Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Feb 28.
Published in final edited form as: Compr Physiol. 2023 Jan 30;13(1):4387–4407. doi: 10.1002/cphy.c220020

Extracellular Matrix (ECM) and Fibrosis in Adipose Tissue: Overview and Perspectives

Kai Sun 1,*, Xin Li 1, Philipp E Scherer 2,3
PMCID: PMC9957663  NIHMSID: NIHMS1872239  PMID: 36715281

Abstract

Fibrosis in adipose tissue is a major driver of obesity-related metabolic dysregulation. It is characterized by an overaccumulation of extracellular matrix (ECM) during unhealthy expansion of adipose tissue in response to over nutrition. In obese adipose-depots, hypoxia stimulates multiple pro-fibrotic signaling pathways in different cell populations, thereby inducing the overproduction of the ECM components, including collagens, noncollagenous proteins, and additional enzymatic components of ECM synthesis. As a consequence, local fibrosis develops. The result of fibrosis-induced mechanical stress not only triggers cell necrosis and inflammation locally in adipose tissue but also leads to system-wide lipotoxicity and insulin resistance. A better understanding of the mechanisms underlying the obesity-induced fibrosis will help design therapeutic approaches to reduce or reverse the pathological changes associated with obese adipose tissue. Here, we aim to summarize the major advances in the field, which include newly identified fibrotic factors, cell populations that contribute to the fibrosis in adipose tissue, as well as novel mechanisms underlying the development of fibrosis. We further discuss the potential therapeutic strategies to target fibrosis in adipose tissue for the treatment of obesity-linked metabolic diseases and cancer.

Introduction

Obesity is a severe epidemic in industrialized and developing countries (242, 263). It has been recognized as a significant risk factor for many chronic diseases, including type 2 diabetes, cardiovascular disease, hypertension, dyslipidemia, and certain types of cancer (68, 242, 263). Obesity is caused by interactions of multiple complex factors, such as overnutrition, reduced physical activity, and environmental and genetic factors. Adipose tissue is the primary organ that is, conveying the negative impact of its unhealthy expansion on the system at large (242). Adipose tissue plasticity means its ability to acquire new structural identities or adopt alternative cellular sizes and compositions in response to different nutritional conditions. In response to excessive caloric intake, adipose tissue experiences a dynamic remodeling process which puts high demands on the plasticity at adipocytes and adipose tissue. The extracellular matrix (ECM) of adipose tissue faces many challenges to accommodate the necessary dynamic changes required for expansion (49, 163, 164, 245). In parallel, the growth of blood vessels via angiogenesis cannot keep pace with the expansion. As a result, local hypoxia develops in obese adipose tissue (79, 163, 241).

Hypoxia initiates multiple pathological changes in the obese adipose tissue (242). Fibrosis is one of the major consequences caused by hypoxia (241, 242), with fibrosis being increasingly appreciated as a predominant player in adipose tissue dysfunction (49, 85, 116, 143, 197, 236, 245). Abnormal ECM accumulation during fibrosis is tightly associated with chronic low-grade inflammation in obese adipose tissue (37, 48, 71, 118, 143). Moreover, the pro-inflammatory factors and free fatty acids (FAs) released from the dysfunctional adipose tissue further circulate to other metabolically active tissues/organs, such as the liver, kidney, and muscles, thereby triggering an elevated degree of lipotoxicity in other organs (131, 258). As a result, the whole system develops insulin resistance and other metabolic disorders.

Below, we offer a brief synopsis of recent findings about the pathological process of fibrosis, underlying mechanisms that govern the whole process, as well as the resulting disorders in adipose tissue. Further, we highlight several recently identified fibrotic factors that play key roles in metabolic dysregulation during obesity. Finally, we discuss the therapeutic perspectives of targeting fibrosis in adipose tissue to treat obesity-related diseases.

ECM Components in Adipose Tissue

The ECM is a three-dimensional network that facilitates the proper structure and function of mature adipocytes, preadipocytes, and other cell populations in the stromal fraction of the adipose tissue (245). It not only provides the mechanical support but also contributes to the cell signaling pathways that are essential for adipogenesis and other proper functions of adipose tissue. The ECM is composed of a variety of highly organized protein factors, including collagens and noncollagenous proteins, multiple regulators that closely interact with ECM, and several other components, such as polysaccharides, glycoproteins, and proteoglycans as well (46). The ECM in adipose tissue is the most flexible structure that experiences dynamic remodeling during the tissue expansion in response to overnutrition (116). While it shares a lot of common components with ECM in other tissues, it also displays several unique structural features, such as the enrichment of collagen VI (Col6) (116). While some of these components only occupy a very small portion of the ECM, they play key roles in maintaining the integrity and normal function of whole adipose tissue.

Collagens

Collagens are the main component of ECM in adipose tissue (116). They occupy significant portion of the noncell mass of adipose tissue (116). Several types of collagens, including Col I, IV, V, VI, VII, VIII, and IX are an integral part of the structure of ECM of adipose tissue. They are secreted by many types of cells in the adipose tissue, including adipocytes, progenitor cells, and other components of the stromal vascular fraction (SVF) (49, 116, 162, 236, 262). Intriguingly, the collagen proteins show diverse distribution patterns in different white adipose tissue (WAT) depots (49). Among them, ColVI is the most abundant subcategory in obese adipose tissue (225). ColVI is a large glycoprotein that is, composed of three subunits—α1, α2, and α3. The three chains are assembled into hetero-tetramers and further form oligomers (116). The complex of ColVI with a high level of tertiary structure is secreted into ECM space where they further associate with other factors to form mature microfibrils that are integrated into the mature ECM (197). The levels of ColVI are tightly regulated during the diet-induced obesity development (116, 150). Clinical studies revealed that the expression levels of ColVIα3 are correlated with fat mass and total body mass during obesity (200). Our recent studies demonstrated that ColVIα3 can be digested by the metalloproteinase MMP14 (150). ColVI and its cleaved product(s) play a key role in adipocyte hypertrophy, local fibrosis and inflammation, and whole-body insulin resistance during obesity (116, 243). In particular, the carboxyterminal cleavage product of Col6 referred to as endotrophin has been found to exert a multitude of functions in adipose tissue as well as other metabolically active tissues/organs and in malignant tumors (197, 198, 244).

Noncollagen proteins

Secreted protein, acidic and rich in cysteine (SPARC)

Secreted protein, acidic and rich in cysteine (SPARC), also known as osteonectin or BM-40, is ubiquitously expressed in adipocytes and stromal cells in adipose tissue. The expression of SPARC is upregulated during obesity (28). Its protein levels are further regulated by insulin, leptin, and circulating glucose under multiple physio/pathological conditions (124). Animal studies reveal that SPARC is involved in growth and differentiation of adipocyte precursors (28). Functional studies indicate that upon being secreted into the ECM lumen, SPARC actively interacts with other factors and hence contributes to the dynamic remodeling of the network (16). Specifically, it modulates the density and the diameters of the type I collagen fibrils (19, 20). Lack of SPARC leads to increased adiposity as well as shortened collagen fibrils and impaired tensile strength of the ECM in adipose tissue (20). This pathological change has been demonstrated to directly affect the expansion capacity of the adipose tissue (20). Clinical observations reveal that the circulating levels of SPARC are elevated in obese patients and SPARC in the plasma may be involved in the progression to cardiovascular disease (12, 247, 256). Furthermore, adipose tissue-specific SPARC expression is tightly linked to obesity-related insulin resistance as well as other diabetic complications (123).

Fibronectin

Fibronectin is one of the essential ECM components in most tissues/organs. Fibronectin is expressed at a high level in mature adipose tissue and preadipocytes as well (177, 287). Intriguingly, clinical studies indicate that fibronectin levels in different adipose depots are dramatically reduced in the obese patients (141). Moreover, its levels are negatively correlated with leptin but positively associated with adiponectin (141). Numerous studies have suggested that fibronectin in adipose tissue contributes to the metabolic dysregulation during obesity. Mechanistically, its regulatory function is through its higher molecular weight oligomers that are assembled in the abundant ECM components in obese adipose tissue. Indeed, oligomerization of fibronectin is required for its cross-linking with other pericellular ECM components, such as thrombospondin (TSP) and type I collagens to stabilize the whole ECM (119, 178, 234). Intriguingly, fibronectin also serves as a key signaling molecule by functioning as a high-affinity ligand for α5β1 integrin (239). The binding of fibronectin may trigger α5β1 integrin-mediated downstream signaling response that is, involved in adipogenesis and maturation of adipocytes. Furthermore, the fibronectin-integrin pathway is also regulated by protein-protein interactions between fibronectin and the soluble protein DLK1 (also known as Pref-1) which activates integrin downstream pathway and inhibits adipocyte differentiation (267).

Thrombospondin (TSP)-1

TSP-1 is a multifunctional matrix protein in adipose tissue. TSP-1 levels are upregulated in obese adipose tissue. It binds to the other ECM molecules and stimulates the production of growth factors and cytokines, thereby triggering cellular signaling that is, involved in ECM remodeling, cell metabolism, and pro-inflammatory responses (122). Specifically, TSP-1 has been demonstrated to activate the latent transforming growth factor-β1 (TGF-β1) pathway which initiates a pro-fibrotic reaction in adipocytes (171). Moreover, its physical interaction with CD36 is involved in enhanced free FA uptake by adipocytes (77, 78). TSP-1 levels are positively correlated with adipose tissue inflammation. Increased TSP-1 further triggers an increase in local proinflammatory responses in obese adipose tissue (151, 171). Intriguingly, the effects on lipid uptake and inflammation for TSP-1 can be reversed by pioglitazone treatment, but the underlying mechanisms remain unclear (259). Loss-of-function of TSP-1 protected the mice from diet-induced inflammation and insulin resistance by reducing macrophage accumulation in adipose tissue (151). Given its positive correlation with obesity and its multifaceted function on adipose tissue remodeling and systemic metabolism, TSP-1 has been considered as a sensitive prognostic and diagnostic indicator for obesity and related type 2 diabetic sequelae (74).

Hyaluronic acid (HA)

Hyaluronic acid (HA) (also known as hyaluronan) is a heterogenous disaccharide polymer that is, an important component of the ECM in most tissues. Historically, its role in the ECM remodeling and metabolic regulation in adipose tissue has been underestimated (294). Most recently, its profound contribution to the metabolic regulation of obese adipose tissue has been better understood with the improvement of more sensitive methods for its isolation, characterization, and visualization, as well as several newly developed animal models (110, 295, 296). As an important structural component, HA actively interacts with other protein factors in the ECM. It may further bind to several receptors on the surface of adipocytes. Through these HA-induced protein-protein interactions, HA triggers multiple downstream signaling pathways that potentially affect adipogenesis as well as lipid and glucose metabolism, cell migration, angiogenesis, fibrosis, apoptosis, and proinflammatory responses (294296). Mechanistically, binding of HA to the cell surface protein CD44 promotes the proliferation of CD44+/PDGFRα+ preadipocytes, while its binding to RHAMM/HMMR receptors blunts the CD44-activated signaling and suppresses adipogenesis (5, 7, 142). Furthermore, given the critical roles of CD44 and RHAMM/HMMR receptors in the regulation of lipid and glucose metabolic pathways, it is suggested that HA is involved in the development of type-2 diabetes (121, 271).

Intriguingly, the level of inflammation is tightly correlated with the size of the HA polymers in different cells. Large HA polymers may suppress the local inflammation, while under pro-inflammatory conditions, the high-molecular-weight-HA may undergo a depolarization process and hence produce the fragments that exert pro-inflammatory functions (40, 175). In diet-induced obese adipose tissue and muscles, the content of HA is significantly increased and its levels are highly correlated with whole-body insulin resistance (110).

Elastin

Many other noncollagenous proteins, such as elastin, have also been found in the ECM of adipose tissue. However, there is limited information about their detailed function and regulation in the adipose tissue. Elastin fibers have been found to be unevenly distributed around adipocytes in different adipose depots, with a dramatic increase of the elastin network in epididymal WAT when compared with subcutaneous WAT. The differences might be caused by specific post-translational modifications of the elastin protein (3, 167). The density of the elastin network is significantly increased in the obese adipose tissue (236). This change is tightly regulated by the increased activity of the cross-linking enzyme Lox (79, 160). Interestingly, in the Col6 knockout adipose tissue, elastin levels were also decreased, suggesting a mutual regulatory effect between elastin and collagen (116). Notably, several peptide products derived from elastin have been demonstrated to induce insulin resistance in both adipose tissue and in muscles, pinpointing its critical role in systemic metabolic regulation (14). The detailed mechanisms leading to cleavage products and the respective enzymes mediating the cleavage remain to be elucidated.

Proteinases and their inhibiting factors

Matrix metalloproteinases (MMPs)

The protein components of the ECM are dynamically remodeled and the process is tightly regulated by enzymes mediating the digestion, including collagenases and other proteinases. Among all the enzymes, matrix metalloproteinases (MMPs) are a large family of proteolytic endopeptidases that are actively involved in the dynamics of ECM remodeling (29, 35, 36, 150). The MMP family is composed of more than 20 members and almost all of the MMPs are produced in an inactive form and need to be activated by other enzymes or by autodigestion (99). Of note, even though theoretically they have the ability to degrade most components in the ECM, they prefer to digest collagens (23, 99). In fact, MMPs exhibit substrate specificities when degrading the proteins during ECM remodeling (165).

Among the family of MMPs, MMP14 (also known as MT1-MMP) is the predominant membrane-bound-type MMP in adipose tissue (35, 36). MMP14 is key for the modulation of stiff pericellular collagens to allow cells to grow out of the stromal regions and is directly involved in overall ECM remodeling under both physiological and pathological conditions (35, 36, 97, 127, 150, 183, 252). It exerts its enzymatic function not only to digest collagen proteins but also to activate other MMPs, such as MMP2 and MMP9, upon tethering onto the plasma membranes of adipocytes (214, 297). During obesity development, the level of MMP14 in adipose tissue is upregulated (29, 150). A genetic variant of the human Mmp14 gene located in proximity to its catalytic domain has been found to be closely associated with obesity and diabetes traits (36). These observations suggest a direct correlation between MMP14 and obesity. Importantly, our recent findings further link MMP14 to the HIF1α-mediated pathological changes in obese adipose tissue (150). We discovered that HIF1α directly upregulates MMP14 expression by binding to its promoter region (150). Meanwhile, HIF1α induces a massive fibrotic program, prominently inducing, amongst many other collagens, ColVI in obese adipose tissue (79, 241). MMP14 cleaves the ColVIα3 chain and releases its carboxy-terminal C5 fragment (197, 243). The C5 fragment, that we refer to as endotrophin, stimulates in turn further wide-spread pro-fibrotic and pro-inflammatory responses in the tissue (see below), ultimately leading to systemic insulin resistance and impaired lipid homeostasis (150). Notably, in addition to MMP14 and other members of the MMP family, a sub-family of MMPs called a disintegrin and metalloproteinase with TSP motif (ADAMTSs) also plays crucial roles in adipose tissue development and various metabolic disease through their ECM remodeling activities (9, 10, 31, 293).

Tissue inhibitors of MMPs (TIMPs)

The counter players for MMPs, the so-called tissue inhibitors of MMPs (TIMPs), have also been demonstrated to be key for the ECM remodeling in obese adipose tissue (23, 67, 161, 183, 224). There are a total of four TIMPs in adipose tissue. Their levels are varied during adipogenesis and adipose tissue remodeling (126, 161). Particularly, TIMP1 is the most widely studied TIMP in adipose tissue. In addition to directly affecting ECM turnover via inhibiting the activity of MMPs, TIMP1 has been reported to have additional profound effects on adipose tissue (67). For example, it negatively regulates adipogenesis in obese mice and in humans (170). TIMP4 is another inhibitor for MMPs that is, enriched in adipose tissue. Based on UniGene analysis, TIMP4 is exclusively expressed in the adipose tissue in humans (165). Its levels are further elevated in response to nutritional stress (273). Lack of TIMP4 leads to reduced hypertrophy and ameliorated fibrosis in obese adipose tissue, demonstrating the effects it exerts on adipose tissue expansion (224).

Other ECM enzymes

In addition to MMPs, there are other important enzymes that function on the formation and remodeling of the ECM in adipose tissue. Among them, lysyl oxidase (LOX) is a copper-containing amine oxidase that has been identified to be expressed in adipose tissue and malignant tumors. It exists in the cytosol or is secreted into the ECM upon synthesis in adipocytes. Depending on its subcellular localization, LOX may play multiple functions in the physiology/pathology of tissues (39). Particularly in the ECM, LOX post-translationally modifies collagens and noncollagen proteins, such as elastin, thereby catalyzing the covalent cross-linking of the fibers formed by the proteins (39, 108). This cross-linking process is essential for the stabilization, elasticity, and flexibility of fibrils and fibers when being integrated into the ECM (39). The levels and function of LOX are highly regulated in obese adipose tissue. We first found that in diet-induced obese adipose tissue, HIF1α upregulates the expression of LOX and the higher LOX levels facilitate the overaccumulation of ECM in the obese adipose tissue (79). In line with this finding, inhibition of LOX significantly reduced the level of local fibrosis (79, 202). The function of another cross-linking enzyme referred to as factor XIII-A (FXIII-A) on adipose tissue remodeling has also drawn attention recently (182). Recent observations demonstrated that FXIII-A in WAT has a causative relationship with obesity in humans (106, 107). In preadipocytes, FXIII-A translocates to the surface of cells where it promotes the assembly of fibronectin to form the ECM surrounding the preadipocytes. As a result, FXIII-A negatively regulates adipogenesis by reducing the proliferation and differentiation of the preadipocytes through remodeling of ECM at the cell surface (182).

Abnormal ECM Remodeling and Fibrosis

The flexibility of the ECM in adipose tissue is illustrated by its ability to accommodate the rapid expansion or shrinking of the tissue in response to a differential nutritional status (219). However, we appreciate that the WAT cannot expand without limits, both in animal models and in humans (27, 70, 260). There exists a threshold for the expansion that keeps the whole tissue in a normal functional range. Beyond this threshold, the extremely stiff ECM gives rise to profound pathological alterations, including the development of local fibrosis (245). Meanwhile, the ECM-laden adipocytes lose their plasticity and undergo pathological changes, including cellular inflammation, ER stress, and apoptosis (218, 242). There are multiple steps that eventually lead to the abnormal ECM remodeling (Figure 1). Hypoxia has been recognized as a key initiating step; fibrosis and its associated inflammation are both pathological consequences not the initiating factors for the further development and progression of a myriad of the obesity-induced metabolic diseases (62, 64, 79, 92, 125, 136, 139).

Figure 1.

Figure 1

Working model for the development of fibrosis and inflammation in obese adipose tissue. In lean adipose tissue, the adipocytes are small and healthy. Adequate blood vessels formed by proper angiogenesis provide oxygen, hormones, nutrients, and adipocyte precursors to support the healthy expansion of the tissue (A); during diet-induced obesity, adipose tissue expands rapidly through hyperplasia and hypertrophy. Meanwhile, new blood vessel formation cannot keep up with the expansion, the adipocytes become larger, and local hypoxia thus develops (B); At the late phase of obesity, hypoxia stimulates massive fibrosis. The mechanical stress induced by the overdeveloped ECM leads to necrosis of the adipocytes. As a result, macrophages are accumulated and polarized to the M1 subtype in the tissue. They form “crown-like” structures in obese adipose depots. The local fibrosis and inflammation further lead to the whole-body insulin resistance (C). Of note, not all the adipose tissue expansion has “unhealthy” consequences. In addition to the calorie excess, genetic variants and environmental factors also have profound effects on the expansion.

Hypoxia: the initiating step

Hypoxia is a challenge that any tissue may face when the local oxygen pressure (pO2) is decreased. Well-known hypoxic conditions persist in the central region of solid tumors. Indeed, hypoxia has been recognized as the major driver for malignant tumor progression in most types of cancer (52, 227). Adipose tissue is the only nontransformed tissue in the body that has the ability to expand over the course of developing obesity to an almost unlimited extent, just like a tumor mass. However, the formation of blood vessels via angiogenesis and vasculogenesis cannot keep pace (79, 246). Indeed, the blood flow rates in an obese fat pad are 30% to 40% lower compared to lean adipose tissue (15). As a result, local hypoxia develops (1, 241).

Detection of hypoxia in obese adipose tissue

The adipocytes are rather large cells and their size can easily excess 200μm in diameter in obese individuals (79, 233). However, the effective diffusion rates of O2 are much less than 200μm in tissue (21). This unique feature of adipocytes further worsens the local hypoxic environment. To detect hypoxia quantitatively in adipose tissue, several approaches have been developed by our group and others, including pimonidazole staining, pO2 tracking in situ, and hypoxia-induced factor (HIF) activity measurements. Experimental results with these techniques have clearly demonstrated that a higher level of hypoxia persists in obese adipose tissue of genetic and diet-induced obese animal models and obese humans (92, 201, 212, 279, 280). Specifically, by tracking the local pO2 with an oxygen sensor using an electron paramagnetic resonance (EPR) system, we revealed that the pO2 in the epidydimal WAT is dynamically decreased during diet-induced obesity (241). Intriguingly, compared to the levels measured in mouse models, the hypoxia level is relative minor in obese human fat tissue (201, 253). However, a recent clinical study argues that highly significant hypoxic conditions exist in severely obese patients (253). More clinical studies with larger populations of obese individuals may be necessary to better define the hypoxic state in obese adipose tissue.

Hypoxia-induced factors (HIFs) and their regulation

Hypoxia exerts profound effects on adipose tissue, ultimately leading to insulin resistance (242). The cells in adipose tissue respond to low oxygen conditions by activating multiple transcriptional factors, such as HIFs, CREB family members, and NF-κB (255). Among these factors, HIF1 has been demonstrated to function as a “Master Regulator” (1, 79, 241). HIF1 is an essential transcription factor that plays a fundamental role in oxygen homeostasis in almost all tissues and in cancer (228). HIF1 contains two subunits, HIF1α and β and they form a basic helix-loop-helix structure when binding to the cis-acting HIF response element (HRE) to exert their function on transcriptional activation/suppression (228). Our groups have utilized adipose-specific gain- and loss-of-function mouse models to study the role of HIF1α in the obesity-associated pathogenesis (79, 241). We have reported that HIF1α plays a pivotal role in shaping the unhealthy microenvironment in obese adipose tissue. During obesity, HIF1 is massively upregulated at both the microRNA (mRNA) and protein level in adipocytes (79, 241). Intriguingly, HIF1 fails to upregulate a typical target gene, VEGF-A in adipocytes, resulting in the lack of angiogenesis in the largely expanded adipose tissue (79, 241). Instead, it triggers a massive “fibrosis program” by upregulating collagen proteins and ECM remodeling enzymes, such as MMP14 and LOX (79, 150, 202, 241). LOX can cross-link lysyl residues in collagens and in elastin. This cross-linking reaction may stabilize the collagens to form the building blocks for the ECM (88, 216). Overexpression of LOX enhances the fibrotic streaks by acting to cross-link the accumulated Col1 and 3 to form the fibrillar collagen fibers (282).

HIF1 is also induced in other cell populations, such as the M1-like polarized macrophages and preadipocytes in obese adipose tissue (208, 231, 232). The function of HIF1α in the macrophages is to drive low-grade inflammation by upregulating IL-1β (208, 232). Interestingly, a recent study demonstrated that inhibition of HIFα in PDGFRβ+ preadipocytes facilitates adipogenesis and healthy expansion of different adipose depots during obesity (231).

The levels and activity of HIF1 are dynamically regulated by multiple signals associated with obesity, such as local oxygen tension, ANT2, insulin, and adipogenesis (65, 84, 229). Intriguingly, MMP14 has been shown to affect HIF1α transcriptional activity by physically retaining its suppressor, factor inhibiting HIF-1α−1 (FIH-1) in the cytoplasm in a subset of cancer cells (159, 204, 205, 221, 223). FIH-1 is an asparaginyl hydroxylase that targets and hence hydroxylates the Asn-803 residue in HIF1α. The hydroxylation (Asn803-OH) significantly blocks the recruitment of P300/CBP to HIF1α, which in turn impairs its overall transcriptional activity (100, 134, 159, 217, 264). The cytosolic region of MMP14 (but not of any other MMPs) interacts with FIH-1, thereby preventing its translocation into the nucleus, and hence allowing HIF1α to evade the suppression by FIH-1 (222). Nevertheless, the importance of FIH-1 in MMP14-mediated HIF1α activation has not yet been directly evaluated in obese adipose tissue. Given that the loss-of-function study of FIH-1 has revealed a profound impact on adipose tissue metabolism (284), we reason that it is an integral component of HIF1α regulation in adipose tissue, mediated by MMP14.

This leads us to propose that HIF1 induction represents an early event during obesity development, while it is a critical step in the sequential processes of obesity-related pathological changes including fibrosis and inflammation.

HIF2α, a protein related to HIF1α, is also broadly expressed in adipocytes and macrophages in adipose tissue (34, 64, 69, 153). It also plays multifaceted roles in metabolism, physiology, and pathology of adipose tissue. Even though it shares similar target genes with HIF1α, it also has some unique functions which are, in some instances, opposite to those of HIF1α (113). Of note, studies have highlighted the important role of HIF2α in protecting adipocytes from dysfunction, predominantly through its proangiogenic actions by upregulating VEGF-A in obese adipose tissue (61, 64, 174, 253). Further insights need to be gained to better understand the mechanistic details of the interactions between HIF1α and HIF2α.

Abnormal ECM formation driven by hypoxia

Fibrosis in adipose tissue is caused by the disproportionate accumulation of ECM proteins. During the process of fibrosis, excessive amounts of ECM proteins are produced, while their degradation is reduced. The overarching pathophysiological role of the ECM is driven by the obesity-induced hypoxic conditions, but the detailed events and underlying mechanisms remain to be further clarified (242, 245). Of note, even though there is an established link between obesity and fibrosis in rodent models, clinical observations reveal that not all obese individuals develop local fibrosis in adipose tissue, suggesting fibrosis is a pathological process that is, controlled by other factors aside from obesity per se, including environmental and genetic factors [reviewed in Sun et al. (242)].

Regulation of collagens in obese adipose tissue

Collagen proteins are massively upregulated in hypoxic adipose tissue (4, 42, 79, 149, 156, 241). Gene profiling data from the WAT of HIF1α transgenic mice show a widespread induction of fibrotic genes. They include multiple types of collagens, such as Col1, 3, 5, 6, and 8 (95). Specifically, Col6 is one of the most abundant collagens and it plays an essential role in shaping dysfunctional ECM (116). Col6 levels are further increased in the obese adipose tissue (116, 225). Clinically, it has been found that the levels of Col6, especially its α3 subunit, strongly correlate with the degree of hypoxia in adipose tissue (136, 200, 201). The excessive accumulation of Col6 may disrupt the normal structure of the ECM and cause increased stiffness of the ECM scaffold. This creates a mechanical stress in the rapidly expanding adipose tissue (245). To support this notion, lack of Col6 results in reduced rigidity which facilitates the expansion of the adipose tissue in both diet-induced obese and ob/ob mice (116). As a result, the lack of Col6 leads to improved metabolic profiles (49, 116).

Regulation of ECM enzymes during obesity

The formation and turnover of the ECM are dynamically regulated by different enzymes that act on ECM factors. One of the key enzymes is LOX. As mentioned above, LOX cross-links collagens and elastin and hence facilitates the formation of the ECM (4, 39, 160, 202). The function of LOX is tightly regulated by hypoxia in obese adipose tissue. Indeed, LOX is a direct transcriptional target for HIF1 induction (79). Furthermore, LOX expression has also been shown to be closely correlated with elevated Col1 expression during the development of adipose tissue (277). Importantly, inhibition of LOX activity not only ameliorates fibrosis but also improves local inflammation and several metabolic parameters (79). Clinical results have confirmed that LOX levels were significantly increased in obese adipose tissue and weight-loss surgery attenuated its expression levels (94, 202).

On the other hand, the ECM digesting enzymes in adipose tissue are also dynamically regulated during obesity. Particularly, MMP14 expression and protein levels are significantly increased in obese adipose tissue (36, 127, 150). We recently demonstrated that HIF1α binds to the promoter regions of MMP14 and hence upregulates its expression (150). Intriguingly, the activated MMP14 exerts dichotomous effects on ECM remodeling depending on the metabolic status: At the early stages during obesity, MMP14 upregulated by HIF1α induction turns over collagen proteins and hence release the mechanical stress on the enlarged adipocytes. In that context, MMP14 brings about metabolically beneficial effects for the adipose tissue expansion; On the other hand, at the later stages of obesity, MMP14 digests Col6 and produces endotrophin, which stimulates local pro-fibrotic and pro-inflammatory reactions in obese adipose tissue, thereby worsening the metabolically unhealthy microenvironment in the tissue. As a direct result, the mice exhibit metabolic dysregulation and insulin resistance (150, 290). Other MMPs and their endogenous inhibitors, the TIMPs, are also dramatically changed during obesity. Specifically, the expression levels of MMP2, 3, 12, 14, and 19 as well as TIMP1 are dramatically upregulated, while MMP7 and TIMP3 were downregulated during obesity in the mice (2, 29, 126, 161, 170, 224). The studies further indicated that the activities of MMP3 and MMP12 are enhanced in the obese adipose tissue (29, 273). TIMP4 levels and activity are also increased in the high fat-diet (HFD)-fed mice while suppression of TIMP4 has been shown to protect the mice from obesity-induced fibrosis in adipose tissue (224, 273). All these observations suggest that there is a finely tuned balance between collagen digesting enzymes and their inhibitors in obese adipose tissue (29). Of note, the dynamic changes of MMPs also exert a profound impact on adipogenesis, angiogenesis, as well as inflammation in adipose tissue [reviewed by Ruiz-Ojeda et al. (218)]. Clinical observations reveal that the levels of MMP7, MMP9, and TIMP1 correlate well with fat mass during obesity. Particularly, MMP9 levels are increased in the insulin-resistant individuals and in the patients with higher body mass index (BMI) as well (2, 136).

Fibrosis and angiogenesis

Healthy expansion of adipose tissue requires proper formation and proliferation of new blood vessels via angiogenesis and vasculogenesis. The functional blood vessels provide nutrients, hormones, growth factors, and stem cells for maintaining the homeostasis of the adipose tissue (24, 25, 195, 242). Pro-angiogenic factors, such as VEGF-A and VEGF receptors 1 and 2, Angiopoietin receptors, and NOX2 are upregulated by HIFs in most tissues, including in the liver, kidney, and tumor tissue (26, 47, 269, 276). In these tissues, the stimulated angiogenesis has been demonstrated to be closely associated with ECM remodeling and the level of fibrosis (51). In that context, targeting angiogenesis has been considered to be an efficient way to reduce local fibrosis (45, 51). However, in obese adipose tissue, HIF1α fails to upregulate VEGF-A for formation of adequate new blood vessels to keep the pace with the expansion of adipose tissue (79). This deficiency further exacerbates the local hypoxic state in the obese adipose tissue (79). Lack of adequate angiogenesis is a unique feature of the obese adipose tissue and the vasculatures may not contribute to the development of fibrosis in the tissue. The underlying mechanisms governing this phenomenon are not yet completely understood.

Fibrosis and inflammation

Fibrosis induces inflammation

The enhanced stiffness during the development of fibrosis causes mechanical stress to the ECM-laden adipocytes, which eventually leads to a pro-inflammatory response in the tissue (242). The detailed molecular events governing the mechanical stress on adipocytes are not completely understood. Several cellular pathways, such as RhoA and NF-κB signals, have been reported to be involved in the whole process. Briefly, RhoA is activated by mechanical shear stress and the increased size of adipocytes (81). Activation of RhoA signaling pathways reduces PPARγ transcriptional activity on adipogenesis, which affects the recruitment of newer adipocytes for healthy expansion (101, 168). Moreover, RhoA and its downstream RhoA kinase activation stimulate multiple pro-inflammatory cytokines, including plasminogen activator inhibitor-1 (PAI-1) and mast cell protease-1 (MCP-1), which trigger local inflammation (148, 184). Enhanced ECM density in adipose tissue also activates the NF-κB pathway, which plays a central role in shaping the inflammatory environment by initiating a pro-inflammatory cascade (8). Specifically, the activated NF-κB induces activation of monocytes and blunts metabolic signaling in obese adipocytes (148).

We and others have observed the fibrosis-induced inflammation in adipose tissue in both diet-induced and genetically obese mouse models. In fibrotic obese adipose tissue, adipocytes experience a significant enlargement through hypertrophy (163, 179, 243). We demonstrated that large lipid droplets rapidly lose their surface covering proteins, such as perilipin-1, upon cell death in adipocytes surrounded by the abnormally high levels of ECM (246) (Figure 1). The dead adipocytes carrying large lipid droplets attract massive infiltration of macrophages, and the accumulated macrophages surrounding the lipid droplets form a typical “crown-like” structure, a characteristic feature of inflammation in the tissue (180). The infiltrated macrophages, upon disposing lipids from the lipid droplets, may polarize into M1-like pro-inflammatory subtypes. They further induce chronic mild inflammation which has been recognized as the root cause of obesity-related insulin resistance and other metabolic disorders (135, 189). Particularly, this phenomenon has been well characterized in our “FAT-ATTAC” (FAT Apoptosis Through Targeted Activation of Caspase 8) mouse model, in which the adipocytes are induced to undergo synchronized apoptosis via Caspase 8 activation (157, 193, 257). In this model, the adipocytes quickly undergo cell death within 2 days upon induction of Caspase 8, while the lipid droplets remain in the original regions for many weeks to form “ghost fat cells.” Meanwhile, the number of “crown-like” structures is significantly increased, reflecting the increased local inflammation (59, 157, 193). Of note, even though the “FAT-ATTAC” mouse model mimics adipocyte death which attracts macrophage accumulation in the obese adipose tissue, it may not fully recapitulate all of the pathological changes associated with obesity. Based on related observations by us and others, we have built a working model in which inflammation happens in the later stages following local hypoxia and fibrosis in adipose tissue (Figure 1).

Inflammation exacerbates fibrosis

Another set of observations support a different working model in which hypoxia may trigger local inflammation before the development of fibrosis. In this model, fibrosis is induced by inflammation [reviewed in Debari and Abbott (46)]. Hypoxia induces infiltration of macrophages and other immune cells in obese adipose tissue (93, 270). Abnormal metabolic signaling, in part due to an increase in free FAs and prolonged exacerbated circulating glucose levels, promotes activation of immune cells. The activated immune cells produce pro-inflammatory cytokines and initiate different levels of inflammation. Particularly, the M1-like macrophages secrete IL-6, TNFα, and several other cytokines that lead to a chronic low-grade inflammatory response (93, 111, 268, 270). Moreover, the innate T cells also exhibit functional abnormalities that further contribute to the overall inflamed state (50, 209). The chronic inflammation offers additional mechanisms for the development of local fibrosis in the obese adipose tissue (245). The pro-fibrotic program is coordinated by a variety of activated or polarized innate and adaptive immune cells (158).

Notably, even though we have two models describing the relationship between fibrosis and inflammation in the obese adipose tissue, they are not contradictory to each other. Instead, the two major pathological changes may promote each other and hence coordinate to shape the unhealthy microenvironment in obese adipose tissue. In the future, with the development of novel tools applied in vitro and in vivo, it will hopefully be possible to better define which is the initial mechanism in response to the stimulation of hypoxia.

Key cellular components involved in fibrosis-induced inflammation

The proinflammatory microenvironment in the fibrotic adipose tissue is shaped by a broad spectrum of inflammatory factors, adipokines, lipid species, and exosomal mRNAs that are produced by different cells (60, 80, 192, 194). Among them, macrophages have been recognized as a major contributor for the factors [reviewed in Sun et al. (242)]. Macrophages infiltrate into obese adipose tissue, polarize into M1-like subtypes and hence secrete pro-inflammatory cytokines, such as TNFα, IL-1β, and IL-6. The cytokines not only exert local effects in the adipose tissue but also circulate into other metabolically active organs, thereby affecting the local inflammatory and metabolic state, ultimately leading to the development of metabolic complications in the whole system (75, 213, 270, 275).

Recently, other immune cells have been appreciated to play critical roles in local inflammation as well as insulin resistance in the obese adipose tissue (169, 181). In the past years, many types of immune cells have been reported to infiltrate unhealthy obese adipose tissue [reviewed in Lackey and Olefsky (133)]. They include both innate and adaptive immune cells, such as T cells, B cells, NK and NKT cells, dendritic cells, mast cells, and neutrophils cells [reviewed in Osborn and Olefsky (191); Ferrante (57)]. Among them, neutrophils belong to innate immune cells (109). Even though their overall numbers are relatively small, they are among the first immune cells that are recruited by adipose tissue where they exert prolonged pro-inflammatory effects via secreting TNFα, MCP-1, and elastase up to 3 months in response to HFD (32, 41, 248, 266). Mast cells also belong to innate immune cells and adipose tissue is a major site of residence of mast cells. Diet-induced obesity dramatically increases the total number of mast cells in the tissue (155, 283). During obesity, mast cells experience a degranulation process which promotes the secretion of multiple pro-inflammatory factors, thereby facilitating a chronic low-grade pro-inflammation microenvironment in the obese adipose tissue (155, 169, 283). T cells and B cells are lymphocytes. Normally they contribute up to 10% of nonadipocytes in adipose tissue (169). Specifically, B cells are actively recruited into obese adipose tissue where they promote the accumulation of other immune cells, including the M1-like macrophages and the T cells (144, 272). T cells represent the second largest population, aside from macrophages in the obese adipose tissue (144). T cells have two subtypes, named CD4-positive and CD8-positive T cells, respectively. CD8-positive T cells are associated with metabolically unhealthy outcomes in obese adipose tissue (120). CD4-positive cells are further subclassified into several groups: pro-inflammatory T helper (Th1) and Th17 cells, anti-inflammatory Th2 cells, and T regulatory (Treg) cells (188). Among them, the Treg cells play a key role in maintaining healthy anti-inflammatory state in lean adipose tissue. Previous studies have demonstrated that decreased numbers of Treg cells are associated with higher level of local inflammation in obese adipose tissue, which might further lead to whole-body insulin resistance (58, 292). More recent research further revealed that other memory T cells increase in the diet-induced obese adipose tissue, leading to severe pathological changes including enhanced lipase activity and calcification of the whole tissue (176).

Even though each immune cell exerts its own function in the inflammatory response, the interplay between them is essential for their contributions to the chronic local inflammation in obese adipose tissue (173). For example, both B cells and T cells, as well as the interplay between them, critically influence the M1-like macrophage infiltration in obese adipose tissue. Moreover, the CD8-positive T cells promote macrophage differentiation and enhance their chemotaxis, thereby leading to their accumulation in the obese adipose tissue (185). Based on previous findings, it is well accepted that infiltrating M1-like macrophages are the end effectors and orchestrate functional communication among all immune cells [reviewed in Lee et al. (144)].

Other pathological consequences of fibrosis

Fibrosis also leads to other effects on adipose tissue. The mechanical stress on the fibrotic adipocytes may enhance de novo lipogenesis and lipolysis in the cells, which further induces the formation of the abnormally large lipid droplets (46, 128, 129, 150, 243, 288). The enlarged lipid droplets caused by fibrosis in adipocytes may further induce the ectopic deposition of lipid in other metabolic tissues, an effect known as lipotoxicity (150, 243). Fibrosis links directly to adipogenesis in adipose tissue. Indeed, ECM factors have been appreciated to play an important role in adipogenesis (6, 38, 140, 289). Consistent with that, an abnormal function of MMP14 during fibrosis has been reported to affect adipogenesis (36, 150). Moreover, ECM proteins upregulated by TGF-β1 or PAI-1 cause the impaired differentiation of adipocytes in in vitro studies (17, 152). Finally, more recent reports argue that the pathologically upregulated HIF1α also drives abnormal adipogenesis in the obese adipose tissue (231).

Nonclassical Fibrotic Factors: Endotrophin as an Example

A number of studies have identified numerous novel factors that potently stimulate fibrosis in adipose tissue (197). Members of these nonclassical “hormone-like” molecules include endostatin, endotrophin, asprosin, and platencin, all of which derive from cleavage events under multiple cellular stress situations (250). Among them, endotrophin has been highlighted to be a potent pro-fibrotic and pro-inflammatory small molecule that triggers profound pathological changes in different tissues and certain types of cancer (226). Endotrophin is the proteolytic product of Col6 α3 chain in the adipose tissue (56, 89, 138, 197199, 211, 215, 243, 244). Its levels are significantly increased in obese and diabetic mice and in humans (243). Endotrophin can be released into circulation and accumulate in other metabolic tissues (Figure 2). Recently, extensive studies have demonstrated that endotrophin has potent bioactivity, stimulating massive fibrosis and inflammation locally in metabolically active tissues, including in adipose tissue, heart, liver, and kidney (54, 56, 112, 117, 138). It is also enriched in certain types of cancer lesions and serves as a driver of malignant tumor growth (22, 196, 197, 199). Particularly in obese adipose tissue, endotrophin is a powerful driving factor for local fibrosis, macrophage infiltration, and other metabolic unfavorable consequences, including lipotoxicity and insulin resistance (243, 290). In line with its function as a necessary and sufficient factor responsible for the diet-induced pathological changes, treatment with an anti-endotrophin neutralizing antibody significantly reverses the metabolically adverse effects induced by HFD in the obese mice (243). Endotrophin has divergent functions on different cell populations in obese adipose tissue (290). However, the details regarding the signaling pathway(s) that endotrophin triggers in cells remain to be further elucidated.

Figure 2.

Figure 2

MMP14 digests Col6 and produce endotrophin. During diet-induced obesity, local hypoxia induces HIF1α in adipose tissue. As a direct target of HIF1α, MMP14 is upregulated. Meanwhile, HIF1α also upregulates Col6 expression. MMP14 digests Col6α3 chain and produces endotrophin. Accumulation of endotrophin further shapes unhealthy microenvironment locally in the adipose tissue via triggering massive fibrosis and inflammation. The local pathological changes ultimately lead to systemic insulin resistance and other metabolic disorders.

While numerous studies have demonstrated endotrophin to be a sensitive biomarker of local fibrosis and inflammation and have highlighted it as a key regulator in adipose tissue dysfunction, insulin resistance, and cancer development, the mechanistic details of the cleavage event, including the identity of the key processing enzyme, were unknown for a long period of time. In that context, we recently made a major breakthrough by finding that MMP14 cleaves Col6 α3 to release endotrophin (150). The cleavage region is located at a consensus-cleavage site for MMP14 (Figure 2). The resulting fragment has been confirmed using an endotrophin-specific antibody (150). We further found that the levels and activity of MMP14 are increased during obesity (150). Aside from MMP14, several other MMPs induced by hypoxia, including MMP2, MMP9, and MMP16 in adipose tissue have recently been reported to cleave Col VI and produce endotrophin (or endotrophin-like molecules) (104).

Importantly, endotrophin is also detected at high levels in many other tissues. Recent reports have demonstrated that endotrophin is a sensitive biomarker for local fibrosis and inflammation in many diseases (Table 1). For example, endotrophin levels in urine correlate with local fibrosis, tubular atrophy, and monocyte infiltration in lupus nephritis patients (66). Studies also established a strong association between the pretransplant plasma levels of endotrophin and the risk of the delayed graft function after kidney transplantation, pinpointing it as a new marker for the prediction of the effects of the transplantation (250). Intriguingly, the levels of endotrophin are significantly higher in the females with polycystic ovary syndrome (PCOS), while other adipose-derived hormones, such as adiponectin and ghrelin, displayed no changes in the patients, suggesting it may also serve as a unique biomarker for the diagnosis of PCOS (72). The circulating levels of endotrophin are also tightly associated with many fibrotic diseases, such as fibrotic interstitial lung disease (ILD) (44). Moreover, the levels of endotrophin in metabolically active tissues, such as adipose tissue, the liver, and the tumor tissues, are also highly correlated with many metabolic diseases, such as obesity and related diabetes, cardiovascular disease, kidney disease, and cancer (summarized in Table 1).

Table 1.

The Correlation Between Levels of Endotrophin and Diseases

Disease Correlation Tissue/Serum/Plasma References

Obesity and diabetes Positive Adipose tissue Sun et al. (243)
Renal fibrosis Positive Kidney Rasmussen et al. (211)
Type 2 diabetes Positive Serum Karsdal et al. (112)
Chronic kidney disease Positive Serum Fenton et al. (56)
Breast cancer Positive Plasma Bu et al. (22)
Heart failure Positive Serum Eruzun et al. (54)
Hepatocellular carcinoma Positive Tumor-neighboring regions Lee et al. (138)
Cirrhotic patients with hepatocellular carcinoma Positive Plasma Leeming et al. (145)
Obese children Positive Plasma Ezzati-Mobaser et al. (55)
Chronic obstructive pulmonary disease (COPD) Positive Serum Rønnow et al. (215)
Non-alcoholic fatty liver disease Positive Serum Kim et al. (117)
Atherosclerotic Positive Atherosclerotic plaque Holm Nielsen et al. (91)
Acute-on-chronic liver failure Positive Plasma Kerbert et al. (115)
Nonalcoholic steatohepatitis Positive Serum Hagström et al. (76)
Chronic multimorbidity Positive Serum Staunstrup et al. (238)
Polycystic ovary syndrome (PCOS) Positive Serum Guney et al. (72)
Crohn’s disease Positive Serum Lindholm et al. (154)
Diabetic nephropathy Positive Serum Yoldemir et al. (281)
Chronic kidney disease Positive Serum Sparding et al. (235)
Kidney fibrosis Positive Serum Genovese et al. (66)
Fibrotic interstitial lung diseases Positive Serum Dasdemir Ilkhan et al. (44)
Kidney transplantation Negative Pretransplant plasma endotrophin Tepel et al. (250)

In summary, endotrophin has drawn a significant attention recently due to its direct link to many vital diseases. It has been demonstrated to have potent bioactivity to trigger local pro-fibrotic and pro-inflammatory reactions and hence systemic metabolic disorders. Its local or circulating levels are highly correlated with the development of these diseases. Therefore, endotrophin bears a great potential to serve as a sensitive biomarker and be targeted to treat pathological aspects of these diseases.

Cellular Regulation of Fibrosis

While it is clear that the root cause of fibrosis in adipose tissue is obesity, the detailed mechanisms governing the development of fibrosis orchestrated by multiple cell populations remain to be further clarified. Furthermore, many other factors, including genetic variants and environmental factors, may profoundly affect the pathological changes during the development of fibrosis. In obese adipose tissue, the large lipid-laden mature adipocytes are surrounded by various cell types which are collectively named SVF (86, 203). The SVF is composed of endothelial cells, preadipocytes, adipose-derived stem cells (ASCs), pericytes, fibroblasts, macrophages, B cells, T cells, and other types of innate immune cells (86, 87). Different types of cells respond to the hypoxia condition during obesity and hence contribute to the fibrotic development respectively (Figure 3) (86, 87, 290). Recently, the development of single-cell or single-nucleus RNA sequencing (specifically for adipocytes) provides a powerful tool to characterize the divergent roles of the subpopulations of adipose tissue in ECM formation and fibrosis (33, 172, 261).

Figure 3.

Figure 3

The divergent function of different cell populations on fibrosis in obese adipose tissue. The fibrotic program is coordinated by multiple cell types in adipose tissue, including adipocytes, macrophages, endothelial cells, ACS, Mast cells and fibroblasts, etc. The cells secrete collagens and non-collagenous proteins, pro-inflammatory factors, ECM enzymes, and multiple unidentified factors which work together to fine-tune the level of fibrosis in response to different cell stimuli. Moreover, the cells interplay with each other and regulate their pro-fibrotic function through the cell-cell communication.

Adipocytes

As the major type of cells in the adipose tissue, adipocytes are embedded in the dense ECM of the adipose depots. Diet-induced obesity induces the upregulation of pro-fibrotic genes in adipocytes (105). A recent transcriptome analysis of adipocytes isolated from diet-induced obese visceral WAT revealed that the adipocytes are switched to a “fibroblast-like” phenotype in lieu of the obese adipose tissue (105). Among all the ECM proteins, Col6 is highly enriched in the adipocytes (116). Metabolically challenged adipocytes express even higher levels of Col6, which has been considered to be a hallmark of adipose tissue fibrosis (116). Even though adipocytes have low metabolic rates and a relatively low demand for the oxygen, they are quite sensitive to hypoxic conditions (241). HIFs are induced in adipocytes in response to diet-induced obesity (1). By using both gain-of-function and loss-of-function genetic tools, we have revealed that HIF1 exerts unique functions on local fibrosis in the obese adipose tissue (79, 241). Particularly, LOX is directly upregulated by HIF1 in the obese adipocytes (4, 79, 208). LOX promotes over-accumulation of ECM by cross-linking collagens and elastin to form the oligomer structures for the collagen and noncollagen fibers (79). Furthermore, HIF1 directly binds to the MMP14 promoter region, thereby activating their transcription (150). The upregulated MMP14 catalyzes the digestion of Col6 and hence produce endotrophin, which triggers massive fibrosis in the unhealthy microenvironments in the obese adipose tissue (150). Recently, we found that inhibition of lipid catabolic enzymes, such as carboxylesterase 1d (CES1) in adipocytes, may induce the upregulation of pro-fibrotic genes including Col3α1, Col6α, and LOX, suggesting the potential role of lipid signaling in fibrosis/inflammation in obese adipocytes (147). On the other hand, the level of fibrosis in the obese adipocytes is downregulated by the PRDM16-containing transcriptional complex, which is mediated by a TFII-I family protein called GTF2IRD1 in a cell-autonomous manner (82). Mechanistically, GTF2IRD1 suppresses the expression level of TGF-β-dependent genes through the recruitment of the PRDM-16 complex (82). Intriguingly, some transmembrane glycoproteins, such as CD248 and decorin, exert their deleterious effects through triggering the local fibrosis in obese adipocytes (43, 206).

Endothelial cells

Endothelial cells represent the most dynamic composition in the SVF and they line up both large and macro vasculature in the adipose tissue. Endothelial cells play a key role in the vascular remodeling of adipose tissue during obesity. Even though at the early stage of obesity, VEGF-A-mediated endothelial cell activation brings about metabolically beneficial effects by counteracting the local hypoxia development via angiogenesis, it eventually leads to exacerbated fibrosis, inflammation, and insulin resistance in the established obese adipose tissue (246). In unhealthy adipose tissue at the later stages of obesity, endothelial cells may form a vascular niche with other cell populations, such as the pericytes, macrophages, and hematopoietic stem cells. In this niche, endothelial cells crosstalk with other cell types, including adipocytes, immune cells, and fibroblasts, thereby contributing to the local fibrosis and inflammation through the so-called “angiocrine pathway” (291). Interestingly, in a bleomycin-induced lung fibrosis model and a cardiac fibrosis model, the endothelial cells transformed to fibroblasts through endothelial-mesenchymal transition (EMT), suggesting that endothelial cells might serve as sources of fibrotic cells under pathological conditions (83, 286). Endothelial cells also recruit macrophages during lung injury (137). Even though the endothelial cells and the vascular niche that they form have been studied in other fibrotic models, it needs to be further examined as to whether they exert similar function on fibrosis in adipose tissue.

Macrophages

The function of macrophages on fibrosis has been well established in many tissues/organs (186, 187, 249, 274). In particular, inflammatory monocytes reside in obese adipose tissue and hence accumulate as mature macrophages in situ. They are the major immune cells that initiate local fibrosis and inflammation, which ultimately leads to systemic insulin resistance (210, 237). Macrophage crosstalk with adipocytes via secreting TNF-α and FFAs which aggravate local inflammation in obese adipose tissue (53, 132, 240). The alternatively activated macrophages have been reported to contribute to tissue fibrosis by stimulating the activation of fibroblasts and the formation of the ECM (63). The function is through multiple pathways and includes: (i) They secrete several pro-fibrotic cytokines including IGF1, CCL17, CCL22, and CTGF; (ii) They overexpress TGF-β which further triggers the downstream pro-fibrotic signaling pathways (166); (iii) They stimulate STAT6 signaling pathways (102, 103); (iv) Finally, the modulation of arginase activity in the M2 macrophages promotes fibrogenesis by regulating the production of collagens (251). Of note, recent work revealed that senescent macrophages accumulating in diet-induced obese adipose tissue promote fibrosis in lieu of the unhealthy microenvironment (210). On the other hand, distinct types of macrophages digest the ECM and hence prevent the development of fibrosis. For example, the classical activating macrophages secret TSLP, MMP2, MMP9, and MMP12, and the enzymes exert their fibrolytic function to release the stiffness of the ECM (146, 167, 220). Therefore, the macrophages play dichotomous roles in fibrosis and their functions are their polarization dependent.

Adipose tissue-derived stem cells (ASCs)

ASCs are dynamically regulated by different metabolic states (86, 87, 231). Particularly, the ASCs in the obese adipose tissue exhibit higher PDGFRα-positive population (98, 162). The progenitors are prone to differentiate to the ECM-synthesizing pro-fibrotic cells in the obese adipose tissue (98). Specifically, a subset of the PDGFα-positive progenitors with high expression of CD9 differentiate into pro-fibrotic cells which directly drive the pathological changes of fibrosis (162). A recent study further revealed that both PDGFα-and β-positive progenitors contribute essentially to the local ECM development in the diet-induced obese adipose tissue (210). Interplays between ASCs and other cell populations also contribute to the ECM remodeling.

Fibroblasts

Fibroblasts are a common cell type in the SVF of the adipose tissue. They provide an important niche for the adipogenesis and the whole tissue homeostasis (87, 231, 285). Fibroblasts are the major source of the ECM and serve as a central regulator for the dynamics of ECM remodeling and pathological fibrosis (114). Particularly, the αSMA-positive myofibroblasts have been recognized to produce the stiff ECM fibers, thereby initiating the kidney, liver, and lung fibrosis (254). In particular, fibroblast-specific protein-1 (FSP-1)-positive fibroblasts play a key role in the ECM remodeling and the whole tissue cellular regulation (285). However, the precise functions of fibroblasts in adipose tissue remain to be further clarified. Moreover, the origins of the myofibroblasts remain to be further defined (164).

Mast cells

Mast cells are a type of immune cell that exist with a large number in connective tissues including adipose tissue (207). In response to different cell stimuli, mast cells secrete histamine, hormones, and cytokines that promote allergic reactions and inflammation (130). Mature mast cells are present in obese adipose tissue in db/db mice where they exert pro-fibrotic function through secretion of MCP-6 (90). Clinically, mast cells are abundant in the subcutaneous WAT in the patients with metabolic syndrome (73). In these patients, the number of mast cells is correlated with increased fibrosis and the local proinflammatory state (73). Moreover, the numbers are also correlated with diabetic parameters, such as insulin resistance (48). While it has been appreciated that mast cell accumulation in the obese adipose tissue accelerates the process of fibrosis and systemic metabolic dysregulation, the details of the whole process remain to be further characterized. Of note, while we identify the roles of each cell type in fibrosis (Figure 3), their functions are tightly regulated by cell-cell communication. The level of fibrosis is fine-tuned by the interplay between them (164). For example, the accrual of macrophages is highly controlled by the perivascular mesenchymal cells (230).

Therapeutic Perspectives on Targeting Fibrosis in Adipose Tissue

Given the severe local and systemic pathological consequences caused by adipose tissue fibrosis, such as loss of the adipose plasticity, increased local inflammation, impaired insulin sensitivity, and the poor prognosis of bariatric surgery (46), targeting fibrosis has become an ideal strategy to combat obesity and related metabolic diseases and cancer (46). Furthermore, the level of fibrosis has been quantitatively scored clinically and the scores have been demonstrated to be reversely correlated with the body-weight loss after the gastric bypass surgery, highlighting its significance in the diagnosis and prognosis of obesity-related dysregulations (11). Unfortunately, no direct therapies to block or reverse adipose tissue fibrosis have been developed to date (163).

HIF1α initiates the whole pathological process of fibrosis and inflammation in the established obese adipose tissue. Therefore, targeting HIF1α might be an efficient way to suppress hypoxia-induced pathological changes. In that context, we tested the effect of a HIF1-specific inhibitor, PX-478 on reversing fibrosis in the diet-induced obese mice. Indeed, we found that PX-478 efficiently suppressed the local pro-fibrotic and pro-inflammatory reactions in the adipose tissue, thereby improving the whole-body metabolism (241). Further probing the efficacy of the PX-478 and other HIF1 inhibitors in obesity and related diseases warrants further studies. Hydroxylase domain (PHD) targets and destabilizes HIF1 (13). Inhibitors for PHDs, which can increase HIF1 expression, such as GSK1278863 and FG-4592, have been in clinical trials to treat kidney anemia. Aside from the observed effect on the kidney disease, the inhibitors also showed ability to lower circulating cholesterol levels, providing new insights into clinical implications for lipid dysregulations induced by obesity (30, 190). However, implication of these agents to directly target adipose tissue fibrosis has yet been examined.

The TGF-β pathway plays a central role in pro-fibrotic reaction during obesity. It is reasonable to design therapeutic strategies to target TGF-β and its downstream signals to block the development of fibrosis, though this is highly challenging. In agreement with the notion, berberine, a natural plant product originally known to exert its antidiabetic effect via stimulating the activation of AMPK (96), has been shown to decrease TGF-β mediated Smad3 phosphorylation, thereby attenuating collagen accumulation and reversing the upregulated fibrotic genes in the diet-induced adipose tissue (265).

Endotrophin is one of the promising targets for consideration. As a molecule that is, produced during the pathological expansion of adipose depots, endotrophin shapes an unhealthy microenvironment in adipose tissue and other metabolically active organs and tumors through triggering local fibrosis and macrophage accumulation in the tissues. It not only serves as a sensitive biomarker for the disease but also provides an ideal target for treatment. Indeed, blockage of bioactivity of endotrophin significantly reduced/reversed local fibrosis in the obese adipose tissue and in the tumors in mice (196, 197, 243). Importantly, we recently developed endotrophin-specific neutralizing antibodies which inhibit tumor growth by blocking the bioactivity of endotrophin (22).

MMP14 has been highlighted to be key for the ECM remodeling during obesity. Findings from us and others suggest the possible reversibility of adipose tissue fibrosis by inhibiting MMP14 (36, 43, 127, 150). Therefore, MMP14 bears a great promise from a therapeutic perspective for obesity and type 2 diabetes. Another key ECM remodeling enzyme, LOX in adipose tissue has been extensively studied in different diseases and it has been considered to be an attractive target for the therapeutic intervention to treat fibrotic diseases [reviewed in Yang et al. (278)].

Conclusion

In the past, obesity-induced fibrosis in adipose tissue has been extensively investigated in vitro by 3-D culture, ex vivo by high-resolution magnetic resonance imaging (MRI), as well as in vivo in different animal models and humans (4, 11, 18, 46, 49, 79, 85, 86, 116, 118, 124, 150, 163, 241, 243, 245, 290). It has been well established that fibrosis developed in obese adipose tissue causes severe pathological changes, including adipocyte necrosis, impaired adipogenesis, metabolic disorders, and inflammation locally in the adipose tissue, ultimately leading to systemic lipotoxicity and insulin resistance (245). Previous findings have demonstrated that different cell populations in obese adipose tissue contribute individually to the development of fibrosis. The interplay between these cells finetunes the whole pathological process. Many additional findings further highlight the central role of hypoxia in the pro-fibrotic and pro-inflammatory reaction in obese adipose tissue (79, 241). While HIF1 stimulates both fibrosis and inflammation, the causal-effect relationship between the two pathological changes remains to be further defined with proper models. Moreover, the detailed mechanisms underlying the overdevelopment of ECM resulting in fibrosis need to be further characterized in depth.

It has been proposed that fibrosis can be reduced or reversed to treat obesity and related metabolic diseases and cancer. Several key fibrotic pathways, such as TGFβ and its downstream signaling, the HIF1-MMP14-endotrophin axis, and HIF1-PHD signaling have been considered to be potential targets to ameliorate fibrosis and related pathological changes. Excitingly, at least in vitro or in preclinical models, they have been demonstrated to be effective to improve metabolic disorders, including dyslipidemia, insulin resistance, and tumor growth (22, 150, 241, 243). In the future, well-designed clinical studies will hopefully validate this therapeutic relevance of these pathways in patients. Of note, even though fibrosis has been demonstrated to be the core constituent of unhealthy microenvironment that further induces local inflammation and other pathological changes in the obese adipose tissue, metabolic dysfunction can be caused by many other profound factors, including nutritional and genetic factors. Therefore, anti-fibrotic therapeutics themselves may be insufficient to reverse the metabolic disorders. In that context, combination of the anti-fibrotic therapeutics with other interventions, such as reduced energy intake, regular physical activity, anti-inflammatory interventions, and/or metabolic (bariatric) surgery bear great promise to synergistically treat obesity and type-2 diabetes.

Didactic Synopsis.

Major teaching points

  • Obesity is caused by interactions of multiple complex factors, such as overnutrition, reduced physical activity, the environmental, and genetic factors.

  • Hypoxia initiates multiple pathological changes in the obese adipose tissue.

    • Fibrosis is one of the major consequences caused byhypoxia.

    • Abnormal ECM accumulation during fibrosis is tightly associated with chronic low-grade inflammation in the obese adipose tissue.

    • The pro-inflammatory factors and free fatty acids released from the dysfunctional adipose tissue further circulate to other metabolically active tissues/organs, thereby triggering an elevated degree of lipotoxicity in the other organs.

  • Fibrosis in adipose tissue is a major driver of obesity-related metabolic dysregulation.

  • The enhanced stiffness during the development of fibrosis causes the mechanical stress to the ECM-laden adipocytes, which eventually leads to necrosis of adipocytes and a pro-inflammatory reaction response in the tissue.

  • The chronic inflammation is an important trigger for the development of the local fibrosis in the obese adipose tissue.

  • The level of fibrosis is fine-tuned by the interplays between multiple cell types in obese adipose tissue.

  • Targeting fibrosis has become a viable strategy to combat obesity and related metabolic diseases and cancer.

Acknowledgements

The authors are grateful to colleagues at UTH and UTSW for the critical discussions during the preparation of the review. We would like to thank the funding supports from NIH/NIDDK (R01-DK109001, R56-DK124419, and R01-DK129815 to Kai Sun; R01DK131537, R01-DK55758, R01-DK099110, RC2-DK118620, R01-DK127274, and R01-DK131537 to Philipp E. Scherer) and the Welsh Foundation Scholar (for Kai Sun). We apologize for omission of any relevant references due to page limitations.

Footnotes

Conflicts of Interest

The authors declare that no competing interests exist.

Related Articles

Contribution of Adipose Tissue Inflammation to the Development of Type 2 Diabetes Mellitus

Extracellular Matrix: A Solid–State Regulator of Cell form, Function, and Tissue Development

References

  • 1.AbdelMassih A, Yacoub E, Husseiny RJ, Kamel A, Hozaien R, El Shershaby M, Rajab M, Yacoub S, Eid MA, Elahmady M, Gadalla M, Mokhtar S, Hassan AA, Abou-Zeid AS, Hussein M, Aboushadi N, Emad N, Zahra N, Hassan A, Hussein E, Ibrahim N, El Nahhas N, Elahmady T, Khallaf M, Mustafa H, Anis N, Albehairy M, Hanna F, Moris L, Ye J. Hypoxia-inducible factor (HIF): The link between obesity and COVID-19. Obes Med 22: 100317, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Akra S, Aksnes TA, Flaa A, Eggesbo HB, Opstad TB, Njerve IU, Seljeflot I. Markers of remodeling in subcutaneous adipose tissue are strongly associated with overweight and insulin sensitivity in healthy non-obese men. Sci Rep 10: 14055, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Alkhouli N, Mansfield J, Green E, Bell J, Knight B, Liversedge N, Tham JC, Welbourn R, Shore AC, Kos K, Winlove CP. The mechanical properties of human adipose tissues and their relationships to the structure and composition of the extracellular matrix. Am J Physiol Endocrinol Metab 305: E1427–E1435, 2013. [DOI] [PubMed] [Google Scholar]
  • 4.Anvari G, Bellas E. Hypoxia induces stress fiber formation in adipocytes in the early stage of obesity. Sci Rep 11: 21473, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Aruffo A, Stamenkovic I, Melnick M, Underhill CB, Seed B. CD44 is the principal cell surface receptor for hyaluronate. Cell 61: 1303–1313, 1990. [DOI] [PubMed] [Google Scholar]
  • 6.Attur M, Lu C, Zhang X, Han T, Alexandre C, Valacca C, Zheng S, Meikle S, Dabovic BB, Tassone E, Yang Q, Kolupaeva V, Yakar S, Abramson S, Mignatti P. Membrane-type 1 matrix metalloproteinase modulates tissue homeostasis by a non-proteolytic mechanism. iScience 23: 101789, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bahrami SB, Tolg C, Peart T, Symonette C, Veiseh M, Umoh JU, Holdsworth DW, McCarthy JB, Luyt LG, Bissell MJ, Yazdani A, Turley EA. Receptor for hyaluronan mediated motility (RHAMM/HMMR) is a novel target for promoting subcutaneous adipogenesis. Integr Biol (Camb) 9: 223–237, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Baker RG, Hayden MS, Ghosh S. NF-kappaB, inflammation, and metabolic disease. Cell Metab 13: 11–22, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bauters D, Bedossa P, Lijnen HR, Hemmeryckx B. Functional role of ADAMTS5 in adiposity and metabolic health. PLoS One 13: e0190595, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bauters D, Cobbaut M, Geys L, Van Lint J, Hemmeryckx B, Lijnen HR. Loss of ADAMTS5 enhances brown adipose tissue mass and promotes browning of white adipose tissue via CREB signaling. Mol. Metab. 6: 715–724, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bel Lassen P, Charlotte F, Liu Y, Bedossa P, Le Naour G, Tordjman J, Poitou C, Bouillot JL, Genser L, Zucker JD, Sokolovska N, Aron-Wisnewsky J, Clement K. The FAT score, a fibrosis score of adipose tissue: Predicting weight-loss outcome after gastric bypass. J Clin Endocrinol Metab 102: 2443–2453, 2017. [DOI] [PubMed] [Google Scholar]
  • 12.Berezin AE, Kremzer AA. Predictive value of circulating osteonectin in patients with ischemic symptomatic chronic heart failure. Biomed J 38: 523–530, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Berra E, Benizri E, Ginouves A, Volmat V, Roux D, Pouyssegur J. HIF prolyl-hydroxylase 2 is the key oxygen sensor setting low steady-state levels of HIF-1alpha in normoxia. EMBO J 22: 4082–4090, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Blaise S, Romier B, Kawecki C, Ghirardi M, Rabenoelina F, Baud S, Duca L, Maurice P, Heinz A, Schmelzer CE, Tarpin M, Martiny L, Garbar C, Dauchez M, Debelle L, Durlach V. Elastin-derived peptides are new regulators of insulin resistance development in mice. Diabetes 62: 3807–3816, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bolinder J, Kerckhoffs DA, Moberg E, Hagstrom-Toft E, Arner P. Rates of skeletal muscle and adipose tissue glycerol release in nonobese and obese subjects. Diabetes 49: 797–802, 2000. [DOI] [PubMed] [Google Scholar]
  • 16.Bornstein P, Sage EH. Matricellular proteins: Extracellular modulators of cell function. Curr Opin Cell Biol 14: 608–616, 2002. [DOI] [PubMed] [Google Scholar]
  • 17.Bortell R, Owen TA, Ignotz R, Stein GS, Stein JL. TGF beta 1 prevents the down-regulation of type I procollagen, fibronectin, and TGF beta 1 gene expression associated with 3T3-L1 pre-adipocyte differentiation. J Cell Biochem 54: 256–263, 1994. [DOI] [PubMed] [Google Scholar]
  • 18.Bouazizi K, Zarai M, Marquet F, Aron-Wisnewsky J, Clement K, Redheuil A, Kachenoura N. Adipose tissue fibrosis assessed by high resolution ex vivo MRI as a hallmark of tissue alteration in morbid obesity. Quant Imaging Med Surg 11: 2162–2168, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bradshaw AD, Graves DC, Motamed K, Sage EH. SPARC-null mice exhibit increased adiposity without significant differences in overall body weight. Proc Natl Acad Sci U S A 100: 6045–6050, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bradshaw AD, Puolakkainen P, Dasgupta J, Davidson JM, Wight TN, Helene Sage E. SPARC-null mice display abnormalities in the dermis characterized by decreased collagen fibril diameter and reduced tensile strength. J Invest Dermatol 120: 949–955, 2003. [DOI] [PubMed] [Google Scholar]
  • 21.Brahimi-Horn MC, Pouyssegur J. Oxygen, a source of life and stress. FEBS Lett 581: 3582–3591, 2007. [DOI] [PubMed] [Google Scholar]
  • 22.Bu D, Crewe C, Kusminski CM, Gordillo R, Ghaben AL, Kim M, Park J, Deng H, Xiong W, Liu XZ, Lonning PE, Halberg N, Rios A, Chang Y, Gonzalez A, Zhang N, An Z, Scherer PE. Human endotrophin as a driver of malignant tumor growth. JCI Insight 5: e125094, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Cabral-Pacheco GA, Garza-Veloz I, Castruita-De la Rosa C, Ramirez-Acuna JM, Perez-Romero BA, Guerrero-Rodriguez JF, Martinez-Avila N, Martinez-Fierro ML. The roles of matrix metalloproteinases and their inhibitors in human diseases. Int J Mol Sci 21: 9739, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cao Y Angiogenesis modulates adipogenesis and obesity. J Clin Invest 117: 2362–2368, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Angiogenesis Cao Y. and vascular functions in modulation of obesity, adipose metabolism, and insulin sensitivity. Cell Metab 18 (4): 478–489, 2013. [DOI] [PubMed] [Google Scholar]
  • 26.Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature 473: 298–307, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Carobbio S, Pellegrinelli V, Vidal-Puig A. Adipose tissue function and expandability as determinants of lipotoxicity and the metabolic syndrome. Adv Exp Med Biol 960: 161–196, 2017. [DOI] [PubMed] [Google Scholar]
  • 28.Chavey C, Boucher J, Monthouel-Kartmann MN, Sage EH, Castan-Laurell I, Valet P, Tartare-Deckert S, Van Obberghen E. Regulation of secreted protein acidic and rich in cysteine during adipose conversion and adipose tissue hyperplasia. Obesity (Silver Spring) 14: 1890–1897, 2006. [DOI] [PubMed] [Google Scholar]
  • 29.Chavey C, Mari B, Monthouel MN, Bonnafous S, Anglard P, Van Obberghen E, Tartare-Deckert S. Matrix metalloproteinases are differentially expressed in adipose tissue during obesity and modulate adipocyte differentiation. J Biol Chem 278: 11888–11896, 2003. [DOI] [PubMed] [Google Scholar]
  • 30.Chen N, Qian J, Chen J, Yu X, Mei C, Hao C, Jiang G, Lin H, Zhang X, Zuo L, He Q, Fu P, Li X, Ni D, Hemmerich S, Liu C, Szczech L, Besarab A, Neff TB, Peony Yu KH, Valone FH. Phase 2 studies of oral hypoxia-inducible factor prolyl hydroxylase inhibitor FG-4592 for treatment of anemia in China. Nephrol Dial Transplant 32: 1373–1386, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Chen SZ, Ning LF, Xu X, Jiang WY, Xing C, Jia WP, Chen XL, Tang QQ, Huang HY. The miR-181d-regulated metalloproteinase Adamts1 enzymatically impairs adipogenesis via ECM remodeling. Cell Death Differ 23: 1778–1791, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Chmelar J, Chung KJ, Chavakis T. The role of innate immune cells in obese adipose tissue inflammation and development of insulin resistance. Thromb Haemost 109: 399–406, 2013. [DOI] [PubMed] [Google Scholar]
  • 33.Cho DS, Lee B, Doles JD. Refining the adipose progenitor cell landscape in healthy and obese visceral adipose tissue using single-cell gene expression profiling. Life Sci Alliance 2: e201900561, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Choe SS, Shin KC, Ka S, Lee YK, Chun JS, Kim JB. Macrophage HIF-2alpha ameliorates adipose tissue inflammation and insulin resistance in obesity. Diabetes 63: 3359–3371, 2014. [DOI] [PubMed] [Google Scholar]
  • 35.Chun TH, Hotary KB, Sabeh F, Saltiel AR, Allen ED, Weiss SJ. A pericellular collagenase directs the 3-dimensional development of white adipose tissue. Cell 125: 577–591, 2006. [DOI] [PubMed] [Google Scholar]
  • 36.Chun TH, Inoue M, Morisaki H, Yamanaka I, Miyamoto Y, Okamura T, Sato-Kusubata K, Weiss SJ. Genetic link between obesity and MMP14-dependent adipogenic collagen turnover. Diabetes 59: 2484–2494, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, Wang S, Fortier M, Greenberg AS, Obin MS. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res 46: 2347–2355, 2005. [DOI] [PubMed] [Google Scholar]
  • 38.Croissandeau G, Chretien M, Mbikay M. Involvement of matrix metalloproteinases in the adipose conversion of 3T3-L1 preadipocytes. Biochem J 364: 739–746, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Csiszar K Lysyl oxidases: A novel multifunctional amine oxidase family. Prog Nucleic Acid Res Mol Biol 70: 1–32, 2001. [DOI] [PubMed] [Google Scholar]
  • 40.Dahl LB, Dahl IM, Engstrom-Laurent A, Granath K. Concentration and molecular weight of sodium hyaluronate in synovial fluid from patients with rheumatoid arthritis and other arthropathies. Ann Rheum Dis 44: 817–822, 1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Dam V, Sikder T, Santosa S. From neutrophils to macrophages: Differences in regional adipose tissue depots. Obes Rev 17: 1–17, 2016. [DOI] [PubMed] [Google Scholar]
  • 42.Dankel SN, Grytten E, Bjune JI, Nielsen HJ, Dietrich A, Bluher M, Sagen JV, Mellgren G. COL6A3 expression in adipose tissue cells is associated with levels of the homeobox transcription factor PRRX1. Sci Rep 10: 20164, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Daquinag AC, Gao Z, Fussell C, Sun K, Kolonin MG. Glycosaminoglycan modification of decorin depends on MMP14 activity and regulates collagen assembly. Cell 9: 2646, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Dasdemir Ilkhan G, Demirci Ucsular F, Celikhisar H, Arman Y, Yalniz E, Tukek T. Original article: Clinical research. Sarcoidosis Vasc Diffuse Lung Dis 38: e2021020, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.de Heer EC, Jalving M, Harris AL. HIFs, angiogenesis, and metabolism: Elusive enemies in breast cancer. J Clin Invest 130: 5074–5087, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.DeBari MK, Abbott RD. Adipose tissue fibrosis: Mechanisms, models, and importance. Int J Mol Sci 21: 6030, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Diebold I, Petry A, Sabrane K, Djordjevic T, Hess J, Gorlach A. The HIF1 target gene NOX2 promotes angiogenesis through urotensin-II. J Cell Sci 125: 956–964, 2012. [DOI] [PubMed] [Google Scholar]
  • 48.Divoux A, Moutel S, Poitou C, Lacasa D, Veyrie N, Aissat A, Arock M, Guerre-Millo M, Clement K. Mast cells in human adipose tissue: Link with morbid obesity, inflammatory status, and diabetes. J Clin Endocrinol Metab 97: E1677–E1685, 2012. [DOI] [PubMed] [Google Scholar]
  • 49.Divoux A, Tordjman J, Lacasa D, Veyrie N, Hugol D, Aissat A, Basdevant A, Guerre-Millo M, Poitou C, Zucker JD, Bedossa P, Clement K. Fibrosis in human adipose tissue: Composition, distribution, and link with lipid metabolism and fat mass loss. Diabetes 59: 2817–2825, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Eljaafari A, Pestel J, Le Magueresse-Battistoni B, Chanon S, Watson J, Robert M, Disse E, Vidal H. Adipose-tissue-derived mesenchymal stem cells mediate PD-L1 overexpression in the white adipose tissue of obese individuals, resulting in T cell dysfunction. Cell 10: 2645, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Elpek GO. Angiogenesis and liver fibrosis. World J Hepatol 7: 377–391, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Emami Nejad A, Najafgholian S, Rostami A, Sistani A, Shojaeifar S, Esparvarinha M, Nedaeinia R, Haghjooy Javanmard S, Taherian M, Ahmadlou M, Salehi R, Sadeghi B, Manian M. The role of hypoxia in the tumor microenvironment and development of cancer stem cell: A novel approach to developing treatment. Cancer Cell Int 21: 62, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Engin AB. Adipocyte-macrophage cross-talk in obesity. Adv Exp Med Biol 960: 327–343, 2017. [DOI] [PubMed] [Google Scholar]
  • 54.Eruzun H, Toprak ID, Arman Y, Yilmaz U, Ozcan M, Kutlu Y, Irmak S, Kutlu O, Yoldemir SA, Altun O, Cil EO, Tukek T. Serum endotrophin levels in patients with heart failure with reduced and mid-range ejection fraction. Eur J Intern Med 64: 29–32, 2019. [DOI] [PubMed] [Google Scholar]
  • 55.Ezzati-Mobaser S, Malekpour-Dehkordi Z, Nourbakhsh M, Tavakoli-Yaraki M, Ahmadpour F, Golpour P, Nourbakhsh M. The up-regulation of markers of adipose tissue fibrosis by visfatin in pre-adipocytes as well as obese children and adolescents. Cytokine 134: 155193, 2020. [DOI] [PubMed] [Google Scholar]
  • 56.Fenton A, Jesky MD, Ferro CJ, Sorensen J, Karsdal MA, Cockwell P, Genovese F. Serum endotrophin, a type VI collagen cleavage product, is associated with increased mortality in chronic kidney disease. PLoS One 12: e0175200, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ferrante AW Jr. The immune cells in adipose tissue. Diabetes Obes Metab 15 (Suppl 3): 34–38, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, Lee J, Goldfine AB, Benoist C, Shoelson S, Mathis D. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med 15: 930–939, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Fischer-Posovszky P, Wang QA, Asterholm IW, Rutkowski JM, Scherer PE. Targeted deletion of adipocytes by apoptosis leads to adipose tissue recruitment of alternatively activated M2 macrophages. Endocrinology 152: 3074–3081, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Flaherty SE 3rd, Grijalva A, Xu X, Ables E, Nomani A, Ferrante AW Jr. A lipase-independent pathway of lipid release and immune modulation by adipocytes. Science 363: 989–993, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Fraisl P, Mazzone M, Schmidt T, Carmeliet P. Regulation of angiogenesis by oxygen and metabolism. Dev Cell 16: 167–179, 2009. [DOI] [PubMed] [Google Scholar]
  • 62.Fujisaka S, Usui I, Ikutani M, Aminuddin A, Takikawa A, Tsuneyama K, Mahmood A, Goda N, Nagai Y, Takatsu K, Tobe K. Adipose tissue hypoxia induces inflammatory M1 polarity of macrophages in an HIF-1alpha-dependent and HIF-1alpha-independent manner in obese mice. Diabetologia 56: 1403–1412, 2013. [DOI] [PubMed] [Google Scholar]
  • 63.Galli SJ, Borregaard N, Wynn TA. Phenotypic and functional plasticity of cells of innate immunity: Macrophages, mast cells and neutrophils. Nat Immunol 12: 1035–1044, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Garcia-Martin R, Alexaki VI, Qin N, Rubin de Celis MF, Economopoulou M, Ziogas A, Gercken B, Kotlabova K, Phieler J, Ehrhart-Bornstein M, Bornstein SR, Eisenhofer G, Breier G, Bluher M, Hampe J, El-Armouche A, Chatzigeorgiou A, Chung KJ, Chavakis T. Adipocyte-specific hypoxia-inducible factor 2alpha deficiency exacerbates obesity-induced brown adipose tissue dysfunction and metabolic dysregulation. Mol Cell Biol 36: 376–393, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Gaspar JM, Velloso LA. Hypoxia inducible factor as a central regulator of metabolism—implications for the development of obesity. Front Neurosci 12: 813, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Genovese F, Akhgar A, Lim SS, Farris AB, Battle M, Cobb J, Sinibaldi D, Karsdal MA, White WI. Collagen type III and VI remodeling biomarkers are associated with kidney fibrosis in lupus nephritis. Kidney360 2: 1473–1481, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Gerin I, Louis GW, Zhang X, Prestwich TC, Kumar TR, Myers MG Jr, Macdougald OA, Nothnick WB. Hyperphagia and obesity in female mice lacking tissue inhibitor of metalloproteinase-1. Endocrinology 150: 1697–1704, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ghaben AL, Scherer PE. Adipogenesis and metabolic health. Nat Rev Mol Cell Biol 20: 242–258, 2019. [DOI] [PubMed] [Google Scholar]
  • 69.Giaccia AJ. HIF-2: The missing link between obesity and cardiomyopathy. J Am Heart Assoc 2: e000710, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Gliniak CM, Scherer PE. A new signal that shrinks fat. Nat Metab 4: 305–307, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol 29: 415–445, 2011. [DOI] [PubMed] [Google Scholar]
  • 72.Guney G, Taskin MI, Baykan O, Adali E, Gul Tezcan S, Sarikaya S, Kaya C, Tolu E. Endotrophin as a novel marker in PCOS and its relation with other adipokines and metabolic parameters: A pilot study. Ther Adv Endocrinol Metab 12: 20420188211049607, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Gurung P, Moussa K, Adams-Huet B, Devaraj S, Jialal I. Increased mast cell abundance in adipose tissue of metabolic syndrome: Relevance to the proinflammatory state and increased adipose tissue fibrosis. Am J Physiol Endocrinol Metab 316: E504–E509, 2019. [DOI] [PubMed] [Google Scholar]
  • 74.Gutierrez LS, Gutierrez J. Thrombospondin 1 in metabolic diseases. Front Endocrinol (Lausanne) 12: 638536, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Haase J, Weyer U, Immig K, Kloting N, Bluher M, Eilers J, Bechmann I, Gericke M. Local proliferation of macrophages in adipose tissue during obesity-induced inflammation. Diabetologia 57: 562–571, 2014. [DOI] [PubMed] [Google Scholar]
  • 76.Hagström H, Bu D, Nasr P, Ekstedt M, Hegmar H, Kechagias S, Zhang N, An Z, Stal P, Scherer PE. Serum levels of endotrophin are associated with nonalcoholic steatohepatitis. Scand J Gastroenterol 56: 437–442, 2021. [DOI] [PubMed] [Google Scholar]
  • 77.Hajri T, Hall AM, Jensen DR, Pietka TA, Drover VA, Tao H, Eckel R, Abumrad NA. CD36-facilitated fatty acid uptake inhibits leptin production and signaling in adipose tissue. Diabetes 56: 1872–1880, 2007. [DOI] [PubMed] [Google Scholar]
  • 78.Hajri T, Han XX, Bonen A, Abumrad NA. Defective fatty acid uptake modulates insulin responsiveness and metabolic responses to diet in CD36-null mice. J Clin Invest 109: 1381–1389, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Halberg N, Khan T, Trujillo ME, Wernstedt-Asterholm I, Attie AD, Sherwani S, Wang ZV, Landskroner-Eiger S, Dineen S, Magalang UJ, Brekken RA, Scherer PE. Hypoxia-inducible factor 1alpha induces fibrosis and insulin resistance in white adipose tissue. Mol Cell Biol 29: 4467–4483, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Han MS, White A, Perry RJ, Camporez JP, Hidalgo J, Shulman GI, Davis RJ. Regulation of adipose tissue inflammation by interleukin 6. Proc Natl Acad Sci U S A 117: 2751–2760, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Hara Y, Wakino S, Tanabe Y, Saito M, Tokuyama H, Washida N, Tatematsu S, Yoshioka K, Homma K, Hasegawa K, Minakuchi H, Fujimura K, Hosoya K, Hayashi K, Nakayama K, Itoh H. Rho and Rho-kinase activity in adipocytes contributes to a vicious cycle in obesity that may involve mechanical stretch. Sci Signal 4: ra3, 2011. [DOI] [PubMed] [Google Scholar]
  • 82.Hasegawa Y, Ikeda K, Chen Y, Alba DL, Stifler D, Shinoda K, Hosono T, Maretich P, Yang Y, Ishigaki Y, Chi J, Cohen P, Koliwad SK, Kajimura S. Repression of adipose tissue fibrosis through a PRDM16-GTF2IRD1 complex improves systemic glucose homeostasis. Cell Metab 27: 180–194 e186, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Hashimoto N, Phan SH, Imaizumi K, Matsuo M, Nakashima H, Kawabe T, Shimokata K, Hasegawa Y. Endothelial-mesenchymal transition in bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol 43: 161–172, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.He Q, Gao Z, Yin J, Zhang J, Yun Z, Ye J. Regulation of HIF-1{alpha} activity in adipose tissue by obesity-associated factors: Adipogenesis, insulin, and hypoxia. Am J Physiol Endocrinol Metab 300: E877–E885, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Henegar C, Tordjman J, Achard V, Lacasa D, Cremer I, Guerre-Millo M, Poitou C, Basdevant A, Stich V, Viguerie N, Langin D, Bedossa P, Zucker JD, Clement K. Adipose tissue transcriptomic signature highlights the pathological relevance of extracellular matrix in human obesity. Genome Biol 9: R14, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Hepler C, Shan B, Zhang Q, Henry GH, Shao M, Vishvanath L, Ghaben AL, Mobley AB, Strand D, Hon GC, Gupta RK. Identification of functionally distinct fibro-inflammatory and adipogenic stromal subpopulations in visceral adipose tissue of adult mice. elife 7: e39636, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hepler C, Vishvanath L, Gupta RK. Sorting out adipocyte precursors and their role in physiology and disease. Genes Dev 31: 127–140, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Herchenhan A, Uhlenbrock F, Eliasson P, Weis M, Eyre D, Kadler KE, Magnusson SP, Kjaer M. Lysyl oxidase activity is required for ordered collagen fibrillogenesis by tendon cells. J Biol Chem 290: 16440–16450, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Heumuller SE, Talantikite M, Napoli M, Armengaud J, Morgelin M, Hartmann U, Sengle G, Paulsson M, Moali C, Wagener R. C-terminal proteolysis of the collagen VI alpha3 chain by BMP-1 and proprotein convertase(s) releases endotrophin in fragments of different sizes. J Biol Chem 294: 13769–13780, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Hirai S, Ohyane C, Kim YI, Lin S, Goto T, Takahashi N, Kim CS, Kang J, Yu R, Kawada T. Involvement of mast cells in adipose tissue fibrosis. Am J Physiol Endocrinol Metab 306: E247–E255, 2014. [DOI] [PubMed] [Google Scholar]
  • 91.Holm Nielsen S, Edsfeldt A, Tengryd C, Gustafsson H, Shore AC, Natali A, Khan F, Genovese F, Bengtsson E, Karsdal M, Leeming DJ, Nilsson J, Goncalves I. The novel collagen matrikine, endotrophin, is associated with mortality and cardiovascular events in patients with atherosclerosis. J Intern Med 290: 179–189, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hosogai N, Fukuhara A, Oshima K, Miyata Y, Tanaka S, Segawa K, Furukawa S, Tochino Y, Komuro R, Matsuda M, Shimomura I. Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes 56: 901–911, 2007. [DOI] [PubMed] [Google Scholar]
  • 93.Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: Direct role in obesity-linked insulin resistance. Science 259: 87–91, 1993. [DOI] [PubMed] [Google Scholar]
  • 94.Huang A, Lin YS, Kao LZ, Chiou YW, Lee GH, Lin HH, Wu CH, Chang CS, Lee KT, Hsueh YY, Tsai PJ, Tang MJ, Tsai YS. Inflammation-induced macrophage lysyl oxidase in adipose stiffening and dysfunction in obesity. Clin Transl Med 11: e543, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Huber J, Loffler M, Bilban M, Reimers M, Kadl A, Todoric J, Zeyda M, Geyeregger R, Schreiner M, Weichhart T, Leitinger N, Waldhausl W, Stulnig TM. Prevention of high-fat diet-induced adipose tissue remodeling in obese diabetic mice by n-3 polyunsaturated fatty acids. Int J Obes 31: 1004–1013, 2007. [DOI] [PubMed] [Google Scholar]
  • 96.Ilyas Z, Perna S, Al-Thawadi S, Alalwan TA, Riva A, Petrangolini G, Gasparri C, Infantino V, Peroni G, Rondanelli M. The effect of Berberine on weight loss in order to prevent obesity: A systematic review. Biomed Pharmacother 127: 110137, 2020. [DOI] [PubMed] [Google Scholar]
  • 97.Itoh Y, Seiki M. MT1-MMP: A potent modifier of pericellular microenvironment. J Cell Physiol 206: 1–8, 2006. [DOI] [PubMed] [Google Scholar]
  • 98.Iwayama T, Steele C, Yao L, Dozmorov MG, Karamichos D, Wren JD, Olson LE. PDGFRalpha signaling drives adipose tissue fibrosis by targeting progenitor cell plasticity. Genes Dev 29: 1106–1119, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Jablonska-Trypuc A, Matejczyk M, Rosochacki S. Matrix metalloproteinases (MMPs), the main extracellular matrix (ECM) enzymes in collagen degradation, as a target for anticancer drugs. J Enzyme Inhib Med Chem 31: 177–183, 2016. [DOI] [PubMed] [Google Scholar]
  • 100.Janke K, Brockmeier U, Kuhlmann K, Eisenacher M, Nolde J, Meyer HE, Mairbaurl H, Metzen E. Factor inhibiting HIF-1 (FIH-1) modulates protein interactions of apoptosis-stimulating p53 binding protein 2 (ASPP2). J Cell Sci 126: 2629–2640, 2013. [DOI] [PubMed] [Google Scholar]
  • 101.Ji Y, Cao M, Liu J, Chen Y, Li X, Zhao J, Qu C. Rock signaling control PPARgamma expression and actin polymerization during adipogenesis. Saudi J Biol Sci 24: 1866–1870, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Jiao B, An C, Du H, Tran M, Wang P, Zhou D, Wang Y. STAT6 deficiency attenuates myeloid fibroblast activation and macrophage polarization in experimental folic acid nephropathy. Cell 10: 3057, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Jiao B, An C, Tran M, Du H, Wang P, Zhou D, Wang Y. Pharmacological inhibition of STAT6 ameliorates myeloid fibroblast activation and alternative macrophage polarization in renal fibrosis. Front Immunol 12: 735014, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Jo W, Kim M, Oh J, Kim CS, Park C, Yoon S, Lee C, Kim S, Nam D, Park J. MicroRNA-29 ameliorates fibro-inflammation and insulin resistance in HIF1alpha-deficient obese adipose tissue by inhibiting endotrophin generation. Diabetes 71: 1746–1762, 2022. [DOI] [PubMed] [Google Scholar]
  • 105.Jones JEC, Rabhi N, Orofino J, Gamini R, Perissi V, Vernochet C, Farmer SR. The adipocyte acquires a fibroblast-like transcriptional signature in response to a high fat diet. Sci Rep 10: 2380, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kaartinen MT, Arora M, Heinonen S, Hang A, Barry A, Lundbom J, Hakkarainen A, Lundholm N, Rissanen A, Kaprio J, Pietilainen KH. F13A1 transglutaminase expression in human adipose tissue increases in acquired excess weight and associates with inflammatory status of adipocytes. Int J Obes 45: 577–587, 2021. [DOI] [PubMed] [Google Scholar]
  • 107.Kaartinen MT, Arora M, Heinonen S, Rissanen A, Kaprio J, Pietilainen KH. Transglutaminases and obesity in humans: Association of F13A1 to adipocyte hypertrophy and adipose tissue immune response. Int J Mol Sci 21: 8289, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kagan HM, Li W. Lysyl oxidase: Properties, specificity, and biological roles inside and outside of the cell. J Cell Biochem 88: 660–672, 2003. [DOI] [PubMed] [Google Scholar]
  • 109.Kane H, Lynch L. Innate immune control of adipose tissue homeostasis. Trends Immunol 40: 857–872, 2019. [DOI] [PubMed] [Google Scholar]
  • 110.Kang L, Lantier L, Kennedy A, Bonner JS, Mayes WH, Bracy DP, Bookbinder LH, Hasty AH, Thompson CB, Wasserman DH. Hyaluronan accumulates with high-fat feeding and contributes to insulin resistance. Diabetes 62: 1888–1896, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Karastergiou K, Mohamed-Ali V. The autocrine and paracrine roles of adipokines. Mol Cell Endocrinol 318: 69–78, 2010. [DOI] [PubMed] [Google Scholar]
  • 112.Karsdal MA, Henriksen K, Genovese F, Leeming DJ, Nielsen MJ, Riis BJ, Christiansen C, Byrjalsen I, Schuppan D. Serum endotrophin identifies optimal responders to PPARgamma agonists in type 2 diabetes. Diabetologia 60: 50–59, 2017. [DOI] [PubMed] [Google Scholar]
  • 113.Keith B, Johnson RS, Simon MC. HIF1alpha and HIF2alpha: Sibling rivalry in hypoxic tumour growth and progression. Nat Rev Cancer 12: 9–22, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Kendall RT, Feghali-Bostwick CA. Fibroblasts in fibrosis: Novel roles and mediators. Front Pharmacol 5: 123, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kerbert AJC, Gupta S, Alabsawy E, Dobler I, Lonsmann I, Hall A, Nielsen SH, Nielsen MJ, Gronbaek H, Amoros A, Yeung D, Macnaughtan J, Mookerjee RP, Macdonald S, Andreola F, Moreau R, Arroyo V, Angeli P, Leeming DJ, Treem W, Karsdal MA, Jalan R. Biomarkers of extracellular matrix formation are associated with acute-on-chronic liver failure. JHEP Rep 3: 100355, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Khan T, Muise ES, Iyengar P, Wang ZV, Chandalia M, Abate N, Zhang BB, Bonaldo P, Chua S, Scherer PE. Metabolic dysregulation and adipose tissue fibrosis: Role of collagen VI. Mol Cell Biol 29: 1575–1591, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Kim M, Lee C, Seo DY, Lee H, Horton JD, Park J, Scherer PE. The impact of endotrophin on the progression of chronic liver disease. Exp Mol Med 52: 1766–1776, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kim M, Neinast MD, Frank AP, Sun K, Park J, Zehr JA, Vishvanath L, Morselli E, Amelotte M, Palmer BF, Gupta RK, Scherer PE, Clegg DJ. ERalpha upregulates Phd3 to ameliorate HIF-1 induced fibrosis and inflammation in adipose tissue. Mol. Metab. 3: 642–651, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Kinsey R, Williamson MR, Chaudhry S, Mellody KT, McGovern A, Takahashi S, Shuttleworth CA, Kielty CM. Fibrillin-1 microfibril deposition is dependent on fibronectin assembly. J Cell Sci 121: 2696–2704, 2008. [DOI] [PubMed] [Google Scholar]
  • 120.Kiran S, Kumar V, Murphy EA, Enos RT, Singh UP. High fat diet-induced CD8(+) T cells in adipose tissue mediate macrophages to sustain low-grade chronic inflammation. Front Immunol 12: 680944, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Kodama K, Horikoshi M, Toda K, Yamada S, Hara K, Irie J, Sirota M, Morgan AA, Chen R, Ohtsu H, Maeda S, Kadowaki T, Butte AJ. Expression-based genome-wide association study links the receptor CD44 in adipose tissue with type 2 diabetes. Proc Natl Acad Sci U S A 109: 7049–7054, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kong P, Gonzalez-Quesada C, Li N, Cavalera M, Lee DW, Frangogiannis NG. Thrombospondin-1 regulates adiposity and metabolic dysfunction in diet-induced obesity enhancing adipose inflammation and stimulating adipocyte proliferation. Am J Physiol Endocrinol Metab 305: E439–E450, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Kos K, Wilding JP. SPARC: A key player in the pathologies associated with obesity and diabetes. Nat Rev Endocrinol 6: 225–235, 2010. [DOI] [PubMed] [Google Scholar]
  • 124.Kos K, Wong S, Tan B, Gummesson A, Jernas M, Franck N, Kerrigan D, Nystrom FH, Carlsson LM, Randeva HS, Pinkney JH, Wilding JP. Regulation of the fibrosis and angiogenesis promoter SPARC/osteonectin in human adipose tissue by weight change, leptin, insulin, and glucose. Diabetes 58: 1780–1788, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Koumenis C, Bi M, Ye J, Feldman D, Koong AC. Hypoxia and the unfolded protein response. Methods Enzymol 435: 275–293, 2007. [DOI] [PubMed] [Google Scholar]
  • 126.Kralisch S, Bluher M, Tonjes A, Lossner U, Paschke R, Stumvoll M, Fasshauer M. Tissue inhibitor of metalloproteinase-1 predicts adiposity in humans. Eur J Endocrinol 156: 257–261, 2007. [DOI] [PubMed] [Google Scholar]
  • 127.Kruglikov IL, Joffin N, Scherer PE. The MMP14-caveolin axis and its potential relevance for lipoedema. Nat Rev Endocrinol 16: 669–674, 2020. [DOI] [PubMed] [Google Scholar]
  • 128.Kruglikov IL, Zhang Z, Scherer PE. Phenotypical conversions of dermal adipocytes as pathophysiological steps in inflammatory cutaneous disorders. Int J Mol Sci 23: 3828, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Kruglikov IL, Zhang Z, Scherer PE. Skin aging: Dermal adipocytes metabolically reprogram dermal fibroblasts. Bioessays 44: e2100207, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Krystel-Whittemore M, Dileepan KN, Wood JG. Mast cell: A multifunctional master cell. Front Immunol 6: 620, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Kusminski CM, Shetty S, Orci L, Unger RH, Scherer PE. Diabetes and apoptosis: Lipotoxicity. Apoptosis 14: 1484–1495, 2009. [DOI] [PubMed] [Google Scholar]
  • 132.Kusunoki H, Taniyama Y, Otsu R, Rakugi H, Morishita R. Anti-inflammatory effects of hepatocyte growth factor on the vicious cycle of macrophages and adipocytes. Hypertens Res 37: 500–506, 2014. [DOI] [PubMed] [Google Scholar]
  • 133.Lackey DE, Olefsky JM. Regulation of metabolism by the innate immune system. Nat Rev Endocrinol 12: 15–28, 2016. [DOI] [PubMed] [Google Scholar]
  • 134.Lando D, Peet DJ, Gorman JJ, Whelan DA, Whitelaw ML, Bruick RK. FIH-1 is an asparaginyl hydroxylase enzyme that regulates the transcriptional activity of hypoxia-inducible factor. Genes Dev 16: 1466–1471, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Lauterbach MA, Wunderlich FT. Macrophage function in obesity-induced inflammation and insulin resistance. Pflugers Arch 469: 385–396, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Lawler HM, Underkofler CM, Kern PA, Erickson C, Bredbeck B, Rasouli N. Adipose tissue hypoxia, inflammation, and fibrosis in obese insulin-sensitive and obese insulin-resistant subjects. J Clin Endocrinol Metab 101: 1422–1428, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Leach HG, Chrobak I, Han R, Trojanowska M. Endothelial cells recruit macrophages and contribute to a fibrotic milieu in bleomycin lung injury. Am J Respir Cell Mol Biol 49: 1093–1101, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Lee C, Kim M, Lee JH, Oh J, Shin HH, Lee SM, Scherer PE, Kwon HM, Choi JH, Park J. COL6A3-derived endotrophin links reciprocal interactions among hepatic cells in the pathology of chronic liver disease. J Pathol 247: 99–109, 2019. [DOI] [PubMed] [Google Scholar]
  • 139.Lee K, Kim HM. A novel approach to cancer therapy using PX-478 as a HIF-1alpha inhibitor. Arch Pharm Res 34: 1583–1585, 2011. [DOI] [PubMed] [Google Scholar]
  • 140.Lee MH, Goralczyk AG, Kriszt R, Ang XM, Badowski C, Li Y, Summers SA, Toh SA, Yassin MS, Shabbir A, Sheppard A, Raghunath M. ECM microenvironment unlocks brown adipogenic potential of adult human bone marrow-derived MSCs. Sci Rep 6: 21173, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Lee SH, Park HS, Lee JA, Song YS, Jang YJ, Kim JH, Lee YJ, Heo Y. Fibronectin gene expression in human adipose tissue and its associations with obesity-related genes and metabolic parameters. Obes Surg 23: 554–560, 2013. [DOI] [PubMed] [Google Scholar]
  • 142.Lee YH, Petkova AP, Granneman JG. Identification of an adipogenic niche for adipose tissue remodeling and restoration. Cell Metab 18: 355–367, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Lee YS, Kim JW, Osborne O, Oh DY, Sasik R, Schenk S, Chen A, Chung H, Murphy A, Watkins SM, Quehenberger O, Johnson RS, Olefsky JM. Increased adipocyte O2 consumption triggers HIF-1alpha, causing inflammation and insulin resistance in obesity. Cell 157: 1339–1352, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Lee YS, Wollam J, Olefsky JM. An integrated view of immunometabolism. Cell 172: 22–40, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Leeming DJ, Nielsen SH, Vongsuvanh R, Uchila P, Nielsen MJ, Reese-Petersen AL, van der Poorten D, Eslam M, Schuppan D, Karsdal MA, George J. Endotrophin, a pro-peptide of Type VI collagen, is a biomarker of survival in cirrhotic patients with hepatocellular carcinoma. Hepat Oncol 8: HEP32, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Li G, Jin F, Du J, He Q, Yang B, Luo P. Macrophage-secreted TSLP and MMP9 promote bleomycin-induced pulmonary fibrosis. Toxicol Appl Pharmacol 366: 10–16, 2019. [DOI] [PubMed] [Google Scholar]
  • 147.Li G, Li X, Yang L, Wang S, Dai Y, Fekry B, Veillon L, Tan L, Berdeaux R, Eckel-Mahan K, Lorenzi PL, Zhao Z, Lehner R, Sun K. Adipose tissue-specific ablation of Ces1d causes metabolic dysregulation in mice. Life Sci Alliance 5: e202101209, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Li Q, Hata A, Kosugi C, Kataoka N, Funaki M. The density of extracellular matrix proteins regulates inflammation and insulin signaling in adipocytes. FEBS Lett 584: 4145–4150, 2010. [DOI] [PubMed] [Google Scholar]
  • 149.Li X, Yang L, Mao Z, Pan X, Zhao Y, Gu X, Eckel-Mahan K, Zuo Z, Tong Q, Hartig SM, Cheng X, Du G, Moore DD, Bellen HJ, Sesaki H, Sun K. Novel role of dynamin-related-protein 1 in dynamics of ER-lipid droplets in adipose tissue. FASEB J 34: 8265–8282, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Li X, Zhao Y, Chen C, Yang L, Lee HH, Wang Z, Zhang N, Kolonin MG, An Z, Ge X, Scherer PE, Sun K. Critical role of matrix metalloproteinase 14 in adipose tissue remodeling during obesity. Mol Cell Biol 40: e00564–19, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Li Y, Tong X, Rumala C, Clemons K, Wang S. Thrombospondin1 deficiency reduces obesity-associated inflammation and improves insulin sensitivity in a diet-induced obese mouse model. PLoS One 6: e26656, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Liang X, Kanjanabuch T, Mao SL, Hao CM, Tang YW, Declerck PJ, Hasty AH, Wasserman DH, Fogo AB, Ma LJ. Plasminogen activator inhibitor-1 modulates adipocyte differentiation. Am J Physiol Endocrinol Metab 290: E103–E113, 2006. [DOI] [PubMed] [Google Scholar]
  • 153.Lin Q, Huang Y, Booth CJ, Haase VH, Johnson RS, Celeste Simon M, Giordano FJ, Yun Z. Activation of hypoxia-inducible factor-2 in adipocytes results in pathological cardiac hypertrophy. J Am Heart Assoc 2: e000548, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Lindholm M, Godskesen LE, Manon-Jensen T, Kjeldsen J, Krag A, Karsdal MA, Mortensen JH. Endotrophin and C6Ma3, serological biomarkers of type VI collagen remodelling, reflect endoscopic and clinical disease activity in IBD. Sci Rep 11: 14713, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Liu J, Divoux A, Sun J, Zhang J, Clement K, Glickman JN, Sukhova GK, Wolters PJ, Du J, Gorgun CZ, Doria A, Libby P, Blumberg RS, Kahn BB, Hotamisligil GS, Shi GP. Genetic deficiency and pharmacological stabilization of mast cells reduce diet-induced obesity and diabetes in mice. Nat Med 15: 940–945, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Liu Y, Aron-Wisnewsky J, Marcelin G, Genser L, Le Naour G, Torcivia A, Bauvois B, Bouchet S, Pelloux V, Sasso M, Miette V, Tordjman J, Clement K. Accumulation and changes in composition of collagens in subcutaneous adipose tissue after bariatric surgery. J Clin Endocrinol Metab 101: 293–304, 2016. [DOI] [PubMed] [Google Scholar]
  • 157.Lizardo K, Ayyappan JP, Oswal N, Weiss LM, Scherer PE, Nagajyothi JF. Fat tissue regulates the pathogenesis and severity of cardiomyopathy in murine chagas disease. PLoS Negl Trop Dis 15: e0008964, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Mack M. Inflammation and fibrosis. Matrix Biol 68–69: 106–121, 2018. [DOI] [PubMed] [Google Scholar]
  • 159.Mahon PC, Hirota K, Semenza GL. FIH-1: A novel protein that interacts with HIF-1alpha and VHL to mediate repression of HIF-1 transcriptional activity. Genes Dev 15: 2675–2686, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Maki JM, Sormunen R, Lippo S, Kaarteenaho-Wiik R, Soininen R, Myllyharju J. Lysyl oxidase is essential for normal development and function of the respiratory system and for the integrity of elastic and collagen fibers in various tissues. Am J Pathol 167: 927–936, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Maquoi E, Munaut C, Colige A, Collen D, Lijnen HR. Modulation of adipose tissue expression of murine matrix metalloproteinases and their tissue inhibitors with obesity. Diabetes 51: 1093–1101, 2002. [DOI] [PubMed] [Google Scholar]
  • 162.Marcelin G, Ferreira A, Liu Y, Atlan M, Aron-Wisnewsky J, Pelloux V, Botbol Y, Ambrosini M, Fradet M, Rouault C, Henegar C, Hulot JS, Poitou C, Torcivia A, Nail-Barthelemy R, Bichet JC, Gautier EL, Clement K. A PDGFRalpha-mediated switch toward CD9(high) adipocyte progenitors controls obesity-induced adipose tissue fibrosis. Cell Metab 25: 673–685, 2017. [DOI] [PubMed] [Google Scholar]
  • 163.Marcelin G, Gautier EL, Clement K. Adipose tissue fibrosis in obesity: Etiology and challenges. Annu Rev Physiol 84: 135–155, 2022. [DOI] [PubMed] [Google Scholar]
  • 164.Marcelin G, Silveira ALM, Martins LB, Ferreira AV, Clement K. Deciphering the cellular interplays underlying obesity-induced adipose tissue fibrosis. J Clin Invest 129: 4032–4040, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Mariman EC, Wang P. Adipocyte extracellular matrix composition, dynamics and role in obesity. Cell Mol Life Sci 67: 1277–1292, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Martinez FO, Helming L, Gordon S. Alternative activation of macrophages: An immunologic functional perspective. Annu Rev Immunol 27: 451–483, 2009. [DOI] [PubMed] [Google Scholar]
  • 167.Martinez-Santibanez G, Singer K, Cho KW, DelProposto JL, Mergian T, Lumeng CN. Obesity-induced remodeling of the adipose tissue elastin network is independent of the metalloelastase MMP-12. Adipocyte 4: 264–272, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.McBeath R, Pirone DM, Nelson CM, Bhadriraju K, Chen CS. Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Dev Cell 6: 483–495, 2004. [DOI] [PubMed] [Google Scholar]
  • 169.McLaughlin T, Ackerman SE, Shen L, Engleman E. Role of innate and adaptive immunity in obesity-associated metabolic disease. J Clin Invest 127: 5–13, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Meissburger B, Stachorski L, Roder E, Rudofsky G, Wolfrum C. Tissue inhibitor of matrix metalloproteinase 1 (TIMP1) controls adipogenesis in obesity in mice and in humans. Diabetologia 54: 1468–1479, 2011. [DOI] [PubMed] [Google Scholar]
  • 171.Memetimin H, Li D, Tan K, Zhou C, Liang Y, Wu Y, Wang S. Myeloid-specific deletion of thrombospondin 1 protects against inflammation and insulin resistance in long-term diet-induced obese male mice. Am J Physiol Endocrinol Metab 315: E1194–E1203, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Merrick D, Sakers A, Irgebay Z, Okada C, Calvert C, Morley MP, Percec I, Seale P. Identification of a mesenchymal progenitor cell hierarchy in adipose tissue. Science 364: eaav2501, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Michailidou Z, Gomez-Salazar M, Alexaki VI. Innate immune cells in the adipose tissue in health and metabolic disease. J Innate Immun 14: 4–30, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Michailidou Z, Morton NM, Moreno Navarrete JM, West CC, Stewart KJ, Fernandez-Real JM, Schofield CJ, Seckl JR, Ratcliffe PJ. Adipocyte pseudohypoxia suppresses lipolysis and facilitates benign adipose tissue expansion. Diabetes 64: 733–745, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Miossec P, Dinarello CA, Ziff M. Interleukin-1 lymphocyte chemotactic activity in rheumatoid arthritis synovial fluid. Arthritis Rheum 29: 461–470, 1986. [DOI] [PubMed] [Google Scholar]
  • 176.Misumi I, Starmer J, Uchimura T, Beck MA, Magnuson T, Whitmire JK. Obesity expands a distinct population of T cells in adipose tissue and increases vulnerability to infection. Cell Rep 27: 514–524 e515, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Mori S, Kiuchi S, Ouchi A, Hase T, Murase T. Characteristic expression of extracellular matrix in subcutaneous adipose tissue development and adipogenesis; comparison with visceral adipose tissue. Int J Biol Sci 10: 825–833, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Mosher DF, Schad PE. Cross-linking of fibronectin to collagen by blood coagulation factor XIIIa. J Clin Invest 64: 781–787, 1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Muir LA, Neeley CK, Meyer KA, Baker NA, Brosius AM, Washabaugh AR, Varban OA, Finks JF, Zamarron BF, Flesher CG, Chang JS, DelProposto JB, Geletka L, Martinez-Santibanez G, Kaciroti N, Lumeng CN, O’Rourke RW. Adipose tissue fibrosis, hypertrophy, and hyperplasia: Correlations with diabetes in human obesity. Obesity (Silver Spring) 24: 597–605, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Murano I, Barbatelli G, Parisani V, Latini C, Muzzonigro G, Castellucci M, Cinti S. Dead adipocytes, detected as crown-like structures, are prevalent in visceral fat depots of genetically obese mice. J Lipid Res 49: 1562–1568, 2008. [DOI] [PubMed] [Google Scholar]
  • 181.Muscogiuri G, Bettini S, Boschetti M, Barrea L, Savastano S, Colao A. Obesity programs of nutrition ER, and assessment g. Low-grade inflammation, CoVID-19, and obesity: Clinical aspect and molecular insights in childhood and adulthood. Int J Obes 46 (7): 1254–1261, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Myneni VD, Hitomi K, Kaartinen MT. Factor XIII-A transglutaminase acts as a switch between preadipocyte proliferation and differentiation. Blood 124: 1344–1353, 2014. [DOI] [PubMed] [Google Scholar]
  • 183.Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res 69: 562–573, 2006. [DOI] [PubMed] [Google Scholar]
  • 184.Nakayama Y, Komuro R, Yamamoto A, Miyata Y, Tanaka M, Matsuda M, Fukuhara A, Shimomura I. RhoA induces expression of inflammatory cytokine in adipocytes. Biochem Biophys Res Commun 379: 288–292, 2009. [DOI] [PubMed] [Google Scholar]
  • 185.Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, Otsu M, Hara K, Ueki K, Sugiura S, Yoshimura K, Kadowaki T, Nagai R. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med 15: 914–920, 2009. [DOI] [PubMed] [Google Scholar]
  • 186.Ogawa T, Shichino S, Ueha S, Matsushima K. Macrophages in lung fibrosis. Int Immunol 33: 665–671, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Ogawa T, Shichino S, Ueha S, Ogawa S, Matsushima K. Complement protein C1q activates lung fibroblasts and exacerbates silica-induced pulmonary fibrosis in mice. Biochem Biophys Res Commun 603: 88–93, 2022. [DOI] [PubMed] [Google Scholar]
  • 188.Oja AE, van Lier RAW, Hombrink P. Two sides of the same coin: Protective versus pathogenic CD4(+) resident memory T cells. Sci Immunol 7: eabf9393, 2022. [DOI] [PubMed] [Google Scholar]
  • 189.Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol 72: 219–246, 2010. [DOI] [PubMed] [Google Scholar]
  • 190.Olson E, Demopoulos L, Haws TF, Hu E, Fang Z, Mahar KM, Qin P, Lepore J, Bauer TA, Hiatt WR. Short-term treatment with a novel HIF-prolyl hydroxylase inhibitor (GSK1278863) failed to improve measures of performance in subjects with claudication-limited peripheral artery disease. Vasc Med 19: 473–482, 2014. [DOI] [PubMed] [Google Scholar]
  • 191.Osborn O, Olefsky JM. The cellular and signaling networks linking the immune system and metabolism in disease. Nat Med 18: 363–374, 2012. [DOI] [PubMed] [Google Scholar]
  • 192.Ouchi N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nat Rev Immunol 11: 85–97, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Pajvani UB, Trujillo ME, Combs TP, Iyengar P, Jelicks L, Roth KA, Kitsis RN, Scherer PE. Fat apoptosis through targeted activation of caspase 8: A new mouse model of inducible and reversible lipoatrophy. Nat Med 11: 797–803, 2005. [DOI] [PubMed] [Google Scholar]
  • 194.Pan Y, Hui X, Hoo RLC, Ye D, Chan CYC, Feng T, Wang Y, Lam KSL, Xu A. Adipocyte-secreted exosomal microRNA-34a inhibits M2 macrophage polarization to promote obesity-induced adipose inflammation. J Clin Invest 129: 834–849, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Park J, Kim M, Sun K, An YA, Gu X, Scherer PE. VEGF-A-expressing adipose tissue shows rapid beiging and enhanced survival after transplantation and confers IL-4-independent metabolic improvements. Diabetes 66: 1479–1490, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Park J, Morley TS, Scherer PE. Inhibition of endotrophin, a cleavage product of collagen VI, confers cisplatin sensitivity to tumours. EMBO Mol Med 5: 935–948, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Park J, Scherer PE. Adipocyte-derived endotrophin promotes malignant tumor progression. J Clin Invest 122: 4243–4256, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Park J, Scherer PE. Endotrophin—a novel factor linking obesity with aggressive tumor growth. Oncotarget 3: 1487–1488, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Park J, Scherer PE. Endotrophin in the tumor stroma: A new therapeutic target for breast cancer? Expert Rev Anticancer Ther 13: 111–113, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Pasarica M, Gowronska-Kozak B, Burk D, Remedios I, Hymel D, Gimble J, Ravussin E, Bray GA, Smith SR. Adipose tissue collagen VI in obesity. J Clin Endocrinol Metab 94: 5155–5162, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Pasarica M, Sereda OR, Redman LM, Albarado DC, Hymel DT, Roan LE, Rood JC, Burk DH, Smith SR. Reduced adipose tissue oxygenation in human obesity: Evidence for rarefaction, macrophage chemotaxis, and inflammation without an angiogenic response. Diabetes 58: 718–725, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Pastel E, Price E, Sjoholm K, McCulloch LJ, Rittig N, Liversedge N, Knight B, Moller N, Svensson PA, Kos K. Lysyl oxidase and adipose tissue dysfunction. Metabolism 78: 118–127, 2018. [DOI] [PubMed] [Google Scholar]
  • 203.Peics J, Vishvanath L, Zhang Q, Shan B, Pedersen TA, Gupta RK. Isolation of adipogenic and fibro-inflammatory stromal cell subpopulations from murine intra-abdominal adipose depots. J Vis Exp, 2020. DOI: 10.3791/61610. [DOI] [PubMed] [Google Scholar]
  • 204.Peng H, Hamanaka RB, Katsnelson J, Hao LL, Yang W, Chandel NS, Lavker RM. MicroRNA-31 targets FIH-1 to positively regulate corneal epithelial glycogen metabolism. FASEB J 26: 3140–3147, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Peng H, Katsnelson J, Yang W, Brown MA, Lavker RM. FIH-1/c-kit signaling: A novel contributor to corneal epithelial glycogen metabolism. Invest Ophthalmol Vis Sci 54: 2781–2786, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Petrus P, Fernandez TL, Kwon MM, Huang JL, Lei V, Safikhan NS, Karunakaran S, O’Shannessy DJ, Zheng X, Catrina SB, Albone E, Laine J, Virtanen K, Clee SM, Kieffer TJ, Noll C, Carpentier AC, Johnson JD, Ryden M, Conway EM. Specific loss of adipocyte CD248 improves metabolic health via reduced white adipose tissue hypoxia, fibrosis and inflammation. EBioMedicine 44: 489–501, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Plotkin JD, Elias MG, Fereydouni M, Daniels-Wells TR, Dellinger AL, Penichet ML, Kepley CL. Human mast cells from adipose tissue target and induce apoptosis of breast cancer cells. Front Immunol 10: 138, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Poblete JMS, Ballinger MN, Bao S, Alghothani M, Nevado JB Jr, Eubank TD, Christman JW, Magalang UJ. Macrophage HIF-1alpha mediates obesity-related adipose tissue dysfunction via interleukin-1 receptor-associated kinase M. Am J Physiol Endocrinol Metab 318: E689–E700, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Porsche CE, Delproposto JB, Geletka L, O’Rourke R, Lumeng CN. Obesity results in adipose tissue T cell exhaustion. JCI Insight 6: e139793, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Rabhi N, Desevin K, Belkina AC, Tilston-Lunel A, Varelas X, Layne MD, Farmer SR. Obesity-induced senescent macrophages activate a fibrotic transcriptional program in adipocyte progenitors. Life Sci Alliance 5: e202101286, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Rasmussen DGK, Fenton A, Jesky M, Ferro C, Boor P, Tepel M, Karsdal MA, Genovese F, Cockwell P. Urinary endotrophin predicts disease progression in patients with chronic kidney disease. Sci Rep 7: 17328, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Rausch ME, Weisberg S, Vardhana P, Tortoriello DV. Obesity in C57BL/6J mice is characterized by adipose tissue hypoxia and cytotoxic T-cell infiltration. Int J Obes 32: 451–463, 2008. [DOI] [PubMed] [Google Scholar]
  • 213.Reilly SM, Saltiel AR. Adapting to obesity with adipose tissue inflammation. Nat Rev Endocrinol 13: 633–643, 2017. [DOI] [PubMed] [Google Scholar]
  • 214.Remacle A, Murphy G, Roghi C. Membrane type I-matrix metalloproteinase (MT1-MMP) is internalised by two different pathways and is recycled to the cell surface. J Cell Sci 116: 3905–3916, 2003. [DOI] [PubMed] [Google Scholar]
  • 215.Rønnow SR, Langholm LL, Karsdal MA, Manon-Jensen T, Tal-Singer R, Miller BE, Vestbo J, Leeming DJ, Sand JMB. Endotrophin, an extracellular hormone, in combination with neoepitope markers of von Willebrand factor improves prediction of mortality in the ECLIPSE COPD cohort. Respir Res 21: 202, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Rossow L, Veitl S, Vorlova S, Wax JK, Kuhn AE, Maltzahn V, Upcin B, Karl F, Hoffmann H, Gatzner S, Kallius M, Nandigama R, Scheld D, Irmak S, Herterich S, Zernecke A, Ergun S, Henke E. LOX-catalyzed collagen stabilization is a proximal cause for intrinsic resistance to chemotherapy. Oncogene 37: 4921–4940, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Ruas JL, Poellinger L, Pereira T. Role of CBP in regulating HIF-1-mediated activation of transcription. J Cell Sci 118: 301–311, 2005. [DOI] [PubMed] [Google Scholar]
  • 218.Ruiz-Ojeda FJ, Mendez-Gutierrez A, Aguilera CM, Plaza-Diaz J. Extracellular matrix remodeling of adipose tissue in obesity and metabolic diseases. Int J Mol Sci 20: 4888, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Rutkowski JM, Stern JH, Scherer PE. The cell biology of fat expansion. J Cell Biol 208: 501–512, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Sabeh F, Li XY, Saunders TL, Rowe RG, Weiss SJ. Secreted versus membrane-anchored collagenases: Relative roles in fibroblast-dependent collagenolysis and invasion. J Biol Chem 284: 23001–23011, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Sakamoto T, Seiki M. A membrane protease regulates energy production in macrophages by activating hypoxia-inducible factor-1 via a non-proteolytic mechanism. J Biol Chem 285: 29951–29964, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Sakamoto T, Seiki M. Integrated functions of membrane-type 1 matrix metalloproteinase in regulating cancer malignancy: Beyond a proteinase. Cancer Sci 108: 1095–1100, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Sakamoto T, Weng JS, Hara T, Yoshino S, Kozuka-Hata H, Oyama M, Seiki M. Hypoxia-inducible factor 1 regulation through cross talk between mTOR and MT1-MMP. Mol Cell Biol 34: 30–42, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Sakamuri S, Watts R, Takawale A, Wang X, Hernandez-Anzaldo S, Bahitham W, Fernandez-Patron C, Lehner R, Kassiri Z. Absence of tissue inhibitor of metalloproteinase-4 (TIMP4) ameliorates high fat diet-induced obesity in mice due to defective lipid absorption. Sci Rep 7: 6210, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Scherer PE, Bickel PE, Kotler M, Lodish HF. Cloning of cell-specific secreted and surface proteins by subtractive antibody screening. Nat Biotechnol 16: 581–586, 1998. [DOI] [PubMed] [Google Scholar]
  • 226.Scherer PE, Gupta OT. Endotrophin: Nominated for best supporting actor in the fibro-inflammatory saga. EBioMedicine 69: 103447, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Sebestyen A, Kopper L, Danko T, Timar J. Hypoxia signaling in cancer: From basics to clinical practice. Pathol Oncol Res 27: 1609802, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Semenza GL. HIF-1 and human disease: One highly involved factor. Genes Dev 14: 1983–1991, 2000. [PubMed] [Google Scholar]
  • 229.Seo JB, Riopel M, Cabrales P, Huh JY, Bandyopadhyay GK, Andreyev AY, Murphy AN, Beeman SC, Smith GI, Klein S, Lee YS, Olefsky JM. Knockdown of Ant2 reduces adipocyte hypoxia and improves insulin resistance in obesity. Nat Metab 1: 86–97, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Shan B, Shao M, Zhang Q, Hepler C, Paschoal VA, Barnes SD, Vishvanath L, An YA, Jia L, Malladi VS, Strand DW, Gupta OT, Elmquist JK, Oh D, Gupta RK. Perivascular mesenchymal cells control adipose-tissue macrophage accrual in obesity. Nat Metab 2: 1332–1349, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Shao M, Hepler C, Zhang Q, Shan B, Vishvanath L, Henry GH, Zhao S, An YA, Wu Y, Strand DW, Gupta RK. Pathologic HIF1alpha signaling drives adipose progenitor dysfunction in obesity. Cell Stem Cell 28: 685–701 e687, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Sharma M, Boytard L, Hadi T, Koelwyn G, Simon R, Ouimet M, Seifert L, Spiro W, Yan B, Hutchison S, Fisher EA, Ramasamy R, Ramkhelawon B, Moore KJ. Enhanced glycolysis and HIF-1alpha activation in adipose tissue macrophages sustains local and systemic interleukin-1beta production in obesity. Sci Rep 10: 5555, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Skurk T, Alberti-Huber C, Herder C, Hauner H. Relationship between adipocyte size and adipokine expression and secretion. J Clin Endocrinol Metab 92: 1023–1033, 2007. [DOI] [PubMed] [Google Scholar]
  • 234.Sottile J, Hocking DC. Fibronectin polymerization regulates the composition and stability of extracellular matrix fibrils and cell-matrix adhesions. Mol Biol Cell 13: 3546–3559, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Sparding N, Genovese F, Rasmussen DGK, Karsdal MA, Neprasova M, Maixnerova D, Satrapova V, Frausova D, Hornum M, Bartonova L, Honsova E, Kollar M, Koprivova H, Hruskova Z, Tesar V. Endotrophin, a collagen type VI-derived matrikine, reflects the degree of renal fibrosis in patients with IgA nephropathy and in patients with ANCA-associated vasculitis. Nephrol Dial Transplant 37 (6): 1099–1108, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Spencer M, Unal R, Zhu B, Rasouli N, McGehee RE Jr, Peterson CA, Kern PA. Adipose tissue extracellular matrix and vascular abnormalities in obesity and insulin resistance. J Clin Endocrinol Metab 96: E1990–E1998, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Spencer M, Yao-Borengasser A, Unal R, Rasouli N, Gurley CM, Zhu B, Peterson CA, Kern PA. Adipose tissue macrophages in insulin-resistant subjects are associated with collagen VI and fibrosis and demonstrate alternative activation. Am J Physiol Endocrinol Metab 299: E1016–E1027, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Staunstrup LM, Bager CL, Frederiksen P, Helge JW, Brunak S, Christiansen C, Karsdal M. Endotrophin is associated with chronic multimorbidity and all-cause mortality in a cohort of elderly women. EBioMedicine 68: 103391, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Strohmeyer N, Bharadwaj M, Costell M, Fassler R, Muller DJ. Fibronectin-bound alpha5beta1 integrins sense load and signal to reinforce adhesion in less than a second. Nat Mater 16: 1262–1270, 2017. [DOI] [PubMed] [Google Scholar]
  • 240.Suganami T, Nishida J, Ogawa Y. A paracrine loop between adipocytes and macrophages aggravates inflammatory changes: Role of free fatty acids and tumor necrosis factor alpha. Arterioscler Thromb Vasc Biol 25: 2062–2068, 2005. [DOI] [PubMed] [Google Scholar]
  • 241.Sun K, Halberg N, Khan M, Magalang UJ, Scherer PE. Selective inhibition of hypoxia-inducible factor 1alpha ameliorates adipose tissue dysfunction. Mol Cell Biol 33: 904–917, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Sun K, Kusminski CM, Scherer PE. Adipose tissue remodeling and obesity. J Clin Invest 121: 2094–2101, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Sun K, Park J, Gupta OT, Holland WL, Auerbach P, Zhang N, Goncalves Marangoni R, Nicoloro SM, Czech MP, Varga J, Ploug T, An Z, Scherer PE. Endotrophin triggers adipose tissue fibrosis and metabolic dysfunction. Nat Commun 5: 3485, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Sun K, Park J, Kim M, Scherer PE. Endotrophin, a multifaceted player in metabolic dysregulation and cancer progression, is a predictive biomarker for the response to PPARgamma agonist treatment. Diabetologia 60: 24–29, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Sun K, Tordjman J, Clement K, Scherer PE. Fibrosis and adipose tissue dysfunction. Cell Metab 18: 470–477, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Sun K, Wernstedt Asterholm I, Kusminski CM, Bueno AC, Wang ZV, Pollard JW, Brekken RA, Scherer PE. Dichotomous effects of VEGF-A on adipose tissue dysfunction. Proc Natl Acad Sci U S A 109: 5874–5879, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Takahashi M, Nagaretani H, Funahashi T, Nishizawa H, Maeda N, Kishida K, Kuriyama H, Shimomura I, Maeda K, Hotta K, Ouchi N, Kihara S, Nakamura T, Yamashita S, Matsuzawa Y. The expression of SPARC in adipose tissue and its increased plasma concentration in patients with coronary artery disease. Obes Res 9: 388–393, 2001. [DOI] [PubMed] [Google Scholar]
  • 248.Talukdar S, Oh DY, Bandyopadhyay G, Li D, Xu J, McNelis J, Lu M, Li P, Yan Q, Zhu Y, Ofrecio J, Lin M, Brenner MB, Olefsky JM. Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase. Nat Med 18: 1407–1412, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Tang PM, Nikolic-Paterson DJ, Lan HY. Macrophages: Versatile players in renal inflammation and fibrosis. Nat Rev Nephrol 15: 144–158, 2019. [DOI] [PubMed] [Google Scholar]
  • 250.Tepel M, Alkaff FF, Kremer D, Bakker SJL, Thaunat O, Nagarajah S, Saleh Q, Berger SP, van den Born J, Krogstrup NV, Nielsen MB, Norregaard R, Jespersen B, Sparding N, Genovese F, Karsdal MA, Rasmussen DGK. Pretransplant endotrophin predicts delayed graft function after kidney transplantation. Sci Rep 12: 4079, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Thompson RW, Pesce JT, Ramalingam T, Wilson MS, White S, Cheever AW, Ricklefs SM, Porcella SF, Li L, Ellies LG, Wynn TA. Cationic amino acid transporter-2 regulates immunity by modulating arginase activity. PLoS Pathog 4: e1000023, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Thrailkill KM, Clay Bunn R, Fowlkes JL. Matrix metalloproteinases: Their potential role in the pathogenesis of diabetic nephropathy. Endocrine 35: 1–10, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Todorcevic M, Manuel AR, Austen L, Michailidou Z, Hazlehurst JM, Neville M, Stradling JR, Karpe F. Markers of adipose tissue hypoxia are elevated in subcutaneous adipose tissue of severely obese patients with obesity hypoventilation syndrome but not in the moderately obese. Int J Obes 45: 1618–1622, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol 3: 349–363, 2002. [DOI] [PubMed] [Google Scholar]
  • 255.Hypoxia Trayhurn P. and adipose tissue function and dysfunction in obesity. Physiol Rev 93: 1–21, 2013. [DOI] [PubMed] [Google Scholar]
  • 256.Trinh K, Julovi SM, Rogers NM. The role of matrix proteins in cardiac pathology. Int J Mol Sci 23: 1338, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Trujillo ME, Pajvani UB, Scherer PE. Apoptosis through targeted activation of caspase 8 (“ATTAC-mice”): Novel mouse models of inducible and reversible tissue ablation. Cell Cycle 4: 1141–1145, 2005. [DOI] [PubMed] [Google Scholar]
  • 258.Unger RH, Clark GO, Scherer PE, Orci L. Lipid homeostasis, lipotoxicity and the metabolic syndrome. Biochim Biophys Acta 1801: 209–214, 2010. [DOI] [PubMed] [Google Scholar]
  • 259.Varma V, Yao-Borengasser A, Bodles AM, Rasouli N, Phanavanh B, Nolen GT, Kern EM, Nagarajan R, Spencer HJ 3rd, Lee MJ, Fried SK, McGehee RE Jr, Peterson CA, Kern PA. Thrombospondin-1 is an adipokine associated with obesity, adipose inflammation, and insulin resistance. Diabetes 57: 432–439, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Verkouter I, Noordam R, Loh NY, van Dijk KW, Zock PL, Mook-Kanamori DO, le Cessie S, Rosendaal FR, Karpe F, Christodoulides C, de Mutsert R. The relation between adult weight gain, adipocyte volume, and the metabolic profile at middle age. J Clin Endocrinol Metab 106: e4438–e4447, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Vijay J, Gauthier MF, Biswell RL, Louiselle DA, Johnston JJ, Cheung WA, Belden B, Pramatarova A, Biertho L, Gibson M, Simon MM, Djambazian H, Staffa A, Bourque G, Laitinen A, Nystedt J, Vohl MC, Fraser JD, Pastinen T, Tchernof A, Grundberg E. Single-cell analysis of human adipose tissue identifies depot and disease specific cell types. Nat Metab 2: 97–109, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Vila IK, Badin PM, Marques MA, Monbrun L, Lefort C, Mir L, Louche K, Bourlier V, Roussel B, Gui P, Grober J, Stich V, Rossmeislova L, Zakaroff-Girard A, Bouloumie A, Viguerie N, Moro C, Tavernier G, Langin D. Immune cell Toll-like receptor 4 mediates the development of obesity- and endotoxemia-associated adipose tissue fibrosis. Cell Rep 7: 1116–1129, 2014. [DOI] [PubMed] [Google Scholar]
  • 263.Visscher TL, Seidell JC. The public health impact of obesity. Annu Rev Public Health 22: 355–375, 2001. [DOI] [PubMed] [Google Scholar]
  • 264.Wang E, Zhang C, Polavaram N, Liu F, Wu G, Schroeder MA, Lau JS, Mukhopadhyay D, Jiang SW, O’Neill BP, Datta K, Li J. The role of factor inhibiting HIF (FIH-1) in inhibiting HIF-1 transcriptional activity in glioblastoma multiforme. PLoS One 9: e86102, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Wang L, Ye X, Hua Y, Song Y. Berberine alleviates adipose tissue fibrosis by inducing AMP-activated kinase signaling in high-fat diet-induced obese mice. Biomed Pharmacother 105: 121–129, 2018. [DOI] [PubMed] [Google Scholar]
  • 266.Wang Y, Nishina PM, Naggert JK. Degradation of IRS1 leads to impaired glucose uptake in adipose tissue of the type 2 diabetes mouse model TALLYHO/Jng. J Endocrinol 203: 65–74, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Wang Y, Zhao L, Smas C, Sul HS. Pref-1 interacts with fibronectin to inhibit adipocyte differentiation. Mol Cell Biol 30: 3480–3492, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Warnberg J, Moreno LA, Mesana MI, Marcos A, group A. Inflammatory mediators in overweight and obese Spanish adolescents. The AVENA Study. Int J Obes Relat Metab Disord 28 (Suppl 3): S59–S63, 2004. [DOI] [PubMed] [Google Scholar]
  • 269.Wei K, Piecewicz SM, McGinnis LM, Taniguchi CM, Wiegand SJ, Anderson K, Chan CW, Mulligan KX, Kuo D, Yuan J, Vallon M, Morton L, Lefai E, Simon MC, Maher JJ, Mithieux G, Rajas F, Annes J, McGuinness OP, Thurston G, Giaccia AJ, Kuo CJ. A liver Hif-2alpha-Irs2 pathway sensitizes hepatic insulin signaling and is modulated by Vegf inhibition. Nat Med 19: 1331–1337, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest 112: 1796–1808, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Weiss L, Slavin S, Reich S, Cohen P, Shuster S, Stern R, Kaganovsky E, Okon E, Rubinstein AM, Naor D. Induction of resistance to diabetes in non-obese diabetic mice by targeting CD44 with a specific monoclonal antibody. Proc Natl Acad Sci U S A 97: 285–290, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, Tsui H, Wu P, Davidson MG, Alonso MN, Leong HX, Glassford A, Caimol M, Kenkel JA, Tedder TF, McLaughlin T, Miklos DB, Dosch HM, Engleman EG. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med 17: 610–617, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Wu Y, Lee MJ, Ido Y, Fried SK. High-fat diet-induced obesity regulates MMP3 to modulate depot- and sex-dependent adipose expansion in C57BL/6J mice. Am J Physiol Endocrinol Metab 312: E58–E71, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Wynn TA, Vannella KM. Macrophages in tissue repair, regeneration, and fibrosis. Immunity 44: 450–462, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, Chen H. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest 112: 1821–1830, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Xu Y, Kong X, Li J, Cui T, Wei Y, Xu J, Zhu Y, Zhu X. Mild hypoxia enhances the expression of HIF and VEGF and triggers the response to injury in rat kidneys. Front Physiol 12: 690496, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Yamazaki Y, Mikami Y, Yuguchi M, Namba Y, Isokawa K. Development of collagen fibres and lysyl oxidase expression in the presumptive dermis of chick limb bud. Anat Histol Embryol 41: 68–74, 2012. [DOI] [PubMed] [Google Scholar]
  • 278.Yang N, Cao DF, Yin XX, Zhou HH, Mao XY. Lysyl oxidases: Emerging biomarkers and therapeutic targets for various diseases. Biomed Pharmacother 131: 110791, 2020. [DOI] [PubMed] [Google Scholar]
  • 279.Ye J, Gao Z, Yin J, He Q. Hypoxia is a potential risk factor for chronic inflammation and adiponectin reduction in adipose tissue of ob/ob and dietary obese mice. Am J Physiol Endocrinol Metab 293: E1118–E1128, 2007. [DOI] [PubMed] [Google Scholar]
  • 280.Yin J, Gao Z, He Q, Zhou D, Guo Z, Ye J. Role of hypoxia in obesity-induced disorders of glucose and lipid metabolism in adipose tissue. Am J Physiol Endocrinol Metab 296: E333–E342, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Yoldemir SA, Arman Y, Akarsu M, Altun O, Ozcan M, Tukek T. Correlation of glycemic regulation and endotrophin in patients with type 2 diabetes; pilot study. Diabetol Metab Syndr 13: 9, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Yu M, Shen W, Shi X, Wang Q, Zhu L, Xu X, Yu J, Liu L. Upregulated LOX and increased collagen content associated with aggressive clinicopathological features and unfavorable outcome in oral squamous cell carcinoma. J Cell Biochem 120: 14348–14359, 2019. [DOI] [PubMed] [Google Scholar]
  • 283.Zelechowska P, Agier J, Kozlowska E, Brzezinska-Blaszczyk E. Mast cells participate in chronic low-grade inflammation within adipose tissue. Obes Rev 19: 686–697, 2018. [DOI] [PubMed] [Google Scholar]
  • 284.Zhang N, Fu Z, Linke S, Chicher J, Gorman JJ, Visk D, Haddad GG, Poellinger L, Peet DJ, Powell F, Johnson RS. The asparaginyl hydroxylase factor inhibiting HIF-1alpha is an essential regulator of metabolism. Cell Metab 11: 364–378, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Zhang R, Gao Y, Zhao X, Gao M, Wu Y, Han Y, Qiao Y, Luo Z, Yang L, Chen J, Ge G. FSP1-positive fibroblasts are adipogenic niche and regulate adipose homeostasis. PLoS Biol 16: e2001493, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Zhang S, Li Y, Huang X, Liu K, Wang QD, Chen AF, Sun K, Lui KO, Zhou B. Seamless genetic recording of transiently activated mesenchymal gene expression in endothelial cells during cardiac fibrosis. Circulation 144: 2004–2020, 2021. [DOI] [PubMed] [Google Scholar]
  • 287.Zhang Z, Cai J, Li Y, He Y, Dong Z, Dai J, Lu F. External volume expansion adjusted adipose stem cell by shifting the ratio of fibronectin to laminin. Tissue Eng Part A 26: 66–77, 2020. [DOI] [PubMed] [Google Scholar]
  • 288.Zhang Z, Kruglikov I, Zhao S, Zi Z, Gliniak CM, Li N, Wang MY, Zhu Q, Kusminski CM, Scherer PE. Dermal adipocytes contribute to the metabolic regulation of dermal fibroblasts. Exp Dermatol 30: 102–111, 2021. [DOI] [PubMed] [Google Scholar]
  • 289.Zhang Z, Qu R, Fan T, Ouyang J, Lu F, Dai J. Stepwise adipogenesis of decellularized cellular extracellular matrix regulates adipose tissue-derived stem cell migration and differentiation. Stem Cells Int 2019: 1845926, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Zhao Y, Gu X, Zhang N, Kolonin MG, An Z, Sun K. Divergent functions of endotrophin on different cell populations in adipose tissue. Am J Physiol Endocrinol Metab 311: E952–E963, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Zhou HY, Sui H, Zhao YJ, Qian HJ, Yang N, Liu L, Guan Q, Zhou Y, Lin HL, Wang DP. The impact of inflammatory immune reactions of the vascular niche on organ fibrosis. Front Pharmacol 12: 750509, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol 11: 136–140, 2010. [DOI] [PubMed] [Google Scholar]
  • 293.Zhu R, Cheng M, Lu T, Yang N, Ye S, Pan YH, Hong T, Dang S, Zhang W. A disintegrin and metalloproteinase with thrombospondin motifs 18 deficiency leads to visceral adiposity and associated metabolic syndrome in mice. Am J Pathol 188: 461–473, 2018. [DOI] [PubMed] [Google Scholar]
  • 294.Zhu Y, Crewe C, Scherer PE. Hyaluronan in adipose tissue: Beyond dermal filler and therapeutic carrier. Sci Transl Med 8: 323ps324, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Zhu Y, Kruglikov IL, Akgul Y, Scherer PE. Hyaluronan in adipogenesis, adipose tissue physiology and systemic metabolism. Matrix Biol 78–79: 284–291, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Zhu Y, Li N, Huang M, Bartels M, Dogne S, Zhao S, Chen X, Crewe C, Straub L, Vishvanath L, Zhang Z, Shao M, Yang Y, Gliniak CM, Gordillo R, Smith GI, Holland WL, Gupta RK, Dong B, Caron N, Xu Y, Akgul Y, Klein S, Scherer PE. Adipose tissue hyaluronan production improves systemic glucose homeostasis and primes adipocytes for CL 316,243-stimulated lipolysis. Nat Commun 12: 4829, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Zucker S, Pei D, Cao J, Lopez-Otin C. Membrane type-matrix metalloproteinases (MT-MMP). Curr Top Dev Biol 54: 1–74, 2003. [DOI] [PubMed] [Google Scholar]

RESOURCES