Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Metab ; 53: 101336, 2021 11.
Article in English | MEDLINE | ID: mdl-34481123

ABSTRACT

OBJECTIVE: Activation of brown adipose tissue (BAT) in humans has been proposed as a new treatment approach for combating obesity and its associated diseases, as BAT participates in the regulation of energy homeostasis as well as glucose and lipid metabolism. Genetic contributors driving brown adipogenesis in humans have not been fully understood. METHODS: Profiling the gene expression of progenitor cells from subcutaneous and deep neck adipose tissue, we discovered new secreted factors with potential regulatory roles in white and brown adipogenesis. Among these, members of the latent transforming growth factor beta-binding protein (LTBP) family were highly expressed in brown compared to white adipocyte progenitor cells, suggesting that these proteins are capable of promoting brown adipogenesis. To investigate this potential, we used CRISPR/Cas9 to generate LTBP-deficient human preadipocytes. RESULTS: We demonstrate that LTBP2 and LTBP3 deficiency does not affect adipogenic differentiation, but diminishes UCP1 expression and function in the obtained mature adipocytes. We further show that these effects are dependent on TGFß2 but not TGFß1 signaling: TGFß2 deficiency decreases adipocyte UCP1 expression, whereas TGFß2 treatment increases it. The activity of the LTBP3-TGFß2 axis that we delineate herein also significantly correlates with UCP1 expression in human white adipose tissue (WAT), suggesting an important role in regulating WAT browning as well. CONCLUSIONS: These results provide evidence that LTBP3, via TGFß2, plays an important role in promoting brown adipogenesis by modulating UCP1 expression and mitochondrial oxygen consumption.


Subject(s)
Latent TGF-beta Binding Proteins/metabolism , Transforming Growth Factor beta2/metabolism , Uncoupling Protein 1/metabolism , Adipose Tissue, White/metabolism , CRISPR-Cas Systems/genetics , Cells, Cultured , Humans , Latent TGF-beta Binding Proteins/deficiency , Uncoupling Protein 1/genetics
2.
Biomech Model Mechanobiol ; 18(1): 261-273, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30306291

ABSTRACT

Fibrillin-1 is an elastin-associated glycoprotein that contributes to the long-term fatigue resistance of elastic fibers as well as to the bioavailability of transforming growth factor-beta (TGFß) in arteries. Altered TGFß bioavailability and/or signaling have been implicated in aneurysm development in Marfan syndrome (MFS), a multi-system condition resulting from mutations to the gene that encodes fibrillin-1. We recently showed that the absence of the latent transforming growth factor-beta binding protein-3 (LTBP-3) in fibrillin-1-deficient mice attenuates the fragmentation of elastic fibers and focal dilatations that are characteristic of aortic root aneurysms in MFS mice, at least to 12 weeks of age. Here, we show further that the absence of LTBP-3 in this MFS mouse model improves the circumferential mechanical properties of the thoracic aorta, which appears to be fundamental in preventing or significantly delaying aneurysm development. Yet, a spinal deformity either remains or is exacerbated in the absence of LTBP-3 and seems to adversely affect the axial mechanical properties of the thoracic aorta, thus decreasing overall vascular function despite the absence of aneurysmal dilatation. Importantly, because of the smaller size of mice lacking LTBP-3, allometric scaling facilitates proper interpretation of aortic dimensions and thus the clinical phenotype. While this study demonstrates that LTBP-3/TGFß directly affects the biomechanical function of the thoracic aorta, it highlights that spinal deformities in MFS might indirectly and adversely affect the overall aortic phenotype. There is a need, therefore, to consider together the vascular and skeletal effects in this syndromic disease.


Subject(s)
Aorta/pathology , Aortic Aneurysm, Thoracic/pathology , Latent TGF-beta Binding Proteins/deficiency , Marfan Syndrome/pathology , Spinal Cord/pathology , Animals , Aorta/physiopathology , Aortic Aneurysm, Thoracic/physiopathology , Biomechanical Phenomena , Genotype , Latent TGF-beta Binding Proteins/metabolism , Male , Mice, Inbred C57BL , Phenotype , Spinal Cord/physiopathology
3.
Matrix Biol ; 71-72: 90-99, 2018 10.
Article in English | MEDLINE | ID: mdl-29217273

ABSTRACT

The latent transforming growth factor (TGF) ß binding proteins (LTBP) are crucial mediators of TGFß function, as they control growth factor secretion, matrix deposition, presentation and activation. Deficiencies in specific LTBP isoforms yield discrete phenotypes representing defects in bone, lung and cardiovascular development mediated by loss of TGFß signaling. Additional phenotypes represent loss of unique TGFß-independent features of LTBP effects on elastogenesis and microfibril assembly. Thus, the LTBPs act as sensors for the regulation of both growth factor activity and matrix function.


Subject(s)
Latent TGF-beta Binding Proteins/deficiency , Latent TGF-beta Binding Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Bone Diseases/metabolism , Cardiovascular Diseases/metabolism , Extracellular Matrix/metabolism , Humans , Lung Diseases/metabolism , Signal Transduction
4.
Matrix Biol ; 59: 109-120, 2017 05.
Article in English | MEDLINE | ID: mdl-27645114

ABSTRACT

Latent transforming growth factor beta binding protein 4 (LTBP4) belongs to the fibrillin/LTBP family of proteins and plays an important role as a structural component of extracellular matrix (ECM) and local regulator of TGFß signaling. We have previously reported that Ltbp4S knock out mice (Ltbp4S-/-) develop centrilobular emphysema reminiscent of late stage COPD, which could be partially rescued by inactivating the antioxidant protein Sestrin 2 (Sesn2). More recent studies showed that Sesn2 knock out mice upregulate Pdgfrß-controlled alveolar maintenance programs that protect against cigarette smoke induced pulmonary emphysema. Based on this, we hypothesized that the emphysema of Ltbp4S-/- mice is primarily caused by defective Pdgfrß signaling. Here we show that LTBP4 induces Pdgfrß signaling by inhibiting the antioxidant Nrf2/Keap1 pathway in a TGFß-dependent manner. Overall, our data identified Ltbp4 as a major player in lung remodeling and injury repair.


Subject(s)
Extracellular Matrix/metabolism , Latent TGF-beta Binding Proteins/genetics , NF-E2-Related Factor 2/genetics , Pulmonary Emphysema/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Transforming Growth Factor beta/genetics , Animals , Cell Line , Epithelial Cells/cytology , Epithelial Cells/metabolism , Extracellular Matrix/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Latent TGF-beta Binding Proteins/deficiency , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Mink , NF-E2-Related Factor 2/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peroxidases , Plasmids/chemistry , Plasmids/metabolism , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Tropoelastin/deficiency , Tropoelastin/genetics
5.
Biochem Biophys Res Commun ; 482(4): 1387-1392, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27956181

ABSTRACT

The four latent transforming growth factor-ß (TGF-ß) binding proteins LTBP1-4 are extracellular matrix-associated proteins playing a critical role in the activation of TGF-ß. The LTBP1 gene forms two major transcript variants (i.e. Ltbp1S and Ltbp1L) that are derived from different promoters. We have previously shown the importance of LTBP1 in vivo by using three different Ltbp1 null mice that were either deleted for exons 1 and 2 (Ltbp1L knockout), exon 5 (Ltbp1ΔEx5), or exon 8 (Ltbp1ΔEx8). While the Ltbp1L knockout and the Ltbp1ΔEx8 are perinatal lethal and die of cardiovascular abnormalities, the Ltbp1ΔEx5 is viable because it expresses a short form of Ltbp1L that lacks 55 amino acids (Δ55 variant of Ltbp1) formed by splicing out exon 5, while lacking the Ltbp1S variant. Since only the Ltbp1ΔEx5 mouse is viable, we have used this model to address aspects of puberty, fertility, age-dependent reproduction, and ovary function. We report for the first time a function of LTBP1 in female reproduction. The Ltbp1ΔEx5 females showed impaired fertility associated with delayed sexual maturity (p = 0.0074) and ovarian cyst formation in females older than 40 weeks (p = 0.0204).


Subject(s)
Infertility, Female/genetics , Latent TGF-beta Binding Proteins/genetics , Ovarian Cysts/metabolism , Alternative Splicing , Animals , Carrier Proteins/metabolism , Cells, Cultured , Estrogens/blood , Exons , Extracellular Matrix/metabolism , Female , Fertility , Fertilization in Vitro , Genotype , Latent TGF-beta Binding Proteins/deficiency , Male , Mice , Mice, Knockout , Polymerase Chain Reaction , Progesterone/blood
6.
Proc Natl Acad Sci U S A ; 112(45): 14012-7, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26494287

ABSTRACT

Marfan syndrome (MFS) is an autosomal dominant disorder of connective tissue, caused by mutations of the microfibrillar protein fibrillin-1, that predisposes affected individuals to aortic aneurysm and rupture and is associated with increased TGFß signaling. TGFß is secreted from cells as a latent complex consisting of TGFß, the TGFß propeptide, and a molecule of latent TGFß binding protein (LTBP). Improper extracellular localization of the latent complex can alter active TGFß levels, and has been hypothesized as an explanation for enhanced TGFß signaling observed in MFS. We previously reported the absence of LTBP-3 in matrices lacking fibrillin-1, suggesting that perturbed TGFß signaling in MFS might be due to defective interaction of latent TGFß complexes containing LTBP-3 with mutant fibrillin-1 microfibrils. To test this hypothesis, we genetically suppressed Ltbp3 expression in a mouse model of progressively severe MFS. Here, we present evidence that MFS mice lacking LTBP-3 have improved survival, essentially no aneurysms, reduced disruption and fragmentation of medial elastic fibers, and decreased Smad2/3 and Erk1/2 activation in their aortas. These data suggest that, in MFS, improper localization of latent TGFß complexes composed of LTBP-3 and TGFß contributes to aortic disease progression.


Subject(s)
Aortic Aneurysm, Thoracic/metabolism , Latent TGF-beta Binding Proteins/metabolism , Marfan Syndrome/complications , Marfan Syndrome/genetics , Multiprotein Complexes/metabolism , Transforming Growth Factor beta/metabolism , Analysis of Variance , Animals , Aortic Aneurysm, Thoracic/etiology , DNA, Complementary/biosynthesis , Fibrillin-1 , Fibrillins , Immunohistochemistry , Latent TGF-beta Binding Proteins/deficiency , Mice , Microfilament Proteins/genetics , Muscle, Smooth, Vascular/cytology , Real-Time Polymerase Chain Reaction
7.
Dis Model Mech ; 8(4): 403-15, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25713297

ABSTRACT

Recent studies have revealed an important role for LTBP-4 in elastogenesis. Its mutational inactivation in humans causes autosomal recessive cutis laxa type 1C (ARCL1C), which is a severe disorder caused by defects of the elastic fiber network. Although the human gene involved in ARCL1C has been discovered based on similar elastic fiber abnormalities exhibited by mice lacking the short Ltbp-4 isoform (Ltbp4S(-/-)), the murine phenotype does not replicate ARCL1C. We therefore inactivated both Ltbp-4 isoforms in the mouse germline to model ARCL1C. Comparative analysis of Ltbp4S(-/-) and Ltbp4-null (Ltbp4(-/-)) mice identified Ltbp-4L as an important factor for elastogenesis and postnatal survival, and showed that it has distinct tissue expression patterns and specific molecular functions. We identified fibulin-4 as a previously unknown interaction partner of both Ltbp-4 isoforms and demonstrated that at least Ltbp-4L expression is essential for incorporation of fibulin-4 into the extracellular matrix (ECM). Overall, our results contribute to the current understanding of elastogenesis and provide an animal model of ARCL1C.


Subject(s)
Cutis Laxa/genetics , Cutis Laxa/pathology , Genes, Recessive , Latent TGF-beta Binding Proteins/genetics , Animals , Animals, Newborn , Aorta/abnormalities , Aorta/pathology , Cardiomegaly/complications , Cardiomegaly/pathology , Elastic Tissue/metabolism , Elastin/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Female , Gene Silencing , Glycosylation , Heart Ventricles/pathology , Humans , Latent TGF-beta Binding Proteins/chemistry , Latent TGF-beta Binding Proteins/deficiency , Latent TGF-beta Binding Proteins/metabolism , Lung/abnormalities , Lung/pathology , Mice, Inbred C57BL , Models, Biological , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Skin/pathology , Weight Loss
8.
Hum Mol Genet ; 23(25): 6722-31, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25070948

ABSTRACT

Disruption of the dystrophin complex causes muscle injury, dysfunction, cell death and fibrosis. Excess transforming growth factor (TGF) ß signaling has been described in human muscular dystrophy and animal models, where it is thought to relate to the progressive fibrosis that characterizes dystrophic muscle. We now found that canonical TGFß signaling acutely increases when dystrophic muscle is stimulated to contract. Muscle lacking the dystrophin-associated protein γ-sarcoglycan (Sgcg null) was subjected to a lengthening protocol to produce maximal muscle injury, which produced rapid accumulation of nuclear phosphorylated SMAD2/3. To test whether reducing SMAD signaling improves muscular dystrophy in mice, we introduced a heterozygous mutation of SMAD4 (S4) into Sgcg mice to reduce but not ablate SMAD4. Sgcg/S4 mice had improved body mass compared with Sgcg mice, which normally show a wasting phenotype similar to human muscular dystrophy patients. Sgcg/S4 mice had improved cardiac function as well as improved twitch and tetanic force in skeletal muscle. Functional enhancement in Sgcg/S4 muscle occurred without a reduction in fibrosis, suggesting that intracellular SMAD4 targets may be important. An assessment of genes differentially expressed in Sgcg muscle focused on those encoding calcium-handling proteins and responsive to TGFß since this pathway is a target for mediating improvement in muscular dystrophy. These data demonstrate that excessive TGFß signaling alters cardiac and muscle performance through the intracellular SMAD pathway.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Dystrophies/metabolism , Myocardium/metabolism , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Body Weight , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation , Heart Function Tests , Humans , Latent TGF-beta Binding Proteins/deficiency , Latent TGF-beta Binding Proteins/genetics , Mice , Mice, Knockout , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Mutation , Myocardium/pathology , Phosphorylation , Sarcoglycans/deficiency , Sarcoglycans/genetics , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Smad4 Protein/genetics , Transforming Growth Factor beta/genetics
9.
J Cell Physiol ; 226(6): 1499-509, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20945348

ABSTRACT

The latent TGF-ß binding proteins (LTBP-1 -3, and -4) assist in the secretion and localization of latent TGF-ß molecules. Ltbp3(-/-) and Ltbp4S(-/-) mice have distinct phenotypes and only in the lungs does deficiency of either Ltbp-3 or Ltbp-4 cause developmental abnormalities. To determine if these two LTBPs have additional common functions, we generated mice deficient for both Ltbp-3 and Ltbp-4S. The only novel defect in Ltbp3(-/-);Ltbp4S(-/-) mice was an early lethality compared to mice with single mutations. In addition lung abnormalities were exacerbated and the terminal air sac septation defect was more severe in Ltbp3(-/-);Ltbp4S(-/-) mice than in Ltbp4S(-/-) mice. Decreased cellularity of Ltbp3(-/-);Ltbp4S(-/-) lungs was correlated with higher rate of apoptosis in newborn lungs of Ltbp3(-/-);Ltbp4S(-/-) animals compared to WT, Ltbp3(-/-), and Ltbp4S(-/-) mice. No differences in the maturation of the major lung cell types were discerned between the single and double mutant mice. However, the distribution of type 2 cells and myofibroblasts was abnormal, and myofibroblast segregation in some areas might be an indication of early fibrosis. We also observed differences in ECM composition between Ltbp3(-/-);Ltbp4S(-/-) and Ltbp4S(-/-) lungs after birth, reflected in decreased incorporation of fibrillin-1 and -2 in Ltbp3(-/-);Ltbp4S(-/-) matrix. The function of the lungs of Ltbp3(-/-);Ltbp4S(-/-) mice after the first week of life was potentially further compromised by macrophage infiltration, as proteases secreted from macrophages might exacerbate developmental emphysema. Together these data indicate that LTBP-3 and -4 perform partially overlapping functions only in the lungs.


Subject(s)
Latent TGF-beta Binding Proteins/metabolism , Lung/embryology , Lung/metabolism , Animals , Apoptosis , Cell Differentiation , Elastin/biosynthesis , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Inflammation/metabolism , Inflammation/pathology , Latent TGF-beta Binding Proteins/deficiency , Latent TGF-beta Binding Proteins/genetics , Lung/pathology , Mice , Microfibrils/metabolism , Phenotype
10.
Dis Model Mech ; 3(3-4): 246-53, 2010.
Article in English | MEDLINE | ID: mdl-20106877

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a leading cause of morbidity and mortality worldwide. Cigarette smoking has been identified as one of the major risk factors and several predisposing genetic factors have been implicated in the pathogenesis of COPD, including a single nucleotide polymorphism (SNP) in the latent transforming growth factor (TGF)-beta binding protein 4 (Ltbp4)-encoding gene. Consistent with this finding, mice with a null mutation of the short splice variant of Ltbp4 (Ltbp4S) develop pulmonary emphysema that is reminiscent of COPD. Here, we report that the mutational inactivation of the antioxidant protein sestrin 2 (sesn2) partially rescues the emphysema phenotype of Ltbp4S mice and is associated with activation of the TGF-beta and mammalian target of rapamycin (mTOR) signal transduction pathways. The results suggest that sesn2 could be clinically relevant to patients with COPD who might benefit from antagonists of sestrin function.


Subject(s)
Gene Silencing , Proteins/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Emphysema/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Alleles , Animals , Disease Models, Animal , Enzyme Induction , Fibroblasts/metabolism , Fibroblasts/pathology , Intracellular Signaling Peptides and Proteins , Latent TGF-beta Binding Proteins/deficiency , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Mutation/genetics , Nuclear Proteins , Peroxidases , Protein Serine-Threonine Kinases/biosynthesis , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Emphysema/complications , Pulmonary Emphysema/enzymology , Pulmonary Emphysema/pathology , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases
11.
Biochim Biophys Acta ; 1783(1): 34-48, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17950478

ABSTRACT

Latent transforming growth factor-beta binding proteins are a family of extracellular matrix proteins comprising four isoforms (LTBP-1, -2, -3, -4) with different structures, tissue expression patterns and affinity for TGF-beta. So far, respective knockout models have highlighted some essential functions for LTBP-2, LTBP-3 and LTBP-4, while the physiological significance of LTBP-1 is only superficially known. Here we report for the first time the generation and characterization of a mouse model lacking both the long and short LTBP-1 isoform. Surprisingly, respective mice are viable and fertile. However, detailed X-ray analysis of the skull revealed a modified facial profile. In addition, the gene disruption induces a reduced biological activity of TGF-beta that became evident in an experimental model of hepatic fibrogenesis in which the LTBP-1 knockout animals were less prone to hepatic fibrogenesis. Furthermore, comparative cDNA microarray gene expression profiling of cultured hepatic stellate cells confirmed that respective nulls were less receptive to cellular activation and transdifferentiation into myofibroblasts. Therefore, we conclude that LTBP-1 has essential functions in the control of TGF-beta activation.


Subject(s)
Face/anatomy & histology , Latent TGF-beta Binding Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cells, Cultured , Down-Regulation , Latent TGF-beta Binding Proteins/deficiency , Latent TGF-beta Binding Proteins/genetics , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phenotype , RNA, Messenger/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...