202307戴宛霖Inflamed Adipose Tissue

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Endocrinology, 2023, 164, 1–13

https://doi.org/10.1210/endocr/bqad094
Advance access publication 8 June 2023
Mini-Review

Inflamed Adipose Tissue: Therapeutic Targets for

Downloaded from https://academic.oup.com/endo/article/164/7/bqad094/7192118 by Chulalongkorn University - Faculty of Medicine user on 27 June 2023
Obesity-related Endothelial Injury
Wanlin Dai,1,2 Ziqi Liu,1,2 Shaojie Yang,3 and Jing Kong3
1
Innovation Institute, China Medical University, Shenyang, 110122, Liaoning, China
2
Health Science Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang,
110122, Liaoning, China
3
Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
Correspondence: Jing Kong, PhD, Department of General Surgery, Shengjing Hospital of China Medical University, Sanhao St, Shenyang, 110004, Liaoning,
China. Email: jingkong@cmu.edu.cn.

Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide and is primarily associated with obesity, visceral adiposity, and unhealthy
perivascular adipose tissue (PVAT). The inflammatory polarization of immune cells residing in adipose tissue and abnormal levels of adipose-
related cytokines are crucial factors contributing to the pathogenesis of metabolic disorders. We reviewed the most relevant papers in the
English literature regarding PVAT and obesity-related inflammation and CVD, aiming to explore potential therapeutic targets for metabolic
alterations related to CV health. Such an understanding will help determine the pathogenetic relationship between obesity and vascular injury
in efforts to ameliorate obesity-related inflammatory responses. In the context of obesity, dysregulation of adipose tissue immune function,
which consists of immune cells and adipose-derived cytokines, plays a crucial role in vascular injury and endothelial dysfunction, especially
the PVAT. Metabolic changes between typical VAT and PVAT in obese conditions would be beneficial in improving the risk of obesity-induced
endothelial dysfunction and CVDs.
Key Words: adipose immunity, endothelial dysfunction, insulin resistance, perivascular adipose tissue
Abbreviations: AdipoR1, adiponectin receptor 1; Ang II, angiotensin II; BAT, brown adipose tissue; CVD, cardiovascular disease; EC, endothelial cell; EnNaC,
endothelial cell Na+ channel; ER, estrogen receptor; FFA, free fatty acid; HIF-1α, hypoxia-inducible factor-1α; IL-33, interleukin 33; IR, insulin resistance; MCP-1,
macrophage chemoattractant protein-1; MR, mineralocorticoid receptor; NK, natural killer; NO, nitric oxide; PI3K, phosphatidylinositol 3-kinase; PVAT,
perivascular adipose tissue; SAT, subcutaneous adipose tissue; Th17, T-helper 17 cells; TNF-α, tumor necrosis factor-α; UCP1, uncoupling protein 1; VAT,
visceral adipose tissue; WAT, white adipose tissue.

Introduction: Inflamed Perivascular Adipose are associated with the immune function of adipose tissue. In
Tissue Is One of the Key Elements in the the obese state, the new formation of adipocytes and broad re­
cruitment of proinflammatory immune cells, especially M2
Progression of Endothelial Injury Under macrophages, give rise to a major source of proinflammatory
Obesity cytokines, contributing to adipose tissue meta-inflammation
Obesity is a major global health issue that must be addressed. (5). Furthermore, the accumulation of dysfunctional adipose
In recent decades, a dramatic increase in the prevalence of tissue leads to adipocyte apoptosis or death, metabolic stress,
obesity has been observed in all life stages. Evidence suggests and hypoxia, subsequently triggering a proinflammatory re­
a strong association between the prevalence of obesity and the sponse (6). The proinflammatory immune response within
growing incidence of metabolic disorders. Obesity, character­ adipose tissue induces high levels of inflammatory serum hall­
ized by meta-inflammation, referred to localized inflamma­ marks accompanied by endothelial dysfunction and vascular
tory responses within the adipose, considerably increases the stiffness, which are strongly related to CVDs (7). Given the
risk of metabolic disorders such as insulin resistance (IR), underlying relationship between obesity, low-grade inflam­
endothelial dysfunction, and cardiovascular diseases (CVDs) mation, and metabolic alterations, targeting imbalanced
(1). Owing to excessive nutrition and inadequate physical ac­ metabolic pathways is a promising therapeutic approach to
tivity, obesity-related chronic inflammation affects people avoid immune-metabolic alterations that contribute to the
worldwide, particularly in developed regions (2, 3). Under progression of the metabolic complications of obesity (8).
conditions of obesity, the quantity and quality of adipocytes Managing the number of immune cells residing in adipose
changes. With an unhealthy expansion of adipose tissue, in­ tissue (9, 10), inhibiting the pathogenic transition form of
sufficient angiogenesis results in an imbalance both in oxygen proinflammatory subtypes (11-13), lowering the levels of in­
supply and lipid metabolism (4). Existing evidence indicates flammatory cytokines derived from the dysfunctional adipose
that cytokines derived from both adipocytes and immune cells tissue (14, 15), and improving the systemic metabolic

Received: 4 February 2023. Editorial Decision: 4 June 2023. Corrected and Typeset: 19 June 2023
© The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail:
journals.permissions@oup.com
2 Endocrinology, 2023, Vol. 164, No. 7

situation may be supportive for obesity therapy and CV content, also known as a stromal vascular fraction, including
health, which is of great significance (16, 17). preadipocytes, fibroblasts, endothelial cells (ECs), adipose
In this review, we first analyze the current understanding of tissue-derived stem cells, and adipose tissue-resident immune
the heterogeneity of adipose tissue and metabolic alterations cells—is highly involved in maintaining adipose tissue
of adipose tissue in the context of obesity, emphasizing the homeostasis (26). Preadipocytes are crucial for regulating
role of adipose tissue-derived immune cells and cytokines in adipocyte differentiation, such as WAT expansion and fat

Downloaded from https://academic.oup.com/endo/article/164/7/bqad094/7192118 by Chulalongkorn University - Faculty of Medicine user on 27 June 2023
inducing obesity-associated inflammatory responses. Second, browning (27). Fibroblasts and ECs provide newly formed
we highlight the importance of inflamed perivascular adipose capillaries and a vasculature microenvironment (28).
tissue (PVAT) in the progression of obesity-induced endothe­ Adipose tissue–derived stem cells are ideal stem cell popula­
lial dysfunction. Finally, we focus on the correlations between tions that have the potential to differentiate into various phe­
abnormal levels of proinflammatory biomarkers and metabol­ notypes regulated by bioactive molecules, which lay the
ic alterations, including IR, endothelial dysfunction, and CV foundation for preventing de novo lipogenesis or obesity
health. Such an understanding will help determine the patho­ (29). Immune cells in adipose tissue include natural killer
genetic relationship between obesity and CVD in efforts to (NK) cells, NKT cells, monocytes, macrophages, dendritic
ameliorate obesity-related inflammation and identify poten­ cells, mast cells, T cells, B cells, and plasma B cells. The great
tial therapeutic approaches that target specific immune- abundance and variety of immune cells resident in the adi­
metabolic dysregulation during obesity. Our paper aims to pose provides the possibility of the fat participating in im­
systematically highlight the association of the inflammatory mune function and inflammatory responses. Existing
phenotype of typical VAT and PVAT with a wide range of evidence has already shown that crosstalk mediated by adi­
metabolic alterations, especially hyperinsulinemia and IR, pokines or cytokines between immune cells and adipocytes
and the increased risk of CVD in the presence of obesity. plays an indispensable role in exerting immune functions
and regulating metabolic processes (25).

White Adipose Tissue: A Crucial Regulator in


the Progression of Obesity-mediated Brown Adipose Tissue: An Important
Inflammation Regulator in Anti-Inflammatory Responses and
Histologically, there are 2 major subsets of adipose tissue: the Pathogenesis of Cardiovascular Risks
white adipose tissue (WAT) and brown adipose tissue (BAT) Adipose tissue is a dynamic organ characterized by the transi­
(Fig. 1). Based on the existing evidence, WAT plays an import­ tion from WAT to BAT via the upregulation of peroxisome
ant role in energy homeostasis and endocrine homeostasis. It proliferators-activated receptors (PPARα/γ), peroxisome
is highly responsive to regulators such as hormones and cyto­ proliferators-activated receptor γ coactivator 1α (PGC-1α),
kines to store excessive energy as neutral lipids, release free and PR domain-containing 16 (Prdm16), while the phosphor­
fatty acids (FFAs), and glycerol during nutrient deprivation. ylation of PPARγ plays an indispensable role in the transition
In addition to energy homeostasis, WAT is considered a func­ from BAT to WAT (30). BAT is a critical endocrine organ,
tional endocrine or immune organ via cytokines or adipokines which is responsible for nonshivering thermogenesis and en­
derived from adipocytes and resident immune cells, such as ergy consumption, especially in infants or during cold expos­
leptin and adiponectin (18, 19). In addition, owing to the dis­ ure, and activation of the sympathetic nervous system.
tribution of WAT, we distinguished between subcutaneous Current evidence has paid additional attention to its promis­
adipose tissue (SAT) found under the skin and visceral adipose ing therapies for anti-inflammatory responses induced by
tissue (VAT) located in the abdominal area. Approximately WAT under obese conditions (31). To put it simply, BAT
80% of the WAT is stored in subcutaneous regions located plays an influential role in enhancing metabolic levels and cor­
in the gluteal, femoral, and abdominal regions in lean, healthy recting metabolic disorders and further exerts beneficial ef­
individuals (20). In addition, the distribution of SAT follows fects on obesity-related inflammation. Previous studies have
sex-dependent trajectories; healthy women tend to accumu­ proposed potential therapeutic strategies for increasing the
late adipose in the lower part of the body, such as gluteofe­ proportion of BAT in the prevention of obesity and related
moral SAT, and men in the abdominal SAT (21). VAT, metabolic diseases. Gunawardana (32) also revealed the pos­
accounting for approximately 20% of the total adipose tissue, sibility for correcting type 1 diabetes via embryonic BAT
surrounds internal organs and includes perirenal, retroperi­ transplants. More important, Becher et al (33) have reported
toneal fat, and omental or mesenteric depots around the intes­ that BAT was independently correlated with lower odds of
tines, which have essential roles in the metabolic obesity-related metabolic disorders and cardiometabolic
microenvironment of visceral organs (22, 23). Unhealthy ex­ risks. The protective role of BAT on CV health is of great im­
pansion of visceral adipocytes may contribute to the impair­ portance in individuals with overweight or obesity. However,
ment of corresponding organs, which is also termed several recent studies have presented conflicting opinions that
adiposopathy, which includes cardiomyopathy, nephropathy, BAT activation may be detrimental to the CV system (34).
and enteropathy (24). Induction of BAT activation is associated with a significant in­
Based on integrated single-cell and spatial transcriptomic crease in heart rate to meet the need to maintain a balance be­
maps of human WAT, Massier et al (25) reported 4 major tween increased energy expenditure and elevated oxygen
cell classes, including adipocytes, fibroblasts, and adipogenic demand levels. A chronic BAT activation via β-adrenergic sig­
progenitors, vascular, and immune cells. Adipocytes, the dom­ naling pathway or thyroid hormone replacement is found to
inant cell type in WAT, account for approximately 80% to be related to serious CV side effects, such as stroke or myocar­
90% of the volume of the tissue and only a third of the cellular dial infarction (35, 36). Additionally, Blondin et al (37) found
composition. The rest of the adipose tissue—nonadipocyte that β3-adrenergic receptor (β3-AR), which used to be
Endocrinology, 2023, Vol. 164, No. 7 3

Downloaded from https://academic.oup.com/endo/article/164/7/bqad094/7192118 by Chulalongkorn University - Faculty of Medicine user on 27 June 2023
Figure 1. Biological characteristics and function of different adipose tissue depots. Comparison of white adipose tissue, brown adipose tissue, and
perivascular adipose tissue with respect to their localization, morphology, and biological function in human-specific depots. Principal metabolic
alterations that occur in the context of obesity, together with adipose tissue remodeling, are highlighted.

thought to be responsible for the activation both of human Abnormal Immune Function in Unhealthy
and rodent BAT, did not play such a critical function in human Adipose Tissue Leads to Endothelial
beings, being displaced by β2-AR. We should note that most
Dysfunction: Focus on Typical Visceral Adipose
of the mechanisms related to the activation of BAT are derived
from rodent studies; taking into account the notable differen­ Tissue and Perivascular Adipose Tissue
ces between species, more clinical research is needed to In obesity, WAT expands unhealthily, leading to increased
confirm the molecular theoretical basis of targeted BAT for oxygen consumption. Existing evidence indicates that ele­
the treatment of obesity-related metabolic disorders. vated expression of hypoxia-inducible factor-1α (HIF-1α) in
Furthermore, given the conflicting conclusions on whether adipocytes mediated by hypoxia is a key trigger of
BAT can be used as an intervention target to improve obesity-related inflammation. Furthermore, intracellular
obesity-related metabolic disorders, including CV risk factors, proinflammatory signals, including c-Jun N-terminal kinase
BAT-targeted therapeutics should be carefully assessed to (JNK) and nuclear factor κB are also activated in response
avoid adverse effects. to excessive neutral lipids accumulated in adipocytes (42,
Uncoupling protein 1 (UCP1) is a major regulator of brown 43). In addition, imbalanced cytokines or adipokines derived
and beige adipocyte energy expenditure and metabolic from failed crosstalk between adipocytes and immune cells re­
homeostasis (38). Cysteine-253 is a regulatory site on UCP1 sult in an initial inflammatory response within adipose tissue
that elevates protein activity on covalent modification. Lack (44). Here, we focused on the primary advances in the latest
of C253 results in adipose tissue redox stress, which drives data on the innate immune cells residing in and recruited to
substantial immune cell infiltration and systemic inflamma­ adipose tissue, especially PVAT, during obesity (Fig. 2).
tory pathology in adipose tissues and liver of male, but not fe­ Notably, emerging evidence on innate immune function in
male, mice. Elevation of systemic estrogen reverses this obese adipose tissue has shown that cytokine profiles secreted
male-specific pathology (39). A decrease in proteins associated by different subsets of immune cells have both anti-
with antigen processing and presentation, phagocytosis, and inflammatory and proinflammatory effects on adipose tissue
B-cell signaling was observed in BAT from mice models fol­ in individuals with obesity. However, the mechanisms of
lowing β-estradiol supplementation, due to estrogen’s roles mediators or effectors upstream and downstream of proin­
in suppressing inflammatory signaling in macrophages.(39) flammatory phenotypes activation, proliferation, and polar­
Furthermore, the intact estrogen signal pathway plays a role ization in obese adipose tissue are still not fully understood.
in increasing the resilience to nutritional/metabolic stress That is, conflicting experimental data are still being observed
and prevents adipose dysfunction via the regulation of key as predictive opinions are being proposed. Targeting these im­
genes involved in antioxidant processes. Estrogen receptors mune cells and regulatory cytokines is expected to be a poten­
(ERs), including ERα, ERβ, and GPR30, are found on the sur­ tial strategy for constraining obesity-associated adipose tissue
face of immune cells and can, thus, modulate the immune re­ inflammation and systemic metabolic alterations.
sponse. 17-β Estradiol inhibits proinflammatory T-helper 17 In contrast to other typical fat depots, PVAT is character­
cells (Th17) and M1 macrophage cell differentiation and en­ ized by dynamic and flexible functional properties between
hances regulatory T cells and M2 macrophage expansion white and brown adipocytes. Adipocytes in PVAT exhibit
(40). Estrogen expressed in adipocytes has been shown to epi­ more WAT histological properties while being less differenti­
genetically promote beiging in mice by promoting demethyla­ ated and having less lipid storage capacity and advanced po­
tion of the Adrb3 and Dio2 promoters, leading to increased tential to release cytokines or nutrient factors as compared
Ucp1 expression (41). to typical WAT (45). Unhealthy PVAT is regarded as a key
4 Endocrinology, 2023, Vol. 164, No. 7

Downloaded from https://academic.oup.com/endo/article/164/7/bqad094/7192118 by Chulalongkorn University - Faculty of Medicine user on 27 June 2023

Figure 2. Immune cells in typical visceral adipose tissue (VAT) and perivascular adipose tissue (PVAT) during childhood obesity. During obesity,
unhealthy adipocytes experience increased quantity and quality. Crown-like structures surrounding dead or apoptotic adipocytes consist of Trem2+/
CD9+ macrophages and adipose tissue dendritic cells, which are the hallmarks of obese adipose tissue. Immune cells infiltrated in adipose tissue
contribute to oxidative stress activation and proinflammatory cytokines secretion, subsequently leading to systemic inflammation and other metabolic
alterations. In addition to the understanding of typical VAT, where the dominant role of infiltrating proinflammatory immune cells has received the most
attention, key regulatory models of accumulation immune cells contributing to vascular remodeling or stiffness via an impaired vasodilation state. In
contrast to other typical fat depots, PVAT is characterized by a significant accumulation both of PVAT M1 and PVAT M2 macrophages that have been
observed during metabolic-induced vascular diseases, which participates in proinflammatory responses and profibrotic responses separately.
Excessive B cells and immunoglobulin deposits lead to the formation of atherosclerotic plaques. Current understanding proposes that PVAT not only
interacts with typical VAT via endocrine proinflammatory signals, but also functions as a source of cytokines and chemokines. Taken together, the
dysfunction of immune cell-mediated adipose tissue remodeling plays an integral role in the development of obesity-related cardiovascular injury. CD,
cluster of differentiation; IFN-γ, interferon-γ; IL-1β, interleukin-1β; MAIT cell, mucosal-associated invariant T cell; NCR1, NK-cell–activating receptor; NK
cell, natural killer cell; Th1, major T helper 1; TNF-α, tumor necrosis factor-α; Trem2; triggering receptor expressed on myeloid cells-2.
Endocrinology, 2023, Vol. 164, No. 7 5

element involved in the microenvironment of the vasculature inflammatory effects of the aldosterone appear to be related
system and plays an indispensable role in vascular pathologies to the induction of osteopontin release in proinflammatory
(7). While the previous view considered PVAT to provide sim­ macrophages and T cells, while the UCP-1 transcription and
ple mechanistic support and protection of vascular structures, thermogenic activity decrease in BAT (56, 57). It is important
current knowledge has clarified the role of PVAT in regulating to note that, in the case of cardiovascular health, the activa­
the metabolic phenotype and physiological function of the tion of the aldosterone system induces an increase in oxidative

Downloaded from https://academic.oup.com/endo/article/164/7/bqad094/7192118 by Chulalongkorn University - Faculty of Medicine user on 27 June 2023
endothelium (46). Data have shown that both the quantity stress and leads to adverse effects (57). Endothelial MR over­
and quality of PVAT change during the pathogenesis of activation under the condition of obesity leads to the overex­
CVDs, characterized by altered cellular composition and pression of the intracellular adhesion molecule 1 (ICAM-1) in
proinflammatory phenotypes. In obesity, PVAT dysfunction a sex-specific manner, thereby enabling leukocyte adhesion to
depends on insufficient angiogenesis, oxidative stress, and in­ coronary artery ECs (58).
flammatory characteristics (47). As mentioned earlier, both the endocrine effect of classical
In the context of obesity, along with low-grade inflamma­ WAT and paracrine factors secreted from inflammatory cells
tion, a large number of immune cells infiltrate the blood ves­ infiltrating the PVAT contribute to endothelial dysfunction,
sels. Stimulated by chemotaxis, immune cells are recruited which is characterized by decreased production or affected
to and further accumulate in the PVAT at high frequency, as sensitivity to vasoprotective regulators. In addition, excessive
well as in the vessel walls, which are critical in the pathogen­ concentrations of FAAs have been well accepted for their
esis of atherosclerosis and vascular disease during obesity effects on vascular IR through serine/threonine phosphoryl­
(47). Numerous differences in the innate and adaptive im­ ation of insulin receptor substrates. These interfere with the
munological properties of PVAT inflammation in vascular downstream activation of the phosphatidylinositol 3-kinase
pathologies, characterized by distinct cellular interactions be­ (PI3K) signaling pathway, subsequently leading to the im­
tween immune cells and the endothelium, have been found pairment of insulin-related vasodilation (48). Consistent
when compared with classic meta-inflammation within the with the prevailing view, studies have provided insight into
VAT in the context of obesity (48). In addition to understand­ the direct action of FAAs on the eNOS enzyme, resulting in
ing typical VAT, for which the dominant role of infiltrating impaired vasodilation (59).
proinflammatory immune cells has received the most atten­ In summary, available experimental results suggest that
tion, key regulatory models of immune cell accumulation con­ PVAT plays an indispensable role in the development of endo­
tribute to hypertensive vascular remodeling via a profibrotic thelial dysfunction. Furthermore, the current understanding
phenotype. In contrast to the M2 polarization of adipose proposes that PVAT not only interacts with typical VAT
tissue macrophages, which is responsible for a potent anti- through endocrine proinflammatory signals but also functions
inflammatory effect, with a decreased frequency during obes­ as a source of cytokines and chemokines. Given the limited
ity, significant accumulation both of PVAT M1 and PVAT M2 mechanistic studies on obesity-induced PVAT inflammation,
macrophages has been observed during metabolic-induced further studies should be performed to investigate the precise
vascular diseases, which participate in proinflammatory re­ function and underlying regulatory processes of inflamed
sponses and profibrotic responses, respectively (49). Data PVAT, which may help identify therapeutic targets for
also illustrate that increased B-cell activation in the PVAT dur­ obesity-related CV injury.
ing obesity is involved in atherosclerotic pathogenesis, pos­
sibly related to aortic antibody deposition and B-cell
infiltration within atherosclerotic plaques (50). Chan et al Metabolic Alterations Related to Adipose
(50) also found that B-cell depletion prevents angiotensin II Tissue Immune Dysfunction Play a Vital Role in
(Ang II)-induced hypertension. In addition, perivascular eosi­
the Pathogenesis of Cardiovascular Diseases
nophils play a vital role in mediating the effects of PVAT on
vascular health (51). Eosinophils are strongly involved in ad­ Inflammatory Factors
renergic signaling, nitric oxide (NO)-related vasodilation, and Increasing observational evidence has focused on alterations
adiponectin-dependent–positive mechanisms within PVAT in circulating inflammatory factors in response to obesity
(52). Eosinophil dysfunction has been observed during obesity (60). Previous cross-sectional studies and longitudinal ana­
or high-fat diet feeding, probably contributing to hyperten­ lyses of individuals with obesity showed that weight gain
sion (52). According to animal model data, manipulating was accompanied by increased levels of macrophage chemo­
the frequency of vascular eosinophils via interleukin 33 attractant protein-1 (MCP-1), IL-6, and TNF-α, which are
(IL-33) during obesity may be a valid target for obesity-related largely related to vascular IR and endothelial dysfunction, es­
vascular dysfunction and CV complications (53). pecially proinflammatory factors derived from perivascular
Furthermore, the obese state increases the amount of min­ adipocytes (Table 1) (61).
eralocorticoid receptors (MRs) and circulating aldosterone, MCP-1 initiates various monocyte-mediated proinflamma­
which subsequently induces a chronic activation of Ang II tory signals and monocyte chemoattractant activities during
type 1 receptors and a suppression of epithelial nitric oxide obesity. It has been established that ECs are well characterized
synthase (eNOS) activity, thus related to the decrease in NO in response to MCP-1 stimulation by binding with CC chemo­
concentration and bioavailability (47, 54). In addition, ele­ kine receptor 2, subsequently facilitating the formation of
vated levels of aldosterone significantly upregulate nicotina­ foam cells and vulnerability to thromboembolism (62).
mide adenine dinucleotide phosphate oxidase (NAPDH Importantly, reducing circulating MCP-1 levels has been pro­
oxidase) components in various cell types (55). In terms of posed as a promising therapeutic approach to ameliorate ath­
the adipose tissue, an increase has been observed in the expres­ erosclerotic and thromboembolic risks (63). TNF-α is a potent
sion of proinflammatory cytokines such as IL-6 and tumor proinflammatory cytokine released by expansive adipocytes,
necrosis factor-α (TNF-α), and the amplification of infiltrated macrophages, and lymphocytes that plays a vital
6 Endocrinology, 2023, Vol. 164, No. 7

Table 1. Changes in circulating inflammatory factors and adipokines during obesity and their potential role in endothelial dysfunction

Marker Change Biological process Potential role in endothelial dysfunction Reference

Serum ↑ Monocyte chemoattractant activities Increased circulating MCP-1 levels with obesity facilitate the (62, 63)
MCP-1 formation of foam cells and vulnerability to thromboembolism via
CCR2

Downloaded from https://academic.oup.com/endo/article/164/7/bqad094/7192118 by Chulalongkorn University - Faculty of Medicine user on 27 June 2023
Serum IL-6 ↑ Pleiotropic but main IL-6 depletion inverses the impairment of endothelial function. (64-66)
proinflammatory effects Increased circulating IL-6 levels inhibit eNOS phosphorylation and
reduce NO bioavailability, further impairing vasodilation
Serum ↑ Proinflammatory effects, induce Increased circulating TNF-α levels interfere with IRS-1 and Akt (67-69)
TNF-α synthesis of acute-phase protein phosphorylation, associated with vascular insulin resistance.
Interaction between TNF-α and IL-6 aggravates vascular
inflammation and vascular stiffness
Leptin ↑ Appetite regulation, Elevated leptin serum levels initiate systemic inflammation via (70, 71-
proinflammatory effects monocyte/macrophage chemotaxis. Leptin potentially functions as 73)
a protective element in cardiovascular tissues
Adiponectin ↓ Anti-inflammatory effects, insulin According to animal models, adiponectin is highly involved in (74-79)
sensitization vasodilation and revascularization through T-cadherin and
transmembrane receptors AdipoR1 and AdipoR2

Abbreviations: AdipoR1, adiponectin receptor 1; CCR2, C-C motif chemokine receptor 2; eNOS, epithelial nitric oxide synthase; IL-6, interleukin-6; IRS-1,
insulin receptor substrate 1; MCP-1, macrophage chemoattractant protein-1; NO, nitric oxide; TNF-α, tumor necrosis factor-α.

role in mediating systemic inflammation, leading to the syn­ target for preventing or treating impaired endothelium-
thesis and secretion of acute-phase proteins, such as high- dependent vasodilation in microcirculation induced by
sensitivity C-reactive protein and fibrinogen. In addition, obesity-related inflammation.
TNF-α is responsible for activating obesity-induced oxidative
stress, leading to the inhibition of insulin signaling (63).
Schinzari et al (67) indicated that administering the monoclo­ Abnormal Levels of Adipokines
nal antibody infliximab to neutralize TNF-α significantly im­ Adipokines, also known as adipose-derived cytokines, are se­
proved vascular IR. Elevated concentrations of circulating creted by adipocytes or produced by immune and mesenchy­
and perivascular TNF-α in response to obesity-related inflam­ mal cells in WAT. Adipokines are considered one of the
mation and IR interfere with insulin receptor substrate-1 and hallmarks of obesity and play prominent roles in the patho­
Akt phosphorylation (68). Moreover, TNF-α has been widely genesis of systemic low-grade inflammation and subsequent
acknowledged for its ability to regulate the mitogen-activated metabolic alterations (70, 81). Here, we focus on major ad­
protein kinase (MAPK) pathway, directly impairing vances in the latest studies on leptin and adiponectin, the first
endothelium-dependent vasodilation and vascular insulin sen­ pair of adipokines to be extracted and identified, that act as
sitivity (69). Lee et al (64) further suggested that TNF-α could antagonistic mediators in regulating inflammatory responses
be a mediator of IL-6, functioning as a contributor to the associated with adipose tissue activity (see Table 1). An imbal­
pathogenesis of endothelial dysfunction of coronary microcir­ ance in circulating leptin and adiponectin levels has been ob­
culation in animal models of type 2 diabetes. Studies on IL-6 served during obesity and is a typical biomarker of
signaling in the context of obesity and obesity-related endo­ metabolic alterations in individuals with obesity (82).
thelial dysfunction have initially focused on experimental ob­ Leptin is a proinflammatory cytokine released from WAT.
servations (65, 66). Furthermore, IL-6 is largely associated Previous data have shown that in children and adults who
with a high risk of atherosclerosis via the upregulation of are overweight, leptin levels increase both in adipose tissue
the Ang II and renin-angiotensin system (80). Recent studies and circulation, whereas the opposite is true in the case of
have reported that IL-6 directly impairs coronary arteriolar weight loss (71). In addition, based on data from clinical trials,
endothelium-dependent vasodilation in mice with type 2 dia­ high baseline leptin levels are associated with a slight reduc­
betes via both overactivation of oxidative stress and inhibition tion in adipose mass in individuals with obesity, which is
of eNOS enzyme phosphorylation, leading to attenuated closely related to IR (72). Furthermore, in obesity, leptin func­
NO-signaling pathways within the endothelium (64). tions as a potent monocyte/macrophage chemoattractant that
Moreover, current evidence emphasizes the possible role of induces the increased release of proinflammatory cytokines
the interactive relationship between TNF-α and IL-6 in vari­ such as TNF-α, IL-6, and MCP-1 by monocytes and
ous vascular inflammation and endothelial dysfunction stages. macrophages (73). However, existing evidence demonstrates
Two hypotheses for the interaction between TNF-α and IL-6 that leptin activates the protective JAK-STAT and
have been proposed: IL-6 expression could be regulated by AMP-activated protein kinase (AMPK) signaling pathways
TNF-α stimulation, or IL-6 could be an essential upregulator in cardiovascular tissues (70). Recent studies have focused
of TNF-α, indicating a strong relationship between IL-6 and on the effects of leptin deprivation on CV injuries. Hence, giv­
TNF-α that contributes to endothelial dysfunction (64). en the leptin-related direct supportive effects and collateral
More research is needed to elucidate the detailed mechanisms adverse effects via obesity-related systemic inflammation in
of reciprocal inhibitory regulation at the molecular level. The CV tissues, further studies are required to reveal the relation­
novel findings of the interactive possibility acting between ship between circulating leptin levels and CV tissue metabol­
IL-6 and TNF-α as an inflamed regulatory site offer a potential ism in the context of obesity. Tissue-targeted therapeutic
Endocrinology, 2023, Vol. 164, No. 7 7

technologies may help address these contradictory metabolic Subsequently, dysfunction of organelles related to metabolic
properties. balance, such as mitochondria, endoplasmic reticulum, and
In contrast, adiponectin is largely associated with anti- lysosomes, has been observed in the progression of lipotoxic­
inflammatory responses, with reduced levels observed in ity, with the regeneration of inflammatory mediators within
individuals with obesity. As circulating adiponectin levels the affected tissues (88). Thereafter, several immune cells res­
decrease in the obese state, recent data further suggest that iding in adipose tissue or other organs, both adaptive and in­

Downloaded from https://academic.oup.com/endo/article/164/7/bqad094/7192118 by Chulalongkorn University - Faculty of Medicine user on 27 June 2023
adiponectin administration alleviates diabetic cardiomyocyte nate immune cells, were found to be closely associated with
apoptosis, exerting a potent protective effect (74). low-grade chronic inflammation and IR in obesity (89, 90).
Adiponectin elicits multiple downstream AMPK and PI3K sig­ In summary, an increasing number of studies have highlighted
naling events, primarily through the transmembrane receptors the crucial role of adipose tissue inflammation and proinflam­
adiponectin receptor 1 (AdipoR1) and AdipoR2 (75, 76). matory cytokines derived from obese adipose tissue as
Studies have proposed that adiponectin may help prevent sys­ contributors to hyperinsulinemia or IR or both (Fig. 3).
tolic and diastolic dysfunction from myocardial infarction and Obesity-related long-term low-grade systemic inflammation
ameliorate atherosclerotic plaque formation and cardiac impairs glucose tolerance and insulin sensitivity, which is
hypertrophy in the pathogenesis of hypertension (77). In add­ one of the initial triggers for individuals with obesity being
ition to AdipoR1 and AdipoR2, T-cadherin has been observed vulnerable to various chronic diseases or metabolic
in the vascular system and is responsible for the cardioprotec­ alterations.
tive role of high-molecular-weight adiponectin (78). Previous Current observational research has shown a higher CV risk
studies have indicated that following T-cadherin ablation, adi­ under obese conditions, or hyperinsulinemia with or without
ponectin fails to exert its short-term and long-term cardiopro­ insulin resistance, or both states, which are highly related to
tective effects during hypertension, myocardial hypertrophy, endothelial injuries and heart failure with preserved ejection
and myocardial ischemia-reperfusion injury (79). fraction (91-93). A previous cohort study of children with
In summary, increasing circulating adiponectin levels is a obesity reported that compared to a group of obese sedentary
promising therapeutic strategy for alleviating obesity, related children, obese children performing physical exercise exhib­
systemic low-grade inflammation, and subsequent obesity- ited improvement in CV risk, estimated by blood pressure,
induced CVDs. However, owing to the limited sensitivity of heart rate, and maximum volume of oxygen, and lower levels
tissue-targeting techniques and potentially severe side effects, of inflammatory biomarkers, such as white blood cells and
drugs that are beneficial for adiponectin synthesis may not be C-reactive protein (94). Similarly, Karason et al (95) also re­
optimally suited for clinical treatment, leading to deviations in ported that individuals with bariatric surgery showed a re­
experimental design and clinical management. duced obesity-related plasma protein profile, associated with
an increased risk of heart failure, compared to controls.
Trouwborst et al (96) revealed in a randomized controlled tri­
Hyperinsulinemia and Insulin Resistance al the positive effects of improved muscle IR and liver IR on
IR is the loss of the role of insulin in maintaining glucose and cardiometabolic health through dietary modification.
lipid homeostasis. In the initial stage of IR, cells become less re­ Compared with healthy controls with normal serum insulin
sponsive to insulin and fail to efficiently use glucose for energy levels, an experimental group with hyperinsulinemia was
metabolism, which in turn leads to increased blood glucose lev­ more susceptible to coronary artery disease. More important,
els. Pancreatic β cells compensate by producing more insulin to both amelioration of insulin sensitivity and weight loss re­
moderate elevated blood glucose levels, resulting in hyperinsu­ versed the increase in CVD risk factors relative to baseline
linemia. As insulin sensitivity worsens, pancreatic β cells (97). Studies have identified related alleles associated with ele­
respond to impaired glucose tolerance by continuously produ­ vated fasting insulin levels or the preclinical stage of IR, which
cing more insulin. Owing to constant overload, the ability of increases the risk of hypertension and CVDs (98). Improving
pancreatic β cells to generate insulin is diminished, further lead­ insulin sensitivity via ketogenic diets reverses the genesis of
ing to an increase in circulating glucose and, finally, IR (83). atherogenic dyslipidemia and systemic inflammation in
It is generally accepted that obesity and long-term low- CVDs (99). In the vasculature, hyperinsulinemia and/or IR
density systemic inflammation are crucial factors in inducing play key roles in the development of atherosclerosis in re­
the affected insulin-signaling pathway, impaired glucose sponse to obesity and systemic low-grade inflammation (93).
homeostasis, and systemic IR. Existing data have shown According to current data from animal models, insulin is re­
that unhealthy enlarged adipose tissue is a key source of in­ sponsible for activating the endothelial cell Na+ channel
flammatory factors in the context of obesity (84). Several po­ (EnNaC), which is highly associated with the stiffening and re­
tential mechanisms underlying the relationship between modeling of the vasculature in a concentration-dependent
obesity-related inflammation and IR have been proposed. manner (100, 101). During feeding a diet rich in refined carbo­
First, with the unhealthy expansion of adipose tissue, expan­ hydrates and saturated fat, mice with EnNaC deletion showed
sive adipocytes release excess FAAs, reactive oxygen species, a significant improvement in vascular stiffness compared with
and proinflammatory cytokines in response to an imbalance that in wild-type controls, while elevated serum insulin levels
in oxygen supply, which plays a key role in inducing the initial were observed (100). Upregulation of EnNaC via overactivity
stage of IR (85). Second, these inflammatory mediators not of insulin stimulation leads to reductions in bioavailable NO
only trigger meta-inflammation within adipose tissues, as through the activation of serum and glucocorticoid-regulated
mentioned before, but also pass through the bloodstream kinase 1 signaling (100). Concisely, in the progression of low­
into other nonadipose organs or tissues, possibly leading to ering insulin sensitivity, the effects of insulin-mediated NO
lipotoxicity (86). In addition, toxic lipids such as ceramides production on vasodilation and further glucose utilization
accumulate in affected tissues, including the liver, heart are impaired, which underlies the pathogenesis of vascular
muscle, and pancreas, further wearing out the cells (87). fibrosis, stiffness, and associated CVDs. In addition,
8 Endocrinology, 2023, Vol. 164, No. 7

Downloaded from https://academic.oup.com/endo/article/164/7/bqad094/7192118 by Chulalongkorn University - Faculty of Medicine user on 27 June 2023
Figure 3. Proinflammatory components of perivascular adipose tissue (PVAT) and metabolic alterations during obesity in the regulation of vascular
homeostasis and vascular dysfunction via hyperinsulinemia or/and insulin resistance. In states of normal body weight (left), healthy PVAT plays a vital
role in maintaining systolic and diastolic activity of blood vessels. Both normal levels of vasodilators, such as adiponectin, and favorable insulin
sensitivity contribute to glucose homeostasis and vascular integrity during the anticontractile process. In the context of obesity (right), inflamed PVAT
and typical visceral adipose tissue (VAT) are highly involved in vascular dysfunction and endothelial stiffness. Increasing levels of free fatty acids,
reduced blood flow, localized hypoxia, apoptosis or necrosis of adipocytes, accumulation of immune cells, and massive production of proinflammatory
cytokines or vasoconstrictors impair insulin sensitivity and subsequently hyperinsulinemia and insulin resistance, resulting in vasoconstriction. In
addition, impaired endothelium shows a decrease in both eNOS activity and NO production, leading to dysfunction of endothelial-dependent
vasodilation. eNOS, epithelial nitric oxide synthase; FFA, free fatty acid; IL-6, interleukin-6; NO, nitric oxide; TNF-α, tumor necrosis factor-α; VAT, visceral
adipose tissue.

renin-angiotensin-aldosterone, and brain melanocortin sys­


tems appear to interact synergistically with direct atherogenic
effects in a state of hyperinsulinemia and/or IR, resulting in
CV system injury (93). In summary, insulin-related signaling
has the potential to provide new therapeutic targets for life­
style interventions and drug development at the intersection
of obesity, systemic inflammation, and CVD pathogenesis.

Outstanding Questions
The prevalence of obesity is particularly high worldwide. In
recent decades, a dramatic increase in the prevalence of obes­
ity has been observed in all life stages. At present, evidence
demonstrates a strong association between the prevalence of
obesity and the growing incidence of unhealthy CV events.
Based on the analysis of the underlying mechanisms or hy­
pothesis, some potential therapies can be attended or maybe
further improved (Fig. 4).

Intervening Early-Life Events Related to Adipose


Immunity and Obesity-related Cardiovascular
Figure 4. Potential therapies for the prevention of obesity-related, Events: The Initial Step of Targeted Therapies
unhealthy cardiovascular events. Specific treatment strategies are
A lack of understanding of the roles of early-life events or child­
summarized in detail in the text.
hood activities that result in high risks for obesity by influencing
adipose tissue immune dysfunction under the pathological con­
hyperinsulinemia may lead to sympathetic overactivity and dition of obesity is a crucial factor of the incidence of
renal sodium retention, further increasing blood pressure. obesity-related CVDs, which occur at younger average ages
Excessive upregulation of the sympathetic nervous, (102, 103). The increased occurrence of obesity during infancy
Endocrinology, 2023, Vol. 164, No. 7 9

Downloaded from https://academic.oup.com/endo/article/164/7/bqad094/7192118 by Chulalongkorn University - Faculty of Medicine user on 27 June 2023
Figure 5. Overview of inflamed adipose tissue and systemic metabolic alterations involved in endothelial dysfunction and cardiovascular injury. First, as
unhealthy adipose tissue expands, hypertrophic adipocytes release excess free fatty acids, reactive oxygen species, and proinflammatory cytokines in
response to an imbalanced oxygen supply. Immune cell infiltration in inflamed adipose tissue is strongly associated with obesity-associated systemic
low-grade inflammation. In addition, toxic lipids, such as ceramides, accumulate in affected tissues, including the liver, heart, muscle, and pancreas,
further wearing out the cells. Second, against enormous nutrients during obesity, pancreatic β cells compensate by producing extra insulin to moderate
the elevated blood glucose levels, which is characterized by hyperinsulinemia. Owing to the constant overload of insulin and the progression of
lipotoxicity, the ability of pancreatic β cells to generate insulin is diminished, which further leads to a vicious cycle with the core of insulin resistance.
Finally, with the accumulation of proinflammatory factors, including vasoconstrictors, and impaired epithelial nitric oxide synthase (eNOS) activity of
endothelial cells, the diastolic process is destroyed, which eventually leads to vasoconstriction, paving the pathogenetic possibility for cardiovascular
disease.

or childhood results in a systemic inflammatory response that is Fig. 2). In addition, inflamed PVAT may play a distinct role
strongly associated with a range of metabolic alterations, in­ in the progression of the pathology of endothelial injury and
cluding unhealthy CV events during adolescence and even vascular stiffness due to the specific location and morphology
into adulthood (104-106). Thus, early intervention taking of the fat depots, which is characterized by different frequen­
into account genetic predisposition, an obesogenic-nurturing cies of infiltrated immune cells compared to typical VAT
environment, and unhealthy lifestyles should receive attention (108). Currently, based on data from animal models, adoptive
(105, 107). However, owing to the limitations of the relevant transfer of anti-inflammatory immune cells to inflamed PVAT,
techniques and the small number of samples from infants and such as regulatory T cells, significantly improves endothelial
children, some interventions may not be fully processed, result­ function and reduces blood pressure (109, 110). Moreover,
ing in low sensitivity of data from some well-designed studies. blocking interactions between proinflammatory immune cells
Further studies should shed light on potential regulators and plays a vital role in ameliorating vascular dysfunction and
signaling pathways in childhood obesity and obesity-related PVAT inflammation, including the crosstalk between macro­
CV events. Simple interpretive language is necessary to facili­ phages, dendritic cells, NK cells, and T cells (108, 111).
tate the generalization of those optimal interventions. However, given that mechanistic studies in inflamed perivas­
cular adipose tissue is still limited, more research should be
performed to investigate the precise relationship between ab­
Control of Inflamed Adipose Tissue in the Context of normal adipose immunity during obesity and inflammatory
Obesity via Regulating the Frequencies of Immune responses of cardiovascular system.
Cells: The Advanced Step of Targeted Therapies
In the context of obesity, crown-like structures surrounding
dead or apoptotic adipocytes consist of Trem2+/CD9+ macro­ Restriction of Abnormal Circulating Inflammatory
phages and adipose tissue dendritic cells, which are the hall­ Factors in the Context of Obesity: The Systemic Step
marks of obese adipose tissue. Both typical VAT and PVAT of Targeted Therapies
are responsible for obesity-related inflammation and subse­ Alterations in circulating inflammatory factors or cytokines
quent inflammatory alterations within other organs (see are reported in previous cross-sectional studies and
10 Endocrinology, 2023, Vol. 164, No. 7

longitudinal analyses of individuals with obesity (see Table 1). Conclusion


Existing animal studies have suggested that TNF-α neutraliza­
In the context of obesity, dysregulation of adipose tissue im­
tion or blockade of TNF-α receptor can promote pathological
mune function, which consists of immune cells and adipose-
changes of myocardial damage, but clinical evidence is still
derived cytokines, plays a crucial role in vascular injury and
lacking. Additionally, leptin repletion possibly reverses
endothelial dysfunction, especially the PVAT (Fig. 5). An in-
ischemia-induced revascularization and the deleterious effects
depth understanding of the roles of adipose tissue immune

Downloaded from https://academic.oup.com/endo/article/164/7/bqad094/7192118 by Chulalongkorn University - Faculty of Medicine user on 27 June 2023
of leptin deficiency. Nonetheless, further studies are required
dysfunction that triggers a series of metabolic alterations dur­
to reveal the relationship between circulating leptin levels
ing obesity, as well as the influence of these inflammatory re­
and CV tissue metabolism in the context of obesity, given
sponses on the pathogenesis of CVDs, will help to identify the
the leptin-related direct supportive effects and collateral ad­
underlying mechanisms involved in adipose tissue immune
verse effects via obesity-related systemic inflammation in CV
dysfunction and CVDs at the molecular level and in the devel­
tissues. Tissue-targeted therapeutic techniques may help ad­
opment of potential targeted therapy. Studies should further
dress these conflicting metabolic properties. Moreover, ac­
focus on the similarities and differences in metabolic changes
cording to data from animal models of adiponectin
between typical VAT and PVAT in obese conditions, which
insufficiency, adiponectin administration plays a critical role
would be beneficial in improving the risk of obesity-induced
in vasculoprotective and angiogenic properties. More basic
endothelial dysfunction and CVDs.
and clinical studies are still needed to identify whether the
beneficial effects of adiponectin repletion exert a direct effect
on cardiovasculature, given abnormal feeding behavior, un­ Search Strategy and Selection Criteria
healthy adipose tissue during obesity (112).
References for this review were identified through searches of
PubMed to identify relevant English-language papers pub­
Potential Aspects of Targeted Therapies: From a lished between January 1, 1980 and July 31, 2022. The search
Comprehensive Perspective term “perivascular adipose tissue” was used in combination
with the “AND” operator for the terms “obesity,” “cardio­
In this paper, we further highlight the potential role of the gut
vascular disease,” “endothelial dysfunction,” “childhood
microbiota and obesity-related metabolic disorders. The gut
obesity,” “obesity-related metabolic alterations,” “early life
microbiome, the largest bacterial colony in the human body,
events,” and “gut microbiota.” The final reference list was
appears to be strongly associated with metabolism (113,
generated based on originality and relevance to the broad
114). The regulatory axis between alterations in gut micro­
scope of this review, with a focus on the most recently pub­
biota and the development or function of adipose tissue may
lished papers.
be a key mediator that plays an essential role in ameliorating
obesity-related inflammation and circulatory risk factors
(115). We note that gut microbiota, under the influence of ex­ Funding
ternal perturbations or endogenous paradigms, could be con­
This work was supported by the Liaoning Science and
sidered as a factor bridging the gap between various lifestyle
Technology Plan Project (grant No. 2021JH2/10300118),
or genetic diversity and subsequent changes in metabolic traits
the Shenyang Science and Technology Innovation Talent
(116). Most CV risk factors, including aging, obesity, in­
Support Program for Youth and Midlife (grant No.
appropriate dietary patterns, and unhealthy lifestyles, have
RC200121), and The 345 Talent Project Program of China
been shown to induce gut dysbiosis, while certain alterations
Medical University Shengjing Hospital (grant No. 2019-40A).
in the gut microbiome further contribute to the pathogenesis
of endothelial injury via adipose tissue dysfunction, sympa­
thetic overactivation, and inflammatory responses in the cir­ Author Contributions
culatory system (117, 118). More important, specific
D.W., K.J., and L.Z. designed the overall study; D.W. and Y.S.
alterations in the gut microbiota under conditions of hyperlip­
collected information and prepared figures; D.W. and K.J.
idemia diets have been shown to result in the recruitment and
performed a systematic literature search and drafted the
activation of B2 cells in PVAT, followed by an increase in cir­
manuscript; and D.W., L.Z., Y.S., and K.J. revised the manu­
culating immunoglobulin G, promoting atherosclerosis (119,
script critically and approved its final version.
120). Studies to date have shown the significance of the com­
mensal microbes and obesity-induced circulatory risks, while
most of existing evidence remains at the phenomenological Disclosures
level (121). There is still a lack of mechanistic studies to ex­
The authors have nothing to disclose.
plore further, particularly in the light of the crosstalk with
PVAT, which is also crucial for estimating or ruling out previ­
ous observations and avoiding potential biases due to con­ Data Availability
founding. In addition, it remains to be further discussed
No new data were generated or analyzed in this research.
whether the study of WAT can be directly extended to
PVAT due to the tissue-specific nature of adipose tissue, and
the relevance of gut microbiome to PVAT should be carefully References
considered. With in-depth understanding of the regulatory 1. Röszer T. Adipose tissue immunometabolism and apoptotic cell
axis among adipose tissue, gut microbiota, and vasculature, clearance. Cells. 2021;10(9):2288.
and the discovering of novel species of gut flora (122-124), 2. Ogden CL, Carroll MD, Kit BK, Flegal KM. Prevalence of child­
treatment of the underlying crosstalk may be available in the hood and adult obesity in the United States, 2011-2012. JAMA.
future. 2014;311(8):806-814.
Endocrinology, 2023, Vol. 164, No. 7 11

3. World Health Organization. Adults in Europe Are Overweight. 24. Thomas MC. The clustering of cardiovascular, renal, adipo-
2010. https://www.euro.who.int/en/about-us/regional-director/ metabolic eye and liver disease with type 2 diabetes. Metab Clin
news/news/2010/12/2570-of-adults-in-europe-are-overweight Exp. 2022;128:154961.
4. Longo M, Zatterale F, Naderi J, et al. Adipose tissue dysfunction 25. Massier L, Jalkanen J, Elmastas M, et al. An integrated single cell
as determinant of obesity-associated metabolic complications. Int and spatial transcriptomic map of human white adipose tissue.
J Mol Sci. 2019;20(9):2358. Nat Commun. 2023;14(1):1438.
5. Lombardo M, Feraco A, Bellia C, et al. Influence of nutritional sta­ 26. Vijay J, Gauthier MF, Biswell RL, et al. Single-cell analysis of hu­

Downloaded from https://academic.oup.com/endo/article/164/7/bqad094/7192118 by Chulalongkorn University - Faculty of Medicine user on 27 June 2023
tus and physical exercise on immune response in metabolic syn­ man adipose tissue identifies depot and disease specific cell types.
drome. Nutrients. 2022;14(10):2054. Nat Metab. 2020;2(1):97-109.
6. Tajbakhsh A, Gheibihayat SM, Karami N, et al. The regulation of 27. Tan X, Zhu T, Zhang L, et al. miR-669a-5p promotes adipogenic
efferocytosis signaling pathways and adipose tissue homeostasis differentiation and induces browning in preadipocytes.
in physiological conditions and obesity: current understanding Adipocyte. 2022;11(1):120-132.
and treatment options. Obes Rev. 2022;23(10):e13487. 28. Ribas-Latre A, Eckel-Mahan K. Nutrients and the circadian clock:
7. Koenen M, Hill MA, Cohen P, Sowers JR. Obesity, adipose tissue a partnership controlling adipose tissue function and health.
and vascular dysfunction. Circ Res. 2021;128(7):951-968. Nutrients. 2022;14(10):2084.
8. Zatterale F, Longo M, Naderi J, et al. Chronic adipose tissue in­ 29. Cruciani S, Garroni G, Pala R, et al. Metformin and vitamin D
flammation linking obesity to insulin resistance and type 2 dia­ modulate adipose-derived stem cell differentiation towards the
betes. Front Physiol. 2019;10:1607. beige phenotype. Adipocyte. 2022;11(1):356-365.
9. Chen H, Sun L, Feng L, Yin Y, Zhang W. Role of innate lymphoid 30. Wang H, Liu L, Lin JZ, Aprahamian TR, Farmer SR. Browning of
cells in obesity and insulin resistance. Front Endocrinol white adipose tissue with roscovitine induces a distinct population
(Lausanne). 2022;13:855197. of UCP1+ adipocytes. Cell Metab. 2016;24(6):835-847.
10. Man K, Kallies A, Vasanthakumar A. Resident and migratory adi­ 31. Xue S, Lee D, Berry DC. Thermogenic adipose tissue in energy
pose immune cells control systemic metabolism and thermogen­ regulation and metabolic health. Front Endocrinol (Lausanne).
esis. Cell Mol Immunol. 2022;19(3):421-431. 2023;14:1150059.
11. SantaCruz-Calvo S, Bharath L, Pugh G, et al. Adaptive immune 32. Gunawardana SC. Therapeutic value of brown adipose tissue: cor­
cells shape obesity-associated type 2 diabetes mellitus and less recting metabolic disease through generating healthy fat.
prominent comorbidities. Nat Rev Endocrinol. 2022;18(1): Adipocyte. 2012;1(4):250-255.
33. Becher T, Palanisamy S, Kramer DJ, et al. Brown adipose tissue is
23-42.
associated with cardiometabolic health. Nat Med. 2021;27(1):
12. Hu RD, Zhang W, Li L, et al. Chromatin accessibility analysis
58-65.
identifies the transcription factor ETV5 as a suppressor of adipose
34. Singh R, Barrios A, Dirakvand G, Pervin S. Human brown adipose
tissue macrophage activation in obesity. Cell Death Dis.
tissue and metabolic health: potential for therapeutic avenues.
2021;12(11):1023.
Cells. 2021;10(11):3030.
13. Yu F, Yu N, Peng J, et al. Emodin inhibits lipid accumulation and
35. Osman F, Franklyn JA, Daykin J, et al. Heart rate variability and
inflammation in adipose tissue of high-fat diet-fed mice by indu­
turbulence in hyperthyroidism before, during, and after treatment.
cing M2 polarization of adipose tissue macrophages. FASEB J.
Am J Cardiol. 2004;94(4):465-469.
2021;35(7):e21730.
36. Scheen AJ. Sibutramine on cardiovascular outcome. Diabetes
14. Flores-Cortez YA, Barragán-Bonilla MI, Mendoza-Bello JM,
Care. 2011;34(Suppl 2):S114-S119.
González-Calixto C, Flores-Alfaro E, Espinoza-Rojo M.
37. Blondin DP, Nielsen S, Kuipers EN, et al. Human brown adipo­
Interplay of retinol binding protein 4 with obesity and associated
cyte thermogenesis is driven by β2-AR stimulation. Cell Metab.
chronic alterations (review). Mol Med Rep. 2022;26(1):244. 2020;32(2):287-300.e7.
15. Santos D, Carvalho E. Adipose-related microRNAs as modulators 38. Sponton CH, de Lima-Junior JC, Leiria LO. What puts the heat on
of the cardiovascular system: the role of epicardial adipose tissue. thermogenic fat: metabolism of fuel substrates. Trends Endocrinol
J Physiol. 2022;600(5):1171-1187. Metab. 2022;33(8):587-599.
16. Cinkajzlová A, Mráz M, Haluzík M. Adipose tissue immune cells 39. Mills EL, Harmon C, Jedrychowski MP, et al. Cysteine 253 of
in obesity, type 2 diabetes mellitus and cardiovascular diseases. UCP1 regulates energy expenditure and sex-dependent adipose
J Endocrinol. 2021;252(1):R1-r22. tissue inflammation. Cell Metab. 2022;34(1):140-157.e8.
17. Queen NJ, Bates R, Huang W, Xiao R, Appana B, Cao L. Visceral 40. Taylor LE, Sullivan JC. Sex differences in obesity-induced hyper­
adipose tissue-directed FGF21 gene therapy improves metabolic tension and vascular dysfunction: a protective role for estrogen in
and immune health in BTBR mice. Mol Ther Methods Clin Dev. adipose tissue inflammation? Am J Physiol Regul Integr Comp
2021;20:409-422. Physiol. 2016;311(4):R714-R720.
18. Frithioff-Bøjsøe C, Lund MAV, Lausten-Thomsen U, et al. Leptin, 41. Bjune JI, Strømland PP, Jersin R, Mellgren G, Dankel SN.
adiponectin, and their ratio as markers of insulin resistance and Metabolic and epigenetic regulation by estrogen in adipocytes.
cardiometabolic risk in childhood obesity. Pediatr Diabetes. Front Endocrinol (Lausanne). 2022;13:828780.
2020;21(2):194-202. 42. Lee YS, Kim JW, Osborne O, et al. Increased adipocyte O2 con­
19. Rafey MF, Abdalgwad R, O’Shea PM, et al. Changes in the leptin sumption triggers HIF-1α, causing inflammation and insulin re­
to adiponectin ratio are proportional to weight loss after meal re­ sistance in obesity. Cell. 2014;157(6):1339-1352.
placement in adults with severe obesity. Front Nutr. 2022;9: 43. Choe SS, Shin KC, Ka S, Lee YK, Chun JS, Kim JB. Macrophage
845574. HIF-2α ameliorates adipose tissue inflammation and insulin resist­
20. Chait A, den Hartigh LJ. Adipose tissue distribution, inflamma­ ance in obesity. Diabetes. 2014;63(10):3359-3371.
tion and its metabolic consequences, including diabetes and car­ 44. Czaja-Stolc S, Potrykus M, Stankiewicz M, Kaska Ł,
diovascular disease. Front Cardiovasc Med. 2020;7:22. Małgorzewicz S. Pro-inflammatory profile of adipokines in obes­
21. Boulet N, Briot A, Galitzky J, Bouloumié A. The sexual dimorph­ ity contributes to pathogenesis, nutritional disorders, and cardio­
ism of human adipose depots. Biomedicines. 2022;10(10):2615. vascular risk in chronic kidney disease. Nutrients. 2022;14(7):
22. Schetz M, De Jong A, Deane AM, et al. Obesity in the critically ill: 1457.
a narrative review. Intensive Care Med. 2019;45(6):757-769. 45. Chang L, Villacorta L, Li R, et al. Loss of perivascular adipose tis­
23. Hwang I, Kim JB. Two faces of white adipose tissue with hetero­ sue on peroxisome proliferator-activated receptor-γ deletion in
geneous adipogenic progenitors. Diabetes Metab J. 2019;43(6): smooth muscle cells impairs intravascular thermoregulation and
752-762. enhances atherosclerosis. Circulation. 2012;126(9):1067-1078.
12 Endocrinology, 2023, Vol. 164, No. 7

46. Chen Y, Qin Z, Wang Y, Li X, Zheng Y, Liu Y. Role of inflamma­ obesity-related metabolic syndrome. Am J Physiol Endocrinol
tion in vascular disease-related perivascular adipose tissue dys­ Metab. 2010;299(6):E947-952.
function. Front Endocrinol (Lausanne). 2021;12:710842. 68. Kirwan JP, del Aguila LF. Insulin signalling, exercise and cellular
47. Stanek A, Broż yna-Tkaczyk K, Myś liń ski W. The role of integrity. Biochem Soc Trans. 2003;31(6):1281-1285.
obesity-induced perivascular adipose tissue (PVAT) dysfunction 69. Li G, Barrett EJ, Barrett MO, Cao W, Liu Z. Tumor necrosis
in vascular homeostasis. Nutrients. 2021;13(11):3843. factor-alpha induces insulin resistance in endothelial cells via a
48. Li M, Qian M, Kyler K, Xu J. Adipose tissue-endothelial cell inter­ p38 mitogen-activated protein kinase-dependent pathway.

Downloaded from https://academic.oup.com/endo/article/164/7/bqad094/7192118 by Chulalongkorn University - Faculty of Medicine user on 27 June 2023
actions in obesity-induced endothelial dysfunction. Front Endocrinology. 2007;148(7):3356-3363.
Cardiovasc Med. 2021;8:681581. 70. Nakamura K, Fuster JJ, Walsh K. Adipokines: a link between
49. Ambrozova G, Martiskova H, Koudelka A, et al. Nitro-oleic acid obesity and cardiovascular disease. J Cardiol. 2014;63(4):
modulates classical and regulatory activation of macrophages and 250-259.
their involvement in pro-fibrotic responses. Free Radic Biol Med. 71. Obradovic M, Sudar-Milovanovic E, Soskic S, et al. Leptin and
2016;90:252-260. obesity: role and clinical implication. Front Endocrinol
50. Chan CT, Sobey CG, Lieu M, et al. Obligatory role for B cells in (Lausanne). 2021;12:585887.
the development of angiotensin II-dependent hypertension. 72. Li G, Feng D, Qu X, et al. Role of adipokines FGF21, leptin and
Hypertension. 2015;66(5):1023-1033. adiponectin in self-concept of youths with obesity. Eur
51. Withers SB, Forman R, Meza-Perez S, et al. Eosinophils are key Neuropsychopharmacol. 2018;28(8):892-902.
regulators of perivascular adipose tissue and vascular functional­ 73. Monteiro L, Pereira J, Palhinha L, Moraes-Vieira PMM. Leptin in
ity. Sci Rep. 2017;7(1):44571. the regulation of the immunometabolism of adipose tissue-
52. Saxton SN, Heagerty AM, Withers SB. Perivascular adipose tissue: macrophages. J Leukoc Biol. 2019;106(3):703-716.
an immune cell metropolis. Exp Physiol. 2020;105(9):1440-1443. 74. Zuo Y, Xiao T, Qiu X, Liu Z, Zhang S, Zhou N. Adiponectin
53. Saxton SN, Whitley AS, Potter RJ, Withers SB, Grencis R, reduces apoptosis of diabetic cardiomyocytes by regulating
Heagerty AM. Interleukin-33 rescues perivascular adipose tissue miR-711/TLR4 axis. Diabetol Metab Syndr. 2022;14(1):131.
anticontractile function in obesity. Am J Physiol Heart Circ 75. Yamauchi T, Kamon J, Ito Y, et al. Cloning of adiponectin recep­
Physiol. 2020;319(6):H1387-h1397. tors that mediate antidiabetic metabolic effects. Nature.
54. Henning RJ. Obesity and obesity-induced inflammatory disease 2003;423(6941):762-769.
contribute to atherosclerosis: a review of the pathophysiology 76. Xu H, Zhao Q, Song N, et al. Adipor1/AdipoR2 dual agonist re­
and treatment of obesity. Am J Cardiovasc Dis. 2021;11(4): covers nonalcoholic steatohepatitis and related fibrosis via endo­
504-529. plasmic reticulum-mitochondria axis. Nat Commun.
55. Belden Z, Deiuliis JA, Dobre M, Rajagopalan S. The role of the 2020;11(1):5807.
mineralocorticoid receptor in inflammation: focus on kidney 77. Matsuda M, Shimomura I, Sata M, et al. Role of adiponectin in
and vasculature. Am J Nephrol. 2017;46(4):298-314. preventing vascular stenosis. The missing link of adipo-vascular
56. Wu C, Zhang H, Zhang J, et al. Inflammation and fibrosis in peri­ axis. J Biol Chem. 2002;277(40):37487-37491.
renal adipose tissue of patients with aldosterone-producing aden­ 78. Pascolutti R, Erlandson SC, Burri DJ, Zheng S, Kruse AC.
oma. Endocrinology. 2018;159(1):227-237. Mapping and engineering the interaction between adiponectin
57. Jia G, Aroor AR, Sowers JR. The role of mineralocorticoid recep­ and T-cadherin. J Biol Chem. 2020;295(9):2749-2759.
tor signaling in the cross-talk between adipose tissue and the 79. Fujishima Y, Maeda N, Matsuda K, et al. Adiponectin association
vascular wall. Cardiovasc Res. 2017;113(9):1055-1063. with T-cadherin protects against neointima proliferation and ath­
58. Moss ME, Lu Q, Iyer SL, et al. Endothelial mineralocorticoid re­ erosclerosis. FASEB J. 2017;31(4):1571-1583.
ceptors contribute to vascular inflammation in atherosclerosis in a 80. Eid RA, Alkhateeb MA, El-Kott AF, et al. A high-fat diet rich in
sex-specific manner. Arterioscler Thromb Vasc Biol. 2019;39(8): corn oil induces cardiac fibrosis in rats by activating JAK2/
1588-1601. STAT3 and subsequent activation of ANG II/TGF-1β/smad3
59. Mallick R, Duttaroy AK. Modulation of endothelium function by pathway: the role of ROS and IL-6 trans-signaling. J Food
fatty acids. Mol Cell Biochem. 2022;477(1):15-38. Biochem. 2019;43(8):e12952.
60. Kwaifa IK, Bahari H, Yong YK, Noor SM. Endothelial dysfunc­ 81. Recinella L, Orlando G, Ferrante C, Chiavaroli A, Brunetti L,
tion in obesity-induced inflammation: molecular mechanisms Leone S. Adipokines: new potential therapeutic target for obesity
and clinical implications. Biomolecules. 2020;10(2):291. and metabolic, rheumatic, and cardiovascular diseases. Front
61. Astha S, Archna S, Sakshi S, Sandeep A, Ishfaq M, Rakhee Y. Physiol. 2020;11:578966.
Study of serum adipocytokines and their association with insulin 82. Frühbeck G, Catalán V, Rodríguez A, Gómez-Ambrosi J.
sensitivity in morbidly obese individuals undergoing bariatric sur­ Adiponectin-leptin ratio: a promising index to estimate adipose
gery. J Obes Metab Syndr. 2020;29(4):303-312. tissue dysfunction. Relation with obesity-associated cardiometa­
62. Bianconi V, Sahebkar A, Atkin SL, Pirro M. The regulation and bolic risk. Adipocyte. 2018;7(1):57-62.
importance of monocyte chemoattractant protein-1. Curr Opin 83. Freeman AM, Pennings N. Insulin Resistance. StatPearls.
Hematol. 2018;25(1):44-51. StatPearls Publishing; 2022.
63. Syarif, Rasyid H, Aman M, Lawrence GS. High-fat diet increases 84. Ahmed B, Sultana R, Greene MW. Adipose tissue and insulin re­
the level of circulating monocyte chemoattractant protein-1 in sistance in obese. Biomed Pharmacother. 2021;137:111315.
Wistar rats, independent of obesity. Ann Med Surg (Lond). 85. Seo JB, Riopel M, Cabrales P, et al. Knockdown of Ant2 reduces
2021;65:102266. adipocyte hypoxia and improves insulin resistance in obesity. Nat
64. Lee J, Lee S, Zhang H, Hill MA, Zhang C, Park Y. Interaction of Metab. 2019;1(1):86-97.
IL-6 and TNF-α contributes to endothelial dysfunction in type 2 86. Farkhondeh T, Llorens S, Pourbagher-Shahri AM, et al. An over­
diabetic mouse hearts. PLoS One. 2017;12(11):e0187189. view of the role of adipokines in cardiometabolic diseases.
65. Huang T, Song J, Gao J, et al. Adipocyte-derived kynurenine pro­ Molecules. 2020;25(21):5218.
motes obesity and insulin resistance by activating the AhR/ 87. Chaurasia B, Summers SA. Ceramides in metabolism: key lipo­
STAT3/IL-6 signaling. Nat Commun. 2022;13(1):3489. toxic players. Annu Rev Physiol. 2021;83(1):303-330.
66. Al-Mansoori L, Al-Jaber H, Prince MS, Elrayess MA. Role of in­ 88. Nishi H, Higashihara T, Inagi R. Lipotoxicity in kidney, heart,
flammatory cytokines, growth factors and adipokines in adipo­ and skeletal muscle dysfunction. Nutrients. 2019;11(7):1664.
genesis and insulin resistance. Inflammation. 2022;45(1):31-44. 89. Michailidou Z, Gomez-Salazar M, Alexaki VI. Innate immune
67. Schinzari F, Tesauro M, Rovella V, et al. Generalized impairment cells in the adipose tissue in health and metabolic disease.
of vasodilator reactivity during hyperinsulinemia in patients with J Innate Immun. 2022;14(1):4-30.
Endocrinology, 2023, Vol. 164, No. 7 13

90. Liu L, Hu J, Wang Y, Lei H, Xu D. The role and research progress 107. Marcos-Pasero H, Aguilar-Aguilar E, de la Iglesia R, et al.
of the balance and interaction between regulatory T cells and oth­ “GENYAL” study to childhood obesity prevention: methodology
er immune cells in obesity with insulin resistance. Adipocyte. and preliminary results. Front Nutr. 2022;9:777384.
2021;10(1):66-79. 108. Hu H, Garcia-Barrio M, Jiang ZS, Chen YE, Chang L. Roles of
91. Li C, Qin D, Hu J, Yang Y, Hu D, Yu B. Inflamed adipose tissue: a perivascular adipose tissue in hypertension and atherosclerosis.
culprit underlying obesity and heart failure with preserved ejection Antioxid Redox Signal. 2021;34(9):736-749.
fraction. Front Immunol. 2022;13:947147. 109. Barhoumi T, Kasal DA, Li MW, et al. T regulatory lymphocytes

Downloaded from https://academic.oup.com/endo/article/164/7/bqad094/7192118 by Chulalongkorn University - Faculty of Medicine user on 27 June 2023
92. He J, Yang W, Wu W, et al. Clinical features, myocardial strain prevent angiotensin II-induced hypertension and vascular injury.
and tissue characteristics of heart failure with preserved ejection Hypertension. 2011;57(3):469-476.
fraction in patients with obesity: a prospective cohort study. 110. Jia J, Yang JQ, Du YR, et al. Transcriptomic profiling reveals
underlying immunoregulation mechanisms of resistant hyperten­
EClinicalMedicine. 2023;55:101723.
sion in injection drug users. J Inflamm Res. 2022;15:3409-3420.
93. da Silva AA, do Carmo JM, Li X, Wang Z, Mouton AJ, Hall JE.
111. Van Beusecum JP, Moreno H, Harrison DG. Innate immunity and
Role of hyperinsulinemia and insulin resistance in hypertension:
clinical hypertension. J Hum Hypertens. 2022;36(6):503-509.
metabolic syndrome revisited. Can J Cardiol. 2020;36(5):
112. Feijóo-Bandín S, Aragón-Herrera A, Moraña-Fernández S, et al.
671-682.
Adipokines and inflammation: focus on cardiovascular diseases.
94. Mennitti C, Ranieri A, Nigro E, et al. The impact of physical ex­
Int J Mol Sci. 2020;21(20):7711.
ercise on obesity in a cohort of southern Italian obese children: im­ 113. Lagier JC, Armougom F, Million M, et al. Microbial culturomics:
provement in cardiovascular risk and immune system biomarkers. paradigm shift in the human gut microbiome study. Clin
Int J Environ Res Public Health. 2022;20(1):602. Microbiol Infect. 2012;18(12):1185-1193.
95. Karason K, Girerd N, Andersson-Asssarsson J, et al. Heart failure 114. Oliveira RA, Pamer EG. Assembling symbiotic bacterial species
in obesity: insights from proteomics in patients treated with or into live therapeutic consortia that reconstitute microbiome func­
without weight-loss surgery. Int J Obes (Lond). 2022;46(12): tions. Cell Host Microbe. 2023;31(4):472-484.
2088-2094. 115. Schinzari F, Tesauro M, Cardillo C. Vasodilator dysfunction in
96. Trouwborst I, Gijbels A, Jardon KM, et al. Cardiometabolic human obesity: established and emerging mechanisms.
health improvements upon dietary intervention are driven by J Cardiovasc Pharmacol. 2021;78(Suppl 6):S40-S52.
tissue-specific insulin resistance phenotype: a precision nutrition 116. Zhernakova A, Kurilshikov A, Bonder MJ, et al. Population-based
trial. Cell Metab. 2023;35(1):71-83.e5. metagenomics analysis reveals markers for gut microbiome com­
97. Janssen J. Hyperinsulinemia and its pivotal role in aging, obesity, position and diversity. Science. 2016;352(6285):565-569.
type 2 diabetes, cardiovascular disease and cancer. Int J Mol Sci. 117. Battson ML, Lee DM, Weir TL, Gentile CL. The gut microbiota as
2021;22(15):7797. a novel regulator of cardiovascular function and disease. J Nutr
98. Yaghootkar H, Scott RA, White CC, et al. Genetic evidence for a Biochem. 2018;56:1-15.
normal-weight “metabolically obese” phenotype linking insulin 118. Rovella V, Rodia G, Di Daniele F, et al. Association of gut hor­
resistance, hypertension, coronary artery disease, and type 2 dia­ mones and microbiota with vascular dysfunction in obesity.
betes. Diabetes. 2014;63(12):4369-4377. Nutrients. 2021;13(2):613.
99. O’Neill BJ. Effect of low-carbohydrate diets on cardiometabolic 119. Chen L, Ishigami T, Doi H, Arakawa K, Tamura K. Gut micro­
risk, insulin resistance, and metabolic syndrome. Curr Opin biota and atherosclerosis: role of B cell for atherosclerosis focusing
on the gut-immune-B2 cell axis. J Mol Med (Berl). 2020;98(9):
Endocrinol Diabetes Obes. 2020;27(5):301-307.
1235-1244.
100. Hill MA, Yang Y, Zhang L, et al. Insulin resistance, cardiovascu­
120. Chen L, Ishigami T, Nakashima-Sasaki R, et al. Commensal
lar stiffening and cardiovascular disease. Metab Clin Exp.
microbe-specific activation of B2 cell subsets contributes to ath­
2021;119:154766.
erosclerosis development independently of lipid metabolism.
101. Hill MA, Jaisser F, Sowers JR. Role of the vascular endothelial so­
EBioMedicine. 2016;13:237-247.
dium channel activation in the genesis of pathologically increased
121. Kim M, Huda MN, Bennett BJ. Sequence meets function-
cardiovascular stiffness. Cardiovasc Res. 2022;118(1):130-140. microbiota and cardiovascular disease. Cardiovasc Res.
102. Weihrauch-Blüher S, Schwarz P, Klusmann JH. Childhood obes­ 2022;118(2):399-412.
ity: increased risk for cardiometabolic disease and cancer in adult­ 122. Le Roy T, de Hase EM, Van Hul M, et al. Dysosmobacter welbio­
hood. Metab Clin Exp. 2019;92:147-152. nis is a newly isolated human commensal bacterium preventing
103. Wühl E. Hypertension in childhood obesity. Acta Paediatr. diet-induced obesity and metabolic disorders in mice. Gut.
2019;108(1):37-43. 2022;71(3):534-543.
104. Drozdz D, Alvarez-Pitti J, Wójcik M, et al. Obesity and cardiome­ 123. Wu TR, Lin CS, Chang CJ, et al. Gut commensal Parabacteroides
tabolic risk factors: from childhood to adulthood. Nutrients. goldsteinii plays a predominant role in the anti-obesity effects of
2021;13(11):4176. polysaccharides isolated from Hirsutella sinensis. Gut.
105. DeBoer MD. Assessing and managing the metabolic syndrome in 2019;68(2):248-262.
children and adolescents. Nutrients. 2019;11(8):1788. 124. Mazier W, Le Corf K, Martinez C, et al. A new strain of
106. Freemark M. Endothelial dysfunction and cardiovascular disease Christensenella minuta as a potential biotherapy for obesity and
in childhood obesity. J Pediatr (Rio J). 2019;95(5):503-505. associated metabolic diseases. Cells. 2021;10(4):823.

You might also like