Fact-checked by Grok 2 weeks ago

Adipogenesis

Adipogenesis is the developmental process by which multipotent mesenchymal cells to the lineage and differentiate into mature, lipid-laden s, forming , , or that plays a central role in and metabolic regulation. This tightly regulated process unfolds in three main stages: ment, where mesenchymal cells or preadipocytes are specified toward the adipogenic lineage; mitotic clonal expansion, involving rapid cell proliferation in response to mitogenic signals; and terminal differentiation, characterized by the expression of adipocyte-specific genes leading to lipid accumulation and functional maturation. Central to adipogenesis is a transcriptional cascade orchestrated by key regulators, including the early-phase factors C/EBPβ and C/EBPδ, which activate the master regulator PPARγ and C/EBPα to drive the expression of genes involved in , insulin sensitivity, and secretion. Signaling pathways such as bone morphogenetic proteins (BMPs) promote , while inhibitors like Wnt, , and TGF-β signaling suppress it to maintain lineage balance. Physiologically, adipogenesis is essential for storing excess energy as triglycerides in , preventing lipotoxicity in non-adipose organs like the liver and muscle, and supporting endocrine functions through adipokines that influence systemic . Dysregulated adipogenesis contributes to by promoting excessive adipose expansion, particularly visceral fat accumulation, and is linked to metabolic disorders such as and ; notably, recent studies highlight age-dependent shifts, with distinct progenitor cells driving heightened adipogenesis in . In humans, adipocytes exhibit a turnover rate of approximately 10% per year, underscoring the process's dynamic role in tissue maintenance and potential therapeutic targeting for intervention.

Overview

Definition and Phases

Adipogenesis is the developmental process by which multipotent mesenchymal stem cells (MSCs) or committed preadipocytes differentiate into mature adipocytes capable of lipid storage. This multi-step is essential for the formation and expansion of , involving sequential cellular commitments and transformations that result in cells specialized for . The initial discovery of adipogenesis as a tractable process occurred in the 1970s through pioneering studies using immortalized embryonic lines, such as 3T3-L1 cells, which demonstrated the inducible conversion of fibroblastic precursors into lipid-filled adipocytes under specific hormonal conditions. These early experiments established adipogenesis as a model for studying , revealing that preadipose cells could be maintained in culture and triggered to mature, laying the groundwork for subsequent molecular analyses. Adipogenesis unfolds in three distinct phases: determination, mitotic clonal expansion, and terminal differentiation. In the determination phase, multipotent MSCs commit irreversibly to the preadipocyte lineage, restricting their potential to differentiate into other mesenchymal cell types such as osteoblasts or chondrocytes. This commitment occurs early in development or in response to physiological cues, marking the point at which precursors adopt an adipocyte-specific fate. Following confluence in culture or appropriate signals, preadipocytes enter mitotic clonal expansion, a proliferative phase characterized by 2–3 rounds of that amplify the committed cell population. This step is crucial for generating sufficient cells prior to maturation and is tightly regulated to ensure proper timing. The final phase, terminal differentiation, involves the morphological and functional maturation of preadipocytes into adipocytes, accompanied by extensive accumulation in the form of triglycerides within cytoplasmic droplets. During this stage, cells express genes that confer insulin sensitivity, lipogenic capacity, and secretory functions, transforming them into fully functional adipose units integrated into tissue depots. Transcription factors orchestrate these changes, driving the coordinated activation of adipocyte-specific programs (detailed in Core Molecular Mechanisms).

Physiological Importance

Adipogenesis plays a central role in by facilitating the storage of excess energy as in during periods of nutrient abundance, thereby preventing in other organs. Mature adipocytes, formed through this process, accumulate in large droplets, providing a high-energy reserve—approximately 38 kJ per gram of , which is roughly double the energy yield from glucose or . During or increased energy demand, these adipocytes release free fatty acids through , regulated by hormones such as insulin (which promotes storage) and catecholamines (which trigger breakdown), ensuring systemic energy balance. Beyond energy storage, adipocytes derived from adipogenesis contribute significantly to endocrine functions by secreting adipokines that modulate whole-body , , and insulin sensitivity. , produced by adipocytes, signals satiety to the , promotes lipid oxidation, and enhances to maintain energy expenditure. , another key , exerts anti-inflammatory effects, improves insulin sensitivity in muscle and liver, and supports glucose utilization, thereby protecting against metabolic dysregulation. These secretions position as an active endocrine organ, influencing peripheral tissues and responses to nutritional status. Dysregulated adipogenesis is implicated in several pathophysiological conditions, particularly those related to metabolic . In , excessive adipogenesis leads to expansion through both (enlargement of existing adipocytes) and (formation of new adipocytes), which can result in , chronic , and , contributing to and . Conversely, impaired adipogenesis, as seen in , reduces expandability, causing ectopic lipid deposition in organs like the liver and muscle, severe , and profound . These imbalances highlight adipogenesis as a critical determinant of metabolic disease risk, where balanced differentiation supports while dysregulation promotes pathology.

Adipocyte Types and Differentiation

White and Brown Adipocytes

White adipocytes and brown adipocytes represent the two primary cell types arising from adipogenesis, each with specialized structures and functions adapted to distinct physiological roles. White adipocytes are primarily derived from Myf5-negative precursor cells within the mesenchymal lineage, enabling their commitment toward rather than myogenic . These cells are characterized by a single large unilocular lipid droplet that occupies most of the , facilitating efficient long-term storage of triglycerides as an energy reserve during periods of nutrient excess. They are predominantly located in subcutaneous depots beneath the skin and visceral depots surrounding internal organs, where they contribute to body insulation and metabolic buffering.00325-7) In contrast, brown adipocytes originate from Myf5-positive precursors that share a developmental trajectory with skeletal muscle cells, highlighting their closer evolutionary tie to myogenic lineages. These cells feature multiple smaller multilocular lipid droplets surrounding numerous mitochondria, which enable their hallmark function of thermogenesis through uncoupled mitochondrial respiration mediated by uncoupling protein 1 (UCP1). This process dissipates energy as heat, independent of ATP production, to maintain body temperature in response to cold exposure. Brown adipocytes are mainly found in specialized brown adipose tissue (BAT) depots, such as the interscapular region in rodents and neonates, though their presence diminishes in adult humans.00464-7) A third adipocyte subtype, beige adipocytes (also known as brite cells), emerges through of mature white adipocytes or recruitment from white adipose progenitors, adopting an intermediate with thermogenic capacity. This plasticity is induced by environmental cues like prolonged cold exposure or pharmacological activation of β-adrenergic receptors, leading to upregulated expression and partial multilocular morphology within white adipose depots.00464-7) Unlike constitutive brown adipocytes, beige cells can revert to a white-like state upon stimulus removal, underscoring their adaptive role in energy expenditure. Both white and brown adipocytes trace their evolutionary and developmental origins to common mesenchymal precursor cells in the embryonic , from which they diverge through lineage-specific commitment signals during adipogenesis. This shared ancestry allows for potential plasticity, such as beige formation, but is governed by factors like PRDM16 that direct Myf5-positive precursors toward brown fate while suppressing , illustrating the intricate balance in differentiation potentials.

In Vivo and In Vitro Models

In vivo models for studying adipogenesis primarily utilize genetically modified to track development and precursor dynamics within physiological contexts. The Cre-loxP recombination system enables conditional of genes in precursors, such as through Adipoq-Cre or aP2-Cre lines, which allow tissue-specific targeting of white and brown adipose depots without affecting other organs. For instance, the AdipoChaser , a triple-transgenic model incorporating promoter-driven rtTA, tetracycline-responsive Cre, and Rosa26-loxP-stop-loxP-lacZ, facilitates inducible and permanent labeling of mature adipocytes upon administration, enabling the quantification of de novo adipogenesis post-labeling. These models reveal depot-specific timing, with epididymal (eWAT) adipogenesis occurring postnatally and subcutaneous (sWAT) initiating embryonically. Obesity models, such as high-fat diet (HFD)-fed mice, mimic diet-induced adipose expansion and are used to investigate versus . In PDGFRα-GFP-Cre-ERT2/tdTomato reporter mice fed HFD for 8 weeks, iWAT expands through of progenitors and of smaller new adipocytes (44% labeled post-HFD versus 13% on chow), while gonadal (gWAT) primarily hypertrophies with limited progenitor (34% labeled post-HFD versus 15% on chow). Human studies employ biopsies for tracing, often via single-nucleus sequencing on subcutaneous and visceral samples from surgical donors, identifying progenitor-to- trajectories and depot-specific patterns without genetic manipulation. In vitro models provide controlled environments for dissecting adipogenesis, with preadipocyte lines and primary cultures as foundational tools. The 3T3-L1 line, derived from mouse embryo fibroblasts and established in 1975 by Green and Kehinde, differentiates into s over 10-14 days when post-confluent cells are treated with a cocktail including 0.5 mM 3-isobutyl-1-methylxanthine (), 0.25-1 µM dexamethasone, and 1-10 µg/mL insulin, often supplemented with 10% fetal bovine serum.90087-2) Similarly, the 3T3-F442A subline, also from Green and Kehinde in 1974, exhibits higher accumulation and requires primarily insulin for , making it suitable for studies of mature traits.90126-3) These lines are selected based on type, with 3T3-L1 favoring white models and adaptations for brown/beige . Primary stromal vascular fraction (SVF) cultures, isolated from or via collagenase digestion, encompass heterogeneous preadipocytes and progenitors that differentiate using the same IBMX-dexamethasone-insulin cocktail over 7-14 days, reflecting depot- and donor-specific variations. approaches offer advantages in mechanistic precision and scalability, allowing genetic manipulations like and , but they lack systemic tissue interactions and vascularization, leading to immature droplets and reduced physiological fidelity compared to systems. Conversely, models provide holistic insights into organismal responses but face ethical constraints, especially in s, and challenges in isolating specific cellular events.

Core Molecular Mechanisms

Transcriptional Regulation: PPARγ and C/EBPs

Peroxisome proliferator-activated receptor gamma (PPARγ) is a that serves as a master regulator of adipogenesis, orchestrating the expression of genes essential for differentiation and lipid storage. The PPARγ2 isoform, which is specifically expressed in adipocytes, is induced early during the differentiation process and plays a pivotal role in committing preadipocytes to the adipogenic lineage. Upon activation, PPARγ forms a heterodimer with (RXR) and binds to peroxisome proliferator response elements (PPREs) in the promoter regions of target genes, thereby promoting the transcription of lipogenic and -specific factors such as binding protein 4 (FABP4) and (LPL). These targets facilitate uptake and storage, hallmarks of mature adipocytes. PPARγ activity is modulated by endogenous ligands like s and synthetic agonists such as , a that potently induces differentiation by stabilizing the receptor's active conformation. The CCAAT/enhancer-binding protein (C/EBP) family of basic transcription factors complements PPARγ by driving sequential phases of adipogenesis. C/EBPβ and C/EBPδ are rapidly upregulated in response to adipogenic stimuli, acting as early inducers that promote mitotic clonal expansion and initiate the expression of downstream factors including PPARγ and C/EBPα. These early C/EBPs bind to specific C/EBP response elements in target gene promoters, facilitating progression and the transition to terminal . In contrast, C/EBPα emerges later and sustains the differentiated state by activating genes involved in , , and exit, such as those encoding adipocyte protein 2 (aP2/FABP4) and insulin-responsive glucose transporter (). C/EBPα expression peaks during mid-to-late adipogenesis, ensuring the maintenance of the . PPARγ and C/EBPα engage in mutual feed-forward loops that amplify and stabilize adipogenic gene expression, forming an interdependent network critical for terminal differentiation. PPARγ directly transactivates the C/EBPα promoter, while C/EBPα reciprocally induces PPARγ2 expression, creating a positive feedback mechanism that reinforces commitment to the adipocyte fate and prevents reversion to a proliferative state. This synergy is essential for the full maturation of adipocytes, as disruptions in either factor impair the other's function. Genetic evidence underscores their indispensability: targeted ablation of PPARγ in mouse embryonic fibroblasts and embryonic stem cells completely blocks adipocyte differentiation in vitro and severely impairs adipose tissue development in vivo, resulting in lipodystrophy-like phenotypes. Similarly, heterozygous dominant-negative mutations in human PPARG, such as those altering the DNA-binding or ligand-binding domains, cause familial partial lipodystrophy characterized by adipose tissue loss, insulin resistance, and metabolic syndrome.

Regulatory Cascade

The regulatory cascade of adipogenesis describes a sequential of transcription factors that drives the of mesenchymal precursor cells into adipocytes, integrating external signals with intrinsic genetic programs. This hierarchical model, first comprehensively outlined in the late 1990s, involves the coordinated expression of key transcription factors that progressively commit cells to the lineage and maintain their differentiated state. In the early phase, external adipogenic stimuli, such as hormonal cues, rapidly induce the expression of CCAAT/enhancer-binding proteins β (C/EBPβ) and δ (C/EBPδ) within hours of . These immediate-early factors to the promoters of peroxisome proliferator-activated receptor γ (PPARγ) and C/EBPα genes, thereby activating their transcription and initiating the core adipogenic program. C/EBPβ and C/EBPδ lack the ability to directly drive terminal but serve as pivotal activators that bridge extrinsic signals to downstream effectors.00031-8) During the mid-phase, PPARγ and C/EBPα form a mutual loop, where each enhances the expression and activity of the other, amplifying the adipogenic signal. This cross-regulation ensures robust of genes essential for , including (SREBP1), which in turn promotes lipogenic enzyme expression such as and . The interplay between PPARγ and C/EBPα is critical, as disruption of either impairs the full execution of the program, with PPARγ acting as the primary driver of identity. In the late phase, sustained expression of PPARγ and C/EBPα maintains the differentiated state by continuously regulating approximately 200 adipocyte-selective genes involved in lipid storage, insulin sensitivity, and metabolic , while suppressing anti-adipogenic factors such as preadipocyte factor 1 (Pref-1). Pref-1, expressed in undifferentiated precursors, inhibits the cascade by blocking C/EBPβ/δ ; its downregulation is essential for progression to maturity. This phase stabilizes the , preventing reversion to a proliferative state. The hierarchical cascade model, proposed by Rosen and Spiegelman in the 1990s, underscores this temporal orchestration, where early inducers activate master regulators that then execute and sustain . Genome-wide studies confirm that PPARγ and C/EBP factors co-occupy most of these target genes, ensuring coordinated expression. Variations exist between white and brown adipocytes; for instance, PRDM16 integrates into the cascade in brown precursors to promote thermogenic genes like alongside PPARγ, diverting the program from white fat characteristics.

Hormonal and Signaling Regulation

Insulin and IGF-1

Insulin and (IGF-1) serve as pivotal hormonal regulators that promote adipogenesis by facilitating preadipocyte proliferation, survival, and differentiation into mature . These hormones exert their effects primarily through binding to their respective receptors, the (IR) and IGF-1 receptor (IGF1R), which are expressed on preadipocytes and . Upon binding, receptor autophosphorylation activates downstream signaling cascades that support the metabolic and proliferative demands of adipocyte development. A central mechanism involves the activation of the phosphoinositide 3-kinase (PI3K)-Akt pathway, which enhances glucose uptake and mitotic clonal expansion essential for adipogenesis. Insulin and IGF-1 stimulate the translocation of glucose transporter 4 (GLUT4) to the plasma membrane, increasing glucose influx to fuel lipogenesis and energy needs during differentiation. This PI3K-Akt signaling also promotes cell cycle progression through mitotic clonal expansion, a synchronous proliferation phase where preadipocytes undergo DNA replication and division prior to terminal differentiation. Furthermore, these hormones induce the expression of CCAAT/enhancer-binding protein delta (C/EBPδ), an early transcription factor that initiates the adipogenic program and links to downstream activation of the core transcriptional cascade. In in vitro models of adipogenesis, insulin is a requisite component of differentiation cocktails, typically combined with agents like 3-isobutyl-1-methylxanthine () and dexamethasone to induce robust formation from preadipocyte cell lines such as 3T3-L1. Optimal insulin concentrations (e.g., 5-10 μg/mL) during the initial 2-3 days of induction drive mitotic expansion and lipid accumulation, with prolonged exposure maintaining mature function. Chronic elevation of insulin, as observed in , further enhances hyperplasia by sustaining proliferative signals and expanding mass. Evidence from genetic models underscores these roles; mice with adipose-specific knockout of the (FIRKO) exhibit markedly reduced mass and impaired fat pad development due to defective preadipocyte . Similarly, combined insulin and IGF-1 receptor knockouts in result in diminished size and number, highlighting overlapping signaling requirements for adipose . For IGF-1, studies demonstrate its promotion of brown adipogenesis in certain contexts, such as enhancing of brown preadipocytes via IGF1R signaling and upregulation of thermogenic markers. Clinically, in drives pathological adipogenesis, contributing to excessive adipose expansion, , and by amplifying pro-adipogenic signals in a dysregulated environment.

Wnt and BMP Signaling

The canonical plays a critical inhibitory role in adipogenesis by stabilizing β-catenin, which translocates to the nucleus and activates transcription factors TCF/LEF to repress the expression of key adipogenic regulators such as PPARγ and C/EBPα. This pathway is initiated through binding of Wnt ligands to receptors and /6 co-receptors on mesenchymal stem cells (MSCs) and preadipocytes, preventing the of these cells into mature adipocytes. In conditions of , suppressed levels of Wnt signaling components, such as Wnt10b, allow for increased by permitting new adipocyte formation, contributing to adipose expansion. In contrast, () signaling generally promotes adipogenesis, particularly through , , and , which bind to type I activin receptor-like kinase (ALK) receptors and trigger the phosphorylation of Smad1/5/8 proteins. These activated Smads form complexes with Smad4 that translocate to the , where they induce the expression of C/EBPβ and facilitate the commitment of multipotent stem cells toward the lineage. BMP signaling is especially vital for brown adipogenesis, as BMP7 activates a comprehensive program including upregulation of brown fat-specific factors like PRDM16 and PGC-1α, driving the differentiation of precursors into thermogenic brown adipocytes. Crosstalk between Wnt and BMP pathways finely tunes the balance between white and brown adipocyte fates during development and differentiation. High Wnt activity favors non-adipogenic lineages or white adipocyte hypertrophy, while BMP dominance shifts precursors toward adipogenesis, with BMP4 preferentially inducing white adipocyte commitment and BMP7 promoting brown. This antagonism has been demonstrated in avian models, such as chick and quail embryos, where modulating BMP signaling enhances adipocyte differentiation in somites, and in human MSCs, where BMP treatment overrides Wnt-mediated inhibition to direct lineage commitment. The interplay integrates with other signals, such as insulin, to modulate overall adipogenic potential in MSCs. Given their opposing effects, targeting these pathways holds therapeutic promise for management; studies, including those from 2022 onward, have explored BMP agonists, particularly BMP7 via , to stimulate brown adipogenesis and increase energy expenditure as a strategy to counteract fat accumulation. For example, AAV-mediated BMP7 delivery has been shown to improve metabolic phenotypes in obese models by enhancing energy expenditure and reducing (as of 2022).

Emerging Regulatory Factors

Senescent Cells

Cellular senescence refers to a state of irreversible growth arrest in preadipocytes, triggered by cellular stresses such as reactive oxygen species (ROS) and DNA damage. This process is characterized by the senescence-associated secretory phenotype (SASP), through which senescent cells release pro-inflammatory factors including interleukin-6 (IL-6) and plasminogen activator inhibitor-1 (PAI-1). In the context of adipose tissue, these senescent preadipocytes accumulate particularly in visceral depots during aging and obesity, contributing to tissue dysfunction. The accumulation of senescent cells in obese exerts inhibitory effects on adipogenesis by secreting paracrine factors, such as activin A, that directly block the of non-senescent progenitors into mature . This SASP-mediated inhibition not only reduces the available pool of functional progenitors but also promotes chronic low-grade , exacerbating metabolic derangements like . Consequently, the impaired adipogenic capacity leads to hypertrophic expansion rather than healthy remodeling, a hallmark of obesity-related adipose dysfunction. Evidence from preclinical models demonstrates that clearing senescent cells can restore adipogenesis and mitigate associated metabolic issues. In a 2015 study using aged mice, genetic or pharmacological elimination of senescent cells enhanced progenitor differentiation and improved adipose tissue function, linking senescence clearance to better metabolic outcomes. Similarly, a 2019 investigation showed that senescent cell ablation in obese mice alleviated inflammation, boosted insulin sensitivity, and promoted adipogenesis in visceral fat, underscoring the causal role of these cells in obesity-driven pathology. These findings highlight senescence as a targetable barrier to adipose plasticity in metabolic disease. The reversibility of senescence-related inhibition has been explored through therapies, which selectively eliminate senescent cells. For instance, treatment with , a , combined with has been shown to reduce senescent preadipocyte burden in , thereby attenuating inflammation and enhancing metabolic function in aging and obese models. Such interventions improve progenitor and , suggesting potential therapeutic avenues for restoring adipogenic capacity in dysfunctional .

Epigenetic Modifications

Epigenetic modifications, including and histone alterations, play a pivotal role in regulating accessibility and during adipogenesis, enabling the transition from mesenchymal progenitors to mature adipocytes. These dynamic changes facilitate the activation of pro-adipogenic programs while silencing anti-adipogenic pathways, distinct from direct transcriptional control by factors like PPARγ and C/EBPs. DNA methylation patterns shift markedly during , with hyper of promoters for anti-adipogenic genes such as Wnt10b suppressing their inhibitory effects and allowing progression toward fat cell commitment. Concurrently, hypomethylation at the PPARγ locus enhances its expression, promoting the initiation of adipogenic through reduced of key regulatory regions. These changes are mediated by DNA methyltransferases (DNMTs) for maintenance and ten-eleven translocation () enzymes for active demethylation, where TETs oxidize to , facilitating gene activation. Histone modifications further fine-tune adipogenic gene accessibility, with acetylation of at 9 (H3K9ac) at enhancers recruited by coactivators CBP/p300 marking active adipogenic loci and correlating positively with progression. Trimethylation of H3 at 4 (H3K4me3) similarly promotes transcriptional activation of these genes, while inhibitors of deacetylases (HDACs), such as , enhance by increasing and boosting expression of markers. Post-2020 studies highlight enzymes' specific roles in adipogenesis; for instance, TET2 promotes white-to-beige conversion through demethylation at pro-browning gene loci, while loss of adipose TET2 and TET3 enhances β-adrenergic responses and thermogenic like Ucp1. Obesity induces lasting epigenetic alterations in adipose progenitors, including hypermethylation and aberrant marks that persist even after significant , impairing normal adipogenic plasticity and contributing to metabolic dysfunction. These reversible epigenetic landscapes, particularly in response to environmental cues, underpin the adaptability of beige activation, allowing metabolic flexibility beyond fixed transcriptional cascades.

Non-coding RNAs

Non-coding RNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), exert post-transcriptional control over adipogenesis by modulating mRNA stability, translation, and interactions with regulatory proteins. These molecules fine-tune the of preadipocytes into , , or adipocytes, influencing accumulation and . miRNAs typically bind to target mRNAs to induce degradation or translational repression, while lncRNAs can act as miRNA sponges, scaffolds for protein complexes, or direct modulators of activity. Several miRNAs play pivotal roles in adipogenic regulation. The miR-17-92 cluster promotes early differentiation by targeting and repressing Rb2/p130, a inhibitor that otherwise restricts the mitotic clonal expansion phase necessary for C/EBPβ . In contrast, miR-143 and miR-145 enhance PPARγ expression and adipogenesis by inhibiting ERK5 signaling, which normally suppresses ; their upregulation during adipogenic accelerates lipid droplet formation in 3T3-L1 cells. miR-130, part of the miR-130/301 family, suppresses adipogenesis, particularly the fate, by directly binding the 3' of PPARγ mRNA to reduce its expression and impair in beige precursors. LncRNAs contribute to adipogenesis through and protein interactions. HOTAIR regulates depot-specific in adipose-derived cells by binding to transcription end sites and interacting with polycomb repressive complex 2 (PRC2) to modulate adipogenic pathways. The steroid receptor RNA activator (SRA) lncRNA modulates PPARγ activity by direct binding, stabilizing the receptor and amplifying its transcriptional output to drive late-stage differentiation and insulin sensitivity in mature adipocytes. These non-coding RNAs exhibit tissue-specific expression patterns, with certain miRNAs and lncRNAs enriched in to favor lipid storage, whereas others, like miR-455, are upregulated in and depots to promote thermogenic programs via targeting HIF1an and activating AMPK signaling. Mechanistically, miRNAs induce mRNA through Argonaute-mediated in RISC complexes, while lncRNAs often as competing endogenous RNAs (ceRNAs) to sequester miRNAs, thereby derepressing pro-adipogenic transcripts. Recent studies from 2021-2023 highlight therapeutic potential, such as CRISPR-mediated knockdown of obesity-promoting lncRNAs like HOTAIR in models, which reduces and improves glucose tolerance without altering core transcriptional cascades. This layered RNA regulation coordinates with epigenetic modifications to ensure precise control of fate.

References

  1. [1]
    Adipogenesis at a glance | Journal of Cell Science
    Aug 15, 2011 · This Cell Science at a Glance article reviews the transition of precursor stem cells into mature lipid-laden adipocytes.
  2. [2]
    Molecular Mechanisms of Adipogenesis: The Anti-adipogenic Role ...
    This review aims to provide a comprehensive knowledge of the key molecular factors (proteins and various signaling pathways) involved in adipocyte ...
  3. [3]
    Modeling Adipogenesis: Current and Future Perspective - PMC - NIH
    Oct 20, 2020 · This review focuses on examining the physiological importance of adipogenesis, the current approaches that are employed to model this tightly controlled ...
  4. [4]
    Distinct adipose progenitor cells emerging with age drive active ...
    Apr 25, 2025 · Adipogenesis is the process by which new adipocytes are generated through the proliferation and differentiation of adipose progenitor cells ( ...
  5. [5]
    Adipogenesis: A Complex Interplay of Multiple Molecular ...
    Jun 16, 2020 · Adipogenesis consists of two phases, namely commitment and terminal differentiation. This review discusses the role of signalling pathways ...Missing: seminal | Show results with:seminal
  6. [6]
    An established preadipose cell line and its differentiation in culture ...
    Cell. 1975 May;5(1):19-27. doi: 10.1016/0092-8674(75)90087-2. Authors. H Green, O Kehinde. PMID: 165899; DOI: 10.1016/0092-8674(75)90087-2. Abstract. When cells ...
  7. [7]
    An established preadipose cell line and its differentiation in culture II ...
    When cells of the established preadipose line 3T3-L1 enter a resting state, they accumulate triglyceride and convert to adipose cells.
  8. [8]
    Adipogenesis: A Necessary but Harmful Strategy - PMC
    Jul 26, 2019 · Adipogenesis is a complex physiological process, necessary to sequester lipids effectively to avoid lipotoxicity in other tissues, like the liver, heart, ...2. Adipose Tissue · 2.1. Wat · 3. Adipogenesis In Response...
  9. [9]
    Adipogenesis - PMC - PubMed Central
    This review will focus on transcriptional control of adipogenesis and its regulation by several endocrine hormones. The general functions and cellular origins ...Missing: seminal | Show results with:seminal
  10. [10]
    Adipogenesis as a Potential Anti-Obesity Target - PubMed Central
    Jan 8, 2021 · This review focuses on available conventional medicine and natural products to prevent or treat obesity by targeting adipogenesis and discussing ...
  11. [11]
    PRDM16 controls a brown fat/skeletal muscle switch - Nature
    Aug 21, 2008 · Here we show by in vivo fate mapping that brown, but not white, fat cells arise from precursors that express Myf5, a gene previously thought to ...Results · Prdm16 Binds To Ppar-γ And... · Online Methods
  12. [12]
    Perilipin1 promotes unilocular lipid droplet formation through the ...
    Mar 12, 2013 · White adipocytes, specialized in energy storage, are characterized by their large unilocular LDs. LD size correlates with the susceptibility to ...
  13. [13]
    Uncoupling Protein 1 of Brown Adipocytes, the Only Uncoupler - NIH
    Uncoupling protein 1 (UCP1), is a unique mitochondrial membranous protein devoted to adaptive thermogenesis, a specialized function performed by brown ...Missing: multilocular seminal<|separator|>
  14. [14]
    The common and distinct features of brown and beige adipocytes
    Both brown fat and beige fat possess thermogenic properties in addition to common morphological and biochemical characteristics.Missing: seminal | Show results with:seminal
  15. [15]
    The developmental origins of adipose tissue - PMC - PubMed Central
    Over the last few decades, the major focus of the field has been cell culture modeling primarily studying in vitro adipogenesis. The term adipogenesis first ...Brown And White Fat: Flavors... · White Adipose Tissue: The... · Adipose Stem Cells: Getting...Missing: 1970s | Show results with:1970s
  16. [16]
  17. [17]
  18. [18]
  19. [19]
  20. [20]
  21. [21]
    Stimulation of adipogenesis in fibroblasts by PPAR gamma 2, a lipid ...
    We demonstrate here that retroviral expression of PPAR gamma 2 stimulates adipose differentiation of cultured fibroblasts.
  22. [22]
    Genome-wide profiling of PPARγ:RXR and RNA polymerase II ...
    The nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) is a key regulator of adipocyte differentiation in vivo and ex vivo and has been shown ...
  23. [23]
    An antidiabetic thiazolidinedione is a high affinity ligand ... - PubMed
    Jun 2, 1995 · We report that thiazolidinediones are potent and selective activators of peroxisome proliferator-activated receptor gamma (PPAR gamma).
  24. [24]
    Cascade regulation of terminal adipocyte differentiation by three ...
    Jan 15, 1995 · Here we provide evidence that C/EBP delta and C/EBP beta play early catalytic roles in the differentiation pathway, relaying the effects of the hormonal ...
  25. [25]
    Regulated expression of three C/EBP isoforms during adipose ...
    The accumulation of C/EBP beta and C/EBP delta reached a maximal level during the first 2 days of differentiation and declined sharply before the onset of C/EBP ...
  26. [26]
    PPARγ Is Required for the Differentiation of Adipose Tissue In Vivo ...
    All of these studies are gain-of-function experiments and have clearly shown that PPARγ is sufficient to induce adipogenesis in a variety of cell types. There ...
  27. [27]
    Human Metabolic Syndrome Resulting From Dominant-Negative ...
    Apr 1, 2003 · Humans with dominant-negative mutations in PPAR-γ represent a novel subtype of inherited partial lipodystrophy. The paucity of limb and ...Research Design And Methods · Studies Of Insulin... · Results
  28. [28]
    Molecular regulation of adipogenesis - PubMed - NIH
    Adipogenesis, or the development of fat cells from preadipocytes, has been one of the most intensely studied models of cellular differentiation.Missing: hierarchical cascade
  29. [29]
    Transcriptional regulation of adipogenesis - Genes & Development
    ... adipocytes when appropriate hormonal inducers were added (Green and Kehinde 1974, 1975, 1976). These lines, designated 3T3-L1 and 3T3-F442A, were isolated ...
  30. [30]
    Article Cross-Regulation of C/EBPα and PPARγ Controls the ...
    Four members of the C/EBP family, α, β, δ, and CHOP-10, are expressed at specific times during adipogenesis in a manner that is consistent with a distinct ...
  31. [31]
    C/EBPα induces adipogenesis through PPARγ: a unified pathway
    These results indicate that C/EBPα and PPARγ participate in a single pathway of fat cell development with PPARγ being the proximal effector of adipogenesis.
  32. [32]
    Function of pref-1 as an inhibitor of adipocyte differentiation - Nature
    Nov 1, 2000 · Downregulation of pref-1 is required for adipose conversion and constitutive expression of pref-1 inhibits adipogenesis. Conversely, decreasing ...
  33. [33]
    Inhibition of adipogenesis and development of glucose intolerance ...
    Preadipocyte factor-1 (Pref-1) is a transmembrane protein highly expressed in preadipocytes. Pref-1 expression is, however, completely abolished in ...
  34. [34]
    Transcriptional Control of Brown Fat Determination by PRDM16 - PMC
    We show here that the zinc-finger protein PRDM16 is highly enriched in brown fat cells compared to white fat cells.
  35. [35]
    The Insulin-Like Growth Factors in Adipogenesis and Obesity
    Insulin-like growth factor (IGF)-I is a critical mediator of preadipocyte proliferation, differentiation, and survival. ... Results from clinical studies on GH ...
  36. [36]
    Differential Roles of Insulin Receptor Substrates in Brown Adipocyte ...
    Insulin promotes adipocyte differentiation via a complex signaling network involving multiple insulin receptor substrates (IRSs).
  37. [37]
    Insulin Stimulates Adipogenesis through the Akt-TSC2-mTORC1 ...
    This study demonstrates that activation of mTORC1 signaling is a critical step in adipocyte differentiation and identifies TSC2 as a primary target of Akt ...
  38. [38]
    Development of insulin-responsive glucose uptake and GLUT4 ...
    May 15, 1998 · Differentiating human fat cells in primary culture develops an insulin-responsive glucose transport system which exhibits a high stability.
  39. [39]
    Insulin-like Growth Factor-1/Insulin Bypasses Pref-1/FA1-mediated ...
    IGF-1/ insulin-dependent signaling plays an important role in trigger- ing the phase of mitotic clonal expansion during adipocyte differentiation.
  40. [40]
    The role of C/EBPδ in the early stages of adipogenesis - ScienceDirect
    These findings show that C/EBPδ is involved in MCE and gene expression in the early stages of adipocyte differentiation.
  41. [41]
    3T3-L1 differentiation into adipocyte cells protocol - Abcam
    This protocol uses insulin, IBMX and dexamethasone to chemically induce the differentiation of 3T3-L1 cells into adipocytes.
  42. [42]
    An optimised protocol for the investigation of insulin signalling ... - NIH
    In this study, we report that adjusting the composition of the adipogenic differentiation cocktail influences adipocyte function, and we propose that hormonal ...
  43. [43]
    Article Hyperinsulinemia Drives Diet-Induced Obesity Independently ...
    Dec 5, 2012 · Collectively, these in vivo data suggest that high-fat diet-induced hyperinsulinemia promotes adult adipocyte hypertrophy and nutrient storage.
  44. [44]
    Article Adipose Tissue Selective Insulin Receptor Knockout Protects ...
    We find that FIRKO mice have markedly reduced fat mass and whole-body triglyceride stores, and are protected from gold thioglucose-induced and age-related ...
  45. [45]
    Impaired Thermogenesis and Adipose Tissue Development in Mice ...
    To investigate the overlapping signaling created by insulin and IGF-1, we created mice with a combined tissue specific knockout of the insulin and IGF-1 ...
  46. [46]
    Insulin/IGF-I Regulation of Necdin and Brown Adipocyte ...
    Based on these results, we propose that insulin/IGF-I act through IRS-1 phosphorylation to stimulate differentiation of brown preadipocytes via two ...
  47. [47]
    Hyperinsulinemia in Obesity, Inflammation, and Cancer
    Mar 29, 2021 · Multiple lines of evidence demonstrate that maximal adipocyte hyperplasia and hypertrophy require fully intact insulin action. The ...
  48. [48]
    Wnt/β-catenin signaling in adipogenesis and metabolism - PMC
    Research conducted over the last decade has established the Wnt/β-catenin signaling pathway as an important regulator of adipocyte differentiation.
  49. [49]
    Wnt Signaling: From Mesenchymal Cell Fate to Lipogenesis and ...
    Jul 28, 2021 · Wnt signaling suppresses adipocyte differentiation by inhibiting expression of PPARγ and C/EBPα, the central regulators of adipogenesis. B: The ...
  50. [50]
    Running Against the Wnt: How Wnt/β-Catenin Suppresses ...
    Feb 8, 2021 · The main role of Wnt/β-catenin signaling is to suppress differentiation while increasing MSC and preadipocytes cell mass.Missing: chick quail
  51. [51]
    How mesenchymal stem cells transform into adipocytes - NIH
    Mar 26, 2024 · BMP2/4/7 use SMAD1/5/8 signaling to regulate adipocyte commitment. BMP2, BMP4, and myostatin ligands affect the adipocyte commitment of MSCs.Missing: ALK seminal
  52. [52]
    Adipose tissue plasticity and the pleiotropic roles of BMP signaling
    BMP4 stimulates angiogenesis, which serves as stem cells niches for adipogenesis; BMP2,4,7 all play roles in commitment of multipotent stem cells to ...Missing: seminal | Show results with:seminal
  53. [53]
    New role of bone morphogenetic protein 7 in brown adipogenesis ...
    BMP-7 activates a full program of brown adipogenesis including induction of early regulators of brown fat fate PRDM16 and PGC-1 (PPARγ coactivator-1) α, ...Missing: ALK seminal
  54. [54]
    Bone Morphogenetic Protein (BMP) Signaling in Adipose Tissue
    BMP4 and BMP7 are the most studied BMPs in adipose tissue, with major roles in white and brown adipogenesis, respectively.
  55. [55]
    Hypertrophy of Adipose Tissues in Quail Embryos by in ovo Injection ...
    As Pparγ and Fabp4 have been used as well-known adipogenic markers, up-regulation of the two genes suggests promotion of adipocyte differentiation in embryonic ...
  56. [56]
    TGF-β/SMAD signaling regulation of mesenchymal stem cells in ...
    Jan 29, 2020 · This review focuses on the roles of TGF-β/SMAD signaling in adipocyte commitment of MSCs. BMP4 and BMP7 signaling are sufficient to induce adipocyte lineage ...Bmp4 Signaling In The... · Bmp2 Signaling In The... · Tgf-β Signaling In The...Missing: seminal | Show results with:seminal<|control11|><|separator|>
  57. [57]
    The impact of cellular senescence in human adipose tissue - PMC
    May 17, 2023 · Senescent AT is characterized by dysfunctional cells, increased inflammation, decreased insulin sensitivity and lipid storage.
  58. [58]
    The role of adipose tissue senescence in obesity - Portland Press
    Adipogenesis is a process by which preadipocytes differentiate into mature adipocytes. Adipogenesis can be regulated by multiple factors including cellular ...
  59. [59]
    Targeting senescent cells enhances adipogenesis and metabolic ...
    Dec 19, 2015 · Our study indicates targeting senescent cells or their products may alleviate age-related dysfunction of progenitors, adipose tissue, and metabolism.
  60. [60]
    Targeting senescent cells alleviates obesity-induced metabolic ...
    Our results implicate cellular senescence as a causal factor in obesity-related inflammation and metabolic derangements.
  61. [61]
    Adipose Stromal Cell Expansion and Exhaustion - PubMed Central
    PMCID: PMC7226766 PMID: 32252348. Abstract. Adipose tissue (AT) is comprised of a diverse number of cell types, including adipocytes, stromal cells ...
  62. [62]
    Senolytic drugs, dasatinib and quercetin, attenuate adipose tissue ...
    Dasatinib is a tyrosine kinase inhibitor that primarily targets senescent adipocyte progenitor cells, while quercetin inhibits anti‐apoptotic BCL‐xL, HIF‐1α, ...
  63. [63]
    Epigenetic Regulation of Adipogenesis in Development of Metabolic ...
    Jan 12, 2021 · Modified cytosine(C) residue, 5-methylcytosine (5mC), established by DNMTs could easily be reverted to unmodified C by ten eleven translocation (TET) enzymes ( ...Adipose Tissue: White, Brown... · Altered Adipokine Production... · Environment-Epigenetic...<|separator|>
  64. [64]
    Regulation of Cyclin D1 and Wnt10b Gene Expression by cAMP ...
    Simultaneously, hypermethylation of the Wnt10b gene promoter suppresses binding of CREB, allowing adipogenesis to proceed. Previous article in issue; Next ...
  65. [65]
    CpG Methylation across the adipogenic PPARγ gene and its ... - NIH
    Jan 26, 2017 · We reported that hypomethylation of the PPARγ CpG site cg10499651 was associated with increased PPARγ expression as measured by both real ...
  66. [66]
    Functional Implications of DNA Methylation in Adipose Biology
    Apr 16, 2019 · During adipogenesis, PPARγ, via the physical interaction with TET1, increases local demethylation around PPARγ-binding sites (27). In mature ...
  67. [67]
    MLL3/MLL4 are required for CBP/p300 binding on enhancers ... - NIH
    Apr 8, 2017 · MLL3/MLL4 are required for CBP/p300 binding on enhancers activated during adipogenesis. Further, MLL4 and CBP identify super-enhancers (SEs) of adipogenesis.
  68. [68]
    Histone modifications in development - Company of Biologists journals
    Jun 13, 2025 · In line with this, H3K9ac levels positively correlate with adipogenesis progression (Yoo et al., 2006). These (and other findings we cannot ...
  69. [69]
    Adipose tissue retains an epigenetic memory of obesity after weight ...
    Nov 18, 2024 · We show that both human and mouse adipose tissues retain cellular transcriptional changes after appreciable weight loss.
  70. [70]
    A negative feedback loop between TET2 and leptin in adipocyte ...
    Apr 1, 2024 · Ten-eleven translocation (TET) 2 is an enzyme that catalyzes DNA demethylation to regulate gene expression by oxidizing 5-methylcytosine to ...
  71. [71]
    LncRNA-Mediated Adipogenesis in Different Adipocytes - PMC
    Jul 5, 2022 · The noncoding RNAs include snRNA (small nuclear RNA), siRNA (small interfering RNA), miRNA (microRNA), lncRNA (long noncoding RNAs), and circRNA ...
  72. [72]
    miR-17-92 cluster accelerates adipocyte differentiation by negatively ...
    These data indicate that miR-17-92 promotes adipocyte differentiation by targeting and negatively regulating Rb2/p130.
  73. [73]
    miRNA regulation of white and brown adipose tissue differentiation ...
    In addition to targeting PTN, miR-143 has previously been shown to promote adipocyte differentiation through targeting of extracellular signal regulated kinase ...
  74. [74]
    miR-130 Suppresses Adipogenesis by Inhibiting Peroxisome ... - NIH
    Among them, miR-130 strongly affected adipocyte differentiation, as overexpressing miR-130 impaired adipogenesis and reducing miR-130 enhanced adipogenesis. A ...
  75. [75]
    Unique role for lncRNA HOTAIR in defining depot-specific gene ...
    May 26, 2022 · The genes selectively activated by HOTAIR are associated with adipogenesis (Fig. 1D) and myogenesis (Supplemental Fig. S1C). Intriguingly ...
  76. [76]
    MicroRNA‐455 regulates brown adipogenesis via a novel HIF1an ...
    These data suggest that miR‐455 may work in concert with sympathetic signals to recruit beige adipocyte differentiation. Figure EV1. miR‐455 induced brown ...Missing: 34654847 | Show results with:34654847
  77. [77]
    Roles of Long Noncoding RNAs in Adipogenesis and Obesity ...
    This review focuses on recent research concerning the functions of lncRNAs in the regulation of white and brown adipogenesis, emphasizing their roles in ...Differentiation · C/ebp Family Proteins · Dlk1/pref1
  78. [78]
    Non-Coding RNAs and Adipogenesis - MDPI
    Jun 10, 2023 · This article outlines the process of adipogenesis, and discusses updated roles and mechanisms of ncRNAs in the development of adipocytes.