Skip to main content

Adipose tissue lipolysis and remodeling during the transition period of dairy cows

Abstract

Elevated concentrations of plasma fatty acids in transition dairy cows are significantly associated with increased disease susceptibility and poor lactation performance. The main source of plasma fatty acids throughout the transition period is lipolysis from adipose tissue depots. During this time, plasma fatty acids serve as a source of calories mitigating the negative energy balance prompted by copious milk synthesis and limited dry matter intake. Past research has demonstrated that lipolysis in the adipose organ is a complex process that includes not only the activation of lipolytic pathways in response to neural, hormonal, or paracrine stimuli, but also important changes in the structure and cellular distribution of the tissue in a process known as adipose tissue remodeling. This process involves an inflammatory response with immune cell migration, proliferation of the cellular components of the stromal vascular fraction, and changes in the extracellular matrix. This review summarizes current knowledge on lipolysis in dairy cattle, expands on the new field of adipose tissue remodeling, and discusses how these biological processes affect transition cow health and productivity.

Background

Adipose tissues, the energy warehouse during the transition period

Adipose tissue (AT) functions as the major body of energy reserve in mammals. During positive energy balance, AT stores energy surplus as fatty acids (FA) incorporated into triacylglycerols (TAG) in a process known as lipogenesis. In de novo lipogenesis, also defined as de novo FA synthesis, FA are derived from carbohydrate and amino acid carbons through acetylCoA. Additionally, AT directly esterifies circulating free FA from dietary or other metabolic origins (e.g. hepatic lipolysis) [1, 2]. The majority of FA incorporated into TAG come from de novo FA synthesis, however, circulating FA may supply up to 50% of FA, especially during early lactation [2, 3]. When energy is limited, the AT releases FA from TAG through lipolysis [4]. Due to its unique lipogenic and lipolytic responses, AT is the only organ capable of extensive growth and reduction at any stage of life [5].

The transition period of dairy cows is defined as the time when the physiological state changes from gestational non-lactating to non-gestational lactating and occurs from 3 wks before through 3 wks after parturition [4]. During the transition period, hormonal changes associated with parturition and the initiation of lactogenesis, including the reduction in progesterone and the surge of prolactin and growth hormone (GH), favor lipolysis over lipogenesis independently of energy balance status [6]. Lipolysis rate is further exacerbated by intense energy requirements, especially glucose, associated with the rapid fetal growth, parturition, the onset of lactation, and reduced dry matter intake, leading to negative energy balance (NEB) [7, 8]. Intense and protracted lipolysis during the transition period leads to AT size reduction and alterations in the secretion pattern of adipokines that favor the use of FA as an energy source for bodily maintenance and glucose for fetal growth and lactation [8, 9]. In addition, lipolysis induces a remodeling process within the adipose organ that is characterized by an inflammatory response, immune cell infiltration, cellular proliferation, and extracellular matrix (ECM) changes [10, 11]. During the transition period, lipolysis-induced AT remodeling coincides with a period of reduced insulin sensitivity in myocytes, hepatocytes, and adipocytes that redirects energy for milk production in the mammary gland [12]. As lactation progresses, bovine adipocytes become more responsive to insulin, resulting in reduced lipolysis rates and enhanced lipogenesis [2]. AT insulin resistance during the transition period ensures a healthy and productive lactation when moderated; however, when intense and protracted, it predisposes cows to inflammatory and metabolic diseases by limiting AT capacity for energy buffering and contributing to chronically increased plasma FA [12, 13]. In this review, we will discuss the different aspects of adipose tissue lipolysis and remodeling during the transition period, and their impact on transition cow health and lactation performance.

Lipolysis

A key characteristic of the transition period is intense lipolysis. In general, lipolysis can be broadly divided into two categories: basal and demand lipolysis [14]. In humans and rodents, the rate of basal lipolysis is determined by adipocyte size and TAG content [10, 15]. Similarly, in dairy cows, basal lipolysis rate is positively associated with adipocyte size and increases steadily throughout lactation [16,17,18,19]. In contrast, demand lipolysis is regulated hormonally in response to energy demands. Independent of the type of lipolysis, TAG within the adipocyte lipid droplet are broken down by the action of three different lipases: adipose triglyceride lipase (ATGL), hormone sensitive lipase (HSL), and monoglyceride lipase (MGL). However, HSL and MGL have a larger role in demand lipolysis than basal lipolysis. The actions of these lipolytic enzymes, explained in subsequent paragraphs, are regulated by different co-activator proteins that also exhibit changes in expression during the transition period.

Lipases

The primary regulator of basal lipolysis in monogastric animals is ATGL (Table 1). This lipase acts exclusively on TAG. ATGL is activated by CGI-58, also known as α/β hydrolase domain-containing protein 5 [20]. Upon lipolytic stimulus and consequent phosphorylation of lipid droplet associated protein perilipin 1 (PLIN1), CGI-58 is released into the cytoplasm and activates ATGL by direct protein-to-protein interaction [20]. In transition dairy cows, ATGL-encoding gene, PNPLA2, is downregulated during the last week of gestation and the first week of lactation compared to the dry period and mid lactation [21, 22]. Similar to the PNPLA2 expression pattern, ATGL abundance remains decreased during the transition period in comparison to the content observed at 4 wks pre-calving [21]. CGI-58 gene expression is also reduced after calving; however, its protein abundance remains unchanged as the transition period progresses [21, 23]. The patterns of gene expression and the protein abundance of ATGL, as well as its coactivator CGI-58, indicate that basal lipolysis is maintained at a steady rate, suggesting that demand lipolysis is the main source of plasma FA during the transition period.

Table 1 Major components of lipolysis pathways in adipose tissue (AT) of dairy cows

HSL is the rate-limiting factor for demand lipolysis ([24, 25], Table 1). This lipase hydrolyses several lipid substrates including TAG, diglycerides, monoglycerides, and cholesterol esters [14]. Upon activation by protein kinase A (PKA), HSL associates with fatty acid binding protein 4 (FABP4) to form a complex that localizes on the lipid droplet. The expression of LIPE, the gene encoding HSL, is reduced during the first 3 wks after parturition compared to the dry period [16, 23]. The protein content of HSL remains unchanged throughout the transition period. However, the rate of phosphorylation at different active sites, and therefore its lipolytic activity, is increased during the first 3 wks after calving [21]. In rodents, HSL requires phosphorylation at Ser563, Ser659, and Ser660 by PKA or protein kinase G (PKG) to initiate TAG hydrolysis. Although HSL serine residue activation sites have not been determined in transition cattle, increased phosphorylation at Ser563 and Ser660 was reported postpartum and during feed restriction protocols [21, 26]. Notably, HSL activity varies depending on anatomical location of adipose tissues. Locher et al. [26] reported higher phosphorylation rates at Ser563 and Ser660 in retroperitoneal AT from dairy cows at 21 d of lactation compared to subcutaneous depots. In the same study [26], phosphorylation rates, especially those at Ser563, were associated with higher lipolytic activity.

A complete activation of the lipolytic process requires not only the activation of ATGL and HSL, but also the phosphorylation of PLIN1. This protein protects the lipid droplet from the lipolytic activity of HSL. PLIN1 is phosphorylated by PKA to allow HSL interaction with TAG. In dairy cows, PLIN1 phosphorylation is increased during the first 3 wks after parturition, compared to other periods of the lactation cycle [21]. In transition dairy cows, lipolysis is modulated by post-transcriptional and allosteric changes in the HSL hydrolase pathway. Although differences in lipolysis rate are reported depending on the adipose tissues’ anatomical site, it is currently unknown if these changes may impact disease susceptibility or lactation efficiency.

The lipolytic pathway is completed by MGL, which acts exclusively on monoglycerides (Table 1). In dairy cows, the gene transcription patterns of MGll, which encodes MGL, follow those of PNPLA2 and LIPE, with lower expression during the first 3 wks of lactation [27]. The dynamics of MGL protein expression are currently unknown in cattle, and it is uncertain if changes in its activity may affect lipolysis rates, as HSL is very active on monoglycerides and MGL hydrolyses these lipid species exclusively.

Regulation of lipolysis during the transition period

In rodents and humans, catecholamines and natriuretic peptides are the most important activators of lipolysis in AT [28]. In the same species, both insulin and catecholamines (acting through alpha adrenergic receptors) are negative regulators of TAG hydrolysis (Table 1). In cattle, the role of catecholamines and insulin in lipolysis modulation is well-documented; however, it is unknown if natriuretic peptides significantly modify lipolytic pathways.

Catecholamines bind to β-adrenergic receptors (βAR) on the adipocyte cell surface, activating adenylyl cyclases that convert ATP to cAMP (Table 1). Accumulation of cAMP induces PKA activation, which in turn phosphorylates PLIN1 and initiates the lipolytic cascade [29]. There are 3 major types of βAR: β1, β2, and β3. In dairy cows, the expression of all βAR-encoding genes was initially reported in mammary gland [30], and,more recently, in subcutaneous AT [16, 27] Only β1 and β2 were demonstrated to have lipolytic responses in the AT of cows, and stimulation of the latter was directly associated with increases in plasma FA during NEB [31]. Similar to the gene expression of lipolytic enzymes, the transcription of B2AR (encoding β2AR) is downregulated immediately after parturition and rises again only after peak lactation [27]. A classical study by Jaster and Wegner [32] revealed that the activity and responses to adrenergic stimuli of the βAR in the subcutaneous AT was increased during the first month after parturition compared to the dry period. The dynamics of AT βAR response during the dry and lactation periods were completely characterized by McNamara and Hillers [17] who demonstrated that lipolytic responses to epinephrine peak at 1 month postpartum and remain elevated 6 months into lactation compared to 1 month pre calving. In the same study, lipolytic responses to norepinephrine stimulation remained higher during lactation compared to the dry period. Strikingly, the lipolytic response triggered by βAR stimulation in early lactation is increased in proportion to milk production [27]. Although not described in dairy cows, βAR density and the lipolytic activity in adipocytes after adrenergic stimulation may vary depending on the anatomical location of the adipose depot. In sheep, there are more βAR present in omental adipocytes, compared to subcutaneous adipocytes, after 2 wks of lactation [6]. The impact of the higher content of βAR in abdominal AT is reflected in the higher rate of lipolysis from these depots during NEB states related to lactation. In fact, the FA profiles of the plasma NEFA fraction in cows with intense lipolysis, such as those with displaced abomasum, are remarkably similar to the FA composition of visceral adipose depots, especially in the content of saturated and monounsaturated FA [33].

Adipocytes are one of the most highly insulin-responsive cell types [34]. In adipocytes, insulin stimulates glucose transport and lipogenesis, promotes the uptake of FA from systemic circulation, and inhibits lipolysis [34, 35]. Insulin binds and activates the insulin receptor tyrosine kinase, resulting in the phosphorylation of insulin receptor substrates 1 and 2 (IRS-1/IRS-2) [36]. These events are followed by activation of phosphatidylinositol 3-kinase (PI3K), phophoinositide-dependent kinase1 (PDK1), and protein kinase B (AKT), which mediate insulin metabolic and mitogenic effects, including glucose uptake through glucose transporters (e.g. GLUT4) [35]. Insulin suppresses lipolysis through the activation of AKT, which results in the inhibition of downstream protein kinase A (PKA) and reduces PKA phosphorylation of PLIN1 [37]. In dairy cows, decreased circulating insulin and AKT phosphorylation in the liver and AT stimulate gluconeogenesis and lipolysis during the transition period and early lactation [8, 12, 38].

During the transition period, changes in the secretion patterns of GH, angiopoietin-like 4 (ANGPTL4), and prolactin further modulate lipolysis. GH activates lipolysis and reduces insulin sensitivity in mammalian AT [39]. In lactating dairy cattle, GH administration augments 2 to 6-fold AT response to adrenergic stimulation [40]. In vitro experiments with AT from lactating animals indicate that GH reduces adipocyte sensitivity to anti-lipolytic molecules, such as adenosine, that inhibit the activity of adenylyl cyclase [41]. Thus, compared to non-lactating cows 4 to 8 wks prior to parturition, cows in the transition period have elevated circulating GH which enhances lipolysis by increasing adipocyte responses to adrenergic stimuli and reducing the inhibitory effects on sympathetic activity through adenosine.

ANGPTL4, also known as fasting-induced adipose factor, is an adipokine secreted during NEB that inhibits adipocyte uptake of FA for esterification [42]. ANGPTL4 plays a key role in enhancing lipolysis in adipocytes, especially during catecholamine stimulation, by increasing cAMP and enhancing the phosphorylation of PKA [43]. ANGPTL4 transcription and synthesis is elevated in response to glucocorticoids [43], possibly indicating that this adipokine facilitates lipolytic response during stress periods, such as parturition. In dairy cows, Koltes and Spurlock [44] described the dynamics of ANGPTLl4 gene expression following lipolysis induction with GH, as well as during NEB states, including the transition period and during feed restriction. ANGPTL4 transcription was inversely associated with the degree of NEB in all three models of limited energy status. Although ANGPTL4 protein content in AT from transition cows has not been determined, it is expected to reflect ANGPTL4 transcription, similar to what has been observed in rodents [45]. Thus, in dairy cattle, ANGPTL4 supports lipolysis during NEB stages, including the transition period.

In addition to supporting lactation, prolactin promotes adipocyte lipolysis in vivo. Furthermore, continuous exposure of fat cells to this pituitary hormone have been shown to reduce AT size in rabbits [46]. However, it is unknown if prolactin exerts the same effect in bovine adipocytes, and its effect on modulating lipolysis intensity during the transition period remains to be elucidated.

Adipokines

The role of AT is now recognized as a major regulator of systemic metabolism which extends beyond energy buffering [47]. This function is accomplished by secreting specialized proteins that exert autocrine, paracrine, and endocrine functions. These proteins are termed adipokines and are produced by the cellular components of AT, such as adipocytes and cells of the stromal vascular fraction (SVF), including immune, vascular, and adipocyte progenitor cells. Despite the ever-expanding list of adipokines, which now accounts for over 300 secretory products [48], few have been studied in dairy cattle (Table 2). Among these signaling molecules, adiponectin and leptin are almost exclusively secreted by AT; while others, such as resistin and retinol binding protein 4, are also produced in the liver. Similarly, interleukin-6 (IL-6) and tumor necrosis factor alpha (TNFα) are not only produced by adipocytes, but also resident immune cells in AT (e.g. macrophages, lymphocytes, polymorphonuclear cells). Recent research provides evidence that the dynamics of adipokine secretion during the transition period drive the homeorhetic potential of dairy cows by redirecting glucose to the mammary gland, increasing FA flow to the liver, and modulating energy intake [49].

Table 2 Adipokine expression dynamics and roles in modulating adipose tissue (AT) lipolysis in transition dairy cows

Adiponectin, also known as ACRP30, is secreted primarily by adipocytes, but is also expressed by cardiomyocytes and skeletal muscle [50]. Adiponectin improves insulin sensitivity and lipogenesis in adipocytes, and FA β-oxidation in myocytes and hepatocytes [9]. These effects are exerted by the activation of its receptors (adipoR1 and adipoR2), which are expressed in liver, AT, and skeletal muscle [51]. In monogastric animals, circulating adiponectin is present in 3 major oligomeric forms (low-molecular weight (LMW), middle molecular weight (MMW), high molecular weight (HMW) and as globular adiponectin [51]. In dairy cows, circulating adiponectin is composed mainly of high molecular weight complexes and its distribution is not affected by stage of lactation [49, 52]. Circulating adiponectin reaches its nadir immediately after calving, then peaks between 40 and 70 d into lactation [53, 54]. Remarkably, concentrations of circulating adiponectin are inversely associated with plasma FA, the main lipolysis biomarker in dairy cows [54]. Similarly, AT expression of genes encoding adipoR1 and adipoR2 is downregulated during the first 3 weeks after calving and then increases steadily to peak at around 100 DIM [55, 56]. The dynamics of the expression and secretion of adiponectin and its receptors may indicate that this adipokine acts as an autocrine, paracrine, and endocrine modulator of the homeorhetic adaptations of AT during the transition period. However, the extent to which adiponectin affects glucose partitioning to the mammary gland in early lactation is currently unknown.

Leptin was one of the first adipokines characterized in mammals. Unlike adiponectin, leptin is expressed and secreted not only by adipocytes, but also by myocytes and mammary gland and gastric mucosa epithelial cells [57]. In dairy cows and other ruminants, the leptin encoding gene (OB) is expressed ubiquitously, including in the rumen, abomasum, duodenum, mammary gland, skeletal muscle, pituitary gland, and AT (reviewed in [58]). Similar to adiponectin, leptin acts in an autocrine, paracrine, and endocrine manner to modulate food intake and energy expenditure. This anorexigenic adipokine reduces lipogenesis and increases lipolysis and FA oxidation [59]. In dairy cows, plasma leptin concentration peaks during the dry period and decreases drastically in the first week after calving [60, 61]. In AT, OB expression reflects leptinemia dynamics, with the highest expression in the dry period and minimal transcription immediately following parturition [60]. Reduction in leptin transcription and secretion during the transition period may promote a rapid return to normal DMI; however, it is presently unclear if, in over-conditioned cows, hyperleptinemia enhances the onset of pre-calving lipolysis and losses of AT reserves [62, 63].

Resistin is an adipokine secreted by adipocytes and macrophages [64]. AT transcription and secretion levels of resistin are linked to the development of insulin resistance in humans, as well as in rodent models of obesity and metabolic syndrome [65]. In transition dairy cows, plasma concentration of resistin peaks during the first week after calving and returns to pre-partum levels after 6 wks of lactation [66, 67]. Plasma resistin is positively associated with plasma NEFA and negatively correlated with milk production [66, 67]. AT resistin transcription and secretion dynamics are similar to that of plasma resistin, with higher expression during the first week post-calving compared to other stages of lactation and the dry period [66]. Remarkably, resistin was associated with higher rate of lipolysis, as well as enhanced transcription of ATGL and HSL, in AT explants from transition cows [66].

RBP4 is a lipocalin transport protein secreted by adipocytes and hepatocytes that serves as a carrier of retinol [68]. Like resistin, RBP4 impairs glucose uptake by adipocytes and hepatocytes by suppressing insulin signaling pathways [69]. Enhanced expression of RBP4 inhibits adipogenesis, thus impairing the FA buffering capacity of AT during obesity or periods of high lipolytic rate [70]. In dairy cows, plasma RBP4 declines sharply after parturition and returns to pre-calving levels by the second week of lactation [71, 72]. In the AT of transition cows, RBP4 transcription was detected and found to be positively associated with TNFα secretion [72].

Adipose tissue remodeling

AT lipolysis not only involves the release of FA, neutral lipids, and glycerol, but also induces a remodeling process of the organ [10, 73]. This remodeling is characterized by an inflammatory response with immune cell migration, proliferation of the cellular components of the SVF, and changes in ECM of AT [5]. Intense research during the past two decades has characterized AT remodeling in human diseases that exhibit high AT lipolysis rates, such as obesity [5, 74], metabolic syndrome [75], and lipodystrophy [76]. More recently, different aspects of AT remodeling in dairy cows during the transition period and metabolic disease were evaluated. We summarize these reports from cattle studies below in the context of the current understanding of adipose tissue remodeling in humans and rodent models.

Immune cell infiltration

Studies in multiple animal species and humans show dynamic populations of immune cells located within the SVF of AT [77,78,79]. Cells of the innate and adaptive immune systems, such as macrophages, mast cells, lymphocytes, neutrophils, eosinophils, dendritic cells, and NK, exhibit active AT trafficking during health and disease in rodents and humans [80], as well as in dairy cows during the transition period and late lactation [81, 82]

Macrophages are the most predominant cell type in monogastric and ruminant SVF [77, 79]. Adipose tissue macrophages (ATM) are a key component of the inflammatory response during lipolysis. The specific inflammatory phenotype of ATM has been broadly classified in classical ATM (M1), which have active pro-inflammatory responses, and alternative phenotype ATM (M2), which promote inflammation resolution. In dairy cows, ATM infiltration occurs as a response to intense lipolysis. In cases of displaced abomasum in transition dairy cows, macrophages exhibiting a M1 phenotype accumulate in aggregates within omental and subcutaneous depots [81]. During induced NEB in late-lactation cows, lipolysis induced ATM infiltration into subcutaneous and visceral AT, yet no phenotypic change was observed [82].

The role of ATM during lipolysis is to remove lipolytic products such as FA, diglycerides, and monoglycerides that, in excess, cause lipotoxicity in AT [83]. ATM are also involved in the recruitment of new adipocyte progenitors by secreting chemotactic proteins such as osteopontin [83]. The transcription of this latter protein was shown to be upregulated in AT of lactating dairy goats during feed deprivation [84]. The ATM phenotype is plastic in response to the milieu of adipocyte-derived molecules, such as adipokines and FA [79]. During lipolysis, ATM bind to saturated FA at toll-like receptors (TLR), resulting in the activation of NF-κB transcription factor pathways and inflammatory gene transcription, leading to M1 polarization [85,86,87]. ATM secretion of cytokines such as TNFα and interleukins 1β and 6 during lipolysis mediates local and systemic inflammation,6 [79, 88]. Interestingly, macrophage infiltration into AT enhances lipolysis, creating a vicious cycle that connects lipolysis, ATM infiltration, and inflammation [89]. To date, this cycle has not been described in cows, but, if exists, could offer a novel explanation of prolonged, severe lipolysis in the transition period.

ATM exhibiting the M2 phenotype possess a restorative and protective function, in contrast to the pro-inflammatory M1 type [90]. The main immune cell-derived inducers of non-classical macrophages are interleukins 4, 10, and 13 [91]. In human and mouse studies, the alternative macrophage phenotype has been induced by omega-3 FA molecules and pharmacological agents through peroxisome proliferator-activated receptors [92,93,94]. Given the intense ATM polarization to M1 during excessive lipolysis in the transition period, the possibility of modulating ATM phenotype in transition dairy cows may offer therapeutic alternatives [81].

Reports on trafficking and inflammatory responses of adipose immune cells, other than ATM, in dairy cows are limited. T-cells were reported to comprise 3–5% of the SVF cells in cows, with or without displaced abomasum, during the transition period and in late lactation [81, 82, 95]. Around 4–7% of SVF cells are B lymphocytes and their numbers are higher in visceral AT than other AT. Interactions among different types of immune cells, adipocytes, and other cells in the SVF of AT modulate metabolic responses during the transition period. Therefore, a better understanding of immune cell population trafficking and inflammatory responses is necessary to develop strategies to modulate AT metabolism, especially lipolysis.

Cell proliferation

AT is a plastic organ that adapts to metabolic challenges by expansion or contraction of adipocyte size and numbers. Furthermore, adipocytes have a constant turnover that requires recruitment of new progenitors regularly [96]. Lipolysis induces the recruitment of new adipocytes in rodent models of obesity and adrenergic signaling [97]. In dairy cows, assessment of cell turnover is difficult because genetic tracing tools are not available for large animals. Markers of cell cycle activation, such as CCNA2, CCNB2, and MKI67 have been used. In late-lactation cows, short-term intense lipolysis did not increase the expression of these genes [82]. In dairy heifers during peak lactation, Häussler et al. detected a 25-fold increase in the number of preadipocytes in the retroperitoneal AT [98], compared to heifers in mid lactation (105 DIM), using immunohistochemistry with antibodies against Ki67, a cell proliferation marker, and Pref-1 (preadipocyte factor). Remarkably, visceral AT apoptosis was also increased in early lactation compared to mid lactation, as demonstrated by the Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay that detects DNA fragments as a measure of apoptosis. The implications of a potentially increased rate of cell proliferation in AT during the transition period is currently unknown. Increased preadipocyte proliferation, and therefore adipogenesis, especially in subcutaneous AT, would likely be beneficial to transition cow metabolism, by, for example, improving the AT capacity to buffer excess FA released during lipolysis, as has been shown in humans and rodent models of insulin signaling dysregulation [99]

Extracellular matrix

The ECM of AT gives structural support to its cellular components. Proteoglycans and fibrous proteins, such as collagens, are the main classes of AT ECM proteins, and their functions and distribution were recently reviewed in detail by Mariman and Wang [100]. In human obesity, characterized by excessive rates of basal lipolysis, there is enhanced deposition of collagens I and VI and thrombospondin-1 [101,102,103]. The presence of these ECM proteins is also associated with ATM infiltration and polarization to the M1 phenotype [102, 104]. High rates of lipolysis observed in cancer cachexia are also associated with changes in the ECM composition that lead to enhanced inflammatory responses by adipocytes [105, 106]. In late-lactation dairy cows, ECM composition is affected by anatomical location, with higher expression of collagens I and VI in subcutaneous fat compared to omental fat [82]. Reports on the dynamics of ECM remodeling during the transition period and early lactation are limited to gene expression evaluation. Akbar et al., [107] reported higher expression of collagen I in subcutaneous AT of transition cows with metritis, compared to healthy animals. In feed-deprived lactating goats, Faulconnier and colleagues described enhanced transcription of COL3A1 [84]. A reanalysis of microarray data by Sumner-Thompson et al., [108] looking for enrichment in different ECM-related groupings via the Database for Annotation, Visualization, and Integrated Discovery revealed that THBS1 expression is upregulated during peak lipolysis at the onset of lactation. Proteome analysis focused on the transition period is necessary to evaluate if lipolytic responses during the transition period induce changes in the composition of ECM that impair AT function.

Impact of AT lipolysis and remodeling

Dysregulated inflammatory responses and oxidative stress are recognized as key components of the metabolic stress syndrome in transition dairy cows [7, 109]. AT remodeling and lipolysis can be considered the third key component, also leading to metabolic stress through two different mechanisms. First, and as demonstrated initially in non-ruminants, lipolysis modulates the phenotype of immune cells [110]. FA and other lipolytic products are potent activators of inflammatory pathways in mononuclear cells, lymphocytes, and polymorphonuclear cells [48, 111]. In dairy cows, FA impair the immune responses of lymphocytes and neutrophils by enhancing the pro-inflammatory response and simultaneously reduce their capacity to clear pathogens [64, 112]. Second, excessive lipolysis is a trigger of oxidative stress. In monogastrics, FA in circulation and in tissues become oxidized by free radicals and rapidly deplete antioxidant reserves, leading to oxidative stress [101]. In dairy cows, specific oxidized products of arachidonic acid, 11-hydroxyeicosatetraenoic acid (11-HETE), and linoleic acid, 9-hydroxyoctadecadienoic acid (9-HODE) and 13-HODE, significantly increase during the transition period coinciding with enhanced lipolytic rate [113]. Modulating the rate and intensity of FA release from AT through nutritional interventions, including supplementation of methyl donors choline and methionine [114], and ω-6 polyunsaturated FA [104, 115, 116], improves immune function, reduces oxidative stress, and enhances adipose insulin sensitivity. These studies further demonstrate the importance of lipolysis as a determinant factor for disease susceptibility in transition dairy cows.

Excessive lipolysis and AT remodeling may also impair lactation performance. Increased concentrations of lipolysis biomarkers NEFA and β-hydroxybutyrate during the transition period are associated with reduced milk production and impaired reproductive function [117]. In monogastrics, AT inflammation, a feature of AT remodeling, is directly linked with the development of AT-specific insulin resistance (IR) [11]. In transition cows a transient state of IR is considered homeorhetic, as it guarantees glucose supply for milk synthesis by limiting glucose use by peripheral tissues and triggering AT lipolysis [12]. However, extended periods of IR during the transition period may increase and prolong AT lipolysis, as observed in women with gestational diabetes and metabolic syndrome patients [118, 119]. During active AT remodeling, the expression and secretion of IL1-β, IL-6, resistin, and TNF-α by human adipocytes and ATM is increased, leading to impaired AT insulin signaling [74, 88]. In transition dairy cows, AT expression of IL-6, resistin, TNF-α, and other bioactive peptides associated with IR is increased compared to other stages of lactation [120,121,122]. Recent studies provide evidence for AT-specific IR during early lactation in high-yielding dairy cows. Zachut et. al [38] demonstrated a significant reduction in the phosphorylation of downstream insulin signaling pathways, such as IRS-1 and AKT, in subcutaneous adipose, while the activation of these pathways remained intact in the liver. Remarkably, adipose-specific IR was only observed in cows that had higher rates of lipolysis and lost more body condition during early lactation. Despite this evidence, the role of periparturient AT remodeling in the development of IR is still a matter of debate, as Mann and colleagues did not observe any changes in insulin signaling pathways in over-conditioned cows with excessive lipolytic rates [123]. Furthermore, throughout lactation the degree of IR is dynamic and was shown to increase during late lactation, a period characterized by reduced lipolytic activity [124]. These reports underscore that lipolysis regulation by insulin signaling and AT remodeling is a complex process that requires further characterization.

Conclusions

The impact of intense and extended periods of AT lipolysis on transition cow disease susceptibility and lactation performance may be directly related to the remodeling process, alterations in the expression of adipokines, and the development of insulin resistance (Fig. 1). Certainly, AT biology research focus on human obesity and the diabetes epidemic in western countries has expanded our understanding of the role of lipolysis in metabolic and immune function. However, further research on AT lipolysis and remodeling is required to elucidate specific inflammatory and metabolic pathways that link adipocyte and immune cell function in dairy cattle. Improving our understanding of AT function in transition dairy cattle will lead to the identification of new biomarkers of disease and productivity, which will allow for improved herd health and profitability.

Fig. 1
figure 1

Lipolysis induces adipose tissue (AT) remodeling. This process is characterized by macrophage infiltration and changes in inflammatory phenotype. During moderate lipolysis, macrophage infiltration is limited and involves mainly the M2 phenotype (anti-inflammatory). In contrast, during excessive lipolysis, most infiltrating macrophages are M1 (pro-inflammatory) and enhance lipolysis and reduce adipocyte insulin sensitivity. Excessive lipolysis and AT remodeling increase disease susceptibility and negatively impact lactation performance

Abbreviations

AT:

Adipose tissue

ATM:

Adipose tissue macrophages

FA:

Fatty acids

M1:

Pro-inflammatory phenotype

M2:

Anti-inflammatory phenotype

NEFA:

Non-esterified fatty acids

TAG:

Triacylglycerols

References

  1. Ameer F, Scandiuzzi L, Hasnain S, Kalbacher H, Zaidi N. De novo lipogenesis in health and disease. Metabolism. 2014;63(7):895–902.

    Article  CAS  PubMed  Google Scholar 

  2. McNamara J. Lipid metabolism in adipose tissue during lactation: a model of a metabolic control system. J Nutr. 1994;124(8 Suppl):1383S–91S.

    CAS  PubMed  Google Scholar 

  3. McNamara J, Hillers J. Regulation of bovine adipose tissue metabolism during lactation. 1. Lipid synthesis in response to increased milk production and decreased energy intake1. J Dairy Sci. 1986;69(12):3032–41.

    Article  CAS  PubMed  Google Scholar 

  4. Contreras GA, Sordillo LM. Lipid mobilization and inflammatory responses during the transition period of dairy cows. Comp Immunol Microbiol Infect Dis. 2011;34(3):281–9.

    Article  PubMed  Google Scholar 

  5. Rutkowski JM, Stern JH, Scherer PE. The cell biology of fat expansion. J Cell Biol. 2015;208(5):501–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Vernon R, Doris R, Finley E, Houslay M, Kilgour E, Lindsay-Watt S. Effects of lactation on the signal transduction systems regulating lipolysis in sheep subcutaneous and omental adipose tissue. Biochem J. 1995;308:291–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sordillo LM, Raphael W. Significance of metabolic stress, lipid mobilization, and inflammation on transition cow disorders. Vet Clin North Am Food Anim Pract. 2013;29(2):267–78.

    Article  PubMed  Google Scholar 

  8. Bell AW, Bauman DE. Adaptations of glucose metabolism during pregnancy and lactation. J Mammary Gland Biol Neoplasia. 1997;2(3):265–78.

    Article  CAS  PubMed  Google Scholar 

  9. Stern J, Rutkowski J, Scherer P. Adiponectin, leptin, and fatty acids in the maintenance of metabolic homeostasis through adipose tissue crosstalk. Cell Metab. 2016;23(5):770–84.

    Article  CAS  PubMed  Google Scholar 

  10. Kosteli A, Sugaru E, Haemmerle G, Martin JF, Lei J, Zechner R, et al. Weight loss and lipolysis promote a dynamic immune response in murine adipose tissue. J Clin Invest. 2010;120(10):3466–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sun K, Kusminski CM, Scherer PE. Adipose tissue remodeling and obesity. J Clin Invest. 2011;121(6):2094–101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. De Koster JD, Opsomer G. Insulin resistance in dairy cows. Vet Clin North Am Food Anim Pract. 2013;29(2):299–322.

    Article  PubMed  Google Scholar 

  13. Faulkner A, Pollock HT. Metabolic responses to euglycaemic hyperinsulinaemia in lactating and non-lactating sheep in vivo. J Endocrinol. 1990;124(1):59–66.

    Article  CAS  PubMed  Google Scholar 

  14. Lafontan M, Langin D. Lipolysis and lipid mobilization in human adipose tissue. Prog Lipid Res. 2009;48(5):275–97.

    Article  CAS  PubMed  Google Scholar 

  15. Magkos F, Fraterrigo G, Yoshino J, Luecking C, Kirbach K, Kelly Shannon C, et al. Effects of moderate and subsequent progressive weight loss on metabolic function and adipose tissue biology in humans with obesity. Cell Metab. 2016;23(4):591–601.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Sumner JM, McNamara JP. Expression of lipolytic genes in the adipose tissue of pregnant and lactating Holstein dairy cattle1. J Dairy Sci. 2007;90(11):5237–46.

    Article  CAS  PubMed  Google Scholar 

  17. McNamara JP, Hillers JK. Adaptations in lipid metabolism of bovine adipose tissue in lactogenesis and lactation. J Lipid Res. 1986;27(2):150–7.

    CAS  PubMed  Google Scholar 

  18. De Koster J, Van den Broeck W, Hulpio L, Claeys E, Van Eetvelde M, Hermans K, et al. Influence of adipocyte size and adipose depot on the in vitro lipolytic activity and insulin sensitivity of adipose tissue in dairy cows at the end of the dry period. J Dairy Sci. 2016;99(3):2319–28.

    Article  PubMed  CAS  Google Scholar 

  19. Yang YT, Baldwin RL. Lipolysis in isolated cow adipose cells. J Dairy Sci. 1973;56(3):366–74.

    Article  CAS  PubMed  Google Scholar 

  20. Granneman JG, Moore H-PH, Krishnamoorthy R, Rathod M. Perilipin controls lipolysis by regulating the interactions of AB-hydrolase containing 5 (Abhd5) and adipose triglyceride lipase (Atgl). J Biol Chem. 2009;284(50):34538–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Koltes DA, Spurlock DM. Coordination of lipid droplet-associated proteins during the transition period of Holstein dairy cows. J Dairy Sci. 2011;94(4):1839–48.

    Article  CAS  PubMed  Google Scholar 

  22. Ji P, Osorio JS, Drackley JK, Loor JJ. Overfeeding a moderate energy diet prepartum does not impair bovine subcutaneous adipose tissue insulin signal transduction and induces marked changes in peripartal gene network expression. J Dairy Sci. 2012;95(8):4333–51.

    Article  CAS  PubMed  Google Scholar 

  23. Khan MJ, Hosseini A, Burrell S, Rocco SM, McNamara JP, Loor JJ. Change in subcutaneous adipose tissue metabolism and gene network expression during the transition period in dairy cows, including differences due to sire genetic merit1. J Dairy Sci. 2013;96(4):2171–82.

    Article  CAS  PubMed  Google Scholar 

  24. Miyoshi H, Perfield JW, Obin MS, Greenberg AS. Adipose triglyceride lipase regulates basal lipolysis and lipid droplet size in adipocytes. J Cell Biochem. 2008;105(6):1430–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lass A, Zimmermann R, Oberer M, Zechner R. Lipolysis – A highly regulated multi-enzyme complex mediates the catabolism of cellular fat stores. Prog Lipid Res. 2011;50(1):14–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Locher LF, Meyer N, Weber EM, Rehage J, Meyer U, Dänicke S, et al. Hormone-sensitive lipase protein expression and extent of phosphorylation in subcutaneous and retroperitoneal adipose tissues in the periparturient dairy cow. J Dairy Sci. 2011;94(9):4514–23.

    Article  CAS  PubMed  Google Scholar 

  27. Rocco SM, McNamara JP. Regulation of bovine adipose tissue metabolism during lactation. 7. Metabolism and gene expression as a function of genetic merit and dietary energy intake1. J Dairy Sci. 2013;96(5):3108–19.

    Article  CAS  PubMed  Google Scholar 

  28. Arner P, Langin D. Lipolysis in lipid turnover, cancer cachexia, and obesity-induced insulin resistance. Trends Endocrinol Metab. 2014;25(5):255–62.

    Article  CAS  PubMed  Google Scholar 

  29. Duncan RE, Ahmadian M, Jaworski K, Sarkadi-Nagy E, Sul HS. Regulation of lipolysis in adipocytes. Annu Rev Nutr. 2007;27(1):79–101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Inderwies T, Pfaffl MW, Meyer HHD, Blum JW, Bruckmaier RM. Detection and quantification of mRNA expression of α- and β-adrenergic receptor subtypes in the mammary gland of dairy cows. Domest Anim Endocrinol. 2003;24(2):123–35.

    Article  CAS  PubMed  Google Scholar 

  31. Ferlay A, Chilliard Y. Effects of the infusion of non-selective beta-, and selective beta1- or beta2-adrenergic agonists, on body fat mobilisation in underfed or overfed non-pregnant heifers. Reprod Nutr Dev. 1999;39(4):409–21.

    Article  CAS  PubMed  Google Scholar 

  32. Jaster EH, Wegner TN. Beta-adrenergic receptor involvement in lipolysis of dairy cattle subcutaneous adipose tissue during dry and lactating state. J Dairy Sci. 1981;64(8):1655–63.

    Article  CAS  PubMed  Google Scholar 

  33. Hostens M, Fievez V, Leroy JLMR, Van Ranst J, Vlaeminck B, Opsomer G. The fatty acid profile of subcutaneous and abdominal fat in dairy cows with left displacement of the abomasum. J Dairy Sci. 2012;95(7):3756–65.

    Article  CAS  PubMed  Google Scholar 

  34. Kahn BB, Flier JS. Obesity and insulin resistance. J Clin Invest. 2000;106(4):473–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Laviola L, Perrini S, Cignarelli A, Natalicchio A, Leonardini A, De Stefano F, et al. Insulin signaling in human visceral and subcutaneous adipose tissue in vivo. Diabetes. 2006;55(4):952–61.

    Article  CAS  PubMed  Google Scholar 

  36. Kahn SE, Prigeon RL, Schwartz RS, Fujimoto WY, Knopp RH, Brunzell JD, et al. Obesity, body fat distribution, insulin sensitivity and islet β-cell function as explanations for metabolic diversity. J Nutr. 2001;131(2):354S–60S.

    CAS  PubMed  Google Scholar 

  37. Choi SM, Tucker DF, Gross DN, Easton RM, DiPilato LM, Dean AS, et al. Insulin regulates adipocyte lipolysis via an akt-independent signaling pathway. Mol Cell Biol. 2010;30(21):5009–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zachut M, Honig H, Striem S, Zick Y, Boura-Halfon S, Moallem U. Periparturient dairy cows do not exhibit hepatic insulin resistance, yet adipose-specific insulin resistance occurs in cows prone to high weight loss. J Dairy Sci. 2013;96(6):5656–69.

    Article  CAS  PubMed  Google Scholar 

  39. Nellemann B, Vendelbo MH, Nielsen TS, Bak AM, Høgild M, Pedersen SB, et al. Growth hormone-induced insulin resistance in human subjects involves reduced pyruvate dehydrogenase activity. Acta Physiol (Oxf). 2014;210(2):392–402.

    Article  CAS  Google Scholar 

  40. Houseknecht KL, Dwyer DA, Lanna DPD, Bauman DE. Effect of somatotropin on adipose tissue metabolism: Ontogeny of the enhanced response to adrenergic challenge in the lactating cow. Domest Anim Endocrinol. 1995;12(1):105–13.

    Article  CAS  PubMed  Google Scholar 

  41. Houseknecht KL, Bauman DE. Regulation of lipolysis by somatotropin: functional alteration of adrenergic and adenosine signaling in bovine adipose tissue. J Endocrinol. 1997;152(3):465–75.

    Article  CAS  PubMed  Google Scholar 

  42. Kersten S. Regulation of lipid metabolism via angiopoietin-like proteins. Biochem Soc Trans. 2005;33(5):1059–62.

    Article  CAS  PubMed  Google Scholar 

  43. Gray NE, Lam LN, Yang K, Zhou AY, Koliwad S, Wang J-C. Angiopoietin-like 4 (Angptl4) protein is a physiological mediator of intracellular lipolysis in murine adipocytes. J Biol Chem. 2012;287(11):8444–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Koltes DA, Spurlock DM. Adipose tissue angiopoietin-like protein 4 messenger RNA changes with altered energy balance in lactating Holstein cows. Domest Anim Endocrinol. 2012;43(4):307–16.

    Article  CAS  PubMed  Google Scholar 

  45. Dutton S, Trayhurn P. Regulation of angiopoietin-like protein 4/fasting-induced adipose factor (Angptl4/FIAF) expression in mouse white adipose tissue and 3 T3-L1 adipocytes. Br J Nutr. 2008;100(01):18–26.

    Article  CAS  PubMed  Google Scholar 

  46. Fortun-Lamothe L, Langin D, Lafontan M. Influence of prolactin on in vivo and in vitro lipolysis in Rabbits. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol. 1996;115(2):141–7.

    Article  CAS  PubMed  Google Scholar 

  47. Ouchi N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nat Rev Immunol. 2011;11(2):85–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lehr S, Hartwig S, Sell H. Adipokines: A treasure trove for the discovery of biomarkers for metabolic disorders. Proteomics Clin Appl. 2012;6(1-2):91–101.

    Article  CAS  PubMed  Google Scholar 

  49. Giesy SL, Yoon B, Currie WB, Kim JW, Boisclair YR. Adiponectin deficit during the precarious glucose economy of early lactation in dairy cows. Endocrinology. 2012;153(12):5834–44.

    Article  CAS  PubMed  Google Scholar 

  50. Jortay J, Senou M, Abou-Samra M, Noel L, Robert A, Many MC, et al. Adiponectin and skeletal muscle: Pathophysiological implications in metabolic stress. Am J Pathol. 2012;181(1):245–56.

    Article  CAS  PubMed  Google Scholar 

  51. Kadowaki T, Yamauchi T. Adiponectin and adiponectin receptors. Endocr Rev. 2005;26(3):439–51.

    Article  CAS  PubMed  Google Scholar 

  52. Sauerwein H, Häusler S. Endogenous and exogenous factors influencing the concentrations of adiponectin in body fluids and tissues in the bovine. Domest Anim Endocrinol. 2016;56(Supplement):S33–43.

    Article  CAS  PubMed  Google Scholar 

  53. Ohtani Y, Takahashi T, Sato K, Ardiyanti A, Song S-H, Sato R, et al. Changes in circulating adiponectin and metabolic hormone concentrations during periparturient and lactation periods in Holstein dairy cows. Anim Sci J. 2012;83(12):788–95.

    Article  CAS  PubMed  Google Scholar 

  54. Kabara E, Sordillo LM, Holcombe S, Contreras GA. Adiponectin links adipose tissue function and monocyte inflammatory responses during bovine metabolic stress. Comp Immunol Microbiol Infect Dis. 2014;37(1):49–58.

    Article  PubMed  Google Scholar 

  55. Lemor A, Hosseini A, Sauerwein H, Mielenz M. Transition 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. Domest Anim Endocrinol. 2009;37(1):37–44.

    Article  CAS  PubMed  Google Scholar 

  56. Saremi B, Winand S, Friedrichs P, Kinoshita A, Rehage J, Dänicke S, et al. 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. 2014;9(1):e86211.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Cava AL, Matarese G. The weight of leptin in immunity. Nat Rev Immunol. 2004;4(5):371–9.

    Article  PubMed  CAS  Google Scholar 

  58. Chilliard Y, Delavaud C, Bonnet M. Leptin expression in ruminants: Nutritional and physiological regulations in relation with energy metabolism. Domest Anim Endocrinol. 2005;29(1):3–22.

    Article  CAS  PubMed  Google Scholar 

  59. William Jr WN, Ceddia RB, Curi R. Leptin controls the fate of fatty acids in isolated rat white adipocytes. J Endocrinol. 2002;175(3):735–44.

    Article  CAS  PubMed  Google Scholar 

  60. Block S, Butler W, Ehrhardt R, Bell A, Van Amburgh M, Boisclair Y. Decreased concentration of plasma leptin in periparturient dairy cows is caused by negative energy balance. J Endocrinol. 2001;171(2):339–48.

    Article  CAS  PubMed  Google Scholar 

  61. Holtenius K, Agenäs S, Delavaud C, Chilliard Y. Effects of feeding intensity during the dry period. 2. Metabolic and hormonal responses. J Dairy Sci. 2003;86(3):883–91.

    Article  CAS  PubMed  Google Scholar 

  62. Weber C, Hametner C, Tuchscherer A, Losand B, Kanitz E, Otten W, et al. Variation in fat mobilization during early lactation differently affects feed intake, body condition, and lipid and glucose metabolism in high-yielding dairy cows. J Dairy Sci. 2013;96(1):165–80.

    Article  CAS  PubMed  Google Scholar 

  63. Ehrhardt RA, Foskolos A, Giesy SL, Wesolowski SR, Krumm CS, Butler WR, et al. Increased plasma leptin attenuates adaptive metabolism in early lactating dairy cows. J Endocrinol. 2016;229(2):145–57.

    Article  CAS  PubMed  Google Scholar 

  64. Schwartz DR, Lazar MA. Human resistin: Found in translation from mouse to man. Trends Endocrinol Metab. 2011;22(7):259–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Ding Q, White SP, Ling C, Zhou W. Resistin and cardiovascular disease. Trends Cardiovasc Med. 2011;21(1):20–7.

    Article  PubMed  CAS  Google Scholar 

  66. Reverchon M, Ramé C, Cognié J, Briant E, Elis S, Guillaume D, et al. Resistin in dairy cows: plasma concentrations during early lactation, expression and potential role in adipose tissue. PLoS One. 2014;9(3):e93198.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Weber M, Locher L, Huber K, Kenéz Á, Rehage J, Tienken R, et al. Longitudinal changes in adipose tissue of dairy cows from late pregnancy to lactation. Part 1: The adipokines apelin and resistin and their relationship to receptors linked with lipolysis. J Dairy Sci. 2016;99(2):1549–59.

    Article  CAS  PubMed  Google Scholar 

  68. Cheng J, Song Z-Y, Pu L, Yang H, Zheng J-M, Zhang Z-Y, et al. Retinol binding protein 4 affects the adipogenesis of porcine preadipocytes through insulin signaling pathways. Biochem Cell Biol. 2013;91(4):236–43.

    Article  CAS  PubMed  Google Scholar 

  69. Klöting N, Graham TE, Berndt J, Kralisch S, Kovacs P, Wason CJ, et al. Serum retinol-binding protein is more highly expressed in visceral than in subcutaneous adipose tissue and is a marker of intra-abdominal fat mass. Cell Metab. 2007;6(1):79–87.

    Article  PubMed  CAS  Google Scholar 

  70. Romacho T, Elsen M, Röhrborn D, Eckel J. Adipose tissue and its role in organ crosstalk. Acta Physiol (Oxf). 2014;210(4):733–53.

    Article  CAS  Google Scholar 

  71. Abd Eldaim MA, Kamikawa A, Soliman MM, Ahmed MM, Okamatsu-Ogura Y, Terao A, et al. Retinol binding protein 4 in dairy cows: its presence in colostrum and alteration in plasma during fasting, inflammation, and the peripartum period. J Dairy Res. 2010;77(01):27–32.

    Article  CAS  PubMed  Google Scholar 

  72. Rezamand P, Watts JS, Hunt KM, Bradford BJ, Mamedova LK, Morey SD. Bovine hepatic and adipose retinol-binding protein gene expression and relationship with tumor necrosis factor-α. J Dairy Sci. 2012;95(12):7097–104.

    Article  CAS  PubMed  Google Scholar 

  73. Granneman JG, Li P, Zhu Z, Lu Y. Metabolic and cellular plasticity in white adipose tissue I: effects of β3-adrenergic receptor activation. Am J Physiol Endocrinol Metab. 2005;289(4):E608–16.

    Article  CAS  PubMed  Google Scholar 

  74. Martinez-Santibañez G, Nien-Kai Lumeng C. Macrophages and the regulation of adipose tissue remodeling. Annu Rev Nutr. 2014;34(1):57–76.

    Article  PubMed  CAS  Google Scholar 

  75. Pope BD, Warren CR, Parker KK, Cowan CA. Microenvironmental control of adipocyte fate and function. Trends Cell Biol. 2016;26(10):745–55.

    Article  CAS  PubMed  Google Scholar 

  76. Bindlish S, Presswala LS, Schwartz F. Lipodystrophy: syndrome of severe insulin resistance. Postgrad Med. 2015;127(5):511–6.

    Article  PubMed  Google Scholar 

  77. Ampem G, Azegrouz H, Bacsadi Á, Balogh L, Schmidt S, Thuróczy J et al. Adipose tissue macrophages in non-rodent mammals: a comparative study. Cell Tissue Res. 2016;363:461. doi:10.1007/s00441-015-2253-1.

  78. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112(12):1796–808.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 2009;15(8):914–20.

    Article  CAS  PubMed  Google Scholar 

  80. Mathis D. Immunological goings-on in visceral adipose tissue. Cell Metab. 2013;17(6):851–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Contreras GA, Kabara E, Brester J, Neuder L, Kiupel M. Macrophage infiltration in the omental and subcutaneous adipose tissues of dairy cows with displaced abomasum. J Dairy Sci. 2015;98(9):6176–87.

    Article  CAS  PubMed  Google Scholar 

  82. Contreras G, Thelen K, Schmidt S, Strieder-Barboza C, Preseault C, Raphael R, et al. Adipose tissue remodeling in late-lactation dairy cows during feed restriction-induced negative energy balance. J Dairy Sci. 2016;99(12):10009–21.

    Article  CAS  PubMed  Google Scholar 

  83. Lee Y-H, Petkova A, Granneman J. Identification of an adipogenic niche for adipose tissue remodeling and restoration. Cell Metab. 2013;18(3):355–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Faulconnier Y, Chilliard Y, Torbati MBM, Leroux C. The transcriptomic profiles of adipose tissues are modified by feed deprivation in lactating goats. Comp Biochem Physiol Part D Genomics Proteomics. 2011;6(2):139–49.

    Article  CAS  PubMed  Google Scholar 

  85. Velloso LA, Folli F, Saad MJ. TLR4 at the crossroads of nutrients, gut microbiota, and metabolic inflammation. Endocr Rev. 2015;36(3):245–71.

    Article  CAS  PubMed  Google Scholar 

  86. Grant RW, Stephens JM. Fat in flames: influence of cytokines and pattern recognition receptors on adipocyte lipolysis. Am J Physiol Endocrinol Metab. 2015;309(3):9.

    Article  CAS  Google Scholar 

  87. Suganami T, Tanimoto-Koyama K, Nishida J, Itoh M, Yuan X, Mizuarai S, et al. Role of the toll-like receptor 4/NF-κB pathway in saturated fatty acid–induced inflammatory changes in the interaction between adipocytes and macrophages. Arterioscler Thromb Vasc Biol. 2007;27(1):84–91.

    Article  CAS  PubMed  Google Scholar 

  88. Wisse BE. The inflammatory syndrome: the role of adipose tissue cytokines in metabolic disorders linked to obesity. J Am Soc Nephrol. 2004;15(11):2792–800.

    Article  CAS  PubMed  Google Scholar 

  89. Gao D, Madi M, Ding C, Fok M, Steele T, Ford C, et al. Interleukin-1beta mediates macrophage-induced impairment of insulin signaling in human primary adipocytes. Am J Physiol Endocrinol Metab. 2014;307(3):10.

    Article  CAS  Google Scholar 

  90. Shaul ME, Bennett G, Strissel KJ, Greenberg AS, Obin MS. Dynamic, M2-like remodeling phenotypes of CD11c + adipose tissue macrophages during high-fat diet–induced obesity in mice. Diabetes. 2010;59(5):1171–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. 2012;122(3):787–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Chang HY, Lee HN, Kim W, Surh YJ. Docosahexaenoic acid induces M2 macrophage polarization through peroxisome proliferator-activated receptor gamma activation. Life Sci. 2015;120:39–47.

    Article  CAS  PubMed  Google Scholar 

  93. Chazaud B. Macrophages: Supportive cells for tissue repair and regeneration. Immunobiology. 2014;219(3):172–8.

    Article  CAS  PubMed  Google Scholar 

  94. Osborn O, Olefsky JM. The cellular and signaling networks linking the immune system and metabolism in disease. Nat Med. 2012;18(3):363–74.

    Article  CAS  PubMed  Google Scholar 

  95. Aylward BAA, Clark M, Barnard A, Jen W, Gittens C, Gressley T, et al. Immune cells populate mesenteric adipose tissues of Holstein Friesian cows. J Dairy Sci. 2015;98(Sppl. 2):292.

    Google Scholar 

  96. Spalding KL, Arner E, Westermark PO, Bernard S, Buchholz BA, Bergmann O, et al. Dynamics of fat cell turnover in humans. Nature. 2008;453(7196):783–7.

    Article  CAS  PubMed  Google Scholar 

  97. Lee Y-H, Petkova A, Mottillo E, Granneman J. In vivo identification of bipotential adipocyte progenitors recruited by β3-adrenoceptor activation and high-fat feeding. Cell Metab. 2012;15(4):480–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Häussler S, Germeroth D, Friedauer K, Akter SH, Dänicke S, Sauerwein H. Characterization of the dynamics of fat cell turnover in different bovine adipose tissue depots. Res Vet Sci. 2013;95(3):1142–50.

    Article  PubMed  CAS  Google Scholar 

  99. Gustafson B, Hedjazifar S, Gogg S, Hammarstedt A, Smith U. Insulin resistance and impaired adipogenesis. Trends Endocrinol Metab. 2015;26(4):193–200.

    Article  CAS  PubMed  Google Scholar 

  100. Mariman ECM, Wang P. Adipocyte extracellular matrix composition, dynamics and role in obesity. Cell Mol Life Sci. 2010;67(8):1277–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Khan T, Muise ES, Iyengar P, Wang ZV, Chandalia M, Abate N, et al. Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol Cell Biol. 2009;29(6):1575–91.

    Article  CAS  PubMed  Google Scholar 

  102. Divoux A, Tordjman J, Lacasa D, Veyrie N, Hugol D, Aissat A, et al. Fibrosis in human adipose tissue: composition, distribution, and link with lipid metabolism and fat mass loss. Diabetes. 2010;59(11):2817–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Varma V, Yao-Borengasser A, Bodles AM, Rasouli N, Phanavanh B, Nolen GT, et al. Thrombospondin-1 Is an adipokine associated with obesity, adipose inflammation, and insulin resistance. Diabetes. 2008;57(2):432–9.

    Article  CAS  PubMed  Google Scholar 

  104. Guglielmi V, Cardellini M, Cinti F, Corgosinho F, Cardolini I, D’Adamo M, et al. Omental adipose tissue fibrosis and insulin resistance in severe obesity. Nutr Diabetes. 2015;5(8):e175.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Dahlman I, Mejhert N, Linder K, Agustsson T, Mutch DM, Kulyte A, et al. Adipose tissue pathways involved in weight loss of cancer cachexia. Br J Cancer. 2010;102(10):1541–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Tsoli M, Schweiger M, Vanniasinghe AS, Painter A, Zechner R, Clarke S, et al. Depletion of white adipose tissue in cancer cachexia syndrome is associated with inflammatory signaling and disrupted circadian regulation. PLoS One. 2014;9(3):e92966.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Akbar H, Cardoso FC, Meier S, Burke C, McDougall S, Mitchell M, et al. Postpartal subclinical endometritis alters transcriptome profiles in liver and adipose tissue of dairy cows. Bioinform Biol Insights. 2014;8:45–63.

    PubMed  PubMed Central  Google Scholar 

  108. Sumner-Thomson JM, Vierck JL, McNamara JP. Differential expression of genes in adipose tissue of first-lactation dairy cattle. J Dairy Sci. 2011;94(1):361–9.

    Article  CAS  PubMed  Google Scholar 

  109. Bradford BJ, Yuan K, Farney JK, Mamedova LK, Carpenter AJ. Invited review: Inflammation during the transition to lactation: New adventures with an old flame. J Dairy Sci. 2015;98(10):6631–50.

    Article  CAS  PubMed  Google Scholar 

  110. Lumeng CN. Innate immune activation in obesity. Mol Aspects Med. 2013;34(1):12–29.

    Article  CAS  PubMed  Google Scholar 

  111. Namgaladze D, Brüne B. Macrophage fatty acid oxidation and its roles in macrophage polarization and fatty acid-induced inflammation. Biochim Biophys Acta. 2016;1861(11):1796–807.

    Article  CAS  PubMed  Google Scholar 

  112. Scalia D, Lacetera N, Bernabucci U, Demeyere K, Duchateau L, Burvenich C. In vitro effects of nonesterified fatty acids on bovine neutrophils oxidative burst and viability1. J Dairy Sci. 2006;89(1):147–54.

    Article  CAS  PubMed  Google Scholar 

  113. Raphael W, Halbert L, Contreras GA, Sordillo LM. Association between polyunsaturated fatty acid-derived oxylipid biosynthesis and leukocyte inflammatory marker expression in periparturient dairy cows. J Dairy Sci. 2014;97(6):3615–25.

    Article  CAS  PubMed  Google Scholar 

  114. Sun F, Cao Y, Cai C, Li S, Yu C, Yao J. Regulation of nutritional metabolism in transition dairy cows: energy homeostasis and health in response to post-ruminal choline and methionine. PLoS One. 2016;11(8):e0160659.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Pires JAA, Pescara JB, Brickner AE, Silva del Rio N, Cunha AP, Grummer RR. Effects of abomasal infusion of linseed oil on responses to glucose and insulin in Holstein Cows. J Dairy Sci. 2008;91(4):1378–90.

    Article  CAS  PubMed  Google Scholar 

  116. Salin S, Taponen J, Elo K, Simpura I, Vanhatalo A, Boston R, et al. Effects of abomasal infusion of tallow or camelina oil on responses to glucose and insulin in dairy cows during late pregnancy. J Dairy Sci. 2012;95(7):3812–25.

    Article  CAS  PubMed  Google Scholar 

  117. Ospina PA, Nydam DV, Stokol T, Overton TR. Associations of elevated nonesterified fatty acids and β-hydroxybutyrate concentrations with early lactation reproductive performance and milk production in transition dairy cattle in the northeastern United States. J Dairy Sci. 2010;93(4):1596–603.

    Article  CAS  PubMed  Google Scholar 

  118. Hernandez TL, Van Pelt RE, Anderson MA, Reece MS, Reynolds RM, de la Houssaye BA, et al. Women with gestational diabetes mellitus randomized to a higher–complex carbohydrate/low-fat diet manifest lower adipose tissue insulin resistance, inflammation, glucose, and free fatty acids: a pilot study. Diabetes Care. 2016;39(1):39–42.

    Article  PubMed  Google Scholar 

  119. Capurso C, Capurso A. From excess adiposity to insulin resistance: The role of free fatty acids. Vascul Pharmacol. 2012;57(2–4):91–7.

    Article  CAS  PubMed  Google Scholar 

  120. Sadri H, Bruckmaier RM, Rahmani HR, Ghorbani GR, Morel I, Van Dorland HA. Gene expression of tumour necrosis factor and insulin signalling-related factors in subcutaneous adipose tissue during the dry period and in early lactation in dairy cows. J Anim Physiol Anim Nutr (Berl). 2010;94(5):e194–202.

    Article  CAS  Google Scholar 

  121. Schmitt E, Ballou MA, Correa MN, DePeters EJ, Drackley JK, Loor JJ. Dietary lipid during the transition period to manipulate subcutaneous adipose tissue peroxisome proliferator-activated receptor-γ co-regulator and target gene expression. J Dairy Sci. 2011;94(12):5913–25.

    Article  CAS  PubMed  Google Scholar 

  122. Zachut M. Defining the adipose tissue proteome of dairy cows to reveal biomarkers related to peripartum insulin resistance and metabolic status. J Proteome Res. 2015;14(7):2863–71.

    Article  CAS  PubMed  Google Scholar 

  123. Mann S, Nydam DV, Abuelo A, Leal Yepes FA, Overton TR, Wakshlag JJ. Insulin signaling, inflammation, and lipolysis in subcutaneous adipose tissue of transition dairy cows either overfed energy during the prepartum period or fed a controlled-energy diet. J Dairy Sci. 2016;99(8):6737–52.

    Article  CAS  PubMed  Google Scholar 

  124. Oliveira LH, Nascimento AB, Monteiro Jr PLJ, Guardieiro MM, Wiltbank MC, Sartori R. Development of insulin resistance in dairy cows by 150 days of lactation does not alter oocyte quality in smaller follicles. J Dairy Sci. 2016;99(11):9174–83.

    Article  CAS  PubMed  Google Scholar 

  125. Mordak R, Stewart PA. Periparturient stress and immune suppression as a potential cause of retained placenta in highly productive dairy cows: examples of prevention. Acta Vet Scand. 2015;57(1):1–8.

    Article  CAS  Google Scholar 

  126. Harvatine KJ, Perfield JW, Bauman DE. expression of enzymes and key regulators of lipid synthesis is upregulated in adipose tissue during CLA-induced milk fat depression in dairy cows. J Nutr. 2009;139(5):849–54.

    Article  CAS  PubMed  Google Scholar 

  127. Zhu P, Goh Yan Y, Chin Hwee Fang A, Kersten S, Tan Nguan S. Angiopoietin-like 4: a decade of research. Biosci Rep. 2012;32(3):211–9.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Authors acknowledge Sarah E. Schmidt for assistance with editing the manuscript.

Funding

This project was supported in part by USDA-NIFA (Washington, DC) grants 2014-68004-21972 and 2015-67015-23207, Department of Large Animal Clinical Sciences (East Lansing, MI), the Michigan State University Elwood Kirkpatrick Dairy Science Research Endowment (East Lansing, MI), Michigan Alliance for Animal Agriculture (East Lansing, Michigan), and Michigan Animal Health Foundation.

Availability of data and materials

Data sharing not applicable to this article as no datasets were generated or analyzed in this review article.

Authors’ contributions

All authors read and approved the final manuscript. GAC, CSB, and WR reviewed literature available and wrote the manuscript.

Competing interests

The authors declare that they have no competing interest.

Consent for publication

Not applicable.

Ethics approval and consent to participate

Not applicable.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to G. Andres Contreras.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Contreras, G.A., Strieder-Barboza, C. & Raphael, W. Adipose tissue lipolysis and remodeling during the transition period of dairy cows. J Animal Sci Biotechnol 8, 41 (2017). https://doi.org/10.1186/s40104-017-0174-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40104-017-0174-4

Keywords