51
|
Role of pigment epithelium-derived factor in the involution of hemangioma: Autocrine growth inhibition of hemangioma-derived endothelial cells. Biochem Biophys Res Commun 2014; 454:282-8. [DOI: 10.1016/j.bbrc.2014.10.052] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 10/11/2014] [Indexed: 01/21/2023]
|
52
|
Adipose-derived stromal cells for osteoarticular repair: trophic function versus stem cell activity. Expert Rev Mol Med 2014; 16:e9. [PMID: 24810570 PMCID: PMC4017835 DOI: 10.1017/erm.2014.9] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The identification of multipotent adipose-derived stromal cells (ASC) has raised hope that tissue regeneration approaches established with bone-marrow-derived stromal cells (BMSC) can be reproduced with a cell-type that is far more accessible in large quantities. Recent detailed comparisons, however, revealed subtle functional differences between ASC and BMSC, stressing the concept of a common mesenchymal progenitor existing in a perivascular niche across all tissues. Focussing on bone and cartilage repair, this review summarises recent in vitro and in vivo studies aiming towards tissue regeneration with ASC. Advantages of good accessibility, high yield and superior growth properties are counterbalanced by an inferiority of ASC to form ectopic bone and stimulate long-bone healing along with their less pronounced osteogenic and angiogenic gene expression signature. Hence, particular emphasis is placed on establishing whether stem cell activity of ASC is so far proven and relevant for successful osteochondral regeneration, or whether trophic activity may largely determine therapeutic outcome.
Collapse
|
53
|
Sart S, Tsai AC, Li Y, Ma T. Three-dimensional aggregates of mesenchymal stem cells: cellular mechanisms, biological properties, and applications. TISSUE ENGINEERING PART B-REVIEWS 2013; 20:365-80. [PMID: 24168395 DOI: 10.1089/ten.teb.2013.0537] [Citation(s) in RCA: 285] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mesenchymal stem cells (MSCs) are primary candidates in cell therapy and tissue engineering and are being tested in clinical trials for a wide range of diseases. Originally isolated and expanded as plastic adherent cells, MSCs have intriguing properties of in vitro self-assembly into three-dimensional (3D) aggregates reminiscent of skeletal condensation in vivo. Recent studies have shown that MSC 3D aggregation improved a range of biological properties, including multilineage potential, secretion of therapeutic factors, and resistance against ischemic condition. Hence, the formation of 3D MSC aggregates has been explored as a novel strategy to improve cell delivery, functional activation, and in vivo retention to enhance therapeutic outcomes. This article summarizes recent reports of MSC aggregate self-assembly, characterization of biological properties, and their applications in preclinical models. The cellular and molecular mechanisms underlying MSC aggregate formation and functional activation are discussed, and the areas that warrant further investigation are highlighted. These analyses are combined to provide perspectives for identifying the controlling mechanisms and refining the methods of aggregate fabrication and expansion for clinical applications.
Collapse
Affiliation(s)
- Sébastien Sart
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | | | | | | |
Collapse
|
54
|
Makridakis M, Roubelakis MG, Vlahou A. Stem cells: Insights into the secretome. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2380-4. [DOI: 10.1016/j.bbapap.2013.01.032] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/19/2013] [Accepted: 01/23/2013] [Indexed: 01/06/2023]
|
55
|
Oñate B, Vilahur G, Camino-López S, Díez-Caballero A, Ballesta-López C, Ybarra J, Moscatiello F, Herrero J, Badimon L. Stem cells isolated from adipose tissue of obese patients show changes in their transcriptomic profile that indicate loss in stemcellness and increased commitment to an adipocyte-like phenotype. BMC Genomics 2013; 14:625. [PMID: 24040759 PMCID: PMC3848661 DOI: 10.1186/1471-2164-14-625] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 09/11/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The adipose tissue is an endocrine regulator and a risk factor for atherosclerosis and cardiovascular disease when by excessive accumulation induces obesity. Although the adipose tissue is also a reservoir for stem cells (ASC) their function and "stemcellness" has been questioned. Our aim was to investigate the mechanisms by which obesity affects subcutaneous white adipose tissue (WAT) stem cells. RESULTS Transcriptomics, in silico analysis, real-time polymerase chain reaction (PCR) and western blots were performed on isolated stem cells from subcutaneous abdominal WAT of morbidly obese patients (ASCmo) and of non-obese individuals (ASCn). ASCmo and ASCn gene expression clustered separately from each other. ASCmo showed downregulation of "stemness" genes and upregulation of adipogenic and inflammatory genes with respect to ASCn. Moreover, the application of bioinformatics and Ingenuity Pathway Analysis (IPA) showed that the transcription factor Smad3 was tentatively affected in obese ASCmo. Validation of this target confirmed a significantly reduced Smad3 nuclear translocation in the isolated ASCmo. CONCLUSIONS The transcriptomic profile of the stem cells reservoir in obese subcutaneous WAT is highly modified with significant changes in genes regulating stemcellness, lineage commitment and inflammation. In addition to body mass index, cardiovascular risk factor clustering further affect the ASC transcriptomic profile inducing loss of multipotency and, hence, capacity for tissue repair. In summary, the stem cells in the subcutaneous WAT niche of obese patients are already committed to adipocyte differentiation and show an upregulated inflammatory gene expression associated to their loss of stemcellness.
Collapse
Affiliation(s)
- Blanca Oñate
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Wu L, Leijten J, van Blitterswijk CA, Karperien M. Fibroblast growth factor-1 is a mesenchymal stromal cell-secreted factor stimulating proliferation of osteoarthritic chondrocytes in co-culture. Stem Cells Dev 2013; 22:2356-67. [PMID: 23557133 PMCID: PMC3749707 DOI: 10.1089/scd.2013.0118] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 04/02/2013] [Indexed: 12/26/2022] Open
Abstract
Previously, we showed that mesenchymal stromal cells (MSCs) in co-culture with primary chondrocytes secrete soluble factors that increase chondrocyte proliferation. The objective of this study is to identify these factors. Human primary chondrocytes (hPCs) isolated from late-stage osteoarthritis patients were co-cultured with human bone marrow-derived MSCs (hMSCs) in pellets. Genome-wide mRNA expression analysis and quantitative polymerase chain reactions (qPCR) were used to identify soluble factors that were specifically induced in co-cultures. Immunofluorescent staining combined with cell tracking and enzyme-linked immunosorbent assay (ELISA) were performed to validate up-regulation at the protein level and to identify the cellular origin of the increased proteins. Chemical blockers and neutralizing antibodies were used to elucidate the role of the identified candidate genes in co-cultures. A number of candidate factors were differentially regulated in co-cultures at the mRNA level. Of these, fibroblast growth factor-1 (FGF-1) mRNA and protein expression were markedly increased in co-cultures predominantly due to up-regulated expression in MSCs. Blocking of FGF signaling in co-culture pellets by specific FGF receptor inhibitors or FGF-1 neutralizing antibodies completely blocked hPCs proliferation. We demonstrate that MSCs increase FGF-1 secretion on co-culture with hPCs, which, in turn, is responsible for increased hPCs proliferation in pellet co-cultures.
Collapse
Affiliation(s)
- Ling Wu
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands
| | - Jeroen Leijten
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands
| | - Clemens A. van Blitterswijk
- Department of Tissue Regeneration, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands
| |
Collapse
|
57
|
Kupcova Skalnikova H. Proteomic techniques for characterisation of mesenchymal stem cell secretome. Biochimie 2013; 95:2196-211. [PMID: 23880644 DOI: 10.1016/j.biochi.2013.07.015] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 07/13/2013] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells with a substantial potential in human regenerative medicine due to their ability to migrate to sites of injury, capability to suppress immune response and accessibility in large amount from patient's own bone marrow or fat tissue. It has been increasingly observed that the transplanted MSCs did not necessarily engraft and differentiate at the site of injury but might exert their therapeutic effects through secreted trophic signals. The MSCs secrete a variety of autocrine/paracrine factors, called secretome, that support regenerative processes in the damaged tissue, induce angiogenesis, protect cells from apoptotic cell death and modulate immune system. The cell culture medium conditioned by MSCs or osteogenic, chondrogenic as well as adipogenic precursors derived from MSCs has become a subject of intensive proteomic profiling in the search for and identification of released factors and microvesicles that might be applicable in regenerative medicine. Jointly with the methods for MSC isolation, expansion and differentiation, proteomic analysis of MSC secretome was enabled recently mainly due to the extensive development in protein separation techniques, mass spectrometry, immunological methods and bioinformatics. This review describes proteomic techniques currently applied or prospectively applicable in MSC secretomics, with a particular focus on preparation of the secretome sample, protein/peptide separation, mass spectrometry and protein quantification techniques, analysis of posttranslational modifications, immunological techniques, isolation and characterisation of secreted vesicles and exosomes, analysis of cytokine-encoding mRNAs and bioinformatics.
Collapse
Affiliation(s)
- Helena Kupcova Skalnikova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, v.v.i., Rumburska 89, 277 21 Libechov, Czech Republic.
| |
Collapse
|
58
|
Paul G, Anisimov SV. The secretome of mesenchymal stem cells: potential implications for neuroregeneration. Biochimie 2013; 95:2246-56. [PMID: 23871834 DOI: 10.1016/j.biochi.2013.07.013] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Accepted: 07/10/2013] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells have shown regenerative properties in many tissues. This feature had originally been ascribed to their multipotency and thus their ability to differentiate into tissue-specific cells. However, many researchers consider the secretome of mesenchymal stem cells the most important player in the observed reparative effects of these cells. In this review, we specifically focus on the potential neuroregenerative effect of mesenchymal stem cells, summarize several possible mechanisms of neuroregeneration and list key factors mediating this effect. We illustrate examples of mesenchymal stem cell treatment in central nervous system disorders including stroke, neurodegenerative disorders (such as Parkinson's disease, Huntington's disease, multiple system atrophy and cerebellar ataxia) and inflammatory disease (such as multiple sclerosis). We specifically highlight studies where mesenchymal stem cells have entered clinical trials.
Collapse
Affiliation(s)
- Gesine Paul
- Translational Neurology Group, Division of Neurology, Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Neurology, Scania University Hospital, Lund, Sweden.
| | | |
Collapse
|
59
|
Inal JM, Kosgodage U, Azam S, Stratton D, Antwi-Baffour S, Lange S. Blood/plasma secretome and microvesicles. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2317-25. [PMID: 23590876 DOI: 10.1016/j.bbapap.2013.04.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 04/04/2013] [Accepted: 04/08/2013] [Indexed: 11/26/2022]
Abstract
A major but hitherto overseen component of the blood/plasma secretome is that of extracellular vesicles (EVs) which are shed from all blood cell types. These EVs are made up of microvesicles (MVs) and exosomes. MVs, 100nm-1μm in diameter, are released from the cell surface, and are a rich source of non-conventionally secreted proteins lacking a conventional signal peptide, and thus not secreted by the classical secretory pathways. Exosomes are smaller vesicles (≤100nm) having an endocytic origin and released upon multivesicular body fusion with the plasma membrane. Both vesicle types play major roles in intercellular cross talk and constitute an important component of the secretome especially in the area of biomarkers for cancer. The release of EVs, which are found in all the bodily fluids, is enhanced in cancer and a major focus of cancer proteomics is therefore targeted at EVs. The blood/plasma secretome is also a source of EVs, potentially diagnostic of infectious disease, whether from EVs released from infected cells or from the pathogens themselves. Despite the great excitement in this field, as is stated here and in other parts of this Special issue entitled: An Updated Secretome, much of the EV research, whether proteomic or functional in nature, urgently needs standardisation both in terms of nomenclature and isolation protocols. This article is part of a Special Issue entitled: An Updated Secretome.
Collapse
Affiliation(s)
- Jameel M Inal
- Cellular and Molecular Immunology Research Centre, School of Human Sciences, London Metropolitan University, 166-220 Holloway Road, London, N7 8DB, UK.
| | | | | | | | | | | |
Collapse
|
60
|
TAKATA TAKANOBU, ISHIGAKI YASUHITO, SHIMASAKI TAKEO, TSUCHIDA HIDEYUKI, MOTOO YOSHIHARU, HAYASHI AKIO, TOMOSUGI NAOHISA. Characterization of proteins secreted by pancreatic cancer cells with anticancer drug treatment in vitro. Oncol Rep 2012; 28:1968-76. [PMID: 22961650 PMCID: PMC3583485 DOI: 10.3892/or.2012.2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 07/23/2012] [Indexed: 12/25/2022] Open
Abstract
Pancreatic cancer is one of the most lethal cancers, with an incidence equaling mortality. It is a heterogeneous group of neoplasms in which pancreatic ductal adenocarcinoma is most common. Pancreatic cancer cannot be cured even if detected early. When treatment is initiated, a suitable method of administration of anticancer drugs must be chosen. Anticancer drugs kill tumor cells. However, side effects including initiation are problematic in anticancer drug therapy. Improved methods for the diagnosis of side effects of pancreatic cancer by using sensitive and specific tumor markers are highly desirable. Therefore, efficient strategies for biomarker discovery are urgently needed. Here, we present an approach based on direct experimental access to proteins released by PANC-1 human pancreatic cancer cells in vitro. A two-dimensional (2-D) map and catalog of this subproteome, herein termed the secretome, were established comprising more than 1,000 proteins observed by '2-D difference in-gel electrophoresis analysis using cyanine dye'. We investigated 22 spots that were 1.20-fold upregulated and 31 spots that were 0.66-fold downregulated by gemcitabine chloride treatment. Proteins in these spots were identified by nano-high-performance liquid chromatography electrospray ionization time of flight mass spectrometry/mass spectrometry. Most secretome constituents were nominally cellular proteins. By mass spectrometry screening, 14-3-3 protein sigma (14-3-3 σ), protein S100-A8, protein S100-A9, galectin-7, lactotransferrin (lactoferrin, LF) precursor, serotransferrin (transferrin) precursor, and vitamin D binding protein precursor were identified. Western blotting confirmed the presence of 14-3-3 σ and LF. We found that upregulation of 14-3-3 σ was associated with apoptosis, and downregulation of LF was found to suppress tumorigenesis.
Collapse
Affiliation(s)
- TAKANOBU TAKATA
- Medical Research Institute, Kanazawa Medical University, Uchinada
| | | | - TAKEO SHIMASAKI
- Medical Research Institute, Kanazawa Medical University, Uchinada
- Department of Medical Oncology, Kanazawa Medical University, Takakura, Hachioji 192-8510
| | - HIDEYUKI TSUCHIDA
- Department of Advanced Medicine, Kanazawa Medical University, Takakura, Hachioji 192-8510
| | - YOSHIHARU MOTOO
- Medical Research Institute, Kanazawa Medical University, Uchinada
- Department of Medical Oncology, Kanazawa Medical University, Takakura, Hachioji 192-8510
| | - AKIO HAYASHI
- Agilent Technologies Japan, Ltd., Takakura, Hachioji 192-8510
| | - NAOHISA TOMOSUGI
- Medical Research Institute, Kanazawa Medical University, Uchinada
- Department of Nephrology, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
61
|
Chavan SS, Hudson LK, Li JH, Ochani M, Harris Y, Patel NB, Katz D, Scheinerman JA, Pavlov VA, Tracey KJ. Identification of pigment epithelium-derived factor as an adipocyte-derived inflammatory factor. Mol Med 2012; 18:1161-8. [PMID: 22714715 DOI: 10.2119/molmed.2012.00156] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 06/14/2012] [Indexed: 12/31/2022] Open
Abstract
Obesity is a major risk factor for insulin resistance, type 2 diabetes mellitus and cardiovascular disease. The pathophysiology of obesity is associated with chronic low-grade inflammation. Adipose tissue in obesity is significantly infiltrated by macrophages that secrete cytokines. The mechanisms of interaction between macrophages and adipocytes, leading to macrophage activation and increased cytokine release, remain to be elucidated. We reasoned that an adipocyte-derived factor might stimulate activation of macrophages. We have identified pigment epithelium-derived factor (PEDF) as a mediator of inflammation that is secreted by adipocytes and mediates macrophage activation. Recombinant PEDF activates macrophages to release tumor necrosis factor (TNF) and interleukin-1 (IL-1). The PEDF receptor adipose triglyceride lipase (ATGL) is required for PEDF-mediated macrophage activation. Selective inhibition of ATGL on macrophages attenuates PEDF-induced TNF production, and PEDF enhances the phosphorylation of p38 and extracellular signal-regulated kinase 1/2 mitogen-activated protein kinases. PEDF administration to rats results in increased serum TNF levels, and insulin resistance. Together, these findings suggest that PEDF secreted by adipocytes contributes to the onset and maintenance of chronic inflammation in obesity, and may be a therapeutic target in ameliorating insulin resistance.
Collapse
Affiliation(s)
- Sangeeta S Chavan
- Laboratory of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, New York 11030, United States of America.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
MSC and Tumors: Homing, Differentiation, and Secretion Influence Therapeutic Potential. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2012; 130:209-66. [PMID: 22990585 DOI: 10.1007/10_2012_150] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
: Mesenchymal stromal/stem cells (MSC) are adult multipotent progenitors with fibroblast-like morphology able to differentiate into adipocytic, osteogenic, chondrogenic, and myogenic lineages. Due to these properties, MSC have been studied and introduced as therapeutics in regenerative medicine. Preliminary studies have also shown a possible involvement of MSC as precursors of cellular elements within tumor microenvironments, in particular tumor-associated fibroblasts (TAF). Among a number of different possible origins, TAF may originate from a pool of circulating progenitors from bone marrow or adipose tissue-derived MSC. There is growing evidence to corroborate that cells immunophenotypically defined as MSC are able to reside as TAF influencing the tumor microenvironment in a potentially bi-phasic and obscure manner: either promoting or inhibiting growth depending on tumor context and MSC sources. Here we focus on relationships between the tumor microenvironment, cancer cells, and MSC, analyzing their diverse ability to influence neoplastic development. Associated activities include MSC homing driven by the secretion of various mediators, differentiation towards TAF phenotypes, and reciprocal interactions with the tumor cells. These are reviewed here with the aim of understanding the biological functions of MSC that can be exploited for innovative cancer therapy.
Collapse
|
63
|
Gurkan UA, Golden R, Kishore V, Riley CP, Adamec J, Akkus O. Immune and inflammatory pathways are involved in inherent bone marrow ossification. Clin Orthop Relat Res 2012; 470:2528-40. [PMID: 22798134 PMCID: PMC3830098 DOI: 10.1007/s11999-012-2459-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Bone marrow plays a key role in bone formation and healing. Although a subset of marrow explants ossifies in vitro without excipient osteoinductive factors, some explants do not undergo ossification. The disparity of outcome suggests a significant heterogeneity in marrow tissue in terms of its capacity to undergo osteogenesis. QUESTIONS/PURPOSES We sought to identify: (1) proteins and signaling pathways associated with osteogenesis by contrasting the proteomes of ossified and poorly ossified marrow explants; and (2) temporal changes in proteome and signaling pathways of marrow ossification in the early and late phases of bone formation. METHODS Explants of marrow were cultured. Media conditioned by ossified (n = 4) and poorly ossified (n = 4) subsets were collected and proteins unique to each group were identified by proteomic analysis. Proteomic data were processed to assess proteins specific to the early phase (Days 1-14) and late phase (Days 15-28) of the culture period. Pathways involved in bone marrow ossification were identified through bioinformatics. RESULTS Twenty-eight proteins were unique to ossified samples and eight were unique to poorly ossified ones. Twelve proteins were expressed during the early phase and 15 proteins were specific to the late phase. Several identified pathways corroborated those reported for bone formation in the literature. Immune and inflammatory pathways were specific to ossified samples. CONCLUSIONS The marrow explant model indicates the inflammatory and immune pathways to be an integral part of the osteogenesis process.
Collapse
Affiliation(s)
- Umut Atakan Gurkan
- />Harvard-MIT Division of Health Sciences and Technology, Brigham and Women’s Hospital, Harvard Medical School, 65 Landsdowne Street, PRB 252, Cambridge, MA 02139 USA
| | - Ryan Golden
- />Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN USA
| | - Vipuil Kishore
- />Department of Mechanical and Aerospace Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106 USA
| | - Catherine P. Riley
- />Department of Research and Development Pathology Associates, Medical Laboratories, Spokane, WA 99204 USA
| | - Jiri Adamec
- />Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE USA
| | - Ozan Akkus
- />Department of Mechanical and Aerospace Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106 USA
- />Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106 USA
- />Department of Orthopaedics, University Hospitals of Cleveland, Cleveland, OH USA
| |
Collapse
|
64
|
Rosenow A, Noben JP, Jocken J, Kallendrusch S, Fischer-Posovszky P, Mariman ECM, Renes J. Resveratrol-induced changes of the human adipocyte secretion profile. J Proteome Res 2012; 11:4733-43. [PMID: 22905912 DOI: 10.1021/pr300539b] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Enlarged white adipose tissue (WAT) is a feature of obesity and leads to changes in its paracrine and endocrine function. Dysfunction of WAT cells is associated with obesity-associated disorders like type 2 diabetes and cardiovascular diseases. Resveratrol (RSV), a natural polyphenolic compound, mimics beneficial effects of calorie restriction. As such, RSV seems a promising therapeutic target for obesity-associated disorders. The effect of RSV on the human adipokine profile is still elusive. Therefore, a proteomic study together with bioinformatical analysis was performed to investigate the effect of RSV on the secretion profile of mature human SGBS adipocytes. RSV incubation resulted in elevated basal glycerol release and reduced intracellular TG content. This increased intracellular lipolysis was accompanied by profound changes in the adipocyte secretion profile. Extracellular matrix proteins were down-regulated while processing proteins were mostly up-regulated after RSV treatment. Interestingly, RSV induced secretion of proteins protective against cellular stress and proteins involved in the regulation of apoptosis. Furthermore, we found a RSV-induced up-regulation of adiponectin and ApoE accompanied by a down-regulation of PAI-1 and PEDF secretion which may improve anti-inflammatory processes and increased insulin sensitivity. These effects may contribute to alleviate obesity-induced metabolic complications. In addition, two novel RSV-regulated adipocyte-secreted proteins were identified.
Collapse
Affiliation(s)
- Anja Rosenow
- Department of Human Biology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University, Maastricht, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
65
|
Bragdon B, Bonor J, Shultz KL, Beamer WG, Rosen CJ, Nohe A. Bone morphogenetic protein receptor type Ia localization causes increased BMP2 signaling in mice exhibiting increased peak bone mass phenotype. J Cell Physiol 2012; 227:2870-9. [PMID: 22170575 DOI: 10.1002/jcp.23028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Bone morphogenetic protein 2 (BMP2) is a growth factor that initiates osteoblast differentiation. Recent studies show that BMP2 signaling regulates bone mineral density (BMD). BMP2 interacts with BMP receptor type Ia (BMPRIa) and type II receptor leading to the activation of the Smad signaling pathway. BMPRIa must shuttle between distinct plasma membrane domains, enriched of Caveolin-1 alpha and Caveolin-1 beta isoforms, and receptor activation occurs in these domains. Yet it remains unknown whether the molecular mechanism that regulates BMP2 signaling is driving mineralization and BMD. Therefore, the B6.C3H-1-12 congenic mouse model with increased BMD and osteoblast mineralization was utilized in this study. Using the family image correlation spectroscopy, we determined if BMP2 led to a significant re-localization of BMPRIa to caveolae of the alpha/beta isoforms in bone marrow stromal cells (BMSCs) isolated from B6.C3H-1-12 mice compared to the C57BL/6J mice, which served as controls. The control, C57BL/6J mice, was selected due to only 4 Mb of chromosome 1 from the C3H/HeJ mouse was backcrossed to a C57BL/6J background. Using reporter gene assays, the B6.C3H-1-12 BMSCs responded to BMP2 with increased Smad activation. Furthermore, disrupting caveolae reduced the BMP2-induced Smad signaling in BMSCs isolated from B6.C3H-1-12 and C57BL/6J. This study suggests for the first time a regulatory mechanism of BMPRIa signaling at the plasma membrane of BMSCs that (i) associated with genetic differences in the distal Chromosome 1 segment carried by the B6.C3H-1-12 congenic and (ii) contributes to increase BMD of the B6.C3H-1-12 compared to the C57BL/6J control mice.
Collapse
Affiliation(s)
- Beth Bragdon
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | | | | | | | | | | |
Collapse
|
66
|
Kristensen LP, Chen L, Nielsen MO, Qanie DW, Kratchmarova I, Kassem M, Andersen JS. Temporal profiling and pulsed SILAC labeling identify novel secreted proteins during ex vivo osteoblast differentiation of human stromal stem cells. Mol Cell Proteomics 2012; 11:989-1007. [PMID: 22801418 DOI: 10.1074/mcp.m111.012138] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
It is well established that bone forming cells (osteoblasts) secrete proteins with autocrine, paracrine, and endocrine function. However, the identity and functional role for the majority of these secreted and differentially expressed proteins during the osteoblast (OB) differentiation process, is not fully established. To address these questions, we quantified the temporal dynamics of the human stromal (mesenchymal, skeletal) stem cell (hMSC) secretome during ex vivo OB differentiation using stable isotope labeling by amino acids in cell culture (SILAC). In addition, we employed pulsed SILAC labeling to distinguish genuine secreted proteins from intracellular contaminants. We identified 466 potentially secreted proteins that were quantified at 5 time-points during 14-days ex vivo OB differentiation including 41 proteins known to be involved in OB functions. Among these, 315 proteins exhibited more than 2-fold up or down-regulation. The pulsed SILAC method revealed a strong correlation between the fraction of isotope labeling and the subset of proteins known to be secreted and involved in OB differentiation. We verified SILAC data using qRT-PCR analysis of 9 identified potential novel regulators of OB differentiation. Furthermore, we studied the biological effects of one of these proteins, the hormone stanniocalcin 2 (STC2) and demonstrated its autocrine effects in enhancing osteoblastic differentiation of hMSC. In conclusion, combining complete and pulsed SILAC labeling facilitated the identification of novel factors produced by hMSC with potential role in OB differentiation. Our study demonstrates that the secretome of osteoblastic cells is more complex than previously reported and supports the emerging evidence that osteoblastic cells secrete proteins with endocrine functions and regulate cellular processes beyond bone formation.
Collapse
Affiliation(s)
- Lars P Kristensen
- Center for Experimental Bioinformatics, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense
| | | | | | | | | | | | | |
Collapse
|
67
|
Faça VM. Human mesenchymal stromal cell proteomics: contribution for identification of new markers and targets for medicine intervention. Expert Rev Proteomics 2012; 9:217-30. [PMID: 22462791 DOI: 10.1586/epr.12.9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem or stromal cells (MSCs) have become of great interest for cell-based therapy owing to their roles in tissue repair and immune suppression. MSCs have the ability to differentiate into specialized tissues, including bone, cartilage and muscle, among several others. Furthermore, it has been found that MSCs can also serve as cellular factories that secrete mediators to stimulate in situ regeneration of injured tissues. Proteomics has contributed significantly to the identification of new proteins to improve cellular characterization of MSCs, to identify new targets for therapeutic intervention and to elucidate important pathways utilized by MSCs to differentiate into distinct tissues. As proteomics technology advances, several studies can be revisited and analyzed in depth, employing state-of-the-art approaches, helping to uncover the cellular mechanisms utilized by MSCs to exert their regenerative functionalities. In this article, we will review the progress made so far and discuss further opportunities for proteomics to contribute to the clinical applications of MSCs.
Collapse
Affiliation(s)
- Vitor Marcel Faça
- Department of Biochemistry & Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Brazil.
| |
Collapse
|
68
|
Tracing putative trafficking of the glycolytic enzyme enolase via SNARE-driven unconventional secretion. EUKARYOTIC CELL 2012; 11:1075-82. [PMID: 22753847 DOI: 10.1128/ec.00075-12] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Glycolytic enzymes are cytosolic proteins, but they also play important extracellular roles in cell-cell communication and infection. We used Saccharomyces cerevisiae to analyze the secretory pathway of some of these enzymes, including enolase, phosphoglucose isomerase, triose phosphate isomerase, and fructose 1,6-bisphosphate aldolase. Enolase, phosphoglucose isomerase, and an N-terminal 28-amino-acid-long fragment of enolase were secreted in a sec23-independent manner. The enhanced green fluorescent protein (EGFP)-conjugated enolase fragment formed cellular foci, some of which were found at the cell periphery. Therefore, we speculated that an overview of the secretory pathway could be gained by investigating the colocalization of the enolase fragment with intracellular proteins. The DsRed-conjugated enolase fragment colocalized with membrane proteins at the cis-Golgi complex, nucleus, endosome, and plasma membrane, but not the mitochondria. In addition, the secretion of full-length enolase was inhibited in a knockout mutant of the intracellular SNARE protein-coding gene TLG2. Our results suggest that enolase is secreted via a SNARE-dependent secretory pathway in S. cerevisiae.
Collapse
|
69
|
Pardo M, Roca-Rivada A, Seoane LM, Casanueva FF. Obesidomics: contribution of adipose tissue secretome analysis to obesity research. Endocrine 2012; 41:374-83. [PMID: 22434412 DOI: 10.1007/s12020-012-9617-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 01/23/2012] [Indexed: 10/28/2022]
Abstract
Obesity is presently reaching pandemic proportions and it is becoming a major health concern in developed and developing countries due to its comorbidities like type II diabetes, cardiovascular pathologies, and some cancers. The discovery of the adipose tissue role as an endocrine gland able to secrete adipokines that affects whole-body energy homeostasis has become a key break-through toward a better molecular understanding of obesity. Among the known adipokines involved in the regulation of energy metabolism very few have been clearly seen as central regulators of insulin sensitivity, metabolism, and energy homeostasis. Thus, the discovery and characterization of new adipocyte-derived factors is still in progress. Proteomics technology has emerged as a useful tool to analyze adipose tissue secretion (secretome) dynamics giving a wider picture into the molecular events that control body weight. Besides the identification of new secreted proteins, the advantage of using this approach is the possibility to detect post-translational modifications and protein interactions that generally cannot be predicted by genome studies. In this review, we summarize the recent efforts to identify new bioactive adipokines by proteomics especially in pathological situations such as obesity.
Collapse
Affiliation(s)
- Maria Pardo
- Grupo Obesidómica, Laboratorio de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Spain.
| | | | | | | |
Collapse
|
70
|
Botta A, Delteil F, Mettouchi A, Vieira A, Estrach S, Négroni L, Stefani C, Lemichez E, Meneguzzi G, Gagnoux-Palacios L. Confluence switch signaling regulates ECM composition and the plasmin proteolytic cascade in keratinocytes. J Cell Sci 2012; 125:4241-52. [PMID: 22641690 DOI: 10.1242/jcs.096289] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In culture, cell confluence generates signals that commit actively growing keratinocytes to exit the cell cycle and differentiate to form a stratified epithelium. Using a comparative proteomic approach, we studied this 'confluence switch' and identified a new pathway triggered by cell confluence that regulates basement membrane (BM) protein composition by suppressing the uPA-uPAR-plasmin pathway. Indeed, confluence triggers adherens junction maturation and enhances TGF-β and activin A activity, resulting in increased deposition of PAI-1 and perlecan in the BM. Extracellular matrix (ECM)-accumulated PAI-1 suppresses the uPA-uPAR-plasmin pathway and further enhances perlecan deposition by inhibiting its plasmin-dependent proteolysis. We show that perlecan deposition in the ECM strengthens cell adhesion, inhibits keratinocyte motility and promotes additional accumulation of PAI-1 in the ECM at confluence. In agreement, during wound-healing, perlecan concentrates at the wound-margin, where BM matures to stabilize keratinocyte adhesion. Our results demonstrate that confluence-dependent signaling orchestrates not only growth inhibition and differentiation, but also controls ECM proteolysis and BM formation. These data suggest that uncontrolled integration of confluence-dependent signaling, might favor skin disorders, including tumorigenesis, not only by promoting cell hyperproliferation, but also by altering protease activity and deposition of ECM components.
Collapse
|
71
|
The emerging role of PEDF in stem cell biology. J Biomed Biotechnol 2012; 2012:239091. [PMID: 22675247 PMCID: PMC3362874 DOI: 10.1155/2012/239091] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 03/15/2012] [Indexed: 01/07/2023] Open
Abstract
Encoded by a single gene, PEDF is a 50 kDa glycoprotein that is highly conserved and is widely expressed among many tissues. Most secreted PEDF deposits within the extracellular matrix, with cell-type-specific functions. While traditionally PEDF is known as a strong antiangiogenic factor, more recently, as this paper highlights, PEDF has been linked with stem cell biology, and there is now accumulating evidence demonstrating the effects of PEDF in a variety of stem cells, mainly in supporting stem cell survival and maintaining multipotency.
Collapse
|
72
|
Scheibe F, Klein O, Klose J, Priller J. Mesenchymal stromal cells rescue cortical neurons from apoptotic cell death in an in vitro model of cerebral ischemia. Cell Mol Neurobiol 2012; 32:567-76. [PMID: 22290155 DOI: 10.1007/s10571-012-9798-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 01/06/2012] [Indexed: 01/01/2023]
Abstract
Cell therapy with mesenchymal stromal cells (MSCs) was found to protect neurons from damage after experimental stroke and is currently under investigation in clinical stroke trials. In order to elucidate the mechanisms of MSC-induced neuroprotection, we used the in vitro oxygen–glucose deprivation (OGD) model of cerebral ischemia. Co-culture of primary cortical neurons with MSCs in a transwell co-culture system for 48 h prior to OGD-reduced neuronal cell death by 30-35%. Similar protection from apoptosis was observed with MSC-conditioned media when added 48 h or 30 min prior to OGD, or even after OGD. Western blot analysis revealed increased phosphorylation of STAT3 and Akt in neuronal cultures after treatment with MSC-conditioned media. Inhibition of the PI3K/Akt pathway completely abolished the neuroprotective potential of MSC-conditioned media, suggesting that MSCs can improve neuronal survival by an Akt-dependent anti-apoptotic signaling cascade. Using mass spectrometry, we identified plasminogen activator inhibitor-1 as an active compound in MSC-conditioned media. Thus, paracrine factors secreted by MSCs protect neurons from apoptotic cell death in the OGD model of cerebral ischemia.
Collapse
Affiliation(s)
- Franziska Scheibe
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | |
Collapse
|
73
|
Ravindran S, Gao Q, Ramachandran A, Sundivakkam P, Tiruppathi C, George A. Expression and distribution of grp-78/bip in mineralizing tissues and mesenchymal cells. Histochem Cell Biol 2012; 138:113-25. [PMID: 22527697 DOI: 10.1007/s00418-012-0952-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2012] [Indexed: 11/30/2022]
Abstract
Glucose-regulated protein 78 (GRP-78) is one of the many endoplasmic reticulum chaperone proteins that have been shown to possess multifunctional roles. We have previously demonstrated that GRP-78 functions as a receptor for dentin matrix protein 1 (DMP1) and is required for DMP1-mediated calcium release; that it is a secreted protein and can bind to type I collagen and DMP1 extracellularly and aid in the nucleation of calcium phosphate. We provide evidence in this study that tyrosine phosphorylation is required for DMP1/GRP-78-mediated calcium release in mesenchymal cells. We further demonstrate that GRP-78 is localized in the nucleus of mesenchymal cells and that the cell surface GRP-78 is not associated with the G-protein Gαq in mesenchymal cells. Results from this study show that during development of mineralized tissues, increased expression of GRP-78 can be observed in condensing cartilage and mesenchymal cells of the alveolar bone, endochondral bone and dental pulp. Additionally, we show that GRP-78 is present in the mineralizing matrices of teeth, bone and in the extracellular matrix of differentiating human marrow stromal cells and dental pulp stem cells. Collectively, our observations provide a new perspective on GRP-78 with respect to mineralized matrix formation.
Collapse
Affiliation(s)
- Sriram Ravindran
- Brodie Tooth Development Genetics and Regenerative Medicine Research Laboratory, Department of Oral Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
74
|
Venkataramani V, Thiele K, Behnes CL, Wulf GG, Thelen P, Opitz L, Salinas-Riester G, Wirths O, Bayer TA, Schweyer S. Amyloid Precursor Protein Is a Biomarker for Transformed Human Pluripotent Stem Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1636-52. [DOI: 10.1016/j.ajpath.2011.12.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 10/30/2011] [Accepted: 12/06/2011] [Indexed: 10/14/2022]
|
75
|
Ramachandran A, Ravindran S, George A. Localization of transforming growth factor beta receptor II interacting protein-1 in bone and teeth: implications in matrix mineralization. J Histochem Cytochem 2012; 60:323-37. [PMID: 22260994 DOI: 10.1369/0022155412436879] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Transforming growth factor beta receptor II (TGFβR-II) interacting protein 1 (TRIP-1) is a WD-40 protein that binds to the cytoplasmic domain of the TGF-β type II receptor in a kinase-dependent manner. To investigate the role of TRIP-1 in mineralized tissues, we examined its pattern of expression in cartilage, bone, and teeth and analyzed the relationship between TRIP-1 overexpression and mineralized matrix formation. Results demonstrate that TRIP-1 was predominantly expressed by osteoblasts, odontoblasts, and chondrocytes in these tissues. Interestingly, TRIP-1 was also localized in the extracellular matrix of bone and at the mineralization front in dentin, suggesting that TRIP-1 is secreted by nonclassical secretory mechanisms, as it is devoid of a signal peptide. In vitro nucleation studies demonstrate a role for TRIP-1 in nucleating calcium phosphate polymorphs. Overexpression of TRIP-1 favored osteoblast differentiation of undifferentiated mesenchymal cells with an increase in mineralized matrix formation. These data indicate an unexpected role for TRIP-1 during development of bone, teeth, and cartilage.
Collapse
Affiliation(s)
- Amsaveni Ramachandran
- Brodie Tooth Development Genetics & Regenerative Medicine Research Laboratory, Department of Oral Biology, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | | |
Collapse
|
76
|
Control of Differentiation of Human Mesenchymal Stem Cells by Altering the Geometry of Nanofibers. JOURNAL OF NANOTECHNOLOGY 2012. [DOI: 10.1155/2012/429890] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Effective differentiation of mesenchymal stem cells (MSCs) is required for clinical applications. To control MSC differentiation, induction media containing different types of soluble factors have been used to date; however, it remains challenging to obtain a uniformly differentiated population of an appropriate quality for clinical application by this approach. We attempted to develop nanofiber scaffolds for effective MSC differentiation by mimicking anisotropy of the extracellular matrix structure, to assess whether differentiation of these cells can be controlled by using geometrically different scaffolds. We evaluated MSC differentiation on aligned and random nanofibers, fabricated by electrospinning. We found that induction of MSCs into adipocytes was markedly more inhibited on random nanofibers than on aligned nanofibers. In addition, adipoinduction on aligned nanofibers was also inhibited in the presence of mixed adipoinduction and osteoinduction medium, although osteoinduction was not affected by a change in scaffold geometry. Thus, we have achieved localized control over the direction of differentiation through changes in the alignment of the scaffold even in the presence of a mixed medium. These findings indicate that precise control of MSC differentiation can be attained by using scaffolds with different geometry, rather than by the conventional use of soluble factors in the medium.
Collapse
|
77
|
Bragdon B, D'Angelo A, Gurski L, Bonor J, Schultz KL, Beamer WG, Rosen CJ, Nohe A. Altered plasma membrane dynamics of bone morphogenetic protein receptor type Ia in a low bone mass mouse model. Bone 2012; 50:189-99. [PMID: 22036911 PMCID: PMC3651650 DOI: 10.1016/j.bone.2011.10.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 09/26/2011] [Accepted: 10/10/2011] [Indexed: 01/28/2023]
Abstract
Bone morphogenetic proteins (BMPs) are growth factors that initiate differentiation of bone marrow stromal cells to osteoblasts and adipocytes, yet the mechanism that decides which lineage the cell will follow is unknown. BMP2 is linked to the development of osteoporosis and variants of BMP2 gene have been reported to increase the development of osteoporosis. Intracellular signaling is transduced by BMP receptors (BMPRs) of type I and type II that are serine/threonine kinase receptors. The BMP type I a receptor (BMPRIa) is linked to osteogenesis and bone mineral density (BMD). BMPRs are localized to caveolae enriched with Caveolin1 alpha/beta and Caveolin beta isoforms to facilitate signaling. BMP2 binding to caveolae was recently found to be crucial for the initiation of the Smad signaling pathway. Here we determined the role of BMP receptor localization within caveolae isoforms and aggregation of caveolae as well as BMPRIa in bone marrow stromal cells (BMSCs) on bone mineral density using the B6.C3H-6T as a model system. The B6.C3H-6T is a congenic mouse with decreased bone mineral density (BMD) with increased marrow adipocytes and decreased osteoprogenitor proliferation. C57BL/6J mice served as controls since only a segment of Chr6 from the C3H/HeJ mouse was backcrossed to a C57BL/6J background. Family of image correlation spectroscopy was used to analyze receptor cluster density and co-localization of BMPRIa and caveolae. It was previously shown that BMP2 stimulation results in an aggregation of caveolae and BMPRIa. Additionally, BMSCs isolated from the B6.C3H-6T mice showed a dispersion of caveolae domains compared to C57BL/6J. The aggregation of BMPRIa that is necessary for signaling to occur was inhibited in BMSCs isolated from B6.C3H-6T. Additionally, we analyzed the co-localization of BMPRIa with caveolin-1 isoforms. There was increased percentage of BMPRIa co-localization with caveolae compared to C57BL/6J. BMP2 stimulation had no effect on the colocalization of BMPRIa with caveolin-1. Disrupting caveolae initiated Smad signaling in the isolated BMSCs from B6.C3H-6T. These data suggest that in congenic 6T mice BMP receptors aggregation is inhibited causing an inhibition of signaling and reduced bone mass.
Collapse
Affiliation(s)
- Beth Bragdon
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Liang H, Hou H, Yi W, Yang G, Gu C, Lau WB, Gao E, Ma X, Lu Z, Wei X, Pei J, Yi D. Increased expression of pigment epithelium-derived factor in aged mesenchymal stem cells impairs their therapeutic efficacy for attenuating myocardial infarction injury. Eur Heart J 2011; 34:1681-90. [PMID: 21606086 PMCID: PMC3675387 DOI: 10.1093/eurheartj/ehr131] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
AIMS Mesenchymal stem cells (MSCs) can ameliorate myocardial infarction (MI) injury. However, older-donor MSCs seem less efficacious than those from younger donors, and the contributing underlying mechanisms remain unknown. Here, we determine how age-related expression of pigment epithelium-derived factor (PEDF) affects MSC therapeutic efficacy for MI. METHODS AND RESULTS Reverse transcriptase-polymerized chain reaction and enzyme-linked immunosorbent assay analyses revealed dramatically increased PEDF expression in MSCs from old mice compared to young mice. Morphological and functional experiments demonstrated significantly impaired old MSC therapeutic efficacy compared with young MSCs in treatment of mice subjected to MI. Immunofluorescent staining demonstrated that administration of old MSCs compared with young MSCs resulted in an infarct region containing fewer endothelial cells, vascular smooth muscle cells, and macrophages, but more fibroblasts. Pigment epithelium-derived factor overexpression in young MSCs impaired the beneficial effects against MI injury, and induced cellular profile changes in the infarct region similar to administration of old MSCs. Knocking down PEDF expression in old MSCs improved MSC therapeutic efficacy, and induced a cellular profile similar to young MSCs administration. Studies in vitro showed that PEDF secreted by MSCs regulated the proliferation and migration of cardiac fibroblasts. CONCLUSIONS This is the first evidence that paracrine factor PEDF plays critical role in the regulatory effects of MSCs against MI injury. Furthermore, the impaired therapeutic ability of aged MSCs is predominantly caused by increased PEDF secretion. These findings indicate PEDF as a promising novel genetic modification target for improving aged MSC therapeutic efficacy.
Collapse
Affiliation(s)
- Hongliang Liang
- Department of Cardiovascular Surgery, Institute of Cardiovascular Disease of Chinese PLA, Xijing Hospital, the Fourth Military Medical University, No.127, West Changle Road, Xi'an, Shaanxi Province 710032, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Ravindran S, Gao Q, Ramachandran A, Blond S, Predescu SA, George A. Stress chaperone GRP-78 functions in mineralized matrix formation. J Biol Chem 2011; 286:8729-39. [PMID: 21239500 PMCID: PMC3059005 DOI: 10.1074/jbc.m110.179341] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 12/16/2010] [Indexed: 11/06/2022] Open
Abstract
Mineralized matrix formation is a well orchestrated event requiring several players. Glucose-regulated protein-78 (GRP-78) is an endoplasmic reticulum chaperone protein that has been implicated in functional roles ranging from involvement in cancer biology to serving as a receptor for viruses. In the present study we explored the role of GRP-78 in mineralized matrix formation. Differential expression of GRP-78 mRNA and protein was observed upon in vitro differentiation of primary mouse calvarial cells. An interesting observation was that GRP-78 was identified in the secretome of these cells and in the bone matrix, suggesting an extracellular function during matrix formation. In vitro nucleation experiments under physiological concentrations of calcium and phosphate ions indicated that GRP-78 can induce the formation of calcium phosphate polymorphs by itself, when bound to immobilized type I collagen and on demineralized collagen wafers. We provide evidence that GRP-78 can bind to DMP1 and type I collagen independent of each other in a simulated extracellular environment. Furthermore, we demonstrate the cell surface localization of GRP-78 and provide evidence that it functions as a receptor for DMP1 endocytosis in pre-osteoblasts and primary calvarial cells. Overall, this study represents a paradigm shift in the biological function of GRP-78.
Collapse
Affiliation(s)
| | - Qi Gao
- From the Departments of Oral Biology and
| | | | - Sylvie Blond
- Pharmaceutical Biotechnology, University of Illinois and
| | - Sanda A. Predescu
- the Department of Pharmacology, Rush University Medical Center, Chicago, Illinois 60612
| | | |
Collapse
|
80
|
Maurer MH. Proteomic definitions of mesenchymal stem cells. Stem Cells Int 2011; 2011:704256. [PMID: 21437194 PMCID: PMC3062154 DOI: 10.4061/2011/704256] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 01/17/2011] [Indexed: 01/09/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are pluripotent cells isolated from the bone marrow and various other organs. They are able to proliferate and self-renew, as well as to give rise to progeny of at least the osteogenic, chondrogenic, and adipogenic lineages. Despite this functional definition, MSCs can also be defined by their expression of a distinct set of cell surface markers. In the current paper, studies investigating the proteome of human MSCs are reviewed with the aim to identify common protein markers of MSCs. The proteomic analysis of MSCs revealed a distinct set of proteins representing the basic molecular inventory, including proteins for (i) cell surface markers, (ii) the responsiveness to growth factors, (iii) the reuse of developmental signaling cascades in adult stem cells, (iv) the interaction with molecules of the extracellular matrix, (v) the expression of genes regulating transcription and translation, (vi) the control of the cell number, and (vii) the protection against cellular stress.
Collapse
Affiliation(s)
- Martin H Maurer
- Department of Physiology and Pathophysiology, University of Heidelberg, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| |
Collapse
|
81
|
Skalnikova H, Motlik J, Gadher SJ, Kovarova H. Mapping of the secretome of primary isolates of mammalian cells, stem cells and derived cell lines. Proteomics 2011; 11:691-708. [PMID: 21241017 DOI: 10.1002/pmic.201000402] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 09/14/2010] [Accepted: 09/20/2010] [Indexed: 01/09/2023]
Abstract
Within a mammalian organism, the interaction among cells both at short and long distances is mediated by soluble factors released by cells into the extracellular environment. The secreted proteins may involve extracellular matrix proteins, proteinases, growth factors, protein hormones, immunoregulatory cytokines, chemokines or other bioactive molecules that have a direct impact on target cell phenotype. Stem cells of mesenchymal, adipose, neural and embryonic origin, fibroblast feeder cells as well as primary isolates of astrocytes, endothelial and muscle cells have recently become targets of intensive secretome profiling with the search for proteins regulating cell survival, proliferation, differentiation or inflammatory response. Recent advances and challenges of the stem cell and primary cell secretome analysis together with the most relevant results are discussed in this review.
Collapse
Affiliation(s)
- Helena Skalnikova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Libechov, Czech Republic
| | | | | | | |
Collapse
|
82
|
Costa CASD, Carlos AS, dos Santos ADS, Monteiro AMV, Moura EGD, Nascimento-Saba CCA. Abdominal adiposity, insulin and bone quality in young male rats fed a high-fat diet containing soybean or canola oil. Clinics (Sao Paulo) 2011; 66:1811-6. [PMID: 22012056 PMCID: PMC3180158 DOI: 10.1590/s1807-59322011001000022] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 05/30/2011] [Accepted: 07/05/2011] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVES A low ratio of omega-6/omega-3 polyunsaturated fatty acids is associated with healthy bone properties. However, fatty diets can induce obesity. Our objective was to evaluate intra-abdominal adiposity, insulin, and bone growth in rats fed a high-fat diet containing low ratios of omega-6/omega-3 provided in canola oil. METHODS After weaning, rats were grouped and fed either a control diet (7S), a high-fat diet containing soybean oil (19S) or a high-fat diet of canola oil (19C) until they were 60 days old. Differences were considered to be significant if p<0.05. RESULTS After 60 days, the 19S and 19C groups showed more energy intake, body density growth and intraabdominal fat mass. However, the 19S group had a higher area (200%) and a lower number (44%) of adipocytes, while the 7S and 19C groups did not differ. The serum concentrations of glucose and insulin and the insulin resistance index were significantly increased in the 19C group (15%, 56%, and 78%, respectively) compared to the 7S group. Bone measurements of the 19S and 19C groups showed a higher femur mass (25%) and a higher lumbar vertebrae mass (11%) and length (5%). Computed tomography analysis revealed more radiodensity in the proximal femoral epiphysis and lumbar vertebrae of 19C group compared to the 7S and 19S groups. CONCLUSIONS Our results suggest that the amount and source of fat used in the diet after weaning increase body growth and fat depots and affect insulin resistance and, consequently, bone health.
Collapse
|
83
|
Polacek M, Bruun JA, Elvenes J, Figenschau Y, Martinez I. The secretory profiles of cultured human articular chondrocytes and mesenchymal stem cells: implications for autologous cell transplantation strategies. Cell Transplant 2010; 20:1381-93. [PMID: 21176404 DOI: 10.3727/096368910x550215] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
This study was undertaken to compare the phenotype of human articular chondrocytes (ACs) and bone marrow-derived mesenchymal stem cells (MSCs) after cell expansion by studying the spectrum of proteins secreted by cells into the culture medium. ACs and MSCs were expanded in monolayer cultures for some weeks, as done in standard cell transplantation procedures. Initially, the expression of cartilage signature genes was compared by real-time PCR. Metabolic labeling of proteins (SILAC) in combination with mass spectrometry (LC/MS-MS) was applied to investigate differences in released proteins. In addition, multiplex assays were carried out to quantify the amounts of several matrix metalloproteases (MMPs) and their natural inhibitors (TIMPs). Expanded chondrocytes showed a slightly higher expression of cartilage-specific genes than MSCs, whereas the overall spectra of released proteins were very similar for the two cell types. In qualitative terms MSCs seemed to secrete similar number of extracellular matrix proteins (43% vs. 45% of total proteins found) and catabolic agents (9% vs. 10%), and higher number of anabolic agents (12 % vs. 7%) compared to ACs. Some matrix-regulatory agents such as serpins, BMP-1, and galectins were detected only in MSC supernatants. Quantitative analyses of MMPs and TIMPs revealed significantly higher levels of MMP-1, MMP-2, MMP-3, and MMP-7 in the medium of ACs. Our data show that after the expansion phase, both ACs and MSCs express a dedifferentiated phenotype, resembling each other. ACs hold a phenotype closer to native cartilage at the gene expression level, whereas MSCs show a more anabolic profile by looking at the released proteins pattern. Our data together with the inherent capability of MSCs to maintain their differentiation potential for longer cultivation periods would favor the use of these cells for cartilage reconstruction.
Collapse
Affiliation(s)
- Martin Polacek
- Orthopaedic Surgery Department, University Hospital of North Norway, Tromsø, Norway
| | | | | | | | | |
Collapse
|
84
|
Zaragosi LE, Wdziekonski B, Villageois P, Keophiphath M, Maumus M, Tchkonia T, Bourlier V, Mohsen-Kanson T, Ladoux A, Elabd C, Scheideler M, Trajanoski Z, Takashima Y, Amri EZ, Lacasa D, Sengenes C, Ailhaud G, Clément K, Bouloumie A, Kirkland JL, Dani C. Activin a plays a critical role in proliferation and differentiation of human adipose progenitors. Diabetes 2010; 59:2513-21. [PMID: 20530742 PMCID: PMC3279533 DOI: 10.2337/db10-0013] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Growth of white adipose tissue takes place in normal development and in obesity. A pool of adipose progenitors is responsible for the formation of new adipocytes and for the potential of this tissue to expand in response to chronic energy overload. However, factors controlling self-renewal of human adipose progenitors are largely unknown. We investigated the expression profile and the role of activin A in this process. RESEARCH DESIGN AND METHODS Expression of INHBA/activin A was investigated in three types of human adipose progenitors. We then analyzed at the molecular level the function of activin A during human adipogenesis. We finally investigated the status of activin A in adipose tissues of lean and obese subjects and analyzed macrophage-induced regulation of its expression. RESULTS INHBA/activin A is expressed by adipose progenitors from various fat depots, and its expression dramatically decreases as progenitors differentiate into adipocytes. Activin A regulates the number of undifferentiated progenitors. Sustained activation or inhibition of the activin A pathway impairs or promotes, respectively, adipocyte differentiation via the C/EBPβ-LAP and Smad2 pathway in an autocrine/paracrine manner. Activin A is expressed at higher levels in adipose tissue of obese patients compared with the expression levels in lean subjects. Indeed, activin A levels in adipose progenitors are dramatically increased by factors secreted by macrophages derived from obese adipose tissue. CONCLUSIONS Altogether, our data show that activin A plays a significant role in human adipogenesis. We propose a model in which macrophages that are located in adipose tissue regulate adipose progenitor self-renewal through activin A.
Collapse
Affiliation(s)
- Laure-Emmanuelle Zaragosi
- UMR6543 Centre National de la Recherche Scientifique, Institute of Developmental Biology and Cancer, University of Nice Sophia-Antipolis, Nice, France
| | - Brigitte Wdziekonski
- UMR6543 Centre National de la Recherche Scientifique, Institute of Developmental Biology and Cancer, University of Nice Sophia-Antipolis, Nice, France
| | - Phi Villageois
- UMR6543 Centre National de la Recherche Scientifique, Institute of Developmental Biology and Cancer, University of Nice Sophia-Antipolis, Nice, France
| | - Mayoura Keophiphath
- INSERM U872, Nutriomique Team 7, UMR S 872, Centre de Recherche des Cordeliers, University Pierre et Marie Curie-Paris 6, Paris, France
| | - Marie Maumus
- INSERM U858, Institute of Molecular Medicine Rangueil, University of Toulouse III Paul-Sabatier, Toulouse, France
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Virginie Bourlier
- INSERM U858, Institute of Molecular Medicine Rangueil, University of Toulouse III Paul-Sabatier, Toulouse, France
| | - Tala Mohsen-Kanson
- UMR6543 Centre National de la Recherche Scientifique, Institute of Developmental Biology and Cancer, University of Nice Sophia-Antipolis, Nice, France
| | - Annie Ladoux
- UMR6543 Centre National de la Recherche Scientifique, Institute of Developmental Biology and Cancer, University of Nice Sophia-Antipolis, Nice, France
| | - Christian Elabd
- UMR6543 Centre National de la Recherche Scientifique, Institute of Developmental Biology and Cancer, University of Nice Sophia-Antipolis, Nice, France
| | | | | | | | - Ez-Zoubir Amri
- UMR6543 Centre National de la Recherche Scientifique, Institute of Developmental Biology and Cancer, University of Nice Sophia-Antipolis, Nice, France
| | - Daniele Lacasa
- INSERM U872, Nutriomique Team 7, UMR S 872, Centre de Recherche des Cordeliers, University Pierre et Marie Curie-Paris 6, Paris, France
| | - Coralie Sengenes
- INSERM U858, Institute of Molecular Medicine Rangueil, University of Toulouse III Paul-Sabatier, Toulouse, France
| | - Gérard Ailhaud
- UMR6543 Centre National de la Recherche Scientifique, Institute of Developmental Biology and Cancer, University of Nice Sophia-Antipolis, Nice, France
| | - Karine Clément
- INSERM U872, Nutriomique Team 7, UMR S 872, Centre de Recherche des Cordeliers, University Pierre et Marie Curie-Paris 6, Paris, France
| | - Anne Bouloumie
- INSERM U858, Institute of Molecular Medicine Rangueil, University of Toulouse III Paul-Sabatier, Toulouse, France
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Christian Dani
- UMR6543 Centre National de la Recherche Scientifique, Institute of Developmental Biology and Cancer, University of Nice Sophia-Antipolis, Nice, France
- Corresponding author: Christian Dani,
| |
Collapse
|
85
|
Sabater M, Moreno-Navarrete JM, Ortega FJ, Pardo G, Salvador J, Ricart W, Frühbeck G, Fernández-Real JM. Circulating pigment epithelium-derived factor levels are associated with insulin resistance and decrease after weight loss. J Clin Endocrinol Metab 2010; 95:4720-8. [PMID: 20631025 DOI: 10.1210/jc.2010-0630] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
OBJECTIVE We aimed to study circulating pigment epithelium-derived factor (PEDF) in vivo in association with insulin resistance and in vitro in human adipocytes. METHODS Circulating PEDF (ELISA) and metabolic profile were assessed in 125 Caucasian men. PEDF levels were also assessed in an independent cohort of subjects (n = 33) to study the effects of changing insulin action. PEDF gene expression and secretion were measured during differentiation of human preadipocytes. RESULTS In all subjects, PEDF was positively associated with body mass index (r = 0.326; P < 0.0001), waist-to-hip ratio (r = 0.380; P < 0.0001), HbA(1c), and fasting triglycerides and negatively with insulin sensitivity (r = -0.320; P < 0.0001). PEDF levels were significantly increased in subjects with altered glucose tolerance and type 2 diabetes. Of the inflammatory markers measured, PEDF levels were positively associated with serum soluble TNF-α receptor 1 and IL-10 in obese subjects. Interestingly, weight loss led to significantly decreased PEDF concentration from 34.8 ± 19.3 to 22.5 ± 14.2 μg/ml (P < 0.0001). Multiple linear regression analyses revealed that insulin sensitivity contributed independently to explain 14% of the variance in PEDF levels after controlling for the effects of body mass index, age, and log fasting triglycerides. Differences in PEDF observed after weight loss were related to changes in obesity, insulin resistance, and blood pressure measures. PEDF gene expression and secretion increased during differentiation of human preadipocytes. CONCLUSION Circulating PEDF is associated with insulin sensitivity. The findings show, for the first time in humans, that PEDF concentrations decrease significantly after weight loss in association with blood pressure. PEDF seems to be involved in human adipocyte biology.
Collapse
Affiliation(s)
- Mònica Sabater
- Department of Diabetes, Endocrinology, and Nutrition, Institut d'Investigació Biomèdica de Girona, Instituto de Salud Carlos III, Girona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Song SY, Chung HM, Sung JH. The pivotal role of VEGF in adipose-derived-stem-cell-mediated regeneration. Expert Opin Biol Ther 2010; 10:1529-37. [PMID: 20860536 DOI: 10.1517/14712598.2010.522987] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
IMPORTANCE OF THE FIELD Several lines of evidence suggest that VEGF is a key regulator of the paracrine effects of adipose-derived stem cells (ASCs), but the mechanism of action remains to be identified. AREAS COVERED IN THIS REVIEW This brief review discusses the following research questions: i) Does VEGF increase the proliferation/migration and differentiation of ASCs?; ii) Does VEGF mediate the paracrine effects of ASCs?; and iii) How is VEGF synthesized, and which factors regulate VEGF secretion? WHAT THE READER WILL GAIN External stimuli such as hypoxia may activate receptor tyrosine kinases in the membrane of ASCs, which, in turn, phosphorylate extracellular signal regulated kinase (ERK) and members of the Akt signaling pathway, stabilizing hypoxia inducible factor 1α (HIF-1α) that are primary regulators of VEGF expression. Secreted VEGF directly stimulates ASCs via VEGF receptors in an autocrine manner and regenerates damaged neighboring cells in a paracrine manner. TAKE HOME MESSAGE Most studies of stem cell regeneration have focused on differentiation of ASCs and their building block function; however, the paracrine effects of ASCs should also be the focus of attention.
Collapse
Affiliation(s)
- Seung-Yong Song
- Department of Plastic & Reconstructive Surgery, CHA University, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Korea
| | | | | |
Collapse
|
87
|
Rosenow A, Arrey TN, Bouwman FG, Noben JP, Wabitsch M, Mariman EC, Karas M, Renes J. Identification of Novel Human Adipocyte Secreted Proteins by Using SGBS Cells. J Proteome Res 2010; 9:5389-401. [DOI: 10.1021/pr100621g] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Anja Rosenow
- NUTRIM School for Nutrition, Toxicology and Metabolism, Department of Human Biology, Maastricht University, Maastricht, The Netherlands, Cluster of Excellence “Macromolecular Complexes”, Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium, and Department of Pediatrics, University of Ulm, Ulm, Germany
| | - Tabiwang N. Arrey
- NUTRIM School for Nutrition, Toxicology and Metabolism, Department of Human Biology, Maastricht University, Maastricht, The Netherlands, Cluster of Excellence “Macromolecular Complexes”, Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium, and Department of Pediatrics, University of Ulm, Ulm, Germany
| | - Freek G. Bouwman
- NUTRIM School for Nutrition, Toxicology and Metabolism, Department of Human Biology, Maastricht University, Maastricht, The Netherlands, Cluster of Excellence “Macromolecular Complexes”, Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium, and Department of Pediatrics, University of Ulm, Ulm, Germany
| | - Jean-Paul Noben
- NUTRIM School for Nutrition, Toxicology and Metabolism, Department of Human Biology, Maastricht University, Maastricht, The Netherlands, Cluster of Excellence “Macromolecular Complexes”, Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium, and Department of Pediatrics, University of Ulm, Ulm, Germany
| | - Martin Wabitsch
- NUTRIM School for Nutrition, Toxicology and Metabolism, Department of Human Biology, Maastricht University, Maastricht, The Netherlands, Cluster of Excellence “Macromolecular Complexes”, Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium, and Department of Pediatrics, University of Ulm, Ulm, Germany
| | - Edwin C.M. Mariman
- NUTRIM School for Nutrition, Toxicology and Metabolism, Department of Human Biology, Maastricht University, Maastricht, The Netherlands, Cluster of Excellence “Macromolecular Complexes”, Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium, and Department of Pediatrics, University of Ulm, Ulm, Germany
| | - Michael Karas
- NUTRIM School for Nutrition, Toxicology and Metabolism, Department of Human Biology, Maastricht University, Maastricht, The Netherlands, Cluster of Excellence “Macromolecular Complexes”, Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium, and Department of Pediatrics, University of Ulm, Ulm, Germany
| | - Johan Renes
- NUTRIM School for Nutrition, Toxicology and Metabolism, Department of Human Biology, Maastricht University, Maastricht, The Netherlands, Cluster of Excellence “Macromolecular Complexes”, Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium, and Department of Pediatrics, University of Ulm, Ulm, Germany
| |
Collapse
|
88
|
Edelman LB, Chandrasekaran S, Price ND. Systems biology of embryogenesis. Reprod Fertil Dev 2010; 22:98-105. [PMID: 20003850 DOI: 10.1071/rd09215] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The development of a complete organism from a single cell involves extraordinarily complex orchestration of biological processes that vary intricately across space and time. Systems biology seeks to describe how all elements of a biological system interact in order to understand, model and ultimately predict aspects of emergent biological processes. Embryogenesis represents an extraordinary opportunity (and challenge) for the application of systems biology. Systems approaches have already been used successfully to study various aspects of development, from complex intracellular networks to four-dimensional models of organogenesis. Going forward, great advancements and discoveries can be expected from systems approaches applied to embryogenesis and developmental biology.
Collapse
Affiliation(s)
- Lucas B Edelman
- Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | | | | |
Collapse
|
89
|
Abstract
Ectomesenchymal dental stem cells could be feasible tools for dental tissue engineering. Dental follicle cells are a promising example, since they are capable of differentiation into various dental tissue cells, such as osteoblasts or cementoblasts. However, cellular mechanisms of cell proliferation and differentiation are not understood in detail. Basic knowledge of these molecular processes may shorten the time before ectomesenchymal dental stem cells can be exploited for bone augmentation in regenerative medicine. Recent developments in proteomics and transcriptomics have made information about genome-wide expression profiles accessible, which can aid in clarifying molecular mechanisms of cells. This review describes the transcriptomes and proteomes of dental follicle cells before and after differentiation, and compares them with differentially expressed populations from dental tissue or bone marrow.
Collapse
Affiliation(s)
- C. Morsczeck
- Department of Operative Dentistry and Periodontology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - G. Schmalz
- Department of Operative Dentistry and Periodontology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
90
|
Choi YA, Lim J, Kim KM, Acharya B, Cho JY, Bae YC, Shin HI, Kim SY, Park EK. Secretome Analysis of Human BMSCs and Identification of SMOC1 as an Important ECM Protein in Osteoblast Differentiation. J Proteome Res 2010; 9:2946-56. [DOI: 10.1021/pr901110q] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Young-Ae Choi
- Department of Oral Pathology, School of Dentistry, BK21, IHBR, Kyungpook National University, Daegu, Korea, Department of Oral Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea, Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University Hospital, Daegu, Korea, and Department of Orthopaedic Surgery, Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu, Korea
| | - Jiwon Lim
- Department of Oral Pathology, School of Dentistry, BK21, IHBR, Kyungpook National University, Daegu, Korea, Department of Oral Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea, Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University Hospital, Daegu, Korea, and Department of Orthopaedic Surgery, Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu, Korea
| | - Kyung Min Kim
- Department of Oral Pathology, School of Dentistry, BK21, IHBR, Kyungpook National University, Daegu, Korea, Department of Oral Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea, Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University Hospital, Daegu, Korea, and Department of Orthopaedic Surgery, Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu, Korea
| | - Bodhraj Acharya
- Department of Oral Pathology, School of Dentistry, BK21, IHBR, Kyungpook National University, Daegu, Korea, Department of Oral Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea, Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University Hospital, Daegu, Korea, and Department of Orthopaedic Surgery, Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu, Korea
| | - Je-Yoel Cho
- Department of Oral Pathology, School of Dentistry, BK21, IHBR, Kyungpook National University, Daegu, Korea, Department of Oral Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea, Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University Hospital, Daegu, Korea, and Department of Orthopaedic Surgery, Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu, Korea
| | - Yong-Chul Bae
- Department of Oral Pathology, School of Dentistry, BK21, IHBR, Kyungpook National University, Daegu, Korea, Department of Oral Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea, Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University Hospital, Daegu, Korea, and Department of Orthopaedic Surgery, Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu, Korea
| | - Hong-In Shin
- Department of Oral Pathology, School of Dentistry, BK21, IHBR, Kyungpook National University, Daegu, Korea, Department of Oral Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea, Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University Hospital, Daegu, Korea, and Department of Orthopaedic Surgery, Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu, Korea
| | - Shin-Yoon Kim
- Department of Oral Pathology, School of Dentistry, BK21, IHBR, Kyungpook National University, Daegu, Korea, Department of Oral Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea, Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University Hospital, Daegu, Korea, and Department of Orthopaedic Surgery, Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu, Korea
| | - Eui Kyun Park
- Department of Oral Pathology, School of Dentistry, BK21, IHBR, Kyungpook National University, Daegu, Korea, Department of Oral Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea, Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University Hospital, Daegu, Korea, and Department of Orthopaedic Surgery, Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu, Korea
| |
Collapse
|
91
|
Germano G, Frapolli R, Simone M, Tavecchio M, Erba E, Pesce S, Pasqualini F, Grosso F, Sanfilippo R, Casali PG, Gronchi A, Virdis E, Tarantino E, Pilotti S, Greco A, Nebuloni M, Galmarini CM, Tercero JC, Mantovani A, D'Incalci M, Allavena P. Antitumor and anti-inflammatory effects of trabectedin on human myxoid liposarcoma cells. Cancer Res 2010; 70:2235-44. [PMID: 20215499 DOI: 10.1158/0008-5472.can-09-2335] [Citation(s) in RCA: 222] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Inflammatory mediators present in the tumor milieu may promote cancer progression and are considered promising targets of novel biological therapies. We previously reported that the marine antitumor agent trabectedin, approved in Europe in 2007 for soft tissue sarcomas and in 2009 for ovarian cancer, was able to downmodulate the production of selected cytokines/chemokines in immune cells. Patients with myxoid liposarcoma (MLS), a subtype characterized by the expression of the oncogenic transcript FUS-CHOP, are highly responsive to trabectedin. The drug had marked antiproliferative effects on MLS cell lines at low nanomolar concentrations. We tested the hypothesis that trabectedin could also affect the inflammatory mediators produced by cancer cells. Here, we show that MLS express several cytokines, chemokines, and growth factors (CCL2, CCL3, CCL5, CXCL8, CXCL12, MIF, VEGF, SPARC) and the inflammatory and matrix-binder protein pentraxin 3 (PTX3), which build up a prominent inflammatory environment. In vitro treatment with noncytotoxic concentrations of trabectedin selectively inhibited the production of CCL2, CXCL8, IL-6, VEGF, and PTX3 by MLS primary tumor cultures and/or cell lines. A xenograft mouse model of human MLS showed marked reduction of CCL2, CXCL8, CD68+ infiltrating macrophages, CD31+ tumor vessels, and partial decrease of PTX3 after trabectedin treatment. Similar findings were observed in a patient tumor sample excised after several cycles of therapy, indicating that the results observed in vitro might have in vivo relevance. In conclusion, trabectedin has dual effects in liposarcoma: in addition to direct growth inhibition, it affects the tumor microenvironment by reducing the production of key inflammatory mediators.
Collapse
Affiliation(s)
- Giovanni Germano
- Department of Immunology and Inflammation, IRCCS Istituto Clinico Humanitas, Rozzano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Wang M, Wang JJ, Li J, Park K, Qian X, Ma JX, Zhang SX. Pigment epithelium-derived factor suppresses adipogenesis via inhibition of the MAPK/ERK pathway in 3T3-L1 preadipocytes. Am J Physiol Endocrinol Metab 2009; 297:E1378-87. [PMID: 19808909 PMCID: PMC2793046 DOI: 10.1152/ajpendo.00252.2009] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We previously reported that circulating levels of pigment epithelium-derived factor (PEDF), a newly identified adipokine, are increased in patients with type 2 diabetes, correlating with body mass index. However, the role of PEDF in adipogenesis remains elusive. In the present study, we have investigated the effects and mechanisms of PEDF on adipocyte differentiation in 3T3-L1 preadipocytes. Differentiation of 3T3-L1 preadipocytes was induced in the presence or absence of human recombinant PEDF protein. The effects of PEDF on adipogenic gene expression, mitotic clonal expansion (MCE), and MAPK activation were investigated. Physiological concentrations of human PEDF protein inhibited adipocyte differentiation, evidenced by decreased lipid accumulation, downregulation of adipocyte markers, and inhibition of master adipogenic transcription factors such as C/EBP-alpha and PPARgamma. The antiadipogenic effects of PEDF were observed only when PEDF was added to the cells on day 0, but not on day 3 during differentiation, suggesting that PEDF targets some early adipogenic events. Similarly, overexpression of PEDF by adenovirus attenuated adipocyte differentiation. Further studies revealed that PEDF, or U-0126, a specific MAPK/ERK inhibitor, sequentially inhibited the early activation of ERK and MCE. Moreover, PEDF attenuated expression and the phosphorylation of C/EBP-beta at Thr(188), an essential step for transcriptional activation of C/EBP-beta. In addition, PEDF expression was decreased significantly in the first 24 h during adipocyte differentiation, suggesting that downregulation of PEDF may be essential for the initiation of MCE and adipogenesis. We conclude that PEDF inhibits adipogenesis in 3T3-L1 preadipocytes partially because of inhibition of the MAPK/ERK signaling pathway and MCE.
Collapse
Affiliation(s)
- Min Wang
- Harold Hamm Oklahoma Diabetes Center and Section of Endocrinology and Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | | | | | | | | | | | | |
Collapse
|
93
|
Copland IB, Lord-Dufour S, Cuerquis J, Coutu DL, Annabi B, Wang E, Galipeau J. Improved autograft survival of mesenchymal stromal cells by plasminogen activator inhibitor 1 inhibition. Stem Cells 2009; 27:467-77. [PMID: 19338064 DOI: 10.1634/stemcells.2008-0520] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mesenchymal stromal cells (MSCs) display robust reparative properties through their ability to limit apoptosis, enhance angiogenesis, and direct positive tissue remodeling. However, low in vivo survival of transplanted cells limits their overall effectiveness and significantly affects their clinical usage. Consequently, identifying strategies to improve cell survival in vivo are a priority. One explanation for their low survival is that MSCs are often transplanted into ischemic tissue, such as infarcted myocardium, where there is poor blood supply and low oxygen tension. Therefore, we examined how MSCs respond to a hypoxic, nutrient-poor stress environment to identify trophic factors that could be manipulated in advance of MSC transplantation. Combining microarray and proteomic screens we identified plasminogen activator inhibitor 1 (PAI-1) as one factor consistently upregulated in our in vitro ischemia-mimicking conditions. Subsequent genetic and chemical manipulation studies define PAI-1 as a negative regulator of MSC survival in vivo. Mechanistically, MSC-derived PAI-1 does not alter MSC survival through a plasmin-dependent mechanism but rather directly impacts on the adhesiveness of MSCs to their surrounding matrices. Thus we can conclude that post-transplantation, PAI-1 negatively impacts MSC survival by promoting anoikis via matrix detachment.
Collapse
Affiliation(s)
- Ian B Copland
- Sir Mortimer B. Davis Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
94
|
Park ES, Cho HS, Kwon TG, Jang SN, Lee SH, An CH, Shin HI, Kim JY, Cho JY. Proteomics Analysis of Human Dentin Reveals Distinct Protein Expression Profiles. J Proteome Res 2009; 8:1338-46. [DOI: 10.1021/pr801065s] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Eun-Sung Park
- Department of Biochemistry & BK 21, Department of Oral & Maxillofacial Surgery, Department of Oral & Maxillofacial Radiology, and Department of Oral Pathology & IHBR, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Hye-Sim Cho
- Department of Biochemistry & BK 21, Department of Oral & Maxillofacial Surgery, Department of Oral & Maxillofacial Radiology, and Department of Oral Pathology & IHBR, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Tae-Geon Kwon
- Department of Biochemistry & BK 21, Department of Oral & Maxillofacial Surgery, Department of Oral & Maxillofacial Radiology, and Department of Oral Pathology & IHBR, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Sin-Nam Jang
- Department of Biochemistry & BK 21, Department of Oral & Maxillofacial Surgery, Department of Oral & Maxillofacial Radiology, and Department of Oral Pathology & IHBR, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Sang-Han Lee
- Department of Biochemistry & BK 21, Department of Oral & Maxillofacial Surgery, Department of Oral & Maxillofacial Radiology, and Department of Oral Pathology & IHBR, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Chang-Hyeon An
- Department of Biochemistry & BK 21, Department of Oral & Maxillofacial Surgery, Department of Oral & Maxillofacial Radiology, and Department of Oral Pathology & IHBR, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Hong-In Shin
- Department of Biochemistry & BK 21, Department of Oral & Maxillofacial Surgery, Department of Oral & Maxillofacial Radiology, and Department of Oral Pathology & IHBR, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Jae-Young Kim
- Department of Biochemistry & BK 21, Department of Oral & Maxillofacial Surgery, Department of Oral & Maxillofacial Radiology, and Department of Oral Pathology & IHBR, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Je-Yoel Cho
- Department of Biochemistry & BK 21, Department of Oral & Maxillofacial Surgery, Department of Oral & Maxillofacial Radiology, and Department of Oral Pathology & IHBR, School of Dentistry, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
95
|
Camafeita E, Lamas JR, Calvo E, López JA, Fernández-Gutiérrez B. Proteomics: New insights into rheumatic diseases. Proteomics Clin Appl 2009; 3:226-241. [DOI: 10.1002/prca.200800146] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
96
|
Bernay B, Gaillard MC, Guryca V, Emadali A, Kuhn L, Bertrand A, Detraz I, Carcenac C, Savasta M, Brouillet E, Garin J, Elalouf JM. Discovering new bioactive neuropeptides in the striatum secretome using in vivo microdialysis and versatile proteomics. Mol Cell Proteomics 2009; 8:946-58. [PMID: 19164277 DOI: 10.1074/mcp.m800501-mcp200] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The striatum, a major component of the brain basal nuclei, is central for planning and executing voluntary movements and undergoes lesions in neurodegenerative disorders such as Huntington disease. To perform highly integrated tasks, the striatum relies on a complex network of communication within and between brain regions with a key role devoted to secreted molecules. To characterize the rat striatum secretome, we combined in vivo microdialysis together with proteomics analysis of trypsin digests and peptidomics studies of native fragments. This versatile approach, carried out using different microdialysis probes and mass spectrometer devices, allowed evidencing with high confidence the expression of 88 proteins and 100 processed peptides. Their secretory pathways were predicted by in silico analysis. Whereas high molecular weight proteins were mainly secreted by the classical mode (94%), low molecular weight proteins equally used classical and non-classical modes (53 and 47%, respectively). In addition, our results suggested alternative secretion mechanisms not predicted by bioinformatics tools. Based on spectrum counting, we performed a relative quantification of secreted proteins and peptides in both basal and neuronal depolarization conditions. This allowed detecting a series of neuropeptide precursors and a 6-fold increase for neurosecretory protein VGF and proenkephalin (PENK) levels. A focused investigation and a long peptide experiment led to the identification of new secreted non-opioid PENK peptides, referred to as PENK 114-133, PENK 239-260, and PENK 143-185. Moreover we showed that injecting synthetic PENK 114-133 and PENK 239-260 into the striatum robustly increased glutamate release in this region. Thus, the combination of microdialysis and versatile proteomics methods shed new light on the secreted protein repertoire and evidenced novel neuropeptide transmitters.
Collapse
Affiliation(s)
- Benoît Bernay
- Laboratoire de PhysioGénomique, Service de Biologie Intégrative et Génétique Moléculaire (SBIGeM), Institut de Biologie et de Technologies de Saclay (iBiTec-S), Commissariat à l'Energie Atomique (CEA), F-91191 Gif-sur-Yvette, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|