Adipose tissue immune cells in obesity, type 2 diabetes mellitus and cardiovascular diseases

in Journal of Endocrinology
Authors:
Anna Cinkajzlová Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic

Search for other papers by Anna Cinkajzlová in
Current site
Google Scholar
PubMed
Close
,
Miloš Mráz Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic

Search for other papers by Miloš Mráz in
Current site
Google Scholar
PubMed
Close
, and
Martin Haluzík Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic

Search for other papers by Martin Haluzík in
Current site
Google Scholar
PubMed
Close

Correspondence should be addressed to M Haluzik: mhalu@lf1.cuni.cz
Free access

Sign up for journal news

Immune cells are an inseparable component of adipose tissue intimately involved in most of its functions. Physiologically, they regulate adipose tissue homeostasis, while in case of adipose tissue stress, immune cells are able to change their phenotype, enhance their count and subsequently contribute to the development and maintenance of local adipose tissue inflammation. Immune cells are an important source of inflammatory cytokines and other pro-inflammatory products that further influence not only surrounding tissues but via systemic circulation also the whole organism being thus one of the main factors responsible for the transition from simple obesity to associated metabolic and cardiovascular complications. The purpose of this review is to summarize current knowledge on different adipose tissue immune cell subsets and their role in the development of obesity, type 2 diabetes mellitus and cardiovascular diseases.

Abstract

Immune cells are an inseparable component of adipose tissue intimately involved in most of its functions. Physiologically, they regulate adipose tissue homeostasis, while in case of adipose tissue stress, immune cells are able to change their phenotype, enhance their count and subsequently contribute to the development and maintenance of local adipose tissue inflammation. Immune cells are an important source of inflammatory cytokines and other pro-inflammatory products that further influence not only surrounding tissues but via systemic circulation also the whole organism being thus one of the main factors responsible for the transition from simple obesity to associated metabolic and cardiovascular complications. The purpose of this review is to summarize current knowledge on different adipose tissue immune cell subsets and their role in the development of obesity, type 2 diabetes mellitus and cardiovascular diseases.

Introduction

Adipose tissue (AT) is an active organ dynamically reacting to the metabolic status of the organism. The whole-body AT can be divided into two major – the subcutaneous (SAT) and the visceral (VAT) – and several minor AT depots (epicardial, perivascular, perirenal, intraosseous, intraarticular, etc.). VAT, which is present mainly in the mesentery and omentum, contains a lower amount of preadipocytes and a higher count of metabolically more active and insulin-resistant adipocytes along with a richer blood supply and innervation than SAT. It also exerts a higher capacity for glucose and lipid uptake and production of free fatty acids, has more receptors for glucocorticoids and androgens and contains more immune cells (Ibrahim 2010). Excessive accumulation of VAT phenotypically presented as android obesity is associated with higher metabolic risk and the development of insulin resistance, arterial hypertension, type 2 diabetes mellitus (T2DM) and cardiovascular diseases (Samsell et al. 2014). One of the main mechanisms responsible for the transition from simple obesity to subsequent cardiometabolic complications is the development of, at first, local and then systemic low-grade inflammation characterized by accumulation of inflammatory immune cells and increased production of pro-inflammatory factors in obese AT, with VAT being again significantly more prone to these effects than SAT (Castro et al. 2016). Furthermore, AT immune cell changes are potentially associated with higher risk of gestational diabetes mellitus and preeclampsia in pregnant women further confirming the role of AT inflammation and its immune cells in obesity-associated morbidity (Huda et al. 2017, Cinkajzlova et al. 2020).

Interestingly, while obesity is primarily associated with phenotypic and functional changes of AT immune cells, recent results from single-cell RNA sequencing techniques showed the existence of several adipocyte progenitor cell subtypes specialized in different functions including thermogenesis, lipid storage and adipokine secretion as well as more subtypes of adipocytes. These adipocyte subtypes, differing in key adipocyte functions such as leptin and adiponectin expression, along with their precursors seem to collectively mediate the multiple diverse functions of AT and contribute to its physiologic and pathophysiologic milieu (Min et al. 2019).

The role of immune cells in adipose tissue physiology

Immune cells in AT serve several functions. Physiologically, they contribute to tissue homeostasis by being involved in tissue repair and apoptosis of damaged or infected cells as they react to damage-associated molecular patterns (DAMPs) such as nuclear or cytosolic proteins and to pathogen-associated molecular patterns (PAMPs) such as bacterial lipopolysaccharide or peptidoglycans. Effective sensing of DAMPs and PAMPs through pathogen recognition receptors rapidly induces immune responses via the activation of complex signaling pathways (Kumar et al. 2011, Roh & Sohn 2018). This process is accompanied by cell metabolic changes and production of different cytokines and other pro-inflammatory factors such as free radicals, nitric oxide and others (Pearce & Pearce 2013).

Immune cell subsets from both main lines – myeloid and lymphoid – can be found in AT, albeit with different frequency and significance for the development of AT inflammation with the most important being macrophages and T lymphocytes and their different phenotype subtypes.

Myeloid cells in adipose tissue

Myeloid cells consist of cells of monocyte–macrophage line, granulocytes, which are further divided into eosinophils, basophils and neutrophils, and some subsets of dendritic cells (DCs), which are crucial for innate immunity. Neutrophils are the most abundant leukocyte subset in mammals and are responsible for pro-inflammatory and antiviral responses. Monocytes are large mononuclear leukocytes involved in the inflammation and clearance of pathogens. They are able to differentiate into macrophages and DCs (Stegelmeier et al. 2019). According to CD14 and CD16 surface expression, monocytes are divided into three subsets – classical (85% of monocytes), non-classical (10% of monocytes) and intermediate monocytes. Classical monocytes are professional phagocytes producing anti- and pro-inflammatory cytokines and reactive oxygen species and engulfing native LDL particles, while non-classical monocytes are weaker phagocytes, engulf oxidized LDL particles and produce pro-inflammatory cytokines (Belge et al. 2002, Ziegler-Heitbrock 2007, Mosig et al. 2009).

Macrophages

The most frequent immune cells in AT derived from the myeloid precursor are macrophages. In general, macrophages are phagocytic cells defending the organism against foreign pathogens and their substances such as lipopolysaccharide, as well as cancer, damaged endogenous cells and cellular debris. They exist either in tissue-resident form (e.g. Kupffer cells in the liver) or can be recruited and developed from blood monocytes in response to cytokine production (Watanabe et al. 2019a). In AT, another source of macrophages might be preadipocytes, as suggested by in vitro studies (Cousin et al. 1999, Charriere et al. 2003). These preadipocyte-derived macrophages were shown to express antigens typical for both monocytes and macrophages and to exert anti-microbial and phagocytic activity (Cousin et al. 2001). Besides this, macrophages are also able to replicate and thus be their own source (Hashimoto et al. 2013, Yona et al. 2013) without any phenotypic changes (Sieweke & Allen 2013). As demonstrated in in vitro studies (Athie-Morales et al. 2004, Mantovani et al. 2004, Iwakura & Ishigame 2006), macrophages can exert two extremely polarized states termed M1 and M2. M2 macrophages represent the anti-inflammatory state characterized by the production of the interleukins (ILs) IL4, IL13 and IL10, while M1 macrophages are deemed pro-inflammatory and produce TNFα, IL12 and IL23, regulating their own and T helper lymphocytes (Table 1). According to the initial hypothesis derived especially from rodent models (Gordon 2003, Gordon & Taylor 2005), the anti-inflammatory M2 macrophages are present predominantly in lean subjects, where they help in keeping AT homeostasis and maintaining physiological insulin sensitivity (Odegaard et al. 2007). Long-term increase in body weight is associated with a transition to the pro-inflammatory M1 phenotype along with enhanced monocyte recruitment into AT. M1 macrophages also represent an important source of reactive oxygen species and nitric oxide (Mantovani et al. 2004), which may contribute to the development of AT fibrosis via mitochondrial dysfunction of preadipocytes and activation of matrix metalloproteinase 9. Macrophages actively communicate with other immune cell types, especially with Th lymphocytes. As demonstrated before, M1 macrophages regulate mostly Th1 and Th17 cell response, while M2 macrophages regulate Th2 lymphocytes (Mantovani et al. 2004). Apart from communication through cytokine production, macrophages and T lymphocytes are able to communicate through direct cell contact by CD154 (CD40 ligand) and CD40 antigens (Elgueta et al. 2009).

Table 1

Signature cytokines of adipose tissue immune cells and their role in obesity, type 2 diabetes mellitus and cardiovascular diseases.

Cytokines

Source

General functions

Association with obesity and T2DM

Association with cardiovascular diseases

TNFα Adipocytes, macrophages (M1/MMe), Th1 lymphocytes, NK cells Induction of systemic inflammation and acute phase response TNFα: correlates with the degree of adiposity and HbA1c, associated with insulin resistance and T2DM, reduces the expression of insulin-regulated glucose transporter type 4 located in adipocytes and skeletal and cardiac muscles TNFα: involved in atherosclerosis progression, stimulates interaction between leukocytes and the endothelium through upregulation of adhesion molecules, stimulates myocyte hypertrophy through generation of reactive oxygen intermediates in cardiac myocytes, induces ventricular remodeling
IL1β IL1β: mediator of metabolic inflammation, represses insulin signaling, contributes to β-cell failure IL1β: increases nitric oxide synthesis leading to lower energy production and reduced myocardial contractility, stimulates post-infarction inflammatory response, involved in cardiac remodeling
IL6 IL6: supports both M2 and Th2 polarization or conversely M1 response in the presence of IFNγ, mediates macrophage–adipocyte crosstalk, increases lipolysis, suppresses adiponectin secretion, increases glycemia by suppressing glucose uptake by adipocytes, decreases hepatic insulin sensitivity and glycogen synthesis IL6: progression of atherosclerosis by induction of endothelial dysfunction and lipoprotein oxidation, independent risk factor for coronary artery disease
IFNγ Th1 cells, Tc cells, NK cells, NKT cells, M1 macrophages Antiviral and antimicrobial immunity Upregulates production of macrophage mediators, induces production of leukocyte adhesion molecules and chemokines, increases antigen-presenting capacity by macrophages and endothelial cells, disrupts cellular metabolism and energy expenditure, increases insulin resistance and negatively influences glucose tolerance Increases accumulation of myeloid cells in infarcted cardiac tissue, activates cardiac macrophages in ischemic myocardium, promotes atherosclerosis and increases plaque vulnerability
IL17 Th17 cells, macrophages, dendritic cells, NK cells, NKT cells, γδ-T cells, neutrophils Host defense against microbes, particularly extracellular bacteria and fungi Inhibits adipogenesis, promotes IL6 production in differentiated adipocytes, increases in obesity and T2DM, impairs glucose uptake and insulin sensitivity, induces free radical production in diabetes Contributes to atherosclerosis and plaque vulnerability, extracellular matrix remodeling through upregulation of angiotensin 2 type 1 receptor, increases in acute coronary syndrome
IL10 M2 macrophages, Th2 cells, Treg cells Suppression of pro-inflammatory reactions Downregulates TNFα production, inhibits expression of adhesion molecules and antigen presentation, controls tissue remodeling, improves insulin sensitivity and glucose transport Downregulates adhesion molecule expression on endothelial cells, protective during myocardium ischemia or reperfusion, low levels lead to development of extensive and unstable atherosclerotic lesions

Associated with higher cardiovascular risk in subjects with chronic kidney disease
IL5 M2 macrophages, Th2 cells, Tregs, mast cells, basophils Homeostatic, anti-parasitic and allergy reactions Associated with alternative macrophage activation and beige fat development IL5: atheroprotective role,facilitates recovery of cardiac function after myocardial infarction
IL4 IL4: improves insulin sensitivity and glucose tolerance by inhibition of adipogenesis and activation of lipolysis IL4: induces pro-inflammatory environments in vascular endothelium by overexpression of inflammatory mediators, increases vascular cell adhesion molecule 1 expression in vascular endothelium, promotes fibrotic tissue formation after myocardial infarction
IL13 IL13: improves insulin secretion, involved in glucose uptake and metabolism in skeletal muscle IL13: inhibits progression of atherosclerosis, facilitates cardiac regeneration after myocardial infarction

IFN, interferon; IL, interleukin; MMe macrophages, metabolically activated macrophages; NK cells, natural killer cells; NKT cells, natural killer T cells; Tc cells, T cytotoxic cells; Th cells, T helper cells; TNF, tumor necrosis factor; Tregs, T regulatory cells; T2DM, type 2 diabetes mellitus.

Nevertheless, currently, it seems that in vivo macrophages in AT show a rather mixed phenotype and functional heterogeneity termed as metabolically activated phenotype (MMe) (Zeyda et al. 2007, Li et al. 2010, Kratz et al. 2014, Martinez & Gordon 2014). According to murine results (Coats et al. 2017), MMe macrophages under the control of toll-like receptor 2 (TLR2), NADPH-oxidase-2 and MYD88 overexpress pro-inflammatory cytokines TNFα, IL1β and IL6 promoting insulin resistance similarly to M1 macrophages, while also being capable to exocytose their lysosomes and clear dead adipocytes, thus limiting insulin resistance similarly to M2 macrophages. Furthermore, metabolic activation of macrophages is associated with the same surface markers as classical activation of M1 macrophages. MMe macrophages specifically overexpress ATP binding cassette subfamily A member 1, CD36 and Perilipin 2 regulated by p62 and peroxisome proliferator-activated receptor-gamma (Kratz et al. 2014). The complex role of macrophages and their derivates in AT inflammation is underscored by recent findings of foam cells – lipid-laden macrophages best known from atherosclerotic lesions – within AT, where they engulf the remains of dead adipocytes (Shapiro et al. 2013) or results from single-cell RNA sequencing describing a population of 'lipid-associated macrophages' in both murine and human VAT. In a mouse model, the proportion of these cells correlated with BMI, and the knockout of their signature gene (Trem2) lead to high-fat diet-induced obesity, insulin resistance and dyslipidemia (Jaitin et al. 2019). These cells similar to M1 macrophages contribute to inflammation by increased IL1β, TNF, IL18, CXCL18 and -platelet-derived growth factor-beta production (Hildreth et al. 2021). To sum up, this macrophage plasticity with the absence of a fixed and static M1/M2 dichotomy might play an important role in the resolution of the actual needs of AT.

Granulocytes

Granulocytes (neutrophils, eosinophils and basophils) play an essential role during microbe-induced and sterile inflammation as they are the terminally differentiated short-lived phagocytes (Geering et al. 2013). Besides that, granulocytes can acquire the function of antigen-presenting cells for T lymphocytes under inflammatory and other pathological conditions (Lin & Lore 2017). While eosinophils and basophils are involved in anti-inflammatory reactions in lean AT, neutrophils act rather in obese AT, where they conversely support pro-inflammatory processes. Eosinophils are generally associated with Th2 responses involved in antiparasitic and allergic reactions with their primary cytokine being IL4 (Jacobsen et al. 2012). Neutrophils are the dominant population among granulocytes and represent one of the first responders during acute inflammation (Geering et al. 2013). In chronic inflammation, they are able to influence adaptive T cell immunity through their effect on DC priming and directly on T (CD4+) cells themselves (Minns et al. 2019). In lean subjects, the actions of neutrophils are suppressed and negatively regulated by adiponectin. Adiponectin inhibits superoxide generation through the regulation of NADPH oxidase (Magalang et al. 2006) as well as neutrophil apoptosis (Rossi & Lord 2013b) and phagocytosis (Rossi & Lord 2013a). Both basophils and mast cells synthetise heparin and histamine and express plasma membrane receptors that bind IgE with high affinity. They both play a crucial role in allergy and asthma (He et al. 2013). Basophils emerge from the bone marrow as mature cells, whereas mast cells are released in an immature form and are fully activated after transmigration into target tissues. Basophils mainly produce Th2 cytokines (IL4 and IL13), while mast cells produce both Th1 and Th2 cytokines (Marone et al. 1997, Geering et al. 2013).

Lymphoid cells in adipose tissue

Cells of the lymphoid line represent a heterogenous cell population involved in both innate and acquired immunity. They develop in bone marrow, thymus and spleen (Caspar-Bauguil et al. 2005). Blood lymphocytes are probably the only source of AT lymphocytes, which are recruited in response to cytokine production of stressed adipocytes and already present immune cells. In AT, blood lymphocytes undergo further maturation in response to tissue needs and actual cytokine milieu (Rocha et al. 2008). Thus, the whole spectrum of lymphocyte subtypes including T helper (Th) and cytotoxic cells, B cells, natural killer (NK), natural killer T (NKT) and γδ-T cells can be found in AT.

T helper lymphocytes

The group of Th lymphocytes consists of several specific T cell subpopulations including Th1, Th2, Th17 and Treg lymphocytes, which reciprocally develop under particular tissue cytokine production and all of which can be found within AT (Mills et al. 2000). According to a simplified concept, Th1 and Th17 lymphocytes represent the pro-inflammatory phenotypes prevalent in obese AT, while Th2 and Treg lymphocytes are considered anti-inflammatory and participate in AT homeostasis in lean subjects (Winer et al. 2009).

Th1 and Th17 cells are stimulated by IFNγ, IL12 and IL6 and are associated with M1 macrophage reactions, while Th2 and Treg cells are stimulated by IL2 and IL4 and are connected with M2 macrophages (Odegaard et al. 2007, Tiemessen et al. 2007). Th1 lymphocytes are a significant source of INFγ and IL2 thus stimulating their own production (Martinez & Gordon 2014). As seen in rodent models (Surendar et al. 2019), adiponectin reduces IFNγ- and IL17-positive T cells and dampens the differentiation of naïve T cells into Th1 and Th17 populations. Th2 lymphocytes produce anti-inflammatory cytokines such as IL4, IL5, IL10 and IL13. They also stimulate IgE production by B lymphocytes as well as eosinophil activation and M2 macrophage responses (Romagnani 1999, Berger 2000). Treg lymphocytes, similarly to Th2 lymphocytes, suppress Th1 and Th17 cell responses by inhibition of IFNγ (Sojka & Fowell 2011, Joller et al. 2014) and by stimulation of IL10 (Chaudhry et al. 2011). Both Th2 and Treg subsets are commonly found in lean AT, but their numbers decrease along with the increase of Th1 and Th1 subsets during the development of obesity and insulin resistance (Feuerer et al. 2009).

T cytotoxic lymphocytes

T cytotoxic (Tc) lymphocytes are cells involved in acquired immunity with the ability to induce apoptosis of infected, damaged or tumor cells by perforin (Prf) and granzyme production. Tc lymphocytes express CD8 antigen and are activated by antigens presented by major histocompatibility complex class I (MHC-I). Their activation is enhanced by cytokine production (notably IL12 and IL18) and can also be triggered by T cell receptor-independent mechanisms (Henry et al. 2008, Freeman et al. 2012). Similarly to Th1 lymphocytes, Tc lymphocytes are increased in obese AT, where they support macrophage accumulation (Jiang et al. 2014), whereas their depletion is associated with enhanced beige adipogenesis as shown in murine experiments (Moysidou et al. 2018). The Tc lymphocyte population also consists of more subpopulations with Tc2 subpopulation producing the anti-inflammatory IL4 and IL5 cytokines, while Tc1 cells being characterized by IFNγ production (Dobrzanski et al. 2004).

B lymphocytes

B lymphocytes are cells of acquired immunity and are the only cell population capable of producing specific antibodies. Besides that, B lymphocytes are antigen-presenting cells for Th lymphocytes (Wortis et al. 1995) and a source of different cytokines (Nishimura et al. 2013). All these three mechanisms were suggested to take part in the regulation of AT inflammation and insulin resistance (Winer et al. 2011). The B cell population consists of B-1 and B-2 cell subpopulations with reciprocal functions. B-1 cells, predominantly found in the peritoneal and pleural cavities, represent long-lived self-renewing cells, which, as a part of innate-like immunity, produce antibodies toward bacterial and self-antigens promoting a rapid response to infection and clearing of apoptotic cells. B-2 cells, which represent the majority of all B lymphocytes, are short-lived circulating cells producing specific antibodies and collaborating with Th lymphocytes (Wong et al. 2019, Mahajan et al. 2020). As seen in murine models, the B-1 to B-2 cell ratio varies between AT departments – being highest in omental VAT (≈0.8:1) and lowest in SAT, also in comparison with perivascular AT (PVAT; ≈0.3:1), epidydimal VAT (≈0.3:1) and brown AT (≈0.2:1) (Srikakulapu & McNamara 2020). Nevertheless, B cell accumulation was shown to promote inflammation, while their global depletion attenuated high-fat diet-induced AT inflammation and insulin resistance (Winer et al. 2011).

Natural killer cells

NK cells belong to the group of innate immune cells and induce apoptosis of cells infected by viruses or transformed by cancer (Smyth et al. 2005). In AT, NK cells are able to communicate with adipocytes by direct cell contact through the NK cell activating receptor 1 (NCR1) (Wensveen et al. 2015). NK cells also induce the differentiation of naïve Th cells into Th1 cells (as they represent an early source of IFNγ) (Martin-Fontecha et al. 2004) and provide cross-presentation of antigens to Tc lymphocytes through DCs (Deauvieau et al. 2015). Single-cell RNA sequencing revealed that healthy SAT contains a unique NK cell subpopulation producing IFNγ (Hildreth et al. 2021). As shown experimentally (O’Sullivan et al. 2016), these cells and their cytokine production could play an important role in the regulation of early phases of obesity development as they are active at the beginning of high-fat feeding directly in AT, where they likely produce the most IFNγ in situ on a per cell basis of all AT cells and are necessary (and sufficient) to drive the pro-inflammatory macrophage polarization.

Natural killer T cells

NKT cells are lymphocytes positioned in between innate and acquired immunity that are capable of pro-inflammatory as well as anti-inflammatory cytokine production. They include three subpopulations: invariant NKT cells (iNKT, type I), diverse NKT cells (dNKT, type II) and NKT-like cells (Park et al. 2018). iNKT cells standardly reside in AT and can be activated by both professional and non-professional antigen-presenting cells (Huh et al. 2017). Of the three NKT lymphocyte subtypes, iNKT cells seem to have the greatest impact on the resolution of AT inflammation due to their ability to recognize lipid molecules loaded onto CD1d receptor of antigen-presenting cells by their semi-invariant T-cell receptor triggering anti-inflammatory cytokine secretion (Huh et al. 2017). dNKT cells are also able to distinguish some types of lipid molecules presented through CD1d (Patel et al. 2012) and possibly exacerbate obesity by controlling the VAT volume and insulin resistance as mice deficient in both iNKT and dNKT show higher weight gain and adipocyte size compared to mice lacking only iNKT cells (Satoh et al. 2012), while also developing hypertriglyceridemia and aggravated inflammation (Subramanian et al. 2018). On the other hand, this population might also have beneficial effects as transferring both iNKT or dNKT cells into obese mice induces transient weight loss and stabilizes glucose homeostasis (Hams et al. 2013).

γδ‐T cells

γδ‐T cells are a specific subpopulation of T cells that develop in the thymus and express a unique receptor composed of one γ‐chain and one δ‐chain. Their activation is elicited mostly by innate signals subsequently triggering cytokine production and cytolytic reactions (Born et al. 2006). Activated γδ‐T cells enhance NK cell cytotoxicity through CD137 engagement, stimulate monocytes and macrophages and promote DC maturation further leading to Th and Tc lymphocyte activation (Leslie et al. 2002, Eberl et al. 2009, Maniar et al. 2010). As found in murine models (Mehta et al. 2015), γδ‐T cells reside in lean AT and increase during diet-induced obesity promoting inflammation and insulin resistance through the regulation of other immune cells such as macrophages. Furthermore, AT-resident γδ-T cells regulate the production of IL33 from stromal cells through the secretion of IL17A, thereby controlling age-dependent Treg expansion as well as core body temperature in response to environmental fluctuations. Potentially, IL17 produced by γδ-T cells could contribute to the Th17 response observed in obese subjects (Kohlgruber et al. 2018).

Dendritic cells in adipose tissue

DCs as professional antigen-presenting cells play a crucial role in the initiation of antigen-specific immunity and tolerance as they drive the maturation and polarization of naïve T cells through MHC complex class II alongside the exposure of appropriate membrane-bound signaling molecules and secretion of specific cytokines. As shown in mouse models (Wu et al. 2001), DCs can be differentiated from both myeloid and lymphoid common progenitor. They drive the regulation of pro-inflammatory as well as anti-inflammatory responses (Steinman 2012). Two major subtypes of DCs have been identified; CD11c-positive conventional (myeloid) DCs (cDCs) involved in the differentiation of CD4+ T cells and CD123-positive plasmacytoid DCs (pDCs) characterized by the production of type I interferon, activation of macrophages and antiviral defense (Tamura et al. 2005, Gilliet et al. 2008, Sundara Rajan & Longhi 2016). As shown in both lean mice and humans (Bertola et al. 2012), their AT contains resident cDCs, which are capable of capturing and presenting antigens to Th lymphocytes in an antigen-specific manner leading to their differentiation – preferentially Treg cells and Th17 lymphocytes in a very low extent.

The role of adipose tissue immune cells in obesity and T2DM

In case of tissue stress induced by long-term overweight and obesity, immune cells probably at first react to changes in the expression profile of the hypertrophied adipocytes (typically increased leptin, TNFα, IL6 and decreased adiponectin production) and try to regulate the commencing inflammation by changing their secretion and polarization profile. In the case of sustained tissue stress, original regulatory reactions apparently overshoot and immune cells themselves, under the influence of adipocyte-produced cytokines and chemokines, start to contribute to the inflammatory milieu (Fig. 1) (Lumeng et al. 2007, McLaughlin et al. 2014).

Figure 1
Figure 1

Immune cells and their main features in obese adipose tissue. Immune cells in adipose tissue react to tissue stress and hypoxia caused by obesity by their transmigration into tissue and pro-inflammatory reaction. CD, cluster of differentiation; IFN, interferon; IL, interleukin; MMe macrophages, metabolically activated macrophages; NK cells, natural killer cells; NKT cells, natural killer T cells; Tc cells, T cytotoxic cells; Th cells, T helper cells; TNF, tumor necrosis factor.

Citation: Journal of Endocrinology 252, 1; 10.1530/JOE-21-0159

As suggested on the basis of mostly rodent studies, lean state is associated with a smaller number of immune cells in AT and their anti-inflammatory profile, while overweight and obesity are characterized by a higher proportion of immune cells with pro-inflammatory status (Weisberg et al. 2003, Bourlier et al. 2008, Lumeng et al. 2008). The lean AT immune cells consist mainly of M2 macrophages, Th2 and Treg lymphocytes, whereas obese AT is dominated by classically activated M1 macrophages, Th1 and Tc lymphocytes and NK cells (Table 2). Additionally, other immune cells such as basophils, eosinophils, B lymphocytes, NKT and DCs can be found in AT with varying frequencies and functions between lean and obese fat (Mraz & Haluzik 2014, Cinkajzlova et al. 2017a). It is currently unclear, if a precise identification and phenotypization of AT immune cells might be used as a tool for the discrimination between different obesity subphenotypes. As determined by prospective epidemiological studies, metabolically healthy obese frequently become metabolically unhealthy leading to elevated risk of obesity-associated comorbidities (Echouffo-Tcheugui et al. 2019). Formerly evaluated methods including determination of peripheral blood leukocyte subclasses aimed at discriminating subjects at potentially increased complication risk seem to be insufficient (Pecht et al. 2014). Therefore, immunophenotyping of AT immune cells might, in spite of its invasiveness, represent an alternative approach to stratifying obese individuals according to their risk profile or serve as a tool for predicting the effects of weight-reducing interventions including bariatric procedures on mitigating the complication risk.

Table 2

The main differences in immune cells between lean and obese adipose tissue and their functions.

Lean adipose tissue Obese adipose tissue
Macrophages
M2 macrophages

• Anti-inflammatory actions

• Homeostatic functions

• Engulfment of free fatty acids

• IL4, IL13, IL10 production
M1/MMe macrophages

• Pro-inflammatory actions

• Reactive oxygen species and nitric oxide production

• TNFα, IL12, IL23 production
T lymphocytes
Th2 lymphocytes

Treg lymphocytes

• Homeostatic functions

• Anti-inflammatory actions

• IL5, IL4, IL13, IL10 production

Tc2 lymphocytes?

• IL2, IL5 production
Th1 lymphocytes

Th17 lymphocytes

• Pro-inflammatory actions

• IFNγ, IL2, IL6, IL17 production

Tc1 lymphocytes

• Cytotoxic functions

• Perforin and granzyme secretion

• IFNγ, TNFα, IL12, IL18 production
B lymphocytes
B1 lymphocytes

• IL10 production

• Antibody (IgM) production
B2 lymphocytes

• Antibody (IgG) production

• Antigen presentation to Th1 lymphocytes

• IL6 production
Non-T/B lymphocytes
iNKT cells

• Anti-inflammatory actions

• Direct cell communication with adipocytes through CD1c antigen

• IL4, IL13, IL10 production
NK cells

• Induction of cell death

• Cytotoxic functions

• IFNγ, TNFα production
Other immune cells
Eosinophils

Basophils

• IL4, IL13, IL10 production

Dendritic cells

• Activation of Th1/Th17 responses
Neutrophils

Mast cells

• IL1β, IL17 production

Dendritic cells

• Activation of Th17 responses

• Macrophage recruitment

• IL6, TGFβ, IL23 production

IFN, interferon; IL, interleukin; MMe macrophages, metabolically activated macrophages; NK cells, natural killer cells; NKT cells, natural killer T cells; Tc cells, T cytotoxic cells; Th cells, T helper cells; TNF, tumor necrosis factor; Treg, T regulatory cells; T2DM, type 2 diabetes mellitus.

Interestingly, loss of metabolic control and increase of inflammatory activity could be also associated with aging, which physiologically leads to immunosenescence (Lee et al. 2021). Immunosenescence is the term used for the decline in innate immunity (mainly in reduced neutrophil and macrophage activation and cytotoxic activity of NK cells) and dysregulated lymphocyte response (increase in anergic memory T cells, decline in naïve T cells, exhaustion of Th, Tc and B lymphocytes). Some hallmarks of obesity-associated changes overlap with immunosenescence, and obesity was proposed to accelerate aging (Perez et al. 2016), which is connected with higher AT abdominal and ectopic accumulation (Mancuso & Bouchard 2019), organelle stress (Ghosh et al. 2016) and with accumulation of senescence cells secreting pro-inflammatory cytokines such as IL6, IL8 or TNFα (Coppe et al. 2008, Baker et al. 2011). According to experimental data (Shirakawa et al. 2016), obesity seems to contribute to immunosenescence by pro-inflammatory activation of immune cells passing through AT microvasculature or pro-inflammatory differentiation and proliferation of cells within AT. Both mechanisms might negatively influence the efficacy of the immune system.

Adipose tissue myeloid cells in obesity and T2DM

Macrophages

As described above, in lean subjects, AT was shown to contain a smaller number of predominantly M2 anti-inflammatory macrophages (approximately 10% of total AT cells), while AT of obese individuals had substantially more macrophages (in extremely obese humans, AT macrophages can represent up to 40% of total AT cells (Weisberg et al. 2003)) with mainly M1 polarization (or the above-described MMe phenotype). As referred above, macrophages of obese are a significant source of pro-inflammatory cytokines such as MCP-1, IL-6 or TNF-α, which drive the development of AT inflammation (Kratz et al. 2014). One of the key players in this process might represent the Wnt signaling pathway, which in adipocytes acts as a modulator of their development (Farmer 2005) and contributes to insulin resistance by supporting AT expansion and lipid accumulation (Fuster et al. 2015, Aamir et al. 2020). In macrophages, Wnt signalization could amplify their pro-inflammatory cytokine production through crosstalk with the TLR-4 pathway, which is activated by lipopolysaccharide and free fatty acids acting via NLRP3 (Nod-like receptor family pyrin domain containing) pathway (Rogero & Calder 2018), as well as in an autocrine fashion through WnT5a expression and its Frizzled 5 receptor (Blumenthal et al. 2006, Catalan et al. 2014).

Nevertheless, the M1/MMe predominance within obese AT can be reversed by weight reduction as obese subjects after weight loss due to bariatric surgery showed lower macrophage count along with a shift back to the anti-inflammatory phenotype in both SAT and VAT (Aron-Wisnewsky et al. 2009, Cinkajzlova et al. 2017b). As shown in an experimental model (Li et al. 2010), phenotypic plasticity and AT macrophage polarization in obese subjects correlate well with the presence or absence of insulin resistance. Thus, insulin resistance might be directly improved by targeting macrophage polarization (Fujisaka et al. 2011).

Granulocytes

In a mouse model, high-fat diet induced eosinophil accumulation in perigonadal white AT. Furthermore, adipocytes promoted the migration of eosinophils and likely also their survival through the expression of IL-3, IL-5 and granulocyte–macrophage colony-stimulating factor. Eosinophil-deficient mice showed impaired glucose tolerance and adipocyte maturation, impairment of energy metabolism and increased inflammatory response (Lee et al. 2018). Eosinophil recruitment into AT might also be regulated by long-lived type 2 innate lymphoid cells (Nussbaum et al. 2013). Eosinophils are capable of IL-4 and other type 2 cytokine production contributing to M2 polarization that results in fat browning through tyrosine hydroxylase expression and catecholamine production (Qiu et al. 2014). On the other hand, a mere presence of eosinophils in AT is insufficient to alter metabolic health (Bolus et al. 2017).

Obesity is associated with increased number of neutrophils in blood, with their activation being also increased (Nascimento et al. 2010, Xu et al. 2015). In lean mice, lipolysis of adipocytes leads to neutrophil accumulation in AT prior to macrophage infiltration, with subsequent neutrophil IL-1β production (Watanabe et al. 2019b). Another murine study (Elgazar-Carmon et al. 2008) confirmed that neutrophils transiently infiltrate intraabdominal AT early (3 and 7 days) after high-fat feeding, where they directly interact with adipocytes (through CD11b and ICAM-1). In humans, neutrophils probably also accumulate in AT during weight gain, while bariatric surgery induces a neutrophil decrease in both SAT and VAT (Garcia-Rubio et al. 2018). Neutrophils could contribute to IL-17 production thus supporting Th17 lymphocyte immune responses (Lin et al. 2011).

Obese subjects (both humans and mice) have increased amount of mast cells in white AT compared to lean individuals, and their deficiency or pharmacological stabilization in mice reduced body weight gain and levels of inflammatory cytokines and proteases in serum and white AT along with improving glucose homeostasis and energy expenditure. Furthermore, mast cells contribute to white AT angiogenesis, associated cell apoptosis and cathepsin activity (Liu et al. 2009). Heparin secreted from stimulated cells may induce uptake of (modified) LDL by macrophages via scavenger receptor-mediated phagocytosis (Lindstedt et al. 1992). It was also suggested that mast cells and basophils possibly react directly with LDL through their own receptors (Virgolini et al. 1995). Surprisingly, bariatric surgery in obese humans was associated with an increase in mast cells in both SAT and VAT (Garcia-Rubio et al. 2018).

Adipose tissue lymphoid cells in obesity and T2DM

T helper lymphocytes

In diet-induced obese (DIO) mice, VAT Th1 lymphocyte population was shown to increase together with growing overweight and obesity, while other subpopulations were static (Winer et al. 2009). Th1 population was also able to produce more INF-γ (Rocha et al. 2008). In contrast, Th2 lymphocytes were suggested to suppress the effects of obesity and insulin resistance as demonstrated by their transplantation into AT of DIO mice (Winer et al. 2009). In obese T2DM subjects, increased circulatory Th1 and Th17 lymphocytes positively correlated with HOMA index of insulin resistance, leptin and insulin levels (Pacifico et al. 2006) and negatively with HDL cholesterol, respectively (Zeng et al. 2012). In experimental animals (Feuerer et al. 2009), Treg lymphocytes were enriched in the abdominal fat of lean subjects, while their number was reduced in insulin-resistant models.

T cytotoxic lymphocytes

Tc lymphocytes that accumulate in obese AT bear the Tc1 phenotype and produce IFN-γ, IL-12 and IL-18 that contribute to a pro-inflammatory milieu (Jiang et al. 2014). Ablation of these cells reduced T cell accumulation, activation and proliferation in AT and protected from insulin resistance. According to murine results (Nishimura et al. 2009), Tc lymphocytes precede macrophage accumulation, and their depletion lowers macrophage infiltration into AT and ameliorates systemic insulin resistance. The percentage of Tc lymphocytes was shown to be higher in VAT compared to SAT and was associated with higher caspase-1 activity leading to IL-1β and IL-18 production, respectively (Koenen et al. 2011). Interestingly, caspase-1 is also activated by free fatty acids which by triggering the NLRP3 pathway initiates the formation of inflammasome and cleavage of pro-caspase-1 into active caspase (Wu et al. 2020). IL-18 in combination with IL-12 and IL-1β, which are also increased in obese AT, participate in increased IFN-γ production by Tc lymphocytes further leading to the activation of M1 macrophages and stimulation of inflammation (McDonnell et al. 2012). Surprisingly, Tc-derived Prf-1 seems to play a protective role as Prf-1 null mice placed on high-fat diet showed increased body weight, adiposity and insulin resistance compared with WT littermates indicating that Prf-1 might be a possible regulator of early body size and growth (Revelo et al. 2015). Besides that, Prf-1 null mice showed higher accumulation of IFN-γ-producing T lymphocytes and M1 macrophages in VAT further suggesting Prf to be involved in homeostatic regulation of T cells and in macrophage infiltration into AT. The role of granzyme is not precisely known, although it might contribute to inflammation by pro-inflammatory cytokine production (Metkar et al. 2008). Furthermore, single-cell RNA sequencing technique revealed that AT of obese subjects contains a subpopulation of CCL5+ Tc lymphocytes positively correlating with the degree of obesity and producing metallothionein (Vijay et al. 2020). Metallothionein seems to prevent the manifestation of obesity-related diseases through suppression of superoxide production, endoplasmic reticulum stress and associated damage (Sato et al. 2013).

B lymphocytes

As shown in DIO mice (Winer et al. 2011), during obesity, B cells infiltrate VAT (starting at 4 weeks of high-fat diet), exhibit maturation in correlation with the development of insulin resistance (without change in proportion between B cell subtypes), produce antibodies supporting inflammation (increase of Ig2c and decrease of IgA) and contribute to T lymphocyte infiltration. IgM production by protective B-1 cells in murine VAT reduced cytokine production by M1 macrophages and ameliorated AT inflammation, insulin resistance and systemic glucose intolerance, while adoptive transfer of B-2 cells into VAT promoted metabolic dysfunction. B-2 cells probably produce pro-inflammatory cytokines (DeFuria et al. 2013) and promote insulin resistance through the secretion of leukotriene B4 (Ying et al. 2017). Furthermore, B-1 (CD20+CD27+CD43+) cells and IgM antibodies were also identified in human VAT, where they inversely correlated with inflammation and insulin resistance in obese subjects (Harmon et al. 2016). Similarly, reduction of B regulatory cells producing the anti-inflammatory IL-10 was identified in both obese mice and humans (Nishimura et al. 2013), and this reduction was possibly associated with Th1/Th17 cytokine (IFN-γ, IL-17) increase (Garcia-Hernandez et al. 2018).

Natural killer cells

Obesity is associated with NK cell accumulation in AT (while their cytotoxic function remains unchanged) and increased NCR-1 expression on adipocytes (Wensveen et al. 2015). NK cells contribute to inflammatory reactions by increased production of IFN-γ and TNF-α, which support M1 macrophage polarization (Lee et al. 2016). Accumulation of NK cells was found to be increased in human VAT in obesity (Wouters et al. 2020), while in mice AT, NK cell depletion was associated with better insulin sensitivity and macrophage reduction (Lee et al. 2016). In contrast to Tc lymphocyte-produced Prf-1, NK-produced Prf-1 possibly does not have any regulatory effects on glucose homeostasis (Revelo et al. 2015).

Natural killer T cells

Resident iNKT cells represent 1–20% of the resident T lymphocytes in lean AT, while obesity and high caloric intake reduce their population (Lynch et al. 2009, 2012, Schipper et al. 2012). In obese AT, iNKT cells are activated by lipids presented by adipocytes and produce IL-4, IL-10 and IL-2 cytokines driving the anti-inflammatory (M2 macrophage, Treg and Th2 lymphocyte) response (Park et al. 2018), but their number significantly declines in parallel with growing inflammation (Lynch et al. 2012). The protective role of iNKT cells was also confirmed by experiments with CD1d deletion in mice models leading to aggravated AT inflammation and insulin resistance (Huh et al. 2017).

γδ‐T cells

In several murine studies (Zuniga et al. 2010, Mehta et al. 2015), the absence of γδ‐T cells was associated with either no difference or a decrease in high-fat diet-induced insulin resistance with partial depletion of γδ‐T cells showing no protection in this regard. In contrast, reduction of αβ/γδ‐T cell ratio (achieved by transgenic overexpression of PPAR-β receptor leading to a partial depletion of αβ-T cells) partially protected the animals from high-fat diet-induced weight gain and decreased insulin resistance and liver steatosis independently of animal weight (Le Menn et al. 2019). γδ‐T cells were also shown to have a crucial role in promoting sympathetic innervation of local AT, ablation of which was associated with defective thermogenesis and the development of obesity (Hu et al. 2020). In another study, mice lacking γδ‐T cells had impaired glucose homeostasis, while ketogenic diet expanded their AT pool which was associated with restrained inflammation. In contrast, long-term ad libitum ketogenic diet depleted AT γδ‐T cells and caused obesity and metabolic derangements (Goldberg et al. 2020).

Dendritic cells

The role of DCs in regulating inflammation in AT seems to be dependent on the actual tissue metabolic state. In lean animals, cDCs promote an anti-inflammatory state and delay the onset of obesity-induced chronic inflammation and insulin resistance (Macdougall et al. 2018), while long-term overnutrition induces a pro-inflammatory switch of cDCs resulting in activation of Th1 and Th17 cell responses (Bertola et al. 2012, Chen et al. 2014). The presence of DCs, which locally increase in murine AT during high-fat diet, seems to be essential for macrophage recruitment and their subsequent activation (Stefanovic-Racic et al. 2012, Cho et al. 2016).

Besides, a minor subpopulation of myeloid DCs with Prf-containing granules might possibly be involved in the regulation of body weight and development of the metabolic syndrome through their control over inflammatory T lymphocytes (Zlotnikov-Klionsky et al. 2015). Interestingly, subjects with T2DM showed increased amounts of pDCs in SAT and even more in EAT relative to non-diabetic subjects regardless of the presence of obesity (Mraz et al. 2019a), while no such changes were seen in cDCs. Single-cell RNA sequencing revealed three populations of cDCs (cDC1, cDC2A and cDC2B) in SAT human samples with thus far unknown roles, although activation of Wnt signalization and tolerogenic programs in AT have been proposed. One population (cDC2B) might also trigger tissue inflammation (Hildreth et al. 2021). Besides the Wnt pathway, IL1β production could be increased by priming NLRP3 inflammasome formation through TLR4 under the exposition to free fatty acids, similarly to macrophages or Tc lymphocytes (Reynolds et al. 2012).

The role of adipose tissue immune cells in cardiovascular diseases

As evidenced by the association of obesity and T2DM with cardiovascular complications, AT and immune cell changes seem to have an eminent role in the development of cardiovascular diseases (Fig. 2). For instance, EAT, whose biological characteristics are similar to VAT, is a potentially important source of inflammatory cytokines that affect surrounding cardiac tissues in autocrine, paracrine and endocrine manner thus contributing to chronic inflammation and myocardium damage (Guzzardi & Iozzo 2011). As shown before (Cheng et al. 2008), TNF-α, IL1β and IL6 tissue levels are higher in patients with coronary artery disease (CAD) compared to non-CAD patients, and their actions are linked to the development or progression of cardiovascular diseases (Ferrari 1999, Volpato et al. 2001, Vicenova et al. 2009). Similarly, PVAT adjacent to human atherosclerotic arteries is more inflamed than PVAT adjacent to non-atherosclerotic vessels (Henrichot et al. 2005), and its thickness around coronary arteries has been associated with coronary artery calcification and increased CVD risk (de Vos et al. 2008).

Figure 2
Figure 2

Major immune cell types associated with cardiovascular risk and their main functions in adjacent adipose tissue depots or atherosclerotic plaques. Although associations between γδ‐T cells, dendritic cells, eosinophils and neutrophils and cardiovascular risk have also been suggested, their significance and functions are still unclear. CD, cluster of differentiation; IFN, interferon; IL, interleukin; MMe macrophages, metabolically activated macrophages; NK cells, natural killer cells; NKT cells, natural killer T cells; Tc cells, T cytotoxic cells; Th cells, T helper cells; TNF, tumor necrosis factor.

Citation: Journal of Endocrinology 252, 1; 10.1530/JOE-21-0159

Myeloid cells in cardiovascular diseases

Macrophages

Human immunochemistry studies (Hirata et al. 2011, Vianello et al. 2016) have shown that EAT macrophages have M1 polarization in patients with CAD, and the percentage of these cells is higher compared to patients without CAD, in which M2 macrophages predominate. This more pro-inflammatory state is accompanied by increased TNFα and decreased adiponectin expression. It is a question if vitamin D supply associating with positive M1 into M2 macrophage transition in EAT of swine could have also beneficial effect during coronary intervention in human (Gunasekar et al. 2018). Besides, macrophage content in PVAT is positively related to the size and characteristics of the atherosclerotic plaque (its lipid core, calcification, collagen and smooth muscle cell content) (Verhagen et al. 2012), and it is also greater near unstable atherosclerosis plaques as compared with stable lesions and positively correlates with the percentage of coronary artery obstruction (Farias‐Itao et al. 2019). Interestingly, the number of PVAT macrophages in subjects with CAD was shown to be similar to individuals with dilated cardiomyopathy (Kralova Lesna et al. 2015). Furthermore, macrophages in pericoronary AT can store oxidized LDL and HDL irrespective of the presence of atherosclerosis, while during its progression, they are able to migrate into the coronary intima and contribute to plaque growth and maturation (Uchida et al. 2017).

Granulocytes

Although evidence about differences in (blood) eosinophils in association with CAD is ambiguous (Verdoia et al. 2015, Gao et al. 2019), a rather positive role of AT eosinophils in protection against atherosclerosis seems plausible given their actions in obesity. However, eosinophil degranulation was shown to be higher in subjects with acute CAD; therefore, their exact function in the development of CAD needs further assessment (Niccoli et al. 2015).

Data about the association between AT neutrophils and cardiovascular risk are limited. In obese females, neutrophils were shown to accumulate in the microvasculature of AT, and their presence positively correlated with BMI and diastolic blood pressure. Similarly, vascular inflammation (evaluated as expression of activated nuclear factor kappaB and cyclooxygenase-2) positively correlated with BMI (Leik & Walsh 2004, Shah et al. 2010). Neutrophils adhere to the endothelium of white AT and are not present in AT parenchyma, suggesting that in vivo they do not transmigrate into the tissue like other immune cells or that their transmigration is limited (Rouault et al. 2013). Nevertheless, neutrophils are potential contributors to atherosclerotic plaque ruptures (Ionita et al. 2010).

In subjects with acute CAD, higher basophil activation (Niccoli et al. 2015) and blood count (Soylu et al. 2014) was found, which might be decreased by statin treatment resulting in a reduction of arterial wall stiffness (Toyama et al. 2012). Mast cells found in the heart were reported to participate in the development of atherosclerosis, coronary inflammation and cardiac ischemia (Kritikou et al. 2016). Moreover, mast cells showed increased prevalence in atherosclerotic lesions and were associated with plaque microvessel density (Willems et al. 2013). Nevertheless, a causal relationship between AT mast cells and basophils and cardiovascular risk has not been confirmed yet.

Lymphoid cells in cardiovascular diseases

T helper lymphocytes

As shown in carotid artery plaques of patients with recent stroke or transient ischemic attack, atherosclerotic plaques are rich in activated Th1 and Th17 lymphocytes (Fernandez et al. 2019). Circulatory Th1 lymphocytes are associated with increased prevalence of acute coronary artery syndrome and atherosclerosis (Methe et al. 2005), while Th2 and Treg responses reduce the risk of myocardial infarction and stroke (Han et al. 2007, Engelbertsen et al. 2013). Interestingly, the protective role of Th2 cells seems to be more prominent in females (Engelbertsen et al. 2013). In humans, EAT and SAT density of Th lymphocytes was not influenced by CAD, although the allover T lymphocyte population was higher in CAD subjects as compared with controls without CAD (Mraz et al. 2019b). As shown by a multi-ethnic study of atherosclerosis (Olson et al. 2013), memory CD4+ T cells are positively related to arterial intimal media thickness, while naïve T cells displayed inverse correlation. Furthermore, higher amount of circulating CD4+ T memory cells was associated with coronary artery calcification. According to rodent results (Bansal et al. 2017), Th lymphocyte expansion in circulation, as well as cardiac and lymphatic tissue, is seen in chronic ischemic heart failure with a predominance of Th2 vs Th1 and Th17 vs Treg cells in the failing myocardium. The function of Th17 in cardiovascular diseases is still unclear, although its master cytokine IL17A expression is increased in atherosclerotic lesions. IL17A is also positively associated with inflammation and plaque vulnerability and negatively with IL10 expression (Erbel et al. 2011). IL17 was also shown to be produced concomitantly with the Th1 signature cytokine IFNγ in coronary artery plaques resulting in synergistical increase of pro-inflammatory responses by vascular smooth muscle cells (Eid et al. 2009). CD28 null (CD28−CD4+) T lymphocytes are another subset of T cells that starts emerging in the context of increased cardiovascular risk. These cells producing IFNγ were found in subjects with acute coronary syndrome, unstable angina and myocardial infarction, where their presence was associated with increased mortality and event recurrence (Liuzzo et al. 1999, 2007). In another study, CAD was associated with CD28−CD4+ T cell increase in circulation and correlated positively with IFNγ lymphocyte production (Liuzzo et al. 2000). Also, increased amount of CD28−CD4+ cells was an independent predictor of future acute coronary events in subjects with unstable angina (Liuzzo et al. 2007).

T cytotoxic lymphocytes

Tc lymphocytes are generally less abundant than Th lymphocytes in human atherosclerotic plaques; however, they might represent up to 50% of leukocytes in advanced lesions, where they dominate over Th lymphocytes and contribute to inflammation by TNFα production and to overall plaque instability (Kyaw et al. 2013). Tc lymphocytes promote the development of vulnerable atherosclerotic plaques by activation of macrophage, smooth muscle and endothelial cell apoptosis resulting in necrotic core formation (van der Wal et al. 1989, Kyaw et al. 2013, Paul et al. 2016). Interestingly, however, Tc lymphocytes seem to have both pro-atherogenic and atheroprotective roles, as their cytotoxic activity toward vascular smooth muscle and endothelial cells together with cytokine production drives the progression and instability of the lesions, while cytotoxic activity toward antigen-presenting cells in turn limits atherosclerosis (Saigusa et al. 2020). In contrast, a cardioprotective Tc cell population characterized by the presence of angiotensin type 2 receptor (AT2R) was identified in rat models. This population increases during ischemic heart injury and contributes to maintaining cardiomyocyte viability and IL10 production, hence suggesting the existence of an AT2R-mediated cellular mechanism involved in modulating the adaptive immune response in the heart (Curato et al. 2010).

B lymphocytes

Experiments with PVAT in mice showed B-1 cells attenuating and B-2 cells aggravating atherosclerosis. B-1 cells limit atherosclerosis development by producing IgM aimed at oxidation of specific epitopes on LDL particles thus blocking oxidized LDL-induced inflammatory cytokine production and foam cell formation (Rosenfeld et al. 2015). As shown by murine transfer studies, B-2 cells are capable of promoting atherosclerosis development entirely on their own without the need of any other lymphocyte population. In the presence of T cells or other lymphocytes, they significantly augment atherosclerosis, while their depletion markedly ameliorates plaque formation (Kyaw et al. 2010). Interestingly, B cells predominate in EAT compared to SAT in human subjects regardless of the presence of CAD, although the role of B-1 and B-2 subtypes in the development of coronary atherosclerosis in humans still needs to be addressed (Mraz et al. 2019b). Besides, the percentage of arterial obstruction increases with the density of B lymphocytes in the periplaque AT, and the density of B cells in the periplaque AT is greater compared with more distant PVAT (Farias‐Itao et al. 2019).

Natural killer cells

Subjects with severe atherosclerosis were shown to have increased amount of circulating NK cells and enhanced the presence of several antigens triggering cytotoxicity and cytokine secretion (e.g. CD160) (Clerc & Rouz 1997, Zuo et al. 2015). Moreover, NK cells were found directly in atherosclerotic plaques (Bobryshev & Lord 2005). However, in other studies, CAD was associated with decreased NK cell levels in blood (Backteman et al. 2014) and in EAT compared to SAT (Mraz et al. 2019b), thus their exact role in the development of atherosclerotic complications requires further elucidation.

Natural killer T cells

As suggested by murine studies (Li et al. 2015) and somehow surprisingly given their anti-inflammatory effects in adipose tissue, NKT cells potently promote atherosclerosis by Prf and granzyme B-dependent apoptosis that increases necrosis and inflammation in atherosclerotic plaques without altering cytokine production. iNKT cell activation during the development of atherosclerosis is at least in part dependent on lipid antigens activating T cell receptors (TCRs) as shown in mice treated by NK cell CD1d-dependent lipid antagonist (Li et al. 2016). Mice lacking both invariant and diverse NKT cells showed increased atherosclerotic lesion area compared to mice lacking only iNKT cell suggesting a possible role of dNKT cells in CAD (Subramanian et al. 2018).

γδ‐T cells

The potential role of γδ‐T cells in the development of atherosclerosis and cardiovascular diseases is currently rather unclear as the available data are scarce (Saigusa et al. 2020). γδ‐T cells were found in atherosclerotic lesions in both mice and humans (Kleindienst et al. 1993, Vu et al. 2014). However, γδ‐T cell-deficient Apoe−/− mice developed atherosclerosis as early as their littermates with preserved γδ-T cells (Cheng et al. 2014). Interestingly, the activation and proliferation of γδ‐T cells is regulated by their intracellular cholesterol content indicating that increased cholesterol content contributes to their hyperactivated phenotype potentially leading to IL17 production and possibly worsening atherosclerosis (Cheng et al. 2013).

Dendritic cells

The presence of chronic CAD did not correlate with total DC content in human EAT (Mraz et al. 2019a). In contrast, in an animal model of acute coronary syndrome, acute ligation of coronary arteries increased the amount of DCs in EAT due to their migration from the infarct site in the myocardium (Horckmans et al. 2018); however, whether this might be a protective or rather a deleterious mechanism remains to be seen.

Conclusion

Immune cells represent an indispensable part of AT. Through their activities (phagocytosis, direct cell contacts, etc.) and cytokine production, they directly and indirectly influence adipocytes and thus also the metabolic and endocrine function of AT that further affects the homeostasis of the whole organism. AT immune cells of both lean and obese subjects include a variety of immune cells (albeit in different proportions), which reciprocally communicate, affect and complement themselves. In obese subjects, the amount of most immune cells in AT increases, and their phenotype changes toward pro-inflammatory state. These changes negatively influence adipocyte function, insulin sensitivity and lipid metabolism and create the basis for the development of T2DM, atherosclerosis and subsequent cardiovascular complications.

Cutting-edge research approaches such as single-cell RNA sequencing enable us to take the characterization of AT immune cells as well as adipocytes and their progenitors to the next level thus providing a novel and exciting look at the AT and its homeostatic functions. Therapeutic strategies aimed at reducing the amount of immune cells, favoring beneficial cell subtypes or modifying their phenotype back to the anti-inflammatory state typical for the lean AT might represent a novel approach in combating obesity-related diseases that would directly affect one of the main pathophysiological mechanisms responsible for the metabolic complications of excessive adiposity; however, the development of such treatment approaches is still in its beginnings.

Declaration of interest

Martin Haluzik is an Editor in Chief of Journal of Endocrinology and Journal of Molecular Endocrinology. Martin Haluzik was not involved in the review or editorial process for this article, on which he is listed as an author. The other authors have nothing to disclose.

Funding

This work was supported by the grants AZV NU20-02-00190, AZV NV19-02-00118 and RVO-VFN64165, and by MH CZ – DRO (Institute for Clinical and Experimental Medicine – IKEM, IN 00023001).

References

  • Aamir K, Khan HU, Sethi G, Hossain MA & & Arya A 2020 Wnt signaling mediates TLR pathway and promote unrestrained adipogenesis and metaflammation: therapeutic targets for obesity and type 2 diabetes. Pharmacological Research 152 104602. (https://doi.org/10.1016/j.phrs.2019.104602)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Aron-Wisnewsky J, Tordjman J, Poitou C, Darakhshan F, Hugol D, Basdevant A, Aissat A, Guerre-Millo M & & Clement K 2009 Human adipose tissue macrophages: m1 and m2 cell surface markers in subcutaneous and omental depots and after weight loss. Journal of Clinical Endocrinology and Metabolism 94 46194623. (https://doi.org/10.1210/jc.2009-0925)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Athie-Morales V, Smits HH, Cantrell DA & & Hilkens CM 2004 Sustained IL-12 signaling is required for Th1 development. Journal of Immunology 172 6169. (https://doi.org/10.4049/jimmunol.172.1.61)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Backteman K, Ernerudh J & & Jonasson L 2014 Natural killer (NK) cell deficit in coronary artery disease: no aberrations in phenotype but sustained reduction of NK cells is associated with low-grade inflammation. Clinical and Experimental Immunology 175 104112. (https://doi.org/10.1111/cei.12210)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, Kirkland JL & & van Deursen JM 2011 Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479 232236. (https://doi.org/10.1038/nature10600)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bansal SS, Ismahil MA, Goel M, Patel B, Hamid T, Rokosh G & & Prabhu SD 2017 Activated T lymphocytes are essential drivers of pathological remodeling in ischemic heart failure. Circulation: Heart Failure 10 e003688. (https://doi.org/10.1161/CIRCHEARTFAILURE.116.003688)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Belge KU, Dayyani F, Horelt A, Siedlar M, Frankenberger M, Frankenberger B, Espevik T & & Ziegler-Heitbrock L 2002 The proinflammatory CD14+CD16+DR++ monocytes are a major source of TNF. Journal of Immunology 168 35363542. (https://doi.org/10.4049/jimmunol.168.7.3536)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Berger A 2000 Th1 and Th2 responses: what are they? BMJ 321 424. (https://doi.org/10.1136/bmj.321.7258.424)

  • Bertola A, Ciucci T, Rousseau D, Bourlier V, Duffaut C, Bonnafous S, Blin-Wakkach C, Anty R, Iannelli A & Gugenheim J et al.2012 Identification of adipose tissue dendritic cells correlated with obesity-associated insulin-resistance and inducing Th17 responses in mice and patients. Diabetes 61 22382247. (https://doi.org/10.2337/db11-1274)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Blumenthal A, Ehlers S, Lauber J, Buer J, Lange C, Goldmann T, Heine H, Brandt E & & Reiling N 2006 The wingless homolog WNT5A and its receptor frizzled-5 regulate inflammatory responses of human mononuclear cells induced by microbial stimulation. Blood 108 965973. (https://doi.org/10.1182/blood-2005-12-5046)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bobryshev YV & & Lord RS 2005 Identification of natural killer cells in human atherosclerotic plaque. Atherosclerosis 180 423427. (https://doi.org/10.1016/j.atherosclerosis.2005.01.046)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bolus WR, Gutierrez D, Anderson-Baucum E, Kennedy A & & Hasty A 2017 The role of eosinophils in adipose tissue inflammation and metabolic function. Journal of Immunology 198 63.1863.18.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Born WK, Reardon CL & & O’Brien RL 2006 The function of gammadelta T cells in innate immunity. Current Opinion in Immunology 18 3138. (https://doi.org/10.1016/j.coi.2005.11.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bourlier V, Zakaroff-Girard A, Miranville A, De Barros S, Maumus M, Sengenes C, Galitzky J, Lafontan M, Karpe F & Frayn KN et al.2008 Remodeling phenotype of human subcutaneous adipose tissue macrophages. Circulation 117 806815. (https://doi.org/10.1161/CIRCULATIONAHA.107.724096)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Caspar-Bauguil S, Cousin B, Galinier A, Segafredo C, Nibbelink M, Andre M, Casteilla L & & Penicaud L 2005 Adipose tissues as an ancestral immune organ: site-specific change in obesity. FEBS Letters 579 34873492. (https://doi.org/10.1016/j.febslet.2005.05.031)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Castro A, Concha LE & & Pantoja C 2017 Low-grade inflammation and its relation to obesity and chronic degenerative diseases . Revista Médica del Hospital General de México 80 101105 . (https://doi.org/10.1016/j.hgmx.2016.06.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Catalan V, Gomez-Ambrosi J, Rodriguez A, Perez-Hernandez AI, Gurbindo J, Ramirez B, Mendez-Gimenez L, Rotellar F, Valenti V & Moncada R et al.2014 Activation of noncanonical Wnt signaling through WNT5A in visceral adipose tissue of obese subjects is related to inflammation. Journal of Clinical Endocrinology and Metabolism 99 E1407E1417. (https://doi.org/10.1210/jc.2014-1191)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Charriere G, Cousin B, Arnaud E, Andre M, Bacou F, Penicaud L & & Casteilla L 2003 Preadipocyte conversion to macrophage. Evidence of plasticity. Journal of Biological Chemistry 278 98509855. (https://doi.org/10.1074/jbc.M210811200)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chaudhry A, Samstein RM, Treuting P, Liang Y, Pils MC, Heinrich JM, Jack RS, Wunderlich FT, Bruning JC & Muller W et al.2011 Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity 34 566578. (https://doi.org/10.1016/j.immuni.2011.03.018)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chen Y, Tian J, Tian X, Tang X, Rui K, Tong J, Lu L, Xu H & & Wang S 2014 Adipose tissue dendritic cells enhances inflammation by prompting the generation of Th17 cells. PLoS ONE 9 e92450. (https://doi.org/10.1371/journal.pone.0092450)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cheng KH, Chu CS, Lee KT, Lin TH, Hsieh CC, Chiu CC, Voon WC, Sheu SH & & Lai WT 2008 Adipocytokines and proinflammatory mediators from abdominal and epicardial adipose tissue in patients with coronary artery disease. International Journal of Obesity 32 268274. (https://doi.org/10.1038/sj.ijo.0803726)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cheng HY, Wu R, Gebre AK, Hanna RN, Smith DJ, Parks JS, Ley K & & Hedrick CC 2013 Increased cholesterol content in gammadelta (gammadelta) T lymphocytes differentially regulates their activation. PLoS ONE 8 e63746. (https://doi.org/10.1371/journal.pone.0063746)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cheng HY, Wu R & & Hedrick CC 2014 Gammadelta (gammadelta) T lymphocytes do not impact the development of early atherosclerosis. Atherosclerosis 234 265269. (https://doi.org/10.1016/j.atherosclerosis.2014.03.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cho KW, Zamarron BF, Muir LA, Singer K, Porsche CE, DelProposto JB, Geletka L, Meyer KA, O’Rourke RW & & Lumeng CN 2016 Adipose tissue dendritic cells are independent contributors to obesity-induced inflammation and insulin resistance. Journal of Immunology 197 36503661. (https://doi.org/10.4049/jimmunol.1600820)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cinkajzlova A, Mraz M & & Haluzik M 2017 a Lymphocytes and macrophages in adipose tissue in obesity: markers or makers of subclinical inflammation? Protoplasma 254 12191232. (https://doi.org/10.1007/s00709-017-1082-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cinkajzlova A, Lacinova Z, Klouckova J, Kavalkova P, Trachta P, Kosak M, Kratky J, Kasalicky M, DoleZalova K & Mraz M et al.2017 b An alternatively activated macrophage marker CD163 in severely obese patients: the influence of very low-calorie diet and bariatric surgery. Physiological Research 66 641652. (https://doi.org/10.33549/physiolres.933522)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cinkajzlova A, Anderlova K, Simjak P, Lacinova Z, Klouckova J, Kratochvilova H, Krejci H, Parizek A, Mraz M & Krsek M et al.2020 Subclinical inflammation and adipose tissue lymphocytes in pregnant females with gestational diabetes mellitus. Journal of Clinical Endocrinology and Metabolism 105 dgaa528. (https://doi.org/10.1210/clinem/dgaa528)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clerc G & & Rouz PM 1997 Lymphocyte subsets in severe atherosclerosis before revascularization. Annals of Internal Medicine 126 10041005. (https://doi.org/10.7326/0003-4819-126-12-199706150-00028)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Coats BR, Schoenfelt KQ, Barbosa-Lorenzi VC, Peris E, Cui C, Hoffman A, Zhou G, Fernandez S, Zhai L & Hall BA et al.2017 Metabolically activated adipose tissue macrophages perform detrimental and beneficial functions during diet-induced obesity. Cell Reports 20 31493161. (https://doi.org/10.1016/j.celrep.2017.08.096)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Coppe JP, Patil CK, Rodier F, Sun Y, Munoz DP, Goldstein J, Nelson PS, Desprez PY & & Campisi J 2008 Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biology 6 28532868. (https://doi.org/10.1371/journal.pbio.0060301)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cousin B, Munoz O, Andre M, Fontanilles AM, Dani C, Cousin JL, Laharrague P, Casteilla L & & Penicaud L 1999 A role for preadipocytes as macrophage-like cells. FASEB Journal 13 305312. (https://doi.org/10.1096/fasebj.13.2.305)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cousin B, Andre M, Casteilla L & & Penicaud L 2001 Altered macrophage-like functions of preadipocytes in inflammation and genetic obesity. Journal of Cellular Physiology 186 380386. (https://doi.org/10.1002/1097-4652(2001)9999:9999<000::AID-JCP1038>3.0.CO;2-T)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Curato C, Slavic S, Dong J, Skorska A, Altarche-Xifro W, Miteva K, Kaschina E, Thiel A, Imboden H & Wang J et al.2010 Identification of noncytotoxic and IL-10-producing CD8+AT2R+ T cell population in response to ischemic heart injury. Journal of Immunology 185 62866293. (https://doi.org/10.4049/jimmunol.0903681)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • de Vos AM, Prokop M, Roos CJ, Meijs MF, van der Schouw YT, Rutten A, Gorter PM, Cramer MJ, Doevendans PA & Rensing BJ et al.2008 Peri-coronary epicardial adipose tissue is related to cardiovascular risk factors and coronary artery calcification in post-menopausal women. European Heart Journal 29 777783. (https://doi.org/10.1093/eurheartj/ehm564)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Deauvieau F, Ollion V, Doffin AC, Achard C, Fonteneau JF, Verronese E, Durand I, Ghittoni R, Marvel J & Dezutter-Dambuyant C et al.2015 Human natural killer cells promote cross-presentation of tumor cell-derived antigens by dendritic cells. International Journal of Cancer 136 10851094. (https://doi.org/10.1002/ijc.29087)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • DeFuria J, Belkina AC, Jagannathan-Bogdan M, Snyder-Cappione J, Carr JD, Nersesova YR, Markham D, Strissel KJ, Watkins AA & Zhu M et al.2013 B cells promote inflammation in obesity and type 2 diabetes through regulation of T-cell function and an inflammatory cytokine profile. PNAS 110 51335138. (https://doi.org/10.1073/pnas.1215840110)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dobrzanski MJ, Reome JB, Hollenbaugh JA & & Dutton RW 2004 Tc1 and Tc2 effector cell therapy elicit long-term tumor immunity by contrasting mechanisms that result in complementary endogenous type 1 antitumor responses. Journal of Immunology 172 13801390. (https://doi.org/10.4049/jimmunol.172.3.1380)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eberl M, Roberts GW, Meuter S, Williams JD, Topley N & & Moser B 2009 A rapid crosstalk of human gammadelta T cells and monocytes drives the acute inflammation in bacterial infections. PLoS Pathogens 5 e1000308. (https://doi.org/10.1371/journal.ppat.1000308)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Echouffo-Tcheugui JB, Short MI, Xanthakis V, Field P, Sponholtz TR, Larson MG & & Vasan RS 2019 Natural history of obesity subphenotypes: dynamic changes over two decades and prognosis in the Framingham heart study. Journal of Clinical Endocrinology and Metabolism 104 738752. (https://doi.org/10.1210/jc.2018-01321)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eid RE, Rao DA, Zhou J, Lo SF, Ranjbaran H, Gallo A, Sokol SI, Pfau S, Pober JS & & Tellides G 2009 Interleukin-17 and interferon-gamma are produced concomitantly by human coronary artery-infiltrating T cells and act synergistically on vascular smooth muscle cells. Circulation 119 14241432. (https://doi.org/10.1161/CIRCULATIONAHA.108.827618)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Elgazar-Carmon V, Rudich A, Hadad N & & Levy R 2008 Neutrophils transiently infiltrate intra-abdominal fat early in the course of high-fat feeding. Journal of Lipid Research 49 18941903. (https://doi.org/10.1194/jlr.M800132-JLR200)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Elgueta R, Benson MJ, de Vries VC, Wasiuk A, Guo Y & & Noelle RJ 2009 Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunological Reviews 229 152172. (https://doi.org/10.1111/j.1600-065X.2009.00782.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Engelbertsen D, Andersson L, Ljungcrantz I, Wigren M, Hedblad B, Nilsson J & & Bjorkbacka H 2013 T-helper 2 immunity is associated with reduced risk of myocardial infarction and stroke. Arteriosclerosis, Thrombosis, and Vascular Biology 33 637644. (https://doi.org/10.1161/ATVBAHA.112.300871)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Erbel C, Dengler TJ, Wangler S, Lasitschka F, Bea F, Wambsganss N, Hakimi M, Bockler D, Katus HA & & Gleissner CA 2011 Expression of IL-17A in human atherosclerotic lesions is associated with increased inflammation and plaque vulnerability. Basic Research in Cardiology 106 125134. (https://doi.org/10.1007/s00395-010-0135-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Farias-Itao DS, Pasqualucci CA, Nishizawa A, Silva LFFd, Campos FM, Bittencourt MS, Silva KCSd, Leite REP, Grinberg LT & Ferretti-Rebustini REdL et al.2019 B lymphocytes and macrophages in the perivascular adipose tissue are associated with coronary atherosclerosis: an autopsy study. Journal of the American Heart Association 8 e013793.(https://doi.org/10.1161/JAHA.119.013793)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Farmer SR 2005 Regulation of PPARgamma activity during adipogenesis. International Journal of Obesity 29 (Supplement 1) S13S16. (https://doi.org/10.1038/sj.ijo.0802907)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fernandez DM, Rahman AH, Fernandez NF, Chudnovskiy A, Amir ED, Amadori L, Khan NS, Wong CK, Shamailova R & Hill CA et al.2019 Single-cell immune landscape of human atherosclerotic plaques. Nature Medicine 25 15761588. (https://doi.org/10.1038/s41591-019-0590-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ferrari R 1999 The role of TNF in cardiovascular disease. Pharmacological Research 40 97105. (https://doi.org/10.1006/phrs.1998.0463)

  • Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, Lee J, Goldfine AB, Benoist C & Shoelson S et al.2009 Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nature Medicine 15 930939. (https://doi.org/10.1038/nm.2002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Freeman BE, Hammarlund E, Raue HP & & Slifka MK 2012 Regulation of innate CD8+ T-cell activation mediated by cytokines. PNAS 109 99719976. (https://doi.org/10.1073/pnas.1203543109)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fujisaka S, Usui I, Kanatani Y, Ikutani M, Takasaki I, Tsuneyama K, Tabuchi Y, Bukhari A, Yamazaki Y & Suzuki H et al.2011 Telmisartan improves insulin resistance and modulates adipose tissue macrophage polarization in high-fat-fed mice. Endocrinology 152 17891799. (https://doi.org/10.1210/en.2010-1312)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fuster JJ, Zuriaga MA, Ngo DT, Farb MG, Aprahamian T, Yamaguchi TP, Gokce N & & Walsh K 2015 Noncanonical Wnt signaling promotes obesity-induced adipose tissue inflammation and metabolic dysfunction independent of adipose tissue expansion. Diabetes 64 12351248. (https://doi.org/10.2337/db14-1164)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gao S, Deng Y, Wu J, Zhang L, Deng F, Zhou J, Yuan Z & & Wang L 2019 Eosinophils count in peripheral circulation is associated with coronary artery disease. Atherosclerosis 286 128134. (https://doi.org/10.1016/j.atherosclerosis.2019.05.027)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Garcia-Hernandez MH, Rodriguez-Varela E, Garcia-Jacobo RE, Hernandez-De la Torre M, Uresti-Rivera EE, Gonzalez-Amaro R & & Portales-Perez DP 2018 Frequency of regulatory B cells in adipose tissue and peripheral blood from individuals with overweight, obesity and normal-weight. Obesity Research and Clinical Practice 12 513519. (https://doi.org/10.1016/j.orcp.2018.07.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Garcia-Rubio J, Leon J, Redruello-Romero A, Pavon E, Cozar A, Tamayo F, Caba-Molina M, Salmeron J & & Carazo Á 2018 Cytometric analysis of adipose tissue reveals increments of adipocyte progenitor cells after weight loss induced by bariatric surgery. Scientific Reports 8 15203. (https://doi.org/10.1038/s41598-018-33488-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Geering B, Stoeckle C, Conus S & & Simon HU 2013 Living and dying for inflammation: neutrophils, eosinophils, basophils. Trends in Immunology 34 398409. (https://doi.org/10.1016/j.it.2013.04.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ghosh AK, Mau T, O’Brien M, Garg S & & Yung R 2016 Impaired autophagy activity is linked to elevated ER-stress and inflammation in aging adipose tissue. Aging 8 25252537. (https://doi.org/10.18632/aging.101083)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gilliet M, Cao W & & Liu YJ 2008 Plasmacytoid dendritic cells: sensing nucleic acids in viral infection and autoimmune diseases. Nature Reviews: Immunology 8 594606. (https://doi.org/10.1038/nri2358)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Goldberg EL, Shchukina I, Asher JL, Sidorov S, Artyomov MN & & Dixit VD 2020 Ketogenesis activates metabolically protective gammadelta T cells in visceral adipose tissue. Nature Metabolism 2 5061. (https://doi.org/10.1038/s42255-019-0160-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gordon S 2003 Alternative activation of macrophages. Nature Reviews: Immunology 3 2335. (https://doi.org/10.1038/nri978)

  • Gordon S & & Taylor PR 2005 Monocyte and macrophage heterogeneity. Nature Reviews: Immunology 5 953964. (https://doi.org/10.1038/nri1733)

  • Gunasekar P, Swier VJ, Fleegel JP, Boosani CS, Radwan MM & & Agrawal DK 2018 Vitamin D and macrophage polarization in epicardial adipose tissue of atherosclerotic swine. PLoS ONE 13 e0199411. (https://doi.org/10.1371/journal.pone.0199411)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guzzardi MA & & Iozzo P 2011 Fatty heart, cardiac damage, and inflammation. Review of Diabetic Studies 8 403417. (https://doi.org/10.1900/RDS.2011.8.403)

  • Hams E, Locksley RM, McKenzie AN & & Fallon PG 2013 Cutting edge: il-25 elicits innate lymphoid type 2 and type II NKT cells that regulate obesity in mice. Journal of Immunology 191 53495353. (https://doi.org/10.4049/jimmunol.1301176)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Han SF, Liu P, Zhang W, Bu L, Shen M, Li H, Fan YH, Cheng K, Cheng HX & Li CX et al.2007 The opposite-direction modulation of CD4+CD25+ Tregs and T helper 1 cells in acute coronary syndromes. Clinical Immunology 124 9097. (https://doi.org/10.1016/j.clim.2007.03.546)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Harmon DB, Srikakulapu P, Kaplan JL, Oldham SN, McSkimming C, Garmey JC, Perry HM, Kirby JL, Prohaska TA & Gonen A et al.2016 Protective role for B-1b B cells and IgM in obesity-associated inflammation, glucose intolerance, and insulin resistance. Arteriosclerosis, Thrombosis, and Vascular Biology 36 682691. (https://doi.org/10.1161/ATVBAHA.116.307166)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hashimoto D, Chow A, Noizat C, Teo P, Beasley MB, Leboeuf M, Becker CD, See P, Price J & Lucas D et al.2013 Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity 38 792804. (https://doi.org/10.1016/j.immuni.2013.04.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • He SH, Zhang HY, Zeng XN, Chen D & & Yang PC 2013 Mast cells and basophils are essential for allergies: mechanisms of allergic inflammation and a proposed procedure for diagnosis. Acta Pharmacologica Sinica 34 12701283. (https://doi.org/10.1038/aps.2013.88)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Henrichot E, Juge-Aubry CE, Pernin A, Pache JC, Velebit V, Dayer JM, Meda P, Chizzolini C & & Meier CA 2005 Production of chemokines by perivascular adipose tissue: a role in the pathogenesis of atherosclerosis? Arteriosclerosis, Thrombosis, and Vascular Biology 25 25942599. (https://doi.org/10.1161/01.ATV.0000188508.40052.35)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Henry CJ, Ornelles DA, Mitchell LM, Brzoza-Lewis KL & & Hiltbold EM 2008 IL-12 produced by dendritic cells augments CD8+ T cell activation through the production of the chemokines CCL1 and CCL17. Journal of Immunology 181 85768584. (https://doi.org/10.4049/jimmunol.181.12.8576)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hildreth AD, Ma F, Wong YY, Sun R, Pellegrini M & & O’Sullivan TE 2021 Single-cell sequencing of human white adipose tissue identifies new cell states in health and obesity. Nature Immunology 22 639653. (https://doi.org/10.1038/s41590-021-00922-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hirata Y, Tabata M, Kurobe H, Motoki T, Akaike M, Nishio C, Higashida M, Mikasa H, Nakaya Y & Takanashi S et al.2011 Coronary atherosclerosis is associated with macrophage polarization in epicardial adipose tissue. Journal of the American College of Cardiology 58 248255. (https://doi.org/10.1016/j.jacc.2011.01.048)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Horckmans M, Bianchini M, Santovito D, Megens RTA, Springael JY, Negri I, Vacca M, Di Eusanio M, Moschetta A & Weber C et al.2018 Pericardial adipose tissue regulates granulopoiesis, fibrosis, and cardiac function after myocardial infarction. Circulation 137 948960. (https://doi.org/10.1161/CIRCULATIONAHA.117.028833)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hu B, Jin C, Zeng X, Resch JM, Jedrychowski MP, Yang Z, Desai BN, Banks AS, Lowell BB & Mathis D et al.2020 2020 gammadelta T cells and adipocyte IL-17RC control fat innervation and thermogenesis. Nature 578 610614. (https://doi.org/10.1038/s41586-020-2028-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huda SS, Jordan F, Bray J, Love G, Payne R, Sattar N & Freeman DJ 2017 Visceral adipose tissue activated macrophage content and inflammatory adipokine secretion is higher in pre-eclampsia than in healthy pregnancys. Clinical Science 131 15291540. (https://doi.org/10.1042/CS20160832)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huh JY, Park J, Kim JI, Park YJ, Lee YK & Kim JB 2017 Deletion of CD1d in adipocytes aggravates adipose tissue inflammation and insulin resistance in obesity. Diabetes 66 835847. (https://doi.org/10.2337/db16-1122)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ibrahim MM 2010 Subcutaneous and visceral adipose tissue: structural and functional differences. Obesity Reviews 11 1118. (https://doi.org/10.1111/j.1467-789X.2009.00623.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ionita MG, van den Borne P, Catanzariti LM, Moll FL, de Vries JP, Pasterkamp G, Vink A & & de Kleijn DP 2010 High neutrophil numbers in human carotid atherosclerotic plaques are associated with characteristics of rupture-prone lesions. Arteriosclerosis, Thrombosis, and Vascular Biology 30 18421848. (https://doi.org/10.1161/ATVBAHA.110.209296)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Iwakura Y & & Ishigame H 2006 The IL-23/IL-17 axis in inflammation. Journal of Clinical Investigation 116 12181222. (https://doi.org/10.1172/JCI28508)

  • Jacobsen EA, Helmers RA, Lee JJ & & Lee NA 2012 The expanding role(s) of eosinophils in health and disease. Blood 120 38823890. (https://doi.org/10.1182/blood-2012-06-330845)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jaitin DA, Adlung L, Thaiss CA, Weiner A, Li B, Descamps H, Lundgren P, Bleriot C, Liu Z & Deczkowska A et al.2019 Lipid-associated macrophages control metabolic homeostasis in a Trem2-dependent manner. Cell 178 686 .e14698.e14. (https://doi.org/10.1016/j.cell.2019.05.054)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jiang E, Perrard XD, Yang D, Khan IM, Perrard JL, Smith CW, Ballantyne CM & & Wu H 2014 Essential role of CD11a in CD8+ T-cell accumulation and activation in adipose tissue. Arteriosclerosis, Thrombosis, and Vascular Biology 34 3443. (https://doi.org/10.1161/ATVBAHA.113.302077)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Joller N, Lozano E, Burkett PR, Patel B, Xiao S, Zhu C, Xia J, Tan TG, Sefik E & Yajnik V et al.2014 Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses. Immunity 40 569581. (https://doi.org/10.1016/j.immuni.2014.02.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kleindienst R, Xu Q, Willeit J, Waldenberger FR, Weimann S & & Wick G 1993 Immunology of atherosclerosis. Demonstration of heat shock protein 60 expression and T lymphocytes bearing alpha/beta or gamma/delta receptor in human atherosclerotic lesions. American Journal of Pathology 142 19271937.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Koenen TB, Stienstra R, van Tits LJ, Joosten LA, van Velzen JF, Hijmans A, Pol JA, van der Vliet JA, Netea MG & Tack CJ et al.2011 The inflammasome and caspase-1 activation: a new mechanism underlying increased inflammatory activity in human visceral adipose tissue. Endocrinology 152 37693778. (https://doi.org/10.1210/en.2010-1480)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kohlgruber AC, Gal-Oz ST, LaMarche NM, Shimazaki M, Duquette D, Koay HF, Nguyen HN, Mina AI, Paras T & Tavakkoli A et al.2018 Γδ T cells producing interleukin-17A regulate adipose regulatory T cell homeostasis and thermogenesis. Nature Immunology 19 464474. (https://doi.org/10.1038/s41590-018-0094-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kralova Lesna I, Tonar Z, Malek I, Maluskova J, Nedorost L, Pirk J, Pitha J, Lanska V & & Poledne R 2015 Is the amount of coronary perivascular fat related to atherosclerosis? Physiological Research 64 S435S443. (https://doi.org/10.33549/physiolres.933151)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kratz M, Coats BR, Hisert KB, Hagman D, Mutskov V, Peris E, Schoenfelt KQ, Kuzma JN, Larson I & Billing PS et al.2014 Metabolic dysfunction drives a mechanistically distinct proinflammatory phenotype in adipose tissue macrophages. Cell Metabolism 20 614625. (https://doi.org/10.1016/j.cmet.2014.08.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kritikou E, Kuiper J, Kovanen PT & & Bot I 2016 The impact of mast cells on cardiovascular diseases. European Journal of Pharmacology 778 103115. (https://doi.org/10.1016/j.ejphar.2015.04.050)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kumar H, Kawai T & & Akira S 2011 Pathogen recognition by the innate immune system. International Reviews of Immunology 30 1634. (https://doi.org/10.3109/08830185.2010.529976)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kyaw T, Tay C, Khan A, Dumouchel V, Cao A, To K, Kehry M, Dunn R, Agrotis A & Tipping P et al.2010 Conventional B2 B cell depletion ameliorates whereas its adoptive transfer aggravates atherosclerosis. Journal of Immunology 185 44104419. (https://doi.org/10.4049/jimmunol.1000033)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kyaw T, Winship A, Tay C, Kanellakis P, Hosseini H, Cao A, Li P, Tipping P, Bobik A & & Toh BH 2013 Cytotoxic and proinflammatory CD8+ T lymphocytes promote development of vulnerable atherosclerotic plaques in apoE-deficient mice. Circulation 127 10281039. (https://doi.org/10.1161/CIRCULATIONAHA.112.001347)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Le Menn G, Sibille B, Murdaca J, Rousseau AS, Squillace R, Vergoni B, Cormont M, Niot I, Grimaldi PA & Mothe-Satney I et al.2019 Decrease in alphabeta/gammadelta T-cell ratio is accompanied by a reduction in high-fat diet-induced weight gain, insulin resistance, and inflammation. FASEB Journal 33 25532562. (https://doi.org/10.1096/fj.201800696RR)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee BC, Kim MS, Pae M, Yamamoto Y, Eberle D, Shimada T, Kamei N, Park HS, Sasorith S & Woo JR et al.2016 Adipose natural killer cells regulate adipose tissue macrophages to promote insulin resistance in obesity. Cell Metabolism 23 685698. (https://doi.org/10.1016/j.cmet.2016.03.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee EH, Itan M, Jang J, Gu HJ, Rozenberg P, Mingler MK, Wen T, Yoon J, Park SY & Roh JY et al.2018 Eosinophils support adipocyte maturation and promote glucose tolerance in obesity. Scientific Reports 8 9894. (https://doi.org/10.1038/s41598-018-28371-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee KA, Robbins PD & & Camell CD 2021 Intersection of immunometabolism and immunosenescence during aging. Current Opinion in Pharmacology 57 107116. (https://doi.org/10.1016/j.coph.2021.01.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leik CE & & Walsh SW 2004 Neutrophils infiltrate resistance-sized vessels of subcutaneous fat in women with preeclampsia. Hypertension 44 7277. (https://doi.org/10.1161/01.HYP.0000130483.83154.37)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leslie DS, Vincent MS, Spada FM, Das H, Sugita M, Morita CT & & Brenner MB 2002 CD1-mediated gamma/delta T cell maturation of dendritic cells. Journal of Experimental Medicine 196 15751584. (https://doi.org/10.1084/jem.20021515)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li P, Lu M, Nguyen MTA, Bae EJ, Chapman J, Feng D, Hawkins M, Pessin JE, Sears DD & Nguyen AK et al.2010 Functional heterogeneity of CD11c-positive adipose tissue macrophages in diet-induced obese mice. Journal of Biological Chemistry 285 1533315345. (https://doi.org/10.1074/jbc.M110.100263)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li Y, To K, Kanellakis P, Hosseini H, Deswaerte V, Tipping P, Smyth MJ, Toh BH, Bobik A & & Kyaw T 2015 CD4+ natural killer T cells potently augment aortic root atherosclerosis by perforin- and granzyme B-dependent cytotoxicity. Circulation Research 116 245254. (https://doi.org/10.1161/CIRCRESAHA.116.304734)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li Y, Kanellakis P, Hosseini H, Cao A, Deswaerte V, Tipping P, Toh BH, Bobik A & & Kyaw T 2016 A CD1d-dependent lipid antagonist to NKT cells ameliorates atherosclerosis in ApoE-/- mice by reducing lesion necrosis and inflammation. Cardiovascular Research 109 305317. (https://doi.org/10.1093/cvr/cvv259)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lin A & & Lore K 2017 Granulocytes: new members of the antigen-presenting cell family. Frontiers in Immunology 8 1781. (https://doi.org/10.3389/fimmu.2017.01781)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lin AM, Rubin CJ, Khandpur R, Wang JY, Riblett M, Yalavarthi S, Villanueva EC, Shah P, Kaplan MJ & & Bruce AT 2011 Mast cells and neutrophils release IL-17 through extracellular trap formation in psoriasis. Journal of Immunology 187 490500. (https://doi.org/10.4049/jimmunol.1100123)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lindstedt KA, Kokkonen JO & & Kovanen PT 1992 Soluble heparin proteoglycans released from stimulated mast cells induce uptake of low density lipoproteins by macrophages via scavenger receptor-mediated phagocytosis. Journal of Lipid Research 33 6575. (https://doi.org/10.1016/S0022-2275(2041884-X)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu J, Divoux A, Sun J, Zhang J, Clement K, Glickman JN, Sukhova GK, Wolters PJ, Du J & Gorgun CZ et al.2009 Genetic deficiency and pharmacological stabilization of mast cells reduce diet-induced obesity and diabetes in mice. Nature Medicine 15 940945. (https://doi.org/10.1038/nm.1994)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liuzzo G, Kopecky SL, Frye RL, O’Fallon WM, Maseri A, Goronzy JJ & & Weyand CM 1999 Perturbation of the T-cell repertoire in patients with unstable angina. Circulation 100 21352139. (https://doi.org/10.1161/01.cir.100.21.2135)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liuzzo G, Goronzy JJ, Yang H, Kopecky SL, Holmes DR, Frye RL & & Weyand CM 2000 Monoclonal T-cell proliferation and plaque instability in acute coronary syndromes. Circulation 101 28832888. (https://doi.org/10.1161/01.cir.101.25.2883)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liuzzo G, Biasucci LM, Trotta G, Brugaletta S, Pinnelli M, Digianuario G, Rizzello V, Rebuzzi AG, Rumi C & Maseri A et al.2007 Unusual CD4+CD28null T lymphocytes and recurrence of acute coronary events. Journal of the American College of Cardiology 50 14501458. (https://doi.org/10.1016/j.jacc.2007.06.040)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lumeng CN, Bodzin JL & & Saltiel AR 2007 Obesity induces a phenotypic switch in adipose tissue macrophage polarization. Journal of Clinical Investigation 117 175184. (https://doi.org/10.1172/JCI29881)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lumeng CN, DelProposto JB, Westcott DJ & & Saltiel AR 2008 Phenotypic switching of adipose tissue macrophages with obesity is generated by spatiotemporal differences in macrophage subtypes. Diabetes 57 32393246. (https://doi.org/10.2337/db08-0872)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lynch L, O’Shea D, Winter DC, Geoghegan J, Doherty DG & & O’Farrelly C 2009 Invariant NKT cells and CD1d(+) cells amass in human omentum and are depleted in patients with cancer and obesity. European Journal of Immunology 39 18931901. (https://doi.org/10.1002/eji.200939349)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lynch L, Nowak M, Varghese B, Clark J, Hogan AE, Toxavidis V, Balk SP, O’Shea D, O’Farrelly C & & Exley MA 2012 Adipose tissue invariant NKT cells protect against diet-induced obesity and metabolic disorder through regulatory cytokine production. Immunity 37 574587. (https://doi.org/10.1016/j.immuni.2012.06.016)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Macdougall CE, Wood EG, Loschko J, Scagliotti V, Cassidy FC, Robinson ME, Feldhahn N, Castellano L, Voisin MB & Marelli-Berg F et al.2018 Visceral adipose tissue immune homeostasis is regulated by the crosstalk between adipocytes and dendritic cell subsets. Cell Metabolism 27 588 .e4601.e4. (https://doi.org/10.1016/j.cmet.2018.02.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Magalang UJ, Rajappan R, Hunter MG, Kutala VK, Kuppusamy P, Wewers MD, Marsh CB & & Parinandi NL 2006 Adiponectin inhibits superoxide generation by human neutrophils. Antioxidants and Redox Signaling 8 21792186. (https://doi.org/10.1089/ars.2006.8.2179)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mahajan VS, Mattoo H, Sun N, Viswanadham V, Yuen GJ, Allard-Chamard H, Ahmad M, Murphy SJ, Cariappa A & Tuncay Y et al.2021 B1 and B2 B cells are characterized by distinct CpG modification states at DNMT3A-maintained enhancers. Nature Communications 12,2208.(https://doi.org/10.1038/s41467-021-22458-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mancuso P & & Bouchard B 2019 The impact of aging on adipose function and adipokine synthesis. Frontiers in Endocrinology 10 137. (https://doi.org/10.3389/fendo.2019.00137)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Maniar A, Zhang X, Lin W, Gastman BR, Pauza CD, Strome SE & & Chapoval AI 2010 Human gammadelta T lymphocytes induce robust NK cell-mediated antitumor cytotoxicity through CD137 engagement. Blood 116 17261733. (https://doi.org/10.1182/blood-2009-07-234211)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A & & Locati M 2004 The chemokine system in diverse forms of macrophage activation and polarization. Trends in Immunology 25 677686. (https://doi.org/10.1016/j.it.2004.09.015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Marone G, Casolaro V, Patella V, Florio G & & Triggiani M 1997 Molecular and cellular biology of mast cells and basophils. International Archives of Allergy and Immunology 114 207217. (https://doi.org/10.1159/000237670)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Martinez FO & & Gordon S 2014 The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000prime Reports 6 13. (https://doi.org/10.12703/P6-13)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Martin-Fontecha A, Thomsen LL, Brett S, Gerard C, Lipp M, Lanzavecchia A & & Sallusto F 2004 Induced recruitment of NK cells to lymph nodes provides IFN-gamma for T(H)1 priming. Nature Immunology 5 12601265. (https://doi.org/10.1038/ni1138)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McDonnell ME, Ganley-Leal LM, Mehta A, Bigornia SJ, Mott M, Rehman Q, Farb MG, Hess DT, Joseph L & Gokce N et al.2012 B lymphocytes in human subcutaneous adipose crown-like structures. Obesity 20 13721378. (https://doi.org/10.1038/oby.2012.54)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McLaughlin T, Liu LF, Lamendola C, Shen L, Morton J, Rivas H, Winer D, Tolentino L, Choi O & Zhang H et al.2014 T-cell profile in adipose tissue is associated with insulin resistance and systemic inflammation in humans. Arteriosclerosis, Thrombosis, and Vascular Biology 34 26372643. (https://doi.org/10.1161/ATVBAHA.114.304636)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mehta P, Nuotio-Antar AM & & Smith CW 2015 Gammadelta T cells promote inflammation and insulin resistance during high fat diet-induced obesity in mice. Journal of Leukocyte Biology 97 121134. (https://doi.org/10.1189/jlb.3A0414-211RR)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Methe H, Brunner S, Wiegand D, Nabauer M, Koglin J & & Edelman ER 2005 Enhanced T-helper-1 lymphocyte activation patterns in acute coronary syndromes. Journal of the American College of Cardiology 45 19391945. (https://doi.org/10.1016/j.jacc.2005.03.040)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Metkar SS, Menaa C, Pardo J, Wang B, Wallich R, Freudenberg M, Kim S, Raja SM, Shi L & Simon MM et al.2008 Human and mouse granzyme A induce a proinflammatory cytokine response. Immunity 29 720733. (https://doi.org/10.1016/j.immuni.2008.08.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mills CD, Kincaid K, Alt JM, Heilman MJ & & Hill AM 2000 M-1/M-2 macrophages and the Th1/Th2 paradigm. Journal of Immunology 164 61666173. (https://doi.org/10.4049/jimmunol.164.12.6166)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Min SY, Desai A, Yang Z, Sharma A, DeSouza T, Genga RMJ, Kucukural A, Lifshitz LM, Nielsen S & Scheele C et al.2019 Diverse repertoire of human adipocyte subtypes develops from transcriptionally distinct mesenchymal progenitor cells. PNAS 116 1797017979. (https://doi.org/10.1073/pnas.1906512116)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Minns D, Smith KJ & & Findlay EG 2019 Orchestration of adaptive T cell responses by neutrophil granule contents. Mediators of Inflammation 2019 8968943. (https://doi.org/10.1155/2019/8968943)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mosig S, Rennert K, Krause S, Kzhyshkowska J, Neunubel K, Heller R & & Funke H 2009 Different functions of monocyte subsets in familial hypercholesterolemia: potential function of CD14+ CD16+ monocytes in detoxification of oxidized LDL. FASEB Journal 23 866874. (https://doi.org/10.1096/fj.08-118240)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moysidou M, Karaliota S, Kodela E, Salagianni M, Koutmani Y, Katsouda A, Kodella K, Tsakanikas P, Ourailidou S & Andreakos E et al.2018 CD8+ T cells in beige adipogenesis and energy homeostasis. JCI Insight 3 e95456. (https://doi.org/10.1172/jci.insight.95456)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mraz M & & Haluzik M 2014 The role of adipose tissue immune cells in obesity and low-grade inflammation. Journal of Endocrinology 222 R113R127. (https://doi.org/10.1530/JOE-14-0283)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mraz M, Cinkajzlova A, Klouckova J, Lacinova Z, Kratochvilova H, Lips M, Porizka M, Kopecky P, Lindner J & Kotulak T et al.2019 a Dendritic cells in subcutaneous and epicardial adipose tissue of subjects with type 2 diabetes, obesity, and coronary artery disease. Mediators of Inflammation 2019 5481725. (https://doi.org/10.1155/2019/5481725)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mraz M, Cinkajzlova A, Klouckova J, Lacinova Z, Kratochvilova H, Lips M, Porizka M, Kopecky P, Pierzynova A & Kucera T et al.2019 b Coronary artery disease is associated with an increased amount of T lymphocytes in human epicardial adipose tissue. Mediators of Inflammation 2019 4075086. (https://doi.org/10.1155/2019/4075086)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nascimento H, Rocha S, Rego C, Mansilha HF, Quintanilha A, Santos-Silva A & Belo L 2010 Leukocyte count versus C-reactive protein levels in obese Portuguese patients aged 6–12 years old. Open Biochemistry Journal 4 7276. (https://doi.org/10.2174/1874091X01004010072)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Niccoli G, Calvieri C, Flego D, Scalone G, Imaeva A, Sabato V, Schiavino D, Liuzzo G & & Crea F 2015 Allergic inflammation is associated with coronary instability and a worse clinical outcome after acute myocardial infarction. Circulation: Cardiovascular Interventions 8 e002554. (https://doi.org/10.1161/CIRCINTERVENTIONS.115.002554)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, Otsu M, Hara K, Ueki K & Sugiura S et al.2009 CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nature Medicine 15 914920. (https://doi.org/10.1038/nm.1964)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nishimura S, Manabe I, Takaki S, Nagasaki M, Otsu M, Yamashita H, Sugita J, Yoshimura K, Eto K & Komuro I et al.2013 Adipose natural regulatory B cells negatively control adipose tissue inflammation. Cell Metabolism 18 759766. (https://doi.org/10.1016/j.cmet.2013.09.017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nussbaum JC, Van Dyken SJ, von Moltke J, Cheng LE, Mohapatra A, Molofsky AB, Thornton EE, Krummel MF, Chawla A & Liang HE et al.2013 Type 2 innate lymphoid cells control eosinophil homeostasis. Nature 502 245248. (https://doi.org/10.1038/nature12526)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, Morel CR, Subramanian V, Mukundan L, Red Eagle A, Vats D, Brombacher F & Ferrante AW et al.2007 Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature 447 11161120. (https://doi.org/10.1038/nature05894)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Olson NC, Doyle MF, Jenny NS, Huber SA, Psaty BM, Kronmal RA & & Tracy RP 2013 Decreased naive and increased memory CD4(+) T cells are associated with subclinical atherosclerosis: the multi-ethnic study of atherosclerosis. PLoS ONE 8 e71498. (https://doi.org/10.1371/journal.pone.0071498)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • O’Sullivan TE, Rapp M, Fan X, Weizman OE, Bhardwaj P, Adams NM, Walzer T, Dannenberg AJ & & Sun JC 2016 Adipose-resident group 1 innate lymphoid cells promote obesity-associated insulin resistance. Immunity 45 428441. (https://doi.org/10.1016/j.immuni.2016.06.016)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pacifico L, Di Renzo L, Anania C, Osborn JF, Ippoliti F, Schiavo E & & Chiesa C 2006 Increased T-helper interferon-gamma-secreting cells in obese children. European Journal of Endocrinology 154 691697. (https://doi.org/10.1530/eje.1.02138)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Park YJ, Park J, Huh JY, Hwang I, Choe SS & & Kim JB 2018 Regulatory roles of invariant natural killer T cells in adipose tissue inflammation: defenders against obesity-induced metabolic complications. Frontiers in Immunology 9 1311. (https://doi.org/10.3389/fimmu.2018.01311)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Patel O, Pellicci DG, Gras S, Sandoval-Romero ML, Uldrich AP, Mallevaey T, Clarke AJ, Le Nours J, Theodossis A & Cardell SL et al.2012 Recognition of CD1d-sulfatide mediated by a type II natural killer T cell antigen receptor. Nature Immunology 13 857863. (https://doi.org/10.1038/ni.2372)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Paul VS, Paul CM & & Kuruvilla S 2016 Quantification of various inflammatory cells in advanced atherosclerotic plaques. Journal of Clinical and Diagnostic Research 10 EC35-38. (https://doi.org/10.7860/JCDR/2016/19354.7879)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pearce EL & & Pearce EJ 2013 Metabolic pathways in immune cell activation and quiescence. Immunity 38 633643. (https://doi.org/10.1016/j.immuni.2013.04.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pecht T, Gutman-Tirosh A, Bashan N & & Rudich A 2014 Peripheral blood leucocyte subclasses as potential biomarkers of adipose tissue inflammation and obesity subphenotypes in humans. Obesity Reviews 15 322337. (https://doi.org/10.1111/obr.12133)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Perez LM, Pareja-Galeano H, Sanchis-Gomar F, Emanuele E, Lucia A & & Galvez BG 2016 ‘Adipaging’: ageing and obesity share biological hallmarks related to a dysfunctional adipose tissue. Journal of Physiology 594 31873207. (https://doi.org/10.1113/JP271691)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qiu Y, Nguyen KD, Odegaard JI, Cui X, Tian X, Locksley RM, Palmiter RD & & Chawla A 2014 Eosinophils and type 2 cytokine signaling in macrophages orchestrate development of functional beige fat. Cell 157 12921308. (https://doi.org/10.1016/j.cell.2014.03.066)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Revelo XS, Tsai S, Lei H, Luck H, Ghazarian M, Tsui H, Shi SY, Schroer S, Luk CT & Lin GH et al.2015 Perforin is a novel immune regulator of obesity-related insulin resistance. Diabetes 64 90103. (https://doi.org/10.2337/db13-1524)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Reynolds CM, McGillicuddy FC, Harford KA, Finucane OM, Mills KH & & Roche HM 2012 Dietary saturated fatty acids prime the NLRP3 inflammasome via TLR4 in dendritic cells-implications for diet-induced insulin resistance. Molecular Nutrition and Food Research 56 12121222