Skip to main content

Lesson on obesity and anatomy of adipose tissue: new models of study in the era of clinical and translational research

Abstract

Obesity is a serious global illness that is frequently associated with metabolic syndrome. Adipocytes are the typical cells of adipose organ, which is composed of at least two different tissues, white and brown adipose tissue. They functionally cooperate, interconverting each other under physiological conditions, but differ in their anatomy, physiology, and endocrine functions. Different cellular models have been proposed to study adipose tissue in vitro. They are also useful for elucidating the mechanisms that are responsible for a pathological condition, such as obesity, and for testing therapeutic strategies. Each cell model has its own characteristics, culture conditions, advantages and disadvantages. The choice of one model rather than another depends on the specific study the researcher is conducting. In recent decades, three-dimensional cultures, such as adipose spheroids, have become very attractive because they more closely resemble the phenotype of freshly isolated cells. The use of such models has developed in parallel with the evolution of translational research, an interdisciplinary branch of the biomedical field, which aims to learn a scientific translational approach to improve human health and longevity. The focus of the present review is on the growing body of data linking the use of new cell models and the spread of translational research. Also, we discuss the possibility, for the future, to employ new three-dimensional adipose tissue cell models to promote the transition from benchside to bedsite and vice versa, allowing translational research to become routine, with the final goal of obtaining clinical benefits in the prevention and treatment of obesity and related disorders.

Introduction

The obesity rate has been increasing worldwide in recent decades, and obesity is often associated with serious comorbidities [1]. A deep knowledge of the anatomy and pathophysiology of adipose organ is fundamental for choosing the best therapeutic strategies.

The goal of this review, after a concise overview of obesity, will be on describing adipose organ and discussing the many different cell models available for studying it. We will describe both two-dimensional and three-dimensional models, considering their respective benefits and constraints, with the ultimate goal of utilizing them for the evaluation of potential new drugs and the provision of information regarding adverse effects. Everything will be framed from the perspective of translational research, which aims to transform laboratory findings into clinical applications and convert clinical evidence into research questions, with the figure of the patient at the center. This means allowing the transition from the laboratory to the clinic and to industry.

Obesity

An overview

Obesity is a metabolic disease that is now considered a worldwide epidemic and represents a global public health problem [1]. It is associated with a reduction in quality of life and a shortened life span and can be described as a multifactorial chronic disease similar to the aging process [2, 3]. In a few simple words, it develops when the energy from the caloric intake exceeds energy expenditure, resulting in a state known as a positive energy balance. Under these conditions, progressive pathological remodeling of adipose tissues occurs because adipocytes accumulate energy in anticipation of a possible period of fasting. During the progression toward obesity, fat cells increase both in size (hypertrophy) and in number (hyperplasia) [4,5,6,7].

Overweight and obesity in adults are usually classified considering the weight-for-height index. It is defined as a person’s weight in kilograms divided by the square of his height in meters (kg/m2) [8]. The result is commonly indicated as the body mass index (BMI). Considering the distribution of white adipose tissue, two kinds of obesity can be described: “apple-shaped obesity”, also called visceral or android obesity, is typical of men and consists of an accumulation of fat in the face, neck, shoulders and, above all, abdominal cavity; “pear-shaped obesity”, typically described as female or gynoid obesity, in which adipose tissue is mainly located beneath the skin (subcutaneous compartment) of hips, buttocks, thighs and belly [9].

Epidemiological evidence

Recent epidemiological studies indicate that the incidence of obesity has increased since 1975 in adults and children; in 2016, more than 1.9 billion adults were classified as overweight. In 2019, these numbers increased, and the overall prevalence of obesity was approximately 14%. Moreover, in the same year, approximately 38.2 million children under 5 years of age and more than 340 million children aged 5 to 19 years were overweight or obese [10, 11].

Overweight and obesity can develop because of unhealthy eating behaviors, such as preferring large amounts of food, which is often high in fat, sugar and refined carbs or is ultra-processed; drinking sugary beverages; and consuming too many alcohol drinks regularly. Moreover, high-calorie food has become less expensive and more convenient and is heavily advertised and promoted. Decreased or no physical activity and a sedentary lifestyle, as well as a shortened sleep duration but also genetic, endocrine, metabolic, and environmental factors, are currently considered the main common causes of the obesity epidemic worldwide [12, 13].

Associated comorbidities

Obesity is considered a risk factor for the onset of other chronic pathologies responsible for 86% of the deaths in Europe (60% worldwide) (https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight). These pathologies include metabolic disorders, such as type 2 diabetes [14], dyslipidemia, insulin resistance, metabolic syndrome, atherosclerosis, hypertension, cardiovascular diseases [15, 16], nonalcoholic fatty liver diseases [17], and other diseases that are not classified as metabolic, such as some forms of cancer [18, 19], osteoarthritis [20], obstructive sleep apnea [21] and neurodegenerative diseases [22, 23]. For all these complications, obesity curtails life expectancy and is associated with a reduction in quality of life and increased healthcare costs [14, 18]. In this regard, a loss of body weight through diet and exercise reduces all causes of mortality in obese individuals [24].

Insulin resistance and type 2 diabetes are complex multifactorial diseases and are the most well-known disorders related to obesity, but the prevalence of endocrine disorders in patients with obesity is considerable [25, 26]. In some cases, hormonal imbalances, including either peptide or steroid hormones, are related to type 2 diabetes. For example, it has been reported that the gastric ghrelin cells of obese patients who are in prediabetic conditions are hyperactive [27]. Considering the known inhibitory effect of ghrelin on insulin secretion [28, 29], its overproduction by the stomach of obese patients may be involved in the pathogenesis of type 2 diabetes.

Adipose organ

General features

The great diffusion of obesity in Western countries has attracted interest in adipose tissue, and a new anatomical and functional concept has been proposed: adipose organ. The interest in adipose organ is related to the discovery, in 1994, of the hormone leptin. It is secreted by a particular type of adipocyte (white adipocyte) and induces the search for food when its levels are low [30]. The discovery of leptin pushed many researchers to deepen their studies on adipose tissue, which was no longer considered a mere energy reserve organ but rather an important endocrine organ capable of interacting with brain centers [31]. In fact, adipose tissue represents a fundamental organ in the life of mammals because it constitutes the main energy source for the approximately one hundred billion cells that require the organism to burn molecules for survival. The presence of adipose tissue allows us to have fasting intervals between one meal and another, and when the interval, due to lack of food, is prolonged, the adipose tissue becomes the tissue of survival.

Anatomy

The adipose organ of all mammals is composed of two main types of cells that can accumulate lipids (triglycerides) but exhibit notable anatomical and functional differences; these two cytotypes are defined as white adipocytes and brown adipocytes. The former are the best known because they constitute the parenchymal element of what is commonly defined as fat, which is abundant in humans and is pathologically increased in obese individuals. White adipocytes are unilocular, accumulate lipids for survival and secrete peptide hormones, cytokines known as adipokines [32], and exosomal miRNAs, which are involved in the control of metabolism [33, 34]. Adipokines produced by white adipocytes regulate energy expenditure, inflammation, and fibrosis. Imbalanced levels of adipokines in blood affect physiological homeostasis, are implicated in the pathogenesis of metabolic syndrome and play a role in metabolic dysfunction-associated steatotic liver disease (MASLD) [35].

Brown adipocytes are less known, and it was commonly believed that their presence in adult humans was negligible, at least from a functional point of view [36]. They are multilocular and rich of big mitochondria, which are typical of these cells. They produce UCP1 (uncoupling protein 1), a thermogenic protein exclusively expressed in these cells and are involved in the mechanism of thermogenesis because they dissipate lipids to produce heat [37, 38]. In recent years, brown adipocytes have also been recognized as endocrine cells that are able to secrete regulatory factors with local autocrine and paracrine effects but also act on distant tissues and organs [39].

Anatomical dissection has demonstrated that adipose cells are contained in subcutaneous (under the skin) and visceral (thoracic and abdominal regions) depots, indicating clear anatomical autonomy from the rest of the organism. A real organ with multiple depots is therefore configured [6]. Each depot is provided with its vascular-nervous peduncles and contains both white adipocytes and brown adipocytes. There are no anatomical barriers between regions containing white adipocytes (white adipose tissue or WAT) and those containing brown adipocytes (brown adipose tissue or BAT).

Plasticity of the adipose organ

WAT and BAT are often present simultaneously; they functionally cooperate and, under physiological conditions, they can interconvert into one another both in mice and humans [40,41,42]. In other words, adipose organ is endowed with remarkable physiological plasticity, which allows for the adaptation of organ function to specific needs [7, 43]. It is possible that under conditions of chronic cold exposure [44], physical exercise [45] and β-adrenergic stimulation [46], the white part of the organ becomes brown to increase thermogenesis, and this phenomenon is called “browning”. On the other hand, the chronic positivity of the energy balance, which is present when the body is subjected to an obesogenic diet, requires an increase in lipid storage capacity, and the conversion of the brown component to the white component contributes to satisfying this need. This phenomenon is called “whitening” and is associated with hypertrophy and hyperplasia [47, 48].

According to different authors, in addition to brown fat cells, in mouse and human there are other fat cells. They are called “brown-in-white” (“brite”) [49] or “beige” [50] adipocytes. As the brown ones, they are rich in mitochondria and express UCP1, but they are dispersed among white adipocytes. As brown adipocytes, they have also thermogenic properties, but these characteristics are UCP1-independent [51]. Beige adipocytes have different embryonic origins respect to brown ones, express a unique gene expression profile, and can derive from progenitors after de novo differentiation or from white adipocytes that can transform under specific conditions [52]. Considering the last option, under the conditions we have described (cold exposure, exercise, adrenergic stimulation), white adipose tissue undergoes morphological, molecular, metabolic and, consequentely, functional changes. These changes affect above all the mitochondria, whose number increases, and lead to obtain beige adipocytes, showing intermediate characteristics between those of white and those of brown adipocytes [53].

Direct transformation of brown/beige adipocytes into white adipocytes and vice versa, depending on the needs of the organism, is known as “transdifferentiation”. It is a physiological phenomenon of cooperation between two adipose tissues in which differentiated mature adipose cells reversibly reprogram their genome without going through a dedifferentiation stage and therefore change phenotype and function in response to physiological stimuli [44, 54, 55]. Another example of transdifferentiation has been reported in breast WAT, which transdifferentiates into milk-secreting glands during pregnancy and lactation. Breast adipocytes change their anatomy, aggregate with each other, incorporating myoepithelial cells and developing a glandular alveolar anatomy. The color of the organ becomes pink because of the presence of pink cells, and they are considered adipocytes because they are rich in cytoplasmic lipids but lack the physiological characteristics of adipocytes. After lactation, the adipoglandular (white‒pink) conversion phenomenon reverses, and pink adipocytes transdifferentiate back into WAT [56]. Recently, gland-BAT (pink‒brown) conversion in the postlactation period has been shown [57].

These data lead to two conclusions:

  • Physiologic white–brown/beige adipose tissue transdifferentiation, given the ability of brown adipose tissue to disperse energy, could be a valid approach for treating obesity, type 2 diabetes and cardiovascular diseases;

  • Mature cells can physiologically reprogram their genome to change their phenotype and, therefore, function. Perhaps, it could be useful to change, for example, tumor cells into normal cells [58].

The obese adipose organ: inflammation

The adipose organ of obese subjects undergoes pathological remodeling through a series of morphological and functional alterations. These events trigger proinflammatory immune cell responses. In particular, chronic low-grade inflammation, mainly supported by macrophages, occurs. Macrophage infiltration is strictly related to weight gain. In fact, under conditions of persistent positive energy balance, adipocytes expand, but their expansion capacity is limited, and the cells die [59, 60]. Because obese adipose tissue contains many dead adipocytes, inflammation becomes prolonged and persistent. Both in animals and in humans, a relationship exists between adiposity and adipose tissue inflammation; that is, the number of macrophages correlates with the number and size of adipocytes. It has been proposed that adipocyte death could occur through necrosis or pyroptosis [61, 62]. In this context, numerous cytokines are released into the microenvironment (TNF, IFNγ, IL-1β and IL-6), inducing innate immune responses [63]. Moreover, hypertrophic and stressed adipose cells secrete chemoattractants such as MCP-1 (monocyte chemoattractant protein-1) and HP (haptoglobin), which recruit macrophages to sequester and ingest adipocyte debris, particularly residual lipid droplets [64]. It has been shown that over 90% of macrophages are localized almost exclusively around dying fat cells to form typical structures called crown-like structures (CLSs), in which macrophages surround the cell residue in a crown-like manner and scavenge lipids and cellular debris. Sometimes, they evolve to form syncytia and multinucleated giant cells, which are much more common in obese animals and individuals than in lean animals [61]. Therefore, the presence of CLSs can be considered a biomarker of adipose tissue inflammation, and their formation is the principal inflammatory lesion. This is also evident for epicardial adipose tissue (EAT). A positive correlation between EAT thickness and cardiac diseases has been shown [65, 66]. Adipose tissue surrounding coronary arteries is considered EAT because no anatomical boundaries separate the two tissues and is a part of perivascular adipose tissue (PVAT) [67]. In general, PVAT is a special type of adipose tissue juxtaposing the outer adventitia of most blood vessels within the vascular tree of different districts of the human body [68]. It contains primarily preadipocytes, mature adipocytes, stem cells, fibroblasts, nerve cells, and various inflammatory cells [69].

From an anatomical point of view, PVAT is a heterogeneous tissue, exhibiting phenotypes from brown to white and beige adipose tissue, depending on the vascular bed where the PVAT is located [70].

As other fat depots, PVAT exerts important endocrine and paracrine functions, secreting adipokines and angiotensin, with a continuous bidirectional communication with the underlying vessels. These secreting properties are crucial for maintaining vascular homeostasis and health under normal physiological conditions [67, 71]. Under pathological conditions (i.e. patients with features of obesity, hyperlipidemia or metabolic syndrome), PVAT becomes dysfunctional and, together with endothelial dysfunction, can play a role in the development and progression of vascular diseases (hypertension or atherosclerotic plaques) [72, 73], predisposing the body to a prothrombotic or a proinflammatory profile.

Weight gain is also associated with cardiovascular diseases, such as myocardial infarction, and it is particularly true if weight gain is rapid. It has been reported that under these conditions, epicardial fat, which has anatomical continuity with the myocardium, is particularly rich in CLS. Therefore, CLS density could also be an important tool for predicting cardiovascular diseases [74]. A variety of other inflammatory changes also occurs, with activation and accumulation of different immune cell types in the white adipose tissue [75, 76].

It has been recently shown that the infiltration of adipose organ by macrophages represents an important early event in the onset of complications such as diabetes 2 and is chronically prolonged. This phenomenon correlates with BMI and with the systemic inflammatory state (measured by C-reactive protein, CRP, levels) and is reversible after weight loss. During this long process, macrophages produce cytokines that exert various side effects. Among these, insulin resistance, type 2 diabetes, and metabolic syndrome can be established because the function of the insulin receptor and the secretion of insulin itself are impaired [59, 60, 77].

Several lines of evidence suggest greater pathogenicity in visceral adipose tissue than in subcutaneous adipose tissue. Individuals with a high WHR (“waist hip-to-ratio”), which is typical of apple-shaped obesity, are more likely to develop comorbidities, such as diabetes, and, in general, have a greater risk of developing metabolic syndrome. This is also true for individuals with a normal body weight and BMI if the body fat percentage is high. In contrast, a low WHR, which is typical of pear-shaped obesity, correlates to a low probability of comorbidity, despite a high BMI [78, 79]. These findings can be explained by the fact that the abdominal visceral fat, which is responsible for apple-shaped obesity in men, can expand less than the subcutaneous fat of the hips, which is typical of pear-shaped obesity in women. Because of this smaller critical death size, visceral adipocytes in men die earlier, visceral obesity is more pathogenic, inflammation is more severe, and comorbidities, such as insulin resistance and diabetes, can easily develop [80,81,82].

The obese adipose organ: remodeling of the extracellular matrix

CLS formation is due to general vascular dysfunction in the adipose tissue microenvironment, with areas of hypoxia and, as we have previously observed, adipocyte death. This vascular inflammation leads to changes in the matrix composition within tissues and in the extracellular matrix (ECM), which naturally embeds adipose cells, maintaining the structural integrity of these cells and therefore providing mechanical support [83, 84]. During the development of obesity, the ECM is subjected to structural remodeling, with an excessive accumulation of collagen fibrils and a change in its protein composition. Some proteins (especially various types of collagen) increase their expression and accumulate in the entire adipose tissue, inducing a state of local fibrosis, which increases the total rigidity of the tissue itself, reducing its expandability [85, 86]. Under physiological conditions, the matrix can remodel and stabilize the normal oscillations of adipocyte size. When fibrosis occurs, this dynamic remodeling is lost, and increased ECM stiffness counteracts the tendency of adipocytes to expand [84, 87]. The result of these two opposing actions could be, over time, an increase in mechanical stress, which, in turn, can determine adipocyte death and the consequent inflammation.

In conclusion, hypoxia and fibrosis are two important hallmarks of dysfunctional adipose tissue in obese individuals and are closely related to each other. A deeper knowledge of the morphology, histology and pathology of obese adipose tissue could help to develop effective strategies and treatment approaches to treat obesity and its related comorbidities.

Cell models for the study of adipose tissue

General features

As in industrialized countries, also in some low-middle-income developing countries the incidence and prevalence of obesity have increased, due to profound urbanization and changes in dietary lifestyle, with an adaptation to the standards of the Western diet. The onset of several obesity-associated diseases has important socioeconomic impacts. For these reasons and considering that adipocytes constitute almost 90% of the volume and up to 40% of the cells present in WAT [88], it is extremely crucial to have translational experimental systems (in vitro models) for functional studies of adipocytes. It is particularly interesting to understand the molecular mechanism of fat cell expansion and obesity, not only for better knowing the pathophysiology of adipose tissue but also because the uptake and release of lipids by adipocytes are likely involved in ectopic lipid deposition in different tissues, which can lead to lipotoxicity and other associated diseases [89, 90]. Moreover, we are still unable to maintain metabolic health in overweight patients by directly targeting their adipose tissue. The use of in vitro models could also allow the development of new treatments that could interfere with the progression of obesity and obesity-induced metabolic disorders [91].

Two-dimensional cell models

Mature white adipocytes are difficult to study because this model has some technical limitations. First, these cells are rich in lipids, and for this reason, they float in suspension [92]. Furthermore, they are difficult to isolate, do not proliferate [93], have a short life span ex vivo, and cannot be frozen without breaking. Finally, they are incompatible with many standard cell biology techniques, such as upright microscopy, cell counting, frozen sectioning, or flow cytometry [94]. To overcome the abovementioned limitations regarding adipocyte buoyancy, 3D matrix cultures or ceiling cultures have been used. Although cell survival and metabolic activity increase, adipocytes undergo progressive dedifferentiation [95]. A variant of the ceiling culture is obtained using a small amount of freshly isolated human mature adipocytes, which aggregate under permeable small-pore transwell membranes (membrane-mature adipocyte aggregate cultures, MAAC). However, adipocytes cannot divide, and a constant supply of fresh samples, for example from surgery, is then required but is not always available [96]. Other primary mature adipocyte models are adipose tissue explant cultures, which are small pieces of WAT digested with collagenase to yield adipocytes [97, 98]. These explants remain metabolically active for several weeks and are used to investigate angiogenesis, inflammation and secretion after pharmacological stimuli [99, 100]. Altered adipocyte marker gene expression, with consequent changes in phenotype, has been described for this model [101, 102]. A modern variant of the above-described method is sandwiched WAT, in which a small piece of adipose tissue is enclosed between two tissue-engineered layers of adipose-derived stromal cells [103] and collagenase allows the release of adipocytes. It is a model of a human three-dimensional organ construct, even though it is described among 2-dimensional (2D) models, maintaining metabolic activity for approximately four weeks without dedifferentiation, and it allows the study of WAT physiology, pathophysiology, personalized medicine, and pharmaceutical development.

Other 2D adipocyte culture models are cell lines that can be derived from mice or humans. Some of these models have been used to test the effects of nutraceuticals and compounds on obesity [104, 105] and are preadipocytes, often derived from fibroblast-like cell lines that undergo differentiation and adipogenesis after specific treatments to mimic beige, brown or white adipocytes [106,107,108,109].

Regarding preadipocyte differentiation, the change of preadipocyte morphology into the shape of mature adipocyte is influenced by epigenetic factors, miRNAs and paracrine factors, as well as by the composition and the microenvironments of the ECM [110]. Adipogenesis in mammals larger than rodents can be easily obtained stimulating transcription of specific factors that regulate the differentiation of preadipocytes into mature adipocytes. It is known that insulin is an adipogenic hormone and it has been reported that its effect can be amplified with a prolonged exposure of cells to a cocktail of specific differentiating agents. Among these, peroxisome proliferator-activated receptor γ (PPARγ) agonists (for example oleic acid, rosiglitazone, cycloglitazone) can be considered. They increase the expression of genes involved in the regulation of fatty acid synthesis and, consequently, of adipogenesis [111, 112]. Previously, MacDougald OA et al. had already shown that rodent 3T3-L1 cells become susceptible to complement of exogenous inducers, as insulin-like growth factor, glucocorticoid, fatty acids, which act as transcriptional activators promoting differentiation [113].

The reproducibility of experiments conducted using cell lines is an important characteristic and is undoubtedly an advantage.

A human 2D cell line model can be generated by the transformation and immortalization of human adipose-derived stem cells (hASCs) present in the adipose tissue stromal vascular fraction (SVF) [114, 115]. The SVF is a part of adipose tissue that includes progenitor cells such as preadipocytes, mesenchymal stem cells (including ASCs), pericytes, endothelial cells, and macrophages. The limitation of this model is that immortalization of the cells is accompanied by decreased adipogenic potential. Similar approaches can be used to isolate preadipocytes from the adipose tissue of patients with different pathologies [95].

When choosing to use the SVF fraction, some important aspects must be taken into account. In particular, adipose progenitor cells, including preadipocytes, are characterized by transcriptomic and proteomic heterogeneity, due to cell type, anatomical localization and sex [116]. Single-cell RNA sequencing (scRNA-seq) analysis has been used to analyze this molecular heterogeneity, allowing to identify, in adipose and progenitor stem cells, several subpopulations, according to different markers they express. This heterogeneity has implications in functional outcomes [117, 118]. Preadipocytes, for example, express a great number of markers, some are cell surface antigen markers and are related to adipogenesis; others are intracellular protein signaling and transcription factors which can influence the preadipocyte maturation process, promoting or repressing it [119].

A particular kind of hASC isolated from the SVF was described by Rodriguez in 2004 [120]. These cells are human multipotent adipose-derived stem (hMADS) cells that can differentiate into adipose cells in serum-free, chemically defined medium, resulting in lipid accumulation. Under other conditions, these cells can differentiate into osteoblasts or myoblasts [121]. They proliferate extensively without being transformed and have been shown to be a valid model for studies of fat tissue development and metabolism. Some of the preadipocyte cell lines that have been developed and used over the years are described in the scientific literature [109, 122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140] and reported in Table 1.

Table 1 Principal preadipocyte cell lines developed and used over the years

Preadipocytes, which can easily differentiate in vitro and are considered new, renewable and cryopreservable cell models [141], can be used to obtain primary cell cultures from both rodents [142] and humans [143]. Adipocyte-derived progenitors, or preadipocytes, isolated from SVFs can be expanded through a couple of passages and are compatible with most standard cell biology methods. After differentiation, these cells can accumulate small lipid droplets and express genes and adipogenic markers of mature adipocytes. A great limitation of primary but also cell line-derived preadipocytes is the lack of a unilocular morphology, that is, only a single large central lipid droplet. In contrast, they retain their multilocular morphology, which is typical of brown adipocytes, despite most cells not showing high thermogenic activity [95]. Small lipid droplets have a different membrane-to-volume ratio than large lipid droplets, and this feature influences the amount of membrane-associated proteins synthetized by adipocytes, as well as the accessibility of the lipolytic machinery to triglycerides and the lipolytic activity of cells. The final effect can be altered adipocyte functionality [144, 145]. The result is that preadipocytes are used mainly for studies of differentiation, such as the use of agents that favor adipose cell maturation. The failure of partial in vitro differentiation is probably due to the lack of an adequate cell microenvironment, that is, the lack of a vascular tree [146, 147] or of cell-cell and cell-matrix contacts [148].

Primary cultures of preadipocytes have also been obtained from pigs. They are useful for studying preadipocyte differentiation because they have adipogenic characteristics similar to those of human preadipocytes [149, 150].

Three-dimensional cell models

Cell lines often encounter genetic instability, and in general, 2D cell models fail to recapitulate the native microenvironment normally present in vivo. Furthermore, adipogenesis is a very complex process [151], and 2D models often cannot precisely reproduce it. On the other hand, 3-dimensional (3D) cell cultures have become a very attractive system for the scientific community [152] because they represent more realistic cell conditions in in vivo tissues.

In available 3D models of adipose tissue, adipocytes more closely resemble the phenotype of freshly isolated cells in terms of morphology, lipid droplet size (10–20 μm), expression pattern and lipid composition [153]. Some of them show obese characteristics such as hypertrophy, increased lipolysis, and insulin resistance, or can mimic an inflammatory microenvironment or fibrosis [154,155,156,157].

Furthermore, they are interesting models for studying gene expression and other biological activities because they closely mirror what occurs in living organisms. Interactions among cells and between cells and the matrix regulate proliferation [158], migration [159], and metabolism [160]. The first paper describing spheroidal self-aggregates, derived from carcinoma cell culture, was published approximately 50 years ago [161], but only in the last decade has the use of this kind of model largely been considered. Among 3D cell cultures, spheroids are likely the most studied and used. Cells of a specific type are seeded at a defined concentration and maintained in suspension. With time (hours or days), cells self-aggregate to form spheroidal structures.

Some of the described 2D adipocyte models can be used to establish 3D cultures. For example, spheroid cultures have been obtained from murine cell lines [154, 162] or from hASCs cultured on top of recombinant elastin-like polypeptide (ELP) conjugated to a charged polyelectrolyte, polyethyleneimine (PEI) [163]. hASCs have also been encapsulated in hydrogels to form spheroids [164, 165]. In a modern approach, hASCs were forced to self-aggregate by a hanging-drop technique, and then spheroids were transferred to agar-coated cell culture dishes [166].

Recently, spheroids from the SVFs of human and murine adipose tissue have been derived [167]. It has also been reported that cells self-organize to form 3D vascularized structures with endogenous endothelial cells self-assembled to form highly organized endothelial networks among stromal cells [94, 168, 169], thus resembling miniature copies of adipose organ.

To conclude, different culture methods have been used to obtain 3D structures from hASCs or SVFs, and in some cases, they have allowed 3D microphysiological systems to be obtained. Either scaffold-based or scaffold-free culture systems have been described [170] (Fig. 1). In the first case, scaffolds are designed to reproduce the native extracellular matrix and can have a biological or a synthetic origin [165, 171]. Moreover, cultures can be set up under static conditions, with changes in the medium every few days, or in a perfusion bioreactor [172]. Unfortunately, the scaffold does not permit homogeneous delivery of nutrients to the cells, and the method has a high risk of contamination and does not accurately recapitulate interstitial flow through adipose tissue in vivo [171]. Culture in a perfusion bioreactor allows better cell viability, but adipogenesis is reduced compared with that in static culture [173].

Fig. 1
figure 1

Schematic overview of the different culture methods available for obtaining spheroid cultures. Created with https://BioRender.com

With respect to scaffold-free culture systems, four main culture mechanisms have been described [173]: magnetic levitation [174], dynamic culture conditions [175], hanging drop, and low-attachment culture [176]. Scaffold-free culture mechanisms have increased adipogenic differentiation potential. Furthermore, they do not restrict adipocyte volume expansion, which could limit the use of scaffolds and could be useful for increasing the physiological properties of adipose tissue engineering applications. Truly, scaffold and scaffold-free 3D culture techniques have been employed to develop microphysiological models of adipose tissue [177] also in pathological states such as obesity [178]. The final goal is to recapitulate the same conditions that are present in a living organism but working with an organ system in vitro because it is now clear that the organotypic microenvironment is a fundamental aspect of 3D models. Microphysiological models of adipose tissue can be regarded as adipose organoids, that is, advanced models for studying adipose tissue development and physiology.

3D models versus in vivo models

The most commonly used animal models are mice. They are easy to find and use, not very expensive, and their genetic manipulation follows well-standardized protocols [179]. However, their use has several limitations. First, the induction of obesity, which is linked to dietary modifications, is often accompanied by genetic and chemical modifications. The introduction of different variables does not always allow reproducibility of experiments in humans [180]. Second, substances and drugs active in mice might not be useful or may even be harmful to humans [181]. Rats have also been used, above all to obtain transgenic and knockout animals for the study of adipose tissue biology, but they have some limitations, and mice remain the preferred animal models for studying the expression of adipose tissue-specific genes [182].

Nonhuman primate models represent a good compromise for considering in vivo research because they are much more similar to humans. Moreover, obesity can be more easily induced in these animals than in mice, and the comorbidities that may occur are similar to those in humans [183, 184]. They can therefore represent a valid link between the simplest studies performed in mice and what is expected to be found in humans. However, their use is limited by their high costs and ethical issues.

In the future, sophisticated adipose organ-like structures could reduce, refine, and replace in vivo experiments and animal models because, more than other models, they can reproduce white and brown/beige adipose depots, avoiding interspecies variability and allowing more accuracy concerning possible outcomes in humans [185]. In general, the aggregation of multipotent mesenchymal stem cells in 3D multicellular spheroids results in improved anti-inflammatory and angiogenic properties beyond increased stemness and survival after experimental transplantation. Their therapeutic-associated properties open the possibility of translating the research achievements toward clinical applications [186]. In fact, adipose tissue is ubiquitous and can be easily obtained in large quantities using a minimally invasive procedure. Appropriate differentiation of autologous precursors of adipocytes could be employed for both regenerative medicine and organ damage caused by injury and disease [187, 188]. However, much work still needs to be done. Variables such as reagents (for example, growth medium or adipogenic factors that are used to routinely induce either beige/brown or white adipose differentiation), culture methods, and analysis methods must be standardized because different protocols are described in the scientific literature. To conclude, even if relevant microphysiological models of adipose tissue using hASCs and SVF cells have been developed [189], studies are still limited, especially regarding the influence of the culture environment on the development of adipose tissue constructs that physiologically mimic the same tissue in vivo.

Translational research: main topics of history, present and future

In recent decades, a new category of science, translational research (TR), has developed. Many definitions can be considered for TR. Briefly, it is “the application of findings from basic research to patient, community and population care and to the advancement of the delivery of health services”, as reported by Cokkinos DV, in 2014 [190]. The term was introduced in the 1990s and referred to novel interventions directed to the treatment of lymphohematopoietic malignancies [191]. It was then extended to other fields and gained wide usage in the early 2000s [192]. TR has developed as a discipline from cross-talk among different scientific specialties, such as clinical pharmacology, cell biology, genetics, chemistry and physiology [193]. In other words, it can be considered an interdisciplinary branch of the biomedical field, supported by three main pillars: benchside, bedside and community, as stated in 2015 by the European Society for Translational Medicine [194]. In this context, patient participation is fundamental.

The principal goals of TR are not only to explore diagnostic and therapeutic interventions and facilitate their application in clinical practice but also to develop a scientific translational research approach to improve human health and longevity, considering the global increase in life expectancy. TR is a bidirectional concept, encompassing not only bench-to-bedside factors (testing new therapeutic strategies developed through research) but also bedside-to-bench factors, which provide feedback about the applications of new treatments and how they can be improved.

In 2003, TR was described as a two-phase process of research, which started from basic science to arriving at public health through clinical science [195,196,197]. Some years ago, a systematic analysis led to the identification of four different stages of TR: T1 is the phase of basic knowledge or basic scientific discovery, in which new methods of diagnosis, treatment and prevention are considered. It starts with basic research and ends with early testing in humans; the T2 phase consists of controlled studies to determine the effectiveness in humans and in establishing clinical guidelines; T3 is the phase of delivery, dissemination, and diffusion research to move evidence-based guidelines into health practice; the T4 phase regards outcomes and effectiveness research to determine the true benefit to the community. It ultimately leads to clinical practices. After a systematic literature review, in 2017, an extra phase, T0, was proposed. It is the phase of basic research in which cell and animal models are used and mechanisms, targets and lead molecules are defined. It is also described as the phase of approaches to health problems [198]. All the phases are strictly connected to each other.

Starting from this description, it is useful to consider a translational science spectrum in which each TR stage is based on and strictly connected to the others, with patient involvement always as a key factor (Fig. 2).

Fig. 2
figure 2

Spectrum showing the interconnections among the stages of translational research, with patient involvement in all the stages. Created with https://BioRender.com

Today, TR, also considered translational science or translational medicine, is an established concept in the medical field and is recognized by the scientific community, research councils and governments. The number of publications referring to “translational medicine” has noticeably increased in the last fifteen years [198, 199], indicative of a wider interest from the medical and scientific network.

The COVID-19 pandemic has presented itself as a case where the translational process has been fully exploited. Preclinical and clinical development, as well as production activities, were planned in parallel, and the global research community made great scientific progress in record time. Experts with necessary and complementary skills, knowledge and experience worked together and created a very collaborative network. Many randomized controlled trials have been proposed and used for confirmed or suspected COVID-19 patients, revealing the possibility of a new integrated future of healthcare and clinical research.

Another aspect to consider during the emergence of the COVID-19 pandemic was the complexity and rising cost of conducting cancer clinical trials. It was necessary to reevaluate all the procedures and to adapt them to the emergent situation. A great challenge was to allow patients continued access to their experimental cancer therapies, ensuring both efficacy and safety.

The adaptation to the new situation paved the way for incorporating changes and novel flexible workflows within clinical trials and for setting more patient-centered clinical trials to easily collect information regarding the effects of new anticancer therapies. The medical and scientific communities now have the opportunity to use these necessitated novel strategies to design a more patient-focused approach to improve impact and efficiency [200, 201].

However, much work is still needed before TR can be incorporated it into everyday health practice and used to achieve optimal patient care and treatment. In this way personalized therapies will provide efficient and cost-effective healthcare. In the future, understanding science and utilizing scientific knowledge will be even more important than they are today [202]. Artificial intelligence, genetically based patient care, use of novel molecular biomarkers and drug testing on organoids/spheroids grown from patient cells will likely be an integral part of healthcare. For example, several institutes and laboratories for metabolism and diabetes already use metabolomics, genomics, and proteomics technologies to combine basic and clinical research [203]. Such a similar approach allows the introduction of new useful biomarkers for the subclassification and management of diabetes in clinical practice [204].

Clinical results can be implemented for the benefit of the patient, even in other fields. The incorporation of new three-dimensional (3D) simulation techniques is highly recommended in orthopedics for articular cartilage repair and regeneration [205] and represents an example of a translational approach in which basic science has a profound effect on clinical practice.

3D simulation, and computer science in general, allows us to obtain image data of anatomical structures of the body, also from the most complex, and helps surgeons to address these problems in depth. This approach translates current discoveries into clinical benefits. Imaging, together with new technologies, such as proteomic, transcriptomic, and metabolomic analyses, can also be used for translational research in bone tumors [206]. In recent years, regenerative and translational dentistry has also been developing to evaluate different new approaches for the implementation of protocols and strategies [207]. TR and precision medicine are also facilitated by the generation of tissue biobanking. Today, biobanking in health care is organized in dynamic units and is extremely useful for modern medical research, for example, in the field of cardiovascular diseases. The use of human biobanks allows us to overcome the limitations related to the intrinsic anatomical, functional and molecular differences in tissue samples that must be considered when animal species are employed [208].

For the examples we have reported and for several other similar reasons, continuous collaborations among universities, hospitals and academies as well as renewed and reviewed healthcare strategies must always be arranged. Continual communication between researchers in shared and different disciplines is fundamental to accelerating scientific progress. It is particularly beneficial when scientific exchanges are multidisciplinary.

3D culture systems and translational research

In vitro 2D cell cultures are still used in several laboratories throughout the world to study, for example, proliferation and/or differentiation. Their use presents the important advantage of being a standardized procedure. However, they also have some disadvantages. For example, the use of fetal bovine serum as an adjuvant can lead to cellular senescence and aging. Furthermore, this kind of model does not always represent the original tissue. For these reasons, 2D cultures are often replaced by 3D culture systems, which more closely resemble in vivo conditions.

Starting from these considerations, the new culture models represent robust and translational experimental systems that could have, in the near future, clinical application, favoring the creation of ad personam therapy.

Spheroids and organoids can be used for bench-to-bedside translation in several research fields, such as tissue engineering and regeneration, cancer cell biology, stem cell research, regenerative therapies, drug testing, and preclinical/clinical models for the treatment of different disorders and pathologies [188, 209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226,227,228,229,230,231,232]. Some examples of the possible applications of 3D multipotent mesenchymal stem cell spheroids are reported in Fig. 3.

Fig. 3
figure 3

Therapeutic potential of 3D multipotent mesenchymal stem cell spheroids and their application in translational medicine. Created with https://BioRender.com

The translation of these findings to obesity and spheroid models is related to the fact that adipose tissue is considered an endocrine organ and obesity is considered an inflammatory chronic disease.

Interestingly, the production of adipose spheroids from the SVF of mouse interscapular adipose tissue has already been reported. After the adipogenic protocol, spheroids had a complex internal structure with fully differentiated adipocytes and are viable, leptin-secreting, and lipolysis-responsive. They expressed the same adipocyte markers as adult mouse adipose tissue, and without an adipogenic stimulus, they maintained leptin secretion and were able to respond to adrenergic lipolytic stimulation by secreting glycerol. It has been reported that after implantation in mouse models, they could sustain leptin production [167].

The biological differences among WAT and SAT depots, leading to functional differences and to a more frequent onset of obesity-induced metabolic complications and comorbidities in visceral adipose tissue, are due not only to differences in their SVF composition but also to the presence of specific adipocyte stem cells and progenitor cells. Specialized adipocyte subtypes are derived from these cells [233,234,235,236] and must be selected for different purposes.

Another aspect to be considered is that some differences may exist not only because of different depot-targeted human tissue samples, as we have described above but also because hASCs can be derived from different sexes or different species. The first aspect may be useful for studying and evaluating sex-related biological differences in WAT expansion and obesity, and the second could be a limitation in establishing obesity cell models derived from species other than humans, limiting the translation of obesity-related findings to humans.

Therefore, experimental systems engineered with human-derived cells with depot-specific profiles are extremely useful for studying every aspect of the physiology and pathology of adipose tissue [166] and developing novel therapies.

However, some limitations must be considered. First, costs, absence of automation, diverse laboratory protocols, poor reproducibility, and limited scale of production are often associated with these procedures. Moreover, new therapies and technologies, chosen according to specific requirements, can be developed and employed only if regulated by approved guidelines to avoid harming the intended beneficiaries, that is, the patients and the public in general.

In terms of modern medicine, recent advances in adipose tissue 3D models (spheroids or organoids, more complex structures) may contribute to a deep understanding of the mechanisms involved in the prevention and treatment of obesity and related disorders (diabetes, metabolic syndrome, nonalcoholic fatty liver disease, cancer, etc.) in humans, reducing the preclinical evaluation process and improving predictive accuracy. Such an approach might also contribute to drug discovery for safe and effective personalized therapy, helping with translation to the pharmaceutical industry and providing efficient and cost-effective healthcare.

Conclusions

The prevalence of obesity is increasing at an alarming rate worldwide, leading to a major increase in the incidence of severe obesity-related conditions such as type 2 diabetes, ischemic heart disease, stroke, and many forms of cancer. The use of different types of study models is crucial for comprehending the pathophysiology of adipose organ and effectively implementing therapeutic strategies to combat obesity, which is now acknowledged as a serious illness. Constant sharing of opinions, information, skills, data and ideas among various professional figures through effective communication and facilitating the flow of knowledge from the laboratory to the bedside, and vice versa, becomes essential to ensure a seamless transition from laboratory settings to clinics and industries. This continuum, with the patient as the focal point, will enable the integration of translational research into routine operations.

Data availability

All the information reported in this review can be found in the scientific literature and can be easily shared.

References

  1. Obesity and overweight [Internet]. WHO. [cited 2024 Jan 16]. https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight

  2. Pararasa C, Bailey CJ, Griffiths HR. Ageing, adipose tissue, fatty acids and inflammation. Biogerontology. 2015;16:235–48.

    Article  CAS  PubMed  Google Scholar 

  3. Pérez LM, Pareja-Galeano H, Sanchis-Gomar F, Emanuele E, Lucia A, Gálvez BG. Adipaging: ageing and obesity share biological hallmarks related to a dysfunctional adipose tissue. J Physiol. 2016;594:3187–207.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Johnson PR, Hirsch J. Cellularity of adipose depots in six strains of genetically obese mice. J Lipid Res. 1972;13:2–11.

    Article  CAS  PubMed  Google Scholar 

  5. Klyde BJ, Hirsch J. Isotopic labeling of DNA in rat adipose tissue: evidence for proliferating cells associated with mature adipocytes. J Lipid Res. 1979;20:691–704.

    Article  CAS  PubMed  Google Scholar 

  6. Cinti S. The adipose organ. Prostaglandins Leukot Essent Fat Acids. 2005;73:9–15.

    Article  CAS  Google Scholar 

  7. Cinti S. Adipose Organ Development and Remodeling. Compr Physiol. 2018;8:1357–431.

    Article  PubMed  Google Scholar 

  8. Roche AF, Sievogel RM, Chumlea WC, Webb P. Grading body fatness from limited anthropometric data. Am J Clin Nutr. 1981;34:2831–8.

    Article  CAS  PubMed  Google Scholar 

  9. Tchernof A, Després J-P. Pathophysiology of human visceral obesity: an update. Physiol Rev. 2013;93:359–404.

    Article  CAS  PubMed  Google Scholar 

  10. Gui S-Y, Wu K-J, Sun Y, Chen Y-N, Liang H-R, Liu W, et al. Traffic noise and adiposity: a systematic review and meta-analysis of epidemiological studies. Environ Sci Pollut Res Int. 2022;29:55707–27.

    Article  PubMed  Google Scholar 

  11. Boutari C, Mantzoros CS. A 2022 update on the epidemiology of obesity and a call to action: as its twin COVID-19 pandemic appears to be receding, the obesity and dysmetabolism pandemic continues to rage on. Metabolism. 2022;133:155217.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Upadhyay J, Farr O, Perakakis N, Ghaly W, Mantzoros C. Obesity as a disease. Med Clin North Am. 2018;102:13–33.

    Article  PubMed  Google Scholar 

  13. Polyzos SA, Mantzoros CS. Obesity: seize the day, fight the fat. Metabolism. 2019;92:1–5.

    Article  CAS  PubMed  Google Scholar 

  14. Singh GM, Danaei G, Farzadfar F, Stevens GA, Woodward M, Wormser D, et al. The age-specific quantitative effects of metabolic risk factors on cardiovascular diseases and diabetes: a pooled analysis. PLoS ONE. 2013;8:e65174.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Emerging Risk Factors Collaboration, Wormser D, Kaptoge S, Di Angelantonio E, Wood AM, Pennells L, et al. Separate and combined associations of body-mass index and abdominal adiposity with cardiovascular disease: collaborative analysis of 58 prospective studies. Lancet. 2011;377:1085–95.

    Article  Google Scholar 

  16. Elagizi A, Kachur S, Carbone S, Lavie CJ, Blair SN. A review of obesity, physical activity, and Cardiovascular Disease. Curr Obes Rep. 2020;9:571–81.

    Article  PubMed  Google Scholar 

  17. Kuang M, Lu S, Xie Q, Peng N, He S, Yu C, et al. Abdominal obesity phenotypes are associated with the risk of developing non-alcoholic fatty liver disease: insights from the general population. BMC Gastroenterol. 2022;22:311.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Lauby-Secretan B, Scoccianti C, Loomis D, Grosse Y, Bianchini F, Straif K, et al. Body fatness and Cancer–viewpoint of the IARC Working Group. N Engl J Med. 2016;375:794–8.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Barb D, Pazaitou-Panayiotou K, Mantzoros CS. Adiponectin: a link between obesity and cancer. Expert Opin Investig Drugs. 2006;15:917–31.

    Article  CAS  PubMed  Google Scholar 

  20. Purcell S, Hossain I, Evans B, Porter G, Richardson G, Ellsmere J. Morbid obesity and severe knee osteoarthritis: which should be treated first? J Gastrointest Surg. 2022;26:1388–93.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Bonsignore MR. Obesity and obstructive sleep apnea. Handb Exp Pharmacol. 2022;274:181–201.

    Article  CAS  PubMed  Google Scholar 

  22. Anstey KJ, Cherbuin N, Budge M, Young J. Body mass index in midlife and late-life as a risk factor for dementia: a meta-analysis of prospective studies. Obes Rev. 2011;12:e426–437.

    Article  CAS  PubMed  Google Scholar 

  23. Alford S, Patel D, Perakakis N, Mantzoros CS. Obesity as a risk factor for Alzheimer’s disease: weighing the evidence. Obes Rev. 2018;19:269–80.

    Article  CAS  PubMed  Google Scholar 

  24. Polyzos SA, Kountouras J, Mantzoros CS. Obesity and nonalcoholic fatty liver disease: from pathophysiology to therapeutics. Metabolism. 2019;92:82–97.

    Article  CAS  PubMed  Google Scholar 

  25. Pasquali R, Casanueva F, Haluzik M, van Hulsteijn L, Ledoux S, Monteiro MP, et al. European Society of Endocrinology Clinical Practice Guideline: endocrine work-up in obesity. Eur J Endocrinol. 2020;182:G1–32.

    Article  CAS  PubMed  Google Scholar 

  26. Wilding JPH. Endocrine testing in obesity. Eur J Endocrinol. 2020;182:C13–5.

    Article  CAS  PubMed  Google Scholar 

  27. Castorina S, Barresi V, Luca T, Privitera G, De Geronimo V, Lezoche G, et al. Gastric ghrelin cells in obese patients are hyperactive. Int J Obes (Lond). 2021;45:184–94.

    Article  CAS  PubMed  Google Scholar 

  28. Müller TD, Nogueiras R, Andermann ML, Andrews ZB, Anker SD, Argente J, et al. Ghrelin Mol Metab. 2015;4:437–60.

    Article  PubMed  Google Scholar 

  29. Poher A-L, Tschöp MH, Müller TD. Ghrelin regulation of glucose metabolism. Peptides. 2018;100:236–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372:425–32.

    Article  CAS  PubMed  Google Scholar 

  31. Schwartz MW, Peskind E, Raskind M, Boyko EJ, Porte D. Cerebrospinal fluid leptin levels: relationship to plasma levels and to adiposity in humans. Nat Med. 1996;2:589–93.

    Article  CAS  PubMed  Google Scholar 

  32. Funcke J-B, Scherer PE. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res. 2019;60:1648–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. J Clin Endocrinol Metab. 2004;89:2548–56.

    Article  CAS  PubMed  Google Scholar 

  34. Cypess AM. Reassessing human adipose tissue. N Engl J Med. 2022;386:768–79.

    Article  CAS  PubMed  Google Scholar 

  35. Pezzino S, Luca T, Castorina M, Puleo S, Latteri S, Castorina S. Role of Perturbated Hemostasis in MASLD and its correlation with Adipokines. Life (Basel). 2024;14:93.

    CAS  PubMed  Google Scholar 

  36. Friedman JM. The function of leptin in nutrition, weight, and physiology. Nutr Rev. 2002;60:85–7. S1-14; discussion S68-84.

    Article  Google Scholar 

  37. Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev. 2004;84:277–359.

    Article  CAS  PubMed  Google Scholar 

  38. Cinti S. UCP1 protein: the molecular hub of adipose organ plasticity. Biochimie. 2017;134:71–6.

    Article  CAS  PubMed  Google Scholar 

  39. Gavaldà-Navarro A, Villarroya J, Cereijo R, Giralt M, Villarroya F. The endocrine role of brown adipose tissue: an update on actors and actions. Rev Endocr Metab Disord. 2022;23:31–41.

    Article  PubMed  Google Scholar 

  40. Cinti S. Reversible physiological transdifferentiation in the adipose organ. Proc Nutr Soc. 2009;68:340–9.

    Article  PubMed  Google Scholar 

  41. Bartelt A, Heeren J. Adipose tissue browning and metabolic health. Nat Rev Endocrinol. 2014;10:24–36.

    Article  CAS  PubMed  Google Scholar 

  42. Abdullahi A, Jeschke MG. White Adipose tissue Browning: a double-edged Sword. Trends Endocrinol Metab. 2016;27:542–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Cinti S. Adipocyte differentiation and transdifferentiation: plasticity of the adipose organ. J Endocrinol Invest. 2002;25:823–35.

    Article  CAS  PubMed  Google Scholar 

  44. Barbatelli G, Murano I, Madsen L, Hao Q, Jimenez M, Kristiansen K, et al. The emergence of cold-induced brown adipocytes in mouse white fat depots is determined predominantly by white to brown adipocyte transdifferentiation. Am J Physiol Endocrinol Metab. 2010;298:E1244–1253.

    Article  CAS  PubMed  Google Scholar 

  45. De Matteis R, Lucertini F, Guescini M, Polidori E, Zeppa S, Stocchi V, et al. Exercise as a new physiological stimulus for brown adipose tissue activity. Nutr Metab Cardiovasc Dis. 2013;23:582–90.

    Article  PubMed  Google Scholar 

  46. Cypess AM, Weiner LS, Roberts-Toler C, Franquet Elía E, Kessler SH, Kahn PA, et al. Activation of human brown adipose tissue by a β3-adrenergic receptor agonist. Cell Metab. 2015;21:33–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Smorlesi A, Frontini A, Giordano A, Cinti S. The adipose organ: white-brown adipocyte plasticity and metabolic inflammation. Obes Rev. 2012;13(Suppl 2):83–96.

    Article  CAS  PubMed  Google Scholar 

  48. Kotzbeck P, Giordano A, Mondini E, Murano I, Severi I, Venema W, et al. Brown adipose tissue whitening leads to brown adipocyte death and adipose tissue inflammation. J Lipid Res. 2018;59:784–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Petrovic N, Walden TB, Shabalina IG, Timmons JA, Cannon B, Nedergaard J. Chronic peroxisome proliferator-activated receptor gamma (PPARgamma) activation of epididymally derived white adipocyte cultures reveals a population of thermogenically competent, UCP1-containing adipocytes molecularly distinct from classic brown adipocytes. J Biol Chem. 2010;285:7153–64.

    Article  CAS  PubMed  Google Scholar 

  50. Wu J, Boström P, Sparks LM, Ye L, Choi JH, Giang A-H, et al. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell. 2012;150:366–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ikeda K, Yamada T. UCP1 dependent and independent thermogenesis in Brown and Beige adipocytes. Front Endocrinol (Lausanne). 2020;11:498.

    Article  PubMed  Google Scholar 

  52. Cheng L, Wang J, Dai H, Duan Y, An Y, Shi L, et al. Brown and beige adipose tissue: a novel therapeutic strategy for obesity and type 2 diabetes mellitus. Adipocyte. 2021;10:48–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Pisani DF, Barquissau V, Chambard J-C, Beuzelin D, Ghandour RA, Giroud M, et al. Mitochondrial fission is associated with UCP1 activity in human brite/beige adipocytes. Mol Metab. 2018;7:35–44.

    Article  CAS  PubMed  Google Scholar 

  54. Cinti S. Transdifferentiation properties of adipocytes in the adipose organ. Am J Physiol Endocrinol Metab. 2009;297:E977–986.

    Article  CAS  PubMed  Google Scholar 

  55. Cinti S. Between brown and white: novel aspects of adipocyte differentiation. Ann Med. 2011;43:104–15.

    Article  PubMed  Google Scholar 

  56. Giordano A, Smorlesi A, Frontini A, Barbatelli G, Cinti S. White, brown and pink adipocytes: the extraordinary plasticity of the adipose organ. Eur J Endocrinol. 2014;170:R159–171.

    Article  CAS  PubMed  Google Scholar 

  57. Giordano A, Perugini J, Kristensen DM, Sartini L, Frontini A, Kajimura S, et al. Mammary alveolar epithelial cells convert to brown adipocytes in post-lactating mice. J Cell Physiol. 2017;232:2923–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Cinti F, Cinti S. The endocrine adipose organ: a System playing a Central Role in COVID-19. Cells. 2022;11:2109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112:1796–808.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112:1821–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, et al. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res. 2005;46:2347–55.

    Article  CAS  PubMed  Google Scholar 

  62. Giordano A, Murano I, Mondini E, Perugini J, Smorlesi A, Severi I, et al. Obese adipocytes show ultrastructural features of stressed cells and die of pyroptosis. J Lipid Res. 2013;54:2423–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Brestoff JR, Artis D. Immune regulation of metabolic homeostasis in health and disease. Cell. 2015;161:146–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Sartipy P, Loskutoff DJ. Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proc Natl Acad Sci U S A. 2003;100:7265–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Malavazos AE, Ermetici F, Coman C, Corsi MM, Morricone L, Ambrosi B. Influence of epicardial adipose tissue and adipocytokine levels on cardiac abnormalities in visceral obesity. Int J Cardiol. 2007;121:132–4.

    Article  PubMed  Google Scholar 

  66. Iacobellis G. Epicardial adipose tissue in contemporary cardiology. Nat Rev Cardiol. 2022;19:593–606.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Ahmadieh S, Kim HW, Weintraub NL. Potential role of perivascular adipose tissue in modulating atherosclerosis. Clin Sci (Lond). 2020;134:3–13.

    Article  CAS  PubMed  Google Scholar 

  68. Cheng CK, Bakar HA, Gollasch M, Huang Y. Perivascular adipose tissue: the Sixth Man of the Cardiovascular System. Cardiovasc Drugs Ther. 2018;32:481–502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Kotanidis CP, Antoniades C. Perivascular fat imaging by computed tomography (CT): a virtual guide. Br J Pharmacol. 2021;178:4270–90.

    Article  CAS  PubMed  Google Scholar 

  70. Gil-Ortega M, Somoza B, Huang Y, Gollasch M, Fernández-Alfonso MS. Regional differences in perivascular adipose tissue impacting vascular homeostasis. Trends Endocrinol Metab. 2015;26:367–75.

    Article  CAS  PubMed  Google Scholar 

  71. Cai M, Zhao D, Han X, Han S, Zhang W, Zang Z, et al. The role of perivascular adipose tissue-secreted adipocytokines in cardiovascular disease. Front Immunol. 2023;14:1271051.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Lu C, Su L-Y, Lee RMKW, Gao Y-J. Alterations in perivascular adipose tissue structure and function in hypertension. Eur J Pharmacol. 2011;656:68–73.

    Article  CAS  PubMed  Google Scholar 

  73. Verhagen SN, Visseren FLJ. Perivascular adipose tissue as a cause of atherosclerosis. Atherosclerosis. 2011;214:3–10.

    Article  CAS  PubMed  Google Scholar 

  74. Malavazos AE, Di Vincenzo A, Iacobellis G, Basilico S, Dubini C, Morricone L, et al. The density of crown-like structures in epicardial adipose tissue could play a role in cardiovascular diseases. Eat Weight Disord. 2022;27:2905–10.

    Article  PubMed  Google Scholar 

  75. Quail DF, Dannenberg AJ. The obese adipose tissue microenvironment in cancer development and progression. Nat Rev Endocrinol. 2019;15:139–54.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Yang Z-H, Chen F-Z, Zhang Y-X, Ou M-Y, Tan P-C, Xu X-W, et al. Therapeutic targeting of white adipose tissue metabolic dysfunction in obesity: mechanisms and opportunities. MedComm. 2020;2024(5):e560.

    Google Scholar 

  77. Mahdi T, Hänzelmann S, Salehi A, Muhammed SJ, Reinbothe TM, Tang Y, et al. Secreted frizzled-related protein 4 reduces insulin secretion and is overexpressed in type 2 diabetes. Cell Metab. 2012;16:625–33.

    Article  CAS  PubMed  Google Scholar 

  78. Ohlson LO, Larsson B, Svärdsudd K, Welin L, Eriksson H, Wilhelmsen L, et al. The influence of body fat distribution on the incidence of diabetes mellitus. 13.5 years of follow-up of the participants in the study of men born in 1913. Diabetes. 1985;34:1055–8.

    Article  CAS  PubMed  Google Scholar 

  79. Oliveros E, Somers VK, Sochor O, Goel K, Lopez-Jimenez F. The concept of normal weight obesity. Prog Cardiovasc Dis. 2014;56:426–33.

    Article  PubMed  Google Scholar 

  80. Camastra S, Vitali A, Anselmino M, Gastaldelli A, Bellini R, Berta R, et al. Muscle and adipose tissue morphology, insulin sensitivity and beta-cell function in diabetic and nondiabetic obese patients: effects of bariatric surgery. Sci Rep. 2017;7:9007.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Björntorp P. Metabolic abnormalities in visceral obesity. Ann Med. 1992;24:3–5.

    Article  PubMed  Google Scholar 

  82. Murano I, Barbatelli G, Parisani V, Latini C, Muzzonigro G, Castellucci M, et al. Dead adipocytes, detected as crown-like structures, are prevalent in visceral fat depots of genetically obese mice. J Lipid Res. 2008;49:1562–8.

    Article  CAS  PubMed  Google Scholar 

  83. Sun K, Tordjman J, Clément K, Scherer PE. Fibrosis and adipose tissue dysfunction. Cell Metab. 2013;18:470–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Khan T, Muise ES, Iyengar P, Wang ZV, Chandalia M, Abate N, et al. Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol Cell Biol. 2009;29:1575–91.

    Article  CAS  PubMed  Google Scholar 

  85. DeBari MK, Abbott RD. Adipose tissue fibrosis: mechanisms, models, and Importance. Int J Mol Sci. 2020;21:6030.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Marcelin G, Silveira ALM, Martins LB, Ferreira AV, Clément K. Deciphering the cellular interplays underlying obesity-induced adipose tissue fibrosis. J Clin Invest. 2019;129:4032–40.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Divoux A, Clément K. Architecture and the extracellular matrix: the still unappreciated components of the adipose tissue. Obes Rev. 2011;12:e494–503.

    Article  CAS  PubMed  Google Scholar 

  88. Rosen ED, Spiegelman BM. What we talk about when we talk about fat. Cell. 2014;156:20–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Carobbio S, Pellegrinelli V, Vidal-Puig A. Adipose tissue function and expandability as determinants of Lipotoxicity and the metabolic syndrome. Adv Exp Med Biol. 2017;960:161–96.

    Article  CAS  PubMed  Google Scholar 

  90. Kusminski CM, Bickel PE, Scherer PE. Targeting adipose tissue in the treatment of obesity-associated diabetes. Nat Rev Drug Discov. 2016;15:639–60.

    Article  CAS  PubMed  Google Scholar 

  91. Williams DM, Nawaz A, Evans M. Drug therapy in obesity: a review of current and emerging treatments. Diabetes Ther. 2020;11:1199–216.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Sugihara H, Yonemitsu N, Miyabara S, Yun K. Primary cultures of unilocular fat cells: characteristics of growth in vitro and changes in differentiation properties. Differentiation. 1986;31:42–9.

    Article  CAS  PubMed  Google Scholar 

  93. Sugihara H, Yonemitsu N, Miyabara S, Toda S. Proliferation of unilocular fat cells in the primary culture. J Lipid Res. 1987;28:1038–45.

    Article  CAS  PubMed  Google Scholar 

  94. Baganha F, Schipper R, Hagberg CE. Towards better models for studying human adipocytes in vitro. Adipocyte. 2022;11:413–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Dufau J, Shen JX, Couchet M, De Castro Barbosa T, Mejhert N, Massier L, et al. In vitro and ex vivo models of adipocytes. Am J Physiol Cell Physiol. 2021;320:C822–41.

    Article  CAS  PubMed  Google Scholar 

  96. Harms MJ, Li Q, Lee S, Zhang C, Kull B, Hallen S, et al. Mature human White adipocytes cultured under membranes maintain identity, function, and can transdifferentiate into Brown-like adipocytes. Cell Rep. 2019;27:213–e2255.

    Article  CAS  PubMed  Google Scholar 

  97. Smith U. Morphologic studies of human subcutaneous adipose tissue in vitro. Anat Rec. 1971;169:97–104.

    Article  CAS  PubMed  Google Scholar 

  98. Smith U, Jacobsson B. Studies of human adipose tissue in culture. II. Effects of insulin and of medium glucose on lipolysis and cell size. Anat Rec. 1973;176:181–3.

    Article  CAS  PubMed  Google Scholar 

  99. Gealekman O, Guseva N, Hartigan C, Apotheker S, Gorgoglione M, Gurav K, et al. Depot-specific differences and insufficient subcutaneous adipose tissue angiogenesis in human obesity. Circulation. 2011;123:186–94.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Moro C, Klimcakova E, Lolmède K, Berlan M, Lafontan M, Stich V, et al. Atrial natriuretic peptide inhibits the production of adipokines and cytokines linked to inflammation and insulin resistance in human subcutaneous adipose tissue. Diabetologia. 2007;50:1038–47.

    Article  CAS  PubMed  Google Scholar 

  101. Gesta S, Lolmède K, Daviaud D, Berlan M, Bouloumié A, Lafontan M, et al. Culture of human adipose tissue explants leads to profound alteration of adipocyte gene expression. Horm Metab Res. 2003;35:158–63.

    Article  CAS  PubMed  Google Scholar 

  102. Huang B, Zhao H, Huang C, Wu L, Xiang L, Chen J, et al. CMKLR1 deficiency attenuates androgen-induced lipid accumulation in mice. Am J Physiol Endocrinol Metab. 2020;318:E371–80.

    Article  PubMed  Google Scholar 

  103. Lau FH, Vogel K, Luckett JP, Hunt M, Meyer A, Rogers CL, et al. Sandwiched White Adipose tissue: a Microphysiological System of Primary Human adipose tissue. Tissue Eng Part C Methods. 2018;24:135–45.

    Article  PubMed  Google Scholar 

  104. Park Y-J, Kim H-Y, Shin S, Lee J, Heo I, Cha Y-Y, et al. Anti-obesity effect of Lythri herba water extracts in vitro and in vivo. J Ethnopharmacol. 2023;317:116789.

    Article  CAS  PubMed  Google Scholar 

  105. Tsai S-C, Huang Y-W, Wu C-C, Wang J-J, Chen Y-T, Singhania RR, et al. Anti-obesity Effect of Nostoc commune ethanol Extract in Vitro and in vivo. Nutrients. 2022;14:968.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Green H, Kehinde O. An established preadipose cell line and its differentiation in culture. II. Factors affecting the adipose conversion. Cell. 1975;5:19–27.

    Article  CAS  PubMed  Google Scholar 

  107. Russell TR, Ho R. Conversion of 3T3 fibroblasts into adipose cells: triggering of differentiation by prostaglandin F2alpha and 1-methyl-3-isobutyl xanthine. Proc Natl Acad Sci U S A. 1976;73:4516–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Zebisch K, Voigt V, Wabitsch M, Brandsch M. Protocol for effective differentiation of 3T3-L1 cells to adipocytes. Anal Biochem. 2012;425:88–90.

    Article  CAS  PubMed  Google Scholar 

  109. 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;88:889–94.

    Article  CAS  PubMed  Google Scholar 

  110. Cawthorn WP, Scheller EL, MacDougald OA. Adipose tissue stem cells meet preadipocyte commitment: going back to the future. J Lipid Res. 2012;53:227–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Pu Y, Veiga-Lopez A. PPARγ agonist through the terminal differentiation phase is essential for adipogenic differentiation of fetal ovine preadipocytes. Cell Mol Biol Lett. 2017;22:6.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Guo PP, Yao XR, Xu YN, Jin X, Li Q, Yan CG, et al. Insulin interacts with PPARγ agonists to promote bovine adipocyte differentiation. Domest Anim Endocrinol. 2024;88:106848.

    Article  CAS  PubMed  Google Scholar 

  113. MacDougald OA, Lane MD. Transcriptional regulation of gene expression during adipocyte differentiation. Annu Rev Biochem. 1995;64:345–73.

    Article  CAS  PubMed  Google Scholar 

  114. Ferrero R, Rainer P, Deplancke B. Toward a Consensus View of mammalian adipocyte stem and progenitor cell heterogeneity. Trends Cell Biol. 2020;30:937–50.

    Article  CAS  PubMed  Google Scholar 

  115. Frazier T, Lee S, Bowles A, Semon J, Bunnell B, Wu X, et al. Gender and age-related cell compositional differences in C57BL/6 murine adipose tissue stromal vascular fraction. Adipocyte. 2018;7:183–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Shan B, Barker CS, Shao M, Zhang Q, Gupta RK, Wu Y. Multilayered omics reveal sex- and depot-dependent adipose progenitor cell heterogeneity. Cell Metab. 2022;34:783–e7997.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Wang T, Sharma AK, Wolfrum C. Novel insights into adipose tissue heterogeneity. Rev Endocr Metab Disord. 2022;23:5–12.

    Article  PubMed  Google Scholar 

  118. Liao X, Zhou H, Deng T. The composition, function, and regulation of adipose stem and progenitor cells. J Genet Genomics. 2022;49:308–15.

    Article  CAS  PubMed  Google Scholar 

  119. Sarantopoulos CN, Banyard DA, Ziegler ME, Sun B, Shaterian A, Widgerow AD. Elucidating the Preadipocyte and its role in adipocyte formation: a Comprehensive Review. Stem Cell Rev Rep. 2018;14:27–42.

    Article  CAS  PubMed  Google Scholar 

  120. Rodriguez A-M, Elabd C, Delteil F, Astier J, Vernochet C, Saint-Marc P, et al. Adipocyte differentiation of multipotent cells established from human adipose tissue. Biochem Biophys Res Commun. 2004;315:255–63.

    Article  CAS  PubMed  Google Scholar 

  121. Rodriguez A-M, Pisani D, Dechesne CA, Turc-Carel C, Kurzenne J-Y, Wdziekonski B, et al. Transplantation of a multipotent cell population from human adipose tissue induces dystrophin expression in the immunocompetent mdx mouse. J Exp Med. 2005;201:1397–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Green H, Meuth M. An established pre-adipose cell line and its differentiation in culture. Cell. 1974;3:127–33.

    Article  CAS  PubMed  Google Scholar 

  123. Green H, Kehinde O. Spontaneous heritable changes leading to increased adipose conversion in 3T3 cells. Cell. 1976;7:105–13.

    Article  CAS  PubMed  Google Scholar 

  124. Négrel R, Grimaldi P, Ailhaud G. Establishment of preadipocyte clonal line from epididymal fat pad of ob/ob mouse that responds to insulin and to lipolytic hormones. Proc Natl Acad Sci U S A. 1978;75:6054–8.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Dani C, Smith AG, Dessolin S, Leroy P, Staccini L, Villageois P, et al. Differentiation of embryonic stem cells into adipocytes in vitro. J Cell Sci. 1997;110(Pt 11):1279–85.

    Article  CAS  PubMed  Google Scholar 

  126. Reznikoff CA, Brankow DW, Heidelberger C. Establishment and characterization of a cloned line of C3H mouse embryo cells sensitive to postconfluence inhibition of division. Cancer Res. 1973;33:3231–8.

    CAS  PubMed  Google Scholar 

  127. Taylor SM, Jones PA. Multiple new phenotypes induced in 10T1/2 and 3T3 cells treated with 5-azacytidine. Cell. 1979;17:771–9.

    Article  CAS  PubMed  Google Scholar 

  128. Konieczny SF, Emerson CP. 5-Azacytidine induction of stable mesodermal stem cell lineages from 10T1/2 cells: evidence for regulatory genes controlling determination. Cell. 1984;38:791–800.

    Article  CAS  PubMed  Google Scholar 

  129. Tang Q-Q, Otto TC, Lane MD. Commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineage. Proc Natl Acad Sci U S A. 2004;101:9607–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Nakano T, Kodama H, Honjo T. Generation of lymphohematopoietic cells from embryonic stem cells in culture. Science. 1994;265:1098–101.

    Article  CAS  PubMed  Google Scholar 

  131. Wolins NE, Quaynor BK, Skinner JR, Tzekov A, Park C, Choi K, et al. OP9 mouse stromal cells rapidly differentiate into adipocytes: characterization of a useful new model of adipogenesis. J Lipid Res. 2006;47:450–60.

    Article  CAS  PubMed  Google Scholar 

  132. Tan YS, Lei YL. Generation and culture of mouse embryonic fibroblasts. Methods Mol Biol. 2019;1960:85–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Yagi K, Kondo D, Okazaki Y, Kano K. A novel preadipocyte cell line established from mouse adult mature adipocytes. Biochem Biophys Res Commun. 2004;321:967–74.

    Article  CAS  PubMed  Google Scholar 

  134. Matsumoto T, Kano K, Kondo D, Fukuda N, Iribe Y, Tanaka N, et al. Mature adipocyte-derived dedifferentiated fat cells exhibit multilineage potential. J Cell Physiol. 2008;215:210–22.

    Article  CAS  PubMed  Google Scholar 

  135. Shah M, George RL, Evancho-Chapman MM, Zhang G. Current challenges in dedifferentiated fat cells research. Organogenesis. 2016;12:119–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Hugo ER, Brandebourg TD, Comstock CES, Gersin KS, Sussman JJ, Ben-Jonathan N. LS14: a novel human adipocyte cell line that produces prolactin. Endocrinology. 2006;147:306–13.

    Article  CAS  PubMed  Google Scholar 

  137. Zilberfarb V, Piétri-Rouxel F, Jockers R, Krief S, Delouis C, Issad T, et al. Human immortalized brown adipocytes express functional beta3-adrenoceptor coupled to lipolysis. J Cell Sci. 1997;110(Pt 7):801–7.

    Article  CAS  PubMed  Google Scholar 

  138. Darimont C, Zbinden I, Avanti O, Leone-Vautravers P, Giusti V, Burckhardt P, et al. Reconstitution of telomerase activity combined with HPV-E7 expression allow human preadipocytes to preserve their differentiation capacity after immortalization. Cell Death Differ. 2003;10:1025–31.

    Article  CAS  PubMed  Google Scholar 

  139. Wabitsch M, Brenner RE, Melzner I, Braun M, Möller P, Heinze E, et al. Characterization of a human preadipocyte cell strain with high capacity for adipose differentiation. Int J Obes Relat Metab Disord. 2001;25:8–15.

    Article  CAS  PubMed  Google Scholar 

  140. Torii I, Morikawa S, Nakano A, Morikawa K. Establishment of a human preadipose cell line, HPB-AML-I: refractory to PPARgamma-mediated adipogenic stimulation. J Cell Physiol. 2003;197:42–52.

    Article  CAS  PubMed  Google Scholar 

  141. Skurk T, Hauner H. Primary culture of human adipocyte precursor cells: expansion and differentiation. Methods Mol Biol. 2012;806:215–26.

    Article  CAS  PubMed  Google Scholar 

  142. Hausman DB, Park HJ, Hausman GJ. Isolation and culture of preadipocytes from rodent white adipose tissue. Methods Mol Biol. 2008;456:201–19.

    Article  PubMed  Google Scholar 

  143. van Harmelen V, Skurk T, Hauner H. Primary culture and differentiation of human adipocyte precursor cells. Methods Mol Med. 2005;107:125–35.

    PubMed  Google Scholar 

  144. Thiam AR, Beller M. The why, when and how of lipid droplet diversity. J Cell Sci. 2017;130:315–24.

    Article  CAS  PubMed  Google Scholar 

  145. Yu J, Li P. The size matters: regulation of lipid storage by lipid droplet dynamics. Sci China Life Sci. 2017;60:46–56.

    Article  CAS  PubMed  Google Scholar 

  146. Cho C-H, Koh YJ, Han J, Sung H-K, Jong Lee H, Morisada T, et al. Angiogenic role of LYVE-1-positive macrophages in adipose tissue. Circ Res. 2007;100:e47–57.

    Article  CAS  PubMed  Google Scholar 

  147. Han J, Lee J-E, Jin J, Lim JS, Oh N, Kim K, et al. The spatiotemporal development of adipose tissue. Development. 2011;138:5027–37.

    Article  CAS  PubMed  Google Scholar 

  148. Louis F, Kitano S, Mano JF, Matsusaki M. 3D collagen microfibers stimulate the functionality of preadipocytes and maintain the phenotype of mature adipocytes for long term cultures. Acta Biomater. 2019;84:194–207.

    Article  CAS  PubMed  Google Scholar 

  149. Pang W-J, Yu T-Y, Bai L, Yang Y-J, Yang G-S. Tissue expression of porcine FoxO1 and its negative regulation during primary preadipocyte differentiation. Mol Biol Rep. 2009;36:165–76.

    Article  CAS  PubMed  Google Scholar 

  150. Suryawan A, Swanson LV, Hu CY. Insulin and hydrocortisone, but not triiodothyronine, are required for the differentiation of pig preadipocytes in primary culture. J Anim Sci. 1997;75:105–11.

    Article  CAS  PubMed  Google Scholar 

  151. Ghaben AL, Scherer PE. Adipogenesis and metabolic health. Nat Rev Mol Cell Biol. 2019;20:242–58.

    Article  CAS  PubMed  Google Scholar 

  152. Abbott A. Cell culture: biology’s new dimension. Nature. 2003;424:870–2.

    Article  CAS  PubMed  Google Scholar 

  153. Shen JX, Couchet M, Dufau J, de Castro Barbosa T, Ulbrich MH, Helmstädter M, et al. 3D adipose tissue Culture Links the Organotypic Microenvironment to Improved adipogenesis. Adv Sci (Weinh). 2021;8:e2100106.

    Article  PubMed  Google Scholar 

  154. Turner PA, Garrett MR, Didion SP, Janorkar AV. Spheroid Culture System confers differentiated Transcriptome Profile and functional advantage to 3T3-L1 adipocytes. Ann Biomed Eng. 2018;46:772–87.

    Article  PubMed  PubMed Central  Google Scholar 

  155. Turner PA, Gurumurthy B, Bailey JL, Elks CM, Janorkar AV. Adipogenic differentiation of human adipose-derived stem cells grown as spheroids. Process Biochem. 2017;59:312–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Pieters VM, Rjaibi ST, Singh K, Li NT, Khan ST, Nunes SS et al. A three-dimensional human adipocyte model of fatty acid-induced obesity. Biofabrication. 2022;14.

  157. Rajangam T, Park MH, Kim S-H. 3D human adipose-derived stem cell clusters as a model for in Vitro Fibrosis. Tissue Eng Part C Methods. 2016;22:679–90.

    Article  CAS  PubMed  Google Scholar 

  158. Loessner D, Rockstroh A, Shokoohmand A, Holzapfel BM, Wagner F, Baldwin J, et al. A 3D tumor microenvironment regulates cell proliferation, peritoneal growth and expression patterns. Biomaterials. 2019;190–191:63–75.

    Article  PubMed  Google Scholar 

  159. Doyle AD, Carvajal N, Jin A, Matsumoto K, Yamada KM. Local 3D matrix microenvironment regulates cell migration through spatiotemporal dynamics of contractility-dependent adhesions. Nat Commun. 2015;6:8720.

    Article  CAS  PubMed  Google Scholar 

  160. Sullivan WJ, Mullen PJ, Schmid EW, Flores A, Momcilovic M, Sharpley MS, et al. Extracellular matrix remodeling regulates glucose metabolism through TXNIP destabilization. Cell. 2018;175:117–e13221.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Sutherland RM, McCredie JA, Inch WR. Growth of multicell spheroids in tissue culture as a model of nodular carcinomas. J Natl Cancer Inst. 1971;46:113–20.

    CAS  PubMed  Google Scholar 

  162. Turner PA, Tang Y, Weiss SJ, Janorkar AV. Three-dimensional spheroid cell model of in vitro adipocyte inflammation. Tissue Eng Part A. 2015;21:1837–47.

    Article  CAS  PubMed  Google Scholar 

  163. Gurumurthy B, Bierdeman PC, Janorkar AV. Spheroid model for functional osteogenic evaluation of human adipose derived stem cells. J Biomed Mater Res A. 2017;105:1230–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Alhadlaq A, Tang M, Mao JJ. Engineered adipose tissue from human mesenchymal stem cells maintains predefined shape and dimension: implications in soft tissue augmentation and reconstruction. Tissue Eng. 2005;11:556–66.

    Article  CAS  PubMed  Google Scholar 

  165. O’Donnell BT, Al-Ghadban S, Ives CJ, L’Ecuyer MP, Monjure TA, Romero-Lopez M, et al. Adipose tissue-derived stem cells retain their adipocyte differentiation potential in three-Dimensional hydrogels and bioreactors †. Biomolecules. 2020;10:1070.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Mandl M, Viertler HP, Hatzmann FM, Brucker C, Großmann S, Waldegger P, et al. An organoid model derived from human adipose stem/progenitor cells to study adipose tissue physiology. Adipocyte. 2022;11:164–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Robledo F, González-Hodar L, Tapia P, Figueroa A-M, Ezquer F, Cortés V. Spheroids derived from the stromal vascular fraction of adipose tissue self-organize in complex adipose organoids and secrete leptin. Stem Cell Res Ther. 2023;14:70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Muller S, Ader I, Creff J, Leménager H, Achard P, Casteilla L, et al. Human adipose stromal-vascular fraction self-organizes to form vascularized adipose tissue in 3D cultures. Sci Rep. 2019;9:7250.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Ioannidou A, Alatar S, Schipper R, Baganha F, Åhlander M, Hornell A, et al. Hypertrophied human adipocyte spheroids as in vitro model of weight gain and adipose tissue dysfunction. J Physiol. 2022;600:869–83.

    Article  CAS  PubMed  Google Scholar 

  170. Fitzgerald SJ, Cobb JS, Janorkar AV. Comparison of the formation, adipogenic maturation, and retention of human adipose-derived stem cell spheroids in scaffold-free culture techniques. J Biomed Mater Res B Appl Biomater. 2020;108:3022–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Gugerell A, Neumann A, Kober J, Tammaro L, Hoch E, Schnabelrauch M, et al. Adipose-derived stem cells cultivated on electrospun l-lactide/glycolide copolymer fleece and gelatin hydrogels under flow conditions - aiming physiological reality in hypodermis tissue engineering. Burns. 2015;41:163–71.

    Article  PubMed  Google Scholar 

  172. Yang F, Carmona A, Stojkova K, Garcia Huitron EI, Goddi A, Bhushan A, et al. A 3D human adipose tissue model within a microfluidic device. Lab Chip. 2021;21:435–46.

    Article  CAS  PubMed  Google Scholar 

  173. Ryu N-E, Lee S-H, Park H. Spheroid Culture System methods and applications for mesenchymal stem cells. Cells. 2019;8:1620.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Labusca L, Herea DD, Minuti AE, Stavila C, Danceanu C, Grigoras M, et al. Magnetic nanoparticle loaded human adipose derived mesenchymal cells spheroids in levitated culture. J Biomed Mater Res B Appl Biomater. 2021;109:630–42.

    Article  CAS  PubMed  Google Scholar 

  175. Zhang S, Liu P, Chen L, Wang Y, Wang Z, Zhang B. The effects of spheroid formation of adipose-derived stem cells in a microgravity bioreactor on stemness properties and therapeutic potential. Biomaterials. 2015;41:15–25.

    Article  PubMed  Google Scholar 

  176. Kapur SK, Wang X, Shang H, Yun S, Li X, Feng G, et al. Human adipose stem cells maintain proliferative, synthetic and multipotential properties when suspension cultured as self-assembling spheroids. Biofabrication. 2012;4:025004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Frazier TP, Hamel K, Wu X, Rogers E, Lassiter H, Robinson J, et al. Adipose-derived cells: building blocks of three-dimensional microphysiological systems. Biomater Transl. 2021;2:301–6.

    PubMed  PubMed Central  Google Scholar 

  178. Yoon H, Seo JK, Park T-E. Microphysiological system recapitulating the pathophysiology of adipose tissue in obesity. Acta Biomater. 2023;159:188–200.

    Article  CAS  PubMed  Google Scholar 

  179. Doulberis M, Papaefthymiou A, Polyzos SA, Katsinelos P, Grigoriadis N, Srivastava DS, et al. Rodent models of obesity. Minerva Endocrinol. 2020;45:243–63.

    Article  PubMed  Google Scholar 

  180. Lutz TA, Woods SC. Overview of animal models of obesity. Curr Protoc Pharmacol. 2012;Chap. 5:Unit5.61.

  181. Vickers SP, Jackson HC, Cheetham SC. The utility of animal models to evaluate novel anti-obesity agents. Br J Pharmacol. 2011;164:1248–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Blüher M. Transgenic animal models for the study of adipose tissue biology. Best Pract Res Clin Endocrinol Metab. 2005;19:605–23.

    Article  PubMed  Google Scholar 

  183. Harwood HJ, Listrani P, Wagner JD. Nonhuman primates and other animal models in diabetes research. J Diabetes Sci Technol. 2012;6:503–14.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Havel PJ, Kievit P, Comuzzie AG, Bremer AA. Use and Importance of Nonhuman Primates in Metabolic Disease Research: current state of the field. ILAR J. 2017;58:251–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. McCarthy M, Brown T, Alarcon A, Williams C, Wu X, Abbott RD, et al. Fat-On-A-Chip Models for Research and Discovery in obesity and its metabolic comorbidities. Tissue Eng Part B Rev. 2020;26:586–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Petrenko Y, Syková E, Kubinová Š. The therapeutic potential of three-dimensional multipotent mesenchymal stromal cell spheroids. Stem Cell Res Ther. 2017;8:94.

    Article  PubMed  PubMed Central  Google Scholar 

  187. Liu J, Zhou L, Zhao F, Zhou C, Yang T, Xu Z, et al. Therapeutic effect of adipose stromal vascular fraction spheroids for partial bladder outlet obstruction induced underactive bladder. Stem Cell Res Ther. 2022;13:68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Sargenti A, Pasqua S, Leu M, Dionisi L, Filardo G, Grigolo B, et al. Adipose stromal cell spheroids for cartilage repair: a Promising Tool for unveiling the critical maturation point. Bioeng (Basel). 2023;10:1182.

    CAS  Google Scholar 

  189. Bender R, McCarthy M, Brown T, Bukowska J, Smith S, Abbott RD, et al. Human adipose derived cells in two- and three-dimensional cultures: functional validation of an in vitro Fat Construct. Stem Cells Int. 2020;2020:4242130.

    Article  PubMed  PubMed Central  Google Scholar 

  190. Cokkinos DV. Introduction: What Is Translational Research. In: Cokkinos DV, editor. Introduction to Translational Cardiovascular Research [Internet]. Cham: Springer International Publishing; 2015 [cited 2024 Feb 8]. pp. 3–16. https://doi.org/10.1007/978-3-319-08798-6_1

  191. Karp JE, McCaffrey RP. New avenues of translational research in leukemia and lymphoma: outgrowth of a Leukemia Society of America-National Cancer Institute workshop. J Natl Cancer Inst. 1994;86:1196–201.

    Article  CAS  PubMed  Google Scholar 

  192. Lee W-H. Translational medical research: present and future. Am J Transl Res. 2009;1:99–100.

    PubMed  PubMed Central  Google Scholar 

  193. Woolf SH. The meaning of translational research and why it matters. JAMA. 2008;299:211–3.

    Article  CAS  PubMed  Google Scholar 

  194. Cohrs RJ, Martin T, Ghahramani P, Bidaut L, Higgins PJ, Shahzad A. Translational Medicine definition by the European Society for Translational Medicine. New Horizons Translational Med. 2015;2:86–8.

    Google Scholar 

  195. Public Confidence and Involvement in Clinical Research: Symposium Summary, Clinical Roundtable, September 2000 [Internet], Washington DC. National Academies Press; 2001 [cited 2024 Feb 12]. http://www.nap.edu/catalog/10155

  196. Exploring the Map of Clinical Research for the Coming Decade: Symposium Summary, Clinical Roundtable, December 2000 [Internet]. Washington, D.C.: National Academies Press. 2001 [cited 2024 Feb 12]. http://www.nap.edu/catalog/10156

  197. Sung NS, Crowley WF, Genel M, Salber P, Sandy L, Sherwood LM, et al. Central challenges facing the national clinical research enterprise. JAMA. 2003;289:1278–87.

    Article  PubMed  Google Scholar 

  198. Fort DG, Herr TM, Shaw PL, Gutzman KE, Starren JB. Mapping the evolving definitions of translational research. J Clin Transl Sci. 2017;1:60–6.

    Article  PubMed  PubMed Central  Google Scholar 

  199. Austin CP. Opportunities and challenges in translational science. Clin Transl Sci. 2021;14:1629–47.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Flaherty KT, Doroshow JH, Galbraith S, Ribas A, Kluetz PG, Pazdur R, et al. Rethinking Cancer Clinical Trial Conduct Induced by COVID-19: an academic Center, Industry, Government, and Regulatory Agency Perspective. Cancer Discov. 2021;11:1881–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Samimi G, House M, Benante K, Bengtson L, Budd T, Dermody B, et al. Lessons learned from the impact of COVID-19 on NCI-sponsored Cancer Prevention clinical trials: moving toward participant-centric study designs. Cancer Prev Res (Phila). 2022;15:279–84.

    Article  CAS  PubMed  Google Scholar 

  202. Hegyi P, Petersen OH, Holgate S, Erőss B, Garami A, Szakács Z, et al. Academia europaea position paper on Translational Medicine: the cycle model for translating scientific results into community benefits. J Clin Med. 2020;9:1532.

    Article  PubMed  PubMed Central  Google Scholar 

  203. Sen S, Chakraborty R. Treatment and diagnosis of diabetes Mellitus and its complication: Advanced approaches. Mini Rev Med Chem. 2015;15:1132–3.

    Article  CAS  PubMed  Google Scholar 

  204. Zhu M, Wu J, Gao J. Exosomes for diabetes syndrome: ongoing applications and perspective. Biomater Sci. 2022;10:2154–71.

    Article  CAS  PubMed  Google Scholar 

  205. Mediouni M. The what, where and why of Orthopaedic Simulation. Int J Surg Res Pract. 2015;2.

  206. Rossi M, Del Fattore A. Molecular and Translational Research on Bone tumors. Int J Mol Sci. 2023;24:1946.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Tatullo M, Codispoti B, Paduano F, Nuzzolese M, Makeeva I. Strategic Tools in Regenerative and Translational Dentistry. Int J Mol Sci. 2019;20:1879.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Zhu Y, Jackson D, Hunter B, Beattie L, Turner L, Hambly BD, et al. Models of cardiovascular surgery biobanking to facilitate translational research and precision medicine. ESC Heart Fail. 2022;9:21–30.

    Article  PubMed  Google Scholar 

  209. Roberto de Barros N, Wang C, Maity S, Peirsman A, Nasiri R, Herland A, et al. Engineered organoids for biomedical applications. Adv Drug Deliv Rev. 2023;203:115142.

    Article  CAS  PubMed  Google Scholar 

  210. Dutta D, Heo I, Clevers H. Disease modeling in stem cell-derived 3D Organoid systems. Trends Mol Med. 2017;23:393–410.

    Article  CAS  PubMed  Google Scholar 

  211. Wang H, Brown PC, Chow ECY, Ewart L, Ferguson SS, Fitzpatrick S, et al. 3D cell culture models: drug pharmacokinetics, safety assessment, and regulatory consideration. Clin Transl Sci. 2021;14:1659–80.

    Article  PubMed  PubMed Central  Google Scholar 

  212. Ong CS, Zhou X, Han J, Huang CY, Nashed A, Khatri S, et al. In vivo therapeutic applications of cell spheroids. Biotechnol Adv. 2018;36:494–505.

    Article  CAS  PubMed  Google Scholar 

  213. Gupta N, Dilmen E, Morizane R. 3D kidney organoids for bench-to-bedside translation. J Mol Med (Berl). 2021;99:477–87.

    Article  PubMed  Google Scholar 

  214. Tekguc M, Gaal RCV, Uzel SGM, Gupta N, Riella LV, Lewis JA, et al. Kidney organoids: a pioneering model for kidney diseases. Transl Res. 2022;250:1–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Miyoshi T, Hiratsuka K, Saiz EG, Morizane R. Kidney organoids in translational medicine: Disease modeling and regenerative medicine. Dev Dyn. 2020;249:34–45.

    Article  CAS  PubMed  Google Scholar 

  216. Kondo J, Inoue M. Application of Cancer Organoid Model for Drug Screening and Personalized Therapy. Cells. 2019;8:470.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Castro F, Leite Pereira C, Helena Macedo M, Almeida A, José Silveira M, Dias S, et al. Advances on colorectal cancer 3D models: the needed translational technology for nanomedicine screening. Adv Drug Deliv Rev. 2021;175:113824.

    Article  CAS  PubMed  Google Scholar 

  218. Lupo F, Piro G, Torroni L, Delfino P, Trovato R, Rusev B, et al. Organoid-Transplant Model systems to study the effects of obesity on the pancreatic carcinogenesis in vivo. Front Cell Dev Biol. 2020;8:308.

    Article  PubMed  PubMed Central  Google Scholar 

  219. Ebner-Peking P, Krisch L, Wolf M, Hochmann S, Hoog A, Vári B, et al. Self-assembly of differentiated progenitor cells facilitates spheroid human skin organoid formation and planar skin regeneration. Theranostics. 2021;11:8430–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Nugraha B, Buono MF, von Boehmer L, Hoerstrup SP, Emmert MY. Human Cardiac Organoids for Disease modeling. Clin Pharmacol Ther. 2019;105:79–85.

    Article  PubMed  Google Scholar 

  221. Figtree GA, Bubb KJ, Tang O, Kizana E, Gentile C. Vascularized Cardiac spheroids as Novel 3D in vitro models to study Cardiac Fibrosis. Cells Tissues Organs. 2017;204:191–8.

    Article  CAS  PubMed  Google Scholar 

  222. Richards DJ, Li Y, Kerr CM, Yao J, Beeson GC, Coyle RC, et al. Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat Biomed Eng. 2020;4:446–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Ingelman-Sundberg M, Lauschke VM. 3D human liver spheroids for translational pharmacology and toxicology. Basic Clin Pharmacol Toxicol. 2022;130(Suppl 1):5–15.

    Article  CAS  PubMed  Google Scholar 

  224. Rose S, Cuvellier M, Ezan F, Carteret J, Bruyère A, Legagneux V, et al. DMSO-free highly differentiated HepaRG spheroids for chronic toxicity, liver functions and genotoxicity studies. Arch Toxicol. 2022;96:243–58.

    Article  CAS  PubMed  Google Scholar 

  225. Hurrell T, Kastrinou-Lampou V, Fardellas A, Hendriks DFG, Nordling Å, Johansson I, et al. Human Liver spheroids as a model to study aetiology and treatment of hepatic fibrosis. Cells. 2020;9:964.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Mannaerts I, Eysackers N, Anne van Os E, Verhulst S, Roosens T, Smout A, et al. The fibrotic response of primary liver spheroids recapitulates in vivo hepatic stellate cell activation. Biomaterials. 2020;261:120335.

    Article  CAS  PubMed  Google Scholar 

  227. Sariyar E, Firtina Karagonlar Z. Modelling the Sorafenib-resistant Liver Cancer Microenvironment by using 3-D spheroids. Altern Lab Anim. 2023;51:301–12.

    Article  PubMed  Google Scholar 

  228. Lee SY, Lee JW. 3D spheroid cultures of stem cells and Exosome applications for cartilage repair. Life (Basel). 2022;12:939.

    CAS  PubMed  Google Scholar 

  229. Muscolino E, Di Stefano AB, Trapani M, Sabatino MA, Giacomazza D, Moschella F, et al. Injectable xyloglucan hydrogels incorporating spheroids of adipose stem cells for bone and cartilage regeneration. Mater Sci Eng C Mater Biol Appl. 2021;131:112545.

    Article  CAS  PubMed  Google Scholar 

  230. Tu C-C, Zheng Z-C, Cheng N-C, Yu J, Tai W-C, Pan Y-X et al. Alveolar mucosal cell spheroids promote extraction socket healing and osseous defect regeneration. J Periodontol. 2023.

  231. Imamura A, Kajiya H, Fujisaki S, Maeshiba M, Yanagi T, Kojima H, et al. Three-dimensional spheroids of mesenchymal stem/stromal cells promote osteogenesis by activating stemness and Wnt/β-catenin. Biochem Biophys Res Commun. 2020;523:458–64.

    Article  CAS  PubMed  Google Scholar 

  232. Yamada Y, Okano T, Orita K, Makino T, Shima F, Nakamura H. 3D-cultured small size adipose-derived stem cell spheroids promote bone regeneration in the critical-sized bone defect rat model. Biochem Biophys Res Commun. 2022;603:57–62.

    Article  CAS  PubMed  Google Scholar 

  233. Silva KR, Côrtes I, Liechocki S, Carneiro JRI, Souza AAP, Borojevic R, et al. Characterization of stromal vascular fraction and adipose stem cells from subcutaneous, preperitoneal and visceral morbidly obese human adipose tissue depots. PLoS ONE. 2017;12:e0174115.

    Article  PubMed  PubMed Central  Google Scholar 

  234. Raajendiran A, Ooi G, Bayliss J, O’Brien PE, Schittenhelm RB, Clark AK, et al. Identification of metabolically distinct adipocyte progenitor cells in human adipose tissues. Cell Rep. 2019;27:1528–e15407.

    Article  CAS  PubMed  Google Scholar 

  235. Yang Loureiro Z, Solivan-Rivera J, Corvera S. Adipocyte heterogeneity underlying adipose tissue functions. Endocrinology. 2022;163:bqab138.

    Article  PubMed  Google Scholar 

  236. Silva KR, Baptista LS. Adipose-derived stromal/stem cells from different adipose depots in obesity development. World J Stem Cells. 2019;11:147–66.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This research received no external funding.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: T.L.; Design: T.L., S.Pe.; Writing-original draft preparation: T.L; Writing-revision and editing: T.L., S.Pe.; Supervision: T.L., S.Pu., S.C.; Project administration: T.L., S.Pe., S.Pu., S.C. All authors have read the manuscript and agreed with its publication.

Corresponding author

Correspondence to Tonia Luca.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Luca, T., Pezzino, S., Puleo, S. et al. Lesson on obesity and anatomy of adipose tissue: new models of study in the era of clinical and translational research. J Transl Med 22, 764 (2024). https://doi.org/10.1186/s12967-024-05547-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-024-05547-3

Keywords