Abstract

During development of type 2 diabetes (T2D), excessive nutritional load is thought to expose pancreatic islets to toxic effects of lipids and reduce β-cell function and mass. However, lipids also play a positive role in cellular metabolism and function. Thus, proper trafficking of lipids is critical for β cells to maximize the beneficial effects of these molecules while preventing their toxic effects. Lipid droplets (LDs) are organelles that play an important role in the storage and trafficking of lipids. In this review, we summarize the discovery of LDs in pancreatic β cells, LD lifecycle, and the effect of LD catabolism on β-cell insulin secretion. We discuss factors affecting LD formation such as age, cell type, species, and nutrient availability. We then outline published studies targeting critical LD regulators, primarily in rat and human β-cell models, to understand the molecular effect of LD formation and degradation on β-cell function and health. Furthermore, based on the abnormal LD accumulation observed in human T2D islets, we discuss the possible role of LDs during the development of β-cell failure in T2D. Current knowledge indicates that proper formation and clearance of LDs are critical to normal insulin secretion, endoplasmic reticulum homeostasis, and mitochondrial integrity in β cells. However, it remains unclear whether LDs positively or negatively affect human β-cell demise in T2D. Thus, we discuss possible research directions to address the knowledge gap regarding the role of LDs in β-cell failure.

The rising incidence of type 2 diabetes (T2D) is attributed to increased exposure to nutritional load that cause insulin resistance, in part by impairing the integrity of insulin target tissues. While pancreatic β cells initially compensate for insulin resistance by increasing insulin secretion and cell mass, T2D develops when β cells fail to secret sufficient insulin to maintain glucose homeostasis (1). Primary islet tissue affected by T2D shows multiple functional and structural changes in glucose sensing (2), hormone secretion (3), islet transcription factor expression (4), secretory machinery protein production (5), β-cell mass, and islet amyloid deposition, among other changes (6). A possible trigger for the changes seen in T2D-affected islets is excessive nutritional exposure, which may come from circulating toxic lipids and increased local pancreatic fat accumulation (7, 8). However, some lipids including free fatty acids (FFAs), also serve as energy sources, building blocks for a biological membrane, and potentiators of glucose-stimulated insulin secretion (GSIS) (9). Thus, regulated trafficking of lipids into and out of lipid droplets (LDs) may aid β cells in maximizing the beneficial effects of lipids while preventing their toxicity. In this review, we focus on recent findings from rodent and human β-cell models to summarize how LD-associated FFA trafficking affects β-cell function. Furthermore, we discuss the possible role LDs play during the development of islet cell failure in T2D.

Lipid Droplets Are Formed in Pancreatic β Cells

What Are Lipid Droplets?

LDs are intracellular organelles present both in prokaryotic and eukaryotic cells, first identified microscopically in the 1880s as discussed in a recent review of LDs (10). LDs consist of a monolayer phospholipid membrane and a neutral lipid core composed of triglycerides (TGs), cholesterol ester, and retinol ester (Fig. 1A) (10, 11). LDs were viewed as an inert fat depot until the late 1980s, when investigations began to reveal the dynamic effect of LDs on cellular metabolism through work initially focused on the adipocytes (10). LDs are now recognized to play an integral part in lipid metabolism in a wide range of cells by allowing the flexible usage of FFAs (12, 13). The lipids stored in LDs can be released and used for fuel, signaling, and the synthesis of structural components at a time and place that fulfills cellular demands (14). LDs can also defend cells by sequestrating potentially toxic forms of lipid molecules (15). Moreover, LDs dynamically interact with the endoplasmic reticulum (ER), mitochondria, autophagosomes, lysosomes, and nuclei to coordinate lipid metabolism together (14). Thus, LDs are now recognized as important regulatory structures in adipocytes, myocytes, hepatocytes, and others.

Islet β-cell lipid droplets (LDs) contain perilipin (PLIN)2, PLIN3, and PLIN5 proteins. A, An LD in a β cell originates from the endoplasmic reticulum (ER) and consists of a core of neutral lipids covered by a monolayer of phospholipids. The PLIN2, PLIN3, and PLIN5 proteins (rectangles) reside on the surface of the LD and have profound effects on activities of LDs. B, Rat insulinoma cell line INS-1 cells were transduced by lentivirus expressing PLIN2-mRuby (red), and neutral lipids were visualized with BODIPY 493 (green) after fixation of the cells. Unpublished data. C, Human islets cultured with BODIPY C12 (BODIPY-FA red) were dispersed and immunostained for PLIN2 (20R-AP002, Fitzgerald) and PLIN5 (green, GP-31, Progen). PLINs surround BODIPY C12 containing LDs in primary human β cells. Reproduced from (24) with permission from the American Diabetes Association. D, Lipid-rich structures identified by BODIPY 493 staining in human pancreatic sections showing PLIN2 (green, ab108323, Abcam) and PLIN3 (green, sc-390968, Santa Cruz) colocalization in pancreatic β cells (marked by INS [white, A0564, DAKO/Agilent]). Top panel, 47-year-old nondiabetic donor; bottom panel, 35-year-old nondiabetic donor. Reproduced from (22) with permission from the American Diabetes Association. B to D, DAPI (4′, 6-diamidino-2-phenylindole) shown in blue. All scale bars: 10 µm.
Figure 1.

Islet β-cell lipid droplets (LDs) contain perilipin (PLIN)2, PLIN3, and PLIN5 proteins. A, An LD in a β cell originates from the endoplasmic reticulum (ER) and consists of a core of neutral lipids covered by a monolayer of phospholipids. The PLIN2, PLIN3, and PLIN5 proteins (rectangles) reside on the surface of the LD and have profound effects on activities of LDs. B, Rat insulinoma cell line INS-1 cells were transduced by lentivirus expressing PLIN2-mRuby (red), and neutral lipids were visualized with BODIPY 493 (green) after fixation of the cells. Unpublished data. C, Human islets cultured with BODIPY C12 (BODIPY-FA red) were dispersed and immunostained for PLIN2 (20R-AP002, Fitzgerald) and PLIN5 (green, GP-31, Progen). PLINs surround BODIPY C12 containing LDs in primary human β cells. Reproduced from (24) with permission from the American Diabetes Association. D, Lipid-rich structures identified by BODIPY 493 staining in human pancreatic sections showing PLIN2 (green, ab108323, Abcam) and PLIN3 (green, sc-390968, Santa Cruz) colocalization in pancreatic β cells (marked by INS [white, A0564, DAKO/Agilent]). Top panel, 47-year-old nondiabetic donor; bottom panel, 35-year-old nondiabetic donor. Reproduced from (22) with permission from the American Diabetes Association. B to D, DAPI (4′, 6-diamidino-2-phenylindole) shown in blue. All scale bars: 10 µm.

The surface of mammalian LDs is typically decorated by members of the perilipin (PLIN) protein family (ie, PLIN1-5, Fig. 1A-B). PLIN composition varies between different cell types and can profoundly affect the behavior of LDs by altering LD size, lipid cargo composition (eg, diacylglycerol, TGs, cholesterol ester), and LD interacting proteins (eg, lipases, colipases, chaperons, transcription factors). PLINs may also directly mediate the tethering of LDs to other organelles (Table 1) (16). In addition to the PLIN family of proteins, the LD membrane houses a variety of proteins that support the diverse functions of LDs. Proteins that regulate lipid metabolism, vesicular trafficking, cell signaling, transcription, and interorganelle communication were demonstrated in a recent proteomic study of LDs in which a proximity labeling strategy was applied to detect LD-associated proteins (17).

Table 1.

Perilipins in mammalian cellsa

Expression pattern Characteristics known in nonislet cells Expression in pancreatic islets and β-cell lines
PLIN1Adipocytes, steroidogenic cellsIncreases lipolysis robustly on PKA activation, and prevents lipolysis during nonstimulated status (16)• Negligible expression in primary mouse and human β cells (7, 83)
• Low levels reported in β-cell lines (INS-1 cells)
PLIN2Ubiquitous; most abundant PLIN in nonadipocytes• Closely correlates with abundance of LDs and neutral lipid contents
• Lacks PKA consensus site
• Passive barrier to block lipolysis and/or lipophagy (16)
Most abundant PLIN in all β-cell lines (mouse MIN6 cells, rat INS1 cells, human EndoC-βH2) and islets (mouse and human) tested (7, 22, 24, 32)
PLIN3Ubiquitous• Distributed in cytosol and on surface of LDs
• Compensates for PLIN2 as an LD protein when PLIN2 is downregulated (89)
• mRNA levels comparable to PLIN2 in human islets and are second most abundant PLIN in mouse islets (7, 22)
• PLIN3/PLIN2 ratio higher in α cells than β cells (83)
• Very low expression in β-cell lines, including MIN6 cells, INS1 cells, and EndoC-βH2 cells (7, 22)
PLIN4Adipocytes, heart, skeletal muscle• Preferentially coats nascent LDs
• Prevents lipolysis
Negligible expression in β-cell lines and islets
PLIN5Highly expressed in oxidative tissues, especially in cardiomyocytes (90, 91)
Highly inducible by fasting in muscle, liver, and pancreatic islets in mice (24, 92)
• Has one PKA consensus site
• Increases lipolysis
• cAMP-dependent translocation to nucleus (16, 75)
• Transports MUFA into nucleus for PGC1a/SIRT1 activation (78)
• Aids regulated transfer of FFAs to mitochondria for FAO (77), through high physical association (76)
Third most abundant PLIN in human and mouse islets but extremely low in β-cell lines (7, 22, 32)
Expression pattern Characteristics known in nonislet cells Expression in pancreatic islets and β-cell lines
PLIN1Adipocytes, steroidogenic cellsIncreases lipolysis robustly on PKA activation, and prevents lipolysis during nonstimulated status (16)• Negligible expression in primary mouse and human β cells (7, 83)
• Low levels reported in β-cell lines (INS-1 cells)
PLIN2Ubiquitous; most abundant PLIN in nonadipocytes• Closely correlates with abundance of LDs and neutral lipid contents
• Lacks PKA consensus site
• Passive barrier to block lipolysis and/or lipophagy (16)
Most abundant PLIN in all β-cell lines (mouse MIN6 cells, rat INS1 cells, human EndoC-βH2) and islets (mouse and human) tested (7, 22, 24, 32)
PLIN3Ubiquitous• Distributed in cytosol and on surface of LDs
• Compensates for PLIN2 as an LD protein when PLIN2 is downregulated (89)
• mRNA levels comparable to PLIN2 in human islets and are second most abundant PLIN in mouse islets (7, 22)
• PLIN3/PLIN2 ratio higher in α cells than β cells (83)
• Very low expression in β-cell lines, including MIN6 cells, INS1 cells, and EndoC-βH2 cells (7, 22)
PLIN4Adipocytes, heart, skeletal muscle• Preferentially coats nascent LDs
• Prevents lipolysis
Negligible expression in β-cell lines and islets
PLIN5Highly expressed in oxidative tissues, especially in cardiomyocytes (90, 91)
Highly inducible by fasting in muscle, liver, and pancreatic islets in mice (24, 92)
• Has one PKA consensus site
• Increases lipolysis
• cAMP-dependent translocation to nucleus (16, 75)
• Transports MUFA into nucleus for PGC1a/SIRT1 activation (78)
• Aids regulated transfer of FFAs to mitochondria for FAO (77), through high physical association (76)
Third most abundant PLIN in human and mouse islets but extremely low in β-cell lines (7, 22, 32)

Abbreviations: cAMP, 3′,5′-cyclic adenosine 5′-monophosphate; FAO, fatty acid oxidation; FFA, free fatty acids; INS, insulin; LD, lipid droplet; mRNA, messenger RNA; MUFA, monounsaturated fatty acid; PGC1a, peroxisome proliferator-activated receptor γ coactivator 1-a; PKA, protein kinase A; PLIN, perilipin; SIRT1, sirtuin 1.

aIn addition to specific references cited, refer to published reviews (7, 93).

Table 1.

Perilipins in mammalian cellsa

Expression pattern Characteristics known in nonislet cells Expression in pancreatic islets and β-cell lines
PLIN1Adipocytes, steroidogenic cellsIncreases lipolysis robustly on PKA activation, and prevents lipolysis during nonstimulated status (16)• Negligible expression in primary mouse and human β cells (7, 83)
• Low levels reported in β-cell lines (INS-1 cells)
PLIN2Ubiquitous; most abundant PLIN in nonadipocytes• Closely correlates with abundance of LDs and neutral lipid contents
• Lacks PKA consensus site
• Passive barrier to block lipolysis and/or lipophagy (16)
Most abundant PLIN in all β-cell lines (mouse MIN6 cells, rat INS1 cells, human EndoC-βH2) and islets (mouse and human) tested (7, 22, 24, 32)
PLIN3Ubiquitous• Distributed in cytosol and on surface of LDs
• Compensates for PLIN2 as an LD protein when PLIN2 is downregulated (89)
• mRNA levels comparable to PLIN2 in human islets and are second most abundant PLIN in mouse islets (7, 22)
• PLIN3/PLIN2 ratio higher in α cells than β cells (83)
• Very low expression in β-cell lines, including MIN6 cells, INS1 cells, and EndoC-βH2 cells (7, 22)
PLIN4Adipocytes, heart, skeletal muscle• Preferentially coats nascent LDs
• Prevents lipolysis
Negligible expression in β-cell lines and islets
PLIN5Highly expressed in oxidative tissues, especially in cardiomyocytes (90, 91)
Highly inducible by fasting in muscle, liver, and pancreatic islets in mice (24, 92)
• Has one PKA consensus site
• Increases lipolysis
• cAMP-dependent translocation to nucleus (16, 75)
• Transports MUFA into nucleus for PGC1a/SIRT1 activation (78)
• Aids regulated transfer of FFAs to mitochondria for FAO (77), through high physical association (76)
Third most abundant PLIN in human and mouse islets but extremely low in β-cell lines (7, 22, 32)
Expression pattern Characteristics known in nonislet cells Expression in pancreatic islets and β-cell lines
PLIN1Adipocytes, steroidogenic cellsIncreases lipolysis robustly on PKA activation, and prevents lipolysis during nonstimulated status (16)• Negligible expression in primary mouse and human β cells (7, 83)
• Low levels reported in β-cell lines (INS-1 cells)
PLIN2Ubiquitous; most abundant PLIN in nonadipocytes• Closely correlates with abundance of LDs and neutral lipid contents
• Lacks PKA consensus site
• Passive barrier to block lipolysis and/or lipophagy (16)
Most abundant PLIN in all β-cell lines (mouse MIN6 cells, rat INS1 cells, human EndoC-βH2) and islets (mouse and human) tested (7, 22, 24, 32)
PLIN3Ubiquitous• Distributed in cytosol and on surface of LDs
• Compensates for PLIN2 as an LD protein when PLIN2 is downregulated (89)
• mRNA levels comparable to PLIN2 in human islets and are second most abundant PLIN in mouse islets (7, 22)
• PLIN3/PLIN2 ratio higher in α cells than β cells (83)
• Very low expression in β-cell lines, including MIN6 cells, INS1 cells, and EndoC-βH2 cells (7, 22)
PLIN4Adipocytes, heart, skeletal muscle• Preferentially coats nascent LDs
• Prevents lipolysis
Negligible expression in β-cell lines and islets
PLIN5Highly expressed in oxidative tissues, especially in cardiomyocytes (90, 91)
Highly inducible by fasting in muscle, liver, and pancreatic islets in mice (24, 92)
• Has one PKA consensus site
• Increases lipolysis
• cAMP-dependent translocation to nucleus (16, 75)
• Transports MUFA into nucleus for PGC1a/SIRT1 activation (78)
• Aids regulated transfer of FFAs to mitochondria for FAO (77), through high physical association (76)
Third most abundant PLIN in human and mouse islets but extremely low in β-cell lines (7, 22, 32)

Abbreviations: cAMP, 3′,5′-cyclic adenosine 5′-monophosphate; FAO, fatty acid oxidation; FFA, free fatty acids; INS, insulin; LD, lipid droplet; mRNA, messenger RNA; MUFA, monounsaturated fatty acid; PGC1a, peroxisome proliferator-activated receptor γ coactivator 1-a; PKA, protein kinase A; PLIN, perilipin; SIRT1, sirtuin 1.

aIn addition to specific references cited, refer to published reviews (7, 93).

Lipid Droplets Are Detected in Pancreatic β Cells

In a 1998 seminal work, Shimabukuro et al (18) proposed that lipid accumulation directly contributes to β-cell demise. Unger and colleagues (19) demonstrated that islet TG content of the diabetic ZDF rat increased 100-fold compared to the lean control. Since then, volumes of work have examined the regulation of lipid homeostasis in β cells under normal and pathological conditions. However, the role of LDs in islets has been untapped for many years, likely because of an inability to visualize LDs in mouse β cells by imaging technique, including electron microscopy (EM) (20-22). TG synthesis in mouse islets is nutritionally regulated, as a high-fat diet (HFD) and fasting in vivo have been shown to increase TGs along with the expression of 2 PLINs (PLIN2 and PLIN5) that are known to associate with TG-rich LDs (23-25). However, this increase in TGs has not been proven to cause visible LD accumulation in mouse islets despite the increase in PLIN expression (20, 23).

Islet LD research began to capture more interest recently when LDs were clearly demonstrated in rat and human adult islet β cells (20, 24, 26). Although the lipid composition of LDs in β cells has not been directly assessed, β-cell LDs appear to be enriched with TGs. BODIPY 558/568 C12 (BODIPY C12) is a fluorescent long-chain fatty acid (LCFA) analogue preferentially incorporated into TG-rich LDs (25, 27). Trevino et al showed that LDs in human β cells are enriched with BODIPY C12 and coated by 2 PLINs (PLIN2 and PLIN5) known to associate with TG-rich LDs (24, 25) (Fig. 1C). In addition, knockdown of adipose triglyceride lipase (ATGL), a key TG catabolic enzyme, has been shown to expand the size and number of LDs in human β cells, further supporting that β-cell LDs are enriched with TGs (21).

The size of LDs in mammalian cells varies widely, from 20 nm to more than 100 µm in diameter, depending on cell type and differentiation status (28). LDs in human β cells are small, with the majority being smaller than 1.2 µm (21). In comparison, mature white adipocytes that specialize in TG storage possess an extremely large unilocular LD occupying most of the cell volume. This unilocular LD is unique to mature adipocytes as immature adipocytes and nonadipocytes usually possess multiple smaller LDs per cell (29). The diameter of the human adipocyte is reported to be 80 to 120 µm on average and may reach up to 300 µm (30). Among nonadipocytes, hepatocytes have a high capacity for TG synthesis and are well studied for pathological accumulation of LDs, known as hepatosteatosis. LD size in hepatocytes is between human β cells and adipocytes, ranging from 60 nm to 5 µm (31). The composition of PLINs expressed in each cell is one likely factor that affects LD size. The expression of PLIN1 is highly specific to mature adipocytes and considered to play a key role in the formation of a large unilocular LD, while PLIN2 is the dominant PLIN in nonadipocytes, including hepatocytes and human β cells (7, 22).

Factors Associated With Lipid Droplet Accumulation in Human β Cells

The number and size of LDs in a given cell also change in response to extrinsic factors, such as stress and the availability of nutrients (12, 15, 29). Indeed, LD accumulation in rat islets and human β cell lines has been shown to increase because of exposure to FFAs and glucose (26, 32). Increased LD accumulation was also a prominent feature of nutritionally stressed and dysfunctional human β cells in vivo (20). In a study by Dai et al (20), human and mouse islets were transplanted into immunodeficient 3-month-old NSG SCID mice to compare their response to chronic nutritional stress. When the recipient mice were placed on a 60% HFD for 3 months after transplantation, transplanted mouse islet β cells showed the expected compensatory increase in proliferation and glucose-stimulated insulin secretion (GSIS). However, β cells in the transplanted human islets did not show a compensatory increase in proliferation or GSIS. Moreover, the expression levels of the key islet transcription factors (MAFB and NKX6.1) were compromised in transplanted human islets but not in mouse islets. Amyloid plaque is a marker associated with dysfunctional human T2D islets and is observed in some young adults with type 1 diabetes (6, 33). Increased deposition of amyloid plaques was seen in human islet transplant on HFD exposure but not in the mouse islet transplant (20). Another unique feature of transplanted human islets was the presence of LDs whose numbers increased on HFD exposure.

As the increase in LDs appears to be a feature in the dysfunctional, transplanted human islets, Tong et al (22) addressed whether LD accumulation is also increased in native T2D islets. When LDs were assessed by neutral lipid staining (BODIPY 493/503 [BODIPY 493]) and PLIN immunostaining in human pancreas, both BODIPY 493 and PLIN2 signals were significantly enriched in α and β cells of T2D islets in relation to surrounding exocrine acinar cells. The increase of β-cell PLIN2 immunoreactivity in T2D islets was also reported independently (34).

In addition to nutritional stress and T2D status, β-cell age and maturation levels appear to be factors associated with LD accumulation in human β cells. Tong et al (22) reported that the number of LDs detected by EM or BODIPY 493 within β cells was low in young human pancreatic donors (age < 10 y) and steadily increased to greater than 6% (LD area/islet area) in older adult donors. LDs have been found to accumulate in human stem cell derived insulin+ β-like cells (35) that were molecularly mature and glucose-responsive when assessed during culture in vitro and after exposure to an in vivo environment by transplantation into NSG mice. In contrast, LDs were absent in less mature, glucose nonresponsive insulin+ β-like cells at an earlier developmental stage (22).

Collectively, LD accumulation in human islets is associated with multiple conditions that also affect β-cell health and function, highlighting the importance of addressing a potential role of LDs in human islets. It also raises the key question of whether abnormal LD accumulation and/or function contribute to the dysfunction of transplanted human islets exposed to an HFD or islets from individuals with T2D.

Distinguishing Lipid Droplets From Lipofuscin

More than 60% of lipid-rich structures visualized by BODIPY493 in human β cells colocalized with PLIN2+ and PLIN3+ immunoreactivity, indicating that they are LDs (Fig. 1D) (22). The remaining neutral lipid staining that lacks PLINs likely represents lysosomal-derived lipofuscins (Fig. 2) (22, 34). First characterized by Cnop et al (36, 37) in human islets, lipofuscins are lipid-laden lysosome-derived structures that show immunoreactivity for cathepsin D and accumulate in postmitotic cells. Lipofuscins react to neutral lipid staining (eg, oil red O and BODIPY 493) and are identified by EM as a heavily pigmented crown structure surrounding a single- or multiple-lipid core. Lipofuscins are found in many different cell types, with abundance positively correlating with cell age (37, 38). In human islets, the number of lipofuscins is substantially lower in α cells compared to β cells (37). Theoretically, BODIPY 493-positive structures that contain lysosomal LAMP1, and not PLIN2 or PLIN3, are expected to be lipofuscins in human β cells (Fig. 2). However, it is plausible that lipofuscins contain lipid cores originating from LDs and delivered to lysosome through autophagy (lipophagy) (39). Unfortunately, detailed information regarding the contribution of LDs to the formation of lipofuscins is lacking and awaits future study.

Lipid droplets (LDs) and lipofuscins are both detectable in human islet β cells. A, Confocal image of lipid-rich structure identified by BODIPY493 (green) in primary human β cells (identified by positive staining with insulin antibody) shown with immunostaining of LAMP1 (red, 1D4B, Developmental Studies Hybridoma Bank). LAMP1– lipid-rich structures (LDs marked by red arrows) and LAMP1+ structures (lipofuscins marked by yellow stars) were both present. Human islets from IIDP (52-year-old nondiabetic donor). B, Representative electron microscopy image of human islet β cells including LDs (red circle) and lipofuscins (yellow rectangle). Human islets from Alberta Diabetes Institute (54-year-old nondiabetic donor). C and D, Details from B, include a cluster of LDs (red arrows) in C, and lipofuscins (yellow stars) in D. A to D, Unpublished data.
Figure 2.

Lipid droplets (LDs) and lipofuscins are both detectable in human islet β cells. A, Confocal image of lipid-rich structure identified by BODIPY493 (green) in primary human β cells (identified by positive staining with insulin antibody) shown with immunostaining of LAMP1 (red, 1D4B, Developmental Studies Hybridoma Bank). LAMP1 lipid-rich structures (LDs marked by red arrows) and LAMP1+ structures (lipofuscins marked by yellow stars) were both present. Human islets from IIDP (52-year-old nondiabetic donor). B, Representative electron microscopy image of human islet β cells including LDs (red circle) and lipofuscins (yellow rectangle). Human islets from Alberta Diabetes Institute (54-year-old nondiabetic donor). C and D, Details from B, include a cluster of LDs (red arrows) in C, and lipofuscins (yellow stars) in D. A to D, Unpublished data.

What Is the Role of β-Cell Lipid Droplets in the Regulation of Insulin Secretion?

Glycerolipid/Free Fatty Acid Cycle and Insulin Secretion

As elegantly summarized by Prentki et al (9, 40), lipolysis of TGs has long been considered to generate metabolites that augment insulin secretion through a process termed the glycerolipid/free fatty acid (GL/FFA) cycle. The GL/FFA cycle starts from the incorporation of FFAs to GLs represented by TGs and is followed by the release of FFAs from TGs through lipolysis. LDs take the central stage of the GL/FFA cycle; newly synthesized TGs are primarily transferred to LDs and lipolysis occurs on LDs. While LD formation has not been studied in depth in β cells, the processes and key proteins involved in LD formation and lipolysis (GL/FFA cycle) are well conserved among mammalian cells and likely shared with β cells (41). Fig. 3 summarizes the current understanding of the process of LD formation and lipolysis, along with the known effects of key lipolytic metabolites on insulin secretory machinery.

 Fate of fatty acids during lipid droplet (LD) formation and lipolysis in β cells. Triglycerides (TG) are synthesized in the endoplasmic reticulum (ER) membrane from fatty acyl-CoA (FA-CoA) and glycerol-3-phosphate (glycerol 3-P), derived from glucose transported through the glucose transporter (GLUT2 for rodents and GLUT1/2 for humans). Sequential addition of a FA chain from FA-CoA to glycerol 3-P forms lysophosphatidic acids (LPA) and phosphatidic acids (PA). Removal of phosphate from PA generates 1,2-diacylglycerol (1,2-DAG) that has 2 FA chains. The addition of a third FA chain to 1,2-DAG by diacylglycerol transferase (DGAT) 1 completes the formation of TG at the ER bilayer. TG accumulated within the ER bilayer first form a lens structure, which eventually grows into an LD and buds off from the ER. FAs can be directly incorporated into TG at LDs by DGAT2. Adipose triglyceride lipase (ATGL) is a major TG lipase that initiates lipolysis by removing FAs from the glycerol backbone at the Sn1 or Sn2- position, creating 2,3-DAG or 1,3-DAG. Hormone-sensitive lipase (HSL) has high activity against DAG and preferentially releases FAs from Sn3, creating 1-monoacylglycerol (1-MAG) or 2-MAG. 1-MAG is known to increase insulin secretion by activating MUNC13-1 (48). FAs can be secreted from β cells and activate cell surface FA receptors such as FFAR1. Alternatively, FAs are converted to FA-CoA for signaling to active PPARδ, palmitoylation of STX1a, or TG synthesis (21, 47).
Figure 3.

Fate of fatty acids during lipid droplet (LD) formation and lipolysis in β cells. Triglycerides (TG) are synthesized in the endoplasmic reticulum (ER) membrane from fatty acyl-CoA (FA-CoA) and glycerol-3-phosphate (glycerol 3-P), derived from glucose transported through the glucose transporter (GLUT2 for rodents and GLUT1/2 for humans). Sequential addition of a FA chain from FA-CoA to glycerol 3-P forms lysophosphatidic acids (LPA) and phosphatidic acids (PA). Removal of phosphate from PA generates 1,2-diacylglycerol (1,2-DAG) that has 2 FA chains. The addition of a third FA chain to 1,2-DAG by diacylglycerol transferase (DGAT) 1 completes the formation of TG at the ER bilayer. TG accumulated within the ER bilayer first form a lens structure, which eventually grows into an LD and buds off from the ER. FAs can be directly incorporated into TG at LDs by DGAT2. Adipose triglyceride lipase (ATGL) is a major TG lipase that initiates lipolysis by removing FAs from the glycerol backbone at the Sn1 or Sn2- position, creating 2,3-DAG or 1,3-DAG. Hormone-sensitive lipase (HSL) has high activity against DAG and preferentially releases FAs from Sn3, creating 1-monoacylglycerol (1-MAG) or 2-MAG. 1-MAG is known to increase insulin secretion by activating MUNC13-1 (48). FAs can be secreted from β cells and activate cell surface FA receptors such as FFAR1. Alternatively, FAs are converted to FA-CoA for signaling to active PPARδ, palmitoylation of STX1a, or TG synthesis (21, 47).

Briefly, LD formation starts as TG accumulation within the ER membrane bilayer at the area marked by the highly conserved, multifunctional protein seipin (41). Diacylglycerol acyltransferase 1 (DGAT1) is an ER-resident protein that catalyzes the last step of TG formation in the ER (12). One function of seipin is to aid the induction of membrane curvature to accommodate the accumulation of synthesized TGs for the formation of a lens structure (see Fig. 3) (42). Seipin and proteins recruited by seipin, such as lipid droplet-associated factor 1 (LDAF1), facilitate further accumulation of TGs to form a lipid core. Then, a lipid core covered by a phospholipid monolayer eventually buds off as a nascent LD (41). Normally, LD formation at the ER membrane is a highly efficient process, with FFAs incorporated into TGs within seconds after entering the cells (14, 43). Glucose drives TG synthesis in β cells by blocking fatty acid oxidation (FAO) and directing FFAs from an energy substrate to TG synthesis (7). PLIN proteins are inserted into LDs soon after budding from the ER and play a key role in regulating lipolysis rate by modulating the access of neutral lipases to the LD surface (see Fig 1A, Fig. 3, and Table 1). After emerging from the ER, LDs can continue to grow by adding TGs using LD-associated enzymes including DGAT2 (12). Even though both DGAT1 and DGAT2 catalyze the last step of TG synthesis, the structural differences lead to their unique localization, with DGAT2 principally residing on the phospholipid monolayer of LD and DGAT1 mostly in the ER (12). Interestingly, the expression of DGAT2 but not DGAT1 was found to be upregulated in human pseudoislets after ATGL downregulation, implicating the role of DGAT2 in the expansion of the LD pool that occurs in ATGL-deficient human β cells (21). It currently remains unknown whether DGAT1 and/or 2 regulate LD levels in an age- or nutrition-dependent manner in human β cells.

Insulin granule biogenesis is one of the most important bilayer vesicle-trafficking events in β cells. It differs from LD biogenesis in the synthesis site of contents, trafficking of vesicles, and the structure of surface membranes. Proinsulin processing starts at the ER lumen followed by transport of proinsulin aggregates to the Golgi apparatus in a COPII (and likely also COPI) vesicle-dependent manner (44, 45). The trans-Golgi network plays a key role in the formation of insulin granules that eventually bud off as vesicles encased by a phospholipid bilayer. Insulin granules continue maturation before delivery to the plasma membrane for exocytosis in response to secretagogue stimulation (46).

How does LD degradation (ie, lipolysis) regulate insulin secretion? It is unlikely that FFAs released from LDs support GSIS through FAO, as FAO is suppressed by glucose and negatively correlates with insulin secretion (9). FFAs released from LDs are thought to augment insulin secretion by activating peroxisome proliferator-activated receptors-δ (PPAR δ) and promoting adenosine 5′-triphosphate production in the mitochondria (47). FFAs secreted locally may activate the FFAR1 cell surface receptor and potentiate GSIS (9). 1-monoacylglyceride (1-MAG) is also generated during lipolysis and activates MUNC13-1 in the SNARE complex to stimulate insulin granule exocytosis (48). Protein modification by FFAs such as palmitoylation (49) is another mechanism by which FFA might affect insulin secretion as discussed in later (21).

Adipose Triglyceride Lipase Regulates Glucose-Stimulated Lipolysis and Lipid Droplet Formation in β Cells

For the GL/FFA cycle to increase insulin secretion effectively, lipolysis must be active in glucose-stimulated β cells. Liu et al (21) showed that ATGL is the enzyme responsible for glucose-stimulated lipolysis in human β cells. ATGL downregulation impaired glucose- and KCl-stimulated insulin secretion in human pseudoislets and rat INS-1 cells. The reduction of insulin secretion in these models was accompanied by a decrease in the syntaxin1a (STX1a) vesicle docking protein without changes in MUNC18 and SNAP25 protein levels. Palmitoylation of STX1a by FFAs was reduced in ATGL-deficient cells, likely accelerating STX1a degradation. In addition to the activation of MUNC13-1 and PPARδ mentioned earlier, STX1a stability regulated by palmitoylation represents another example of how lipolytic metabolites support insulin secretion (see Fig. 3) (47, 48). Unlike human islets and INS-1cells, mouse islets fail to increase lipolysis in response to glucose, demonstrating the differences in the regulation of lipolysis between human, rat, and mouse islets (21). In general, lipolysis of LDs is regulated by multiple factors, including phosphorylation of PLIN1/5, recruitment of lipases to LDs, and interaction between lipases and lipase activators and inhibitors (eg, ABHD5, G0S2, HILPDA) (16, 39, 50). Currently, a mechanism by which glucose activates lipolysis in β cells has not been determined. Interestingly, glucose-stimulated lipolysis is impaired in human T2D islets (21), a defect that could disrupt the GL/FFA cycle, increase LD accumulation, and impair insulin secretion.

Is Lipid Droplet Accumulation in Aging and Diabetic β Cells a Defender or a Contributor of β-Cell Demise?

LD accumulation in adult human β cells is potentially important for insulin secretion by providing metabolic intermediates through regulated lipolytic processes (21). In contrast, the LD accumulation seen in diabetic and possibly aging β cells may reflect an imbalance in LD formation and catabolism (22). Here, we discuss how dysregulated LD accumulation may affect β cells.

Lipid Droplets Appear to Defend β Cells Under Glucolipotoxic Conditions

Exposure to saturated LCFA (C13-21) in the presence of high glucose levels causes cytotoxic stress in β-cell lines and cultured islets, a phenomenon termed glucolipotoxicity (51). Palmitic acid (PA, C16:0) is the most abundant LCFA in the human body (52), and its level is significantly elevated in the blood of individuals with obesity or metabolic syndromes (53, 54). PA impairs rodent β-cell viability and function by inducing oxidative, ER, and inflammatory stress pathways (7, 51). In addition, PA compromises insulin secretion and causes apoptosis in human islets (55, 56). In contrast, monounsaturated FAs (MUFAs), represented by oleic acid (OA), are well tolerated by β cells. Interestingly, the toxicity of FFAs in rat INS-1 cells and rat islets inversely correlates with their ability to be incorporated into TGs. OA promotes TG synthesis and LD formation, whereas PA is much less efficiently incorporated into TGs (57). The cytotoxic effects of PA are greatly alleviated by coincubation with OA, which might be due to improved sequestration of PA into LDs (57, 58). Stearoyl-CoA desaturase (SCD) is an enzyme that catalyzes double bond formation, and its activation increases the OA to PA ratio favoring TG synthesis and LD biogenesis (59). In fact, overexpression of SCD2 reduces, while downregulation/inhibition of SCD1 exacerbates, PA toxicity in mouse MIN6 cells and rat INS-1 cells (59-61).

Interestingly, the expression of SCD1 is high in the human EndoC-βH1 cell line, a cell line that shows efficient LD formation and resistance to PA-induced stress (62, 63). Downregulation of SCD1 has been shown to make EndoC-βH1 cells susceptible to inflammation and ER stress after PA exposure (64). Collectively, these results imply that glucolipotoxicity in β cells can be prevented/reduced by increasing TG synthesis and expanding the LD pool size. However, results obtained in vitro using a single FA species may not reproduce changes that islets undergo in vivo, as FFA milieu in vivo is composed of a complex mixture of FFAs. Also, FFA toxicity is not always diminished by the efficiency of FFAs to induce LD biogenesis; monounsaturated erucic acid (C22:1) is highly cytotoxic to EndoC-βH1/2 cells despite efficient promotion of LD formation (32, 65).

Possible Function of Stress-induced Lipid Droplets in β Cells

Multiple stress pathways are activated in β cells undergoing aging- or T2D-related cell demise (1, 66), appreciating that age alone increases the risk of T2D (67-69). Interestingly, the same stress conditions are reported to promote LD accumulation in many different cellular contexts (11). While a detailed description of the relationship between LDs and each stress pathway is beyond the scope of this review, LD buildup correlates with oxidative stress in cancer cells and hepatocytes (11). Moreover, ER stress induces LD accumulation in hepatocytes, which is attributed both to increased lipogenesis and reduced FAO activity (70, 71).

In general, the accumulation of LDs is considered to be important in neutralizing the stress response (15, 72). During Drosophila development, glial cells generate polyunsaturated FAs (PUFAs) at the plasma membrane by oxidative stress. Sequestration of generated PUFA into LDs is shown to protect the glial cells and adjacent neuronal stem cells from reactive oxygen species generated from free PUFA peroxidation (73). Promotion of FFA storage into LDs also appears to mitigate ER stress in the mouse liver (11). TGs stored in LDs provide a precursor for eicosanoid production to modulate the cellular response to inflammation (43). Healthy neurons typically contain very few LDs as they transfer lipids to surrounding microglia cells. Microglia cells incorporate transferred lipids into LDs, but eventually reach an LD-saturation threshold. This coincides with the timing when adjacent neurons start to accumulate LDs and develop signs of dysfunction (74). Thus, the transfer of lipids from neurons to LDs forming microglia cells appears to be a protective mechanism for neurons. Currently, the specific role(s) of LDs in human β cells under stress is still unclear, although targeting of PLIN protein production in β-cell models has begun to provide some insight (Table 2).

Table 2.

Studies of perilipin overexpression/downregulation in β-cell models

Models Changes in lipid metabolism Changes in β-cell health and function References
Overexpression
PLIN1INS1 cell• Reduced lipolysis
• No change in FAO
GSIS reduced under regular culture condition but conferred protection against chronic PA-induced GSIS reduction(84)
PLIN2MIN6 cells mouse islet• Increased TG accumulation
• No change in lipolysis or FAO
No effect on GSIS under regular culture conditions(23)
MIN6 cellsN/TIncreased ATF4 and CHOP expression with or without tunicamycin(88)
EndoC-βH2 cellsIncreased LD levels• Prevented erucic acid induced GSIS reduction
• Increased expression of positive effectors of insulin secretion
(32)
PLIN5MIN6 cells mouse islet• Increased TG accumulation
• Increased lipolysis
Increased augmentation of GSIS by cAMP and PA in vitro and by GLP-1 receptor agonist in vivo(24)
INS-1 cells• Increased LD levels
• Reduced ATGL expression and increased CPT1a expression
• Prevented chronic PA-induced GSIS reduction
• Reduced PA induced markers of ER stress and oxidative stress
(81, 82)
Overexpression in miceaN/TImproved glucose tolerance under HFD(81)
Downregulation
PLIN1RIN-m5F cells, INS-1 cellsReduced LD levelsNo protection against apoptosis by chronic FA(87)
PLIN2MIN6 cellsReduced TG accumulation and lipolysisReduced acute augmentation of GSIS by PA(23)
RIN-m5F cells, INS-1 cellsReduced LD levelsNo protection against apoptosis by chronic FA(87)
MIN6 cells, whole-body PLIN2 knockout miceN/T• Protection against ER stress caused by PA, tunicamycin, and INS mutation in Akita mice
• Upregulated autophagy
(88)
INS1 cells, β-cell–specific PLIN2 knockout mice, human pseudoislets• Reduced FAO
• Increased medium-chain acylcarnitine levels
• Increased influx of fluorescent OA analogue to mitochondria
• Impaired GSIS
• Reduced OXPHOS proteins
(85)
EndoC-βH2 cellsReduced LD levels• Impaired GSIS
• Increased ER stress markers
• ER stress inhibitor TUDCA partially restores GSIS
• Reduced expression of β-cell–specific genes and positive regulators of insulin secretion
(32)
PLIN5INS-1 cellsN/TIncreased ER stress markers. This change can be prevented by CPT1a overexpression(81)
Models Changes in lipid metabolism Changes in β-cell health and function References
Overexpression
PLIN1INS1 cell• Reduced lipolysis
• No change in FAO
GSIS reduced under regular culture condition but conferred protection against chronic PA-induced GSIS reduction(84)
PLIN2MIN6 cells mouse islet• Increased TG accumulation
• No change in lipolysis or FAO
No effect on GSIS under regular culture conditions(23)
MIN6 cellsN/TIncreased ATF4 and CHOP expression with or without tunicamycin(88)
EndoC-βH2 cellsIncreased LD levels• Prevented erucic acid induced GSIS reduction
• Increased expression of positive effectors of insulin secretion
(32)
PLIN5MIN6 cells mouse islet• Increased TG accumulation
• Increased lipolysis
Increased augmentation of GSIS by cAMP and PA in vitro and by GLP-1 receptor agonist in vivo(24)
INS-1 cells• Increased LD levels
• Reduced ATGL expression and increased CPT1a expression
• Prevented chronic PA-induced GSIS reduction
• Reduced PA induced markers of ER stress and oxidative stress
(81, 82)
Overexpression in miceaN/TImproved glucose tolerance under HFD(81)
Downregulation
PLIN1RIN-m5F cells, INS-1 cellsReduced LD levelsNo protection against apoptosis by chronic FA(87)
PLIN2MIN6 cellsReduced TG accumulation and lipolysisReduced acute augmentation of GSIS by PA(23)
RIN-m5F cells, INS-1 cellsReduced LD levelsNo protection against apoptosis by chronic FA(87)
MIN6 cells, whole-body PLIN2 knockout miceN/T• Protection against ER stress caused by PA, tunicamycin, and INS mutation in Akita mice
• Upregulated autophagy
(88)
INS1 cells, β-cell–specific PLIN2 knockout mice, human pseudoislets• Reduced FAO
• Increased medium-chain acylcarnitine levels
• Increased influx of fluorescent OA analogue to mitochondria
• Impaired GSIS
• Reduced OXPHOS proteins
(85)
EndoC-βH2 cellsReduced LD levels• Impaired GSIS
• Increased ER stress markers
• ER stress inhibitor TUDCA partially restores GSIS
• Reduced expression of β-cell–specific genes and positive regulators of insulin secretion
(32)
PLIN5INS-1 cellsN/TIncreased ER stress markers. This change can be prevented by CPT1a overexpression(81)

Abbreviations: ATF4, activating transcription factor 4; ATGL, adipose triglyceride lipase; cAMP, 3′,5′-cyclic adenosine 5′-monophosphate; CHOP, C/EBP homologous protein; CPT1a, carnitine palmitoyltransferase 1a; ER, endoplasmic reticulum; FAO, fatty acid oxidation; FFA, free fatty acid; GLP-1, glucagon-like peptide 1; GSIS, glucose-stimulated insulin secretion; HFD, high-fat diet; INS, insulin; LD, lipid droplet; N/T, not tested; OA, oleic acid; OXPHOS, oxidative phosphorylation; PA, palmitic acid; PKA, protein kinase A; PLIN, perilipin; MUFA, monounsaturated fatty acid; TGs, triglycerides; TUDCA, tauroursodeoxycholic acid.

aExpression of PLIN5 enriched in mouse islets but PLIN5 expression was also increased in other tissues.

Table 2.

Studies of perilipin overexpression/downregulation in β-cell models

Models Changes in lipid metabolism Changes in β-cell health and function References
Overexpression
PLIN1INS1 cell• Reduced lipolysis
• No change in FAO
GSIS reduced under regular culture condition but conferred protection against chronic PA-induced GSIS reduction(84)
PLIN2MIN6 cells mouse islet• Increased TG accumulation
• No change in lipolysis or FAO
No effect on GSIS under regular culture conditions(23)
MIN6 cellsN/TIncreased ATF4 and CHOP expression with or without tunicamycin(88)
EndoC-βH2 cellsIncreased LD levels• Prevented erucic acid induced GSIS reduction
• Increased expression of positive effectors of insulin secretion
(32)
PLIN5MIN6 cells mouse islet• Increased TG accumulation
• Increased lipolysis
Increased augmentation of GSIS by cAMP and PA in vitro and by GLP-1 receptor agonist in vivo(24)
INS-1 cells• Increased LD levels
• Reduced ATGL expression and increased CPT1a expression
• Prevented chronic PA-induced GSIS reduction
• Reduced PA induced markers of ER stress and oxidative stress
(81, 82)
Overexpression in miceaN/TImproved glucose tolerance under HFD(81)
Downregulation
PLIN1RIN-m5F cells, INS-1 cellsReduced LD levelsNo protection against apoptosis by chronic FA(87)
PLIN2MIN6 cellsReduced TG accumulation and lipolysisReduced acute augmentation of GSIS by PA(23)
RIN-m5F cells, INS-1 cellsReduced LD levelsNo protection against apoptosis by chronic FA(87)
MIN6 cells, whole-body PLIN2 knockout miceN/T• Protection against ER stress caused by PA, tunicamycin, and INS mutation in Akita mice
• Upregulated autophagy
(88)
INS1 cells, β-cell–specific PLIN2 knockout mice, human pseudoislets• Reduced FAO
• Increased medium-chain acylcarnitine levels
• Increased influx of fluorescent OA analogue to mitochondria
• Impaired GSIS
• Reduced OXPHOS proteins
(85)
EndoC-βH2 cellsReduced LD levels• Impaired GSIS
• Increased ER stress markers
• ER stress inhibitor TUDCA partially restores GSIS
• Reduced expression of β-cell–specific genes and positive regulators of insulin secretion
(32)
PLIN5INS-1 cellsN/TIncreased ER stress markers. This change can be prevented by CPT1a overexpression(81)
Models Changes in lipid metabolism Changes in β-cell health and function References
Overexpression
PLIN1INS1 cell• Reduced lipolysis
• No change in FAO
GSIS reduced under regular culture condition but conferred protection against chronic PA-induced GSIS reduction(84)
PLIN2MIN6 cells mouse islet• Increased TG accumulation
• No change in lipolysis or FAO
No effect on GSIS under regular culture conditions(23)
MIN6 cellsN/TIncreased ATF4 and CHOP expression with or without tunicamycin(88)
EndoC-βH2 cellsIncreased LD levels• Prevented erucic acid induced GSIS reduction
• Increased expression of positive effectors of insulin secretion
(32)
PLIN5MIN6 cells mouse islet• Increased TG accumulation
• Increased lipolysis
Increased augmentation of GSIS by cAMP and PA in vitro and by GLP-1 receptor agonist in vivo(24)
INS-1 cells• Increased LD levels
• Reduced ATGL expression and increased CPT1a expression
• Prevented chronic PA-induced GSIS reduction
• Reduced PA induced markers of ER stress and oxidative stress
(81, 82)
Overexpression in miceaN/TImproved glucose tolerance under HFD(81)
Downregulation
PLIN1RIN-m5F cells, INS-1 cellsReduced LD levelsNo protection against apoptosis by chronic FA(87)
PLIN2MIN6 cellsReduced TG accumulation and lipolysisReduced acute augmentation of GSIS by PA(23)
RIN-m5F cells, INS-1 cellsReduced LD levelsNo protection against apoptosis by chronic FA(87)
MIN6 cells, whole-body PLIN2 knockout miceN/T• Protection against ER stress caused by PA, tunicamycin, and INS mutation in Akita mice
• Upregulated autophagy
(88)
INS1 cells, β-cell–specific PLIN2 knockout mice, human pseudoislets• Reduced FAO
• Increased medium-chain acylcarnitine levels
• Increased influx of fluorescent OA analogue to mitochondria
• Impaired GSIS
• Reduced OXPHOS proteins
(85)
EndoC-βH2 cellsReduced LD levels• Impaired GSIS
• Increased ER stress markers
• ER stress inhibitor TUDCA partially restores GSIS
• Reduced expression of β-cell–specific genes and positive regulators of insulin secretion
(32)
PLIN5INS-1 cellsN/TIncreased ER stress markers. This change can be prevented by CPT1a overexpression(81)

Abbreviations: ATF4, activating transcription factor 4; ATGL, adipose triglyceride lipase; cAMP, 3′,5′-cyclic adenosine 5′-monophosphate; CHOP, C/EBP homologous protein; CPT1a, carnitine palmitoyltransferase 1a; ER, endoplasmic reticulum; FAO, fatty acid oxidation; FFA, free fatty acid; GLP-1, glucagon-like peptide 1; GSIS, glucose-stimulated insulin secretion; HFD, high-fat diet; INS, insulin; LD, lipid droplet; N/T, not tested; OA, oleic acid; OXPHOS, oxidative phosphorylation; PA, palmitic acid; PKA, protein kinase A; PLIN, perilipin; MUFA, monounsaturated fatty acid; TGs, triglycerides; TUDCA, tauroursodeoxycholic acid.

aExpression of PLIN5 enriched in mouse islets but PLIN5 expression was also increased in other tissues.

Perilipins Control β-Cell Lipid Droplet Levels

Perilipin 5 Augments Insulin Secretion and Protects β Cells Under Stress

PLIN5 is highly expressed in oxidative tissues and possesses features that promote mitochondrial activity and FAO (16). Protein kinase A (PKA)-dependent phosphorylation of PLIN5 increases LD lipolysis (16, 75). PLIN5 also promotes the association of LDs to the mitochondria and facilitates the transfer of FFAs released from lipolysis to the mitochondria (76, 77). PLIN5 can bind FAs and translocate to the nuclei to allosterically activate peroxisome proliferator-activated receptor gamma coactivator 1-a (PGC1a) (78). While PLIN5 is expressed at moderate levels both in human and mouse islets, levels in human islets are higher than in mouse islets (24).

Currently, functional information about PLIN5 in human β cells is very limited (7). However, PLIN5 actions have been investigated in overexpression experiments in rodent β-cell models (24). Trevino and colleagues (24) found that the lipolysis rate was higher on PLIN5 overexpression than PLIN2 overexpression in MIN6 cells when TG synthesis was increased to a similar extent by overexpression of PLIN2 and PLIN5. The role of PLIN5 in promoting lipolysis was further confirmed, as a cell-permeable 3′,5′-cyclic adenosine 5′-monophosphate (cAMP) analogue bromoadenosine (8Br-cAMP) increased lipolysis in PLIN5 overexpressing MIN6 cells. The change in lipolysis correlated with insulin secretion in MIN6 cells and mouse islets overexpressing PLIN2 or PLIN5. GSIS under acute PA and 8Br-cAMP exposure was increased in β-cell models overexpressing PLIN5, but not in those overexpressing PLIN2. Furthermore, GSIS in vivo was augmented by the glucagon-like peptide-1 (GLP-1) receptor agonist in mice with β-cell–specific PLIN5 overexpression. A physiological role of islet PLIN5 in vivo was implicated from the induction of islet PLIN5 expression by fasting in mice. Adipocytes release FFAs during fasting that are redistributed to the heart, liver, and other tissues where FFAs are stored in LDs (79, 80). In the study by Trevino et al (24), fasting induced a marked increase of PLIN5 expression and TG accumulation in mouse islets, implicating that PLIN5 upregulation during fasting allows β cells to store TGs in PLIN5-coated LDs that can release lipolytic metabolites on refeeding to facilitate insulin secretion.

Overexpression of PLIN5 is also reported to improve GSIS in INS-1 cells chronically exposed to PA as well as in HFD-fed mice (81, 82). Moreover, PLIN5 overexpression was found to prevent PA-induced apoptosis and ER stress in INS-1 cells (81). Improvement in viability and reduction in apoptosis after chronic exposure to PA in PLIN5 overexpressed INS-1 cells was shown to be associated with activation of the nuclear erythroid 2-related factor (Nrf2)-antioxidant response element pathway, which defends cells against oxidative stress (82). Although data in human β cells are lacking, it appears that PLIN5 stimulates GSIS and protects β cells from nutritional stress, at least in rodent models. It is necessary to remember that studies performed in mice did not specify if visible LDs are formed in β cells overexpressing PLIN5 (24, 81).

Currently, little is known about PLIN1, PLIN3, or PLIN4 function in β cells. According to human islet single-cell RNA sequencing data, PLIN1 and PLIN4 are expressed at very low levels relative to PLIN2, PLIN3, or PLIN5 (83). Overexpressing PLIN1 in INS-1 cells has been shown to reduce GSIS under regular culture conditions but prevent chronic PA-induced GSIS reduction (84). This could be another example of an increase in LD formation mediated by PLIN overexpression being protective to β cells.

Perilipin 2 Is Crucial to the Health and Activity of Pancreatic β Cells

PLIN2 is the most abundant PLIN in nonadipocytes, including pancreatic β cells (see Table 1) and promotes LD pool expansion by preventing lipolysis and lipophagy (16). PLIN2 expression levels closely correlate with cellular LDs and neutral lipid content, while its downregulation is sufficient to reduce LD accumulation (7, 32). PLIN2 protein levels show the most robust increase in response to FFA load relative to other PLIN family members in human islet cells (23, 24). Both PLIN2 and PLIN3 expression markedly increase in human EndoC-βH2 cells on cessation of cell proliferation and induction of glucose-responsive insulin secretion, indicating that the proliferation and maturation status of the β cell may regulate PLIN gene/protein expression (32).

PLIN2 appears to be crucial for insulin secretion and the prevention of β-cell stress. Mishra et al (85) studied the effect of PLIN2 downregulation on INS1 cells, human pseudoislets, and β-cell–specific PLIN2 knockout mice. Downregulating PLIN2 in INS-1 cells impaired GSIS under regular culture conditions and after chronic incubation with PA. In addition, reduction of PLIN2 levels impaired GSIS in human pseudoislets. In β-cell–specific PLIN2 knockout mice placed on an HFD, GSIS was compromised in vivo and in isolated islets, in which islet TG content was reduced. All 3 models of PLIN2-deficient β cells/islets showed reduced levels of the oxidative phosphorylation (OXPHOS) complex components and impaired mitochondrial integrity. This included reduction in oxygen consumption rate, an alteration in the acylcarnitine profile, decreased FAO levels, and increased mitochondrial fragmentation in PLIN2-deficient INS-1 cells. Metabolic labeling with BODIPY C12 showed that PLIN2 deficiency increased trafficking of FFAs into the mitochondria, possibly contributing to mitochondrial dysfunction and impaired insulin secretion. However, ER stress markers were not altered in these 3 models, nor were islet-enriched PDX1, MAFA, and NKX6-1 transcription factor levels in PLIN2-deficient human pseudoislets.

More recently, the EndoC-βH2 cell line was used to study how LD regulation might influence human β cells (32). In nonproliferative EndoC-βH2-Cre cells (86), PLIN2 knockdown reduced LD levels and impaired GSIS, whereas overexpression of PLIN2 increased LD levels and improved GSIS. RNA sequencing revealed that more genes were differentially expressed after PLIN2 downregulation (1479 genes increased and 493 genes reduced) than overexpression (237 genes increased and 48 genes reduced), which included activated levels of ER stressors (inositol requiring enzyme 1 A [IRE1 A] and sXbp1), non–β-cell hormones (somatostatin and glucagon), and progenitor markers (SOX9, FEV) associated with T2D islets. In contrast, overexpression of PLIN2 increased GSIS and effectors of insulin secretion, including exocytosis complex component 5 (EXOC5), potassium inwardly rectifying channel, subfamily J, member 11 (KCNJ11), STX1a, and sarcoplasmic reticulum calcium-ATPase 2b (SERCA2b). Treatment with tauroursodeoxycholic acid (TUDCA), an ER stress inhibitor, restored GSIS on PLIN2 deficiency in EndoC-βH2-Cre cells, while PLIN2 overexpression protected against erucic acid-induced toxicity. Mitochondrial gene expression was also compromised on downregulation of PLIN2 in EndoC-βH2-Cre cells, in agreement with the reduction of the OXPHOS complex reported in PLIN2-deficient human pseudoislets (32, 85). However, it is presently unclear why β-cell identity regulators and ER stressors are altered by PLIN2 downregulation in a human β-cell line context but not in human pseudoislets (32, 85). These moderate discrepancies could be due to differences in cellular context (eg, cell line vs primary islet cells), the collective effect from other LD-containing islet cell types (eg, α cells) on global deletion of PLIN2 in pseudoislets, and/or the difference in the duration of PLIN2 downregulation.

Although the aforementioned studies indicate that proper maintenance of PLIN2-coated LDs in human β cells supports cell health and function, results from some rodent β-cell models suggest that downregulation of PLIN2 has either neutral or protective effects against stress (see Table 2). One study reported that downregulation of PLIN1 or PLIN2 did not affect the viability of rat RIN-mM5F or INS-1 cells cultured with PA and OA (87). Another demonstrated that whole-body PLIN2 knockout prevented ER stress, preserved β-cell mass, and was protective against hyperglycemia in mice carrying the Akita insulin mutation, which causes severe ER stress and apoptosis in β cells (88). In the same study, ER stress induced by tunicamycin or PA was reduced in PLIN2-deficient mouse MIN6 cells and mouse islets. While it is presently difficult to explain the different results obtained on compromising PLIN2 levels in rodent models, all existing data strongly suggest that LD accumulation controls human β-cell function and integrity under both normal and pathological circumstances.

Conclusions and Future Directions

Our general understanding of the effect of LDs on multiple aspects of cellular activity has increased considerably in recent years. The importance of β-cell LDs has attracted little attention until recent studies reported their presence in human β cells and enrichment in T2D islets. Studies of ATGL and PLIN2 in human islet β-cell models strongly indicate that the formation and degradation of LDs are equally critical for β cells in maintaining normal insulin secretion, ER homeostasis, and mitochondrial integrity.

A number of critical questions regarding the regulation of α- and β-cell function by LDs remain. Why are LDs not visible in mouse islets even when PLIN protein levels and TG contents increase in response to lipid load? How does age regulate LD accumulation in human islets? How do LDs regulate adult human islet α- and β-cell activity? Which LD-associated proteins regulate the functions of LDs in human islet cells, and how? Does LD accumulation influence the functional and molecular heterogeneity of the human islet α- and β-cell populations? Why are LDs enriched in T2D islets; do they protect or accelerate β-cell demise?

We propose examining the effect of LDs on human islet cell health using models translatable to human pathophysiology. Embryonically derived islet cells and human pseudoislets represent relevant and tractable model systems, especially when combined with xenotransplantation, for investigating the effect of LDs on human islet cells in vivo under normal and HFD stress conditions.

Abbreviations

    Abbreviations
     
  • ATGL

    adipose triglyceride lipase

  •  
  • cAMP

    3′,5′-cyclic adenosine 5′-monophosphate

  •  
  • DGAT1

    diacylglycerol acyltransferase 1

  •  
  • EM

    electron microscopy

  •  
  • ER

    endoplasmic reticulum

  •  
  • FAO

    fatty acid oxidation

  •  
  • FFA

    free fatty acid

  •  
  • GL

    glycerolipid

  •  
  • GLP-1

    glucagon-like peptide 1

  •  
  • GSIS

    glucose-stimulated insulin secretion

  •  
  • HFD

    high-fat diet

  •  
  • INS

    insulin

  •  
  • LCFA

    long-chain fatty acid

  •  
  • LD

    lipid droplet

  •  
  • MUFA

    monounsaturated fatty acid

  •  
  • Nrf2

    nuclear erythroid 2-related factor

  •  
  • OA

    oleic acid

  •  
  • OXPHOS

    oxidative phosphorylation

  •  
  • PA

    palmitic acid

  •  
  • PKA

    protein kinase A

  •  
  • PLIN

    perilipin

  •  
  • PUFA

    polyunsaturated fatty acid

  •  
  • SCD

    stearoyl-CoA desaturase

  •  
  • STX1a, syntaxin 1a; T2D

    type 2 diabetes

  •  
  • TGs

    triglycerides

  •  
  • TUDCA

    tauroursodeoxycholic acid

Acknowledgments

Human pancreatic islets provided by the National Institute of Diabetes and Digestive and Kidney Diseases–funded Integrated Islet Distribution Program (IIDP) at City of Hope (No. 2UC4DK098085) were used for this review. Fig. 1A and 3 were created with BioRender.com. Materials from (24) have been used for Fig. 1C and from (22) for Fig. 1D with the permission of the American Diabetes Association, which holds the copyright of these materials and all rights reserved.

Financial Support

This work was supported by the National Institutes of Health (grant Nos. R01-DK090490 to Y.I. and RO1 DK050203, RO1 DK090570, RO1 DK126482, RO1 DK065949 to R.S.), the Department of Veterans Affairs (grant No. I01 BX005107), the Fraternal Orders of Eagles Diabetes Research Center at the University of Iowa, the Mark Collie endowment for diabetes research, and a JDRF (No. 3-PDF-2019-738-A-N to X.T.).

Disclosures

The authors have nothing to disclose.

Data Availability

Some or all data sets generated during and/or analyzed during the present study are not publicly available but are available from the corresponding author on reasonable request.

References

1.

Prentki
M
,
Nolan
CJ
.
Islet β cell failure in type 2 diabetes
.
J Clin Invest.
2006
;
116
(
7
):
1802
-
1812
.

2.

Ferrannini
E
,
Gastaldelli
A
,
Miyazaki
Y
,
Matsuda
M
,
Mari
A
,
DeFronzo
RA
.
β-Cell function in subjects spanning the range from normal glucose tolerance to overt diabetes: a new analysis
.
J Clin Endocrinol Metab.
2005
;
90
(
1
):
493
-
500
.

3.

Del Guerra
S
,
Lupi
R
,
Marselli
L
, et al. 
Functional and molecular defects of pancreatic islets in human type 2 diabetes
.
Diabetes.
2005
;
54
(
3
):
727
-
735
.

4.

Hunter
CS
,
Stein
RW
.
Evidence for loss in identity, de-differentiation, and trans-differentiation of islet β-cells in type 2 diabetes
.
Front Genet.
2017
;
8
:
35
.

5.

Thurmond
DC
,
Gaisano
HY
.
Recent insights into beta-cell exocytosis in type 2 diabetes
.
J Mol Biol.
2020
;
432
(
5
):
1310
-
1325
.

6.

Spijker
HS
,
Song
H
,
Ellenbroek
JH
, et al. 
Loss of β-cell identity occurs in type 2 diabetes and is associated with islet amyloid deposits
.
Diabetes.
2015
;
64
(
8
):
2928
-
2938
.

7.

Imai
Y
,
Cousins
RS
,
Liu
S
,
Phelps
BM
,
Promes
JA
.
Connecting pancreatic islet lipid metabolism with insulin secretion and the development of type 2 diabetes
.
Ann N Y Acad Sci.
2020
;
1461
(
1
):
53
-
72
.

8.

Wigger
L
,
Barovic
M
,
Brunner
AD
, et al. 
Multi-omics profiling of living human pancreatic islet donors reveals heterogeneous beta cell trajectories towards type 2 diabetes
.
Nat Metab.
2021
;
3
(
7
):
1017
-
1031
.

9.

Prentki
M
,
Corkey
BE
,
Madiraju
SRM
.
Lipid-associated metabolic signalling networks in pancreatic beta cell function
.
Diabetologia.
2020
;
63
(
1
):
10
-
20
.

10.

Coleman
RA
.
The “discovery” of lipid droplets: a brief history of organelles hidden in plain sight
.
Biochim Biophys Acta Mol Cell Biol Lipids.
2020
;
1865
(
9
):
158762
.

11.

Welte
MA
,
Gould
AP
.
Lipid droplet functions beyond energy storage
.
Biochim Biophys Acta Mol Cell Biol Lipids
.
2017
;
1862
(
10 Pt B
):
1260
-
1272
.

12.

Gluchowski
NL
,
Becuwe
M
,
Walther
TC
,
Farese
RV
Jr
.
Lipid droplets and liver disease: from basic biology to clinical implications
.
Nat Rev Gastroenterol Hepatol.
2017
;
14
(
6
):
343
-
355
.

13.

Gemmink
A
,
Goodpaster
BH
,
Schrauwen
P
,
Hesselink
MKC
.
Intramyocellular lipid droplets and insulin sensitivity, the human perspective
.
Biochim Biophys Acta Mol Cell Biol Lipids.
2017
;
1862
(
10 Pt B
):
1242
-
1249
.

14.

Bosch
M
,
Parton
RG
,
Pol
A
.
Lipid droplets, bioenergetic fluxes, and metabolic flexibility
.
Semin Cell Dev Biol.
2020
;
108
:
33
-
46
.

15.

Jarc
E
,
Petan
T
.
Lipid droplets and the management of cellular stress
.
Yale J Biol Med.
2019
;
92
(
3
):
435
-
452
.

16.

Sztalryd
C
,
Brasaemle
DL
.
The perilipin family of lipid droplet proteins: gatekeepers of intracellular lipolysis
.
Biochim Biophys Acta Mol Cell Biol Lipids.
2017
;
1862
(
10 Pt B
):
1221
-
1232
.

17.

Bersuker
K
,
Peterson
CWH
,
To
M
, et al. 
A proximity labeling strategy provides insights into the composition and dynamics of lipid droplet proteomes
.
Dev Cell.
2018
;
44
(
1
):
97
-
112.e7
.

18.

Shimabukuro
M
,
Zhou
YT
,
Levi
M
,
Unger
RH
.
Fatty acid-induced beta cell apoptosis: a link between obesity and diabetes
.
Proc Natl Acad Sci U S A.
1998
;
95
(
5
):
2498
-
2502
.

19.

Unger
RH
,
Zhou
YT
,
Orci
L
.
Regulation of fatty acid homeostasis in cells: novel role of leptin
.
Proc Natl Acad Sci U S A.
1999
;
96
(
5
):
2327
-
2332
.

20.

Dai
C
,
Kayton
NS
,
Shostak
A
, et al. 
Stress-impaired transcription factor expression and insulin secretion in transplanted human islets
.
J Clin Invest.
2016
;
126
(
5
):
1857
-
1870
.

21.

Liu
S
,
Promes
JA
,
Harata
M
, et al. 
Adipose triglyceride lipase is a key lipase for the mobilization of lipid droplets in human β-cells and critical for the maintenance of syntaxin 1a levels in β-cells
.
Diabetes.
2020
;
69
(
6
):
1178
-
1192
.

22.

Tong
X
,
Dai
C
,
Walker
JT
, et al. 
Lipid droplet accumulation in human pancreatic islets is dependent on both donor age and health
.
Diabetes.
2020
;
69
(
3
):
342
-
354
.

23.

Faleck
DM
,
Ali
K
,
Roat
R
, et al. 
Adipose differentiation-related protein regulates lipids and insulin in pancreatic islets
.
Am J Physiol Endocrinol Metab.
2010
;
299
(
2
):
E249
-
E257
.

24.

Trevino
MB
,
Machida
Y
,
Hallinger
DR
, et al. 
Perilipin 5 regulates islet lipid metabolism and insulin secretion in a cAMP-dependent manner: implication of its role in the postprandial insulin secretion
.
Diabetes.
2015
;
64
(
4
):
1299
-
1310
.

25.

Hsieh
K
,
Lee
YK
,
Londos
C
,
Raaka
BM
,
Dalen
KT
,
Kimmel
AR
.
Perilipin family members preferentially sequester to either triacylglycerol-specific or cholesteryl-ester-specific intracellular lipid storage droplets
.
J Cell Sci.
2012
;
125
(
Pt 17
):
4067
-
4076
.

26.

Vernier
S
,
Chiu
A
,
Schober
J
, et al. 
β-Cell metabolic alterations under chronic nutrient overload in rat and human islets
.
Islets
2012
;
4
(
6
):
379
-
392
.

27.

Quinlivan
VH
,
Wilson
MH
,
Ruzicka
J
,
Farber
SA
.
An HPLC-CAD/fluorescence lipidomics platform using fluorescent fatty acids as metabolic tracers
.
J Lipid Res.
2017
;
58
(
5
):
1008
-
1020
.

28.

Guo
Y
,
Cordes
KR
,
Farese
RV
Jr
,
Walther
TC
.
Lipid droplets at a glance
.
J Cell Sci.
2009
;
122
(
Pt 6
):
749
-
752
.

29.

Suzuki
M
,
Shinohara
Y
,
Ohsaki
Y
,
Fujimoto
T
.
Lipid droplets: size matters
.
J Electron Microsc.
2011
;
60
(
Suppl 1
):
S101
-
S116
.

30.

Stenkula
KG
,
Erlanson-Albertsson
C
.
Adipose cell size: importance in health and disease
.
Am J Physiol Regul Integr Comp Physiol.
2018
;
315
(
2
):
R284
-
R295
.

31.

Schott
MB
,
Weller
SG
,
Schulze
RJ
, et al. 
Lipid droplet size directs lipolysis and lipophagy catabolism in hepatocytes
.
J Cell Biol.
2019
;
218
(
10
):
3320
-
3335
.

32.

Tong
X
,
Stein
R
.
Lipid droplets protect human β-cells from lipotoxicity-induced stress and cell identity changes
.
Diabetes.
2021
;
70
(
11
):
2595
-
2607
.

33.

Westermark
GT
,
Krogvold
L
,
Dahl-Jørgensen
K
,
Ludvigsson
J
.
Islet amyloid in recent-onset type 1 diabetes—the DiViD study
.
Ups J Med Sci.
2017
;
122
(
3
):
201
-
203
.

34.

Ji
J
,
Petropavlovskaia
M
,
Khatchadourian
A
, et al. 
Type 2 diabetes is associated with suppression of autophagy and lipid accumulation in β-cells
.
J Cell Mol Med.
2019
;
23
(
4
):
2890
-
2900
.

35.

Nair
GG
,
Liu
JS
,
Russ
HA
, et al. 
Recapitulating endocrine cell clustering in culture promotes maturation of human stem-cell-derived β cells
.
Nat Cell Biol.
2019
;
21
(
2
):
263
-
274
.

36.

Cnop
M
,
Grupping
A
,
Hoorens
A
,
Bouwens
L
,
Pipeleers-Marichal
M
,
Pipeleers
D
.
Endocytosis of low-density lipoprotein by human pancreatic beta cells and uptake in lipid-storing vesicles, which increase with age
.
Am J Pathol.
2000
;
156
(
1
):
237
-
244
.

37.

Cnop
M
,
Hughes
SJ
,
Igoillo-Esteve
M
, et al. 
The long lifespan and low turnover of human islet beta cells estimated by mathematical modelling of lipofuscin accumulation
.
Diabetologia.
2010
;
53
(
2
):
321
-
330
.

38.

Schmucker
DL
,
Sachs
H
.
Quantifying dense bodies and lipofuscin during aging: a morphologist’s perspective
.
Arch Gerontol Geriatr.
2002
;
34
(
3
):
249
-
261
.

39.

Zechner
R
,
Madeo
F
,
Kratky
D
.
Cytosolic lipolysis and lipophagy: two sides of the same coin
.
Nat Rev Mol Cell Biol.
2017
;
18
(
11
):
671
-
684
.

40.

Prentki
M
,
Matschinsky
FM
,
Madiraju
SR
.
Metabolic signaling in fuel-induced insulin secretion
.
Cell Metab.
2013
;
18
(
2
):
162
-
185
.

41.

Nettebrock
NT
,
Bohnert
M
.
Born this way—biogenesis of lipid droplets from specialized ER subdomains
.
Biochim Biophys Acta Mol Cell Biol Lipids.
2020
;
1865
(
1
):
158448
.

42.

Zoni
V
,
Khaddaj
R
,
Lukmantara
I
, et al. 
Seipin accumulates and traps diacylglycerols and triglycerides in its ring-like structure
.
Proc Natl Acad Sci U S A.
2021
;
118
(
10
):
e2017205118
.

43.

Bosch
M
,
Sweet
MJ
,
Parton
RG
,
Pol
A
.
Lipid droplets and the host-pathogen dynamic: FATal attraction?
J Cell Biol.
2021
;
220
(
8
):
e202104005
.

44.

Sugawara
T
,
Kano
F
,
Murata
M
.
Rab2A is a pivotal switch protein that promotes either secretion or ER-associated degradation of (pro)insulin in insulin-secreting cells
.
Sci Rep.
2014
;
4
:
6952
.

45.

Fang
J
,
Liu
M
,
Zhang
X
, et al. 
COPII-dependent ER export: a critical component of insulin biogenesis and β-cell ER homeostasis
.
Mol Endocrinol.
2015
;
29
(
8
):
1156
-
1169
.

46.

Omar-Hmeadi
M
,
Idevall-Hagren
O
.
Insulin granule biogenesis and exocytosis
.
Cell Mol Life Sci.
2021
;
78
(
5
):
1957
-
1970
.

47.

Tang
T
,
Abbott
MJ
,
Ahmadian
M
,
Lopes
AB
,
Wang
Y
,
Sul
HS
.
Desnutrin/ATGL activates PPARδ to promote mitochondrial function for insulin secretion in islet β cells
.
Cell Metab.
2013
;
18
(
6
):
883
-
895
.

48.

Zhao
S
,
Mugabo
Y
,
Iglesias
J
, et al.  α
/β-Hydrolase domain-6-accessible monoacylglycerol controls glucose-stimulated insulin secretion
.
Cell Metab.
2014
;
19
(
6
):
993
-
1007
.

49.

Lanyon-Hogg
T
,
Faronato
M
,
Serwa
RA
,
Tate
EW
.
Dynamic protein acylation: new substrates, mechanisms, and drug targets
.
Trends Biochem Sci.
2017
;
42
(
7
):
566
-
581
.

50.

Kulminskaya
N
,
Oberer
M
.
Protein-protein interactions regulate the activity of adipose triglyceride lipase in intracellular lipolysis
.
Biochimie.
2020
;
169
:
62
-
68
.

51.

Oh
YS
,
Bae
GD
,
Baek
DJ
,
Park
EY
,
Jun
HS
.
Fatty acid-induced lipotoxicity in pancreatic beta-cells during development of type 2 diabetes
.
Front Endocrinol (Lausanne).
2018
;
9
:
384
.

52.

Carta
G
,
Murru
E
,
Banni
S
,
Manca
C
.
Palmitic acid: physiological role, metabolism and nutritional implications
.
Front Physiol.
2017
;
8
:
902
.

53.

Feng
R
,
Luo
C
,
Li
C
, et al. 
Free fatty acids profile among lean, overweight and obese non-alcoholic fatty liver disease patients: a case-control study
.
Lipids Health Dis.
2017
;
16
(
1
):
165
.

54.

Sobczak
AIS
,
Blindauer
CA
,
Stewart
AJ
.
Changes in plasma free fatty acids associated with type-2 diabetes
.
Nutrients.
2019
;
11
(
9
):
2022
.

55.

Staaf
J
,
Ubhayasekera
SJKA
,
Sargsyan
E
, et al. 
Initial hyperinsulinemia and subsequent β-cell dysfunction is associated with elevated palmitate levels
.
Pediatr Res.
2016
;
80
(
2
):
267
-
274
.

56.

El-Assaad
W
,
Buteau
J
,
Peyot
ML
, et al. 
Saturated fatty acids synergize with elevated glucose to cause pancreatic beta-cell death
.
Endocrinology.
2003
;
144
(
9
):
4154
-
4163
.

57.

Cnop
M
,
Hannaert
JC
,
Hoorens
A
,
Eizirik
DL
,
Pipeleers
DG
.
Inverse relationship between cytotoxicity of free fatty acids in pancreatic islet cells and cellular triglyceride accumulation
.
Diabetes.
2001
;
50
(
8
):
1771
-
1777
.

58.

Nemecz
M
,
Constantin
A
,
Dumitrescu
M
, et al. 
The distinct effects of palmitic and oleic acid on pancreatic beta cell function: the elucidation of associated mechanisms and effector molecules
.
Front Pharmacol.
2018
;
9
:
1554
.

59.

Busch
AK
,
Gurisik
E
,
Cordery
DV
, et al. 
Increased fatty acid desaturation and enhanced expression of stearoyl coenzyme A desaturase protects pancreatic beta-cells from lipoapoptosis
.
Diabetes.
2005
;
54
(
10
):
2917
-
2924
.

60.

Thörn
K
,
Hovsepyan
M
,
Bergsten
P
.
Reduced levels of SCD1 accentuate palmitate-induced stress in insulin-producing β-cells
.
Lipids Health Dis.
2010
;
9
:
108
.

61.

Green
CD
,
Olson
LK
.
Modulation of palmitate-induced endoplasmic reticulum stress and apoptosis in pancreatic β-cells by stearoyl-CoA desaturase and Elovl6
.
Am J Physiol Endocrinol Metab.
2011
;
300
(
4
):
E640
-
E649
.

62.

Krizhanovskii
C
,
Kristinsson
H
,
Elksnis
A
, et al. 
EndoC-βH1 cells display increased sensitivity to sodium palmitate when cultured in DMEM/F12 medium
.
Islets.
2017
;
9
(
3
):
e1296995
.

63.

Thomas
P
,
Leslie
KA
,
Welters
HJ
,
Morgan
NG
.
Long-chain saturated fatty acid species are not toxic to human pancreatic β-cells and may offer protection against pro-inflammatory cytokine induced β-cell death
.
Nutr Metab (Lond).
2021
;
18
(
1
):
9
.

64.

Oshima
M
,
Pechberty
S
,
Bellini
L
, et al. 
Stearoyl CoA desaturase is a gatekeeper that protects human beta cells against lipotoxicity and maintains their identity
.
Diabetologia.
2020
;
63
(
2
):
395
-
409
.

65.

Plötz
T
,
von Hanstein
AS
,
Krümmel
B
,
Laporte
A
,
Mehmeti
I
,
Lenzen
S
.
Structure-toxicity relationships of saturated and unsaturated free fatty acids for elucidating the lipotoxic effects in human EndoC-βH1 beta-cells
.
Biochim Biophys Acta Mol Basis Dis.
2019
;
1865
(
11
):
165525
.

66.

Imai
Y
,
Dobrian
AD
,
Morris
MA
,
Nadler
JL
.
Islet inflammation: a unifying target for diabetes treatment?
Trends Endocrinol Metab.
2013
;
24
(
7
):
351
-
360
.

67.

Zhu
M
,
Liu
X
,
Liu
W
,
Lu
Y
,
Cheng
J
,
Chen
Y
.
β Cell aging and age-related diabetes
.
Aging (Albany NY).
2021
;
13
(
5
):
7691
-
7706
.

68.

De Tata
V
.
Age-related impairment of pancreatic beta-cell function: pathophysiological and cellular mechanisms
.
Front Endocrinol (Lausanne).
2014
;
5
:
138
.

69.

Mihailidou
C
,
Chatzistamou
I
,
Papavassiliou
AG
,
Kiaris
H
.
Modulation of pancreatic islets’ function and survival during aging involves the differential regulation of endoplasmic reticulum stress by p21 and CHOP
.
Antioxid Redox Signal.
2017
;
27
(
4
):
185
-
200
.

70.

DeZwaan-McCabe
D
,
Sheldon
RD
,
Gorecki
MC
, et al. 
ER stress inhibits liver fatty acid oxidation while unmitigated stress leads to anorexia-induced lipolysis and both liver and kidney steatosis
.
Cell Rep.
2017
;
19
(
9
):
1794
-
1806
.

71.

Lee
JS
,
Mendez
R
,
Heng
HH
,
Yang
ZQ
,
Zhang
K
.
Pharmacological ER stress promotes hepatic lipogenesis and lipid droplet formation
.
Am J Transl Res.
2012
;
4
(
1
):
102
-
113
.

72.

Geltinger
F
,
Schartel
L
,
Wiederstein
M
, et al. 
Friend or foe: lipid droplets as organelles for protein and lipid storage in cellular stress response, aging and disease
.
Molecules.
2020
;
25
(
21
):
5053
.

73.

Bailey
AP
,
Koster
G
,
Guillermier
C
, et al. 
Antioxidant role for lipid droplets in a stem cell niche of drosophila
.
Cell.
2015
;
163
(
2
):
340
-
353
.

74.

Ralhan
I
,
Chang
CL
,
Lippincott-Schwartz
J
,
Ioannou
MS
.
Lipid droplets in the nervous system
.
J Cell Biol.
2021
;
220
(
7
):
e202102136
.

75.

Gallardo-Montejano
VI
,
Saxena
G
,
Kusminski
CM
, et al. 
Nuclear Perilipin 5 integrates lipid droplet lipolysis with PGC-1α/SIRT1-dependent transcriptional regulation of mitochondrial function
.
Nat Commun.
2016
;
7
:
12723
.

76.

Wang
H
,
Sreenevasan
U
,
Hu
H
, et al. 
Perilipin 5, a lipid droplet-associated protein, provides physical and metabolic linkage to mitochondria
.
J Lipid Res.
2011
;
52
(
12
):
2159
-
2168
.

77.

Kuramoto
K
,
Okamura
T
,
Yamaguchi
T
, 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
(
28
):
23852
-
23863
.

78.

Najt
CP
,
Khan
SA
,
Heden
TD
, et al. 
Lipid droplet-derived monounsaturated fatty acids traffic via PLIN5 to allosterically activate SIRT1
.
Mol Cell.
2020
;
77
(
4
):
810
-
824.e8
.

79.

Kramer
DA
,
Quiroga
AD
,
Lian
J
,
Fahlman
RP
,
Lehner
R
.
Fasting and refeeding induces changes in the mouse hepatic lipid droplet proteome
.
J Proteomics.
2018
;
181
:
213
-
224
.

80.

Jacob
S
.
Lipid droplet accumulation in the heart during fasting
.
Acta Histochem.
1987
;
82
(
2
):
149
-
152
.

81.

Zhu
Y
,
Zhang
X
,
Zhang
L
, et al. 
Perilipin5 protects against lipotoxicity and alleviates endoplasmic reticulum stress in pancreatic β-cells
.
Nutr Metab (Lond)
2019
;
16
:
50
.

82.

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 β-cells
.
Front Endocrinol (Lausanne).
2020
;
11
:
166
.

83.

Benner
C
,
van der Meulen
T
,
Cacéres
E
,
Tigyi
K
,
Donaldson
CJ
,
Huising
MO
.
The transcriptional landscape of mouse beta cells compared to human beta cells reveals notable species differences in long non-coding RNA and protein-coding gene expression
.
BMC Genomics.
2014
;
15
(
1
):
620
.

84.

Borg
J
,
Klint
C
,
Wierup
N
, et al. 
Perilipin is present in islets of Langerhans and protects against lipotoxicity when overexpressed in the beta-cell line INS-1
.
Endocrinology.
2009
;
150
(
7
):
3049
-
3057
.

85.

Mishra
AL
,
Liu
S
,
Promes
J
, et al. 
Perilipin2 downregulation in β cells impairs insulin secretion under nutritional stress and damages mitochondria
.
JCI Insight.
2021
;
6
(
9
):
e144341
.

86.

Scharfmann
R
,
Pechberty
S
,
Hazhouz
Y
, et al. 
Development of a conditionally immortalized human pancreatic β cell line
.
J Clin Invest.
2014
;
124
(
5
):
2087
-
2098
.

87.

Plötz
T
,
Hartmann
M
,
Lenzen
S
,
Elsner
M
.
The role of lipid droplet formation in the protection of unsaturated fatty acids against palmitic acid induced lipotoxicity to rat insulin-producing cells
.
Nutr Metab (Lond).
2016
;
13
:
16
.

88.

Chen
E
,
Tsai
TH
,
Li
L
,
Saha
P
,
Chan
L
,
Chang
BH
.
PLIN2 is a key regulator of the unfolded protein response and endoplasmic reticulum stress resolution in pancreatic β cells
.
Sci Rep.
2017
;
7
:
40855
.

89.

Sztalryd
C
,
Bell
M
,
Lu
X
, et al. 
Functional compensation for adipose differentiation-related protein (ADFP) by Tip47 in an ADFP null embryonic cell line
.
J Biol Chem.
2006
;
281
(
45
):
34341
-
34348
.

90.

Wolins
NE
,
Quaynor
BK
,
Skinner
JR
, et al. 
OXPAT/PAT-1 is a PPAR-induced lipid droplet protein that promotes fatty acid utilization
.
Diabetes.
2006
;
55
(
12
):
3418
-
3428
.

91.

Dalen
KT
,
Dahl
T
,
Holter
E
, et al. 
LSDP5 is a PAT protein specifically expressed in fatty acid oxidizing tissues
.
Biochim Biophys Acta.
2007
;
1771
(
2
):
210
-
227
.

92.

Zhang
E
,
Cui
W
,
Lopresti
M
, et al. 
Hepatic PLIN5 signals via SIRT1 to promote autophagy and prevent inflammation during fasting
.
J Lipid Res.
2020
;
61
(
3
):
338
-
350
.

93.

Itabe
H
,
Yamaguchi
T
,
Nimura
S
,
Sasabe
N
.
Perilipins: a diversity of intracellular lipid droplet proteins
.
Lipids Health Dis.
2017
;
16
(
1
):
83
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)