Next Article in Journal
Targeting Interactions between Fibroblasts and Macrophages to Treat Cardiac Fibrosis
Previous Article in Journal
Neuroreceptor Inhibition by Clozapine Triggers Mitohormesis and Metabolic Reprogramming in Human Blood Cells
Previous Article in Special Issue
Bone Marrow Adipose Tissue
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Impaired Remodeling of White Adipose Tissue in Obesity and Aging: From Defective Adipogenesis to Adipose Organ Dysfunction

by
Carla Iacobini
,
Martina Vitale
,
Jonida Haxhi
,
Stefano Menini
and
Giuseppe Pugliese
*,‡
Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work.
Cells 2024, 13(9), 763; https://doi.org/10.3390/cells13090763
Submission received: 2 April 2024 / Revised: 22 April 2024 / Accepted: 25 April 2024 / Published: 30 April 2024
(This article belongs to the Special Issue Molecular Mechanisms of Adipose Organ Remodelling)

Abstract

:
The adipose organ adapts and responds to internal and environmental stimuli by remodeling both its cellular and extracellular components. Under conditions of energy surplus, the subcutaneous white adipose tissue (WAT) is capable of expanding through the enlargement of existing adipocytes (hypertrophy), followed by de novo adipogenesis (hyperplasia), which is impaired in hypertrophic obesity. However, an impaired hyperplastic response may result from various defects in adipogenesis, leading to different WAT features and metabolic consequences, as discussed here by reviewing the results of the studies in animal models with either overexpression or knockdown of the main molecular regulators of the two steps of the adipogenesis process. Moreover, impaired WAT remodeling with aging has been associated with various age-related conditions and reduced lifespan expectancy. Here, we delve into the latest advancements in comprehending the molecular and cellular processes underlying age-related changes in WAT function, their involvement in common aging pathologies, and their potential as therapeutic targets to influence both the health of elderly people and longevity. Overall, this review aims to encourage research on the mechanisms of WAT maladaptation common to conditions of both excessive and insufficient fat tissue. The goal is to devise adipocyte-targeted therapies that are effective against both obesity- and age-related disorders.

1. Introduction

Adipose tissue (AT) is a fascinating and multifaceted organ. From a morphological and spatial–topographic perspective, it can delineate the contour of a physically fit individual, accentuating the elegant curves and captivating lines with artistic precision. Conversely, in the case of a long-lasting unhealthy lifestyle, particularly when coupled with predisposing genetic factors, AT can become a burdensome and disabling factor, leading to physical and psychological distress. Alongside the physical challenges associated with obesity, such as severely restricted mobility and diminished quality of life, exists the psychological burden of perceiving AT as a catalyst for social judgment and isolation.
The eclectic nature of this large organ is also evident from a functional perspective. While it was traditionally perceived as an inert depot for storing triacylglycerol, a mounting body of evidence over the past 50 years indicates that AT functions as a dynamically active metabolic and endocrine organ [1]. Its functions range from providing mechanical and thermal insulation to protect other organs to playing a critical role in energy storage, heat production, and immune function. To accomplish these functions, adipocytes are organized in specific areas known as white AT (WAT) and brown adipose tissue (BAT), with further divisions based on their location in the body. As we delve deeper into the origins and functions of adipocytes, including their metabolic, bioenergetic, and immune functions, it becomes evident that these cells play diverse roles even within the same AT depot [2]. Furthermore, the AT is not solely composed of adipocytes; rather, it comprises the extracellular matrix and various cell types, including adipocyte precursor cells, preadipocytes, endothelial cells, vascular precursor cells, and immune cells [3], collectively referred to as the stromal vascular fraction (SVF), also known as the AT microenvironment [4]. Advanced techniques like single-cell RNA sequencing (scRNA-seq) have offered deeper insights into WAT complexity, but they also raised new questions about the functions of its cellular subtypes [5,6].
The AT is able to adapt and respond to internal and environmental stimuli by remodeling both its cellular and extracellular components, which makes it a key player in health and disease [7]. This ability to undergo phenotypic, metabolic, and structural adaptations is called AT plasticity [8]. Dynamic AT changes in response to cold and warm temperatures involve the processes of beiging/browning and whitening, respectively, of WAT and BAT, the corresponding switch from a “storage and release” to an “uptake and oxidation” metabolic program and vice versa, and de novo adipogenesis [8]. This issue exceeds this review’s scope, and readers are referred to the vast literature on BAT plasticity and its role in thermogenesis.
Focusing on WAT plasticity, it is referred to as the ability of this fat depot to undergo adaptations to several stimuli, including food availability and intake, which trigger differentiation from precursors to mature adipocytes (i.e., adipogenesis), switching from a “fat storage” to a “fat mobilization” metabolic program, and WAT expansion through adipocyte hypertrophy and hyperplasia and vice versa, but also morphological and functional changes in various stromal vascular components [8]. These adaptations are essential for maintaining the health and function of WAT as both an energy storage and an endocrine organ. In addition, they have profound implications for modulating whole-body metabolism by regulating other important metabolic tissues, such as the liver, muscle, and pancreas, and for maintaining the health of cardiovascular, renal, nervous, and osteoarticular tissues.
The accumulation of visceral WAT (vWAT) around and within internal organs is linked to metabolic derangements, like insulin resistance and type 2 diabetes (T2D), which contrasts with subcutaneous WAT (sWAT), which is often viewed as protective, although this view is simplistic [2]. Furthermore, the aging process influences WAT through alterations in distribution patterns and composition, concomitant with diminished functionality of adipocyte precursor cells and an increase in the presence of senescent cells [9].
The focus of this review is the role of maladaptive WAT remodeling in obesity and associated comorbidities, with a special emphasis on impaired adipogenesis. In particular, we will discuss how WAT maladaptation may result from various defects in adipogenesis, leading to different WAT features and metabolic consequences. Additionally, we offer insights into the latest advancements in comprehending the molecular and cellular processes underlying age-related WAT maladaptation, highlighting its importance as a core element in aging and related chronic disorders, such as osteoarthritis, osteoporosis, cognitive dysfunction, and cachexia, and examining its potential as a therapeutic target to impact both elderly well-being and lifespan. By identifying significant unresolved questions, our objective is to highlight the hurdles that remain in envisioning and developing novel therapeutic interventions to target maladaptive adipocytes in obesity and age-related diseases.

2. Impaired White Adipose Tissue Remodeling in Obesity

The sWAT is the largest and safest WAT depot of the body, as it is capable of expanding under conditions of energy surplus through de novo adipogenesis, i.e., through recruitment of new adipocytes (hyperplasia), in addition to enlargement of existing adipocytes (hypertrophy), thus allowing accommodation of large amounts of lipids without compromising metabolic health [10]. Adipocyte size was shown to increase with expanding body fat up to a maximum, after which a more rapid increase in adipocyte numbers occurred [11]. Indeed, although the number of adipocytes in human sWAT is established around puberty, there is an annual turnover of ~10% [12].
However, once this adipogenic capacity is overwhelmed, sWAT expansion proceeds exclusively through hypertrophy, and when adipocytes achieve a critical cell volume, they degenerate and die through pyroptosis [13], with the formation of crown-like structures of dead/dying adipocytes surrounded by infiltrating macrophages and fibrotic tissue [14]. This results in sWAT dysfunction (adiposopathy) with fat diversion to vWAT and ectopic sites, including the liver, muscle, pancreas, and heart, ultimately leading to tissue and systemic inflammation and insulin resistance, development of T2D, and increased risk of cardiovascular disease (CVD) and cancer [15,16]. Thus, at variance with hyperplastic obesity, hypertrophic obesity is associated with increased cardiometabolic risk (Figure 1).
It is worth noting that the adipogenic capacity of sWAT is not only limited but also extremely variable among individuals, as shown by the existence of persons with obesity either with or without insulin resistance and components of the metabolic syndrome, including impaired glucose regulation, atherogenic dyslipidemia, and hypertension, despite the same body mass index (BMI). In addition to these two obese phenotypes, which are called metabolic unhealthy obese (MUO) and metabolic healthy obese (MHO), respectively [17,18], there are also lean phenotypes characterized by the presence or absence of metabolic derangement, i.e., the metabolic unhealthy normal weight (MUNW), also called metabolically obese normal weight [MONW], and the metabolic healthy normal weight (MHNW), respectively [19]. This indicates that it is not just a matter of absolute quantity of fat and shifts the focus on sWAT dysfunction and remodeling of fat depots, which is dependent on the individual capacity to expand and accommodate the surplus of energy [20]. However, it is debated whether MHO is a transient condition that is destined, sooner or later, to shift towards MUO [18], and studies in people with the MHO phenotype have shown that they still have an increased risk of T2D and CVD [21,22]. Nonetheless, there is no doubt that these individuals have a higher adipogenic capacity than those with the MUO and especially the MUNW phenotypes due to genetic and epigenetic factors [10].

2.1. Adipocyte Size as a Marker of White Adipose Tissue Dysfunction

The concept of impaired sWAT adipogenesis and expandability, linking obesity to its complications [23], points to the importance of adipocyte size (hypertrophy) as opposed to adipocyte numbers (hyperplasia) as a marker of sWAT dysfunction and predictor of increased cardiometabolic risk [24]. Adipocyte size varies according to several factors, including sex and fat depot (lower in vWAT than in sWAT in women, but not in men) [24], as well as the method of measurement [25]. Adipocytes are generally classified by size into three categories, i.e., small (<70 μm), large (70–120 μm), and very large (>120 μm) [25], or four categories, i.e., small, medium, large, and very large [26]. In this section, we will briefly summarize the evidence supporting the concept that the higher the size (or the lower the number) of sWAT adipocytes, the higher the risk of fat diversion to vWAT and ectopic sites and the impairment of insulin sensitivity and glucose homeostasis predisposing to CVD and cancer. In particular, we will focus on the studies showing that metabolic dysfunction correlates with “large adipocytes”, indicating the predominance of hypertrophy over hyperplasia, but also on the reports highlighting the importance of a population of “small adipocytes” in the development of insulin resistance and T2D, possibly suggesting impaired adipocyte maturation.

2.1.1. Large Adipocytes

Adipocyte size was shown to increase with increasing body fat mass, with a curvilinear relationship [24]. Likewise, adipocyte size was found to increase with increasing BMI until reaching a plateau at a BMI ~30 kg/m2, which, however, was lower in men (~25 BMI kg/m2) than in women (~35 kg/m2), suggesting a greater propensity to develop hypertrophic obesity in the male sex compared to the female sex [24]. Several studies have shown that adipocyte size is increased in people with obesity [11,27,28] and T2D [29,30,31], and even in lean individuals with a genetic predisposition to T2D [32,33] but not to obesity [32]. Moreover, adipocyte size correlated positively with lipolysis, WAT, systemic inflammation, and insulin resistance [27,29,30,31,34] and negatively with lipogenesis [34], and it predicted the incidence of T2D [30,31] as well as the persistence of T2D or T2D risk after gastric by-pass surgery [35]. A number of studies have reported an association between adipocyte size and dyslipidemia, with differences according to sex and fat depot [36,37,38,39,40]. Conversely, adipocyte numbers correlated positively with insulin sensitivity and HDL-cholesterol and negatively with insulin and triglycerides [36], and a decreased adipogenic potential of sWAT, but not vWAT, was found to be associated with visceral obesity [41] and impaired glucose regulation [42].
When using the deviation of the measured volume from the expected volume according to body fat mass, with positive and negative values corresponding to hypertrophy and hyperplasia, respectively, this marker of adipocyte morphology was independent of sex and body weight and correlated positively with fasting plasma insulin levels and insulin resistance, irrespective of adipocyte size, and negatively with total adipocyte numbers [41]. Moreover, a negative correlation was observed between adipocyte hypertrophy and hyperplasia when adjusted for the predicted adipocyte volume at a given body fat mass, indicating that the higher the predicted adipocyte size, the lower the rate of adipogenesis. In fact, the absolute number of new adipocytes generated each year was 70% lower in hypertrophy than in hyperplasia, and individual values correlated strongly with morphology but not adipocyte size [43].
However, other studies did not find a correlation between sWAT adipocyte size and metabolic derangement [28,44,45] or reported an association in non-diabetic but not in T2D individuals [46]. Furthermore, in Asian Indians, size was higher but less strongly correlated with measures of adiposity and metabolic parameters in sWAT versus vVAT adipocytes [47].

2.1.2. Small Adipocytes

A number of studies have shown a bimodal cell size distribution in WAT, with about the same proportion of large and small adipocytes and the small rather than the large cell population being related to metabolic derangement associated with obesity. In fact, McLaughlin et al. reported that among individuals with obesity, those with insulin resistance had fewer large adipocytes with a similar or larger mean diameter and a higher ratio of small to large cells compared with their insulin-sensitive counterparts. It is worth noting that the fraction of small adipocytes was associated with insulin resistance, decreased expression of adipogenic genes, and increased expression of inflammatory genes, whereas the diameter of the large adipocytes was associated with percent body fat [48,49,50]. Moreover, Pasarica et al. showed that obese individuals with T2D had lower total adipocyte numbers, with an increase in the fraction of small adipocytes as well as a decrease in the size of small and medium adipocytes and a marked increase in the size of very large adipocytes compared to those without T2D [51]. Likewise, Fang et al. found an increase in the percentage of small cells and a corresponding decrease in the percentage of large cells in both sWAT and vWAT of individuals with versus without T2D; the fraction of small adipocytes, but not that of large adipocytes, correlated positively with indices of glucose homeostasis, though this association was significant only for vWAT [52]. Finally, two studies assessed the impact of short-term overfeeding on metabolic derangement according to adipocyte size. Johannsen et al. showed no association between sWAT cell size and ectopic fat accumulation, but individuals with smaller adipocytes at baseline had a greater decrease in insulin sensitivity, which was related to skeletal muscle tissue inflammation [53]. McLaughlin et al. reported that insulin-resistant individuals, who had a higher adipocyte diameter and percentage of small adipocytes associated with a worse metabolic profile, exhibited no change in cell size and no (or only minor) further fat diversion to vWAT and ectopic sites and deterioration of insulin sensitivity with diet-induced body weight gain. This was at variance with insulin-sensitive individuals, who demonstrated significant enlargement of adipocytes and reduction of the percentage of small adipocytes associated with a marked impairment of insulin sensitivity, which was predicted by the smaller baseline adipocyte size [54].
These findings were thought to challenge the AT expandability hypothesis linking adipocyte hypertrophy to metabolic derangement associated with obesity [55]. However, they have also been interpreted as the consequence of an impaired ability of sWAT to generate fully mature adipocytes capable of accommodating surplus energy [10]. This implies that sWAT dysfunction limiting tissue plasticity and fat storage capacity may arise not only from a reduced hyperplastic WAT response due to impaired recruitment and expansion of adipocyte precursors (the first step of adipogenesis) but also from a failure of adipocytes to accumulate lipids due to defective terminal differentiation of preadipocytes into fully mature cells (the second step of adipogenesis) [56].

2.2. Adipogenesis as a Central Mechanism for White Adipose Tissue Plasticity

As discussed above, adipogenesis represents a safe mechanism for WAT expansion under conditions of energy surplus, as it allows for accommodating large amounts of fat without achieving the critical adipocyte volume that drives cell degeneration and death as well as fat diversion to ectopic sites and related metabolic derangement [8].
Adipogenesis is the process by which pluripotent mesenchymal stem cells (MSCs) commit to the adipose lineage to become preadipocytes, which then differentiate into adipocytes [57]. Preadipocytes differentiate by undergoing mitotic clonal expansion, followed by growth arrest and terminal differentiation with acquisition of a mature phenotype, i.e., expression of adipocyte genes and accumulation of triglycerides (Figure 2) [57].
Studies of pluripotent stem cells or preadipocytes, such as the C3H10T1/2 and 3T3-L1/F442A cell lines, respectively, have enabled the characterization of the two steps of adipogenesis and the identification of the molecular regulators of this process. The results of these studies have been extensively reviewed elsewhere [57,58] and will be briefly summarized here.

2.2.1. Commitment of Pluripotent Stem Cells to Adipocyte Lineage

Pluripotent MSCs, which are located in the SVF of AT and the bone marrow, can undergo a multistep process of commitment in the adipocyte, myocyte, chondrocyte, or osteocyte lineage, depending on specific signals. Commitment to the adipocyte lineage with the generation of preadipocytes is triggered by chronic overfeeding through increased GLUT4-dependsent glucose uptake in the AT [59]. Several factors have been identified that modulate commitment to the adipocyte lineage, including the members of the bone morphogenetic protein (BMP) family BMP-2 and 4 [60,61], the wingless-type MMTV integration site (Wnt) family [62,63,64], and the Hedgehog (Hh) protein family [65]. Interestingly, these factors are members of developmental signaling pathways that have been conserved from invertebrates to vertebrates, like fat function and distribution [66].
The BMPs play a fundamental role in the commitment to adipocyte lineage by signaling through the BMP receptors (BMPr) 2 and 1, which form cell surface complexes with serine/threonine kinase activity. Upon binding to BMPs, the BMPr1 kinase is phosphorylated and activated, thus phosphorylating the transcription factor Smad 1/5/8, which forms a complex with Smad4 that translocates into the nucleus and regulates gene expression [61]. The non-canonical BMP signaling through the mitogen-activated protein kinases (MAPK) extracellular signal-regulated kinase (ERK), Jun N-terminal kinase (JNK), and p38 is also involved in the promotion of adipogenesis [67]. Downstream targets of BMP signaling include three cytoskeleton-associated proteins, i.e., lysyl oxidase (Lox), translationally controlled tumor protein 1 (Tpt1), and αβ-crystallin, which promote F-actin fibers’ breakdown and rearrangement to the preadipocyte periphery, resulting in cell shape changes [68]. Signaling through the BMP pathway is inhibited by several substances, including gremlin, noggin, matrix Gla protein (MGP), BMP, and activin membrane bound inhibitor (BAMBI), chordin-like, follistatin, and follistatin-like 3 [67].
The Wnt family of secreted signaling glycoproteins also participates in stimulating adipocyte lineage commitment via the canonical pathway by acting upstream of BMPs. The activation of Wnt signaling through the binding of Wnt ligands to the cell surface frizzled receptor and the associated low-density lipoprotein receptor-related coactivator 5/6 results in the dissociation of the destruction complex consisting of adenomatous polyposis coli (APC), axin, and glycogen synthase kinase-3β (GSK-3β). This prevents phosphorylation by GSK-3β and consequent ubiquitination and proteasomal degradation of β-catenin, which allows it to translocate into the nucleus and activate the transcription factors lymphoid enhancer factor 1 (Lef1) and T-cell factor (Tcf) [69]. Using microarray technology, the activators R-spondins-2 and -3 and the downstream transcription factors Lef1 and Tcf of the canonical Wnt signaling pathway were found to be upregulated in proliferating A33 preadipocytes but not in C3H10T1/2 progenitor cells, together with β-catenin nuclear accumulation [62]. Though these findings might suggest a favorable effect of the Wnt pathway on adipocyte commitment, the latter was shown to be inhibited by the Wnt inducible signaling pathway protein (WISP2) via negative regulation of the BMP pathway (see below) [70]. Moreover, the activation of the Wnt pathway plays an inhibitory role in adipogenesis by maintaining preadipocytes in undifferentiated states by suppressing PPARγ and C/EBPα, thus promoting diversion of precursor cells to other lineages [53,64].
Finally, it is generally agreed that expression of Hh proteins inhibits adipocyte lineage commitment. These proteins bind to a cell surface receptor complex consisting of the Patched (Ptc) receptors 1 and 2 and the seven-transmembrane receptor Smoothened (Smo), thus relieving Smo from Ptc inhibition and allowing for the activation of the transcription factors Gli1, Gli2, and Gli3 [71]. In C3H10T1/2 progenitor cells, sonic Hh protein was shown to enhance BMP-2-stimulated osteoblast commitment while suppressing adipocyte commitment [65].

2.2.2. Preadipocyte Differentiation into Mature Adipocytes

Mitotic clonal expansion is a prerequisite for preadipocyte differentiation [72]. In vitro, when exposed to a differentiation medium containing insulin, dexamethasone, and isobutyl-methyl-xanthine, a non-selective phosphodiesterase inhibitor raising intracellular cyclic AMP, preadipocyte cell lines, such as 3T3-L1 and 3T3-F442A, reenter the cell cycle and initiate a proliferative burst. Cell cycle progression is mediated by cyclin-dependent kinases (CDKs) that phosphorylate the retinoblastoma cell-cycle-related protein (Rb), thus releasing E2 transcription factor (E2F) 1, which becomes available to promote the expression of genes involved in S-phase entry, DNA synthesis, and mitosis [73]. The protein kinase A-dependent phosphorylation of cyclic AMP response element-binding protein (CREB) is a critical step in this process, as phosphorylated CREB binds to the promoter and increases the expression of cyclin D1 [74] and the transcription factor CCATT enhancer binding protein (C/EBP) β [75]. While cyclin D1 is required for proliferation [74], C/EBPβ undergoes dual phosphorylation sequentially induced by MAPK and GSK-3β [76], which produces a conformational change that renders the leucine zipper of monomeric C/EBPβ accessible for dimerization, thus allowing acquisition of DNA-binding activity [77]. Though C/EBPβ is required for mitotic clonal expansion in vitro [78], concomitant expression and activation of C/EBPδ also play a role in this process [79]. Preadipocyte factor 1 (Pref-1)/delta-like non-canonical notch ligand 1 (Dlk1) is a transmembrane protein expressed by preadipocytes that inhibits mitotic clonal expansion and hence differentiation into mature adipocytes [80].
Induction of terminal differentiation requires termination of mitotic clonal expansion by exiting the cell cycle, and it is mediated by C/EBPβ-induced transactivation of C/EBPα and peroxisome proliferator-activated receptor (PPAR) γ [81]. The genes coding for these transcription factors have C/EBP regulatory elements for C/EBPβ binding [82], through which C/EBPα and PPARγ cross-activate each other [83] and C/EBPα maintains its expression (autoactivation) [84]. In vitro adipocyte differentiation requires PPARγ [85], which acts as the master regulator of the adipogenesis program through a unified pathway [86]. It was in fact shown to promote adipogenesis in C/EBPα-deficient cells [73], which is at variance with C/EBPα in cells lacking PPARγ [87]. However, repression of E2F1-dependent transcription by C/EBPα was found to be critical for the suppression of cell proliferation and the induction of terminal differentiation [88]. Transactivated C/EBPα and PPARγ serve as pleiotropic transcriptional activators of a group of genes promoting the acquisition of the morphological and functional features of mature adipocytes [89]. Among these genes, the transcription factor sterol regulatory element-binding protein (SREBP) 1c, primarily regulated at the transcriptional level by insulin, plays a central role by controlling the expression of genes coding for enzymes of fatty acid (FA) and triacylglycerol biosynthesis and catabolism, such as acetyl-CoA carboxylase (ACC), fatty acid synthase (FAS), and stearoyl CoA desaturase (SCD1) [90]. In addition, it activates PPARy expression through the production of an endogenous ligand [91]. For this reason, it is also called adipocyte determination and differentiation factor-1 (ADD-1). As a result of de novo lipogenesis, preadipocytes lose their fibroblast-like appearance and acquire that of mature adipocytes by accumulating cytoplasmic triglycerides. Other genes that are induced during adipocyte differentiation include those coding for the lipolytic enzymes adipocyte-triacylglycerol lipase (ATGL) and hormone-sensitive lipase (HSL) [92], the gluconeogenic enzyme phosphoenolpyruvate carboxykinase (PEPCK), which provides glycerol 3-phosphate for FA re-esterification [93], the carrier protein fatty acid binding protein (FABP) 4 or 422/adipocyte protein 2 (aP2), which is involved in FA uptake, transport, and metabolism [94], the insulin receptor and the insulin-responsive glucose transporter GLUT4, and the adipokines leptin, resistin, and adiponectin [57].
Several factors were shown to regulate adipogenesis by stimulating or inhibiting terminal differentiation via modulation of the adipogenic transcription factors PPARγ and C/EBPα. Activation of BMP signaling has a stimulatory role, as phosphorylated Smad 1/5/8 releases zinc finger protein (Zfp) 423 from the complex with WISP2, thus enabling Zfp423 to translocate into the nucleus and activate PPARγ transcription [70]. Moreover, the transcription factor Schnurri2 was also found to mediate BMP-dependent induction of PPARγ expression [65]. The Wnt signaling pathways was shown to exert an inhibitory effect on the differentiation of preadipocytes into mature adipocytes not only via formation of the WISP2-Zpf423 complex but also through the stabilization and nuclear translocation of β-catenin [63,64]. Likewise, an inhibitory role in adipocyte differentiation was also reported for the Hh signaling pathway, which is mediated by GATA 2and 3 [66]. At variance with E2F1, E2F4 was found to negatively regulate adipogenesis by repressing PPARγ expression during terminal adipocyte differentiation [95]. Hypophosphorylated RB proteins are also involved by playing a dual role, as they were shown to either stimulate adipogenesis by enhancing the binding and transcription of C/EBPβ [96] or inhibit adipogenesis by forming a complex with PPARγ, which contains the histone deacetylase (HDAC) 3, which attenuates PPARγ-induced expression of adipogenic genes [97]. The Krupel-like factors (KLFs) are deoxyribonucleic acid-binding transcriptional factors that regulate adipogenesis both positively and negatively [98]. Activators of adipogenesis include KLF4, early growth response 2 (Egr2)/Krox20, KLF5, KLF6, KLF8, KLF9, KLF13, and KLF15, which act via C/EBPs and PPARγ, except KLF6, which inhibits the expression of Pref-1/Dlk1 [99]. Suppressors of adipogenesis include KLF2, KLF3, and KLF7, which also act through C/EBPs and PPARγ [99]. Finally, galectins are multifunctional lectins belonging to an ancient family defined by recognition of β-galactoside structures through a carbohydrate-recognition-binding domain (CRD). Three of the mammalian galectins, galectin-1 [100], 3 [101,102], and 12 [103], have been shown to be required for in vitro adipocyte differentiation. Moreover, it was found that (a) galectin-3 stimulates proliferation of preadipocytes [104]; (b) both galectin-1 and 3 interact with PPARγ during differentiation of preadipocytes [100,101]; and (c) galectin-12 causes cell cycle arrest when expressed in cultured cells [105], negatively regulates lipolysis in adipocytes [106], and promotes apoptosis in WAT [107].

2.3. Defective Adipogenesis and White Adipose Tissue Dysfunction

The classical view linking impaired sWAT expandability due to defective adipogenesis to tissue and systemic inflammation and associated metabolic derangement has been tested in in vivo studies using genetically modified mice maintained under standard conditions or challenged with energy surplus by feeding a high-fat diet (HFD) or crossing with mice with genetic obesity. These animals were either lacking or overexpressing one of the numerous factors that were shown to be involved in the formation of mature adipocytes from uncommitted precursors through the two steps of adipogenesis in in vitro studies. In some cases, the genetic modification was responsible for a phenotype resembling human lipodystrophy, which allowed for investigating the effect of reduced AT development on the occurrence of metabolic derangement and the response to overfeeding. In some others, it resulted in a silent phenotype that required exposure to increased energy/fat intake to unmask the impaired WAT capacity to safely accommodate lipids. However, studies in animal models have been difficult for several reasons. The most important one is that ablation of genes coding for some of the factors involved in adipogenesis is lethal, and alternative strategies had to be used. One of these strategies was generating adipocyte-specific knockout mice using the Cre-loxP system, which, however, showed different specificity and efficacy depending on the gene promoter. Here, we describe the results of a number of these studies investigating the role of various molecular regulators, which might help to understand the mechanisms operating in the human obese and lean phenotypes (Table 1).

2.3.1. Conserved Developmental Signaling Pathways

Mice overexpressing BMP4 in adipocytes from the FABP4/aP2 promoter showed reduced sWAT mass and adipocyte size along with an increased number of white adipocytes with brown characteristics, increased energy expenditure, improved insulin sensitivity and glucose tolerance, and protection against HFD-induced obesity and metabolic derangement [108]. Conversely, adipocyte-specific BMP4-deficient mice exhibited increased WAT mass and adipocyte size along with brown adipocytes with white characteristics and impaired insulin sensitivity and glucose tolerance [108]. Consistent with these results, ablation of the BMP enhancer kielin/chordin-like protein aggravated HFD-induced obesity, whereas its overexpression was protective [109], and deletion of the BMP inhibitor follistatin-like 3 was shown to reduce body fat and improve insulin sensitivity and glucose tolerance [110]. However, combined loss of follistatin and follistatin-like 3 was associated with increased fat mass, insulin resistance, and glucose intolerance [110].
Mice overexpressing the Wnt ligand Wnt10b in adipocytes from the FABP4/aP2 promoter were shown to have impaired WAT (and BAT) development, with a reduction in the number but not in the size of adipocytes [111] and protection from dietary and genetic obesity [111,112]. The finding that these mice were more insulin-sensitive and glucose-tolerant than their wild-type counterparts despite the impaired WAT plasticity was attributed to decreased levels of resistin and/or inflammatory cytokines [111,112]. Resistance to HFD-induced obesity was also observed in mice with ablation of β-catenin [113] or the chaperone protein Wntless [114] in mature adipocytes as well as in mice with global deficiency of functional secreted frizzled-related protein (SFRP) 5, an inhibitor of Wnt signaling [115]. Interestingly, WAT of mice lacking functional SFRP-5 had an unchanged number of adipocytes associated with an increase in small cells and a decrease in large cells [115]. Conversely, increased body weight, WAT mass, and adipocyte size associated with impaired insulin sensitivity and glucose tolerance were observed in HFD-fed mice with adipocyte-specific deletion of transcription factor 7-like 2 (TCF7L2), a key intracellular effector of the Wnt-signaling pathway [116].
The naturally occurring spontaneous mesenchymal dysplasia (mes) in adult mice (Ptc1mes/mes) carrying a deletion of Ptc1 at the carboxyl-terminal cytoplasmic region, which is essential for the inhibition of Hh signaling, showed a significant reduction in epididymal WAT mass and adipocyte size (and possibly number) under standard feeding conditions. In addition, the gene expressions of PPARγ and CEBPα and adipocyte markers were reduced, with no change in glucose tolerance [117]. However, previous studies have provided opposite results, which contrast with the inhibitory role of the Hh proteins on adipocyte commitment and differentiation. In fact, body weight and fat mass were significantly increased by injection of the Hh agonist sonic Hh-IgG fusion protein in adult mice [118] and decreased by injection of anti-Hh antibodies in mice with dietary or genetic obesity [119].

2.3.2. CCATT Enhancer Binding Proteins

Studies in animal models lacking C/EBPs were severely limited by the embryonic lethality of mice deficient in these transcription factors.
Mice knockout for C/EBPβ or C/EBPδ were shown to have virtually normal WAT mass, whereas a marked defect due to a reduction in the number more than in the size of adipocytes was found in double C/EBPβ and C/EBPδ knockout mice, suggesting a functional redundancy of members of the C/EBP family. However, it should be considered that only 15% of cebpb−/−/cebpd−/− animals survived until adulthood [120].
As mice deficient in C/EBPα failed to accumulate lipids in sWAT but died shortly after birth from hypoglycemia due to impaired liver gluconeogenesis [121], transgenic cebpa−/− animals expressing C/EBPα only in the liver were generated [122]. These mice also showed an absence of WAT associated with postprandial hyperlipidemia, fatty liver, and increased insulin but normal glucose levels [122].

2.3.3. Peroxisome Proliferator-Activated Receptor γ

Ablation of PPARγ in vivo is also lethal; thus, alternative mouse models with either global or adipocyte-specific PPARγ deficiency have been used for dissecting the role of this transcription factor in adipogenesis [123].
Rescuing of embryonic lethality despite global PPARγ deletion was obtained by preserving PPARγ expression in the trophoblasts or generating hypomorphic, heterozygous, chimera, or isoform-specific knockout animals as well as mice carrying dominant negative human mutations [123]. Inactivation of PPARγ in the embryo but not in trophoblasts caused severe lipodystrophy, organomegaly, and insulin resistance, whereas glucose tolerance was decreased in male and increased in female mice [124]. Mice carrying a hypomorphic mutation at the PPARγ2 locus also showed no or only rare and hypertrophic adipocytes due to the elimination of PPARγ before WAT development, in addition to reduced premature lethality. However, surviving mice had no hepatic steatosis but rather fat accumulation at the muscle level, with only mild impairment of glucose tolerance and elevation of glucose and insulin levels only in the fed state, indicating modest insulin resistance. The discrepancy between the severe lipodystrophy resembling human autosomal dominant familial partial lipodystrophy and the mild metabolic phenotype was attributed to the induction of a compensatory gene expression program in the muscle promoting oxidation of excess lipids [125]. In contrast, mice heterozygous for PPARγ were shown to have comparable body weight, fat-pad mass, and free fatty acid (FFA) levels and to be more insulin-sensitive than their wild-type littermates, a finding that was explained by the lack of PPARγ-dependent transcriptional repression of genes impairing insulin sensitivity and/or increased expression of leptin [126]. The use of pparg+/+ and pparg−/− chimeras demonstrated the central role of PPARγ in AT development, as only pparg+/+ adipocytes were detected in these animals [85]. Compared with wild-type animals, mice with deletion of PPARγ2 showed comparable body weight but reduced WAT mass due to a decrease in both adipocyte numbers and size, together with severe insulin resistance in male but not female animals and normal glucose tolerance. Moreover, these mice were protected from HFD-induced obesity [127]. In contrast, another study reported that pparg2−/− mice fed standard chow exhibited no difference versus wild-type mice in body weight, WAT mass, adipocyte numbers and size, FFA and triglyceride levels, and ectopic fat accumulation, but they showed peripheral insulin resistance and impaired glucose tolerance [128]. Conversely, when fed an HFD, knockout mice showed increased WAT area and adipocyte size despite comparable body weight but similarly reduced insulin sensitivity [128]. These findings indicate that PPARγ2 regulates glucose homeostasis and insulin sensitivity independent of its effect on adipogenesis [127,128]. Heterozygous knock-in mouse models carrying the dominant negative PPARγ mutations L466A and P465L, which were found in humans with lipodystrophy and/or the metabolic syndrome, recapitulated some of the features of the human phenotypes. Heterozygous PPARγ L466A knock-in mice had lower body weight gain and fat mass with a heterogenous population of adipocytes of predominantly small and medium size instead of a homogenous population of large adipocytes, as in wild-type animals. This was associated with decreased adiponectin and increased FFA levels, hepatic steatosis, and hypertension but normal insulin sensitivity. However, on HFD, these mice gained less weight and developed a more severe insulin resistance than wild-type animals [129]. In contrast, heterozygous PPARγ P465L knock-in mice had normal body weight, fat mass, and insulin sensitivity but altered AT distribution with increased sWAT and a more homogeneous population of adipocytes of a larger size than in wild-type mice, associated with increased glucose tolerance and hypertension. When fed an HFD, these animals showed a comparable reduction in whole-body, hepatic, and peripheral insulin sensitivity compared to wild-type mice [130].
Mice with adipocyte-specific PPARγ deletion also showed a lipodystrophic phenotype, though the results of these studies were not univocal with respect to the extent of the reduction of fat mass and the metabolic consequences of impaired WAT development and/or maintenance. A less severe phenotype was observed in adipocyte-specific PPARγ knockout mice generated using a FABP4/aP2 gene-promoter-regulated Cre-loxP system, a finding that was attributed to the deletion of PPARγ at a later developmental stage. He et al. showed that these mice had reduced WAT (and BAT) mass due to a marked decrease in adipocyte numbers because of apoptosis, associated with macrophage infiltration and fibrosis [131]. It is worth noting that while most of the remaining cells were highly hypertrophic, there was also a population of small adipocyte-like cells, suggesting the recruitment of adipocyte precursors that failed to maturate, a feature that mimics that reported in obese humans [48,49,50,51]. Under standard chow feeding, these animals showed liver steatosis with decreased leptin and increased FFA, triglyceride levels, and hepatic glucose production compared with wild-type mice, though insulin sensitivity and glucose tolerance were comparable. However, when fed an HFD, they gained significantly less weight, and their residual WAT (and BAT) became more lipoatrophic as in aging mice on standard chow; moreover, glucose and insulin levels were two-fold higher and insulin sensitivity was lower than in wild-type animals. These findings highlight the essential role of PPARγ in the survival of mature adipocytes and the terminal differentiation/maturation of new fat cells. Using the same approach but a different FABP4/aP2-Cre transgene, Jones et al. also found that compared with wild-type animals, mice with adipocyte-specific PPARγ ablation had decreased weight gain, WAT mass, and adipokine levels when fed an HFD, together with WAT inflammation and fibrosis and fatty liver [132]. However, in contrast to He et al., they reported that adipocyte-specific PPARγ knockout mice were completely protected from HFD-induced insulin resistance and impaired glucose tolerance. In euglycemic–hyperinsulinemic clamp studies, the whole-body glucose disposal rate was in fact higher in adipocyte-specific PPARγ knockout versus wild-type mice fed an HFD. This was due to higher glucose disposal at the liver but not the muscle level, as hepatic glycogen synthesis and content were enhanced in knockout versus wild-type mice, whereas muscle glucose uptake was similar in the two groups. These findings suggest a central role for the liver in protecting from whole-body insulin resistance by increasing glucose metabolism despite enhanced fat content, possibly due to a compensatory increase in hepatic PPARγ expression. However, a comparison of different Cre lines suggested that both the mild WAT phenotype and the contrasting data regarding the associated metabolic derangement might be due to the limited specificity and efficacy of the FABP4/aP2-Cre approach in mediating adipocyte-specific recombination [133]. An adipocyte-specific PPARγ knockout mouse model generated using a transgenic Cre line driven by the regulatory region of adiponectin (Adipoq–Cre) was in fact shown to have nearly absent WAT (and BAT) and a small number of adipocytes dispersed among a great number of stromal cells [134]. This was associated with a severe reduction in adipokine levels, massive liver steatosis, dramatic elevation of glucose and insulin levels, and extreme insulin resistance. The finding that selective ablation of PPARγ in mature adipocytes by using a tamoxifen-inducible recombination system caused a rapid and massive death of white and brown pparg−/− adipocytes, which were subsequently replaced by newly formed pparg+/+ adipocytes, confirmed the critical role of PPARγ for the survival of mature adipocytes [135].

2.3.4. Other Factors

Mice knockouts for E2F1 were also found to be resistant to HFD-induced obesity, as the lower increase in body weight versus wild-type mice was entirely explained by a lower increase in WAT mass [95].
In contrast with the in vitro studies showing a compelling role for SREBP-1c in promoting adipogenesis, in vivo studies provided different results. In fact, the surviving SREBP-1 knockout mice (only 15%) showed no change in WAT development or the expression of key adipogenic genes when compared to wild-type mice, suggesting a compensation from the upregulation of SREBP-2 or other factors [136]. Even more surprisingly, transgenic mice overexpressing the nuclear form of SREBP-1c (nSREBP) in adipocytes under the control of the FABP4/Ap2 enhancer/promoter showed a phenotype resembling human congenital generalized lipodystrophy. The differentiation of AT was markedly impaired, with the decreased WAT consisting almost entirely of small immature adipocytes and the increased BAT containing hypertrophic, fat-laden brown adipocytes resembling immature white cells as well as reduced expression of adipogenic genes. This was associated with fatty liver, severe insulin resistance, and impaired glucose homeostasis. A possible explanation for these findings was the interference with adipocyte differentiation by the very high nuclear levels of nSREBP-1c, as the FABP4/aP2-nSREBP-1c transgene encodes a truncated version of SREBP-1c that enters the nucleus directly with no requirement for proteolysis and no feedback regulation [137].
Mice overexpressing PEPCK in adipocytes from the FABP4/aP2 promoter showed increased body weight, fat mass, and adipocyte size as a consequence of increased glycerol 3-phosphate synthesis and FA re-esterification. This was associated with decreased FFA and normal leptin levels in the circulation, which were likely responsible for the preserved insulin sensitivity and glucose tolerance [138].
Mice lacking Schnurri-2 showed a reduced body weight and epidydimal WAT mass with a decrease in the size but not in the number of adipocytes; however, insulin sensitivity and glucose tolerance were increased despite lipodystrophy [139]. These findings are similar to those observed in mice heterozygous for CREB-binding protein, a co-activator for several transcriptions, including SREBPs, C/EBPs, and PPARs, which were also completely protected from HFD-induced obesity [140].
Mice lacking Pref-1/Dlk1 showed accelerated adiposity associated with growth retardation; this phenotype was also observed in mice heterozygous for paternally but not maternally expressed pref1-null allele [141]. Mice expressing Pref-1 in adipocytes from the FABP4/aP2 promoter showed severely reduced fat mass due to decreased adipocyte size and reduced expression of adipocyte markers and adipokines, which are associated with increased triglycerides and impaired insulin sensitivity and glucose tolerance [142]. These mice were also found to be resistant to HFD-induced obesity, as they gained less weight and fat mass but showed higher FFA levels and more severe insulin resistance and glucose intolerance than their wild-type counterparts [143].
Mice deficient for KLFs showed no or only a minor phenotype. In fact, heterozygous KLF5 knockout mice exhibited impaired development of WAT characterized by a smaller adipocyte size, which, however, reversed by 4 weeks of age [144], whereas mice lacking KLF13 showed normal adipogenesis [145].
Table 1. Studies in animal models investigating the role of molecular regulators of adipogenesis.
Table 1. Studies in animal models investigating the role of molecular regulators of adipogenesis.
Study
[Reference]
Mouse ModelNormal Diet/Genetic BackgroundDietary/Genetic Obesity
MoleculeManipulationFat Mass/CellsMetabolism↑ Body Weight/Fat MassMetabolic Derangement
Conserved developmental signaling pathways
Qian et al.
[108]
BMP-4Adipocyte-specific (FABP4) overexpression↓ WAT mass
↓ size ↑ number
↑ insulin sensitivity
↑ glucose tolerance
Adipocyte-specific (FABP4) deletion↑ WAT mass
↑ size ↓ number
↓ insulin sensitivity
↓ glucose tolerance
NANA
Soofi et al.
[109]
Kielin/chordin-like proteinGlobal overexpression↓ WAT mass= insulin sensitivity
= glucose tolerance
Global deletion= WAT mass= insulin sensitivity
= glucose tolerance
↑/=
Brown et al.
[110]
Follistatin-like 3Global deletion↓ fat mass↑ insulin sensitivity
↑ glucose tolerance
NANA
Follistatin and follistatin-likeGlobal deletion↑ fat mass↓ insulin sensitivity
↓ glucose tolerance
NANA
Longo et al.
[111]
Wnt10bAdipocyte-specific (FABP4) overexpression↓ WAT mass
= size ↓ number
↑ insulin sensitivity
↑ glucose tolerance
Wright et al.
[112]
Wnt10bAdipocyte-specific (FABP4) overexpression↓ WAT massNA
Chen et al.
[113]
β-cateninAdipocyte-specific (Adipoq) deletion↓ WAT massNA
Bagchi et al.
[114]
WntlessAdipocyte-specific (Adipoq) deletion==
Mori et al.
[115]
SFRP-5Global deletion↑ WAT mass
↑ size = number
NA
Geoghegan et al.
[116]
TCF7L2Adipocyte-specific (Adipoq) deletionNANA
Li et al.
[117]
Ptc1Global deletion at the carboxyl-terminal cytoplasmic region↓ fat mass
↓ size ↓ number
= glucose toleranceNANA
Martin et al.
[118]
HhSonic Hh-IgG fusion protein injection↑ fat massNANANA
Buhman et al.
[119]
HhAnti-Hh antibody injectionNANANA
CCATT enhancer binding proteins
Tanaka et al.
[120]
C/EBPβGlobal deletionNon-significant
↓ WAT mass
NANANA
C/EBPδGlobal deletionNon-significant
↓ WAT mass
NANANA
C/EBPβ and C/EBPδGlobal deletionSevere ↓ WAT massNANANA
Wang et al.
[121]
C/EBPαGlobal deletionAbsent WAThypoglycemiaNANA
Linhart et al.
[122]
C/EBPαGlobal deletion (except liver)Absent WAT↑ insulin
= glucose
NANA
Peroxisome proliferator-activated receptor γ
Duan et al.
[124]
PPARγGlobal deletion (except trophoblasts)Severe ↓ WAT mass↓ insulin sensitivity
↓ (M) ↑ (F) glucose tolerance
NANA
Koutnikova et al.
[125]
PPARγ2Hypomorphic mutationSevere ↓ WAT massmild ↓ insulin sensitivity
mild ↓ glucose tolerance
NANA
Miles et al.
[126]
PPARγGlobal deletion (heterozygous)= WAT mass↑ insulin sensitivityNANA
Rosen et al.
[85]
PPARγChimera= WAT mass with only pparg+/+ cellsNANANA
Zhang et al.
[127]
PPARγ2Global deletion↓ WAT mass
↓ size ↓ number
severe ↓ insulin sensitivity (M)
= glucose tolerance
NA
Medina-Gomez et al. [128]PPARγ2Global deletion= WAT mass
= size = number
↓ insulin sensitivity ↓ glucose tolerance=
Freedman et al.
[129]
PPARγDominant negative mutation L466A↓ WAT mass
↓ size (small–medium)
=I nsulin sensitivity
Tsai et al.
[130]
PPARγDominant negative mutation P465L↑ WAT mass
↑ size (large)
= insulin sensitivity
↑ glucose tolerance
=
He et al.
[131]
PPARγAdipocyte-specific (FABP4) overexpression↓ WAT mass
size (very large, small) ↓ number
= insulin sensitivity
= glucose tolerance
Jones et al.
[132]
PPARγAdipocyte-specific (FABP4) overexpression↓ WAT mass= insulin sensitivity
= glucose tolerance
Wang et al.
[134]
PPARγAdipocyte-specific (Adipoq) deletionsevere ↓ WAT mass
severe ↓ number
severe ↓ insulin sensitivity
severe ↓ glucose tolerance
NANA
Other factors
Fajas et al.
[95]
E2F1Global deletionNANANA
Shimano et al.
[136]
SREBP-1Global deletion= WAT massNANANA
Shimomura et al. [137]SREBP-1Global overexpression (nuclear form)↓ WAT mass
↓ size
↓ insulin sensitivity
↓ glucose tolerance
NANA
Franckhauser et al. [138]PEPCKAdipocyte-specific (FABP4) overexpression↑ WAT mass
↑ size
= insulin sensitivity
= glucose tolerance
NANA
Jin et al.
[139]
Schnurri-2Global deletion↓ WAT mass
↓ size = number
↑ insulin sensitivity
↑ glucose tolerance
NANA
Yamauchi et al.
[140]
CREBpGlobal deletion (heterozygous)↓ WAT mass↑ glucose tolerance
Moon et al.
[141]
Pref-1/Dlk1Global deletion↑ WAT mass
↑ size
NANANA
Lee et al.
[142]
Pref-1/Dlk1Adipocyte-specific (FABP4) overexpression↓ WAT mass
↓ size
↓ insulin sensitivity
↓ glucose tolerance
NANA
Villena et al.
[143]
Pref-1/Dlk1Adipocyte-specific (FABP4) overexpressionNANA
Oishi et al.
[144]
KLF5Global deletion↓ WAT mass
↓ size (transient)
NANANA
Gordon et al.
[145]
KLF13Global deletion= fat massNANANA
Yang et al.
[106]
Galectin 12Global deletion↓ WAT mass
↓ size = number
NA
Baek et al.
[100]
Galectin 1Global deletion= WAT mass
= size
= glucose
Baek et al.
[101]
Galectin 3Global deletion↓ WAT mass
↓ size
= glucose
Blasetti Fantauzzi et al. [102]Galectin 3Global deletion= WAT mass
↓ size ↑ number
↓ insulin sensitivity (transient)
↓ glucose tolerance
NANA
Pejnovic
et al. [146]
Galectin 3Global deletion= WAT mass= insulin sensitivity
= glucose tolerance
Li et al.
[147]
Galectin 3Global deletion= WAT mass↑ insulin sensitivity
↑ glucose tolerance
=
Pang et al.
[148]
Galectin 3Global deletion↑ WAT mass↑ glucose
Takada et al.
[149]
Galectin 3Global deletion↓ muscle fatNANANA
Martínez-Martínez et al. [150]Galectin 3Inhibitor (modified citrus pectin) injection= WAT massNA=NA
BMP = bone morphogenetic protein; Wnt = wingless-type; SFRP = secreted frizzled-related protein; TCF7L2 = transcription factor 7-like 2; Ptc = Patched; Hh = Hedgehog; C/EBP = CCATT enhancer binding protein; PPAR = peroxisome proliferator-activated receptor; E2F = E2 transcription factor; SREBP = sterol regulatory element-binding protein; PEPCK = phosphoenolpyruvate carboxykinase; CREBp = phosphorylated cyclic AMP response element-binding protein; Pref = preadipocyte factor; Dlk = delta-like non-canonical notch ligand; KLF = Krupel-like factor.
Mice knockouts for galectin-12, which is preferentially expressed in adipocytes and localized to lipid droplets, were shown to have a similar body weight but substantially lower sWAT and vWAT mass and triglyceride content compared with wild-type mice due to a lower size but not number of adipocytes. Moreover, Lgals12−/− mice exhibited significant protection from dietary and genetic obesity, as shown by the lower weight gain, insulin resistance, and glucose intolerance compared to Lgals12+/+ animals, consistent with increased lipolysis enhancing mitochondrial respiration and whole-body energy expenditure [106]. No effect on body weight, adiposity, or metabolism was reported with global deletion of galectin-1, which is expressed in several tissues, including adipocytes, where it localizes to the nucleus during differentiation. However, when fed an HFD, Lgals1−/− mice showed significantly lower increases in body weight, WAT mass, adipocyte size, and expression of lipogenic and inflammatory genes and glucose but not FFA or triglyceride levels compared with Lgals1+/+ animals, consistent with protection from dietary obesity [100]. At variance with galectin-12 and -1, several studies have investigated the effect of ablation or inhibition of galectin-3, which shows a widespread expression in the body and serves multiple functions, thus possibly explaining the somewhat contrasting data reported on WAT morphology and function in mice fed standard chow or an HFD. Both body weight and sWAT and/or vWAT mass were consistently found to be normal in young Lgals3−/− mice under standard chow conditions [102,146,147], except in one study showing higher fat mass at 20 weeks of age [148]. Blasetti Fantauzzi et al. showed that the unchanged WAT mass at 2 months of age was the result of an increased cell number compensating for a reduced size of adipocytes, which showed signs of immaturity, such as decreased expression of genes coding for adipogenic transcription factors, lipogenic enzymes, and adipokines, indicating impaired terminal differentiation [102]. Moreover, despite no sign of adipocyte degeneration/death or fat diversion to ectopic sites, this WAT phenotype was associated with tissue and systemic inflammation and insulin resistance but normal glucose levels, which, however, became elevated one month later [102]. These findings suggested that adipocyte immaturity may directly cause metabolic derangement, i.e., in the absence of an increased demand for fat storage and, hence, independently of increased fat accumulation and redistribution. This interpretation was supported by the finding that maturation of adipocytes was observed in Lgals3−/− mice at 5 months of age, along with resolution of inflammation and restoration of insulin sensitivity, though glucose levels remained abnormal due to reduced insulin secretion, suggesting that ablation of galectin-3 also enhances age-dependent impairment in β-cell functional reserve [102]. Pang et al. also found higher blood glucose and glycated hemoglobin levels in Lgals3−/− versus Lgals3+/+ mice on a standard diet, suggesting that galectin-3 modulates β-cell function [148]. In contrast, decreased body weight, WAT mass, and adipogenic and lipogenic gene expression were observed in 17-month-old Lgals3−/− mice on a standard chow, without any sign of inflammation or abnormal glucose homeostasis [101]. Moreover, Li et al. found that 8-month-old Lgals3−/− mice had better insulin sensitivity and glucose tolerance than Lgals3+/+ animals despite similar body weight, suggesting that (macrophage-derived) galectin-3 impairs insulin action, as also shown by the in vitro finding of direct galactin-3 binding to the insulin receptor and inhibition of downstream signaling [147]. Finally, mice knockouts for galectin-3 showed suppression of intermuscular AT formation during muscle regeneration, consistent with the observation that this lectin promotes adipogenic differentiation of MSCs in muscle, which is a mechanism by which obesity favors the development of sarcopenia [149]. However, response to HFD was also very different among studies, despite the fact that in three out of four of those testing this response the source and background of the Lgals3−/− mice were the same [101,146,147,148]. In particular, mice knockouts for galectin-3 or treated with an inhibitor of this lectin were found to have increased [146,148], unchanged [147,150], or decreased [101] weight gain and WAT mas, associated with increased [146,148] or decreased [150] inflammation and impaired [146,148], unchanged [101], or improved [147] insulin resistance and/or glucose tolerance. However, these contrasting results might be, at least in part, attributed to the use of models of global inhibition of galectin-3, either genetic or pharmacological, and hence the lack of other functions of this lectin, involving the regulation of β-cell function and the immune system [151] and the advanced glycation end-product (AGE)-receptor function exerted by this lectin, which is opposite to that of the receptor for AGEs (RAGE) [152].

2.3.5. Considerations from Animals Studies

The results of the above studies in animals lacking or overexpressing factors known to exert a stimulatory or inhibitory effect on adipogenesis showed that defects in this process translated into a wide range of phenotypes, which could be detected under standard conditions or in response to overfeeding using models of dietary or genetic obesity.
Apart from the opposite results obtained when increasing or decreasing the same factor using different approaches or compared with the in vitro studies, which may be attributed to methodological reasons, the sequence of events linking defective adipogenesis to metabolic derangement through the predominance of the hypertrophic over the hyperplastic WAT response was not consistently observed. In fact, mice showing impaired WAT development had either unchanged or reduced body weight along with a decrease in WAT mass of various extent, which was due to a reduction in adipocyte numbers and/or size. This phenotype resembling human lipodystrophy was protected from dietary or genetic obesity, as body weight gain was less, but the reduced WAT capacity to accommodate excess fat was associated with not only impaired, as expected, but unchanged or even improved insulin sensitivity and glucose tolerance. In some of these studies, the favourable metabolic profile was attributed to various factors, including increased energy expenditure, decreased inflammatory cytokines, increased leptin or decreased resistin levels, or compensation by the liver or muscle. Conversely, in some other instances, a normal WAT mass in the absence of fat diversion to ectopic sites was associated with significant metabolic derangement.
Taken together, these findings highlight the extreme complexity and heterogeneity of defects of adipogenesis, which may end up with different phenotypes in terms of WAT mass and adipocyte number and size, depending on the type of defect and the stage at which the process is blocked or retarded. In addition, the discrepancies between the WAT phenotype and the metabolic profile point to a role for additional factors operating in excess fat disposal.

3. Impaired White Adipose Tissue Remodeling in Aging

Serving as a crucial energy reservoir and hormonal regulator vital for systemic energy metabolism and immune homeostasis, WAT plays an active role in the aging process and the development of age-related illnesses [3,153]. Consistently, interventions aimed at delaying or preventing adipocyte dysfunction have shown potential for extending both health span and lifespan [154,155].

3.1. Significance of Changes in White Adipose Tissue Distribution, Cellular and Lipid Composition, and Molecular Signature in Aging

Appropriate remodeling of WAT with aging is important for maintaining health and function. Research conducted in humans, especially among healthy centenarians, has revealed the critical role of WAT in extreme old age [156]. This tissue secretes various circulating factors, including adiponectin, known for their association with a protective metabolic and anti-inflammatory profile during aging [157,158]. Furthermore, research conducted on mice infected with bacteria has demonstrated that WAT serves as a reservoir for memory T cell populations, facilitating a protective memory response to infections and thereby contributing to the immune system’s defense mechanisms [159]. On the other hand, there is experimental and clinical evidence suggesting that changes in WAT with advancing age may contribute to age-related dysfunctions in other organs and systems, rather than merely serving as a marker of WAT aging (Figure 3).
First of all, aging is associated with the redistribution of WAT. Subcutaneous WAT experiences a general decline as individuals age, particularly in the abdomen and limb region [160,161]. The decrease in both the number and differentiation potential of adipocyte precursors cells (APCs) contributes to this phenomenon [162]. In parallel, there is an accumulation of visceral fat and intramuscular fat. These changes are linked to the development of the metabolic syndrome accompanied by an increased risk of T2D and CVD during aging [163]. Notably, middle-aged mice already display impaired plasticity in sWAT, which could contribute to the early onset of insulin resistance [164]. Furthermore, the accumulation of T cells and B cells, which has been demonstrated as a typical feature of aging in WAT, becomes noticeable from middle age [165]. The process of aging is further linked to changes in the balance of macrophages within AT, favoring a shift towards a pro-inflammatory state. This is indicated by the decline in the relative abundance of M2-like macrophages compared to M1-like macrophages, which is connected to a reduction in the expression of PPARγ [166]. Overall, besides anticipating aging-related changes in other tissues [165], the inflammatory cell signature observed during the physiological aging of WAT is unique and differs from that induced in the context of diet-induced obesity [166].
From a molecular standpoint, targeted proteomic analysis found significant age-related changes in murine WAT, including shifts in lipid and central carbon metabolism, electron transport chain complexes, and inflammation [167]. RNA sequencing of 17 organs confirmed that WAT ages early, with increased expression of certain age-related genes during midlife, preceding similar changes in other organs. This heightened gene expression is associated with immune responses and cytokine-mediated inflammation [165]. WAT was also identified as a significant source of age-associated plasma proteins, potentially accelerating aging throughout the body [165]. It follows that interventions targeting WAT aging may help suppress age-related diseases and prolong the lifespan.
Finally, lipidomic research exploring the alteration of WAT in mice as they age reveals substantial modifications in lipid composition as well. These changes encompass modifications in both phospholipid and sphingolipid profiles within WAT [168,169]. Specifically, as observed in the gonadal WAT of 24-month-old mice, aging leads to elevated levels of phosphatidylethanolamine, phosphatidylcholine, phosphatidylinositol, and ether-phospholipids [169]. Regarding sphingolipids, the increased activity of enzymes involved in ceramide production, such as serine palmitoyl transferase and ceramide synthase, likely contributes to the rise in ceramide species like ceramide 16:0 and 20:0 observed in the WAT of aging mice [170]. The overall levels of sphingomyelin also undergo alterations during aging, with an increase in abundance observed within WAT of rats aged 24 months [170]. Changes in the lipid composition of aging WAT in rodent models have been linked to heightened vulnerability to oxidative stress [171], reduced fluidity of cell membranes [172], and compromised thermogenic capability due to impaired differentiation into beige/brown adipocytes [168]. Significantly, changes in the profiles of phospholipids and sphingolipids detected in WAT of rodents seem to mirror changes observed in human plasma [173,174]. This suggests that the remodeling of lipid composition in WAT during aging substantially contributes to the observed variations in plasma lipid composition [175]. In human studies, elevated plasma concentrations of sphingolipids have been suggested as potential indicators for insulin resistance and Alzheimer’s disease (AD) [176,177]. Moreover, ceramide 16:0 has been singled out as a promising biomarker for aging [178]. Together, these findings suggest that plasma lipid composition may serve as a biomarker of age-related changes in AT and an indicator of healthy or pathological aging.

3.2. Cellular Senescence, Autophagy, and Fibrosis of White Adipose Tissue in Aging

Cellular senescence is characterized by cells entering a state of permanent cell cycle arrest while remaining metabolically active, alive, and resistant to apoptosis [179]. While cellular senescence serves beneficial functions in tissue development, repair processes, and tumor suppression, senescence in adipocytes leads to various dysfunctions. These include compromised lipid handling, abnormal production of adipocytokines, insulin resistance, and hindered adaptive thermogenesis [180].
Senescent adipocytes, aside from their inability to divide, develop what is known as a senescence-associated secretory phenotype (SASP), characterized by the persistent secretion of elevated levels of extracellular matrix-degrading enzymes, pro-inflammatory cytokines, and immune modulators, such as tumor necrosis factor-alpha (TNFα), interleukin (IL)-6, and IL-8 [181]. Thus, SASP plays a role in the development of localized and systemic chronic low-grade inflammation, known as inflammaging, through autocrine and paracrine mechanisms [182,183,184]. Additionally, the capacity of senescent APCs to differentiate into functional mature white, beige, or brown adipocytes decreases with aging [183,185,186,187,188,189]. In particular, the acquisition of a SASP phenotype during the aging process contributes to the inability to induce WAT beiging in response to cold temperatures. This hindrance can be reversed by genetic or pharmacological means aimed at targeting the p38/MAPK-p16Ink4a pathway in aged mouse and human beige progenitor cells [185].
Both sWAT and vWAT WAT exhibit varying correlations with aging-related changes, including cellular senescence. Telomere length, a marker indicating the risk of cellular senescence, consistently appears shorter in sWAT compared to vWAT, implying that sWAT is more susceptible to age-related damage [190]. Recently, a subset of aging-dependent regulatory cells, termed ARCs, was identified in peripheral sWAT of aged mice [191]. These cells, characterized by markers typical of APCs, such as platelet-derived growth factor receptor α (PDGFRα) and Pref-1, also exhibited elevated expression of inflammatory markers, including CD163, Adgre1 (F4/80), and Nuclear Factor Kappa B Subunit 1, with heightened levels observed in the oldest cohort of mice [191]. Notably, ARCs also exhibit elevated PU.1 expression, a transcription factor encoded by the Spi1 gene, which is known to inhibit preadipocyte differentiation by suppressing PPAR-γ [192]. Therefore, in addition to their inability to differentiate, ARCs exhibit a secretory profile that hinders the differentiation of nearby adipose progenitors, ultimately contributing to age-related sWAT loss.
Autophagy, a dysregulated process in senescent cells [193], has shown associations with various features of aging WAT. These include but are not limited to blunted adipogenesis, reduced adipocyte functionality, inflammatory reactions, and fibrosis. However, the precise role of autophagy in regulating lipid metabolism, sustaining energy balance, and impacting insulin sensitivity remains incompletely understood. In contrast to the widespread reduction in autophagic activity associated with aging, which leads to disruptions of homeostasis in several tissues, aged adipocytes exhibit increased autophagy. This augmentation is attributed to diminished levels of Rubicon, a protein known to negatively regulate autophagy [194]. Increased autophagic activity in aged adipocytes suppresses PPARγ by promoting the degradation of Steroid Receptor Coactivator-1 and Transcriptional Intermediary Factor 2, essential coactivators of this master regulator of adipocyte differentiation, lipogenesis, function, and insulin sensitivity [195]. Consistently, a reduction in Rubicon leads to adverse outcomes, such as glucose intolerance and the accumulation of fat in the liver [194].
Another feature of aging WAT is fibrosis, characterized by an excessive deposition of extracellular matrix components. This process disrupts tissue architecture and reduces the WAT’s capacity to accommodate lipids. In general, fibrosis is a signature of unhealthy WAT [196]. Within WAT, myofibroblasts with fibrosis-inducing properties develop from APCs expressing PDGFRα, which typically undergo differentiation into beige adipocytes [197,198]. Recent experimental findings indicate that the fibrogenic-to-adipogenic transition of APCs is compromised in aging WAT. While mature adipocytes from WAT of young mice secrete β-hydroxybutyrate to promote the differentiation of APCs into beige adipocytes and suppress fibrosis, the age-related reduction of the transcription factor PRDM16 is responsible for the decreased secretion of this ketone body by adipocytes [192]. Consequently, this leads to the promotion of WAT fibrosis and the suppression of beige fat formation. Notably, targeting fat cells by increasing PRDM16 expression [192] or by promoting the formation of a complex with the cold-inducible transcription factor GTF2IRD1 [199] is sufficient to enhance systemic glucose homeostasis and WAT insulin sensitivity through the repression of WAT fibrosis. Importantly, these improvements occur regardless of changes in body mass [199].
To summarize, all of these findings indicate that the molecular and structural changes that WAT undergoes during aging, including senescence, abnormal autophagy, and the fibrosis process, play a role in systemic dysfunction, such as inflammaging and glucose homeostasis alterations, rather than merely being a consequence of adipose maladaptation.

3.3. Potential Therapeutic Interventions for Age-Related Adipocyte Dysfunction

The potential to delay the processes that characterize the aging of WAT with drugs capable of correcting age-associated adipocyte dysfunction is an area of increasing interest. Several studies employing emerging or established systemic therapies have produced intriguing results in mitigating age-related changes in the adipose organ (Figure 4), which have been associated with lifespan extension in certain experimental studies.
The removal of senescent cells by using senolytics has been demonstrated to delay age-related decline and the progression of chronic diseases in both animal models and humans [200,201,202,203]. A recent discovery has identified the glycoprotein nonmetastatic melanoma protein B as a key molecular target for senolytic treatment. Indeed, vaccination aimed at this seno-antigen has been shown to diminish senescence in WAT and ameliorate age-related pathological features in mice [204].
Even the experimental use of conventional medications prescribed for managing T2D has shown interesting effects on the aging process of WAT. Biguanides have long been suggested to offer advantages for both health span and life span in experimental research [205]. Interestingly, metformin has been observed to enhance adipogenesis, glucose uptake, and mitochondrial fatty acid oxidation in adipose stromal cells obtained from the sWAT of women aged over 55 years by activating AMP-activated protein kinase [205]. Additional mechanisms for the beneficial effects of metformin on WAT aging are represented by the improvement of PPARγ and sterol regulatory element-binding protein signaling as well as collagen trimerization in WAT [206].
Finally, thiazolidinediones (TZDs) are known to improve WAT and whole-body insulin sensitivity by acting as ligands for PPARγ, thereby promoting adipocyte differentiation, enhancing adipocyte function, and reducing the release of free fatty acids into circulation [207,208]. A recent study has revealed that administering low doses of TZDs led to improvements in age-related decline of WAT. In particular, TZDs were found to decrease inflammation and fibrosis in aging WAT, thereby playing a role in preserving overall AT homeostasis [209].

3.4. White Adipose Tissue Maladaptation in Age-Related Disorders

In addition to the increased cardiometabolic risk, there is evidence suggesting that WAT maladaptation influences typical pathologies affecting the elderly by disrupting metabolic homeostasis and the regenerative capabilities of other organs and tissues [163]. For instance, the accumulation of ectopic fat in various tissues due to the diminished function of WAT, including the muscle, liver, dermis, and bone marrow, is associated with age-related disorders, such as sarcopenia, hepatic steatosis, diminished dermal elasticity, and osteoporosis [187,210,211,212,213]. In the following sections, we will explore WAT maladaptation in common pathologies of the elderly, including osteoarthritis (OA), osteoporosis, cognitive decline and dementia, and cancer cachexia (CC) (Figure 4). We will examine its potential role as both a contributor to and a target of chronic and cancer processes, suggesting it as a therapeutic target to mitigate the progression of frailty.

3.4.1. Osteoarthritis

OA stands as the predominant type of arthritis and is acknowledged as a primary source of pain and disability [214,215]. Its prevalence encompasses 3.3% of the global population, a statistic as of 2015 [216,217], with a tendency to increase with age. Among individuals aged 60 and above, approximately 10% of males and 18% of females experience this condition [218].
The risk of OA is increased among individuals who are overweight. However, emerging evidence suggests that alterations in biomechanical joint loading resulting from higher body mass are insufficient to fully explain the severity of knee OA associated with obesity [214,219,220,221,222]. These findings imply that additional factors associated with dysfunction in AT, primarily WAT, including abnormal release of adipokines, inflammation, and ensuing metabolic disorders, may significantly contribute to this process [214,215,220,223,224]. This topic is of utmost importance, especially considering the current lack of disease-modifying drugs for OA.
To support the hypothesis that WAT contributes to OA, a recent study has demonstrated that transgenic mice exhibiting lipodystrophy, regardless of whether they were fed a standard chow or a high-fat diet, exhibit resistance to both spontaneous and post-traumatic OA. This resistance persists despite the presence of numerous factors formerly associated with OA, including body weight and inflammation. Additionally, the implantation of a mixture of sWAT and vWAT from wild-type mice reintroduced susceptibility to cartilage damage and post-traumatic OA in this preclinical animal model of lipodystrophy [225]. This indicates a direct correlation between WAT and cartilage degradation, irrespective of body weight, and implies that paracrine signaling from WAT contributes to joint degeneration.
Previous and subsequent studies confirm a correlation between AT and joint health, elucidating insights into the natural history of OA and providing clues for treatment. Of interest is the observation that the therapeutic efficacy of injectable WAT-derived products for OA treatment could depend on the health status of the adipose organ and the metabolic condition of the patient [226]. Injectable therapies utilizing WAT-derived products have shown potential in modifying disease outcomes in joint tissues according to preclinical investigations conducted on animal models of OA [227]. However, limited clinical evidence exists for this autologous WAT-derived cell therapy. While some studies suggest safety and benefits in joint function and pain relief [228,229,230], findings from high-quality trials often vary widely, frequently resulting in unsatisfactory clinical outcomes [231,232,233]. The variability in results can be attributed directly to the substantial variances in the quality of WAT utilized in the preparation of injectable products [234]. Dysfunctional WAT from patients with obesity and OA, characterized by senescence, abnormal adipokine production, inflammation, and fibrosis may be the reason for the unsatisfactory clinical outcomes of this cell-based treatment [226]. This suggests that products derived from WAT of patients with obesity may possess inadequate properties for treating OA. Similarly, advancing age has been recognized as a significant risk factor for the ineffectiveness of autologous WAT-derived cell therapy [235], owing to its influence on the paracrine and inflammatory functions of WAT-derived mesenchymal stem cells [236].
Finally, age-related changes in the infrapatellar fat pad may also play a role in knee OA [237,238]. Lower total collagen deposition and elastic fibers around the adipocytes were found in elderly donors compared to young donors [239]. Combined with the finding that the infrapatellar fat pad serves as a significant source of IL-6 and other inflammatory modulators [237] and is affected by fibrotic and vascular changes in OA patients [238], this local WAT depot has attracted attention for its active involvement in joint degeneration. However, further investigation is required to explore the functions of the various WAT depots in OA, particularly concerning the impact of adipose-derived stem cells from both healthy and unhealthy WAT on OA.

3.4.2. Senile Osteoporosis

Osteoporosis is a metabolic bone condition characterized by a gradual decline in bone mass, microstructure, and biomechanical properties. This results in a weakening of bone strength, increasing the likelihood of fractures, particularly from minor impacts (i.e., brittle fractures) [240]. The relation between body fat and the skeleton is as complex as it is contentious. Each type of AT, including BAT, sWAT, and particularly vWAT, has been associated with variations in bone mineral density (BMD) and microarchitecture [241]. On the one hand, obesity-related excess vWAT alters adipokine secretion, increasing pro-inflammatory cytokines that may disrupt bone remodeling [242]. Consistently, an inverse correlation was observed between spinal cortical and trabecular BMD and vWAT content, even after adjusting for BMI [243]. On the other hand, increased mechanical loading resulting from elevated BMI has been linked to higher BMD and improved microstructure, thus positively impacting both cortical and trabecular bone health [244]. To reconcile conflicting results, a U-shaped correlation between visceral WAT and BMD has been proposed [245], thus further increasing the level of complexity.
In addition to WAT and BAT, bone marrow AT (BMAT) has gained recognition as an extra fat reservoir involved in bone-fat interaction. It has been suggested to be associated with BMD loss during aging (i.e., senile osteoporosis) [246,247]. From a morphological and functional perspective, the fat cells constituting the BMAT, with their unilocular appearance and the production of adipokines, such as leptin and adiponectin [248], exhibit characteristics more akin to white adipocytes [249,250]. BMAT could constitute approximately 5% of the total fat mass in adult individuals. Bone marrow adiposity increases with age, occupying about 50% of the bone marrow volume at the age of 25 [251]. It further increases throughout adulthood because of the conversion of bone marrow to BMAT [247,252]. Importantly, while BMAT formation is distinct from that of other fat depots, it is conceivable that BMAT accretion may not be entirely independent of the distribution of WAT, particularly when sWAT redistribution occurs during aging [160,161,253]. Interestingly, the buildup of BMAT is linked to a reduction in bone formation in its vicinity, leading to a decline in bone mechanical strength [254,255].
In senile osteoporosis, bone loss primary results from an altered bone remodeling with either decreased number and/or mineralizing function of osteoblasts, thereby resulting in a reduction in bone mechanical strength and eventually an increased risk of fractures [256]. There is experimental evidence suggesting that WAT, including BMAT, could contribute to bone loss through mechanisms involving lipotoxicity, impairing osteoblast function and survival [257,258,259]. Several animal models of progeria exhibit osteoporosis [260], including the senescence accelerated prone 8 (SAMP8) mouse strain, which mimics clinical features of aging in humans, including senile osteoporosis [261,262]. Interestingly, lifelong dietary supplementation with fish oil, rich in ω3 (n-3) fatty acids, has been shown to partially prevent age-related bone loss in mice [262]. This effect was associated with reduced volume of BMAT [263]. The study did not investigate the mechanisms by which fish oil aids in osteoblast function and bone health, while also inhibiting BMAT expansion. The authors suggest that it may suppress lipotoxicity resulting from age-related BMAT accumulation, in which fatty acid composition, high in saturated fatty acids, is emerging as a key signature of this fat depot compared to other WAT depots [247]. This hypothesis finds partial support in prior research demonstrating fish oil’s ability to reduce PPAR-γ expression, thereby promoting stem cell differentiation towards osteoblastic rather than adipogenic lineages [262].
To conclude, besides the necessity of conducting well-designed clinical trials to acquire robust and conclusive data on the relationship between WAT and bone, further research must be undertaken to validate whether BMAT can truly be regarded as a robust and reliable indicator of bone health and fracture risk when compared to other fat depots. This confirmation could potentially reveal new clinical perspectives in the management of age-related osteoporosis and, possibly, bone loss associated with metabolic conditions [264].

3.4.3. Cognitive Decline and Dementia

As our societies age, the prevalence of dementia worldwide is projected to increase significantly, with estimates suggesting a rise from 55 million individuals affected in 2019 to a staggering 139 million by 2050 [265]. Concurrently, the associated expenses are anticipated to surpass a twofold increase over the same timeframe.
An important interplay exists between AT and the brain. It is well known that the brain influences various aspects of AT function, including lipolysis [266], cytokine production [267], and, through neuroimmune interactions, plays a role in thermoregulation [268]. However, several hints suggest that the reverse may also be true. In particular, a great deal of research indicates that AT influences brain function via adipokines, which can cross the blood-brain barrier to affect target areas or regulate its function. Adipokines play a significant role in various brain functions, including synaptic plasticity, memory formation, neurogenesis, inflammatory responses, and the maintenance of the blood-brain barrier [269]. Consistently, obesity, T2DM, and other metabolic disorders associated with altered adipokine production increase the risk of cognitive impairment and numerous neurodegenerative diseases, including AD [270,271,272,273]. Accordingly, adiposity has been proposed as an independent factor favoring the development of AD [274].
However, as observed with osteoporosis, the connection between obesity and dementia, particularly AD, is intricate and controversial [275]. On one hand, magnetic resonance imaging and computed tomography research revealed brain structure changes in overweight/obese individuals [276,277]. Further studies found a link between visceral WAT and decreased brain volume in middle-aged adults, particularly impacting the temporal lobe and hippocampus size [278]. Additionally, a connection has been established linking obesity, neuroinflammation, autoimmune diseases, such as multiple sclerosis, and neurodegenerative disorders, including AD and Parkinson’s disease [279,280,281,282,283,284,285]. Contrary to these studies, other research suggests that individuals classified as underweight (BMI < 20 kg/m2) show a heightened susceptibility to dementia, while those categorized as severely obese (BMI > 40 kg/m2) have a decreased risk of dementia compared to those with a healthy BMI [275,286]. Interestingly, a more significant decrease in BMI during aging, as well as a low BMI in later life, have been associated with an increased risk of dementia and AD [286]. These results underscore the significance of considering a life-course perspective when examining the relationship between BMI and cognitive function. They also suggest that the inappropriate expression of factors derived from AT, either due to dysfunction in adipocytes caused by body weight issues or the unhealthy remodeling of WAT during aging, could potentially disrupt brain homeostasis and functions.
The role of adipokines in regulating adipose-brain cross-talk and the associated risk of developing cognitive decline and dementia has recently been comprehensively reviewed [269]. However, adipokines are not the sole adipose factor capable of regulating the brain. Preclinical studies have established an association between visceral WAT and impaired memory in obese mice through IL-1 microglial activation, mediated by the NLR family Pyrin Domain Containing 3 [287]. Furthermore, genetically modified mice engineered to release a 20-amino acid peptide called NaKtide in adipocytes demonstrated enhanced hippocampal memory by inhibiting Na,K-ATPase signaling [288]. Notably, recent research in humans has uncovered previously unidentified connections between the transcriptome of WAT and brain function. In particular, RNA sequencing of WAT identified genes linked to cognitive domains in human cohorts. Among them, solute carrier family 18 member A2 (SLC18A2) and regulating synaptic membrane exocytosis 1 (RIMS1) mRNA in WAT were examined for their functional relevance. Briefly, conditional knockdown of SLC18A2 in AT reversed memory impairment induced by HFD in mice. Furthermore, downregulation of Vmat (the Drosophila orthologue of SLC18A2) improved short-term memory, while overexpression of Rim (RIMS1 orthologue) enhanced learning abilities in Drosophila [289]. In the search for accessible biomarkers with prognostic and predictive value, gene expression in peripheral blood mononuclear cells was assessed, revealing that genes associated with cognition in AT (NUDT2, AMPH, UNC5B, OAT, EZR, and NR4A2) exhibited similar associations with cognitive traits across 816 subjects [289]. These findings reinforce the concept of targeting AT as a therapeutic strategy for cognitive dysfunction, by identifying potential biomarkers and clinically relevant therapeutic targets. Delivering miRNAs targeted at AT and influencing cognitive functions appears to offer a safer approach compared to directly targeting the brain.
Unfortunately, this study [289] solely concentrated on individuals with severe obesity (BMI > 35 kg/m2), thereby limiting its generalizability. To bolster these findings and broaden their applicability regarding the association between fat and cognitive dysfunction during aging, further research utilizing larger, longitudinal studies is imperative, especially with regards to older adults.

3.4.4. Cancer Cachexia

In addition to excess fat, conditions characterized by the deficiency of WAT, such as cachexia, are associated with notable metabolic issues including insulin resistance, impaired glucose tolerance, and inflammation [290]. This suggests similar or even identical molecular mechanisms linking obesity and scarcity of WAT with metabolic conditions [291], likely reflecting comparable dysfunctions of WAT in conditions of both extreme abundance and deficiency.
Cachexia is a progressive multifactorial condition marked by the loss of skeletal muscle and adipose depots, which does not respond to nutritional interventions [292]. Wasting can result from a variety of chronic conditions such as kidney disease, heart failure, and HIV infection [293]. Significantly, cachexia affects 30% of patients with cancer, contributing to approximately 20% of cancer-related deaths [294]. Originally believed to be solely a result of the increased energy demands of the tumor, the understanding of CC has evolved to recognize it as the outcome of tumor-secreted factors inducing a cachectic metabolism favoring catabolic processes [295]. Another paradigm that has evolved over time is that, while muscle wasting remains a significant aspect of the neoplastic cachectic state, the depletion of WAT may play a decisive role, particularly after the observation that the reduction in fat mass precedes that of lean mass in the progression of CC [292,296], and the demonstration of a direct link between fat breakdown and muscle wasting [297].
By demonstrating elevated gene expression of uncoupling protein 1 and browning markers in cachectic WAT of murine models of lung, liver, and pancreatic cancers, it has been proposed that browning-induced increased energy expenditure contributes to muscle wasting in CC [290]. In particular, Kir et al. [298] have shown that blocking parathyroidhormone-related protein (PTHrP) on adipose tissue prevented CC in mice treated with the cachexia-inducing Lewis lung cancer cells. Nevertheless, heightened resting energy expenditure is not frequently noted in CC, in either patients or mouse models [299,300]. This may suggest a distinct function of PTHrP in CC, separate from its involvement in stimulating the thermogenic process of adipose tissue.
From another perspective, peculiar changes in fat cell metabolism were described in CC. Generally, the reduction in fat mass induced by nutrient deficiency (e.g., fasting) is associated with an increase in lipolysis and a reduction in lipogenesis [301]. However, CC appears to deviate from this general law of balance between lipogenesis and lipolysis when determining fat loss. In fact, while increased WAT lipolysis has definitely been recognized as a key element in the development of CC, some recent evidence suggests that, contrary to previous findings [302,303], lipogenesis is also increased in CC [300,304]. Regarding lipolysis, the main enzymes responsible for the hydrolysis of triacylglycerols and diacylglycerols, ATGL and HSL, respectively, are increased in patients with CC [297]. In alignment with the pivotal role of lipolysis in CC progression, the ablation of HSL and, particularly, ATGL has been demonstrated to protect mice against the loss of WAT and, to some extent, muscle, even in the presence of unchanged levels of other cachexokines, such as TNF-α and IL-6 [297]. Accordingly, therapies targeting adipocyte lipolysis, particularly the ATGL enzyme, have been proposed for the treatment of CC [305]. However, increased lipolysis is not the only metabolic change affecting the adipocyte in CC, and WAT wasting could actually be a result of more complex metabolic rearrangements. In particular, increased lipogenesis has also been reported [300,304]. Increased triglyceride synthesis, along with a high rate of lipolysis, indicates elevated substrate cycling, implying a rapid turnover of the fat mass and ongoing remodeling of the WAT depots. This process necessitates heightened energy demand, potentially resulting in a net loss of WAT. Consistently, exposure to tumor cells and tumor growth in mice has been shown to trigger a futile energy-depleting cycle in cultured white adipocytes and WAT, respectively, resulting in a reduction of WAT ATP levels by more than 50% [300].
In addition to the findings from preclinical models [297], in vitro studies also indicate that a direct effect of tumor-derived factors, rather than indirect effects, is responsible for cachectic fat loss [305]. Recently, compelling evidence supporting the cancer’s ability to induce WAT wasting by impacting the angiocrine signals of WAT endothelium has been generated [305]. As demonstrated using human and mouse cancer models, tumors alter the transcription of endothelial genes in WAT even in the precachectic state, leading to an overactivation of Notch1 signaling in distant WAT endothelium. This subsequently exacerbates tumor-induced WAT loss. Mechanistically, Notch1 signaling increases the synthesis of retinoic acid and IL-33 in endothelial cells, and pharmacological inhibition of retinoic acid signaling was sufficient to prevent WAT wasting in a mouse CC model [306].
To summarize, the relationship between cancer and WAT in CC has not been fully defined. Recent data indicate the need to direct research efforts towards identifying tumor factors that may regulate WAT remodeling, in order to pinpoint targetable pathways to inhibit the progression of tissue wasting.

4. Conclusions

The adipose organ has a unique ability to adapt and respond to stimuli, such as energy surplus, by remodeling and expanding through hypertrophy and hyperplasia. While enlargement of existing adipocytes (hypertrophy) beyond a certain threshold is detrimental, de novo adipogenesis, i.e., recruitment of new adipocytes from uncommitted precursors (hyperplasia), allows for accommodating large amounts of lipids without compromising metabolic health. Therefore, adipocyte size is considered a marker of subcutaneous WAT hypertrophy and dysfunction and a predictor of increased cardiometabolic risk. However, studies in individuals with obesity and T2D have shown a bimodal cell size distribution in WAT, with about the same proportion of large and small adipocytes, suggesting that a defect may occur at any stage of the process in which MSCs commit to the adipose lineage to become preadipocytes, which then differentiate into adipocytes. The extreme complexity and heterogeneity of defects of adipogenesis may explain the different phenotypes observed in animal models and humans in terms of WAT mass and adipocyte numbers and size. Additional factors play a role in modulating excess fat disposal resulting in the metabolic derangement associated with WAT dysfunction.
Both an excess and scarcity of WAT can pose health threats to the elderly. The remodeling of WAT during aging is a multifaceted process crucial for maintaining metabolic homeostasis involving both its cellular and lipid components. When this process is disrupted, it can precipitate the onset of cardiometabolic conditions and may contribute to several age-related diseases. While significant progress has been achieved in researching adipose organ pathophysiology, there remains a need for a deeper comprehension of how WAT remodeling affects systemic and specific tissue homeostasis during aging. In particular, understanding the mechanisms underlying adipose organ senescence is crucial, as it exhibits signs of aging earlier and is intricately linked to virtually all organs and systems. This understanding holds paramount importance for realizing the actual therapeutic benefits achievable by targeting adipocytes to delay the aging process and mitigate age-related disorders.
More generally, in consideration of the clinical and experimental observations demonstrating comparable metabolic effects resulting from both excess and insufficiency of WAT, likely indicative of shared molecular mechanisms, there arises a necessity to prioritize research endeavors towards the enhancement of WAT quality rather than quantity. This involves supporting the maintenance and development of fully functional adipocytes to foster healthy WAT during aging and address metabolic challenges.

Funding

S.M. acknowledges the support of grant PE00000003 (decree 1550, 11.10.2022) (“ON Foods—Research and innovation network on food and nutrition Sustainability, Safety and Security—Working ON Foods”) from the Italian Ministry of University and Research (CUP D93C22000890001) under the National Recovery and Resilience Plan (NRRP), funded by the European Union—NextGenerationEU; GP acknowledges the support of grant PE0000015 (decree 1550, 11.10.2022) (“Age-It—A novel public-private alliance to generate socioeconomic, biomedical and technological solutions for an inclusive Italian ageing society) from the Italian Ministry of University and Research (CUP B53C22004090006) under the National Recovery and Resilience Plan (NRRP), funded by the European Union—NextGenerationEU.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Ahima, R.S.; Flier, J.S. Adipose tissue as an endocrine organ. Trends Endocrinol. Metab. 2000, 11, 327–332. [Google Scholar] [CrossRef]
  2. Hamdy, O.; Porramatikul, S.; Al-Ozairi, E. Metabolic obesity: The paradox between visceral and subcutaneous fat. Curr. Diabetes Rev. 2006, 2, 367–373. [Google Scholar] [CrossRef]
  3. Corvera, S. Cellular Heterogeneity in Adipose Tissues. Annu. Rev. Physiol. 2021, 83, 257–278. [Google Scholar] [CrossRef]
  4. Jeffery, E.; Wing, A.; Holtrup, B.; Sebo, Z.; Kaplan, J.L.; Saavedra-Peña, R.; Church, C.D.; Colman, L.; Berry, R.; Rodeheffer, M.S. The Adipose Tissue Microenvironment Regulates Depot-Specific Adipogenesis in Obesity. Cell Metab. 2016, 24, 142–150. [Google Scholar] [CrossRef]
  5. Hildreth, A.D.; Ma, F.; Wong, Y.Y.; Sun, R.; Pellegrini, M.; O’Sullivan, T.E. Single-cell sequencing of human white adipose tissue identifies new cell states in health and obesity. Nat. Immunol. 2021, 22, 639–653. [Google Scholar] [CrossRef]
  6. Vijay, J.; Gauthier, M.F.; Biswell, R.L.; Louiselle, D.A.; Johnston, J.J.; Cheung, W.A.; Belden, B.; Pramatarova, A.; Biertho, L.; Gibson, M.; et al. Single-cell analysis of human adipose tissue identifies depot and disease specific cell types. Nat. Metab. 2020, 2, 97–109. [Google Scholar] [CrossRef]
  7. Pan, J.; Yin, J.; Gan, L.; Xue, J. Two-sided roles of adipose tissue: Rethinking the obesity paradox in various human diseases from a new perspective. Obes. Rev. 2023, 24, e13521. [Google Scholar] [CrossRef]
  8. Sakers, A.; De Siqueira, M.K.; Seale, P.; Villanueva, C.J. Adipose-tissue plasticity in health and disease. Cell 2022, 185, 419–446. [Google Scholar] [CrossRef]
  9. Von Bank, H.; Kirsh, C.; Simcox, J. Aging adipose: Depot location dictates age-associated expansion and dysfunction. Ageing Res. Rev. 2021, 67, 101259. [Google Scholar] [CrossRef]
  10. Hammarstedt, A.; Gogg, S.; Hedjazifar, S.; Nerstedt, A.; Smith, U. Impaired Adipogenesis and Dysfunctional Adipose Tissue in Human Hypertrophic Obesity. Physiol. Rev. 2018, 98, 1911–1941. [Google Scholar] [CrossRef]
  11. Krotkiewski, M.; Björntorp, P.; Sjöström, L.; Smith, U. Impact of obesity on metabolism in men and women. Importance of regional adipose tissue distribution. J. Clin. Investig. 1983, 72, 1150–1162. [Google Scholar] [CrossRef] [PubMed]
  12. Spalding, K.L.; Arner, E.; Westermark, P.O.; Bernard, S.; Buchholz, B.A.; Bergmann, O.; Blomqvist, L.; Hoffstedt, J.; Näslund, E.; Britton, T.; et al. Dynamics of fat cell turnover in humans. Nature 2008, 453, 783–787. [Google Scholar] [CrossRef] [PubMed]
  13. Giordano, A.; Murano, I.; Mondini, E.; Perugini, J.; Smorlesi, A.; Severi, I.; Barazzoni, R.; Scherer, P.E.; Cinti, S. Obese adipocytes show ultrastructural features of stressed cells and die of pyroptosis. J. Lipid Res. 2013, 54, 2423–2436. [Google Scholar] [CrossRef] [PubMed]
  14. Cinti, S. Adipose Organ Development and Remodeling. Compr. Physiol. 2018, 8, 1357–1431. [Google Scholar] [CrossRef] [PubMed]
  15. Smith, U. Abdominal obesity: A marker of ectopic fat accumulation. J. Clin. Investig. 2015, 125, 1790–1792. [Google Scholar] [CrossRef] [PubMed]
  16. Lim, S.; Meigs, J.B. Ectopic fat and cardiometabolic and vascular risk. Int. J. Cardiol. 2013, 169, 166–176. [Google Scholar] [CrossRef]
  17. Zhao, J.Y.; Zhou, L.J.; Ma, K.L.; Hao, R.; Li, M. MHO or MUO? White adipose tissue remodeling. Obes. Rev. 2024, 25, e13691. [Google Scholar] [CrossRef]
  18. Iacobini, C.; Pugliese, G.; Blasetti Fantauzzi, C.; Federici, M.; Menini, S. Metabolically healthy versus metabolically unhealthy obesity. Metabolism 2019, 92, 51–60. [Google Scholar] [CrossRef]
  19. Agius, R.; Pace, N.P.; Fava, S. Phenotyping obesity: A focus on metabolically healthy obesity and metabolically unhealthy normal weight. Diabetes Metab. Res. Rev. 2024, 40, e3725. [Google Scholar] [CrossRef]
  20. Pellegrinelli, V.; Carobbio, S.; Vidal-Puig, A. Adipose tissue plasticity: How fat depots respond differently to pathophysiological cues. Diabetologia 2016, 59, 1075–1088. [Google Scholar] [CrossRef]
  21. Twig, G.; Afek, A.; Derazne, E.; Tzur, D.; Cukierman-Yaffe, T.; Gerstein, H.C.; Tirosh, A. Diabetes risk among overweight and obese metabolically healthy young adults. Diabetes Care 2014, 37, 2989–2995. [Google Scholar] [CrossRef]
  22. Opio, J.; Croker, E.; Odongo, G.S.; Attia, J.; Wynne, K.; McEvoy, M. Metabolically healthy overweight/obesity are associated with increased risk of cardiovascular disease in adults.; even in the absence of metabolic risk factors: A systematic review and meta-analysis of prospective cohort studies. Obes. Rev. 2020, 21, e13127. [Google Scholar] [CrossRef]
  23. Carobbio, S.; Pellegrinelli, V.; Vidal-Puig, A. Adipose Tissue Function and Expandability as Determinants of Lipotoxicity and the Metabolic Syndrome. Adv. Exp. Med. Biol. 2017, 960, 161–196. [Google Scholar] [CrossRef] [PubMed]
  24. Laforest, S.; Labrecque, J.; Michaud, A.; Cianflone, K.; Tchernof, A. Adipocyte size as a determinant of metabolic disease and adipose tissue dysfunction. Crit. Rev. Clin. Lab. Sci. 2015, 52, 301–313. [Google Scholar] [CrossRef]
  25. Stenkula, K.G.; Erlanson-Albertsson, C. Adipose cell size: Importance in health and disease. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2018, 315, R284–R295. [Google Scholar] [CrossRef]
  26. Skurk, T.; Alberti-Huber, C.; Herder, C.; Hauner, H. Relationship between adipocyte size and adipokine expression and secretion. J. Clin. Endocrinol. Metab. 2007, 92, 1023–1033. [Google Scholar] [CrossRef] [PubMed]
  27. Landgraf, K.; Rockstroh, D.; Wagner, I.V.; Weise, S.; Tauscher, R.; Schwartze, J.T.; Löffler, D.; Bühligen, U.; Wojan, M.; Till, H.; et al. Evidence of early alterations in adipose tissue biology and function and its association with obesity-related inflammation and insulin resistance in children. Diabetes 2015, 64, 1249–1261. [Google Scholar] [CrossRef] [PubMed]
  28. Joffe, B.I.; Goldberg, R.B.; Feinstein, J.; Kark, A.; Seftel, H.C. Adipose cell size in obese Africans: Evidence against the existence of insulin resistance in some patients. J. Clin. Pathol. 1979, 32, 471–474. [Google Scholar] [CrossRef]
  29. Weyer, C.; Foley, J.E.; Bogardus, C.; Tataranni, P.A.; Pratley, R.E. Enlarged subcutaneous abdominal adipocyte size, but not obesity itself, predicts type II diabetes independent of insulin resistance. Diabetologia 2000, 43, 1498–1506. [Google Scholar] [CrossRef]
  30. Lönn, M.; Mehlig, K.; Bengtsson, C.; Lissner, L. Adipocyte size predicts incidence of type 2 diabetes in women. FASEB J. 2010, 24, 326–331. [Google Scholar] [CrossRef]
  31. Acosta, J.R.; Douagi, I.; Andersson, D.P.; Bäckdahl, J.; Rydén, M.; Arner, P.; Laurencikiene, J. Increased fat cell size: A major phenotype of subcutaneous white adipose tissue in non-obese individuals with type 2 diabetes. Diabetologia 2016, 59, 560–570. [Google Scholar] [CrossRef] [PubMed]
  32. Arner, P.; Arner, E.; Hammarstedt, A.; Smith, U. Genetic predisposition for Type 2 diabetes, but not for overweight/obesity, is associated with a restricted adipogenesis. PLoS ONE 2011, 6, e18284. [Google Scholar] [CrossRef]
  33. Yang, J.; Eliasson, B.; Smith, U.; Cushman, S.W.; Sherman, A.S. The size of large adipose cells is a predictor of insulin resistance in first-degree relatives of type 2 diabetic patients. Obesity 2012, 20, 932–938. [Google Scholar] [CrossRef]
  34. Roberts, R.; Hodson, L.; Dennis, A.L.; Neville, M.J.; Humphreys, S.M.; Harnden, K.E.; Micklem, K.J.; Frayn, K.N. Markers of de novo lipogenesis in adipose tissue: Associations with small adipocytes and insulin sensitivity in humans. Diabetologia 2009, 52, 882–890. [Google Scholar] [CrossRef] [PubMed]
  35. Cotillard, A.; Poitou, C.; Torcivia, A.; Bouillot, J.L.; Dietrich, A.; Klöting, N.; Grégoire, C.; Lolmede, K.; Blüher, M.; Clément, K. Adipocyte size threshold matters: Link with risk of type 2 diabetes and improved insulin resistance after gastric bypass. J. Clin. Endocrinol. Metab. 2014, 99, E1466–E1470. [Google Scholar] [CrossRef]
  36. Rydén, M.; Andersson, D.P.; Bergström, I.B.; Arner, P. Adipose tissue and metabolic alterations: Regional differences in fat cell size and number matter, but differently: A cross-sectional study. J. Clin. Endocrinol. Metab. 2014, 99, E1870–E1876. [Google Scholar] [CrossRef] [PubMed]
  37. Hoffstedt, J.; Arner, E.; Wahrenberg, H.; Andersson, D.P.; Qvisth, V.; Löfgren, P.; Rydén, M.; Thörne, A.; Wirén, M.; Palmér, M.; et al. Regional impact of adipose tissue morphology on the metabolic profile in morbid obesity. Diabetologia 2010, 53, 2496–2503. [Google Scholar] [CrossRef]
  38. Henninger, A.M.; Eliasson, B.; Jenndahl, L.E.; Hammarstedt, A. Adipocyte hypertrophy.; inflammation and fibrosis characterize subcutaneous adipose tissue of healthy, non-obese subjects predisposed to type 2 diabetes. PLoS ONE 2014, 9, e105262. [Google Scholar] [CrossRef]
  39. Imbeault, P.; Lemieux, S.; Prud’homme, D.; Tremblay, A.; Nadeau, A.; Després, J.P.; Mauriège, P. Relationship of visceral adipose tissue to metabolic risk factors for coronary heart disease: Is there a contribution of subcutaneous fat cell hypertrophy? Metabolism 1999, 48, 355–362. [Google Scholar] [CrossRef]
  40. Heinonen, S.; Saarinen, L.; Naukkarinen, J.; Rodríguez, A.; Frühbeck, G.; Hakkarainen, A.; Lundbom, J.; Lundbom, N.; Vuolteenaho, K.; Moilanen, E.; et al. Adipocyte morphology and implications for metabolic derangements in acquired obesity. Int. J. Obes. 2014, 38, 1423–1431. [Google Scholar] [CrossRef]
  41. Lessard, J.; Laforest, S.; Pelletier, M.; Leboeuf, M.; Blackburn, L.; Tchernof, A. Low abdominal subcutaneous preadipocyte adipogenesis is associated with visceral obesity.; visceral adipocyte hypertrophy, and a dysmetabolic state. Adipocyte 2014, 3, 197–205. [Google Scholar] [CrossRef] [PubMed]
  42. Belligoli, A.; Compagnin, C.; Sanna, M.; Favaretto, F.; Fabris, R.; Busetto, L.; Foletto, M.; Dal Prà, C.; Serra, R.; Prevedello, L.; et al. Characterization of subcutaneous and omental adipose tissue in patients with obesity and with different degrees of glucose impairment. Sci. Rep. 2019, 9, 11333. [Google Scholar] [CrossRef] [PubMed]
  43. Arner, E.; Westermark, P.O.; Spalding, K.L.; Britton, T.; Rydén, M.; Frisén, J.; Bernard, S.; Arner, P. Adipocyte turnover: Relevance to human adipose tissue morphology. Diabetes 2010, 59, 105–109. [Google Scholar] [CrossRef] [PubMed]
  44. Ledoux, S.; Coupaye, M.; Essig, M.; Msika, S.; Roy, C.; Queguiner, I.; Clerici, C.; Larger, E. Traditional anthropometric parameters still predict metabolic disorders in women with severe obesity. Obesity 2010, 18, 1026–1032. [Google Scholar] [CrossRef] [PubMed]
  45. Veilleux, A.; Caron-Jobin, M.; Noël, S.; Laberge, P.Y.; Tchernof, A. Visceral adipocyte hypertrophy is associated with dyslipidemia independent of body composition and fat distribution in women. Diabetes 2011, 60, 1504–1511. [Google Scholar] [CrossRef] [PubMed]
  46. Lundgren, M.; Svensson, M.; Lindmark, S.; Renström, F.; Ruge, T.; Eriksson, J.W. Fat cell enlargement is an independent marker of insulin resistance and ‘hyperleptinaemia’. Diabetologia 2007, 50, 625–633. [Google Scholar] [CrossRef] [PubMed]
  47. Meena, V.P.; Seenu, V.; Sharma, M.C.; Mallick, S.R.; Bhalla, A.S.; Gupta, N.; Mohan, A.; Guleria, R.; Pandey, R.M.; Luthra, K.; et al. Relationship of adipocyte size with adiposity and metabolic risk factors in Asian Indians. PLoS ONE 2014, 9, e108421. [Google Scholar] [CrossRef] [PubMed]
  48. McLaughlin, T.; Sherman, A.; Tsao, P.; Gonzalez, O.; Yee, G.; Lamendola, C.; Reaven, G.M.; Cushman, S.W. Enhanced proportion of small adipose cells in insulin-resistant vs insulin-sensitive obese individuals implicates impaired adipogenesis. Diabetologia 2007, 50, 1707–1715. [Google Scholar] [CrossRef] [PubMed]
  49. McLaughlin, T.; Deng, A.; Yee, G.; Lamendola, C.; Reaven, G.; Tsao, P.S.; Cushman, S.W.; Sherman, A. Inflammation in subcutaneous adipose tissue: Relationship to adipose cell size. Diabetologia 2010, 53, 369–377. [Google Scholar] [CrossRef]
  50. McLaughlin, T.; Lamendola, C.; Coghlan, N.; Liu, T.C.; Lerner, K.; Sherman, A.; Cushman, S.W. Subcutaneous adipose cell size and distribution: Relationship to insulin resistance and body fat. Obesity 2014, 22, 673–680. [Google Scholar] [CrossRef]
  51. Pasarica, M.; Xie, H.; Hymel, D.; Bray, G.; Greenway, F.; Ravussin, E.; Smith, S.R. Lower total adipocyte number but no evidence for small adipocyte depletion in patients with type 2 diabetes. Diabetes Care 2009, 32, 900–902. [Google Scholar] [CrossRef] [PubMed]
  52. Fang, L.; Guo, F.; Zhou, L.; Stahl, R.; Grams, J. The cell size and distribution of adipocytes from subcutaneous and visceral fat is associated with type 2 diabetes mellitus in humans. Adipocyte 2015, 4, 273–279. [Google Scholar] [CrossRef] [PubMed]
  53. Johannsen, D.L.; Tchoukalova, Y.; Tam, C.S.; Covington, J.D.; Xie, W.; Schwarz, J.M.; Bajpeyi, S.; Ravussin, E. Effect of 8 weeks of overfeeding on ectopic fat deposition and insulin sensitivity: Testing the “adipose tissue expandability” hypothesis. Diabetes Care 2014, 37, 2789–2797. [Google Scholar] [CrossRef] [PubMed]
  54. McLaughlin, T.; Craig, C.; Liu, L.F.; Perelman, D.; Allister, C.; Spielman, D.; Cushman, S.W. Adipose Cell Size and Regional Fat Deposition as Predictors of Metabolic Response to Overfeeding in Insulin-Resistant and Insulin-Sensitive Humans. Diabetes 2016, 65, 1245–1254. [Google Scholar] [CrossRef]
  55. White, U.; Ravussin, E. Dynamics of adipose tissue turnover in human metabolic health and disease. Diabetologia 2019, 62, 17–23. [Google Scholar] [CrossRef] [PubMed]
  56. Menini, S.; Iacobini, C.; Blasetti Fantauzzi, C.; Pesce, C.M.; Pugliese, G. Role of Galectin-3 in Obesity and Impaired Glucose Homeostasis. Oxid. Med. Cell Longev. 2016, 2016, 9618092. [Google Scholar] [CrossRef] [PubMed]
  57. Tang, Q.Q.; Lane, M.D. Adipogenesis: From stem cell to adipocyte. Annu. Rev. Biochem. 2012, 81, 715–736. [Google Scholar] [CrossRef] [PubMed]
  58. Ambele, M.A.; Dhanraj, P.; Giles, R.; Pepper, M.S. Adipogenesis: A Complex Interplay of Multiple Molecular Determinants and Pathways. Int. J. Mol. Sci. 2020, 21, 4283. [Google Scholar] [CrossRef] [PubMed]
  59. Shepherd, P.R.; Gnudi, L.; Tozzo, E.; Yang, H.; Leach, F.; Kahn, B.B. Adipose cell hyperplasia and enhanced glucose disposal in transgenic mice overexpressing GLUT4 selectively in adipose tissue. J. Biol. Chem. 1993, 268, 22243–22246. [Google Scholar] [CrossRef]
  60. Tang, Q.Q.; Otto, T.C.; Lane, M.D. Commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineage. Proc. Natl. Acad. Sci. USA 2004, 101, 9607–9711. [Google Scholar] [CrossRef]
  61. Huang, H.; Song, T.J.; Li, X.; Hu, L.; He, Q.; Liu, M.; Lane, M.D.; Tang, Q.Q. BMP signaling pathway is required for commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineage. Proc. Natl. Acad. Sci. USA 2009, 106, 12670–12675. [Google Scholar] [CrossRef] [PubMed]
  62. Bowers, R.R.; Lane, M.D. Wnt signaling and adipocyte lineage commitment. Cell Cycle 2008, 7, 1191–1196. [Google Scholar] [CrossRef] [PubMed]
  63. Ross, S.E.; Hemati, N.; Longo, K.A.; Bennett, C.N.; Lucas, P.C.; Erickson, R.L.; MacDougald, O.A. Inhibition of adipogenesis by Wnt signaling. Science 2000, 289, 950–953. [Google Scholar] [CrossRef] [PubMed]
  64. Kang, S.; Bennett, C.N.; Gerin, I.; Rapp, L.A.; Hankenson, K.D.; Macdougald, O.A. Wnt signaling stimulates osteoblastogenesis of mesenchymal precursors by suppressing CCAAT/enhancer-binding protein alpha and peroxisome proliferator-activated receptor gamma. J. Biol. Chem. 2007, 282, 14515–14524. [Google Scholar] [CrossRef] [PubMed]
  65. Spinella-Jaegle, S.; Rawadi, G.; Kawai, S.; Gallea, S.; Faucheu, C.; Mollat, P.; Courtois, B.; Bergaud, B.; Ramez, V.; Blanchet, A.M.; et al. Sonic hedgehog increases the commitment of pluripotent mesenchymal cells into the osteoblastic lineage and abolishes adipocytic differentiation. J. Cell Sci. 2001, 114, 2085–2094. [Google Scholar] [CrossRef] [PubMed]
  66. Suh, J.M.; Gao, X.; McKay, J.; McKay, R.; Salo, Z.; Graff, J.M. Hedgehog signaling plays a conserved role in inhibiting fat formation. Cell Metab. 2006, 3, 25–34. [Google Scholar] [CrossRef] [PubMed]
  67. Blázquez-Medela, A.M.; Jumabay, M.; Boström, K.I. Beyond the bone: Bone morphogenetic protein signaling in adipose tissue. Obes. Rev. 2019, 20, 648–658. [Google Scholar] [CrossRef] [PubMed]
  68. Huang, H.Y.; Hu, L.L.; Song, T.J.; Li, X.; He, Q.; Sun, X.; Li, Y.M.; Lu, H.J.; Yang, P.Y.; Tang, Q.Q. Involvement of cytoskeleton-associated proteins in the commitment of C3H10T1/2 pluripotent stem cells to adipocyte lineage induced by BMP2/4. Mol. Cell Proteomics 2011, 10, M110.002691. [Google Scholar] [CrossRef] [PubMed]
  69. Kikuchi, A.; Yamamoto, H.; Sato, A. Selective activation mechanisms of Wnt signaling pathways. Trends Cell Biol. 2009, 19, 119–129. [Google Scholar] [CrossRef]
  70. Li, S.N.; Wu, J.F. TGF-β/SMAD signaling regulation of mesenchymal stem cells in adipocyte commitment. Stem Cell Res. Ther. 2020, 11, 41. [Google Scholar] [CrossRef]
  71. Rosen, E.D. New drugs from fat bugs? Cell Metab. 2006, 3, 1–2. [Google Scholar] [CrossRef] [PubMed]
  72. Tang, Q.Q.; Otto, T.C.; Lane, M.D. Mitotic clonal expansion: A synchronous process required for adipogenesis. Proc. Natl. Acad. Sci. USA 2003, 100, 44–49. [Google Scholar] [CrossRef] [PubMed]
  73. Denechaud, P.D.; Fajas, L.; Giralt, A. E2F1, a Novel Regulator of Metabolism. Front. Endocrinol. 2017, 8, 311. [Google Scholar] [CrossRef] [PubMed]
  74. Fox, K.E.; Colton, L.A.; Erickson, P.F.; Friedman, J.E.; Cha, H.C.; Keller, P.; MacDougald, O.A.; Klemm, D.J. Regulation of cyclin D1 and Wnt10b gene expression by cAMP-responsive element-binding protein during early adipogenesis involves differential promoter methylation. J. Biol. Chem. 2008, 283, 35096–35105. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, J.W.; Klemm, D.J.; Vinson, C.; Lane, M.D. Role of CREB in transcriptional regulation of CCAAT/enhancer-binding protein beta gene during adipogenesis. J. Biol. Chem. 2004, 279, 4471–4478. [Google Scholar] [CrossRef] [PubMed]
  76. Tang, Q.Q.; Grønborg, M.; Huang, H.; Kim, J.W.; Otto, T.C.; Pandey, A.; Lane, M.D. Sequential phosphorylation of CCAAT enhancer-binding protein beta by MAPK and glycogen synthase kinase 3beta is required for adipogenesis. Proc. Natl. Acad. Sci. USA 2005, 102, 9766–9771. [Google Scholar] [CrossRef] [PubMed]
  77. Kim, J.W.; Tang, Q.Q.; Li, X.; Lane, M.D. Effect of phosphorylation and S-S bond-induced dimerization on DNA binding and transcriptional activation by C/EBPbeta. Proc. Natl. Acad. Sci. USA 2007, 104, 1800–1804. [Google Scholar] [CrossRef] [PubMed]
  78. Tang, Q.Q.; Otto, T.C.; Lane, M.D. CCAAT/enhancer-binding protein beta is required for mitotic clonal expansion during adipogenesis. Proc. Natl. Acad. Sci. USA 2003, 100, 850–855. [Google Scholar] [CrossRef]
  79. Yeh, W.C.; Cao, Z.; Classon, M.; McKnight, S.L. Cascade regulation of terminal adipocyte differentiation by three members of the C/EBP family of leucine zipper proteins. Genes Dev. 1995, 9, 168–181. [Google Scholar] [CrossRef]
  80. da Silva, C.; Durandt, C.; Kallmeyer, K.; Ambele, M.A.; Pepper, M.S. The Role of Pref-1 during Adipogenic Differentiation: An Overview of Suggested Mechanisms. Int. J. Mol. Sci. 2020, 21, 4104. [Google Scholar] [CrossRef]
  81. Wu, Z.; Bucher, N.L.; Farmer, S.R. Induction of peroxisome proliferator-activated receptor gamma during the conversion of 3T3 fibroblasts into adipocytes is mediated by C/EBPbeta, C/EBPdelta, and glucocorticoids. Mol. Cell Biol. 1996, 16, 4128–4136. [Google Scholar] [CrossRef]
  82. Christy, R.J.; Kaestner, K.H.; Geiman, D.E.; Lane, M.D. CCAAT/enhancer binding protein gene promoter: Binding of nuclear factors during differentiation of 3T3-L1 preadipocytes. Proc. Natl. Acad. Sci. USA 1991, 88, 2593–2597. [Google Scholar] [CrossRef] [PubMed]
  83. Wu, Z.; Rosen, E.D.; Brun, R.; Hauser, S.; Adelmant, G.; Troy, A.E.; McKeon, C.; Darlington, G.J.; Spiegelman, B.M. Cross-regulation of C/EBP alpha and PPAR gamma controls the transcriptional pathway of adipogenesis and insulin sensitivity. Mol. Cell 1999, 3, 151–158. [Google Scholar] [CrossRef] [PubMed]
  84. Clarke, S.L.; Robinson, C.E.; Gimble, J.M. CAAT/enhancer binding proteins directly modulate transcription from the peroxisome proliferator-activated receptor gamma 2 promoter. Biochem. Biophys. Res. Commun. 1997, 240, 99–103. [Google Scholar] [CrossRef] [PubMed]
  85. Rosen, E.D.; Sarraf, P.; Troy, A.E.; Bradwin, G.; Moore, K.; Milstone, D.S.; Spiegelman, B.M.; Mortensen, R.M. PPAR gamma is required for the differentiation of adipose tissue in vivo and in vitro. Mol. Cell 1999, 4, 611–617. [Google Scholar] [CrossRef] [PubMed]
  86. Rosen, E.D.; Hsu, C.H.; Wang, X.; Sakai, S.; Freeman, M.W.; Gonzalez, F.J.; Spiegelman, B.M. C/EBPalpha induces adipogenesis through PPARgamma: A unified pathway. Genes Dev. 2002, 16, 22–26. [Google Scholar] [CrossRef]
  87. Rosen, E.D.; Spiegelman, B.M. Molecular regulation of adipogenesis. Annu. Rev. Cell Dev. Biol. 2000, 16, 145–171. [Google Scholar] [CrossRef]
  88. Porse, B.T.; Pedersen, T.A.; Xu, X.; Lindberg, B.; Wewer, U.M.; Friis-Hansen, L.; Nerlov, C. E2F repression by C/EBPalpha is required for adipogenesis and granulopoiesis in vivo. Cell 2001, 107, 247–258. [Google Scholar] [CrossRef] [PubMed]
  89. Kuri-Harcuch, W.; Velez-delValle, C.; Vazquez-Sandoval, A.; Hernández-Mosqueira, C.; Fernandez-Sanchez, V. A cellular perspective of adipogenesis transcriptional regulation. J. Cell Physiol. 2019, 234, 1111–1129. [Google Scholar] [CrossRef]
  90. Eberlé, D.; Hegarty, B.; Bossard, P.; Ferré, P.; Foufelle, F. SREBP transcription factors: Master regulators of lipid homeostasis. Biochimie 2004, 86, 839–848. [Google Scholar] [CrossRef]
  91. Kim, J.B.; Wright, H.M.; Wright, M.; Spiegelman, B.M. ADD1/SREBP1 activates PPARgamma through the production of endogenous ligand. Proc. Natl. Acad. Sci. USA 1998, 95, 4333–4337. [Google Scholar] [CrossRef] [PubMed]
  92. Ahmadian, M.; Wang, Y.; Sul, H.S. Lipolysis in adipocytes. Int. J. Biochem. Cell Biol. 2010, 42, 555–559. [Google Scholar] [CrossRef] [PubMed]
  93. Forest, C.; Tordjman, J.; Glorian, M.; Duplus, E.; Chauvet, G.; Quette, J.; Beale, E.G.; Antoine, B. Fatty acid recycling in adipocytes: A role for glyceroneogenesis and phosphoenolpyruvate carboxykinase. Biochem. Soc. Trans. 2003, 31, 1125–1129. [Google Scholar] [CrossRef]
  94. Furuhashi, M.; Hotamisligil, G.S. Fatty acid-binding proteins: Role in metabolic diseases and potential as drug targets. Nat. Rev. Drug Discov. 2008, 7, 489–503. [Google Scholar] [CrossRef] [PubMed]
  95. Fajas, L.; Landsberg, R.L.; Huss-Garcia, Y.; Sardet, C.; Lees, J.A.; Auwerx, J. E2Fs regulate adipocyte differentiation. Dev. Cell 2002, 3, 39–49. [Google Scholar] [CrossRef] [PubMed]
  96. Chen, P.L.; Riley, D.J.; Chen, Y.; Lee, W.H. Retinoblastoma protein positively regulates terminal adipocyte differentiation through direct interaction with C/EBPs. Genes Dev. 1996, 10, 2794–2804. [Google Scholar] [CrossRef] [PubMed]
  97. Fajas, L.; Egler, V.; Reiter, R.; Hansen, J.; Kristiansen, K.; Debril, M.B.; Miard, S.; Auwerx, J. The retinoblastoma-histone deacetylase 3 complex inhibits PPARgamma and adipocyte differentiation. Dev. Cell 2002, 3, 903–910. [Google Scholar] [CrossRef] [PubMed]
  98. Ali, A.T.; Hochfeld, W.E.; Myburgh, R.; Pepper, M.S. Adipocyte and adipogenesis. Eur. J. Cell Biol. 2013, 92, 229–236. [Google Scholar] [CrossRef] [PubMed]
  99. Pollak, N.M.; Hoffman, M.; Goldberg, I.J.; Drosatos, K. Krüppel-like factors: Crippling and un-crippling metabolic pathways. JACC Basic Transl. Sci. 2018, 3, 132–156. [Google Scholar] [CrossRef]
  100. Baek, J.H.; Kim, D.H.; Lee, J.; Kim, S.J.; Chun, K.H. Galectin-1 accelerates high-fat diet-induced obesity by activation of peroxisome proliferator-activated receptor gamma (PPARγ) in mice. Cell Death Dis. 2021, 12, 66. [Google Scholar] [CrossRef]
  101. Baek, J.H.; Kim, S.J.; Kang, H.G.; Lee, H.-W.; Kim, J.H.; Hwang, K.A.; Song, J.; Chun, K.H. Galectin-3 activates PPARγ and supports white adipose tissue formation and high-fat diet-induced obesity. Endocrinology 2015, 156, 147–156. [Google Scholar] [CrossRef]
  102. Blasetti Fantauzzi, C.; Iacobini, C.; Menini, S.; Vitale, M.; Sorice, G.P.; Mezza, T.; Cinti, S.; Giaccari, A.; Pugliese, G. Galectin-3 gene deletion results in defective adipose tissue maturation and impaired insulin sensitivity and glucose homeostasis. Sci. Rep. 2020, 10, 20070. [Google Scholar] [CrossRef] [PubMed]
  103. Yang, R.Y.; Hsu, D.K.; Yu, L.; Chen, H.Y.; Liu, F.T. Galectin-12 is required for adipogenic signaling and adipocyte differentiation. J. Biol. Chem. 2004, 279, 29761–29766. [Google Scholar] [CrossRef]
  104. Kiwaki, K.; Novak, C.M.; Hsu, D.K.; Liu, F.T.; Levine, J.A. Galectin-3 stimulates preadipocyte proliferation and is up-regulated in growing adipose tissue. Obesity 2007, 15, 32–39. [Google Scholar] [CrossRef]
  105. Yang, R.Y.; Hsu, D.K.; Yu, L.; Ni, J.; Liu, F.T. Cell cycle regulation by galectin-12.; a new member of the galectin superfamily. J. Biol. Chem. 2001, 276, 20252–20260. [Google Scholar] [CrossRef] [PubMed]
  106. Yang, R.Y.; Yu, L.; Graham, J.L.; Hsu, D.K.; Lloyd, K.C.; Havel, P.J.; Liu, F.T. Ablation of a galectin preferentially expressed in adipocytes increases lipolysis, reduces adiposity, and improves insulin sensitivity in mice. Proc. Natl. Acad. Sci. USA 2011, 108, 18696–18701. [Google Scholar] [CrossRef] [PubMed]
  107. Hotta, K.; Funahashi, T.; Matsukawa, Y.; Takahashi, M.; Nishizawa, H.; Kishida, K.; Matsuda, M.; Kuriyama, H.; Kihara, S.; Nakamura, T.; et al. Galectin-12, an Adipose-expressed Galectin-like Molecule Possessing Apoptosis-inducing Activity. J. Biol. Chem. 2001, 276, 34089–34097. [Google Scholar] [CrossRef]
  108. Qian, S.W.; Tang, Y.; Li, X.; Liu, Y.; Zhang, Y.Y.; Huang, H.Y.; Xue, R.D.; Yu, H.Y.; Guo, L.; Gao, H.D.; et al. BMP4-mediated brown fat-like changes in white adipose tissue alter glucose and energy homeostasis. Proc. Natl. Acad. Sci. USA 2013, 110, E798–E807. [Google Scholar] [CrossRef]
  109. Soofi, A.; Wolf, K.I.; Emont, M.P.; Qi, N.; Martinez-Santibanez, G.; Grimley, E.; Ostwani, W.; Dressler, G.R. The kielin/chordin-like protein (KCP) attenuates high-fat diet-induced obesity and metabolic syndrome in mice. J. Biol. Chem. 2017, 292, 9051–9062. [Google Scholar] [CrossRef]
  110. Brown, M.L.; Bonomi, L.; Ungerleider, N.; Zina, J.; Kimura, F.; Mukherjee, A.; Sidis, Y.; Schneyer, A. Follistatin and follistatin like-3 differentially regulate adiposity and glucose homeostasis. Obesity 2011, 19, 1940–1949. [Google Scholar] [CrossRef]
  111. Longo, K.A.; Wright, W.S.; Kang, S.; Gerin, I.; Chiang, S.H.; Lucas, P.C.; Opp, M.R.; MacDougald, O.A. Wnt10b inhibits development of white and brown adipose tissues. J. Biol. Chem. 2004, 279, 35503–35509. [Google Scholar] [CrossRef]
  112. Wright, W.S.; Longo, K.A.; Dolinsky, V.W.; Gerin, I.; Kang, S.; Bennett, C.N.; Chiang, S.H.; Prestwich, T.C.; Gress, C.; Burant, C.F.; et al. Wnt10b inhibits obesity in ob/ob and agouti mice. Diabetes 2007, 56, 295–303. [Google Scholar] [CrossRef]
  113. Chen, M.; Lu, P.; Ma, Q.; Cao, Y.; Chen, N.; Li, W.; Zhao, S.; Chen, B.; Shi, J.; Sun, Y.; et al. CTNNB1/β-catenin dysfunction contributes to adiposity by regulating the cross-talk of mature adipocytes and preadipocytes. Sci. Adv. 2020, 6, eaax9605. [Google Scholar] [CrossRef]
  114. Bagchi, D.P.; Li, Z.; Corsa, C.A.; Hardij, J.; Mori, H.; Learman, B.S.; Lewis, K.T.; Schill, R.L.; Romanelli, S.M.; MacDougald, O.A. Wntless regulates lipogenic gene expression in adipocytes and protects against diet-induced metabolic dysfunction. Mol. Metab. 2020, 39, 100992. [Google Scholar] [CrossRef] [PubMed]
  115. Mori, H.; Prestwich, T.C.; Reid, M.A.; Longo, K.A.; Gerin, I.; Cawthorn, W.P.; Susulic, V.S.; Krishnan, V.; Greenfield, A.; Macdougald, O.A. Secreted frizzled-related protein 5 suppresses adipocyte mitochondrial metabolism through WNT inhibition. J. Clin. Investig. 2012, 122, 2405–2416. [Google Scholar] [CrossRef] [PubMed]
  116. Geoghegan, G.; Simcox, J.; Seldin, M.M.; Parnell, T.J.; Stubben, C.; Just, S.; Begaye, L.; Lusis, A.J.; Villanueva, C.J. Targeted deletion of Tcf7l2 in adipocytes promotes adipocyte hypertrophy and impaired glucose metabolism. Mol. Metab. 2019, 24, 44–63. [Google Scholar] [CrossRef] [PubMed]
  117. Li, Z.; Zhang, H.; Denhard, L.A.; Liu, L.H.; Zhou, H.; Lan, Z.J. Reduced white fat mass in adult mice bearing a truncated Patched 1. Int. J. Biol. Sci. 2008, 4, 29–36. [Google Scholar] [CrossRef]
  118. Martin, P.L.; Lane, J.; Pouliot, L.; Gains, M.; Stejskal, R.; Smith, S.Y.; Galdes, A.; Green, J. Increases in Adipose and Total Body Weight, but Not in Lean Body Mass, Associated with Subcutaneous Administration of Sonic Hedgehog-Ig Fusion Protein to Mice. Drug Dev. Res. 2002, 57, 107–114. [Google Scholar] [CrossRef]
  119. Buhman, K.K.; Wang, L.C.; Tang, Y.; Swietlicki, E.A.; Kennedy, S.; Xie, Y.; Liu, Z.Y.; Burkly, L.C.; Levin, M.S.; Rubin, D.C.; et al. Inhibition of Hedgehog signaling protects adult mice from diet-induced weight gain. J. Nutr. 2004, 134, 2979–2984. [Google Scholar] [CrossRef]
  120. Tanaka, T.; Yoshida, N.; Kishimoto, T.; Akira, S. Defective adipocyte differentiation in mice lacking the C/EBPbeta and/or C/EBPdelta gene. EMBO J. 1997, 16, 7432–7443. [Google Scholar] [CrossRef]
  121. Wang, N.D.; Finegold, M.J.; Bradley, A.; Ou, C.N.; Abdelsayed, S.V.; Wilde, M.D.; Taylor, L.R.; Wilson, D.R.; Darlington, G.J. Impaired energy homeostasis in C/EBP alpha knockout mice. Science 1995, 269, 1108–1112. [Google Scholar] [CrossRef] [PubMed]
  122. Linhart, H.G.; Ishimura-Oka, K.; DeMayo, F.; Kibe, T.; Repka, D.; Poindexter, B.; Bick, R.J.; Darlington, G.J. C/EBPalpha is required for differentiation of white, but not brown, adipose tissue. Proc. Natl. Acad. Sci. USA 2001, 98, 12532–12537. [Google Scholar] [CrossRef]
  123. Gray, S.L.; Dalla Nora, E.; Vidal-Puig, A.J. Mouse models of PPAR-gamma deficiency: Dissecting PPAR-gamma’s role in metabolic homoeostasis. Biochem. Soc. Trans. 2005, 33, 1053–1058. [Google Scholar] [CrossRef]
  124. Duan, S.Z.; Ivashchenko, C.Y.; Whitesall, S.E.; D’Alecy, L.G.; Duquaine, D.C.; Brosius, F.C., 3rd; Gonzalez, F.J.; Vinson, C.; Pierre, M.A.; Milstone, D.S.; et al. Hypotension, lipodystrophy, and insulin resistance in generalized PPARgamma-deficient mice rescued from embryonic lethality. J. Clin. Investig. 2007, 117, 812–822. [Google Scholar] [CrossRef] [PubMed]
  125. Koutnikova, H.; Cock, T.A.; Watanabe, M.; Houten, S.M.; Champy, M.F.; Dierich, A.; Auwerx, J. Compensation by the muscle limits the metabolic consequences of lipodystrophy in PPAR gamma hypomorphic mice. Proc. Natl. Acad. Sci. USA 2003, 100, 14457–14462. [Google Scholar] [CrossRef]
  126. Miles, P.D.; Barak, Y.; He, W.; Evans, R.M.; Olefsky, J.M. Improved insulin-sensitivity in mice heterozygous for PPAR-gamma deficiency. J. Clin. Investig. 2000, 105, 287–292. [Google Scholar] [CrossRef]
  127. Zhang, J.; Fu, M.; Cui, T.; Xiong, C.; Xu, K.; Zhong, W.; Xiao, Y.; Floyd, D.; Liang, J.; Li, E.; et al. Selective disruption of PPARgamma 2 impairs the development of adipose tissue and insulin sensitivity. Proc. Natl. Acad. Sci. USA 2004, 101, 10703–10708. [Google Scholar] [CrossRef] [PubMed]
  128. Medina-Gomez, G.; Virtue, S.; Lelliott, C.; Boiani, R.; Campbell, M.; Christodoulides, C.; Perrin, C.; Jimenez-Linan, M.; Blount, M.; Dixon, J.; et al. The link between nutritional status and insulin sensitivity is dependent on the adipocyte-specific peroxisome proliferator-activated receptor-gamma2 isoform. Diabetes 2005, 54, 1706–1716. [Google Scholar] [CrossRef] [PubMed]
  129. Freedman, B.D.; Lee, E.J.; Park, Y.; Jameson, J.L. A dominant negative peroxisome proliferator-activated receptor-gamma knock-in mouse exhibits features of the metabolic syndrome. J. Biol. Chem. 2005, 280, 17118–17125. [Google Scholar] [CrossRef]
  130. Tsai, Y.S.; Kim, H.J.; Takahashi, N.; Kim, H.S.; Hagaman, J.R.; Kim, J.K.; Maeda, N. Hypertension and abnormal fat distribution but not insulin resistance in mice with P465L PPARgamma. J. Clin. Investig. 2004, 114, 240–249. [Google Scholar] [CrossRef]
  131. He, W.; Barak, Y.; Hevener, A.; Olson, P.; Liao, D.; Le, J.; Nelson, M.; Ong, E.; Olefsky, J.M.; Evans, R.M. Adipose-specific peroxisome proliferator-activated receptor gamma knockout causes insulin resistance in fat and liver but not in muscle. Proc. Natl. Acad. Sci. USA 2003, 100, 15712–15717. [Google Scholar] [CrossRef]
  132. Jones, J.R.; Barrick, C.; Kim, K.A.; Lindner, J.; Blondeau, B.; Fujimoto, Y.; Shiota, M.; Kesterson, R.A.; Kahn, B.B.; Magnuson, M.A. Deletion of PPARgamma in adipose tissues of mice protects against high fat diet-induced obesity and insulin resistance. Proc. Natl. Acad. Sci. USA 2005, 102, 6207–6212. [Google Scholar] [CrossRef]
  133. Lee, K.Y.; Russell, S.J.; Ussar, S.; Boucher, J.; Vernochet, C.; Mori, M.A.; Smyth, G.; Rourk, M.; Cederquist, C.; Rosen, E.D.; et al. Lessons on conditional gene targeting in mouse adipose tissue. Diabetes 2013, 62, 864–874. [Google Scholar] [CrossRef]
  134. Wang, F.; Mullican, S.E.; DiSpirito, J.R.; Peed, L.C.; Lazar, M.A. Lipoatrophy and severe metabolic disturbance in mice with fat-specific deletion of PPARγ. Proc. Natl. Acad. Sci. USA 2013, 110, 18656–18661. [Google Scholar] [CrossRef]
  135. Imai, T.; Takakuwa, R.; Marchand, S.; Dentz, E.; Bornert, J.M.; Messaddeq, N.; Wendling, O.; Mark, M.; Desvergne, B.; Wahli, W.; et al. Peroxisome proliferator-activated receptor gamma is required in mature white and brown adipocytes for their survival in the mouse. Proc. Natl. Acad. Sci. USA 2004, 101, 4543–4547. [Google Scholar] [CrossRef] [PubMed]
  136. Shimano, H.; Shimomura, I.; Hammer, R.E.; Herz, J.; Goldstein, J.L.; Brown, M.S.; Horton, J.D. Elevated levels of SREBP-2 and cholesterol synthesis in livers of mice homozygous for a targeted disruption of the SREBP-1 gene. J. Clin. Investig. 1997, 100, 2115–2124. [Google Scholar] [CrossRef]
  137. Shimomura, I.; Hammer, R.E.; Richardson, J.A.; Ikemoto, S.; Bashmakov, Y.; Goldstein, J.L.; Brown, M.S. Insulin resistance and diabetes mellitus in transgenic mice expressing nuclear SREBP-1c in adipose tissue: Model for congenital generalized lipodystrophy. Genes Dev. 1998, 12, 3182–3194. [Google Scholar] [CrossRef] [PubMed]
  138. Franckhauser, S.; Muñoz, S.; Pujol, A.; Casellas, A.; Riu, E.; Otaegui, P.; Su, B.; Bosch, F. Increased fatty acid re-esterification by PEPCK overexpression in adipose tissue leads to obesity without insulin resistance. Diabetes 2002, 51, 624–630. [Google Scholar] [CrossRef]
  139. Jin, W.; Takagi, T.; Kanesashi, S.N.; Kurahashi, T.; Nomura, T.; Harada, J.; Ishii, S. Schnurri-2 controls BMP-dependent adipogenesis via interaction with Smad proteins. Dev. Cell. 2006, 10, 461–471. [Google Scholar] [CrossRef]
  140. Yamauchi, T.; Oike, Y.; Kamon, J.; Waki, H.; Komeda, K.; Tsuchida, A.; Date, Y.; Li, M.X.; Miki, H.; Akanuma, Y.; et al. Increased insulin sensitivity despite lipodystrophy in Crebbp heterozygous mice. Nat. Genet. 2002, 30, 221–226. [Google Scholar] [CrossRef]
  141. Moon, Y.S.; Smas, C.M.; Lee, K.; Villena, J.A.; Kim, K.H.; Yun, E.J.; Sul, H.S. Mice lacking paternally expressed Pref-1/Dlk1 display growth retardation and accelerated adiposity. Mol. Cell Biol. 2002, 22, 5585–5592. [Google Scholar] [CrossRef]
  142. Lee, K.; Villena, J.A.; Moon, Y.S.; Kim, K.H.; Lee, S.; Kang, C.; Sul, H.S. Inhibition of adipogenesis and development of glucose intolerance by soluble preadipocyte factor-1 (Pref-1). J. Clin. Investig. 2003, 111, 453–461. [Google Scholar] [CrossRef] [PubMed]
  143. Villena, J.A.; Choi, C.S.; Wang, Y.; Kim, S.; Hwang, Y.J.; Kim, Y.B.; Cline, G.; Shulman, G.I.; Sul, H.S. Resistance to high-fat diet-induced obesity but exacerbated insulin resistance in mice overexpressing preadipocyte factor-1 (Pref-1): A new model of partial lipodystrophy. Diabetes 2008, 57, 3258–3266. [Google Scholar] [CrossRef] [PubMed]
  144. Oishi, Y.; Manabe, I.; Tobe, K.; Tsushima, K.; Shindo, T.; Fujiu, K.; Nishimura, G.; Maemura, K.; Yamauchi, T.; Kubota, N.; et al. Krüppel-like transcription factor KLF5 is a key regulator of adipocyte differentiation. Cell Metab. 2005, 1, 27–39. [Google Scholar] [CrossRef]
  145. Gordon, A.R.; Outram, S.V.; Keramatipour, M.; Goddard, C.A.; Colledge, W.H.; Metcalfe, J.C.; Hager-Theodorides, A.L.; Crompton, T.; Kemp, P.R. Splenomegaly and modified erythropoiesis in KLF13−/− mice. J. Biol. Chem. 2008, 283, 11897–11904. [Google Scholar] [CrossRef]
  146. Pejnovic, N.N.; Pantic, J.M.; Jovanovic, I.P.; Radosavljevic, G.D.; Milovanovic, M.Z.; Nikolic, I.G.; Zdravkovic, N.S.; Djukic, A.L.; Arsenijevic, N.N.; Lukic, M.L. Galectin-3 deficiency accelerates high-fat diet-induced obesity and amplifies inflammation in adipose tissue and pancreatic islets. Diabetes 2013, 62, 1932–1944. [Google Scholar] [CrossRef] [PubMed]
  147. Li, P.; Liu, S.; Lu, M.; Bandyopadhyay, G.; Oh, D.; Imamura, T.; Johnson, A.M.F.; Sears, D.; Shen, Z.; Cui, B.; et al. Hematopoietic-Derived Galectin-3 Causes Cellular and Systemic Insulin Resistance. Cell 2016, 167, 973–984. [Google Scholar] [CrossRef]
  148. Pang, J.; Rhodes, D.H.; Pini, M.; Akasheh, R.T.; Castellanos, K.J.; Cabay, R.J.; Cooper, D.; Perretti, M.; Fantuzzi, G. Increased adiposity.; dysregulated glucose metabolism and systemic inflammation in Galectin-3 KO mice. PLoS ONE 2013, 8, e57915. [Google Scholar] [CrossRef] [PubMed]
  149. Takada, N.; Takasugi, M.; Nonaka, Y.; Kamiya, T.; Takemura, K.; Satoh, J.; Ito, S.; Fujimoto, K.; Uematsu, S.; Yoshida, K.; et al. Galectin-3 promotes the adipogenic differentiation of PDGFRα+ cells and ectopic fat formation in regenerating muscle. Development 2022, 149, dev199443. [Google Scholar] [CrossRef]
  150. Martínez-Martínez, E.; Calvier, L.; Rossignol, P.; Rousseau, E.; Fernández-Celis, A.; Jurado-López, R.; Laville, M.; Cachofeiro, V.; López-Andrés, N. Galectin-3 inhibition prevents adipose tissue remodelling in obesity. Int. J. Obes. 2016, 40, 1034–1038. [Google Scholar] [CrossRef]
  151. Pugliese, G.; Iacobini, C.; Pesce, C.M.; Menini, S. Galectin-3: An emerging all-out player in metabolic disorders and their complications. Glycobiology 2015, 25, 136–150. [Google Scholar] [CrossRef] [PubMed]
  152. Pricci, F.; Leto, G.; Amadio, L.; Iacobini, C.; Romeo, G.; Cordone, S.; Gradini, R.; Barsotti, P.; Liu, F.T.; Di Mario, U.; et al. Role of galectin-3 as a receptor for advanced glycosylation end products. Kidney Int. Suppl. 2000, 77, S31–S39. [Google Scholar] [CrossRef] [PubMed]
  153. Trim, W.V.; Walhin, J.P.; Koumanov, F.; Bouloumié, A.; Lindsay, M.A.; Chen, Y.C.; Travers, R.L.; Turner, J.E.; Thompson, D. Divergent immunometabolic changes in adipose tissue and skeletal muscle with ageing in healthy humans. J. Physiol. 2022, 600, 921–947. [Google Scholar] [CrossRef] [PubMed]
  154. López-Otín, C.; Galluzzi, L.; Freije, J.M.P.; Madeo, F.; Kroemer, G. Metabolic Control of Longevity. Cell 2016, 166, 802–821. [Google Scholar] [CrossRef] [PubMed]
  155. Barzilai, N.; Gupta, G. Revisiting the role of fat mass in the life extension induced by caloric restriction. J. Gerontol. A Biol. Sci. Med. Sci. 1999, 54, B89–B96, discussion B97–B98. [Google Scholar] [CrossRef] [PubMed]
  156. Ageing and Health. Available online: https://www.who.int/news-room/fact-sheets/detail/ageing-and-health (accessed on 4 March 2024).
  157. Roszkowska-Gancarz, M.; Bartoszewicz, Z.; Połosak, J.; Kuryłowicz, A.; Jonas, M.; Mossakowska, M.; Franek, E.; Puzianowska-Kuźnicka, M. Total and High Molecular Weight Adiponectin and Level-Modifying Polymorphisms of ADIPOQ in Centenarians. Endokrynol. Pol. 2012, 63, 439–446. [Google Scholar] [PubMed]
  158. Arai, Y.; Kamide, K.; Hirose, N. Adipokines and Aging: Findings from Centenarians and the Very Old. Front. Endocrinol. 2019, 10, 440966. [Google Scholar] [CrossRef]
  159. Han, S.-J.; Zaretsky, A.G.; Andrade-Oliveira, V.; Brenchley, J.M.; Brodsky, I.E.; Belkaid, Y.; Collins, N.; Dzutsev, A.; Shaik, J.; Morais Da Fonseca, D.; et al. White Adipose Tissue Is a Reservoir for Memory T Cells and Promotes Protective Memory Responses to Infection. Immunity 2017, 47, 1154–1168.e6. [Google Scholar] [CrossRef] [PubMed]
  160. Cinti, S. Obesity, Type 2 Diabetes and the Adipose Organ; Springer International Publishing AG: Cham, Switzerland, 2018. [Google Scholar]
  161. Schwartz, R.S.; Jaeger, L.F.; Veith, R.C.; Lakshminarayan, S. The Effect of Diet or Exercise on Plasma Norepinephrine Kinetics in Moderately Obese Young Men. Int. J. Obes. 1990, 14, 1–11. [Google Scholar]
  162. Caso, G.; McNurlan, M.A.; Mileva, I.; Zemlyak, A.; Mynarcik, D.C.; Gelato, M.C. Peripheral Fat Loss and Decline in Adipogenesis in Older Humans. Metabolism 2013, 62, 337–340. [Google Scholar] [CrossRef]
  163. Tchkonia, T.; Morbeck, D.E.; Von Zglinicki, T.; Van Deursen, J.; Lustgarten, J.; Scrable, H.; Khosla, S.; Jensen, M.D.; Kirkland, J.L. Fat Tissue, Aging, and Cellular Senescence. Aging Cell 2010, 9, 667–684. [Google Scholar] [CrossRef]
  164. Corrales, P.; Vivas, Y.; Izquierdo-Lahuerta, A.; Horrillo, D.; Seoane-Collazo, P.; Velasco, I.; Torres, L.; Lopez, Y.; Martínez, C.; López, M.; et al. Long-Term Caloric Restriction Ameliorates Deleterious Effects of Aging on White and Brown Adipose Tissue Plasticity. Aging Cell 2019, 18, e12948. [Google Scholar] [CrossRef]
  165. Schaum, N.; Lehallier, B.; Hahn, O.; Pálovics, R.; Hosseinzadeh, S.; Lee, S.E.; Sit, R.; Lee, D.P.; Losada, P.M.; Zardeneta, M.E.; et al. Ageing Hallmarks Exhibit Organ-Specific Temporal Signatures. Nature 2020, 583, 596–602. [Google Scholar] [CrossRef] [PubMed]
  166. Lumeng, C.N.; Liu, J.; Geletka, L.; Delaney, C.; Delproposto, J.; Desai, A.; Oatmen, K.; Martinez-Santibanez, G.; Julius, A.; Garg, S.; et al. Aging Is Associated with an Increase in T Cells and Inflammatory Macrophages in Visceral Adipose Tissue. J. Immunol. 2011, 187, 6208–6216. [Google Scholar] [CrossRef] [PubMed]
  167. Yu, Q.; Xiao, H.; Jedrychowski, M.P.; Schweppe, D.K.; Navarrete-Perea, J.; Knott, J.; Rogers, J.; Chouchani, E.T.; Gygi, S.P. Sample multiplexing for targeted pathway proteomics in aging mice. Proc. Natl. Acad. Sci. USA 2020, 117, 9723–9732. [Google Scholar] [CrossRef]
  168. Gohlke, S.; Zagoriy, V.; Cuadros Inostroza, A.; Méret, M.; Mancini, C.; Japtok, L.; Schumacher, F.; Kuhlow, D.; Graja, A.; Stephanowitz, H.; et al. Identification of Functional Lipid Metabolism Biomarkers of Brown Adipose Tissue Aging. Mol. Metab. 2019, 24, 1–17. [Google Scholar] [CrossRef]
  169. Held, N.M.; Buijink, M.R.; Elfrink, H.L.; Kooijman, S.; Janssens, G.E.; Luyf, A.C.M.; Pras-Raves, M.L.; Vaz, F.M.; Michel, S.; Houtkooper, R.H.; et al. Aging Selectively Dampens Oscillation of Lipid Abundance in White and Brown Adipose Tissue. Sci. Rep. 2021, 11, 5932. [Google Scholar] [CrossRef]
  170. Bonzón-Kulichenko, E.; Moltó, E.; Pintado, C.; Fernández, A.; Arribas, C.; Schwudke, D.; Gallardo, N.; Shevchenko, A.; Andrés, A. Changes in Visceral Adipose Tissue Plasma Membrane Lipid Composition in Old Rats Are Associated with Adipocyte Hypertrophy with Aging. J. Gerontol. A Biol. Sci. Med. Sci. 2018, 73, 1139–1146. [Google Scholar] [CrossRef]
  171. Pamplona, R. Membrane Phospholipids, Lipoxidative Damage and Molecular Integrity: A Causal Role in Aging and Longevity. Biochim. Biophys. Acta 2008, 1777, 1249–1262. [Google Scholar] [CrossRef]
  172. Fajardo, V.A.; McMeekin, L.; Leblanc, P.J. Influence of Phospholipid Species on Membrane Fluidity: A Meta-Analysis for a Novel Phospholipid Fluidity Index. J. Membr. Biol. 2011, 244, 97–103. [Google Scholar] [CrossRef]
  173. Slade, E.; Irvin, M.R.; Xie, K.; Arnett, D.K.; Claas, S.A.; Kind, T.; Fardo, D.W.; Graf, G.A. Age and Sex Are Associated with the Plasma Lipidome: Findings from the GOLDN Study. Lipids Health Dis. 2021, 20, 30. [Google Scholar] [CrossRef] [PubMed]
  174. Collino, S.; Montoliu, I.; Martin, F.P.J.; Scherer, M.; Mari, D.; Salvioli, S.; Bucci, L.; Ostan, R.; Monti, D.; Biagi, E.; et al. Metabolic Signatures of Extreme Longevity in Northern Italian Centenarians Reveal a Complex Remodeling of Lipids, Amino Acids, and Gut Microbiota Metabolism. PLoS ONE 2013, 8, e56564. [Google Scholar] [CrossRef] [PubMed]
  175. Cho, Y.K.; Lee, S.; Lee, J.; Doh, J.; Park, J.H.; Jung, Y.S.; Lee, Y.H. Lipid Remodeling of Adipose Tissue in Metabolic Health and Disease. Exp. Mol. Med. 2023, 55, 1955–1973. [Google Scholar] [CrossRef] [PubMed]
  176. Mielke, M.M.; Haughey, N.J.; Bandaru, V.V.R.; Weinberg, D.D.; Darby, E.; Zaidi, N.; Pavlik, V.; Doody, R.S.; Lyketsos, C.G. Plasma Sphingomyelins Are Associated with Cognitive Progression in Alzheimer’s Disease. J. Alzheimer’s Dis. 2011, 27, 259–269. [Google Scholar] [CrossRef] [PubMed]
  177. Iqbal, J.; Walsh, M.T.; Hammad, S.M.; Hussain, M.M. Sphingolipids and Lipoproteins in Health and Metabolic Disorders. Trends Endocrinol. Metab. 2017, 28, 506–518. [Google Scholar] [CrossRef] [PubMed]
  178. Wennberg, A.M.V.; Schafer, M.J.; Lebrasseur, N.K.; Savica, R.; Bui, H.H.; Hagen, C.E.; Hollman, J.H.; Petersen, R.C.; Mielke, M.M. Plasma Sphingolipids Are Associated with Gait Parameters in the Mayo Clinic Study of Aging. J. Gerontol. A Biol. Sci. Med. Sci. 2018, 73, 960–965. [Google Scholar] [CrossRef] [PubMed]
  179. Kumari, R.; Jat, P. Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype. Front. Cell Dev. Biol. 2021, 9, 645593. [Google Scholar] [CrossRef] [PubMed]
  180. Liu, Z.; Wu, K.K.L.; Jiang, X.; Xu, A.; Cheng, K.K.Y. The Role of Adipose Tissue Senescence in Obesity- and Ageing-Related Metabolic Disorders. Clin. Sci. 2020, 134, 315–330. [Google Scholar] [CrossRef] [PubMed]
  181. Campisi, J. Aging, Cellular Senescence, and Cancer. Annu. Rev. Physiol. 2013, 75, 685–705. [Google Scholar] [CrossRef]
  182. Baker, D.J.; Wijshake, T.; Tchkonia, T.; Lebrasseur, N.K.; Childs, B.G.; Van De Sluis, B.; Kirkland, J.L.; Van Deursen, J.M. Clearance of P16Ink4a-Positive Senescent Cells Delays Ageing-Associated Disorders. Nature 2011, 479, 232–236. [Google Scholar] [CrossRef]
  183. Xu, M.; Palmer, A.K.; Ding, H.; Weivoda, M.M.; Pirtskhalava, T.; White, T.A.; Sepe, A.; Johnson, K.O.; Stout, M.B.; Giorgadze, N.; et al. Targeting Senescent Cells Enhances Adipogenesis and Metabolic Function in Old Age. Elife 2015, 4, e12997. [Google Scholar] [CrossRef]
  184. Xu, M.; Tchkonia, T.; Ding, H.; Ogrodnik, M.; Lubbers, E.R.; Pirtskhalava, T.; White, T.A.; Johnson, K.O.; Stout, M.B.; Mezera, V.; et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc. Natl. Acad. Sci. USA 2015, 112, E6301–E6310. [Google Scholar] [CrossRef]
  185. Berry, D.C.; Jiang, Y.; Arpke, R.W.; Close, E.L.; Uchida, A.; Reading, D.; Berglund, E.D.; Kyba, M.; Graff, J.M. Cellular Aging Contributes to Failure of Cold-Induced Beige Adipocyte Formation in Old Mice and Humans. Cell Metab. 2017, 25, 166–181. [Google Scholar] [CrossRef]
  186. Stout, M.B.; Tchkonia, T.; Pirtskhalava, T.; Palmer, A.K.; List, E.O.; Berryman, D.E.; Lubbers, E.R.; Escande, C.; Spong, A.; Masternak, M.M.; et al. Growth Hormone Action Predicts Age-Related White Adipose Tissue Dysfunction and Senescent Cell Burden in Mice. Aging 2014, 6, 575–586. [Google Scholar] [CrossRef]
  187. Palmer, A.K.; Jensen, M.D. Metabolic Changes in Aging Humans: Current Evidence and Therapeutic Strategies. J. Clin. Investig. 2022, 132, e158451. [Google Scholar] [CrossRef]
  188. Karagiannides, I.; Tchkonia, T.; Dobson, D.E.; Steppan, C.M.; Cummins, P.; Chan, G.; Salvatori, K.; Hadzopoulou-Cladaras, M.; Kirkland, J.L. Altered Expression of C/EBP Family Members Results in Decreased Adipogenesis with Aging. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2001, 280, R1772–R1780. [Google Scholar] [CrossRef] [PubMed]
  189. Le Pelletier, L.; Mantecon, M.; Gorwood, J.; Auclair, M.; Foresti, R.; Motterlini, R.; Laforge, M.; Atlan, M.; Fève, B.; Capeau, J.; et al. Metformin Alleviates Stress-Induced Cellular Senescence of Aging Human Adipose Stromal Cells and the Ensuing Adipocyte Dysfunction. Elife 2021, 10, e62635. [Google Scholar] [CrossRef]
  190. Lakowa, N.; Trieu, N.; Flehmig, G.; Lohmann, T.; Schön, M.R.; Dietrich, A.; Zeplin, P.H.; Langer, S.; Stumvoll, M.; Blüher, M.; et al. Telomere Length Differences between Subcutaneous and Visceral Adipose Tissue in Humans. Biochem. Biophys. Res. Commun. 2015, 457, 426–432. [Google Scholar] [CrossRef]
  191. Nguyen, H.P.; Lin, F.; Yi, D.; Xie, Y.; Dinh, J.; Xue, P.; Sul, H.S. Aging-Dependent Regulatory Cells Emerge in Subcutaneous Fat to Inhibit Adipogenesis. Dev. Cell 2021, 56, 1437–1451.e3. [Google Scholar] [CrossRef] [PubMed]
  192. Wang, W.; Ishibashi, J.; Trefely, S.; Shao, M.; Cowan, A.J.; Sakers, A.; Lim, H.W.; O’Connor, S.; Doan, M.T.; Cohen, P.; et al. A PRDM16-Driven Metabolic Signal from Adipocytes Regulates Precursor Cell Fate. Cell Metab. 2019, 30, 174–189.e5. [Google Scholar] [CrossRef]
  193. Wiley, C.D.; Campisi, J. The Metabolic Roots of Senescence: Mechanisms and Opportunities for Intervention. Nat. Metab. 2021, 3, 1290–1301. [Google Scholar] [CrossRef] [PubMed]
  194. Yamamuro, T.; Kawabata, T.; Fukuhara, A.; Saita, S.; Nakamura, S.; Takeshita, H.; Fujiwara, M.; Enokidani, Y.; Yoshida, G.; Tabata, K.; et al. Age-Dependent Loss of Adipose Rubicon Promotes Metabolic Disorders via Excess Autophagy. Nat. Commun. 2020, 11, 4150. [Google Scholar] [CrossRef] [PubMed]
  195. Ahmadian, M.; Suh, J.M.; Hah, N.; Liddle, C.; Atkins, A.R.; Downes, M.; Evans, R.M. PPARγ Signaling and Metabolism: The Good, the Bad and the Future. Nat. Med. 2013, 19, 557–566. [Google Scholar] [CrossRef] [PubMed]
  196. Sun, K.; Tordjman, J.; Clément, K.; Scherer, P.E. Fibrosis and Adipose Tissue Dysfunction. Cell Metab. 2013, 18, 470–477. [Google Scholar] [CrossRef] [PubMed]
  197. Berry, D.C.; Jiang, Y.; Graff, J.M. Mouse Strains to Study Cold-Inducible Beige Progenitors and Beige Adipocyte Formation and Function. Nat. Commun. 2016, 7, 10184. [Google Scholar] [CrossRef] [PubMed]
  198. Marcelin, G.; Ferreira, A.; Liu, Y.; Atlan, M.; Aron-Wisnewsky, J.; Pelloux, V.; Botbol, Y.; Ambrosini, M.; Fradet, M.; Rouault, C.; et al. A PDGFRα-Mediated Switch toward CD9high Adipocyte Progenitors Controls Obesity-Induced Adipose Tissue Fibrosis. Cell Metab. 2017, 25, 673–685. [Google Scholar] [CrossRef]
  199. Hasegawa, Y.; Ikeda, K.; Chen, Y.; Alba, D.L.; Stifler, D.; Shinoda, K.; Hosono, T.; Maretich, P.; Yang, Y.; Ishigaki, Y.; et al. Repression of Adipose Tissue Fibrosis through a PRDM16-GTF2IRD1 Complex Improves Systemic Glucose Homeostasis. Cell Metab. 2018, 27, 180–194.e6. [Google Scholar] [CrossRef]
  200. Xu, M.; Pirtskhalava, T.; Farr, J.N.; Weigand, B.M.; Palmer, A.K.; Weivoda, M.M.; Inman, C.L.; Ogrodnik, M.B.; Hachfeld, C.M.; Fraser, D.G.; et al. Senolytics Improve Physical Function and Increase Lifespan in Old Age. Nat. Med. 2018, 24, 1246–1256. [Google Scholar] [CrossRef]
  201. Hickson, L.T.J.; Langhi Prata, L.G.P.; Bobart, S.A.; Evans, T.K.; Giorgadze, N.; Hashmi, S.K.; Herrmann, S.M.; Jensen, M.D.; Jia, Q.; Jordan, K.L.; et al. Senolytics Decrease Senescent Cells in Humans: Preliminary Report from a Clinical Trial of Dasatinib plus Quercetin in Individuals with Diabetic Kidney Disease. EBioMedicine 2019, 47, 446–456. [Google Scholar] [CrossRef]
  202. Justice, J.N.; Nambiar, A.M.; Tchkonia, T.; LeBrasseur, N.K.; Pascual, R.; Hashmi, S.K.; Prata, L.; Masternak, M.M.; Kritchevsky, S.B.; Musi, N.; et al. Senolytics in Idiopathic Pulmonary Fibrosis: Results from a First-in-Human, Open-Label, Pilot Study. EBioMedicine 2019, 40, 554–563. [Google Scholar] [CrossRef]
  203. Crespo-Garcia, S.; Tsuruda, P.R.; Dejda, A.; Ryan, R.D.; Fournier, F.; Chaney, S.Y.; Pilon, F.; Dogan, T.; Cagnone, G.; Patel, P.; et al. Pathological Angiogenesis in Retinopathy Engages Cellular Senescence and Is Amenable to Therapeutic Elimination via BCL-XL Inhibition. Cell Metab. 2021, 33, 818–832.e7. [Google Scholar] [CrossRef] [PubMed]
  204. Suda, M.; Shimizu, I.; Katsuumi, G.; Yoshida, Y.; Hayashi, Y.; Ikegami, R.; Matsumoto, N.; Yoshida, Y.; Mikawa, R.; Katayama, A.; et al. Senolytic Vaccination Improves Normal and Pathological Age-Related Phenotypes and Increases Lifespan in Progeroid Mice. Nat. Aging 2021, 1, 1117–1126. [Google Scholar] [CrossRef] [PubMed]
  205. Martin-Montalvo, A.; Mercken, E.M.; Mitchell, S.J.; Palacios, H.H.; Mote, P.L.; Scheibye-Knudsen, M.; Gomes, A.P.; Ward, T.M.; Minor, R.K.; Blouin, M.J.; et al. Metformin Improves Healthspan and Lifespan in Mice. Nat. Commun. 2013, 4, 2192. [Google Scholar] [CrossRef] [PubMed]
  206. Kulkarni, A.S.; Brutsaert, E.F.; Anghel, V.; Zhang, K.; Bloomgarden, N.; Pollak, M.; Mar, J.C.; Hawkins, M.; Crandall, J.P.; Barzilai, N. Metformin Regulates Metabolic and Nonmetabolic Pathways in Skeletal Muscle and Subcutaneous Adipose Tissues of Older Adults. Aging Cell 2018, 17, e12723. [Google Scholar] [CrossRef]
  207. Goltsman, I.; Khoury, E.E.; Winaver, J.; Abassi, Z. Does Thiazolidinedione Therapy Exacerbate Fluid Retention in Congestive Heart Failure? Pharmacol. Ther. 2016, 168, 75–97. [Google Scholar] [CrossRef] [PubMed]
  208. Guan, H.P.; Li, Y.; Jensen, M.V.; Newgard, C.B.; Steppan, C.M.; Lazar, M.A. A Futile Metabolic Cycle Activated in Adipocytes by Antidiabetic Agents. Nat. Med. 2002, 8, 1122–1128. [Google Scholar] [CrossRef] [PubMed]
  209. Xu, L.; Ma, X.; Verma, N.; Perie, L.; Pendse, J.; Shamloo, S.; Marie Josephson, A.; Wang, D.; Qiu, J.; Guo, M.; et al. PPARγ Agonists Delay Age-Associated Metabolic Disease and Extend Longevity. Aging Cell 2020, 19, e13267. [Google Scholar] [CrossRef] [PubMed]
  210. Hogrel, J.Y.; Barnouin, Y.; Azzabou, N.; Butler-Browne, G.; Voit, T.; Moraux, A.; Leroux, G.; Behin, A.; McPhee, J.S.; Carlier, P.G. NMR Imaging Estimates of Muscle Volume and Intramuscular Fat Infiltration in the Thigh: Variations with Muscle, Gender, and Age. Age 2015, 37, 60. [Google Scholar] [CrossRef] [PubMed]
  211. Ezure, T. Subcutaneous Fat Infiltration into the Dermal Layer Induces Wrinkle Formation. Skin Res. Technol. 2023, 29, e13296. [Google Scholar] [CrossRef]
  212. Ezure, T.; Amano, S. Influence of Subcutaneous Adipose Tissue Mass on Dermal Elasticity and Sagging Severity in Lower Cheek. Skin Res. Technol. 2010, 16, 332–338. [Google Scholar] [CrossRef]
  213. Pachón-Peña, G.; Bredella, M.A. Bone Marrow Adipose Tissue in Metabolic Health. Trends Endocrinol. Metab. 2022, 33, 401–408. [Google Scholar] [CrossRef] [PubMed]
  214. Berenbaum, F.; Griffin, T.M.; Liu-Bryan, R. Review: Metabolic Regulation of Inflammation in Osteoarthritis. Arthritis Rheumatol. 2017, 69, 9–21. [Google Scholar] [CrossRef] [PubMed]
  215. Collins, K.H.; Herzog, W.; MacDonald, G.Z.; Reimer, R.A.; Rios, J.L.; Smith, I.C.; Zernicke, R.F.; Hart, D.A. Obesity, Metabolic Syndrome, and Musculoskeletal Disease: Common Inflammatory Pathways Suggest a Central Role for Loss of Muscle Integrity. Front. Physiol. 2018, 9, 112. [Google Scholar] [CrossRef]
  216. Vos, T.; Allen, C.; Arora, M.; Barber, R.M.; Brown, A.; Carter, A.; Casey, D.C.; Charlson, F.J.; Chen, A.Z.; Coggeshall, M.; et al. Global, Regional, and National Incidence, Prevalence, and Years Lived with Disability for 310 Diseases and Injuries, 1990-2015: A Systematic Analysis for the Global Burden of Disease Study 2015. Lancet 2016, 388, 1545–1602. [Google Scholar] [CrossRef]
  217. March, L.; Smith, E.U.R.; Hoy, D.G.; Cross, M.J.; Sanchez-Riera, L.; Blyth, F.; Buchbinder, R.; Vos, T.; Woolf, A.D. Burden of Disability Due to Musculoskeletal (MSK) Disorders. Best Pract. Res. Clin. Rheumatol. 2014, 28, 353–366. [Google Scholar] [CrossRef]
  218. Glyn-Jones, S.; Palmer, A.J.R.; Agricola, R.; Price, A.J.; Vincent, T.L.; Weinans, H.; Carr, A.J. Osteoarthritis. Lancet 2015, 386, 376–387. [Google Scholar] [CrossRef]
  219. Tang, R.; Harasymowicz, N.S.; Wu, C.L.; Collins, K.H.; Choi, Y.R.; Oswald, S.J.; Guilak, F. Gene Therapy for Follistatin Mitigates Systemic Metabolic Inflammation and Post-Traumatic Arthritis in High-Fat Diet-Induced Obesity. Sci. Adv. 2020, 6, eaaz7492. [Google Scholar] [CrossRef] [PubMed]
  220. Griffin, T.M.; Huebner, J.L.; Kraus, V.B.; Guilak, F. Extreme Obesity Due to Impaired Leptin Signaling in Mice Does Not Cause Knee Osteoarthritis. Arthritis Rheum. 2009, 60, 2935–2944. [Google Scholar] [CrossRef]
  221. Harasymowicz, N.S.; Choi, Y.R.; Wu, C.L.; Iannucci, L.; Tang, R.; Guilak, F. Intergenerational Transmission of Diet-Induced Obesity, Metabolic Imbalance, and Osteoarthritis in Mice. Arthritis Rheumatol. 2020, 72, 632–644. [Google Scholar] [CrossRef]
  222. Wu, C.L.; Jain, D.; McNeill, J.N.; Little, D.; Anderson, J.A.; Huebner, J.L.; Kraus, V.B.; Rodriguiz, R.M.; Wetsel, W.C.; Guilak, F. Dietary Fatty Acid Content Regulates Wound Repair and the Pathogenesis of Osteoarthritis Following Joint Injury. Ann. Rheum. Dis. 2015, 74, 2076–2083. [Google Scholar] [CrossRef]
  223. Griffin, T.M.; Fermor, B.; Huebner, J.L.; Kraus, V.B.; Rodriguiz, R.M.; Wetsel, W.C.; Cao, L.; Setton, L.A.; Guilak, F. Diet-Induced Obesity Differentially Regulates Behavioral, Biomechanical, and Molecular Risk Factors for Osteoarthritis in Mice. Arthritis Res. Ther. 2010, 12, R130. [Google Scholar] [CrossRef] [PubMed]
  224. Mengsteab, P.Y.; Otsuka, T.; McClinton, A.; Shemshaki, N.S.; Shah, S.; Kan, H.M.; Obopilwe, E.; Vella, A.T.; Nair, L.S.; Laurencin, C.T. Mechanically Superior Matrices Promote Osteointegration and Regeneration of Anterior Cruciate Ligament Tissue in Rabbits. Proc. Natl. Acad. Sci. USA 2020, 117, 28655–28666. [Google Scholar] [CrossRef] [PubMed]
  225. Collins, K.H.; Lenz, K.L.; Pollitt, E.N.; Ferguson, D.; Hutson, I.; Springer, L.E.; Oestreich, A.K.; Tang, R.; Choi, Y.R.; Meyer, G.A.; et al. Adipose Tissue Is a Critical Regulator of Osteoarthritis. Proc. Natl. Acad. Sci. USA 2020, 118, e2021096118. [Google Scholar] [CrossRef]
  226. Miller, D.; Grant, A.; Durgam, S.; El-Hayek, K.; Flanigan, D.C.; Malanga, G.; Vasileff, W.K.; Baria, M.R. Adipose-Derived Stem Cells, Obesity, and Inflammation: A Systematic Review and Implications for Osteoarthritis Treatment. Am. J. Phys. Med. Rehabil. 2022, 101, 879–887. [Google Scholar] [CrossRef] [PubMed]
  227. Perucca Orfei, C.; Boffa, A.; Sourugeon, Y.; Laver, L.; Magalon, J.; Sánchez, M.; Tischer, T.; Filardo, G.; de Girolamo, L. Cell-Based Therapies Have Disease-Modifying Effects on Osteoarthritis in Animal Models. A Systematic Review by the ESSKA Orthobiologic Initiative. Part 1: Adipose Tissue-Derived Cell-Based Injectable Therapies. Knee Surg. Sports Traumatol. Arthrosc. 2023, 31, 641–655. [Google Scholar] [CrossRef] [PubMed]
  228. Lu, L.; Dai, C.; Zhang, Z.; Du, H.; Li, S.; Ye, P.; Fu, Q.; Zhang, L.; Wu, X.; Dong, Y.; et al. Treatment of Knee Osteoarthritis with Intra-Articular Injection of Autologous Adipose-Derived Mesenchymal Progenitor Cells: A Prospective, Randomized, Double-Blind, Active-Controlled, Phase IIb Clinical Trial. Stem Cell Res. Ther. 2019, 10, 143. [Google Scholar] [CrossRef] [PubMed]
  229. Freitag, J.; Bates, D.; Wickham, J.; Shah, K.; Huguenin, L.; Tenen, A.; Paterson, K.; Boyd, R. Adipose-Derived Mesenchymal Stem Cell Therapy in the Treatment of Knee Osteoarthritis: A Randomized Controlled Trial. Regen. Med. 2019, 14, 213–230. [Google Scholar] [CrossRef] [PubMed]
  230. Garza, J.R.; Campbell, R.E.; Tjoumakaris, F.P.; Freedman, K.B.; Miller, L.S.; Santa Maria, D.; Tucker, B.S. Clinical Efficacy of Intra-Articular Mesenchymal Stromal Cells for the Treatment of Knee Osteoarthritis: A Double-Blinded Prospective Randomized Controlled Clinical Trial. Am. J. Sports Med. 2020, 48, 588–598. [Google Scholar] [CrossRef] [PubMed]
  231. Gadelkarim, M.; Abd Elmegeed, A.; Allam, A.H.; Awad, A.K.; Shehata, M.A.; AbouEl-Enein, A.; Alsadek, M.E.; Abo Deeb, M.; Afifi, A.M. Safety and Efficacy of Adipose-Derived Mesenchymal Stem Cells for Knee Osteoarthritis: A Systematic Review and m-Analysis. Jt. Bone Spine 2022, 89, 105404. [Google Scholar] [CrossRef]
  232. Zaffagnini, S.; Andriolo, L.; Boffa, A.; Poggi, A.; Cenacchi, A.; Busacca, M.; Kon, E.; Filardo, G.; Di Martino, A. Microfragmented Adipose Tissue Versus Platelet-Rich Plasma for the Treatment of Knee Osteoarthritis: A Prospective Randomized Controlled Trial at 2-Year Follow-Up. Am. J. Sports Med. 2022, 50, 2881–2892. [Google Scholar] [CrossRef]
  233. Van Genechten, W.; Vuylsteke, K.; Martinez, P.R.; Swinnen, L.; Sas, K.; Verdonk, P. Autologous Micro-Fragmented Adipose Tissue (MFAT) to Treat Symptomatic Knee Osteoarthritis: Early Outcomes of a Consecutive Case Series. J. Clin. Med. 2021, 10, 2231. [Google Scholar] [CrossRef] [PubMed]
  234. Cavallo, C.; Boffa, A.; Salerno, M.; Merli, G.; Grigolo, B.; Filardo, G. Adipose Tissue-Derived Products May Present Inflammatory Properties That Affect Chondrocytes and Synoviocytes from Patients with Knee Osteoarthritis. Int. J. Mol. Sci. 2023, 24, 12401. [Google Scholar] [CrossRef] [PubMed]
  235. Gobbi, A.; Dallo, I.; Rogers, C.; Striano, R.D.; Mautner, K.; Bowers, R.; Rozak, M.; Bilbool, N.; Murrell, W.D. Two-Year Clinical Outcomes of Autologous Microfragmented Adipose Tissue in Elderly Patients with Knee Osteoarthritis: A Multi-Centric, International Study. Int. Orthop. 2021, 45, 1179–1188. [Google Scholar] [CrossRef]
  236. Park, J.S.; Park, G.; Hong, H.S. Age Affects the Paracrine Activity and Differentiation Potential of Human Adipose-derived Stem Cells. Mol. Med. Rep. 2021, 23, 160. [Google Scholar] [CrossRef] [PubMed]
  237. Distel, E.; Cadoudal, T.; Durant, S.; Poignard, A.; Chevalier, X.; Benelli, C. The Infrapatellar Fat Pad in Knee Osteoarthritis: An Important Source of Interleukin-6 and Its Soluble Receptor. Arthritis Rheum. 2009, 60, 3374–3377. [Google Scholar] [CrossRef]
  238. Favero, M.; El-Hadi, H.; Belluzzi, E.; Granzotto, M.; Porzionato, A.; Sarasin, G.; Rambaldo, A.; Iacobellis, C.; Cigolotti, A.; Fontanella, C.G.; et al. Infrapatellar Fat Pad Features in Osteoarthritis: A Histopathological and Molecular Study. Rheumatology 2017, 56, 1784–1793. [Google Scholar] [CrossRef] [PubMed]
  239. Stocco, E.; Belluzzi, E.; Contran, M.; Boscolo-Berto, R.; Picardi, E.; Guidolin, D.; Fontanella, C.G.; Olivotto, E.; Filardo, G.; Borile, G.; et al. Age-Dependent Remodeling in Infrapatellar Fat Pad Adipocytes and Extracellular Matrix: A Comparative Study. Front. Med. 2021, 8, 1403. [Google Scholar] [CrossRef] [PubMed]
  240. Anam, A.K.; Insogna, K. Update on Osteoporosis Screening and Management. Med. Clin. 2021, 105, 1117–1134. [Google Scholar] [CrossRef]
  241. Hardouin, P.; Rharass, T.; Lucas, S. Bone Marrow Adipose Tissue: To Be or Not To Be a Typical Adipose Tissue? Front. Endocrinol. 2016, 7, 85. [Google Scholar] [CrossRef]
  242. Shapses, S.A.; Sukumar, D. Bone Metabolism in Obesity and Weight Loss. Annu. Rev. Nutr. 2012, 32, 287–309. [Google Scholar] [CrossRef]
  243. Zhang, P.; Peterson, M.; Su, G.L.; Wang, S.C. Visceral adiposity is negatively associated with bone density and muscle attenuation. Am. J. Clin. Nutr. 2015, 101, 337–343. [Google Scholar] [CrossRef] [PubMed]
  244. Liu, C.T.; Broe, K.E.; Zhou, Y.; Boyd, S.K.; Cupples, L.A.; Hannan, M.T.; Lim, E.; McLean, R.R.; Samelson, E.J.; Bouxsein, M.L.; et al. Visceral Adipose Tissue Is Associated with Bone Microarchitecture in the Framingham Osteoporosis Study. J. Bone Miner. Res. 2017, 32, 143–150. [Google Scholar] [CrossRef] [PubMed]
  245. Lin, Y.; Zhong, X.; Lu, D.; Yao, W.; Zhou, J.; Wu, R.; Feng, F. Association of Visceral and Subcutaneous Fat with Bone Mineral Density in US Adults: A Cross-Sectional Study. Sci. Rep. 2023, 13, 10682. [Google Scholar] [CrossRef] [PubMed]
  246. Justesen, J.; Stenderup, K.; Ebbesen, E.N.; Mosekilde, L.; Steiniche, T.; Kassem, M. Adipocyte Tissue Volume in Bone Marrow Is Increased with Aging and in Patients with Osteoporosis. Biogerontology 2001, 2, 165–171. [Google Scholar] [CrossRef] [PubMed]
  247. Griffith, J.F.; Yeung, D.K.W.; Antonio, G.E.; Lee, F.K.H.; Hong, A.W.L.; Wong, S.Y.S.; Lau, E.M.C.; Leung, P.C. Vertebral Bone Mineral Density, Marrow Perfusion, and Fat Content in Healthy Men and Men with Osteoporosis: Dynamic Contrast-Enhanced MR Imaging and MR Spectroscopy. Radiology 2005, 236, 945–951. [Google Scholar] [CrossRef] [PubMed]
  248. Sadie-Van Gijsen, H.; Crowther, N.J.; Hough, F.S.; Ferris, W.F. The Interrelationship between Bone and Fat: From Cellular See-Saw to Endocrine Reciprocity. Cell Mol. Life Sci. 2013, 70, 2331–2349. [Google Scholar] [CrossRef] [PubMed]
  249. Rozman, C.; Feliu, E.; Berga, L.; Reverter, J.C.; Climent, C.; Ferran, M.J. Age-Related Variations of Fat Tissue Fraction in Normal Human Bone Marrow Depend Both on Size and Number of Adipocytes: A Stereological Study. Exp. Hematol. 1989, 17, 34–37. [Google Scholar] [PubMed]
  250. Allen, J.E.; Henshaw, D.L.; Keitch, P.A.; Fews, A.P.; Eatough, J.P. Fat Cells in Red Bone Marrow of Human Rib: Their Size and Spatial Distribution with Respect to the Radon-Derived Dose to the Haemopoietic Tissue. Int. J. Radiat. Biol. 1995, 68, 669–678. [Google Scholar] [CrossRef] [PubMed]
  251. Blebea, J.S.; Houseni, M.; Torigian, D.A.; Fan, C.; Mavi, A.; Zhuge, Y.; Iwanaga, T.; Mishra, S.; Udupa, J.; Zhuang, J.; et al. Structural and Functional Imaging of Normal Bone Marrow and Evaluation of Its Age-Related Changes. Semin. Nucl. Med. 2007, 37, 185–194. [Google Scholar] [CrossRef]
  252. Pansini, V.; Monnet, A.; Salleron, J.; Hardouin, P.; Cortet, B.; Cotten, A. 3 Tesla (1) H MR Spectroscopy of Hip Bone Marrow in a Healthy Population, Assessment of Normal Fat Content Values and Influence of Age and Sex. J. Magn. Reson. Imaging 2014, 39, 369–376. [Google Scholar] [CrossRef]
  253. Schwartz, R.S.; Shuman, W.P.; Bradbury, V.L.; Cain, K.C.; Fellingham, G.W.; Beard, J.C.; Kahn, S.E.; Stratton, J.R.; Cerqueira, M.D.; Abrass, I.B. Body Fat Distribution in Healthy Young and Older Men. J. Gerontol. 1990, 45, M181–M185. [Google Scholar] [CrossRef] [PubMed]
  254. Schwartz, A.V.; Sigurdsson, S.; Hue, T.F.; Lang, T.F.; Harris, T.B.; Rosen, C.J.; Vittinghoff, E.; Siggeirsdottir, K.; Sigurdsson, G.; Oskarsdottir, D.; et al. Vertebral Bone Marrow Fat Associated with Lower Trabecular BMD and Prevalent Vertebral Fracture in Older Adults. J. Clin. Endocrinol. Metab. 2013, 98, 2294–2300. [Google Scholar] [CrossRef] [PubMed]
  255. Martin, R.B.; Chow, B.D.; Lucas, P.A. Bone Marrow Fat Content in Relation to Bone Remodeling and Serum Chemistry in Intact and Ovariectomized Dogs. Calcif. Tissue Int. 1990, 46, 189–194. [Google Scholar] [CrossRef] [PubMed]
  256. Marie, P.J.; Kassem, M. Extrinsic mechanisms involved in age-related defective bone formation. J. Clin. Endocrinol. Metab. 2011, 96, 600–609. [Google Scholar] [CrossRef] [PubMed]
  257. Gasparrini, M.; Rivas, D.; Elbaz, A.; Duque, G. Differential Expression of Cytokines in Subcutaneous and Marrow Fat of Aging C57BL/6J Mice. Exp. Gerontol. 2009, 44, 613–618. [Google Scholar] [CrossRef] [PubMed]
  258. Singh, L.; Tyagi, S.; Myers, D.; Duque, G. Good, Bad, or Ugly: The Biological Roles of Bone Marrow Fat. Curr. Osteoporos. Rep. 2018, 16, 130–137. [Google Scholar] [CrossRef] [PubMed]
  259. Gunaratnam, K.; Vidal, C.; Gimble, J.M.; Duque, G. Mechanisms of Palmitate-Induced Lipotoxicity in Human Osteoblasts. Endocrinology 2014, 155, 108–116. [Google Scholar] [CrossRef] [PubMed]
  260. Kõks, S.; Dogan, S.; Tuna, B.G.; González-Navarro, H.; Potter, P.; Vandenbroucke, R.E. Mouse Models of Ageing and Their Relevance to Disease. Mech. Ageing Dev. 2016, 160, 41–53. [Google Scholar] [CrossRef] [PubMed]
  261. Gao, J.; Wang, Z.; Gao, P.; Fan, Q.; Zhang, T.; Cui, L.; Shi, L.; Liu, Z.; Yang, Z.; He, L.; et al. Daphnetin Alleviates Senile and Disuse Osteoporosis by Distinct Modulations of Bone Formation and Resorption. Antioxidants 2022, 11, 2365. [Google Scholar] [CrossRef]
  262. Wauquier, F.; Barquissau, V.; Léotoing, L.; Davicco, M.J.; Lebecque, P.; Mercier, S.; Philippe, C.; Miot-Noirault, E.; Chardigny, J.M.; Morio, B.; et al. Borage and Fish Oils Lifelong Supplementation Decreases Inflammation and Improves Bone Health in a Murine Model of Senile Osteoporosis. Bone 2012, 50, 553–561. [Google Scholar] [CrossRef]
  263. Hassan, E.B.; Alderghaffar, M.; Wauquier, F.; Coxam, V.; Demontiero, O.; Vogrin, S.; Wittrant, Y.; Duque, G. The Effects of Dietary Fatty Acids on Bone, Hematopoietic Marrow and Marrow Adipose Tissue in a Murine Model of Senile Osteoporosis. Aging 2019, 11, 7938–7947. [Google Scholar] [CrossRef] [PubMed]
  264. Paccou, J.; Hardouin, P.; Cotton, A.; Penel, G.; Cortet, B. The Role of Bone Marrow Fat in Skeletal Health: Usefulness and Perspectives for Clinicians. J. Clin. Endocrinol. Metab. 2015, 100, 3613–3621. [Google Scholar] [CrossRef] [PubMed]
  265. Dementia. Available online: https://www.who.int/news-room/fact-sheets/detail/dementia (accessed on 13 March 2024).
  266. Zeng, W.; Pirzgalska, R.M.; Pereira, M.M.A.; Kubasova, N.; Barateiro, A.; Seixas, E.; Lu, Y.H.; Kozlova, A.; Voss, H.; Martins, G.G.; et al. Sympathetic Neuro-Adipose Connections Mediate Leptin-Driven Lipolysis. Cell 2015, 163, 84–94. [Google Scholar] [CrossRef] [PubMed]
  267. Cardoso, F.; Klein Wolterink, R.G.J.; Godinho-Silva, C.; Domingues, R.G.; Ribeiro, H.; da Silva, J.A.; Mahú, I.; Domingos, A.I.; Veiga-Fernandes, H. Neuro-Mesenchymal Units Control ILC2 and Obesity via a Brain-Adipose Circuit. Nature 2021, 597, 410–414. [Google Scholar] [CrossRef] [PubMed]
  268. Morrison, S.F.; Nakamura, K. Central Mechanisms for Thermoregulation. Annu. Rev. Physiol. 2019, 81, 285–308. [Google Scholar] [CrossRef] [PubMed]
  269. Bettinetti-Luque, M.; Trujillo-Estrada, L.; Garcia-Fuentes, E.; Andreo-Lopez, J.; Sanchez-Varo, R.; Garrido-Sánchez, L.; Gómez-Mediavilla, Á.; López, M.G.; Garcia-Caballero, M.; Gutierrez, A.; et al. Adipose Tissue as a Therapeutic Target for Vascular Damage in Alzheimer’s Disease. Br. J. Pharmacol. 2024, 181, 840–878. [Google Scholar] [CrossRef] [PubMed]
  270. Biessels, G.J.; Despa, F. Cognitive Decline and Dementia in Diabetes Mellitus: Mechanisms and Clinical Implications. Nat. Rev. Endocrinol. 2018, 14, 591–604. [Google Scholar] [CrossRef] [PubMed]
  271. Dove, A.; Shang, Y.; Xu, W.; Grande, G.; Laukka, E.J.; Fratiglioni, L.; Marseglia, A. The Impact of Diabetes on Cognitive Impairment and Its Progression to Dementia. Alzheimers Dement. 2021, 17, 1769–1778. [Google Scholar] [CrossRef] [PubMed]
  272. de Bruijn, R.F.; Ikram, M.A. Cardiovascular risk factors and future risk of Alzheimer’s disease. BMC Med. 2014, 12, 130. [Google Scholar] [CrossRef]
  273. Kiliaan, A.J.; Arnoldussen, I.A.C.; Gustafson, D.R. Adipokines: A Link between Obesity and Dementia? Lancet Neurol. 2014, 13, 913–923. [Google Scholar] [CrossRef]
  274. Beydoun, M.A.; Beydoun, H.A.; Wang, Y. Obesity and central obesity as risk factors for incident dementia and its subtypes: A systematic review and meta-analysis. Obes. Rev. 2008, 9, 204–218. [Google Scholar] [CrossRef] [PubMed]
  275. Qizilbash, N.; Gregson, J.; Johnson, M.E.; Pearce, N.; Douglas, I.; Wing, K.; Evans, S.J.W.; Pocock, S.J. BMI and Risk of Dementia in Two Million People over Two Decades: A Retrospective Cohort Study. Lancet Diabetes Endocrinol. 2015, 3, 431–436. [Google Scholar] [CrossRef] [PubMed]
  276. Ward, M.A.; Carlsson, C.M.; Trivedi, M.A.; Sager, M.A.; Johnson, S.C. The Effect of Body Mass Index on Global Brain Volume in Middle-Aged Adults: A Cross Sectional Study. BMC Neurol. 2005, 5, 23. [Google Scholar] [CrossRef] [PubMed]
  277. Taki, Y.; Kinomura, S.; Sato, K.; Inoue, K.; Goto, R.; Okada, K.; Uchida, S.; Kawashima, R.; Fukuda, H. Relationship between Body Mass Index and Gray Matter Volume in 1,428 Healthy Individuals. Obesity 2008, 16, 119–124. [Google Scholar] [CrossRef] [PubMed]
  278. Debette, S.; Beiser, A.; Hoffmann, U.; DeCarli, C.; O’Donnell, C.J.; Massaro, J.M.; Au, R.; Himali, J.J.; Wolf, P.A.; Fox, C.S.; et al. Visceral Fat Is Associated with Lower Brain Volume in Healthy Middle-Aged Adults. Ann. Neurol. 2010, 68, 136–144. [Google Scholar] [CrossRef] [PubMed]
  279. Letra, L.; Santana, I.; Seiça, R. Obesity as a Risk Factor for Alzheimer’s Disease: The Role of Adipocytokines. Metab. Brain Dis. 2014, 29, 563–568. [Google Scholar] [CrossRef] [PubMed]
  280. Lee, E.B. Obesity, Leptin, and Alzheimer’s Disease. Ann. N. Y. Acad. Sci. 2011, 1243, 15–29. [Google Scholar] [CrossRef] [PubMed]
  281. Gustafson, D. Adiposity Indices and Dementia. Lancet Neurol. 2006, 5, 713–720. [Google Scholar] [CrossRef] [PubMed]
  282. Zhang, P.; Tian, B. Metabolic Syndrome: An Important Risk Factor for Parkinson’s Disease. Oxid. Med. Cell Longev. 2014, 2014, 729194. [Google Scholar] [CrossRef]
  283. Versini, M.; Jeandel, P.Y.; Rosenthal, E.; Shoenfeld, Y. Obesity in autoimmune diseases: Not a passive bystander. Autoimmun. Rev. 2014, 13, 981–1000. [Google Scholar] [CrossRef]
  284. Gustafson, D.; Rothenberg, E.; Blennow, K.; Steen, B.; Skoog, I. An 18-year follow-up of overweight and risk of Alzheimer disease. Arch. Intern. Med. 2003, 163, 1524–1528. [Google Scholar] [CrossRef]
  285. Guerrero-García, J.J.; Carrera-Quintanar, L.; López-Roa, R.I.; Márquez-Aguirre, A.L.; Rojas-Mayorquín, A.E.; Ortuño-Sahagún, D. Multiple Sclerosis and Obesity: Possible Roles of Adipokines. Mediat. Inflamm. 2016, 2016, 4036232. [Google Scholar] [CrossRef]
  286. Tolppanen, A.M.; Ngandu, T.; Kåreholt, I.; Laatikainen, T.; Rusanen, M.; Soininen, H.; Kivipelto, M. Midlife and Late-Life Body Mass Index and Late-Life Dementia: Results from a Prospective Population-Based Cohort. J. Alzheimer’s Dis. 2014, 38, 201–209. [Google Scholar] [CrossRef] [PubMed]
  287. Guo, D.H.; Yamamoto, M.; Hernandez, C.M.; Khodadadi, H.; Baban, B.; Stranahan, A.M. Visceral adipose NLRP3 impairs cognition in obesity via IL-1R1 on CX3CR1+ cells. J. Clin. Investig. 2020, 130, 1961–1976. [Google Scholar] [CrossRef]
  288. Sodhi, K.; Pratt, R.; Wang, X.; Lakhani, H.V.; Pillai, S.S.; Zehra, M.; Wang, J.; Grover, L.; Henderson, B.; Denvir, J.; et al. Role of Adipocyte Na,K-ATPase Oxidant Amplification Loop in Cognitive Decline and Neurodegeneration. iScience 2021, 24, 103262. [Google Scholar] [CrossRef] [PubMed]
  289. Oliveras-Cañellas, N.; Castells-Nobau, A.; de la Vega-Correa, L.; Latorre-Luque, J.; Motger-Albertí, A.; Arnoriaga-Rodriguez, M.; Garre-Olmo, J.; Zapata-Tona, C.; Coll-Martínez, C.; Ramió-Torrentà, L.; et al. Adipose tissue coregulates cognitive function. Sci. Adv. 2023, 9, eadg4017. [Google Scholar] [CrossRef]
  290. Petruzzelli, M.; Schweiger, M.; Schreiber, R.; Campos-Olivas, R.; Tsoli, M.; Allen, J.; Swarbrick, M.; Rose-John, S.; Rincon, M.; Robertson, G.; et al. A Switch from White to Brown Fat Increases Energy Expenditure in Cancer-Associated Cachexia. Cell Metab. 2014, 20, 433–447. [Google Scholar] [CrossRef]
  291. Vegiopoulos, A.; Rohm, M.; Herzig, S. Adipose Tissue: Between the Extremes. EMBO J. 2017, 36, 1999–2017. [Google Scholar] [CrossRef] [PubMed]
  292. Fearon, K.; Strasser, F.; Anker, S.D.; Bosaeus, I.; Bruera, E.; Fainsinger, R.L.; Jatoi, A.; Loprinzi, C.; MacDonald, N.; Mantovani, G.; et al. Definition and Classification of Cancer Cachexia: An International Consensus. Lancet Oncol. 2011, 12, 489–495. [Google Scholar] [CrossRef]
  293. Morley, J.E.; Thomas, D.R.; Wilson, M.M.G. Cachexia: Pathophysiology and Clinical Relevance. Am. J. Clin. Nutr. 2006, 83, 735–743. [Google Scholar] [CrossRef]
  294. von Haehling, S.; Anker, S.D. Prevalence, Incidence and Clinical Impact of Cachexia: Facts and Numbers-Update 2014. J. Cachexia Sarcopenia Muscle 2014, 5, 261–263. [Google Scholar] [CrossRef] [PubMed]
  295. Fearon, K.C.; Glass, D.J.; Guttridge, D.C. Cancer cachexia: Mediators, signaling, and metabolic pathways. Cell Metab. 2012, 16, 153–166. [Google Scholar] [CrossRef] [PubMed]
  296. Fouladiun, M.; Körner, U.; Bosaeus, I.; Daneryd, P.; Hyltander, A.; Lundholm, K.G. Body Composition and Time Course Changes in Regional Distribution of Fat and Lean Tissue in Unselected Cancer Patients on Palliative Care--Correlations with Food Intake, Metabolism, Exercise Capacity, and Hormones. Cancer 2005, 103, 2189–2198. [Google Scholar] [CrossRef] [PubMed]
  297. Das, S.K.; Eder, S.; Schauer, S.; Diwoky, C.; Temmel, H.; Guertl, B.; Gorkiewicz, G.; Tamilarasan, K.P.; Kumari, P.; Trauner, M.; et al. Adipose Triglyceride Lipase Contributes to Cancer-Associated Cachexia. Science 2011, 333, 233–238. [Google Scholar] [CrossRef] [PubMed]
  298. Kir, S.; White, J.P.; Kleiner, S.; Kazak, L.; Cohen, P.; Baracos, V.E.; Spiegelman, B.M. Tumour-derived PTH-related protein triggers adipose tissue browning and cancer cachexia. Nature 2014, 513, 100–104. [Google Scholar] [CrossRef] [PubMed]
  299. Blum, D.; Omlin, A.; Baracos, V.E.; Solheim, T.S.; Tan, B.H.; Stone, P.; Kaasa, S.; Fearon, K.; Strasser, F.; European Palliative Care Research Collaborative. Cancer cachexia: A systematic literature review of items and domains associated with involuntary weight loss in cancer. Crit. Rev. Oncol. Hematol. 2011, 80, 114–144. [Google Scholar] [CrossRef] [PubMed]
  300. Rohm, M.; Schäfer, M.; Laurent, V.; Üstünel, B.E.; Niopek, K.; Algire, C.; Hautzinger, O.; Sijmonsma, T.P.; Zota, A.; Medrikova, D.; et al. An AMP-activated protein kinase-stabilizing peptide ameliorates adipose tissue wasting in cancer cachexia in mice. Nat. Med. 2016, 22, 1120–1130. [Google Scholar] [CrossRef] [PubMed]
  301. Kersten, S. Mechanisms of Nutritional and Hormonal Regulation of Lipogenesis. EMBO Rep. 2001, 2, 282–286. [Google Scholar] [CrossRef]
  302. Trew, J.A.; Begg, R.W. In Vitro Incorporation of Acetate-1-C14 into Adipose Tissue from Normal and Tumor-Bearing Rats. Cancer Res. 1959, 19, 1014–1019. [Google Scholar]
  303. Lanza-Jacoby, S.; Lansey, S.C.; Miller, E.E.; Cleary, M.P. Sequential Changes in the Activities of Lipoprotein Lipase and Lipogenic Enzymes during Tumor Growth in Rats. Cancer Res. 1984, 44, 5062–5067. [Google Scholar]
  304. Mulligan, H.D.; Tisdale, M.J. Lipogenesis in Tumour and Host Tissues in Mice Bearing Colonic Adenocarcinomas. Br. J. Cancer 1991, 63, 719–722. [Google Scholar] [CrossRef] [PubMed]
  305. Mayer, N.; Schweiger, M.; Romauch, M.; Grabner, G.F.; Eichmann, T.O.; Fuchs, E.; Ivkovic, J.; Heier, C.; Mrak, I.; Lass, A.; et al. Development of Small-Molecule Inhibitors Targeting Adipose Triglyceride Lipase. Nat. Chem. Biol. 2013, 9, 785–787. [Google Scholar] [CrossRef] [PubMed]
  306. Taylor, J.; Uhl, L.; Moll, I.; Hasan, S.S.; Wiedmann, L.; Morgenstern, J.; Giaimo, B.D.; Friedrich, T.; Alsina-Sanchis, E.; De Angelis Rigotti, F.; et al. Endothelial Notch1 Signaling in White Adipose Tissue Promotes Cancer Cachexia. Nat. Cancer 2023, 4, 1544–1560. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the sequence of events occurring in hyperplastic and hypertrophic obesity. sWAT = subcutaneous white adipose tissue; vWAT = visceral white adipose tissue; T2D = type 2 diabetes; CVD = cardiovascular disease.
Figure 1. Schematic representation of the sequence of events occurring in hyperplastic and hypertrophic obesity. sWAT = subcutaneous white adipose tissue; vWAT = visceral white adipose tissue; T2D = type 2 diabetes; CVD = cardiovascular disease.
Cells 13 00763 g001
Figure 2. Schematic representation of the adipogenesis process. BMP = bone morphogenetic protein; Wnt = wingless-type MMTV integration site; Hh = Hedgehog; KLF = Krupel-like factors; Pref = preadipocyte factor; Rb = retinoblastoma cell-cycle-related protein; E2F = E2 transcription factor; CREB = cyclic AMP response element-binding protein; C/EBP = CCATT enhancer binding protein; PPAR = peroxisome proliferator-activated receptor; SREBP = sterol regulatory element-binding protein; MSC = mesenchymal stem cell. Positive and negative regulators of adipogenesis are shown in green and red ovals, respectively.
Figure 2. Schematic representation of the adipogenesis process. BMP = bone morphogenetic protein; Wnt = wingless-type MMTV integration site; Hh = Hedgehog; KLF = Krupel-like factors; Pref = preadipocyte factor; Rb = retinoblastoma cell-cycle-related protein; E2F = E2 transcription factor; CREB = cyclic AMP response element-binding protein; C/EBP = CCATT enhancer binding protein; PPAR = peroxisome proliferator-activated receptor; SREBP = sterol regulatory element-binding protein; MSC = mesenchymal stem cell. Positive and negative regulators of adipogenesis are shown in green and red ovals, respectively.
Cells 13 00763 g002
Figure 3. WAT changes during aging and their significance for its function and overall health. WAT, white adipose tissue; APCs, adipocyte precursor cells; T2D, type 2 diabetes.
Figure 3. WAT changes during aging and their significance for its function and overall health. WAT, white adipose tissue; APCs, adipocyte precursor cells; T2D, type 2 diabetes.
Cells 13 00763 g003
Figure 4. Involvement of WAT dysfunction in common pathologies of the elderly and potential therapies aimed at mitigating age-related changes in WAT or preventing aging pathologies. WAT, white adipose tissue; MSCs, mesenchymal stem cells; TZDs, thiazolidinediones; ATGL, adipose triglyceride lipase.
Figure 4. Involvement of WAT dysfunction in common pathologies of the elderly and potential therapies aimed at mitigating age-related changes in WAT or preventing aging pathologies. WAT, white adipose tissue; MSCs, mesenchymal stem cells; TZDs, thiazolidinediones; ATGL, adipose triglyceride lipase.
Cells 13 00763 g004
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

Iacobini, C.; Vitale, M.; Haxhi, J.; Menini, S.; Pugliese, G. Impaired Remodeling of White Adipose Tissue in Obesity and Aging: From Defective Adipogenesis to Adipose Organ Dysfunction. Cells 2024, 13, 763. https://doi.org/10.3390/cells13090763

AMA Style

Iacobini C, Vitale M, Haxhi J, Menini S, Pugliese G. Impaired Remodeling of White Adipose Tissue in Obesity and Aging: From Defective Adipogenesis to Adipose Organ Dysfunction. Cells. 2024; 13(9):763. https://doi.org/10.3390/cells13090763

Chicago/Turabian Style

Iacobini, Carla, Martina Vitale, Jonida Haxhi, Stefano Menini, and Giuseppe Pugliese. 2024. "Impaired Remodeling of White Adipose Tissue in Obesity and Aging: From Defective Adipogenesis to Adipose Organ Dysfunction" Cells 13, no. 9: 763. https://doi.org/10.3390/cells13090763

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