Previous Article in Journal
Functional HER1/HER2-Expressing Murine Tumor Models for Preclinical Evaluation of Targeted Therapies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Critical Evaluation of Adipogenic Cell Models: Impact of the Receptor Toolkit on Adipogenic Potential

by
Andrea Gutiérrez-García
1,†,
Francisco Javier Olivas-Aguirre
2,3,† and
Miguel Olivas-Aguirre
1,4,*
1
Laboratory of Cancer Pathophysiology, University Center for Biomedical Research, University of Colima, Colima 28040, Mexico
2
Conahcyt National Laboratory of Body Composition and Energetic Metabolism (LaNCoCoME), Tijuana 22390, Mexico
3
Medical and Psychology School, Autonomous University of Baja California, Tijuana 22390, Mexico
4
Secretaría de Ciencia, Humanidades, Tecnología e Innovación (Secihti) Programa de Investigadores e Investigadoras por México, México City 03940, Mexico
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Receptors 2025, 4(4), 19; https://doi.org/10.3390/receptors4040019
Submission received: 11 February 2025 / Revised: 23 July 2025 / Accepted: 28 September 2025 / Published: 15 October 2025

Abstract

Adipocyte cell models are essential for investigating adipogenesis, yet methodological inconsistencies pose challenges to obtaining reproducible and physiologically relevant results. Murine cell lines, such as 3T3-L1 and OP9, are commonly utilized due to their established adipogenic capabilities. However, differences in its metabolic, genetic regulation, and receptor signaling raise concerns about their applicability to human adipose biology. Human-derived models, including mesenchymal stem cells (hMSCs) and preadipocyte cell lines, offer a closer approximation to in vivo adipogenesis but display significant variability in differentiation efficiency. This variability is often compounded by heterogeneous differentiation protocols, variations in cell confluence, and unstandardized pharmacological induction strategies. A pivotal factor influencing adipogenic potential is the receptor toolkit, which dictates cellular responses to differentiation stimuli. This study systematically evaluates key receptors—PPARγ, glucocorticoid receptors (GR), insulin receptor (IR), thyroid hormone receptors (TR), estrogen receptors (ER), and adenosine receptors (AR)—across commonly used adipocyte models to assess their roles in adipogenic regulation. Additionally, we examine the impact of pharmacological agents capable of inducing adipogenesis (adipogens) and the methodological inconsistencies that contribute to variations in adipocyte differentiation. By addressing these factors, we aim to elucidate the extent to which receptor variability influences experimental outcomes and propose a more structured approach to interpreting adipogenesis research. This critical assessment underscores the need for greater methodological transparency and receptor profiling to enhance the reliability of adipocyte models in metabolic research. Standardizing differentiation methodologies while accounting for receptor diversity will be essential for refining in vitro models and improving their translational potential in the study of obesity, diabetes, and other metabolic disorders.

1. Introduction

Adipogenesis is the physiological process through which precursor cells differentiate into mature adipocytes, playing a fundamental role in energy storage, endocrine function, and metabolic homeostasis. Given its central regulatory role, adipose tissue is closely linked to the onset and progression of metabolic disorders, including obesity, insulin resistance, type 2 diabetes mellitus, and cardiovascular disease [1]. The differentiation of adipocytes is controlled by a complex interplay of transcription factors, signaling pathways, and extracellular regulators that collectively determine cellular fate and function [2]. Although critical regulators such as PPARγ, C/EBPα, and insulin-mediated pathways have been extensively studied, a key gap persists in understanding how variability in receptor expression modulates adipogenic potential. A more refined characterization of the molecular determinants governing adipogenesis is essential to develop targeted therapeutic strategies aimed at addressing the underlying causes of metabolic disease.
While several comprehensive reviews have provided a foundational framework for the study of adipogenic models—notably addressing their utility and limitations [3,4,5]—emerging evidence continues to expand and enrich the field. In particular, there is growing interest in investigating additional variables, such as receptor expression profiles, which may critically influence the efficacy of the induction protocols. Our review aims to integrate this evolving knowledge and highlight receptor-driven mechanisms as a means to address methodological challenges that limit the reproducibility and clinical applicability of in vitro adipogenic models.
Historically, murine-derived preadipocyte cell lines, such as 3T3-L1 and OP9, have been widely used due to their well-characterized adipogenic responses and differentiation capacity. However, species–specific differences in metabolic regulation, transcriptional activity, and receptor-mediated signaling impose significant limitations on their relevance to human adipose tissue biology [6]. While human MSCs and preadipocyte-derived cell lines provide a more physiologically relevant alternative, they face several limitations: many adipogenic human cell lines origin from pathologies, their differentiation potential is highly variable and lacks standardization. Furthermore, commonly employed adipogenic differentiation protocols rely on a diverse set of pharmacological inducers—including insulin, dexamethasone (DEX), isobutyl-1-methylxanthine (IBMX), and peroxisome proliferator-activated receptor gamma (PPARγ) agonists—without adequately considering the extent to which receptor expression influences differentiation efficiency [7]. The assumption that all preadipocyte models share comparable receptor profiles has led to discrepancies in lipid accumulation, gene expression patterns, and overall differentiation outcomes, complicating cross-study comparability and hindering mechanistic insights into adipogenesis.
To address key methodological limitations in adipogenesis research, this study systematically profiles the expression of critical receptors—PPARγ, glucocorticoid receptors (GR), thyroid receptors (TR), estrogen receptors (ER), insulin receptors (IR), retinoid receptors (RXR), and adrenergic receptors (AR)—across widely used adipogenic models. By evaluating how receptor expression influences differentiation dynamics, we aim to optimize adipocyte differentiation protocols and enhance their reproducibility. To strengthen this framework, we also critically assess existing literature on receptor-mediated regulation, incorporating comparative data from studies using techniques such as flow cytometry, Western blotting, RNA sequencing, and PCR. This integrated approach seeks to clarify current inconsistencies, improve the physiological relevance of in vitro models, and advance the translational potential of adipogenesis research.

2. Critical Evaluation of In Vitro Adipogenesis Models: Methodological Considerations and Regulatory Mechanisms

2.1. Human and Murine Models of Adipogenesis: Implications and Limitations

Numerous scientific publications report the generation of mature adipocytes from precursor cells of both human and murine origin. However, a major complication with these primary models lies in the inherent limitations of accessibility and the high cost and challenges associated with culturing primary cells. To address this need, commercial providers offer human mesenchymal stem cells (hMSCs) or human multipotent adipose-derived stem cells (hMADS) sourced from various tissues, including adipose tissue (AT), bone marrow (BM), and umbilical cord matrix from individual donors. However, the source of each sample can vary, and crucial aspects such as the donor’s race, ethnicity, gender, age, or health status remain undetermined, as these details are protected. Research has shown that these factors can significantly influence the phenotype of the cells being studied, leading to variability in results. Consequently, the lack of control over these variables makes it challenging to draw consistent and reproducible conclusions across studies. Furthermore, while these commercial providers claim that the cells are tested to demonstrate their differentiation capacity, the efficiency of differentiation can vary significantly between batches, even when cultured under identical conditions.
Additionally, many suppliers suggest differentiation protocols based on their proprietary products, which typically claim to improve differentiation efficiency [8]. However, the composition of these protocols is often confidential for commercial reasons. This situation complicates the reproducibility of results, as researchers worldwide are unable to replicate the experiments due to the lack of standardized, well-characterized cellular models.
Therefore, it is imperative that adipocyte research be conducted using more universally applicable and reproducible models, such as well-characterized cell lines. To identify the most commonly used and commercially available models, we conducted a comprehensive search through multiple databases, including Google Scholar, Scopus, and PubMed, using a Boolean strategy with “adipogenesis” as the primary keyword combined with cell line names as search variants. The subsequent results, analysis and discussion in this work focused on the most prominent models with sufficient characterization to allow for meaningful comparison and interpretation. This selection criterion was arbitrarily defined by the authors but is supported by the frequency and popularity of use of these models in the scientific literature. While this may be considered a source of bias and a limitation, the contribution of alternative models to the field remains disproportionately low, which reinforces the relevance and focus of our approach. As part of our exclusion criteria, we deliberately omitted non-commercially available cells and hMSCs and hMADS, acknowledging that although some of these models are available and valuable for adipogenesis research, they are derived from individual donors and thus inherently subject to inter-individual, source, racial, genetic, age, and sex-related variability. Such donor-specific information is often inaccessible to researchers, particularly when obtained from commercial sources, which imposes significant limitations for studies focused on receptor-dependent mechanisms.
The analysis of the adipogenic cells lines revealed that approximately 75% of the models used in adipocyte research are murine, with the minority derived from human sources (Figure 1A). Additionally, we observed significant disparities in the tissue origins and the phenotypic and functional characteristics of these cell lines. Among human cell lines, only HS-5 is derived from BM, while the others originate from subcutaneous/abdominal adipose tissue (SGBS, PAZ6, LS14, ASC52telo) and the retroperitoneal region (LiSa-2).
Regarding murine cell lines, most are derived from various tissues such as BM, subcutaneous and epididymal AT, calvaria, with a smaller number originating from embryonic or perinatal sources (Figure 1A, right). Among human-derived cell lines, the SGBS cell line is the most widely used for adipogenesis. However, its origin, from a pediatric donor with Berardinelli–Seip syndrome, raises questions about the representativeness of this model. On the other hand, LiSa-2 and LS14 cells, which are derived from liposarcoma, further highlight that 50% of the most commonly used human cell lines in adipogenesis research originate from pathological conditions.
Furthermore, the database analysis, revealed that the 3T3 cell line (parental and derivatives such as 3T3-L1 and 3T3-F442A) is by far the most widely used for adipogenesis studies. Despite discrepancies in the number of results across databases (due to varying search algorithms), the trend remained consistent across all platforms, clearly indicating that murine (embryionic) 3T3 cells are overwhelmingly the predominant cell line used for adipogenesis research. It also highlights the need for a more standardized and reproducible approach using well-characterized human cell lines to address the limitations and inconsistencies in current adipogenesis models.
In this regard, efforts have been made to develop human-derived systems such as hMADS cells (hMADS 1–6), which exhibit high proliferative capacity and can maintain their adipogenic potential for up to 200 passages. However, these cells are not commercially available through major international suppliers, which significantly restricts their accessibility and global reproducibility. Moreover, hMADS cells were established from pediatric donors between 4 months and 4 years old, undergoing various surgical procedures, and not necessarily classified as healthy [9]. The adipose tissue was obtained from umbilical cord and pubic regions, which, combined with the donor age, confers these cells biological characteristics distinct from adult-derived adipose progenitors.
Indeed, infant-derived MSCs display enhanced resistance to senescence and replicative stress, as well as constitutively elevated expression of genes such as SOD1-3, c-MYC, p16, p21, p53, PPARγ, and LPL [10]. These features, while experimentally advantageous, limit the extrapolation of findings to adult adipogenesis and raise concerns about the physiological relevance of this model in contexts seeking to emulate adult adipose tissue behavior.
The overreliance on models with unknown or pathological origins complicates the comparability and applicability of findings across different research groups. Moreover, it is imperative to consider the limitations associated with using cell lines that harbor diverse chromosomal abnormalities when compared to primary cells. For instance, the 3T3-L1 cell line, which is widely employed in adipogenesis studies, possesses an aneuploid and unstable karyotype. Such aberrations may potentially influence cellular behavior and differentiation capacity, theoretically favoring or limiting adipogenesis, and thereby affecting the accurate extrapolation of results to a healthy physiological context. Detailed karyotypic information and other key cellular observations are provided and will be discussed later in Table S1.
Moreover, the source from which adipocytes are isolated plays a crucial role, as these cells exhibit distinct gene expression patterns and demonstrate variable responses to signaling pathways in vitro. The microenvironmental context in which these cells were originally present has been shown to significantly influence their differentiation capacity. For instance, adipocytes derived from subcutaneous tissue exhibit a much higher differentiation potential compared to those from omental adipose tissue. This distinction is often overlooked by many researchers who use and compare cell lines without considering that adipogenesis efficiency may vary depending on the tissue source [11,12,13].
Taken together, the observations discussed throughout this section underscore the need to refine the selection of adipogenic cell models toward systems that more accurately reflect the biology of adult human adipocytes. The most widely used models today—whether murine, pediatric, or tumor-derived—present significant limitations, including genetic instability, donor-related variability, and restricted physiological relevance. These factors contribute to marked differences in adipogenic capacity that do not necessarily correspond to the behavior of adipose tissue in adult humans. Furthermore, inconsistencies in tissue origin, undefined donor metadata, and non-standardized differentiation protocols hinder reproducibility and limit the extrapolation of results. Moving forward, it is essential that the field prioritize models with well-characterized origins and stable phenotypes, derived from adult sources, in order to strengthen the translational value of adipogenesis research and reduce variability across studies.

2.2. From Precursor Cells to Adipocytes: Stages and Molecular Regulation of Adipogenesis

Adipogenesis is a multistage process by which precursor cells (MSC, fibroblasts or preadipocytes) progressively acquire the characteristics required to become mature adipocytes. This differentiation follows a defined sequence of genetic, morphological, and functional changes, which can be broadly divided into the following stages: commitment, induction, early differentiation, and late differentiation (Figure 2). During the commitment phase, MSCs, identified by the expression of surface markers such as CD73, CD90, and CD105, while lacking CD14, CD11b, CD34, CD45, CD79a, CD19, and HLA-DR [14], transition into preadipocytes. MSCs exhibit self-renewal potential and the ability to differentiate into multiple lineages (multipotency), including adipocytes, chondrocytes, and osteoblasts. Specific lineage regulators dictate MSC fate, such as Runx2 for osteoblastogenesis [15], Sox9 for chondrogenesis [16], and for adipogenesis, the CAAT/enhancer-binding protein (C/EBP) family—particularly C/EBPα—and the peroxisome proliferator-activated receptor gamma (PPARγ), both of which are essential positive regulators.
The transformation of MSCs into adipocytes is not direct but involves the generation of an intermediate cell type called the preadipocyte. Compared to MSCs, preadipocytes lack multipotency and their differentiation potential is restricted to adipocytes under optimal conditions. Morphologically, both MSCs and preadipocytes are heterogeneous fusiform cells, devoid of significant cytoplasmic complexity, and exhibit high proliferative potential. As a result, markers such as Ki67 are commonly used to identify preadipocytes before their terminal differentiation, along with other early differentiation markers such as C/EBPβ, C/EBPδ, Pref-1, and ZFP423 [17].
While the specific molecular events driving the differentiation of MSCs into preadipocytes are not fully understood, recent reviews have discussed the role of several pathways and microenvironmental factors in this process [14]. During the commitment phase, preadipocytes proliferate extensively, and this step is considered critical for adipocyte generation, as reported by multiple independent groups, despite the variability in their adipogenic models. It is important to note that high cell confluence promotes cell cycle arrest in adherent cells when the culture surface is saturated, leading to inhibition of proliferation upon cell-to-cell contact. Once at high confluence, preadipocytes, restricted to a specific lineage and arrested in the cell cycle, can be supplemented with an adipogenic cocktail to initiate early differentiation. This stage is characterized by the appearance of small lipid droplets in the cytoplasm and an increase in metabolic activity. The duration of this stage is highly dependent on the culture conditions and models used, but it typically lasts for the first 7 days of culture. Subsequently, adipocyte maturation involves further specialization, during which the cell demonstrates its capacity for lipid storage, leading to noticeable changes in morphology. The cell becomes ellipsoid with larger lipid droplets, which can be single or multiple, depending on the type of adipocyte generated. It is important to recognize that the classical classification of unilocular white adipocytes and multilocular brown adipocytes does not reliably recapitulate in vitro. Rather than serving as a clear distinction, this morphological criterion often complicates the interpretation of results, as many immortalized and even primary white adipocyte models display a multilocular phenotype. Therefore, studies claiming to generate specific adipocyte types should validate their identity through a more precise and systematic approach using well-established markers. Functional markers (e.g., enzymes and regulatory proteins), as well as specific surface markers, have been proposed for decades and continue to be refined to improve the accuracy of adipocyte characterization in vitro [18,19,20].
The molecular determinants driving adipogenic differentiation from pre-adipocytes are graphically shown in Figure 2B. PPARγ was initially identified as the essential factor for adipogenesis. It is considered the master regulator because it is necessary for differentiation and, in some models, sufficient by itself to induce differentiation. Various transcription factors and receptors work together to collaborate with PPARγ, including members of the C/EBP family, such as C/EBPβ and C/EBPδ, which are early factors associated with early differentiation. These are activated in pre-adipocytes to promote the transcription of PPARγ itself, which in turn enhances the expression of other C/EBP family members, particularly C/EBPα. This creates a positive feedback loop, further enhancing the expression of PPARγ. Adipogenesis research has been focused on PPARγ, as it has been experimentally shown that pre-adipocytes can differentiate into adipocytes in the absence of C/EBPα, but not without PPARγ [21]. The loop generated by C/EBPα is considered a later-stage process and a means to consolidate differentiation by regulating genes involved in adipocyte specialization and function. The activity of PPARγ is mediated by ligands but is also enhanced by interaction with other nuclear receptor members, such as retinoid receptors (RXR) [22]. The formation of the PPARγ/RXR heterodimer induces conformational changes that facilitate ligand binding and interaction with PPAR response elements (PPRE) in the promoters of target genes, promoting gene expression and mediating the transition from pre-adipocyte to mature adipocyte. Other nuclear receptors have been linked to the adipogenic differentiation process, such as TR; however, their role in differentiation is not fully understood and may vary positively or negatively depending on the experimental context (discussed later). Nonetheless, TR ligands often promote PPARγ expression. Finally, other receptors, such as GR and insulin receptors, are responsible for promoting the expression of early differentiation molecules, such as C/EBPδ and C/EBPβ, which cooperatively favor PPAR γ expression. These are, in general, the primary regulatory factors of the adipogenic program (Figure 2B).

2.3. Pharmacological Regulators of Adipogenesis In Vitro

It was initially shown that PPAR receptors could be physiologically activated by micromolar concentrations of fatty acids. However, three decades ago, a family of antidiabetic compounds known as thiazolidinediones (TZDs), including rosiglitazone, pioglitazone, and troglitazone, was identified as potent PPARγ activators, with nanomolar affinity for its isoforms [23]. Since then, the inclusion of TZDs in adipogenic cocktails has become widely used. Additionally, insulin is employed in the adipogenic cocktails, as it regulates early expression of key transcription factors such as C/EBPβ and C/EBPδ, while downregulating C/EBPα through the insulin receptor (InsR) [24]. Insulin also promotes glucose uptake via GLUT4 expression, facilitating triglyceride accumulation in adipocytes. Notably, insulin sensitivity varies across models, with OP9 cells being more sensitive and effective than 3T3-L1 cells [25]. This is significant because high, chronic insulin concentrations can lead to reprogramming that negatively impacts insulin signaling, glucose uptake, lipid droplet size, and induces basal glucose uptake and lipolysis disruptions [26]. Isobutylmethylxanthine (IBMX), a non-selective, non-competitive phosphodiesterase (PDE) inhibitor, promotes increased cAMP levels, thereby activating protein kinase A, which is essential for the transcriptional activation of PPARγ and C/EBPβ [27,28]. Finally, glucocorticoids (GCs) have proven to be effective inducers of early differentiation, contributing to the expression of PPARγ and various C/EBP family members, such as C/EBPβ, C/EBPδ, and C/EBPα [29,30,31]. In some studies, triiodothyronine (T3) has been added to adipogenic cocktails, although its use is less common. It has been shown to promote not differentiation, but rather the transition from white adipocytes to brown adipocytes (browning), enhancing mitochondrial biogenesis and the expression of thermogenic proteins like UCP-1 [32,33,34]. Similarly, indomethacin has been used as an adjunct in adipogenic cocktails, as it has been demonstrated to partially activate the PPARγ receptor directly [35]. More recently, indomethacin has been shown to promote browning during adipogenesis, warranting cautious use and thorough evaluation of the phenotypic outcomes of the generated adipocytes [36].

2.4. Current Protocols for Adipocyte Differentiation: Methodological Approaches and Key Limitations

The differentiation of adipocytes in vitro has become a crucial model for understanding adipogenesis and related metabolic processes. Various protocols have been developed, utilizing a range of pharmacological agents to induce differentiation. A comprehensive search of protocols related to adipocyte differentiation was conducted using the terms “Adipocytes differentiation AND…” across several adipogenic cell lines, without temporal restrictions. This approach aimed to provide a broad understanding of the key factors influencing adipogenic differentiation. A total of 55 protocols were included, from which essential parameters were extracted, including starting cell confluence, the composition of the adipogenic cocktail, protocol duration, and differentiation confirmation methods (Table 1).
Table 1. Characteristics of the protocols employed for adipocyte differentiation from several cell lines.
Table 1. Characteristics of the protocols employed for adipocyte differentiation from several cell lines.
Murine Cell Lines
White Adipocyte Cells
Cell lineOrganismInitial Cell Confluency
(n Well in Plate/cm2)
Culture Medium EmployedDifferentiation Cocktail EmployedDifferentiation DaysDifferentiation EfficiencyValidation MethodReference
3T3-L1Mouse50,000
(24)
DMEM-HG
10% BCS
100 U/mL penicillin
100 μg/mL streptomycin
0.25 μg/mL amphotericin B
1 μg/mL insulin
0.025 µM DEX
0.5 mM IBMX
7NDNile Red staining
mRNA expression of Pparg, Fabp4 & Plin1
[37]
NSDMEM-HG
10% FBS
100 U/mL penicillin
100 mg/mL streptomycin
0.02 μg/mL insulin
0.250 µM DEX
0.5 mM IBMX
7NDOxygen consumption[38]
NSDMEM-HG
110 mg/L sodium pyruvate
10% BCS
6 μg/mL insulin
0.1 µM DEX
0.5 mM IBMX
1 µM ROSI
250 µM INDO
1080% Oil Red O staining[39]
2000–3000
(cm2)
DMEM-HG
10% BCS
100 U/mL penicillin
100 mg/mL streptomycin
5 μg/mL insulin
1 μM of DEX
0.5 μM IBMX
2 μM ROSI
1490% Oil Red O staining[40]
30,000
(96)
DMEM-HG
10% BCS
100 U/mL penicillin
100 mg/mL streptomycin
1.0 μg/mL insulin
0.5 mM IBMX
10NDNile Red staining[41]
10,000
(24)
DMEM/F12
100 U/mL penicillin
100 mg/mL streptomycin
10% FBS
10 μg/mL insulin
1 μM DEX
0.5 mM IBMX
20 μM rosiglitazone
895% Oil Red O staining[42]
NSDMEM
100 mg/mL of kanamycin
10% FBS
10 µg/mL insulin
1µM DEX
0.5 mM IBMX
570% Oil Red O staining[43]
30,000
(cm2)
DMEM with 1 g/L glucose
10% FBS
100 U/mL penicillin
100 mg/mL streptomycin
10 g/L-glutamine
1 μg/mL insulin
0.25 μM DEX
0.5 mM IBMX
1490% Oil Red O staining
PLIN-1 expression
FABP expression
[44]
NSDMEM F-12
10% BCS
100 U/mL penicillin
100 mg/mL streptomycin
0.5 μg/mL insulin
5 μM DEX
0.5 mM IBMX
1 μM ROSI
1 nM T3
7100% Lipid quantification[45]
30,000
(96)
DMEM-HG
10% BCS
100 U/mL penicillin
100 mg/mL streptomycin
1.0 μg/mL insulin
0.5 mM IBMX
1080% Triglyceride Content[46]
NSDMEM-HG
L-Glutamine
10% BCS
100 U/mL penicillin
2 µM insulin
1 µM DEX
0.025 mM IBMX
16–2090% Oil Red O staining[47]
NSDMEM-HG
10% BCS
100 U/mL penicillin
10 μg/mL insulin
1 μM DEX
0.5 mM IBMX
6100% Oil Red O staining
Triglyceride accumulation assay
[48]
NSDMEM
10% FBS
100 μg/mL streptomycin
100 U/mL penicillin
250 ng/mL fungizone
10 μg/mL insulin
0.1 μM DEX
0.5 mM IBMX
890% Oil Red O staining[49]
NSDMEM
10% BCS
2 mM l-glutamine
100 U/mL penicillin
100 U/mL streptomycin
0.01 µg/mL insulin
1 μM DEX
0.25 mM IBMX
255% PPARG expression[50]
NSDMEM
10% FBS
500 mM IBMX
200 mM indomethacin
1 μM DEX
10 μM Insulin
10>50%Oil Red O staining[51]
3T3-F442AMouse5000
(6)
DMEM-HG
4 mmol/L L-glutamine
1.5 g/L sodium bicarbonate
10% BCS
100 μg/mL streptomycin
100 U/mL penicillin
10 μg/mL insulin
1 μM rosiglitazone
780% Oil Red O staining[52]
NS
(48)
DMEM
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
10 μg/mL insulin 2NDTriglyceride accumulation assay[53]
3300
(cm2)
DMEM
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
5 μg/mL insulin
1 μM DEX
5 mM IBMX
6–890% Oil Red O staining[54]
NS
(48)
DMEM
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
10 μg/mL insulin 3NDLipidTox staining
Upregulation of PPARγ and C/EBP-α
[55]
70,000
(6)
DMEM
10% FBS
100 U/mL penicillin
100 μg/mL amphotericin
5 μg/mL insulin 8NDOil Red O staining
Cygb expression
[56]
NSDMEM-HG
10% FBS
50 μg/mL streptomycin
100 U/mL penicillin
58 μg/mL insulin
1 μM DEX
0.5 mM IBMX
12NDOil Red O staining [57]
OP9Mouse50,000
(24)
αMEM
20% FBS
26 mM sodium bicarbonate
100 U/mL penicillin
100 μg/mL streptomycin
0.25 μg/mL amphotericin B
1 μg/mL insulin
0.025 μM DEX
0.5 mM IBMX
7100% Nile Red staining
mRNA expression of Pparg, Fabp4 & Plin1
[37]
NSαMEM
5% FBS
100 U/mL penicillin
100 μg/mL streptomycin
1 mM rosiglitazone1080%Oil Red O staining[58]
60,000
(48)
αMEM
20% FBS
100/mL penicillin
100 µg/mL streptomycin
2 M L-glutamine
10 µg/mL insulin
0.25 µM DEX
0.25 mM IBMX
6NDOil Red O staining[59]
2000
(384)
αMEM
10% FBS
100/mL penicillin
100 µg/mL streptomycin
GLUTAMAX
5 µg/mL insulin
10 µM DEX
0.5 mM IBMX
6NDNile Red imaging
Trygliceride content assay
[60]
NSDMEM
5% FBS
1 mM rosiglitazone
10 µg/mL insulin
1 μM DEX
0.500 mM IBMX
1 μM rosiglitazone
8NDOil Red O staining
mRNA expression of Pparg, Fabp4 & Plin1
[61]
NSαMEM
20% FBS
100 U/mL Penicillin
100 mg/mL Streptomycin
292 mg/mL L-glutamate
10 µg/mL insulin
1 µM DEX
0.25 mM IBMX
280% PPARG expression[50]
NSαMEM
20% FBS
100 U/mL Penicillin
100 mg/mL Streptomycin
292 mg/mL L-glutamate
10 µg/mL insulin
1 µM DEX
250 mM IBMX
2NDPPARG expression[62]
400,000
(24)
DMEM
10% FBS
10 µg/mL insulin570% Oil Red O staining[63]
NSαMEM
20% FBS
1 μg/mL of insulin
1 μM DEX
0.5 mM IBMX
580% NS[64]
D1-ORL-UVAMouse500
(24)
DMEM
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
5 µg/mL insulin
0.01 μM DEXA
50 µM INDO
940%Oil Red O staining[65]
1000
(24)
DMEM
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
5 µg/mL insulin
0.01 μM DEXA
50 µM INDO
15NDNS[66]
BMS2Mouse50,000
(NS)
DMEM
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
1 µg/mL insulin
0.25 μM DEXA
0.5 mM IBMX
880%Oil Red O staining[67]
20,000
(24)
DMEM-HG
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
5 µM hydrocortisone
0.5 mM IBMX
5 μM Rosiglitazone
25 μM Pioglitazone
60 µM INDO
680%Oil Red O staining[68]
OB17Mouse NSDMEM
10% FBS
33 μM biotin
17 μM sodium pantothenate
Penicilin
Streptomycin
17 nM insulin
1.5 nM T3
14NDNS[69]
3000/cm2DMEM
200 U/mL penicilin
50 μg Streptomycin
33 μM biotin
17 μM pantothenate
17 nM Insulin
2 nM T3
19NDND[70]
NSDMEM
antibiotics
33μM biotin
17 μM pantothenate
170 nM Insulin
1.5 nM T3
18NDND[71]
NSDMEM
antibiotics
33 μM biotin
17 μM pantothenate
170 nM Insulin
1.5 nM T3
16NDND[72]
MS5 NS aMEM
10% FCS
5 µg/mL insulin 15NDND [73]
19,000–38,000
(12)
DMEM
10% FBS
100 U/mL Penicilin
100 μg/mL streptomycin
2 mM L-glutamine
10 mM HEPES
1 mM sodium piruvate
50 µM mercaptoethanol
STEMPro adipogenesis Kit14–2195%Oil Red O staining [74]
Brown adipocyte cells
C3H/10T1/2 Mouse20,000
(cm2)
DMEM
10% FBS
5 µg/mL insulin
0.25 µM DEX
0.5 mM IBMX
1290% Oil Red O staining[75]
NSDMEM
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
1% L-glutamine
5 µg/mL insulin
0.2 µM DEX
0.5 mM IBMX
5 µM rosiglitazone
6–880% Oil Red O staining[76]
NSDMEM
10% FBS
5 μg/mL of insulin
0.5 μM DEX
0.25 mM IBMX
50 μM INDO
535% Oil Red O staining[51]
5000
(96)
DMEM
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
1% L-glutamine
10 µM insulin
1 μM DEX
0.5 mM IBMX
0.5 µM rosiglitazone
0.25 μM INDO
780% Oil Red O staining[77]
NSDMEM
10% FBS
500 mM IBMX
200 mM INDO
1 μM DEX
10 μM Insulin
10>50%Oil Red O staining[51]
T37iMouseNSDMEM/Ham’s F12
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
0.058 µg/mL insulin
2 nM T3
8NDmRNA expression[78]
10,000
(24)
DMEM/Ham’s F12
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
0.6 µg/mL insulin
2 nM T3
995%Oil Red O staining
Trygliceride content assay
[79]
NSDMEM/Ham’s F12
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
0.12 µg/mL insulin
2 nM T3
735%mRNA expression[80]
HIB1BMouseNSDMEM
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
10 µg/mL insulin
0.25 μM DEX
0.5 mM IBMX
8NDlipolytic protein markers[81]
NSDMEM
10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
0.12 µg/mL insulin
0.25 μM DEX
0.5 mM IBMX
1 nM T3
7 90%Oil Red O staining[82]
BFC-1MouseNSDMEM
10% FBS
200 U/mL penicillin
50 μg/mL streptomycin
0.058 µg/mL insulin
2 nM T3
NSNDmRNA expression[83]
Human in vitro cellular model
White adipocyte cells
Cell lineOrganismInitial cell confluency
(n well in plate/cm2)
Culture medium employedDifferentiation cocktail employedDifferentiation daysDifferentiation Efficiency (based on ORO staining and microscopic analysis)Validation MethodReference
HS-5HumanNS10% FBS
100 U/mL penicillin
100 μg/mL streptomycin
1% L-glutamine
OriCellTM Supplement For Human Related Stem Cells Adipogenic Differentiation A-I, A-II & BNS80%Oil Red O staining[84]
20,000
(75 cm2)
DMEM
10% FBS
1% antibiotic/antimycotic
10 µg/mL insulin
500 µM DEX
0.5 mM IBMX
100 µM INDO
1990%Oil Red O staining[85]
300,000
(cm2)
αMEM
10% FBS
5 µg/mL insulin
0.5 µM DEX
0.25 mM IBMX
50 µM INDO
1480%Oil Red O staining[86]
LiSa-2Human10,000
(cm2)
DMEM/Ham’s F12
10 mg/mL transferrin
15 mM NaHCO3
15 mM HEPES
33 mM biotin
17 mM pantothenate
100 U/mL penicillin
100 μg/mL streptomycin
5 µg/mL insulin
0.02 nM T3
1 µM cortisol
2030%Sudan red and hematoxylin[87]
NSDMEM/Ham’s F12
2% FBS
100 U/mL penicillin
100 μg/mL streptomycin
5 µg/mL insulin
1 mM DEX
1 nM T3
9NDOil Red O staining[88]
NSDMEM-F12
33 µM biotin
17 µM pantothenic acid
100 U/mL penicillin/streptomycin
10% FBS
0.005 µg/mL insulin
0.02 nM T3
1 µM cortisol
NSNDMicroscopy[89]
SGBSHumanNSDMEM-F12
33 µM biotin
17 µM pantothenic acid
100 U/mL penicillin/streptomycin
10% FBS
0.1 µg/mL insulin
0.025 μM DEX
0.5 mM IBMX
2 µM rosiglitazone
0.02 nM T3
0.1 µM cortisol
28NDMicroscopy[89]
800,000
(15 cm2)
DMEM-F12
66 µM biotin
33 nM pantothenic acid
2% penicillin/streptomycin antibiotics
0.5 µg/mL insulin
0.095 µM DEX
1.9 mM IBMX
7.6 µM rosiglitazone
0.76 nM T3
380 nM cortisol
1290%Secretome[90]
NS
(6)
DMEM-F12
3 µM biotin
17 µM pantothenic acid
0.5% penicillin
0.5% streptomycin
0.5% amphotericin B
0.116 µg/mL insulin
0.25 µM DEX
0.5 mM IBMX
2 µM troglitazone
0.2 nM T3
100 nM cortisol
7NDTriglyceride accumulation assay[91]
NS
(12)
DMEM-F12
33 µM biotin
17 µM pantothenic acid
100 U/mL penicillin/streptomycin
10% FBS
0.1 µg/mL insulin
0.025 µM DEX
0.5 mM IBMX
2 µM rosiglitazone
0.02 nM T3
100 nM cortisol
12NDRNAm and Oxygen consumption[92]
LS14Human500
(96)
DMEM-F-12
33 µM biotin
17 µM pantothenic acid
100 U/mL penicillin/streptomycin
1 µM insulin
0.25 mM IBMX
2 µM rosiglitazone
1 nM T3
No glucocorticoids
1058%Oil Red O staining[93]
Human100,000
(24)
DMEM/F12 (1:1)
33 µM biotin
0.5% penicillin
0.5% streptomycin
17 µM pantothenic acid
10 µg/mL apotransferrin
200 µM ascorbate phosphate
4 µM oleic acid/BSA
4 µM linoleic acid/BSA
1 µM human insulin
1 nM T3
2 µM rosiglitazone
1 µM methoprene acid (RXR ligand)
1 µM T0901317 (LXR ligand)
250 µM IBMX
No glucocorticoids
1070–80% mentioned in the text. ND[94]
ASC52TeloHumanNDDMEM/F12 (1:1)
GlutaMAX 1x
250 mM sodium piruvate
10% FBS
100 nM insulin
500 µM DEX
0.25 mM IBMX
2 µM rosiglitazone
21<10%
Oil red O staining[95]
ND
(12)
DMEM
Low glucose
10% FBS

10 µg/mL insulin
1 µM DEX
0.5 mM IBMX
2128%PPARy activation in comparison to MSCs[96]
Brown adipocyte cells
PAZ6HumanNSDMEM/Ham’s F12
2% FBS
100 U/mL penicillin
100 μg/mL streptomycin
5 µg/mL insulin
1 μM DEX
1 nM T3
9NDOil Red O staining[88]
NSDMEM-Ham’s F-12
8% FBS
15 mmol/l HEPES
5 µg/mL insulin
0.1 μM DEX
0.25 mM IBMX
1 mM pioglitazone
1 nM T3
14NDOil Red O staining[97]
1,500,000
(NS)
DMEM-Ham’s F-12
8% FBS
15 mmol/l HEPES
5 µg/mL insulin
0.1 μM DEX
0.25 mM IBMX
1 mM pioglitazone
1 nM T3
14NDOil Red O staining[98]
10,000
(cm2)
DMEM-Ham’s F-12
8% FBS
15 mmol/l HEPES
100 U/mL penicillin
100 μg/mL streptomycin
0.25 mM IBMX
0.1 nM T3
14NDNS[99]
NSDMEM-Ham’s F-12
8% FBS
15 mmol/l HEPES
100 U/mL penicillin
100 μg/mL streptomycin
3 µg/mL insulin
0.1 μM DEX
0.25 mM IBMX
1000 nM pioglitazone
1 nM T3
14NDNS[100]
WT-1Human NSDMEM
10% FBS
penicillin
streptomycin
20 nM insulin
1 μM DEX
0.5 mM IBMX
0.125 mM INDO
1 nM T3
6NDNS[101]
DMEM
10% FBS
5 μg/mL insulin
1 μM DEX
0.5 mM IBMX
0.5 μM rosiglitazone
880–90%Oil Red O Staining [102]
NS: Not Specified; ND: Not determined; FBS: Bovine Fetal Serum; BCS: Bovine Calf Serum; DEX: dexamethasone; DMEM: Dulbecco’s Modified Eagle Medium; DMEM-HG: Dulbecco’s Modified Eagle Medium with high glucose; HEPES: 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid; INDO: indomethacine; IBX: isobutylmethylxanthin; Pparγ: PPARγ1/2; T3: triiodothyronine.
The analysis of adipogenic differentiation protocols across multiple murine and human cell lines reveals substantial heterogeneity in both the composition and concentrations of the compounds used—even among protocols applied to the same cell line. While insulin, DEX, and IBMX remain core components (Figure 2C), their concentrations vary dramatically, ranging from nanomolar to millimolar levels, often without clear pharmacological justification. Additional agents such as rosiglitazone, pioglitazone, indomethacin (INDO), T3, and cortisol are selectively included, further diversifying differentiation strategies. No species–specific trends in compound selection or protocol design were consistently observed, with the notable exception of T3, which appears almost exclusively in protocols targeting brown or beige adipogenesis rather than classical white adipocytes. Interestingly, some protocols—such as those used for the SGBS or LS14 cell line—employ complex cocktails containing up to six non-redundant pharmacological agents. In contrast, in other models like 3T3-F442A, OP9, or MS5 effective adipogenic differentiation has been achieved even with minimalistic approach, often relying on a single modulator such as insulin or, in the case of OP9, rosiglitazone alone (Table 1). These observations highlight not only the diversity of strategies employed but also the adaptability of certain cell lines to simplified induction conditions.
It is imperative to objectively justify the use of different drug concentrations in adipogenic differentiation protocols. While many compounds are indispensable for the induction and early or late stages of differentiation, their use at excessively high concentrations or for prolonged periods can lead to cytotoxic effects. This issue becomes particularly evident in protocols that feature atypical combinations or unreferenced concentrations of adipogenic inducers. For instance, IBMX, commonly used at concentrations between 250 and 500 µM, has been reported at a questionably high concentration of 500 mM in 3T3-L1 and C3H/10T1/2 cells [51]. Similarly, indomethacin has been used across a wide range—from 250 nM [77] to 250 µM [39], and even up to 200 mM [51]. DEX, typically used at low micromolar concentrations (0.01–1 µM), has been employed at significantly higher levels, such as 10 µM [60], 100 µM [85], and up to 500 µM [95], which can be cytotoxic depending on the cell type. Due to such toxicity, some researchers have even chosen to exclude DEX from their protocols [93]. In addition, PPARγ agonists, which have nanomolar affinity, are sometimes used at unnecessarily high micromolar concentrations and in redundant combinations, such as 5 µM rosiglitazone with 25 µM pioglitazone [42]. Or even 1 mM for OP9 cells [58]. These inconsistencies highlight the need for careful optimization and justification of experimental conditions to ensure reproducibility and facilitate meaningful comparisons across different cellular models.

2.5. Impact of the Culture Conditions on Adipogenic Differentiation

Considering that confluence is a determinant factor for the commitment of progenitor cells to specific differentiation lineages, understanding and standardizing confluence-related conditions is essential. High cell density alone has been shown to induce transcriptomic changes affecting thousands of genes relevant to differentiation potential [103], including those involved in membrane receptor expression, signal transduction, metabolic activity, and secretome composition. Also, it has been found that confluence levels directly influence lineage commitment in primary BM MSCs [104]. These findings highlight that confluence is not merely a preparatory condition, but a biological signal that can critically shape cell fate.
In this sense, a significant heterogeneity in adipogenic differentiation conditions was identified among multiple studies, particularly regarding the number of cells seeded, which ranged from 200 to 280,000 cells/cm2 (Table 1 and Figure 3A). Although one might assume that authors wait for cultures to reach confluence before initiating adipogenesis, this is not always the case. In several studies, the adipogenic cocktail was applied immediately after seeding, without allowing time for cells to reach confluence. This is relevant for two reasons: first, it may limit cell cycle arrest—a critical step in preadipocyte commitment to the adipogenic lineage—and second, it alters the theoretical drug-to-cell ratio, introducing an additional layer of variability to the differentiation outcome. While our comparative analysis of seeding density and differentiation efficiency (estimated from representative microscopic images) did not reveal a consistent correlation, these findings highlight the need to better define the specific dependencies of each cell type. For example, 3T3-L1 cells lose adipogenic potential at high confluence or after repeated passaging [25], whereas OP9 cells retain this capacity, regardless of confluence level. These intrinsic differences suggest that although high confluence is often a prerequisite, it must be carefully standardized. Overconfluent cultures, for instance, may trigger stress responses or lead to cell death, thereby compromising differentiation.
We strongly recommend that future studies report both the seeding density and the conditions under which adipogenic induction is initiated—whether by indicating the confluence level at day 0 or the number of days post-confluence. Standardizing and clearly documenting these parameters would greatly enhance the reproducibility and comparability of results across different models.
In this context, it is also important to highlight that even basic experimental conditions—such as the type of culture plate used—can substantially influence adipogenic differentiation efficiency. Mehra and colleagues, demonstrated that the surface properties of culture plates, despite being often overlooked, can alter differentiation outcomes by as much as 50%, even under otherwise optimal conditions. This finding underscores the critical role of seemingly minor technical details in shaping experimental results. We therefore consider it not only relevant but necessary to include such parameters in future protocols and discussions aimed at optimizing adipogenic differentiation strategies [105]. Furthermore, in line with this, adipogenic induction media itself can regulate cell cycle genes to promote arrest and facilitate differentiation, which may explain why, under appropriate induction conditions, high differentiation efficiencies can still be achieved regardless of initial cell number [106]. These findings underscore the need for further research on each cell line, focusing on the transcriptional and proteomic mechanisms governing adipogenesis, as a significant knowledge gap remains in this field.

2.6. Impact of the Receptor Toolkit on the Adipogenic Potential of Cellular Models

From a critical perspective, the rationale behind the considerable disparity in the number of compounds included in the adipogenic cocktails and the highly variable concentrations used remains unclear. In our view, the use and replication of differentiation protocols have been passed down over decades, adapting and transposing them from model to model without due consideration of the underlying principles governing each of the components and their molecular targets. There is a wrong assumption that all models—whether murine or human, normal or disease-derived, primary or cell line-based—possess comparable adipogenic machinery, proliferate equally, reach confluence at the same time, and exhibit identical affinities for ligands.
A thorough analysis of publications employing adipogenic cocktails reveals that most studies use the full array of adipogenic compounds without exploring the presence of their molecular targets, assuming their existence (Table S1). While many of the receptors discussed earlier (Section 2.3; Figure 2) are widely expressed across various tissues, the analysis conducted shows that, for the cell lines used in adipogenesis studies, characterization is often limited, and extremely scarce for comparative studies across different cell models. In general, it is evident that the most thoroughly characterized cell line in terms of adipogenic receptor profiling is the murine 3T3-L1 line, followed by the OP9 cells, which partly explains their popularity and consideration as useful and effective models for differentiation. However, other well-characterized models, such as the murine C3H/10T1/2 cells, have not gained comparable popularity. For the remaining cell lines, the situation is largely underexplored, and the available evidence is either nonexistent or extremely limited, as is the case for MS-5, D1 ORL UVA, BMS2, LiSa-2, PAZ 6, T37i, or WT-1 cells (Figure 3B).

2.6.1. PPARγ Receptor

PPARγ is unequivocally recognized as the master regulator of adipogenic differentiation, as it is both necessary and sufficient, in most of the cases, to drive the commitment of precursor cells into adipocytes. This pivotal role is underscored by its ability to orchestrate a complex transcriptional program, binding to thousands of sites as revealed by genome-wide profiling studies [107,108]. The essential requirement of PPARγ has been rigorously demonstrated through both in vivo and in vitro knockout models, where its absence results in a complete failure of adipogenic differentiation [109].
In line with this, our receptor expression analysis (Figure 3B) confirms that PPARγ is the most extensively studied and documented receptor across all adipogenic cellular models, except for MS-5 cells, where a targeted knockout (KO) of PPARγ has been reported. Notably, authors describe the MS-5 PPARγ KO as incapable of differentiation, reinforcing its indispensable nature. However, they fail to present comparative data from wild-type and KO conditions, limiting a comprehensive evaluation [110]. Given its prominence, numerous recent reviews continue to explore the role of PPARγ in adipogenesis. While PPARγ binding sites exhibit remarkably low conservation between human and murine models, the genes regulated by PPARγ remain conserved at approximately 50–60% [107]. While this suggests a significant evolutionary conservation of PPARγ function, it raises critical questions about the validity of widely used adipogenic models such as 3T3 or OP9 in fully capturing human adipocyte biology.
Despite the extensive documentation of PPARγ, it is imperative to acknowledge that its transcriptional activity depends on its heterodimerization with RXRs. Our analysis (Figure 3B) highlights that only a limited number of studies have thoroughly investigated RXR isoform expression in adipogenic models. This is important, as PPARγ-mediated transcriptional regulation is highly dependent upon RXR interactions, which may be model-dependent. For instance, in MSCs, RXRs act as pro-adipogenic co-regulators through PPARγ activation. However, in preadipocyte models such as 3T3 cells, RXR agonists paradoxically inhibit adipogenesis [111]. These recent findings suggest that while PPARγ expression is well-documented, its mere presence should not be interpreted as a definitive indicator of positive adipogenic regulation. Instead, deeper investigations into the interplay between nuclear receptors in specific cellular contexts are required to accurately predict a model’s responsiveness to adipogenic stimuli and its differentiation potential in response to agonists.

2.6.2. Glucocorticoid Receptors

The role of GRs in cellular differentiation has been firmly established in both 3T3-L1 cells and embryonic murine fibroblasts with GR silencing [112]. The absence of GRs significantly compromises the ability to activate the adipogenic program, reducing CEBPα by 66%, CEBPβ by 50%, CEBPδ by 66%, and PPARγ by 55%, even in the presence of insulin and IBMX. While these fibroblasts retain a residual capacity for differentiation, which can be rescued via PPARγ inducers, the notion that GR is non-essential for adipogenesis in vitro is misleading. Instead, it is clear that GR plays a substantial and non-redundant role in orchestrating the adipogenic cascade, favoring adipogenic fate instead of osteogenic differentiation [112,113].
Our previous analyses reinforce this notion (Figure 3B and Table 1), demonstrating a direct correlation between GR expression, DEX sensitivity, and differentiation capacity. Notably, comparative studies reveal that 3T3-L1 cells exhibit twice the GR abundance compared to OP9 cells, a distinction that aligns with the differential GCs concentrations required for adipogenesis induction in each cell line (Table 1).
Understanding the mechanisms by which GR mediates adipogenic responses is critical, as adipogenic induction efficiency is not merely dictated by receptor presence or abundance but by a deeper assessment of its functional state. GRs are typically sequestered in the cytoplasm by chaperone proteins, awaiting ligand binding to facilitate nuclear translocation. However, emerging evidence suggests that multiple cell types harbor membrane-bound GRs capable of engaging GCs to trigger rapid, non-genomic signaling events [114,115]. The presence and functional significance of these membrane GRs in preadipocytes or MSCs remain unexplored, yet their potential role as glucocorticoid buffers warrants further investigation.
A similar paradigm may explain glucocorticoid sensitivity discrepancies across cell models, particularly in the context of human GR isoforms. The GRα isoform exists in multiple splice variants (A, B, C1-3, D1-3), whereas GRβ is an alternative, permanently nuclear isoform that cannot bind glucocorticoids yet exerts dominant-negative effects by interfering with the transcriptional activity of GRα isoforms [116]. However, the ratio between GRs isoforms in adipose precursors and its significance in adipogenesis remains unexplored.
Given that the osteoblast–adipocyte balance in MSC differentiation is profoundly influenced by GC dosage and GR signaling dynamics [117], precise characterization of GR expression levels and isoform distribution in each cellular model is highly encouraged. This knowledge is essential for optimizing adipogenic differentiation protocols and advancing our understanding of GC-mediated lineage commitment in MSCs.

2.6.3. Thyroid Hormone Receptors

Thyroid hormones (predominantly T3) play a critical role in regulating the adipogenic program and the specialization of adipocytes toward thermogenesis, particularly in the formation of brown adipose tissue. T3 exerts its effects through both direct action on thyroid response elements and through diverse thyroid receptor isoforms, as well as by regulating accessory genes such as PPARγ and C/EBPα [118,119,120]. The variability and accessibility of these determinants across different cell lines may influence the capacity of T3 to promote adipogenesis and adipocyte specialization.
Recent studies have demonstrated the role of T3 in white adipocytes derived from mice. Specifically, knockout (KO) and expression of unique isoforms of TRα or TRβ have shown that TRβ is the predominant isoform mediating T3 action on genes involved in metabolism and the adipogenic program [121]. This highlights the importance of receptor isoforms in mediating T3’s effects on adipogenesis.
In line with this, independent research has demonstrated in standard cell lines such as 3T3-L1 that dominant negative mutant isoforms of TRα or TRβ limit adipogenesis, in 94% or 54%, respectively, compared to wild-type 3T3 cells, indicating a pro-adipogenic role for both receptors [119]. Interestingly, TRα appears to play a more dominant role in this model. It is noteworthy that the abundance of TR isoforms in specific cell lines may determine their differentiation efficiency, particularly when using isoform-specific ligands.
As discussed previously, thyroid receptors are not only essential for promoting adipogenesis but also play a pivotal role in the browning of white adipose tissue, which involves the adaptive thermogenic program [122]. This is an important consideration when adding thyroid hormone to adipogenic cocktails, as while T3 enhances adipogenesis through PPARγ and C/EBPα, its effects on browning cannot be overlooked.
The expression of TRα and TRβ in commonly used models such as OP9 and 3T3-L1 further underscores the relevance of these receptors in regulating key genes and factors essential for the adipogenic program. The differential expression of these receptors may offer greater or more direct regulation over the adipogenic process. However, whether this provides an advantage or redundancy with other receptors requires further investigation, as does the role of these receptors in other cellular models that remain underexplored in the context of adipogenesis.

2.6.4. Adrenoreceptors (AdrR)

Adrenoceptors constitute central regulators of adipose tissue biology, particularly in the control of lipolysis, thermogenesis, and the sympathetic adaptation of energy metabolism [123]. Yet, their role in adipocyte differentiation itself remains elusive. The evidence presents conflicting data depending on the model used, the differentiation stage, the adrenergic receptor subtype, and the experimental conditions.
Among the β-adrenoceptors, β3-AR is the most frequently associated with adipogenesis, particularly in murine white adipocyte models. In 3T3-L1 cells, β3-AR expression increases robustly during terminal differentiation, and its activation enhances triglyceride accumulation and promotes transcriptional programs involving PPARγ and C/EBPα, partly via phosphorylation and nuclear translocation of STAT-5. [124,125]. This suggests a possibly instructive role of β3-AR signaling in late-stage adipogenesis. In contrast, β1- and β2-ARs are expressed at lower levels, and their functional relevance in adipogenesis remains less defined. While both β1- and β2-adrenoceptors are constitutively expressed in adipocyte models, their regulation during differentiation diverges significantly across systems. While β1-AR expression tends to decrease during adipogenesis—a pattern observed both in murine models and primary human adipocytes—β2-AR displays a more complex profile. In primary adipocytes, differentiation is typically accompanied by upregulation of both β2-AR and β3-AR, supporting their role in metabolic maturation and responsiveness to catecholamines. In contrast, LS14 cells, derived from human liposarcoma, exhibit a decline in β2-AR expression during differentiation and do not express β3-AR at all. This deviation underscores the limitations of tumor-derived adipocyte models in recapitulating canonical adrenergic signaling patterns observed in healthy adipose tissue [93,94]. What does appear consistent across several models is that β3-AR activity is more closely linked to metabolic maturation and lipid storage, while β1- and β2-ARs are better characterized for their roles in lipolysis and acute stress response.
In human white adipocyte cell models, particularly in LS14 and ASC52telo cells, β3-AR is either absent or expressed in a small, functionally unresponsive subpopulation. This casts doubt on the translational relevance of murine findings to human adipogenesis and indicates a need for more refined, single-cell resolution analyses of receptor functionality. Intriguingly, in ASC52telo, less than 2% of the cells responded to β-AR agonists in calcium mobilization assays, despite molecular detection of the receptors—suggesting a disconnection between expression and downstream signaling [93,94,126].
Additionally, in the HS-5 human stromal cell line, pharmacological studies have documented the presence of functional β2-ARs, which appear to mediate prosurvival signaling. However, their specific role in adipogenesis remains undefined and requires further experimental exploration. These findings raise the possibility that adrenergic signaling in stromal-derived adipogenic models may influence not only differentiation but also cell viability.
The role of β-AR expression in adipogenesis posses another layer of complexity. Given that some components of the adipogenic cocktails, such as T3, enhance β3-AR expression in murine adipocytes [127], GC, present in nearly all adipogenic differentiation cocktails, induces β2-AR while repressing both β3-AR and UCP1 [128]. This glucocorticoid-driven reprogramming may interfere with the browning potential of adipocytes in vitro, a relevant confounder considering that β3-AR agonism is a well-established inducer of UCP1 expression and beige adipocyte recruitment. Yet, experimental validation of this effect has been inconsistent and limited to a minority of cell lines, further challenging the assumption of a conserved β-AR–UCP1 axis across models.
In stromal and bone marrow–derived cells (e.g., MS-5), β2- and β3-ARs are functionally present but their role in adipogenesis remains ambiguous. However, activation of these receptors leads to downregulation of CXCL12, a chemokine crucial for hematopoietic cell retention. Given that mature adipocytes also secrete significant amounts of CXCL12 [129], this link may be particularly relevant when adipogenic models are employed to study cancer–adipocyte interactions.
Taken together, current evidence suggests that adrenoceptor expression and function in adipogenesis are model- and context-dependent. The role of β3-AR appears most closely tied to browning or terminal lipid filling and metabolic maturation, rather than commitment or early differentiation per se. Meanwhile, β2-AR activity may reflect a glucocorticoid-influenced lipolytic program rather than an adipogenic one. Functional validation remains scarce; for instance, in ASC52telo cells, although molecular detection confirms the presence of β1-, β2-, and β3-ARs, only about 20% of the population expresses each isoform, and fewer than 2% of cells respond to non-selective β-AR agonists in single-cell calcium imaging assays. This indicates that receptor presence does not guarantee functional responsiveness, underscoring the need for combined molecular and functional assessments when interpreting adrenergic signaling in adipogenesis especially in human systems, and the widespread use of GCs in differentiation protocols complicates the intepretation of the possible role of adrenoreceptors in adipogenesis.

2.6.5. Liver X Receptors (LxRs)

LxRs are members of the nuclear receptor superfamily and exist in two isoforms: LxRα and LxRβ. They act as sensors of cholesterol and oxidized lipids, regulating genes involved in lipid metabolism and energy homeostasis. In the context of adipogenesis, both isoforms are considered permissive, as their activation enhances the activity of other nuclear receptors, such as RxRs, through the formation of functional heterodimers.
In murine cells, the role of LxR in adipogenesis has been documented more extensively, particularly in the OP9 and 3T3 (3T3-L1 and 3T3-F442A) cell lines. In OP9, LxRα is expressed approximately fourfold higher than in 3T3-L1, while LxRβ is expressed at similar levels in both lines [130]. LxRα has been shown to promote the expression of PPARγ; however, LxRα agonism for two days does not significantly increase PPARγ levels in OP9 cells, in contrast to what is observed in 3T3-L1. This difference may be attributed to the fact that OP9 cells represent a different stage of adipocyte differentiation and already express high basal levels of PPARγ. Nevertheless, LxRα activation increases triglyceride accumulation in OP9, indicating a relevant metabolic role independent of further PPARγ induction [130].
In contrast, in 3T3-L1 and 3T3-F442A cells—where LxRα expression is similar to LxRβ levels—LxRα agonism clearly promotes adipogenic differentiation. This can be explained by the receptor’s ability to induce PPARγ, whose basal levels are lower in these cells compared to OP9, and by its synergistic action with RxRs. Although retinoid receptors are also expressed in OP9, some isoforms are more abundant in 3T3 cells (Figure 3B), highlighting the importance of receptor abundance and cooperative interactions in the final outcome of adipogenesis. This distinction supports the idea that LxR activation does not act independently, but rather in coordination with the nuclear receptor network.
In murine brown adipocytes, LxR’s role has not been associated with the induction of adipogenesis per se, but rather with their functional specialization. In BAT the receptor acts as a transcriptional repressor of adrenergic receptors and, consequently, of cAMP-mediated UCP1 expression [131]. Thus, LxR functions as a browning repressor in this context, underscoring a functional distinction between adipocyte types.
In human models, cell lines remain poorly characterized with respect to LxR. However, administration of LxR ligands has been shown to increase insulin sensitivity in both the LiSa-2 cell line and in primary human adipocytes [132], which may partially explain the observed increase in triglyceride content in other models. Whether this effect is specific to LiSa-2 or can be generalized to other human adipocyte models remains unknown.

2.6.6. Adenosin Receptors (AdoRs)

AdoRs belong to the family of G protein-coupled receptors (GPCRs) and are involved in multiple physiological processes, including inflammation, energy metabolism, and cellular differentiation. In mammals and rodents, four subtypes have been identified: A1, A2A, A2B, and A3 with similar homology. These are encoded by distinct genes and differ in their affinity for adenosine as well as their coupling to different G proteins. Subtypes A1R and A3R primarily couple to Gαi/o proteins, which inhibit adenylate cyclase and reduce intracellular cyclic AMP (cAMP) levels. Conversely, A2AR and A2BR couple to Gαs proteins, stimulating adenylate cyclase and increasing cAMP, which is a key second messenger and regulator of adipogenesis, and modulation of its intracellular levels by AdoRs influences adipocyte differentiation depending on receptor subtype, cellular context, and expression patterns.
In hMSCs, the A2BR subtype is most abundantly expressed under basal conditions [133]. During adipogenic differentiation, the expression of A1R and A2AR increases, facilitating the induction of transcription factors C/EBPα and PPARγ, which are critical for adipocyte commitment and lipid accumulation. Further studies of the same group employing adipogenic cell lines suggest a differential role between AdoR types, whereas A2AR plays a more prominent role in early differentiation stages, A1R predominantly promotes lipogenesis [134,135].
Expression and function of AdoRs vary across experimental models. Murine cell lines such as 3T3-L1 and 3T3-F442A are the most well documented. In such models the overexpression of A1R and A2AR enhances the expression of adipocyte genes like lipoprotein lipase (LPL) and glycerol-3-phosphate dehydrogenase, while suppressing osteoblastic markers, thereby promoting adipogenesis. In contrast, A2BR activation inhibits adipogenesis and favors osteogenesis [134,136,137].
As previously mentioned, functional divergence among subtypes is attributed not only to their differential G protein coupling, but also to the efficiency to activate intracellular pathways, e.g., both A2AR and A2BR elevate cAMP, generally favoring adipogenesis. However, in 3T3-F442A cells, forskolin-induced cAMP elevation shows a biphasic effect: low concentrations (1 nM) stimulate adipogenesis, while high concentrations (1 µM) inhibit it [138]. These observations imply that although some receptor subtypes may share redundant signaling pathways—such as the Gαs-cAMP axis—their affinity for adenosine and their intrinsic ability to effectively transduce signaling can be critical determinants in adipogenic outcomes. Subtypes with high ligand affinity and efficient coupling may promote sustained activation of key transcriptional regulators, whereas others may trigger transient or insufficient signaling, leading to divergent cell fate decisions.
Additionally, for Ob17 cells, the stable transfection of A1R expression has been linked to reduced proliferation and enhanced lipid accumulation, suggesting that AdoRs may play dual roles in promoting adipocyte maturation and arresting cell growth. However, these functions remain understudied across most models [137].
In human models the expression of AdoRs is limited and mostly restricted to the SGBS cell line, where functional expression of ARs has been detected and indicates that A1R and A2AR contribute to regulation of differentiation and lipid accumulation, similar to hMSCs [126].

2.6.7. Estrogen Receptors (ERs)

ERs have been extensively studied for their roles in the epigenetic and metabolic regulation of adipocytes, as well as in adipose tissue function. However, their involvement in adipogenic precursor cells remains relatively underexplored. Among these receptors, ERα, ERβ, and GPER have been implicated in modulating adipogenic processes. In murine models, estrogens acting through these receptors have been shown to negatively regulate the differentiation of MSCs by suppressing the expression of key adipogenic transcription factors such as PPARγ and C/EBPα [139]. In contrast, in human MSCs, estrogens not only inhibit adipogenesis but also promote osteogenic differentiation programs [140].
The presence of estrogens in the culture medium has been associated with altered expression of specific receptor isoforms. For instance, in murine MSCs, estrogen exposure enhances ERα expression [139], whereas in human adipocyte precursors, ERβ expression is upregulated approximately threefold, while GPER expression is downregulated [141]. In the widely used 3T3-L1 cell line, ERα is the predominant isoform, followed by ERβ, although variations in receptor expression levels have been reported depending on the cell source (e.g., ATCC vs. ZenBio). Higher receptor expression has also been observed in OP9 cells, suggesting these cells may be particularly susceptible to estrogen-mediated inhibition of adipogenesis [130].
In C3H/10T1/2 cells, both ERα and ERβ are expressed, and estrogen treatment leads to a significant upregulation of bone morphogenetic protein-2 (BMP-2), with ERα producing a ninefold increase compared to a threefold increase by ERβ [142]. This finding is notable because BMP-2 has been shown to act as a repressor of adipogenesis in this cell model. It promotes osteogenic differentiation while limiting lipid accumulation and downregulating adipocyte-specific markers [143,144]. mRNA expression of both ER isoforms has also been reported in other cell lines, including LiSa-2, SGBS, and HS-5 [89,145]. However, the functional roles of these receptors in MSCs and their involvement in adipogenic differentiation remain hypothetical and largely untested.
Overall, current evidence suggests a predominantly suppressive role of ERs in the adipogenic models studied. A theoretical exception may exist in the BMS2 cell line, where estrogens have been shown to regulate 85 genes through ERα, including the upregulation of SFRP1–3 [73]. These secreted frizzled-related proteins are known to act as osteogenic inhibitors, thereby potentially favoring adipogenic differentiation. However, the direct role of ERα in the differentiation of BMS2 cells has not yet been experimentally confirmed.
When designing adipogenic experiments, it is important to consider the potential influence of culture components such as serum and phenol red, both of which have been reported to exhibit estrogenic activity [146,147]. This may partly explain the variability observed in adipogenic outcomes across different cell lines. Given these considerations, estrogen should be viewed not only as a physiological regulator but also as a potential experimental confounder or control in adipogenesis studies.

3. Perspectives and Critical Considerations

Current adipogenesis models provide critical insights into human adipose tissue biology, yet their relevance is highly dependent on the specific metabolic and physiological processes being investigated. Treating adipocyte models as universally representative of adipose biology overlooks key differences in species-specific regulation, depot-specific functions, and metabolic properties. For example, murine 3T3-L1 cells have long been the standard for studying adipocyte differentiation due to their well-characterized adipogenic pathways, yet their transcriptional regulation, insulin signaling, and receptor expression differ significantly from human adipocytes. In contrast, SGBS cells, derived from human subcutaneous fat, exhibit higher insulin sensitivity, greater GLUT4 expression, and a metabolic profile closer to human physiology. However, their differentiation is highly dependent on pharmacological activation of PPARγ, which may not accurately reflect adipogenesis in vivo [148]. These differences underscore the importance of selecting adipocyte models that align with the specific research question rather than assuming a single model can universally represent adipose tissue function.
Each adipocyte model offers a distinct perspective on adipose biology based on its tissue origin and metabolic properties. Subcutaneous-derived models (e.g., SGBS) provide insight into insulin-sensitive adipose depots, as they display reduced lipolysis and an anti-inflammatory cytokine profile [149,150]. In contrast, while also evaluating subcutaneous-derived murine models (e.g., 3T3-L1), they exhibit greater insulin resistance, significant increases of leptin expression and overall higher lipolytic activity [148], making them relevant for studying obesity-related metabolic dysfunction. Meanwhile, brown (e.g., T37i) serve as powerful tools for investigating adaptive thermogenesis, mitochondrial function, and energy expenditure, due to their high UCP1 expression and ability to undergo browning [151]. Recognizing these depot-specific and metabolic differences is essential for ensuring that in vitro adipogenesis research produces physiologically relevant findings that can be translated effectively to human metabolic health and disease.
The study of adipogenesis has heavily relied on in vitro models that, while useful, often introduce methodological biases affecting the interpretation and relevance of results to human physiology. One of the most significant sources of bias arises from the overrepresentation of specific cell lines, such as 3T3-L1, which exhibit a strong dependence on PPARγ for differentiation. This has led to an oversimplified view of adipogenesis, where PPARγ is often considered the primary driver of adipocyte differentiation, neglecting the roles of other nuclear receptors. The assumption that all adipocyte models respond uniformly to differentiation stimuli is problematic, as receptor expression and metabolic responses vary significantly between murine and human-derived models.
To compensate for these differences, researchers frequently adjust differentiation protocols by increasing the concentration of adipogenic inducers or prolonging induction times. While these modifications can enhance differentiation efficiency, they artificially override intrinsic cellular differences, potentially leading to non-physiological adipocyte phenotypes. For instance, the use of high doses of rosiglitazone to compensate for the low PPARγ sensitivity in SGBS cells leads to excessive activation of key pathways such as IRS-2, p85-PI3K, and GLUT4 [152], amplifying glucose uptake and lipid accumulation beyond normal physiological levels. This overstimulation not only artificially accelerates adipogenic differentiation but also biases the interpretation of model efficiency, as pharmacologically induced lipid accumulation does not necessarily reflect functionally adequate adipogenesis. Furthermore, by forcing differentiation, intrinsic depot-specific differences may be masked, causing SGBS adipocytes to exhibit characteristics similar to visceral adipocytes despite their subcutaneous origin.
Given that adipogenesis in human tissues is a gradual and highly regulated process, such pharmacological interventions can compromise the translational relevance of the model, leading to misinterpretations regarding adipocyte biology and metabolic function. Therefore, it is crucial to consider the effects of PPARγ supraphysiological activation when evaluating adipogenic differentiation in SGBS cells, avoiding inappropriate extrapolations that could distort our understanding of physiological adipogenesis mechanisms.
Similarly, low-insulin sensitivity models, such as LiSa-2 or ASC52telo, often require prolonged differentiation periods, which may induce metabolic shifts that resemble pathological states rather than normal adipogenesis. As previously demonstrated, during LiSa-2 differentiation, hyperinsulinemia led to insulin resistance and alterations in glucose uptake and lipid metabolism, highlighting that extended differentiation protocols may promote metabolic dysfunction rather than physiologically representative adipocyte characteristics [153].
Other perspectives include the urgent exploration of models such as T37i, which exhibit exceptionally high adipogenic efficiency—even under low confluence conditions—and differentiate rapidly within a short timeframe. These features offer notable experimental advantages, such as reduced resource requirements, lower cell density dependence, and high reproducibility. However, despite their practical appeal, T37i cells remain poorly characterized at the molecular level. Moreover, their origin from a malignant hibernoma raises concerns, as their neoplastic nature may result in atypical receptor profiles or altered signaling pathways. While these characteristics enhance their utility in vitro, they warrant caution when extrapolating findings to healthy brown adipose tissue physiology.
Thus, beyond inducers, factors like cell source, differentiation duration, and receptor profiling significantly influence experimental outcomes. The lack of standardization in these variables can lead to heterogeneous results, making it difficult to compare findings across different studies. This highlights the importance of characterizing receptor expression before inducing differentiation, as well as ensuring that experimental conditions align with the physiological properties of human adipose tissue. Moving forward, the selection of adipocyte models should be guided by the specific research question, rather than relying on a one-size-fits-all approach.

4. Conclusions

Despite decades of research, the field of adipogenesis continues to rely on assumptions that often remain unverified. One of the most prevalent assumptions is the constitutive expression of key receptors targeted by differentiation cocktails. Our review highlights that in most commonly used adipogenic models, the presence and abundance of nuclear and membrane receptors—particularly those mediating insulin, glucocorticoid, thyroid, and PPARγ signaling—is frequently presumed rather than experimentally confirmed. This gap undermines the reproducibility and interpretability of findings across studies and models.
Available evidence suggests that receptor expression levels and subtypes profiles play a decisive role in the differentiation potential of adipose precursors. However, even when receptor proteins are detected, functional characterization is essential, as several examples demonstrate that receptors may be present but fail to signal effectively, or may lead to unexpected anti- or pro-adipogenic effects depending on the cellular context.
It is increasingly evident that no universal differentiation strategy will suit all adipogenic models. Differences in donor origin, species, cell lineage, and intended adipocyte phenotype (e.g., white, beige, or brown) demand tailored approaches. Current adipogenic cocktails—often designed based on historical precedent rather than molecular rationale—should be re-evaluated and adjusted according to the specific receptor toolkit of the selected model. Doing so could significantly improve the efficiency, consistency, and physiological relevance of in vitro adipogenesis studies.
Ultimately, integrating receptor profiling as a standard step in model validation may pave the way for more accurate, reproducible, and translatable adipogenesis research.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/receptors4040019/s1, Table S1—Receptors’ expression across different cell lines. References [154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200] are cited in the Supplementary Materials.

Author Contributions

Conceptualization, M.O.-A.; methodology, M.O.-A., F.J.O.-A. and A.G.-G.; software, M.O.-A. and F.J.O.-A.; validation, M.O.-A., F.J.O.-A. and A.G.-G.; formal analysis, M.O.-A., F.J.O.-A. and A.G.-G.; investigation, M.O.-A., F.J.O.-A. and A.G.-G.; resources, M.O.-A.; data curation, M.O.-A., F.J.O.-A. and A.G.-G.; writing—original draft preparation, M.O.-A., and F.J.O.-A.; writing—review and editing, M.O.-A., F.J.O.-A. and A.G.-G.; visualization, M.O.-A., and F.J.O.-A.; supervision, M.O.-A.; project administration, M.O.-A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), grant number 303072 and the Secretaría de Ciencia, Humanidades, Tecnología e Innovación (Secihti) grant number CBF-2025-I-795 to MO-A.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data used in this study are included in the main text and Supplementary Material.

Acknowledgments

We would like to express our sincere gratitude to the Secretaría de Ciencia, Humanidades, Tecnología e Innovación (Secihti) for their support to M.O.-A through the “Investigadores por México” program. We thank the Secihti for the scholarship granted to A.G.-G. (CVU 204337), which supported this research. Additionally, we are deeply grateful for the support provided to F.J.O.-A through the Postdoctoral Fellowship Program (Accuracy of commercial heart rate monitors in overweight and obese young adults: an exploratory study for determining the maximum fat oxidation rate during exercise), which has significantly contributed to the success of this project. We also thank Oxana Dobrovinskaya for her support through her PRONACES project (grant number 303072).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sakers, A.; De Siqueira, M.K.; Seale, P.; Villanueva, C.J. Adipose-tissue plasticity in health and disease. Cell 2022, 185, 419–446. [Google Scholar] [CrossRef]
  2. Ambele, M.A.; Dhanraj, P.; Giles, R.; Pepper, M.S. Adipogenesis: A complex interplay of multiple molecular determinants and pathways. Int. J. Mol. Sci. 2020, 21, 4283. [Google Scholar] [CrossRef]
  3. Poulos, S.P.; Dodson, M.V.; Hausman, G.J. Cell line models for differentiation: Preadipocytes and adipocytes. Exp. Biol. Med. 2010, 235, 1185–1193. [Google Scholar] [CrossRef]
  4. Ruiz-Ojeda, F.J.; Rupérez, A.I.; Gomez-Llorente, C.; Gil, A.; Aguilera, C.M. Cell models and their application for studying adipogenic differentiation in relation to obesity: A review. Int. J. Mol. Sci. 2016, 17, 1040. [Google Scholar] [CrossRef] [PubMed]
  5. Vohra, M.S.; Ahmad, B.; Serpell, C.J.; Parhar, I.S.; Wong, E.H. Murine in vitro cellular models to better understand adipogenesis and its potential applications. Differentiation 2020, 115, 62–84. [Google Scholar] [CrossRef] [PubMed]
  6. Bahmad, H.F.; Daouk, R.; Azar, J.; Sapudom, J.; Teo, J.C.; Abou-Kheir, W.; Al-Sayegh, M. Modeling adipogenesis: Current and future perspective. Cells 2020, 9, 2326. [Google Scholar] [CrossRef] [PubMed]
  7. Kim, S.; Reed, E.; Monti, S.; Schlezinger, J.J. A data-driven transcriptional taxonomy of adipogenic chemicals to identify white and brite adipogens. Environ. Health Perspect. 2021, 129, 077006. [Google Scholar] [CrossRef]
  8. Cyagen. Available online: https://www.cyagen.com/us/en/product/Human-Related-Mesenchymal-Stem-Cells-Adipogenic-Differentiation-Kit.html (accessed on 10 February 2025).
  9. Rodriguez, A.M.; Pisani, D.; Dechesne, C.A.; Turc-Carel, C.; Kurzenne, J.Y.; Wdziekonski, B.; Dani, C. Transplantation of a multipotent cell population from human adipose tissue induces dystrophin expression in the immunocompetent mdx mouse. J. Exp. Med. 2005, 201, 1397–1405. [Google Scholar] [CrossRef]
  10. Wu, S.H.; Yu, J.H.; Liao, Y.T.; Liu, K.H.; Chiang, E.R.; Chang, M.C.; Wang, J.P. Comparison of the infant and adult adipose-derived mesenchymal stem cells in proliferation, senescence, anti-oxidative ability and differentiation potential. Tissue Eng. Regen. Med. 2022, 19, 589–601. [Google Scholar] [CrossRef]
  11. Zhang, P.; Zhang, B.; Shang, P.; Fu, Y.; Nie, R.; Chamba, Y.; Zhang, H. Comparative transcriptomic profiles of differentiated adipocytes provide insights into adipogenesis mechanisms of subcutaneous and intramuscular fat tissues in pigs. Cells 2022, 11, 499. [Google Scholar] [CrossRef]
  12. Nandy, A.; Rendina-Ruedy, E. Bone marrow adipocytes—Good, bad, or just different? Best Pract. Res. Clin. Endocrinol. Metab. 2021, 35, 101550. [Google Scholar] [CrossRef] [PubMed]
  13. Toyoda, M.; Matsubara, Y.; Lin, K.; Sugimachi, K.; Furue, M. Characterization and comparison of adipose tissue-derived cells from human subcutaneous and omental adipose tissues. Cell Biochem. Funct. 2009, 27, 229–235. [Google Scholar] [CrossRef] [PubMed]
  14. Liu, S.-S.; Fang, X.; Wen, X.; Liu, J.-S.; Alip, M.; Sun, T.; Wang, Y.-Y.; Chen, H.-W. How mesenchymal stem cells transform into adipocytes: Overview of the current understanding of adipogenic differentiation. World J. Stem Cells 2024, 16, 245–256. [Google Scholar] [CrossRef]
  15. Chan, W.C.W.; Tan, Z.; To, M.K.T.; Chan, D. Regulation and role of transcription factors in osteogenesis. Int. J. Mol. Sci. 2021, 22, 5445. [Google Scholar] [CrossRef]
  16. Yang, L.; Ren, Z.; Yan, S.; Zhao, L.; Liu, J.; Zhao, L.; Chen, D. Nsun4 and Mettl3 mediated translational reprogramming of Sox9 promotes BMSC chondrogenic differentiation. Commun. Biol. 2022, 5, 495. [Google Scholar] [CrossRef]
  17. Cawthorn, W.P.; Scheller, E.L.; MacDougald, O.A. Adipose tissue stem cells meet preadipocyte commitment: Going back to the future. J. Lipid Res. 2012, 53, 227–246. [Google Scholar] [CrossRef]
  18. Ntambi, J.M.; Young-Cheul, K. Adipocyte differentiation and gene expression. J. Nutr. 2000, 130, 3122S–3126S. [Google Scholar] [CrossRef]
  19. Ussar, S.; Lee, K.Y.; Dankel, S.N.; Boucher, J.; Haering, M.F.; Kleinridders, A.; Thomou, T.; Xue, R.; Macotela, Y.; Kahn, C.R. ASC-1, PAT2, and P2RX5 are cell surface markers for white, beige, and brown adipocytes. Sci. Transl. Med. 2014, 6, 247ra103. [Google Scholar] [CrossRef]
  20. Pilkington, A.C.; Paz, H.A.; Wankhade, U.D. Beige adipose tissue identification and marker specificity—Overview. Front. Endocrinol. 2021, 12, 599134. [Google Scholar] [CrossRef]
  21. Rosen, E.D.; Hsu, C.-H.; Wang, X.; Sakai, S.; Freeman, M.W.; Gonzalez, F.J.; Spiegelman, B.M. C/EBPalpha induces adipogenesis through PPARgamma: A unified pathway. Genes Dev. 2002, 16, 22–26. [Google Scholar] [CrossRef]
  22. Dubuquoy, L.; Dharancy, S.; Nutten, S.; Pettersson, S.; Auwerx, J.; Desreumaux, P. Role of peroxisome proliferator-activated receptor gamma and retinoid X receptor heterodimer in hepatogastroenterological diseases. Lancet 2002, 360, 1410–1418. [Google Scholar] [CrossRef]
  23. Lehmann, J.M.; Moore, L.B.; Smith-Oliver, T.A.; Wilkison, W.O.; Willson, T.M.; Kliewer, S.A. An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor γ (PPARγ). J. Biol. Chem. 1995, 270, 12953–12956. [Google Scholar] [CrossRef] [PubMed]
  24. MacDougald, O.A.; Cornelius, P.; Liu, R.; Lane, M.D. Insulin regulates transcription of the CCAAT/enhancer binding protein (C/EBP) alpha, beta, and delta genes in fully-differentiated 3T3-L1 adipocytes. J. Biol. Chem. 1995, 270, 647–654. [Google Scholar] [CrossRef] [PubMed]
  25. Wolins, N.E.; Quaynor, B.K.; Skinner, J.R.; Tzekov, A.; Park, C.; Choi, K.; Bickel, P.E. OP9 mouse stromal cells rapidly differentiate into adipocytes: Characterization of a useful new model of adipogenesis. J. Lipid Res. 2006, 47, 450–460. [Google Scholar] [CrossRef] [PubMed]
  26. Rossi, A.; Eid, M.; Dodgson, J.; Davies, G.; Musial, B.; Wabitsch, M.; Church, C.; Hornigold, D.C. In vitro characterization of the effects of chronic insulin stimulation in mouse 3T3-L1 and human SGBS adipocytes. Adipocyte 2020, 9, 415–426. [Google Scholar] [CrossRef]
  27. Kim, S.P.; Ha, J.M.; Yun, S.J.; Kim, E.K.; Chung, S.W.; Hong, K.W.; Kim, C.D.; Bae, S.S. Transcriptional activation of peroxisome proliferator-activated receptor-γ requires activation of both protein kinase A and Akt during adipocyte differentiation. Biochem. Biophys. Res. Commun. 2010, 399, 55–59. [Google Scholar] [CrossRef]
  28. Lee, H.-L.; Qadir, A.S.; Park, H.-J.; Chung, E.; Lee, Y.-S.; Woo, K.M.; Ryoo, H.-M.; Kim, H.J.; Baek, J.-H. cAMP/Protein Kinase A signaling inhibits Dlx5 expression via activation of CREB and subsequent C/EBPβ induction in 3T3-L1 preadipocytes. Int. J. Mol. Sci. 2018, 19, 3161. [Google Scholar] [CrossRef]
  29. Sai, S.; Esteves, C.L.; Kelly, V.; Michailidou, Z.; Anderson, K.; Coll, A.P.; Nakagawa, Y.; Ohzeki, T.; Seckl, J.R.; Chapman, K.E. Glucocorticoid regulation of the promoter of 11beta-hydroxysteroid dehydrogenase type 1 is indirect and requires CCAAT/enhancer-binding protein-beta. Mol. Endocrinol. 2008, 22, 2049–2060. [Google Scholar] [CrossRef]
  30. Ayala-Sumuano, J.-T.; Velez-delValle, C.; Beltrán-Langarica, A.; Marsch-Moreno, M.; Hernandez-Mosqueira, C.; Kuri-Harcuch, W. Glucocorticoid paradoxically recruits adipose progenitors and impairs lipid homeostasis and glucose transport in mature adipocytes. Sci. Rep. 2013, 3, 2573. [Google Scholar] [CrossRef]
  31. Zubiría, M.G.; Giordano, A.P.; Gambaro, S.E.; Alzamendi, A.; Frontini-López, Y.R.; Moreno, G.; Spinedi, E.; Giovambattista, A. Dexamethasone primes adipocyte precursor cells for differentiation by enhancing adipogenic competency. Life Sci. 2020, 261, 118363. [Google Scholar] [CrossRef]
  32. Yau, W.W.; Singh, B.K.; Lesmana, R.; Zhou, J.; Sinha, R.A.; Wong, K.A.; Wu, Y.; Bay, B.-H.; Sugii, S.; Sun, L.; et al. Thyroid hormone (T3) stimulates brown adipose tissue activation via mitochondrial biogenesis and MTOR-mediated mitophagy. Autophagy 2018, 15, 131–150. [Google Scholar] [CrossRef]
  33. Liu, S.; Shen, S.; Yan, Y.; Sun, C.; Lu, Z.; Feng, H.; Ma, Y.; Tang, Z.; Yu, J.; Wu, Y.; et al. Triiodothyronine (T3) promotes brown fat hyperplasia via thyroid hormone receptor α mediated adipocyte progenitor cell proliferation. Nat. Commun. 2022, 13, 3394. [Google Scholar] [CrossRef]
  34. Huang, L.H.; Guo, Z.F.; Huang, M.J.; Zeng, X.Y.; Huang, H.B. Triiodothyronine (T3) promotes browning of white adipose through inhibition of the PI3K/AKT signalling pathway. Sci. Rep. 2024, 14, 20370. [Google Scholar] [CrossRef] [PubMed]
  35. Lehmann, J.M.; Lenhard, J.M.; Oliver, B.B.; Ringold, G.M.; Kliewer, S.A. Peroxisome proliferator-activated receptors alpha and gamma are activated by indomethacin and other non-steroidal anti-inflammatory drugs. J. Biol. Chem. 1997, 272, 3406–3410. [Google Scholar] [CrossRef] [PubMed]
  36. Overby, H.; Yang, Y.; Xu, X.; Wang, S.; Zhao, L. Indomethacin promotes browning and brown adipogenesis in both murine and human fat cells. Pharmacol. Res. Perspect. 2020, 8, e00592. [Google Scholar] [CrossRef] [PubMed]
  37. Andrews, F.V.; Kim, S.M.; Edwards, L.; Schlezinger, J.J. Identifying adipogenic chemicals: Disparate effects in 3T3-L1, OP9 and primary mesenchymal multipotent cell models. Toxicol. Vitr. 2020, 67, 104904. [Google Scholar] [CrossRef]
  38. Morrison, S.; McGee, S.L. 3T3-L1 adipocytes display phenotypic characteristics of multiple adipocyte lineages. Adipocyte 2015, 4, 295–302. [Google Scholar] [CrossRef]
  39. Kraus, N.A.; Ehebauer, F.; Zapp, B.; Rudolphi, B.; Kraus, B.J.; Kraus, D. Quantitative assessment of adipocyte differentiation in cell culture. Adipocyte 2016, 5, 351–358. [Google Scholar] [CrossRef]
  40. Zhao, X.; Hu, H.; Wang, C.; Bai, L.; Wang, Y.; Wang, W.; Wang, J. A comparison of methods for effective differentiation of the frozen-thawed 3T3-L1 cells. Anal. Biochem. 2019, 568, 57–64. [Google Scholar] [CrossRef]
  41. Kassotis, C.D.; Hoffman, K.; Völker, J.; Pu, Y.; Veiga-Lopez, A.; Kim, S.M.; Stapleton, H.M. Reproducibility of adipogenic responses to metabolism disrupting chemicals in the 3T3-L1 pre-adipocyte model system: An interlaboratory study. Toxicology 2021, 461, 152900. [Google Scholar] [CrossRef]
  42. Pu, Y.; Veiga-Lopez, A. PPARγ agonist through the terminal differentiation phase is essential for adipogenic differentiation of fetal ovine preadipocytes. Cell Biol. Int. 2017, 41, 1231–1239. [Google Scholar] [CrossRef] [PubMed]
  43. Ukita, M.; Yamaguchi, T.; Ohata, N.; Tamura, M. Sclerostin enhances adipocyte differentiation in 3T3-L1 cells. J. Cell. Biochem. 2016, 117, 1419–1428. [Google Scholar] [CrossRef]
  44. Josan, C.; Kakar, S.; Raha, S. Matrigel® enhances 3T3-L1 cell differentiation. Adipocyte 2021, 10, 361–377. [Google Scholar] [CrossRef] [PubMed]
  45. Miller, C.N.; Yang, J.Y.; England, E.; Yin, A.; Baile, C.A.; Rayalam, S. Isoproterenol increases uncoupling, glycolysis, and markers of beiging in mature 3T3-L1 adipocytes. PLoS ONE 2015, 10, e0138344. [Google Scholar] [CrossRef]
  46. Luz, A.L.; Kassotis, C.D.; Stapleton, H.M.; Meyer, J.N. The high-production volume fungicide pyraclostrobin induces triglyceride accumulation associated with mitochondrial dysfunction, and promotes adipocyte differentiation independent of PPARγ activation, in 3T3-L1 cells. Toxicology 2018, 393, 150–159. [Google Scholar] [CrossRef]
  47. Teixeira, C.; Sousa, A.P.; Santos, I.; Rocha, A.C.; Alencastre, I.; Pereira, A.C.; Fernandes, R. Enhanced 3T3-L1 differentiation into adipocytes by pioglitazone pharmacological activation of peroxisome proliferator activated receptor-gamma (PPAR-γ). Biology 2022, 11, 806. [Google Scholar] [CrossRef]
  48. Li, Y.; Rong, Y.; Bao, L.; Nie, B.; Ren, G.; Zheng, C.; Huggins, K.W. Suppression of adipocyte differentiation and lipid accumulation by stearidonic acid (SDA) in 3T3-L1 cells. Lipids Health Dis. 2017, 16, 181. [Google Scholar] [CrossRef]
  49. Martini, C.N.; Gabrielli, M.; Bonifacino, G.; Codesido, M.M.; Vila, M.D.C. Lead enhancement of 3T3-L1 fibroblasts differentiation to adipocytes involves ERK, C/EBPβ and PPARγ activation. Mol. Cell. Biochem. 2018, 437, 37–44. [Google Scholar] [CrossRef]
  50. Bahrami-Nejad, Z.; Zhao, M.L.; Tholen, S.; Hunerdosse, D.; Tkach, K.E.; van Schie, S.; Teruel, M.N. A transcriptional circuit filters oscillating circadian hormonal inputs to regulate fat cell differentiation. Cell Metab. 2018, 27, 854–868. [Google Scholar] [CrossRef]
  51. Zhang, S.; You, Y.; Li, Y.; Yuan, H.; Zhou, J.; Tian, L.; Zhu, E. Foxk1 stimulates adipogenic differentiation via a peroxisome proliferator-activated receptor gamma 2-dependent mechanism. FASEB J. 2023, 37, e23266. [Google Scholar] [CrossRef]
  52. Torabi, S.; Mo, H. Trans, trans-farnesol as a mevalonate-derived inducer of murine 3T3-F442A pre-adipocyte differentiation. Exp. Biol. Med. 2016, 241, 493–500. [Google Scholar] [CrossRef]
  53. Chakrabarti, S.; Wu, J. Milk-derived tripeptides IPP (Ile-Pro-Pro) and VPP (Val-Pro-Pro) promote adipocyte differentiation and inhibit inflammation in 3T3-F442A cells. PLoS ONE 2015, 10, e0117492. [Google Scholar] [CrossRef]
  54. Khalilpourfarshbafi, M.; Devi Murugan, D.; Abdul Sattar, M.Z.; Sucedaram, Y.; Abdullah, N.A. Withaferin A inhibits adipogenesis in 3T3-F442A cell line, improves insulin sensitivity and promotes weight loss in high fat diet-induced obese mice. PLoS ONE 2019, 14, e0218792. [Google Scholar] [CrossRef]
  55. Jahandideh, F.; Chakrabarti, S.; Davidge, S.T.; Wu, J. Egg white hydrolysate shows insulin mimetic and sensitizing effects in 3T3-F442A pre-adipocytes. PLoS ONE 2017, 12, e0185653. [Google Scholar] [CrossRef]
  56. Doğan, A.; Demirci, S.; Kıratlı, B.; Şahin, F. Cytoglobin: A potential marker for adipogenic differentiation in preadipocytes in vitro. Cytotechnology 2017, 69, 157–165. [Google Scholar] [CrossRef] [PubMed]
  57. Zhang, Y.; Yu, H.; Gao, P.; Chen, J.; Yu, C.; Zong, C.; Wang, X. The effect of growth hormone on lipid accumulation or maturation in adipocytes. Cell. Physiol. Biochem. 2016, 39, 2135–2148. [Google Scholar] [CrossRef] [PubMed]
  58. Zhu, S.; Wang, W.; Zhang, J.; Ji, S.; Jing, Z.; Chen, Y.Q. Slc25a5 regulates adipogenesis by modulating ERK signaling in OP9 cells. Cell. Mol. Biol. Lett. 2022, 27, 11. [Google Scholar] [CrossRef] [PubMed]
  59. Seo, Y.-S.; Kang, O.-H.; Kim, S.-B.; Mun, S.-H.; Kang, D.-H.; Yang, D.-W.; Kwon, D.-Y. Quercetin prevents adipogenesis by regulation of transcriptional factors and lipases in OP9 cells. Int. J. Mol. Med. 2015, 35, 1779–1785. [Google Scholar] [CrossRef]
  60. Campos, V.; Rappaz, B.; Kuttler, F.; Turcatti, G.; Naveiras, O. High-throughput, nonperturbing quantification of lipid droplets with digital holographic microscopy. J. Lipid Res. 2018, 59, 1301–1310. [Google Scholar] [CrossRef]
  61. Zhang, J.; Wang, W.; Feng, N.; Jiang, X.; Zhu, S.; Chen, Y.Q. Ndufa6 regulates adipogenic differentiation via Scd1. Adipocyte 2021, 10, 646–657. [Google Scholar] [CrossRef]
  62. Taylor, B.; Shah, A.; Bielczyk-Maczyńska, E. TGF-β is insufficient to induce adipocyte state loss without concurrent PPARγ downregulation. Sci. Rep. 2020, 10, 14084. [Google Scholar] [CrossRef] [PubMed]
  63. Kato, S.; Kato, Y.; Shibata, H.; Saitoh, Y.; Miwa, N. Repressive effects of oat extracts on intracellular lipid-droplet formation in adipocytes and a three-dimensional subcutaneous adipose tissue model. Mater. Sci. Eng. C Mater. Biol. Appl. 2015, 49, 269–273. [Google Scholar] [CrossRef] [PubMed]
  64. Thunen, A.; La Placa, D.; Zhang, Z.; Shively, J.E. Role of lncRNA LIPE-AS1 in adipogenesis. Adipocyte 2022, 11, 11–27. [Google Scholar] [CrossRef] [PubMed]
  65. Baskan, O.; Mese, G.; Ozcivici, E. Low-intensity vibrations normalize adipogenesis-induced morphological and molecular changes of adult mesenchymal stem cells. Proc. Inst. Mech. Eng. Part H J. Eng. Med. 2017, 231, 160–168. [Google Scholar] [CrossRef]
  66. Sarigil, O.; Anil-Inevi, M.; Yilmaz, E.; Mese, G.; Tekin, H.C.; Ozcivici, E. Label-free density-based detection of adipocytes of bone marrow origin using magnetic levitation. Analyst 2019, 144, 2942–2953. [Google Scholar] [CrossRef]
  67. Larsen, M.C.; Almeldin, A.; Tong, T.; Rondelli, C.M.; Maguire, M.; Jaskula-Sztul, R.; Jefcoate, C.R. Cytochrome P4501B1 in bone marrow is co-expressed with key markers of mesenchymal stem cells. BMS2 cell line models PAH disruption of bone marrow niche development functions. Toxicol. Appl. Pharmacol. 2020, 401, 115111. [Google Scholar] [CrossRef]
  68. Kelly, K.A.; Gimble, J.M. 1, 25-Dihydroxy vitamin D3 inhibits adipocyte differentiation and gene expression in murine bone marrow stromal cell clones and primary cultures. Endocrinology 1998, 139, 2622–2628. [Google Scholar] [CrossRef]
  69. Dace, A.; Martin-El Yazidi, C.; Bonne, J.; Planells, R.; Torresani, J. Calcitriol is a positive effector of adipose differentiation in the OB 17 cell line: Relationship with the adipogenic action of triiodothyronine. Biochem. Biophys. Res. Commun. 1997, 232, 771–776. [Google Scholar] [CrossRef]
  70. Amri, E.-Z.; Grimaldi, P.; Négrel, R.; Ailhaud, G. Adipose conversion of ob17 cells: Insulin acts solely as a modulator in the expression of the differentiation program. Exp. Cell Res. 1984, 152, 368–377. [Google Scholar] [CrossRef]
  71. Grimaldi, P.; Djian, P.; Poli, P.; Negrel, R.; Ailhaud, G. Lipogenic and mitogenic effects of insulin during conversion of Ob17 cells to adipose-like cells. Mol. Cell. Endocrinol. 1983, 29, 271–285. [Google Scholar] [CrossRef]
  72. Djian, P.; Grimaldi, P.; Négrel, R.; Ailhaud, G. Adipose conversion of Ob17 preadipocytes: Relationships between cell division and fat cell cluster formation. Exp. Cell Res. 1982, 142, 273–281. [Google Scholar] [CrossRef]
  73. Yokota, T.; Meka, C.S.R.; Medina, K.L.; Igarashi, H.; Comp, P.C.; Takahashi, M.; Nishida, M.; Oritani, K.; Miyagawa, J.; Funahashi, T.; et al. Paracrine Regulation of Fat Cell Formation in Bone Marrow Cultures via Adiponectin and Prostaglandins. J. Clin. Investig. 2002, 109, 1303–1310. [Google Scholar] [CrossRef] [PubMed]
  74. Schelker, R.C.; Iberl, S.; Müller, G.; Hart, C.; Herr, W.; Grassinger, J. TGF-β1 and CXCL12 Modulate Proliferation and Chemotherapy Sensitivity of Acute Myeloid Leukemia Cells Co-Cultured with Multipotent Mesenchymal Stromal Cells. Leukemia 2017, 31, 337–345. [Google Scholar] [CrossRef] [PubMed]
  75. Schwind, L.; Schetting, S.; Montenarh, M. Inhibition of protein kinase CK2 prevents adipogenic differentiation of mesenchymal stem cells like C3H/10T1/2 cells. Pharmaceuticals 2017, 10, 22. [Google Scholar] [CrossRef] [PubMed]
  76. Cricrí, D.; Coppi, L.; Pedretti, S.; Mitro, N.; Caruso, D.; De Fabiani, E.; Crestani, M. Histone deacetylase 3 regulates adipocyte phenotype at early stages of differentiation. Int. J. Mol. Sci. 2021, 22, 9300. [Google Scholar] [CrossRef]
  77. Alashi, A.M.; Blanchard, C.L.; Mailer, R.J.; Agboola, S.O.; Mawson, A.J.; Aluko, R.E.; Strappe, P. Effects of canola proteins and hydrolysates on adipogenic differentiation of C3H10T/2 mesenchymal stem cells. Food Chem. 2015, 185, 226–232. [Google Scholar] [CrossRef]
  78. Dimitriadis, G.K.; Adya, R.; Tan, B.K.; Jones, T.A.; Menon, V.S.; Ramanjaneya, M.; Randeva, H.S. Effects of visfatin on brown adipose tissue energy regulation using T37i cells. Cytokine 2019, 113, 248–255. [Google Scholar] [CrossRef]
  79. Yun, Y.R.; Lee, J.E.; Lee, S.; Hong, S.W. Exploring the anti-obesity effects of kimchi through enhanced thermogenesis in differentiated T37i brown adipocytes. Food Nutr. Res. 2024, 68, 10–29219. [Google Scholar] [CrossRef]
  80. Marzolla, V.; Feraco, A.; Gorini, S.; Mammi, C.; Marrese, C.; Mularoni, V.; Caprio, M. The novel non-steroidal MR antagonist finerenone improves metabolic parameters in high-fat diet-fed mice and activates brown adipose tissue via AMPK-ATGL pathway. FASEB J. 2020, 34, 12450–12465. [Google Scholar] [CrossRef]
  81. Pham, H.G.; Mukherjee, S.; Choi, M.J.; Yun, J.W. BMP11 regulates thermogenesis in white and brown adipocytes. Cell Biochem. Funct. 2021, 39, 496–510. [Google Scholar] [CrossRef]
  82. Abdul Majeed, S.; Dunzendorfer, H.; Weiner, J.; Heiker, J.T.; Kiess, W.; Körner, A.; Landgraf, K. COBL, MKX and MYOC are potential regulators of brown adipose tissue development associated with obesity-related metabolic dysfunction in children. Int. J. Mol. Sci. 2023, 24, 3085. [Google Scholar] [CrossRef]
  83. Zovich, D.C.; Orologa, A.; Okuno, M.; Kong, L.W.; Talmage, D.A.; Piantedosi, R.; Blaner, W.S. Differentiation-dependent expression of retinoid-binding proteins in BFC-1 beta adipocytes. J. Biol. Chem. 1992, 267, 13884–13889. [Google Scholar] [CrossRef] [PubMed]
  84. Sun, Y.; Li, X.; Yang, X.; Chen, B.; Zhang, W. Small extracellular vesicles derived from adipocytes attenuate intervertebral disc degeneration in rats by rejuvenating senescent nucleus pulposus cells and endplate cells by delivering exogenous NAMPT. Oxid. Med. Cell. Longev. 2021, 2021, 9955448. [Google Scholar] [CrossRef] [PubMed]
  85. Raffaele, M.; Barbagallo, I.; Licari, M.; Carota, G.; Sferrazzo, G.; Spampinato, M.; Sorrenti, V.; Vanella, L. N-Acetylcysteine (NAC) ameliorates lipid-related metabolic dysfunction in bone marrow stromal cells-derived adipocytes. Evid.-Based Complement. Altern. Med. 2018, 2018, 5310961. [Google Scholar] [CrossRef] [PubMed]
  86. Chen, M.; Dong, Y.; Tian, L.; Zhou, J.; Zhu, E.; Yuan, H.; Wang, B. Metastasis suppressor 1 interacts with protein tyrosine phosphatase receptor-δ to regulate adipogenesis. FASEB J. 2023, 37, e22857. [Google Scholar] [CrossRef]
  87. Wabitsch, M.; Brüderlein, S.; Melzner, I.; Braun, M.; Mechtersheimer, G.; Möller, P. LiSa-2, a novel human liposarcoma cell line with a high capacity for terminal adipose differentiation. Int. J. Cancer 2000, 88, 889–894. [Google Scholar] [CrossRef]
  88. Van Beek, E.A.; Bakker, A.H.; Kruyt, P.M.; Vink, C.; Saris, W.H.; Franssen-van Hal, N.L.W.; Keijer, J. Comparative expression analysis of isolated human adipocytes and the human adipose cell lines LiSa-2 and PAZ6. Int. J. Obes. 2008, 32, 912–921. [Google Scholar] [CrossRef]
  89. Monastra, G.; Gambioli, R.; Unfer, V.; Forte, G.; Maymo-Masip, E.; Comitato, R. D-Chiro-Inositol and Myo-Inositol Induce WAT/BAT Trans-Differentiation in Two Different Human Adipocyte Models (SGBS and LiSa-2). Int. J. Mol. Sci. 2023, 24, 7421. [Google Scholar] [CrossRef]
  90. Rosenow, A.; Arrey, T.N.; Bouwman, F.G.; Noben, J.P.; Wabitsch, M.; Mariman, E.C.; Renes, J. Identification of novel human adipocyte secreted proteins by using SGBS cells. J. Proteome Res. 2010, 9, 5389–5401. [Google Scholar] [CrossRef]
  91. Takahashi, A.; Koike, R.; Watanabe, S.; Kuribayashi, K.; Wabitsch, M.; Miyamoto, M.; Iwata, T. Polypeptide N-acetylgalactosaminyltransferase-15 regulates adipogenesis in human SGBS cells. Sci. Rep. 2024, 14, 20049. [Google Scholar] [CrossRef]
  92. Klusóczki, Á.; Veréb, Z.; Vámos, A.; Fischer-Posovszky, P.; Wabitsch, M.; Bacso, Z.; Kristóf, E. Differentiating SGBS adipocytes respond to PPARγ stimulation, irisin and BMP7 by functional browning and beige characteristics. Sci. Rep. 2019, 9, 5823. [Google Scholar] [CrossRef]
  93. Hugo, E.R.; Brandebourg, T.D.; Comstock, C.E.; Gersin, K.S.; Sussman, J.J.; Ben-Jonathan, N. LS14: A novel human adipocyte cell line that produces prolactin. Endocrinology 2006, 147, 306–313. [Google Scholar] [CrossRef]
  94. Hugo, E.R.; Borcherding, D.C.; Gersin, K.S.; Loftus, J.; Ben-Jonathan, N. Prolactin release by adipose explants, primary adipocytes, and LS14 adipocytes. J. Clin. Endocrinol. Metab. 2008, 93, 4006–4012. [Google Scholar] [CrossRef]
  95. Masnikov, D.; Stafeev, I.; Michurina, S.; Zubkova, E.; Mamontova, E.; Ratner, E.; Menshikov, M.; Parfyonova, Y. hTERT-immortalized adipose-derived stem cell line ASC52Telo demonstrates limited potential for adipose biology research. Anal. Biochem. 2021, 628, 114268. [Google Scholar] [CrossRef]
  96. Primak, A.; Kalinina, N.; Skryabina, M.; Usachev, V.; Chechekhin, V.; Vigovskiy, M.; Chechekhina, E.; Voloshin, N.; Kulebyakin, K.; Kulebyakina, M. Novel Immortalized Human Multipotent Mesenchymal Stromal Cell Line for Studying Hormonal Signaling. Int. J. Mol. Sci. 2024, 25, 2421. [Google Scholar] [CrossRef] [PubMed]
  97. Strobel, A.; Siquier, K.; Zilberfarb, V.; Strosberg, A.D.; Issad, T. Effect of thiazolidinediones on expression of UCP2 and adipocyte markers in human PAZ6 adipocytes. Diabetologia 1999, 42, 527–533. [Google Scholar] [CrossRef] [PubMed]
  98. Zilberfarb, V.; Siquier, K.; Strosberg, A.D.; Issad, T. Effect of dexamethasone on adipocyte differentiation markers and tumour necrosis factor-α expression in human PAZ6 cells. Diabetologia 2001, 44, 377–386. [Google Scholar] [CrossRef] [PubMed]
  99. Jockers, R.; Issad, T.; Zilberfarb, V.; De Coppet, P.; Marullo, S.; Strosberg, A.D. Desensitization of the β-adrenergic response in human brown adipocytes. Endocrinology 1998, 139, 2676–2684. [Google Scholar] [CrossRef]
  100. Brydon, L.; Petit, L.; Delagrange, P.; Strosberg, A.D.; Jockers, R. Functional expression of MT2 (Mel1b) melatonin receptors in human PAZ6 adipocytes. Endocrinology 2001, 142, 4264–4271. [Google Scholar] [CrossRef]
  101. Bai, X.; Zhu, Q.; Combs, M.; Wabitsch, M.; Mack, C.P.; Taylor, J.M. GRAF1 deficiency leads to defective brown adipose tissue differentiation and thermogenic response. Sci. Rep. 2024, 14, 28692. [Google Scholar] [CrossRef]
  102. Murholm, M.; Dixen, K.; Hansen, J.B. Ras signalling regulates differentiation and UCP1 expression in models of brown adipogenesis. Biochim. Biophys. Acta Gen. Subj. 2010, 1800, 617–627. [Google Scholar] [CrossRef] [PubMed]
  103. Ren, J.; Wang, H.; Tran, K.; Civini, S.; Jin, P.; Castiello, L.; Feng, J.; Kuznetsov, S.A.; Robey, P.G.; Sabatino, M.; et al. Human Bone Marrow Stromal Cell Confluence: Effects on Cell Characteristics and Methods of Assessment. Cytotherapy 2015, 17, 897–911. [Google Scholar] [CrossRef] [PubMed]
  104. Abo-Aziza, F.A.M.; Zaki, A.A. The Impact of Confluence on Bone Marrow Mesenchymal Stem (BMMSC) Proliferation and Osteogenic Differentiation. Int. J. Hematol. Oncol. Stem Cell Res. 2017, 11, 121–132. [Google Scholar] [PubMed]
  105. Mehra, A.; Macdonald, I.; Pillay, T.S. Variability in 3T3-L1 adipocyte differentiation depending on cell culture dish. Anal. Biochem. 2007, 362, 281–283. [Google Scholar] [CrossRef]
  106. Marcon, B.H.; Shigunov, P.; Spangenberg, L.; Pereira, I.T.; de Aguiar, A.M.; Amorín, R.; Rebelatto, C.K.; Correa, A.; Dallagiovanna, B. Cell cycle genes are downregulated after adipogenic triggering in human adipose tissue-derived stem cells by regulation of mRNA abundance. Sci. Rep. 2019, 9, 5611. [Google Scholar]
  107. Lefterova, M.I.; Haakonsson, A.K.; Lazar, M.A.; Mandrup, S. PPARγ and the global map of adipogenesis and beyond. Trends Endocrinol. Metab. 2014, 25, 293–302. [Google Scholar] [CrossRef]
  108. Tontonoz, P.; Hu, E.; Spiegelman, B.M. Stimulation of adipogenesis in fibroblasts by PPARγ2, a lipid-activated transcription factor. Cell 1994, 79, 1147–1156. [Google Scholar] [CrossRef]
  109. Rosen, E.D.; Sarraf, P.; Troy, A.E.; Bradwin, G.; Moore, K.; Milstone, D.S.; Spiegelman, B.M.; Mortensen, R.M. PPARγ is required for the differentiation of adipose tissue in vivo and in vitro. Mol. Cell 1999, 4, 611–617. [Google Scholar] [CrossRef]
  110. Zhu, J.; Guérineau, H.; Lefebvre-Fortané, A.-M.; Largeaud, L.; Lambert, J.; Rousselot, P.; Boudouin, M.; Calvo, J.; Prost, S.; Clauser, S.; et al. The AXL inhibitor bemcentinib overcomes microenvironment-mediated resistance to pioglitazone in acute myeloid leukemia. FEBS J. 2025, 292, 115–128. [Google Scholar] [CrossRef]
  111. Nakashima, K.-I.; Okamura, M.; Matsumoto, I.; Kameda, N.; Tsuboi, T.; Yamaguchi, E.; Itoh, A.; Inoue, M. Regulation of adipogenesis through retinoid X receptor and/or peroxisome proliferator-activated receptor by designed lignans based on natural products in 3T3-L1 cells. J. Nat. Med. 2023, 77, 315–326. [Google Scholar]
  112. Bauerle, K.T.; Hutson, I.; Scheller, E.L.; Harris, C.A. Glucocorticoid Receptor Signaling Is Not Required for In Vivo Adipogenesis. Endocrinology 2018, 159, 2050–2061. [Google Scholar] [CrossRef]
  113. Liu, H.; Wei, N.; Joshi, V.; Yu, Y.; Kim, N.; Krishnamachari, Y.; Zhang, Q.; Salem, A.K. Effects of Glucocorticoid Receptor Small Interfering RNA Delivered Using Poly Lactic-Co-Glycolic Acid Microparticles on Proliferation and Differentiation Capabilities of Human Mesenchymal Stromal Cells. Tissue Eng. Part A 2012, 18, 775–784. [Google Scholar]
  114. Vernocchi, S.; Battello, N.; Schmitz, S.; Revets, D.; Billing, A.M.; Turner, J.D.; Muller, C.P. Membrane Glucocorticoid Receptor Activation Induces Proteomic Changes Aligning with Classical Glucocorticoid Effects. Mol. Cell. Proteom. 2013, 12, 1764–1779. [Google Scholar] [CrossRef] [PubMed]
  115. Gametchu, B.; Watson, C.S.; Wu, S. Use of Receptor Antibodies to Demonstrate Membrane Glucocorticoid Receptor in Cells from Human Leukemic Patients. FASEB J. 1993, 7, 1283–1292. [Google Scholar] [CrossRef] [PubMed]
  116. Lewis-Tuffin, L.J.; Jewell, C.M.; Bienstock, R.J.; Collins, J.B.; Cidlowski, J.A. Human glucocorticoid receptor beta binds RU-486 and is transcriptionally active. Mol. Cell. Biol. 2007, 27, 2266–2282. [Google Scholar] [CrossRef]
  117. Han, L.; Wang, B.; Wang, R.; Gong, S.; Chen, G.; Xu, W. The shift in the balance between osteoblastogenesis and adipogenesis of mesenchymal stem cells mediated by glucocorticoid receptor. Stem Cell Res. Ther. 2019, 10, 377. [Google Scholar] [CrossRef]
  118. Obregon, M.-J. Thyroid hormone and adipocyte differentiation. Thyroid 2008, 18, 185–195. [Google Scholar] [CrossRef]
  119. Mishra, A.; Zhu, X.-G.; Ge, K.; Cheng, S.-Y. Adipogenesis is differentially impaired by thyroid hormone receptor mutant isoforms. J. Mol. Endocrinol. 2010, 44, 247–255. [Google Scholar] [CrossRef]
  120. Roth, L.; Johann, K.; Hönes, G.S.; Oelkrug, R.; Wagner, L.; Hoffmann, A.; Krohn, K.; Moeller, L.C.; Weiner, J.; Heiker, J.T.; et al. Thyroid hormones regulate Zfp423 expression in regionally distinct adipose depots through direct and cell-autonomous action. Cell Rep. 2023, 42, 112088. [Google Scholar] [CrossRef]
  121. Ma, Y.; Shen, S.; Yan, Y.; Zhang, S.; Liu, S.; Tang, Z.; Yu, J.; Ma, M.; Niu, Z.; Li, Z.; et al. Adipocyte Thyroid Hormone β Receptor-Mediated Hormone Action Fine-tunes Intracellular Glucose and Lipid Metabolism and Systemic Homeostasis. Diabetes 2023, 72, 562–574. [Google Scholar] [CrossRef]
  122. Lin, J.Z.; Martagón, A.J.; Cimini, S.L.; Gonzalez, D.D.; Tinkey, D.W.; Biter, A.; Baxter, J.D.; Webb, P.; Gustafsson, J.-Å.; Hartig, S.M.; et al. Pharmacological Activation of Thyroid Hormone Receptors Elicits a Functional Conversion of White to Brown Fat. Cell Rep. 2015, 13, 1528–1537. [Google Scholar] [CrossRef] [PubMed]
  123. Evans, B.A.; Merlin, J.; Bengtsson, T.; Hutchinson, D.S. Adrenoceptors in white, brown, and brite adipocytes. Br. J. Pharmacol. 2019, 176, 2416–2432. [Google Scholar] [CrossRef] [PubMed]
  124. Guest, S.J.; Hadcock, J.R.; Watkins, D.C.; Malbon, C.C. Beta 1- and beta 2-adrenergic receptor expression in differentiating 3T3-L1 cells. Independent regulation at the level of mRNA. J. Biol. Chem. 1990, 265, 5370–5375. [Google Scholar] [CrossRef] [PubMed]
  125. Roshanzadeh, A.; Yadav, A.K.; Pydi, S.P.; Kimura, T.; Jang, B.C. Expression and role of β3-adrenergic receptor during the differentiation of 3T3-L1 preadipocytes into adipocytes. Biology 2022, 11, 772. [Google Scholar] [CrossRef]
  126. Tyurin-Kuzmin, P.A.; Chechekhin, V.I.; Ivanova, A.M.; Dyikanov, D.T.; Sysoeva, V.Y.; Kalinina, N.I.; Tkachuk, V.A. Noradrenaline sensitivity is severely impaired in immortalized adipose-derived mesenchymal stem cell line. Int. J. Mol. Sci. 2018, 19, 3712. [Google Scholar] [CrossRef]
  127. Khadija, E.; Pairault, J.; Fève, B. Triiodothyronine regulates β3-adrenoceptor expression in 3T3-F442A differentiating adipocytes. Eur. J. Biochem. 1996, 239, 519–525. [Google Scholar] [CrossRef]
  128. Bakopanos, E.; Silva, J.E. Opposing effects of glucocorticoids on β3-adrenergic receptor expression in HIB-1B brown adipocytes. Mol. Cell. Endocrinol. 2002, 190, 29–37. [Google Scholar] [CrossRef]
  129. Agueda-Oyarzabal, M.; Isidor, M.S.; Plucińska, K.; Ingerslev, L.R.; Dmytriyeva, O.; Petersen, P.S.; Laftih, S.; Pontoppidan, A.B.; Henningsen, J.B.; Rupar, K.; et al. Transcriptomic signatures of cold acclimated adipocytes reveal CXCL12 as a Brown autocrine and paracrine chemokine. Mol. Metab. 2025, 93, 102102. [Google Scholar] [CrossRef]
  130. Kassotis, C.D.; Masse, L.; Kim, S.; Schlezinger, J.J.; Webster, T.F.; Stapleton, H.M. Characterization of adipogenic chemicals in three different cell culture systems: Implications for reproducibility based on cell source and handling. Sci. Rep. 2017, 7, 42104. [Google Scholar] [CrossRef]
  131. Wang, H.; Zhang, Y.; Yehuda-Shnaidman, E.; Medvedev, A.V.; Kumar, N.; Daniel, K.W.; Robidoux, J.; Czech, M.P.; Mangelsdorf, D.J.; Collins, S. Liver X receptor α is a transcriptional repressor of the uncoupling protein 1 gene and the brown fat phenotype. Mol. Cell. Biol. 2008, 28, 2187–2200. [Google Scholar] [CrossRef]
  132. Laurencikiene, J.; Rydén, M. Liver X receptors and fat cell metabolism. Int. J. Obes. 2012, 36, 1494–1502. [Google Scholar] [CrossRef]
  133. Gharibi, B.; Abraham, A.A.; Ham, J.; Evans, B.A. Adenosine receptor subtype expression and activation influence the differentiation of mesenchymal stem cells to osteoblasts and adipocytes. J. Bone Min. Res. 2011, 26, 2112–2124. [Google Scholar] [CrossRef]
  134. Gharibi, B.; Abraham, A.A.; Ham, J.; Evans, B.A.J. Contrasting effects of A1 and A2b adenosine receptors on adipogenesis. Int. J. Obes. 2012, 36, 397–406. [Google Scholar] [CrossRef]
  135. Fuggetta, M.P.; Zonfrillo, M.; Villivà, C.; Bonmassar, E.; Ravagnan, G. Inflammatory microenvironment and adipogenic differentiation in obe-1238 sity: The inhibitory effect of theobromine in a model of human obesity in vitro. Mediat. Inflamm. 2019, 2019, 1515621. [Google Scholar] [CrossRef]
  136. Eisenstein, A.; Ravid, K. G protein-coupled receptors and adipogenesis: A focus on adenosine receptors. J. Cell Physiol. 2014, 229, 414–421. [Google Scholar] [CrossRef]
  137. Tatsis-Kotsidis, I.; Erlanger, B.F. Initiation of a process of differentiation by stable transfection of ob17 preadipocytes with the cDNA of human A1 adenosine receptor. Biochem. Pharmacol. 1999, 58, 167–170. [Google Scholar] [CrossRef] [PubMed]
  138. Yarwood, S.J. The Cyclic AMP Signalling System as a Regulator of Preadipocyte Differentiation; University of Glasgow: Glasgow, UK, 1997. [Google Scholar]
  139. Bitirim, C.V.; Ozer, Z.B.; Akcali, K.C. Estrogen receptor alpha regulates the expression of adipogenic genes genetically and epigenetically in rat bone marrow-derived mesenchymal stem cells. PeerJ 2021, 9, e12071. [Google Scholar] [CrossRef] [PubMed]
  140. Zhao, J.W.; Gao, Z.L.; Mei, H.; Li, Y.L.; Wang, Y. Differentiation of human mesenchymal stem cells: The potential mechanism for estrogen-induced preferential osteoblast versus adipocyte differentiation. Am. J. Med. Sci. 2011, 341, 460–468. [Google Scholar] [CrossRef] [PubMed]
  141. Al-Ghadban, S.; Isern, S.U.; Herbst, K.L.; Bunnell, B.A. The expression of adipogenic marker is significantly increased in estrogen-treated lipedema adipocytes differentiated from adipose stem cells in vitro. Biomedicines 2024, 12, 1042. [Google Scholar] [CrossRef]
  142. Zhou, S.; Turgeman, G.; Harris, S.E.; Leitman, D.C.; Komm, B.S.; Bodine, P.V.; Gazit, D. Estrogens activate bone morphogenetic protein-2 gene transcription in mouse mesenchymal stem cells. Mol. Endocrinol. 2003, 17, 56–66. [Google Scholar] [CrossRef]
  143. Tang, Q.Q.; Otto, T.C.; Lane, M.D. Commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineage. Proc. Natl. Acad. Sci. USA 2004, 101, 9607–9611. [Google Scholar] [CrossRef]
  144. Ahrens, M.; Ankenbauer, T.; Schröder, D.; Hollnagel, A.; Mayer, H.; Gross, G. Expression of human bone morphogenetic proteins-2 or -4 in murine mesenchymal progenitor C3H10T½ cells induces differentiation into distinct mesenchymal cell lineages. DNA Cell Biol. 1993, 12, 871–880. [Google Scholar] [CrossRef]
  145. Huang, J.; Woods, P.; Normolle, D.; Goff, J.P.; Benos, P.V.; Stehle, C.J.; Steinman, R.A. Downregulation of estrogen receptor and modulation of growth of breast cancer cell lines mediated by paracrine stromal cell signals. Breast Cancer Res. Treat. 2017, 161, 229–243. [Google Scholar] [CrossRef] [PubMed]
  146. Ruedl, C.; Cappelletti, V.; Coradini, D.; Granata, G.; Di Fronzo, G. Influence of culture conditions on the estrogenic cell growth stimulation of human breast cancer cells. J. Steroid Biochem. Mol. Biol. 1990, 37, 195–200. [Google Scholar] [CrossRef] [PubMed]
  147. Berthois, Y.; Katzenellenbogen, J.A.; Katzenellenbogen, B.S. Phenol red in tissue culture media is a weak estrogen: Implications concerning the study of estrogen-responsive cells in culture. Proc. Natl. Acad. Sci. USA 1986, 83, 2496–2500. [Google Scholar] [CrossRef] [PubMed]
  148. Tews, D.; Brenner, R.E.; Siebert, R.; Debatin, K.M.; Fischer-Posovszky, P.; Wabitsch, M. 20 Years with SGBS cells—A versatile in vitro model of human adipocyte biology. Int. J. Obes. 2022, 46, 1939–1947. [Google Scholar] [CrossRef]
  149. Do, M.S.; Jeong, H.S.; Choi, B.H.; Hunter, L.; Langley, S.; Pazmany, L.; Trayhurn, P. Inflammatory gene expression patterns revealed by DNA microarray analysis in TNF-α-treated SGBS human adipocytes. Yonsei Med. J. 2006, 47, 729–736. [Google Scholar] [CrossRef]
  150. Schaedlich, K.; Beier, L.S.; Kolbe, J.; Wabitsch, M.; Ernst, J. Pro-inflammatory effects of DEHP in SGBS-derived adipocytes and THP-1 macrophages. Sci. Rep. 2021, 11, 7928. [Google Scholar] [CrossRef]
  151. Lee, J.Y.; Takahashi, N.; Yasubuchi, M.; Kim, Y.I.; Hashizaki, H.; Kim, M.J.; Kawada, T. Triiodothyronine induces UCP-1 expression and mitochondrial biogenesis in human adipocytes. Am. J. Physiol.-Cell Physiol. 2012, 302, C463–C472. [Google Scholar] [CrossRef]
  152. Standaert, M.L.; Kanoh, Y.; Sajan, M.P.; Bandyopadhyay, G.; Farese, R.V. Cbl, IRS-1, and IRS-2 mediate effects of rosiglitazone on PI3K, PKC-λ, and glucose transport in 3T3/L1 adipocytes. Endocrinology 2002, 143, 1705–1716. [Google Scholar] [CrossRef]
  153. Fernández-Veledo, S.; Nieto-Vazquez, I.; de Castro, J.; Ramos, M.P.; Brüderlein, S.; Möller, P.; Lorenzo, M. Hyperinsulinemia induces insulin resistance on glucose and lipid metabolism in a human adipocytic cell line: Paracrine interaction with myocytes. J. Clin. Endocrinol. Metab. 2008, 93, 2866–2876. [Google Scholar] [CrossRef] [PubMed]
  154. Ahonen, M.A.; Höring, M.; Nguyen, V.D.; Qadri, S.; Taskinen, J.H.; Nagaraj, M.; Olkkonen, V.M. Insulin-inducible THRSP maintains mitochondrial function and regulates sphingolipid metabolism in human adipocytes. Mol. Med. 2022, 28, 68. [Google Scholar] [CrossRef] [PubMed]
  155. Gao, J.; Yan, X.-L.; Li, R.; Liu, Y.; He, W.; Sun, S.; Zhang, Y.; Liu, B.; Xiong, J.; Mao, N. Characterization of OP9 as authentic mesenchymal stem cell line. J. Genet. Genom. 2010, 37, 475–482. [Google Scholar] [CrossRef] [PubMed]
  156. Lane, J.M.; Doyle, J.R.; Fortin, J.P.; Kopin, A.S.; Ordovás, J.M. Development of an OP9 derived cell line as a robust model to rapidly study adipocyte differentiation. PLoS ONE 2014, 9, e112123. [Google Scholar] [CrossRef]
  157. Li, X.; Shridas, P.; Forrest, K.; Bailey, W.; Webb, N.R. Group X secretory phospholipase A2 negatively regulates adipogenesis in murine models. FASEB J. 2010, 24, 4313. [Google Scholar] [CrossRef]
  158. Rubin, C.S.; Hirsch, A.; Fung, C.; Rosen, O.M. Development of hormone receptors and hormonal responsiveness in vitro: Insulin receptors and insulin sensitivity in the preadipocyte and adipocyte forms of 3T3-L1 cells. J. Biol. Chem. 1978, 253, 7570–7578. [Google Scholar] [CrossRef]
  159. Groeneveld, M.P.; Brierley, G.V.; Rocha, N.M.; Siddle, K.; Semple, R.K. Acute knockdown of the insulin receptor or its substrates Irs1 and 2 in 3T3-L1 adipocytes suppresses adiponectin production. Sci. Rep. 2016, 6, 21105. [Google Scholar] [CrossRef]
  160. Fu, M.; Sun, T.; Bookout, A.L.; Downes, M.; Yu, R.T.; Evans, R.M.; Mangelsdorf, D.J. A Nuclear Receptor Atlas: 3T3-L1 Adipogenesis. Mol. Endocrinol. 2005, 19, 2437–2450. [Google Scholar] [CrossRef]
  161. Rizzatti, V.; Boschi, F.; Pedrotti, M.; Zoico, E.; Sbarbati, A.; Zamboni, M. Lipid droplets characterization in adipocyte differentiated 3T3-L1 cells: Size 1108 and optical density distribution. Eur. J. Histochem. 2013, 57, e24. [Google Scholar] [CrossRef]
  162. Brackett, L.E.; Daly, J.W. Functional characterization of the A2b adenosine receptor in NIH 3T3 fibroblasts. Biochem. Pharmacol. 1994, 47, 801–814. [Google Scholar] [CrossRef]
  163. Choudhary, S.A.; Bora, N.; Banerjee, D.; Arora, L.; Das, A.S.; Yadav, R.; Dasgupta, S. A novel small molecule A2A adenosine receptor agonist, indirubin-3′-monoxime, alleviates lipid-induced inflammation and insulin resistance in 3T3-L1 adipocytes. Biochem. J. 2019, 476, 2371–2391. [Google Scholar] [CrossRef]
  164. Saad, M.; Folli, F.; Araki, E.; Hashimoto, N.; Csermely, P.; Kahn, C. Regulation of insulin receptor, insulin receptor substrate-1 and phosphatidylinositol 3-kinase in 3T3-F442A adipocytes. Effects of differentiation, insulin, and dexamethasone. Mol. Endocrinol. 1994, 8, 545–557. [Google Scholar]
  165. Hainque, B.; Moustaid, N.; Quignard-Boulange, A.; Ardouin, B.; Lavau, M. Glucocorticoid binding during the differentiation of 3T3-F442A fibroblasts into adipocytes. A possible regulatory effect of insulin. Biochim. Biophys. Acta Mol. Cell Res. 1987, 931, 347–353. [Google Scholar] [CrossRef] [PubMed]
  166. Seo, J.B.; Moon, H.M.; Kim, W.S.; Lee, Y.S.; Jeong, H.W.; Yoo, E.J.; Kim, J.B. Activated liver X receptors stimulate adipocyte differentiation through induction of peroxisome proliferator-activated receptor γ expression. Mol. Cell. Biol. 2004, 24, 3430–3444. [Google Scholar] [CrossRef] [PubMed]
  167. Flores-Delgado, G.; Marsch-Moreno, M.; Kuri-Harcuch, W. Thyroid hormone stimulates adipocyte differentiation of 3T3 cells. Mol. Cell. Biochem. 1987, 76, 35–43. [Google Scholar] [CrossRef] [PubMed]
  168. Khadija, E.; Courtalon, A.; Gauthereau, X.; Chambaut-Guérin, A.M.; Pairault, J.; Fève, B. Differential regulation by tumor necrosis factor-α of β1-, β2-, and β3-adrenoreceptor gene expression in 3T3-F442A adipocytes. J. Biol. Chem. 1997, 272, 24514–24521. [Google Scholar]
  169. Grimaldi, P.A. The roles of PPARs in adipocyte differentiation. Prog. Lipid Res. 2001, 40, 269–281. [Google Scholar] [CrossRef]
  170. Safonova, I.; Darimont, C.; Amri, E.-Z.; Grimaldi, P.; Ailhaud, G.; Reichert, U.; Shroot, B. Retinoids are positive effectors of adipose cell differentiation. Biochem. Pharmacol. 1994, 104, 201–211. [Google Scholar] [CrossRef]
  171. Schneider, L.; El-Yazidi, C.; Dace, A.; Maraninchi, M.; Planells, R.; Margotat, A.; Torresani, J. Expression of the 1,25-(OH)2 vitamin D3 receptor gene during the differentiation of mouse Ob17 preadipocytes and cross talk with the thyroid hormone receptor signalling pathway. J. Mol. Endocrinol. 2005, 34, 221–235. [Google Scholar] [CrossRef]
  172. Teboul, M.; Bismuth, J.; Ghiringhelli, O.; Bonne, J.; Torresani, J. Developmental and thyroidal regulation of the nuclear T3 receptors/c-erb A oncogene products in the Ob 17 preadipocyte cell line. J. Recept. Res. 1991, 11, 865–882. [Google Scholar] [CrossRef]
  173. Mendez-Ferrer, S.; Lucas, D.; Battista, M.; Jang, J.; Frenette, P. Circadian traffic of hematopoietic stem cells is orchestrated by the molecular clock and mediated by β3-adrenergic signals from the sympathetic nervous system. Blood 2007, 110, 219. [Google Scholar] [CrossRef]
  174. Buyse, M.; Viengchareun, S.A.Y.; Bado, A.; Lombès, M. Insulin and glucocorticoids differentially regulate leptin transcription and secretion in brown adipocytes. FASEB J. 2001, 15, 1357–1366. [Google Scholar] [CrossRef] [PubMed]
  175. Gimble, J.M.; Dorheim, M.A.; Cheng, Q.; Medina, K.; Wang, C.S.; Jones, R.; Koren, E.; Pietrangeli, C.; Kincade, P.W. Adipogenesis in a murine bone marrow stromal cell line capable of supporting B lineage lymphocyte growth and proliferation: Biochemical and molecular characterization. Eur. J. Immunol. 1990, 20, 379–387. [Google Scholar] [CrossRef] [PubMed]
  176. Horowitz, M.C.; Berry, R.; Holtrup, B.; Sebo, Z.; Nelson, T.; Fretz, J.A.; Lindskog, D.; Kaplan, J.L.; Ables, G.; Rodeheffer, M.S.; et al. Bone marrow adipocytes. Adipocyte 2017, 6, 193–204. [Google Scholar] [CrossRef]
  177. Klaus, S.; Choy, L.; Champigny, O.; Cassard-Doulcier, A.M.; Ross, S.; Spiegelman, B.; Ricquier, D. Characterization of the novel brown adipocyte cell line HIB 1B: Adrenergic pathways involved in regulation of uncoupling protein gene expression. J. Cell Sci. 1994, 107, 313–319. [Google Scholar] [CrossRef]
  178. Hondares, E.; Rosell, M.; Díaz-Delfín, J.; Olmos, Y.; Monsalve, M.; Iglesias, R.; Villarroya, F.; Giralt, M. Peroxisome Proliferator-activated Receptor α (PPARα) Induces PPARγ Coactivator 1α (PGC-1α) Gene Expression and Contributes to Thermogenic Activation of Brown Fat: Involvement of PRDM16. J. Biol. Chem. 2011, 286, 43112–43122. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  179. Paz, K.; Liu, Y.F.; Shorer, H.; Hemi, R.; LeRoith, D.; Quan, M.; Zick, Y. Phosphorylation of insulin receptor substrate-1 (IRS-1) by protein kinase B positively regulates IRS-1 function. J. Biol. Chem. 1999, 274, 28816–28822. [Google Scholar] [CrossRef]
  180. Sheng, X.; Zhu, X.; Zhang, Y.; Cui, G.; Peng, L.; Lu, X.; Zang, Y.Q. Rhein protects against obesity and related metabolic disorders through liver X receptor-mediated uncoupling protein 1 upregulation in brown adipose tissue. Int. J. Biol. Sci. 2012, 8, 1375. [Google Scholar] [CrossRef]
  181. Soumano, K.; Desbiens, S.; Rabelo, R.; Bakopanos, E.; Camirand, A.; Silva, J.E. Glucocorticoids inhibit the transcriptional response of the uncoupling protein-1 gene to adrenergic stimulation in a brown adipose cell line. Life Sci. 2000, 165, 15–26. [Google Scholar] [CrossRef]
  182. Penfornis, P.; Viengchareun, S.; Le Menuet, D.; Cluzeaud, F.; Zennaro, M.-C.; Lombès, M. The mineralocorticoid receptor mediates aldosterone-induced differentiation of T37i cells into brown adipocytes. Am. J. Physiol. Endocrinol. Metab. 2000, 279, E386–E394. [Google Scholar] [CrossRef]
  183. Held, N.M.; Kuipers, E.N.; van Weeghel, M.; van Klinken, J.B.; Denis, S.W.; Lombès, M.; Wanders, R.J.; Vaz, F.M.; Rensen, P.C.; Verhoeven, A.J.; et al. Pyruvate dehydrogenase complex plays a central role in brown adipocyte energy expenditure and fuel utilization during short-term beta-adrenergic activation. Sci. Rep. 2018, 8, 9562. [Google Scholar] [CrossRef]
  184. Chida, K.; Hashiba, H.; Sasaki, K.; Kuroki, T. Activation of Protein Kinase C and specific phosphorylation of a Mr 90,000 membrane protein of promotable BALB/3T3 and C3H/10T½ cells by tumor promoters. Cancer Res. 1986, 46, 1055–1062. [Google Scholar]
  185. Guernsey, D.L.; Schmidt, T.J. Corticosterone effects on differentiation and X-ray-induced transformation of C3H/10T1/2 mouse cells. Cell Differ. 1988, 24, 159–164. [Google Scholar] [CrossRef]
  186. Rong, J.X.; Klein, J.L.D.; Qiu, Y.; Xie, M.; Johnson, J.H.; Waters, K.M.; Zhang, V.; Kashatus, J.A.; Remlinger, K.S.; Bing, N.; et al. Rosiglitazone induces mitochondrial biogenesis in differentiated murine 3T3-L1 and C3H/10T1/2 adipocytes. PPAR Res. 2011, 2011, 179454. [Google Scholar] [CrossRef]
  187. Ogawa, A.; Ohba, K.I.; Uchida, Y.; Wada, K. New adipogenic cell lines derived from C3H10T1/2. Vitr. Cell. Dev. Biol.-Anim. 1999, 35, 307–310. [Google Scholar] [CrossRef]
  188. Lindsey, R.C.; Mohan, S. Thyroid hormone acting via TRβ induces expression of browning genes in mouse bone marrow adipose tissue. Endocrine 2017, 56, 109–120. [Google Scholar] [CrossRef]
  189. Takahata, Y.; Takarada, T.; Iemata, M.; Yamamoto, T.; Nakamura, Y.; Kodama, A.; Yoneda, Y. Functional expression of β2 adrenergic receptors responsible for protection against oxidative stress through promotion of glutathione synthesis after Nrf2 upregulation in undifferentiated mesenchymal C3H10T1/2 stem cells. J. Cell. Physiol. 2009, 218, 268–275. [Google Scholar] [CrossRef]
  190. Tchivileva, I.E.; Tan, K.S.; Gambarian, M.; Nackley, A.G.; Medvedev, A.V.; Romanov, S.; Diatchenko, L. Signaling pathways mediating β3-adrenergic receptor-induced production of interleukin-6 in adipocytes. Mol. Immunol. 2009, 46, 2256–2266. [Google Scholar] [CrossRef] [PubMed]
  191. Adamo, A.; Delfino, P.; Gatti, A.; Bonato, A.; Takam Kamga, P.; Bazzoni, R.; Ugel, S.; Mercuri, A.; Caligola, S.; Krampera, M. HS-5 and HS-27A stromal cell lines to study bone marrow mesenchymal stromal cell-mediated support to cancer development. Front. Cell Dev. Biol. 2020, 8, 584232. [Google Scholar] [CrossRef] [PubMed]
  192. Satilmis, H.; Verheye, E.; Vlummens, P.; Oudaert, I.; Vandewalle, N.; Fan, R.; De Veirman, K. Targeting the β2-adrenergic receptor increases chemosensitivity in multiple myeloma by induction of apoptosis and modulating cancer cell metabolism. J. Pathol. 2023, 259, 69–80. [Google Scholar] [CrossRef] [PubMed]
  193. Chen, Z.; Yu, H.; Shi, X.; Warren, C.R.; Lotta, L.A.; Friesen, M.; Meissner, T.B.; Langenberg, C.; Wabitsch, M.; Wareham, N.; et al. Functional screening of candidate causal genes for insulin resistance in human preadipocytes and adipocytes. Circ. Res. 2020, 126, 330–346. [Google Scholar] [CrossRef]
  194. Yeo, C.R.; Agrawal, M.; Hoon, S.; Shabbir, A.; Shrivastava, M.K.; Huang, S.; Toh, S.A. SGBS cells as a model of human adipocyte browning: A comprehensive comparative study with primary human white subcutaneous adipocytes. Sci. Rep. 2017, 7, 4031. [Google Scholar] [CrossRef] [PubMed]
  195. Lahnalampi, M.; Heinäniemi, M.; Sinkkonen, L.; Wabitsch, M.; Carlberg, C. Time-resolved expression profiling of the nuclear receptor superfamily in human adipogenesis. PLoS ONE 2010, 5, e12991. [Google Scholar] [CrossRef]
  196. Ceperuelo-Mallafre, V.; Ejarque, M.; Duran, X.; Pachon, G.; Vazquez-Carballo, A.; Roche, K.; Nunez-Roa, C.; Garrido-Sanchez, L.; Tinahones, F.J.; Vendrell, J.; et al. Zinc-α2-glycoprotein modulates AKT-dependent insulin signaling in human adipocytes by activation of the PP2A phosphatase. PLoS ONE 2015, 10, e0129644. [Google Scholar] [CrossRef] [PubMed]
  197. Kulebyakin, K.; Tyurin-Kuzmin, P.; Efimenko, A.; Voloshin, N.; Kartoshkin, A.; Karagyaur, M.; Grigorieva, O.; Novoseletskaya, E.; Sysoeva, V.; Makarevich, P.; et al. Decreased Insulin Sensitivity in Telomerase-Immortalized Mesenchymal Stem Cells Affects Efficacy and Outcome of Adipogenic Differentiation in vitro. Front. Cell Dev. Biol. 2021, 9, 662078. [Google Scholar] [CrossRef]
  198. Lewandowski, M.; Busch, R.; Marschner, J.A.; Merk, D. Comparative Evaluation and Profiling of Chemical Tools for the Nuclear Hormone Receptor Family 2. ACS Pharmacol. Transl. Sci. 2025, 8, 854–870. [Google Scholar] [CrossRef]
  199. Kazantzis, M.; Takahashi, V.; Hinkle, J.; Kota, S.; Zilberfarb, V.; Issad, T.; Strosberg, A.D. PAZ6 cells constitute a representative model for human brown pre-adipocytes. Front. Endocrinol. 2012, 3, 13. [Google Scholar] [CrossRef]
  200. Oberkofler, H.; Schraml, E.; Krempler, F.; Patsch, W. Restoration of sterol-regulatory-element-binding protein-1c gene expression in HepG2 cells by peroxisome-proliferator-activated receptor-γ co-activator-1α. Biochem. J. 2004, 381, 357–363. [Google Scholar] [CrossRef]
Figure 1. Human and murine cell lines in adipogenesis research. (A). Tissue origins of selected human (left) and murine (right) cell lines. (B). Analysis of the publication frequency of these selected cell lines in adipogenesis research. Data were retrieved from Scholar (Google: https://scholar.google.com/), PubMed (NIH: https://pubmed.ncbi.nlm.nih.gov/), and Scopus (Elsevier: https://www.scopus.com/) databases. Exact frequency values for each cell line are publicly available via the respective databases by entering the cell line name.
Figure 1. Human and murine cell lines in adipogenesis research. (A). Tissue origins of selected human (left) and murine (right) cell lines. (B). Analysis of the publication frequency of these selected cell lines in adipogenesis research. Data were retrieved from Scholar (Google: https://scholar.google.com/), PubMed (NIH: https://pubmed.ncbi.nlm.nih.gov/), and Scopus (Elsevier: https://www.scopus.com/) databases. Exact frequency values for each cell line are publicly available via the respective databases by entering the cell line name.
Receptors 04 00019 g001
Figure 2. Stages of adipogenesis. (A). Schematic representation of the multistage process of adipocyte differentiation, illustrating the transition from MSCs to preadipocytes and mature adipocytes. Commonly used adipogenic cell lines are listed based on their phenotype and predisposition to adipose differentiation (upper left). (B). A flowchart outlining the molecular events driving adipogenesis. Events promoting adipogenesis are indicated by black arrows, while inhibitory interactions are denoted in red. The text follows a color code: black for receptors, enzymes, or events; green for positive regulators of adipogenesis; and red for examples of adipogenic inhibitors. (C). Word cloud representation of main adipogens employed; the font size represents the relative frequency of use in the analyzed reports. Original data for this analysis are presented in Table 1.
Figure 2. Stages of adipogenesis. (A). Schematic representation of the multistage process of adipocyte differentiation, illustrating the transition from MSCs to preadipocytes and mature adipocytes. Commonly used adipogenic cell lines are listed based on their phenotype and predisposition to adipose differentiation (upper left). (B). A flowchart outlining the molecular events driving adipogenesis. Events promoting adipogenesis are indicated by black arrows, while inhibitory interactions are denoted in red. The text follows a color code: black for receptors, enzymes, or events; green for positive regulators of adipogenesis; and red for examples of adipogenic inhibitors. (C). Word cloud representation of main adipogens employed; the font size represents the relative frequency of use in the analyzed reports. Original data for this analysis are presented in Table 1.
Receptors 04 00019 g002
Figure 3. Characteristics of culture seeding conditions and receptor toolkit in adipogenic models. (A). The cell numbers from the reported protocols were normalized to cells/cm2. The segment on the x-axis highlights the heterogeneity in cell culture conditions. whereas the y-axis includes the differentiation efficiency. A color code is used to identifies the different cell lines employed. the value next to each datum refers to the reported days needed to assess the differentiation (B). Heatmap showing the receptor diversity expressed in the most commonly used adipogenic models. The color code is as follows: white: expression has not been reported; light green: expression reported in independent studies (non-comparative); dark green (++): receptor expression documented in studies involving multiple cell lines under the same experimental conditions with corresponding comparisons. Receptor expression was considered only when validated by ligand binding assays, Western blot, Northern blot, PCR, or another confirmatory methods such as immunocytochemistry or flow cytometry. This figure was generated exclusively from previously published studies. References supporting Figure 3A are listed in Table 1, while those related to receptor expression patterns in the cell lines are provided in Table S1. The figure represents an integrative analysis of the literature, and no unpublished or original experimental data are included.
Figure 3. Characteristics of culture seeding conditions and receptor toolkit in adipogenic models. (A). The cell numbers from the reported protocols were normalized to cells/cm2. The segment on the x-axis highlights the heterogeneity in cell culture conditions. whereas the y-axis includes the differentiation efficiency. A color code is used to identifies the different cell lines employed. the value next to each datum refers to the reported days needed to assess the differentiation (B). Heatmap showing the receptor diversity expressed in the most commonly used adipogenic models. The color code is as follows: white: expression has not been reported; light green: expression reported in independent studies (non-comparative); dark green (++): receptor expression documented in studies involving multiple cell lines under the same experimental conditions with corresponding comparisons. Receptor expression was considered only when validated by ligand binding assays, Western blot, Northern blot, PCR, or another confirmatory methods such as immunocytochemistry or flow cytometry. This figure was generated exclusively from previously published studies. References supporting Figure 3A are listed in Table 1, while those related to receptor expression patterns in the cell lines are provided in Table S1. The figure represents an integrative analysis of the literature, and no unpublished or original experimental data are included.
Receptors 04 00019 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gutiérrez-García, A.; Olivas-Aguirre, F.J.; Olivas-Aguirre, M. Critical Evaluation of Adipogenic Cell Models: Impact of the Receptor Toolkit on Adipogenic Potential. Receptors 2025, 4, 19. https://doi.org/10.3390/receptors4040019

AMA Style

Gutiérrez-García A, Olivas-Aguirre FJ, Olivas-Aguirre M. Critical Evaluation of Adipogenic Cell Models: Impact of the Receptor Toolkit on Adipogenic Potential. Receptors. 2025; 4(4):19. https://doi.org/10.3390/receptors4040019

Chicago/Turabian Style

Gutiérrez-García, Andrea, Francisco Javier Olivas-Aguirre, and Miguel Olivas-Aguirre. 2025. "Critical Evaluation of Adipogenic Cell Models: Impact of the Receptor Toolkit on Adipogenic Potential" Receptors 4, no. 4: 19. https://doi.org/10.3390/receptors4040019

APA Style

Gutiérrez-García, A., Olivas-Aguirre, F. J., & Olivas-Aguirre, M. (2025). Critical Evaluation of Adipogenic Cell Models: Impact of the Receptor Toolkit on Adipogenic Potential. Receptors, 4(4), 19. https://doi.org/10.3390/receptors4040019

Article Metrics

Back to TopTop