Next Article in Journal
Effects of Nesting Material Provision and High-Dose Vitamin C Supplementation during the Peripartum Period on Prepartum Nest-Building Behavior, Farrowing Process, Oxidative Stress Status, Cortisol Levels, and Preovulatory Follicle Development in Hyperprolific Sows
Previous Article in Journal
Iron, Oxidative Stress, and Metabolic Dysfunction—Associated Steatotic Liver Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Perilipins in Oxidative Stress—Implications for Cardiovascular Disease

by
Mathieu Cinato
1,
Linda Andersson
1,
Azra Miljanovic
1,
Marion Laudette
1,
Oksana Kunduzova
2,
Jan Borén
1 and
Malin C. Levin
1,*
1
Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
2
Institute of Metabolic and Cardiovascular Diseases (I2MC), National Institute of Health and Medical Research (INSERM) 1297, Toulouse III University—Paul Sabatier, 31432 Toulouse, France
*
Author to whom correspondence should be addressed.
Antioxidants 2024, 13(2), 209; https://doi.org/10.3390/antiox13020209
Submission received: 9 December 2023 / Revised: 12 January 2024 / Accepted: 1 February 2024 / Published: 7 February 2024

Abstract

:
Oxidative stress is the imbalance between the production of reactive oxygen species (ROS) and antioxidants in a cell. In the heart, oxidative stress may deteriorate calcium handling, cause arrhythmia, and enhance maladaptive cardiac remodeling by the induction of hypertrophic and apoptotic signaling pathways. Consequently, dysregulated ROS production and oxidative stress have been implicated in numerous cardiac diseases, including heart failure, cardiac ischemia–reperfusion injury, cardiac hypertrophy, and diabetic cardiomyopathy. Lipid droplets (LDs) are conserved intracellular organelles that enable the safe and stable storage of neutral lipids within the cytosol. LDs are coated with proteins, perilipins (Plins) being one of the most abundant. In this review, we will discuss the interplay between oxidative stress and Plins. Indeed, LDs and Plins are increasingly being recognized for playing a critical role beyond energy metabolism and lipid handling. Numerous reports suggest that an essential purpose of LD biogenesis is to alleviate cellular stress, such as oxidative stress. Given the yet unmet suitability of ROS as targets for the intervention of cardiovascular disease, the endogenous antioxidant capacity of Plins may be beneficial.

1. Introduction

Myocardial disease remains the leading cause of death and disability worldwide. Despite advances in medical and interventional therapy for myocardial pathologies such as ischemia, valvular heart disease, or hypertension, many surviving patients still develop heart failure. In addition, recent studies show that the incidence of heart failure is increasing in the younger population in parallel with the increasing prevalence of diabetes and obesity [1,2,3].
Oxidative stress is a common denominator in the pathogenesis of myocardial disease. Oxidative stress is the imbalance between the production of reactive oxygen species (ROS) and antioxidants in a cell. In patients with myocardial disease, oxidative stress occurs in the myocardium and is associated with left ventricular dysfunction [4,5,6]. Within the heart, oxidative stress can impair calcium handling, cause arrhythmia, and enhance maladaptive cardiac remodeling by the induction of hypertrophy and apoptosis [7].
Currently, emerging evidence indicates that lipid droplets (LDs) play a critical role in the cellular response to oxidative stress. In this review, we focus on the role of the LD proteins perilipins (Plins) and their role in oxidative stress. We will also discuss the implications of Plins and oxidative stress in cardiovascular disease. For a schematic summary, please see Figure 1.

1.1. ROS within the Heart

Oxidative stress occurs when there is an excessive production of ROS in relation to antioxidant defense. ROS are oxygen-containing reactive species and comprise oxygen free radicals (e.g., superoxide anion radical O2•−, hydroxyl radicals, and peroxyl radicals) as well as non-radicals (e.g., hydrogen peroxide H2O2, hypochlorous acid, and ozone) [22].
In most cell types, and especially in cardiomyocytes, the mitochondrial electron transport chain is the main endosource of ROS production [23]. A fraction of the electrons running through the electron transport chain in the mitochondrial inner membrane are partially reduced to O2•− and are rapidly dismutated to H2O2 by superoxide dismutase (SOD) and then further reduced to H2O by antioxidative enzymes (gluthatione peroxidase, peroxiredoxin, and catalase). In addition to mitochondria, ROS can also be generated by cytosolic sources. One of the most important sources of cytosolic ROS are the NADPH oxidases (NOX) enzymes [24]. NOX proteins produce O2•− through NADPH electron exchange, and NOX-dependent ROS production regulates many metabolic processes and has been implicated in cardiovascular disease [25]. Furthermore, cytosolic ROS are also produced by xanthine oxidase, nitric oxide synthase, monoamine oxidase, cyclooxygenases, and cytochrome P450 enzymes [26].
The tight equilibrium between ROS production and neutralization is ensured either by the regulation of the expression/activity of enzymes producing free radicals or by the endogenous antioxidant system. The latter comprises antioxidant enzymes, such as SOD, catalase, or glutathione peroxidase, as well as small molecules, such as hydrophilic antioxidants and lipophilic radical antioxidants [27]. To protect itself from oxidative stress, the cell can activate the antioxidant response element found within the promoter region of many cytoprotective antioxidants [28]. The antioxidant response element is transcriptionally activated through nuclear translocation/accumulation and the binding of its transcription factor, Nrf2 (NF-E2-related), and thus it is responsible for the regulation of a large panel of antioxidant enzymes [29].
Under physiological conditions, ROS regulate many cellular processes when present at low concentrations, including gene expression, energetic production, substrate oxidation, hormone production, and cellular defenses [30]. Elevated ROS levels overpassing the antioxidant defense can also be beneficial in certain situations (i.e., antimicrobial defense, exercise adaptation). However, excessive ROS production leading to oxidative stress is mainly toxic, resulting in damaged cell constituents and impaired cellular function. In the heart, multiple studies have shown that ROS impair a broad range of cellular functions, including mitochondrial function and biogenesis, mitochondrial permeability transition pore opening, energy metabolism, calcium handling, excitation–contraction coupling, cardiac fibroblast activation, and cell death [31,32,33,34,35,36]. Additionally, oxidative stress due to a reduction in the antioxidant defense has also been identified as a contributing factor to cardiomyocyte dysfunction [26].
Certainly, dysregulated ROS production and oxidative stress trigger maladaptive cardiac remodeling in numerous cardiac diseases, including cardiac ischemia–reperfusion injury, arrhythmia, hypertrophy, and diabetic cardiomyopathy potentially progressing to heart failure [26,37,38].

1.2. Cardiac Dysfunction Promotes Metabolic Abnormalities

In cardiomyocytes, lipid homeostasis depends on a dynamic balance between fatty acid uptake from the surroundings and consumption by mitochondrial β-oxidation. Cardiac dysfunction and remodeling are known to promote metabolic abnormalities [39]. The heart has a very high energy demand and must maintain a continuous production of ATP to sustain contractile function. The healthy heart is metabolically flexible and can easily switch between different energy substrates, such as fatty acids and glucose. However, the failing heart loses this flexibility, resulting in a decreased ability to produce energy through ATP and other high-energy phosphate compounds [40,41,42], by up to 40% [43]. The energy deficit in the failing heart is associated with profound metabolic reprogramming, including an increased uptake of lipids and glucose and a subsequent accumulation of lipids [44]. It is well known that neutral lipids (triglycerides and cholesteryl esters) accumulate in LDs in the diseased heart [45]. In addition, we and others have shown that other lipid intermediates (potentially lipotoxic and/or bioactive lipids) also accumulate in the remodeling heart in response to a pathological insult [46,47,48,49,50]. In response to metabolic reprogramming, the abnormal accumulation of myocardial LDs may impact the redox state of the heart.

1.3. LDs and Plins in the Heart

LDs are conserved intracellular organelles found in almost all cell types [51,52]. This dynamic organelle consists of a core of neutral lipids such as cholesterol esters and triglycerides [53]. LDs store lipids that can be used as metabolic fuel and for membrane components, posttranslational protein modifications, and signaling molecules within the cell [52,54]. They can vary in size from 100 nm up to 100 µm in white adipose tissue (WAT), filling up the entire adipocyte [52,55]. The membrane that surrounds the core consists of phospholipids, cholesterol, and proteins with different functions [51,54,55,56,57].
Proteomic studies have identified more than 200 proteins that are associated with LDs [51,58]. The proteins that coat the LDs can vary between droplets within the cell, between metabolic conditions, and between cell types, and the limited capacity for proteins to bind to LDs further regulates this [52,54,59]. One of the major LD protein families are the Plins [59,60]. To date, five different Plins have been identified in mammals, Plins1–5 [58]. Plins sequester lipids by protecting LDs from lipases. Plin2 and Plin3 are ubiquitously expressed in many tissues and cells, whereas Plin1, 4, and 5 have more specialized tissue expression [54].
Within the heart, LD storage is normally low, with small and few LDs. However, abundant and enlarged LDs are found in the hearts of patients with diabetes, obesity, and metabolic syndrome, as well as with cardiovascular disease [61]. In the heart, four Plins are present (Plin2, Plin3, Plin4, and Plin5). However, Plin2 and Plin5 are by far the most abundant LD proteins in cardiomyocytes [60,62,63].

1.4. LDs and Oxidative Stress

Emerging evidence indicates that LDs, in addition to mere energy metabolism and lipid handling, play important roles in the cellular stress response [64,65]. Numerous reports suggest that an essential purpose of LD biogenesis is to relieve cellular lipotoxic stress, as well as oxidative stress.
Oxidative stress resulting from the overproduction of ROS often correlates with the increased biogenesis of LDs. The causal mechanisms are still not clarified, but potential mechanisms may be the activation of lipogenesis mediated by sterol regulatory element-binding protein (SREBP) as well as altered phospholipid turnover [66,67]. In addition, previous studies have shown that a reduction in a cell’s ability to form LDs may result in severe oxidative stress. Cheng et al. reported that the inhibition of DGAT1 (a major triglycerides-synthesizing enzyme; diacylglycerol-acyltransferase 1) disrupted LD accumulation and led to an increased flux of FAs to mitochondria, resulting in mitochondrial damage, oxidative stress, and apoptosis [68]. Furthermore, Bailey et al. described the antioxidant role of LDs, which limit ROS production from the peroxidation of polyunsaturated FAs in the neuroblasts of drosophila. Their results clearly indicate that LDs comprise a multifunctional organelle that controls energy metabolism, signaling molecules, and intracellular lipotoxicity [66].
The physical association between mitochondria and LDs is a growing research area. Accumulating reports show that this inter-organelle association crucially defines a metabolically distinct subset of mitochondria called peridroplet mitochondria [69]. Beyond the mere bioenergetics, peridroplet mitochondria show a distinct proteome and cristae organization as well as dynamics with reduced fusion and motility [69,70]. Although the mechanisms remain unclear, this interaction may have important implications in the antioxidative properties of LDs. In response to ROS, Tan et al. described the increased incidence of these contact sites in line with increased Plin5 expression through JNK-p38-ATF signaling. In this context, Plin5 could regulate the expression levels of mitochondrial cytochrome c oxidases and alleviate ROS production [17]. In addition, emerging evidence suggests that LDs are required for the autophagic processes [71]. Mitophagy is a selective degradation pathway for defective mitochondria in the lysosomes [72]. Yet incompletely understood, the importance of DGAT1-dependent LD biosynthesis in mitophagy may provide additional evidence on LD-dependent ROS and cell stress management [73,74].
Lastly, LDs have been found to play a key role in driving inflammation by modulating immune cell function. They can provide energy and structural components to produce inflammatory mediators such as prostaglandins, leukotrienes, and cytokines [75]. LDs also interact with the inflammasome, a multiprotein complex that activates the proinflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18) in response to a multitude of pathogen-associated molecular patterns and host-derived signals, including ROS [76]. Among all the described inflammasomes, the nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) inflammasome has been the most investigated due to its implication in a wide array of inflammatory human diseases [77]. Interestingly, NLRP3 affects mitochondrial ROS production by regulating LD formation [78]. NLRP3 activation was also found to be responsible for TREM1-mediated neuroinflammation and ROS production in microglial cells [79]. In this context, TREM1 colocalized with Plin2-positive LDs accumulated through impaired lipophagy. The LDs would thus act as a shield to this pro-oxidant factor. However, the nature of the interaction and how it affects the function of TREM1 remain to be determined. TREM1 plays a key role in oxidative stress, and its inhibition has been suggested as an anti-atherosclerotic therapy [80]. In line with a maladaptive lipid-handling profile, macrophages transitioning to an inflammatory state in human atherosclerosis display high levels of both TREM1 and Plin2 [81]. The activation of the NLRP3 inflammasome is an interesting focus for many cardiovascular injuries [82,83,84] and, in particular, in non-immune cardiac cells (i.e., cardiomyocytes and cardiac fibroblasts) [85,86]. The mechanisms underlying the role of the cardiac inflammasome in relation to lipid accumulation and oxidative stress in metabolic cardiac disease remain unknown. It may provide a better understanding of the key mediators and mechanisms underlying the switch between adaptive and maladaptive LD storage.

2. Plins in the Context of Oxidative Stress and Cardiac Dysfunction

2.1. Plin1

Plin1 is preferentially expressed in adipocytes and steroidogenic cells where it acts as a barrier to prevent LD triglycerides from hydrolysis by lipase [87]. From the current knowledge, the role of Plin1 on cardiac oxidative stress is thus limited to a perturbed metabolic crosstalk between adipose tissue and the myocardium. Indeed, an increased supply of free fatty acids to cardiomyocytes in Plin1-knockout mice results in the accumulation of ROS species and the induction of oxidative stress. This is suggested to be the result of an imbalanced superoxide generation and the reduced ability of antioxidants to detoxify excess ROS [8]. More recently, a dual role of Plin1 has been described in modulating the immune deficiency signaling in the fat body of the fruit fly, Drosophila melanogaster. Plin1 was shown to be downregulated in the early stages of the immune response, leading to the formation of large LDs and thereby taking part in an antioxidative function, efficiently eliminating ROS accumulation after bacterial infection [9]. However, in accordance with a recent report on mice, the prolonged downregulation of Plin1 during persistent immune hyperactivation in Drosophila was critical in promoting large LD-higher rate of Bmm/ATGL-mediated lipolysis leading to excessive lipotoxicity [10].

2.2. Plin2

Plin2 is an LD-associated protein abundantly expressed in nonadipose tissues. It is constitutively associated with intracellular LDs. Plin2 is linked to LD storage in ectopic tissues, and its increased expression is associated with numerous metabolic diseases (insulin resistance, type 2 diabetes, and cardiovascular diseases) in humans as well as in animal models [88]. Global Plin2-knockout mice display reduced liver triglycerides levels and are resistant to diet-induced obesity [89]. Roberts et al. performed an impressive CRISPR-Cas9 screen to identify the regulators of Plin2 expression and stability. They identified canonical genes that control lipid metabolism as well as genes involved in ubiquitination, transcription, and mitochondrial function [90]. In addition, the expression of Plin2 has been shown to be upregulated in the context of oxidative stress. Jin et al. have shown that ROS can induce LD accumulation in hepatocytes by inducing the expression of Plin2 [11]. The endogenous upregulation of Plin2 can alleviate UVA-induced oxidative stress in dermal fibroblasts [12]. Ramosaj et al. elegantly showed that in neural progenitor/stem cells, Plin2-induced LDs generated elevated ROS production but that the higher ROS levels did not result in increased lipid peroxidation [13]. In breast cancer, upregulation of Plin2 and the promotion of lipid storage mediates the adaptation to oxidative stress [14]. An accumulation of LDs, accompanied by an increased expression of Plin2, was also observed in stress-activated microglia [91]. However, in this case, elevated Plin2 levels were supporting oxidative stress in the rostral ventrolateral medulla of stressed rats through phospholipid biosynthesis and metabolism dysregulation. In addition, Plin2 was found to play a crucial role in cerebral ischemia–reperfusion by impacting proinflammatory cytokines and the NLRP3 inflammasome [92].
Plin2 is highly expressed in the heart. In mouse heart, Plin2 is upregulated during fasting-induced steatosis [93]. Plin2-knockout mice had increased myocardial triglyceride levels and an increased myocardial abundance of Plin3 and Plin5 compared with littermate mice. We showed that the increased triglyceride accumulation in Plin2-deficient hearts was caused by reduced lipophagy. Thus, our results suggest that Plin2 is important for the proper hydrolysis of LDs. Western blots showed that the fusion marker mitofusin2 was significantly upregulated in Plin2-deficient hearts, but the fission marker Drp1 was not affected. The expression of mitochondrial proteins OXPHOS proteins complex IV and complex I were reduced in Plin2-deficient hearts. However, basal and maximal mitochondrial respiration was not affected by the lack of Plin2 [63]. Thus, mitochondrial function is intact in Plin2−/− cardiomyocytes, and the increased triglyceride accumulation in Plin2−/− cardiomyocytes is not due to differences in respiration.
Ueno et al. investigated the pathophysiological role of myocardial Plin2 by generating a transgenic mouse model with cardiomyocyte-specific overexpression of Plin2. Plin2-overexpressing hearts displayed massive triglyceride accumulation but preserved myocardial morphology and cardiac function in young mice [94]. In another overexpression model, Sato et al. found that Plin2-induced cardiac steatosis resulted in deteriorated gap junctions in the intercalated discs, impaired conduction propagation, and a higher incidence of atrial fibrillation in aged mice [95].

2.3. Plin3

Plin3 is ubiquitously distributed among tissues [96,97]. Plin3 binds to newly synthesized LDs but is replaced by other Plins, such as Plin2 and Plin5, when the LD matures [98]. Plin3 has the ability to move on and off the LD and is stable in the cytoplasm [96]. The depletion of hepatic Plin3 by antisense oligos suppresses hepatic steatosis and improves glucose homeostasis in mice [99]. In response to oxidative stress-induced apoptosis, Plin3 has been shown to be recruited to the mitochondria where it can protect mitochondrial membrane activity without affecting ROS generation [15].

2.4. Plin4

Plin4 is the least studied member of the Plin family, and knowledge about its regulation and function is still scarce. Plin4 is mainly found in preadipocytes and in membranes of newly synthesized LDs [100]. Pourteymour et al. have also shown that Plin4 is expressed in human skeletal muscle [101]. Indeed, genetic variation in the PLIN4 gene was recently identified in an Italian cohort, resulting in the accumulation of Plin4 within muscle fibers, disrupted fiber organization, and reduced muscle contractility [102,103]. Recently, the increased expression of Plin4 during chemically induced oxidative stress has been reported [16].

2.5. Plin5

Plin5 is highly expressed in oxidative tissue [62] and the most studied member of the Plin family in the context of oxidative stress [104]. Cellular ROS levels have been shown to promote Plin5 expression in hepatic cells [17]. Furthermore, a recent report highlighted Plin5 as being the only Plin with a reduced expression on LDs from LPS-treated livers, suggesting that the ROS burden could also affect its location/function [105]. However, even if reports accumulate about the role of Plin5 in reducing oxidative damage, the knowledge about its regulation in cells facing an oxidative burden is still scarce. Zhu et al. showed that Plin5 regulates protection against oxidative damage by mediating the Nrf-antioxidant response element signaling pathway in pancreatic β-cells [18].
Plin5 is essential for maintaining LDs in oxidative tissues by antagonizing lipases [60,106,107]. Plin5-knockout mice lack detectable LDs in cardiomyocytes and have markedly reduced triglycerides accumulation in the heart [19,48]. Plin5-deficient cardiomyocytes undergo a metabolic shift by decreasing the fatty acid uptake and instead increasing the glucose uptake and are thereby able to maintain their energy metabolism. Plin5-knockout mice maintain a close-to-normal heart function under baseline conditions. However, during stress or myocardial ischemia, Plin5 deficiency results in myocardial reduced substrate availability, severely impaired cardiac function, and increased mortality [48]. Kuramoto et al. further showed that the production of ROS was increased in the Plin5−/− mouse hearts, leading to a reduced heart function with age. This was, however, reduced by the administration of N-acetylcysteine, a precursor of an antioxidant, glutathione. In addition, Plin5-deficient mice displayed aggravated cardiac hypertrophy and elevated myocardial oxidative stress following transaortic constriction [108]. Thus, LDs prevent excess ROS production by sequestering fatty acids from oxidation and hence suppress oxidative burden to the heart [19]. Moreover, Zheng et al. have shown that Plin5 reduces oxidative stress following myocardial ischemia–reperfusion injury, through the inhibition of the lipolysis of LDs [20]. The authors showed that Plin5 deficiency increased the myocardial infarct area and reduced the heart function. Furthermore, Plin5-deficient myocardium displayed damaged mitochondria, increased ROS and malondialdehyde levels, and reduced SOD activity [20].
Plin5 provides a physical linkage to mitochondria by anchoring the mitochondria to the LD by the C-terminal region of Plin5 [109]. A deficiency in Plin5 in cardiomyocytes has also been shown to result in reduced mitochondrial function [110]. In mitochondria isolated from Plin5-deficient hearts, the oxidative capacity was reduced. However, there was no effect on the mitochondrial oxidative stress or the generation of ROS [110]. Miner et al. recently showed that FATP4 is a mitochondrial interactor of Plin5, enabling fatty acid channeling from LDs to mitochondria [111]. Kien et al. further investigated the impact of LD–mitochondria coupling by comparing a truncated form of Plin5 (that lacked the ability to couple to mitochondria) with wildtype Plin5. They found that efficient coupling between Plin5 and mitochondria had no effect on fatty acid oxidation but significantly improved the respiratory capacity of mitochondria [112]. Higher mitochondrial respiration may involve the increased production of mitochondrial ROS without deleterious effects. In this context, Plin5 overexpression may thus enhance ROS detoxification and/or improve their usage towards beneficial pathways. Future studies of the mitochondrial interactors of Plin5 will improve the understanding of how Plin5 manages mitochondrial ROS production in pathophysiological conditions.
Additional studies have suggested a role for Plin5 in atherosclerosis and oxidative stress. Plin5 deficiency leads to accelerated atherosclerosis progression and oxidative stress in ApoE−/− mice [113]. The inactivation of Plin5 in macrophages resulted in elevated inflammation and oxidative stress [113]. Moreover, Plin5 also regulates vascular smooth muscle cell proliferation by modulating ROS generation [21].
In humans, genetic variation in PLIN5 is associated with impaired cardiac function after myocardial ischemia. Patients carrying the allele rs884164 are at higher risk of cardiovascular morbidity and mortality after myocardial ischemia [48].

2.6. Transcriptional and Posttranslational Regulation of Plins in Response to ROS

While it is well recognized that oxidative stress leads to the increased expression of Plins in various tissues, the mechanisms of the transcriptional and posttranslational regulation of Plins by oxidative stress need further investigation. The PPAR family of transcription factors is key for the regulation of most of mammalian Plins (i.e., Plin1–2 and 4–5) [114]. In combination with specific transcriptional cofactors, such as PGC1 family members, the three PPAR isoforms (α, β/δ, and γ) regulate the differential cell-specific activations of Plin1, 2, 4, and 5 in different tissues. ROS levels can regulate transcriptional activity, including PPARγ function [115,116]. With transcriptional regulation, the stability of Plins is the determining factor for their tissue-specific expression levels and function. One of the most important targets of redox-based modifications is the redox-sensitive thiols of cysteines [117]. The C-terminus of Plin1 protein is a relatively conserved hydrophobic domain with five cysteine residues that have been shown to be sulfhydrated, thereby stabilizing the protein [118]. The recent development of a broad and quantitative analysis of the cysteine proteome in living tissues [119] represents an exciting approach to better understand how persistent ROS exposure could affect Plin stability and regulate their expression during oxidative stress. In addition, several reports show that posttranslational modifications of Plins are crucial for their location and function [59,120,121]. Unbiased screening approaches could facilitate more thorough investigations of the dynamic of oxidative stress-linked posttranslational modifications of Plins. Studies of human single-nucleotide polymorphisms in combination with genetically engineered mice would therefore help the understanding of the specific role of each Plin in the management of cardiac oxidative stress.

3. Concluding Remarks and Perspectives

One of the limitations in studying the isoform-specific functions of Plins in ROS management is the tight complexity in the regulation of Plins. It has been established that most mouse-engineered models for one specific Plin result in consequent regulation and compensation by other members of the family. For example, Plin3 and Plin5 are increased in Plin2-knockout mice, suggesting that they may be able to compensate for some Plin2 functions that are lost [63,122]. Above all, all Plins regulate LD turnover and access to lipases [59], resulting in the dysregulation of metabolic by-products of Plin-regulated lipolysis in genetically engineered models. Consequently, the feedback regulation of non-targeted Plins may be promoted, complicating the interpretation of their isoform-specific role.
Even though extensive experimental studies suggest that ROS are suitable targets for the intervention of cardiovascular disease, clinical trials have unfortunately shown negative results [123]. Contributing factors to the lack of clinical effects may be the different primary pathophysiologic mechanisms depending on the etiology of the disease and the large number of exogenous and endogenous antioxidant players, which implies a high interpatient variability [124]. ROS generation and interactions with other signaling molecules have been shown to occur in a compartmentalized manner, and thus there are still opportunities for novel approaches, especially targeting the endogenous antioxidant capacity. Preclinical models have provided strong evidence that LDs can function as ROS scavengers; however, knowledge about the specific role of LD proteins such as Plins in this context needs to be further investigated.
Specific modulation of Plin levels is challenging and limits the therapeutic options for ROS mitigation. However, there are increasing reports showing that increased levels of Plin2 or Plin5 reduce cellular oxidative damage. Therefore, testing the endogenous antioxidant capacity of Plins to alleviate oxidative stress in heart failure appears to be an exciting opportunity in ROS reduction during cardiovascular disease. The difficulties in specifically modulating Plin levels represent the major limitation to success in this unmet scientific challenge. However, a recent report showed that LDs can be released in the extracellular space and exchanged between cells [125]. This raises the question of the biological role of this mechanism and the potential role of LD proteins in extracellular communication. This has huge potential for the use of LDs as extracellular vesicles as novel drug delivery vehicles but most importantly as a therapy itself. In this context, promising work from Zhao et al. validated that Plin-coated artificial LDs could be taken up by cells, significantly reducing hydrogen peroxide-induced ROS and alleviating cellular lipotoxicity caused by excess fatty acids [126].
Recently, we have shown that cardiac-specific Plin5 overexpression promotes physiological-like hypertrophy with preserved/improved cardiac function [127]. This study emphasized the therapeutic potential of the Plins to maintain cardiac physiology in challenging settings. Contrary to maladaptive remodeling, physiological cardiac hypertrophy is considered harmless, completely reversible, and mostly occurs in response to increased workload such as exercise. Mounting evidence supports the notion that ROS as well as Plin levels are tightly regulated during exercise-induced adaptations [128,129,130,131,132]. Further investigations are needed to elucidate the interplay between Plins and ROS in the context of exercise and physiological hypertrophy.

Author Contributions

Conceptualization, M.C. and M.C.L.; writing—original draft preparation, M.C. and M.C.L.; writing—review and editing, M.C., L.A., A.M., M.L., O.K., J.B. and M.C.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Swedish Society for Medical Research (Svenska Sällskapet för Medicinsk Forskning, SSMF), the Swedish Heart and Lung Foundation, and the Swedish Research Council.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No data were used for the research described in this article.

Conflicts of Interest

The authors declare that they have no conflict of interest.

References

  1. Christiansen, M.N.; Kober, L.; Weeke, P.; Vasan, R.S.; Jeppesen, J.L.; Smith, J.G.; Gislason, G.H.; Torp-Pedersen, C.; Andersson, C. Age-Specific Trends in Incidence, Mortality, and Comorbidities of Heart Failure in Denmark, 1995 to 2012. Circulation 2017, 135, 1214–1223. [Google Scholar] [CrossRef] [PubMed]
  2. Glynn, P.; Lloyd-Jones, D.M.; Feinstein, M.J.; Carnethon, M.; Khan, S.S. Disparities in Cardiovascular Mortality Related to Heart Failure in the United States. J. Am. Coll. Cardiol. 2019, 73, 2354–2355. [Google Scholar] [CrossRef] [PubMed]
  3. Rosengren, A.; Edqvist, J.; Rawshani, A.; Sattar, N.; Franzen, S.; Adiels, M.; Svensson, A.M.; Lind, M.; Gudbjornsdottir, S. Excess risk of hospitalisation for heart failure among people with type 2 diabetes. Diabetologia 2018, 61, 2300–2309. [Google Scholar] [CrossRef] [PubMed]
  4. Hori, M.; Nishida, K. Oxidative stress and left ventricular remodelling after myocardial infarction. Cardiovasc. Res. 2009, 81, 457–464. [Google Scholar] [CrossRef]
  5. Karimi Galougahi, K.; Antoniades, C.; Nicholls, S.J.; Channon, K.M.; Figtree, G.A. Redox biomarkers in cardiovascular medicine. Eur. Heart J. 2015, 36, 1576–1582. [Google Scholar] [CrossRef]
  6. Tsutsui, H.; Kinugawa, S.; Matsushima, S. Oxidative stress and heart failure. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H2181–H2190. [Google Scholar] [CrossRef]
  7. Burgoyne, J.R.; Mongue-Din, H.; Eaton, P.; Shah, A.M. Redox signaling in cardiac physiology and pathology. Circ. Res. 2012, 111, 1091–1106. [Google Scholar] [CrossRef] [PubMed]
  8. Liu, S.; Geng, B.; Zou, L.; Wei, S.; Wang, W.; Deng, J.; Xu, C.; Zhao, X.; Lyu, Y.; Su, X.; et al. Development of hypertrophic cardiomyopathy in perilipin-1 null mice with adipose tissue dysfunction. Cardiovasc. Res. 2015, 105, 20–30. [Google Scholar] [CrossRef]
  9. Wang, L.; Lin, J.; Yu, J.; Yang, K.; Sun, L.; Tang, H.; Pan, L. Downregulation of Perilipin1 by the Immune Deficiency Pathway Leads to Lipid Droplet Reconfiguration and Adaptation to Bacterial Infection in Drosophila. J. Immunol. 2021, 207, 2347–2358. [Google Scholar] [CrossRef]
  10. Wang, L.; Lin, J.; Yang, K.; Wang, W.; Lv, Y.; Zeng, X.; Zhao, Y.; Yu, J.; Pan, L. Perilipin1 deficiency prompts lipolysis in lipid droplets and aggravates the pathogenesis of persistent immune activation in Drosophila. J. Innate Immun. 2023, 15, 697–708. [Google Scholar] [CrossRef]
  11. Jin, Y.; Tan, Y.; Chen, L.; Liu, Y.; Ren, Z. Reactive Oxygen Species Induces Lipid Droplet Accumulation in HepG2 Cells by Increasing Perilipin 2 Expression. Int. J. Mol. Sci. 2018, 19, 3445. [Google Scholar] [CrossRef]
  12. Lu, Y.S.; Jiang, Y.; Yuan, J.P.; Jiang, S.B.; Yang, Y.; Zhu, P.Y.; Sun, Y.Z.; Qi, R.Q.; Liu, T.; Wang, H.X.; et al. UVA Induced Oxidative Stress Was Inhibited by Paeoniflorin/Nrf2 Signaling or PLIN2. Front. Pharmacol. 2020, 11, 736. [Google Scholar] [CrossRef]
  13. Ramosaj, M.; Madsen, S.; Maillard, V.; Scandella, V.; Sudria-Lopez, D.; Yuizumi, N.; Telley, L.; Knobloch, M. Lipid droplet availability affects neural stem/progenitor cell metabolism and proliferation. Nat. Commun. 2021, 12, 7362. [Google Scholar] [CrossRef]
  14. Cadenas, C.; Vosbeck, S.; Edlund, K.; Grgas, K.; Madjar, K.; Hellwig, B.; Adawy, A.; Glotzbach, A.; Stewart, J.D.; Lesjak, M.S.; et al. LIPG-promoted lipid storage mediates adaptation to oxidative stress in breast cancer. Int. J. Cancer 2019, 145, 901–915. [Google Scholar] [CrossRef]
  15. Hocsak, E.; Racz, B.; Szabo, A.; Mester, L.; Rapolti, E.; Pozsgai, E.; Javor, S.; Bellyei, S.; Gallyas, F., Jr.; Sumegi, B.; et al. TIP47 protects mitochondrial membrane integrity and inhibits oxidative-stress-induced cell death. FEBS Lett. 2010, 584, 2953–2960. [Google Scholar] [CrossRef]
  16. Bernier, F.; Kuhara, T.; Xiao, J. Probiotic Bifidobacterium breve MCC1274 Protects against Oxidative Stress and Neuronal Lipid Droplet Formation via PLIN4 Gene Regulation. Microorganisms 2023, 11, 791. [Google Scholar] [CrossRef]
  17. Tan, Y.; Jin, Y.; Wang, Q.; Huang, J.; Wu, X.; Ren, Z. Perilipin 5 Protects against Cellular Oxidative Stress by Enhancing Mitochondrial Function in HepG2 Cells. Cells 2019, 8, 1241. [Google Scholar] [CrossRef]
  18. Zhu, Y.; Ren, C.; Zhang, M.; Zhong, Y. Perilipin 5 Reduces Oxidative Damage Associated with Lipotoxicity by Activating the PI3K/ERK-Mediated Nrf2-ARE Signaling Pathway in INS-1 Pancreatic beta-Cells. Front. Endocrinol. 2020, 11, 166. [Google Scholar] [CrossRef] [PubMed]
  19. Kuramoto, K.; Okamura, T.; Yamaguchi, T.; Nakamura, T.Y.; Wakabayashi, S.; Morinaga, H.; Nomura, M.; Yanase, T.; Otsu, K.; Usuda, N.; et al. Perilipin 5, a lipid droplet-binding protein, protects heart from oxidative burden by sequestering fatty acid from excessive oxidation. J. Biol. Chem. 2012, 287, 23852–23863. [Google Scholar] [CrossRef] [PubMed]
  20. 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] [PubMed]
  21. Gan, X.; Zhao, J.; Chen, Y.; Li, Y.; Xuan, B.; Gu, M.; Feng, F.; Yang, Y.; Yang, D.; Sun, X. Plin5 inhibits proliferation and migration of vascular smooth muscle cell through interacting with PGC-1alpha following vascular injury. Bioengineered 2022, 13, 10665–10678. [Google Scholar] [CrossRef]
  22. Sies, H.; Belousov, V.V.; Chandel, N.S.; Davies, M.J.; Jones, D.P.; Mann, G.E.; Murphy, M.P.; Yamamoto, M.; Winterbourn, C. Defining roles of specific reactive oxygen species (ROS) in cell biology and physiology. Nat. Rev. Mol. Cell Biol. 2022, 23, 499–515. [Google Scholar] [CrossRef]
  23. Brand, M.D. Mitochondrial generation of superoxide and hydrogen peroxide as the source of mitochondrial redox signaling. Free Radic. Biol. Med. 2016, 100, 14–31. [Google Scholar] [CrossRef]
  24. Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef] [PubMed]
  25. Cave, A.C.; Brewer, A.C.; Narayanapanicker, A.; Ray, R.; Grieve, D.J.; Walker, S.; Shah, A.M. NADPH oxidases in cardiovascular health and disease. Antioxid. Redox Signal 2006, 8, 691–728. [Google Scholar] [CrossRef]
  26. D’Oria, R.; Schipani, R.; Leonardini, A.; Natalicchio, A.; Perrini, S.; Cignarelli, A.; Laviola, L.; Giorgino, F. The Role of Oxidative Stress in Cardiac Disease: From Physiological Response to Injury Factor. Oxidative Med. Cell. Longev. 2020, 2020, 5732956. [Google Scholar] [CrossRef]
  27. He, L.; He, T.; Farrar, S.; Ji, L.; Liu, T.; Ma, X. Antioxidants Maintain Cellular Redox Homeostasis by Elimination of Reactive Oxygen Species. Cell. Physiol. Biochem. 2017, 44, 532–553. [Google Scholar] [CrossRef] [PubMed]
  28. Rushmore, T.H.; Morton, M.R.; Pickett, C.B. The antioxidant responsive element. Activation by oxidative stress and identification of the DNA consensus sequence required for functional activity. J. Biol. Chem. 1991, 266, 11632–11639. [Google Scholar] [CrossRef]
  29. Ngo, V.; Duennwald, M.L. Nrf2 and Oxidative Stress: A General Overview of Mechanisms and Implications in Human Disease. Antioxidants 2022, 11, 2345. [Google Scholar] [CrossRef] [PubMed]
  30. Halliwell, B.; Gutteridge, J.M. Free Radicals in Biology and Medicine, 5th ed.; Oxford Academic Press: Oxford, UK, 2015. [Google Scholar]
  31. Ong, S.B.; Samangouei, P.; Kalkhoran, S.B.; Hausenloy, D.J. The mitochondrial permeability transition pore and its role in myocardial ischemia reperfusion injury. J. Mol. Cell. Cardiol. 2015, 78, 23–34. [Google Scholar] [CrossRef] [PubMed]
  32. Quijano, C.; Trujillo, M.; Castro, L.; Trostchansky, A. Interplay between oxidant species and energy metabolism. Redox Biol. 2016, 8, 28–42. [Google Scholar] [CrossRef]
  33. Lennicke, C.; Cocheme, H.M. Redox metabolism: ROS as specific molecular regulators of cell signaling and function. Mol. Cell 2021, 81, 3691–3707. [Google Scholar] [CrossRef]
  34. Humeres, C.; Frangogiannis, N.G. Fibroblasts in the Infarcted, Remodeling, and Failing Heart. JACC Basic. Transl. Sci. 2019, 4, 449–467. [Google Scholar] [CrossRef]
  35. Gorlach, A.; Bertram, K.; Hudecova, S.; Krizanova, O. Calcium and ROS: A mutual interplay. Redox Biol. 2015, 6, 260–271. [Google Scholar] [CrossRef] [PubMed]
  36. Xu, T.; Ding, W.; Ji, X.; Ao, X.; Liu, Y.; Yu, W.; Wang, J. Oxidative Stress in Cell Death and Cardiovascular Diseases. Oxidative Med. Cell. Longev. 2019, 2019, 9030563. [Google Scholar] [CrossRef] [PubMed]
  37. Takimoto, E.; Kass, D.A. Role of oxidative stress in cardiac hypertrophy and remodeling. Hypertension 2007, 49, 241–248. [Google Scholar] [CrossRef]
  38. De Geest, B.; Mishra, M. Role of Oxidative Stress in Diabetic Cardiomyopathy. Antioxidants 2022, 11, 784. [Google Scholar] [CrossRef] [PubMed]
  39. Wende, A.R.; Brahma, M.K.; McGinnis, G.R.; Young, M.E. Metabolic Origins of Heart Failure. JACC Basic. Transl. Sci. 2017, 2, 297–310. [Google Scholar] [CrossRef] [PubMed]
  40. Hochachka, P.W.; Buck, L.T.; Doll, C.J.; Land, S.C. Unifying theory of hypoxia tolerance: Molecular/metabolic defense and rescue mechanisms for surviving oxygen lack. Proc. Natl. Acad. Sci. USA 1996, 93, 9493–9498. [Google Scholar] [CrossRef]
  41. Lopaschuk, G.D.; Ussher, J.R.; Folmes, C.D.; Jaswal, J.S.; Stanley, W.C. Myocardial fatty acid metabolism in health and disease. Physiol. Rev. 2010, 90, 207–258. [Google Scholar] [CrossRef] [PubMed]
  42. Ritterhoff, J.; Tian, R. Metabolism in cardiomyopathy: Every substrate matters. Cardiovasc. Res. 2017, 113, 411–421. [Google Scholar] [CrossRef]
  43. Ingwall, J.S. Energy metabolism in heart failure and remodelling. Cardiovasc. Res. 2009, 81, 412–419. [Google Scholar] [CrossRef]
  44. Schulze, P.C.; Drosatos, K.; Goldberg, I.J. Lipid Use and Misuse by the Heart. Circ. Res. 2016, 118, 1736–1751. [Google Scholar] [CrossRef]
  45. Sharma, S.; Adrogue, J.V.; Golfman, L.; Uray, I.; Lemm, J.; Youker, K.; Noon, G.P.; Frazier, O.H.; Taegtmeyer, H. Intramyocardial lipid accumulation in the failing human heart resembles the lipotoxic rat heart. FASEB J. 2004, 18, 1692–1700. [Google Scholar] [CrossRef]
  46. Andersson, L.; Cinato, M.; Mardani, I.; Miljanovic, A.; Arif, M.; Koh, A.; Lindbom, M.; Laudette, M.; Bollano, E.; Omerovic, E.; et al. Glucosylceramide Synthase Deficiency in the Heart Compromises β1-Adrenergic Receptor Trafficking. Eur. Heart J. 2021, 42, 4481–4492. [Google Scholar] [CrossRef]
  47. Chokshi, A.; Drosatos, K.; Cheema, F.H.; Ji, R.; Khawaja, T.; Yu, S.; Kato, T.; Khan, R.; Takayama, H.; Knoll, R.; et al. Ventricular assist device implantation corrects myocardial lipotoxicity, reverses insulin resistance, and normalizes cardiac metabolism in patients with advanced heart failure. Circulation 2012, 125, 2844–2853. [Google Scholar] [CrossRef]
  48. Drevinge, C.; Dalen, K.T.; Mannila, M.N.; Tang, M.S.; Stahlman, M.; Klevstig, M.; Lundqvist, A.; Mardani, I.; Haugen, F.; Fogelstrand, P.; et al. Perilipin 5 is protective in the ischemic heart. Int. J. Cardiol. 2016, 219, 446–454. [Google Scholar] [CrossRef]
  49. Drevinge, C.; Karlsson, L.O.; Stahlman, M.; Larsson, T.; Perman Sundelin, J.; Grip, L.; Andersson, L.; Boren, J.; Levin, M.C. Cholesteryl esters accumulate in the heart in a porcine model of ischemia and reperfusion. PLoS ONE 2013, 8, e61942. [Google Scholar] [CrossRef]
  50. Klevstig, M.; Stahlman, M.; Lundqvist, A.; Scharin Tang, M.; Fogelstrand, P.; Adiels, M.; Andersson, L.; Kolesnick, R.; Jeppsson, A.; Boren, J.; et al. Targeting acid sphingomyelinase reduces cardiac ceramide accumulation in the post-ischemic heart. J. Mol. Cell. Cardiol. 2016, 93, 69–72. [Google Scholar] [CrossRef]
  51. Onal, G.; Kutlu, O.; Gozuacik, D.; Dokmeci Emre, S. Lipid Droplets in Health and Disease. Lipids Health Dis. 2017, 16, 128. [Google Scholar] [CrossRef]
  52. Walther, T.C.; Farese, R.V., Jr. Lipid droplets and cellular lipid metabolism. Annu. Rev. Biochem. 2012, 81, 687–714. [Google Scholar] [CrossRef]
  53. 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]
  54. 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]
  55. Guo, Y.; Cordes, K.R.; Farese, R.V., Jr.; Walther, T.C. Lipid droplets at a glance. J. Cell Sci. 2009, 122, 749–752. [Google Scholar] [CrossRef]
  56. Meyers, A.; Weiskittel, T.M.; Dalhaimer, P. Lipid Droplets: Formation to Breakdown. Lipids 2017, 52, 465–475. [Google Scholar] [CrossRef]
  57. Walther, T.C.; Chung, J.; Farese, R.V., Jr. Lipid Droplet Biogenesis. Annu. Rev. Cell Dev. Biol. 2017, 33, 491–510. [Google Scholar] [CrossRef]
  58. Sztalryd, C.; Brasaemle, D.L. The perilipin family of lipid droplet proteins: Gatekeepers of intracellular lipolysis. Biochim. Biophys. Acta 2017, 1862, 1221–1232. [Google Scholar] [CrossRef]
  59. 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]
  60. 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]
  61. Goldberg, I.J.; Reue, K.; Abumrad, N.A.; Bickel, P.E.; Cohen, S.; Fisher, E.A.; Galis, Z.S.; Granneman, J.G.; Lewandowski, E.D.; Murphy, R.; et al. Deciphering the Role of Lipid Droplets in Cardiovascular Disease: A Report From the 2017 National Heart, Lung, and Blood Institute Workshop. Circulation 2018, 138, 305–315. [Google Scholar] [CrossRef]
  62. Dalen, K.T.; Dahl, T.; Holter, E.; Arntsen, B.; Londos, C.; Sztalryd, C.; Nebb, H.I. LSDP5 is a PAT protein specifically expressed in fatty acid oxidizing tissues. Biochim. Biophys. Acta 2007, 1771, 210–227. [Google Scholar] [CrossRef]
  63. Mardani, I.; Tomas Dalen, K.; Drevinge, C.; Miljanovic, A.; Stahlman, M.; Klevstig, M.; Scharin Tang, M.; Fogelstrand, P.; Levin, M.; Ekstrand, M.; et al. Plin2-deficiency reduces lipophagy and results in increased lipid accumulation in the heart. Sci. Rep. 2019, 9, 6909. [Google Scholar] [CrossRef]
  64. Jarc, E.; Petan, T. Lipid Droplets and the Management of Cellular Stress. Yale J. Biol. Med. 2019, 92, 435–452. [Google Scholar]
  65. 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]
  66. Bailey, A.P.; Koster, G.; Guillermier, C.; Hirst, E.M.; MacRae, J.I.; Lechene, C.P.; Postle, A.D.; Gould, A.P. Antioxidant Role for Lipid Droplets in a Stem Cell Niche of Drosophila. Cell 2015, 163, 340–353. [Google Scholar] [CrossRef]
  67. Liu, L.; Zhang, K.; Sandoval, H.; Yamamoto, S.; Jaiswal, M.; Sanz, E.; Li, Z.; Hui, J.; Graham, B.H.; Quintana, A.; et al. Glial lipid droplets and ROS induced by mitochondrial defects promote neurodegeneration. Cell 2015, 160, 177–190. [Google Scholar] [CrossRef]
  68. Cheng, X.; Geng, F.; Pan, M.; Wu, X.; Zhong, Y.; Wang, C.; Tian, Z.; Cheng, C.; Zhang, R.; Puduvalli, V.; et al. Targeting DGAT1 Ameliorates Glioblastoma by Increasing Fat Catabolism and Oxidative Stress. Cell Metab. 2020, 32, 229–242.e8. [Google Scholar] [CrossRef]
  69. Benador, I.Y.; Veliova, M.; Liesa, M.; Shirihai, O.S. Mitochondria Bound to Lipid Droplets: Where Mitochondrial Dynamics Regulate Lipid Storage and Utilization. Cell Metab. 2019, 29, 827–835. [Google Scholar] [CrossRef]
  70. Veliova, M.; Petcherski, A.; Liesa, M.; Shirihai, O.S. The biology of lipid droplet-bound mitochondria. Semin. Cell Dev. Biol. 2020, 108, 55–64. [Google Scholar] [CrossRef]
  71. Xu, C.; Fan, J. Links between autophagy and lipid droplet dynamics. J. Exp. Bot. 2022, 73, 2848–2858. [Google Scholar] [CrossRef]
  72. Schofield, J.H.; Schafer, Z.T. Mitochondrial Reactive Oxygen Species and Mitophagy: A Complex and Nuanced Relationship. Antioxid. Redox Signal 2021, 34, 517–530. [Google Scholar] [CrossRef]
  73. Long, M.; McWilliams, T.G. Lipid droplets promote efficient mitophagy. Autophagy 2023, 19, 724–725. [Google Scholar] [CrossRef]
  74. Long, M.; Sanchez-Martinez, A.; Longo, M.; Suomi, F.; Stenlund, H.; Johansson, A.I.; Ehsan, H.; Salo, V.T.; Montava-Garriga, L.; Naddafi, S.; et al. DGAT1 activity synchronises with mitophagy to protect cells from metabolic rewiring by iron depletion. EMBO J. 2022, 41, e109390. [Google Scholar] [CrossRef]
  75. Monson, E.A.; Trenerry, A.M.; Laws, J.L.; Mackenzie, J.M.; Helbig, K.J. Lipid droplets and lipid mediators in viral infection and immunity. FEMS Microbiol. Rev. 2021, 45, fuaa066. [Google Scholar] [CrossRef]
  76. Zheng, D.; Liwinski, T.; Elinav, E. Inflammasome activation and regulation: Toward a better understanding of complex mechanisms. Cell Discov. 2020, 6, 36. [Google Scholar] [CrossRef]
  77. Blevins, H.M.; Xu, Y.; Biby, S.; Zhang, S. The NLRP3 Inflammasome Pathway: A Review of Mechanisms and Inhibitors for the Treatment of Inflammatory Diseases. Front. Aging Neurosci. 2022, 14, 879021. [Google Scholar] [CrossRef]
  78. Zhang, Z.; Guo, P.; Liang, L.; Jila, S.; Ru, X.; Zhang, Q.; Chen, J.; Chen, Z.; Feng, H.; Chen, Y. NLRP3-dependent lipid droplet formation contributes to posthemorrhagic hydrocephalus by increasing the permeability of the blood-cerebrospinal fluid barrier in the choroid plexus. Exp. Mol. Med. 2023, 55, 574–586. [Google Scholar] [CrossRef]
  79. 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, 19, 2639–2656. [Google Scholar] [CrossRef]
  80. Panagopoulos, A.; Samant, S.; Bakhos, J.J.; Liu, M.; Khan, B.; Makadia, J.; Muhammad, F.; Kievit, F.M.; Agrawal, D.K.; Chatzizisis, Y.S. Triggering receptor expressed on myeloid cells-1 (TREM-1) inhibition in atherosclerosis. Pharmacol. Ther. 2022, 238, 108182. [Google Scholar] [CrossRef]
  81. Dib, L.; Koneva, L.A.; Edsfeldt, A.; Zurke, Y.X.; Sun, J.; Nitulescu, M.; Attar, M.; Lutgens, E.; Schmidt, S.; Lindholm, M.W.; et al. Lipid-associated macrophages transition to an inflammatory state in human atherosclerosis, increasing the risk of cerebrovascular complications. Nat. Cardiovasc. Res. 2023, 2, 656–672. [Google Scholar] [CrossRef]
  82. Toldo, S.; Abbate, A. The NLRP3 inflammasome in acute myocardial infarction. Nat. Rev. Cardiol. 2018, 15, 203–214. [Google Scholar] [CrossRef]
  83. Zhou, W.; Chen, C.; Chen, Z.; Liu, L.; Jiang, J.; Wu, Z.; Zhao, M.; Chen, Y. NLRP3: A Novel Mediator in Cardiovascular Disease. J. Immunol. Res. 2018, 2018, 5702103. [Google Scholar] [CrossRef]
  84. Abbate, A.; Toldo, S.; Marchetti, C.; Kron, J.; Van Tassell, B.W.; Dinarello, C.A. Interleukin-1 and the Inflammasome as Therapeutic Targets in Cardiovascular Disease. Circ. Res. 2020, 126, 1260–1280. [Google Scholar] [CrossRef]
  85. Wu, J.; Dong, E.; Zhang, Y.; Xiao, H. The Role of the Inflammasome in Heart Failure. Front. Physiol. 2021, 12, 709703. [Google Scholar] [CrossRef]
  86. Zheng, Y.; Xu, L.; Dong, N.; Li, F. NLRP3 inflammasome: The rising star in cardiovascular diseases. Front. Cardiovasc. Med. 2022, 9, 927061. [Google Scholar] [CrossRef]
  87. Brasaemle, D.L.; Subramanian, V.; Garcia, A.; Marcinkiewicz, A.; Rothenberg, A. Perilipin A and the control of triacylglycerol metabolism. Mol. Cell. Biochem. 2009, 326, 15–21. [Google Scholar] [CrossRef]
  88. 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]
  89. McManaman, J.L.; Bales, E.S.; Orlicky, D.J.; Jackman, M.; MacLean, P.S.; Cain, S.; Crunk, A.E.; Mansur, A.; Graham, C.E.; Bowman, T.A.; et al. Perilipin-2-null mice are protected against diet-induced obesity, adipose inflammation, and fatty liver disease. J. Lipid Res. 2013, 54, 1346–1359. [Google Scholar] [CrossRef]
  90. Roberts, M.A.; Deol, K.K.; Mathiowetz, A.J.; Lange, M.; Leto, D.E.; Stevenson, J.; Hashemi, S.H.; Morgens, D.W.; Easter, E.; Heydari, K.; et al. Parallel CRISPR-Cas9 screens identify mechanisms of PLIN2 and lipid droplet regulation. Dev. Cell 2023, 58, 1782–1800.E10. [Google Scholar] [CrossRef]
  91. 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]
  92. Liu, X.Y.; Li, Q.S.; Yang, W.H.; Qiu, Y.; Zhang, F.F.; Mei, X.H.; Yuan, Q.W.; Sui, R.B. Inhibition of perilipin 2 attenuates cerebral ischemia/reperfusion injury by blocking NLRP3 inflammasome activation both in vivo and in vitro. Cell. Dev. Biol. Anim. 2023, 59, 204–213. [Google Scholar] [CrossRef]
  93. Suzuki, K.; Takahashi, K.; Nishimaki-Mogami, T.; Kagechika, H.; Yamamoto, M.; Itabe, H. Docosahexaenoic acid induces adipose differentiation-related protein through activation of retinoid x receptor in human choriocarcinoma BeWo cells. Biol. Pharm. Bull. 2009, 32, 1177–1182. [Google Scholar] [CrossRef]
  94. Ueno, M.; Suzuki, J.; Hirose, M.; Sato, S.; Imagawa, M.; Zenimaru, Y.; Takahashi, S.; Ikuyama, S.; Koizumi, T.; Konoshita, T.; et al. Cardiac overexpression of perilipin 2 induces dynamic steatosis: Prevention by hormone-sensitive lipase. Am. J. Physiol. Endocrinol. Metab. 2017, 313, E699–E709. [Google Scholar] [CrossRef]
  95. Sato, S.; Suzuki, J.; Hirose, M.; Yamada, M.; Zenimaru, Y.; Nakaya, T.; Ichikawa, M.; Imagawa, M.; Takahashi, S.; Ikuyama, S.; et al. Cardiac overexpression of perilipin 2 induces atrial steatosis, connexin 43 remodeling, and atrial fibrillation in aged mice. Am. J. Physiol. Endocrinol. Metab. 2019, 317, E1193–E1204. [Google Scholar] [CrossRef]
  96. Bulankina, A.V.; Deggerich, A.; Wenzel, D.; Mutenda, K.; Wittmann, J.G.; Rudolph, M.G.; Burger, K.N.; Honing, S. TIP47 functions in the biogenesis of lipid droplets. J. Cell Biol. 2009, 185, 641–655. [Google Scholar] [CrossRef]
  97. Wolins, N.E.; Rubin, B.; Brasaemle, D.L. TIP47 associates with lipid droplets. J. Biol. Chem. 2001, 276, 5101–5108. [Google Scholar] [CrossRef]
  98. Wilson, M.H.; Ekker, S.C.; Farber, S.A. Imaging cytoplasmic lipid droplets in vivo with fluorescent perilipin 2 and perilipin 3 knock-in zebrafish. eLife 2021, 10, e66393. [Google Scholar] [CrossRef]
  99. Carr, R.M.; Patel, R.T.; Rao, V.; Dhir, R.; Graham, M.J.; Crooke, R.M.; Ahima, R.S. Reduction of TIP47 improves hepatic steatosis and glucose homeostasis in mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2012, 302, R996–R1003. [Google Scholar] [CrossRef]
  100. 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]
  101. Pourteymour, S.; Lee, S.; Langleite, T.M.; Eckardt, K.; Hjorth, M.; Bindesboll, C.; Dalen, K.T.; Birkeland, K.I.; Drevon, C.A.; Holen, T.; et al. Perilipin 4 in human skeletal muscle: Localization and effect of physical activity. Physiol. Rep. 2015, 3, e12481. [Google Scholar] [CrossRef]
  102. Di Blasi, C.; Moghadaszadeh, B.; Ciano, C.; Negri, T.; Giavazzi, A.; Cornelio, F.; Morandi, L.; Mora, M. Abnormal lysosomal and ubiquitin-proteasome pathways in 19p13.3 distal myopathy. Ann. Neurol. 2004, 56, 133–138. [Google Scholar] [CrossRef]
  103. Maggi, L.; Gibertini, S.; Iannibelli, E.; Gallone, A.; Bonanno, S.; Cazzato, D.; Gerevini, S.; Moscatelli, M.; Blasevich, F.; Riolo, G.; et al. PLIN4-related myopathy: Clinical, histological and imaging data in a large cohort of patients. J. Neurol. 2023, 270, 4538–4543. [Google Scholar] [CrossRef]
  104. 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]
  105. Bosch, M.; Sanchez-Alvarez, M.; Fajardo, A.; Kapetanovic, R.; Steiner, B.; Dutra, F.; Moreira, L.; Lopez, J.A.; Campo, R.; Mari, M.; et al. Mammalian lipid droplets are innate immune hubs integrating cell metabolism and host defense. Science 2020, 370, eaay8085. [Google Scholar] [CrossRef]
  106. Granneman, J.G.; Moore, H.H.; Mottillo, E.P.; Zhu, Z. Functional interactions between Mldp (LSDP5) and Abhd5 in the control of intracellular lipid accumulation. J. Biol. Chem. 2009, 284, 3049–3057. [Google Scholar] [CrossRef]
  107. Wang, H.; Bell, M.; Sreenivasan, U.; Sreenevasan, U.; Hu, H.; Liu, J.; Dalen, K.; Londos, C.; Yamaguchi, T.; Rizzo, M.A.; et al. Unique regulation of adipose triglyceride lipase (ATGL) by perilipin 5, a lipid droplet-associated protein. J. Biol. Chem. 2011, 286, 15707–15715. [Google Scholar] [CrossRef]
  108. Wang, C.; Yuan, Y.; Wu, J.; Zhao, Y.; Gao, X.; Chen, Y.; Sun, C.; Xiao, L.; Zheng, P.; Hu, P.; et al. Plin5 deficiency exacerbates pressure overload-induced cardiac hypertrophy and heart failure by enhancing myocardial fatty acid oxidation and oxidative stress. Free Radic. Biol. Med. 2019, 141, 372–382. [Google Scholar] [CrossRef]
  109. Wang, H.; Sreenivasan, U.; Hu, H.; Saladino, A.; Polster, B.M.; Lund, L.M.; Gong, D.W.; Stanley, W.C.; Sztalryd, C. Perilipin 5, a lipid droplet-associated protein, provides physical and metabolic linkage to mitochondria. J. Lipid Res. 2011, 52, 2159–2168. [Google Scholar] [CrossRef]
  110. Andersson, L.; Drevinge, C.; Mardani, I.; Dalen, K.T.; Stahlman, M.; Klevstig, M.; Lundqvist, A.; Haugen, F.; Adiels, M.; Fogelstrand, P.; et al. Deficiency in perilipin 5 reduces mitochondrial function and membrane depolarization in mouse hearts. Int. J. Biochem. Cell Biol. 2017, 91, 9–13. [Google Scholar] [CrossRef]
  111. Miner, G.E.; So, C.M.; Edwards, W.; Ragusa, J.V.; Wine, J.T.; Wong Gutierrez, D.; Airola, M.V.; Herring, L.E.; Coleman, R.A.; Klett, E.L.; et al. PLIN5 interacts with FATP4 at membrane contact sites to promote lipid droplet-to-mitochondria fatty acid transport. Dev. Cell 2023, 58, 1250–1265.E6. [Google Scholar] [CrossRef]
  112. Kien, B.; Kolleritsch, S.; Kunowska, N.; Heier, C.; Chalhoub, G.; Tilp, A.; Wolinski, H.; Stelzl, U.; Haemmerle, G. Lipid droplet-mitochondria coupling via perilipin 5 augments respiratory capacity but is dispensable for FA oxidation. J. Lipid Res. 2022, 63, 100172. [Google Scholar] [CrossRef] [PubMed]
  113. Zhou, P.L.; Li, M.; Han, X.W.; Bi, Y.H.; Zhang, W.G.; Wu, Z.Y.; Wu, G. Perilipin 5 deficiency promotes atherosclerosis progression through accelerating inflammation, apoptosis, and oxidative stress. J. Cell. Biochem. 2019, 120, 19107–19123. [Google Scholar] [CrossRef] [PubMed]
  114. Poulsen, L.; Siersbaek, M.; Mandrup, S. PPARs: Fatty acid sensors controlling metabolism. Semin. Cell Dev. Biol. 2012, 23, 631–639. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, H.; Colavitti, R.; Rovira, I.I.; Finkel, T. Redox-dependent transcriptional regulation. Circ. Res. 2005, 97, 967–974. [Google Scholar] [CrossRef]
  116. Trumper, V.; Wittig, I.; Heidler, J.; Richter, F.; Brune, B.; von Knethen, A. Redox Regulation of PPARgamma in Polarized Macrophages. PPAR Res. 2020, 2020, 8253831. [Google Scholar] [CrossRef]
  117. Herker, E.; Ott, M. Emerging role of lipid droplets in host/pathogen interactions. J. Biol. Chem. 2012, 287, 2280–2287. [Google Scholar] [CrossRef]
  118. Ding, Y.; Wang, H.; Geng, B.; Xu, G. Sulfhydration of perilipin 1 is involved in the inhibitory effects of cystathionine gamma lyase/hydrogen sulfide on adipocyte lipolysis. Biochem. Biophys. Res. Commun. 2020, 521, 786–790. [Google Scholar] [CrossRef]
  119. Xiao, H.; Jedrychowski, M.P.; Schweppe, D.K.; Huttlin, E.L.; Yu, Q.; Heppner, D.E.; Li, J.; Long, J.; Mills, E.L.; Szpyt, J.; et al. A Quantitative Tissue-Specific Landscape of Protein Redox Regulation during Aging. Cell 2020, 180, 968–983.E24. [Google Scholar] [CrossRef]
  120. Macpherson, R.E.; Vandenboom, R.; Roy, B.D.; Peters, S.J. Skeletal muscle PLIN3 and PLIN5 are serine phosphorylated at rest and following lipolysis during adrenergic or contractile stimulation. Physiol. Rep. 2013, 1, e00084. [Google Scholar] [CrossRef]
  121. 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]
  122. Doncheva, A.I.; Li, Y.; Khanal, P.; Hjorth, M.; Kolset, S.O.; Norheim, F.A.; Kimmel, A.R.; Dalen, K.T. Altered hepatic lipid droplet morphology and lipid metabolism in fasted Plin2-null mice. J. Lipid Res. 2023, 64, 100461. [Google Scholar] [CrossRef]
  123. Van der Pol, A.; van Gilst, W.H.; Voors, A.A.; van der Meer, P. Treating oxidative stress in heart failure: Past, present and future. Eur. J. Heart Fail. 2019, 21, 425–435. [Google Scholar] [CrossRef]
  124. Forrester, S.J.; Kikuchi, D.S.; Hernandes, M.S.; Xu, Q.; Griendling, K.K. Reactive Oxygen Species in Metabolic and Inflammatory Signaling. Circ. Res. 2018, 122, 877–902. [Google Scholar] [CrossRef]
  125. Amarasinghe, I.; Phillips, W.; Hill, A.F.; Cheng, L.; Helbig, K.J.; Willms, E.; Monson, E.A. Cellular communication through extracellular vesicles and lipid droplets. J. Extracell. Biol. 2023, 2, e77. [Google Scholar] [CrossRef]
  126. Zhao, P.; Jin, Y.; Wu, X.; Huang, J.; Chen, L.; Tan, Y.; Yuan, H.; Wu, J.; Ren, Z. Artificial Lipid Droplets: Novel Effective Biomaterials to Protect Cells against Oxidative Stress and Lipotoxicity. Nanomaterials 2022, 12, 672. [Google Scholar] [CrossRef]
  127. Cinato, M.; Mardani, I.; Miljanovic, A.; Drevinge, C.; Laudette, M.; Bollano, E.; Henricsson, M.; Tolo, J.; Bauza Thorbrugge, M.; Levin, M.; et al. Cardiac Plin5 interacts with SERCA2 and promotes calcium handling and cardiomyocyte contractility. Life Sci. Alliance 2023, 6, e202201690. [Google Scholar] [CrossRef]
  128. Merry, T.L.; Ristow, M. Do antioxidant supplements interfere with skeletal muscle adaptation to exercise training? J. Physiol. 2016, 594, 5135–5147. [Google Scholar] [CrossRef]
  129. Gomez-Cabrera, M.C.; Domenech, E.; Romagnoli, M.; Arduini, A.; Borras, C.; Pallardo, F.V.; Sastre, J.; Vina, J. Oral administration of vitamin C decreases muscle mitochondrial biogenesis and hampers training-induced adaptations in endurance performance. Am. J. Clin. Nutr. 2008, 87, 142–149. [Google Scholar] [CrossRef]
  130. Bajpeyi, S.; Apaflo, J.N.; Rosas, V.; Sepulveda-Rivera, K.; Varela-Ramirez, A.; Covington, J.D.; Galgani, J.E.; Ravussin, E. Effect of an acute long-duration exercise bout on skeletal muscle lipid droplet morphology, GLUT 4 protein, and perilipin protein expression. Eur. J. Appl. Physiol. 2023, 123, 2771–2778. [Google Scholar] [CrossRef]
  131. Jevons, E.F.P.; Gejl, K.D.; Strauss, J.A.; Ortenblad, N.; Shepherd, S.O. Skeletal muscle lipid droplets are resynthesized before being coated with perilipin proteins following prolonged exercise in elite male triathletes. Am. J. Physiol. Endocrinol. Metab. 2020, 318, E357–E370. [Google Scholar] [CrossRef]
  132. De Almeida, M.E.; Ortenblad, N.; Petersen, M.H.; Schjerning, A.N.; Wentorf, E.K.; Jensen, K.; Hojlund, K.; Nielsen, J. Acute exercise increases the contact between lipid droplets and mitochondria independently of obesity and type 2 diabetes. J. Physiol. 2023, 601, 1797–1815. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of the interplay between oxidative stress and perilipins (Plins). The effect of each plin on oxidative stress is shown in thick orange lines, and the effect of oxidative stress on perilipin levels and location is shown in thin arrows. Studied organs and cells as well as references (Plin1: [8,9,10]; Plin2: [11,12,13,14]; Plin3: [15]; Plin4: [16]; Plin5: [17,18,19,20,21]) are highlighted in grey for each perilipin. Grey dotted arrows highlight the current gap in knowledge.
Figure 1. Schematic illustration of the interplay between oxidative stress and perilipins (Plins). The effect of each plin on oxidative stress is shown in thick orange lines, and the effect of oxidative stress on perilipin levels and location is shown in thin arrows. Studied organs and cells as well as references (Plin1: [8,9,10]; Plin2: [11,12,13,14]; Plin3: [15]; Plin4: [16]; Plin5: [17,18,19,20,21]) are highlighted in grey for each perilipin. Grey dotted arrows highlight the current gap in knowledge.
Antioxidants 13 00209 g001
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

Cinato, M.; Andersson, L.; Miljanovic, A.; Laudette, M.; Kunduzova, O.; Borén, J.; Levin, M.C. Role of Perilipins in Oxidative Stress—Implications for Cardiovascular Disease. Antioxidants 2024, 13, 209. https://doi.org/10.3390/antiox13020209

AMA Style

Cinato M, Andersson L, Miljanovic A, Laudette M, Kunduzova O, Borén J, Levin MC. Role of Perilipins in Oxidative Stress—Implications for Cardiovascular Disease. Antioxidants. 2024; 13(2):209. https://doi.org/10.3390/antiox13020209

Chicago/Turabian Style

Cinato, Mathieu, Linda Andersson, Azra Miljanovic, Marion Laudette, Oksana Kunduzova, Jan Borén, and Malin C. Levin. 2024. "Role of Perilipins in Oxidative Stress—Implications for Cardiovascular Disease" Antioxidants 13, no. 2: 209. https://doi.org/10.3390/antiox13020209

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