Next Article in Journal
Editorial: ‘Engineering the Tumor Immune Microenvironment’ Special Issue
Previous Article in Journal
YY1 Knockdown Relieves the Differentiation Block and Restores Apoptosis in AML Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Challenges in Pharmacological Intervention in Perilipins (PLINs) to Modulate Lipid Droplet Dynamics in Obesity and Cancer

by
Victória Bombarda-Rocha
1,2,
Dany Silva
1,2,
Allal Badr-Eddine
1,
Patrícia Nogueira
1,2,
Jorge Gonçalves
1,2,* and
Paula Fresco
1,2
1
Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
2
UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(15), 4013; https://doi.org/10.3390/cancers15154013
Submission received: 29 June 2023 / Revised: 1 August 2023 / Accepted: 4 August 2023 / Published: 7 August 2023
(This article belongs to the Section Cancer Pathophysiology)

Abstract

:

Simple Summary

This review highlights the importance of perilipins in lipid metabolism and their potential as therapeutic targets for lipid-associated diseases like cancer and obesity. Perilipins are proteins found in lipid droplets that regulate lipase activity and play a crucial role in maintaining the balance between lipid synthesis and breakdown. Modulating perilipins could improve existing treatments or offer new therapeutic opportunities for addressing these diseases.

Abstract

Perilipins (PLINs) are the most abundant proteins in lipid droplets (LD). These LD-associated proteins are responsible for upgrading LD from inert lipid storage structures to fully functional organelles, fundamentally integrated in the lipid metabolism. There are five distinct perilipins (PLIN1–5), each with specific expression patterns and metabolic activation, but all capable of regulating the activity of lipases on LD. This plurality creates a complex orchestrated mechanism that is directly related to the healthy balance between lipogenesis and lipolysis. Given the essential role of PLINs in the modulation of the lipid metabolism, these proteins can become interesting targets for the treatment of lipid-associated diseases. Since reprogrammed lipid metabolism is a recognized cancer hallmark, and obesity is a known risk factor for cancer and other comorbidities, the modulation of PLINs could either improve existing treatments or create new opportunities for the treatment of these diseases. Even though PLINs have not been, so far, directly considered for pharmacological interventions, there are many established drugs that can modulate PLINs activity. Therefore, the aim of this study is to assess the involvement of PLINs in diseases related to lipid metabolism dysregulation and whether PLINs can be viewed as potential therapeutic targets for cancer and obesity.

1. Introduction

In recent years, evidence is being presented to support a major dependence of cancer cells on fatty acid (FA) oxidation to support cell proliferation, survival, stemness, and metastatic progression [1,2,3]. Cancer cells may rely on scavenging the FA extracellular sources of lipids (from neighbor or apoptotic cells) or on endogenous de novo lipid synthesis to face their higher dependence on FA oxidation [4]. To prevent lipotoxicity resulting from the presence of free FA in the cytoplasm, cancer cells need to store these FAs efficiently in compartments that can be easily mobilized according to the cell’s needs [2,4,5,6,7,8]; lipid droplets (LDs) are believed to be such compartments [9].
LDs are nano- to micro-sized organelles, composed of a neutral lipid core surrounded by a phospholipid monolayer [10,11]. For a long time, LDs were considered inert lipid storage organelles but, after the discovery of proteins anchored to their surfaces [12], they became recognized as functional organelles, with key roles in lipid metabolism, energy homeostasis, and communication between distinct cellular signaling pathways [11,13,14].
Perilipins (PLINs), the most abundant proteins in LDs, regulate LD stability and lipid turnover, consequently controlling the overall lipid metabolism [15]. Given the involvement of lipid droplets in cancer pathophysiology [9], understanding the roles of perilipins could help clarify how changes in the lipid metabolism affect cancer cells and shed light on the potential impact of these metabolic alterations on cancer progression. The goal of this study is to examine the primary roles of perilipins, especially in cancer, with the aim of exploring their potential as therapeutic targets for cancer treatment. PLINs play a role in maintaining the equilibrium among various FA stores within the body and contribute to the onset of conditions like obesity and irregular fat accumulation in non-adipose tissues, as seen in liver steatosis and atheroma plaque formation [16]. Given that obesity is a known risk factor for cancer, the link between conditions predisposing to obesity and cancer will also be explored.

2. LD Biogenesis

The biogenesis of LDs mainly takes place within the endoplasmic reticulum (ER), culminating in the creation of a fully functional structure [10]. A schematic representation of this process is depicted in Figure 1.
The LD structure comprises a core of neutral lipids enveloped by a single layer of phospholipids with proteins [10]. The core of neutral lipids is composed of esterified cholesterol or FAs, which can be obtained either through external uptake or via de novo synthesis [17].
The uptake of FA occurs through transporters such as CD36, the fatty acid transport protein family (FATP), and plasma membrane fatty acid-binding proteins (FABPpm) [18]. CD36 is recognized as a multifunctional membrane scavenger receptor [18]. When free FA binds to CD36, the resulting complex is internalized, forming an endosome. This endosome then transports the FA into the cell, possibly directly to the LD, where it is esterified and stored as triacylglycerol (TAG) [19]. FABPs function as enzymes, being able to convert FA into FA-acyl-CoA. The FA-acyl-CoA formed can translocate to the inner side of the membrane (aqueous phase) and bind to the cytoplasmic fatty acid-binding protein (FABPc) [20], which will shuttle FAs to different cellular compartments [21].
De novo lipogenesis, the process of producing new FAs, originates from citrate and acetate, which are produced by pyruvate oxidation via the TCA cycle [22]. This process primarily occurs in hepatocytes and adipocytes under normal physiological conditions [22]. However, this process is reactivated in cancer cells, seemingly as an adaptive strategy to produce large amounts of FA, which can then be lengthened or desaturated for various critical functions of cancer cells survival [22].
Within the ER, FAs are transformed into neutral lipids, specifically in the form of TAG [23]. The synthesis of TAG starts with the activation of FA through the addition of acyl-CoA, a reaction catalyzed by the enzyme acyl-CoA synthetase (ACS) [23]. This reaction produces FA-acyl-CoA, which is then esterified with glycerol-3-phosphate by the enzyme glycerol-P acyltransferase (GPAT), resulting in the formation of lysophosphatidic acid (LPA) [23]. LPA acts as a substrate for acylglycerol-P acyltransferase (AGPAT), which catalyzes the addition of another FA-acyl-CoA, yielding phosphatidic acid (PA) [23]. PA is then used by phosphatidic acid phosphohydrolase (PAP) to create diacylglycerol (DAG), which is finally esterified into TAG by the enzyme diacylglycerol acyltransferase (DGAT) [23]. DGAT, the enzyme responsible for the final step of TAG synthesis, has two isoforms, DGAT1 and DGAT2 [24]. DGAT2, found in LD membranes, handles the esterification of FA within the LD [24]. This isoform forms a complex with another enzyme, monoacylglycerol acyltransferase 2 (MGAT2), which is instrumental in converting monoacylglycerol into DAG [24].
The TAG synthesized by DGAT starts to accumulate between the bilayer membrane of the ER, marking the beginning of LD biogenesis [11,25]. Continuous accumulation of TAG builds lens-like structures which eventually break away, or bud, from the ER [11,25]. This budding process is driven by the interfacial tensions between the LD and the cytosol [25]. This stage requires the involvement of additional proteins that facilitate the expansion of the neutral lipid core and help stabilize the TAG aggregates [25]. The proteins involved in this process primarily consist of fat storage-inducing transmembrane proteins (FIT or FITM) and SEIPIN [26,27]. FIT proteins are localized to the ER, and both of their isoforms, FIT1 and FIT2, participate in LD biogenesis [26,27]. Rather than synthesizing neutral lipids, FIT1 and FIT2 assist in integrating TAG into LDs [26,27]. When FIT proteins are overexpressed, they result in LDs enriched with TAG [26]. SEIPIN, also known as Berardinelli–Seip congenital lipodystrophy type 2 or BSCL2 protein, is a transmembrane protein in the ER and it plays a pivotal role in shaping LD [28]. It is suggested that SEIPIN stabilizes the membrane bridges between the ER and LD, acting as a contact point and enabling the transfer of neutral lipids into LD [29,30]. In the absence of SEIPIN, the budding process is delayed and becomes heterogeneous [31]. In humans, deficiency in SEIPIN can cause lipodystrophy (an abnormal distribution and/or partial loss of adipose tissue), neurological defects, and multi-organ problems [31].
A group of other proteins, the PLINs, also participate in the regulation of the rate of lipolysis in the LD [32]. These proteins are transferred from the Golgi to the ER via the coat complex protein I (COPI) and ADP-ribosylation factor 1 (ARF1) machinery [33]. When LDs complete their formation, they detach from the ER already fully functional and able to toggle between lipogenic and lipolytic pathways as per the cell’s needs [34].
The balance between lipogenesis and lipolysis is directly influenced by the accessibility of lipases to the lipid core of the LD [11]. PLINs play a crucial role in regulating this accessibility and thus are key regulators of lipid metabolism [32].

3. PLINs as LD Gatekeepers

PLINs, also known as PAT proteins, represent the most prevalent proteins found in LD membranes [32]. Five PLINs have been identified in mammals (PLINs 1–5) and all serve as crucial proteins for the stabilization and protection of LDs, responding to either lipogenic or lipolytic stimuli [35]. They function by preventing lipases from accessing the content of LDs [32,35,36,37]. The regulation of PLINs’ efficacy or activity is influenced by both the expression levels of individual variants and the activity of kinases [32]. The latter is dependent on the metabolic state of the cell or the energy requirements of the organism [32]. Given that each PLIN has a unique expression pattern [38], the distribution of lipids throughout the human body is determined by the collective contribution of each PLIN, in accordance with its specific control mechanisms and tissue distribution. To deepen the role of perilipins in systemic lipid distribution, a more comprehensive overview of each perilipin is provided as follows.

3.1. Perilipin 1

PLIN1 is mainly found in mature adipocytes, where it acts as a lipolysis modulator, thereby regulating lipid redistribution from adipose tissues to other organs and tissues [39]. PLIN1 regulates the access of the lipases to the LD content [39]. PLIN1 is bonded to comparative gene identification-58 (CGI-58), a co-activation factor of adipose triglyceride lipase (ATGL), preventing ATGL activation and lipase access to stored TAG [39]. The activity of CGI-58 depends on the energy state of the organism. When in the fed state, CGI-58 remains combined with PLIN1 [39,40]. Conversely, after prolonged fasting, CGI-58 is released from PLIN1, binds with ATGL, and activates ATGL lipase activity [39,40].
The lipolytic pathway of PLIN1 is controlled by receptors linked to the cAMP-dependent protein kinase (PKA) pathway [39]. Phosphorylation of PLIN1 induced by PKA disrupts the bond between PLIN1 and CGI-58, setting free CGI-58 to bind to ATGL; PKA also phosphorylates ATGL, thereby increasing its affinity for free CGI-58 [40]. Additionally, PKA phosphorylation uncovers the hormone-sensitive lipase (HSL) binding site in PLIN1, which allows the lipase to access the diacylglycerol (DAG) and monoacylglycerol (MAG) within the LDs [41,42]. Figure 2 shows a schematic representation of PLIN1 in basal and stimulated states.
PLIN1 is an important regulator of lipolysis in white adipose tissue (WAT) and it is associated with both basal and adrenergic-induced lipolysis [43]. The PLIN1 gene locus has been associated with variability in weight loss [44]. Polymorphisms [45], epigenetic modifications [46], and varying levels of PLIN1 expression [47,48,49] have also been associated with obesity phenotypes and with the success rate of weight loss following bariatric surgeries [50].
In cancer, PLIN1 overexpression has been reported to favor cancer development in tissues which normally express PLIN1 (adipose tissue) [51], being highly expressed in sebaceous adenomas and carcinomas [52] and proposed to be a potential biomarker for liposarcomas [53]. PLIN1 overexpression was also found in hepatic tumors, including hepatocellular adenoma and carcinoma [52]. In breast cancer cells, PLIN1 expression was shown to prevent cancer cell evasion and tumorigenesis and reduced PLIN1 expression was correlated with a poorer prognosis in breast cancer patients [54,55]. These studies seem to indicate the distinct role of PLIN1 in cancer according to the FA management profile of each cell type. In cells that function as FA storage cells (adipocytes and, to some extent, hepatocytes), PLIN1 overexpression may favor tumorigenesis whereas in cells that are end-users of FA (like cancer cells), a poor expression of PLIN1 may limit the cell’s ability to store FA and, hypothetically, force the cell’s evasion to find a place more favorable to have access to energy.

3.2. Perilipin 2

PLIN2, also known as adipophilin, is ubiquitous and continuously expressed [56,57,58]. PLIN2 cell levels are controlled by its rates of synthesis and degradation [56]. PLIN2 binds to the LD membrane and acts as a barrier to lipolysis by preventing the access of the lipases to the lipids stored in the LD, meaning the LD permeability to lipolytic enzymes is proportional to the cellular PLIN2 [32,58,59,60].
Expression of PLIN2 is induced by peroxisome proliferator-activated receptor gamma (PPARγ) activation [61,62,63]. PPARγ activators and high fat diets were reported to increase PLIN2 expression [61,63,64,65,66,67]. PPARγ is predominantly expressed in adipose tissue [68,69]. Therefore, these stimuli may promote the retention of TAG inside the LDs of adipocytes by a PPARγ-induced increase in PLIN2 expression.
The ubiquitin–proteasome pathway consistently degrades expressed PLIN2, a process that involves the E3 ubiquitin ligase (Ubr1) [70,71]. Ubr1 targets PLIN2 for degradation in an amino acid-dependent manner [71]. This increase in Ubr1 activity results in enhanced ubiquitination of PLIN2, which facilitates the access of lipolytic enzymes to the LD core. In turn, this leads to an elevated bioavailability of FA for β-oxidation, thereby fueling the increase in the cell’s anabolic activity triggered by the availability of amino acids [72]. Reciprocally, PLIN2 overexpression was shown to refrain protein synthesis translated in decreased muscle thickness and strength [72].
PLIN2 bound to the LD membrane is degraded by chaperone-mediated autophagy (CMA), as illustrated in Figure 3 [73,74]. CMA involves a coordinate action of heat-shock cognate protein of 70 kDa (Hsc70) and lysosome-associated membrane protein type 2A (LAMP-2A), which are also present in LDs [75]. CMA of PLIN2 starts with the binding of PLIN2 to Hsc70, which interacts thereafter with LAMP-2A, culminating with PLIN2 degradation [76].
CMA of PLIN2 may be increased by its phosphorylation by AMP-activated protein kinase (AMPK) [74]. Under conditions of starvation, specifically when glucose is low, ATP concentrations will drop, increasing the AMP/ATP ratio [74]. This elevated ratio is detected by starvation sensors like AMPK, which promote CMA of PLIN2 [74]. This degradation allows lipolytic enzymes access to the content of LDs, resulting in the mobilization of FA as alternative energy sources to counteract starvation [74,77]. However, the response to an increase in AMP/ATP ratio seems to be context-dependent, as is demonstrated by the alterations triggered by hypoxia.
Hypoxia induces an extensive metabolic reprogramming of the cell to face the decrease in nutrients and oxygen supply. The metabolic reprogramming is coordinated by the hypoxia-inducible factors (HIF) 1-alpha (HIF-1α) and 2-alpha (HIF-2α) [78]. Under normoxic conditions, HIF-1α and HIF-2α are continuously ubiquitinated for proteasomal degradation [79,80]. Hypoxia prevents HIFs degradation and allows their translocation to the nucleus, where they combine with HIF-1β altering gene transcription so the cell can endure hypoxic conditions [78]. The PLIN2 gene, also known as ADRP (Adipose Differentiation-Related Protein) gene, is one of the genes induced by HIF-1α and by HIF-2α [81,82]. Additionally, HIF-1α also induces the expression of hypoxia-inducible lipid droplet-associated (hilpda), the gene that encodes the hypoxia-inducible protein 2 (HIG-2) [83] whose function is the inhibition of ATGL-mediated lipolysis [84,85]. Hypoxia ends up promoting an increase in lipid storage in LDs, by increasing PLIN2 and inhibiting the activity of lipolytic enzymes.
The interplay between PLIN2 and hypoxia seems to be particularly relevant as PLIN2 has been shown to be involved in a positive loop that further inhibits the degradation of HIF-1α and sustains its expression and that of hilpda and, therefore, the increase in LD load promoted by hypoxia [86]. The pathophysiological consequences of this loop may be vast and are far from being totally understood as hypoxia or “pseudo-hypoxic conditions” (i.e., conditions that prevent HIF-1α and HIF-2α degradation even in the presence of oxygen), and may be relevant in promoting obesity [87,88] or cancer [89]. The association between PLIN2 and cancer is extensively documented. PLIN2 has been associated with tumorigenesis and is often considered as a poor prognosis indicator in cancers of the colon [90,91], breast [92,93], prostate [94,95], lung [96,97], bladder [95], kidney [82,98,99,100,101,102,103,104,105], thyroid [106], gastric [107], and melanoma [108].
PLIN2 was also shown to be involved in the interaction between LDs and the plasma membrane [109,110,111], and to play a pivotal role in both the formation and secretion of milk lipids [112]. Therefore, PLIN2 may also participate in intercellular signaling. PLIN2 was shown to act as an adipokine [113,114,115], promoting the activation of the inflammatory and fibrotic processes of progressive liver injury [116], the development of atherosclerotic arterial plaques in the cardiovascular system [117], and alterations of the phenotype of microglia and of other macrophages towards a more inflammatory phenotype in the brain [118]. The alterations in the phenotype of brain inflammatory cells induced by PLIN2 have been related to neuroinflammation and oxidative stress [118], neurodegeneration [119], the suppression of remyelination [120,121], cognitive impairment [122] and accelerated aging [119,123].

3.3. Perilipin 3

PLIN3, also known as TIP47, is ubiquitously expressed [124]. During the nucleation phase of LD biogenesis, PLIN3 is mobilized to the nascent LD [35,125]. Over time, PLIN3 may be progressively replaced by PLIN2 [35,126]. Functionally, PLIN3 behaves much like PLIN2, restricting lipases’ access to the LD core. Its degradation is also mediated by CMA and regulated by AMPK [73].
PLIN3 seems to play a pivotal role in promoting the transition from brown to WAT and, consequently, in obesity. The depletion of PLIN3 in WAT triggers the generation of brown adipocytes and the expression of genes responsible for thermogenesis [127].
PLIN3 also appears to play a significant role in cancer pathogenesis. Studies have reported abnormal PLIN3 expression in several types of cancer, including cervical [128], clear cell renal cell [129], breast [93], lung [130], and prostate [131,132,133]. The prevalence of high PLIN3 levels in these cancer types could suggest an increased rate of LD biogenesis or potentially hindered conditions for PLIN3′s CMA. It is noteworthy that the upregulation of PLIN3 expression has been observed to result in decreased efficacy of enzalutamide treatment and radiotherapy (thus increasing resistance) [131,133]. On the contrary, it has been demonstrated that PLIN3 can enhance the therapeutic efficacy of docetaxel by reducing resistance to therapy [132]. These seemingly contradictory effects might shed light on intricate aspects of cancer metabolism, underscoring the proposed significance of LD in both cellular metabolism (availability of cholesterol for androgen synthesis and of FA to compensate blockade of endocytic pathways) and in stress management (protecting the cells from the stress induced by the lipid peroxidation caused by radiotherapy) [134].

3.4. Perilipin 4

PLIN4, also known as S3-12, is the least studied protein of the perilipin family [135]. It is mostly identified in preadipocytes and in membranes of nascent LDs [135,136]. It is suggested that PLIN4 is involved with adipocyte differentiation and, to promote stability of LD membrane, acts as a surfactant [59]. PLIN4 location has also been correlated to cholesterol ester rich LDs [137,138] and its expression is also induced by PPARγ activation [61].
The depletion of PLIN4 has been linked to diminished PLIN5 expression in the heart and a reduction in heart LDs. This suggests that the functions of PLIN4 and PLIN5 may be closely interconnected [139].
The most solid evidence pointing to a relationship between PLIN4 and obesity was provided by the observation that single nucleotide polymorphisms of PLIN4 were correlated with obese phenotypes [140]. In cancer, PLIN4 is highly expressed in luminal A and B breast carcinomas [93] and its expression is associated with triple-negative breast cancer resistance to cytotoxic chemotherapy [141].

3.5. Perilipin 5

PLIN5, also known as OXPAT, predominantly occurs in tissues that rely on β-oxidation for energy generation, such as skeletal muscle, cardiac muscle, and brown adipose tissue [142]. Its expression has also been documented in the epithelial cells of the gastrointestinal and urogenital tracts, hepatocytes, renal tubular cells, ductal cells of the salivary glands, and pancreatic cells [143].
PLIN5 is found at the contact site between LDs and mitochondria [32,37]. The relevance of such location raises the question of whether LD–mitochondria contact sites exist to promote the transfer of FAs from LDs to mitochondria, or to shield the cell from an overload of FAs that the mitochondria cannot process [32,37]. Findings that PLIN5 overexpression induces cardiac steatosis, and that PLIN5 ablation reduces cardiac LD formation [144], suggest that PLIN5 plays a role in a cellular adaptive response to high lipid oxidative metabolism [144,145,146,147]. These observations that support the protective role of PLIN5 from excessive FA load are further supported by the observation that PLIN5 is regulated by AMPK and that AMPK activation increases PLIN5 expression and LD formation, and mitigates cellular oxidative stress by lowering the levels of reactive oxygen species in hepatic stellate cells [146]. However, PLIN5 has also been shown to interact with both CGI-58 and ATGL, inhibiting the lipolytic activity of ATGL through a dual mechanism regulated by PKA [148,149], as illustrated in Figure 4. Any stimuli that result in PKA activation—such as cold exposure, physical activity, fasting, or other stress factors—will induce PLIN5 phosphorylation. PLIN5 phosphorylation leads to its detachment from CGI-58 and ATGL, subsequently activating ATGL’s lipolytic activity [144]. Besides its role in regulating access to LD reserves, PLIN5, when phosphorylated by PKA, translocate to the nucleus. There, it interacts with sirtuin 1 (SIRT1), activates peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), and stimulates the transcription of genes of FA catabolism, mitochondrial biogenesis, and respiration [144]. Through this PKA-mediated regulation, PLIN5 not only facilitates FA oxidation but also increases the cell’s FA oxidative capacity [144,149].
Therefore, the function of PLIN5 seems to participate in a delicate balance to preserve lipid homeostasis [149]. It favors β-oxidation metabolism by allowing the release of FA and promoting the synthesis of the required enzymatic machinery when the cell needs to meet a higher energy demand [149]. Yet, it is also capable of preventing excessive lipolysis, shielding the cell against lipotoxicity when the concentration of FAs in the cytoplasm exceeds the amount that mitochondria can handle, or when signs of oxidative stress are present [149].
In the context of cancer, the overexpression of PLIN5 has been observed in patients with breast cancer, renal carcinomas, liposarcoma, rhabdomyosarcoma, and leiomyosarcoma [93,143]. It is also significantly prevalent in hepatocellular carcinomas [150], where it serves as a biomarker linked to unfavorable prognosis for patients suffering from this type of liver cancer, and it has been associated with the evolution of non-alcoholic fatty liver disease to liver cancer [151].

4. Coordination of PLINs’ Function

The diverse roles and tissue distribution of PLINs raise the question about how their actions are coordinated to act both as gatekeepers of LD and to allow the release of LD contents when required [32]. The orchestration of PLINs’ function can be achieved through the concerted action systems that monitor the availability of energy and nutrients for metabolic processes [38].
Peroxisome proliferator-activated receptors (PPARs) are transcriptional factors that control the expression of genes that code for proteins involved in FA storage, glucose metabolism, and adipocyte differentiation [152], including PLINs, as described above [142,153,154]. Out of the three known PPAR isoforms (α, β/δ, and γ), PPARγ and PPARα have the most significant impact on the function of PLINs. PPARγ is ubiquitously expressed but the tissue distribution can vary among its three mRNA splice variants: PPARγ1, PPARγ2, and PPAγ3. PPARγ1 is also ubiquitously expressed, while PPARγ2 is predominantly found in adipose tissues and the liver and PPARγ3 is mainly expressed in the colon and adipose tissue [155]. On the other hand, PPARα is primarily expressed in skeletal muscle, brown adipose tissue, and the liver, playing a key role in the regulation of β-oxidation [156].
PPARγ activity can be influenced by substances like polyunsaturated FAs, eicosanoids, and oxidized lipid components [157]. Elevated levels of FAs activate PPARγ, which in turn boosts the loading capacity of LDs. This boost operates through a dual mechanism: first, by promoting adipocyte differentiation, which increases the number of cells capable of storing FAs in LDs and, second, by directly enhancing LD loading [158].
Two specific mechanisms seem to underpin the enhancement of LD loading mediated by PPARγ. One is the upregulation of PLIN1 expression, which improves the capacity to restrict lipolytic enzymes from acting primarily on LDs in adipocytes [153]. The other involves an increase in PLIN2 expression, which bolsters the ability to block lipolytic enzymes’ access to the LD core [61,63,64,65,66,67].
In addition to the influence on PPARγ, free FAs also stimulate PPARα, resulting in increased β-oxidation in skeletal muscle and the liver [156,159]. Thus, when free FA levels rise, PLINs in adipose tissue, liver, and other target organs coordinate their activities to promote a balanced redistribution of FAs between the adipose tissue and the target organs. This coordination allows these tissues to expand their capacity to store more FAs within LDs and to utilize more FAs for β-oxidation.
Any stimulus that activates PKA can lead to PLIN1 phosphorylation by PKA [39]. PLIN1 phosphorylation inhibits its ability to restrict the access of lipolytic enzymes to the LD’s core in adipocytes, which in turn increases the levels of FAs in the plasma [39]. These FAs can activate PPARs [159], triggering a process like the one described above.
The key distinction between PKA-induced regulation and PPARγ-induced regulation lies in the effect on adipocytes’ ability to store FAs in LDs. Under PKA regulation, this ability is diminished due to PKA’s action on PLIN1 [39]. This results in an imbalanced distribution of FAs, causing a preferential flow from the adipose tissue to the target organs. Furthermore, PKA modulates PLIN5, which also facilitates the transfer of FAs to mitochondria. This not only enhances the use of FAs as energy substrates but also creates an FA gradient to favor the mobilization of FAs from adipose tissues.
In situations where a cell has the necessary conditions for proliferation (such as the availability of sufficient amino acids), this mechanism may also be triggered. By sensing the abundance of these anabolic components, the cell promotes PLIN2 degradation as well [72]. This process increases the available energy sources to meet the heightened demands imposed by anabolic activity [72].
This coordinated regulation can fail if either the sensitivity of PLIN1 to phosphorylation decreases or the control over PLIN2 degradation lessens. In such cases, obesity or ectopic fat deposits may occur, leading to dysfunctions in lipid metabolism and the potential development of associated diseases [116]. In fact, studies have shown that the absence of PLIN2 in mice fed a high-fat diet (HFD) prevented HFD-induced obesity [116]. This outcome was associated with an increase in the formation of subcutaneous beige adipocytes expressing uncoupling protein 1 (UCP-1), as well as reduced formation of inflammatory foci in WAT and the reduction of liver steatosis [116]. Moreover, it was also observed that a loss of PLIN2 resulted in reduced energy intake and increased physical activity in mice subjected to HFD feeding [116].
In the context of cancer, the overexpression of PLIN2 and PLIN3 is often observed [90,91,92,93,95,96,99,101,105,106,107,108,128,129,130,131,132,133,160,161,162,163,164]. Considering their individual roles, this suggests that cancer cells might possess an enhanced ability to generate new LDs, as PLIN3 typically associates with nascent lipid droplets, and a heightened capability for lipid storage, since overexpression of PLIN2 could make lipid droplets less accessible to lipolytic enzymes [35]. Therefore, the overexpression of PLIN3 and PLIN2 in cancer cells could extend beyond an augmented generation of lipid droplets, as it may also denote heightened efficacy in lipid preservation. This could provide a competitive advantage over ‘normal’ cells that do not overexpress PLIN3 and PLIN2, such as adipocytes.
Several prevalent types of cancer, such as breast, colorectal, esophageal, kidney, gallbladder, uterine, pancreatic, and liver, have been associated with obesity, which also increases the likelihood of cancer mortality [165,166]. The population-attributable fraction, a measure of the burden of cancer attributable to obesity, is 11.9% for men and 13.1% for women [165]. Proximity to adipose tissue has been observed to favor the growth or development of metastasis in many epithelial tumors [167,168].
Adipocytes and cancer cells appear to establish a metabolic symbiosis, transforming adipocytes into cancer-associated adipocytes and promoting cancer growth [167]. This transformation involves the mobilization of FAs from adipocytes, a process that contributes to tumor progression [168]. Various factors likely play a role in this mobilization, including alterations in the expression and activity of PLINs. The coordination of PLINs by both PPARs and PKA pathways may be particularly relevant.
Adrenaline is the endogenous ligand of β2-adrenoceptors, which are prototypically coupled to the Gs/cAMP/PKA pathway [169]. Both our own research [170,171] and that of others [172,173] have demonstrated that tumorigenic cancer cells have the capacity to synthesize and release adrenaline, creating conditions for adrenergic activation in the tumor microenvironment independent of sympathetic or hormonal stimulation. Therefore, the presence of this PKA regulation, which enables the release of adipocyte LD control by PLIN1, combined with the increased capacity of PLIN2 to retain FAs in the LDs of cancer cells via the FA/PPARγ pathway, shifts the adipocyte/cancer cell balance in favor of FA retention by cancer cells.
Ultimately, the accumulation of FAs in the LDs of cancer cells, along with their ability to mobilize FAs from adipocytes in the tumor microenvironment, may enhance the ability of cancer cells to resist the starvation conditions signaled by AMPK activation [2].

5. Pharmacological Interventions

The significance of PLINs in regulating lipid metabolism is undeniable. However, they have not yet been prioritized as targets for pharmacological intervention. Nevertheless, there are a variety of drug categories, some of which are approved for clinical use, that operate on the regulators of PLIN function or expression. This repertoire includes PPAR activators [174], SIRT1 [175,176], and AMPK activators [177], as well as drugs that interact with receptors linked to the Gs/cAMP/PKA pathway [178].
These regulatory pathways exert unique influences on different PLINs [41,142,146,149,153,154]. Therefore, their impact on the coordination of PLIN functions and on the allocation of FAs between adipocytes and recipient cells may also be distinct. A comparative analysis of these different scenarios will be undertaken, juxtaposing theoretical expectations with empirical data concerning the established effects of these drugs on obesity and cancer risk. Figure 5 illustrates the coordination of perilipins, with a focus on PLINs 1, 2 and 5, and possible pharmacological interventions.

5.1. PPAR Activators

As previously described, PPARs play a significant role in maintaining nutrient homeostasis and they are the main regulators of PLIN expression [142,153,154]. Therefore, the pharmacological modulation of PPAR activity is directly associated with the modulation of the energy metabolism [174,179]. Even though this effect is already vastly explored in clinical practice for the treatment of hyperglycemia and dyslipidemias, it is also a target of interest for metabolic diseases such as obesity and cancer [180].
Fibrates are high affinity PPARα activators used for the treatment of dyslipidemias [181,182]. Through the activation of PPARα, these drugs enhance the cellular uptake of FA and their breakdown through the β-oxidation pathways [182]. Besides activating other genes related to peroxisomal and mitochondrial FA oxidation, PPARα induces PLIN5 expression [183], which can explain the weight lowering effect of fibrates, since PLIN5 is involved in the PKA-mediated lipolytic mechanisms and β-oxidation [149]. The prioritization of the oxidation of FAs over glycolysis and the disturbance of the equilibrium between glucose and lipid metabolism are a possible explanation of the potential anti-tumorigenic activity of PPARα agonists in cancer treatment [184].
Thiazolidinediones (TZDs), recognized as PPARγ activators, are utilized in the management of diabetes [185]. Considering the established tendency of PPARγ activators to induce adipogenesis [186] and upregulate PLIN2 [61,62,63] expression in non-adipose cells, it is plausible that these medications might foster fat accumulation [187], which would explain the commonly observed weight gain associated with thiazolidinedione therapy [34].
Some cancer types are known to express PLIN2 [90,91,92,93,95,96,99,101,105,106,107,108,160,161,162,163,164]. Consequently, PPARγ activators might heighten the expression of these PLIN isoforms, shielding FAs in the LDs more efficiently, thereby challenging the cancer cell’s ability to utilize FAs to meet its metabolic demands [157,188]. A more difficult access to FAs within the LDs could potentially suppress cancer cell proliferation. This hypothesis aligns with findings that TZDs have demonstrated a reduction in cancer cell proliferation [189] and that PPARγ activation is seen as a possible anticancer strategy [190,191,192].
TZDs are also known to have anti-inflammatory properties [193,194]. Whether the anti-inflammatory effects of thiazolidinediones could also be attributed to the inhibition of PLIN2 release and, hence, a consequential suppression of its function as an adipokine, is a possibility that merits further investigation.

5.2. AMPK Activators

Given AMPK function in promoting the CMA of PLIN2, PLIN3, and the increasing expression of PLIN5 [146], the stimulation of AMPK affords a potential route to altering the coordination of PLINs in the oversight of FA trafficking.
Pharmacologically, AMPK may be activated by drugs such as salicylic acid or by its precursor, acetylsalicylic acid [195] and metformin [177]. Therefore, it is expected that AMPK activators would ease the mobilization of FAs from LDs and prepare target cells for a more extensive use of FAs for mitochondrial β-oxidation. In 3T3-L1 preadipocytes, aspirin treatment was shown to inhibit adipocyte differentiation [196] and lipid accumulation [197]. In animal models of obesity, treatment with a low dose of aspirin resulted in a significant reduction of body weight, visceral fat mass and serum total cholesterols, and serum and adipose tissue TAGs [198]. In humans, the influence of aspirin on obesity has not yet been systematically studied but, in healthy volunteers, a low dose of aspirin was shown to increase β-oxidation [199].
Metformin was also shown to reduce LD accumulation in human chorionic villous mesenchymal stem cells [200], an observation consistent with what was expected from the effects of an AMPK activator. However, the inconsistency of the effects of metformin in human subjects did not support the approval for its clinical use for weight loss in non-diabetic patients [201]. The inconsistency observed could potentially be attributed to the mechanism underlying the activation of AMPK [201]. Metformin activates AMPK indirectly, inhibiting the respiratory chain complex I [202], ATP production and, consequently, causing a putative increase in the AMP/ATP ratio needed for AMPK activation. Weight loss caused by AMPK activation would also require FA β-oxidation, which will be limited in cases where respiratory chain complex I is inhibited.
Adiponectin is a glycoprotein primarily produced in adipocytes [203]. The expression of adiponectin and its plasma levels are influenced by circadian rhythms and by the activity of several hormones and transcription factors [204]. PPARγ is the major positive regulator of adiponectin gene expression [205], which provides a link between the availability of FAs for β-oxidation. The effects of adiponectin are mediated by adiponectin receptors, AdipoR1, and AdipoR2 [206]. Adiponectin receptors are membrane receptors with seven transmembrane domains like the G-protein coupled receptors [207]. However, they have an atypical structure and function, acting as catalytic receptors [208]. AMPK is one of the targets of adiponectin receptors [209]. Therefore, activation of adiponectin receptors by endogenous adiponectin or by adiponectin receptor agonists [210] may activate AMPK and cause the pattern of PLIN alterations expected from a direct AMPK activation: facilitating FA mobilization as a result of CMA of PLIN2 and PLIN3 [73] and promoting FA β-oxidation, as a result of the increase in PLIN5 expression [32]. Adiponectin also increases HDL (high-density lipoprotein) levels and its function [211] and the expression of apolipoprotein A-I (Apo A-I), the main apolipoprotein of HDL [212]. Single nucleotide polymorphisms in the PLIN1 and PLIN2 genes have been associated with changes in Apo A-I levels (PLIN1 in boys and PLIN2 in girls) and HDL-cholesterol levels (PLIN1 in girls), which was seen as a putative risk factor for obesity and cardiovascular disease depending on sex across life [213]. The influence of adiponectin in obesity and in cancer is widely accepted [214,215,216,217,218,219,220]. The role of adiponectin in preventing and protecting against the development of multiple disorders related to obesity, especially in metabolic syndromes, diabetes mellitus, cardiovascular diseases [221], inflammation [222], and cancers [217,223] has been extensively documented and may have a contribution from an effect on HDL levels, since it has been demonstrated that HDL can potentially reduce the risk and progression of cancer [213].

5.3. SIRT1 Activators

SIRT1 is a post-translational regulator that affects multiple biological processes according to the cell energy status [224], and its activity is induced by PLIN5 [225]. Therefore, SIRT1 activators may interfere with PLINs pathways by reinforcing the role of PLIN5 in promoting β-oxidation [149].
Resveratrol is one of the first SIRT1 activators studied, but the list of SIRT activators is being extended with the inclusion of other activators of natural (quercetin, sulforaphane) and synthetic origin (selisistat, SRT2104) [175,176]. Sulforaphane has been reported to decrease body weight and adipose tissue increases induced by HFD in mice, by reducing the number and size of LDs and the expression of PLIN2 and PLIN5 in 3T3-L1 cells [226]. The activity of SIRT1 activators in obesity and in related metabolic syndromes is being studied and some works revealed improvements in metabolic markers and mitochondrial function [227], which may indicate the involvement of SIRT1 activation in the effects of PLIN5 in preserving mitochondrial β-oxidation. Several studies have also demonstrated the potential effect of resveratrol in chemoprevention in some cancers [228]. Although several mechanisms have been proposed to explain the anti-cancer properties of resveratrol, the activation of PLIN5/SIRT1 pathway represents one likely explanation.

5.4. Modulators of cAMP/PKA Pathway

The cAMP/PKA pathway is a complex pathway that presents several possibilities of pharmacological intervention. One of the most obvious and extensively explored is the intervention at the membrane G-protein coupled receptors that lead to adenylyl cyclase activation [229]. The β-adrenoceptors (β-AR) are one of the most studied receptors [230].
β-AR activation promotes the lipolysis of stored triglyceride in both white and brown adipocytes [231]. The mechanism involved is PKA-induced PLIN1 phosphorylation and the consequent promotion of lipolysis [39]. However, this effect may not culminate in overall weight loss since β-AR activation also increases protein synthesis [232]. The anabolic effect explains the off-label use of β-AR agonists in animal production to increase growth and carcass protein composition [233]. It is also one of the reasons why β-AR agonists are included in the Prohibited List issued by the World Anti-Doping Agency [234,235].
In cancer, activation of the PKA pathway may also provide the energy to fuel the higher anabolic activity associated with these cells. This assumption is supported by the observation that clenbuterol, a long-acting β-AR agonist, was shown to induce cell proliferation [236] and that β-AR activation caused an increase in LD number and lipid content [170] in breast cancer cell lines. The putative clinical relevance of a pro-tumorigenic effect of β-AR activation is further supported by reports showing that the chronic use of β-AR antagonists decreases the incidence or mortality of many cancer types [237,238,239,240,241,242,243].

5.5. Other Drugs

Knowing the relevance of PLINs in controlling FA distribution and use, its putative involvement may be considered when trying to explain the effects caused by several drugs on body weight. For instance, the chronic use of antipsychotic drugs, such as olanzapine, clozapine, and chlorpromazine, causes an increase in body weight and fat mass [244,245]. These effects have been ascribed to increased expression of PLIN1 in human adipocytes [245], PLIN2, and PLIN4 [244].
The opposite may also occur. The following clinically approved drugs were described to decrease lipid accumulation or cause weight loss as an adverse drug reaction by a mechanism that involves a reduction of PLIN1 expression or an increase PLIN1 degradation: adapalene, used for topical treatment of mild to moderate acne [246]; artesunate, an antimalarial drug [247]; menadione, a synthetic form of vitamin K [248]; tetrandrine, an alkaloid with anti-inflammatory and anti-cancerous activities [249]; and nelfinavir, an antiviral agent [250].
Other drugs may cause a reaction pattern more complex than an increase/decrease of body weight. For instance, vitamin D [251] or dihydroartemisinin, an antimalarial drug, may reduce PLIN2 expression and LD formation [252].

6. Conclusions and Future Directions

This review summarizes the role of LDs and PLINs in the management of lipids in our body. The coordinated activity of PLINs promotes a healthy balance between storage, when supply exceeds demand, and release, when demand increases. However, this coordination is very fragile and can be easily disrupted.
Alterations in the balance of PLINs that cause an excessive retention of lipids in the LD may promote WAT expansion and obesity. This alteration may favor LD expansion in cancer tissues. Pharmacological interventions to prevent this type of imbalance could be applicable to the treatment of obesity-related diseases and cancer. The challenge to develop drugs that target the activity of PLINs may be facilitated by the knowledge that some drugs already used in clinical practice are known to alter body weight by altering PLINs activity. The present review is intended to provide new insights into the coordination of PLINs activity to foster the development of PLINs pharmacology, particularly with new classes of drugs that are able to directly target each PLIN.

Author Contributions

Conceptualization, J.G., P.F. and V.B.-R.; methodology, J.G., P.F. and V.B.-R.; software, V.B.-R. and D.S.; validation, J.G., P.F., D.S. and V.B.-R.; formal analysis, J.G. and P.F.; investigation, V.B.-R., D.S., A.B.-E. and P.N.; resources, J.G.; data curation, J.G. and P.F.; writing—original draft preparation, V.B.-R., D.S., A.B.-E. and P.N.; writing—review and editing, P.F., J.G., V.B.-R. and A.B.-E.; visualization, V.B.-R., D.S., A.B.-E. and P.N.; supervision, P.F. and J.G.; project administration, P.F.; funding acquisition, J.G. All authors have read and agreed to the published version of the manuscript.

Funding

D.S. has a Ph.D. grant scholarship (Grant number: 2020.08563.BD) financed by Fundação para a Ciência e Tecnologia (FCT) through the European Social Fund (ESF) and national funds.

Acknowledgments

All figures were created on https://www.biorender.com (accessed on 1 August 2023).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Royo-Garcia, A.; Courtois, S.; Parejo-Alonso, B.; Espiau-Romera, P.; Sancho, P. Lipid droplets as metabolic determinants for stemness and chemoresistance in cancer. World J. Stem Cells 2021, 13, 1307–1317. [Google Scholar] [CrossRef] [PubMed]
  2. Petan, T.; Jarc, E.; Jusovic, M. Lipid Droplets in Cancer: Guardians of Fat in a Stressful World. Molecules 2018, 23, 1941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Ma, Y.; Temkin, S.M.; Hawkridge, A.M.; Guo, C.; Wang, W.; Wang, X.Y.; Fang, X. Fatty acid oxidation: An emerging facet of metabolic transformation in cancer. Cancer Lett. 2018, 435, 92–100. [Google Scholar] [CrossRef] [PubMed]
  4. Cheng, C.; Geng, F.; Cheng, X.; Guo, D. Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun. 2018, 38, 27. [Google Scholar] [CrossRef]
  5. Maan, M.; Peters, J.M.; Dutta, M.; Patterson, A.D. Lipid metabolism and lipophagy in cancer. Biochem. Biophys. Res. Commun. 2018, 504, 582–589. [Google Scholar] [CrossRef]
  6. Matsushita, Y.; Nakagawa, H.; Koike, K. Lipid Metabolism in Oncology: Why It Matters, How to Research, and How to Treat. Cancers 2021, 13, 474. [Google Scholar] [CrossRef]
  7. Wang, C.W. Lipid droplets, lipophagy, and beyond. Biochim. Biophys. Acta 2016, 1861, 793–805. [Google Scholar] [CrossRef]
  8. Petan, T. Lipid Droplets in Cancer. Rev. Physiol. Biochem. Pharmacol. 2023, 185, 53–86. [Google Scholar] [CrossRef]
  9. Cruz, A.L.S.; Barreto, E.A.; Fazolini, N.P.B.; Viola, J.P.B.; Bozza, P.T. Lipid droplets: Platforms with multiple functions in cancer hallmarks. Cell Death Dis. 2020, 11, 105. [Google Scholar] [CrossRef] [Green Version]
  10. Farese, R.V., Jr.; Walther, T.C. Lipid droplets finally get a little R-E-S-P-E-C-T. Cell 2009, 139, 855–860. [Google Scholar] [CrossRef] [Green Version]
  11. Walther, T.C.; Farese, R.V., Jr. Lipid droplets and cellular lipid metabolism. Annu. Rev. Biochem. 2012, 81, 687–714. [Google Scholar] [CrossRef] [Green Version]
  12. Greenberg, A.S.; Egan, J.J.; Wek, S.A.; Garty, N.B.; Blanchette-Mackie, E.J.; Londos, C. Perilipin, a major hormonally regulated adipocyte-specific phosphoprotein associated with the periphery of lipid storage droplets. J. Biol. Chem. 1991, 266, 11341–11346. [Google Scholar] [CrossRef]
  13. Bosch, M.; Pol, A. Eukaryotic lipid droplets: Metabolic hubs, and immune first responders. Trends Endocrinol. Metab. 2022, 33, 218–229. [Google Scholar] [CrossRef]
  14. Welte, M.A.; Gould, A.P. Lipid droplet functions beyond energy storage. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2017, 1862, 1260–1272. [Google Scholar] [CrossRef]
  15. Kimmel, A.R.; Sztalryd, C. The Perilipins: Major Cytosolic Lipid Droplet-Associated Proteins and Their Roles in Cellular Lipid Storage, Mobilization, and Systemic Homeostasis. Annu. Rev. Nutr. 2016, 36, 471–509. [Google Scholar] [CrossRef]
  16. Zhang, P.; Meng, L.; Song, L.; Du, J.; Du, S.; Cui, W.; Liu, C.; Li, F. Roles of Perilipins in Diseases and Cancers. Curr. Genom. 2018, 19, 247–257. [Google Scholar] [CrossRef]
  17. Wilfling, F.; Haas, J.T.; Walther, T.C.; Farese, R.V., Jr. Lipid droplet biogenesis. Curr. Opin. Cell Biol. 2014, 29, 39–45. [Google Scholar] [CrossRef] [Green Version]
  18. Silverstein, R.L.; Li, W.; Park, Y.M.; Rahaman, S.O. Mechanisms of cell signaling by the scavenger receptor CD36: Implications in atherosclerosis and thrombosis. Trans. Am. Clin. Clim. Assoc. 2010, 121, 206–220. [Google Scholar]
  19. Hao, J.W.; Wang, J.; Guo, H.; Zhao, Y.Y.; Sun, H.H.; Li, Y.F.; Lai, X.Y.; Zhao, N.; Wang, X.; Xie, C.; et al. CD36 facilitates fatty acid uptake by dynamic palmitoylation-regulated endocytosis. Nat. Commun. 2020, 11, 4765. [Google Scholar] [CrossRef]
  20. Glatz, J.F.C.; Luiken, J. Dynamic role of the transmembrane glycoprotein CD36 (SR-B2) in cellular fatty acid uptake and utilization. J. Lipid Res. 2018, 59, 1084–1093. [Google Scholar] [CrossRef] [Green Version]
  21. Boord, J.B.; Fazio, S.; Linton, M.F. Cytoplasmic fatty acid-binding proteins: Emerging roles in metabolism and atherosclerosis. Curr. Opin. Lipidol. 2002, 13, 141–147. [Google Scholar] [CrossRef] [PubMed]
  22. Koundouros, N.; Poulogiannis, G. Reprogramming of fatty acid metabolism in cancer. Br. J. Cancer 2020, 122, 4–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Coleman, R.A.; Lee, D.P. Enzymes of triacylglycerol synthesis and their regulation. Prog. Lipid Res. 2004, 43, 134–176. [Google Scholar] [CrossRef] [PubMed]
  24. Jin, Y.; McFie, P.J.; Banman, S.L.; Brandt, C.; Stone, S.J. Diacylglycerol acyltransferase-2 (DGAT2) and monoacylglycerol acyltransferase-2 (MGAT2) interact to promote triacylglycerol synthesis. J. Biol. Chem. 2014, 289, 28237–28248. [Google Scholar] [CrossRef] [Green Version]
  25. Walther, T.C.; Farese, R.V., Jr. The life of lipid droplets. Biochim. Biophys. Acta 2009, 1791, 459–466. [Google Scholar] [CrossRef]
  26. Kadereit, B.; Kumar, P.; Wang, W.J.; Miranda, D.; Snapp, E.L.; Severina, N.; Torregroza, I.; Evans, T.; Silver, D.L. Evolutionarily conserved gene family important for fat storage. Proc. Natl. Acad. Sci. USA 2008, 105, 94–99. [Google Scholar] [CrossRef]
  27. Wang, G.; Chen, A.; Wu, Y.; Wang, D.; Chang, C.; Yu, G. Fat storage-inducing transmembrane proteins: Beyond mediating lipid droplet formation. Cell Mol. Biol. Lett. 2022, 27, 98. [Google Scholar] [CrossRef]
  28. Salo, V.T.; Belevich, I.; Li, S.; Karhinen, L.; Vihinen, H.; Vigouroux, C.; Magre, J.; Thiele, C.; Holtta-Vuori, M.; Jokitalo, E.; et al. Seipin regulates ER-lipid droplet contacts and cargo delivery. EMBO J. 2016, 35, 2699–2716. [Google Scholar] [CrossRef]
  29. Barneda, D.; Christian, M. Lipid droplet growth: Regulation of a dynamic organelle. Curr. Opin. Cell Biol. 2017, 47, 9–15. [Google Scholar] [CrossRef] [Green Version]
  30. Jin, Y.; Tan, Y.; Zhao, P.; Ren, Z. SEIPIN: A Key Factor for Nuclear Lipid Droplet Generation and Lipid Homeostasis. Int. J. Mol. Sci. 2020, 21, 8208. [Google Scholar] [CrossRef]
  31. Rao, M.J.; Goodman, J.M. Seipin: Harvesting fat and keeping adipocytes healthy. Trends Cell Biol. 2021, 31, 912–923. [Google Scholar] [CrossRef]
  32. Sztalryd, C.; Brasaemle, D.L. The perilipin family of lipid droplet proteins: Gatekeepers of intracellular lipolysis. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2017, 1862, 1221–1232. [Google Scholar] [CrossRef]
  33. D’Souza-Schorey, C.; Chavrier, P. ARF proteins: Roles in membrane traffic and beyond. Nat. Rev. Mol. Cell Biol. 2006, 7, 347–358. [Google Scholar] [CrossRef]
  34. Ahmadian, M.; Suh, J.M.; Hah, N.; Liddle, C.; Atkins, A.R.; Downes, M.; Evans, R.M. PPARgamma signaling and metabolism: The good, the bad and the future. Nat. Med. 2013, 19, 557–566. [Google Scholar] [CrossRef] [Green Version]
  35. Wolins, N.E.; Brasaemle, D.L.; Bickel, P.E. A proposed model of fat packaging by exchangeable lipid droplet proteins. FEBS Lett. 2006, 580, 5484–5491. [Google Scholar] [CrossRef] [Green Version]
  36. Bersuker, K.; Olzmann, J.A. Establishing the lipid droplet proteome: Mechanisms of lipid droplet protein targeting and degradation. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2017, 1862, 1166–1177. [Google Scholar] [CrossRef]
  37. Schuldiner, M.; Bohnert, M. A different kind of love—Lipid droplet contact sites. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2017, 1862, 1188–1196. [Google Scholar] [CrossRef]
  38. Sztalryd, C.; Kimmel, A.R. Perilipins: Lipid droplet coat proteins adapted for tissue-specific energy storage and utilization, and lipid cytoprotection. Biochimie 2014, 96, 96–101. [Google Scholar] [CrossRef] [Green Version]
  39. Hansen, J.S.; de Mare, S.; Jones, H.A.; Goransson, O.; Lindkvist-Petersson, K. Visualization of lipid directed dynamics of perilipin 1 in human primary adipocytes. Sci. Rep. 2017, 7, 15011. [Google Scholar] [CrossRef] [Green Version]
  40. Yamaguchi, T.; Omatsu, N.; Morimoto, E.; Nakashima, H.; Ueno, K.; Tanaka, T.; Satouchi, K.; Hirose, F.; Osumi, T. CGI-58 facilitates lipolysis on lipid droplets but is not involved in the vesiculation of lipid droplets caused by hormonal stimulation. J. Lipid Res. 2007, 48, 1078–1089. [Google Scholar] [CrossRef] [Green Version]
  41. Itabe, H.; Yamaguchi, T.; Nimura, S.; Sasabe, N. Perilipins: A diversity of intracellular lipid droplet proteins. Lipids Health Dis. 2017, 16, 83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Malfacini, D.; Pfeifer, A. GPCR in Adipose Tissue Function-Focus on Lipolysis. Biomedicines 2023, 11, 588. [Google Scholar] [CrossRef] [PubMed]
  43. Mottagui-Tabar, S.; Ryden, M.; Lofgren, P.; Faulds, G.; Hoffstedt, J.; Brookes, A.J.; Andersson, I.; Arner, P. Evidence for an important role of perilipin in the regulation of human adipocyte lipolysis. Diabetologia 2003, 46, 789–797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Garaulet, M.; Vera, B.; Bonnet-Rubio, G.; Gomez-Abellan, P.; Lee, Y.C.; Ordovas, J.M. Lunch eating predicts weight-loss effectiveness in carriers of the common allele at PERILIPIN1: The ONTIME (Obesity, Nutrigenetics, Timing, Mediterranean) study. Am. J. Clin. Nutr. 2016, 104, 1160–1166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Smith, C.E.; Ordovas, J.M. Update on perilipin polymorphisms and obesity. Nutr. Rev. 2012, 70, 611–621. [Google Scholar] [CrossRef] [Green Version]
  46. Bialesova, L.; Kulyte, A.; Petrus, P.; Sinha, I.; Laurencikiene, J.; Zhao, C.; Wright, K.D.; Arner, P.; Dahlman, I. Epigenetic Regulation of PLIN 1 in Obese Women and its Relation to Lipolysis. Sci. Rep. 2017, 7, 10152. [Google Scholar] [CrossRef] [Green Version]
  47. Kern, P.A.; Di Gregorio, G.; Lu, T.; Rassouli, N.; Ranganathan, G. Perilipin expression in human adipose tissue is elevated with obesity. J. Clin. Endocrinol. Metab. 2004, 89, 1352–1358. [Google Scholar] [CrossRef] [Green Version]
  48. Miyoshi, H.; Souza, S.C.; Endo, M.; Sawada, T.; Perfield, J.W., 2nd; Shimizu, C.; Stancheva, Z.; Nagai, S.; Strissel, K.J.; Yoshioka, N.; et al. Perilipin overexpression in mice protects against diet-induced obesity. J. Lipid Res. 2010, 51, 975–982. [Google Scholar] [CrossRef] [Green Version]
  49. Wang, Y.; Sullivan, S.; Trujillo, M.; Lee, M.J.; Schneider, S.H.; Brolin, R.E.; Kang, Y.H.; Werber, Y.; Greenberg, A.S.; Fried, S.K. Perilipin expression in human adipose tissues: Effects of severe obesity, gender, and depot. Obes. Res. 2003, 11, 930–936. [Google Scholar] [CrossRef]
  50. De Oliveira, B.A.P.; de Souza Pinhel, M.A.; Nicoletti, C.F.; de Oliveira, C.C.; Quinhoneiro, D.C.G.; Noronha, N.Y.; Fassini, P.G.; da Silva Junior, W.A.; Junior, W.S.; Nonino, C.B. UCP2 and PLIN1 Expression Affects the Resting Metabolic Rate and Weight Loss on Obese Patients. Obes. Surg. 2017, 27, 343–348. [Google Scholar] [CrossRef]
  51. Meng, L.X.; Zheng, Y.X.; He, M.L.; Zhou, X.M.; Sun, S.Y.; Ding, Z.J.; Meng, Q.; Li, B.C.; Sun, Y.W. Silencing of perilipin by short hairpin RNA inhibits proliferation and induces apoptosis in liposarcoma cells. Mol. Med. Rep. 2018, 18, 4571–4576. [Google Scholar] [CrossRef] [Green Version]
  52. Straub, B.K.; Herpel, E.; Singer, S.; Zimbelmann, R.; Breuhahn, K.; Macher-Goeppinger, S.; Warth, A.; Lehmann-Koch, J.; Longerich, T.; Heid, H.; et al. Lipid droplet-associated PAT-proteins show frequent and differential expression in neoplastic steatogenesis. Mod. Pathol. 2010, 23, 480–492. [Google Scholar] [CrossRef] [Green Version]
  53. Westhoff, C.C.; Mrozinski, J.; Riedel, I.; Heid, H.W.; Moll, R. Perilipin 1 is a highly specific marker for adipocytic differentiation in sarcomas with intermediate sensitivity. J. Cancer Res. Clin. Oncol. 2017, 143, 225–232. [Google Scholar] [CrossRef]
  54. Kim, S.; Lee, Y.; Koo, J.S. Differential expression of lipid metabolism-related proteins in different breast cancer subtypes. PLoS ONE 2015, 10, e0119473. [Google Scholar] [CrossRef] [Green Version]
  55. Zhou, C.; Wang, M.; Zhou, L.; Zhang, Y.; Liu, W.; Qin, W.; He, R.; Lu, Y.; Wang, Y.; Chen, X.Z.; et al. Prognostic significance of PLIN1 expression in human breast cancer. Oncotarget 2016, 7, 54488–54502. [Google Scholar] [CrossRef] [Green Version]
  56. Brasaemle, D.L.; Barber, T.; Wolins, N.E.; Serrero, G.; Blanchette-Mackie, E.J.; Londos, C. Adipose differentiation-related protein is an ubiquitously expressed lipid storage droplet-associated protein. J. Lipid Res. 1997, 38, 2249–2263. [Google Scholar] [CrossRef]
  57. Imamura, M.; Inoguchi, T.; Ikuyama, S.; Taniguchi, S.; Kobayashi, K.; Nakashima, N.; Nawata, H. ADRP stimulates lipid accumulation and lipid droplet formation in murine fibroblasts. Am. J. Physiol. Endocrinol. Metab. 2002, 283, E775–E783. [Google Scholar] [CrossRef] [Green Version]
  58. Listenberger, L.L.; Ostermeyer-Fay, A.G.; Goldberg, E.B.; Brown, W.J.; Brown, D.A. Adipocyte differentiation-related protein reduces the lipid droplet association of adipose triglyceride lipase and slows triacylglycerol turnover. J. Lipid Res. 2007, 48, 2751–2761. [Google Scholar] [CrossRef] [Green Version]
  59. Najt, C.P.; Devarajan, M.; Mashek, D.G. Perilipins at a glance. J. Cell Sci. 2022, 135, jcs259501. [Google Scholar] [CrossRef]
  60. Patel, S.; Yang, W.; Kozusko, K.; Saudek, V.; Savage, D.B. Perilipins 2 and 3 lack a carboxy-terminal domain present in perilipin 1 involved in sequestering ABHD5 and suppressing basal lipolysis. Proc. Natl. Acad. Sci. USA 2014, 111, 9163–9168. [Google Scholar] [CrossRef]
  61. Dalen, K.T.; Schoonjans, K.; Ulven, S.M.; Weedon-Fekjaer, M.S.; Bentzen, T.G.; Koutnikova, H.; Auwerx, J.; Nebb, H.I. Adipose tissue expression of the lipid droplet-associating proteins S3-12 and perilipin is controlled by peroxisome proliferator-activated receptor-gamma. Diabetes 2004, 53, 1243–1252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Hodgkinson, C.P.; Ye, S. Microarray analysis of peroxisome proliferator-activated receptor-gamma induced changes in gene expression in macrophages. Biochem. Biophys. Res. Commun. 2003, 308, 505–510. [Google Scholar] [CrossRef] [PubMed]
  63. Schadinger, S.E.; Bucher, N.L.; Schreiber, B.M.; Farmer, S.R. PPARgamma2 regulates lipogenesis and lipid accumulation in steatotic hepatocytes. Am. J. Physiol. Endocrinol. Metab. 2005, 288, E1195–E1205. [Google Scholar] [CrossRef] [PubMed]
  64. Gao, J.; Ye, H.; Serrero, G. Stimulation of adipose differentiation related protein (ADRP) expression in adipocyte precursors by long-chain fatty acids. J. Cell Physiol. 2000, 182, 297–302. [Google Scholar] [CrossRef]
  65. Lee, Y.J.; Ko, E.H.; Kim, J.E.; Kim, E.; Lee, H.; Choi, H.; Yu, J.H.; Kim, H.J.; Seong, J.K.; Kim, K.S.; et al. Nuclear receptor PPARgamma-regulated monoacylglycerol O-acyltransferase 1 (MGAT1) expression is responsible for the lipid accumulation in diet-induced hepatic steatosis. Proc. Natl. Acad. Sci. USA 2012, 109, 13656–13661. [Google Scholar] [CrossRef]
  66. Motomura, W.; Inoue, M.; Ohtake, T.; Takahashi, N.; Nagamine, M.; Tanno, S.; Kohgo, Y.; Okumura, T. Up-regulation of ADRP in fatty liver in human and liver steatosis in mice fed with high fat diet. Biochem. Biophys. Res. Commun. 2006, 340, 1111–1118. [Google Scholar] [CrossRef] [Green Version]
  67. Sikder, K.; Shukla, S.K.; Patel, N.; Singh, H.; Rafiq, K. High Fat Diet Upregulates Fatty Acid Oxidation and Ketogenesis via Intervention of PPAR-gamma. Cell Physiol. Biochem. 2018, 48, 1317–1331. [Google Scholar] [CrossRef]
  68. Sharma, A.M.; Staels, B. Review: Peroxisome proliferator-activated receptor gamma and adipose tissue--understanding obesity-related changes in regulation of lipid and glucose metabolism. J. Clin. Endocrinol. Metab. 2007, 92, 386–395. [Google Scholar] [CrossRef]
  69. Spiegelman, B.M. Peroxisome proliferator-activated receptor gamma: A key regulator of adipogenesis and systemic insulin sensitivity. Eur. J. Med. Res. 1997, 2, 457–464. [Google Scholar]
  70. Zhang, Y.; Lin, S.; Peng, J.; Liang, X.; Yang, Q.; Bai, X.; Li, Y.; Li, J.; Dong, W.; Wang, Y.; et al. Amelioration of hepatic steatosis by dietary essential amino acid-induced ubiquitination. Mol. Cell 2022, 82, 1528–1542.e1510. [Google Scholar] [CrossRef]
  71. Zhao, W.; Zhang, Y.; Lin, S.; Li, Y.; Zhu, A.J.; Shi, H.; Liu, M. Identification of Ubr1 as an amino acid sensor of steatosis in liver and muscle. J. Cachexia Sarcopenia Muscle 2023, 14, 1454–1467. [Google Scholar] [CrossRef]
  72. Conte, M.; Vasuri, F.; Trisolino, G.; Bellavista, E.; Santoro, A.; Degiovanni, A.; Martucci, E.; D’Errico-Grigioni, A.; Caporossi, D.; Capri, M.; et al. Increased Plin2 expression in human skeletal muscle is associated with sarcopenia and muscle weakness. PLoS ONE 2013, 8, e73709. [Google Scholar] [CrossRef] [Green Version]
  73. Kaushik, S.; Cuervo, A.M. Degradation of lipid droplet-associated proteins by chaperone-mediated autophagy facilitates lipolysis. Nat. Cell Biol. 2015, 17, 759–770. [Google Scholar] [CrossRef] [Green Version]
  74. Kaushik, S.; Cuervo, A.M. AMPK-dependent phosphorylation of lipid droplet protein PLIN2 triggers its degradation by CMA. Autophagy 2016, 12, 432–438. [Google Scholar] [CrossRef] [Green Version]
  75. Casey, C.A.; Donohue, T.M., Jr.; Kubik, J.L.; Kumar, V.; Naldrett, M.J.; Woods, N.T.; Frisbie, C.P.; McNiven, M.A.; Thomes, P.G. Lipid droplet membrane proteome remodeling parallels ethanol-induced hepatic steatosis and its resolution. J. Lipid Res. 2021, 62, 100049. [Google Scholar] [CrossRef]
  76. Kaushik, S.; Cuervo, A.M. The coming of age of chaperone-mediated autophagy. Nat. Rev. Mol. Cell Biol. 2018, 19, 365–381. [Google Scholar] [CrossRef]
  77. Liu, R.; Lee, J.H.; Li, J.; Yu, R.; Tan, L.; Xia, Y.; Zheng, Y.; Bian, X.L.; Lorenzi, P.L.; Chen, Q.; et al. Choline kinase alpha 2 acts as a protein kinase to promote lipolysis of lipid droplets. Mol. Cell 2021, 81, 2722–2735.e2729. [Google Scholar] [CrossRef]
  78. Bracken, C.P.; Whitelaw, M.L.; Peet, D.J. The hypoxia-inducible factors: Key transcriptional regulators of hypoxic responses. Cell Mol. Life Sci. 2003, 60, 1376–1393. [Google Scholar] [CrossRef]
  79. Fong, G.H.; Takeda, K. Role and regulation of prolyl hydroxylase domain proteins. Cell Death Differ. 2008, 15, 635–641. [Google Scholar] [CrossRef] [Green Version]
  80. Salceda, S.; Caro, J. Hypoxia-inducible factor 1alpha (HIF-1alpha) protein is rapidly degraded by the ubiquitin-proteasome system under normoxic conditions. Its stabilization by hypoxia depends on redox-induced changes. J. Biol. Chem. 1997, 272, 22642–22647. [Google Scholar] [CrossRef] [Green Version]
  81. Bensaad, K.; Favaro, E.; Lewis, C.A.; Peck, B.; Lord, S.; Collins, J.M.; Pinnick, K.E.; Wigfield, S.; Buffa, F.M.; Li, J.L.; et al. Fatty acid uptake and lipid storage induced by HIF-1alpha contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep. 2014, 9, 349–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Qiu, B.; Ackerman, D.; Sanchez, D.J.; Li, B.; Ochocki, J.D.; Grazioli, A.; Bobrovnikova-Marjon, E.; Diehl, J.A.; Keith, B.; Simon, M.C. HIF2alpha-Dependent Lipid Storage Promotes Endoplasmic Reticulum Homeostasis in Clear-Cell Renal Cell Carcinoma. Cancer Discov. 2015, 5, 652–667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Gimm, T.; Wiese, M.; Teschemacher, B.; Deggerich, A.; Schodel, J.; Knaup, K.X.; Hackenbeck, T.; Hellerbrand, C.; Amann, K.; Wiesener, M.S.; et al. Hypoxia-inducible protein 2 is a novel lipid droplet protein and a specific target gene of hypoxia-inducible factor-1. FASEB J. 2010, 24, 4443–4458. [Google Scholar] [CrossRef]
  84. DiStefano, M.T.; Danai, L.V.; Roth Flach, R.J.; Chawla, A.; Pedersen, D.J.; Guilherme, A.; Czech, M.P. The Lipid Droplet Protein Hypoxia-inducible Gene 2 Promotes Hepatic Triglyceride Deposition by Inhibiting Lipolysis. J. Biol. Chem. 2015, 290, 15175–15184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Povero, D.; Johnson, S.M.; Liu, J. Hypoxia, hypoxia-inducible gene 2 (HIG2)/HILPDA, and intracellular lipolysis in cancer. Cancer Lett. 2020, 493, 71–79. [Google Scholar] [CrossRef]
  86. Liu, W.; Liu, X.; Liu, Y.; Ling, T.; Chen, D.; Otkur, W.; Zhao, H.; Ma, M.; Ma, K.; Dong, B.; et al. PLIN2 promotes HCC cells proliferation by inhibiting the degradation of HIF1alpha. Exp. Cell Res. 2022, 418, 113244. [Google Scholar] [CrossRef]
  87. Trayhurn, P. Hypoxia and adipose tissue function and dysfunction in obesity. Physiol. Rev. 2013, 93, 1–21. [Google Scholar] [CrossRef] [Green Version]
  88. Wang, R.; Sun, Q.; Wu, X.; Zhang, Y.; Xing, X.; Lin, K.; Feng, Y.; Wang, M.; Wang, Y.; Wang, R. Hypoxia as a Double-Edged Sword to Combat Obesity and Comorbidities. Cells 2022, 11, 3735. [Google Scholar] [CrossRef]
  89. Muz, B.; de la Puente, P.; Azab, F.; Azab, A.K. The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy. Hypoxia 2015, 3, 83–92. [Google Scholar] [CrossRef] [Green Version]
  90. Matsubara, J.; Honda, K.; Ono, M.; Sekine, S.; Tanaka, Y.; Kobayashi, M.; Jung, G.; Sakuma, T.; Nakamori, S.; Sata, N.; et al. Identification of adipophilin as a potential plasma biomarker for colorectal cancer using label-free quantitative mass spectrometry and protein microarray. Cancer Epidemiol. Biomark. Prev. 2011, 20, 2195–2203. [Google Scholar] [CrossRef] [Green Version]
  91. Qi, W.; Fitchev, P.S.; Cornwell, M.L.; Greenberg, J.; Cabe, M.; Weber, C.R.; Roy, H.K.; Crawford, S.E.; Savkovic, S.D. FOXO3 growth inhibition of colonic cells is dependent on intraepithelial lipid droplet density. J. Biol. Chem. 2013, 288, 16274–16281. [Google Scholar] [CrossRef] [Green Version]
  92. Pucer, A.; Brglez, V.; Payre, C.; Pungercar, J.; Lambeau, G.; Petan, T. Group X secreted phospholipase A(2) induces lipid droplet formation and prolongs breast cancer cell survival. Mol. Cancer 2013, 12, 111. [Google Scholar] [CrossRef] [Green Version]
  93. Zhang, X.; Su, L.; Sun, K. Expression status and prognostic value of the perilipin family of genes in breast cancer. Am. J. Transl. Res. 2021, 13, 4450–4463. [Google Scholar]
  94. Lin, L.C.; Gao, A.C.; Lai, C.H.; Hsieh, J.T.; Lin, H. Induction of neuroendocrine differentiation in castration resistant prostate cancer cells by adipocyte differentiation-related protein (ADRP) delivered by exosomes. Cancer Lett. 2017, 391, 74–82. [Google Scholar] [CrossRef]
  95. Morrissey, J.J.; Mobley, J.; Figenshau, R.S.; Vetter, J.; Bhayani, S.; Kharasch, E.D. Urine aquaporin 1 and perilipin 2 differentiate renal carcinomas from other imaged renal masses and bladder and prostate cancer. Mayo Clin. Proc. 2015, 90, 35–42. [Google Scholar] [CrossRef]
  96. Fujimoto, M.; Yoshizawa, A.; Sumiyoshi, S.; Sonobe, M.; Menju, T.; Hirata, M.; Momose, M.; Date, H.; Haga, H. Adipophilin expression in lung adenocarcinoma is associated with apocrine-like features and poor clinical prognosis: An immunohistochemical study of 328 cases. Histopathology 2017, 70, 232–241. [Google Scholar] [CrossRef]
  97. Meng, X.; Sun, R.; Wang, W.; Zhang, N.; Cao, S.; Liu, B.; Fang, P.; Deng, S.; Yang, S. ADFP promotes cell proliferation in lung adenocarcinoma via Akt phosphorylation. J. Cell Mol. Med. 2021, 25, 827–839. [Google Scholar] [CrossRef]
  98. Cao, Q.; Ruan, H.; Wang, K.; Song, Z.; Bao, L.; Xu, T.; Xiao, H.; Wang, C.; Cheng, G.; Tong, J.; et al. Overexpression of PLIN2 is a prognostic marker and attenuates tumor progression in clear cell renal cell carcinoma. Int. J. Oncol. 2018, 53, 137–147. [Google Scholar] [CrossRef] [Green Version]
  99. Morrissey, J.J.; London, A.N.; Luo, J.; Kharasch, E.D. Urinary biomarkers for the early diagnosis of kidney cancer. Mayo Clin. Proc. 2010, 85, 413–421. [Google Scholar] [CrossRef] [Green Version]
  100. Morrissey, J.J.; Mellnick, V.M.; Luo, J.; Siegel, M.J.; Figenshau, R.S.; Bhayani, S.; Kharasch, E.D. Evaluation of Urine Aquaporin-1 and Perilipin-2 Concentrations as Biomarkers to Screen for Renal Cell Carcinoma: A Prospective Cohort Study. JAMA Oncol. 2015, 1, 204–212. [Google Scholar] [CrossRef]
  101. Morrissey, J.J.; Mobley, J.; Song, J.; Vetter, J.; Luo, J.; Bhayani, S.; Figenshau, R.S.; Kharasch, E.D. Urinary concentrations of aquaporin-1 and perilipin-2 in patients with renal cell carcinoma correlate with tumor size and stage but not grade. Urology 2014, 83, 256.e9–256.e14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Song, J.B.; Morrissey, J.J.; Mobley, J.M.; Figenshau, K.G.; Vetter, J.M.; Bhayani, S.B.; Kharasch, E.D.; Figenshau, R.S. Urinary aquaporin 1 and perilipin 2: Can these novel markers accurately characterize small renal masses and help guide patient management? Int. J. Urol. 2019, 26, 260–265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Song, Y.; Zhong, L.; Zhou, J.; Lu, M.; Xing, T.; Ma, L.; Shen, J. Data-Independent Acquisition-Based Quantitative Proteomic Analysis Reveals Potential Biomarkers of Kidney Cancer. Proteom. Clin. Appl. 2017, 11, 1700066. [Google Scholar] [CrossRef] [PubMed]
  104. Tolkach, Y.; Luders, C.; Meller, S.; Jung, K.; Stephan, C.; Kristiansen, G. Adipophilin as prognostic biomarker in clear cell renal cell carcinoma. Oncotarget 2017, 8, 28672–28682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Yao, M.; Huang, Y.; Shioi, K.; Hattori, K.; Murakami, T.; Nakaigawa, N.; Kishida, T.; Nagashima, Y.; Kubota, Y. Expression of adipose differentiation-related protein: A predictor of cancer-specific survival in clear cell renal carcinoma. Clin. Cancer Res. 2007, 13, 152–160. [Google Scholar] [CrossRef] [Green Version]
  106. Takada, N.; Hirokawa, M.; Ito, A.; Suzuki, A.; Higuchi, M.; Kuma, S.; Hayashi, T.; Daa, T.; Miyauchi, A. Cytoplasmic Lipid Accumulation Characteristic of the Cribriform Variant of Papillary Thyroid Carcinoma. Pathobiology 2017, 84, 251–257. [Google Scholar] [CrossRef]
  107. Sun, X.; Yang, S.; Feng, X.; Zheng, Y.; Zhou, J.; Wang, H.; Zhang, Y.; Sun, H.; He, C. The modification of ferroptosis and abnormal lipometabolism through overexpression and knockdown of potential prognostic biomarker perilipin2 in gastric carcinoma. Gastric Cancer 2020, 23, 241–259. [Google Scholar] [CrossRef]
  108. Fujimoto, M.; Matsuzaki, I.; Yamamoto, Y.; Yoshizawa, A.; Warigaya, K.; Iwahashi, Y.; Kojima, F.; Furukawa, F.; Murata, S.I. Adipophilin expression in cutaneous malignant melanoma. J. Cutan. Pathol. 2017, 44, 228–236. [Google Scholar] [CrossRef]
  109. Fortunato, D.; Giuffrida, M.G.; Cavaletto, M.; Garoffo, L.P.; Dellavalle, G.; Napolitano, L.; Giunta, C.; Fabris, C.; Bertino, E.; Coscia, A.; et al. Structural proteome of human colostral fat globule membrane proteins. Proteomics 2003, 3, 897–905. [Google Scholar] [CrossRef]
  110. Heid, H.W.; Schnolzer, M.; Keenan, T.W. Adipocyte differentiation-related protein is secreted into milk as a constituent of milk lipid globule membrane. Biochem. J. 1996, 320 Pt 3, 1025–1030. [Google Scholar] [CrossRef] [Green Version]
  111. Monks, J.; Orlicky, D.J.; Libby, A.E.; Dzieciatkowska, M.; Ladinsky, M.S.; McManaman, J.L. Perilipin-2 promotes lipid droplet-plasma membrane interactions that facilitate apocrine lipid secretion in secretory epithelial cells of the mouse mammary gland. Front. Cell Dev. Biol. 2022, 10, 958566. [Google Scholar] [CrossRef]
  112. Chong, B.M.; Reigan, P.; Mayle-Combs, K.D.; Orlicky, D.J.; McManaman, J.L. Determinants of adipophilin function in milk lipid formation and secretion. Trends Endocrinol. Metab. 2011, 22, 211–217. [Google Scholar] [CrossRef] [Green Version]
  113. Karasek, D.; Vaverkova, H.; Frysak, Z.; Orsag, J.; Novotny, D.; Halenka, M.; Slavik, L. Relationship between serum adipocyte fatty acid-binding protein and endothelial/hemostatic markers in dyslipidemic subjects. Neuro Endocrinol. Lett. 2012, 33 (Suppl. 2), 26–31. [Google Scholar]
  114. Liao, B.; Geng, L.; Zhang, F.; Shu, L.; Wei, L.; Yeung, P.K.K.; Lam, K.S.L.; Chung, S.K.; Chang, J.; Vanhoutte, P.M.; et al. Adipocyte fatty acid-binding protein exacerbates cerebral ischaemia injury by disrupting the blood-brain barrier. Eur. Heart J. 2020, 41, 3169–3180. [Google Scholar] [CrossRef]
  115. Wei, C.; Liu, Y.; Xing, E.; Ding, Z.; Tian, Y.; Zhao, Z.; Fan, W.; Sun, L. Association Between Novel Pro- and Anti- Inflammatory Adipocytokines in Patients with Acute Coronary Syndrome. Clin. Appl. Thromb. Hemost. 2022, 28, 10760296221128021. [Google Scholar] [CrossRef]
  116. Orlicky, D.J.; Libby, A.E.; Bales, E.S.; McMahan, R.H.; Monks, J.; La Rosa, F.G.; McManaman, J.L. Perilipin-2 promotes obesity and progressive fatty liver disease in mice through mechanistically distinct hepatocyte and extra-hepatocyte actions. J. Physiol. 2019, 597, 1565–1584. [Google Scholar] [CrossRef] [Green Version]
  117. Cho, K.Y.; Miyoshi, H.; Nakamura, A.; Greenberg, A.S.; Atsumi, T. Lipid Droplet Protein PLIN1 Regulates Inflammatory Polarity in Human Macrophages and is Involved in Atherosclerotic Plaque Development by Promoting Stable Lipid Storage. J. Atheroscler. Thromb. 2023, 30, 170–181. [Google Scholar] [CrossRef]
  118. Zhang, S.; Hu, L.; Han, C.; Huang, R.; Ooi, K.; Qian, X.; Ren, X.; Chu, D.; Zhang, H.; Du, D.; et al. PLIN2 Mediates Neuroinflammation and Oxidative/Nitrosative Stress via Downregulating Phosphatidylethanolamine in the Rostral Ventrolateral Medulla of Stressed Hypertensive Rats. J. Inflamm. Res. 2021, 14, 6331–6348. [Google Scholar] [CrossRef]
  119. Conte, M.; Medici, V.; Malagoli, D.; Chiariello, A.; Cirrincione, A.; Davin, A.; Chikhladze, M.; Vasuri, F.; Legname, G.; Ferrer, I.; et al. Expression pattern of perilipins in human brain during aging and in Alzheimer’s disease. Neuropathol. Appl. Neurobiol. 2022, 48, e12756. [Google Scholar] [CrossRef]
  120. Bogie, J.F.J.; Grajchen, E.; Wouters, E.; Corrales, A.G.; Dierckx, T.; Vanherle, S.; Mailleux, J.; Gervois, P.; Wolfs, E.; Dehairs, J.; et al. Stearoyl-CoA desaturase-1 impairs the reparative properties of macrophages and microglia in the brain. J. Exp. Med. 2020, 217, e20191660. [Google Scholar] [CrossRef]
  121. Loix, M.; Wouters, E.; Vanherle, S.; Dehairs, J.; McManaman, J.L.; Kemps, H.; Swinnen, J.V.; Haidar, M.; Bogie, J.F.J.; Hendriks, J.J.A. Perilipin-2 limits remyelination by preventing lipid droplet degradation. Cell Mol. Life Sci. 2022, 79, 515. [Google Scholar] [CrossRef] [PubMed]
  122. Li, Q.; Zhao, Y.; Guo, H.; Li, Q.; Yan, C.; Li, Y.; He, S.; Wang, N.; Wang, Q. Impaired lipophagy induced-microglial lipid droplets accumulation contributes to the buildup of TREM1 in diabetes-associated cognitive impairment. Autophagy 2023, 1–18. [Google Scholar] [CrossRef] [PubMed]
  123. Conte, M.; Franceschi, C.; Sandri, M.; Salvioli, S. Perilipin 2 and Age-Related Metabolic Diseases: A New Perspective. Trends Endocrinol. Metab. 2016, 27, 893–903. [Google Scholar] [CrossRef] [PubMed]
  124. Wolins, N.E.; Rubin, B.; Brasaemle, D.L. TIP47 associates with lipid droplets. J. Biol. Chem. 2001, 276, 5101–5108. [Google Scholar] [CrossRef] [Green Version]
  125. Choi, Y.M.; Ajjaji, D.; Fleming, K.D.; Borbat, P.P.; Jenkins, M.L.; Moeller, B.E.; Fernando, S.; Bhatia, S.R.; Freed, J.H.; Burke, J.E.; et al. Structural insights into perilipin 3 membrane association in response to diacylglycerol accumulation. Nat. Commun. 2023, 14, 3204. [Google Scholar] [CrossRef]
  126. Skinner, J.R.; Shew, T.M.; Schwartz, D.M.; Tzekov, A.; Lepus, C.M.; Abumrad, N.A.; Wolins, N.E. Diacylglycerol enrichment of endoplasmic reticulum or lipid droplets recruits perilipin 3/TIP47 during lipid storage and mobilization. J. Biol. Chem. 2009, 284, 30941–30948. [Google Scholar] [CrossRef] [Green Version]
  127. Lee, Y.K.; Sohn, J.H.; Han, J.S.; Park, Y.J.; Jeon, Y.G.; Ji, Y.; Dalen, K.T.; Sztalryd, C.; Kimmel, A.R.; Kim, J.B. Perilipin 3 Deficiency Stimulates Thermogenic Beige Adipocytes Through PPARalpha Activation. Diabetes 2018, 67, 791–804. [Google Scholar] [CrossRef]
  128. Szigeti, A.; Minik, O.; Hocsak, E.; Pozsgai, E.; Boronkai, A.; Farkas, R.; Balint, A.; Bodis, J.; Sumegi, B.; Bellyei, S. Preliminary study of TIP47 as a possible new biomarker of cervical dysplasia and invasive carcinoma. Anticancer Res. 2009, 29, 717–724. [Google Scholar]
  129. Wang, K.; Ruan, H.; Song, Z.; Cao, Q.; Bao, L.; Liu, D.; Xu, T.; Xiao, H.; Wang, C.; Cheng, G.; et al. PLIN3 is up-regulated and correlates with poor prognosis in clear cell renal cell carcinoma. Urol. Oncol. 2018, 36, 343.e9–343.e19. [Google Scholar] [CrossRef]
  130. Lung, J.; Hung, M.S.; Wang, T.Y.; Chen, K.L.; Luo, C.W.; Jiang, Y.Y.; Wu, S.Y.; Lee, L.W.; Lin, P.Y.; Chen, F.F.; et al. Lipid Droplets in Lung Cancers Are Crucial for the Cell Growth and Starvation Survival. Int. J. Mol. Sci. 2022, 23, 12533. [Google Scholar] [CrossRef]
  131. Lamprou, I.; Kakouratos, C.; Tsolou, A.; Pavlidis, P.; Xanthopoulou, E.T.; Nanos, C.; Tsaroucha, A.; Sivridis, E.; Giatromanolaki, A.; Koukourakis, M.I. Lipophagy-Related Protein Perilipin-3 and Resistance of Prostate Cancer to Radiation Therapy. Int. J. Radiat. Oncol. Biol. Phys. 2022, 113, 401–414. [Google Scholar] [CrossRef]
  132. Lamprou, I.; Tsolou, A.; Kakouratos, C.; Mitrakas, A.G.; Xanthopoulou, E.T.; Kassela, K.; Karakasiliotis, I.; Zois, C.E.; Giatromanolaki, A.; Koukourakis, M.I. Suppressed PLIN3 frequently occurs in prostate cancer, promoting docetaxel resistance via intensified autophagy, an event reversed by chloroquine. Med. Oncol. 2021, 38, 116. [Google Scholar] [CrossRef]
  133. Zhou, L.; Song, Z.; Hu, J.; Liu, L.; Hou, Y.; Zhang, X.; Yang, X.; Chen, K. ACSS3 represses prostate cancer progression through downregulating lipid droplet-associated protein PLIN3. Theranostics 2021, 11, 841–860. [Google Scholar] [CrossRef]
  134. Jarc, E.; Petan, T. Lipid Droplets and the Management of Cellular Stress. Yale J. Biol. Med. 2019, 92, 435–452. [Google Scholar]
  135. Wolins, N.E.; Skinner, J.R.; Schoenfish, M.J.; Tzekov, A.; Bensch, K.G.; Bickel, P.E. Adipocyte protein S3-12 coats nascent lipid droplets. J. Biol. Chem. 2003, 278, 37713–37721. [Google Scholar] [CrossRef] [Green Version]
  136. Scherer, P.E.; Bickel, P.E.; Kotler, M.; Lodish, H.F. Cloning of cell-specific secreted and surface proteins by subtractive antibody screening. Nat. Biotechnol. 1998, 16, 581–586. [Google Scholar] [CrossRef]
  137. Hsieh, K.; Lee, Y.K.; Londos, C.; Raaka, B.M.; Dalen, K.T.; Kimmel, A.R. Perilipin family members preferentially sequester to either triacylglycerol-specific or cholesteryl-ester-specific intracellular lipid storage droplets. J. Cell Sci. 2012, 125, 4067–4076. [Google Scholar] [CrossRef] [Green Version]
  138. Kraemer, F.B.; Khor, V.K.; Shen, W.J.; Azhar, S. Cholesterol ester droplets and steroidogenesis. Mol. Cell Endocrinol. 2013, 371, 15–19. [Google Scholar] [CrossRef] [Green Version]
  139. Chen, W.; Chang, B.; Wu, X.; Li, L.; Sleeman, M.; Chan, L. Inactivation of Plin4 downregulates Plin5 and reduces cardiac lipid accumulation in mice. Am. J. Physiol. Endocrinol. Metab. 2013, 304, E770–E779. [Google Scholar] [CrossRef] [Green Version]
  140. Richardson, K.; Louie-Gao, Q.; Arnett, D.K.; Parnell, L.D.; Lai, C.Q.; Davalos, A.; Fox, C.S.; Demissie, S.; Cupples, L.A.; Fernandez-Hernando, C.; et al. The PLIN4 variant rs8887 modulates obesity related phenotypes in humans through creation of a novel miR-522 seed site. PLoS ONE 2011, 6, e17944. [Google Scholar] [CrossRef] [Green Version]
  141. Sirois, I.; Aguilar-Mahecha, A.; Lafleur, J.; Fowler, E.; Vu, V.; Scriver, M.; Buchanan, M.; Chabot, C.; Ramanathan, A.; Balachandran, B.; et al. A Unique Morphological Phenotype in Chemoresistant Triple-Negative Breast Cancer Reveals Metabolic Reprogramming and PLIN4 Expression as a Molecular Vulnerability. Mol. Cancer Res. 2019, 17, 2492–2507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Bickel, P.E.; Tansey, J.T.; Welte, M.A. PAT proteins, an ancient family of lipid droplet proteins that regulate cellular lipid stores. Biochim. Biophys. Acta 2009, 1791, 419–440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Hashani, M.; Witzel, H.R.; Pawella, L.M.; Lehmann-Koch, J.; Schumacher, J.; Mechtersheimer, G.; Schnolzer, M.; Schirmacher, P.; Roth, W.; Straub, B.K. Widespread expression of perilipin 5 in normal human tissues and in diseases is restricted to distinct lipid droplet subpopulations. Cell Tissue Res. 2018, 374, 121–136. [Google Scholar] [CrossRef] [PubMed]
  144. Kimmel, A.R.; Sztalryd, C. Perilipin 5, a lipid droplet protein adapted to mitochondrial energy utilization. Curr. Opin. Lipidol. 2014, 25, 110–117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Laurens, C.; Bourlier, V.; Mairal, A.; Louche, K.; Badin, P.M.; Mouisel, E.; Montagner, A.; Marette, A.; Tremblay, A.; Weisnagel, J.S.; et al. Perilipin 5 fine-tunes lipid oxidation to metabolic demand and protects against lipotoxicity in skeletal muscle. Sci. Rep. 2016, 6, 38310. [Google Scholar] [CrossRef] [Green Version]
  146. Lin, J.; Chen, A. Perilipin 5 restores the formation of lipid droplets in activated hepatic stellate cells and inhibits their activation. Lab. Investig. 2016, 96, 791–806. [Google Scholar] [CrossRef] [Green Version]
  147. Zheng, P.; Xie, Z.; Yuan, Y.; Sui, W.; Wang, C.; Gao, X.; Zhao, Y.; Zhang, F.; Gu, Y.; Hu, P.; et al. Plin5 alleviates myocardial ischaemia/reperfusion injury by reducing oxidative stress through inhibiting the lipolysis of lipid droplets. Sci. Rep. 2017, 7, 42574. [Google Scholar] [CrossRef] [Green Version]
  148. Pollak, N.M.; Jaeger, D.; Kolleritsch, S.; Zimmermann, R.; Zechner, R.; Lass, A.; Haemmerle, G. The interplay of protein kinase A and perilipin 5 regulates cardiac lipolysis. J. Biol. Chem. 2015, 290, 1295–1306. [Google Scholar] [CrossRef] [Green Version]
  149. Zhang, X.; Xu, W.; Xu, R.; Wang, Z.; Zhang, X.; Wang, P.; Peng, K.; Li, M.; Li, J.; Tan, Y.; et al. Plin5 Bidirectionally Regulates Lipid Metabolism in Oxidative Tissues. Oxid. Med. Cell Longev. 2022, 2022, 4594956. [Google Scholar] [CrossRef]
  150. Asimakopoulou, A.; Vucur, M.; Luedde, T.; Schneiders, S.; Kalampoka, S.; Weiss, T.S.; Weiskirchen, R. Perilipin 5 and Lipocalin 2 Expression in Hepatocellular Carcinoma. Cancers 2019, 11, 385. [Google Scholar] [CrossRef] [Green Version]
  151. Huang, R.; Yan, G.; Sun, H.; Zhang, J.; Song, D.; Kong, R.; Yan, P.; Hu, P.; Xie, A.; Wang, S.; et al. Identification of prognostic and metastasis-related alternative splicing signatures in hepatocellular carcinoma. Biosci. Rep. 2020, 40, BSR20201001. [Google Scholar] [CrossRef]
  152. Tyagi, S.; Gupta, P.; Saini, A.S.; Kaushal, C.; Sharma, S. The peroxisome proliferator-activated receptor: A family of nuclear receptors role in various diseases. J. Adv. Pharm. Technol. Res. 2011, 2, 236–240. [Google Scholar] [CrossRef]
  153. Arimura, N.; Horiba, T.; Imagawa, M.; Shimizu, M.; Sato, R. The peroxisome proliferator-activated receptor gamma regulates expression of the perilipin gene in adipocytes. J. Biol. Chem. 2004, 279, 10070–10076. [Google Scholar] [CrossRef] [Green Version]
  154. Wolins, N.E.; Quaynor, B.K.; Skinner, J.R.; Tzekov, A.; Croce, M.A.; Gropler, M.C.; Varma, V.; Yao-Borengasser, A.; Rasouli, N.; Kern, P.A.; et al. OXPAT/PAT-1 is a PPAR-induced lipid droplet protein that promotes fatty acid utilization. Diabetes 2006, 55, 3418–3428. [Google Scholar] [CrossRef] [Green Version]
  155. Choudhary, N.S.; Kumar, N.; Duseja, A. Peroxisome Proliferator-Activated Receptors and Their Agonists in Nonalcoholic Fatty Liver Disease. J. Clin. Exp. Hepatol. 2019, 9, 731–739. [Google Scholar] [CrossRef] [Green Version]
  156. Burri, L.; Thoresen, G.H.; Berge, R.K. The Role of PPARα Activation in Liver and Muscle. PPAR Res. 2010, 2010, 542359. [Google Scholar] [CrossRef] [Green Version]
  157. Hernandez-Quiles, M.; Broekema, M.F.; Kalkhoven, E. PPARgamma in Metabolism, Immunity, and Cancer: Unified and Diverse Mechanisms of Action. Front. Endocrinol. 2021, 12, 624112. [Google Scholar] [CrossRef]
  158. Hamza, M.S.; Pott, S.; Vega, V.B.; Thomsen, J.S.; Kandhadayar, G.S.; Ng, P.W.; Chiu, K.P.; Pettersson, S.; Wei, C.L.; Ruan, Y.; et al. De-novo identification of PPARgamma/RXR binding sites and direct targets during adipogenesis. PLoS ONE 2009, 4, e4907. [Google Scholar] [CrossRef] [Green Version]
  159. Grygiel-Gorniak, B. Peroxisome proliferator-activated receptors and their ligands: Nutritional and clinical implications—A review. Nutr. J. 2014, 13, 17. [Google Scholar] [CrossRef] [Green Version]
  160. Giulitti, F.; Petrungaro, S.; Mandatori, S.; Tomaipitinca, L.; de Franchis, V.; D’Amore, A.; Filippini, A.; Gaudio, E.; Ziparo, E.; Giampietri, C. Anti-tumor Effect of Oleic Acid in Hepatocellular Carcinoma Cell Lines via Autophagy Reduction. Front. Cell Dev. Biol. 2021, 9, 629182. [Google Scholar] [CrossRef]
  161. Azukisawa, S.; Zheng, J.; Guo, X.; Ura, H.; Niida, Y.; Itoh, T.; Yamada, S. The differential expression of perilipin-2 in hepatoblastoma and its association with prognosis. Histol. Histopathol. 2021, 36, 1169–1178. [Google Scholar] [CrossRef] [PubMed]
  162. De Lima-Souza, R.A.; Rodrigues, N.M.; Scarini, J.F.; Silva, M.F.S.; Tincani, A.J.; Egal, E.S.A.; Altemani, A.; Mariano, F.V. Metabolic alterations in carcinoma ex pleomorphic adenoma development of lacrimal glands. Int. Ophthalmol. 2022, 42, 1101–1109. [Google Scholar] [CrossRef] [PubMed]
  163. Shang, C.; Li, Y.; He, T.; Liao, Y.; Du, Q.; Wang, P.; Qiao, J.; Guo, H. The prognostic miR-532-5p-correlated ceRNA-mediated lipid droplet accumulation drives nodal metastasis of cervical cancer. J. Adv. Res. 2022, 37, 169–184. [Google Scholar] [CrossRef] [PubMed]
  164. Rios Garcia, M.; Meissburger, B.; Chan, J.; de Guia, R.M.; Mattijssen, F.; Roessler, S.; Birkenfeld, A.L.; Raschzok, N.; Riols, F.; Tokarz, J.; et al. Trip13 Depletion in Liver Cancer Induces a Lipogenic Response Contributing to Plin2-Dependent Mitotic Cell Death. Adv. Sci. 2022, 9, e2104291. [Google Scholar] [CrossRef] [PubMed]
  165. Avgerinos, K.I.; Spyrou, N.; Mantzoros, C.S.; Dalamaga, M. Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metabolism 2019, 92, 121–135. [Google Scholar] [CrossRef]
  166. Pati, S.; Irfan, W.; Jameel, A.; Ahmed, S.; Shahid, R.K. Obesity and Cancer: A Current Overview of Epidemiology, Pathogenesis, Outcomes, and Management. Cancers 2023, 15, 485. [Google Scholar] [CrossRef]
  167. Mukherjee, A.; Bilecz, A.J.; Lengyel, E. The adipocyte microenvironment and cancer. Cancer Metastasis Rev. 2022, 41, 575–587. [Google Scholar] [CrossRef]
  168. Nieman, K.M.; Romero, I.L.; Van Houten, B.; Lengyel, E. Adipose tissue and adipocytes support tumorigenesis and metastasis. Biochim. Biophys. Acta 2013, 1831, 1533–1541. [Google Scholar] [CrossRef] [Green Version]
  169. McGraw, D.W.; Liggett, S.B. Molecular mechanisms of beta2-adrenergic receptor function and regulation. Proc. Am. Thorac. Soc. 2005, 2, 292–296, discussion 292–311. [Google Scholar] [CrossRef] [Green Version]
  170. Silva, D.; Kacprzak, K.; Quintas, C.; Goncalves, J.; Fresco, P. Activation of beta-Adrenoceptors Promotes Lipid Droplet Accumulation in MCF-7 Breast Cancer Cells via cAMP/PKA/EPAC Pathways. Int. J. Mol. Sci. 2023, 24, 767. [Google Scholar] [CrossRef]
  171. Amaro, F.; Silva, D.; Reguengo, H.; Oliveira, J.C.; Quintas, C.; Vale, N.; Goncalves, J.; Fresco, P. beta-Adrenoceptor Activation in Breast MCF-10A Cells Induces a Pattern of Catecholamine Production Similar to that of Tumorigenic MCF-7 Cells. Int. J. Mol. Sci. 2020, 21, 7968. [Google Scholar] [CrossRef]
  172. Silva, D.; Quintas, C.; Goncalves, J.; Fresco, P. Contribution of adrenergic mechanisms for the stress-induced breast cancer carcinogenesis. J. Cell Physiol. 2022, 237, 2107–2127. [Google Scholar] [CrossRef]
  173. Montoya, A.; Amaya, C.N.; Belmont, A.; Diab, N.; Trevino, R.; Villanueva, G.; Rains, S.; Sanchez, L.A.; Badri, N.; Otoukesh, S.; et al. Use of non-selective beta-blockers is associated with decreased tumor proliferative indices in early stage breast cancer. Oncotarget 2017, 8, 6446–6460. [Google Scholar] [CrossRef] [Green Version]
  174. Gervois, P.; Fruchart, J.C.; Staels, B. Drug Insight: Mechanisms of action and therapeutic applications for agonists of peroxisome proliferator-activated receptors. Nat. Clin. Pract. Endocrinol. Metab. 2007, 3, 145–156. [Google Scholar] [CrossRef]
  175. Curry, A.M.; White, D.S.; Donu, D.; Cen, Y. Human Sirtuin Regulators: The “Success” Stories. Front. Physiol. 2021, 12, 752117. [Google Scholar] [CrossRef]
  176. Chen, M.; Huang, L.; Lv, Y.; Li, L.; Dong, Q. Sulforaphane protects against oxidative stress-induced apoptosis via activating SIRT1 in mouse osteoarthritis. Mol. Med. Rep. 2021, 24, 612. [Google Scholar] [CrossRef]
  177. Zhou, G.; Myers, R.; Li, Y.; Chen, Y.; Shen, X.; Fenyk-Melody, J.; Wu, M.; Ventre, J.; Doebber, T.; Fujii, N.; et al. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Investig. 2001, 108, 1167–1174. [Google Scholar] [CrossRef]
  178. Smith, U. Adrenergic control of lipid metabolism. Acta Med. Scand. Suppl. 1983, 672, 41–44. [Google Scholar] [CrossRef]
  179. Hong, F.; Pan, S.; Guo, Y.; Xu, P.; Zhai, Y. PPARs as Nuclear Receptors for Nutrient and Energy Metabolism. Molecules 2019, 24, 2545. [Google Scholar] [CrossRef] [Green Version]
  180. Hong, F.; Xu, P.; Zhai, Y. The Opportunities and Challenges of Peroxisome Proliferator-Activated Receptors Ligands in Clinical Drug Discovery and Development. Int. J. Mol. Sci. 2018, 19, 2189. [Google Scholar] [CrossRef] [Green Version]
  181. Lalloyer, F.; Staels, B. Fibrates, glitazones, and peroxisome proliferator-activated receptors. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 894–899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Staels, B.; Dallongeville, J.; Auwerx, J.; Schoonjans, K.; Leitersdorf, E.; Fruchart, J.C. Mechanism of action of fibrates on lipid and lipoprotein metabolism. Circulation 1998, 98, 2088–2093. [Google Scholar] [CrossRef] [Green Version]
  183. Rakhshandehroo, M.; Knoch, B.; Muller, M.; Kersten, S. Peroxisome proliferator-activated receptor alpha target genes. PPAR Res. 2010, 2010, 612089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Tan, Y.; Wang, M.; Yang, K.; Chi, T.; Liao, Z.; Wei, P. PPAR-alpha Modulators as Current and Potential Cancer Treatments. Front. Oncol. 2021, 11, 599995. [Google Scholar] [CrossRef] [PubMed]
  185. Ahsan, W. The Journey of Thiazolidinediones as Modulators of PPARs for the Management of Diabetes: A Current Perspective. Curr. Pharm. Des. 2019, 25, 2540–2554. [Google Scholar] [CrossRef]
  186. Barak, Y.; Nelson, M.C.; Ong, E.S.; Jones, Y.Z.; Ruiz-Lozano, P.; Chien, K.R.; Koder, A.; Evans, R.M. PPAR gamma is required for placental, cardiac, and adipose tissue development. Mol. Cell 1999, 4, 585–595. [Google Scholar] [CrossRef]
  187. American Association of Neurological Surgeons (AANS); American Society of Neuroradiology (ASNR); Cardiovascular and Interventional Radiology Society of Europe (CIRSE); Canadian Interventional Radiology Association (CIRA); Congress of Neurological Surgeons (CNS); European Society of Minimally Invasive Neurological Therapy (ESMINT); European Society of Neuroradiology (ESNR); European Stroke Organization (ESO); Society for Cardiovascular Angiography and Interventions (SCAI); Society of Interventional Radiology (SIR); et al. Multisociety Consensus Quality Improvement Revised Consensus Statement for Endovascular Therapy of Acute Ischemic Stroke. Int. J. Stroke 2018, 13, 612–632. [Google Scholar]
  188. De la Rosa Rodriguez, M.A.; Kersten, S. Regulation of lipid droplet-associated proteins by peroxisome proliferator-activated receptors. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2017, 1862, 1212–1220. [Google Scholar] [CrossRef]
  189. Biondo, L.A.; Teixeira, A.A.S.; de OS Ferreira, K.C.; Neto, J.C.R. Pharmacological Strategies for Insulin Sensitivity in Obesity and Cancer: Thiazolidinediones and Metformin. Curr. Pharm. Des. 2020, 26, 932–945. [Google Scholar] [CrossRef]
  190. Reka, A.K.; Goswami, M.T.; Krishnapuram, R.; Standiford, T.J.; Keshamouni, V.G. Molecular cross-regulation between PPAR-gamma and other signaling pathways: Implications for lung cancer therapy. Lung Cancer 2011, 72, 154–159. [Google Scholar] [CrossRef] [Green Version]
  191. Augimeri, G.; Gelsomino, L.; Plastina, P.; Giordano, C.; Barone, I.; Catalano, S.; Ando, S.; Bonofiglio, D. Natural and Synthetic PPARgamma Ligands in Tumor Microenvironment: A New Potential Strategy against Breast Cancer. Int. J. Mol. Sci. 2020, 21, 9721. [Google Scholar] [CrossRef]
  192. Chi, T.; Wang, M.; Wang, X.; Yang, K.; Xie, F.; Liao, Z.; Wei, P. PPAR-gamma Modulators as Current and Potential Cancer Treatments. Front. Oncol. 2021, 11, 737776. [Google Scholar] [CrossRef]
  193. Jiang, C.; Ting, A.T.; Seed, B. PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature 1998, 391, 82–86. [Google Scholar] [CrossRef]
  194. Consoli, A.; Devangelio, E. Thiazolidinediones and inflammation. Lupus 2005, 14, 794–797. [Google Scholar] [CrossRef]
  195. Hawley, S.A.; Fullerton, M.D.; Ross, F.A.; Schertzer, J.D.; Chevtzoff, C.; Walker, K.J.; Peggie, M.W.; Zibrova, D.; Green, K.A.; Mustard, K.J.; et al. The ancient drug salicylate directly activates AMP-activated protein kinase. Science 2012, 336, 918–922. [Google Scholar] [CrossRef] [Green Version]
  196. Su, Y.F.; Yang, S.H.; Lee, Y.H.; Wu, B.C.; Huang, S.C.; Liu, C.M.; Chen, S.L.; Pan, Y.F.; Chou, S.S.; Chou, M.Y.; et al. Aspirin-induced inhibition of adipogenesis was p53-dependent and associated with inactivation of pentose phosphate pathway. Eur. J. Pharmacol. 2014, 738, 101–110. [Google Scholar] [CrossRef]
  197. Hsieh, C.C.; Huang, Y.S. Aspirin Breaks the Crosstalk between 3T3-L1 Adipocytes and 4T1 Breast Cancer Cells by Regulating Cytokine Production. PLoS ONE 2016, 11, e0147161. [Google Scholar] [CrossRef]
  198. Roy, S.; Bhowmik, D.R.; Begum, R.; Amin, M.T.; Islam, M.A.; Ahmed, F.; Hossain, M.S. Aspirin attenuates the expression of adhesion molecules, risk of obesity, and adipose tissue inflammation in high-fat diet-induced obese mice. Prostaglandins Other Lipid Mediat. 2022, 162, 106664. [Google Scholar] [CrossRef]
  199. Di Minno, A.; Porro, B.; Turnu, L.; Manega, C.M.; Eligini, S.; Barbieri, S.; Chiesa, M.; Poggio, P.; Squellerio, I.; Anesi, A.; et al. Untargeted Metabolomics to Go beyond the Canonical Effect of Acetylsalicylic Acid. J. Clin. Med. 2019, 9, 51. [Google Scholar] [CrossRef] [Green Version]
  200. Gu, Q.; Gu, Y.; Yang, H.; Shi, Q. Metformin Enhances Osteogenesis and Suppresses Adipogenesis of Human Chorionic Villous Mesenchymal Stem Cells. Tohoku J. Exp. Med. 2017, 241, 13–19. [Google Scholar] [CrossRef] [Green Version]
  201. Yerevanian, A.; Soukas, A.A. Metformin: Mechanisms in Human Obesity and Weight Loss. Curr. Obes. Rep. 2019, 8, 156–164. [Google Scholar] [CrossRef] [PubMed]
  202. Owen, M.R.; Doran, E.; Halestrap, A.P. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem. J. 2000, 348 Pt 3, 607–614. [Google Scholar] [CrossRef] [PubMed]
  203. Stefan, N.; Stumvoll, M. Adiponectin—Its role in metabolism and beyond. Horm. Metab. Res. 2002, 34, 469–474. [Google Scholar] [CrossRef] [PubMed]
  204. Fang, H.; Judd, R.L. Adiponectin Regulation and Function. Compr. Physiol. 2018, 8, 1031–1063. [Google Scholar] [CrossRef] [PubMed]
  205. Sun, C.; Mao, S.; Chen, S.; Zhang, W.; Liu, C. PPARs-Orchestrated Metabolic Homeostasis in the Adipose Tissue. Int. J. Mol. Sci. 2021, 22, 8974. [Google Scholar] [CrossRef]
  206. Lee, M.H.; Klein, R.L.; El-Shewy, H.M.; Luttrell, D.K.; Luttrell, L.M. The adiponectin receptors AdipoR1 and AdipoR2 activate ERK1/2 through a Src/Ras-dependent pathway and stimulate cell growth. Biochemistry 2008, 47, 11682–11692. [Google Scholar] [CrossRef] [Green Version]
  207. Almabouada, F.; Diaz-Ruiz, A.; Rabanal-Ruiz, Y.; Peinado, J.R.; Vazquez-Martinez, R.; Malagon, M.M. Adiponectin receptors form homomers and heteromers exhibiting distinct ligand binding and intracellular signaling properties. J. Biol. Chem. 2013, 288, 3112–3125. [Google Scholar] [CrossRef] [Green Version]
  208. Vasiliauskaite-Brooks, I.; Sounier, R.; Rochaix, P.; Bellot, G.; Fortier, M.; Hoh, F.; De Colibus, L.; Bechara, C.; Saied, E.M.; Arenz, C.; et al. Structural insights into adiponectin receptors suggest ceramidase activity. Nature 2017, 544, 120–123. [Google Scholar] [CrossRef] [Green Version]
  209. Kubota, N.; Yano, W.; Kubota, T.; Yamauchi, T.; Itoh, S.; Kumagai, H.; Kozono, H.; Takamoto, I.; Okamoto, S.; Shiuchi, T.; et al. Adiponectin stimulates AMP-activated protein kinase in the hypothalamus and increases food intake. Cell Metab. 2007, 6, 55–68. [Google Scholar] [CrossRef] [Green Version]
  210. Nehme, R.; Diab-Assaf, M.; Decombat, C.; Delort, L.; Caldefie-Chezet, F. Targeting Adiponectin in Breast Cancer. Biomedicines 2022, 10, 2958. [Google Scholar] [CrossRef]
  211. Zocchi, M.; Della Porta, M.; Lombardoni, F.; Scrimieri, R.; Zuccotti, G.V.; Maier, J.A.; Cazzola, R. A Potential Interplay between HDLs and Adiponectin in Promoting Endothelial Dysfunction in Obesity. Biomedicines 2022, 10, 1344. [Google Scholar] [CrossRef]
  212. Tanyanskiy, D.A.; Shavva, V.S.; Dizhe, E.B.; Oleinikova, G.N.; Lizunov, A.V.; Nekrasova, E.V.; Mogilenko, D.A.; Larionova, E.E.; Orlov, S.V.; Denisenko, A.D. Adiponectin Stimulates Apolipoprotein A-1 Gene Expression in HepG2 Cells via AMPK, PPARalpha, and LXRs Signaling Mechanisms. Biochemistry 2022, 87, 1252–1259. [Google Scholar] [CrossRef]
  213. Vales-Villamarin, C.; Lumpuy-Castillo, J.; Gavela-Perez, T.; de Dios, O.; Perez-Nadador, I.; Soriano-Guillen, L.; Garces, C. Sex-Dependent Mediation of Leptin in the Association of Perilipin Polymorphisms with BMI and Plasma Lipid Levels in Children. Nutrients 2022, 14, 3072. [Google Scholar] [CrossRef]
  214. Byeon, J.S.; Jeong, J.Y.; Kim, M.J.; Lee, S.M.; Nam, W.H.; Myung, S.J.; Kim, J.G.; Yang, S.K.; Kim, J.H.; Suh, D.J. Adiponectin and adiponectin receptor in relation to colorectal cancer progression. Int. J. Cancer 2010, 127, 2758–2767. [Google Scholar] [CrossRef]
  215. Karnati, H.K.; Panigrahi, M.K.; Li, Y.; Tweedie, D.; Greig, N.H. Adiponectin as a Potential Therapeutic Target for Prostate Cancer. Curr. Pharm. Des. 2017, 23, 4170–4179. [Google Scholar] [CrossRef] [Green Version]
  216. Engin, A. Adiponectin-Resistance in Obesity. Adv. Exp. Med. Biol. 2017, 960, 415–441. [Google Scholar] [CrossRef]
  217. Parida, S.; Siddharth, S.; Sharma, D. Adiponectin, Obesity, and Cancer: Clash of the Bigwigs in Health and Disease. Int. J. Mol. Sci. 2019, 20, 2519. [Google Scholar] [CrossRef] [Green Version]
  218. Matsuzawa, Y. Adiponectin: A key player in obesity related disorders. Curr. Pharm. Des. 2010, 16, 1896–1901. [Google Scholar] [CrossRef]
  219. Shehzad, A.; Iqbal, W.; Shehzad, O.; Lee, Y.S. Adiponectin: Regulation of its production and its role in human diseases. Hormones 2012, 11, 8–20. [Google Scholar] [CrossRef]
  220. Nagaraju, G.P.; Rajitha, B.; Aliya, S.; Kotipatruni, R.P.; Madanraj, A.S.; Hammond, A.; Park, D.; Chigurupati, S.; Alam, A.; Pattnaik, S. The role of adiponectin in obesity-associated female-specific carcinogenesis. Cytokine Growth Factor Rev. 2016, 31, 37–48. [Google Scholar] [CrossRef]
  221. Padmalayam, I.; Suto, M. Role of adiponectin in the metabolic syndrome: Current perspectives on its modulation as a treatment strategy. Curr. Pharm. Des. 2013, 19, 5755–5763. [Google Scholar] [CrossRef] [PubMed]
  222. Robinson, K.; Prins, J.; Venkatesh, B. Clinical review: Adiponectin biology and its role in inflammation and critical illness. Crit. Care 2011, 15, 221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Dalamaga, M.; Diakopoulos, K.N.; Mantzoros, C.S. The role of adiponectin in cancer: A review of current evidence. Endocr. Rev. 2012, 33, 547–594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Yang, Y.; Liu, Y.; Wang, Y.; Chao, Y.; Zhang, J.; Jia, Y.; Tie, J.; Hu, D. Regulation of SIRT1 and Its Roles in Inflammation. Front. Immunol. 2022, 13, 831168. [Google Scholar] [CrossRef] [PubMed]
  225. Gallardo-Montejano, V.I.; Saxena, G.; Kusminski, C.M.; Yang, C.; McAfee, J.L.; Hahner, L.; Hoch, K.; Dubinsky, W.; Narkar, V.A.; Bickel, P.E. Nuclear Perilipin 5 integrates lipid droplet lipolysis with PGC-1alpha/SIRT1-dependent transcriptional regulation of mitochondrial function. Nat. Commun. 2016, 7, 12723. [Google Scholar] [CrossRef] [Green Version]
  226. Tian, S.; Lei, P.; Teng, C.; Sun, Y.; Song, X.; Li, B.; Shan, Y. Targeting PLIN2/PLIN5-PPARgamma: Sulforaphane Disturbs the Maturation of Lipid Droplets. Mol. Nutr. Food Res. 2019, 63, e1900183. [Google Scholar] [CrossRef]
  227. Hoffmann, E.; Wald, J.; Lavu, S.; Roberts, J.; Beaumont, C.; Haddad, J.; Elliott, P.; Westphal, C.; Jacobson, E. Pharmacokinetics and tolerability of SRT2104, a first-in-class small molecule activator of SIRT1, after single and repeated oral administration in man. Br. J. Clin. Pharmacol. 2013, 75, 186–196. [Google Scholar] [CrossRef] [Green Version]
  228. Ko, J.H.; Sethi, G.; Um, J.Y.; Shanmugam, M.K.; Arfuso, F.; Kumar, A.P.; Bishayee, A.; Ahn, K.S. The Role of Resveratrol in Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 2589. [Google Scholar] [CrossRef] [Green Version]
  229. Ishikawa, Y.; Homcy, C.J. The adenylyl cyclases as integrators of transmembrane signal transduction. Circ. Res. 1997, 80, 297–304. [Google Scholar] [CrossRef]
  230. Vasudevan, N.T.; Mohan, M.L.; Goswami, S.K.; Naga Prasad, S.V. Regulation of beta-adrenergic receptor function: An emphasis on receptor resensitization. Cell Cycle 2011, 10, 3684–3691. [Google Scholar] [CrossRef] [Green Version]
  231. Collins, S. beta-Adrenoceptor Signaling Networks in Adipocytes for Recruiting Stored Fat and Energy Expenditure. Front. Endocrinol. 2011, 2, 102. [Google Scholar] [CrossRef] [Green Version]
  232. Smith, G.I.; Atherton, P.; Reeds, D.N.; Mohammed, B.S.; Rankin, D.; Rennie, M.J.; Mittendorfer, B. Omega-3 polyunsaturated fatty acids augment the muscle protein anabolic response to hyperinsulinaemia-hyperaminoacidaemia in healthy young and middle-aged men and women. Clin. Sci. 2011, 121, 267–278. [Google Scholar] [CrossRef] [Green Version]
  233. Marchant-Forde, J.N.; Lay, D.C., Jr.; Marchant-Forde, R.M.; McMunn, K.A.; Richert, B.T. The effects of R-salbutamol on growth, carcass measures, and health of finishing pigs. J. Anim. Sci. 2012, 90, 4081–4089. [Google Scholar] [CrossRef]
  234. Hostrup, M.; Jacobson, G.A.; Jessen, S.; Lemminger, A.K. Anabolic and lipolytic actions of beta(2) -agonists in humans and antidoping challenges. Drug Test. Anal. 2020, 12, 597–609. [Google Scholar] [CrossRef]
  235. Piribauer, M.; Jiang, L.; Kostov, T.; Parr, M.; Steidel, S.; Bizjak, D.A.; Steinacker, J.M.; Diel, P. Combinatory in vitro effects of the beta2-agonists salbutamol and formoterol in skeletal muscle cells. Toxicol. Lett. 2023, 378, 10–18. [Google Scholar] [CrossRef]
  236. Re, G.; Badino, P.; Girardi, C.; Di Carlo, F. Effects of a beta 2-agonist (clenbuterol) on cultured human (CG-5) breast cancer cells. Pharmacol. Res. 1992, 26, 377–384. [Google Scholar] [CrossRef]
  237. Montoya, A.; Varela-Ramirez, A.; Dickerson, E.; Pasquier, E.; Torabi, A.; Aguilera, R.; Nahleh, Z.; Bryan, B. The beta adrenergic receptor antagonist propranolol alters mitogenic and apoptotic signaling in late stage breast cancer. Biomed. J. 2019, 42, 155–165. [Google Scholar] [CrossRef]
  238. Phadke, S.; Clamon, G. Beta blockade as adjunctive breast cancer therapy: A review. Crit. Rev. Oncol. Hematol. 2019, 138, 173–177. [Google Scholar] [CrossRef]
  239. Chang, H.; Lee, S.H. Beta-adrenergic receptor blockers and hepatocellular carcinoma survival: A systemic review and meta-analysis. Clin. Exp. Med. 2023, 23, 853–858. [Google Scholar] [CrossRef]
  240. Kocak, M.Z.; Er, M.; Ugrakli, M.; Hendem, E.; Araz, M.; Eryilmaz, M.K.; Artac, M. Could the concomitant use of beta blockers with bevacizumab improve survival in metastatic colon cancer? Eur. J. Clin. Pharmacol. 2023, 79, 485–491. [Google Scholar] [CrossRef]
  241. Falcinelli, M.; Al-Hity, G.; Baron, S.; Mampay, M.; Allen, M.C.; Samuels, M.; Jones, W.; Cilibrasi, C.; Flaherty, R.L.; Giamas, G.; et al. Propranolol reduces IFN-gamma driven PD-L1 immunosuppression and improves anti-tumour immunity in ovarian cancer. Brain Behav. Immun. 2023, 110, 1–12. [Google Scholar] [CrossRef] [PubMed]
  242. Knight, J.M.; Kerswill, S.A.; Hari, P.; Cole, S.W.; Logan, B.R.; D’Souza, A.; Shah, N.N.; Horowitz, M.M.; Stolley, M.R.; Sloan, E.K.; et al. Repurposing existing medications as cancer therapy: Design and feasibility of a randomized pilot investigating propranolol administration in patients receiving hematopoietic cell transplantation. BMC Cancer 2018, 18, 593. [Google Scholar] [CrossRef] [PubMed]
  243. Shamim, M.A.; Shahid, A.; Sardar, P.K.; Yeung, S.; Reyes, J.; Kim, J.; Parsa, C.; Orlando, R.; Wang, J.; Kelly, K.M.; et al. Transfersome Encapsulated with the R-carvedilol Enantiomer for Skin Cancer Chemoprevention. Nanomaterials 2023, 13, 929. [Google Scholar] [CrossRef] [PubMed]
  244. Nimura, S.; Yamaguchi, T.; Ueda, K.; Kadokura, K.; Aiuchi, T.; Kato, R.; Obama, T.; Itabe, H. Olanzapine promotes the accumulation of lipid droplets and the expression of multiple perilipins in human adipocytes. Biochem. Biophys. Res. Commun. 2015, 467, 906–912. [Google Scholar] [CrossRef] [PubMed]
  245. Cottingham, C.M.; Patrick, T.; Richards, M.A.; Blackburn, K.D. Tricyclic antipsychotics promote adipogenic gene expression to potentiate preadipocyte differentiation in vitro. Hum. Cell 2020, 33, 502–511. [Google Scholar] [CrossRef]
  246. Sato, T.; Akimoto, N.; Kitamura, K.; Kurihara, H.; Hayashi, N.; Ito, A. Adapalene suppresses sebum accumulation via the inhibition of triacylglycerol biosynthesis and perilipin expression in differentiated hamster sebocytes in vitro. J. Dermatol. Sci. 2013, 70, 204–210. [Google Scholar] [CrossRef]
  247. Jang, B.C. Artesunate inhibits adipogeneis in 3T3-L1 preadipocytes by reducing the expression and/or phosphorylation levels of C/EBP-alpha, PPAR-gamma, FAS, perilipin A, and STAT-3. Biochem. Biophys. Res. Commun. 2016, 474, 220–225. [Google Scholar] [CrossRef]
  248. Funk, M.I.; Conde, M.A.; Piwien-Pilipuk, G.; Uranga, R.M. Novel antiadipogenic effect of menadione in 3T3-L1 cells. Chem. Biol. Interact. 2021, 343, 109491. [Google Scholar] [CrossRef]
  249. Jang, B.C. Tetrandrine has anti-adipogenic effect on 3T3-L1 preadipocytes through the reduced expression and/or phosphorylation levels of C/EBP-alpha, PPAR-gamma, FAS, perilipin A, and STAT-3. Biochem. Biophys. Res. Commun. 2016, 476, 481–486. [Google Scholar] [CrossRef]
  250. Kovsan, J.; Ben-Romano, R.; Souza, S.C.; Greenberg, A.S.; Rudich, A. Regulation of adipocyte lipolysis by degradation of the perilipin protein: Nelfinavir enhances lysosome-mediated perilipin proteolysis. J. Biol. Chem. 2007, 282, 21704–21711. [Google Scholar] [CrossRef] [Green Version]
  251. Li, J.; Mihalcioiu, M.; Li, L.; Zakikhani, M.; Camirand, A.; Kremer, R. Vitamin D prevents lipid accumulation in murine muscle through regulation of PPARgamma and perilipin-2 expression. J. Steroid Biochem. Mol. Biol. 2018, 177, 116–124. [Google Scholar] [CrossRef]
  252. Hao, L.; Guo, Y.; Peng, Q.; Zhang, Z.; Ji, J.; Liu, Y.; Xue, Y.; Li, C.; Zheng, K.; Shi, X. Dihydroartemisinin reduced lipid droplet deposition by YAP1 to promote the anti-PD-1 effect in hepatocellular carcinoma. Phytomedicine 2022, 96, 153913. [Google Scholar] [CrossRef]
Figure 1. Overview of LD biogenesis in cancer cells. The figure illustrates the possible sources of FAs and the mechanisms involved in LD biogenesis: Free FA uptake can occur through FABPpm (1) and, when combined with lipoproteins, uptake occurs through CD36-mediated internalization, through a clathrin-mediated endocytosis (2). These FA scavenging mechanisms may be complemented by FA de novo synthesis (3). The FAs absorbed/synthetized will be esterified into neutral lipids and can be transported to the ER or to LD. TAG synthesis starts with the activation of FA into Acyl-CoA and follows in the ER by action of the esterification enzymes (4). The newly formed neutral lipids accumulate between the ER bilayers, where LD biogenesis occurs. SEIPIN stabilizes the LD structure while FIT proteins help in the portioning of neutral lipids (5). Once fully formed, the LD will be released into the cytosol, carrying a set of proteins and enzymes for managing the lipid cargo in response to lipolytic stimuli (6). Abbreviations: ACS (acyl-CoA synthetase); AGPAT (acylglycerol-P acyltransferase); ATGL (adipose triacylglyceride lipase); CGI-58 (comparative gene identification 58 protein); DAG (diacylglyceride); DGAT (diacylglycerol acyltransferase); FABPpm (plasma membrane fatty acid-binding proteins); FA (fatty acid); FASN (fatty acid synthase); FIT (fat-storage inducing transmembrane); GLUT (glucose transporter); GPAT (glycerol-P acyltransferase); HSL (hormone sensitive lipase); LPA (lysophosphatidic acid); PA (phosphatidic acid); PAP (phosphatidic acid phosphohydrolase); PLIN (perilipin); TAG (triacylglyceride); TCA cycle (tricarboxylic acid cycle).
Figure 1. Overview of LD biogenesis in cancer cells. The figure illustrates the possible sources of FAs and the mechanisms involved in LD biogenesis: Free FA uptake can occur through FABPpm (1) and, when combined with lipoproteins, uptake occurs through CD36-mediated internalization, through a clathrin-mediated endocytosis (2). These FA scavenging mechanisms may be complemented by FA de novo synthesis (3). The FAs absorbed/synthetized will be esterified into neutral lipids and can be transported to the ER or to LD. TAG synthesis starts with the activation of FA into Acyl-CoA and follows in the ER by action of the esterification enzymes (4). The newly formed neutral lipids accumulate between the ER bilayers, where LD biogenesis occurs. SEIPIN stabilizes the LD structure while FIT proteins help in the portioning of neutral lipids (5). Once fully formed, the LD will be released into the cytosol, carrying a set of proteins and enzymes for managing the lipid cargo in response to lipolytic stimuli (6). Abbreviations: ACS (acyl-CoA synthetase); AGPAT (acylglycerol-P acyltransferase); ATGL (adipose triacylglyceride lipase); CGI-58 (comparative gene identification 58 protein); DAG (diacylglyceride); DGAT (diacylglycerol acyltransferase); FABPpm (plasma membrane fatty acid-binding proteins); FA (fatty acid); FASN (fatty acid synthase); FIT (fat-storage inducing transmembrane); GLUT (glucose transporter); GPAT (glycerol-P acyltransferase); HSL (hormone sensitive lipase); LPA (lysophosphatidic acid); PA (phosphatidic acid); PAP (phosphatidic acid phosphohydrolase); PLIN (perilipin); TAG (triacylglyceride); TCA cycle (tricarboxylic acid cycle).
Cancers 15 04013 g001
Figure 2. Influence of PLIN1 activation on the assembly of lipolytic enzymes in the lipid droplet (LD). Panel (A): In basal state, CGI-58 remains attached to PLIN1 avoiding interaction with ATGL and consequent co-activation of the lipase. The lipid content of the LD stays protected from lipolytic activity. Panel (B): In stimulated state, PKA phosphorylates PLIN1 and the interaction with CGI-58 is broken. CGI-58 binds to phosphorylated ATGL, and the lipase is fully activated. Phosphorylated HSL binds to phosphorylated PLIN1, which allows access to the lipid content of the LD. Abbreviations: ATGL (adipose triacylglyceride lipase); CGI-58 (comparative gene identification 58 protein); HSL (hormone sensitive lipase); P (phosphate); PKA (protein kinase A); PLIN1 (perilipin 1).
Figure 2. Influence of PLIN1 activation on the assembly of lipolytic enzymes in the lipid droplet (LD). Panel (A): In basal state, CGI-58 remains attached to PLIN1 avoiding interaction with ATGL and consequent co-activation of the lipase. The lipid content of the LD stays protected from lipolytic activity. Panel (B): In stimulated state, PKA phosphorylates PLIN1 and the interaction with CGI-58 is broken. CGI-58 binds to phosphorylated ATGL, and the lipase is fully activated. Phosphorylated HSL binds to phosphorylated PLIN1, which allows access to the lipid content of the LD. Abbreviations: ATGL (adipose triacylglyceride lipase); CGI-58 (comparative gene identification 58 protein); HSL (hormone sensitive lipase); P (phosphate); PKA (protein kinase A); PLIN1 (perilipin 1).
Cancers 15 04013 g002
Figure 3. Influence of PLIN2 activation on the assembly and access of lipolytic enzymes in the lipid droplet (LD). Panel (A): In basal state, PLIN2 stays attached to the LD surface, protecting the LD content from the lipase activity as a barrier. Panel (B): In stimulated state, AMPK phosphorylates PLIN2. Phosphorylated PLIN2 binds to Hsc70 and is subsequently carried to the lysosome for chaperone-mediated autophagy degradation. Without PLIN2, the LD is vulnerable to the lipolytic activity of the lipases. Abbreviations: AMPK (AMP-activated protein kinase); ATGL (adipose triacylglyceride lipase); CGI-58 (comparative gene identification 58 protein); Hsc70 (heat shock cognate 70 kDa protein); HSL (hormone sensitive lipase); P (phosphate); PLIN2 (perilipin 2).
Figure 3. Influence of PLIN2 activation on the assembly and access of lipolytic enzymes in the lipid droplet (LD). Panel (A): In basal state, PLIN2 stays attached to the LD surface, protecting the LD content from the lipase activity as a barrier. Panel (B): In stimulated state, AMPK phosphorylates PLIN2. Phosphorylated PLIN2 binds to Hsc70 and is subsequently carried to the lysosome for chaperone-mediated autophagy degradation. Without PLIN2, the LD is vulnerable to the lipolytic activity of the lipases. Abbreviations: AMPK (AMP-activated protein kinase); ATGL (adipose triacylglyceride lipase); CGI-58 (comparative gene identification 58 protein); Hsc70 (heat shock cognate 70 kDa protein); HSL (hormone sensitive lipase); P (phosphate); PLIN2 (perilipin 2).
Cancers 15 04013 g003
Figure 4. Influence of PLIN5 activation on the assembly and access of lipolytic enzymes in the lipid droplet (LD). Panel (A): In basal state, PLIN5 binds to CGI-58 and ATGL, preventing their interaction and consequent lipolytic activity. Panel (B): In stimulated state, PKA phosphorylates PLIN5 and its interaction with CGI-58 and ATGL is undone. CGI-58 binds to phosphorylated ATGL and the lipase is fully activated. Phosphorylated HSL is active and it keeps bonded to phosphorylated PLIN5, which allows access to the lipid content of the LD. Phosphorylated PLIN5 can also travel to the nucleus, where it binds to sirtuin1 and peroxisome proliferator-activated receptor gamma coactivator 1-alpha to activate the transcription of genes related FA catabolism, mitochondrial biogenesis, and respiration. PLIN5 is also known to be a contact site between LDs and mitochondria during β-oxidation. Abbreviations: ATGL (adipose triacylglyceride lipase); CGI-58 (comparative gene identification 58 protein); HSL (hormone sensitive lipase); P (phosphate); PKA (protein kinase A); PLIN5 (perilipin 5).
Figure 4. Influence of PLIN5 activation on the assembly and access of lipolytic enzymes in the lipid droplet (LD). Panel (A): In basal state, PLIN5 binds to CGI-58 and ATGL, preventing their interaction and consequent lipolytic activity. Panel (B): In stimulated state, PKA phosphorylates PLIN5 and its interaction with CGI-58 and ATGL is undone. CGI-58 binds to phosphorylated ATGL and the lipase is fully activated. Phosphorylated HSL is active and it keeps bonded to phosphorylated PLIN5, which allows access to the lipid content of the LD. Phosphorylated PLIN5 can also travel to the nucleus, where it binds to sirtuin1 and peroxisome proliferator-activated receptor gamma coactivator 1-alpha to activate the transcription of genes related FA catabolism, mitochondrial biogenesis, and respiration. PLIN5 is also known to be a contact site between LDs and mitochondria during β-oxidation. Abbreviations: ATGL (adipose triacylglyceride lipase); CGI-58 (comparative gene identification 58 protein); HSL (hormone sensitive lipase); P (phosphate); PKA (protein kinase A); PLIN5 (perilipin 5).
Cancers 15 04013 g004
Figure 5. PLIN’s coordination of FA storage and possible pharmacological intervention strategies. Increase in the body’s need for FAs for β-oxidation is signaled through hormone-dependent PKA activation, which leads to PKA-mediated lipolysis and increase of β-oxidation to fulfill the body’s energy demands. This mechanism is partly orchestrated by the type of PLIN expressed in each cell type. In adipocytes (Box A), PKA induces PLIN1 phosphorylation, allowing lipolytic action over the lipid droplet (LD) lipid content. In β-oxidative cells (Box B), PKA induces PLIN5 phosphorylation, favoring FA transfer to mitochondria and β-oxidation. Conditions of starvation/nutrient scarcity (low ATP/AMP ratio) are signaled through AMPK. AMPK activation induces PLIN2 phosphorylation, allowing the lipolytic action over the LD lipid content (see text for details). The increase in free FA availability will stimulate PPARs. PPARγ activation in adipocytes will stimulate adipogenesis (preadipocyte differentiation) and expression of PLIN1 and PLIN2, increasing the storage capacity and their capacity to react to hormone-induced FA mobilization. PPARγ activation in β-oxidative cells will increase mainly PLIN2 expression, favoring FA storage in LDs and the capacity of these cells to react to local starvation conditions. Increased FA availability will also activate PPARα, which stimulates PLIN5 expression in β-oxidative cells, increasing their β-oxidative capacity to react to hormone stimulation. Pharmacologically, PPARs, PKA, and AMPK can be modulated by widely used drugs such as TZDs (PPARγ agonists), fibrates (PPARα agonists), metformin (AMPK activator), and adrenoceptor agonists and antagonists (modulation of PKA pathway). In the figure, green lines indicate activation and red lines indicate inhibition. Abbreviations: AMPK (AMP-activated protein kinase); FA (fatty acid); LD (lipid droplet); P (phosphate); PKA (protein kinase A); PLIN1 (perilipin 1); PLIN2 (perilipin 2); PLIN5 (perilipin 5); PPARα (peroxisome proliferator-activated receptor alpha); PPARγ (peroxisome proliferator-activated receptor gamma); TZDs (thiazolidinediones).
Figure 5. PLIN’s coordination of FA storage and possible pharmacological intervention strategies. Increase in the body’s need for FAs for β-oxidation is signaled through hormone-dependent PKA activation, which leads to PKA-mediated lipolysis and increase of β-oxidation to fulfill the body’s energy demands. This mechanism is partly orchestrated by the type of PLIN expressed in each cell type. In adipocytes (Box A), PKA induces PLIN1 phosphorylation, allowing lipolytic action over the lipid droplet (LD) lipid content. In β-oxidative cells (Box B), PKA induces PLIN5 phosphorylation, favoring FA transfer to mitochondria and β-oxidation. Conditions of starvation/nutrient scarcity (low ATP/AMP ratio) are signaled through AMPK. AMPK activation induces PLIN2 phosphorylation, allowing the lipolytic action over the LD lipid content (see text for details). The increase in free FA availability will stimulate PPARs. PPARγ activation in adipocytes will stimulate adipogenesis (preadipocyte differentiation) and expression of PLIN1 and PLIN2, increasing the storage capacity and their capacity to react to hormone-induced FA mobilization. PPARγ activation in β-oxidative cells will increase mainly PLIN2 expression, favoring FA storage in LDs and the capacity of these cells to react to local starvation conditions. Increased FA availability will also activate PPARα, which stimulates PLIN5 expression in β-oxidative cells, increasing their β-oxidative capacity to react to hormone stimulation. Pharmacologically, PPARs, PKA, and AMPK can be modulated by widely used drugs such as TZDs (PPARγ agonists), fibrates (PPARα agonists), metformin (AMPK activator), and adrenoceptor agonists and antagonists (modulation of PKA pathway). In the figure, green lines indicate activation and red lines indicate inhibition. Abbreviations: AMPK (AMP-activated protein kinase); FA (fatty acid); LD (lipid droplet); P (phosphate); PKA (protein kinase A); PLIN1 (perilipin 1); PLIN2 (perilipin 2); PLIN5 (perilipin 5); PPARα (peroxisome proliferator-activated receptor alpha); PPARγ (peroxisome proliferator-activated receptor gamma); TZDs (thiazolidinediones).
Cancers 15 04013 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bombarda-Rocha, V.; Silva, D.; Badr-Eddine, A.; Nogueira, P.; Gonçalves, J.; Fresco, P. Challenges in Pharmacological Intervention in Perilipins (PLINs) to Modulate Lipid Droplet Dynamics in Obesity and Cancer. Cancers 2023, 15, 4013. https://doi.org/10.3390/cancers15154013

AMA Style

Bombarda-Rocha V, Silva D, Badr-Eddine A, Nogueira P, Gonçalves J, Fresco P. Challenges in Pharmacological Intervention in Perilipins (PLINs) to Modulate Lipid Droplet Dynamics in Obesity and Cancer. Cancers. 2023; 15(15):4013. https://doi.org/10.3390/cancers15154013

Chicago/Turabian Style

Bombarda-Rocha, Victória, Dany Silva, Allal Badr-Eddine, Patrícia Nogueira, Jorge Gonçalves, and Paula Fresco. 2023. "Challenges in Pharmacological Intervention in Perilipins (PLINs) to Modulate Lipid Droplet Dynamics in Obesity and Cancer" Cancers 15, no. 15: 4013. https://doi.org/10.3390/cancers15154013

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop