Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cells ; 10(5)2021 05 07.
Article in English | MEDLINE | ID: mdl-34067093

ABSTRACT

Adiposity is strongly associated with cardiovascular (CV) morbidity. Uncoupling protein 1 (UCP1) increases energy expenditure in adipocytes and may counteract adiposity. Our objective was to investigate a connection between UCP1 expression and cardiovascular health in patients with rheumatoid arthritis (RA) in a longitudinal observational study. Transcription of UCP1 was measured by qPCR in the subcutaneous adipose tissue of 125 female RA patients and analyzed with respect to clinical parameters and the estimated CV risk. Development of new CV events and diabetes mellitus was followed for five years. Transcription of UCP1 was identified in 89 (71%) patients. UCP1 positive patients had often active RA disease (p = 0.017), high serum levels of IL6 (p = 0.0025) and were frequently overweight (p = 0.015). IL-6hiBMIhi patients and patients treated with IL6 receptor inhibitor tocilizumab had significantly higher levels of UCP1 compared to other RA patients (p < 0.0001, p = 0.032, respectively). Both UCP1hi groups displayed unfavorable metabolic profiles with high plasma glucose levels and high triglyceride-to-HDL ratios, which indicated insulin resistance. Prospective follow-up revealed no significant difference in the incidence of new CV and metabolic events in the UCP1hi groups and remaining RA patients. The study shows that high transcription of UCP1 in adipose tissue is related to IL6-driven processes and reflects primarily metabolic CV risk in female RA patients.


Subject(s)
Arthritis, Rheumatoid/complications , Biomarkers/metabolism , Cardiovascular Diseases/pathology , Gene Expression Regulation , Interleukin-6/blood , Uncoupling Protein 1/metabolism , Aged , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Female , Follow-Up Studies , Humans , Middle Aged , Prognosis , Prospective Studies , Risk Factors , Uncoupling Protein 1/genetics
2.
FASEB J ; 35(4): e21357, 2021 04.
Article in English | MEDLINE | ID: mdl-33710685

ABSTRACT

First-degree relatives (FDRs) of type 2 diabetics (T2D) feature dysfunction of subcutaneous adipose tissue (SAT) long before T2D onset. miRNAs have a role in adipocyte precursor cells (APC) differentiation and in adipocyte identity. Thus, impaired miRNA expression may contribute to SAT dysfunction in FDRs. In the present work, we have explored changes in miRNA expression associated with T2D family history which may affect gene expression in SAT APCs from FDRs. Small RNA-seq was performed in APCs from healthy FDRs and matched controls and omics data were validated by qPCR. Integrative analyses of APC miRNome and transcriptome from FDRs revealed down-regulated hsa-miR-23a-5p, -193a-5p and -193b-5p accompanied by up-regulated Insulin-like Growth Factor 2 (IGF2) gene which proved to be their direct target. The expression changes in these marks were associated with SAT adipocyte hypertrophy in FDRs. APCs from FDRs further demonstrated reduced capability to differentiate into adipocytes. Treatment with IGF2 protein decreased APC adipogenesis, while over-expression of hsa-miR-23a-5p, -193a-5p and -193b-5p enhanced adipogenesis by IGF2 targeting. Indeed, IGF2 increased the Wnt Family Member 10B gene expression in APCs. Down-regulation of the three miRNAs and IGF2 up-regulation was also observed in Peripheral Blood Leukocytes (PBLs) from FDRs. In conclusion, APCs from FDRs feature a specific miRNA/gene profile, which associates with SAT adipocyte hypertrophy and appears to contribute to impaired adipogenesis. PBL detection of this profile may help in identifying adipocyte hypertrophy in individuals at high risk of T2D.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Genetic Predisposition to Disease , Insulin-Like Growth Factor II/metabolism , MicroRNAs/metabolism , Adipogenesis , Cloning, Molecular , Diabetes Mellitus, Type 2/genetics , Family , Gene Expression Regulation , Humans , Insulin-Like Growth Factor II/genetics , MicroRNAs/genetics
3.
PLoS One ; 16(2): e0247300, 2021.
Article in English | MEDLINE | ID: mdl-33606810

ABSTRACT

OBJECTIVE: Gremlin 1 (GREM1) is a secreted BMP2/4 inhibitor which regulates commitment and differentiation of human adipose precursor cells and prevents the browning effect of BMP4. GREM1 is an insulin antagonist and serum levels are high in type 2 diabetes (T2D). We here examined in vivo effects of AAV8 (Adeno-Associated Viral vectors of serotype eight) GREM 1 targeting the liver in mature mice to increase its systemic secretion and also, in a separate study, injected recombinant GREM 1 intraperitoneally. The objective was to characterize systemic effects of GREM 1 on insulin sensitivity, glucose tolerance, body weight, adipose cell browning and other local tissue effects. METHODS: Adult mice were injected with AAV8 vectors expressing GREM1 in the liver or receiving regular intra-peritoneal injections of recombinant GREM1 protein. The mice were fed with a low fat or high fat diet (HFD) and followed over time. RESULTS: Liver-targeted AAV8-GREM1 did not alter body weight, whole-body glucose and insulin tolerance, or adipose tissue gene expression. Although GREM1 protein accumulated in liver cells, GREM1 serum levels were not increased suggesting that it may not have been normally processed for secretion. Hepatic lipid accumulation, inflammation and fibrosis were also not changed. Repeated intraperitoneal rec-GREM1 injections for 5 weeks were also without effects on body weight and insulin sensitivity. UCP1 was slightly but significantly reduced in both white and brown adipose tissue but this was not of sufficient magnitude to alter body weight. We validated that recombinant GREM1 inhibited BMP4-induced pSMAD1/5/9 in murine cells in vitro, but saw no direct inhibitory effect on insulin signalling and pAkt (ser 473 and thr 308) activation. CONCLUSION: GREM1 accumulates intracellularly when overexpressed in the liver cells of mature mice and is apparently not normally processed/secreted. However, also repeated intraperitoneal injections were without effects on body weight and insulin sensitivity and adipose tissue UCP1 levels were only marginally reduced. These results suggest that mature mice do not readily respond to GREMLIN 1 but treatment of murine cells with GREMLIN 1 protein in vitro validated its inhibitory effect on BMP4 signalling while insulin signalling was not altered.


Subject(s)
Dependovirus/genetics , Diet, High-Fat/adverse effects , Insulin Resistance/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , 3T3-L1 Cells , Animals , Body Weight , Cell Line , Disease Models, Animal , Genetic Therapy , Genetic Vectors/administration & dosage , Glucose Tolerance Test , Humans , Injections, Intraperitoneal , Intercellular Signaling Peptides and Proteins/administration & dosage , Male , Mice , Recombinant Proteins/administration & dosage
4.
Epigenomics ; 12(10): 873-888, 2020 05.
Article in English | MEDLINE | ID: mdl-32483983

ABSTRACT

Aim: First-degree relatives (FDR) of individuals with Type 2 diabetes (T2D) feature restricted adipogenesis, which render them more vulnerable to T2D. Epigenetics may contribute to these abnormalities. Methods: FDR pre-adipocyte Methylome and Transcriptome were investigated by MeDIP- and RNA-Seq, respectively. Results:Methylome analysis revealed 2841 differentially methylated regions (DMR) in FDR. Most DMR localized into gene-body and were hypomethylated. The strongest hypomethylation signal was identified in an intronic-DMR at the PTPRD gene. PTPRD hypomethylation in FDR was confirmed by bisulphite sequencing and was responsible for its upregulation. Interestingly, Ptprd-overexpression in 3T3-L1 pre-adipocytes inhibited adipogenesis. Notably, the validated PTPRD-associated DMR was significantly hypomethylated in peripheral blood leukocytes from the same FDR individuals. Finally, PTPRD methylation pattern was also replicated in obese individuals. Conclusion: Our findings indicated a previously unrecognized role of PTPRD in restraining adipogenesis. This abnormality may contribute to increase FDR proclivity toward T2D.


Subject(s)
Adipogenesis/genetics , DNA Methylation , Diabetes Mellitus, Type 2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , 3T3-L1 Cells , Adult , Animals , Epigenesis, Genetic , Female , Humans , Male , Mice
5.
Mol Metab ; 32: 15-26, 2020 02.
Article in English | MEDLINE | ID: mdl-32029225

ABSTRACT

OBJECTIVE: Bone morphogenetic protein 4 (BMP4) adeno-associated viral vectors of serotype 8 (AAV8) gene therapy targeting the liver prevents the development of obesity in initially lean mice by browning the large subcutaneous white adipose tissue (WAT) and enhancing energy expenditure. Here, we examine whether this approach could also reduce established obesity. METHODS: Dietary-induced obese C57BL6/N mice received AAV8 BMP4 gene therapy at 17-18 weeks of age. They were kept on a high-fat diet and phenotypically characterized for an additional 10-12 weeks. Following termination, the mice underwent additional characterization in vitro. RESULTS: Surprisingly, we observed no effect on body weight, browning of WAT, or energy expenditure in these obese mice, but whole-body insulin sensitivity and glucose tolerance were robustly improved. Insulin signaling and insulin-stimulated glucose uptake were increased in both adipose cells and skeletal muscle. BMP4 also decreased hepatic glucose production and reduced gluconeogenic enzymes in the liver, but not in the kidney, in addition to enhancing insulin action in the liver. CONCLUSIONS: Our findings show that BMP4 prevents, but does not reverse, established obesity in adult mice, while it improves insulin sensitivity independent of weight reduction. The BMP antagonist Noggin was increased in WAT in obesity, which may account for the lack of browning.


Subject(s)
Adipose Tissue, Brown , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/therapeutic use , Genetic Therapy , Insulin/metabolism , Obesity/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Diet, High-Fat/adverse effects , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/chemically induced , Signal Transduction
6.
Diabetes ; 69(3): 331-341, 2020 03.
Article in English | MEDLINE | ID: mdl-31882566

ABSTRACT

The BMP2/4 antagonist and novel adipokine Gremlin 1 is highly expressed in human adipose cells and increased in hypertrophic obesity. As a secreted antagonist, it inhibits the effect of BMP2/4 on adipose precursor cell commitment/differentiation. We examined mRNA levels of Gremlin 1 in key target tissues for insulin and also measured tissue and serum levels in several carefully phenotyped human cohorts. Gremlin 1 expression was high in adipose tissue, higher in visceral than in subcutaneous tissue, increased in obesity, and further increased in type 2 diabetes (T2D). A similar high expression was seen in liver biopsies, but expression was considerably lower in skeletal muscles. Serum levels were increased in obesity but most prominently in T2D. Transcriptional activation in both adipose tissue and liver as well as serum levels were strongly associated with markers of insulin resistance in vivo (euglycemic clamps and HOMA of insulin resistance), and the presence of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). We also found Gremlin 1 to antagonize insulin signaling and action in human primary adipocytes, skeletal muscle, and liver cells. Thus, Gremlin 1 is a novel secreted insulin antagonist and biomarker as well as a potential therapeutic target in obesity and its complications T2D and NAFLD/NASH.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Intercellular Signaling Peptides and Proteins/genetics , Non-alcoholic Fatty Liver Disease/genetics , Obesity/genetics , RNA, Messenger/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adipokines/genetics , Adipokines/metabolism , Adult , Aged , Case-Control Studies , Diabetes Mellitus, Type 2/metabolism , Female , Glucose Clamp Technique , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Insulin/metabolism , Insulin Antagonists/pharmacology , Insulin Resistance/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Intra-Abdominal Fat/metabolism , Liver/metabolism , Male , Middle Aged , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Primary Cell Culture , Subcutaneous Fat/metabolism
7.
Physiol Rev ; 98(4): 1911-1941, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30067159

ABSTRACT

The subcutaneous adipose tissue (SAT) is the largest and best storage site for excess lipids. However, it has a limited ability to expand by recruiting and/or differentiating available precursor cells. When inadequate, this leads to a hypertrophic expansion of the cells with increased inflammation, insulin resistance, and a dysfunctional prolipolytic tissue. Epi-/genetic factors regulate SAT adipogenesis and genetic predisposition for type 2 diabetes is associated with markers of an impaired SAT adipogenesis and development of hypertrophic obesity also in nonobese individuals. We here review mechanisms for the adipose precursor cells to enter adipogenesis, emphasizing the role of bone morphogenetic protein-4 (BMP-4) and its endogenous antagonist gremlin-1, which is increased in hypertrophic SAT in humans. Gremlin-1 is a secreted and a likely important mechanism for the impaired SAT adipogenesis in hypertrophic obesity. Transiently increasing BMP-4 enhances adipogenic commitment of the precursor cells while maintained BMP-4 signaling during differentiation induces a beige/brown oxidative phenotype in both human and murine adipose cells. Adipose tissue growth and development also requires increased angiogenesis, and BMP-4, as a proangiogenic molecule, may also be an important feedback regulator of this. Hypertrophic obesity is also associated with increased lipolysis. Reduced lipid storage and increased release of FFA by hypertrophic SAT are important mechanisms for the accumulation of ectopic fat in the liver and other places promoting insulin resistance. Taken together, the limited expansion and storage capacity of SAT is a major driver of the obesity-associated metabolic complications.


Subject(s)
Adipogenesis/physiology , Adipose Tissue/pathology , Obesity/pathology , Adipocytes/pathology , Animals , Cell Differentiation/physiology , Diabetes Mellitus, Type 2/pathology , Humans , Inflammation/pathology , Insulin Resistance/physiology
8.
J Cell Commun Signal ; 12(1): 309-318, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29247377

ABSTRACT

Obesity and type 2 diabetes increase worldwide at an epidemic rate. It is expected that by the year 2030 around 500 million people will have diabetes; predominantly type 2 diabetes. The CCN family of proteins has become of interest in both metabolic and other common human diseases because of their effects on mesenchymal stem cell (MSCs) proliferation and differentiation as well as being important regulators of fibrosis. We here review current knowledge of the WNT1 inducible signaling pathway protein 2 (CCN5/WISP2). It has been shown to be an important regulator of both these processes through effects on both the canonical WNT and the TGFß pathways. It is also under normal regulation by the adipogenic commitment factor BMP4, in contrast to conventional canonical WNT ligands, and allows MSCs to undergo normal adipose cell differentiation. CCN5/WISP2 is highly expressed in, and secreted by, MSCs and is an important regulator of MSCs growth. In a transgenic mouse model overexpressing CCN5/WISP2 in the adipose tissue, we have shown that it is secreted and circulating in the blood, the mice develop hypercellular white and brown adipose tissue, have increased lean body mass and enlarged hypercellular hearts. Obese transgenic mice had improved insulin sensitivity. Interestingly, the anti-fibrotic effect of CCN5/WISP2 is protective against heart failure by inhibition of the TGFß pathway. Understanding how CCN5/WISP2 is regulated and signals is important and may be useful for developing new treatment strategies in obesity and metabolic diseases and it can also be a target in regenerative medicine.

9.
Diabetologia ; 61(2): 369-380, 2018 02.
Article in English | MEDLINE | ID: mdl-29067487

ABSTRACT

AIMS/HYPOTHESIS: Subcutaneous adipocyte hypertrophy is associated with insulin resistance and increased risk of type 2 diabetes, and predicts its future development independent of obesity. In humans, subcutaneous adipose tissue hypertrophy is a consequence of impaired adipocyte precursor cell recruitment into the adipogenic pathway rather than a lack of precursor cells. The zinc finger transcription factor known as zinc finger protein (ZFP) 423 has been identified as a major determinant of pre-adipocyte commitment and maintained white adipose cell function. Although its levels do not change during adipogenesis, ectopic expression of Zfp423 in non-adipogenic murine cells is sufficient to activate expression of the gene encoding peroxisome proliferator-activated receptor γ (Pparγ; also known as Pparg) and increase the adipogenic potential of these cells. We investigated whether the Zfp423 gene is under epigenetic regulation and whether this plays a role in the restricted adipogenesis associated with hypertrophic obesity. METHODS: Murine 3T3-L1 and NIH-3T3 cells were used as fibroblasts committed and uncommitted to the adipocyte lineage, respectively. Human pre-adipocytes were isolated from the stromal vascular fraction of subcutaneous adipose tissue of 20 lean non-diabetic individuals with a wide adipose cell size range. mRNA levels were measured by quantitative real-time PCR, while methylation levels were analysed by bisulphite sequencing. Chromatin structure was analysed by micrococcal nuclease protection assay, and DNA-methyltransferases were chemically inhibited by 5-azacytidine. Adipocyte differentiation rate was evaluated by Oil Red O staining. RESULTS: Comparison of uncommitted (NIH-3T3) and committed (3T3-L1) adipose precursor cells revealed that Zfp423 expression increased (p < 0.01) in parallel with the ability of the cells to differentiate into mature adipocytes owing to both decreased promoter DNA methylation (p < 0.001) and nucleosome occupancy (nucleosome [NUC] 1 p < 0.01; NUC2 p < 0.001) in the 3T3-L1 compared with NIH-3T3 cells. Interestingly, non-adipogenic epigenetic profiles can be reverted in NIH-3T3 cells as 5-azacytidine treatment increased Zfp423 mRNA levels (p < 0.01), reduced DNA methylation at a specific CpG site (p < 0.01), decreased nucleosome occupancy (NUC1, NUC2: p < 0.001) and induced adipocyte differentiation (p < 0.05). These epigenetic modifications can also be initiated in response to changes in the pre-adipose cell microenvironment, in which bone morphogenetic protein 4 (BMP4) plays a key role. We finally showed that, in human adipocyte precursor cells, impaired epigenetic regulation of zinc nuclear factor (ZNF)423 (the human orthologue of murine Zfp423) was associated with inappropriate subcutaneous adipose cell hypertrophy. As in NIH-3T3 cells, the normal ZNF423 epigenetic profile was rescued by 5-azacytidine exposure. CONCLUSIONS/INTERPRETATION: Our results show that epigenetic events regulate the ability of precursor cells to commit and differentiate into mature adipocytes by modulating ZNF423, and indicate that dysregulation of these mechanisms accompanies subcutaneous adipose tissue hypertrophy in humans.


Subject(s)
Adipogenesis/physiology , Diabetes Mellitus, Type 2/metabolism , Obesity/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , DNA Methylation/genetics , DNA Methylation/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Humans , Mice , NIH 3T3 Cells , Obesity/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Cell Rep ; 20(5): 1038-1049, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28768190

ABSTRACT

We examined the effect of Bone Morphogenetic Protein 4 (BMP4) on energy expenditure in adult mature mice by targeting the liver with adeno-associated viral (AAV) BMP4 vectors to increase circulating levels. We verified the direct effect of BMP4 in inducing a brown oxidative phenotype in differentiating preadipocytes in vitro. AAV-BMP4-treated mice display marked browning of subcutaneous adipocytes, with increased mitochondria and Uncoupling Protein 1 (UCP1). These mice are protected from obesity on a high-fat diet and have increased whole-body energy expenditure, improved insulin sensitivity, reduced liver fat, and reduced adipose tissue inflammation. On a control diet, they show unchanged body weight but improved insulin sensitivity. In contrast, AAV-BMP4-treated mice showed beiging of BAT with reduced UCP1, increased lipids, and reduced hormone-sensitive lipase (HSL). Thus, BMP4 exerts different effects on WAT and BAT, but the overall effect is to enhance insulin sensitivity and whole-body energy expenditure by browning subcutaneous adipose tissue.


Subject(s)
Adipocytes, Brown/metabolism , Bone Morphogenetic Protein 4/biosynthesis , Dependovirus , Genetic Therapy/methods , Obesity/prevention & control , Subcutaneous Fat/metabolism , Animals , Bone Morphogenetic Protein 4/genetics , Energy Metabolism , Male , Mice , Obesity/genetics , Obesity/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
11.
Am J Physiol Endocrinol Metab ; 313(4): E450-E462, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28655716

ABSTRACT

Females are, in general, more insulin sensitive than males. To investigate whether this is a direct effect of sex-steroids (SS) in white adipose tissue (WAT), we developed a male mouse model overexpressing the aromatase enzyme, converting testosterone (T) to estradiol (E2), specifically in WAT (Ap2-arom mice). Adipose tissue E2 levels were increased while circulating SS levels were unaffected in male Ap2-arom mice. Importantly, male Ap2-arom mice were more insulin sensitive compared with WT mice and exhibited increased serum adiponectin levels and upregulated expression of Glut4 and Irs1 in WAT. The expression of markers of macrophages and immune cell infiltration was markedly decreased in WAT of male Ap2-arom mice. The adipogenesis was enhanced in male Ap2-arom mice, supported by elevated Pparg expression in WAT and enhanced differentiation of preadipocyte into mature adipocytes. In summary, increased adipose tissue aromatase activity reduces adipose tissue inflammation and improves insulin sensitivity in male mice. We propose that estrogen increases insulin sensitivity via a local effect in WAT on adiponectin expression, adipose tissue inflammation, and adipogenesis.


Subject(s)
Adipose Tissue, White/metabolism , Aromatase/genetics , Estradiol/metabolism , Insulin Resistance/genetics , Testosterone/metabolism , Adipocytes , Adipogenesis/genetics , Adiponectin/metabolism , Adipose Tissue, White/immunology , Animals , Gene Knock-In Techniques , Glucose Transporter Type 4/metabolism , Inflammation , Insulin Receptor Substrate Proteins/metabolism , Macrophages/immunology , Male , Mice , PPAR gamma/metabolism , Up-Regulation
12.
Sci Rep ; 7: 43515, 2017 02 27.
Article in English | MEDLINE | ID: mdl-28240264

ABSTRACT

WISP2 is a novel adipokine, most highly expressed in the adipose tissue and primarily in undifferentiated mesenchymal cells. As a secreted protein, it is an autocrine/paracrine activator of canonical WNT signaling and, as an intracellular protein, it helps to maintain precursor cells undifferentiated. To examine effects of increased WISP2 in vivo, we generated an aP2-WISP2 transgenic (Tg) mouse. These mice had increased serum levels of WISP2, increased lean body mass and whole body energy expenditure, hyperplastic brown/white adipose tissues and larger hyperplastic hearts. Obese Tg mice remained insulin sensitive, had increased glucose uptake by adipose cells and skeletal muscle in vivo and ex vivo, increased GLUT4, increased ChREBP and markers of adipose tissue lipogenesis. Serum levels of the novel fatty acid esters of hydroxy fatty acids (FAHFAs) were increased and transplantation of Tg adipose tissue improved glucose tolerance in recipient mice supporting a role of secreted FAHFAs. The growth-promoting effect of WISP2 was shown by increased BrdU incorporation in vivo and Tg serum increased mesenchymal precursor cell proliferation in vitro. In contrast to conventional canonical WNT ligands, WISP2 expression was inhibited by BMP4 thereby allowing normal induction of adipogenesis. WISP2 is a novel secreted regulator of mesenchymal tissue cellularity.


Subject(s)
Adipose Tissue/metabolism , Gene Expression , Insulin Resistance/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Autocrine Communication , Biomarkers , Body Composition , Body Weight , Bone Morphogenetic Protein 4/metabolism , Cell Count , Cell Proliferation/drug effects , Cell Size , Energy Metabolism , Genotype , Glucose/metabolism , Glucose Tolerance Test , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Hyperplasia , Insulin/metabolism , Lipogenesis/genetics , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Transforming Growth Factor beta/metabolism
13.
Trends Endocrinol Metab ; 26(4): 193-200, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25703677

ABSTRACT

The adipose tissue is crucial in regulating insulin sensitivity and risk for diabetes through its lipid storage capacity and thermogenic and endocrine functions. Subcutaneous adipose tissue (SAT) stores excess lipids through expansion of adipocytes (hypertrophic obesity) and/or recruitment of new precursor cells (hyperplastic obesity). Hypertrophic obesity in humans, a characteristic of genetic predisposition for diabetes, is associated with abdominal obesity, ectopic fat accumulation, and the metabolic syndrome (MS), while the ability to recruit new adipocytes prevents this. We review the regulation of adipogenesis, its relation to SAT expandability and the risks of ectopic fat accumulation, and insulin resistance. The actions of GLUT4 in SAT, including a novel family of lipids enhancing insulin sensitivity/secretion, and the function of bone morphogenetic proteins (BMPs) in white and beige/brown adipogenesis in humans are highlighted.


Subject(s)
Adipogenesis , Down-Regulation , Glucose Transporter Type 4/metabolism , Insulin Resistance , Metabolic Syndrome/metabolism , Models, Biological , Subcutaneous Fat, Abdominal/metabolism , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adiposity , Animals , Glucose Transporter Type 4/genetics , Humans , Hypertrophy , Lipid Metabolism , Metabolic Syndrome/genetics , Metabolic Syndrome/immunology , Metabolic Syndrome/pathology , Organ Specificity , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/pathology
14.
Diabetes ; 64(5): 1670-81, 2015 May.
Article in English | MEDLINE | ID: mdl-25605802

ABSTRACT

The limited expandability of subcutaneous adipose tissue, due to reduced ability to recruit and differentiate new adipocytes, prevents its buffering effect in obesity and is characterized by expanded adipocytes (hypertrophic obesity). Bone morphogenetic protein-4 (BMP4) plays a key role in regulating adipogenic precursor cell commitment and differentiation. We found BMP4 to be induced and secreted by differentiated (pre)adipocytes, and BMP4 was increased in large adipose cells. However, the precursor cells exhibited a resistance to BMP4 owing to increased secretion of the BMP inhibitor Gremlin-1 (GREM1). GREM1 is secreted by (pre)adipocytes and is an inhibitor of both BMP4 and BMP7. BMP4 alone, and/or silencing GREM1, increased transcriptional activation of peroxisome proliferator-activated receptor γ and promoted the preadipocytes to assume an oxidative beige/brown adipose phenotype including markers of increased mitochondria and PGC1α. Driving white adipose differentiation inhibited the beige/brown markers, suggesting the presence of multipotent adipogenic precursor cells. However, silencing GREM1 and/or adding BMP4 during white adipogenic differentiation reactivated beige/brown markers, suggesting that increased BMP4 preferentially regulates the beige/brown phenotype. Thus, BMP4, secreted by white adipose cells, is an integral feedback regulator of both white and beige adipogenic commitment and differentiation, and resistance to BMP4 by GREM1 characterizes hypertrophic obesity.


Subject(s)
Adipogenesis/physiology , Adipose Tissue, Brown/metabolism , Adipose Tissue/metabolism , Bone Morphogenetic Protein 4/metabolism , Adipogenesis/genetics , Adult , Bone Morphogenetic Protein 4/antagonists & inhibitors , Bone Morphogenetic Protein 4/genetics , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Expression Regulation/physiology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Middle Aged , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Obesity/metabolism , RNA Interference , RNA, Small Interfering
15.
J Biol Chem ; 289(10): 6899-6907, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24451367

ABSTRACT

WNT1-inducible-signaling pathway protein 2 (WISP2) is primarily expressed in mesenchymal stem cells, fibroblasts, and adipogenic precursor cells. It is both a secreted and cytosolic protein, the latter regulating precursor cell adipogenic commitment and PPARγ induction by BMP4. To examine the effect of the secreted protein, we expressed a full-length and a truncated, non-secreted WISP2 in NIH3T3 fibroblasts. Secreted, but not truncated WISP2 activated the canonical WNT pathway with increased ß-catenin levels, its nuclear targeting phosphorylation, and LRP5/6 phosphorylation. It also inhibited Pparg activation and the effect of secreted WISP2 was reversed by the WNT antagonist DICKKOPF-1. Differentiated 3T3-L1 adipose cells were also target cells where extracellular WISP2 activated the canonical WNT pathway, inhibited Pparg and associated adipose genes and, similar to WNT3a, promoted partial dedifferentiation of the cells and the induction of a myofibroblast phenotype with activation of markers of fibrosis. Thus, WISP2 exerts dual actions in mesenchymal precursor cells; secreted WISP2 activates canonical WNT and maintains the cells in an undifferentiated state, whereas cytosolic WISP2 regulates adipogenic commitment.


Subject(s)
Adipogenesis , Adipokines/metabolism , CCN Intercellular Signaling Proteins/metabolism , Mesenchymal Stem Cells/cytology , Repressor Proteins/metabolism , Wnt Signaling Pathway , 3T3-L1 Cells , Adipokines/antagonists & inhibitors , Adipokines/genetics , Animals , CCN Intercellular Signaling Proteins/antagonists & inhibitors , CCN Intercellular Signaling Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mice , NIH 3T3 Cells , PPAR gamma/genetics , PPAR gamma/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Wnt3 Protein/metabolism
17.
Proc Natl Acad Sci U S A ; 110(7): 2563-8, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23359679

ABSTRACT

Inability to recruit new adipose cells following weight gain leads to inappropriate enlargement of existing cells (hypertrophic obesity) associated with inflammation and a dysfunctional adipose tissue. We found increased expression of WNT1 inducible signaling pathway protein 2 (WISP2) and other markers of WNT activation in human abdominal s.c. adipose tissue characterized by hypertrophic obesity combined with increased visceral fat accumulation and insulin resistance. WISP2 activation in the s.c. adipose tissue, but not in visceral fat, identified the metabolic syndrome in equally obese individuals. WISP2 is a novel adipokine, highly expressed and secreted by adipose precursor cells. Knocking down WISP2 induced spontaneous differentiation of 3T3-L1 and human preadipocytes and allowed NIH 3T3 fibroblasts to become committed to the adipose lineage by bone morphogenetic protein 4 (BMP4). WISP2 forms a cytosolic complex with the peroxisome proliferator-activated receptor γ (PPARγ) transcriptional activator zinc finger protein 423 (Zfp423), and this complex is dissociated by BMP4 in a SMAD-dependent manner, thereby allowing Zfp423 to enter the nucleus, activate PPARγ, and commit the cells to the adipose lineage. The importance of intracellular Wisp2 protein for BMP4-induced adipogenic commitment and PPARγ activation was verified by expressing a mutant Wisp2 protein lacking the endoplasmic reticulum signal and secretion sequence. Secreted Wnt/Wisp2 also inhibits differentiation and PPARγ activation, albeit not through Zfp423 nuclear translocation. Thus adipogenic commitment and differentiation is regulated by the cross-talk between BMP4 and canonical WNT signaling and where WISP2 plays a key role. Furthermore, they link WISP2 with hypertrophic obesity and the metabolic syndrome.


Subject(s)
Adipose Tissue/metabolism , Bone Morphogenetic Protein 4/metabolism , CCN Intercellular Signaling Proteins/metabolism , Mesenchymal Stem Cells/physiology , PPAR gamma/metabolism , Repressor Proteins/metabolism , Analysis of Variance , Animals , CCN Intercellular Signaling Proteins/genetics , Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Gene Knockdown Techniques , Humans , Immunoblotting , Immunoprecipitation , Mice , Microscopy, Fluorescence , NIH 3T3 Cells , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric , Transcription Factors/metabolism
18.
Am J Physiol Endocrinol Metab ; 297(5): E999-E1003, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19622783

ABSTRACT

Obesity is associated mainly with adipose cell enlargement in adult man (hypertrophic obesity), whereas the formation of new fat cells (hyperplastic obesity) predominates in the prepubertal age. Adipose cell size, independent of body mass index, is negatively correlated with whole body insulin sensitivity. Here, we review recent findings linking hypertrophic obesity with inflammation and a dysregulated adipose tissue, including local cellular insulin resistance with reduced IRS-1 and GLUT4 protein content. In addition, the number of preadipocytes in the abdominal subcutaneous adipose tissue capable of undergoing differentiation to adipose cells is reduced in hypertrophic obesity. This is likely to promote ectopic lipid accumulation, a well-known finding in these individuals and one that promotes insulin resistance and cardiometabolic risk. We also review recent results showing that TNFα, but not MCP-1, resistin, or IL-6, completely prevents normal adipogenesis in preadipocytes, activates Wnt signaling, and induces a macrophage-like phenotype in the preadipocytes. In fact, activated preadipocytes, rather than macrophages, may completely account for the increased release of chemokines and cytokines by the adipose tissue in obesity. Understanding the molecular mechanisms for the impaired preadipocyte differentiation in the subcutaneous adipose tissue in hypertrophic obesity is a priority since it may lead to new ways of treating obesity and its associated metabolic complications.


Subject(s)
Adipogenesis/physiology , Inflammation/physiopathology , Obesity/physiopathology , Adipocytes/drug effects , Adipocytes/physiology , Adipose Tissue/physiology , Cell Differentiation/physiology , Humans , Tumor Necrosis Factor-alpha/pharmacology , Wnt Proteins/physiology
19.
Diabetes Metab Res Rev ; 24(8): 595-603, 2008.
Article in English | MEDLINE | ID: mdl-18756581

ABSTRACT

Type 2 diabetes is the most common metabolic disorder today and has reached epidemic proportions in many countries. Insulin resistance and inflammation play a central role in the pathogenesis of type 2 diabetes and are present long before the onset of the disease. During this time, many of the complications associated with type 2 diabetes are initiated. Of major concern is the two- to fourfold increase in cardiovascular morbidity and mortality in this group compared to a nondiabetic population. Obesity, characterized by enlarged fat cells, and insulin resistance are, like type 2 diabetes, associated with impaired adipogenesis and a low-grade chronic inflammation that to a large extent emanates from the adipose tissue. Both these processes contribute to unfavourable alterations of the circulating levels of several bioactive molecules (adipokines) that are secreted from the adipose tissue, many of which have documented inhibitory effects on insulin sensitivity in the liver and peripheral tissues and, in addition, have negative effects on the cardiovascular system.Here we review current knowledge of the adipose tissue as an endocrine organ, the local and systemic effects of a chronic state of low-grade inflammation residing in the adipose tissue, and, in particular, the effects of inflammation and circulating adipokines on the vascular wall.


Subject(s)
Adipokines/physiology , Adipose Tissue/blood supply , Adipose Tissue/physiopathology , Inflammation/physiopathology , Insulin Resistance , Vascular Diseases/physiopathology , 3T3 Cells , Adipocytes/pathology , Adipocytes/physiology , Animals , Diabetes Mellitus, Type 2/etiology , Humans , Macrophages/physiology , Mice
20.
Exp Cell Res ; 307(1): 259-75, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15922745

ABSTRACT

Signalling from the growth factor receptor subunit and proto-oncogene c-erbB2 has been shown to inhibit the adhesive function of the collagen receptor integrin alpha(2)beta(1) in human mammary epithelial cells. This anti-adhesive effect is mediated by the MAP ERK kinase 1/2 (MEK1/2) and protein kinase B (PKB) pathways. Here, we show that both pathways mediate suppression of matrix adhesion by causing the extracellular domain of the beta(1) integrin subunit to adopt an inactive conformation. The conformational switch was also dependent on rapid and extensive actin depolymerisation. While neither activation nor inhibition of the Rho GTPase affected this rearrangement, Rho was found to be activated by c-erbB2 and to be necessary for conformation-dependent integrin inactivation and, apparently by a different mechanism, a delayed re-formation of stress fibers which did not restore integrin function. Interestingly, the initial actin depolymerisation as well as its effects on integrin function was shown to be mediated by PKB. These results demonstrate how oncogenic growth factor signalling inhibits matrix adhesion by multiple pathways converging on integrin conformation and how Rho signalling can profoundly influence integrin activation in a cytoskeleton-independent manner.


Subject(s)
Actins/metabolism , Integrin beta1/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, ErbB-2/metabolism , rho GTP-Binding Proteins/metabolism , Antibodies, Monoclonal/metabolism , Breast Neoplasms/pathology , Carcinoma/pathology , Cell Adhesion , Cell Line, Tumor , Enzyme Activation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Fluorescent Antibody Technique, Indirect , Humans , Integrin beta1/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt , RNA, Small Interfering/metabolism , Receptor, ErbB-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...