Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Language
Publication year range
1.
J Plast Reconstr Aesthet Surg ; 83: 69-76, 2023 08.
Article in English | MEDLINE | ID: mdl-37270997

ABSTRACT

BACKGROUND: Although autologous fat grafting is considered a successful method for the management of contour deformities, the fat graft could potentially induce cancer reappearance by fueling dormant breast cancer cells. Our aim was to characterize the role of adipose-derived stem cells on active and dormant breast cancer cell growth. METHODS: Cobalt chloride was used to induce dormancy in MCF-7 cancer cells. Proliferation of active and dormant cancer cells was determined in the presence of adipose-derived stem cells. A proteome array was used to detect cancer-related protein expression in the cell-conditioned medium. The migration of cancer cells was measured in response to conditioned medium from the adipose-derived stem cells. RESULTS: The adipose-derived stem cells showed variable effects on active MCF-7 cells growth and inhibited MCF-7 proliferation after the withdrawal of cobalt chloride. Of the 84 different proteins measured in the conditioned medium, only tenascin-C was differentially expressed in the co-cultures. MCF-7 cells alone did not express tenascin-C, whereas co-cultures between MCF-7 and adipose-derived stem cells expressed more tenascin-C versus adipose-derived stem cells alone. The conditioned medium from co-cultures significantly increased the migration of the cancer cells. CONCLUSIONS: Adipose-derived stem cells themselves neither increased the growth or migration of cancer cells, suggesting that autologous fat grafting may be oncologically safe if reconstruction is postponed until there is no evidence of active disease. However, interactions between adipose-derived stem cells and MCF-7 cancer cells could potentially lead to the production of factors, which further promote cancer cell migration.


Subject(s)
Adipose Tissue , Breast Neoplasms , Humans , Female , Adipose Tissue/transplantation , Culture Media, Conditioned/pharmacology , Culture Media, Conditioned/metabolism , Tenascin/metabolism , Tenascin/pharmacology , Stem Cells , Cell Proliferation
2.
Matrix Biol ; 112: 90-115, 2022 09.
Article in English | MEDLINE | ID: mdl-35963565

ABSTRACT

Obesity triggers skeletal muscle physio-pathological alterations. However, the crosstalk between adipose tissue and myogenic cells remains poorly understood during obesity. We identified NID-1 among the adipose tissue secreted factors impairing myogenic potential of human myoblasts and murine muscle stem cells in vitro. Mice under High Fat Diet (HFD) displayed increased NID-1 expression in the skeletal muscle endomysium associated with intramuscular fat adipose tissue expansion and compromised muscle stem cell function. We show that NID-1 is highly secreted by skeletal muscle fibro-adipogenic/mesenchymal progenitors (FAPs) during obesity. We demonstrate that increased muscle NID-1 impairs muscle stem cells proliferation and primes the fibrogenic differentiation of FAPs, giving rise to an excessive deposition of extracellular matrix. Finally, we propose a model in which obesity leads to skeletal muscle extracellular matrix remodeling by FAPs, mediating the alteration of myogenic function by adipose tissue and highlighting the key role of NID-1 in the crosstalk between adipose tissue and skeletal muscle.


Subject(s)
Adipogenesis , Muscle Development , Animals , Cell Differentiation , Extracellular Matrix , Humans , Mice , Muscle, Skeletal/metabolism , Obesity/genetics , Obesity/metabolism
3.
J. physiol. biochem ; 74(4): 613-622, nov. 2018. ilus, graf
Article in English | IBECS | ID: ibc-179039

ABSTRACT

Adipose tissue (AT) expands under obesogenic conditions. Yet, when the growth exceeds a certain limit, AT becomes dysfunctional and surplus lipids start depositing ectopically. Polymerase I and transcription release factor (PTRF) has been proposed as a mechanism leading to a dysfunctional AT by decreasing the adipogenic potential of human adipocyte precursors. However, whether or not PTRF can be secreted by the adipocytes into the bloodstream is not yet known. For this work, PTRF presence was investigated in plasma. We also produced a recombinant PTRF (rPTRF) and examined its impact on the functional interactions between the adipocyte and the hepatocyte in vitro. We demonstrated that PTRF can be found in human plasma, and is at least in part, carried by exosomes. In vitro treatment with rPTRF increased the hypertrophy and senescence of 3T3-L1 adipocytes. In turn, those rPTRF-treated adipocytes increased lipid accumulation in hepatocytes. Lastly, we found a positive correlation between circulating PTRF and the concentration of PTRF in the visceral fat depot. All these findings point toward the presence of an enlarged and dysfunctional visceral adipose tissue which secretes PTRF. This circulating PTRF behaves as an adipokine and may partially contribute to the well-known detrimental effects of visceral fat accumulation


Subject(s)
Humans , Animals , Male , Female , Mice , Exosomes/metabolism , Intra-Abdominal Fat/metabolism , Lipid Metabolism , Membrane Proteins/metabolism , Obesity/metabolism , RNA-Binding Proteins/metabolism , 3T3-L1 Cells , Absorption, Physiological , Cellular Senescence , Cohort Studies , Intra-Abdominal Fat/pathology , Intra-Abdominal Fat/ultrastructure
4.
J Physiol Biochem ; 74(4): 613-622, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29869069

ABSTRACT

Adipose tissue (AT) expands under obesogenic conditions. Yet, when the growth exceeds a certain limit, AT becomes dysfunctional and surplus lipids start depositing ectopically. Polymerase I and transcription release factor (PTRF) has been proposed as a mechanism leading to a dysfunctional AT by decreasing the adipogenic potential of human adipocyte precursors. However, whether or not PTRF can be secreted by the adipocytes into the bloodstream is not yet known. For this work, PTRF presence was investigated in plasma. We also produced a recombinant PTRF (rPTRF) and examined its impact on the functional interactions between the adipocyte and the hepatocyte in vitro. We demonstrated that PTRF can be found in human plasma, and is at least in part, carried by exosomes. In vitro treatment with rPTRF increased the hypertrophy and senescence of 3T3-L1 adipocytes. In turn, those rPTRF-treated adipocytes increased lipid accumulation in hepatocytes. Lastly, we found a positive correlation between circulating PTRF and the concentration of PTRF in the visceral fat depot. All these findings point toward the presence of an enlarged and dysfunctional visceral adipose tissue which secretes PTRF. This circulating PTRF behaves as an adipokine and may partially contribute to the well-known detrimental effects of visceral fat accumulation.


Subject(s)
Exosomes/metabolism , Intra-Abdominal Fat/metabolism , Lipid Metabolism , Membrane Proteins/metabolism , Obesity/metabolism , RNA-Binding Proteins/metabolism , 3T3-L1 Cells , Absorption, Physiological , Animals , Cell Size , Cellular Senescence , Cohort Studies , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/metabolism , Exosomes/pathology , Exosomes/ultrastructure , Female , Glucose/metabolism , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/ultrastructure , Humans , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/pathology , Intra-Abdominal Fat/ultrastructure , Male , Membrane Proteins/genetics , Mice , Microscopy, Electron, Transmission , Obesity/blood , Obesity/pathology , RNA-Binding Proteins/blood , RNA-Binding Proteins/genetics , Recombinant Proteins/metabolism , Subcutaneous Fat, Abdominal/metabolism , Subcutaneous Fat, Abdominal/pathology , Subcutaneous Fat, Abdominal/ultrastructure
5.
Am J Physiol Cell Physiol ; 312(1): C83-C91, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27856429

ABSTRACT

Healthy expansion of human adipose tissue requires mesenchymal stem cells (hMSC) able to proliferate and differentiate into mature adipocytes. Hence, characterization of those factors that coordinate hMSC-to-adipocyte transition is of paramount importance to modulate the adipose tissue expansion. It has been previously reported that the adipogenic program of hMSC can be disrupted by upregulating caveolar proteins, and polymerase I and transcript release factor (PTRF) is an integral component of caveolae, highly expressed in adipose tissue. Here, we hypothesized that the role of PTRF in adipocyte functionality might stem from an effect on hMSC. To test this hypothesis, we isolated hMSC from the subcutaneous fat depot. We found an upregulated expression of the PTRF associated with decreased adipogenic potential of hMSC, likely due to the existence of senescent adipocyte precursors. Employing short hairpin RNA-based constructs to stably reduce PTRF, we were able to restore insulin sensitivity and reduced basal lipolysis and leptin levels in human adipocytes with high levels of PTRF. Additionally, we pinpointed the detrimental effect caused by PTRF on the adipose tissue to the existence of senescent adipocyte precursors unable to proliferate and differentiate into adipocytes. This study provides evidence that impaired adipocyte functionality can be corrected, at least partially, by PTRF downregulation and warrants further in vivo research in patients with dysfunctional adipose tissue to prevent metabolic complications.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Cell Differentiation/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , RNA-Binding Proteins/metabolism , Adipogenesis/physiology , Adult , Aged , Aged, 80 and over , Cells, Cultured , Female , Gene Knockdown Techniques , Humans , Male , Middle Aged
6.
FASEB J ; 28(8): 3769-79, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24812087

ABSTRACT

Impaired adipogenesis renders an adipose tissue unable to expand, leading to lipotoxicity and conditions such as diabetes and cardiovascular disease. While factors important for adipogenesis have been studied extensively, those that set the limits of adipose tissue expansion remain undetermined. Feeding a Western-type diet to apolipoprotein E2 knock-in mice, a model of metabolic syndrome, produced 3 groups of equally obese mice: mice with normal glucose tolerance, hyperinsulinemic yet glucose-tolerant mice, and prediabetic mice with impaired glucose tolerance and reduced circulating insulin. Using proteomics, we compared subcutaneous adipose tissues from mice in these groups and found that the expression of PTRF (polymerase I and transcript release factor) associated selectively with their glucose tolerance status. Lentiviral and pharmacologically overexpressed PTRF, whose function is critical for caveola formation, compromised adipocyte differentiation of cultured 3T3-L1cells. In human adipose tissue, PTRF mRNA levels positively correlated with markers of lipolysis and cellular senescence. Furthermore, a negative relationship between telomere length and PTRF mRNA levels was observed in human subcutaneous fat. PTRF is associated with limited adipose tissue expansion underpinning the key role of caveolae in adipocyte regulation. Furthermore, PTRF may be a suitable adipocyte marker for predicting pathological obesity and inform clinical management.


Subject(s)
Adipocytes/pathology , Adipogenesis/physiology , Caveolae/physiology , Diet/adverse effects , Glucose Intolerance/etiology , Hyperinsulinism/etiology , Obesity/etiology , Prediabetic State/etiology , RNA-Binding Proteins/physiology , Subcutaneous Fat/metabolism , 3T3-L1 Cells , Adiponectin/blood , Animals , Aorta/pathology , Apolipoprotein E2/genetics , Cellular Senescence , Female , Gene Expression Profiling , Gene Knock-In Techniques , Glucose Intolerance/blood , Glucose Intolerance/pathology , Humans , Hyperinsulinism/blood , Hyperinsulinism/pathology , Insulin Resistance , Lipolysis , Liver/chemistry , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/classification , Obesity/pathology , Prediabetic State/blood , Prediabetic State/pathology , Pregnancy , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins/metabolism , Subcutaneous Fat/pathology , Telomere Shortening , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...