPubMed
Ohtsuka, H, Koiwa, M, Hatsugaya, A, Kudo, K, Hoshi, F, Itoh, N, Yokota, H, Okada, H and Kawamura, S-I 2001. Relationship between serum TNF activity and insulin resistance in dairy cows affected with naturally occurring fatty liver. Journal of Veterinary Medical Science 63, 10211025.CrossRefGoogle ScholarPubMed
Palomer, X, Pizarro-Delgado, J, Barroso, E and Vázquez-Carrera, M 2018. Palmitic and oleic acid: the yin and yang of fatty acids in type 2 diabetes mellitus. Trends in Endocrinology and Metabolism 29, 178190.CrossRefGoogle ScholarPubMed
Peraldi, P, Hotamisligil, GS, Buurman, WA, White, MF and Spiegelman, BM 1996. Tumor necrosis factor (TNF)-alpha inhibits insulin signaling through stimulation of the p55 TNF receptor and activation of sphingomyelinase. Journal of Biological Chemistry 271, 1301813022.CrossRefGoogle ScholarPubMed
Perfield, JW, Ortinau, LC, Pickering, RT, Ruebel, ML, Meers, GM and Rector, RS 2013. Altered hepatic lipid metabolism contributes to nonalcoholic fatty liver disease in leptin-deficient Ob/Ob mice. Journal of Obesity 2013, 296537.CrossRefGoogle ScholarPubMed
Petersen, MC and Jurczak, MJ 2016. CrossTalk opposing view: intramyocellular ceramide accumulation does not modulate insulin resistance. Journal of Physiology 594, 31713174.CrossRefGoogle Scholar
Pires, JAA, Pescara, JB, Brickner, AE, Silva del Rio, N, Cunha, AP and Grummer, RR 2008. Effects of abomasal infusion of linseed oil on responses to glucose and insulin in holstein cows. Journal of Dairy Science 91, 13781390.CrossRefGoogle ScholarPubMed
Pires, JAA, Souza, AH and Grummer, RR 2007. Induction of hyperlipidemia by intravenous infusion of tallow emulsion causes insulin resistance in Holstein cows. Journal of Dairy Science 90, 27352744.CrossRefGoogle ScholarPubMed
Poitout, V, Hagman, D, Artner, I, Stein, R, Harmon, JS and Robertson, RP 2006. Regulation of the insulin gene by glucose and fatty acids. Journal of Nutrition 136, 873876.CrossRefGoogle ScholarPubMed
Pullen, D, Liesman, J and Emery, R 1990. A species comparison of liver slice synthesis and secretion of triacylglycerol from nonesterified fatty acids in media. Journal of Animal Science 68, 13951399.CrossRefGoogle ScholarPubMed
Pushkareva, M, Obeid, LM and Hannun, YA 1995. Ceramide: an endogenous regulator of apoptosis and growth suppresion. Immunology Today 16, 294297.CrossRefGoogle Scholar
Qin, N, Kokkonen, T, Salin, S, Seppänen-Laakso, T, Taponen, J, Vanhatalo, A and Elo, K 2017. Prepartal overfeeding alters the lipidomic profiles in the liver and the adipose tissue of transition dairy cows. Metabolomics 13, 21.CrossRefGoogle Scholar
Raphael, W, Halbert, L, Contreras, GA and Sordillo, LM 2014. Association between polyunsaturated fatty acid-derived oxylipid biosynthesis and leukocyte inflammatory marker expression in periparturient dairy cows. Journal of Dairy Science 97, 36153625.CrossRefGoogle ScholarPubMed
Rico, JE, Bandaru, VVR, Dorskind, JM, Haughey, NJ and McFadden, JW 2015. Plasma ceramides are elevated in overweight Holstein dairy cows experiencing greater lipolysis and insulin resistance during the transition from late pregnancy to early lactation. Journal of Dairy Science 98, 77577770.CrossRefGoogle ScholarPubMed
Rico, JE, Giesy, SL, Haughey, NJ, Boisclair, YR and McFadden, JW 2018a. Intravenous triacylglycerol infusion promotes ceramide accumulation and hepatic steatosis in dairy cows. Journal of Nutrition 148, 15291535.CrossRefGoogle ScholarPubMed
Rico, JE, Mathews, AT, Lovett, J, Haughey, NJ and McFadden, JW 2016. Palmitic acid feeding increases ceramide supply in association with increased milk yield, circulating nonesterified fatty acids, and adipose tissue responsiveness to a glucose challenge. Journal of Dairy Science 99, 88178830.CrossRefGoogle ScholarPubMed
Rico, JE, Myers, WA, Laub, DJ, Davis, AN, Zeng, Q and McFadden, JW 2018c. Hot topic: Ceramide inhibits insulin sensitivity in primary bovine adipocytes. Journal of Dairy Science 101, 34283432.CrossRefGoogle ScholarPubMed
Rico, JE, Rico, DE, Phipps, ZC, Zeng, Q, Corl, BA, Chouinard, PY, Gervais, R and McFadden, JW 2017b. Circulating ceramide concentrations are influenced by saturated fatty acid chain length in mid-lactation dairy cows. Journal of Dairy Science 100 (E-Suppl. 2), 394.Google Scholar
Rico, JE, Saed Samii, S, Mathews, AT, Lovett, J, Haughey, NJ and McFadden, JW 2017a. Temporal changes in sphingolipids and systemic insulin sensitivity during the transition from gestation to lactation. PloS ONE 12, e0176787.CrossRefGoogle ScholarPubMed
Rico, JE, Zang, Y, Haughey, N, Rius, A and McFadden, JW 2018b. Short communication: circulating fatty acylcarnitines are elevated in overweight periparturient dairy cows in association with sphingolipid biomarkers of insulin resistance. Journal of Dairy Science 101, 812819.CrossRefGoogle ScholarPubMed
Roche, JR, Bell, AW, Overton, TR, and Loor, JJ 2013. Nutritional management of the transition cow in the 21st century - a paradigm shift in thinking. Animal Production Science 53, 10001023.CrossRefGoogle Scholar
Rodríguez-Gutiérrez, R, Lavalle-González, FJ, Martínez-Garza, LE, Landeros-Olvera, E, López-Alvarenga, JC, Torres-Sepúlveda, MR, González-González, JG, Mancillas-Adame, LG, Salazar-Gonzalez, B and Villarreal-Pérez, JZ 2012. Impact of an exercise program on acylcarnitines in obesity: a prospective controlled study. Journal of the International Society of Sports Nutrition 9, 22.CrossRefGoogle ScholarPubMed
Rukkwamsuk, T, Geelen, MJH, Kruip, TAM and Wensing, T 2000. Interrelation of fatty acid composition in adipose tissue, serum, and liver of dairy cows during the development of fatty liver postpartum. Journal of Dairy Science 83, 5259.CrossRefGoogle ScholarPubMed
Rutkowsky, JM, Knotts, TA, Ono-Moore, KD, McCoin, CS, Huang, S, Schneider, D, Singh, S, Adams, SH and Hwang, DH 2014. Acylcarnitines activate pro-inflammatory signaling pathways. American Journal of Physiology-Heart and Circulatory Physiology 306, E1378E1387.Google Scholar
Ryman, VE, Pighetti, GM, Lippolis, JD, Gandy, JC, Applegate, CM and Sordillo, LM 2015. Quantification of bovine oxylipids during intramammary Streptococcus uberis infection. Prostaglandins and Other Lipid Mediators 121, 207217.CrossRefGoogle ScholarPubMed
Saed Samii, S, Zang, Y, Myers, WA, Grilli, E and McFadden, JW 2018. A lipidomic analysis of bovine liver during metabolic disease. Journal of Dairy Science 101 (Suppl. 2), 104.Google Scholar
Saremi, B, Winand, S, Friedrichs, P, Kinoshita, A, Rehage, J, Dänicke, S, Häussler, S, Breves, G, Mielenz, M and Sauerwein, H 2014. Longitudinal profiling of the tissue-specific expression of genes related with insulin sensitivity in dairy cows during lactation focusing on different fat depots. PloS ONE 9, e86211.CrossRefGoogle ScholarPubMed
Schäff, C, Börner, S, Hacke, S, Kautzsch, U, Sauerwein, H, Spachmann, SK, Schweigel-Röntgen, M, Hammon, HM and Kuhla, B 2013. Increased muscle fatty acid oxidation in dairy cows with intensive body fat mobilization during early lactation. Journal of Dairy Science 96, 64496460.CrossRefGoogle ScholarPubMed
Schoenberg, KM, Waldron, MR, Giesy, SL, Boisclair, YR, Harvatine, KJ, Kharitonenkov, A and Cheng, C 2011. Plasma FGF21 is elevated by the intense lipid mobilization of lactation. Endocrinology 152, 46524661.CrossRefGoogle ScholarPubMed
Schooneman, MG, Achterkamp, N, Argmann, CA, Soeters, MR and Houten, SM 2014. Plasma acylcarnitines inadequately reflect tissue acylcarnitine metabolism. Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids 1841, 987994.CrossRefGoogle ScholarPubMed
Schooneman, MG, Vaz, FM, Houten, SM and Soeters, MR 2013. Acylcarnitines: reflecting or inflicting insulin resistance? Diabetes 62, 18.CrossRefGoogle ScholarPubMed
Shimabukuro, M, Higa, M, Zhou, Y-T, Wang, M-Y, Newgard, CB and Unger, RH 1998. Lipoapoptosis in beta-cells of obese prediabetic fa/fa rats role of serine palmitoyltransferase overexpression. Journal of Biological Chemistry 273, 3248732490.CrossRefGoogle ScholarPubMed
Shimabukuro, M, Koyama, K, Chen, G, Wang, M-Y, Trieu, F, Lee, Y, Newgard, CB and Unger, RH 1997. Direct antidiabetic effect of leptin through triglyceride depletion of tissues. Proceedings of the National Academy of Sciences 94, 46374641.CrossRefGoogle ScholarPubMed
Song, Y, Li, N, Gu, J, Fu, S, Peng, Z, Zhao, C, Zhang, Y, Li, X, Wang, Z, Li, X and Liu, G 2016. β-Hydroxybutyrate induces bovine hepatocyte apoptosis via an ROS-p38 signaling pathway. Journal of Dairy Science 99, 91849198.CrossRefGoogle ScholarPubMed
Sordillo, LM and Aitken, SL 2009. Impact of oxidative stress on the health and immune function of dairy cattle. Veterinary Immunology and Immunopathology 128, 104109.CrossRefGoogle ScholarPubMed
Sordillo, LM and Raphael, W 2013. Significance of metabolis stress, lipid mobilization, and inflammation on transition cow disorders. Veterinary Clinics: Food Animal Practice 29, 267278.Google Scholar
Sordillo, LM, Weaver, JA, Cao, Y-Z, Corl, C, Sylte, MJ and Mullarky, IK 2005. Enhanced 15-HPETE production during oxidant stress induces apoptosis of endothelial cells. Prostaglandins and Other Lipid Mediators 76, 1934.CrossRefGoogle ScholarPubMed
Stein, DT, Stevenson, BE, Chester, MW, Basit, M, Daniels, MB, Turley, SD and McGarry, JD 1997. The insulinotropic potency of fatty acids is influenced profoundly by their chain length and degree of saturation. Journal of Clinical Investigation 100, 398403.CrossRefGoogle ScholarPubMed
Summers, SA and Goodpaster, BH 2016. CrossTalk proposal: intramyocellular ceramide accumulation does modulate insulin resistance. Journal of Physiology 594, 31673170.CrossRefGoogle ScholarPubMed
Turpin, S, Ryall, J, Southgate, R, Darby, I, Hevener, A, Febbraio, M, Kemp, B, Lynch, G and Watt, M 2009. Examination of ‘lipotoxicity’ in skeletal muscle of high-fat fed and ob/ob mice. Journal of Physiology 587, 15931605.CrossRefGoogle ScholarPubMed
Turpin, SM, Nicholls, HT, Willmes, DM, Mourier, A, Brodesser, S, Wunderlich, CM, Mauer, J, Xu, E, Hammerschmidt, P and Brönneke, HS 2014. Obesity-induced CerS6-dependent C16:0 ceramide production promotes weight gain and glucose intolerance. Cell Metabolism 20, 678686.CrossRefGoogle Scholar
Urh, C, Denißen, J, Harder, I, Koch, C, Gerster, E, Ettle, T, Kraus, N, Schmitz, R, Kuhla, B, Stamer, E, Spiekers, H and Sauerwein, H 2019. Circulating adiponectin concentrations during the transition from pregnancy to lactation in high-yielding dairy cows: testing the effects of farm, parity, and dietary energy level in large animal numbers. Domestic Animal Endocrinology 69, 112.CrossRefGoogle ScholarPubMed
Videla, LA, Rodrigo, R, Araya, J and Poniachik, J 2004. Oxidative stress and depletion of hepatic long-chain polyunsaturated fatty acids may contribute to nonalcoholic fatty liver disease. Free Radical Biology and Medicine 37, 14991507.CrossRefGoogle ScholarPubMed
Videla, LA, Rodrigo, R, Araya, J and Poniachik, J 2006. Insulin resistance and oxidative stress interdependency in non-alcoholic fatty liver disease. Trends in Molecular Medicine 12, 555558.CrossRefGoogle ScholarPubMed
Wang, J, Zhu, X, Chen, C, Li, X, Gao, Y, Li, P, Zhang, Y, Long, M and Wang, Z 2012. Effect of insulin-like growth factor-1 (IGF-1) on the gluconeogenesis in calf hepatocytes cultured in vitro. Molecular and Cellular Biochemistry 362, 8791.CrossRefGoogle ScholarPubMed
Whatling, C, McPheat, W and Herslof, M 2007. The potential link between atherosclerosis and the 5-lipoxygenase pathway: investigational agents with new implications for the cardiovascular field. Expert Opinion on Investigational Drugs 16, 18791893.CrossRefGoogle ScholarPubMed
Wiesner, P, Leidl, K, Boettcher, A, Schmitz, G and Liebisch, G 2009. Lipid profiling of FPLC-separated lipoprotein fractions by electrospray ionization tandem mass spectrometry. Journal of Lipid Research 50, 574585.CrossRefGoogle ScholarPubMed
Yadav, A, Kataria, MA, Saini, V and Yadav, A 2013. Role of leptin and adiponectin in insulin resistance. Clinica Chimica Acta 417, 8084.CrossRefGoogle ScholarPubMed
Yang, G, Badeanlou, L, Bielawski, J, Roberts, AJ, Hannun, YA and Samad, F 2009. Central role of ceramide biosynthesis in body weight regulation, energy metabolism, and the metabolic syndrome. American Journal of Physiology-Endocrinology and Metabolism 297, E211E224.CrossRefGoogle ScholarPubMed
Yang, Y, Sadri, H, Prehn, C, Adamski, J, Rehage, J, Dänicke, S, Saremi, B and Sauerwein, H 2019. Acylcarnitine profiles in serum and muscle of dairy cows receiving conjugated linoleic acids or a control fat supplement during early lactation. Journal of Dairy Science 102, 754767.CrossRefGoogle ScholarPubMed
Zachut, M, Honig, H, Striem, S, Zick, Y, Boura-Halfon, S and Moallem, U 2013. Periparturient dairy cows do not exhibit hepatic insulin resistance, yet adipose-specific insulin resistance occurs in cows prone to high weight loss. Journal of Dairy Science 96, 56565669.CrossRefGoogle Scholar
Zhou, Y, Zhou, Z, Batistel, F, Martinez-Cortés, I, Pate, R, Luchini, D and Loor, J 2018. Methionine and choline supply alter transmethylation, transsulfuration, and cytidine 5′-diphosphocholine pathways to different extents in isolated primary liver cells from dairy cows. Journal of Dairy Science 101, 1138411395.CrossRefGoogle ScholarPubMed
Zhou, Z, Bulgari, O, Vailati-Riboni, M, Trevisi, E, Ballou, M, Cardoso, F, Luchini, D and Loor, J 2016. Rumen-protected methionine compared with rumen-protected choline improves immunometabolic status in dairy cows during the peripartal period. Journal of Dairy Science 99, 89568969.CrossRefGoogle ScholarPubMed
Zom, R, Van Baal, J, Goselink, R, Bakker, J, De Veth, M and Van Vuuren, A 2011. Effect of rumen-protected choline on performance, blood metabolites, and hepatic triacylglycerols of periparturient dairy cattle. Journal of Dairy Science 94, 40164027.CrossRefGoogle ScholarPubMed