Next Article in Journal
Opposing Effects of Interleukin-36γ and Interleukin-38 on Trained Immunity
Next Article in Special Issue
Long-Term Ingestion of Sicilian Black Bee Chestnut Honey and/or D-Limonene Counteracts Brain Damage Induced by High Fat-Diet in Obese Mice
Previous Article in Journal
Aqua/Mechanochemical Mediated Synthesis of Novel Spiro [Indole–Pyrrolidine] Derivatives
Previous Article in Special Issue
Preliminary Evidence of the Potent and Selective Adenosine A2B Receptor Antagonist PSB-603 in Reducing Obesity and Some of Its Associated Metabolic Disorders in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Management of Obesity and Obesity-Related Disorders: From Stem Cells and Epigenetics to Its Treatment

by
Sara Cruciani
1,2,
Alessandro Palmerio Delitala
3,
Maria Laura Cossu
4,
Carlo Ventura
5 and
Margherita Maioli
1,2,6,*
1
Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
2
Consorzio Interuniversitario “Istituto Nazionale Biostrutture e Biosistemi” (INBB), Viale delle Medaglie d’Oro 305, 00136 Roma, Italy
3
Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
4
General Surgery Unit 2 “Clinica Chirurgica” Medical, Surgical and Experimental Sciences Department, University of Sassari, 07100 Sassari, Italy
5
National Laboratory of Molecular Biology and Stem Cell Engineering, Eldor Lab, Istituto Nazionale di Biostrutture e Biosistemi (INBB), Via di Corticella 183, 40128 Bologna, Italy
6
Center for Developmental Biology and Reprogramming (CEDEBIOR), Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2310; https://doi.org/10.3390/ijms24032310
Submission received: 30 December 2022 / Revised: 17 January 2023 / Accepted: 20 January 2023 / Published: 24 January 2023

Abstract

:
Obesity is a complex worldwide disease, characterized by an abnormal or excessive fat accumulation. The onset of this pathology is generally linked to a complex network of interactions among genetic and environmental factors, aging, lifestyle, and diets. During adipogenesis, several regulatory mechanisms and transcription factors are involved. As fat cells grow, adipose tissue becomes increasingly large and dysfunctional, losing its endocrine function, secreting pro-inflammatory cytokines, and recruiting infiltrating macrophages. This long-term low-grade systemic inflammation results in insulin resistance in peripheral tissues. In this review we describe the main mechanisms involved in adipogenesis, from a physiological condition to obesity. Current therapeutic strategies for the management of obesity and the related metabolic syndrome are also reported.

1. Introduction

Lifestyle changes, such as the consumption of calorie-dense foods and sedentary living, have progressively led to a profound imbalance between calorie intake and consumption [1]. Obesity is a direct consequence of these changes and is closely related to many metabolic disorders, including type 2 diabetes, insulin resistance, hyperglycemia, dyslipidemia, hypertension, and non-metabolic diseases, such as heart disease and many types of cancer [2,3,4]. The prevalence of childhood/adolescent obesity is increasing worldwide, rising from 4.5 percent in 1990 to 6.7 percent in 2010 [5]. A survey of the obesity epidemic shows that as of 2022 more than 39 million children worldwide are obese [6]. Obesity is strictly dependent on body composition rather than body weight and particularly on the number of adipocytes. Adipose tissue can be classified into different types, white (WAT) and brown (BAT) [7]. The primary function of WAT is energy storage in the form of triglycerides (TG) [8]. On the other hand, BAT dissipates energy to produce heat, suggesting its possible anti-obesity role [9,10]. In addition to brown and white adipocytes, another class of adipocytes, called beige/brown adipocytes, has recently been described [11]. Preadipocytes are converted into mature adipocytes in the final stage of differentiation when they are exposed to certain stimuli [12]. Thus, identifying the molecular mechanisms underlying resident mesenchymal stem cell (MSCs) differentiation could add new insights for the identification of future therapeutic approaches against obesity [13]. Mesenchymal stem cells (MSCs) are a type of adult stem cells known for their high plasticity and ability to generate mesodermal and non-mesodermal tissues [14]. Cell proliferation and differentiation are two opposing processes. Adipogenesis is regulated by a complex and highly orchestrated gene expression program, which occurs in stages [15]. The first phase involves the commitment of pluripotent stem cells into preadipocytes. Preadipocytes cannot be morphologically distinguished from their precursor cells, but they have also lost the potential to differentiate into other cell types [16]. Therefore, in the second stage, known as terminal differentiation, preadipocytes gradually acquire the features of mature adipocytes and become able to regulate lipid transport and synthesis, insulin sensitivity, and secretion of adipocyte-specific proteins [17]. During these phases, specific adipogenic-related genes are induced, and, at the same time, a series of epigenetic and chromatin modifications also occurs, leading ultimately to the silencing of the stemness-related genes [18,19]. Several natural or synthetic molecules can therefore act directly on these transcription factors, thus representing promising therapeutic agents to modulate uncontrolled adipose tissue hyperproliferation [20,21]. In this review, we focus on the biological features of adipose tissue, analyzing the transcription factors and key proteins involved in stem cell differentiation and the major surgical and pharmacological interventions for the control of obesity and its related metabolic syndrome.

2. Epigenetic Programming of Adipose-Derived Stem Cells (ADSCs)

Adipose-derived stem cells (ADSCs) have emerged in recent years as the most sought-after source of cells for the treatment of obesity, metabolic and degenerative diseases, due to their availability, rapid expansion, and differentiative potential toward several phenotypes [22,23]. ADSCs are mainly found in the so-called stromal vascular fraction (SVF) of adipose tissue, from which they can be easily isolated by mechanical and enzymatic procedures [24,25]. The application of adult stem cells in regenerative medicine enables the repair of many tissues, including vessels, muscles, nerves, cartilage, and skin [26]. Stem cell differentiation requires the precise activation of genes involved in the development of a definite cell type. On the other hand, differentiation itself involves the suppression of specific stemness-related genes, such as octamer-binding transcription factor 4 (Oct-4), sex determining region Y-box 2 (SOX2), and Nanog Homeobox protein (NANOG) [27,28]. The differentiation of preadipocytes into mature adipocytes is regulated by a complex network of transcription factors [29,30]. Any dysregulation in this process causes lipodystrophy, which impairs glucose and lipid homeostasis [31]. While Runt-related transcription factor 2 (RUNX2), the key osteogenic transcription factor, triggers an osteogenic differentiation program in MSCs, adipogenic differentiation is mainly promoted by peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT/enhancer binding protein alpha (C/EBPα) [32,33,34]. PPARγ, in particular, is considered the master regulator of adipogenesis [35]. On the other hand, C/EBPα-deficient cells are able to differentiate as adipocytes; however, these C/EBPα-deficient cells are resistant to insulin [36]. In addition to PPARγ, other genes are also involved in adipogenic commitment, such as uncoupling protein 1 (UCP1), which distinguishes BAT from WAT, whose function is thermogenesis in response to cold stress or β-adrenergic stimulus [37]. Insulin-Like Growth Factor-1 (IGF-1), Transforming Growth Factor beta (TGF-β), or cyclic AMP (cAMP) signaling pathways also play key roles in adipocyte differentiation [38]. Moreover, differential gene expression appears to be strongly influenced by epigenetic factors. DNA demethylation and methylation, acetylation or ubiquitylation of histones, as well as various noncoding RNAs, such as microRNAs, are the most studied epigenetic factors involved in modulating cellular organization [29,39]. Epigenetic changes can directly modulate gene promoters, thus facilitating (or preventing) the recruitment of additional chromatin-modifying enzymes or transcriptional regulators that would drive stem cell differentiation [40,41]. Post-translational modifications (PTMs) of histones by histone deacetylases (HDACs) or histone methyltransferases (HMTs) have been reported to be crucial in shaping the process of adipogenesis [42,43]. Reduced expression of Sirt1 and Sirt2, for example, has been associated with increased differentiation capacity of visceral adipose stem cells [44]. In addition, altered global DNA methylation pattern was observed during metabolic disorders on various genes involved in adipocyte differentiation, lipid metabolism, and inflammation [45,46]. Decreased expression of some epigenetic factors, such as HDAC1, promotes adipogenesis and visceral fat accumulation in human obesity [47,48]. Within this context, it has been demonstrated that exposure of ADSCs to metformin and vitamin D increases the expression of HDAC1 and other epigenetic modulators [49]. Furthermore, several microRNAs (miRNAs) are indeed found to be involved in the inactivation of the adipogenesis process. After treatment with metformin and vitamin D, for example, ADSCs showed upregulated levels of miR-145 and a downregulated expression of miR-148 [49]. miR-145 is downregulated during adipogenesis, while its upregulation inhibits adipogenesis by reducing the activity of PI3K/Akt and MAPK signaling pathways [50]. The expression of miR-148a is also affected by lipid accumulation. When upregulated, miR-148a promotes adipogenic differentiation, while it inhibits preadipocyte differentiation when miR-145 is upregulated and miR-148 is downregulated [51] (Figure 1). These molecules are also able to modulate inflammation and the expression of other key genes involved in adipogenic differentiation, counteracting WAT formation, and inducing a “brown-like” phenotype [52,53]. The use of bioactive molecules and chemical stimuli can then control the de-novo lipogenesis, differentiation, and physiology of adipose tissue, for the in vivo treatment of chronic pathological conditions of difficult resolution.

3. Biological Features of Adipose Tissue

Adipose tissue comprises 15–25% of the body weight and it can be classified into subcutaneous and visceral tissue [54]. Adipose tissue represents the main site of energy storage playing an important role as an endocrine organ, and directly modulating systemic lipid, glucose metabolism, and insulin sensitivity [55]. Within adipose tissue, in addition to the adipocytes, the stromal vascular fraction (SVF), containing a type of stem cells called adipose-derived stem cells (ADSCs), can be found [56,57]. These cells are able to differentiate into mature adipocytes following a process called adipogenesis [58]. Within healthy adipose stores, ADSCs have extensive immunomodulatory functions, such as the inhibition of natural killer (NK) cell and T cell proliferation. Moreover, they exert key secretory functions by releasing inflammatory cytokines, as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) in response to inflammation [59,60]. Lipid storage is determined by the balance between lipogenesis and lipolysis. The volume of the adipocyte reflects its specific function of storing energy in the form of lipids and, therefore, the ability of the cell to drastically modulate its size in response to changes in energy balance [61]. Adipose tissue expansion is determined by hyperplasia and/or hypertrophy of adipocytes. Hyperplasia refers to the formation of new adipocytes from preadipocytes at-hormone regulation and mediated by a series of transcription factors [30]. Unlike hyperplasia, hypertrophy is the enlargement of adipocytes by accumulation of lipids, either by uptake from the circulation or through the fatty acid synthesis pathway, known as “de novo lipogenesis” [62]. Generally, adipocyte hypertrophy is mainly associated with insulin resistance, hepatic steatosis, and other markers of metabolic dysfunction [63]. Besides the WAT and BAT, beige/brite adipose tissue shows intermediate characteristics between white and brown fat, with a central nuclei, multilocular lipid droplets, and is rich in mitochondria [64,65]. In addition to being highly insulin-responsive, adipose tissue also secretes several adipokines involved in glucose regulation and metabolic health [66]. These molecules can act as endocrine regulators, influencing different tissues and organs and regulating local signaling in a paracrine or autocrine manner [67,68]. Beige adipocytes are formed by a process named “WAT browning”, following stimulation of sympathetic activity during chronic cold exposure or administration of β3-adrenergic receptor agonists or exercise [69]. Several pharmacological and nonpharmacological strategies have consequently been developed to induce WAT browning as a possible mechanism to control weight gain and obesity [70]. In fact, adipose organ dysfunction can lead to age-related metabolic alterations, resulting in increased production of inflammatory peptides and macrophage infiltration, and a decrease in anti-inflammatory activity [71]. The production of pro-inflammatory mediators and the infiltration of immune cells inside the tissue generate a state of chronic inflammation. This inflammatory state that worsens with age is defined as “inflammaging” [72]. With aging, there is an increase in the number of white adipocytes and a decreased activity of brown adipocytes. Aging is also associated with a decrease in the formation of beige adipocytes [73]. During obesity, if compared to WAT, brown and beige adipose tissue are less likely to undergo local inflammation, even though an increased production of pro-inflammatory cytokines, such as TNF-alpha and MCP-1 [74], can be observed. However, the inflammation that is generated can compromise the thermogenic activity in BAT due to impaired insulin sensitivity and reduced glucose absorption [75].

4. Obesity and Obesity-Related Metabolic Syndrome

Obesity is an abnormal or excessive increase in body fat, classified according to the determination of Body Mass Index (BMI) [76]. According to the World Health Organization (WHO), a BMI greater than or equal to 30 kg/m2 is consistent with obesity, which is divided into different grades according to severity, and a BMI between 25 and 30 kg/m2 identifies overweight [77]. Excess body fat, especially visceral, is also related to a major risk of numerous chronic diseases including cardiovascular disease, type 2 diabetes, hepatic steatosis, several types of malignancies, and muscular and osteoarticular disorders [78,79]. These diseases are responsible for nearly 3 million deaths per year worldwide. Obesity is the fifth leading cause of death after hypertension, smoking, hyperglycemia, and physical inactivity. A real epidemic of overweight and obese individuals can be detected, especially in undeveloped countries [80,81]. The WHO has predicted a global “obesity epidemic” by 2030, in which 1 in 5 women and 1 in 7 men will be living with obesity. Moreover, this overweight/obesity epidemic and all its complications, have such negative implications on public health, that it can be considered a pandemic disease [82]. Several studies are still underway to better understand the causal factors of obesity. It is a complex hereditary disease whose pathophysiology seems dependent on the interaction between genetics, epigenetics, metagenomics, and environment factors [83,84]. In addition, since the early 1980s, various environmental changes have fostered an “obesogenic environment” with an abundance of high-calorie food, poor-quality food, and, not least, a sedentary lifestyle with reduced physical activity [85]. A long-term imbalance between energy intake and energy expenditure alters the metabolism and functions of WAT. During over-nutrition, lipids are stored within adipocytes [86]. When an enlarged hypertrophic adipocyte reaches maximum capacity, it is no longer able to store excess lipids and becomes fibrotic and inflammatory. Obesity disrupts physiological homeostasis and alters microenvironments by altering stem cell plasticity and impairing regenerative capacity [87,88]. The decreased plasticity of ADSCs exposed to the obese environment could significantly limit their therapeutic potential and ultimately reduce their therapeutic efficacy [89,90]. Excessive adiposity leads to hyperplasia of adipocytes and the secretion of growth factors, such as insulin-like growth factor-1 (IGF-1), tumor necrosis factor-α (TNF-α), angiotensin II (Ang II), and macrophage colony-stimulating factor (M-CSF) [91,92]. Macrophages and other immune cells produce pro-inflammatory cytokines and reactive oxygen species (ROS) that contribute to the development of a state of chronic low-grade inflammation and insulin resistance [93]. In addition, specific adipokines secreted by adipocytes increase vasomotor endothelial tone and consequently hypertension in obese patients [94]. Obesity can lead to increased synthesis and secretion of low-density lipoprotein (LDL) and very low-density lipoprotein (VLDL), which in turn release triglycerides into extrahepatic tissues. High plasma levels of free fatty acids (FFA) inhibit lipogenesis, preventing proper clearance of serum triacylglycerol levels, leading to hypertriglyceridemia and may result in insulin receptor dysfunction [95,96]. Epigenetic changes are crucial for several key biological processes, including cellular differentiation. Recent evidence suggests that obesity may result from the complex interplay between environmental changes and the epigenome [97,98,99]. Indeed, many genes are activated or inhibited to regulate energy metabolism. For example, PPARγ promoter methylation is increased in the subcutaneous adipose tissue of obese women [100]. DNMT1-deficient mice exhibit reduced energy expenditure, increased body weight, and susceptibility to diet-induced obesity [101]. HDAC1 deletion has been found to significantly increase the expression of the thermogenic genes UCP1 and Peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α) by increasing acetylation and decreasing methylation of histone H3 lysine 27 (H3K27) [102]. Overexpression of HDAC4 in adipocytes leads to the expansion of beige adipocytes and a reduction in adiposity [103]. Stem cell or brown adipose tissue transplantation, cell lysates, and exosomes have been tested in obese mouse models [104,105]. Overall, ADSCs were found to be effective in treating obesity-associated diabetes and inflammation and protective against cardiovascular disease [106,107]. Therefore, stem cell therapy represents a promising treatment strategy for obesity and obesity-related comorbidities [108].

5. Surgical Management of Obesity

Obesity represents an emerging worldwide disease, with an increased incidence in younger people, that has prompted the development of new and even more effective therapies [109]. Bariatric surgery, also called weight loss surgery, represents one of the approaches for the treatment of severe obesity, with the development of less invasive methods than in the past (Figure 2) [110,111]. Bariatric surgery procedures work by modifying the digestive system, trying to prevent many metabolic obesity-related diseases. The history of obesity surgery is studded with surgical techniques [112]. However, a few main groups can be identified: (1) pure malabsorption (jejunum-ileal bypass or biliointestinal bypass); (2) restrictive (gastroplasties and gastric banding); (3) mixed (bilio-pancreatic diversion and gastric bypass); and (4) alternatives (intragastric balloon and gastric stimulator) [113]. These kinds of procedures are indicated in patients with BMI >40 kg/m2 or >35 kg/m2 with complications associated with obesity, and in those who do not improve with medical therapy [114,115]. The gastric sleeve, also called sleeve gastrectomy, is one of the most performed bariatric surgeries [116]. It is a simple procedure that removes a large portion of the stomach, leaving behind a small, tubular portion, like a sleeve. This reduces the intake of food and the secretion of hunger hormones [117]. Gastric bypasses modify the stomach, making it Y-shaped by creating a small pouch in the upper part of the stomach [118]. Food flows through the new smaller stomach and the lower segment of the small intestine, bypassing the rest. The small intestine restriction makes this method more effective than gastric restriction alone [119]. Biliopancreatic diversion with duodenal switch (BPD-DS) is similar to gastric bypass but more extreme [120]. This surgery bypasses most of the small intestine, significantly reducing the hunger hormones produced in the small intestine and stomach, and greatly limiting the amount of nutrition the small intestine can absorb [121]. Stomach Intestinal Pylorus Sparing Surgery (SIPS) is a modified version of the original duodenal switch [122]. It begins with a sleeve gastrectomy, and divides the first part of the small intestine by closing it back into a loop; in this type of surgery, a smaller part of the small intestine is bypassed, allowing for greater nutrient absorption [123]. These kinds of procedures involve several complications that occur in the short and long term. Early complications include thromboembolism, pulmonary or respiratory failure, hemorrhage, peritonitis, and wound infection [124]. Late complications include gastrointestinal obstruction, marginal ulceration, band malfunction, steatorrhea, diarrhea, micronutrient nutritional deficiencies, and neurological complications [125,126]. This also occurs in general surgery. Excess subcutaneous adipose tissue leads to impaired healing due to low regional perfusion and oxygen tension. Second, the operative time for the obese is quite long and is a significant predictor of postoperative wound infections [127]. The new style of thinking aims to develop less invasive methods in approaching the obese surgical patients [128]. Almost all bariatric surgeons follow a sequence of application of the various techniques available, in which these are ordered according to the criteria of increasing invasiveness but at the same time effectiveness [129,130]. The beginning of this course is represented by the endoscopic placement of an endogastric board (BIB), which is followed in almost all patients by restrictive surgery [131]. This intervention is represented in many cases by adjustable gastric banding (LAGB), which has the advantage of being completely reversible, as it does not involve mutilation of any part of the digestive tract or anastomosis, and of being easily performed laparoscopically [132,133]. Laparoscopic procedures are associated with shorter operative time, less postoperative pain, and earlier recovery, as well as better respiratory function and aesthetic results [134]. The Swedish Obese Subjects (SOS) study showed that weight loss achieved after bariatric surgery significantly improves all obesity-related risk factors such as diabetes, hypercholesterolemia, low lipoprotein levels, hypertension, and hyperuricemia [135,136]. Weight loss after bariatric surgery is also associated with a significant reduction in overall mortality [137].

6. Pharmacological Interventions for Obesity

Pharmacological treatment of obesity became a hot topic in the scientific community (Figure 2), due to the possible wide number of subjects that could potentially benefit, also because behavioral approaches commonly failed and indications for bariatric surgery are becoming narrower [138]. Drugs used to treat obesity can be divided accordingly to their mechanisms of action [139].

6.1. Sympathicomimetic

These compounds are generally used for a short-term treatment of obesity. A weight regain after their discontinuation has been also reported [140]. Among these drugs, two compounds are the most used in the clinical practice: diethylpropion, is an amphetamine derivate and a sympathomimetic stimulant which stimulates the endogenous release of dopamine and norepinephrine causing a suppression of appetite. Further, diethylpropion indirectly elevated leptin level in the brain, thus inhibiting the production of neuropeptide Y [141]. The second compound is phentermine that, likewise to diethylpropion, acts through appetite suppression and by increasing basal energy expenditure. Approved by Food and Drug Administration for short-term use (3 months), a recent trial reported an 8.1 Kg loss after 12 weeks of clinical trial with phentermine, as compared to the placebo [142]. Due to the presence of specific side effects (irritability, mood changes, insomnia, elevation in mean blood pressure and palpitations), its use is discouraged in patients with cardiovascular disease, hyperthyroidism, and anxiety.

6.2. Sympathicomimetic and Anticonvulsivant

Phentermine–topiramate: this combined therapy was approved by FDA in 2012 for the chronic treatment of weight control. The appetite suppressant property of phentermine has been associated with the anti-convulsing drug topiramate which showed weight loss potential. Indeed, it has been hypothesized that topiramate has an appetite suppressant effect by modulating GABA receptor activation [143]. The recommend dose for this combined therapy is 7.5 mg/46 mg every day and was shown to induce a mean weight loss of 9.6 Kg after 108 weeks of treatment. The use of higher dosage (15 mg/92 mg) was associated with a further reduction in body weight [144].

6.3. Pancreatic Lipase Inhibitor

Orlistat was approved both by FDA and European Medicines Agency (EMA) for chronic weight management. Orlistat inhibits pancreas and stomach lipase, thus causing a decrease in fat absorption and a reduction in caloric intake [145]. Common side effects have been reported to be diarrhea, flatulence, and abdominal pains, as well as fat-soluble vitamin deficiencies [146] and scanty cases of liver-injuries [147].

6.4. 5-HT2c Serotonin Agonist

Lorcaserin is a serotonin receptor agonist with high affinity for 5-HT2C receptor, thus limiting the onset of hallucinations and side effects at the cardiac level, respectively caused by the binding of 5-HT2A and 5-HT2B receptors [148]. Despite the possibility of additional side effect (such as headache, nausea, dry mouth, dizziness, and constipation), the use of lorcaserin at 10 mg twice a day was shown to decrease the body weight by at least 5% in 47.5% of the patients, while 22.6% of them had a weight loss ≥ 10% after 1 year of follow-up [149].

6.5. Glucagon-like Peptide 1 Agonists

Liraglutide is a glucagon-like peptide 1 (GLP-1) agonist, approved by both FDA and EMA for chronic weight management. GLP-1 is physiologically secreted by the enteroendocrine L-cells of distal ileum and proximal colon after oral meal consumption, but a certain basal rate of secretion has been recently postulated [150]. GLP-1 is an incretin hormone which increases the glucose-dependent insulin secretion but has shown additional effects: decreases in appetite and food consumption [151], and delays in gastric emptying, thus increasing postprandial feelings of satiety and fullness [152]. The use of Liraglutide was associated with a mean reduction of 8.4 Kg in body weight after 1 year of follow-up and a reduction in cardiovascular risk factors [153]. Gastrointestinal side effects have been reported in some studies [154].

6.6. Opioid Receptor Antagonist/Dopamine and Noradrenaline Reuptake Inhibitor

Naltrexone–bupropion: naltrexone is an opioid antagonist prescribed for the management of alcohol and opioid use disorder. Bupropion is a dopamine and norepinephrine neuronal reuptake inhibitor and was firstly prescribed as an antidepressant. Bupropion also stimulates the α-MSH release, thus inducing appetite regulation. While the exact underlying mechanism is still unknown, it has been suggested that naltrexone and bupropion synergistically act at the melanocortin system [155]. Their combined use is approved by EMA and FDA and has been associated with a mean reduction of 6.1% in body weight [156].

7. Conclusions

Dysfunction of adipocytes and adipose tissue is the main feature of obesity, resulting in an increased risk of insulin resistance, type 2 diabetes, fatty liver disease, hypertension, dyslipidemia, and cardiovascular disorders. In most obese subjects, the compromised physiology of the adipose tissue depends on the hypertrophy of the adipocytes and on the interaction of genetic, epigenetic, and environmental factors. Several surgical procedures, such as biliointestinal bypass or gastric banding, and pharmacological interventions, such as pancreatic lipase inhibitors or Glucagon-Like Peptide 1 agonists, are applied for the management of obesity. Therefore, stem cell therapy and the use of targeted treatment acting on cell differentiation or epigenetic modifications of key target genes, may represent a promising strategy for the management of obesity and obesity-related metabolic syndrome.

Author Contributions

Conceptualization, S.C. and M.M.; validation, C.V. and M.M.; investigation, S.C.; A.P.D. and M.M.; writing—original draft preparation, S.C.; A.P.D. and M.L.C.; writing—review and editing, M.M.; visualization, C.V.; supervision, M.M; funding acquisition, M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by “Fondo di Ateneo per la ricerca 2022” (Margherita Maioli).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Vicente, A.J.-L.; Beltrán-Carrillo, J.; Megías, Á.; González-Cutre, D. Elements behind sedentary lifestyles and unhealthy eating habits in individuals with severe obesity. Int. J. Qual. Stud. Health Well-Being 2022, 17, 2056967. [Google Scholar]
  2. Scully, T.; Ettela, A.; LeRoith, D.; Gallagher, E.J. Obesity, Type 2 Diabetes, and Cancer Risk. Front. Oncol. 2021, 10, 615375. [Google Scholar] [CrossRef] [PubMed]
  3. Jung, U.J.; Choi, M.S. Obesity and its metabolic complications: The role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. Int. J. Mol. Sci. 2014, 15, 6184–6223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Hajer, G.R.; Van Haeften, T.W.; Visseren, F.L.J. Adipose tissue dysfunction in obesity, diabetes, and vascular diseases. Eur. Heart J. 2008, 29, 2959–2971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. De Onis, M.; Blössner, M.; Borghi, E. Global prevalence and trends of overweight and obesity among preschool children. Am. J. Clin. Nutr. 2010, 92, 1257–1264. [Google Scholar] [CrossRef] [Green Version]
  6. Ling, T.-S.A.K.J.; Chen, S.; Zahry, N.R. Economic burden of childhood overweight and obesity: A systematic review and meta-analysis. Obes. Rev. 2022, 24, e13535. [Google Scholar] [CrossRef]
  7. Lee, Y.H.; Mottillo, E.P.; Granneman, J.G. Adipose tissue plasticity from WAT to BAT and in between. Biochim. Biophys. Acta Mol. Basis Dis. 2014, 1842, 358–369. [Google Scholar] [CrossRef] [Green Version]
  8. Welte, M.A.; Gould, A.P. Lipid droplet functions beyond energy storage. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2017, 1862, 1260–1272. [Google Scholar] [CrossRef] [PubMed]
  9. Seale, P.; Lazar, M.A. Brown fat in humans: Turning up the heat on obesity. Diabetes 2009, 58, 1482–1484. [Google Scholar] [CrossRef] [Green Version]
  10. Maliszewska, K.; Kretowski, A. Brown adipose tissue and its role in insulin and glucose homeostasis. Int. J. Mol. Sci. 2021, 22, 1530. [Google Scholar] [CrossRef]
  11. Pilkington, A.C.; Paz, H.A.; Wankhade, U.D. Beige Adipose Tissue Identification and Marker Specificity—Overview. Front. Endocrinol. 2021, 12, 599134. [Google Scholar] [CrossRef]
  12. Audano, M.; Pedretti, S.; Caruso, D.; Crestani, M.; De Fabiani, E.; Mitro, N. Regulatory mechanisms of the early phase of white adipocyte differentiation: An overview. Cell. Mol. Life Sci. 2022, 79, 1–14. [Google Scholar] [CrossRef]
  13. Glenn, J.D. Mesenchymal stem cells: Emerging mechanisms of immunomodulation and therapy. World, J. Stem Cells 2014, 6, 526–539. [Google Scholar] [CrossRef] [PubMed]
  14. Ullah, I.; Subbarao, R.B.; Rho, G.J. Human mesenchymal stem cells—Current trends and future prospective. Biosci. Rep. 2015, 35, e00191. [Google Scholar] [CrossRef] [PubMed]
  15. Moseti, D.; Regassa, A.; Kim, W.K. Molecular regulation of adipogenesis and potential anti-adipogenic bioactive molecules. Int. J. Mol. Sci. 2016, 17, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Cawthorn, W.P.; Scheller, E.L.; MacDougald, O.A. Adipose tissue stem cells meet preadipocyte commitment: Going back to the future. J. Lipid Res. 2012, 53, 227–246. [Google Scholar] [CrossRef] [Green Version]
  17. Gregoire, F.M.; Smas, C.M.; Sul, H.S. Understanding adipocyte differentiation. Physiol. Rev. 1998, 78, 783–809. [Google Scholar] [CrossRef] [Green Version]
  18. Nic-Can, G.I.; Rodas-Junco, B.A.; Carrillo-Cocom, L.M.; Zepeda-Pedreguera, A.; Peñaloza-Cuevas, R.; Aguilar-Ayala, F.J.; Rojas-Herrera, R.A. Epigenetic regulation of adipogenic differentiation by histone lysine demethylation. Int. J. Mol. Sci. 2019, 20, 3918. [Google Scholar] [CrossRef] [Green Version]
  19. Ying, H.; Pan, R.; Chen, Y. Epigenetic Control of Mesenchymal Stromal Cell Fate Decision. In Post-Translational Modifications in Cell. Functions and Diseases; InTechOpen: London, UK, 2021. [Google Scholar] [CrossRef]
  20. Szymczak-Pajor, I.; Miazek, K.; Selmi, A.; Balcerczyk, A.; Śliwińska, A. The Action of Vitamin D in Adipose Tissue: Is There the Link between Vitamin D Deficiency and Adipose Tissue-Related Metabolic Disorders? Int. J. Mol. Sci. 2022, 23, 956. [Google Scholar] [CrossRef]
  21. Santaniello, S.; Cruciani, S.; Basoli, V.; Balzano, F.; Bellu, E.; Garroni, G.; Ginesu, G.C.; Cossu, M.L.; Facchin, F.; Delitala, A.P.; et al. Melatonin and Vitamin D Orchestrate Adipose Derived Stem Cell Fate by Modulating Epigenetic Regulatory. Genes 2018, 15, 1631–1639. [Google Scholar] [CrossRef]
  22. Fathi, E.; Farahzadi, R. Isolation, culturing, characterization and aging of adipose tissue-derived mesenchymal stem cells: A brief overview. Braz. Arch. Biol. Technol. 2016, 59. [Google Scholar] [CrossRef] [Green Version]
  23. Schäffler, A.; Büchler, C. Concise Review: Adipose Tissue-Derived Stromal Cells—Basic and Clinical Implications for Novel Cell-Based Therapies. Stem Cells 2007, 25, 818–827. [Google Scholar] [CrossRef]
  24. Basoli, V.; Santaniello, S.; Cruciani, S.; Ginesu, G.C.; Cossu, M.L.; Delitala, A.P.; Serra, P.A.; Ventura, C.; Maioli, M. Melatonin and vitamin D interfere with the adipogenic fate of adipose-derived stem cells. Int. J. Mol. Sci. 2017, 18, 981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Bianchi, F.; Maioli, M.; Leonardi, E.; Olivi, E.; Pasquinelli, G.; Valente, S.; Mendez, A.J.; Ricordi, C.; Raffaini, M.; Tremolada, C.; et al. A new nonenzymatic method and device to obtain a fat tissue derivative highly enriched in pericyte-like elements by mild mechanical forces from human lipoaspirates. Cell Transplant. 2013, 22, 2063–2077. [Google Scholar] [CrossRef] [PubMed]
  26. Naderi, N.; Combellack, E.J.; Griffin, M.; Sedaghati, T.; Javed, M.; Findlay, M.W.; Wallace, C.G.; Mosahebi, A.; Butler, P.E.; Seifalian, A.M.; et al. The regenerative role of adipose-derived stem cells (ADSC) in plastic and reconstructive surgery. Int. Wound, J. 2017, 14, 112–124. [Google Scholar] [CrossRef]
  27. Rooj, A.K.; Bronisz, A.; Godlewski, J. The role of octamer binding transcription factors in glioblastoma multiforme. Biochim. Biophys. Acta Gene Regul. Mech. 2016, 18, 1093–1108. [Google Scholar] [CrossRef] [Green Version]
  28. Kashyap, V.; Rezende, N.C.; Scotland, K.B.; Shaffer, S.M.; Persson, J.L.; Gudas, L.J.; Mongan, A.P. Regulation of Stem cell pluripotency and differentiation involves a mutual regulatory circuit of the Nanog, OCT4, and SOX2 pluripotency transcription factors with polycomb Repressive Complexes and Stem Cell microRNAs. Stem Cells Dev. 2009, 18, 1093–1108. [Google Scholar] [CrossRef]
  29. Perez-Campo, F.; Riancho, J. Epigenetic Mechanisms Regulating Mesenchymal Stem Cell Differentiation. Curr. Genom. 2015, 16, 368–383. [Google Scholar] [CrossRef] [Green Version]
  30. Ambele, M.A.; Dhanraj, P.; Giles, R.; Pepper, M.S. Adipogenesis: A complex interplay of multiple molecular determinants and pathways. Int. J. Mol. Sci. 2020, 21, 4283. [Google Scholar] [CrossRef]
  31. Moreno-Indias, I.; Tinahones, F.J. Impaired adipose tissue expandability and lipogenic capacities as ones of the main causes of metabolic disorders. J. Diabetes Res. 2015, 2015, 970375. [Google Scholar] [CrossRef]
  32. Chen, Q.; Shou, P.; Zheng, C.; Jiang, M.; Cao, G.; Yang, Q.; Cao, J.; Xie, N.; Velletri, T.; Zhang, X.; et al. Fate decision of mesenchymal stem cells: Adipocytes or osteoblasts? Cell Death Differ. 2016, 23, 1128–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Meyer, M.B.; Benkusky, N.A.; Sen, B.; Rubin, J.; Pike, J.W. Epigenetic plasticity drives adipogenic and osteogenic differentiation of marrow-derived mesenchymal stem cells. J. Biol. Chem. 2016, 291, 17829–17847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Hu, L.; Yin, C.; Zhao, F.; Ali, A.; Ma, J.; Qian, A. Mesenchymal stem cells: Cell fate decision to osteoblast or adipocyte and application in osteoporosis treatment. Int. J. Mol. Sci. 2018, 19, 360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Yang, D.; Li, N.; Zhang, G. Spontaneous adipogenic differentiation potential of adipose-derived stem cells decreased with increasing cell passages. Mol. Med. Rep. 2018, 17, 6109–6115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Rosen, E.D.; Hsu, C.H.; Wang, X.; Sakai, S.; Freeman, M.W.; Gonzalez, F.J.; Spiegelman, B.M. C/EBPα induces adipogenesis through PPARγ: A unified pathway. Genes Dev. 2002, 16, 22–26. [Google Scholar] [CrossRef] [Green Version]
  37. Yi, D.; Nguyen, H.P.; Sul, H.S. Epigenetic dynamics of the thermogenic gene program of adipocytes. Biochem. J. 2020, 477, 1137–1148. [Google Scholar] [CrossRef] [Green Version]
  38. Farmer, S.R. Transcriptional control of adipocyte formation. Cell Metab. 2006, 4, 263–273. [Google Scholar] [CrossRef] [Green Version]
  39. Zhou, Y.; Kim, J.; Yuan, X.; Braun, T. Epigenetic modifications of stem cells: A paradigm for the control of cardiac progenitor cells. Circ. Res. 2011, 109, 1067–1081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Krämer, A.I.; Handschin, C. How epigenetic modifications drive the expression and mediate the action of PGC-1α in the regulation of metabolism. Int. J. Mol. Sci. 2019, 20, 5449. [Google Scholar] [CrossRef] [Green Version]
  41. Jang, S.; Hwang, J.; Jeong, H.-S. The Role of Histone Acetylation in Mesenchymal Stem Cell Differentiation. Chonnam Med. J. 2022, 58, 6–12. [Google Scholar] [CrossRef]
  42. Li, H.X.; Xiao, L.; Wang, C.; Gao, J.L.; Zhai, Y.G. Epigenetic regulation of adipocyte differentiation and adipogenesis. J. Zhejiang Univ. Sci. B 2010, 11, 784–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Pant, R.; Firmal, P.; Shah, V.K.; Alam, A.; Chattopadhyay, S. Epigenetic Regulation of Adipogenesis in Development of Metabolic Syndrome. Front. Cell Dev. Biol. 2021, 8, 619888. [Google Scholar] [CrossRef] [PubMed]
  44. Perrini, S.; Porro, S.; Nigro, P.; Cignarelli, A.; Caccioppoli, C.; Genchi, V.A.; Martines, G.; De Fazio, M.; Capuano, P.; Natalicchio, A.; et al. Reduced SIRT1 and SIRT2 expression promotes adipogenesis of human visceral adipose stem cells and associates with accumulation of visceral fat in human obesity. Int. J. Obes. 2020, 44, 307–319. [Google Scholar] [CrossRef]
  45. Castellano-Castillo, D.; Moreno-Indias, I.; Sanchez-Alcoholado, L.; Ramos-Molina, B.; Alcaide-Torres, J.; Morcillo, S.; Ocaña-Wilhelmi, L.; Tinahones, F.; Queipo-Ortuño, M.I.; Cardonao, F. Altered adipose tissue DNA methylation status in metabolic syndrome: Relationships between global DNA methylation and specific methylation at adipogenic, lipid metabolism and inflammatory candidate genes and metabolic variables. J. Clin. Med. 2019, 8, 87. [Google Scholar] [CrossRef] [Green Version]
  46. Mirzaeicheshmeh, E.; Zerrweck, C.; Centeno-Cruz, F.; Baca-Peynado, P.; Martinez-Hernandez, A.; García-Ortiz, H.; Contreras-Cubas, C.; Salas-Martínez, M.G.; Saldaña-Alvarez, Y.; Mendoza-Caamal, E.C.; et al. Alterations of DNA methylation during adipogenesis differentiation of mesenchymal stem cells isolated from adipose tissue of patients with obesity is associated with type 2 diabetes. Adipocyte 2021, 10, 493–504. [Google Scholar] [CrossRef]
  47. Mayoral, R.; Osborn, O.; McNelis, J.; Johnson, A.M.; Oh, D.Y.; Izquierdo, C.L.; Chung, H.; Li, P.; Traves, P.G.; Bandyopadhyay, G.; et al. Adipocyte SIRT1 knockout promotes PPARγ activity, adipogenesis and insulin sensitivity in chronic-HFD and obesity. Mol. Metab. 2015, 4, 378–391. [Google Scholar] [CrossRef]
  48. Eung, J.Y.; Chung, J.J.; Sung, S.C.; Kang, H.K.; Jae, B.K. Down-regulation of histone deacetylases stimulates adipocyte differentiation. J. Biol. Chem. 2006, 281, 6608–6615. [Google Scholar] [CrossRef] [Green Version]
  49. Cruciani, S.; Garroni, G.; Balzano, F.; Pala, R.; Bellu, E.; Cossu, M.L.; Ginesu, G.C.; Ventura, C.; Maioli, M. Tuning adipogenic differentiation in adscs by metformin and vitamin d: Involvement of mirnas. Int. J. Mol. Sci. 2020, 21, 6181. [Google Scholar] [CrossRef] [PubMed]
  50. Wang, L.L.; Zhang, S.; Mei, G.C.C.; Li, S.; Zhang, W.; Junjvlieke, Z. MiR-145 reduces the activity of PI3K/Akt and MAPK signaling pathways and inhibits adipogenesis in bovine preadipocytes. Genomics 2020, 112, 2688–2694. [Google Scholar] [CrossRef]
  51. Wang, N.; He, L.; Lin, H.; Tan, L.; Sun, Y.; Zhang, X.; Danser, A.H.J.; Lu, H.S.; He, Y.; Lu, X. MicroRNA-148a regulates low-density lipoprotein metabolism by repressing the (pro)renin receptor. PLoS ONE 2020, 15, e0225356. [Google Scholar] [CrossRef]
  52. Cruciani, S.; Garroni, G.; Pala, R.; Cossu, M.L.; Ginesu, G.C.; Ventura, C.; Maioli, M. Metformin and vitamin d modulate inflammation and autophagy during adipose-derived stem cell differentiation. Int. J. Mol. Sci. 2021, 22, 6686. [Google Scholar] [CrossRef] [PubMed]
  53. Cruciani, M.M.S.; Garroni, G.; Pala, R.; Coradduzza, D.; Cossu, M.L.; Ginesu, G.C.; Capobianco, G.; Dessole, S.; Ventura, C. Metformin and vitamin D modulate adipose-derived stem cell differentiation towards the beige phenotype. Adipocyte 2022, 11, 356–365. [Google Scholar] [CrossRef] [PubMed]
  54. Harvey, I.; Boudreau, A.; Stephens, J.M. Adipose tissue in health and disease: Adipose Tissue in Health and Disease. Open Biol. 2020, 10, 200291. [Google Scholar] [CrossRef] [PubMed]
  55. Booth, A.; Magnuson, A.; Fouts, J.; Foster, M.T. Adipose tissue: An endocrine organ playing a role in metabolic regulation. Horm. Mol. Biol. Clin. Investig. 2016, 26, 25–42. [Google Scholar] [CrossRef] [PubMed]
  56. Bora, P.; Majumdar, A.S. Adipose tissue-derived stromal vascular fraction in regenerative medicine: A brief review on biology and translation. Stem Cell Res. Ther. 2017, 8, 145. [Google Scholar] [CrossRef]
  57. Ramakrishnan, V.M.; Boyd, N.L. The Adipose Stromal Vascular Fraction as a Complex Cellular Source for Tissue Engineering Applications. Tissue Eng. Part B Rev. 2018, 24, 289–299. [Google Scholar] [CrossRef]
  58. Song, T.; Kuang, S. Adipocyte dedifferentiation in health and diseases. Clinical Sci. 2019, 133, 2107–2119. [Google Scholar] [CrossRef]
  59. Ceccarelli, S.; Pontecorvi, P.; Anastasiadou, E.; Napoli, C.; Marchese, C. Immunomodulatory Effect of Adipose-Derived Stem Cells: The Cutting Edge of Clinical Application. Front. Cell Dev. Biol. 2020, 8, 236. [Google Scholar] [CrossRef]
  60. Abbasi, B.; Shamsasenjan, K.; Ahmadi, M.; Beheshti, S.A.; Saleh, M. Mesenchymal stem cells and natural killer cells interaction mechanisms and potential clinical applications. Stem Cell Res. Ther. 2022, 13, 97. [Google Scholar] [CrossRef]
  61. Rosen, E.D.; Spiegelman, B.M. Adipocytes as regulators of energy balance and glucose homeostasis. Nature 2006, 444, 847–853. [Google Scholar] [CrossRef]
  62. Hsiao, W.Y.; Guertin, D.A. De Novo Lipogenesis as a Source of Second Messengers in Adipocytes. Curr. Diabetes Rep. 2019, 19, 138. [Google Scholar] [CrossRef] [PubMed]
  63. Chait, A.; den Hartigh, L.J. Adipose Tissue Distribution, Inflammation and Its Metabolic Consequences, Including Diabetes and Cardiovascular Disease. Front. Cardiovasc. Med. 2020, 7, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Giralt, M.; Villarroya, F. White, brown, beige/brite: Different adipose cells for different functions? Endocrinology 2013, 154, 2992–3000. [Google Scholar] [CrossRef] [Green Version]
  65. Rui, L. Brown and beige adipose tissues in health and disease. Compr. Physiol. 2017, 7, 1281–1306. [Google Scholar] [CrossRef]
  66. Al-Mansoori, L.; Al-Jaber, H.; Prince, M.S.; Elrayess, M.A. Role of Inflammatory Cytokines, Growth Factors and Adipokines in Adipogenesis and Insulin Resistance. Inflammation 2022, 45, 31–44. [Google Scholar] [CrossRef]
  67. Smith, U.; Kahn, B.B. Adipose tissue regulates insulin sensitivity: Role of adipogenesis, de novo lipogenesis and novel lipids. J. Intern. Med. 2016, 280, 465–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Kahn, D.E.; Bergman, B.C. Keeping It Local in Metabolic Disease: Adipose Tissue Paracrine Signaling and Insulin Resistance. Diabetes 2022, 71, 599–609. [Google Scholar] [CrossRef] [PubMed]
  69. Lizcano, F.; Arroyave, F. Control of adipose cell browning and its therapeutic potential. Metabolites 2020, 10, 471. [Google Scholar] [CrossRef]
  70. Cheng, L.; Wang, J.; Dai, H.; Duan, Y.; An, Y.; Shi, L.; Lv, Y.; Li, H.; Wang, C.; Ma, Q.; et al. Brown and beige adipose tissue: A novel therapeutic strategy for obesity and type 2 diabetes mellitus. Adipocyte 2021, 10, 48–65. [Google Scholar] [CrossRef]
  71. Li, Y.; Yun, K.; Mu, R. A review on the biology and properties of adipose tissue macrophages involved in adipose tissue physiological and pathophysiological processes. Lipids Health Dis. 2020, 19, 164. [Google Scholar] [CrossRef]
  72. Zoico, E.; Rubele, S.; De Caro, A.; Nori, N.; Mazzali, G.; Fantin, F.; Rossi, A.; Zamboni, M. Brown and beige adipose tissue and aging. Front. Endocrinol. 2019, 10, 368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Von Bank, H.; Kirsh, C.; Simcox, J. Aging adipose: Depot location dictates age-associated expansion and dysfunction. Ageing Res. Rev. 2021, 67, 101259. [Google Scholar] [CrossRef] [PubMed]
  74. Villarroya, F.; Cereijo, R.; Gavaldà-Navarro, A.; Villarroya, J.; Giralt, M. Inflammation of brown/beige adipose tissues in obesity and metabolic disease. J. Intern. Med. 2018, 284, 492–504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Omran, F.; Christian, M. Inflammatory Signaling and Brown Fat Activity. Front. Endocrinol. 2020, 11, 156. [Google Scholar] [CrossRef] [PubMed]
  76. Purnell, J.Q. Definitions, Classification, and Epidemiology of Obesity; MDText.com, Inc.: South Dartmouth, MA, USA, 2018. [Google Scholar]
  77. Flegal, K.M.; Graubard, B.I. Estimates of excess deaths associated with body mass index and other anthropometric variables. Am. J. Clin. Nutr. 2009, 89, 1213–1219. [Google Scholar] [CrossRef] [Green Version]
  78. Gutiérrez-Cuevas, J.; Santos, A.; Armendariz-Borunda, J. Pathophysiological molecular mechanisms of obesity: A link between mafld and nash with cardiovascular diseases. Int. J. Mol. Sci. 2021, 22, 11629. [Google Scholar] [CrossRef]
  79. Caussy, C.; Aubin, A.; Loomba, R. The Relationship Between Type 2 Diabetes, NAFLD, and Cardiovascular Risk. Curr. Diabetes Rep. 2021, 21, 15. [Google Scholar] [CrossRef]
  80. Hruby, A.; Hu, F.B. The Epidemiology of Obesity: A Big Picture. PharmacoEconomics 2015, 33, 673–689. [Google Scholar] [CrossRef]
  81. Ryan, D.; Barquera, S.; Cavalcanti, O.B.; Ralston, J. The Global Pandemic of Overweight and Obesity. In Handbook of Global Health; Springer: Cham, Switzerland, 2020. [Google Scholar] [CrossRef]
  82. Lagerros, Y.T.; Rössner, S. Obesity management: What brings success? Ther. Adv. Gastroenterol. 2013, 6, 77–88. [Google Scholar] [CrossRef]
  83. Pigeyre, M.; Yazdi, F.T.; Kaur, Y.; Meyre, D. Recent progress in genetics, epigenetics and metagenomics unveils the pathophysiology of human obesity. Clin. Sci. 2016, 130, 943–986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Ghaben, A.L.; Scherer, P.E. Adipogenesis and metabolic health. Nat. Rev. Mol. Cell Biol. 2019, 20, 242–258. [Google Scholar] [CrossRef] [PubMed]
  85. Townshend, T.; Lake, A. Obesogenic environments: Current evidence of the built and food environments. Perspect. Public Health 2017, 137, 38–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Longo, M.; Zatterale, F.; Naderi, J.; Parrillo, L.; Formisano, P.; Raciti, G.A.; Beguinot, F.; Miele, C. Adipose tissue dysfunction as determinant of obesity-associated metabolic complications. Int. J. Mol. Sci. 2019, 20, 2358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Fuster, J.J.; Ouchi, N.; Gokce, N.; Walsh, K. Obesity-induced changes in adipose tissue microenvironment and their impact on cardiovascular disease. Circ. Res. 2016, 118, 1786–1807. [Google Scholar] [CrossRef] [Green Version]
  88. Baptista, L.S. Obesity and weight loss could alter the properties of adipose stem cells? World, J. Stem Cells 2015, 7, 165–173. [Google Scholar] [CrossRef]
  89. Payab, M.; Goodarzi, P.; Foroughi Heravani, N.; Hadavandkhani, M.; Zarei, Z.; Falahzadeh, K.; Larijani, B.; Rahim, F.; Arjmand, B. Stem cell and obesity: Current state and future perspective. In Advances in Experimental Medicine and Biology; Springer: Cham, Switzerland, 2018. [Google Scholar] [CrossRef]
  90. Oestreich, A.K.; Collins, K.H.; Harasymowicz, N.S.; Wu, C.L.; Guilak, F. Is Obesity a Disease of Stem Cells? Cell Stem Cell 2020, 27, 15–18. [Google Scholar] [CrossRef]
  91. Caruso, C.; Balistreri, C.R.; Candore, G. The role of adipose tissue and adipokines in obesity-related inflammatory diseases. Mediat. Inflamm. 2010, 2010, 802078. [Google Scholar] [CrossRef] [Green Version]
  92. Ahmad, B.; Serpell, C.J.; Fong, I.L.; Wong, E.H. Molecular Mechanisms of Adipogenesis: The Anti-adipogenic Role of AMP-Activated Protein Kinase. Front. Mol. Biosci. 2020, 7, 76. [Google Scholar] [CrossRef]
  93. Benomar, Y.; Taouis, M. Molecular mechanisms underlying obesity-induced hypothalamic inflammation and insulin resistance: Pivotal role of resistin/tlr4 pathways. Front. Endocrinol. 2019, 10, 140. [Google Scholar] [CrossRef] [Green Version]
  94. Campia, U.; Tesauro, M.; Cardillo, C. Human obesity and endothelium-dependent responsiveness. Br. J. Pharmacol. 2012, 165, 561–573. [Google Scholar] [CrossRef] [Green Version]
  95. Klop, B.; Elte, J.W.F.; Cabezas, M.C. Dyslipidemia in Obesity: Mechanisms and Potential Targets. Nutrients 2013, 5, 1218–1240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Wang, H.; Peng, D.Q. New insights into the mechanism of low high-density lipoprotein cholesterol in obesity. Lipids Health Dis. 2011, 10, 176. [Google Scholar] [CrossRef] [Green Version]
  97. Gao, W.; Liu, J.L.; Lu, X.; Yang, Q. Epigenetic regulation of energy metabolism in obesity. J. Mol. Cell Biol. 2021, 13, 480–499. [Google Scholar] [CrossRef]
  98. Park, Y.J.; Han, S.M.; Huh, J.Y.; Kim, J.B. Emerging roles of epigenetic regulation in obesity and metabolic disease. J. Biol. Chem. 2021. [CrossRef] [PubMed]
  99. Van Dijk, S.J.; Molloy, P.L.; Varinli, H.; Morrison, J.L.; Muhlhausler, B.S. Members of EpiSCOPE. Epigenetics and human obesity. Int. J. Obes. 2015, 39, 85–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Ma, X.; Kang, S. Functional implications of DNA methylation in adipose biology. Diabetes 2019, 68, 871–878. [Google Scholar] [CrossRef] [Green Version]
  101. Bruggeman, E.C.; Garretson, J.T.; Wu, R.; Shi, H.; Xue, B. Neuronal Dnmt1 deficiency attenuates diet-Induced obesity in mice. Endocrinology 2018, 159, 145–162. [Google Scholar] [CrossRef] [Green Version]
  102. Li, F.; Wu, R.; Cui, X.; Zha, L.; Yu, L.; Shi, H.; Xue, B. Histone deacetylase 1 (HDAC1) negatively regulates thermogenic program in brown adipocytes via coordinated regulation of histone H3 lysine 27 (H3K27) deacetylation and methylation. J. Biol. Chem. 2016, 291, 4523–4536. [Google Scholar] [CrossRef]
  103. Paulo, E.; Wu, D.; Hecker, P.; Zhang, Y.; Wang, B. Adipocyte HDAC4 activation leads to beige adipocyte expansion and reduced adiposity. J. Endocrinol. 2018, 239, 153–165. [Google Scholar] [CrossRef] [Green Version]
  104. Mikłosz, A.; Nikitiuk, B.E.; Chabowski, A. Using adipose-derived mesenchymal stem cells to fight the metabolic complications of obesity: Where do we stand? Obes. Rev. 2022, 23, e13413. [Google Scholar] [CrossRef] [PubMed]
  105. Calvo, E.; Keiran, N.; Núñez-Roa, C.; Maymó-Masip, E.; Ejarque, M.; Sabadell-Basallote, J.; Del Mar Rodríguez-Peña, M.; Ceperuelo-Mallafré, V.; Seco, J.; Benaiges, E.; et al. Effects of stem cells from inducible brown adipose tissue on diet-induced obesity in mice. Sci. Rep. 2021, 11, 13923. [Google Scholar] [CrossRef] [PubMed]
  106. Rochette, L.; Mazini, L.; Malka, G.; Zeller, M.; Cottin, Y.; Vergely, C. The crosstalk of adipose-derived stem cells (Adsc), oxidative stress, and inflammation in protective and adaptive responses. Int. J. Mol. Sci. 2020, 21, 9262. [Google Scholar] [CrossRef] [PubMed]
  107. Yang, H.; Li, C.; Li, Y.; Tai, R.; Sun, C. Adipose-derived stem cells and obesity: The spear and shield relationship. Genes Dis. 2021; in press. [Google Scholar] [CrossRef]
  108. Jaber, H.; Issa, K.; Eid, A.; Saleh, F.A. The therapeutic effects of adipose-derived mesenchymal stem cells on obesity and its associated diseases in diet-induced obese mice. Sci. Rep. 2021, 11, 6291. [Google Scholar] [CrossRef] [PubMed]
  109. Lin, X.; Li, H. Obesity: Epidemiology, Pathophysiology, and Therapeutics. Front. Endocrinol. 2021, 12, 706978. [Google Scholar] [CrossRef] [PubMed]
  110. Wolfe, B.M.; Kvach, E.; Eckel, R.H. Treatment of Obesity: Weight Loss and Bariatric Surgery. Circ. Res. 2016, 118, 1844–1855. [Google Scholar] [CrossRef] [PubMed]
  111. Hlinnik, A.A.; Aulas, S.D.; Stebounov, S.S.; Rummo, O.O.; Hermanovich, V.I. Bariatric Surgery for Morbid Obesity. Nov. Khirurgii 2021, 29, 662–670. [Google Scholar] [CrossRef]
  112. Nguyen, N.T.; Varela, J.E. Bariatric surgery for obesity and metabolic disorders: State of the art. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 160–169. [Google Scholar] [CrossRef]
  113. Eisenberg, D.; Duffy, A.J.; Bell, R.L. Update on obesity surgery. World, J. Gastroenterol. 2006, 12, 3196–3203. [Google Scholar] [CrossRef]
  114. De Luca, M.; Angrisani, L.; Himpens, J.; Busetto, L.; Scopinaro, N.; Weiner, R.; Sartori, A.; Stier, C.; Lakdawala, M.; Bhasker, A.G.; et al. Indications for Surgery for Obesity and Weight-Related Diseases: Position Statements from the International Federation for the Surgery of Obesity and Metabolic Disorders (IFSO). Obes. Surg. 2016, 26, 1659–1696. [Google Scholar] [CrossRef]
  115. Cummings, D.E.; Rubino, F. Metabolic surgery for the treatment of type 2 diabetes in obese individuals. Diabetologia 2018. [CrossRef] [PubMed] [Green Version]
  116. Santarpia, L.; Del Piano, C.; Amato, V.; Marra, M.; De Caprio, C.; De Rosa, E.; Contaldo, F.; Pasanisi, F. Impact of laparoscopic sleeve gastrectomy on metabolism and liver structure. Obes. Facts 2014. [Google Scholar]
  117. Bužga, M.; Zavadilová, V.; Holéczy, P.; Švagera, Z.; Švorc, P.; Foltys, A.; Zonča, P. Dietary intake and ghrelin and leptin changes after sleeve gastrectomy. Wideochir. Inne Tech. Maloinwazyjne 2014, 9, 554–561. [Google Scholar] [CrossRef] [Green Version]
  118. Chen, G.; Zhang, G.; Peng, B.; Cheng, Z.; Du, X. Roux-En-Y Gastric Bypass Versus Sleeve Gastrectomy Plus Procedures for Treatment of Morbid Obesity: Systematic Review and Meta-Analysis. Obes. Surg. 2021, 31, 3303–3311. [Google Scholar] [CrossRef] [PubMed]
  119. Laurenius, A.M.A.; Wallengren, O.; Alaraj, A.; Forslund, H.B.; Thorell, A.; Wallenius, V. Resolution of diabetes, gastrointestinal symptoms, and self-reported dietary intake after gastric bypass versus sleeve gastrectomy: A randomized study. Surg. Obes. Relat. Dis. 2022; in press. [Google Scholar] [CrossRef]
  120. Strain, G.W.; Torghabeh, M.H.; Gagner, M.; Ebel, F.; Dakin, G.F.; Abelson, J.S.; Connolly, D.; Pomp, A. The Impact of Biliopancreatic Diversion with Duodenal Switch (BPD/DS) Over 9 Years. Obes. Surg. 2017, 27, 787–794. [Google Scholar] [CrossRef] [PubMed]
  121. Pereira, S.S.; Guimarães, M.; Almeida, R.; Pereira, A.M.; Lobato, C.B.; Hartmann, B.; Hilsted, L.; Holst, J.J.; Nora, M. Biliopancreatic diversion with duodenal switch (BPD-DS) and single-anastomosis duodeno-ileal bypass with sleeve gastrectomy (SADI-S) result in distinct post-prandial hormone profiles. Int. J. Obes. 2019, 43, 2518–2527. [Google Scholar] [CrossRef] [PubMed]
  122. Mitzman, B.; Cottam, D.; Goriparthi, R.; Cottam, S.; Zaveri, H.; Surve, A.; Roslin, M.S. Stomach Intestinal Pylorus Sparing (SIPS) Surgery for Morbid Obesity: Retrospective Analyses of Our Preliminary Experience. Obes. Surg. 2016, 26, 2098–2104. [Google Scholar] [CrossRef]
  123. Neichoy, B.T.; Schniederjan, B.; Cottam, D.R.; Surve, A.K.; Zaveri, H.M.; Cottam, A.; Cottam, S. Stomach intestinal pylorus-sparing surgery for morbid obesity. J. Soc. Laparoendosc. Surg. 2018, 22, e2017.00063. [Google Scholar] [CrossRef] [PubMed]
  124. Monkhouse, S.J.W.; Morgan, J.D.T.; Norton, S.A. Complication of bariatric surgery: Presentation and emergency management—A review. Ann. R. Coll. Surg. Engl. 2009, 91, 280–286. [Google Scholar] [CrossRef]
  125. Decker, G.A.; Swain, J.M.; Crowell, M.D.; Scolapio, J.S. Gastrointestinal and nutritional complications after bariatric surgery. Am. J. Gastroenterol. 2007, 102, 2571–2580. [Google Scholar] [CrossRef] [PubMed]
  126. Collazo-Clavell, M.L.; Shah, M. Common and Rare Complications of Bariatric Surgery. Endocrinol. Metab. Clin. N. Am. 2020, 49, 329–346. [Google Scholar] [CrossRef] [PubMed]
  127. Tjeertes, E.E.K.M.; Hoeks, S.S.E.; Beks, S.S.B.J.C.; Valentijn, T.T.M.; Hoofwijk, A.A.G.M.; Stolker, R.J.R.J. Obesity—A risk factor for postoperative complications in general surgery? BMC Anesthesiol. 2015, 15, 112. [Google Scholar] [CrossRef] [Green Version]
  128. Rashti, F.; Gupta, E.; Ebrahimi, S.; Shope, T.R.; Koch, T.R.; Gostout, C.J. Development of minimally invasive techniques for management of medically-complicated obesity. World J. Gastroenterol. 2014, 20, 13424–13445. [Google Scholar] [CrossRef] [PubMed]
  129. Bhandari, M.; Fobi, M.A.L.; Buchwald, J.N. Bariatric Metabolic Surgery Standardization (BMSS) Working Group. Standardization of Bariatric Metabolic Procedures: World Consensus Meeting Statement. Obes. Surg. 2019, 29, 309–345. [Google Scholar] [CrossRef] [Green Version]
  130. Al-Mulhim, A.S.; Al-Hussaini, H.A.; Al-Jalal, B.A.; Al-Moagal, R.O.; Al-Najjar, S.A. Obesity Disease and Surgery. Int. J. Chronic Dis. 2014, 2014, 652341. [Google Scholar] [CrossRef]
  131. Král, J.; Machytka, E.; Horká, V.; Selucká, J.; Doleček, F.; Špičák, J.; Kovářová, V.; Haluzík, M.; Bužga, M. Endoscopic treatment of obesity and nutritional aspects of bariatric endoscopy. Nutrients 2021, 13, 4268. [Google Scholar] [CrossRef]
  132. Picot, J.; Jones, J.; Colquitt, J.L.; Gospodarevskaya, E.; Loveman, E.; Baxter, L.; Clegg, A.J. The clinical effectiveness and cost-effectiveness of bariatric (weight loss) surgery for obesity: A systematic review and economic evaluation. Health Technol. Assess. 2009, 13. [Google Scholar] [CrossRef] [Green Version]
  133. Zurawel, R.; Gluck, M.; Piecuch, J.; Nowowiejska-Wiewiora, A.; Niedziela, J.; Wiewiora, M. Effectiveness and Safety of Adjustable Gastric Banding in Morbidly Obese Patients After 5 Years of Follow-up. Indian, J. Surg. 2021, 77, 853–862. [Google Scholar] [CrossRef]
  134. Mandrioli, M.; Inaba, K.; Piccinini, A.; Biscardi, A.; Sartelli, M.; Agresta, F.; Catena, F.; Cirocchi, R.; Jovine, E.; Tugnoli, G.; et al. Advances in laparoscopy for acute care surgery and trauma. World J. Gastroenterol. 2016, 22, 668–680. [Google Scholar] [CrossRef]
  135. Torgerson, J.S.; Sjöström, L. The Swedish Obese Subjects (SOS) study—Rationale and results. Int. J. Obes. 2001, 25, S2–S4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Scheen, A.J.; Letiexhe, M.; Rorive, M.; De Flines, J.; Luyckx, F.H.; Desaive, C. Bariatric surgery: 10-Year results of Swedish Obese Subjects Study. Rev. Med. 2005, 60, 121–125. [Google Scholar]
  137. Wiggins, T.; Guidozzi, N.; Welbourn, R.; Ahmed, A.R.; Markar, S.R. Association of bariatric surgery with all-cause mortality and incidence of obesity-related disease at a population level: A systematic review and meta-analysis. PLoS Med. 2020, 17, e1003206. [Google Scholar] [CrossRef] [PubMed]
  138. Mechanick, J.I.; Apovian, C.; Brethauer, S.; Garvey, W.T.; Joffe, A.M.; Kim, J.; Kushner, R.F.; Lindquist, R.; Pessah-Pollack, R.; Seger, J.; et al. Clinical Practice Guidelines for the Perioperative Nutrition, Metabolic, and Nonsurgical Support of Patients Undergoing Bariatric Procedures—2019 Update: Cosponsored by American Association of Clinical Endocrinologists/American College of Endocrinology. Endocr. Pract. 2019, 25, 1–75. [Google Scholar] [CrossRef] [PubMed]
  139. Müller, T.D.; Blüher, M.; Tschöp, M.H.; DiMarchi, R.D. Anti-obesity drug discovery: Advances and challenges. Nat. Rev. Drug Discov. 2022, 21, 201–223. [Google Scholar] [CrossRef]
  140. Apovian, C.M.; Aronne, L.J.; Bessesen, D.H.; McDonnell, M.E.; Murad, M.H.; Pagotto, U.; Ryan, D.H.; Still, C.D.; Endocrine Society. Pharmacological management of obesity: An endocrine society clinical practice guideline. J. Clin. Endocrinol. Metab. 2015, 100, 342–362. [Google Scholar] [CrossRef] [Green Version]
  141. Bray, G.A. Drug treatment of obesity. Psychiatr. Clin. N. Am. 2005, 28, 193–217. [Google Scholar] [CrossRef]
  142. Van Dieren, S.; Czernichow, S.; Chalmers, J.; Kengne, A.P.; de Galan, B.E.; Poulter, N.; Woodward, M.; Beulens, J.W.; Grobbee, D.E.; van der Schouw, Y.T.; et al. Weight changes and their predictors amongst 11 140 patients with type 2 diabetes in the ADVANCE trial. Diabetes Obes. Metab. 2012, 14, 464–469. [Google Scholar] [CrossRef]
  143. Turenius, C.I.; Htut, M.M.; Prodon, D.A.; Ebersole, P.L.; Ngo, P.T.; Lara, R.N.; Wilczynski, J.L.; Stanley, B.G. GABAA receptors in the lateral hypothalamus as mediators of satiety and body weight regulation. Brain Res. 2009, 1262, 16–24. [Google Scholar] [CrossRef]
  144. Garvey, W.T.; Ryan, D.H.; Look, M.; Gadde, K.M.; Allison, D.B.; Peterson, C.A.; Schwiers, M.; Day, W.W.; Bowden, C.H. Two-year sustained weight loss and metabolic benefits with controlled-release phentermine/topiramate in obese and overweight adults (SEQUEL): A randomized, placebo-controlled, phase 3 extension study. Am. J. Clin. Nutr. 2012, 95, 297–308. [Google Scholar] [CrossRef]
  145. Drent, M.L.; Popp-Snijders, C.; Adér, H.J.; Jansen, J.B.M.J.; van der Veen, E.A. Lipase Inhibition and Hormonal Status, Body Composition and Gastrointestinal Processing of a Liquid High-Fat Mixed Meal in Moderately Obese Subjects. Obes. Res. 1995, 3, 573–581. [Google Scholar] [CrossRef] [PubMed]
  146. McDuffie, J.R.; Calis, K.A.; Booth, S.L.; Uwaifo, G.I.; Yanovski, J.A. Effects of orlistat on fat-soluble vitamins in obese adolescents. Pharmacotherapy 2002, 22, 814–822. [Google Scholar] [CrossRef] [PubMed]
  147. Ioannides-Demos, L.L.; Piccenna, L.; McNeil, J.J. Pharmacotherapies for obesity: Past, current, and future therapies. J. Obes. 2011, 2011, 179674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Thomsen, W.J.; Grottick, A.J.; Menzaghi, F.; Reyes-Saldana, H.; Espitia, S.; Yuskin, D.; Whelan, K.; Martin, M.; Morgan, M.; Chen, W.; et al. Lorcaserin, a novel selective human 5-hydroxytryptamine2C agonist: In vitro and in vivo pharmacological characterization. J. Pharmacol. Exp. Ther. 2008, 325, 577–587. [Google Scholar] [CrossRef] [Green Version]
  149. Smith, S.R.; Weissman, N.J.; Anderson, C.M.; Sanchez, M.; Chuang, E.; Stubbe, S.; Bays, H.; Shanahan, W.R. Behavioral Modification and Lorcaserin for Overweight and Obesity Management (BLOOM) Study Group. Multicenter, Placebo-Controlled Trial of Lorcaserin for Weight Management. N. Engl. J. Med. 2010, 363, 245–256. [Google Scholar] [CrossRef] [Green Version]
  150. Toft-Nielson, M.; Madsbad, S.; Holst, J.J. The Effect of Glucagon-Like Peptide I (GLP-I) on Glucose Elimination in Healthy Subjects Depends on the Pancreatic Glucoregulatory Hormones. Diabetes 1996, 45, 552–556. [Google Scholar] [CrossRef]
  151. Flint, A.; Raben, A.; Astrup, A.; Holst, J.J. Glucagon-like peptide 1 promotes satiety and suppresses energy intake in humans. J. Clin. Investig. 1998, 101, 515–520. [Google Scholar] [CrossRef]
  152. Schirra, J.; Nicolaus, M.; Roggel, R.; Katschinski, M.; Storr, M.; Woerle, H.J.; Göke, B. Endogenous glucagon-like peptide 1 controls endocrine pancreatic secretion and antro-pyloro-duodenal motility in humans. Gut 2006, 55, 243–251. [Google Scholar] [CrossRef] [Green Version]
  153. Pi-Sunyer, X.; Astrup, A.; Fujioka, K.; Greenway, F.; Halpern, A.; Krempf, M.; Lau, D.C.; le Roux, C.W.; Violante Ortiz, R.; Jensen, C.B.; et al. A Randomized, Controlled Trial of 3.0 mg of Liraglutide in Weight Management. N. Engl. J. Med. 2015, 373, 11–22. [Google Scholar] [CrossRef]
  154. Pi-Sunyer, X. The medical risks of obesity. Postgrad. Med. 2009, 121, 21–33. [Google Scholar] [CrossRef]
  155. Pan, A.; Sun, Q.; Czernichow, S.; Kivimaki, M.; Okereke, O.I.; Lucas, M.; Manson, J.E.; Ascherio, A.; Hu, F.B. Bidirectional association between depression and obesity in middle-aged and older women. Int. J. Obes. 2012, 36, 595–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Greenway, F.L.; Fujioka, K.; Plodkowski, R.A.; Mudaliar, S.; Guttadauria, M.; Erickson, J.; Kim, D.D.; Dunayevich, E.; COR-I Study Group. Effect of naltrexone plus bupropion on weight loss in overweight and obese adults (COR-I): A multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2010, 376, 595–605. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Interaction between molecules and miRNAs. ADSC exposure to metformin and vitamin D modulate miRNAs profile, inducing the upregulation of miR-145 and the simultaneously downregulation of miR-148. The final effect is the inhibition of adipogenesis. Created with BioRender.com accessed on 30 December 2022.
Figure 1. Interaction between molecules and miRNAs. ADSC exposure to metformin and vitamin D modulate miRNAs profile, inducing the upregulation of miR-145 and the simultaneously downregulation of miR-148. The final effect is the inhibition of adipogenesis. Created with BioRender.com accessed on 30 December 2022.
Ijms 24 02310 g001
Figure 2. Some therapeutical approaches for management of obesity. Created with BioRender.com accessed on 30 December 2022.
Figure 2. Some therapeutical approaches for management of obesity. Created with BioRender.com accessed on 30 December 2022.
Ijms 24 02310 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cruciani, S.; Delitala, A.P.; Cossu, M.L.; Ventura, C.; Maioli, M. Management of Obesity and Obesity-Related Disorders: From Stem Cells and Epigenetics to Its Treatment. Int. J. Mol. Sci. 2023, 24, 2310. https://doi.org/10.3390/ijms24032310

AMA Style

Cruciani S, Delitala AP, Cossu ML, Ventura C, Maioli M. Management of Obesity and Obesity-Related Disorders: From Stem Cells and Epigenetics to Its Treatment. International Journal of Molecular Sciences. 2023; 24(3):2310. https://doi.org/10.3390/ijms24032310

Chicago/Turabian Style

Cruciani, Sara, Alessandro Palmerio Delitala, Maria Laura Cossu, Carlo Ventura, and Margherita Maioli. 2023. "Management of Obesity and Obesity-Related Disorders: From Stem Cells and Epigenetics to Its Treatment" International Journal of Molecular Sciences 24, no. 3: 2310. https://doi.org/10.3390/ijms24032310

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop