The process by which multipotent cells commit to differentiate into distinct cell types, eventually forming functional tissues and organisms, has fascinated scientists for decades. Consequently, numerous studies have contributed to our understanding of how transcription factors and signaling molecules regulate differentiation. A growing area of interest in the field centers around the role of nutrients and metabolic pathways in cell fate determination. This review focuses on adipogenesis (also termed hyperplasia), the formation of adipocytes, which are key sensors of nutrient availability. We will examine recent findings that reshape our understanding of how nucleotide metabolism regulates adipogenesis.

The identification of fat tissue as a distinct anatomical and physiological entity dates back to ancient medical traditions, including the Ayurvedic concept of meda dhatu and Hippocratic discussions of corpulence as a disease state [1,2]. Scientific recognition of adipose tissue as a specific tissue type emerged during the nineteenth century, particularly with Arthur Hassall’s 1849 microscopic description of fat cells—then termed ‘fat vesicles’—marking the beginning of its formal anatomical classification [3,4]. Adipose tissue’s role was thought to be limited to energy storage and insulation until the mid-twentieth century, when the metabolic importance of adipose tissue began to emerge. In 1948, Shapiro and Wertheimer, in a review article, highlighted evidence that adipose tissue is innervated by sympathetic nerve fibers, possesses a dense network of capillaries, and that lipid storage and mobilization are highly regulated within the tissue [5]. This characterization introduced an important concept that adipose tissue is an active participant in organismal physiology.

Throughout the latter half of the twentieth century, the physiological relevance of adipose tissue was further revealed. A pivotal breakthrough came in 1994 when Friedman and colleagues identified leptin, the first known adipokine, while studying the ob (obese) gene, which encodes this hormone. Mutations in the leptin gene were found to cause severe obesity in mice [6]. This discovery redefined adipose tissue as a key endocrine organ, highlighting its role in regulating energy homeostasis, appetite, and metabolism.

Building on the importance of adipose tissue as an endocrine organ, another fundamental role is its ability to prevent hepatic and muscular lipotoxicity by safely storing excess nutrients. Excess energy intake, when not matched by energy expenditure, leads to the storage of surplus nutrients in adipose tissue, expanding fat depots. This expansion occurs through two mechanisms: hypertrophy, the enlargement of preexisting adipocytes through lipid accumulation, and adipogenesis, the formation of new adipocytes from the proliferation and differentiation of precursor cells, both of which are extensively characterized in the literature [7]. The balance between these mechanisms is governed by interactions between diet and genetics [8]. In obesity-mediated metabolic dysfunction, hypertrophy is linked to negative health effects, such as insulin resistance and fibrosis [9]. In contrast, increased adipogenesis is associated with improved insulin sensitivity and overall metabolic health [10]. Given the beneficial aspects of adipogenesis, many research studies have focused on identifying the regulatory mechanisms that govern this process, with the goal of mitigating the adverse effects of obesity.

Transcriptional and signaling regulation of adipogenesis

Adipogenesis involves two distinct phases that include mitotic clonal expansion (MCE) and lipid accumulation (Figure 1). These events are temporally co-ordinated by key transcriptional and signaling events. Preadipocytes, stimulated by insulin, glucocorticoids, or other proliferative agents [11,12], undergo multiple rounds of MCE [13,14] before terminal differentiation [15,16]. MCE is controlled at multiple levels, including regulation by transcription factors, DNA replication, cell cycle progression, and mitogenic signaling. The transcription factors c-Jun, c-Fos, and c-Myc play distinct, yet complementary, roles in promoting adipogenesis. c-Myc primarily facilitates DNA replication by up-regulating genes involved in DNA synthesis and chromatin remodeling. c-Jun and c-Fos, components of the AP-1 complex, drive cell cycle progression by enhancing the transcription of cyclins and other cell cycle regulators. Together, these factors contribute to mitogenic signaling, integrating extracellular growth cues to activate downstream pathways, such as mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/Akt, that support adipocyte proliferation and differentiation [17–19]. Cyclins and cyclin-dependent kinases regulate cell cycle progression, and their activity influences adipogenesis [20]. The expression of genes that regulate cell cycle progression is mediated by CCAAT/enhancer binding proteins (C/EBPs), C/EBPβ, and C/EBPδ, early transcriptional regulators.

Regulation of adipogenesis by nucleotides.

Figure 1:
Regulation of adipogenesis by nucleotides.

In early adipogenesis, cAMP regulates transcription through the activation of protein kinase A (PKA), leading to the phosphorylation of cAMP response element-binding protein (CREB). Nicotinamide mononucleotide (NMN) accumulation promotes the activity of nicotinamide mononucleotide adenylyl transferases (NMNAT2), resulting in the inhibition of anti-adipogenic enzyme polymerase 1 (PARP1) and the increased production of NAD+. NAD acts as a cofactor for sirtuin 4 (SIRT4), an enzyme that promotes mitochondrial fuel oxidation and promotes proliferator-activated receptor gamma (PPARγ). NAD accumulation results in increased α-ketoglutarate (αKG), leading to the demethylation of histone H3K9 and increased adipogenic transcription. During MCE, cyclic nucleotides and nucleobases inhibit Wnt and Yap signaling to increase adipogenic transcription and support DNA replication, respectively. In late differentiation, nucleotides act as cofactors for enzymes that promote insulin receptor signaling, enhance PPARγ and CEBPα, and support lipogenesis. ATP also provides energy to demanding processes like protein synthesis. Nicotinamide adenine dinucleotide phosphate (NADPH) may support adipogenesis by modulating the levels of reactive oxygen species (ROS). cGMP promotes adipogenic transcription via cGMP-dependent kinase (PKG) activation.

Figure 1:
Regulation of adipogenesis by nucleotides.

In early adipogenesis, cAMP regulates transcription through the activation of protein kinase A (PKA), leading to the phosphorylation of cAMP response element-binding protein (CREB). Nicotinamide mononucleotide (NMN) accumulation promotes the activity of nicotinamide mononucleotide adenylyl transferases (NMNAT2), resulting in the inhibition of anti-adipogenic enzyme polymerase 1 (PARP1) and the increased production of NAD+. NAD acts as a cofactor for sirtuin 4 (SIRT4), an enzyme that promotes mitochondrial fuel oxidation and promotes proliferator-activated receptor gamma (PPARγ). NAD accumulation results in increased α-ketoglutarate (αKG), leading to the demethylation of histone H3K9 and increased adipogenic transcription. During MCE, cyclic nucleotides and nucleobases inhibit Wnt and Yap signaling to increase adipogenic transcription and support DNA replication, respectively. In late differentiation, nucleotides act as cofactors for enzymes that promote insulin receptor signaling, enhance PPARγ and CEBPα, and support lipogenesis. ATP also provides energy to demanding processes like protein synthesis. Nicotinamide adenine dinucleotide phosphate (NADPH) may support adipogenesis by modulating the levels of reactive oxygen species (ROS). cGMP promotes adipogenic transcription via cGMP-dependent kinase (PKG) activation.

Close modal

C/EBPβ and C/EBPδ also support the lipid accumulation part of adipogenesis by activating peroxisome proliferator-activated receptor gamma (PPARγ). PPARγ exists in two isoforms, with PPARγ2 demonstrating increased fat specificity compared with PPARγ1. PPARγ induces C/EBPα expression, which then generates a positive feedback loop through a C/EBPα-mediated increase in PPARγ expression. This interaction between PPARγ and C/EBPα ensures the maintained expression of adipocyte-specific genes, for example, adiponectin [AdipoQ, a hormone that enhances insulin sensitivity and fatty acid oxidation (FAO)], fatty acid binding protein 4 (FABP4, facilitates intracellular lipid transport), and lipoprotein lipase (essential for hydrolyzing circulating triglycerides into free fatty acids for uptake and storage) [21].

Hormonal cues play a critical role in reinforcing this transcriptional network. In addition to these core factors, insulin—secreted by pancreatic β-cells in response to rising blood glucose levels—is a key inducer of adipogenesis, primarily acting through the insulin receptor and activating downstream signaling pathways involving IRS1/2, PI3K, and AKT. This cascade promotes glucose uptake and lipogenesis, supporting the growth and function of adipocytes. Insulin also stimulates the expression of key adipogenic transcription factors [22,23] and is important for maintaining adipocyte function and suppressing lipolysis [24]. Insulin induces sterol regulatory element-binding protein, promoting PPARγ expression and adipogenesis [21]. As insulin levels rise postprandially, they signal nutrient availability, triggering adipocyte differentiation to increase lipid storage capacity.

Glucocorticoids, including cortisol, signal to regulate adipogenesis through the glucocorticoid receptor (GR). Glucocorticoid-GR signaling induces CEBPβ, which subsequently activates PPARγ. However, the effects of glucocorticoids are complex. While insulin may enhance the pro-adipogenic effects of glucocorticoids, excessive glucocorticoid signaling can disrupt normal lipid metabolism and promote lipolysis in mature adipocytes [25,26].

Additional signaling pathways also influence adipogenesis. Bone morphogenetic proteins (BMPs) and Wingless-related integration site (WNT) signaling have opposing functions in adipogenesis. For example, BMP4 signaling activates Sma and Mad related (SMAD) proteins that increase the expression of PPARγ [27]. In contrast, Wnt10b stabilizes β-catenin, suppressing the activity of PPARγ [28,29].

Finally, cytoskeletal remodeling supports morphological maturation of adipocytes. As preadipocytes differentiate, changes in cytoskeletal structure, including the disassembly of actin filaments and microtubules, facilitate the morphological changes required for adipocyte formation. Ras homolog family member A (RhoA) signaling promotes cytoskeletal tension, and disruption of RhoA activity can enhance adipogenesis, suggesting that cytoskeletal reorganization is an important early event in the differentiation process [30,31].

Metabolic regulation of adipogenesis

Metabolic regulators also contribute to adipogenesis [32], co-ordinating adipocyte differentiation with the organism’s energy status. Through the pentose phosphate pathway (PPP), glucose promotes adipocyte maturation by generating cofactors essential for lipogenesis [33]. Branched-chain amino acids (BCAAs)—leucine, isoleucine, and valine—serve as a source of lipogenic acetyl-CoA through mitochondrial transamination and oxidation [34,35]. BCAA oxidation is regulated by the mitochondrial sirtuin, SIRT4, which enhances adipocyte differentiation by increasing PPARγ activity [36]. Nuclear acetyl-CoA levels modulate histone acetylation, and its reduced availability inhibits adipocyte differentiation [37]. These studies highlight the mechanisms by which nutrient oxidation supports adipogenesis.

Nucleotides take on many functional roles: precursors for DNA/RNA synthesis, cofactors for metabolic redox reactions, and signaling molecules. Nucleotides are important for the synthesis of DNA and RNA, and as signaling molecules that promote cellular proliferation required to support adipogenesis. DNA replication is required for proliferation [38–40], and impairing DNA synthesis or regulators of DNA replication impairs adipogenesis [41–43]. Across cell types, glutamine supports DNA replication in proliferation, and glutamine-depleted culture medium impairs proliferation [44]. The addition of adenine, adenosine, inosine, and hypoxanthine overcomes glutamine depletion, restoring DNA synthesis and proliferation in fibroblasts [38]. In adipogenesis, inhibition of de novo purine and pyrimidine biosynthesis reduces cell number, suppressing adipogenesis, consistent with impaired MCE [45,46]. These studies suggest that nucleotide availability regulates proliferation in adipogenesis. Extracellular purines can also bind to receptors on the plasma membrane and induce signaling cascades in an autocrine or paracrine manner [47]. Nucleotides regulate many adipogenic processes, such as the induction of adipogenesis, MCE, and lipogenesis (Figure 1).

In quiescent or non-proliferating cells, the nucleotide salvage pathway is primarily responsible for maintaining intracellular nucleotide levels, whereas during times of growth, cells increase de novo nucleotide synthesis [48,49]. In de novo purine synthesis, phosphoribosyl pyrophosphate (PRPP) contributes a ribose sugar and a phosphate, while the addition of carbons and nitrogen from glycine, glutamine, aspartate, and N10-formyl-tetrahydrofolate ultimately leads to the generation of inosine monophosphate (IMP) (Figure 2A). IMP can then be converted to adenosine monophosphate (AMP) or to guanosine monophosphate (GMP) [50]. Through the salvage pathway, nucleosides and nucleobases can be recycled to produce AMP, GMP, or IMP. In the de novo synthesis of pyrimidines, the pyrimidine ring is first synthesized before the incorporation of PRPP to generate uridine monophosphate (UMP) [51,52]. UMP is then used to generate cytidine monophosphate (CMP) and thymidine monophosphate (TMP) [50,53]. Pyrimidines can also be generated via the salvage pathway, which involves the recycling of nucleosides. These nucleosides can be derived from the catabolism of nucleotides within the cell or obtained through extracellular uptake mediated by concentrative nucleoside transporters and equilibrative nucleoside transporters [54–57].

Nucleotide biosynthesis in adipogenesis.

Figure 2:
Nucleotide biosynthesis in adipogenesis.

(A) Purine and pyrimidine biosynthetic pathways. Red arrows indicate the salvage pathway, and blue arrows indicate the de novo pathway. (B) Under conditions that stimulate adipogenesis, acetyl-CoA is derived from glucose and BCAAs, generating ATP via oxidative phosphorylation (OXPHOS), and performing lipogenesis using citrate from the TCA cycle. Under purine and pyrimidine inhibition, flux of pyruvate to acetyl-CoA is disrupted and FAO is elevated. Likewise, lipogenesis and OXPHOS are suppressed. Nucleotide inhibition results in arrested adipogenesis.

Figure 2:
Nucleotide biosynthesis in adipogenesis.

(A) Purine and pyrimidine biosynthetic pathways. Red arrows indicate the salvage pathway, and blue arrows indicate the de novo pathway. (B) Under conditions that stimulate adipogenesis, acetyl-CoA is derived from glucose and BCAAs, generating ATP via oxidative phosphorylation (OXPHOS), and performing lipogenesis using citrate from the TCA cycle. Under purine and pyrimidine inhibition, flux of pyruvate to acetyl-CoA is disrupted and FAO is elevated. Likewise, lipogenesis and OXPHOS are suppressed. Nucleotide inhibition results in arrested adipogenesis.

Close modal

cAMP

Cyclic adenosine monophosphate (cAMP) is synthesized from adenosine triphosphate (ATP) by the enzyme adenylate cyclase [58]. cAMP acts as a second messenger in signal transduction to mediate the effects of hormones, neurotransmitters, and inflammatory molecules to promote adipogenesis [59,60]. G-protein coupled receptors, activated during adipogenesis, enhance cAMP synthesis [61], promoting adipogenesis and altering adipocyte metabolism [62]. Elevated cAMP levels suppress the expression of anti-adipogenic factors like WNTs and Yes-associated protein (YAP), allowing the progression of adipogenesis after MCE [63–66]. cAMP also activates protein kinase A (PKA), promoting its nuclear translocation and allowing for the phosphorylation of cAMP response element-binding protein (CREB) [67,68]. Phosphorylated CREB then promotes the expression of the adipogenic transcriptional cascade [69,70].

cGMP

cGMP, another cyclic nucleotide, mediates nitric oxide signaling [71], which regulates adipogenesis through distinct mechanisms from those of cAMP. Nitric oxide is synthesized from l-arginine by nitric oxide synthase enzymes [72]. NO diffuses freely across membranes and activates soluble guanylyl cyclase (sGC), leading to the production of cGMP [73]. Elevated cGMP levels activate PKG, up-regulating the expression of PPARγ, CEBPα, and genes in lipid metabolism, increasing triglyceride content in differentiating adipocytes [74].

ATP

In adipogenesis, ATP is primarily generated through increased glycolysis and mitochondrial oxidation of BCAAs, which are up-regulated to meet the heightened energy demands. In addition, mitochondrial biogenesis and its enhanced activity also contribute to the elevated ATP supply [75]. ATP is not only an essential energy source for the biosynthetic processes involved in adipogenesis but also functions as a key cofactor for various enzymes and as a signaling molecule that modulates adipogenic processes [76]. Protein translation is one of the most energetically demanding processes [77], and adipocytes display a two-fold increase in protein levels during terminal differentiation, demonstrating a need for ATP [78]. ATP is required as a cofactor by several enzymes involved in de novo lipogenesis, including ATP citrate lyase (ACLY) and acetyl-CoA carboxylase, both of which are central to lipid biosynthesis and adipocyte maturation [79]. Finally, ATP can also regulate adipogenesis through purinergic signaling via the P2Y receptor, for example [76].

NADPH

Nicotinamide adenine dinucleotide phosphate (NADPH) is generated through several pathways in adipogenesis, including the oxidative phase of the PPP [80], malic enzyme, which produces NADPH while converting malate to pyruvate, and isocitrate dehydrogenase, which generates NADPH during the conversion of isocitrate to α-ketoglutarate. A key function of NADPH is its role as an essential cofactor for lipogenesis [33]. Additionally, NADPH contributes to maintaining redox homeostasis by supporting glutathione and thioredoxin production, which are critical for mitigating ROS during adipogenesis [81].

NAD

Nicotinamide adenine dinucleotide (NAD+) plays a key role in redox reactions, acting as a cofactor for various enzymes that regulate metabolism and transcription [82]. NAD+ is salvaged from nicotinamide (NAM), nicotinic acid, and nicotinamide ribose and is also generated de novo from tryptophan. In the NAD salvage pathway, NAM phosphoribosyltransferase (NAMPT) catalyzes the conversion of NAM into nicotinamide mononucleotide (NMN) using 5′-PRPP, which is then converted to NAD+ by nicotinamide mononucleotide adenylyl transferases (NMNATs) [83].

NAMPT not only aids in reprogramming metabolism in adipogenesis, promoting both glycolysis and mitochondrial respiration, but also regulates adipogenesis through epigenetic modifications. By increasing NAD+ levels and enhancing α-ketoglutarate availability, NAMPT promotes demethylation of histone H3K9 and increases the transcription of adipogenic transcription factors PPARγ and CEBPα [84].

NMNATs exhibit differential subcellular localizations: NMNAT-1 is nuclear, NMNAT-2 is cytoplasmic, and NMNAT-3 is mitochondrial. The synthesis of NAD+ in different cellular compartments has significant effects on adipogenesis. Knockdown of NMNAT1 promotes adipogenesis and lipid accumulation by decreasing the activity of poly(ADP-ribose) polymerase 1 (PARP1) [85], an inhibitor of CEBPβ [86]. NMNAT2 is up-regulated early in adipogenesis and supports differentiation by both limiting substrate availability for nuclear NMNAT1 and providing NAD+ to support increased glucose metabolism [85]. Additionally, increased mitochondrial NAD+ may increase substrate availability for SIRT4, a positive regulator of PPARγ in early adipogenesis [36]. In contrast, reduced nuclear NAD+ down-regulates SIRT1, a negative adipogenic regulator [87].

Given the significance of nucleotides in adipogenesis and the therapeutic use of nucleotide biosynthesis inhibitors, several studies evaluated how blocking nucleotide biosynthesis altered cellular metabolism and the effect thereof on adipogenesis. First, Shinde and Nunn showed that pyrimidine and purine metabolites are elevated during adipogenesis. Blocking purine or pyrimidine biosynthesis blocked adipogenesis by suppressing the PPARγ−C/EBPα transcriptional program [88] (Figure 2B). While purine depletion has been shown to block the mechanistic target of rapamycin complex 1 (mTORC1) activation in proliferating cells, it had no effect on mTORC1 activity in adipogenic models [88–90]. Rather, inhibition of purine biosynthesis activated AMP-activated protein kinase (AMPK), although the phosphorylation of its substrates was not induced. Furthermore, the expression of lipogenesis and lipolysis genes was reduced, likely due to the suppression of PPARγ [88]. Importantly, interfering with purine availability through genetic deletion of inosine monophosphate dehydrogenase 2 disrupted adipose expansion in vivo [91].

Next, the Ben-Sahra group has highlighted that pyrimidines maintain pyruvate oxidation and the tricarboxylic citric acid (TCA) cycle in mitochondria through the regulation of pyruvate dehydrogenase (PDH) to support de novo lipogenesis and adipocyte differentiation [92]. Importantly, the enzyme thiamine pyrophosphate kinase 1 (TPK1), which is crucial for synthesizing thiamine pyrophosphate (TPP) from thiamine, was found to be regulated by pyrimidine concentrations [92] (Figure 2B). This contrasts with previous assumptions that TPK1 predominantly uses ATP for this reaction, revealing that the pyrimidine UTP—and not purines or other pyrimidines like CTP—is key in limiting cellular TPP synthesis [92–94]. Given that TPP is an essential cofactor for PDH, the enzyme that converts pyruvate into acetyl-CoA, UTP indirectly regulates pyruvate catabolism [92]. Through this mechanism, UTP significantly contributes to modulating metabolic processes vital for energy homeostasis and cell growth. When UTP is depleted (either by inhibiting pyrimidine synthesis with small molecules like brequinar or by knocking down genes in the pyrimidine pathway), PDH activity is impaired, and less acetyl-CoA is generated, thereby curtailing the substrate availability for fatty acid synthesis [92]. Furthermore, inhibition of pyrimidine biosynthesis increases FAO, suggesting compensation for the energy shortfall due to impaired pyruvate processing [92]. These findings are further supported by a study that showed that blocking purine or pyrimidine biosynthesis decreased glucose utilization for mitochondrial oxidative phosphorylation (OXPHOS) while increasing FAO [45] (Figure 2B). Inhibition of FAO in the presence of purine or pyrimidine biosynthesis inhibitors restored adipogenesis and mitochondrial activity [45].

Nucleotide-mediated activation of mitochondrial metabolism also drives citrate production. Citrate exported from the mitochondria into the cytosol is cleaved by ACLY to generate acetyl-CoA, a key substrate for de novo fatty acid synthesis, as well as histone acetylation [37]. The availability of acetyl-CoA directly influences lipid accumulation in differentiating adipocytes and modulates gene expression through epigenetic mechanisms that support the adipogenic transcriptional program. As such, ACLY-knockout results in repressed expression of PPARγ, adiponectin, and FABP4 [95]. Beyond histone acetylation, other epigenetic modifications, such as histone and DNA methylation, are also influenced by cellular metabolism and contribute significantly to adipogenesis. For instance, the methyl donor S-adenosylmethionine (SAM), derived from ATP and methionine, is required for both DNA and histone methylation and has been shown to regulate adipocyte gene expression. SAM enhances PPARγ expression and promotes lipid accumulation, partly through the suppression of the Wnt/β-catenin and Hedgehog signaling pathways [96]. Moreover, dynamic changes in histone methylation at key regulatory loci—such as demethylation of H3K27me3 and enrichment of H3K4me3—have been shown to facilitate transcriptional activation of adipogenic genes during differentiation [97,98]. DNA methylation also plays a repressive role early in adipogenesis, with demethylation at promoters of Cebpα and Pparγ correlating with their transcriptional activation [99]. Together, these studies reveal that nucleotide availability is not only essential for sustaining mitochondrial metabolism and biosynthetic processes but also critically governs the transcriptional and epigenetic networks that drive adipocyte differentiation and lipid accumulation.

In summary, the process of adipogenesis is orchestrated through a complex interplay of transcriptional regulation, signaling pathways, and metabolism. Metabolic regulators, including the PPP and the metabolism of BCAAs, integrate the energy status of the cell with adipocyte differentiation. Likewise, nucleotides play significant signaling roles beyond their canonical functions in DNA and RNA synthesis, including regulating critical processes such as lipogenesis (Table 1). Recent findings have shown that pyrimidine and purine levels distinctly affect TCA cycle and glycolytic intermediates, with pyrimidine depletion impairing pyruvate processing and purine depletion altering mitochondrial function.

Table 1:
Roles of nucleotide metabolism in adipogenesis: Summary of critical mechanisms by which nucleotides modulate adipogenesis.
Metabolite/metabolic pathwayRegulatory roleEffect on adipogenesis
De novo purine biosynthesis Generates IMP used to produce AMP and GMP. Inhibition reduces adipocyte proliferation and suppresses PPARγ, impairing adipogenesis [45,46,88]. 
De novo pyrimidine biosynthesis Produces UMP for CMP and TMP synthesis; regulates TCA cycle via UTP and TPP. Pyrimidine depletion reduces pyruvate oxidation, impairs lipogenesis, and shifts cells toward FAO [92]. 
Pentose phosphate pathway Generates NADPH and ribose sugars for nucleotide synthesis and lipogenesis. Enhances fatty acid synthesis during adipocyte maturation [33]. 
ATP Provides energy and cofactors for biosynthetic processes and purinergic signaling. Required for lipogenesis, protein translation, and adipocyte differentiation [76,78]. 
NADPH Essential cofactor for lipogenesis; maintains redox homeostasis during differentiation. Supports lipid biosynthesis and mitigates ROS in adipocytes [33,80]. 
NAD+ Acts as a cofactor for redox reactions and regulates epigenetic modifications. Promotes metabolism through NAMPT and NMNAT activity; supports PPARγ and CEBPα expression via histone remodeling [84]. 
cAMP Second messenger generated by adenylate cyclase; activates PKA and CREB. Promotes adipogenic transcriptional cascades, suppresses anti-adipogenic factors like WNTs, and supports differentiation [60–70]. 
cGMP Second messenger generated by NO-sGC signaling; activates PKG. Up-regulates lipid metabolism genes and PPARγ/CEBPα expression, increasing triglyceride content in adipocytes [71–73]. 
Thiamine pyrophosphate (TPP) Cofactor for PDH regulated by UTP concentration via TPK1 activity. UTP controls acetyl-CoA production through PDH, modulating fatty acid synthesis [92
Metabolite/metabolic pathwayRegulatory roleEffect on adipogenesis
De novo purine biosynthesis Generates IMP used to produce AMP and GMP. Inhibition reduces adipocyte proliferation and suppresses PPARγ, impairing adipogenesis [45,46,88]. 
De novo pyrimidine biosynthesis Produces UMP for CMP and TMP synthesis; regulates TCA cycle via UTP and TPP. Pyrimidine depletion reduces pyruvate oxidation, impairs lipogenesis, and shifts cells toward FAO [92]. 
Pentose phosphate pathway Generates NADPH and ribose sugars for nucleotide synthesis and lipogenesis. Enhances fatty acid synthesis during adipocyte maturation [33]. 
ATP Provides energy and cofactors for biosynthetic processes and purinergic signaling. Required for lipogenesis, protein translation, and adipocyte differentiation [76,78]. 
NADPH Essential cofactor for lipogenesis; maintains redox homeostasis during differentiation. Supports lipid biosynthesis and mitigates ROS in adipocytes [33,80]. 
NAD+ Acts as a cofactor for redox reactions and regulates epigenetic modifications. Promotes metabolism through NAMPT and NMNAT activity; supports PPARγ and CEBPα expression via histone remodeling [84]. 
cAMP Second messenger generated by adenylate cyclase; activates PKA and CREB. Promotes adipogenic transcriptional cascades, suppresses anti-adipogenic factors like WNTs, and supports differentiation [60–70]. 
cGMP Second messenger generated by NO-sGC signaling; activates PKG. Up-regulates lipid metabolism genes and PPARγ/CEBPα expression, increasing triglyceride content in adipocytes [71–73]. 
Thiamine pyrophosphate (TPP) Cofactor for PDH regulated by UTP concentration via TPK1 activity. UTP controls acetyl-CoA production through PDH, modulating fatty acid synthesis [92

CREB, cAMP response element-binding protein; FAO, fatty acid oxidation; NADPH, nicotinamide adenine dinucleotide phosphate; PDH, pyruvate dehydrogenase; PKA, protein kinase A; PPARγ, proliferator-activated receptor gamma; PKG, cGMP-dependent kinase; ROS, reactive oxygen species; TPP, thiamine pyrophosphate; TPK1, thiamine pyrophosphate kinase 1.

Disrupting nucleotide biosynthesis arrests adipogenesis, which may be detrimental to metabolic health given adipogenesis’ protective effects [10]. However, recent work has shown that in vivo pharmacological inhibition of purine biosynthesis blocks fat tissue expansion by decreasing caloric intake and increasing energy expenditure through skeletal muscle thermogenesis [100]. This may confer protection against the adverse effects of chronic caloric surplus, such as insulin resistance and ectopic lipid accumulation. Several small-molecule inhibitors of purine and pyrimidine metabolism are currently in clinical use for treating rheumatoid arthritis, Crohn’s disease, ulcerative colitis, and numerous cancers [101–108]. The effects of these drugs on body weight extend beyond their intended therapeutic use as immunosuppressants and anticancer agents, and therefore, further research is needed to understand the risks and benefits associated with their use. Additional studies are needed to examine the specific cell type-dependent effects of purine and pyrimidine depletion on cellular physiology.

Perspectives

  • Adipogenesis is an important protective mechanism that allows for the safe storage of excess nutrients. Given the alarming rise in obesity, there is much interest in understanding the regulation of this process. Here, we summarize the roles of nucleotides in adipogenesis.

  • Beyond supporting DNA replication and cellular proliferation, studies have also shown the role of nucleotides in the regulation of signaling and transcription mechanisms required for adipogenesis. More recently, purines and pyrimidines were found to be essential for metabolic reprogramming that stimulates adipogenesis.

  • Inhibitors of nucleotide biosynthesis are used to treat various disease states. Future studies should investigate how the use of these inhibitors affects metabolic health, particularly whether there are risks or benefits associated with their use in obese or cachectic individuals.

The authors declare no competing interests.

E.Z. was supported by an AHA Career Development Award 23CDA1050768 and NIGMS R35GM154684.

All authors contributed to conceptualization, writing, and editing.

Figures were created in BioRender.com.

ACLY

ATP citrate lyase

BCAAs

branched-chain amino acids

BMPs

bone morphogenetic proteins

C/EBPs

CCAAT/enhancer binding proteins

CREB

cAMP response element-binding protein

FABP4

fatty acid binding protein 4

FAO

fatty acid oxidation

GR

glucocorticoid receptor

MCE

mitotic clonal expansion

NADPH

nicotinamide adenine dinucleotide phosphate

NAM

nicotinamide

NAMPT

nicotinamide phosphoribosyltransferase

NMATs

nicotinamide mononucleotide adenylyl transferases

NMN

nicotinamide mononucleotide

PARP1

polymerase 1

PDH

pyruvate dehydrogenase

PKA

protein kinase A

PKG

cGMP-dependent kinase

PPARγ

proliferator-activated receptor gamma

PPP

pentose phosphate pathway

PRPP

phosphoribosyl pyrophosphate

ROS

reactive oxygen species

RhoA

Ras homolog family member A

SIRT4

sirtuin 4

SMAD

Sma and Mad related proteins

TPK1

thiamine pyrophosphate kinase 1

TPP

thiamine pyrophosphate

WNT

wingless-related integration site

YAP

yes-associated protein

mTORC1

mechanistic target of rapamycin complex 1

sGC

soluble guanylyl cyclase

1
Jaiswal
,
Y.S.
and
Williams
,
L.L
. (
2017
)
A glimpse of ayurveda - the forgotten history and principles of Indian traditional medicine
.
J. Tradit. Complement. Med.
7
,
50
53
https://doi.org/10.1016/j.jtcme.2016.02.002
2
Haslam
,
D
. (
2007
)
Obesity: a medical history
.
Obes. Rev.
8 Suppl 1
,
31
36
https://doi.org/10.1111/j.1467-789X.2007.00314.x
3
Bray
,
G.A
. (
1993
)
Commentary on Classics in Obesity 5. Fat Cell Theory and Units of Knowledge
4
Hassall
,
A
. (
1993
)
Observations on the development of the fat vesicle. 1849
.
Obes. Res.
1
,
419
420
https://doi.org/10.1002/j.1550-8528.1993.tb00022.x
5
Wertheimer
,
E.
and
Shapiro
,
B
. (
1948
)
The physiology of adipose tissue
.
Physiol. Rev.
28
,
451
464
https://doi.org/10.1152/physrev.1948.28.4.451
6
Zhang
,
Y.
,
Proenca
,
R.
,
Maffei
,
M.
,
Barone
,
M.
,
Leopold
,
L.
and
Friedman
,
J.M
. (
1994
)
Positional cloning of the mouse obese gene and its human homologue
.
Nature
372
,
425
432
https://doi.org/10.1038/372425a0
7
Ghaben
,
A.L.
and
Scherer
,
P.E
. (
2019
)
Adipogenesis and metabolic health
.
Nat. Rev. Mol. Cell Biol.
20
,
242
258
https://doi.org/10.1038/s41580-018-0093-z
8
Marti
,
A.
and
Martinez-González
,
M.A
. (
2008
)
Interaction between genes and lifestyle factors on obesity: Nutrition Society Silver Medal Lecture
.
PNAS
67
,
1
8
https://doi.org/10.1017/S002966510800596X
9
Gliniak
,
C.M.
,
Pedersen
,
L.
and
Scherer
,
P.E
. (
2023
)
Adipose tissue fibrosis: the unwanted houseguest invited by obesity
.
J. Endocrinol.
259
, e230180 https://doi.org/10.1530/JOE-23-0180
10
Vishvanath
,
L.
and
Gupta
,
R.K
. (
2019
)
Contribution of adipogenesis to healthy adipose tissue expansion in obesity
.
J. Clin. Invest.
129
,
4022
4031
https://doi.org/10.1172/JCI129191
11
Schmidt
,
W.
,
Pöll-Jordan
,
G.
and
Löffler
,
G
. (
1990
)
Adipose conversion of 3T3-L1 cells in a serum-free culture system depends on epidermal growth factor, insulin-like growth factor I, corticosterone, and cyclic AMP
.
J. Biol. Chem.
265
,
15489
15495
https://doi.org/10.1016/S0021-9258(18)55422-3
12
Smith
,
P.J.
,
Wise
,
L.S.
,
Berkowitz
,
R.
,
Wan
,
C.
and
Rubin
,
C.S
. (
1988
)
Insulin-like growth factor-I is an essential regulator of the differentiation of 3T3-L1 adipocytes
.
J. Biol. Chem.
263
,
9402
9408
https://doi.org/10.1016/S0021-9258(19)76555-7
13
Bernlohr
,
D.A.
,
Bolanowski
,
M.A.
,
Kelly
,
T.J.
and
Lane
,
M.D
. (
1985
)
Evidence for an increase in transcription of specific mRNAs during differentiation of 3T3-L1 preadipocytes
.
J. Biol. Chem.
260
,
5563
5567
https://doi.org/10.1016/S0021-9258(18)89059-7
14
MacDougald
,
O.A.
and
Lane
,
M.D
. (
1995
)
Transcriptional regulation of gene expression during adipocyte differentiation
.
Annu. Rev. Biochem
64
,
345
373
https://doi.org/10.1146/annurev.bi.64.070195.002021
15
Patel
,
Y.M.
and
Lane
,
M.D
. (
1999
)
Role of calpain in adipocyte differentiation
.
Proc. Natl. Acad. Sci. U.S.A.
96
,
1279
1284
https://doi.org/10.1073/pnas.96.4.1279
16
Tang
,
Q.Q.
,
Otto
,
T.C.
and
Lane
,
M.D
. (
2003
)
Mitotic clonal expansion: a synchronous process required for adipogenesis
.
Proc. Natl. Acad. Sci. U.S.A.
100
,
44
49
https://doi.org/10.1073/pnas.0137044100
17
Gurland
,
G.
,
Ashcom
,
G.
,
Cochran
,
B.H.
and
Schwartz
,
J
. (
1990
)
Rapid events in growth hormone action. Induction of c-fos and c-jun transcription in 3T3-F442A preadipocytes
.
Endocrinology
127
,
3187
3195
https://doi.org/10.1210/endo-127-6-3187
18
Johmura
,
Y.
,
Osada
,
S.
,
Nishizuka
,
M.
and
Imagawa
,
M
. (
2008
)
FAD24 acts in concert with histone acetyltransferase HBO1 to promote adipogenesis by controlling DNA replication
.
J. Biol. Chem.
283
,
2265
2274
https://doi.org/10.1074/jbc.M707880200
19
Kim
,
S.J.
and
Kahn
,
C.R
. (
1994
)
Insulin stimulates phosphorylation of c-Jun, c-Fos, and Fos-related proteins in cultured adipocytes
.
J. Biol. Chem.
269
,
11887
11892
https://doi.org/10.1016/S0021-9258(17)32656-X
20
Li
,
X.
,
Kim
,
J.W.
,
Grønborg
,
M.
,
Urlaub
,
H.
,
Lane
,
M.D.
and
Tang
,
Q.Q
. (
2007
)
Role of cdk2 in the sequential phosphorylation/activation of C/EBPbeta during adipocyte differentiation
.
Proc. Natl. Acad. Sci. U.S.A.
104
,
11597
11602
https://doi.org/10.1073/pnas.0703771104
21
Rosen
,
E.D.
and
Spiegelman
,
B.M
. (
2000
)
Molecular regulation of adipogenesis
.
Annu. Rev. Cell Dev. Biol.
16
,
145
171
https://doi.org/10.1146/annurev.cellbio.16.1.145
22
Boucher
,
J.
,
Kleinridders
,
A.
and
Kahn
,
C.R
. (
2014
)
Insulin receptor signaling in normal and insulin-resistant states
.
Cold Spring Harb. Perspect. Biol.
6
, a009191 https://doi.org/10.1101/cshperspect.a009191
23
Kahn
,
B.B.
and
Flier
,
J.S
. (
2000
)
Obesity and insulin resistance
.
J. Clin. Invest.
106
,
473
481
https://doi.org/10.1172/JCI10842
24
Lawrence
,
J.C
. (
1992
)
Signal transduction and protein phosphorylation in the regulation of cellular metabolism by insulin
.
Annu. Rev. Physiol.
54
,
177
193
https://doi.org/10.1146/annurev.ph.54.030192.001141
25
Gathercole
,
L.L.
,
Morgan
,
S.A.
,
Bujalska
,
I.J.
,
Hauton
,
D.
,
Stewart
,
P.M.
and
Tomlinson
,
J.W
. (
2011
)
Regulation of lipogenesis by glucocorticoids and insulin in human adipose tissue
.
Plos One
6
, e26223 https://doi.org/10.1371/journal.pone.0026223
26
Lee
,
R.A.
,
Harris
,
C.A.
and
Wang
,
J.C
. (
2018
)
Glucocorticoid receptor and adipocyte biology
.
Nucl. Receptor Res.
5
, 101373 https://doi.org/10.32527/2018/101373
27
Blázquez-Medela
,
A.M.
,
Jumabay
,
M.
and
Boström
,
K.I
. (
2019
)
Beyond the bone: bone morphogenetic protein signaling in adipose tissue
.
Obes. Rev.
20
,
648
658
https://doi.org/10.1111/obr.12822
28
Huang
,
H.
,
Song
,
T.J.
,
Li
,
X.
,
Hu
,
L.
,
He
,
Q.
,
Liu
,
M.
et al
. (
2009
)
BMP signaling pathway is required for commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineage
.
Proc. Natl. Acad. Sci. U.S.A.
106
,
12670
12675
https://doi.org/10.1073/pnas.0906266106
29
Kanazawa
,
A.
,
Tsukada
,
S.
,
Kamiyama
,
M.
,
Yanagimoto
,
T.
,
Nakajima
,
M.
and
Maeda
,
S
. (
2005
)
Wnt5b partially inhibits canonical Wnt/beta-catenin signaling pathway and promotes adipogenesis in 3T3-L1 preadipocytes
.
Biochem. Biophys. Res. Commun.
330
,
505
510
https://doi.org/10.1016/j.bbrc.2005.03.007
30
Arnsdorf
,
E.J.
,
Tummala
,
P.
,
Kwon
,
R.Y.
and
Jacobs
,
C.R
. (
2009
)
Mechanically induced osteogenic differentiation--the role of RhoA, ROCKII and cytoskeletal dynamics
.
J. Cell. Sci.
122
,
546
553
https://doi.org/10.1242/jcs.036293
31
Sordella
,
R.
,
Classon
,
M.
,
Hu
,
K.Q.
,
Matheson
,
S.F.
,
Brouns
,
M.R.
,
Fine
,
B.
et al
. (
2002
)
Modulation of CREB activity by the Rho GTPase regulates cell and organism size during mouse embryonic development
.
Dev. Cell
2
,
553
565
https://doi.org/10.1016/s1534-5807(02)00162-4
32
Nunn
,
E.R.
,
Shinde
,
A.B.
and
Zaganjor
,
E
. (
2022
)
Weighing in on adipogenesis
.
Front. Physiol.
13
, 821278 https://doi.org/10.3389/fphys.2022.821278
33
Jackson
,
R.M.
,
Griesel
,
B.A.
,
Gurley
,
J.M.
,
Szweda
,
L.I.
and
Olson
,
A.L
. (
2017
)
Glucose availability controls adipogenesis in mouse 3T3-L1 adipocytes via up-regulation of nicotinamide metabolism
.
J. Biol. Chem.
292
,
18556
18564
https://doi.org/10.1074/jbc.M117.791970
34
Green
,
C.R.
,
Wallace
,
M.
,
Divakaruni
,
A.S.
,
Phillips
,
S.A.
,
Murphy
,
A.N.
,
Ciaraldi
,
T.P.
et al
. (
2016
)
Branched-chain amino acid catabolism fuels adipocyte differentiation and lipogenesis
.
Nat. Chem. Biol.
12
,
15
21
https://doi.org/10.1038/nchembio.1961
35
Neinast
,
M.D.
,
Jang
,
C.
,
Hui
,
S.
,
Murashige
,
D.S.
,
Chu
,
Q.
,
Morscher
,
R.J.
et al
. (
2019
)
Quantitative analysis of the whole-body metabolic fate of branched-chain amino acids
.
Cell Metab.
29
,
417
429
https://doi.org/10.1016/j.cmet.2018.10.013
36
Zaganjor
,
E.
,
Yoon
,
H.
,
Spinelli
,
J.B.
,
Nunn
,
E.R.
,
Laurent
,
G.
,
Keskinidis
,
P.
et al
. (
2021
)
SIRT4 is an early regulator of branched-chain amino acid catabolism that promotes adipogenesis
.
Cell Rep.
36
, 109345 https://doi.org/10.1016/j.celrep.2021.109345
37
Wellen
,
K.E.
,
Hatzivassiliou
,
G.
,
Sachdeva
,
U.M.
,
Bui
,
T.V.
,
Cross
,
J.R.
and
Thompson
,
C.B
. (
2009
)
ATP-citrate lyase links cellular metabolism to histone acetylation
.
Science
324
,
1076
1080
https://doi.org/10.1126/science.1164097
38
Engström
,
W.
and
Zetterberg
,
A
. (
1984
)
The relationship between purines, pyrimidines, nucleosides, and glutamine for fibroblast cell proliferation
.
J. Cell. Physiol.
120
,
233
241
https://doi.org/10.1002/jcp.1041200218
39
Kim
,
J.K.
,
Rathbone
,
M.P.
,
Middlemiss
,
P.J.
,
Hughes
,
D.W.
and
Smith
,
R.W
. (
1991
)
Purinergic stimulation of astroblast proliferation: guanosine and its nucleotides stimulate cell division in chick astroblasts
.
J. Neurosci. Res.
28
,
442
455
https://doi.org/10.1002/jnr.490280318
40
Rathbone
,
M.P.
,
Deforge
,
S.
,
Deluca
,
B.
,
Gabel
,
B.
,
Laurenssen
,
C.
,
Middlemiss
,
P.
et al
. (
1992
)
Purinergic stimulation of cell division and differentiation: mechanisms and pharmacological implications
.
Med. Hypotheses
37
,
213
219
https://doi.org/10.1016/0306-9877(92)90190-n
41
Helin
,
K
. (
1998
)
Regulation of cell proliferation by the E2F transcription factors
.
Curr. Opin. Genet. Dev.
8
,
28
35
https://doi.org/10.1016/s0959-437x(98)80058-0
42
Reichert
,
M.
and
Eick
,
D
. (
1999
)
Analysis of cell cycle arrest in adipocyte differentiation
.
Oncogene
18
,
459
466
https://doi.org/10.1038/sj.onc.1202308
43
Sardet
,
C.
,
Vidal
,
M.
,
Cobrinik
,
D.
,
Geng
,
Y.
,
Onufryk
,
C.
,
Chen
,
A.
et al
. (
1995
)
E2F-4 and E2F-5, two members of the E2F family, are expressed in the early phases of the cell cycle
.
Proc. Natl. Acad. Sci. U.S.A.
92
,
2403
2407
https://doi.org/10.1073/pnas.92.6.2403
44
Zetterberg
,
A.
and
Engström
,
W
. (
1981
)
Glutamine and the regulation of DNA replication and cell multiplication in fibroblasts
.
J. Cell. Physiol.
108
,
365
373
https://doi.org/10.1002/jcp.1041080310
45
Pinette
,
J.A.
,
Myers
,
J.W.
,
Park
,
W.Y.
,
Bryant
,
H.G.
,
Eddie
,
A.M.
,
Wilson
,
G.A.
et al
. (
2024
)
Disruption of nucleotide biosynthesis reprograms mitochondrial metabolism to inhibit adipogenesis
.
J. Lipid Res.
65
, 100641 https://doi.org/10.1016/j.jlr.2024.100641
46
Wilson
,
S.M.
,
Barsoum
,
M.J.
,
Wilson
,
B.W.
and
Pappone
,
P.A
. (
1999
)
Purine nucleotides modulate proliferation of brown fat preadipocytes
.
Cell Prolif.
32
,
131
140
https://doi.org/10.1046/j.1365-2184.1999.32230131.x
47
Tozzi
,
M.
and
Novak
,
I
. (
2017
)
Purinergic receptors in adipose tissue as potential targets in metabolic disorders
.
Front. Pharmacol.
8
, 878 https://doi.org/10.3389/fphar.2017.00878
48
Yamaoka
,
T.
,
Kondo
,
M.
,
Honda
,
S.
,
Iwahana
,
H.
,
Moritani
,
M.
,
Ii
,
S.
et al
. (
1997
)
Amidophosphoribosyltransferase limits the rate of cell growth-linked de novo purine biosynthesis in the presence of constant capacity of salvage purine biosynthesis
.
J. Biol. Chem.
272
,
17719
17725
https://doi.org/10.1074/jbc.272.28.17719
49
Yamaoka
,
T
. (
2001
)
Feedback inhibition of amidophosphoribosyltransferase regulates the rate of cell growth via purine nucleotide, DNA, and protein syntheses
.
J. Biol. Chem.
276
,
21285
21291
https://doi.org/10.1074/jbc.272.28.17719
50
Lane
,
A.N.
and
Fan
,
T.W.M
. (
2015
)
Regulation of mammalian nucleotide metabolism and biosynthesis
.
Nucleic Acids Res.
43
,
2466
2485
https://doi.org/10.1093/nar/gkv047
51
Aleman
,
V.
and
Handler
,
P
. (
1967
)
Dihydroorotate dehydrogenase
.
Journal of Biological Chemistry
242
,
4087
4096
https://doi.org/10.1016/S0021-9258(18)95783-2
52
Jones
,
M.E
. (
1980
)
Pyrimidine nucleotide biosynthesis in animals: genes, enzymes, and regulation of UMP biosynthesis
.
Annu. Rev. Biochem.
49
,
253
279
https://doi.org/10.1146/annurev.bi.49.070180.001345
53
Simon
,
E.S.
,
Bednarski
,
M.D.
and
Whitesides
,
G.M
. (
1988
)
Synthesis of CMP-NeuAc from N-acetylglucosamine: generation of CTP from CMP using adenylate kinase
.
J. Am. Chem. Soc.
110
,
7159
7163
https://doi.org/10.1021/ja00229a033
54
Baldwin
,
S.A.
,
Beal
,
P.R.
,
Yao
,
S.Y.M.
,
King
,
A.E.
,
Cass
,
C.E.
and
Young
,
J.D
. (
2004
)
The equilibrative nucleoside transporter family, SLC29
.
Pflugers Arch.
447
,
735
743
https://doi.org/10.1007/s00424-003-1103-2
55
Guallar
,
J.P.
,
Cano-Soldado
,
P.
,
Aymerich
,
I.
,
Domingo
,
J.C.
,
Alegre
,
M.
,
Domingo
,
P.
et al
. (
2007
)
Altered expression of nucleoside transporter genes (SLC28 and SLC29) in adipose tissue from HIV-1–infected patients
.
Antivir. Ther. (Lond.)
12
,
853
864
https://doi.org/10.1177/135965350701200601
56
Pastor-Anglada
,
M.
and
Perez-Torras
,
S
. (
2018
)
Who is who in adenosine transport
.
Front. Pharmacol.
9
, 627 https://doi.org/10.3389/fphar.2018.00606
57
Pfeifer
,
A.
,
Mikhael
,
M.
and
Niemann
,
B
. (
2024
)
Inosine: novel activator of brown adipose tissue and energy homeostasis
.
Trends Cell Biol.
34
,
72
82
https://doi.org/10.1016/j.tcb.2023.04.007
58
Sadana
,
R.
and
Dessauer
,
C.W
. (
2009
)
Physiological roles for G protein-regulated adenylyl cyclase isoforms: insights from knockout and overexpression studies
.
Neurosignals
17
,
5
22
https://doi.org/10.1159/000166277
59
Berthet
,
J.
,
Rall
,
T.W.
and
Sutherland
,
E.W
. (
1957
)
The relationship of epinephrine and glucagon to liver phosphorylase. IV. Effect of epinephrine and glucagon on the reactivation of phosphorylase in liver homogenates
.
J. Biol. Chem.
224
,
463
475
https://doi.org/10.1016/S0021-9258(18)65045-8
60
McKnight
,
G.S.
,
Cummings
,
D.E.
,
Amieux
,
P.S.
,
Sikorski
,
M.A.
,
Brandon
,
E.P.
,
Planas
,
J.V.
et al
. (
1998
)
Cyclic AMP, PKA, and the physiological regulation of adiposity
.
Recent Prog. Horm. Res.
53
,
139
159
https://pubmed.ncbi.nlm.nih.gov/9769707
61
Rogne
,
M.
and
Taskén
,
K
. (
2014
)
Compartmentalization of cAMP signaling in adipogenesis, lipogenesis, and lipolysis
.
Horm. Metab. Res.
46
,
833
840
https://doi.org/10.1055/s-0034-1389955
62
Pfeifer
,
A.
and
Hoffmann
,
L.S
. (
2015
)
Brown, beige, and white: the new color code of fat and its pharmacological implications
.
Annu. Rev. Pharmacol. Toxicol.
55
,
207
227
https://doi.org/10.1146/annurev-pharmtox-010814-124346
63
Bennett
,
C.N.
,
Ross
,
S.E.
,
Longo
,
K.A.
,
Bajnok
,
L.
,
Hemati
,
N.
,
Johnson
,
K.W.
et al
. (
2002
)
Regulation of Wnt signaling during adipogenesis
.
J. Biol. Chem.
277
,
30998
31004
https://doi.org/10.1074/jbc.M204527200
64
Ross
,
S.E.
,
Hemati
,
N.
,
Longo
,
K.A.
,
Bennett
,
C.N.
,
Lucas
,
P.C.
,
Erickson
,
R.L.
et al
. (
2000
)
Inhibition of adipogenesis by Wnt signaling
.
Science
289
,
950
953
https://doi.org/10.1126/science.289.5481.950
65
Tang
,
Q.Q.
,
Jiang
,
M.S.
and
Lane
,
M.D
. (
1999
)
Repressive effect of Sp1 on the C/EBPalpha gene promoter: role in adipocyte differentiation
.
Mol. Cell. Biol.
19
,
4855
4865
https://doi.org/10.1128/MCB.19.7.4855
66
Yu
,
F.X.
,
Zhang
,
Y.
,
Park
,
H.W.
,
Jewell
,
J.L.
,
Chen
,
Q.
,
Deng
,
Y.
et al
. (
2013
)
Protein kinase A activates the Hippo pathway to modulate cell proliferation and differentiation
.
Genes Dev.
27
,
1223
1232
https://doi.org/10.1101/gad.219402.113
67
Gonzalez
,
G.A.
and
Montminy
,
M.R
. (
1989
)
Cyclic AMP stimulates somatostatin gene transcription by phosphorylation of CREB at serine 133
.
Cell
59
,
675
680
https://doi.org/10.1016/0092-8674(89)90013-5
68
Hagiwara
,
M.
,
Brindle
,
P.
,
Harootunian
,
A.
,
Armstrong
,
R.
,
Rivier
,
J.
,
Vale
,
W.
et al
. (
1993
)
Coupling of hormonal stimulation and transcription via the cyclic AMP-responsive factor CREB is rate limited by nuclear entry of protein kinase A
.
Mol. Cell. Biol.
13
,
4852
4859
https://doi.org/10.1128/mcb.13.8.4852-4859.1993
69
Sordella
,
R
. (
2003
)
Modulation of Rho GTPase signaling regulates a switch between adipogenesis and myogenesis
.
Cell
113
,
147
158
https://doi.org/10.1016/S1534-5807(02)00162-4
70
Zhang
,
J.W.
,
Klemm
,
D.J.
,
Vinson
,
C.
and
Lane
,
M.D
. (
2004
)
Role of CREB in transcriptional regulation of CCAAT/enhancer-binding protein beta gene during adipogenesis
.
J. Biol. Chem.
279
,
4471
4478
https://doi.org/10.1074/jbc.M311327200
71
Tojima
,
T.
,
Itofusa
,
R.
and
Kamiguchi
,
H
. (
2009
)
The nitric oxide-cGMP pathway controls the directional polarity of growth cone guidance via modulating cytosolic Ca2+ signals
.
J. Neurosci.
29
,
7886
7897
https://doi.org/10.1523/JNEUROSCI.0087-09.2009
72
Knowles
,
R.G.
and
Moncada
,
S
. (
1994
)
Nitric oxide synthases in mammals
.
Biochem. J.
298 ( Pt 2)
,
249
258
https://doi.org/10.1042/bj2980249
73
Arnold
,
W.P.
,
Mittal
,
C.K.
,
Katsuki
,
S.
and
Murad
,
F
. (
1977
)
Nitric oxide activates guanylate cyclase and increases guanosine 3′:5′-cyclic monophosphate levels in various tissue preparations.
.
Proc Natl Acad Sci USA
74
,
3203
3207
https://doi.org/10.1073/pnas.74.8.3203
74
Zhang
,
X.
,
Ji
,
J.
,
Yan
,
G.
,
Wu
,
J.
,
Sun
,
X.
,
Shen
,
J.
et al
. (
2010
)
Sildenafil promotes adipogenesis through a PKG pathway
.
Biochem. Biophys. Res. Commun.
396
,
1054
1059
https://doi.org/10.1016/j.bbrc.2010.05.064
75
Wilson-Fritch
,
L.
,
Burkart
,
A.
,
Bell
,
G.
,
Mendelson
,
K.
,
Leszyk
,
J.
,
Nicoloro
,
S.
et al
. (
2003
)
Mitochondrial biogenesis and remodeling during adipogenesis and in response to the insulin sensitizer rosiglitazone
.
Mol. Cell. Biol.
23
,
1085
1094
https://doi.org/10.1128/MCB.23.3.1085-1094.2003
76
Omatsu-Kanbe
,
M.
,
Inoue
,
K.
,
Fujii
,
Y.
,
Yamamoto
,
T.
,
Isono
,
T.
,
Fujita
,
N.
et al
. (
2006
)
Effect of ATP on preadipocyte migration and adipocyte differentiation by activating P2Y receptors in 3T3-L1 cells
.
Biochem. J.
393
,
171
180
https://doi.org/10.1042/BJ20051037
77
Pontes
,
M.H.
,
Sevostyanova
,
A.
and
Groisman
,
E.A
. (
2015
)
When too much ATP is bd for protein synthesis
.
J. Mol. Biol.
427
,
2586
2594
https://doi.org/10.1016/j.jmb.2015.06.021
78
Mackall
,
J.C.
,
Student
,
A.K.
,
Polakis
,
S.E.
and
Lane
,
M.D
. (
1976
)
Induction of lipogenesis during differentiation in a “preadipocyte” cell line
.
Journal of Biological Chemistry
251
,
6462
6464
https://doi.org/10.1016/S0021-9258(20)81883-3
79
Solinas
,
G.
,
Borén
,
J.
and
Dulloo
,
A.G
. (
2015
)
De novo lipogenesis in metabolic homeostasis: more friend than foe?
Mol. Metab.
4
,
367
377
https://doi.org/10.1016/j.molmet.2015.03.004
80
Jiang
,
P.
,
Du
,
W.
,
Wang
,
X.
,
Mancuso
,
A.
,
Gao
,
X.
,
Wu
,
M.
et al
. (
2011
)
p53 regulates biosynthesis through direct inactivation of glucose-6-phosphate dehydrogenase
.
Nat. Cell Biol.
13
,
310
316
https://doi.org/10.1038/ncb2172
81
Ying
,
W
. (
2008
)
NAD+/NADH and NADP+/NADPH in cellular functions and cell death: regulation and biological consequences
.
Antioxid. Redox Signal.
10
,
179
206
https://doi.org/10.1089/ars.2007.1672
82
Verdin
,
E
. (
2015
)
NAD+ in aging, metabolism, and neurodegeneration
.
Science
350
,
1208
1213
https://doi.org/10.1126/science.aac4854
83
Chiarugi
,
A.
,
Dölle
,
C.
,
Felici
,
R.
and
Ziegler
,
M
. (
2012
)
The NAD metabolome--a key determinant of cancer cell biology
.
Nat. Rev. Cancer
12
,
741
752
https://doi.org/10.1038/nrc3340
84
Okabe
,
K.
,
Nawaz
,
A.
,
Nishida
,
Y.
,
Yaku
,
K.
,
Usui
,
I.
,
Tobe
,
K.
et al
. (
2020
)
NAD+ metabolism regulates preadipocyte differentiation by enhancing α-Ketoglutarate-mediated histone H3K9 demethylation at the PPARγ promoter
.
Front. Cell Dev. Biol.
8
, 586179 https://doi.org/10.3389/fcell.2020.586179
85
Ryu
,
K.W.
,
Nandu
,
T.
,
Kim
,
J.
,
Challa
,
S.
,
DeBerardinis
,
R.J.
and
Kraus
,
W.L
. (
2018
)
Metabolic regulation of transcription through compartmentalized NAD+ biosynthesis
.
Science
360
, eaan5780 https://doi.org/10.1126/science.aan5780
86
Luo
,
X.
,
Ryu
,
K.W.
,
Kim
,
D.-S.
,
Nandu
,
T.
,
Medina
,
C.J.
,
Gupte
,
R.
et al
. (
2017
)
PARP-1 controls the adipogenic transcriptional program by PARylating C/EBPβ and modulating its transcriptional activity
.
Mol. Cell
65
,
260
271
https://doi.org/10.1016/j.molcel.2016.11.015
87
Katsyuba
,
E.
,
Romani
,
M.
,
Hofer
,
D.
and
Auwerx
,
J
. (
2020
)
NAD+ homeostasis in health and disease
.
Nat. Metab.
2
,
9
31
https://doi.org/10.1038/s42255-019-0161-5
88
Shinde
,
A.B.
,
Nunn
,
E.R.
,
Wilson
,
G.A.
,
Chvasta
,
M.T.
,
Pinette
,
J.A.
,
Myers
,
J.W.
et al
. (
2023
)
Inhibition of nucleotide biosynthesis disrupts lipid accumulation and adipogenesis
.
J. Biol. Chem.
299
, 104635 https://doi.org/10.1016/j.jbc.2023.104635
89
Emmanuel
,
N.
,
Ragunathan
,
S.
,
Shan
,
Q.
,
Wang
,
F.
,
Giannakou
,
A.
,
Huser
,
N.
et al
. (
2017
)
Purine nucleotide availability regulates mTORC1 activity through the Rheb GTPase
.
Cell Rep.
19
,
2665
2680
https://doi.org/10.1016/j.celrep.2017.05.043
90
Hoxhaj
,
G.
,
Hughes-Hallett
,
J.
,
Timson
,
R.C.
,
Ilagan
,
E.
,
Yuan
,
M.
,
Asara
,
J.M.
et al
. (
2017
)
The mTORC1 signaling network senses changes in cellular purine nucleotide levels
.
Cell Rep.
21
,
1331
1346
https://doi.org/10.1016/j.celrep.2017.10.029
91
Hu
,
J.
,
Zheng
,
S.
,
Hua
,
M.
,
Ding
,
M.
,
Hu
,
Z.
and
Jiang
,
H
. (
2024
)
Deletion of Impdh2 in adipocyte precursors limits the expansion of white adipose tissue and enhances metabolic health with overnutrition
.
Biochem. Biophys. Res. Commun.
716
, 149998 https://doi.org/10.1016/j.bbrc.2024.149998
92
Sahu
,
U.
,
Villa
,
E.
,
Reczek
,
C.R.
,
Zhao
,
Z.
,
O’Hara
,
B.P.
,
Torno
,
M.D.
et al
. (
2024
)
Pyrimidines maintain mitochondrial pyruvate oxidation to support de novo lipogenesis
.
Science
383
,
1484
1492
https://doi.org/10.1126/science.adh2771
93
Baker
,
L.J.
,
Dorocke
,
J.A.
,
Harris
,
R.A.
and
Timm
,
D.E
. (
2001
)
The crystal structure of yeast thiamin pyrophosphokinase
.
Structure
9
,
539
546
https://doi.org/10.1016/s0969-2126(01)00615-3
94
Sambon
,
M.
,
Pavlova
,
O.
,
Alhama-Riba
,
J.
,
Wins
,
P.
,
Brans
,
A.
and
Bettendorff
,
L
. (
2022
)
Product inhibition of mammalian thiamine pyrophosphokinase is an important mechanism for maintaining thiamine diphosphate homeostasis
.
Biochimica et Biophysica Acta (BBA) - General Subjects
1866
,
130071
https://doi.org/10.1016/j.bbagen.2021.130071
95
Zhao
,
S.
,
Torres
,
A.
,
Henry
,
R.A.
,
Trefely
,
S.
,
Wallace
,
M.
,
Lee
,
J.V.
et al
. (
2016
)
ATP-citrate lyase controls a glucose-to-acetate metabolic switch
.
Cell Rep.
17
,
1037
1052
https://doi.org/10.1016/j.celrep.2016.09.069
96
Liu
,
Y.
,
Lv
,
W.
,
Yu
,
B.
,
Ju
,
T.
,
Yang
,
F.
,
Jiang
,
M.
et al
. (
2013
)
S-Adenosylmethionine-induced adipogenesis is accompanied by suppression of Wnt/β-catenin and Hedgehog signaling pathways
.
Mol Cell Biochem
382
,
59
73
http://link.springer.com/10.1007/s11010-013-1718-3
97
Cho
,
Y.W.
,
Hong
,
S.
,
Jin
,
Q.
,
Wang
,
L.
,
Lee
,
J.E.
,
Gavrilova
,
O.
et al
. (
2009
)
Histone methylation regulator PTIP is required for PPARγ and C/EBPα expression and adipogenesis
.
Cell Metab.
10
,
27
39
https://doi.org/10.1016/j.cmet.2009.05.010
98
Wang
,
L.
,
Jin
,
Q.
,
Lee
,
J.-E.
,
Su
,
I.
and
Ge
,
K
. (
2010
)
Histone H3K27 methyltransferase Ezh2 represses Wnt genes to facilitate adipogenesis
.
Proc. Natl. Acad. Sci. U.S.A.
107
,
7317
7322
https://doi.org/10.1073/pnas.1000031107
99
Noer
,
A.
,
Boquest
,
A.C.
and
Collas
,
P
. (
2007
)
Dynamics of adipogenic promoter DNA methylation during clonal culture of human adipose stem cells to senescence
.
BMC Cell Biol.
8
, 18 https://doi.org/10.1186/1471-2121-8-18
100
Myers
,
J.W.
,
Park
,
W.Y.
,
Eddie
,
A.M.
,
Shinde
,
A.B.
,
Prasad
,
P.
,
Murphy
,
A.C.
et al
. (
2024
)
Systemic inhibition of de novo purine biosynthesis prevents weight gain and improves metabolic health by increasing thermogenesis and decreasing food intake
.
bioRxiv
2024.10.28.620705 https://doi.org/10.1101/2024.10.28.620705
101
Crohn’s disease: management
. (
2019
)
London: National Institution for Health and Care Excellence
102
Ulcerative colitis: management
. (
2019
)
London: National Institution for Health and Care Excellence
103
Donahue
,
K.E.
,
Gartlebner
,
G.
,
Schulman
,
E.R.
,
Jonas
,
B.
et al
. (
2018
)
Comparative effectiveness review, No. 211
.
18-EHC015-EF
.
Rockville, MD: Agency for Healthcare Research and Quality
104
PDQ Adult Treatment Editorial Board
. (
2024
)
Colon cancer treatment (PDQ): health professional version
.
National Cancer Institute
.
In: PDQ Cancer Information Summaries
. https://www.cancer.gov/types/colorectal/hp/colon-treatment-pdq
105
PDQ Adult Treatment Editorial Board
. (
2024
)
Gastric cancer treatment (PDQ): health professional version
.
Bethesda, MD: National Cancer Institute
.
In: PDQ Cancer Information Summaries
. https://www.ncbi.nlm.nih.gov/books/NBK65766/
106
PDQ Adult Treatment Editorial Board
. (
2024
)
Rectal cancer treatment (PDQ): health professional version
.
National Cancer Institute
.
In: PDQ Cancer Information Summaries
. https://www.cancer.gov/types/colorectal/hp/rectal-treatment-pdq
107
PDQ Adult Treatment Editorial Board
. (
2024
)
Breast cancer treatment (PDQ): health professional version
.
National Cancer Institute
.
In: PDQ Cancer Information Summaries
. https://www.cancer.gov/types/breast/hp/breast-treatment-pdq
108
PDQ Adult Treatment Editorial Board
. (
2024
)
Esophageal cancer treatment (PDQ): health professional version
.
National Cancer Institute
.
In: PDQ Cancer Information
. https://www.cancer.gov/types/esophageal/hp/esophageal-treatment-pdq
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).