Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Cell Physiol ; 233(12): 9077-9086, 2018 12.
Article in English | MEDLINE | ID: mdl-29932216

ABSTRACT

Adipose tissue is a dynamic endocrine organ playing a pivotal role in metabolism modulation. Adipocytes differentiation requires a highly orchestrated series of changes of gene expression in precursor cells. At the same time, white mature adipocytes are plastic cells able to reversibly transdifferentiate toward fibroblast-like cells via the liposecretion process, returning back to a non-committed status of the cells. In particular, adipose tissue microenvironment along with external signaling molecules such as adipokines, cytokines and growth factors can regulate adipocytes physiology through complex molecular networks. MicroRNAs (miRNAs), a type of non-coding RNA, acting as fine regulators of biological processes and their expression is sensible to the environment and cellular status changes. MiRNAs are thought to play a pivotal role in regulating the physiology of adipose tissue as well as in the development of obesity and associated metabolic disturbances, although the underlying mechanisms have not been identified so far. Elucidating the molecular mechanisms orchestrating adipose tissue biology is required to better characterize obesity and its associated diseases. In this respect, the review aims to analyze the microRNAs potentially involved in adipogenesis highlighting their role in the process of liposecretion, adipocyte proliferation, and adipokines secretion. The role of microRNAs in the development of obesity and obesity-associated disorders is also discussed.


Subject(s)
Adipocytes/cytology , Adipose Tissue/growth & development , Cell Differentiation/genetics , MicroRNAs/genetics , Adipocytes/metabolism , Adipocytes, White/cytology , Adipocytes, White/metabolism , Adipose Tissue/metabolism , Animals , Cell Proliferation/genetics , Gene Expression Regulation/genetics , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Signal Transduction/genetics
2.
J Cell Physiol ; 233(5): 3784-3793, 2018 05.
Article in English | MEDLINE | ID: mdl-28926092

ABSTRACT

White mature adipocytes (MAs) are plastic cells able to reversibly transdifferentiate toward fibroblast-like cells maintaining stem cell gene signatures. The main morphologic aspect of this transdifferentiation process, called liposecretion, is the secretion of large lipid droplets and the development of organelles necessary for exocrine secretion. There is a considerable interest in the adipocyte plastic properties involving liposecretion process, but the molecular details are incompletely explored. This review analyzes the gene expression of MAs isolated from human subcutaneous fat tissue with respect to bone marrow (BM)-derived mesenchymal stem cells (MSC) focusing on gene regulatory pathways involved into cellular morphology changes, cellular proliferation and transports of molecules through the membrane, suggesting potential ways to guide liposecretion. In particular, Wnt, MAPK/ERK, and AKT pathways were accurately described, studying up- and down-stream molecules involved. Moreover, adipogenic extra- and intra-cellular interactions were analyzed studying the role of CDH2, CDH11, ITGA5, E-Syt1, PAI-1, IGF1, and INHBB genes. Additionally, PLIN1 and PLIN2 could be key-genes of liposecretion process regulating molecules transport through the membrane. All together data demonstrated that liposecretion is regulated through a complex molecular networks that are able to respond to microenvironment signals, cytokines, and growth factors. Autocrine as well as external signaling molecules might activate liposecretion affecting adipocytes physiology.


Subject(s)
Adipocytes/cytology , Adipogenesis/genetics , Cell Differentiation/genetics , Mesenchymal Stem Cells/cytology , Animals , Bone Marrow Cells/cytology , Cell Proliferation/genetics , Humans
3.
J Cell Physiol ; 233(1): 88-97, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28181253

ABSTRACT

Obesity is a condition likely associated with several dysmetabolic conditions or worsening of cardiovascular and other chronic disturbances. A key role in this mechanism seem to be played by the onset of low-grade systemic inflammation, highlighting the importance of the interplay between adipocytes and immune system cells. Adipocytes express a complex and highly adaptive biological profile being capable to selectively activate different metabolic pathways in order to respond to environmental stimuli. It has been demonstrated how adipocytes, under appropriate stimulation, can easily differentiate and de-differentiate thereby converting themselves into different phenotypes according to metabolic necessities. Although underlying mechanisms are not fully understood, growing in adipocyte size and the inability of storing triglycerides under overfeeding conditions seem to be crucial for the switching to a dysfunctional metabolic profile, which is characterized by inflammatory and apoptotic pathways activation, and by the shifting to pro-inflammatory adipokines secretion. In obesity, changes in adipokines secretion along with adipocyte deregulation and fatty acids release into circulation contribute to maintain immune cells activation as well as their infiltration into regulatory organs. Over the well-established role of macrophages, recent findings suggest the involvement of new classes of immune cells such as T regulatory lymphocytes and neutrophils in the development inflammation and multi systemic worsening. Deeply understanding the pathways of adipocyte regulation and the de-differentiation process could be extremely useful for developing novel strategies aimed at curbing obesity-related inflammation and related metabolic disorders.


Subject(s)
Adipocytes/metabolism , Cell Communication , Immune System/metabolism , Inflammation/etiology , Obesity/complications , Adipocytes/immunology , Adipocytes/pathology , Adipogenesis , Adipokines/metabolism , Animals , Apoptosis , Cytokines/metabolism , Humans , Immune System/immunology , Immune System/pathology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Lipid Metabolism , Obesity/immunology , Obesity/metabolism , Obesity/pathology , Phenotype , Signal Transduction
4.
J Cell Physiol ; 233(2): 1500-1511, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28574591

ABSTRACT

In bone marrow (BM), hematopoietic elements are mingled with adipocytes (BM-A), which are the most abundant stromal component in the niche. BM-A progressively increase with aging, eventually occupying up to 50% of BM cavities. In this work, the role played by BM-A was explored by studying primary human BM-A isolated from hip surgery patients at the molecular level, through microarray analysis, and at the functional level, by assessing their relationship with primary human hematopoietic stem cells (HSC) by the long-term culture initiating cell (LTC-IC) assay. Findings demonstrated that BM-A are capable of supporting HSC survival in the LTC-IC assay, since after 5 weeks of co-culture, HSC were still able to proliferate and differentiate. Furthermore, critical molecules such as C-X-C motif chemokine 12 (CXCL12), interleukin (IL)-8, colony-stimulating factor 3 (CSF3), and leukaemia inhibitory factor (LIF), were expressed at similar levels in BM-A and in primary human BM mesenchymal stromal cells (BM-MSC), whereas IL-3 was higher in BM-A. Interestingly, BM-A displayed a different gene expression profile compared with subcutaneous adipose tissue adipocytes (AT-A) collected from abdominal surgery patients, especially in terms of regulation of lipid metabolism, stemness genes, and white-to-brown differentiation pathways. Accordingly, analysis of the gene pathways involved in hematopoiesis regulation showed that BM-A are more closely related to BM-MSC than to AT-A. The present data suggest that BM-A play a supporting role in the hematopoietic niche and directly sustain HSC survival.


Subject(s)
Adipocytes/physiology , Bone Marrow Cells/physiology , Cell Communication , Hematopoietic Stem Cells/physiology , Adipocytes/metabolism , Aged , Aged, 80 and over , Bone Marrow Cells/metabolism , Cell Proliferation , Cell Survival , Cells, Cultured , Chemokine CXCL12/metabolism , Coculture Techniques , Colony-Stimulating Factors/metabolism , Female , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Humans , Interleukin-8/metabolism , Leukemia Inhibitory Factor/metabolism , Male , Middle Aged , Phenotype , Signal Transduction , Stem Cell Niche , Subcutaneous Fat/cytology , Subcutaneous Fat/physiology , Time Factors , Transcriptome
5.
Stem Cells ; 35(12): 2340-2350, 2017 12.
Article in English | MEDLINE | ID: mdl-28905448

ABSTRACT

The transcription factor (TF) yes-associated protein 1 (YAP1) is a major effector of the tumor suppressive Hippo signaling pathway and is also necessary to maintain pluripotency in embryonic stem cells. Elevated levels of YAP1 expression antagonize the tumor suppressive effects of the Hippo pathway that normally represses YAP1 function. High YAP1 expression is observed in several types of human cancers and is particularly prominent in cancer stem cells (CSCs). The stem cell TF Sox2, which marks and maintains CSCs in osteosarcomas (OSs), promotes YAP1 expression by binding to an intronic enhancer element and YAP1 expression is also crucial for the maintainance of OS stem cells. To further understand the regulation of YAP1 expression in OSs, we subjected the YAP1 intronic enhancer to scanning mutagenesis to identify all DNA cis-elements critical for enhancer function. Through this approach, we identified two novel TFs, GA binding protein (GABP) and myeloid zinc finger 1 (MZF1), which are essential for basal YAP1 transcription. These factors are highly expressed in OSs and bind to distinct sites in the YAP1 enhancer. Depletion of either factor leads to drastically reduced YAP1 expression and thus a reversal of stem cell properties. We also found that YAP1 can regulate the expression of Sox2 by binding to two distinct DNA binding sites upstream and downstream of the Sox2 gene. Thus, Sox2 and YAP1 reinforce each others expression to maintain stemness and tumorigenicity in OSs, but the activity of MZF1 and GABP is essential for YAP1 transcription. Stem Cells 2017;35:2340-2350.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , GA-Binding Protein Transcription Factor/metabolism , Kruppel-Like Transcription Factors/metabolism , Phosphoproteins/metabolism , SOXB1 Transcription Factors/metabolism , Cell Line, Tumor , Humans , Neoplastic Stem Cells/metabolism , Signal Transduction/physiology , Transcription Factors , YAP-Signaling Proteins
6.
J Cell Physiol ; 232(10): 2887-2899, 2017 Oct.
Article in English | MEDLINE | ID: mdl-27987321

ABSTRACT

White adipocytes are plastic cells able to reversibly transdifferentiate into brown adipocytes and into epithelial glandular cells under physiologic stimuli in vivo. These plastic properties could be used in future for regenerative medicine, but are incompletely explored in their details. Here, we focused on plastic properties of human mature adipocytes (MA) combining gene expression profile through microarray analysis with morphologic data obtained by electron and time lapse microscopy. Primary MA showed the classic morphology and gene expression profile of functional mature adipocytes. Notably, despite their committed status, MA expressed high levels of reprogramming genes. MA from ceiling cultures underwent transdifferentiation toward fibroblast-like cells with a well-differentiated morphology and maintaining stem cell gene signatures. The main morphologic aspect of the transdifferentiation process was the secretion of large lipid droplets and the development of organelles necessary for exocrine secretion further supported the liposecretion process. Of note, electron microscope findings suggesting liposecretion phenomena were found also in explants of human fat and rarely in vivo in fat biopsies from obese patients. In conclusion, both MA and post-liposecretion adipocytes show a well-differentiated phenotype with stem cell properties in line with the extraordinary plasticity of adipocytes in vivo. J. Cell. Physiol. 232: 2887-2899, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Adipogenesis , Cell Plasticity , Lipid Metabolism , Mesenchymal Stem Cells/metabolism , Obesity/metabolism , Adipocytes, Brown/ultrastructure , Adipocytes, White/ultrastructure , Aged , Aged, 80 and over , Cell Lineage , Cell Shape , Cells, Cultured , Cellular Reprogramming , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Genetic Markers , Humans , Lipid Droplets/metabolism , Mesenchymal Stem Cells/ultrastructure , Microscopy, Confocal , Microscopy, Electron , Microscopy, Video , Middle Aged , Obesity/pathology , Obesity/physiopathology , Oligonucleotide Array Sequence Analysis , Phenotype , Time Factors , Time-Lapse Imaging
7.
J Cell Physiol ; 230(7): 1525-33, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25641257

ABSTRACT

Mature adipocytes have shown dynamic plasticity to be converted into fibroblast-like and lipid-free cells. After the dedifferentiation process, these cells re-entered the cell cycle and acquired a high proliferation potential, becoming a valid source of stem cells. However, many aspects of the cellular biosafety about dedifferentiated fat cells remained unclear. This study aimed to elucidate their potential susceptibility to malignant transformation and to ascertain the safety of these cells for clinical use. To evaluate the genomic stability of dedifferentiated adipocytes, telomere length, hTERT gene transcription, the capacity of these cells to grow in an anchorage-independent manner and the presence of DNA damage by single cell gel electrophoresis assay were studied. Spontaneous chromosomal alterations were excluded by cytogenetic analysis and the expression level of c-myc and p53, tumor associated genes, were assessed, evaluating also p53 loss of function mutations. Despite the high proliferation capacity of dedifferentiated adipocytes, these cells showed stable telomere length compared with mature adipocytes, no hTERT transcriptions and consequently no telomerase activity, suggesting that both transformation and senescence were avoided. A constant expression level of c-myc and p53, the inability of dedifferentiated adipocytes to grow in an anchorage-independent manner, the absence of DNA damage suggested the safety of these cells. Moreover, a normal karyotype was preserved throughout the dedifferentiation process. Data in vivo showed that dedifferentiated adipocytes analyzed for tumorigenicity did not develop tumors. In conclusion, our data indicated that dedifferentiated adipocytes could be a relatively easily accessible resource for cell therapy and regenerative medicine.


Subject(s)
Adipocytes/cytology , Cell Dedifferentiation/physiology , Adipocytes/physiology , Aged , Aged, 80 and over , Animals , Carcinogenesis , Cell Line, Tumor , Comet Assay , Gene Expression Regulation/physiology , Humans , Karyotype , Mice , Mice, Nude , Middle Aged , Neoplasms, Experimental , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Telomerase/genetics , Telomerase/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
Cytotherapy ; 17(9): 1292-301, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26276010

ABSTRACT

BACKGROUND AIMS: Adipose tissue is a critical organ that plays a major role in energy balance regulation and the immune response through intricate signals. METHODS: We report on the inter-relation between mature adipocytes and lymphocytes in terms of adipocyte-derived T-cell chemo-attractants and adipocyte metabolic effects on lymphocytes. RESULTS: During the culture time, mature adipocytes changed their structural and functional properties into de-differentiated cells. Isolated mature adipocytes expressed significantly higher levels of CIITA, major histocompatibility complex II (human leukocyte antigen [HLA]-DR) and costimulatory signal molecule CD80 compared with adipocytes after the de-differentiation process. Moreover, human leukocyte antigen-G, which may prevent the immune responses of mesenchymal stromal cells, was expressed at lower level in mature adipocytes compared with de-differentiated adipocytes. In line with these molecular data, functional results showed different immunoregulatory properties between adipocytes before and after the de-differentiation process. Mature adipocytes stimulated the proliferation of total lymphocytes and immunoselected cell populations CD3+, CD4+ and CD8+ in a direct contact-dependent way that involved the major histocompatibility complex I and II pathways. Moreover, adipocytes secreted potential chemo-attractant factors, but data showed that adipocyte-derived culture medium was not sufficient to activate lymphocyte proliferation, suggesting that a direct contact between adipocytes and immune cells was needed. However, specific mature adipocyte cytokines enhanced lymphocyte proliferation in a mixed lymphocyte reaction. CONCLUSIONS: In conclusion, cross-talk occurs between adipocytes and lymphocytes within adipose tissue involving T-cell chemo-attraction by mature adipocytes. Our findings, together with current observations in the field, provide a rationale to identify adipocyte-lymphocyte cross-talk that instigates adipose inflammation.


Subject(s)
Adipocytes/immunology , Adipose Tissue/immunology , Cell Communication/immunology , Inflammation/immunology , T-Lymphocytes/immunology , Adipocytes/cytology , Adipose Tissue/cytology , Adipose Tissue/physiology , Aged , Aged, 80 and over , B7-1 Antigen/immunology , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , HLA-DR Antigens/immunology , HLA-G Antigens/biosynthesis , Histocompatibility Antigens Class I/metabolism , Humans , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Mesenchymal Stem Cells/immunology , Middle Aged , Nuclear Proteins/immunology , T-Lymphocytes/cytology , Trans-Activators/immunology
9.
Stem Cells ; 30(5): 965-74, 2012 May.
Article in English | MEDLINE | ID: mdl-22367678

ABSTRACT

Mature adipocytes are generally considered terminally differentiated because they have lost their proliferative abilities. Here, we studied the gene expression and functional properties of mature adipocytes isolated from human omental and subcutaneous fat tissues. We also focused on dedifferentiated adipocytes in culture and their morphologies and functional changes with respect to mature adipocytes, stromal-vascular fraction (SVF)-derived mesenchymal stem cells (MSCs) and bone marrow (BM)-derived MSCs. Isolated mature adipocytes expressed stem cell and reprogramming genes. They replicated in culture after assuming a fibroblast-like shape and expanded similarly to SVF- and BM-derived MSCs. During the dedifferentiation process, mature adipocytes lost their lineage gene expression profile, assumed the typical mesenchymal morphology and immunophenotype, expressed stem cell genes and differentiated into multilineage cells. Moreover, during the dedifferentiation process, we showed changes in the epigenetic status of mature adipocytes, which led dedifferentiated adipocytes to display a similar DNA methylation condition to BM-derived MSCs. Like SVF- and BM-derived MSCs, dedifferentiated adipocytes were able to inhibit the proliferation of stimulated lymphocytes in coculture while mature adipocytes stimulated their growth. Furthermore, dedifferentiated adipocytes maintained the survival and complete differentiation characteristic of hematopoietic stem cells. This is the first study that in addition to characterizing isolated and dedifferentiated adipocytes also reports on the immunoregulatory and hematopoietic supporting functions of these cells. This structural and functional characterization might have clinical applications of both mature and dedifferentiated adipocytes in such fields, as regenerative medicine.


Subject(s)
Adipocytes/metabolism , Bone Marrow Cells/metabolism , Cell Dedifferentiation/physiology , Mesenchymal Stem Cells/metabolism , Adipocytes/ultrastructure , Aged , Aged, 80 and over , Bone Marrow Cells/ultrastructure , Cell Proliferation , Cell Survival/physiology , Cells, Cultured , DNA Methylation/physiology , Epigenesis, Genetic/physiology , Female , Humans , Lymphocytes/metabolism , Lymphocytes/ultrastructure , Male , Mesenchymal Stem Cells/ultrastructure , Middle Aged
10.
Mol Metab ; 78: 101813, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37777008

ABSTRACT

OBJECTIVE: Hepatic steatosis is a key initiating event in the pathogenesis of alcohol-associated liver disease (ALD), the most detrimental organ damage resulting from alcohol use disorder. However, the mechanisms by which alcohol induces steatosis remain incompletely understood. We have previously found that alcohol binging impairs brain insulin action, resulting in increased adipose tissue lipolysis by unrestraining sympathetic nervous system (SNS) outflow. Here, we examined whether an impaired brain-SNS-adipose tissue axis drives hepatic steatosis through unrestrained adipose tissue lipolysis and increased lipid flux to the liver. METHODS: We examined the role of lipolysis, and the brain-SNS-adipose tissue axis and stress in alcohol induced hepatic triglyceride accumulation in a series of rodent models: pharmacological inhibition of the negative regulator of insulin signaling protein-tyrosine phosphatase 1ß (PTP1b) in the rat brain, tyrosine hydroxylase (TH) knockout mice as a pharmacogenetic model of sympathectomy, adipocyte specific adipose triglyceride lipase (ATGL) knockout mice, wildtype (WT) mice treated with ß3 adrenergic agonist or undergoing restraint stress. RESULTS: Intracerebral administration of a PTP1b inhibitor, inhibition of adipose tissue lipolysis and reduction of sympathetic outflow ameliorated alcohol induced steatosis. Conversely, induction of adipose tissue lipolysis through ß3 adrenergic agonism or by restraint stress worsened alcohol induced steatosis. CONCLUSIONS: Brain insulin resistance through upregulation of PTP1b, increased sympathetic activity, and unrestrained adipose tissue lipolysis are key drivers of alcoholic steatosis. Targeting these drivers of steatosis may provide effective therapeutic strategies to ameliorate ALD.


Subject(s)
Fatty Liver, Alcoholic , Fatty Liver , Liver Diseases, Alcoholic , Rats , Mice , Animals , Lipolysis , Rodentia/metabolism , Fatty Liver/pathology , Insulin/metabolism , Ethanol/adverse effects , Mice, Knockout , Obesity
12.
Cell Transplant ; 27(5): 754-764, 2018 05.
Article in English | MEDLINE | ID: mdl-29682980

ABSTRACT

Hematopoietic stem and progenitor cells reside within the bone marrow (BM) microenvironment. By a well-balanced interplay between self-renewal and differentiation, they ensure a lifelong supply of mature blood cells. Physiologically, multiple different cell types contribute to the regulation of stem and progenitor cells in the BM microenvironment by cell-extrinsic and cell-intrinsic mechanisms. During the last decades, mesenchymal stromal cells (MSCs) have been identified as one of the main cellular components of the BM microenvironment holding an indispensable role for normal hematopoiesis. During aging, MSCs diminish their functional and regenerative capacities and in some cases encounter replicative senescence, promoting inflammation and cancer progression. It is now evident that alterations in specific stromal cells that comprise the BM microenvironment can contribute to hematologic malignancies, and there is growing interest regarding the contribution of MSCs to the pathogenesis of myelodysplastic syndromes (MDSs), a clonal hematological disorder, occurring mostly in the elderly, characterized by ineffective hematopoiesis and increased tendency to acute myeloid leukemia evolution. The pathogenesis of MDS has been associated with specific genetic and epigenetic events occurring both in hematopoietic stem cells (HSCs) and in the whole BM microenvironment with an aberrant cross talk between hematopoietic elements and stromal compartment. This review highlights the role of MSCs in MDS showing functional and molecular alterations such as altered cell-cycle regulation with impaired proliferative potential, dysregulated cytokine secretion, and an abnormal gene expression profile. Here, the current knowledge of impaired functional properties of both aged MSCs and MSCs in MDS have been described with a special focus on inflammation and senescence induced changes in the BM microenvironment. Furthermore, a better understanding of aberrant BM microenvironment could improve future potential therapies.


Subject(s)
Cellular Senescence , Mesenchymal Stem Cells/pathology , Myelodysplastic Syndromes/pathology , Cellular Microenvironment , Humans , Models, Biological , Phenotype
13.
Oncogene ; 37(33): 4626-4632, 2018 08.
Article in English | MEDLINE | ID: mdl-29743593

ABSTRACT

The stem cell transcription factor Sox2 is highly expressed in many cancers where it is thought to mark cancer stem cells (CSCs). In osteosarcomas, the most common bone malignancy, high Sox2 expression marks and maintains a fraction of tumor-initiating cells that show all the properties of CSC. Knockdown of Sox2 expression abolishes tumorigenicity and suppresses the CSC phenotype. Here we show that, in a mouse model of osteosarcoma, osteoblast-specific Sox2 conditional knockout (CKO) causes a drastic reduction in the frequency and onset of tumors. The rare tumors detected in the Sox2 CKO animals were all Sox2 positive, indicating that they arose from cells that had escaped Sox2 deletion. Furthermore, Sox2 inactivation in cultured osteosarcoma cells by CRISPR/CAS technology leads to a loss of viability and proliferation of the entire cell population. Inactivation of the YAP gene, a major Hippo pathway effector which is a direct Sox2 target, causes similar results and YAP overexpression rescues cells from the lethality caused by Sox2 inactivation. These effects were osteosarcoma-specific, suggesting a mechanism of cell "addiction" to Sox2-initiated pathways. The requirement of Sox2 for osteosarcoma formation as well as for the survival of the tumor cells suggests that disruption of Sox2-initiated pathways could be an effective strategy for the treatment of osteosarcoma.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Proliferation/genetics , Osteosarcoma/genetics , Osteosarcoma/pathology , SOXB1 Transcription Factors/genetics , Animals , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Mice , Mice, Knockout , Neoplastic Stem Cells/pathology , Signal Transduction/genetics
14.
Oncotarget ; 7(38): 60954-60970, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27528232

ABSTRACT

Osteosarcoma (OS) is a highly aggressive pediatric bone cancer in which most tumor cells remain immature and fail to differentiate into bone-forming osteoblasts. However, OS cells readily respond to adipogenic stimuli suggesting they retain mesenchymal stem cell-like properties. Here we demonstrate that nuclear receptor PPARγ agonists such as the anti-diabetic, thiazolidinedione (TZD) drugs induce growth arrest and cause adipogenic differentiation in human, mouse and canine OS cells as well as in tumors in mice. Gene expression analysis reveals that TZDs induce lipid metabolism pathways while suppressing targets of the Hippo-YAP pathway, Wnt signaling and cancer-related proliferation pathways. Significantly, TZD action appears to be restricted to the high Sox2 expressing cancer stem cell population and is dependent on PPARγ expression. TZDs also affect growth and cell fate by causing the cytoplasmic sequestration of the transcription factors SOX2 and YAP that are required for tumorigenicity. Finally, we identify a TZD-regulated gene signature based on Wnt/Hippo target genes and PPARγ that predicts patient outcomes. Together, this work highlights a novel connection between PPARγ agonist in inducing adipogenesis and mimicking the tumor suppressive hippo pathway. It also illustrates the potential of drug repurposing for TZD-based differentiation therapy for osteosarcoma.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Osteosarcoma/metabolism , PPAR gamma/agonists , PPAR gamma/metabolism , Phosphoproteins/metabolism , Adipocytes/cytology , Adipogenesis , Animals , Cell Cycle , Cell Cycle Proteins , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Dogs , Hippo Signaling Pathway , Humans , Lipid Metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Osteosarcoma/genetics , Protein Serine-Threonine Kinases/metabolism , Rosiglitazone , Signal Transduction , Thiazolidinediones/chemistry , Transcription Factors , Wnt Proteins/metabolism , YAP-Signaling Proteins
15.
Exp Hematol ; 43(2): 137-46, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25448487

ABSTRACT

The process of cellular differentiation in terminally differentiated cells is thought to be irreversible, and these cells are thought to be incapable of differentiating into distinct cell lineages. Our previous study showed that mature adipocytes represent an alternative source of mesenchymal stem cells. Here, results showed the capacity of mature adipocytes to differentiate into endothelial-like cells, using the ability of these cells to revert into an immature phase without any relievable chromosomal alterations. Mature adipocytes were isolated from human omental and subcutaneous fat and were dedifferentiated in vitro. The resulting cells were subcultivated for endothelial differentiation and were analyzed for their expression of specific genes and proteins. Endothelial-like cells were harvested from the differentiation medium and were traditionally cultured to evaluate the endothelial markers and the karyotype. Cells cultured in specific medium formed tube-like structures and expressed several endothelial marker genes and proteins. The endothelial-like cells expressed significantly higher levels of vascular endothelium growth factor receptor 2, vascular endothelial cadherin, Von Willebrand factor, and CD133 than the untreated cells. These cells were positively stained for CD31 and vascular endothelial cadherin, markers of mature endothelial cells. Moreover, the low-density lipoprotein-uptake assay demonstrated a functionally endothelial differentiation of these cells. When these cells were harvested and reseeded in basal medium, they lost the endothelial markers and reacquired the typical mesenchymal stem cell markers and the ability to expand in a short time period. Moreover, karyotype analysis showed that these cells reverted into an immature phase without any karyotype alterations. In conclusion, the results showed that adipocytes exhibited a great plasticity toward the endothelial lineage, suggesting their possible use in cell therapy applications for vascular disease.


Subject(s)
Adipocytes/cytology , Endothelial Cells/cytology , Mesenchymal Stem Cells/cytology , Omentum/cytology , Subcutaneous Fat, Abdominal/cytology , AC133 Antigen , Adipocytes/drug effects , Adipocytes/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Biological Transport , Biomarkers/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Dedifferentiation/drug effects , Cell Proliferation/drug effects , Culture Media/chemistry , Culture Media/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Karyotype , Lipoproteins, LDL/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Omentum/metabolism , Peptides/genetics , Peptides/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Primary Cell Culture , Subcutaneous Fat, Abdominal/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
16.
J Mol Neurosci ; 55(1): 91-98, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25007949

ABSTRACT

The potential ability to differentiate dedifferentiated adipocytes into a neural lineage is attracting strong interest as an emerging method of producing model cells for the treatment of a variety of neurological diseases. Here, we describe the efficient conversion of dedifferentiated adipocytes into a neural-like cell population. These cells grew in neurosphere-like structures and expressed a high level of the early neuroectodermal marker Nestin. These neurospheres could proliferate and express stemness genes, suggesting that these cells could be committed to the neural lineage. After neural induction, NeuroD1, Sox1, Double Cortin, and Eno2 were not expressed. Patch clamp data did not reveal different electrophysiological properties, indicating the inability of these cells to differentiate into mature neurons. In contrast, the differentiated cells expressed a high level of CLDN11, as demonstrated using molecular method, and stained positively for the glial cell markers CLDN11 and GFAP, as demonstrated using immunocytochemistry. These data were confirmed by quantitative results for glial cell line-derived neurotrophic factor production, which showed a higher secretion level in neurospheres and the differentiated cells compared with the untreated cells. In conclusion, our data demonstrate morphological, molecular, and immunocytochemical evidence of initial neural differentiation of mature adipocytes, committing to a glial lineage.


Subject(s)
Adipocytes/cytology , Neural Stem Cells/cytology , Neurogenesis , Neuroglia/cytology , Action Potentials , Adipocytes/metabolism , Adipocytes/physiology , Aged , Aged, 80 and over , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Claudins/genetics , Claudins/metabolism , Doublecortin Domain Proteins , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Middle Aged , Nestin/genetics , Nestin/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Neuroglia/metabolism , Neuroglia/physiology , Neuropeptides/genetics , Neuropeptides/metabolism , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
17.
Exp Hematol ; 41(6): 558-566.e2, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23435314

ABSTRACT

Adipocytes are a cell population largely located in the human bone marrow cavity. In this specific microenvironment where adipocytes can interact with a variety of different cells, the role of fat is mainly unknown. To our knowledge, this report is the first to characterize mature adipocytes isolated from human bone marrow (BM-A) molecularly and functionally to better understand their roles into the hematopoietic microenvironment. Healthy BM-A were isolated after collagenase digestion and filtration. We studied the morphology of BM-A, their gene expression and immunophenotypic profile and their functional ability in the hematopoietic microenvironment, comparing them with adipocytes derived from adipose tissue (AT-A). BM-A showed a unilocular lipid morphology similar to AT-A and did not lose their morphology in culture; they showed a comparable pattern of stem cell-surface antigens to AT-A. In line with these observations, molecular data showed that BM-A expressed some embryonic stem cells genes, such as Oct4, KLf4, c-myc, Gata4, Tbx1, and Sox17, whereas they did not express the stem cell markers Sox2 and Nanog. Moreover, BM-A had long telomeres that were similar to bone marrow mesenchymal stem cells. Notably, BM-A supported the survival and differentiation of hematopoietic stem cells in long-term cultures. These results showed that BM-A are stromal cells with a gene expression pattern that distinguished them from AT-A. BM-A showed stem cell properties through their hematopoietic supporting function, which was certainly linked to their role in the maintenance of the bone marrow microenvironment. Depending on specific demands, BM-A may acquire different functions based on their local environment.


Subject(s)
Adipocytes/cytology , Bone Marrow Cells/cytology , Cellular Microenvironment/physiology , Adipocytes/metabolism , Adipocytes/ultrastructure , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Adult , Antigens, Differentiation/analysis , Cell Differentiation , Cell Lineage , Cell Shape , Cytokines/biosynthesis , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Kruppel-Like Factor 4 , Mesoderm/cytology , Middle Aged , Organ Specificity , Organelles/ultrastructure , Stromal Cells/cytology , Subcutaneous Fat/cytology , Telomere/ultrastructure , Transcription Factors/metabolism
18.
Leuk Res ; 37(11): 1538-44, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24095110

ABSTRACT

Telomere dysfunction might generate genomic instability leading to the progression of myelodysplastic syndromes (MDS) into acute myeloid leukemia (AML). We investigated telomere length (TL), telomerase activity (TA) and hTERT, c-myc, mad1, and p53 expression in the bone marrow of patients with MDS (n=109), AML (n=47) and in controls (n=24). TL was lower in MDS patients than in controls (p<0.001) and higher in L-MDS (low, intermediate-1 IPSS, p<0.01) respect H-MDS (high, intermediate-2 IPSS, p<0.01) patients. Mad-1 expression was higher in MDS patients than in controls (p<0.01), c-myc expression was highest in AML and in H-MDS patients. Our results show that the telomere dynamics might be useful for stratifying patients according to a risk scoring system.


Subject(s)
Biomarkers, Tumor/genetics , Cell Cycle Proteins/genetics , Myelodysplastic Syndromes/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Telomere Homeostasis/genetics , Aged , Blotting, Western , Case-Control Studies , Cell Cycle , Female , Follow-Up Studies , Humans , Male , Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/pathology , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/genetics
19.
Cell Transplant ; 20(5): 643-54, 2011.
Article in English | MEDLINE | ID: mdl-21054927

ABSTRACT

Mesenchymal stem cells (MSCs) are promising candidates for cell therapy and tissue engineering. Increasing evidence suggests that MSCs isolated from fetal tissues are more plastic and grow faster than adult MSCs. In this study, we characterized human mesenchymal progenitor cells from chorionic villi (CV) and amniotic fluid (AF) isolated during the first and second trimesters, respectively, and compared them with adult bone marrow-derived MSCs (BM). We evaluated 10 CV, 10 AF, and 6 BM samples expanded until the MSCs reached senescence. We used discarded cells from prenatal analyses for all the experiments. To evaluate the replicative stability of these cells, we studied the telomerase activity, hTERT gene transcription, and telomere length in these cells. Spontaneous chromosomal alterations were excluded by cytogenetic analysis. We studied the expression of c-myc and p53, tumor-associated genes, at different passage in culture and the capacity of these cells to grow in an anchorage-independent manner by using soft agar assay. We isolated homogeneous populations of spindle-shaped CV, AF, and BM cells expressing mesenchymal immunophenotypic markers throughout the period of expansion. CV cells achieved 14 ± 0.9 logs of expansion in 118 days and AF cells achieved 21 ± 0.9 logs in 118 days, while BM cells achieved 11 × 0.4 logs in 84 days. Despite their high proliferation capacity, fetal MSCs showed no telomerase activity, no hTERT and c-myc transcriptions, and maintained long, stable telomeres. A constant expression level of p53 and a normal karyotype were preserved throughout long-term expansion, suggesting the safety of fetal MSCs. In conclusion, our results indicate that fetal MSCs could be an alternative, more accessible resource for cell therapy and regenerative medicine.


Subject(s)
Amniotic Fluid/cytology , Chorionic Villi , Mesenchymal Stem Cells/cytology , Adolescent , Adult , Cell Differentiation , Cells, Cultured , Female , Humans , Karyotyping , Mesenchymal Stem Cells/metabolism , Pregnancy , Proto-Oncogene Proteins c-myc/metabolism , Telomerase/genetics , Telomerase/metabolism , Telomere/physiology , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL