Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 13: 809106, 2022.
Article in English | MEDLINE | ID: mdl-35720339

ABSTRACT

Background and Aims: Myasthenia gravis (MG) is a T-cell dependent antibody-mediated autoimmune disease in which the nicotinic acetylcholine receptor (AChR) is the major autoantigen, comprising several T and B cell auto-epitopes. We hypothesized that an efficacious drug candidate for antigen-specific therapy in MG should comprise a broad range of these auto-epitopes and be administered in a noninflammatory and tolerogenic context. Methods: We used a soluble mutated form of the extracellular domain of the α1 chain of the AChR (α1-ECDm), which represents the major portion of auto-epitopes involved in MG, and investigated, in a well-characterized rat model of experimental autoimmune myasthenia gravis (EAMG) whether its intravenous administration could safely and efficiently treat the autoimmune disease. Results: We demonstrated that intravenous administration of α1-ECDm abrogates established EAMG, in a dose and time dependent manner, as assessed by clinical symptoms, body weight, and compound muscle action potential (CMAP) decrement. Importantly, the effect was more pronounced compared to drugs representing current standard of care for MG. The protein had a short plasma half-life, most of what could be recovered was sequestered in the liver, kidneys and spleen. Further, we did not observe any signs of toxicity or intolerability in animals treated with α1-ECDm. Conclusion: We conclude that intravenous treatment with α1-ECDm is safe and effective in suppressing EAMG. α1-ECDm is in preclinical development as a promising new drug candidate for MG.


Subject(s)
Myasthenia Gravis, Autoimmune, Experimental , Receptors, Nicotinic , Animals , Epitopes, B-Lymphocyte , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Rats , Receptors, Cholinergic , Receptors, Nicotinic/genetics , T-Lymphocytes
2.
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
3.
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.

4.
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
5.
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
6.
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
7.
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
8.
PLoS One ; 6(3): e17957, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21437251

ABSTRACT

BACKGROUND: Notch and Wnt pathways are key regulators of intestinal homeostasis and alterations in these pathways may lead to the development of colorectal cancer (CRC). In CRC the Apc/ß-catenin genes in the Wnt signaling pathway are frequently mutated and active Notch signaling contributes to tumorigenesis by keeping the epithelial cells in a proliferative state. These pathways are simultaneously active in proliferative adenoma cells and a crosstalk between them has previously been suggested in normal development as well as in cancer. PRINCIPAL FINDINGS: In this study, in silico analysis of putative promoters involved in transcriptional regulation of genes coding for proteins in the Notch signaling pathway revealed several putative LEF-1/TCF sites as potential targets for ß-catenin and canonical Wnt signaling. Further results from competitive electrophoretic mobility-shift assay (EMSA) studies suggest binding of several putative sites in Notch pathway gene promoters to in vitro translated ß-catenin/Lef-1. Wild type (wt)-Apc negatively regulates ß-catenin. By induction of wt-Apc or ß-catenin silencing in HT29 cells, we observed that several genes in the Notch pathway, including Notch-2, were downregulated. Finally, active Notch signaling was verified in the Apc(Min/+) mouse model where Hes-1 mRNA levels were found significantly upregulated in intestinal tumors compared to normal intestinal mucosa. Luciferase assays showed an increased activity for the core and proximal Notch-2 promoter upon co-transfection of HCT116 cells with high expression recombinant Tcf-4, Lef-1 or ß-catenin. CONCLUSIONS: In this paper, we identified Notch-2 as a novel target for ß-catenin-dependent Wnt signaling. Furthermore our data supports the notion that additional genes in the Notch pathway might be transcriptionally regulated by Wnt signaling in colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Receptor, Notch2/genetics , Signal Transduction/genetics , Wnt Proteins/metabolism , Adenoma/genetics , Animals , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Binding Sites , Computational Biology , HCT116 Cells , HT29 Cells , Humans , Luciferases/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding , Receptor, Notch2/metabolism , Transcription Factor 4 , Transcription Factors/metabolism , Transcriptional Activation/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...