Next Article in Journal
A Pilot Study of Multiplex Ligation-Dependent Probe Amplification Evaluation of Copy Number Variations in Romanian Children with Congenital Heart Defects
Next Article in Special Issue
FOXP3: A Player of Immunogenetic Architecture in Lung Cancer
Previous Article in Journal
Differences in Immune Characteristics and Related Gene Expression in Spleen among Ningxiang, Berkshire Breeds and Their Hybrid Pigs
Previous Article in Special Issue
CCND1 Overexpression in Idiopathic Dilated Cardiomyopathy: A Promising Biomarker?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunogenetic Aspects of Sarcopenic Obesity

by
Łukasz Mazurkiewicz
,
Krystian Czernikiewicz
and
Bogna Grygiel-Górniak
*
Department of Rheumatology, Rehabilitation and Internal Diseases, Poznan University of Medical Sciences, 61-701 Poznan, Poland
*
Author to whom correspondence should be addressed.
Genes 2024, 15(2), 206; https://doi.org/10.3390/genes15020206
Submission received: 2 January 2024 / Revised: 28 January 2024 / Accepted: 1 February 2024 / Published: 5 February 2024
(This article belongs to the Special Issue New Advances in Immunogenetics of Disease)

Abstract

:
Sarcopenic obesity (SO) is a combination of obesity and sarcopenia, with diagnostic criteria defined as impaired skeletal muscle function and altered body composition (e.g., increased fat mass and reduced muscle mass). The mechanism of SO is not yet perfectly understood; however, the pathogenesis includes aging and its complications, chronic inflammation, insulin resistance (IR), and hormonal changes. Genetic background is apparent in the pathogenesis of isolated obesity, which is most often polygenic and is characterized by the additive effect of various genetic factors. The genetic etiology has not been strictly established in SO. Still, many data confirm the existence of pathogenic gene variants, e.g., Fat Mass and Obesity Associated Gene (FTO), beta-2-adrenergic receptor (ADRB2) gene, melanocortin-4 receptor (MC4R) and others with obesity. The literature on the role of these genes is scarce, and their role has not yet been thoroughly established. On the other hand, the involvement of systemic inflammation due to increased adipose tissue in SO plays a significant role in its pathophysiology through the synthesis of various cytokines such as monocyte chemoattractant protein-1 (MCP-1), IL-1Ra, IL-15, adiponectin or CRP. The lack of anti-inflammatory cytokine (e.g., IL-15) can increase SO risk, but further studies are needed to evaluate the exact mechanisms of implications of various cytokines in SO individuals. This manuscript analyses various immunogenetic and non-genetic factors and summarizes the recent findings on immunogenetics potentially impacting SO development.

1. Introduction

Sarcopenic obesity (SO) involves both the gain of fat mass (obesity), and sarcopenia is defined as low muscle strength resulting from low muscle quantity or quality [1,2]. The previous lack of universal diagnostic criteria made it difficult to estimate SO’s prevalence and to plan treatment strategies [1].
The published standards by the European Society for Clinical Nutrition and Metabolism (ESPEN) and the European Association for the Study of Obesity (EASO) allow us to gather more information about this condition, which is typical for an aging society [1]. The current definition of SO includes the coexistence of obesity and sarcopenia, and the diagnostic criteria are based on impaired skeletal muscle function and altered body composition–like increased fat mass and reduced muscle mass [1]. Two recent meta-analyses estimated the prevalence of SO as between 9% and 11%; however, the lack of universal SO definition and the incoherence of definitions used to define obesity or sarcopenia separately, make the SO prevalence still inconclusive and needing further investigation [3,4,5]. The prevalence of SO increases with age and is associated with accumulation of chronic diseases—for example fractures, cardiovascular diseases or cancer [1,6,7]. Higher all-cause mortality rate in women suffering from SO was observed and a higher risk of death within 10 years in 75-years-old SO women was reported [7,8]. Pathogenesis of SO includes aging, strictly associated with physical inactivity and often malnutrition, as well as chronic inflammation, insulin resistance (IR), and hormonal changes [9]. The mechanism of SO is not yet perfectly understood; however, it is a complex condition and is characterized by both obesity and sarcopenia, with overlapping features [10]. The aim of this review is to indicate the multifactorial and immunogenetic nature of SO, which involves both genetic predisposition and immune system involvement in the pathophysiology of the disease.

2. Materials and Methods

Publicly available databases on medical sciences, like PubMed or Google Scholar, were searched through to identify records published between 2014–2023. The search strategy included main words—sarcopenia and obesity—and other keywords related to the pathogenesis of obesity, sarcopenia, and SO, reaching the major areas such as prevalence, mechanisms, immunology, and genetics. Various connections between keywords were made, combined with obesity or sarcopenia, using the conjunction words like AND or OR. All article types were considered if only the main search criteria were met. At first, titles were screened for accuracy. Articles that qualified for evaluation were analyzed through the abstract and the whole text. Manuscripts in English and Polish language were included in this analysis. Exceptionally, besides the search criteria, additional articles from the research field describing crucial findings were included regardless of the publication year. The search strategy is presented in Figure 1.

3. Obesity and Sarcopenia—What Came First: The Egg or the Chicken?

Obesity is a complex multifactorial disease characterized by accumulated excess body fat, negatively affecting health [11]. Obesity in most of the global population is defined as a BMI value equal to or over 30 kg/m2, while for Asians, it is equal to or above 25 kg/m2 [11,12]. In turn, sarcopenia is defined as loss of muscle mass and strength or physical function, which naturally occurs in aging. This condition is typical for a geriatric population, and its prevalence increases with age [2,13,14]. Sarcopenia is related to frailty and multi-morbidity and is often observed in users of residential aged care (nursing home) services [15,16]. Since sarcopenia is a highly prevalent condition in older people and is related to disability, hospitalization, and death, quick diagnosis and adequate prevention and treatment should be implemented [17].
The definition of sarcopenia has developed over the last two decades. The original meaning of this condition only included reduced muscle mass. With modern anthropometric techniques and imaging capabilities, the definition of sarcopenia has evolved (Table 1).
New tools and methods increase the possibility of sarcopenia diagnosis more precisely, permitting comparisons between different social groups [22] (Table 2). This was a milestone that opened up the possibility of diagnosing sarcopenia in younger age groups and, consequently, increased scientific curiosity about which risk factors are related to its development (Figure 2). Since then, sarcopenia has also been reported in young people who had low physical activity associated with long-term immobilization [2,23,24]. However, determining the prevalence of sarcopenia in the young population is challenging; however, based on the evidence, more than one subject in every ten young adults of most ethnicities is estimated to have sarcopenia.
Conversely to obesity assessment, defining sarcopenia is difficult because of technical problems in the accurate evaluation of muscle mass and muscle quality [25,26,27,28]. In the literature, there is a broad discussion about the techniques of muscle mass and strength estimation; however, recent data confirm that the estimation of muscle strength better than mass predicts adverse outcomes of sarcopenia [24,29]. Unfortunately, there is no international consensus on a definition of sarcopenia. Many reports attempt to describe this phenomenon, as exemplified by consensuses published by the European Working Group on Sarcopenia in Older People (EWGSOP1 and EWGSOP2) [2,13], the Foundation for the National Institutes of Health (FNIH) [20] or Australian and New Zealand Society for Sarcopenia and Frailty Research (ANZSSFR) [21] (Table 1). Therefore, there are many delineations in sarcopenia definitions, although most are convergent and consider low muscle mass and low muscle strength or performance as the exponent of sarcopenia. Uncertainties in sarcopenia diagnosis are also related to the evaluation methods, which differ in how they assess the quantity and quality of muscles and propose different points (Table 2). The EWGSOP suggests that muscle mass should be expressed relative to height, while the FNIH recommends adjustment by BMI. Such discrepancies contribute to inconsistencies in estimating the prevalence of sarcopenia and cause difficulties in comparison of various populations [30,31,32].
Table 2. Methods of muscle mass estimation.
Table 2. Methods of muscle mass estimation.
Muscle Mass Estimation and Cut-Off Points in Sarcopenic Patients
MethodCriteriaReference
AnthropometricReference population: Rosetta study [18]—a population-based survey of 883 elderly Hispanic and non-Hispanic white men and women living in New Mexico

ALM/ht2
  • M ≤ 7.26 kg/m2
  • F ≤ 5.45 k/m2
[18,19]
Dual X-ray absorptiometry—DXAreference population: Rosetta study [18]

Sarcopenia Index
ALM/ht2
  • M ≤ 7.26 kg/m2
  • F ≤ 5.45 k/m2
[33,34,35]
STUDY: Health ABC—the Health Aging and Body Composition
Sarcopenia index: ALM/ht2
  • M ≤ 7.23 kg/m2
  • F ≤ 5.67 kg/m2
[36]
DXA—less than the twentieth percentile of healthy adults
Sarcopenia index: ALM/ht2
  • M ≤ 7.23 kg/m2
  • F ≤ 5.67 kg/m2
[17]
Bioelectrical impedance—BIASTUDY: NHANES III—the Third National Health and Nutrition Examination Survey
Sarcopenia definition: ratio of muscle mass/total body mass
  • M ≤ 31.5%
  • F ≤ 22.1%
[37]
Study population: subjects who attended a 1988–1992 Rancho Bernardo Study clinic

Sarcopenia definition: the value of FFM of > or =2.0 standard deviations below the gender-specific mean of a young reference population (Mean FFM was 43.5 kg for women and 61.7 kg for men.)

Sarcopenia index—Total lean mass/ht2
M ≤ 8.50 kg/m2
F ≤ 5.75 kg/m2
[38]
STUDY: NHANES III—the Third National Health and Nutrition Examination Survey

Sarcopenia definition: total muscle mass/ht2:
  • M ≤ 8.50 kg/m2
  • F ≤ 5.75 kg/m2
[39]
STUDY: Cardiovascular Health Study

Sarcopenia definition: fat-free mass:
  • M ≤ 47.9 kg
  • F ≤ 34.7 kg
[40]
Estimation of cut-off points for SO
dual X-ray absorptiometry—DXAAnalysis of two definitions of SO:

New Mexico Elder Health Survey
  • Obesity definition: BMI ≥ 30 kg/m2
  • Sarcopenia assessment: ALM divided by height squared (M < 7.23 kg/m2 and F < 5.67 kg/m2)

Health ABC study
  • Obesity definition: BMI ≥ 30 kg/m2
  • ALM divided by height and fat mass
  • DXA—lowest twentieth percentile of residuals (sex-specific)
Sarcopenia, defined by these two definitions, has differential prevalence rates by obesity status, which underlines the necessity of fat mass assessment in evaluating sarcopenia.
[36]
New Mexico Elder Health Survey:
  • Obesity definition: body fat: M ≥ 28% and F ≥ 40%
  • Sarcopenia assessment: ALM divided by height squared
  • DXA: M < 7.26 kg/m2 and F < 5.45 kg/m2
[41]
Nutrition as a Determinant of Successful Aging study
  • Obesity definition: Body fat (M ≥28% and F ≥35%)
  • Sarcopenia assessment: ALM divided by height squared
  • DXA: M < 8.51 kg/m2 and F < 6.29 kg/m2
[42]
ALM = appendicular lean mass; ALM/ht2—appendicular lean mass divided by height squared; DXA—dual X-ray absorptiometry; FFM—free fat mass; M—male; F—female.
In the literature SO is defined as excess fat mass and sarcopenia [43]. Nevertheless, obesity is a separate syndrome, and not each type of obesity is related to sarcopenia. Since body mass loss results in a decrease in both fat mass and lean body mass, it is possible that decreasing weight in obese older persons could worsen the age-related loss of muscle mass and increase sarcopenia [43]. Furthermore, there is a synergistic adverse effect of sarcopenia and obesity, particularly in older populations, which results in an increasing prevalence of SO [44]. It is also worth underlining that synergistic problems of sarcopenia and obesity lead to impairments in instrumental activities of daily living [41].
The definitions of SO have been proposed by two associations: the European Society for Clinical Nutrition and Metabolism (ESPEN) and the European Association for the Study of Obesity (EASO). According to the initiative of these societies, SO is defined as the co-existence of excess adiposity and low muscle mass/function. The diagnosis of SO should be considered in at-risk individuals with elevated BMI or waist circumference and markers of low skeletal muscle mass and function. It is extremely important to determine associated comorbidities. Diagnosis of SO should be confirmed by decreased skeletal muscle function and improper body composition (excess adiposity and low skeletal muscle mass). Each SO patient should have been screened for possible related-body composition clinical complications [1].
The synergistic risk factors of the development of obesity and sarcopenia at a young age are similar in adults and older people and include physical inactivity, a not-well-balanced diet (e.g., a pro-inflammatory diet deficient in antioxidant vitamins A and E), metabolic syndrome, vitamin D deficiency, inherent and perinatal factors, gut microbiota dysbiosis, endocrinopathy, malignancy, and other inflammatory disorders [23]. The similar pathological pathways suggest the relationship between both obesity and sarcopenia, and the development of one condition may increase the risk of another one; however, this hypothesis requires future studies. It is worthy to underline that the phenotype of sarcopenia has many contributing causes unrelated to aging [45,46]. Such implications indicate that the well-described obesity-related genes may also influence the sarcopenic genotype. Such implications suggest that well-described obesity-related genes may be valuable information for searching for the causes of SO and enabling individualized therapy planning.

4. Immune Involvement in Sarcopenic Obesity

4.1. Pro-Inflammatory Cytokine Synthesis and Their Relation to Sarcopenic Obesity

Increased adipose tissue causes systemic inflammation, which plays a major role in the pathophysiology of SO due to the effect on skeletal muscles and the decrease in oxidative capacity [47]. Both obesity and sarcopenia are strictly connected with chronic inflammation [48,49]. Obesity leads to increased adipocyte macrophage infiltration, resulting in further adipokine secretion. Adipose tissues change their physiological activity and release excess free fatty acids, reactive oxygen species, and pro-inflammatory cytokines [50]. Ectopic fat deposition results in organelle damage and dysfunction, causing IR [50]. Additionally, both the production and efficiency of insulin decrease with age. Additionally, insulin low-grade inflammation and cytokine synthesis decrease insulin sensitivity. With aging, obesity associated with insulin resistance leads to muscle catabolism, reduction in muscle strength, and, finally, SO [9].
MCP-1 plays a major role in the pathogenesis of obesity [51]. A study by Huber et al. demonstrated elevated gene expression of CC chemokines (a family of chemokines that attract inflammatory mononuclear cells to the inflammation sites) and their receptors, including MCP-1 and C-C motif chemokine receptor 2 (CCR2) in subcutaneous and visceral adipose tissue in obese patients [52]. MCP-1 helps the CCR2-positive monocytes to recruit and infiltrate the adipose tissues, which are transformed into M1 macrophages [50,53,54]. M1 macrophages are responsible for synthesizing TNF-α, IL-6, and MCP-1 [54]. This results in further cytokine expression [50]. IL-6 levels, IL-1 receptor antagonist (IL-1Ra), and soluble IL-6 receptor levels are elevated, which is crucial in SO development [49].
Physiologically, muscle-produced IL-6 positively impacts muscle regeneration and hypertrophy; however, in persistent inflammatory conditions, like the obese state, it acts as a pro-inflammatory, activating the MAP3K8 expression, further influencing NF-kB and JNK transcription factors, leading to IR [10,55,56]. Excessive secretion of IL-6 is observed in adiposity. Moreover, the IL-6-174G>C polymorphism in the IL-6 promoter region significantly increases the risk of obesity [57]. Other IL-6 gene polymorphisms, such as rs1800795(G/C) and rs1800796(G/C), are the risk factors for obesity and are considered potential new targets for drug inventions in obesity treatment [58].
Another cytokine secreted in obesity is IL-1Ra. This cytokine is a natural antagonist in the IL-1 family and reveals a counterregulatory (anti-inflammatory) function. The disproportions between pro-inflammatory IL-1 and anti-inflammatory IL-1Ra lead to the type 1 diabetes mellitus (T1DM) [59].
The plasma level of IL-15 decreases in sarcopenia [10]. The function of this cytokine impacts the anabolic effect on muscle growth [56]. It has been documented that the older people (above 95 years of age) living independently have greater levels of IL-15 than the two control groups—those between 30–59 years of age and 60–89 years of age. Thus, IL-15 has a positive impact on aging [60]. This cytokine stimulates the upregulation of the pro-oxidative PPARδ and SIRT1 benefit against obesity and IR [61]. Thus, IL-15 can be one of the beneficial factors during sarcopenia treatment related to advanced aging [10].
Adiponectin, a crucial player in obesity prevention, is responsible for insulin activity and regulating further pro- and anti-inflammatory cytokines. It enhances the production of IL-10 and IL-1Ra and reduces the amount of TNF-α [55,62,63]. The plasma concentration of adiponectin negatively correlates with body fat mass [63]. Another adipocytokine is leptin, which reveals pro-inflammatory activity and stimulates the production of IL-6, IL-12, and TNF-α. Its plasma concentration is positively correlated with body fat mass [62,63]. High levels of TNF-α and IL-6 are observed in sarcopenia and muscle weakness [56].
In summary, many cytokines participate in obesity pathogenesis. Some of them (like IL-6) have anti-inflammatory activity in the physiological state, while they gain pro-inflammatory properties in the obesity state [10]. The lack of anti-inflammatory cytokine (e.g., IL-15) can be one of the mechanisms leading to increased SO risk. Further investigations are needed to evaluate the exact mechanisms of various cytokines in SO individuals.

4.2. Hormonal Factors Related to Immune Response and Genetic Background

SO has been associated with hormonal factors. With age, the functioning of the human body, including its humoral aspects, undergoes changes that affect hormonal regulation. These systemic changes can be an underlying factor leading to the development of sarcopenia and SO [64].
Sex-specific hormonal changes characteristic of older adults are associated with SO [64,65]. Low estrogen levels after menopause are related to increased body weight. Also, the localization of adipose tissue is altered from subcutaneous to visceral [65]. Low estrogen levels are also associated with bone mass reduction [64]. In males, the testosterone level stimulates muscle synthesis [66]. In older males, decreasing testosterone levels correlate with lower muscle strength, muscle mass, poor physical performance, and SO [64,65,67].
Not only sex hormones, but also cortisol, growth hormone, insulin, and insulin-like hormones play a role in SO pathophysiology [64,66,68]. Insulin resistance and inflammation are present due to changes in signaling in hormonal pathways, leading to the elevation of cytokines and causing oxidative stress [66]. Insulin also increases muscle mass. This fact explains why patients with diabetes develop sarcopenia more rapidly [64].
Ghrelin is a hormone produced in the stomach, precisely in its funds. It is most notable for its stimulatory effect on growth hormone release, food intake increase, and regulation of fat deposition [69]. Ghrelin, ghrelin analogs, growth hormone, and insulin growth factor-1 (IGF-1) are reported to increase muscle mass but not muscle strength [64]. Thus, the decreased ghrelin level can influence the risk of SO. Another factor participating in muscle condition is myostatin, which acts as a regulator for skeletal muscle growth. Its main effects are muscle atrophy and cachexia [70]. Low myostatin levels are associated with muscle development [64]. Thus, anti-myostatin antibodies can used for SO therapy [68].
Due to the role of hormones in the development of SO, experimental hormonal therapies have been undertaken to treat SO. Recent reports show the experimental attempts to use growth hormone, growth hormone-releasing hormone analogs, and hormone replacement therapy (HRT) [68]. In postmenopausal women, HRT increases muscle strength (especially estrogen use) [64].

4.3. Infections during Childhood and the Risk of Obesity

Obesity results from the interaction between genetic, environmental factors, and diet [71]. Unfortunately, childhood infections treated using antibiotics are related to microbial resistance to available agents [72]. Thus, appropriate management of antibiotic therapy is of first importance to prevent potential drug resistance, but that is an important modifiable risk factor for obesity during childhood exposure [71,73]. The crucial criteria include the first six months after delivery and the repeated use of more than one antibiotic during this period [73]. Antibiotic use influences gut microbiota composition and maturation and changes the metabolism path of carbohydrates to short-chain fatty acids [74,75]. Another hypothesis is the disturbed mitochondrial function, which leads to the reduction of mitofusin 2 (Mfn2) [71,76]. A decrease in the amount of Mfn2, which plays a role in obesity development, could have an impact on the epidemic of childhood obesity worldwide [71,73].
An inflammatory marker implicated in SO is C-reactive protein (CRP), and its relationship with SO in specific populations was observed [47,77]. CRP levels, as well as IL-6 levels, increase with age. Fortunately, increased physical activity reduces the CRP level [78]. CRP may also be a mortality predictor, especially in older patients unrelated to SO [79].
Ohler and Braddock suggest that infections are stronger risk factors for childhood obesity than the use of antibiotics [80]. Some of the adenoviruses increase adiposity in animals and affect obesity in humans. For example, a protein of Ad-36—namely the early 4 open reading frame 1 (E4-ORF1)—is necessary for causing acute adipogenic effects [81,82]. One of the most important mechanisms is that the virus activates PPARγ, leading to increased adiposity by committing adult stem cells into adipocytes [82,83]. Another important protein necessary for maintaining Ad-36-induced obesity is monocyte chemoattractant protein-1 (MCP-1) [82]. Another example of viral infections affecting obesity are those caused by herpesviridae—especially CMV, being lipogenic and affecting various parameters of metabolic syndrome, like blood pressure after infection [82].

4.4. The Role of Maternal HLA in Sarcopenic Obesity

The human leukocyte antigen (HLAs) complex mediates a chronic inflammatory pathway. It is widely known to be associated with specific autoimmune diseases, including spondyloarthritis, Behçet’s disease, or multiple sclerosis [84]. A study concerning older adults with autoimmune diseases revealed a strong association between sarcopenia and autoimmune disease. However, investigating various HLA carriers with no autoimmune disease diagnosis unveiled an association of various HLA types, like HLA-DQA1*03:01 and HLA-DRB4*01:03, with sarcopenia. Additionally, some of them were more prevalent in women [84]. A strong association with muscle weakness was also found near the HLA-DQA1 gene, known to be a rheumatoid entity [85]. Single nucleotide polymorphisms associated with SO were also found in genes related to immune surveillance: HLA-DRB1 and HLA-DRB5 [67]. HLA-DQB1-AS1 seems to be associated with hand grip strength and is one of the genes that need further study [86].

4.5. The Link between Genetic Background and Inflammation in Sarcopenic Obesity

Though the macrophage population is a mix of M1 and M2 macrophages, the larger degree of obesity is associated with more accumulated adipose tissue macrophages and their activation, which is triggered by various inflammatory mediators—like CCR2—chemokine leading to organizing into an inflammatory M1 phenotype [87]. FTO plays a positive role in macrophage activation. However, FTO is downregulated in both subtypes but promotes polarization in both groups, which is essential for maintaining both phenotypes [88]. Another study demonstrated that FTO is a transcriptional suppressor of IFN-stimulated genes by suppressing STAT3 transcription factor activation. This path also refers to some genes encoding pro-inflammatory factors [89]. STAT3 itself was reported not only to both promote and inhibit oncogenesis in various mechanisms but also to have an impact on immune suppression and inflammation [90]. For example, it enhances the production of IL-10, which suppresses the production of pro-inflammatory cytokines [90]. A study by Dubey et al. demonstrated that in mice models, increased methylation of m6A-RNA and a decrease in FTO expression in the myocardium led to an increase in the expression of inflammatory myocardial cytokine genes—like IL-6, TNF-α, and IL-1β [91].
The expression of PPARγ is recently described to be essential in the differentiation processes of various immune cells—like macrophages or dendritic cells [92]. Like FTO, PPARγ plays a role in the polarization process of macrophages, favoring the alternative M2 phenotype [93].
Specific MTHFR SNPs have been linked to both sarcopenia and obesity (Figure 3), leading to a hypothesis that these polymorphisms might play a role in SO. An MTHFR polymorphism has been associated with elevated IL-6 levels in a study by Araki et al. [94], suggesting a potential role of MTHFR polymorphism in pro-inflammatory cytokine secretion. An MTHFR polymorphism (rs1803311) has been positively linked to higher homocysteine levels, which promotes the elevation of pro-inflammatory markers such as TNF-α, IL-6, and IL-1β. Building on the established connection between folate levels and pro-inflammatory markers, it has been observed that supplementing folate through natural foods for an eight-week period resulted in a reduction of inflammatory markers and homocysteine levels [95].
IL-6 polymorphisms have been linked to obesity in numerous studies. Researchers place IL-6 polymorphisms, such as rs1800795 and rs1800796, as risk factors for obesity [58,96]. It is worth mentioning that some SNPs (rs1800797) are associated with a reduced risk of obesity [96].
Interleukin 1 gene family has also been associated with the risk of obesity. As shown in the study by Melo et al., there has been a positive link between eleven variants of the IL1B gene and an increased risk of obesity in children [97]. Research conducted by Maculewicz et al. showed the involvement of IL-1 family genes in obesity but emphasized that interactions between different polymorphisms should be sought [98].
Overall, the evidence on the correlation of gene polymorphisms (as depicted in Figure 3) and pro-inflammatory cytokine secretion is relatively scarce, leading to the conclusion that this should be a subject for further investigation.

5. Genetic Aspects of Sarcopenic Obesity

5.1. Genetic Susceptibility to Sarcopenic Obesity

Genetic factors play a major role in the pathogenesis of obesity. The cause of obesity is most often polygenic and is characterized by the additive effect of various genetic factors [99,100]. Genome Wide Association Studies (GWAS) are widely used to estimate genetic risk markers [101]. The difference between monogenic and polygenic obesity does not concern particular genes—the majority of the same genes play a role in both types of obesity. However, in polygenic obesity, the dysfunction of protein synthesis varies [102]. Obesity predisposition leads further to a pro-inflammatory state, resulting in the secretion of cytokines.
The identified genetic variants affecting the body mass are being used to make a polygenic risk score (PRO) to more accurately calculate overall genetic risks for obesity, simultaneously taking into account multiple gene variants [103,104]. For example, Tan and Mitra discovered higher odds of obesity in the third tertile of PRO regarding Fat Mass and Obesity Associated Gene (FTO) and beta-2-adrenergic receptor (ADRB2) gene variants [104]. Another study noticed the synergistic impact of other genes such as peroxisome proliferator-activated receptor gamma (PPARγ), FTO, and melanocortin-4 receptor (MC4R)—in the predisposition to overweight and obesity [105].
FTO initiates the onset of DM2 by increasing BMI value [100,106]. Its function is probably to regulate appetite and energy expenditure, as the highest levels are expressed in the hypothalamic regions [106,107]. It encodes a demethylase, being a family of AlkB family—ALKBH9 [107]. The main substrate is N6-methyladenosine (m6A), which plays a role in post-transcriptional regulatory processes [107]. An often studied SNP is rs9939609—homozygous for this variant in adults has a 1.67-fold increased risk of obesity [107]. Park and Choi found this dependence, especially in women [108]. Studies demonstrate that FTO SNPs may also be associated with lean mass index (LMI) and sarcopenia [109]. FTO rs9939609 AA homozygotes have been related to the risk for sarcopenia in older women [110]. These findings indicate this variant as a possible major factor in SO; however, to our knowledge, it is still unknown if FTO rs9939609 AA homozygotes are related to SO [109,110]. The same authors described endothelial nitric oxide synthetase gene—NOS3 (rs1799983 GG genotype) to increase the sarcopenia risk almost twofold. It is postulated that this genotype makes the nitric oxide effect on muscles less effective, which explains the GG genotype’s potential complicity in sarcopenia [110].
Another genetic factor that may influence SO development is MC4R gene. This gene mutation is a flagship gene determining monogenic obesity [99]. Leptin, together with neuropeptide Y (NPY) and agouti-related peptide (AgRP), inhibits the appetite-stimulating system and activates the MC4R pathway, regulating hunger [111,112]. Most discovered MC4R mutations lead to a decrease in their function and result in obesity [99]. The melanocyte-stimulating hormone (MSH), a proopiomelanocortin (POMC) derived hormone, is stimulated by leptin, which helps reduce food intake. The POMC deficiency is rare [112]. MC4R agonist—setmelanotide is a promising agent in MC4R deficiency treatment, leading to significant weight loss [111,112]. To our knowledge, this genetic factor is not described in sarcopenia or SO. The rs17782313 T-allele reduces the incidence of lipid metabolism disorders and protects against metabolic disorders in Polish postmenopausal women [113]. A Chinese study confirms that the MC4R rs17782313 C/C genotype is associated with higher triglyceride (TG) levels in older Chinese women [114].
The beta-3 adrenergic receptor (ADRB3) gene regulates thermogenesis, and its expression is limited in the obese population [115]. Specific polymorphism of ADRB3 gene—Trp64Arg impacts adipokines and lipid levels (mainly LDL-cholesterol) and is associated with obesity [116,117]. SO is regulated by adipose tissue and skeletal muscle amount ratio, which changes during aging, and in this process, ADRB3 gene variants seem to play a crucial role. The function of this gene increases with age when adipose inflammation leads to the redistribution of fat to the intra-abdominal area and fatty infiltrations in skeletal muscles, resulting in decreased overall strength and functionality. Therefore, various metabolic alterations are observed, including insulin resistance. The expression of ADRB3 is regulated by the diet. The study of animal models has shown that 3-week supplementation with a phenolic-rich olive leaf extract (100 mg/kg to Wistar rats orally) attenuated the aging-induced alterations in body composition and insulin resistance. Olive leaf extract treatment downregulated the expression of ADRB3 and attenuated the aging-induced changes in the mRNA levels of insulin receptor and PPAR-γ [118].
ADRB3 gene changes are observed in human studies. In non-obese adolescents, the less frequent allele (Trp64Arg and Arg64Arg) is related to higher LDL-cholesterol levels and lower maximal fat oxidation rates when compared with non-carriers (Trp64Trp) [117]. With age, the role of this ADRB3 genotype increases, which reflects the tendency for increased worldwide obesity prevalence in an aging society. In the study of women with breast cancer, it was observed that homozygous individuals carrying the ADRB3 wild-type allele exhibited significantly higher mean visceral fat levels compared to those with the variant allele, indicating a greater degree of obesity [119]. Overweight and obese women with Trp64Trp of ADRB gene polymorphism and normal TG levels are mainly normoglycemic, indicating this polymorphism’s beneficial influence on metabolic parameters [120]. Conversely, Arg64/X polymorphism of the ADRB3 gene is related to dyslipidemia, particularly if nutrient intake is high which has proatherogenic effect due to overconsumption of fat, arachidic acid and overall high energy intake [121]. A similar relation is presented in the de Luis DA et al. study, which shows that obese subjects with the Arg64/X allele are predisposed to lipid disorders (higher TC, LDL, and TG) [122]. Not only in Caucasian populations [123] but also in Asian entities [124], the Arg64 allele is associated with obesity development.
Nevertheless, epigenetic changes influencing the ADRB3 gene are also crucial. DNA hypermethylation has been discovered to be related to being overweight, obese, and having a higher waist-hip ratio [115]. Thus, specific ADRB3 gene polymorphism and epigenetic changes may impact sarcopenic development; however, data on the genetic involvement of ADRB3 in sarcopenia or SO is scarce.
The potential roles of amino acid transporters are constantly investigated, and several hypotheses are suggested [125]. One regards the solute carrier 6, subfamily A of member 14 gene, which encodes SLC6A14—a Na+/Cl-transporter for amino acids (AA) [101]. The polymorphism of this gene, for example, SNP rs2011162; 22510 C>G, is associated with obesity [126,127]. SLC6A19 is another type of Na+-coupled transporter for neutral amino acids. It is involved in the intestinal absorption of dietary protein-derived amino acids and the renal reabsorption of circulating amino acids [128]. It also controls appetite by regulating tryptophan availability for serotonin synthesis [126]. Deleting SLC6A19 protects against obesity and metabolic syndrome development and improves glycemic control. Thus, pharmacological inhibition of the transporter can benefit the control of metabolic parameters [129,130]. Recent findings show that SNP in the 3′UTR-region of its mRNA suppresses the transporter expression and causes metabolic disorders. In animal studies, such mutations predispose to obesity during a high-fat diet [101]. The same polymorphism is associated with reduced fat oxidation [131]. Unfortunately, data on SLC6A14’s role in sarcopenia are not available.
PPARγ belongs to the nuclear receptor family and is activated by various fatty acid metabolites [132]. It is expressed mainly in the adipose tissue (both white and brown) and other organs, such as the liver, spleen, and large intestine [133]. PPARγ heterodimerizes with retinoid X receptor (RXR) in the nucleus, enabling gene transcription and expression regulation [132,133,134]. PPARγ-2 gene acts as a regulator of metabolism and is strongly upregulated during adipogenesis. PPARγ-2 is expressed in adipose tissue [92]. A study by Ren et al. demonstrated that PPARγ-2 prevents lipotoxicity and prompts the increase of adipose tissue [133,135].
As a result of PPARγ stimulation, adipokines such as adiponectin, interleukin-6 (IL-6), leptin, or monocyte chemoattractant protein-1 (MCP-1) are released [133]. A missense mutation of PPARγ (rs1801282; C>G; Pro12Ala) plays an essential role in the pathogenesis of obesity. However, the impact of this polymorphism on obesity-related parameters is not clarified [136,137,138,139,140], which may result from the influence of diet and the genetic heterogeneity of various populations [141,142]. Postmenopausal overweight and obese women with Pro12Pro polymorphism of the PPARγ gene and higher lean body mass (>58% of body mass) have a bigger chance of recommended glucose levels compared to subjects with lower lean body mass [120].
A recent meta-analysis discovered worse metabolic parameters in obesity for the G allele carriers—including higher BMI, waist circumference, and total cholesterol [140]. Other common polymorphisms of the PPARγ gene are rs3856806; C>T; His447His, which is involved in an increased risk of obesity, coronary heart disease, or colorectal cancer, and rs1800571; Pro115Gln [137,143,144]. As SO is more common in older adults, there is a need to investigate the obesity and sarcopenia risk factors in this group. Some of them—like PPARγ mutation (rs1801282; C>G; Pro12Ala)—were described in elderly subjects to be a risk factor for increased obesity, especially in subjects with high carbohydrate intake with the co-presence of one rs9939609 allele of FTO gene [145].
Jones et al. discovered several genes responsible for low grip strength among 60+ patients. One is the growth/differentiation factor 5 (GDF5) protein. It concerns a 5′ untranslated region mutation of GDF5. GDF5 is a member of the transforming growth factor beta (TGF-β) family, impacting bone and joint development and the formation of osteoarthritis [85].
Genetic risk factors of sarcopenia were also assessed in the Korean population described by Park et al. Sarcopenia risk is associated with SNPs in fatty acid desaturase 2 (FADS2) (rs97384), MYO10 (rs31574), KCNQ5 (rs6453647), DOCK5 (rs11135857), and low-density lipoprotein Receptor Related Protein 1B (LRP1B) (rs74659977) genes. However, it was assessed that the genetic impact might be masked by factors such as metabolic syndrome, high serum total cholesterol levels, and high grip strength. Lifestyle factors such as smoking, coffee, and alcohol intake did not interact with polygenic risk scores for sarcopenia. Exercise was highlighted as the only lifestyle change capable of overcoming genetic factors [66].
Common variants of FADS2 rs1535 major alleles (A-alleles) are reported to be associated with higher BMI in children and adolescents [146]. FADS2 polymorphisms affect the obesity risk in the adult population as well [147]. Moreover—some SNPs can manifest by enhancing circulating fatty acids, influencing predispositions of developing metabolic syndrome, or interacting with dietary non-enzymatic antioxidant capacity—thereby influencing cardio-metabolic risk factors [147,148]. Intronic or intragenic FADS2 SNPs are associated with different levels of polyunsaturated fatty acids (PUFA) ω-6 and ω-3 in pregnant women [149]. LRP1B is a protein with an important role in lipid metabolism, including lipoprotein catabolism, and is linked to BMI and WHR in children [150]. These findings force further investigations on these genes and their possible link to SO.
Studies have shown that in women with co-occurring sarcopenia and obesity, mutations occurred in genes ACTN3 and MTHFR [151]. However, it is worth noting that despite the identification of specific SNPs, the definition of SO adopted by the authors may vary, significantly affecting the study’s outcomes and potentially including polymorphisms whose association with SO, according to the current definition, is insufficient [151]. For instance, drawing upon the research by Khanal et al., for patients with sarcopenia diagnosed in compliance with the low percent of skeletal muscle mass (%SSM) definition, a positive correlation with SNPs in FTO, ESR1, and NOS3 was found. However, females diagnosed with decreased SMI (skeletal muscle index) showed TRHR polymorphism only [110].
Another mechanism associated with SO is telomere shortening, analyzed by Goddard et al. Individuals with SO have significantly shorter telomeres than the control group [152]. Some genetic risk factors for obesity, sarcopenia, or SO are presented in Table 3.

5.2. Epigenetic Factors in Sarcopenic Obesity

The literature mentions various individual, environmental, and genetic risk factors for sarcopenia. Individual risk factors such as age [68,153], male gender [153], higher BMI [153], high monocyte level [153], and sleep duration exceeding 9 h [153] have been positively associated with SO. Advanced age (defined as ≥75 years old) is also associated with a reduction of motor function, resulting in muscle immobility, and predisposing to sarcopenia [68].
Another important risk factor of SO is food insecurity [67,154]. Food insecurity is poor food quality, lack of variety, and simultaneous excessive food consumption. Older people are generally more vulnerable to various factors such as functional disability or impairment, chronic health conditions, lower economic income, and a sedentary lifestyle. All those factors interact, leading to food insecurity [154]. Lowered protein intake, combined with higher calorie intake as well as insufficient vitamin and mineral intake, is part of food insecurity but is also defined as a modifiable risk factor for SO alone [66,68,154].
Sufficient fiber, zinc, vitamin D, calcium, and folate intake is associated with a lower risk of sarcopenia [66,67,68]. Sarcopenia is also associated with high serum glucose, total cholesterol, LDL-cholesterol, and triglyceride concentration [66]. The metabolic disorders related to inflammation, insulin resistance, and oxidative stress are proposed [68].
Recent research by Semenova et al. showed the association of sarcopenia with genetic risk factors. The study observed that the same polymorphisms contribute partially to both obesity and muscle loss, potentially explaining why most individuals affected by sarcopenia are also obese. According to the study, these identified polymorphisms are located in 73 genes with diverse functions, including those related to protein metabolism (BCKDHB, BTRC, COMMD4, SERPINA1, WWP2), carbohydrate metabolism (ADPGK, CDKAL1, GIP, PRRC2A), and lipid metabolism (ADCY3, E2F3, HMGA2, MTCH2, NCOA1, NMT1, PPARδ, immune system activity (BTNL2), myogenesis (SFMBT1), and intracellular transport (GBF1, KIF1B, RIN3, SLC39A8, XPO4) [67]. The same study identified risk alleles associated with sarcopenia. It was assessed that risk alleles are associated with traits such as tiredness, falls in the last 12 months, lowered physical activity, low mineral bone density, neuroticism, time spent watching television, smoking, and poor diet [67].

6. Conclusions

SO is a complex entity comprising immunogenetics as the major cause. The previous use of different definitions and criteria of SO components results in difficulties in estimating the disease’s prevalence, however researchers indicate a higher prevalence in older, as well as chronically ill patients. Both immune and genetic involvement is crucial. Increased adipose tissue leads to systemic inflammation, changes skeletal muscle metabolism, and results in SO and decreased oxidative capacity [47]. Pro-inflammatory cytokine synthesis is of significant importance in the pathophysiology of SO, further—many SNPs were discovered to play a crucial role in obesity (e.g., MC4R or β3) or sarcopenia separately; however, the exact genetic polymorphisms in SO are not well-established yet but remain suspected. More studies are needed to establish the exact genetic alterations leading to SO, leading to the possibilities of early prevention. Nevertheless, the issue of SO is often underestimated, and there is a need to enhance awareness among medical professionals.

Author Contributions

Data acquisition, data analysis, manuscript writing—Ł.M., K.C. and B.G.-G.; Conceptualization, supervision, manuscript writing, and editing—B.G.-G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Donini, L.M.; Busetto, L.; Bischoff, S.C.; Cederholm, T.; Ballesteros-Pomar, M.D.; Batsis, J.A.; Bauer, J.M.; Boirie, Y.; Cruz-Jentoft, A.J.; Dicker, D.; et al. Definition and Diagnostic Criteria for Sarcopenic Obesity: ESPEN and EASO Consensus Statement. Obes. Facts 2022, 15, 321–335. [Google Scholar] [CrossRef]
  2. Cruz-Jentoft, A.J.; Bahat, G.; Bauer, J.; Boirie, Y.; Bruyère, O.; Cederholm, T.; Cooper, C.; Landi, F.; Rolland, Y.; Sayer, A.A.; et al. Sarcopenia: Revised European Consensus on Definition and Diagnosis. Age Ageing 2019, 48, 16–31. [Google Scholar] [CrossRef] [PubMed]
  3. Liu, C.; Wong, P.Y.; Chung, Y.L.; Chow, S.K.-H.; Cheung, W.H.; Law, S.W.; Chan, J.C.N.; Wong, R.M.Y. Deciphering the “Obesity Paradox” in the Elderly: A Systematic Review and Meta-Analysis of Sarcopenic Obesity. Obes. Rev. Off. J. Int. Assoc. Study Obes. 2023, 24, e13534. [Google Scholar] [CrossRef] [PubMed]
  4. Gao, Q.; Mei, F.; Shang, Y.; Hu, K.; Chen, F.; Zhao, L.; Ma, B. Global Prevalence of Sarcopenic Obesity in Older Adults: A Systematic Review and Meta-Analysis. Clin. Nutr. 2021, 40, 4633–4641. [Google Scholar] [CrossRef] [PubMed]
  5. Donini, L.M.; Busetto, L.; Bauer, J.M.; Bischoff, S.; Boirie, Y.; Cederholm, T.; Cruz-Jentoft, A.J.; Dicker, D.; Frühbeck, G.; Giustina, A.; et al. Critical Appraisal of Definitions and Diagnostic Criteria for Sarcopenic Obesity Based on a Systematic Review. Clin. Nutr. 2020, 39, 2368–2388. [Google Scholar] [CrossRef] [PubMed]
  6. Wagenaar, C.A.; Dekker, L.H.; Navis, G.J. Prevalence of Sarcopenic Obesity and Sarcopenic Overweight in the General Population: The Lifelines Cohort Study. Clin. Nutr. 2021, 40, 4422–4429. [Google Scholar] [CrossRef] [PubMed]
  7. Wei, S.; Nguyen, T.T.; Zhang, Y.; Ryu, D.; Gariani, K. Sarcopenic Obesity: Epidemiology, Pathophysiology, Cardiovascular Disease, Mortality, and Management. Front. Endocrinol. 2023, 14, 1185221. [Google Scholar] [CrossRef] [PubMed]
  8. Von Berens, Å.; Obling, S.R.; Nydahl, M.; Koochek, A.; Lissner, L.; Skoog, I.; Frändin, K.; Skoglund, E.; Rothenberg, E.; Cederholm, T. Sarcopenic Obesity and Associations with Mortality in Older Women and Men—A Prospective Observational Study. BMC Geriatr. 2020, 20, 199. [Google Scholar] [CrossRef] [PubMed]
  9. Wang, M.; Tan, Y.; Shi, Y.; Wang, X.; Liao, Z.; Wei, P. Diabetes and Sarcopenic Obesity: Pathogenesis, Diagnosis, and Treatments. Front. Endocrinol. 2020, 11, 568. [Google Scholar] [CrossRef]
  10. Guo, A.; Li, K.; Xiao, Q. Sarcopenic Obesity: Myokines as Potential Diagnostic Biomarkers and Therapeutic Targets? Exp. Gerontol. 2020, 139, 111022. [Google Scholar] [CrossRef]
  11. World Health Organization. Obesity: Preventing and Managing the Global Epidemic: Report of a WHO Consultation. World Health; WHO Technical Report Series 894; World Health Organization: Geneva, Switzerland, 2000; pp. i–xii, 1–253. [Google Scholar]
  12. World Health Organization Regional Office for the Western. The Asia-Pacific Perspective: Redefining Obesity and Its Treatment; Health Communications Australia: Sydney, Australia, 2000; ISBN 978-0-9577082-1-1. [Google Scholar]
  13. Cruz-Jentoft, A.J.; Landi, F.; Topinková, E.; Michel, J.-P. Understanding Sarcopenia as a Geriatric Syndrome. Curr. Opin. Clin. Nutr. Metab. Care 2010, 13, 1–7. [Google Scholar] [CrossRef]
  14. Makizako, H. Frailty and Sarcopenia as a Geriatric Syndrome in Community-Dwelling Older Adults. Int. J. Environ. Res. Public Health 2019, 16, 4013. [Google Scholar] [CrossRef]
  15. Dennison, E.M.; Sayer, A.A.; Cooper, C. Epidemiology of Sarcopenia and Insight into Possible Therapeutic Targets. Nat. Rev. Rheumatol. 2017, 13, 340–347. [Google Scholar] [CrossRef]
  16. Papadopoulou, S.K.; Tsintavis, P.; Potsaki, P.; Papandreou, D. Differences in the Prevalence of Sarcopenia in Community-Dwelling, Nursing Home and Hospitalized Individuals. A Systematic Review and Meta-Analysis. J. Nutr. Health Aging 2020, 24, 83–90. [Google Scholar] [CrossRef] [PubMed]
  17. Fielding, R.A.; Vellas, B.; Evans, W.J.; Bhasin, S.; Morley, J.E.; Newman, A.B.; Abellan van Kan, G.; Andrieu, S.; Bauer, J.; Breuille, D.; et al. Sarcopenia: An Undiagnosed Condition in Older Adults. Current Consensus Definition: Prevalence, Etiology, and Consequences. International Working Group on Sarcopenia. J. Am. Med. Dir. Assoc. 2011, 12, 249–256. [Google Scholar] [CrossRef] [PubMed]
  18. Gallagher, D.; Visser, M.; De Meersman, R.E.; Sepúlveda, D.; Baumgartner, R.N.; Pierson, R.N.; Harris, T.; Heymsfield, S.B. Appendicular Skeletal Muscle Mass: Effects of Age, Gender, and Ethnicity. J. Appl. Physiol. 1997, 83, 229–239. [Google Scholar] [CrossRef] [PubMed]
  19. Baumgartner, R.N.; Koehler, K.M.; Gallagher, D.; Romero, L.; Heymsfield, S.B.; Ross, R.R.; Garry, P.J.; Lindeman, R.D. Epidemiology of Sarcopenia among the Elderly in New Mexico. Am. J. Epidemiol. 1998, 147, 755–763. [Google Scholar] [CrossRef]
  20. Studenski, S.A.; Peters, K.W.; Alley, D.E.; Cawthon, P.M.; McLean, R.R.; Harris, T.B.; Ferrucci, L.; Guralnik, J.M.; Fragala, M.S.; Kenny, A.M.; et al. The FNIH Sarcopenia Project: Rationale, Study Description, Conference Recommendations, and Final Estimates. J. Gerontol. A Biomed. Sci. Med. Sci. 2014, 69, 547–558. [Google Scholar] [CrossRef]
  21. Zanker, J.; Sim, M.; Anderson, K.; Balogun, S.; Brennan-Olsen, S.L.; Dent, E.; Duque, G.; Girgis, C.M.; Grossmann, M.; Hayes, A.; et al. Consensus Guidelines for Sarcopenia Prevention, Diagnosis and Management in Australia and New Zealand. J. Cachexia Sarcopenia Muscle 2023, 14, 142–156. [Google Scholar] [CrossRef]
  22. Sayer, A.A.; Syddall, H.; Martin, H.; Patel, H.; Baylis, D.; Cooper, C. The Developmental Origins of Sarcopenia. J. Nutr. Health Aging 2008, 12, 427–432. [Google Scholar] [CrossRef]
  23. Jung, H.N.; Jung, C.H.; Hwang, Y.-C. Sarcopenia in Youth. Metabolism 2023, 144, 155557. [Google Scholar] [CrossRef]
  24. Schaap, L.A.; van Schoor, N.M.; Lips, P.; Visser, M. Associations of Sarcopenia Definitions, and Their Components, with the Incidence of Recurrent Falling and Fractures: The Longitudinal Aging Study Amsterdam. J. Gerontol. A Biol. Sci. Med. Sci. 2018, 73, 1199–1204. [Google Scholar] [CrossRef]
  25. Masanés, F.; Rojano I Luque, X.; Salvà, A.; Serra-Rexach, J.A.; Artaza, I.; Formiga, F.; Cuesta, F.; López Soto, A.; Ruiz, D.; Cruz-Jentoft, A.J. Cut-off Points for Muscle Mass—Not Grip Strength or Gait Speed—Determine Variations in Sarcopenia Prevalence. J. Nutr. Health Aging 2017, 21, 825–829. [Google Scholar] [CrossRef]
  26. Buckinx, F.; Landi, F.; Cesari, M.; Fielding, R.A.; Visser, M.; Engelke, K.; Maggi, S.; Dennison, E.; Al-Daghri, N.M.; Allepaerts, S.; et al. Pitfalls in the Measurement of Muscle Mass: A Need for a Reference Standard. J. Cachexia Sarcopenia Muscle 2018, 9, 269–278. [Google Scholar] [CrossRef]
  27. Treviño-Aguirre, E.; López-Teros, T.; Gutiérrez-Robledo, L.; Vandewoude, M.; Pérez-Zepeda, M. Availability and Use of Dual Energy X-ray Absorptiometry (DXA) and Bio-Impedance Analysis (BIA) for the Evaluation of Sarcopenia by Belgian and Latin American Geriatricians. J. Cachexia Sarcopenia Muscle 2014, 5, 79–81. [Google Scholar] [CrossRef]
  28. McGregor, R.A.; Cameron-Smith, D.; Poppitt, S.D. It Is Not Just Muscle Mass: A Review of Muscle Quality, Composition and Metabolism during Ageing as Determinants of Muscle Function and Mobility in Later Life. Longev. Healthspan 2014, 3, 9. [Google Scholar] [CrossRef]
  29. Leong, D.P.; Teo, K.K.; Rangarajan, S.; Lopez-Jaramillo, P.; Avezum, A.; Orlandini, A.; Seron, P.; Ahmed, S.H.; Rosengren, A.; Kelishadi, R.; et al. Prognostic Value of Grip Strength: Findings from the Prospective Urban Rural Epidemiology (PURE) Study. Lancet 2015, 386, 266–273. [Google Scholar] [CrossRef]
  30. Pagotto, V.; Silveira, E.A. Applicability and Agreement of Different Diagnostic Criteria for Sarcopenia Estimation in the Elderly. Arch. Gerontol. Geriatr. 2014, 59, 288–294. [Google Scholar] [CrossRef]
  31. Dam, T.-T.; Peters, K.W.; Fragala, M.; Cawthon, P.M.; Harris, T.B.; McLean, R.; Shardell, M.; Alley, D.E.; Kenny, A.; Ferrucci, L.; et al. An Evidence-Based Comparison of Operational Criteria for the Presence of Sarcopenia. J. Gerontol. A Biomed. Sci. Med. Sci. 2014, 69, 584–590. [Google Scholar] [CrossRef] [PubMed]
  32. Sui, S.X.; Holloway-Kew, K.L.; Hyde, N.K.; Williams, L.J.; Tembo, M.C.; Leach, S.; Pasco, J.A. Definition-Specific Prevalence Estimates for Sarcopenia in an Australian Population: The Geelong Osteoporosis Study. JCSM Clin. Rep. 2020, 5, 89–98. [Google Scholar] [CrossRef]
  33. Melton, L.J.; Khosla, S.; Crowson, C.S.; O’Connor, M.K.; O’Fallon, W.M.; Riggs, B.L. Epidemiology of Sarcopenia. J. Am. Geriatr. Soc. 2000, 48, 625–630. [Google Scholar] [CrossRef]
  34. Morley, J.E.; Baumgartner, R.N.; Roubenoff, R.; Mayer, J.; Nair, K.S. Sarcopenia. J. Lab. Clin. Med. 2001, 137, 231–243. [Google Scholar] [CrossRef]
  35. Iannuzzi-Sucich, M.; Prestwood, K.M.; Kenny, A.M. Prevalence of Sarcopenia and Predictors of Skeletal Muscle Mass in Healthy, Older Men and Women. J. Gerontol. A Biol. Sci. Med. Sci. 2002, 57, M772–M777. [Google Scholar] [CrossRef]
  36. Newman, A.B.; Kupelian, V.; Visser, M.; Simonsick, E.; Goodpaster, B.; Nevitt, M.; Kritchevsky, S.B.; Tylavsky, F.A.; Rubin, S.M.; Harris, T.B.; et al. Sarcopenia: Alternative Definitions and Associations with Lower Extremity Function. J. Am. Geriatr. Soc. 2003, 51, 1602–1609. [Google Scholar] [CrossRef]
  37. Janssen, I.; Heymsfield, S.B.; Ross, R. Low Relative Skeletal Muscle Mass (Sarcopenia) in Older Persons Is Associated with Functional Impairment and Physical Disability. J. Am. Geriatr. Soc. 2002, 50, 889–896. [Google Scholar] [CrossRef] [PubMed]
  38. Castillo, E.M.; Goodman-Gruen, D.; Kritz-Silverstein, D.; Morton, D.J.; Wingard, D.L.; Barrett-Connor, E. Sarcopenia in Elderly Men and Women: The Rancho Bernardo Study. Am. J. Prev. Med. 2003, 25, 226–231. [Google Scholar] [CrossRef] [PubMed]
  39. Janssen, I.; Baumgartner, R.N.; Ross, R.; Rosenberg, I.H.; Roubenoff, R. Skeletal Muscle Cutpoints Associated with Elevated Physical Disability Risk in Older Men and Women. Am. J. Epidemiol. 2004, 159, 413–421. [Google Scholar] [CrossRef] [PubMed]
  40. Janssen, I. Influence of Sarcopenia on the Development of Physical Disability: The Cardiovascular Health Study. J. Am. Geriatr. Soc. 2006, 54, 56–62. [Google Scholar] [CrossRef]
  41. Baumgartner, R.N.; Wayne, S.J.; Waters, D.L.; Janssen, I.; Gallagher, D.; Morley, J.E. Sarcopenic Obesity Predicts Instrumental Activities of Daily Living Disability in the Elderly. Obes. Res. 2004, 12, 1995–2004. [Google Scholar] [CrossRef]
  42. Bouchard, D.R.; Dionne, I.J.; Brochu, M. Sarcopenic/Obesity and Physical Capacity in Older Men and Women: Data from the Nutrition as a Determinant of Successful Aging (NuAge)-the Quebec Longitudinal Study. Obesity 2009, 17, 2082–2088. [Google Scholar] [CrossRef]
  43. Barazzoni, R.; Bischoff, S.; Boirie, Y.; Busetto, L.; Cederholm, T.; Dicker, D.; Toplak, H.; Van Gossum, A.; Yumuk, V.; Vettor, R. Sarcopenic Obesity: Time to Meet the Challenge. Obes. Facts 2018, 11, 294–305. [Google Scholar] [CrossRef]
  44. Roubenoff, R. Sarcopenic Obesity: The Confluence of Two Epidemics. Obes. Res. 2004, 12, 887–888. [Google Scholar] [CrossRef]
  45. Dodds, R.M.; Syddall, H.E.; Cooper, R.; Benzeval, M.; Deary, I.J.; Dennison, E.M.; Der, G.; Gale, C.R.; Inskip, H.M.; Jagger, C.; et al. Grip Strength across the Life Course: Normative Data from Twelve British Studies. PLoS ONE 2014, 9, e113637. [Google Scholar] [CrossRef]
  46. Sayer, A.A.; Syddall, H.E.; Gilbody, H.J.; Dennison, E.M.; Cooper, C. Does Sarcopenia Originate in Early Life? Findings from the Hertfordshire Cohort Study. J. Gerontol. A Biol. Sci. Med. Sci. 2004, 59, M930–M934. [Google Scholar] [CrossRef]
  47. Gonzalez, A.; Simon, F.; Achiardi, O.; Vilos, C.; Cabrera, D.; Cabello-Verrugio, C. The Critical Role of Oxidative Stress in Sarcopenic Obesity. Oxidative Med. Cell. Longev. 2021, 2021, 4493817. [Google Scholar] [CrossRef]
  48. Lynch, G.M.; Murphy, C.H.; Castro, E.d.M.; Roche, H.M. Inflammation and Metabolism: The Role of Adiposity in Sarcopenic Obesity. Proc. Nutr. Soc. 2020, 79, 435–447. [Google Scholar] [CrossRef]
  49. Schrager, M.A.; Metter, E.J.; Simonsick, E.; Ble, A.; Bandinelli, S.; Lauretani, F.; Ferrucci, L. Sarcopenic Obesity and Inflammation in the InCHIANTI Study. J. Appl. Physiol. 2007, 102, 919–925. [Google Scholar] [CrossRef]
  50. Ahmed, B.; Sultana, R.; Greene, M.W. Adipose Tissue and Insulin Resistance in Obese. Biomed. Pharmacother. 2021, 137, 111315. [Google Scholar] [CrossRef]
  51. Panee, J. Monocyte Chemoattractant Protein 1 (MCP-1) in Obesity and Diabetes. Cytokine 2012, 60, 1–12. [Google Scholar] [CrossRef] [PubMed]
  52. Huber, J.; Kiefer, F.W.; Zeyda, M.; Ludvik, B.; Silberhumer, G.R.; Prager, G.; Zlabinger, G.J.; Stulnig, T.M. CC Chemokine and CC Chemokine Receptor Profiles in Visceral and Subcutaneous Adipose Tissue Are Altered in Human Obesity. J. Clin. Endocrinol. Metab. 2008, 93, 3215–3221. [Google Scholar] [CrossRef] [PubMed]
  53. Lauterbach, M.A.R.; Wunderlich, F.T. Macrophage Function in Obesity-Induced Inflammation and Insulin Resistance. Pflug. Arch. 2017, 469, 385–396. [Google Scholar] [CrossRef]
  54. Chylikova, J.; Dvorackova, J.; Tauber, Z.; Kamarad, V. M1/M2 Macrophage Polarization in Human Obese Adipose Tissue. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc Czechoslov. 2018, 162, 79–82. [Google Scholar] [CrossRef]
  55. Hong, S.-H.; Choi, K.M. Sarcopenic Obesity, Insulin Resistance, and Their Implications in Cardiovascular and Metabolic Consequences. Int. J. Mol. Sci. 2020, 21, 494. [Google Scholar] [CrossRef]
  56. Alizadeh Pahlavani, H. Exercise Therapy for People with Sarcopenic Obesity: Myokines and Adipokines as Effective Actors. Front. Endocrinol. 2022, 13, 811751. [Google Scholar] [CrossRef]
  57. Hu, M.; Yu, Z.; Luo, D.; Zhang, H.; Li, J.; Liang, F.; Chen, R. Association between -174G>C Polymorphism in the IL-6 Promoter Region and the Risk of Obesity: A Meta-Analysis. Medicine 2018, 97, e11773. [Google Scholar] [CrossRef] [PubMed]
  58. Koc, G.; Doran, T.; Uygur, M.M.; Kirac, D. Obesity Is Associated with IL-6 Gene Polymorphisms Rs1800795 and Rs1800796 but Not SOCS3 Rs4969170. Mol. Biol. Rep. 2023, 50, 2041–2048. [Google Scholar] [CrossRef]
  59. Luotola, K. IL-1 Receptor Antagonist (IL-1Ra) Levels and Management of Metabolic Disorders. Nutrients 2022, 14, 3422. [Google Scholar] [CrossRef]
  60. Gangemi, S.; Basile, G.; Monti, D.; Alba Merendino, R.; Di Pasquale, G.; Bisignano, U.; Nicita-Mauro, V.; Franceschi, C. Age-Related Modifications in Circulating IL-15 Levels in Humans. Mediat. Inflamm. 2005, 2005, 245–247. [Google Scholar] [CrossRef] [PubMed]
  61. Quinn, L.S.; Anderson, B.G.; Conner, J.D.; Wolden-Hanson, T.; Marcell, T.J. IL-15 Is Required for Postexercise Induction of the pro-Oxidative Mediators PPARδ and SIRT1 in Male Mice. Endocrinology 2014, 155, 143–155. [Google Scholar] [CrossRef]
  62. Tilg, H.; Moschen, A.R. Adipocytokines: Mediators Linking Adipose Tissue, Inflammation and Immunity. Nat. Rev. Immunol. 2006, 6, 772–783. [Google Scholar] [CrossRef] [PubMed]
  63. Kob, R.; Bollheimer, L.C.; Bertsch, T.; Fellner, C.; Djukic, M.; Sieber, C.C.; Fischer, B.E. Sarcopenic Obesity: Molecular Clues to a Better Understanding of Its Pathogenesis? Biogerontology 2015, 16, 15–29. [Google Scholar] [CrossRef]
  64. Morley, J.E. Hormones and Sarcopenia. Curr. Pharm. Des. 2017, 23, 4484–4492. [Google Scholar] [CrossRef]
  65. Roh, E.; Choi, K.M. Health Consequences of Sarcopenic Obesity: A Narrative Review. Front. Endocrinol. 2020, 11, 332. [Google Scholar] [CrossRef]
  66. Park, S. Association between Polygenetic Risk Scores Related to Sarcopenia Risk and Their Interactions with Regular Exercise in a Large Cohort of Korean Adults. Clin. Nutr. 2021, 40, 5355–5364. [Google Scholar] [CrossRef]
  67. Semenova, E.A.; Pranckevičienė, E.; Bondareva, E.A.; Gabdrakhmanova, L.J.; Ahmetov, I.I. Identification and Characterization of Genomic Predictors of Sarcopenia and Sarcopenic Obesity Using UK Biobank Data. Nutrients 2023, 15, 758. [Google Scholar] [CrossRef]
  68. Evans, K.; Abdelhafiz, D.; Abdelhafiz, A.H. Sarcopenic Obesity as a Determinant of Cardiovascular Disease Risk in Older People: A Systematic Review. Postgrad. Med. 2021, 133, 831–842. [Google Scholar] [CrossRef]
  69. Pradhan, G.; Samson, S.L.; Sun, Y. Ghrelin: Much More than a Hunger Hormone. Curr. Opin. Clin. Nutr. Metab. Care 2013, 16, 619–624. [Google Scholar] [CrossRef]
  70. Esposito, P.; Picciotto, D.; Battaglia, Y.; Costigliolo, F.; Viazzi, F.; Verzola, D. Myostatin: Basic Biology to Clinical Application. Adv. Clin. Chem. 2022, 106, 181–234. [Google Scholar] [CrossRef]
  71. Aghaali, M.; Hashemi-Nazari, S.S. Association between Early Antibiotic Exposure and Risk of Childhood Weight Gain and Obesity: A Systematic Review and Meta-Analysis. J. Pediatr. Endocrinol. Metab. JPEM 2019, 32, 439–445. [Google Scholar] [CrossRef]
  72. Bassetti, S.; Tschudin-Sutter, S.; Egli, A.; Osthoff, M. Optimizing Antibiotic Therapies to Reduce the Risk of Bacterial Resistance. Eur. J. Intern. Med. 2022, 99, 7–12. [Google Scholar] [CrossRef]
  73. Rasmussen, S.H.; Shrestha, S.; Bjerregaard, L.G.; Ängquist, L.H.; Baker, J.L.; Jess, T.; Allin, K.H. Antibiotic Exposure in Early Life and Childhood Overweight and Obesity: A Systematic Review and Meta-Analysis. Diabetes Obes. Metab. 2018, 20, 1508–1514. [Google Scholar] [CrossRef]
  74. Cho, I.; Yamanishi, S.; Cox, L.; Methé, B.A.; Zavadil, J.; Li, K.; Gao, Z.; Mahana, D.; Raju, K.; Teitler, I.; et al. Antibiotics in Early Life Alter the Murine Colonic Microbiome and Adiposity. Nature 2012, 488, 621–626. [Google Scholar] [CrossRef]
  75. Wilkins, A.T.; Reimer, R.A. Obesity, Early Life Gut Microbiota, and Antibiotics. Microorganisms 2021, 9, 413. [Google Scholar] [CrossRef]
  76. Bach, D.; Pich, S.; Soriano, F.X.; Vega, N.; Baumgartner, B.; Oriola, J.; Daugaard, J.R.; Lloberas, J.; Camps, M.; Zierath, J.R.; et al. Mitofusin-2 Determines Mitochondrial Network Architecture and Mitochondrial Metabolism. A Novel Regulatory Mechanism Altered in Obesity. J. Biol. Chem. 2003, 278, 17190–17197. [Google Scholar] [CrossRef]
  77. Dutra, M.T.; Avelar, B.P.; Souza, V.C.; Bottaro, M.; Oliveira, R.J.; Nóbrega, O.T.; Moreno Lima, R. Relationship between Sarcopenic Obesity-Related Phenotypes and Inflammatory Markers in Postmenopausal Women. Clin. Physiol. Funct. Imaging 2017, 37, 205–210. [Google Scholar] [CrossRef]
  78. Sakuma, K.; Yamaguchi, A. Sarcopenic Obesity and Endocrinal Adaptation with Age. Int. J. Endocrinol. 2013, 2013, 204164. [Google Scholar] [CrossRef]
  79. Mazurkiewicz, M.; Lizurej, W.; Mazurkiewicz, L.; Majewski, A.; Tobis, S.; Cymerys, M. C-Reactive Protein and Albumin Levels as Useful Tools for Predicting In-Hospital Outcomes in Polish Nonagenarians. Clin. Interv. Aging 2023, 18, 1547–1554. [Google Scholar] [CrossRef]
  80. Ohler, A.M.; Braddock, A. Infections and Antibiotic Use in Early Life, and Obesity in Early Childhood: A Mediation Analysis. Int. J. Obes. 2005 2022, 46, 1608–1614. [Google Scholar] [CrossRef]
  81. Voss, J.D.; Atkinson, R.L.; Dhurandhar, N.V. Role of Adenoviruses in Obesity. Rev. Med. Virol. 2015, 25, 379–387. [Google Scholar] [CrossRef]
  82. Voss, J.D.; Dhurandhar, N.V. Viral Infections and Obesity. Curr. Obes. Rep. 2017, 6, 28–37. [Google Scholar] [CrossRef]
  83. Pasarica, M.; Mashtalir, N.; McAllister, E.J.; Kilroy, G.E.; Koska, J.; Permana, P.; de Courten, B.; Yu, M.; Ravussin, E.; Gimble, J.M.; et al. Adipogenic Human Adenovirus Ad-36 Induces Commitment, Differentiation, and Lipid Accumulation in Human Adipose-Derived Stem Cells. Stem Cells 2008, 26, 969–978. [Google Scholar] [CrossRef]
  84. Jones, G.; Pilling, L.C.; Kuo, C.-L.; Kuchel, G.; Ferrucci, L.; Melzer, D. Sarcopenia and Variation in the Human Leukocyte Antigen Complex. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 301–308. [Google Scholar] [CrossRef]
  85. Jones, G.; Trajanoska, K.; Santanasto, A.J.; Stringa, N.; Kuo, C.-L.; Atkins, J.L.; Lewis, J.R.; Duong, T.; Hong, S.; Biggs, M.L.; et al. Genome-Wide Meta-Analysis of Muscle Weakness Identifies 15 Susceptibility Loci in Older Men and Women. Nat. Commun. 2021, 12, 654. [Google Scholar] [CrossRef]
  86. Singh, A.N.; Gasman, B. Disentangling the Genetics of Sarcopenia: Prioritization of NUDT3 and KLF5 as Genes for Lean Mass & HLA-DQB1-AS1 for Hand Grip Strength with the Associated Enhancing SNPs & a Scoring System. BMC Med. Genet. 2020, 21, 40. [Google Scholar] [CrossRef]
  87. Sica, A.; Erreni, M.; Allavena, P.; Porta, C. Macrophage Polarization in Pathology. Cell. Mol. Life Sci. 2015, 72, 4111–4126. [Google Scholar] [CrossRef]
  88. Gu, X.; Zhang, Y.; Li, D.; Cai, H.; Cai, L.; Xu, Q. N6-Methyladenosine Demethylase FTO Promotes M1 and M2 Macrophage Activation. Cell. Signal. 2020, 69, 109553. [Google Scholar] [CrossRef]
  89. McFadden, M.J.; Sacco, M.T.; Murphy, K.A.; Park, M.; Gokhale, N.S.; Somfleth, K.Y.; Horner, S.M. FTO Suppresses STAT3 Activation and Modulates Proinflammatory Interferon-Stimulated Gene Expression. J. Mol. Biol. 2022, 434, 167247. [Google Scholar] [CrossRef]
  90. Carpenter, R.L.; Lo, H.-W. STAT3 Target Genes Relevant to Human Cancers. Cancers 2014, 6, 897–925. [Google Scholar] [CrossRef]
  91. Dubey, P.K.; Patil, M.; Singh, S.; Dubey, S.; Ahuja, P.; Verma, S.K.; Krishnamurthy, P. Increased m6A-RNA Methylation and FTO Suppression Is Associated with Myocardial Inflammation and Dysfunction during Endotoxemia in Mice. Mol. Cell. Biochem. 2022, 477, 129–141. [Google Scholar] [CrossRef]
  92. Hernandez-Quiles, M.; Broekema, M.F.; Kalkhoven, E. PPARgamma in Metabolism, Immunity, and Cancer: Unified and Diverse Mechanisms of Action. Front. Endocrinol. 2021, 12, 624112. [Google Scholar] [CrossRef]
  93. Parisi, L.; Gini, E.; Baci, D.; Tremolati, M.; Fanuli, M.; Bassani, B.; Farronato, G.; Bruno, A.; Mortara, L. Macrophage Polarization in Chronic Inflammatory Diseases: Killers or Builders? J. Immunol. Res. 2018, 2018, 8917804. [Google Scholar] [CrossRef]
  94. Araki, A.; Hosoi, T.; Orimo, H.; Ito, H. Association of Plasma Homocysteine with Serum Interleukin-6 and C-Peptide Levels in Patients with Type 2 Diabetes. Metabolism 2005, 54, 809–814. [Google Scholar] [CrossRef]
  95. Lisboa, J.V.d.C.; Ribeiro, M.R.; Luna, R.C.P.; Lima, R.P.A.; do Nascimento, R.A.F.; Monteiro, M.G.C.A.; Lima, K.Q.d.F.; Fechine, C.P.N.d.S.; de Oliveira, N.F.P.; Persuhn, D.C.; et al. Food Intervention with Folate Reduces TNF-α and Interleukin Levels in Overweight and Obese Women with the MTHFR C677T Polymorphism: A Randomized Trial. Nutrients 2020, 12, 361. [Google Scholar] [CrossRef]
  96. Gholami, M.; Sharifi, F.; Shahriari, S.; Khoshnevisan, K.; Larijani, B.; Amoli, M.M. Association of Interleukin-6 Polymorphisms with Obesity: A Systematic Review and Meta-Analysis. Cytokine 2019, 123, 154769. [Google Scholar] [CrossRef]
  97. Melo, A.P.C.; Teixeira, H.M.P.; Coelho, R.S.; De Jesus, T.D.S.; Queiroz, G.A.; Silva, H.D.S.; De Almeida, Y.C.F.; Alcantara-Neves, N.M.; De Matos, S.M.A.; D’innocenzo, S.; et al. Variants in Proinflammatory Genes IL1RL1, IL1B and IRF4 Are Associated with Overweight in a Pediatric Brazilian Population. Gene 2022, 828, 146478. [Google Scholar] [CrossRef]
  98. Maculewicz, E.; Antkowiak, B.; Antkowiak, O.; Borecka, A.; Mastalerz, A.; Leońska-Duniec, A.; Humińska-Lisowska, K.; Michałowska-Sawczyn, M.; Garbacz, A.; Lorenz, K.; et al. The Interactions between Interleukin-1 Family Genes: IL1A, IL1B, IL1RN, and Obesity Parameters. BMC Genom. 2022, 23, 112. [Google Scholar] [CrossRef]
  99. Hinney, A.; Körner, A.; Fischer-Posovszky, P. The Promise of New Anti-Obesity Therapies Arising from Knowledge of Genetic Obesity Traits. Nat. Rev. Endocrinol. 2022, 18, 623–637. [Google Scholar] [CrossRef]
  100. Mahmoud, R.; Kimonis, V.; Butler, M.G. Genetics of Obesity in Humans: A Clinical Review. Int. J. Mol. Sci. 2022, 23, 11005. [Google Scholar] [CrossRef]
  101. Sivaprakasam, S.; Sikder, M.O.F.; Ramalingam, L.; Kaur, G.; Dufour, J.M.; Moustaid-Moussa, N.; Wachtel, M.S.; Ganapathy, V. SLC6A14 Deficiency Is Linked to Obesity, Fatty Liver, and Metabolic Syndrome but Only under Conditions of a High-Fat Diet. Biochim. Biophys. Acta Mol. Basis Dis. 2021, 1867, 166087. [Google Scholar] [CrossRef]
  102. Męczekalski, B.; Czyżyk, A.; Warenik-Szymankiewicz, A. Rola Genów w Powstawaniu Otyłości. Współczesne Poglądy, Patogeneza, Aspekty Kliniczne. Endokrynol. Otyłość Zaburzenia Przemiany Mater. 2008, 4, 27–37. [Google Scholar]
  103. Shabana; Shahid, S.U.; Hasnain, S. Use of a Gene Score of Multiple Low-Modest Effect Size Variants Can Predict the Risk of Obesity Better than the Individual SNPs. Lipids Health Dis. 2018, 17, 155. [Google Scholar] [CrossRef]
  104. Tan, P.Y.; Mitra, S.R. The Combined Effect of Polygenic Risk from FTO and ADRB2 Gene Variants, Odds of Obesity, and Post-Hipcref Diet Differences. Lifestyle Genom. 2020, 13, 84–98. [Google Scholar] [CrossRef]
  105. Castro, G.V.; Latorre, A.F.S.; Korndorfer, F.P.; de Carlos Back, L.K.; Lofgren, S.E. The Impact of Variants in Four Genes: MC4R, FTO, PPARG and PPARGC1A in Overweight and Obesity in a Large Sample of the Brazilian Population. Biochem. Genet. 2021, 59, 1666–1679. [Google Scholar] [CrossRef]
  106. Frayling, T.M.; Timpson, N.J.; Weedon, M.N.; Zeggini, E.; Freathy, R.M.; Lindgren, C.M.; Perry, J.R.B.; Elliott, K.S.; Lango, H.; Rayner, N.W.; et al. A Common Variant in the FTO Gene Is Associated with Body Mass Index and Predisposes to Childhood and Adult Obesity. Science 2007, 316, 889–894. [Google Scholar] [CrossRef]
  107. Lan, N.; Lu, Y.; Zhang, Y.; Pu, S.; Xi, H.; Nie, X.; Liu, J.; Yuan, W. FTO—A Common Genetic Basis for Obesity and Cancer. Front. Genet. 2020, 11, 559138. [Google Scholar] [CrossRef]
  108. Park, H.-G.; Choi, J.-H. Genetic Variant Rs9939609 in FTO Is Associated with Body Composition and Obesity Risk in Korean Females. BMJ Open Diabetes Res. Care 2023, 11, e003649. [Google Scholar] [CrossRef]
  109. Ran, S.; Jiang, Z.-X.; He, X.; Liu, Y.; Zhang, Y.-X.; Zhang, L.; Pei, Y.-F.; Zhang, M.; Hai, R.; Gu, G.-S.; et al. Replication of FTO Gene Associated with Lean Mass in a Meta-Analysis of Genome-Wide Association Studies. Sci. Rep. 2020, 10, 5057. [Google Scholar] [CrossRef] [PubMed]
  110. Khanal, P.; He, L.; Stebbings, G.; Onambele-Pearson, G.L.; Degens, H.; Williams, A.; Thomis, M.; Morse, C.I. Prevalence and Association of Single Nucleotide Polymorphisms with Sarcopenia in Older Women Depends on Definition. Sci. Rep. 2020, 10, 2913. [Google Scholar] [CrossRef] [PubMed]
  111. Clément, K.; Biebermann, H.; Farooqi, I.S.; Van der Ploeg, L.; Wolters, B.; Poitou, C.; Puder, L.; Fiedorek, F.; Gottesdiener, K.; Kleinau, G.; et al. MC4R Agonism Promotes Durable Weight Loss in Patients with Leptin Receptor Deficiency. Nat. Med. 2018, 24, 551–555. [Google Scholar] [CrossRef] [PubMed]
  112. Collet, T.-H.; Dubern, B.; Mokrosinski, J.; Connors, H.; Keogh, J.M.; Mendes de Oliveira, E.; Henning, E.; Poitou-Bernert, C.; Oppert, J.-M.; Tounian, P.; et al. Evaluation of a Melanocortin-4 Receptor (MC4R) Agonist (Setmelanotide) in MC4R Deficiency. Mol. Metab. 2017, 6, 1321–1329. [Google Scholar] [CrossRef] [PubMed]
  113. Brodowski, J.; Szkup, M.; Jurczak, A.; Wieder-Huszla, S.; Brodowska, A.; Laszczyńska, M.; Karakiewicz, B.; Kęcka, K.; Grochans, E. Searching for the Relationship between the Parameters of Metabolic Syndrome and the Rs17782313 (T>C) Polymorphism of the MC4R Gene in Postmenopausal Women. Clin. Interv. Aging 2017, 12, 549–555. [Google Scholar] [CrossRef]
  114. Yang, J.; Gao, Q.; Gao, X.; Tao, X.; Cai, H.; Fan, Y.; Zhang, N.; Zhang, Y.; Li, L.; Li, H. Melanocortin-4 Receptor Rs17782313 Polymorphisms Are Associated with Serum Triglycerides in Older Chinese Women. Asia Pac. J. Clin. Nutr. 2016, 25, 213–219. [Google Scholar] [CrossRef] [PubMed]
  115. Lima, R.P.A.; Ribeiro, M.R.; de Farias Lima, K.Q.; de Sena, E.A.; de Oliveira Costa, D.; Luna, R.C.P.; do Nascimento, R.A.F.; da Conceição Rodrigues Gonçalves, M.; de Toledo Vianna, R.P.; de Moraes, R.M.; et al. Methylation Profile of the ADRB3 Gene and Its Association with Lipid Profile and Nutritional Status in Adults. Biol. Res. 2019, 52, 21. [Google Scholar] [CrossRef] [PubMed]
  116. Luo, Z.; Zhang, T.; Wang, S.; He, Y.; Ye, Q.; Cao, W. The Trp64Arg Polymorphism in Β3 Adrenergic Receptor (ADRB3) Gene Is Associated with Adipokines and Plasma Lipids: A Systematic Review, Meta-Analysis, and Meta-Regression. Lipids Health Dis. 2020, 19, 99. [Google Scholar] [CrossRef] [PubMed]
  117. De Jesus, Í.C.; Alle, L.F.; Munhoz, E.C.; da Silva, L.R.; Lopes, W.A.; Tureck, L.V.; Purim, K.S.M.; Titski, A.C.K.; Leite, N. Trp64Arg Polymorphism of the ADRB3 Gene Associated with Maximal Fat Oxidation and LDL-C Levels in Non-Obese Adolescents. J. Pediatr. 2018, 94, 425–431. [Google Scholar] [CrossRef] [PubMed]
  118. González-Hedström, D.; Priego, T.; Amor, S.; de la Fuente-Fernández, M.; Martín, A.I.; López-Calderón, A.; Inarejos-García, A.M.; García-Villalón, Á.L.; Granado, M. Olive Leaf Extract Supplementation to Old Wistar Rats Attenuates Aging-Induced Sarcopenia and Increases Insulin Sensitivity in Adipose Tissue and Skeletal Muscle. Antioxidants 2021, 10, 737. [Google Scholar] [CrossRef] [PubMed]
  119. McKean-Cowdin, R.; Li, X.; Bernstein, L.; McTiernan, A.; Ballard-Barbash, R.; Gauderman, W.J.; Gilliland, F. The ADRB3 Trp64Arg Variant and Obesity in African-American Breast Cancer Cases. Int. J. Obes. 2005 2007, 31, 1110–1118. [Google Scholar] [CrossRef] [PubMed]
  120. Grygiel-Górniak, B.; Kaczmarek, E.; Mosor, M.; Przysławski, J.; Nowak, J. Gene-Diet-Related Factors of Hyperglycaemia in Postmenopausal Women. J. Appl. Genet. 2018, 59, 169–177. [Google Scholar] [CrossRef]
  121. Grygiel-Górniak, B.; Kaczmarek, E.; Mosor, M.; Przysławski, J.; Nowak, J. The Gene-Diet Associations in Postmenopausal Women with Newly Diagnosed Dyslipidemia. J. Nutr. Health Aging 2017, 21, 1031–1037. [Google Scholar] [CrossRef]
  122. de Luis, D.A.; Aller, R.; Izaola, O.; Gonzalez-Sagrado, M.; Conde, R. Relation of Trp64Arg Polymorphism of β3-Adrenoreceptor Gene with Cardiovascular Risk Factors in Presurgical Morbidly Obese Patients. Arch. Med. Res. 2008, 39, 791–795. [Google Scholar] [CrossRef]
  123. Clément, K.; Vaisse, C.; Manning, B.S.; Basdevant, A.; Guy-Grand, B.; Ruiz, J.; Silver, K.D.; Shuldiner, A.R.; Froguel, P.; Strosberg, A.D. Genetic Variation in the β3-Adrenergic Receptor and an Increased Capacity to Gain Weight in Patients with Morbid Obesity. N. Engl. J. Med. 1995, 333, 352–354. [Google Scholar] [CrossRef]
  124. Kurokawa, N.; Young, E.H.; Oka, Y.; Satoh, H.; Wareham, N.J.; Sandhu, M.S.; Loos, R.J.F. The ADRB3 Trp64Arg Variant and BMI: A Meta-Analysis of 44 833 Individuals. Int. J. Obes. 2005 2008, 32, 1240–1249. [Google Scholar] [CrossRef]
  125. Bhutia, Y.D.; Mathew, M.; Sivaprakasam, S.; Ramachandran, S.; Ganapathy, V. Unconventional Functions of Amino Acid Transporters: Role in Macropinocytosis (SLC38A5/SLC38A3) and Diet-Induced Obesity/Metabolic Syndrome (SLC6A19/SLC6A14/SLC6A6). Biomolecules 2022, 12, 235. [Google Scholar] [CrossRef]
  126. Suviolahti, E.; Oksanen, L.J.; Öhman, M.; Cantor, R.M.; Ridderstråle, M.; Tuomi, T.; Kaprio, J.; Rissanen, A.; Mustajoki, P.; Jousilahti, P.; et al. The SLC6A14 Gene Shows Evidence of Association with Obesity. J. Clin. Investig. 2003, 112, 1762–1772. [Google Scholar] [CrossRef]
  127. Durand, E.; Boutin, P.; Meyre, D.; Charles, M.A.; Clement, K.; Dina, C.; Froguel, P. Polymorphisms in the Amino Acid Transporter Solute Carrier Family 6 (Neurotransmitter Transporter) Member 14 Gene Contribute to Polygenic Obesity in French Caucasians. Diabetes 2004, 53, 2483–2486. [Google Scholar] [CrossRef]
  128. Bröer, S. Epithelial Neutral Amino Acid Transporters: Lessons from Mouse Models. Curr. Opin. Nephrol. Hypertens. 2013, 22, 539–544. [Google Scholar] [CrossRef]
  129. Cheng, Q.; Shah, N.; Bröer, A.; Fairweather, S.; Jiang, Y.; Schmoll, D.; Corry, B.; Bröer, S. Identification of Novel Inhibitors of the Amino Acid Transporter B0 AT1 (SLC6A19), a Potential Target to Induce Protein Restriction and to Treat Type 2 Diabetes. Br. J. Pharmacol. 2017, 174, 468–482. [Google Scholar] [CrossRef] [PubMed]
  130. Javed, K.; Cheng, Q.; Carroll, A.J.; Truong, T.T.; Bröer, S. Development of Biomarkers for Inhibition of SLC6A19 (B0AT1)-A Potential Target to Treat Metabolic Disorders. Int. J. Mol. Sci. 2018, 19, 3597. [Google Scholar] [CrossRef] [PubMed]
  131. Corpeleijn, E.; Petersen, L.; Holst, C.; Saris, W.H.; Astrup, A.; Langin, D.; MacDonald, I.; Martinez, J.A.; Oppert, J.-M.; Polak, J.; et al. Obesity-Related Polymorphisms and Their Associations with the Ability to Regulate Fat Oxidation in Obese Europeans: The NUGENOB Study. Obesity 2010, 18, 1369–1377. [Google Scholar] [CrossRef] [PubMed]
  132. Auwerx, J. PPARgamma, the Ultimate Thrifty Gene. Diabetologia 1999, 42, 1033–1049. [Google Scholar] [CrossRef] [PubMed]
  133. Laganà, A.S.; Vitale, S.G.; Nigro, A.; Sofo, V.; Salmeri, F.M.; Rossetti, P.; Rapisarda, A.M.C.; La Vignera, S.; Condorelli, R.A.; Rizzo, G.; et al. Pleiotropic Actions of Peroxisome Proliferator-Activated Receptors (PPARs) in Dysregulated Metabolic Homeostasis, Inflammation and Cancer: Current Evidence and Future Perspectives. Int. J. Mol. Sci. 2016, 17, 999. [Google Scholar] [CrossRef]
  134. Wang, S.; Lin, Y.; Gao, L.; Yang, Z.; Lin, J.; Ren, S.; Li, F.; Chen, J.; Wang, Z.; Dong, Z.; et al. PPAR-γ Integrates Obesity and Adipocyte Clock through Epigenetic Regulation of Bmal1. Theranostics 2022, 12, 1589–1606. [Google Scholar] [CrossRef] [PubMed]
  135. Ren, D.; Collingwood, T.N.; Rebar, E.J.; Wolffe, A.P.; Camp, H.S. PPARgamma Knockdown by Engineered Transcription Factors: Exogenous PPARgamma2 but Not PPARgamma1 Reactivates Adipogenesis. Genes Dev. 2002, 16, 27–32. [Google Scholar] [CrossRef] [PubMed]
  136. Deeb, S.S.; Fajas, L.; Nemoto, M.; Pihlajamäki, J.; Mykkänen, L.; Kuusisto, J.; Laakso, M.; Fujimoto, W.; Auwerx, J. A Pro12Ala Substitution in PPARgamma2 Associated with Decreased Receptor Activity, Lower Body Mass Index and Improved Insulin Sensitivity. Nat. Genet. 1998, 20, 284–287. [Google Scholar] [CrossRef] [PubMed]
  137. Muntean, C.; Sasaran, M.O.; Crisan, A.; Banescu, C. Effects of PPARG and PPARGC1A Gene Polymorphisms on Obesity Markers. Front. Public Health 2022, 10, 962852. [Google Scholar] [CrossRef] [PubMed]
  138. Franks, P.W.; Jablonski, K.A.; Delahanty, L.; Hanson, R.L.; Kahn, S.E.; Altshuler, D.; Knowler, W.C.; Florez, J.C.; Diabetes Prevention Program Research Group. The Pro12Ala Variant at the Peroxisome Proliferator-Activated Receptor γ Gene and Change in Obesity-Related Traits in the Diabetes Prevention Program. Diabetologia 2007, 50, 2451–2460. [Google Scholar] [CrossRef]
  139. Dedoussis, G.V.; Vidra, N.; Butler, J.; Papoutsakis, C.; Yannakoulia, M.; Hirschhorn, J.N.; Lyon, H.N. Peroxisome Proliferator-Activated Receptor-γ (PPARγ) Pro12Ala Polymorphism and Risk for Pediatric Obesity. Clin. Chem. Lab. Med. 2009, 47, 1047–1050. [Google Scholar] [CrossRef]
  140. Li, S.; He, C.; Nie, H.; Pang, Q.; Wang, R.; Zeng, Z.; Song, Y. G Allele of the Rs1801282 Polymorphism in PPARγ Gene Confers an Increased Risk of Obesity and Hypercholesterolemia, While T Allele of the Rs3856806 Polymorphism Displays a Protective Role Against Dyslipidemia: A Systematic Review and Meta-Analysis. Front. Endocrinol. 2022, 13, 919087. [Google Scholar] [CrossRef] [PubMed]
  141. Wu, Z.; Lou, Y.; Jin, W.; Liu, Y.; Lu, L.; Lu, G. The Pro12Ala Polymorphism in the Peroxisome Proliferator-Activated Receptor Gamma-2 Gene (PPARγ2) Is Associated with Increased Risk of Coronary Artery Disease: A Meta-Analysis. PLoS ONE 2012, 7, e53105. [Google Scholar] [CrossRef]
  142. Chmielewska-Kassassir, M.; Woźniak, L.A.; Ogrodniczek, P.; Wójcik, M. The role of peroxisome proliferator-activated receptors γ (PPARγ) in obesity and insulin resistance. Postęp. Hig. Med. Dośw. 2013, 67, 1283–1299. [Google Scholar] [CrossRef]
  143. Ristow, M.; Müller-Wieland, D.; Pfeiffer, A.; Krone, W.; Kahn, C.R. Obesity Associated with a Mutation in a Genetic Regulator of Adipocyte Differentiation. N. Engl. J. Med. 1998, 339, 953–959. [Google Scholar] [CrossRef]
  144. Kurnaz-Gomleksiz, O.; Torun, B.C.; Isbir, T.; Bulut, T.; Sokucu, N.; Yilmaz-Aydogan, H.; Canbay, E. The Role of PPAR-Gamma C161T Polymorphism in Colorectal Cancer Susceptibility. In Vivo 2022, 36, 1911–1915. [Google Scholar] [CrossRef]
  145. Galbete, C.; Toledo, J.; Martínez-González, M.Á.; Martínez, J.A.; Guillén-Grima, F.; Marti, A. Lifestyle Factors Modify Obesity Risk Linked to PPARG2 and FTO Variants in an Elderly Population: A Cross-Sectional Analysis in the SUN Project. Genes Nutr. 2013, 8, 61–67. [Google Scholar] [CrossRef] [PubMed]
  146. Maguolo, A.; Zusi, C.; Giontella, A.; Miraglia Del Giudice, E.; Tagetti, A.; Fava, C.; Morandi, A.; Maffeis, C. Influence of Genetic Variants in FADS2 and ELOVL2 Genes on BMI and PUFAs Homeostasis in Children and Adolescents with Obesity. Int. J. Obes. 2005 2021, 45, 56–65. [Google Scholar] [CrossRef] [PubMed]
  147. Khamlaoui, W.; Mehri, S.; Hammami, S.; Hammouda, S.; Chraeif, I.; Elosua, R.; Hammami, M. Association Between Genetic Variants in FADS1-FADS2 and ELOVL2 and Obesity, Lipid Traits, and Fatty Acids in Tunisian Population. Clin. Appl. Thromb. Off. J. Int. Acad. Clin. Appl. Thromb. 2020, 26, 1076029620915286. [Google Scholar] [CrossRef] [PubMed]
  148. Khodarahmi, M.; Javidzade, P.; Farhangi, M.A.; Hashemzehi, A.; Kahroba, H. Interplay between Fatty Acid Desaturase2 (FADS2) Rs174583 Genetic Variant and Dietary Antioxidant Capacity: Cardio-Metabolic Risk Factors in Obese Individuals. BMC Endocr. Disord. 2022, 22, 167. [Google Scholar] [CrossRef] [PubMed]
  149. Santana, J.d.M.; Pereira, M.; Carvalho, G.Q.; Gouveia Peluzio, M.d.C.; Drumond Louro, I.; Santos, D.B.d.; Oliveira, A.M. FADS1 and FADS2 Gene Polymorphisms Modulate the Relationship of Omega-3 and Omega-6 Fatty Acid Plasma Concentrations in Gestational Weight Gain: A NISAMI Cohort Study. Nutrients 2022, 14, 1056. [Google Scholar] [CrossRef]
  150. Lee, S. The Genetic and Epigenetic Association of LDL Receptor Related Protein 1B (LRP1B) Gene with Childhood Obesity. Sci. Rep. 2019, 9, 1815. [Google Scholar] [CrossRef]
  151. Khanal, P.; Williams, A.G.; He, L.; Stebbings, G.K.; Onambele-Pearson, G.L.; Thomis, M.; Degens, H.; Morse, C.I. Sarcopenia, Obesity, and Sarcopenic Obesity: Relationship with Skeletal Muscle Phenotypes and Single Nucleotide Polymorphisms. J. Clin. Med. 2021, 10, 4933. [Google Scholar] [CrossRef]
  152. Goddard, T.; Tsintzas, K.; Stephan, B.C.M.; Prado, C.M.; Mazidi, M.; Siervo, M. Sarcopenic Obesity Is Associated with Telomere Shortening: Findings from the NHANES 1999–2002. Int. J. Obes. 2005 2022, 46, 437–440. [Google Scholar] [CrossRef]
  153. Lu, L.; He, X.; Song, Y.; Zhuang, M.; Wu, X.; Chen, N. Prevalence and Risk Factors of Sarcopenia without Obesity and Sarcopenic Obesity among Chinese Community Older People in Suburban Area of Shanghai: A Cross-Sectional Study. Front. Aging Neurosci. 2022, 14, 1034542. [Google Scholar] [CrossRef] [PubMed]
  154. Fonseca-Pérez, D.; Arteaga-Pazmiño, C.; Maza-Moscoso, C.P.; Flores-Madrid, S.; Álvarez-Córdova, L. Food Insecurity as a Risk Factor of Sarcopenic Obesity in Older Adults. Front. Nutr. 2022, 9, 1040089. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Graphical presentation of the search strategy used in this review.
Figure 1. Graphical presentation of the search strategy used in this review.
Genes 15 00206 g001
Figure 2. Multi-factorial causes of obesity, sarcopenia and SO.
Figure 2. Multi-factorial causes of obesity, sarcopenia and SO.
Genes 15 00206 g002
Figure 3. Genes related to obesity and sarcopenia (green—confirmed obesity-related gene polymorphisms; red—sarcopenia-related gene polymorphisms; yellow—probable gene polymorphisms in SO). The hypothesis concerns evidence from studies where the role of various selected polymorphisms is reported in the context of obesity and sarcopenia.
Figure 3. Genes related to obesity and sarcopenia (green—confirmed obesity-related gene polymorphisms; red—sarcopenia-related gene polymorphisms; yellow—probable gene polymorphisms in SO). The hypothesis concerns evidence from studies where the role of various selected polymorphisms is reported in the context of obesity and sarcopenia.
Genes 15 00206 g003
Table 1. Criteria of sarcopenia according to national consensuses.
Table 1. Criteria of sarcopenia according to national consensuses.
Criteria of Sarcopenia
OrganizationCriteriaReference
The New Mexico Elder Health Survey, 1993–1995 (1997) (1998)Reference population: Rosetta study (n = 883) [18]
Sarcopenia definition: an appendicular skeletal muscle mass (kg)/height2 (m2) less than two standard deviations below the mean of a young reference group.
[18,19]
International working group on sarcopenia—consensus of geriatricians and scientists (2011)Sarcopenia definition: the age-associated loss of skeletal muscle mass and function.

Diagnosis of sarcopenia:
  • gait speed < than 1 m·s−1
  • objectively measured low muscle mass—appendicular mass relative to ht2 (M ≤ 7.23 kg/m2 and F ≤ 5.67 kg/m2)
[17]
The Foundation for the National Institutes of Health Biomarkers Consortium Sarcopenia Project, (2014)1. Weakness
  • Handgrip strength M < 26 kg; F < 16 kg
  • Handgrip strength adjusted for BMI: M < 1.0 and F < 0.56
2. ALM: M < 19.75 kg and F < 15.02 kg
3. ALM adjusted for BMI: M < 0.789 and F < 0.512
[20]
The European Working Group on Sarcopenia in Older People 2 (EWGSOP2) (2019)Criteria
1. Low muscle strength
2. Low muscle quantity or quality
3. Low physical performance

Sarcopenia:
  • probable—fulfilled criterion 1
  • diagnosed—fulfilled criterion 1 + documented criterion 2
  • severe—all criteria fulfilled
[2]
Australia and New Zealand. The Australian and New Zealand Society for Sarcopenia and Frailty Research (ANZSSFR) Sarcopenia, (2023)1. Screening tools
  • SARC-F may (is consistent with the EWGSOP2 algorithm)
  • clinical suspicion (falls, weight loss, feeling weak, ↓ mobility
2. low muscle strength (handgrip strength, chair stand test) or clinical performance (decreed gait seed)
3. sarcopenia conformation: DXA, BIA, CT, MRI
[21]
↓—decrease; ALM = appendicular lean mass; BMI = body mass index; M—male; F—female; DXA—dual X-ray absorptiometry, BIA—bioelectrical impedance analysis; CT—computer tomography; MRI—magnetic resonance; EWGSOP2—the European Working Group on Sarcopenia in Older People.
Table 3. Genetic background of obesity, sarcopenia, and SO.
Table 3. Genetic background of obesity, sarcopenia, and SO.
Disease and/or Its Definitions UsedGenetic Risk FactorStudied Population; nDefinition Used/ConclusionCitation
Obesity or impaired metabolic functionsFTO (rs9939609)Korean adult women; n = 3335
-
association of FTO with body fat markers
[108]
ADRB3adults; n = 265
-
correlation of ADRB3 hypermethylation with overweight, obesity, higher waist-hip ratio, trans-fat intake and altered lipid profile
[115]
schoolchildren; n = 72Trp64Arg and Arg64Arg allele carriers present with higher LDL-c levels and lover maximal fat oxidation rates[117]
Obesity or impaired metabolic functions
(BMI ≥ 30 kg/m2)
SLC6A14French-Caucasian adults; n = 1267, controls = 64920649 C > T and 22510 C > G polymorphisms are associated with obesity[127]
Finnish men; n = 117, controls = 182association of SLC6A14 polymorphisms with obesity[126]
n = 722 Europeans22510 C > G allele reduces fasting fat oxidation[131]
SarcopeniaFTOn = 2207↓ LMI in 29 SNP of FTO[109]
Sarcopenia %SMM sarcopenia definition usedFTO (rs9939609)women > 60 years; n = 307↑ sarcopenia risk[110]
NOS3 (rs1799983)
SarcopeniaGDF5 (rs143384)Europeans > 60 yeas; n = 256.523↓ grip strength[85]
Sarcopenia
SMI < 29.0% in men and SMI < 22.8% in women
FADS2 (rs97384)men and women > 50 years;
n = 1599
controls = 23,391
-
various factors might offset the genetic impact
-
exercise might overcome the genetic effect
[66]
MYO10 (rs3157)
KCNQ5 (rs6453647)
DOCK5 (rs11135857)
LRP1B (rs74659977)
SO
SMI < 6.76 kg/m2, HGS < 28.5 kg and BF% > 38%
ACTN3 (rs1815739)older women in England; n = 307Sarcopenia in obese women[151]
MTHFR (rs1801131)
MTHFR (rs1537516)
↓—decrease; ↑—increase; LMI—lean mass index (lean soft tissue/square of height); %SMM—skeletal muscle mass; SMI—skeletal muscle index (SMM/height2); HGS—handgrip strength; BF%—body fat%.
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

Mazurkiewicz, Ł.; Czernikiewicz, K.; Grygiel-Górniak, B. Immunogenetic Aspects of Sarcopenic Obesity. Genes 2024, 15, 206. https://doi.org/10.3390/genes15020206

AMA Style

Mazurkiewicz Ł, Czernikiewicz K, Grygiel-Górniak B. Immunogenetic Aspects of Sarcopenic Obesity. Genes. 2024; 15(2):206. https://doi.org/10.3390/genes15020206

Chicago/Turabian Style

Mazurkiewicz, Łukasz, Krystian Czernikiewicz, and Bogna Grygiel-Górniak. 2024. "Immunogenetic Aspects of Sarcopenic Obesity" Genes 15, no. 2: 206. https://doi.org/10.3390/genes15020206

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