Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Stem Cells ; 40(4): 385-396, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35262736

ABSTRACT

Lin28A is an RNA-binding protein that controls mammalian development and maintenance of the pluripotency of embryonic stem cells (ESCs) via regulating the processing of the microRNA let-7. Lin28A is highly expressed in ESCs, and ectopic expression of this protein facilitates reprogramming of somatic cells to induced pluripotent stem cells. However, the mechanisms underlying the post-translational regulation of Lin28A protein stability in ESCs remain unclear. In the present study, we identified Kap1 (KRAB-associated protein 1) as a novel Lin28A-binding protein using affinity purification and mass spectrometry. Kap1 specifically interacted with the N-terminal region of Lin28A through its coiled-coil domain. Kap1 overexpression significantly attenuated Lin28A ubiquitination and increased its stability. However, small interfering RNA-mediated knockdown of Kap1 promoted the ubiquitination of Lin28A, leading to its proteasomal degradation. Trim71, an E3 ubiquitin ligase, induced Lin28A degradation and Kap1 knockdown accelerated the Trim71-dependent degradation of Lin28A. Mutation of the lysine 177 residue of Lin28A to arginine abrogated the ubiquitination and degradation of Lin28A which were accelerated by Kap1 silencing. Moreover, Kap1 overexpression led to the accumulation of Lin28A in the cytoplasm, but not in the nucleus, and reduced the levels of let-7 subtypes. These results suggest that Kap1 plays a key role in regulation of the stability of Lin28A by modulating the Trim71-mediated ubiquitination and subsequent degradation of Lin28A, thus playing a pivotal role in the regulation of ESC self-renewal and pluripotency.


Subject(s)
Embryonic Stem Cells , Induced Pluripotent Stem Cells , Animals , Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Mammals , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Ubiquitination
2.
Mol Cells ; 44(7): 481-492, 2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34326276

ABSTRACT

Tribbles homolog 2 (TRIB2) is implicated in tumorigenesis and drug resistance in various types of cancers. However, the role of TRIB2 in the regulation of tumorigenesis and drug resistance of cancer stem cells (CSCs) is still elusive. In the present study, we showed increased expression of TRIB2 in spheroid-forming and aldehyde dehydrogenase-positive CSC populations of A2780 epithelial ovarian cancer cells. Short hairpin RNA-mediated silencing of TRIB2 expression attenuates the spheroid-forming, migratory, tumorigenic, and drug-resistant properties of A2780 cells, whereas overexpression of TRIB2 increases the CSC-like characteristics. TRIB2 overexpression induced GSK3ß inactivation by augmenting AKT-dependent phosphorylation of GSK3ß at Ser9, followed by increasing ß-catenin level via reducing the GSK3ß-mediated phosphorylation of ß-catenin. Treatment of TRIB2-ovexpressed A2780 cells with the phosphoinositide-3-kinase inhibitor LY294002 abrogated TRIB2-stimulated proliferation, migration, drug resistance of A2780 cells. These results suggest a critical role for TRIB2 in the regulation of CSC-like properties by increasing the stability of ß-catenin protein via the AKT-GSK3ß-dependent pathways.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , beta Catenin/metabolism , Humans , Signal Transduction
3.
Cell Death Differ ; 28(2): 685-699, 2021 02.
Article in English | MEDLINE | ID: mdl-32895487

ABSTRACT

Oct4 plays a crucial role in the regulation of self-renewal of embryonic stem cells (ESCs) and reprogramming of somatic cells to induced pluripotent stem cells. However, the molecular mechanisms underlying posttranslational regulation and protein stability of Oct4 remain unclear. Using affinity purification and mass spectrometry analysis, we identified Kap1 as an Oct4-binding protein. Silencing of Kap1 reduced the protein levels of Oct4 in ESCs, whereas the overexpression of Kap1 stimulated the levels of Oct4. In addition, Kap1 overexpression stimulated the self-renewal of ESCs and attenuated the spontaneous differentiation of ESCs in response to LIF withdrawal. Kap1 overexpression increased the stability of Oct4 by inhibiting the Itch-mediated ubiquitination of Oct4. Silencing of Kap1 augmented Itch-mediated ubiquitination and inhibited the stability of Oct4. We identified the lysine 133 (K133) residue in Oct4 as a ubiquitination site responsible for the Kap1-Itch-dependent regulation of Oct4 stability. Preventing ubiquitination at the lysine residue by mutation to arginine augmented the reprogramming of mouse embryonic fibroblasts to induced pluripotent stem cells. These results suggest that Kap1 plays a crucial role in the regulation of the pluripotency of ESCs and somatic cell reprogramming by preventing Itch-mediated ubiquitination and the subsequent degradation of Oct4.


Subject(s)
Cellular Reprogramming/physiology , Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Animals , Cells, Cultured , Female , Fibroblasts/metabolism , Lysine/metabolism , Mice , Mice, Inbred C3H , Protein Stability , Ubiquitination
4.
Stem Cell Res Ther ; 9(1): 197, 2018 07 18.
Article in English | MEDLINE | ID: mdl-30021650

ABSTRACT

BACKGROUND: Endothelial cells have been shown to mediate angiogenesis in ischemic injury sites and contribute to the repair of damaged tissues. However, the treatment of ischemic disease requires a significant number of endothelial cells, which are difficult to isolate from patients. Embryonic stem cells have been considered a potential source of therapeutic cells due to their unlimited self-renewal and pluripotent properties. With regard to vascular development, Notch1 has been established as a key regulator of the specification of arterial endothelial cells. METHODS: Using a doxycycline-induced expression system of the intracellular domain of Notch1, we explored the role of Notch1 in the differentiation of embryonic stem cells to arterial endothelial cells. The therapeutic effect of the arterial endothelial cells was investigated in a murine hindlimb ischemia model. The blood perfusion rate in the ischemic limb was determined by laser Doppler perfusion imaging, and vasculogenesis was quantified using immunocytochemistry. RESULTS: Induced expression of the intracellular domain of Notch1 increased the levels of endothelial markers, such as CD31 and VE-cadherin, in differentiated endothelial cells. Induction of intracellular domain of Notch1 stimulated expression of the arterial-type endothelial cell markers (Nrp1 and Ephrin B2), but not the venous-type endothelial cell markers (Nrp2 and Coup-TFII). In addition, overexpression of intracellular domain of Notch1 resulted in increased expression of CXCR4, a chemokine receptor involved in vascular development. Induction of intracellular domain of Notch1 increased endothelial tube formation and migration of differentiated endothelial cells. Intramuscular administration of Notch1-induced arterial endothelial cells was more effective than administration of the control endothelial cells in restoring the blood flow in an ischemic hindlimb mouse model. Transplantation of Notch1-induced arterial endothelial cells augmented the number of blood vessels and incorporation of endothelial cells into newly formed blood vessels. CONCLUSIONS: These results suggest that Notch1 promotes endothelial maturation and arterial specification during the differentiation of embryonic stem cells to endothelial cells and increases the angiogenic potential of endothelial cells.


Subject(s)
Endothelial Cells/metabolism , Mouse Embryonic Stem Cells/metabolism , Receptor, Notch1/metabolism , Animals , Cell Movement , Flow Cytometry , Humans , Male , Mice
5.
Wound Repair Regen ; 26(2): 116-126, 2018 03.
Article in English | MEDLINE | ID: mdl-29802745

ABSTRACT

Atrial natriuretic peptide (ANP) is a powerful vasodilating peptide secreted by cardiac muscle cells, and endothelial progenitor cells (EPCs) have been reported to stimulate cutaneous wound healing by mediating angiogenesis. To determine whether ANP can promote the EPC-mediated repair of injured tissues, we examined the effects of ANP on the angiogenic properties of EPCs and on cutaneous wound healing. In vitro, ANP treatment enhanced the migration, proliferation, and endothelial tube-forming abilities of EPCs. Furthermore, small interfering RNA-mediated silencing of natriuretic peptide receptor-1, which is a receptor for ANP, abrogated ANP-induced migration, tube formation, and proliferation of EPCs. In a murine cutaneous wound model, administration of either ANP or EPCs had no significant effect on cutaneous wound healing or angiogenesis in vivo, whereas the coadministration of ANP and EPCs synergistically potentiated wound healing and angiogenesis. In addition, ANP promoted the survival and incorporation of transplanted EPCs into newly formed blood vessels in wounds. These results suggest ANP accelerates EPC-mediated cutaneous wound healing by promoting the angiogenic properties and survival of transplanted EPCs.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Endothelial Progenitor Cells/physiology , Neovascularization, Physiologic/drug effects , Wound Healing/drug effects , Wounds and Injuries/pathology , Animals , Cell Proliferation/physiology , Cells, Cultured , Disease Models, Animal , Endothelial Progenitor Cells/drug effects , Humans , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction
6.
Exp Mol Med ; 49(11): e401, 2017 11 24.
Article in English | MEDLINE | ID: mdl-29170476

ABSTRACT

Embryonic stem (ES) cells are pluripotent cells characterized by self-renewability and differentiation potential. Induced pluripotent stem (iPS) cells are ES cell-equivalent cells derived from somatic cells by the introduction of core reprogramming factors. ES and iPS cells are important sources for understanding basic biology and for generating therapeutic cells for clinical applications. Tribbles homolog 2 (Trib2) functions as a scaffold in signaling pathways. However, the relevance of Trib2 to the pluripotency of ES and iPS cells is unknown. In the present study, we elucidated the importance of Trib2 in maintaining pluripotency in mouse ES cells and in generating iPS cells from somatic cells through the reprogramming process. Trib2 expression decreased as ES cells differentiated, and Trib2 knockdown in ES cells changed their colony morphology while reducing the activity of alkaline phosphatase and the expression of the pluripotency marker genes Oct4, Sox2, Nanog and Klf4. Trib2 directly interacted with Oct4 and elevated Oct4 promoter activity. During the generation of iPS cells, Trib2 knockdown decreased the reprogramming efficiency of mouse embryonic fibroblasts, whereas Trib2 overexpression significantly increased their reprogramming efficiency. In summary, our results suggest that Trib2 is important for maintaining self-renewal in ES cells and for pluripotency induction during the reprogramming process.


Subject(s)
Cellular Reprogramming , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Biomarkers , Cell Differentiation/genetics , Cells, Cultured , Cellular Reprogramming Techniques , Gene Expression Regulation, Developmental , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kruppel-Like Factor 4 , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
7.
Blood ; 125(9): 1418-26, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25587036

ABSTRACT

Hematopoietic and vascular development share many common features, including cell surface markers and sites of origin. Recent lineage-tracing studies have established that definitive hematopoietic stem and progenitor cells arise from vascular endothelial-cadherin(+) hemogenic endothelial cells of the aorta-gonad-mesonephros region, but the genetic programs underlying the specification of hemogenic endothelial cells remain poorly defined. Here, we discovered that Notch induction enhances hematopoietic potential and promotes the specification of hemogenic endothelium in differentiating cultures of mouse embryonic stem cells, and we identified Foxc2 as a highly upregulated transcript in the hemogenic endothelial population. Studies in zebrafish and mouse embryos revealed that Foxc2 and its orthologs are required for the proper development of definitive hematopoiesis and function downstream of Notch signaling in the hemogenic endothelium. These data establish a pathway linking Notch signaling to Foxc2 in hemogenic endothelial cells to promote definitive hematopoiesis.


Subject(s)
Embryonic Stem Cells/cytology , Endothelium, Vascular/cytology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Receptor, Notch1/metabolism , Animals , Apoptosis , Blotting, Western , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Embryonic Stem Cells/metabolism , Endothelium, Vascular/metabolism , Forkhead Transcription Factors/genetics , Hematopoietic Stem Cells/metabolism , Mice , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Notch1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
8.
Stem Cells ; 32(12): 3126-36, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25185564

ABSTRACT

Oct4 has been implicated in regulation of pluripotency in embryonic stem cells (ESCs) and reprogramming of somatic cells into induced pluripotent stem cells. However, the molecular mechanisms involved in Oct4-dependent regulation of pluripotency and reprogramming have not been clear. To gain insight into the mechanism of regulation of Oct4-mediated self-renewal of ESCs and reprogramming of somatic cells, we attempted to identify Oct4-binding proteins using affinity purification and mass spectrometry. We identified Reptin, a key component of ATP-dependent chromatin remodeling complexes, as an Oct4-binding protein. Depletion of endogenous Reptin using lentiviral short hairpin RNA (shRNA) led to a decrease in the number and size of alkaline phosphatase-positive colonies of mouse ESCs. In addition, shRNA-mediated silencing of Reptin resulted in decreased expression of pluripotency-specific marker genes, including Oct4, Sox2, Nanog, and SSEA-1. Results of the Oct4 reporter assay showed synergism between Oct4 and Reptin, and depletion of endogenous Reptin abolished Oct4 transcriptional activity. Results of a chromatin immunoprecipitation assay showed the overlapping interaction of Reptin and Oct4 to CR4 in the Oct4 enhancer in ESCs. Knockdown of Reptin using shRNA suppressed the reprogramming of mouse embryonic fibroblasts to induced pluripotent stem cells, whereas overexpression of Reptin resulted in enhanced efficiency of induced pluripotent stem cell generation. These results strongly suggest that Reptin plays a key role in maintaining the pluripotency of ESCs and in establishing the pluripotency during reprogramming of somatic cells by regulation of Oct4-mediated gene regulation.


Subject(s)
Cell Differentiation/physiology , Cellular Reprogramming , DNA Helicases/metabolism , Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , Animals , Cells, Cultured , Cellular Reprogramming/physiology , Mice
9.
PLoS One ; 8(9): e76875, 2013.
Article in English | MEDLINE | ID: mdl-24098810

ABSTRACT

Reprogramming of somatic cells to pluripotent cells requires the introduction of factors driving fate switches. Viral delivery has been the most efficient method for generation of induced pluripotent stem cells. Transfection, which precedes virus production, is a commonly-used process for delivery of nucleic acids into cells. The aim of this study is to evaluate the efficiency of PLGA/ bPEI nanoparticles in transfection and virus production. Using a modified method of producing PLGA nanoparticles, PLGA/bPEI-DNA nanoparticles were examined for transfection efficiency and virus production yield in comparison with PLGA-DNA, bPEI-DNA nanoparticles or liposome-DNA complexes. After testing various ratios of PLGA, bPEI, and DNA, the ratio of 6:3:1 (PLGA:bPEI:DNA, w/w/w) was determined to be optimal, with acceptable cellular toxicity. PLGA/bPEI-DNA (6:3:1) nanoparticles showed superior transfection efficiency, especially in multiple gene transfection, and viral yield when compared with liposome-DNA complexes. The culture supernatants of HEK293FT cells transfected with PLGA/bPEI-DNA of viral constructs containing reprogramming factors (Oct4, Sox2, Klf4, or c-Myc) successfully and more efficiently generated induced pluripotent stem cell colonies from mouse embryonic fibroblasts. These results strongly suggest that PLGA/bPEI-DNA nanoparticles can provide significant advantages in studying the effect of multiple factor delivery such as in reprogramming or direct conversion of cell fate.


Subject(s)
Cell Dedifferentiation/genetics , DNA/chemistry , Lactic Acid/chemistry , Nanoparticles/chemistry , Polyethyleneimine/chemistry , Polyglycolic Acid/chemistry , Retroviridae/genetics , Transfection/methods , Animals , Flow Cytometry , Genetic Therapy/methods , HEK293 Cells , Humans , Immunohistochemistry , Kruppel-Like Factor 4 , Mice , Microscopy, Fluorescence , NIH 3T3 Cells , Polylactic Acid-Polyglycolic Acid Copolymer
10.
Int J Biochem Cell Biol ; 45(8): 1869-77, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23735324

ABSTRACT

Oncostatin M, a member of the interleukin-6 family of cytokines, has been implicated in tumorigenesis of human prostate cancer. In the current study, we demonstrate that oncostatin M promotes human adipose tissue-derived mesenchymal stem cell-stimulated tumor growth in an in vivo xenograft transplantation model of the human prostate cancer cell line PC-3M-luc-C6, a PC3M cell line expressing the luciferase gene. Conditioned medium derived from oncostatin M-treated mesenchymal stem cells stimulated adhesion of PC-3M-luc-C6 cells. We identified TGFBI and periostin, extracellular matrix proteins implicated in tumorigenesis and metastasis, as oncostatin M-induced secreted proteins in mesenchymal stem cells. Treatment with oncostatin M stimulated secretion of periostin and TGFBI from mesenchymal stem cells in a time-dependent manner. Immunodepletion of TGFBI and periostin from conditioned medium derived from oncostatin M-treated mesenchymal stem cells resulted in abrogation of adhesion of PC-3M-luc-C6 cells stimulated by oncostatin M-conditioned medium. In addition, small interfering RNA-mediated silencing of TGFBI and periostin resulted in abrogation of cell adhesion stimulated by oncostatin M-conditioned medium. These results suggest that mesenchymal stem cell-derived TGFBI and periostin play a key role in tumorigenesis by stimulating adhesion of prostate cancer cells.


Subject(s)
Cell Adhesion Molecules/metabolism , Extracellular Matrix Proteins/metabolism , Mesenchymal Stem Cells/cytology , Oncostatin M/pharmacology , Paracrine Communication/drug effects , Prostatic Neoplasms/pathology , Transforming Growth Factor beta/metabolism , Adipose Tissue/cytology , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Gene Silencing/drug effects , Humans , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , RNA, Small Interfering/metabolism , Recombinant Proteins/pharmacology , Stem Cell Transplantation , Xenograft Model Antitumor Assays
11.
Arterioscler Thromb Vasc Biol ; 32(11): 2733-40, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22962328

ABSTRACT

OBJECTIVE: Mesenchymal stem cells are useful for vascular regeneration of injured tissues. Macrophages are involved in acute or chronic inflammatory diseases, and interleukin-1ß (IL-1ß), a proinflammatory cytokine, plays a key role in the activation of macrophages within injured tissues. To explore the role of macrophages on mesenchymal stem cell-mediated vascular regeneration, we examined the effects of IL-1ß-activated macrophages on differentiation of human adipose tissue-derived mesenchymal stem cells (hASCs) to smooth muscle cells (SMCs) and the vascular regenerative capacity of the differentiated SMCs in a hindlimb ischemia animal model. METHODS AND RESULTS: We demonstrate that IL-1ß-conditioned medium from RAW 264.7 macrophages induces differentiation of human adipose tissue-derived mesenchymal stem cells to α-smooth muscle actin-positive SMCs, and the differentiated SMCs exhibited increased contractility in response to KCl and carbachol treatment. Transplantation of the differentiated SMCs attenuated severe hindlimb ischemia and promoted vascular regeneration. IL-1ß treatment stimulated secretion of prostaglandin F(2α) from RAW 264.7 cells. Small interfering RNA-mediated silencing of the prostaglandin F(2α) receptor completely abrogated IL-1ß conditioned medium-stimulated α-smooth muscle actin expression. Moreover, prostaglandin F(2α) treatment stimulated expression of α-smooth muscle actin in human adipose tissue-derived mesenchymal stem cells. CONCLUSIONS: These results suggest that IL-1ß-activated macrophages promote differentiation of human adipose tissue-derived mesenchymal stem cells to SMCs through a prostaglandin F(2α)-mediated paracrine mechanism.


Subject(s)
Cell Differentiation , Dinoprost/metabolism , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Muscle, Skeletal/blood supply , Myocytes, Smooth Muscle/metabolism , Paracrine Communication , Actins/metabolism , Adipose Tissue/cytology , Animals , Biomarkers/metabolism , Cell Line , Coculture Techniques , Culture Media, Conditioned/metabolism , Disease Models, Animal , Hindlimb , Humans , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Ischemia/immunology , Ischemia/metabolism , Ischemia/physiopathology , Ischemia/surgery , Macrophage Activation , Macrophages/immunology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Mice , Mice, Nude , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/transplantation , Neovascularization, Physiologic , RNA Interference , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Regional Blood Flow , Time Factors , Transfection
12.
Int J Biochem Cell Biol ; 44(11): 2069-76, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22903068

ABSTRACT

Lysophosphatidic acid (LPA) is involved in mesenchymal stem cell-stimulated tumor growth in vivo. However, the molecular mechanism by which mesenchymal stem cells promote tumorigenesis remains elusive. In the present study, we demonstrate that conditioned medium from A549 human lung adenocarcinoma cells (A549 CM) induced the expression of ADAM12, a disintegrin and metalloproteases family member, in human adipose tissue-derived mesenchymal stem cells (hASCs). A549 CM-stimulated ADAM12 expression was abrogated by pretreatment of hASCs with the LPA receptor 1 inhibitor Ki16425 or by small interfering RNA-mediated silencing of LPA receptor 1, suggesting a key role for the LPA-LPA receptor 1 signaling axis in A549 CM-stimulated ADAM12 expression. Silencing of ADAM12 expression using small interfering RNA or short hairpin RNA abrogated LPA-induced expression of both α-smooth muscle actin, a marker of carcinoma-associated fibroblasts, and ADAM12 in hASCs. Using a xenograft transplantation model of A549 cells, we demonstrated that silencing of ADAM12 inhibited the hASC-stimulated in vivo growth of A549 xenograft tumors and the differentiation of transplanted hASCs to α-smooth muscle actin-positive carcinoma-associated fibroblasts. LPA-conditioned medium from hASCs induced the adhesion of A549 cells and silencing of ADAM12 inhibited LPA-induced expression of extracellular matrix proteins, periostin and ßig-h3, in hASCs and LPA-conditioned medium-stimulated adhesion of A549 cells. These results suggest a pivotal role for LPA-stimulated ADAM12 expression in tumor growth and the differentiation of hASCs to carcinoma-associated fibroblasts expressing α-smooth muscle actin, periostin, and ßig-h3.


Subject(s)
ADAM Proteins/metabolism , Adipose Tissue/cytology , Lysophospholipids/pharmacology , Membrane Proteins/metabolism , Mesenchymal Stem Cells/enzymology , Neoplasms/pathology , ADAM12 Protein , Actins/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Extracellular Matrix Proteins/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Silencing/drug effects , Humans , Lung Neoplasms/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Nude , Neoplasms/metabolism , RNA, Small Interfering/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
13.
PLoS One ; 7(7): e40820, 2012.
Article in English | MEDLINE | ID: mdl-22808268

ABSTRACT

BACKGROUND: Transforming growth factor-ß1 (TGF-ß1) induces the differentiation of human adipose tissue-derived mesenchymal stem cells (hASCs) into smooth muscle cells. Lipid rafts are cholesterol-rich microdomains in cell membranes that reportedly play a key role in receptor-mediated signal transduction and cellular responses. In order to clarify whether lipid rafts are involved in TGF-ß1-induced differentiation of hASCs into smooth muscle cells, we analyzed the lipid raft proteome of hASCs. METHODS AND RESULTS: Pretreatment of hASCs with the lipid raft disruptor methyl-ß-cyclodextrin abrogated TGF-ß1-induced expression of α-smooth muscle actin, a smooth muscle cell marker, suggesting a pivotal role of lipid rafts in TGF-ß1-induced differentiation of hASCs to smooth muscle cells. Sucrose density gradient centrifugation along with a shotgun proteomic strategy using liquid chromatography-tandem mass spectrometry identified 1002 individual proteins as the lipid raft proteome, and 242 of these were induced by TGF-ß1 treatment. ADAM12, a disintegrin and metalloproteases family member, was identified as the most highly up-regulated protein in response to TGF-ß1 treatment. TGF-ß1 treatment of hASCs stimulated the production of both ADAM12 protein and mRNA. Silencing of endogenous ADAM12 expression using lentiviral small hairpin RNA or small interfering RNA abrogated the TGF-ß1-induced differentiation of hASCs into smooth muscle cells. CONCLUSIONS: These results suggest a pivotal role for lipid raft-associated ADAM12 in the TGF-ß1-induced differentiation of hASCs into smooth muscle cells.


Subject(s)
ADAM Proteins/metabolism , Cell Differentiation/drug effects , Membrane Proteins/metabolism , Mesenchymal Stem Cells/cytology , Myocytes, Smooth Muscle/cytology , Proteomics/methods , Transforming Growth Factor beta1/pharmacology , ADAM12 Protein , Actins/metabolism , Adipose Tissue/cytology , Caveolin 1/metabolism , Cholesterol/deficiency , Cholesterol/metabolism , Humans , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Smad2 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...