Next Article in Journal
An Immunological Approach to the Biocompatibility of Mesoporous SiO2-CaO Nanospheres
Next Article in Special Issue
Transamidation Down-Regulates Intestinal Immunity of Recombinant α-Gliadin in HLA-DQ8 Transgenic Mice
Previous Article in Journal
PARP Inhibitor Olaparib Causes No Potentiation of the Bleomycin Effect in VERO Cells, Even in the Presence of Pooled ATM, DNA-PK, and LigIV Inhibitors
Previous Article in Special Issue
Interplay between Type 2 Transglutaminase (TG2), Gliadin Peptide 31-43 and Anti-TG2 Antibodies in Celiac Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Transglutaminases and Obesity in Humans: Association of F13A1 to Adipocyte Hypertrophy and Adipose Tissue Immune Response

1
Faculty of Medicine (Experimental Medicine), McGill University, Montreal, QC H3A 0J7, Canada
2
Faculty of Dentistry (Biomedical Sciences), McGill University, Montreal, QC H3A 0J7, Canada
3
Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
4
Department of Public Health, University of Helsinki, 00100 Helsinki, Finland
5
Abdominal Center, Obesity Center, Endocrinology, University of Helsinki and Helsinki University Central Hospital, 00014 Helsinki, Finland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(21), 8289; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21218289
Submission received: 31 August 2020 / Revised: 30 October 2020 / Accepted: 1 November 2020 / Published: 5 November 2020

Abstract

:
Transglutaminases TG2 and FXIII-A have recently been linked to adipose tissue biology and obesity, however, human studies for TG family members in adipocytes have not been conducted. In this study, we investigated the association of TGM family members to acquired weight gain in a rare set of monozygotic (MZ) twins discordant for body weight, i.e., heavy–lean twin pairs. We report that F13A1 is the only TGM family member showing significantly altered, higher expression in adipose tissue of the heavier twin. Our previous work linked adipocyte F13A1 to increased weight, body fat mass, adipocyte size, and pro-inflammatory pathways. Here, we explored further the link of F13A1 to adipocyte size in the MZ twins via a previously conducted TWA study that was further mined for genes that specifically associate to hypertrophic adipocytes. We report that differential expression of F13A1 (ΔHeavy–Lean) associated with 47 genes which were linked via gene enrichment analysis to immune response, leucocyte and neutrophil activation, as well as cytokine response and signaling. Our work brings further support to the role of F13A1 in the human adipose tissue pathology, suggesting a role in the cascade that links hypertrophic adipocytes with inflammation.

Graphical Abstract

1. Introduction

Transglutaminases (protein-glutamine γ-glutamyltransferase, TGase, TG, EC 2.3.2.13) are a family of enzymes that catalyze a Ca2+- and thiol-dependent posttranslational modification of glutamine residues [1,2,3,4]. These reactions include acyl transfer reaction with primary amines, glutamine deamination if the second substrate is water, and transamidation, i.e., isopeptide crosslinking, if the second substrate is a lysine of a protein polypeptide chain. The isopeptide formation between glutamine and lysine residues, which is by far the most studied reaction of TGs [1,2,3,4], stabilizes proteins and induces formation of protein networks and alterations in substrate protein biochemistry, stiffness, solubility, and cell adhesive features [2,4,5,6,7]. TGs also have cell signaling functions that do not require crosslinking activity [8].
The TG family consists of nine gene products: TG1–TG7 and Factor XIII-A [1,2,4] (Genes: TGM (human), Tgm (mouse)). Transglutaminases TG2 and FXIII-A have been studied widely in the context of connective tissue hemostasis and differentiation of their resident cell types. While much focus has been on cartilage and bone [9,10,11,12,13,14,15], new research also shows their involvement in adipose tissue. White adipose tissue (WAT) is a form of connective tissue capable of thermoregulation and energy storage and it plays a central role in maintenance of balanced energy metabolism [16]. In caloric excess, WAT expands via preadipocyte proliferation (hyperplasia) and by their differentiation to lipid storing adipocytes followed by their growth (hypertrophy) [16]. The current understanding is that adipocyte size is associated with metabolic consequences of obesity in a manner where smaller adipocyte phenotype predicts healthier outcome, and larger adipocytes are linked to insulin resistance and other comorbidities of excess weight [17,18]. It is not fully understood why the presence of larger adipocytes presents a worse metabolic outcome in obesity; however, this may at least not link to adipocyte mitochondrial dysfunction [19] but rather on hypertrophic adipocyte-mediated initiation of innate immune reaction [20,21,22]. In obesity, prolonged inflammation compromises lipid storage and healthy WAT function, which lead to lipid accumulation to non-metabolic tissues [16,23,24]. This is linked to the development of insulin resistance and T2D [23].
TG2, which is a ubiquitously expressed TG with a plethora of functions in health and disease, has been linked to adipogenesis and obesity by us [25] and by others [26]. We demonstrated that the absence of TG2 in mouse embryonic fibroblasts increased adipogenesis via several pathways, which included decreased Pref-1 production, canonical Wnt/β-catenin signaling, and increased Akt phosphorylation [25]. The research group of Szondy showed that Tgm2-deficient mice do not gain more weight compared to their controls, however they exhibit increased inflammation of adipose tissue.
A recent genome-wide association study by Pietiläinen’s group linked FXIII-A to obesity in weight discordant monozygotic (MZ) twins, and further studies showed that seven small nucleotide polymorphisms (SNPs) in the F13A1 gene are associated with body mass index (BMI) [27]. The work by Kaartinen’s group in turn examined the role and mechanisms of action of FXIII-A in 3T3-L1 adipocytes and investigated the metabolic phenotype of high-fat diet-fed F13a1-/- mice. We reported that in cell cultures, the FXIII-A enzyme can act as a switch to modulate insulin signaling from pro-proliferative to pro-differentiating function via regulating plasma fibronectin accumulation into extracellular fibrils [28]. We also showed that F13a1 deficiency in mice alters the adipose tissue cellularity and increases small and large adipocytes but decreases macrophage infiltration and improves insulin sensitivity of obese adipose tissue [29], suggesting that FXIII-A reacts to weight gain and may have an adverse effect on adipose tissue health. In our most recent work, we examined the association of F13A1 to human obesity in weight discordant MZ twin pairs, where the association of F13A1 to human obesity was originally discovered [27]. In this clinical study, we reported that the expression of F13A1 is significantly increased in the adipose tissue and adipocyte-enriched fraction of the heavier co-twin, and its differential expression (ΔHeavy–Lean) shows a strong, significant linear correlation with increase in weight, fat mass, and adipocyte volume, and a negative correlation with circulating adiponectin [30]. A further transcriptome-wide association study (TWAS) between Heavy–Lean ΔF13A1 and changes in full adipocyte transcriptome between the co-twins identified 182 highly F13A1 correlating transcripts, for which gene enrichment analysis linked cell stress, inflammation, extracellular matrix remodeling, and angiogenesis of the adipose tissue.
The aim of this study was first to investigate the expression of the whole TGM family in acquired excess weight in the MZ twins, and secondly to elucidate details of the links of F13A1 to pathways associated with adipocyte size that may be relevant to metabolism. We report that F13A1 is the only TGM family gene transcript that is significantly linked to acquired weight gain, showing an increase in the heavier twin. F13A1 associated with 47 gene transcripts that also showed a link to large adipocyte size, which mostly represented genes of neutrophil attraction, inflammasome, and secretion of pro-inflammatory cytokines. Our study brings further evidence to the relevance of FXIII-A/F13A1 in adipose tissue health.

2. Results

Our first aim was to examine potential links of TGM family members to excess acquired weight in the 12 weight discordant MZ twins. The significant weight and adipocity discordance of the twins are presented in Table 1. Differential (ΔHeavy–Lean twin) gene expression in adipose tissue and adipocyte-enriched fraction is reflective of cellular changes in the tissue upon its expansion during weight gain. As presented in Figure 1A,B, F13A1 is the only TGM family gene that reacts to excess weight in a significant manner and shows increased expression in both adipose tissue and adipocyte-enriched fraction of the heavier twin [30].
Our previous work linked increase in F13A1 expression to increase in adipocyte size/volume [30]. This, with previous mouse data, suggests that F13A1 may be increasingly produced by larger adipocytes or may regulate the cell size. In the same study, we identified 182 F13A1- and weight-associated genes which showed significant (p < 0.05) and strong Pearson correlation (r > 0.7) for Heavy–Lean ΔF13A1 [30]. Gene enrichment analysis showed that the 182 genes linked to cell stress, inflammation, tissue remodeling, hemostasis, and angiogenesis [30]. To further examine the link of F13A1 to adipocyte size, we applied the following strategy (Figure 2). First, the pool of 182 genes was further screened for genes that also showed correlation to Heavy–Lean ΔAdipocyte Diameter in the 12 MZ twin pairs. This resulted in a total of 283 genes from which 42 were found as shared with the original 182 gene set. Second, we screened the set for adipocyte size correlating (positive and negative) genes that were identified from 15 MZ twins, some of which were shared with this study [17]. This resulted in no additional genes (Supplemental Tables S1 and S2). Third, we screened 14 reported ‘large adipocyte genes’ from a study where adipocytes, isolated from human SAT, were separated via 70 μm mesh into small (57.8 ± 0.23 μm) and large (95 ± 0.38 μm) adipocytes, followed by mRNA expression analysis and identification of genes that were expressed more than four-fold in large adipocytes versus small ones [18,31] (Supplemental Table S3). From this set, three additional F13A1- and weight-correlated genes were identified (SELE, IL8, and LPCAT1). Fourth, we searched the literature for genes that have been published to regulate adipocyte size. A literature search (‘adipocyte size’ and ‘regulation’ and ‘gene’) resulted in 835 papers, from which 17 reported on genes with strong links to regulation of adipocyte size (associations were not included) (Supplemental Table S4). Only one out of these genes was found in the set of 182 genes (REPIN1). Thus, a total of 383 ‘adipocyte size’ genes were screened, which resulted in a final set of 47 gene transcripts that showed significant correlation to ΔF13A1 and were differentially expressed in the Heavy vs. Lean twin. Classical regulators of adipogenesis and lipid storage, PPARG, CEBPA, CEBPB, and PLIN1, did not show correlation. The set of 47 genes are listed in Table 2. From this set, 12 genes showed strong negative correlation with ΔWeight-, ΔF13A1-, and ΔAdipocyte Diameter. The remaining 35 genes were positive correlators.
Gene enrichment analysis for the 47 genes, with GOnet and Panther gene ontology tools, resulted in a total of 61 GOterms for Biological Process (BP), 13 Cellular Component (CC), and no terms for Molecular Functions. Figure 3 shows a selection [28] of the most significant, over-represented Biological Process GOterms and all 13 Cellular Component terms. The full list of GOterms is listed in Supplemental Tables S5 and S6. Further cluster analysis (Figure 4) for the GOterms using the GOnet tool revealed three clear functional clusters related to immune response, inflammation, and cytokine response and signaling. A detailed examination of each of the clusters revealed central GOterms, suggesting inflammatory response to external factors via neutrophil and leucocyte activation as well as cytokine secretion (Figure 5, Figure 6 and Figure 7). Each of the Biological Process and Cellular Component terms and some relevant genes are discussed below in more detail.

3. Discussion

Adipose tissue expansion through hypertrophy, where large adipocytes predominate in the expanding tissue, correlates with adipose tissue low-grade systemic inflammation and worse metabolic consequences of obesity [33,34,35,36]. The mechanisms and pathways that precede and follow the hypertrophic expansion and inflammation are numerous. Accumulating data points to sequela that involves free fatty acid (FFA)-mediated initiation of adipocyte hypertrophy, which is followed by initiation of innate immune response by the hypertrophic adipocytes [34]. Previous studies have linked two TGM family members, TGM2 and F13A1, to adipose tissue biology and inflammation [25,26,28,29,30,37,38,39]. In this study, we have investigated the differential expression of the whole TGM family in adipose tissue of MZ twins that are discordant in weight (Heavy–Lean twin pairs). The MZ twins are a unique model of acquired weight gain where background involving genetic, age, sex, and early environmental influences can be controlled. The twins’ adipocity parameters and metabolism are well characterized and include adipocyte morphology, mitochondrial, fibronolytic and coagulatory, and inflammatory status [40,41,42,43,44,45,46,47]. We report that F13A1 is the sole TGM family member that shows significantly altered, and in this case increased, expression in the heavier twin. Despite robust data from mouse studies linking TG2 to adipose tissue function in obesity [25,26], TGM2 showed no altered expression between the Heavy–Lean co-twins. Further studies in larger cohorts of more severely obese individuals could shed more light into the role of TG2 in human adipose tissue pathology.
The presence of large versus small adipocytes in adipose tissue in obesity has been associated with worse metabolic outcome [18]. Our previous work linked increase in F13A1 in adipose tissue to increased adipocyte size [30]. The aim of this current study was to further examine through which pathways F13A1 associates to large adipocytes. This was done by screening the pool of the previously identified 182 F13A1- and weight-correlating genes [30] in the adipocyte-enriched fraction for: (1) genes that have been reported to associate positively or negatively to adipocyte size [17], and (2) genes that are expressed significantly more in larger adipocytes or demonstrated in the literature to regulate adipocyte size [18,31]. Here, we demonstrate tight association of F13A1 to 47 ‘adipocyte size’ genes that via gene enrichment analysis linked to Biological Processes involving response to external stimulus, secretion, exocytosis, vesicle-mediated transport, cell surface receptor signaling, neutrophil activation, and response to interleukin 1. The GOterms for Cellular Components suggested association of F13A1 in lysosome, secretory vesicles, plasma membrane rafts, and extracellular space. Network and cluster analysis of the GOterms link these to immune response/inflammation and cytokine response (Figure 4), which jointly suggest that F13A1 may be involved in pro-inflammatory response from large adipocytes. In our previous study, we discussed several genes that are also among the current 47 genes and they were associated with adipocyte oxidative stress, extracellular matrix (ECM) organization and remodeling, angiogenesis, and macrophages [30]. Associations to all the above processes suggests that F13A1 may be involved throughout in molecular events of the tissue expansion.
The adipocyte-enriched fraction used in this study was prepared from SAT biopsies by a method involving collagenase digestion followed by collecting of the released, buoyant adipocytes. This method is considered to separate adipocytes from the stromal vascular fraction that contain pre-adipocytes, mesenchymal stem cells, endothelial and vascular smooth muscle cells, and immune cells [48], however, other cell types do co-purify and contribute to the transcripts observed. Thus, the preparation enriches adipocytes, but is not void of other cell types. It has become apparent that pure adipocyte preparations require flow cytometry sorting and is more difficult than previously considered [49]. The other cell types have likely co-purified and may include macrophages whose marker ITGAM (Integrin αM; CD11b) is present also in this final set of 47 genes. The preparation also likely includes endothelial cells, which as suggested by the expression of genes like SELE (Selectin E) [30], is a product of cytokine-stimulated endothelial cells and mediates endothelial cell adhesion to leucocytes [50]. Macrophages and endothelial cells also both produce FXIII-A enzyme [3,51,52,53,54,55,56,57,58,59]. Our major future aims include detailing the cell types that are responsible for F13A1/F13a1 production at different phases of adipose tissue expansion, which is critical to understanding where in the inflammatory sequela F13A1/F13a1 may be involved.
Of the 47 gene transcripts that correlate with weight, F13A1, and adipocyte size, we have discussed four in our previous study (CYBB, CTSS, ITMG, and VCAN) [30]. Below, we will focus mostly on the new ones identified here. The most numerous GOterms for Biological Processes were associated with neutrophil and leucocyte activation (Figure 5). One of the highest F13A1 correlating genes in this set was PYCARD (r2 = 0.89) (Figure 5B), which encodes for ASC (apoptosis-associated speck-like protein containing a CARD). PYCARD/ASC is made up by macrophages, where it activates inflammasome via acting as an adaptor protein between Nlrp3 and caspase-1 [60]. Caspase-1 is critical for proteolytic activation and secretion of pro-inflammatory cytokines IL1β and IL-18 [61,62]. The presence of FFAs in adipose tissue can induce activation of inflammasome [61] and its essential role in metabolic disturbances has become clear [61,63,64]. A very recent work demonstrated that the absence of these three critical inflammasome components, Nlrp3, ASC, and caspase-1, in mice protects mice from HFD-induced obesity, insulin resistance, and adipocyte hypertrophy [64]. The Heavy–Lean increase in ΔIL1B also shows strong linear correlation to ΔF13A1 (r2 = 0.62) (Figure 5B). IL1β is one of the major pro-inflammatory cytokines whose role in metabolic dysfunction is well documented [65].
Neutrophils are the first responders of acute inflammatory reaction and are the first cells to be recruited to the site of inflammation [66,67,68,69]. They are the most populous but short-lived cell type in early inflammation and are capable of producing large quantities of cytokines and chemokines, including TNF-α, IL-1β, and IL-8, which can initiate the second wave of immune cells, such as infiltration of macrophages and lymphocytes [66,67,68,69]. The role of neutrophils in adipose tissue inflammation has also become apparent [33,70,71]. The links of F13A1 to both neutrophil and leucocyte activation (Figure 5) supports a role in early inflammation as well as perpetuation of the pro-inflammatory milieu. The genes contributing to GOterms that link F13A1 to neutrophil activation are, among others, CYBB (NADP oxidase) (Nox2), which is made by neutrophils and macrophages and responsible for generation of reactive oxygen species and oxidative stress, which in turn can activate neutrophils and neutrophil extracellular trap formation [72]. In adipose tissue, CYBB/Nox2 contributes to adipose tissue inflammation and development of insulin resistance [73]. The presence of TNFSF13B (TNF Superfamily Member 13b) (also known as BAFF) in the set of 47 weight and F13A1 correlating genes also supports neutrophil links (Figure 5B). TNFSF13B/BAFF is a neutrophil-derived cytokine that supports B lymphocyte survival. B lymphocytes can tune adipose tissue inflammation via secreting cytokines and immunoglobulins, and they contribute to metabolic failure of adipose tissue [63,74,75]. BAFF is also produced by adipocytes [76]. The role of F13A1 in inflammation in many tissues is supported by vast literature and it is expressed by macrophages and dendritic cells [51,52,77,78,79,80,81,82]. The mechanisms by which FXIII-A contributes to inflammation involve its fibrin-stabilizing function [83,84] and monocyte/macrophage adhesion [79,85]. It may also crosslink other substrates such as plasma fibronectin [28,29]. It is not known if F13A1 is produced by neutrophils themselves.
Cluster analysis of GOterms also revealed that F13A1 links to cytokine-mediated signaling pathways (Figure 6). Here, we demonstrate two indicators that show that F13A1 is also negatively associated with important insulin modifiers, which is apparent from its negative correlation to PTPRF and STAT5A. The ΔF13A1PTPRF linear regression graph in Figure 6B shows that the high increase in F13A1 correlated with a smaller increase in PTPRF. PTPRF encodes for Protein Tyrosine Phosphatase Receptor Type F (also known as LAR), which inhibits the insulin receptor activation [86,87]. PTPRF/LAR knockdown in adipocytes increases adipogenesis [88] and in mice, disturbances in glucose homeostasis [89]. This data interestingly links high F13A1 to promotion to insulin signaling, which we demonstrated in 3T3-L1 adipocytes [28]; however, in mice, the F13A1 absence improves insulin sensitivity [29]. Also, negative association to STAT5A (Figure 6B) (Signal Transducer and Activator of Transcription 5A), a known promoter of adipogenesis and adipocyte differentiation, supports our previous cell culture results, where FXIII-A was required for pre-adipocyte proliferation but was rapidly downregulated during adipogenesis [28]. The link to large adipocytes suggests that it is also made later during hypertrophy, however this could also mean that large adipocytes induce F13A1 in other adipose tissue cells. Interestingly, recent advances in understanding the phenotypes and functionality of large, hypertrophic adipocytes in adipose tissue in obesity has shown that they likely function as antigen-presenting cells to active innate immunity/immune cells via MHCII and play an important role in the initiation of adipose tissue inflammation [20,90]. Mature adipocytes have also been demonstrated to express leucocyte chemo-attractants that via direct adipocyte–leucocyte interaction stimulate T-cell proliferation [91]. The correlation of F13A1 to HLA-DRA (Figure 5B), an HLA class II histocompatibility complex component, found on the surface of antigen presenting cells, could suggest a link to large adipocytes specialized to initiation of inflammation.
The stimuli initiating F13A1 production, cell types producing it, and cellular location/externalization and activation in adipose tissue are major questions arising from this study. Here, the increase of F13A1 is linked to GOterms for cellular response to external stimuli and response to lipopolysaccharide (LPS), supporting the idea that F13A1 could be induced by those factors that initiate the inflammatory reaction. The stimuli that can initiate adipose tissue inflammation are complex and numerous [33,34] and include fibrosis and alterations in extracellular matrix components and stiffness [83,92,93,94,95,96], apoptosis and necrosis of hypertrophic adipocytes [97], hypoxia from defective angiogenesis [24,98], free fatty acids (lipotoxicity) [21], and exogenous, as well as gut microbiota-derived LPS [36]. The GOterms for this cluster of Biological Processes are presented in Figure 7, and there are two genes of particular interest PLA2G16 and MAPK1.PLA2G16—phospholipase 2A—a lipolytic enzyme made in hypertrophic adipocytes that produces FFAs and induces hyperlipidemia [99,100]. Coregulation between PLA2G16 and F13A1 could thus imply a link to FFAs. Positive association with MAPK1 (Figure 6B) is also worthy of mention as we demonstrated that in osteoblasts, extracellular collagen type I, also produced by adipocytes, is a major regulator of FXIII-A expression and externalization [15]. MAPK1 is also needed for lipolysis in rat and mouse adipocytes [101,102].
Our original screen contained a set of regulatory genes demonstrated to modulate adipocyte size. From the final set of genes, 46 of the 47 were large adipocyte-associated genes and only one, REPIN1, was a gene that has been demonstrated to regulate adipocyte size and whole-body insulin sensitivity [103]. The association between F13A1 and REPIN1 is negative (r = −0.77; r2 = 0.70; p = 0.0002), i.e., higher ΔREPIN1 levels link to lower ΔF13A1. This and the positive association of ΔF13A1 to ΔAdipocyte Diameter and ΔAdipocyte Volume [30] strongly suggests that majority of F13A1 is a result of adipocyte hypertrophy-induced events in weight gain, rather than it being a gene that influences the adipocyte size. Detailed weight gain studies in mice can address when F13a1 transcription and protein production is initiated, when and how it is activated during adipose tissue expansion, and what are the consequences of its absence or its overexpression to adipocytes and adipose tissue inflammatory cells and processes. These studies are ongoing in our laboratory.
In conclusion, our study brings further evidence to the involvement of F13A1 in the pathological response of adipose tissue to expansion in weight gain in humans. The concepts revealed in our study suggest novel pathways linked to F13A1 and inflammation providing many new hypotheses to be explored that will hopefully show if FXIII-A could be a target to control inflammation of adipose tissue.

4. Materials and Methods

4.1. Weight-Discordant MZ Twins

This study involved 12 rare, healthy MZ twin pairs discordant for weight (intra-pair difference in acquired excess weight ΔBMI (body mass index) ≥ 3 kg/m2 (males n = 2, females n = 10, aged 27, 7 ± 1.4 years)) which were identified from two population-based twin cohorts, FinnTwin16 (n = 2839 pairs) and FinnTwin12 (n = 2578 pairs) [104]. A detailed description of the twin material has been published previously [45,105]. The twins were healthy and were not on any regular medication, excluding contraceptives. Table 1 describes the clinical characteristics of the 12 twin pairs regarding weight and adipocity. This data table has been also presented in Reference [30]. All participants have given a written informed consent. The study design was approved by the Ethical Committee of the Helsinki University Central Hospital.

4.2. Adipose Tissue Biopsies, Adipocyte and Tissue Preparations, and RNA Extraction

Abdominal subcutaneous adipose tissue (SAT) biopsies were obtained surgically under the umbilicus under local anesthesia. Specimens were flash frozen in −80 °C liquid nitrogen. Adipocyte-enriched fraction was prepared by a previously reported protocol which involved adipose tissue digestion in 2% collagenase dissolved in DMEM/F-12 with supplements and 2% bovine serum albumin [17]. This was followed by adipocyte mRNA extraction using RNeasy Lipid Tissue Mini Kit (Qiagen, Nordic, Solletuna, Sweden) with a DNase I (Qiagen) digestion according to the manufacturer’s instructions.

4.3. Adipocyte Morphology

Fresh SAT biopsy specimens were digested with 2% collagenase in DMEM/F12 with supplements [17] and 2% bovine serum albumin (as above) and were used for measurement and calculation of adipocyte size in all 12 discordant twin pairs, as before [17]. After collagenase treatment, the digests were washed and maintained in the above medium and images of live adipocytes were taken with a light microscope (Zeiss, Axioplan2) using 50× magnification. A minimum of 200 cells were measured for diameter using an image processing and analysis software ImageJ (ImageJ 1.42q/Java 1. 6.0_10 32-bit), as previously described [17].

4.4. Affymetrix Transcriptomics

A total of 500 ng of RNA from adipocyte-enriched fraction, isolated as described above, was used for gene expression analysis on an Affymetrix U133 Plus 2.0 array (Thermo Fischer, Santa Clara, CA, USA) according to the manufacturer’s instructions and validated as described previously [45]. Hybridization, staining, and washing were performed using the Affymetrics Fluidics Station 450 and Hybridization oven 640 under standard conditions. Expression data was pre-processed with BioConductor software (open source, www.bioconductor.org) and the GC-RMA algorithm [106].

4.5. Transcriptome-Wide Association Study, Statistics, and Bioinformatics

Intra-pair values (ΔHeavy–Lean) for twins’ transcriptome (differential gene expression, Δ) were calculated and screened for correlation to intra-pair ΔF13A1 expression in adipocyte-enriched fraction using R Statistical Software (https://www.r-project.org/) [107]. Significance within the TGM family (TGM1-7 and F13A1) expression in twin pairs was calculated via analysis of variance (ANOVA) with Bonferroni post-test using Prism GraphPad software (release 8.0, San Diego, CA, USA). p-Values < 0.05 were considered significant. Pearson (r) correlation and r2 parameters were obtained for ΔF13A1 and ΔAdipocyte Diameter, and those with r2 > 0.5 and associated p-values < 0.05 were considered for further screening. Normal distribution of F13A1 mRNA expression values and adipocyte diameter values were assessed as before [30]. Significance of differential gene expression between the co-twins was tested with a paired T-test and associated p-values < 0.05 were considered significant. Further linear regression analysis was performed for selected genes with significant associations to ΔF13A1 with Prism GraphPad software (release 8.0, San Diego, CA, USA). Here, p-values were considered significant as follows: * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001. Gene Ontology enrichment analysis and GOterm cluster analysis for over-represented genes from transcriptome screening was performed using Panther, GOnet, and Gorilla online tools. Significance for GOterms is represented in False Discovery Rate (FDR) adjusted p-values.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/1422-0067/21/21/8289/s1. References [17,31,108,109,110,111,112,113,114,115,116,117,118,119,120] are cited in the supplementary materials.

Author Contributions

Conceptualization, Writing, Review and Editing, Data Analysis and Funding Acquisition, M.T.K.; Conceptualization, Writing, Review and Editing, Clinical Data Collection, Data Analysis, K.H.P.; Data Analysis, Writing, S.H.; Data Analysis, M.A.; Clinical Data Collection, A.R. and J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This study has been supported by a grant to M.T.K. from the Canadian Institutes of Health Research (CIHR) (MOP-119403). M.A. is supported by an internal stipend, Nyman Award, from the Faculty of Medicine of McGill University. S.H. was supported by Finnish Diabetes Research Foundation, Emil Aaltonen Foundation, Finnish Medical Foundation, Maud Kuistila Foundation, Orion Foundation, Paulo foundation and Helsinki University Hospital funds. K.H.P. is funded by the Academy of Finland, grant numbers 314383, 266286, and the Academy of Finland, Centre of Excellence in Research on Mitochondria, Metabolism and Disease (FinMIT), grant number 272376, the Finnish Medical Foundation, the Gyllenberg Foundation, the Novo Nordisk Foundation, grant numbers NNF17OC0027232, NNF10OC1013354, the Finnish Diabetes Research Foundation, the Finnish Foundation for Cardiovascular Research, the University of Helsinki, Government Research Funds and Helsinki University Hospital.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Eckert, R.L.; Kaartinen, M.T.; Nurminskaya, M.; Belkin, A.M.; Colak, G.; Johnson, G.V.W.; Mehta, K. Transglutaminase Regulation of Cell Function. Physiol. Rev. 2014, 94, 383–417. [Google Scholar] [CrossRef] [Green Version]
  2. Iismaa, S.E.; Mearns, B.M.; Lorand, L.; Graham, R.M. Transglutaminases and Disease: Lessons from Genetically Engineered Mouse Models and Inherited Disorders. Physiol. Rev. 2009, 89, 991–1023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Muszbek, L.; Bereczky, Z.; Bagoly, Z.; Komáromi, I.; Katona, É. Factor XIII: A Coagulation Factor with Multiple Plasmatic and Cellular Functions. Physiol. Rev. 2011, 91, 931–972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Lorand, L.; Graham, R.M. Transglutaminases: Crosslinking enzymes with pleiotropic functions. Nat. Rev. Mol. Cell Biol. 2003, 4, 140–156. [Google Scholar] [CrossRef] [PubMed]
  5. Forsprecher, J.; Wang, Z.; Goldberg, H.A.; Kaartinen, M.T. Transglutaminase-mediated oligomerization promotes osteoblast adhesive properties of osteopontin and bone sialoprotein. Cell Adhes. Migr. 2011, 5, 65–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Kaartinen, M.T.; Pirhonen, A.; Linnala-Kankkunen, A.; Mäenpää, P.H. Cross-linking of Osteopontin by Tissue Transglutaminase Increases Its Collagen Binding Properties. J. Biol. Chem. 1999, 274, 1729–1735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Nelea, V.; Nakano, Y.; Kaartinen, M.T. Size Distribution and Molecular Associations of Plasma Fibronectin and Fibronectin Crosslinked by Transglutaminase. Protein J. 2008, 27, 223–233. [Google Scholar] [CrossRef]
  8. Kanchan, K.; Fuxreiter, M.; Fésüs, L. Physiological, pathological, and structural implications of non-enzymatic protein–protein interactions of the multifunctional human transglutaminase. Cell. Mol. Life Sci. 2015, 72, 3009–3035. [Google Scholar] [CrossRef] [PubMed]
  9. Kaartinen, M.T.; El-Maadawy, S.; Räsänen, N.H.; McKee, M.D. Tissue Transglutaminase and Its Substrates in Bone. J. Bone Miner. Res. 2002, 17, 2161–2173. [Google Scholar] [CrossRef] [PubMed]
  10. Mousa, A.; Cui, C.; Song, A.; Myneni, V.D.; Sun, H.; Li, J.J.; Murshed, M.; Melino, G.; Kaartinen, M.T. Transglutaminases factor XIII-A and TG2 regulate resorption, adipogenesis and plasma fibronectin homeostasis in bone and bone marrow. Cell Death Differ. 2017, 24, 844–854. [Google Scholar] [CrossRef] [Green Version]
  11. Nurminskaya, M. Transglutaminases in mineralized tissues. Front. Biosci. 2006, 11, 1591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Al-Jallad, H.F.; Myneni, V.D.; Piercy-Kotb, S.A.; Chabot, N.; Mulani, A.; Keillor, J.W.; Kaartinen, M.T. Plasma Membrane Factor XIIIA Transglutaminase Activity Regulates Osteoblast Matrix Secretion and Deposition by Affecting Microtubule Dynamics. PLoS ONE 2011, 6, e15893. [Google Scholar] [CrossRef] [Green Version]
  13. Cui, C.; Wang, S.; Myneni, V.D.; Hitomi, K.; Kaartinen, M.T. Transglutaminase activity arising from Factor XIIIA is required for stabilization and conversion of plasma fibronectin into matrix in osteoblast cultures. Bone 2014, 59, 127–138. [Google Scholar] [CrossRef] [PubMed]
  14. Heath, D.J.; Downes, S.; Verderio, E.; Griffin, M. Characterization of Tissue Transglutaminase in Human Osteoblast-like Cells. J. Bone Miner. Res. 2001, 16, 1477–1485. [Google Scholar] [CrossRef]
  15. Piercy-Kotb, S.A.; Mousa, A.; Al-Jallad, H.F.; Myneni, V.D.; Chicatun, F.; Nazhat, S.N.; Kaartinen, M.T. Factor XIIIA transglutaminase expression and secretion by osteoblasts is regulated by extracellular matrix collagen and the MAP kinase signaling pathway. J. Cell. Physiol. 2012, 227, 2936–2946. [Google Scholar] [CrossRef]
  16. Cristancho, A.G.; Lazar, M.A. Forming functional fat: A growing understanding of adipocyte differentiation. Nat. Rev. Mol. Cell Biol. 2011, 12, 722–734. [Google Scholar] [CrossRef] [PubMed]
  17. Heinonen, S.; Saarinen, L.; Naukkarinen, J.; Rodríguez, A.; Frühbeck, G.; Hakkarainen, A.; Lundbom, J.; Lundbom, N.; Vuolteenaho, K.; Moilanen, E.; et al. Adipocyte morphology and implications for metabolic derangements in acquired obesity. Int. J. Obes. 2014, 38, 1423–1431. [Google Scholar] [CrossRef]
  18. Tandon, P.; Wafer, R.; Minchin, J.E.N. Adipose morphology and metabolic disease. J. Exp. Biol. 2018, 221, jeb164970. [Google Scholar] [CrossRef] [Green Version]
  19. Yin, X.; Lanza, I.R.; Swain, J.M.; Sarr, M.G.; Nair, K.S.; Jensen, M.D. Adipocyte Mitochondrial Function Is Reduced in Human Obesity Independent of Fat Cell Size. J. Clin. Endocrinol. Metab. 2014, 99, E209–E216. [Google Scholar] [CrossRef] [Green Version]
  20. Xiao, L.; Yang, X.; Lin, Y.; Li, S.; Jiang, J.; Qian, S.; Tang, Q.; He, R.; Li, X. Large adipocytes function as antigen-presenting cells to activate CD4+ T cells via upregulating MHCII in obesity. Int. J. Obes. 2015, 40, 112–120. [Google Scholar] [CrossRef] [Green Version]
  21. Engin, A.B. What Is Lipotoxicity? Adv. Exp. Med. Biol. 2017, 960, 197–220. [Google Scholar]
  22. Engin, A.B. Adipocyte-Macrophage Cross-Talk in Obesity. Adv. Exp. Med. Biol. 2017, 960, 327–343. [Google Scholar] [CrossRef] [PubMed]
  23. Kahn, S.E.; Hull, R.L.; Utzschneider, K.M. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nat. Cell Biol. 2006, 444, 840–846. [Google Scholar] [CrossRef] [PubMed]
  24. Sun, K.; Kusminski, C.M.; Scherer, P.E. Adipose tissue remodeling and obesity. J. Clin. Investig. 2011, 121, 2094–2101. [Google Scholar] [CrossRef] [Green Version]
  25. Myneni, V.D.; Melino, G.; Kaartinen, M.T. Transglutaminase 2—A novel inhibitor of adipogenesis. Cell Death Dis. 2015, 6, e1868. [Google Scholar] [CrossRef] [Green Version]
  26. Sághy, T.; Köröskényi, K.; Hegedűs, K.; Antal, M.; Bankó, C.; Bacsó, Z.; Papp, A.; Stienstra, R.; Szondy, Z. Loss of transglutaminase 2 sensitizes for diet-induced obesity-related inflammation and insulin resistance due to enhanced macrophage c-Src signaling. Cell Death Dis. 2019, 10, 439. [Google Scholar] [CrossRef] [Green Version]
  27. Naukkarinen, J.; Surakka, I.; Pietiläinen, K.H.; Rissanen, A.; Salomaa, V.; Ripatti, S.; Yki-Jarvinen, H.; Van Duijn, C.M.; Wichmann, H.-E.; Kaprio, J.; et al. Use of Genome-Wide Expression Data to Mine the “Gray Zone” of GWA Studies Leads to Novel Candidate Obesity Genes. PLoS Genet. 2010, 6, e1000976. [Google Scholar] [CrossRef] [PubMed]
  28. Myneni, V.D.; Hitomi, K.; Kaartinen, M.T. Factor XIII-A transglutaminase acts as a switch between preadipocyte proliferation and differentiation. Blood 2014, 124, 1344–1353. [Google Scholar] [CrossRef]
  29. Myneni, V.D.; Mousa, A.; Kaartinen, M.T. Factor XIII-A transglutaminase deficient mice show signs of metabolically healthy obesity on high fat diet. Sci. Rep. 2016, 6, 35574. [Google Scholar] [CrossRef] [Green Version]
  30. Kaartinen, A.; Heinonen, L.; Hakkarainen, L.; Rissanen, K.; Pietiläinen, K.H. F13A1 transglutaminase expression in human adipose tissue increases in acquired excess weight and associates with inflammatory status of adipocytes. Int. J. Obes. in press.
  31. Jernås, M.; Palming, J.; Sjöholm, K.; Jennische, E.; Svensson, P.-A.; Gabrielsson, B.G.; Levin, M.; Sjögren, A.; Rudemo, M.; Lystig, T.C.; et al. Separation of human adipocytes by size: Hypertrophic fat cells display distinct gene expression. FASEB J. 2006, 20, 1540–1542. [Google Scholar] [CrossRef]
  32. Lee, B.-C.; Lee, J. Cellular and molecular players in adipose tissue inflammation in the development of obesity-induced insulin resistance. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2014, 1842, 446–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Burhans, M.S.; Hagman, D.K.; Kuzma, J.N.; Schmidt, K.A.; Kratz, M. Contribution of Adipose Tissue Inflammation to the Development of Type 2 Diabetes Mellitus. Compr. Physiol. 2018, 9, 1–58. [Google Scholar] [CrossRef]
  34. Reilly, S.M.; Saltiel, A.R. Adapting to obesity with adipose tissue inflammation. Nat. Rev. Endocrinol. 2017, 13, 633–643. [Google Scholar] [CrossRef]
  35. Stolarczyk, E. Adipose tissue inflammation in obesity: A metabolic or immune response? Curr. Opin. Pharmacol. 2017, 37, 35–40. [Google Scholar] [CrossRef]
  36. Trim, W.; Turner, J.E.; Thompson, D. Parallels in Immunometabolic Adipose Tissue Dysfunction with Ageing and Obesity. Front. Immunol. 2018, 9, 169. [Google Scholar] [CrossRef] [Green Version]
  37. Caccamo, D.; Currò, M.; Ferlazzo, N.; Condello, S.; Ientile, R. Monitoring of transglutaminase2 under different oxidative stress conditions. Amino Acids 2011, 42, 1037–1043. [Google Scholar] [CrossRef] [PubMed]
  38. Currò, M.; Ferlazzo, N.; Risitano, R.; Condello, S.; Vecchio, M.; Caccamo, D.; Ientile, R. Transglutaminase 2 and phospholipase A2 interactions in the inflammatory response in human Thp-1 monocytes. Amino Acids 2013, 46, 759–766. [Google Scholar] [CrossRef]
  39. Ientile, R.; Caccamo, D.; Griffin, M. Tissue transglutaminase and the stress response. Amino Acids 2007, 33, 385–394. [Google Scholar] [CrossRef]
  40. Heinonen, S.; Buzkova, J.; Muniandy, M.; Kaksonen, R.; Ollikainen, M.; Ismail, K.; Hakkarainen, A.; Lundbom, J.; Lundbom, N.; Vuolteenaho, K.; et al. Impaired Mitochondrial Biogenesis in Adipose Tissue in Acquired Obesity. Diabetes 2015, 64, 3135–3145. [Google Scholar] [CrossRef] [Green Version]
  41. Heinonen, S.; Jokinen, R.; Rissanen, A.; Pietiläinen, K.H. White adipose tissue mitochondrial metabolism in health and in obesity. Obes. Rev. 2019, 21, e12958. [Google Scholar] [CrossRef]
  42. Heinonen, S.; Muniandy, M.; Buzkova, J.; Mardinoglu, A.; Rodríguez, A.; Frühbeck, G.; Hakkarainen, A.; Lundbom, J.; Lundbom, N.; Kaprio, J.; et al. Mitochondria-related transcriptional signature is downregulated in adipocytes in obesity: A study of young healthy MZ twins. Diabetology 2016, 60, 169–181. [Google Scholar] [CrossRef] [Green Version]
  43. Jukarainen, S.; Heinonen, S.; Rämö, J.T.; Rinnankoski-Tuikka, R.; Rappou, E.; Tummers, M.; Muniandy, M.; Hakkarainen, A.; Lundbom, J.; Lundbom, N.; et al. Obesity Is Associated With Low NAD(+)/SIRT Pathway Expression in Adipose Tissue of BMI-Discordant Monozygotic Twins. J. Clin. Endocrinol. Metab. 2016, 101, 275–283. [Google Scholar] [CrossRef] [Green Version]
  44. Kaye, S.M.; Pietiläinen, K.H.; Kotronen, A.; Joutsi-Korhonen, L.; Kaprio, J.; Yki-Järvinen, H.; Silveira, A.; Hamsten, A.; Lassila, R.; Rissanen, A. Obesity-Related Derangements of Coagulation and Fibrinolysis: A Study of Obesity-Discordant Monozygotic Twin Pairs. Obesity 2012, 20, 88–94. [Google Scholar] [CrossRef]
  45. Naukkarinen, J.; Heinonen, S.; Hakkarainen, A.; Lundbom, J.; Vuolteenaho, K.; Saarinen, L.; Hautaniemi, S.; Rodríguez, A.; Frühbeck, G.; Pajunen, P.; et al. Characterising metabolically healthy obesity in weight-discordant monozygotic twins. Diabetology 2013, 57, 167–176. [Google Scholar] [CrossRef] [Green Version]
  46. Pietiläinen, K.H.; Ismail, K.; Järvinen, E.; Heinonen, S.; Tummers, M.; Bollepalli, S.; Lyle, R.; Muniandy, M.; Moilanen, E.; Hakkarainen, A.; et al. DNA methylation and gene expression patterns in adipose tissue differ significantly within young adult monozygotic BMI-discordant twin pairs. Int. J. Obes. 2015, 40, 654–661. [Google Scholar] [CrossRef]
  47. Sahebekhtiari, N.; Saraswat, M.; Joenväärä, S.; Jokinen, R.; Lovric, A.; Kaye, S.; Mardinoglu, A.; Rissanen, A.; Kaprio, J.; Renkonen, R.; et al. Plasma Proteomics Analysis Reveals Dysregulation of Complement Proteins and Inflammation in Acquired Obesity—A Study on Rare BMI-Discordant Monozygotic Twin Pairs. Proteom. Clin. Appl. 2019, 13, e1800173. [Google Scholar] [CrossRef]
  48. Church, C.D.; Berry, R.; Rodeheffer, M.S. Isolation and Study of Adipocyte Precursors. Methods Enzymol. 2014, 537, 31–46. [Google Scholar] [CrossRef] [Green Version]
  49. Hagberg, C.E.; Li, Q.; Kutschke, M.; Bhowmick, D.; Kiss, E.; Shabalina, I.G.; Harms, M.J.; Shilkova, O.; Kozina, V.; Nedergaard, J.; et al. Flow Cytometry of Mouse and Human Adipocytes for the Analysis of Browning and Cellular Heterogeneity. Cell Rep. 2018, 24, 2746–2756.e5. [Google Scholar] [CrossRef] [Green Version]
  50. Martin, J.; Collot-Teixeira, S.; McGregor, L.; McGregor, J.L. The dialogue between endothelial cells and monocytes/macrophages in vascular syndromes. Curr. Pharm. Des. 2007, 13, 1751–1759. [Google Scholar] [CrossRef]
  51. Bagoly, Z.; Katona, É.; Muszbek, L. Factor XIII and inflammatory cells. Thromb. Res. 2012, 129, S77–S81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Sun, H.; Kaartinen, M.T. Transglutaminases in Monocytes and Macrophages. Med. Sci. 2018, 6, 115. [Google Scholar] [CrossRef] [Green Version]
  53. Mitchell, J.L.; Mutch, N.J. Let’s cross-link: Diverse functions of the promiscuous cellular transglutaminase factor XIII-A. J. Thromb. Haemost. 2019, 17, 19–30. [Google Scholar] [CrossRef] [Green Version]
  54. Dardik, R.; Inbal, A. Complex formation between tissue transglutaminase II (tTG) and vascular endothelial growth factor receptor 2 (VEGFR-2): Proposed mechanism for modulation of endothelial cell response to VEGF. Exp. Cell Res. 2006, 312, 2973–2982. [Google Scholar] [CrossRef]
  55. Dardik, R.; Leor, J.; Skutelsky, E.; Castel, D.; Holbova, R.; Schiby, G.; Shaish, A.; Dickneite, G.; Loscalzo, J.; Inbal, A. Evaluation of the pro-angiogenic effect of factor XIII in heterotopic mouse heart allografts and FXIII-deficient mice. Thromb. Haemost. 2006, 95, 546–550. [Google Scholar] [CrossRef] [PubMed]
  56. Dardik, R.; Loscalzo, J.; Inbal, A. Factor XIII (FXIII) and angiogenesis. J. Thromb. Haemost. 2005, 4, 19–25. [Google Scholar] [CrossRef]
  57. Dardik, R.; Shenkman, B.; Tamarin, I.; Eskaraev, R.; Harsfalvi, J.; Varon, D.; Inbal, A. Factor XIII mediates adhesion of platelets to endothelial cells through alpha(v)beta(3) and glycoprotein IIb/IIIa integrins. Thromb. Res. 2002, 105, 317–323. [Google Scholar] [CrossRef]
  58. Dardik, R.; Solomon, A.; Loscalzo, J.; Eskaraev, R.; Bialik, A.; Goldberg, I.; Schiby, G.; Inbal, A. Novel Proangiogenic Effect of Factor XIII Associated With Suppression of Thrombospondin 1 Expression. Arter. Thromb. Vasc. Biol. 2003, 23, 1472–1477. [Google Scholar] [CrossRef] [Green Version]
  59. Inbal, A.; Dardik, R. Role of Coagulation Factor XIII (FXIII) in Angiogenesis and Tissue Repair. Pathophysiol. Haemost. Thromb. 2006, 35, 162–165. [Google Scholar] [CrossRef]
  60. Akira, S.; Misawa, T.; Satoh, T. Macrophages control innate inflammation. Diabetes Obes. Metab. 2013, 15, 10–18. [Google Scholar] [CrossRef]
  61. Wen, H.; Gris, D.; Lei, Y.; Jha, S.; Zhang, L.; Huang, M.T.-H.; Brickey, W.J.; Ting, J.P.-Y. Fatty acid–induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat. Immunol. 2011, 12, 408–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Rheinheimer, J.; De Souza, B.M.; Cardoso, N.S.; Bauer, A.C.; Crispim, D. Current role of the NLRP3 inflammasome on obesity and insulin resistance: A systematic review. Metabolism 2017, 74, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Camell, C.D.; Günther, P.; Lee, A.; Goldberg, E.L.; Spadaro, O.; Youm, Y.-H.; Bartke, A.; Hubbard, G.B.; Ikeno, Y.; Ruddle, N.H.; et al. Aging Induces an Nlrp3 Inflammasome-Dependent Expansion of Adipose B Cells That Impairs Metabolic Homeostasis. Cell Metab. 2019, 30, 1024–1039.e6. [Google Scholar] [CrossRef]
  64. Stienstra, R.; Van Diepen, J.A.; Tack, C.J.; Zaki, M.H.; Van De Veerdonk, F.L.; Perera, D.; Neale, G.A.; Hooiveld, G.J.; Hijmans, A.; Vroegrijk, I.; et al. Inflammasome is a central player in the induction of obesity and insulin resistance. Proc. Natl. Acad. Sci. USA 2011, 108, 15324–15329. [Google Scholar] [CrossRef] [Green Version]
  65. Ballak, D.B.; Stienstra, R.; Tack, C.J.; Dinarello, C.A.; Van Diepen, J.A. IL-1 family members in the pathogenesis and treatment of metabolic disease: Focus on adipose tissue inflammation and insulin resistance. Cytokine 2015, 75, 280–290. [Google Scholar] [CrossRef] [Green Version]
  66. Castanheira, F.V.S.; Kubes, P. Neutrophils and NETs in modulating acute and chronic inflammation. Blood 2019, 133, 2178–2185. [Google Scholar] [CrossRef] [PubMed]
  67. Kolaczkowska, E.; Kubes, P. Neutrophil recruitment and function in health and inflammation. Nat. Rev. Immunol. 2013, 13, 159–175. [Google Scholar] [CrossRef]
  68. Liew, P.X.; Kubes, P. The Neutrophil’s Role During Health and Disease. Physiol. Rev. 2019, 99, 1223–1248. [Google Scholar] [CrossRef]
  69. Mantovani, A.; Cassatella, M.A.; Costantini, C.; Jaillon, S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat. Rev. Immunol. 2011, 11, 519–531. [Google Scholar] [CrossRef]
  70. Maurizi, G.; Della Guardia, L.; Maurizi, A.; Poloni, A. Adipocytes properties and crosstalk with immune system in obesity-related inflammation. J. Cell. Physiol. 2017, 233, 88–97. [Google Scholar] [CrossRef]
  71. Watanabe, Y.; Nagai, Y.; Honda, H.; Okamoto, N.; Yanagibashi, T.; Ogasawara, M.; Yamamoto, S.; Imamura, R.; Takasaki, I.; Hara, H.; et al. Bidirectional crosstalk between neutrophils and adipocytes promotes adipose tissue inflammation. FASEB J. 2019, 33, 11821–11835. [Google Scholar] [CrossRef] [Green Version]
  72. Hook, J.S.; Cao, M.; Potera, R.M.; Alsmadi, N.Z.; Schmidtke, D.W.; Moreland, J.G. Nox2 Regulates Platelet Activation and NET Formation in the Lung. Front. Immunol. 2019, 10, 1472. [Google Scholar] [CrossRef]
  73. Han, C.Y. Roles of Reactive Oxygen Species on Insulin Resistance in Adipose Tissue. Diabetes Metab. J. 2016, 40, 272–279. [Google Scholar] [CrossRef]
  74. Srikakulapu, P.; McNamara, C.A. B Lymphocytes and Adipose Tissue Inflammation. Arter. Thromb. Vasc. Biol. 2020, 40, 1110–1122. [Google Scholar] [CrossRef]
  75. Winer, D.A.; Winer, S.; Chng, M.H.Y.; Shen, L.; Engleman, E.G. B Lymphocytes in obesity-related adipose tissue inflammation and insulin resistance. Cell. Mol. Life Sci. 2013, 71, 1033–1043. [Google Scholar] [CrossRef] [Green Version]
  76. Kim, Y.-H.; Choi, B.-H.; Cheon, H.-G.; Do, M.-S. B cell activation factor (BAFF) is a novel adipokine that links obesity and inflammation. Exp. Mol. Med. 2009, 41, 208–216. [Google Scholar] [CrossRef] [Green Version]
  77. Esnault, S.; Kelly, E.A.; Sorkness, R.L.; Evans, M.D.; Busse, W.W.; Jarjour, N.N. Airway factor XIII associates with type 2 inflammation and airway obstruction in asthmatic patients. J. Allergy Clin. Immunol. 2016, 137, 767–773.e6. [Google Scholar] [CrossRef] [Green Version]
  78. Gualtierotti, R.; Ingegnoli, F.; Boscolo, M.; Griffini, S.; Grovetti, E.; Cugno, M. Tocilizumab Effects on Coagulation Factor XIII in Patients with Rheumatoid Arthritis. Adv. Ther. 2019, 36, 3494–3502. [Google Scholar] [CrossRef] [Green Version]
  79. Ichinose, A. Factor XIII is a key molecule at the intersection of coagulation and fibrinolysis as well as inflammation and infection control. Int. J. Hematol. 2012, 95, 362–370. [Google Scholar] [CrossRef] [PubMed]
  80. Kovar, F.M.; Marsik, C.L.; Jilma, B.; Mannhalter, C.; Joukhadar, C.; Wagner, O.F.; Endler, G. The inflammatory response is influenced by FXIII VAL 34 LEU polymorphism in a human LPS model. Wien. Klin. Wochenschr. 2009, 121, 515–519. [Google Scholar] [CrossRef]
  81. Raghu, H.; Cruz, C.; Rewerts, C.L.; Frederick, M.D.; Thornton, S.; Mullins, E.S.; Schoenecker, J.G.; Degen, J.L.; Flick, M.J. Transglutaminase factor XIII promotes arthritis through mechanisms linked to inflammation and bone erosion. Blood 2015, 125, 427–437. [Google Scholar] [CrossRef] [Green Version]
  82. Soendergaard, C.; Kvist, P.H.; Seidelin, J.B.; Pelzer, H.; Nielsen, O.H. Systemic and intestinal levels of factor XIII-A: The impact of inflammation on expression in macrophage subtypes. J. Gastroenterol. 2015, 51, 796–807. [Google Scholar] [CrossRef]
  83. Kopec, A.K.; Abrahams, S.R.; Thornton, S.; Palumbo, J.S.; Mullins, E.S.; Divanovic, S.; Weiler, H.; Owens, A.P.; Mackman, N.; Goss, A.M.; et al. Thrombin promotes diet-induced obesity through fibrin-driven inflammation. J. Clin. Investig. 2017, 127, 3152–3166. [Google Scholar] [CrossRef] [PubMed]
  84. Porrello, A.; Leslie, P.L.; Harrison, E.B.; Gorentla, B.K.; Kattula, S.; Ghosh, S.K.; Azam, S.H.; Holtzhausen, A.; Chao, Y.L.; Hayward, M.C.; et al. Factor XIIIA-expressing inflammatory monocytes promote lung squamous cancer through fibrin cross-linking. Nat. Commun. 2018, 9, 1988. [Google Scholar] [CrossRef] [Green Version]
  85. Lanir, N.; Ciano, P.S.; Van De Water, L.; McDonagh, J.; Dvorak, A.M.; Dvorak, H.F. Macrophage migration in fibrin gel matrices. II. Effects of clotting factor XIII, fibronectin, and glycosaminoglycan content on cell migration. J. Immunol. 1988, 140, 2340–2349. [Google Scholar] [PubMed]
  86. Goldstein, B.J. Protein-Tyrosine phosphatases and the regulation of insulin action. J. Cell. Biochem. 1992, 48, 33–42. [Google Scholar] [CrossRef] [PubMed]
  87. Goldstein, B.J.; Ahmad, F.; Ding, W.; Li, P.-M.; Zhang, W.-R. Regulation of the insulin signalling pathway by cellular protein-tyrosine phosphatases. Mol. Cell. Biochem. 1998, 182, 91–99. [Google Scholar] [CrossRef]
  88. Kim, W.-K.; Jung, H.; Kim, D.-H.; Kim, E.-Y.; Chung, J.-W.; Cho, Y.-S.; Park, S.G.; Park, B.-C.; Ko, Y.; Bae, K.-H.; et al. Regulation of adipogenic differentiation by LAR tyrosine phosphatase in human mesenchymal stem cells and 3T3-L1 preadipocytes. J. Cell Sci. 2009, 122, 4160–4167. [Google Scholar] [CrossRef] [Green Version]
  89. Ren, J.M.; Li, P.M.; Zhang, W.R.; Sweet, L.J.; Cline, G.; Shulman, G.I.; Livingston, J.N.; Goldstein, B.J. Transgenic mice deficient in the LAR protein-tyrosine phosphatase exhibit profound defects in glucose homeostasis. Diabetes 1998, 47, 493–497. [Google Scholar] [CrossRef]
  90. Deng, T.; Lyon, C.J.; Minze, L.J.; Lin, J.; Zou, J.; Liu, J.Z.; Ren, Y.; Yin, Z.; Hamilton, D.J.; Reardon, P.R.; et al. Class II Major Histocompatibility Complex Plays an Essential Role in Obesity-Induced Adipose Inflammation. Cell Metab. 2013, 17, 411–422. [Google Scholar] [CrossRef] [Green Version]
  91. Poloni, A.; Maurizi, G.; Ciarlantini, M.; Medici, M.; Mattiucci, D.; Mancini, S.; Maurizi, A.; Falconi, M.; Olivieri, A.; Leoni, P. Interaction between human mature adipocytes and lymphocytes induces T-cell proliferation. Cytotherapy 2015, 17, 1292–1301. [Google Scholar] [CrossRef]
  92. Datta, R.; Podolsky, M.J.; Atabai, K. Fat fibrosis: Friend or foe? JCI Insight 2018, 3, e122289. [Google Scholar] [CrossRef]
  93. Khan, T.; Muise, E.S.; Iyengar, P.; Wang, Z.V.; Chandalia, M.; Abate, N.; Zhang, B.B.; Bonaldo, P.; Chua, S.; Scherer, P.E. Metabolic Dysregulation and Adipose Tissue Fibrosis: Role of Collagen VI. Mol. Cell. Biol. 2008, 29, 1575–1591. [Google Scholar] [CrossRef] [Green Version]
  94. Lin, D.; Chun, T.-H.; Kang, L. Adipose extracellular matrix remodelling in obesity and insulin resistance. Biochem. Pharmacol. 2016, 119, 8–16. [Google Scholar] [CrossRef] [Green Version]
  95. Crewe, C.; An, Y.A.; Scherer, P.E. The ominous triad of adipose tissue dysfunction: Inflammation, fibrosis, and impaired angiogenesis. J. Clin. Investig. 2017, 127, 74–82. [Google Scholar] [CrossRef] [Green Version]
  96. Ruiz-Ojeda, F.J.; Méndez-Gutiérrez, A.; Aguilera, C.M.; Plaza-Díaz, J. Extracellular Matrix Remodeling of Adipose Tissue in Obesity and Metabolic Diseases. Int. J. Mol. Sci. 2019, 20, 4888. [Google Scholar] [CrossRef] [Green Version]
  97. Cinti, S.; Mitchell, G.; Barbatelli, G.; Murano, I.; Ceresi, E.; Faloia, E.; Wang, S.; Fortier, M.; Greenberg, A.S.; Obin, M.S. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J. Lipid Res. 2005, 46, 2347–2355. [Google Scholar] [CrossRef] [Green Version]
  98. Trayhurn, P. Hypoxia and Adipose Tissue Function and Dysfunction in Obesity. Physiol. Rev. 2013, 93, 1–21. [Google Scholar] [CrossRef] [Green Version]
  99. Sato, H.; Taketomi, Y.; Murakami, M. Metabolic regulation by secreted phospholipase A. Inflamm. Regen. 2016, 36, 7. [Google Scholar] [CrossRef] [Green Version]
  100. Iyer, A.; Lim, J.; Poudyal, H.; Reid, R.C.; Suen, J.Y.; Webster, J.; Prins, J.B.; Whitehead, J.P.; Fairlie, D.; Brown, L. An Inhibitor of Phospholipase A2 Group IIA Modulates Adipocyte Signaling and Protects Against Diet-Induced Metabolic Syndrome in Rats. Diabetes 2012, 61, 2320–2329. [Google Scholar] [CrossRef] [Green Version]
  101. Zhang, T.; He, J.; Xu, C.; Zu, L.; Jiang, H.; Pu, S.; Guo, X.; Xu, G. Mechanisms of metformin inhibiting lipolytic response to isoproterenol in primary rat adipocytes. J. Mol. Endocrinol. 2008, 42, 57–66. [Google Scholar] [CrossRef]
  102. Greenberg, A.S.; Shen, W.-J.; Muliro, K.; Patel, S.; Souza, S.C.; Roth, R.A.; Kraemer, F.B. Stimulation of Lipolysis and Hormone-sensitive Lipase via the Extracellular Signal-regulated Kinase Pathway. J. Biol. Chem. 2001, 276, 45456–45461. [Google Scholar] [CrossRef] [Green Version]
  103. Hesselbarth, N.; Kunath, A.; Kern, M.; Gericke, M.; Mejhert, N.; Rydén, M.; Stumvoll, M.; Blüher, M.; Klöting, N. Repin1 deficiency in adipose tissue improves whole-body insulin sensitivity, and lipid metabolism. Int. J. Obes. 2017, 41, 1815–1823. [Google Scholar] [CrossRef]
  104. Kaprio, J. Twin studies in Finland Twin research and human genetics. Off. J. Int. Soc. Twin Stud. 2006, 9, 772–777. [Google Scholar]
  105. Granér, M.; Seppälä-Lindroos, A.; Rissanen, A.; Hakkarainen, A.; Lundbom, N.; Kaprio, J.; Nieminen, M.S.; Pietiläinen, K.H. Epicardial Fat, Cardiac Dimensions, and Low-Grade Inflammation in Young Adult Monozygotic Twins Discordant for Obesity. Am. J. Cardiol. 2012, 109, 1295–1302. [Google Scholar] [CrossRef]
  106. Gentleman, R.C.; Carey, V.; Bates, D.; Bolstad, B.; Dettling, M.; Dudoit, S.; Ellis, B.; Gautier, L.; Ge, Y.; Gentry, J.; et al. Bioconductor: Open software development for computational biology and bioinformatics. Genome Biol. 2004, 5, R80. [Google Scholar] [CrossRef] [Green Version]
  107. R Core Team. R: A Language and Environment for Statistical Computing; R Foundation for Statistical Computing: Vienna, Austria, 2011. [Google Scholar]
  108. Zhang, Y.; Xie, L.; Gunasekar, S.K.; Tong, D.; Mishra, A.; Gibson, W.J.; Wang, C.; Fidler, T.; Marthaler, B.; Klingelhutz, A.; et al. SWELL1 is a regulator of adipocyte size, insulin signalling and glucose homeostasis. Nat. Cell Biol. 2017, 19, 504–517. [Google Scholar] [CrossRef] [Green Version]
  109. Abreu-Vieira, G.; Fischer, A.W.; Mattsson, C.; De Jong, J.M.; Shabalina, I.G.; Rydén, M.; Laurencikiene, J.; Arner, P.; Cannon, B.; Nedergaard, J.; et al. Cidea improves the metabolic profile through expansion of adipose tissue. Nat. Commun. 2015, 6, 7433. [Google Scholar] [CrossRef] [Green Version]
  110. Weingarten, A.; Turchetti, L.; Krohn, K.; Klöting, I.; Kern, M.; Kovacs, P.; Stumvoll, M.; Blüher, M.; Klöting, N. Novel genes on rat chromosome 10 are linked to body fat mass, preadipocyte number and adipocyte size. Int. J. Obes. (2005) 2016, 40, 1832–1840. [Google Scholar] [CrossRef]
  111. Lundbäck, V.; Kulyté, A.; Strawbridge, R.J.; Ryden, M.; Arner, P.; Marcus, C.; Dahlman, I. FAM13A and POM121C are candidate genes for fasting insulin: Functional follow-up analysis of a genome-wide association study. Diabetol. 2018, 61, 1112–1123. [Google Scholar] [CrossRef] [Green Version]
  112. Krautbauer, S.; Neumeier, M.; Rein-Fischboeck, L.; Haberl, E.M.; Tilg, H.; Eisinger, K.; Buechler, C. Adipocyte Hypertrophy and Improved Postprandial Lipid Response in Beta 2 Syntrophin Deficient Mice. Cell. Physiol. Biochem. 2019, 52, 1151–1165. [Google Scholar] [CrossRef]
  113. Geoghegan, G.; Simcox, J.; Seldin, M.M.; Parnell, T.J.; Stubben, C.; Just, S.; Begaye, L.; Lusis, A.J.; Villanueva, C.J. Targeted deletion of Tcf7l2 in adipocytes promotes adipocyte hypertrophy and impaired glucose metabolism. Mol. Metab. 2019, 24, 44–63. [Google Scholar] [CrossRef] [PubMed]
  114. Kilian, T.M.; Klöting, N.; Blüher, M.; Beck-Sickinger, A.G. Prenatal notch1 receptor blockade by protein delta homolog 1 (DLK1) modulates adipocyte size in vivo. Int. J. Obes. 2015, 40, 698–705. [Google Scholar] [CrossRef]
  115. Longo, M.; Raciti, G.A.; Zatterale, F.; Parrillo, L.; Desiderio, A.; Spinelli, R.; Hammarstedt, A.; Hedjazifar, S.; Hoffmann, J.M.; Nigro, C.; et al. Epigenetic modifications of the Zfp/ZNF423 gene control murine adipogenic commitment and are dysregulated in human hypertrophic obesity. Diabetologia 2018, 61, 369–380. [Google Scholar] [CrossRef] [Green Version]
  116. Huang, Z.H.; Gu, D.; Mazzone, T. Role of adipocyte-derived apoE in modulating adipocyte size, lipid metabolism, and gene expression in vivo. Am. J. Physiol. Metab. 2009, 296, E1110–E1119. [Google Scholar] [CrossRef] [Green Version]
  117. Acosta, J.R.; Douagi, I.; Andersson, D.P.; Bäckdahl, J.; Rydén, M.; Arner, P.; Laurencikiene, J. Increased fat cell size: A major phenotype of subcutaneous white adipose tissue in non-obese individuals with type 2 diabetes. Diabetologia 2015, 59, 560–570. [Google Scholar] [CrossRef] [Green Version]
  118. Ruschke, K.; Illes, M.; Kern, M.; Klöting, I.; Fasshauer, M.; Schön, M.; Kosacka, J.; Fitzl, G.; Kovacs, P.; Stumvoll, M.; et al. Repin1 maybe involved in the regulation of cell size and glucose transport in adipocytes. Biochem. Biophys. Res. Commun. 2010, 400, 246–251. [Google Scholar] [CrossRef] [PubMed]
  119. Takahashi, M.; Kamei, Y.; Ezaki, O. Mest/Peg1 imprinted gene enlarges adipocytes and is a marker of adipocyte size. Am. J. Physiol. Metab. 2005, 288, E117–E124. [Google Scholar] [CrossRef] [Green Version]
  120. Yvan-Charvet, L.; Even, P.C.; Bloch-Faure, M.; Guerre-Millo, M.; Moustaid-Moussa, N.; Ferré, P.; Quignard-Boulange, A. Deletion of the Angiotensin Type 2 Receptor (AT2R) Reduces Adipose Cell Size and Protects from Diet-Induced Obesity and Insulin Resistance. Diabetes 2005, 54, 991–999. [Google Scholar] [CrossRef]
Figure 1. Gene expression of transglutaminase family genes and association of F13A1 mRNA expression in acquired excess weight. Increase in transglutaminase family (TGM) members in excess weight represented by expression difference (Δ) between heavy and lean monozygotic (MZ) weight discordant twins (n = 12 twin pairs). (A) Differential expression in adipocyte-enriched fraction. (B) Differential expression in adipose tissue. Analysis of variance (ANOVA) with Bonferroni post-test shows that F13A1 is the sole TGM family member that reacts to acquired excess weight in both preparations. No significant changes were observed in other TGM family members. NS, not significant.
Figure 1. Gene expression of transglutaminase family genes and association of F13A1 mRNA expression in acquired excess weight. Increase in transglutaminase family (TGM) members in excess weight represented by expression difference (Δ) between heavy and lean monozygotic (MZ) weight discordant twins (n = 12 twin pairs). (A) Differential expression in adipocyte-enriched fraction. (B) Differential expression in adipose tissue. Analysis of variance (ANOVA) with Bonferroni post-test shows that F13A1 is the sole TGM family member that reacts to acquired excess weight in both preparations. No significant changes were observed in other TGM family members. NS, not significant.
Ijms 21 08289 g001
Figure 2. Transcriptome-wide association study (TWAS) strategy to investigate the association of F13A1 with acquired excess weight and adipocyte size in weight-discordant monozygotic twins. To examine what weight gain and adipocyte size-associated pathways F13A1 is linked to, we performed a transcriptome-wide association study (TWAS) of differentially expressed and correlated genes (Heavy–Lean ΔF13A1 vs. Heavy–Lean Δtranscriptome). The initial screen identified 182 genes whose differential expression showed significant linear correlation with F13A1 (r2 > 0.5 and p < 0.05) and significant differential expression between the Heavy and Lean co-twins (p < 0.05) [32]. This set was screened for previously published adipocyte size correlating (positive and negative) genes in the MZ twins, some of which were shared with this study. This resulted in no additional genes. The set was then screened for 14 reported ‘large adipocyte genes’ and 17 genes from the literature that have been published to regulate adipocyte size (PubMed search (‘adipocyte size’ and ‘regulation’ and ‘gene’)). All screened genes are listed in Supplemental Tables S1–S4. The resulting final set of 47 genes that correlated with F13A1, adipocyte diameter, and were differentially expressed in the co-twin pairs are listed in Table 2.
Figure 2. Transcriptome-wide association study (TWAS) strategy to investigate the association of F13A1 with acquired excess weight and adipocyte size in weight-discordant monozygotic twins. To examine what weight gain and adipocyte size-associated pathways F13A1 is linked to, we performed a transcriptome-wide association study (TWAS) of differentially expressed and correlated genes (Heavy–Lean ΔF13A1 vs. Heavy–Lean Δtranscriptome). The initial screen identified 182 genes whose differential expression showed significant linear correlation with F13A1 (r2 > 0.5 and p < 0.05) and significant differential expression between the Heavy and Lean co-twins (p < 0.05) [32]. This set was screened for previously published adipocyte size correlating (positive and negative) genes in the MZ twins, some of which were shared with this study. This resulted in no additional genes. The set was then screened for 14 reported ‘large adipocyte genes’ and 17 genes from the literature that have been published to regulate adipocyte size (PubMed search (‘adipocyte size’ and ‘regulation’ and ‘gene’)). All screened genes are listed in Supplemental Tables S1–S4. The resulting final set of 47 genes that correlated with F13A1, adipocyte diameter, and were differentially expressed in the co-twin pairs are listed in Table 2.
Ijms 21 08289 g002
Figure 3. Gene enrichment analysis. Selected over-represented GOterms among the 47 genes whose differential expression correlated with F13A1, adipocyte diameter, and showed significantly altered expression between heavy and lean co-twins. Gene ontology analysis was done using GOnet and confirmed with Panther. A total of 61 GOterms were found for Biological Process, and 28 highly relevant are represented here. All 13 Cellular Component terms are shown. No terms for Molecular Functions arose in the search. Significance of the terms is represented as –log10 (p-value) and number of genes in each GOterm is indicated with the bar. Full set of the GOterms with p-values, as well as false discovery rate adjusted p-values and list of genes can be found in Supplemental Tables S5 and S6.
Figure 3. Gene enrichment analysis. Selected over-represented GOterms among the 47 genes whose differential expression correlated with F13A1, adipocyte diameter, and showed significantly altered expression between heavy and lean co-twins. Gene ontology analysis was done using GOnet and confirmed with Panther. A total of 61 GOterms were found for Biological Process, and 28 highly relevant are represented here. All 13 Cellular Component terms are shown. No terms for Molecular Functions arose in the search. Significance of the terms is represented as –log10 (p-value) and number of genes in each GOterm is indicated with the bar. Full set of the GOterms with p-values, as well as false discovery rate adjusted p-values and list of genes can be found in Supplemental Tables S5 and S6.
Ijms 21 08289 g003
Figure 4. Network of gene ontology terms from 47 genes correlating with F13A1, adipocyte size, and acquired excess weight links F13A1 to inflammatory status of adipose tissue. Gene ontology term, gene clustering, and mapping analysis was performed using GOnet web-application (http://tools.dice-database.org/GOnet/). Analysis is presented as modified Euler layout which reveals three clear functional clusters linked to inflammatory response. The darker color of the GOterm node represents higher significance. The full set of values and GOterms are presented in Supplemental Tables S5 and S6.
Figure 4. Network of gene ontology terms from 47 genes correlating with F13A1, adipocyte size, and acquired excess weight links F13A1 to inflammatory status of adipose tissue. Gene ontology term, gene clustering, and mapping analysis was performed using GOnet web-application (http://tools.dice-database.org/GOnet/). Analysis is presented as modified Euler layout which reveals three clear functional clusters linked to inflammatory response. The darker color of the GOterm node represents higher significance. The full set of values and GOterms are presented in Supplemental Tables S5 and S6.
Ijms 21 08289 g004
Figure 5. F13A1 expression in adipocyte-enriched fraction associates with neutrophil activation and early immune response. (A) Map of GOterm nodes representing Biological Processes involved in immune response. Node clusters for neutrophil activation, secretion, and regulation of immune response are evident. Gene lists for representative nodes are shown. (B) Linear correlation of ΔF13A1 (Heavy–Lean twin) with differential expression Δgene (Heavy–Lean twin) of selected genes from GOterms. F13A1 levels increase in adipocyte-enriched fraction together with PYCARD (an inflammasome and caspase-1 activator), IL1B (interleukin 1b) (early pro-inflammatory cytokine), and HLA-DRA (HLA class II histocompatibility antigen presenting factor, and TNFSF13B (TNF Superfamily Member 13b and B-cell activating factor)).
Figure 5. F13A1 expression in adipocyte-enriched fraction associates with neutrophil activation and early immune response. (A) Map of GOterm nodes representing Biological Processes involved in immune response. Node clusters for neutrophil activation, secretion, and regulation of immune response are evident. Gene lists for representative nodes are shown. (B) Linear correlation of ΔF13A1 (Heavy–Lean twin) with differential expression Δgene (Heavy–Lean twin) of selected genes from GOterms. F13A1 levels increase in adipocyte-enriched fraction together with PYCARD (an inflammasome and caspase-1 activator), IL1B (interleukin 1b) (early pro-inflammatory cytokine), and HLA-DRA (HLA class II histocompatibility antigen presenting factor, and TNFSF13B (TNF Superfamily Member 13b and B-cell activating factor)).
Ijms 21 08289 g005
Figure 6. F13A1 expression in adipocyte-enriched fraction associates with cytokine-mediated signaling pathways. (A) Map of GOterm nodes representing Biological Processes involved in responding to external stimuli. Gene lists for representative nodes are shown. (B) Linear correlation of ΔF13A1 (Heavy–Lean twin) with differential expression Δgene (Heavy–Lean twin) of selected genes from GOterms. F13A1 levels in adipocyte-enriched fraction negatively correlate with PTPRF, a positive inhibitor of insulin signaling, and to STAT5A, a promoter of adipocyte differentiation.
Figure 6. F13A1 expression in adipocyte-enriched fraction associates with cytokine-mediated signaling pathways. (A) Map of GOterm nodes representing Biological Processes involved in responding to external stimuli. Gene lists for representative nodes are shown. (B) Linear correlation of ΔF13A1 (Heavy–Lean twin) with differential expression Δgene (Heavy–Lean twin) of selected genes from GOterms. F13A1 levels in adipocyte-enriched fraction negatively correlate with PTPRF, a positive inhibitor of insulin signaling, and to STAT5A, a promoter of adipocyte differentiation.
Ijms 21 08289 g006
Figure 7. F13A1 expression in adipocyte-enriched fraction associates with immune response to external factors and stimulus. (A) Map of GOterm nodes representing Biological Processes involved in responding to external stimuli. Gene lists for representative nodes are shown. (B) Linear correlation of ΔF13A1 (Heavy–Lean twin) with differential expression Δgene (Heavy–Lean twin) of selected genes from GOterms. F13A1 levels in adipocyte-enriched fraction correlate negatively with PLA2G16 (phospholipase A2), a lipolytic enzyme, and positively with expression of MAPK1.
Figure 7. F13A1 expression in adipocyte-enriched fraction associates with immune response to external factors and stimulus. (A) Map of GOterm nodes representing Biological Processes involved in responding to external stimuli. Gene lists for representative nodes are shown. (B) Linear correlation of ΔF13A1 (Heavy–Lean twin) with differential expression Δgene (Heavy–Lean twin) of selected genes from GOterms. F13A1 levels in adipocyte-enriched fraction correlate negatively with PLA2G16 (phospholipase A2), a lipolytic enzyme, and positively with expression of MAPK1.
Ijms 21 08289 g007
Table 1. Clinical characteristics of the twin pairs (n = 12) used in this study to obtain transcriptomics data from adipose tissue and adipocyte-enriched fraction. Student’s T-test was used to calculate the differences between the twin pairs, and Wilcoxon Signed rank’s test to determine the p-values.
Table 1. Clinical characteristics of the twin pairs (n = 12) used in this study to obtain transcriptomics data from adipose tissue and adipocyte-enriched fraction. Student’s T-test was used to calculate the differences between the twin pairs, and Wilcoxon Signed rank’s test to determine the p-values.
Clinical and Adipocity ParametersHeavier Co-twinLeaner Co-twinp-Value
Height (cm)169.3 ± 2.89169.2 ± 3.00.7530
Weight (kg)83.8 ± 4.667.0 ± 4.10.0022
BMI (kg/m2)29.0 ± 0.8523.2 ± 0.70.0022
Body fat (%)41.9 ± 2.032.8 ±1.80.0029
Body fat (kg)34.7 ± 2.322.0 ± 1.90.0022
Fat-free mass (kg)46.6 ± 3.742.8 ± 2.90.0029
Subcutaneous fat (dm3)5762.4 ± 450.63303.6 ± 275.40.0022
Intra-abdominal fat (dm3)1109.8 ± 186.4502.2 ± 91.20.0037
Liver fat (%)2.4 ± 0.740.60 ± 0.060.0076
Triglycerides (mmol/L)1.14 ± 0.070.96 ± 0.110.1823
fP-Leptin (pg/mL)34,229.0 ± 5931.318,331.8 ± 4412.60.0037
fP-Adiponectin (ng/mL)2844.0 ± 382.13520.1 ± 380.10.0186
Adipocyte diameter (mm) 87.9 ± 2.8276.6 ± 2.770.0022
Adipocyte number8.8e + 13 ± 5.7e + 128.5e + 13 ± 8.2e + 120.4328
BMI: body mass index, fP: fasting plasma. Data are presented as mean ± SEM.
Table 2. List of 47 genes showing strong and significant correlation Heavy–Lean ΔF13A1, ΔAdipocyte Diameter, and significantly altered expression between the 12 MZ co-twins. Genes are ranked based on smallest p-value.
Table 2. List of 47 genes showing strong and significant correlation Heavy–Lean ΔF13A1, ΔAdipocyte Diameter, and significantly altered expression between the 12 MZ co-twins. Genes are ranked based on smallest p-value.
Positive Correlation
Gene SymbolGene IDGene NamePearson (r)p-Value
CPVLENSG00000106066Carboxypeptidase, vitellogenic-like0.9460.00000097
PYCARDENSG00000103490PYD and CARD domain containing0.9410.00000167
CTSSENSG00000163131Cathepsin S0.9020.00002544
ITGAMENSG00000169896Integrin subunit alpha M0.8850.00005774
DCLK1ENSG00000133083Doublecortin-like kinase 10.8670.00012405
CYBBENSG00000165168Cytochrome b-245 beta chain0.8560.00018955
SRGNENSG00000122862Serglycin0.8420.0003081
TM4SF1ENSG00000169908Transmembrane 4 L six family member 180.8340.00039092
PDE4BENSG00000184588Phosphodiesterase 4B0.8340.00039317
TFPIENSG00000003436Tissue factor pathway inhibitor0.830.00044582
ANXA4ENSG00000196975Annexin A40.8270.00049387
IL8ENSG00000169429Interleukin 80.8060.00087858
VCANENSG00000038427Versican0.8060.00088439
BCL2A1ENSG00000140379BCL2-related protein A10.8020.00097386
LCP1ENSG00000136167Lymphocyte cytosolic protein 1/Plastin-20.8020.00097956
RECKENSG00000122707Reversion inducing cysteine-rich protein with kazal motifs0.7990.00105162
ABI3BPENSG00000154175ABI family member 3 binding protein0.7950.00115971
IL1BENSG00000125538Interleukin 1 beta0.7860.00145102
SAMSN1ENSG00000155307SAM domain, SH3 domain, and nuclear localization signals 10.780.00165797
RGS1ENSG00000090104Regulator of G protein signaling 10.7750.00186205
CCDC109BENSG00000005059Mitochondrial calcium uniporter dominant negative beta subunit0.7740.0018858
NCEH1ENSG00000144959Neutral cholesterol ester hydrolase 10.7660.0022702
ARPC5ENSG00000162704Actin-related protein 2/3 complex subunit 50.7640.00235294
LYZENSG00000090382Lysozyme0.7620.00247521
MAPK1ENSG00000100030Mitogen-activated protein kinase 10.7430.00359262
HLA-DRAENSG00000204287Major histocompatibility complex, class II, DR alpha0.7430.00361775
ARHGAP18ENSG00000146376Rho GTPase activating protein 180.7430.00364017
TNFSF13BENSG00000102524TNF superfamily member 13b/B cell activation factor0.7390.00393293
WIPF1ENSG00000115935WAS/WASL interacting protein family, member 10.7280.00481724
CNKSR3ENSG00000153721CNKSR family member 30.7250.00503039
MGPENSG00000111341Matrix Gla protein0.720.00555039
EVI2AENSG00000126860Ecotropic viral integration site 2A0.7170.00579394
NPTNENSG00000156642Neuroplastin0.7150.00596749
SELEENSG00000007908Selectin-E0.7780.00174649
LPCAT1ENSG00000153395Lysophosphatidylcholine acyltransferase 10.7320.00445358
Negative Correlation
Gene SymbolGene IDGene NamePearson (r)p-Value
SCRN2ENSG00000141295Secernin 2–0.8760.000086
PTPRFENSG00000142949Protein tyrosine phosphatase receptor type F–0.840.00033
EMC3ENSG00000125037ER membrane protein complex subunit 3–0.8160.000673
ABCD1ENSG00000101986ATP binding cassette subfamily D member 1–0.7930.001229
PLA2G16ENSG00000176485Phospholipase A and acyltransferase 3–0.7840.001527
NFRKBENSG00000170322Nuclear factor related to kappaB binding protein–0.740.00386
HEPACAMENSG00000165478Hepatic and glial cell adhesion molecule–0.7340.00426
RBP1ENSG00000114115Retinol binding protein 1, cellular–0.7250.00501
STAT5AENSG00000126561Signal transducer and activator of transcription 5A–0.720.00556
TUSC5ENSG00000184811Tumor suppressor candidate 5/Trafficking regulator of GLUT4–0.7170.005843
COX5AENSG00000178741Cytochrome c oxidase subunit Va–0.7120.006346
REPIN1ENSG00000214022Replication initiator 1–0.7720.001987
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kaartinen, M.T.; Arora, M.; Heinonen, S.; Rissanen, A.; Kaprio, J.; Pietiläinen, K.H. Transglutaminases and Obesity in Humans: Association of F13A1 to Adipocyte Hypertrophy and Adipose Tissue Immune Response. Int. J. Mol. Sci. 2020, 21, 8289. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21218289

AMA Style

Kaartinen MT, Arora M, Heinonen S, Rissanen A, Kaprio J, Pietiläinen KH. Transglutaminases and Obesity in Humans: Association of F13A1 to Adipocyte Hypertrophy and Adipose Tissue Immune Response. International Journal of Molecular Sciences. 2020; 21(21):8289. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21218289

Chicago/Turabian Style

Kaartinen, Mari T., Mansi Arora, Sini Heinonen, Aila Rissanen, Jaakko Kaprio, and Kirsi H. Pietiläinen. 2020. "Transglutaminases and Obesity in Humans: Association of F13A1 to Adipocyte Hypertrophy and Adipose Tissue Immune Response" International Journal of Molecular Sciences 21, no. 21: 8289. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21218289

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

Article Metrics

Back to TopTop