1
|
Wei F, Hughes M, Omer M, Ngo C, Pugazhendhi AS, Kolanthai E, Aceto M, Ghattas Y, Razavi M, Kean TJ, Seal S, Coathup M. A Multifunctional Therapeutic Strategy Using P7C3 as A Countermeasure Against Bone Loss and Fragility in An Ovariectomized Rat Model of Postmenopausal Osteoporosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308698. [PMID: 38477537 PMCID: PMC11151083 DOI: 10.1002/advs.202308698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Indexed: 03/14/2024]
Abstract
By 2060, an estimated one in four Americans will be elderly. Consequently, the prevalence of osteoporosis and fragility fractures will also increase. Presently, no available intervention definitively prevents or manages osteoporosis. This study explores whether Pool 7 Compound 3 (P7C3) reduces progressive bone loss and fragility following the onset of ovariectomy (OVX)-induced osteoporosis. Results confirm OVX-induced weakened, osteoporotic bone together with a significant gain in adipogenic body weight. Treatment with P7C3 significantly reduced osteoclastic activity, bone marrow adiposity, whole-body weight gain, and preserved bone area, architecture, and mechanical strength. Analyses reveal significantly upregulated platelet derived growth factor-BB and leukemia inhibitory factor, with downregulation of interleukin-1 R6, and receptor activator of nuclear factor kappa-B (RANK). Together, proteomic data suggest the targeting of several key regulators of inflammation, bone, and adipose turnover, via transforming growth factor-beta/SMAD, and Wingless-related integration site/be-catenin signaling pathways. To the best of the knowledge, this is first evidence of an intervention that drives against bone loss via RANK. Metatranscriptomic analyses of the gut microbiota show P7C3 increased Porphyromonadaceae bacterium, Candidatus Melainabacteria, and Ruminococcaceae bacterium abundance, potentially contributing to the favorable inflammatory, and adipo-osteogenic metabolic regulation observed. The results reveal an undiscovered, and multifunctional therapeutic strategy to prevent the pathological progression of OVX-induced bone loss.
Collapse
Affiliation(s)
- Fei Wei
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
| | - Megan Hughes
- School of BiosciencesCardiff UniversityWalesCF10 3ATUK
| | - Mahmoud Omer
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
| | - Christopher Ngo
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | | | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC)University of Central FloridaOrlandoFL32826USA
| | - Matthew Aceto
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | - Yasmine Ghattas
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | - Mehdi Razavi
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | - Thomas J Kean
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | - Sudipta Seal
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC)University of Central FloridaOrlandoFL32826USA
| | - Melanie Coathup
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| |
Collapse
|
2
|
Le C, Hu X, Tong L, Ye X, Zhang J, Yan J, Sherchan P, Zhang JH, Gao F, Tang J. Inhibition of LAR attenuates neuroinflammation through RhoA/IRS-1/Akt signaling pathway after intracerebral hemorrhage in mice. J Cereb Blood Flow Metab 2023; 43:869-881. [PMID: 36802818 PMCID: PMC10196755 DOI: 10.1177/0271678x231159352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/03/2023] [Accepted: 01/28/2023] [Indexed: 02/23/2023]
Abstract
Leukocyte common antigen-related phosphatase (LAR) is widely expressed in the central nervous system and is known to regulate a variety of processes including cell growth, differentiation, and inflammation. However, little is currently known about LAR signaling mediated neuroinflammation after intracerebral hemorrhage (ICH). The objective of this study was to investigate the role of LAR in ICH using autologous blood injection-induced ICH mouse model. Expression of endogenous proteins, brain edema and neurological function after ICH were evaluated. Extracellular LAR peptide (ELP), an inhibitor of LAR, was administered to ICH mice and outcomes were evaluated. LAR activating-CRISPR or IRS inhibitor NT-157 was administered to elucidate the mechanism. The results showed that expressions of LAR, its endogenous agonist chondroitin sulfate proteoglycans (CSPGs) including neurocan and brevican, and downstream factor RhoA increased after ICH. Administration of ELP reduced brain edema, improved neurological function, and decreased microglia activation after ICH. ELP decreased RhoA and phosphorylated serine-IRS1, increased phosphorylated tyrosine-IRS1 and p-Akt, and attenuated neuroinflammation after ICH, which was reversed by LAR activating-CRISPR or NT-157. In conclusion, this study demonstrated that LAR contributed to neuroinflammation after ICH via RhoA/IRS-1 pathway, and ELP may be a potential therapeutic strategy to attenuate LAR mediated neuroinflammation after ICH.
Collapse
Affiliation(s)
- Chensheng Le
- Department of Neurology, The Second
Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou,
China
- Department of Physiology and
Pharmacology, Loma Linda University, Loma Linda, CA, USA
- Department of Neurology, Ningbo
Medical Center Lihuili Hospital, Ningbo, China
| | - Xin Hu
- Department of Physiology and
Pharmacology, Loma Linda University, Loma Linda, CA, USA
- Department of Neurosurgery, West
China Hospital, Sichuan University, Chengdu, China
| | - Lusha Tong
- Department of Neurology, The Second
Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou,
China
- Department of Physiology and
Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Xianghua Ye
- Department of Neurology, The Second
Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou,
China
| | - Junyi Zhang
- Department of Physiology and
Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Jun Yan
- Department of Physiology and
Pharmacology, Loma Linda University, Loma Linda, CA, USA
- Department of Neurosurgery, Guangxi
Medical University Cancer Hospital, Nanning, China
| | - Prativa Sherchan
- Department of Physiology and
Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology and
Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Feng Gao
- Department of Neurology, The Second
Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou,
China
| | - Jiping Tang
- Department of Physiology and
Pharmacology, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
3
|
Nartey MNN, Jisaka M, Syeda PK, Nishimura K, Shimizu H, Yokota K. Prostaglandin D 2 Added during the Differentiation of 3T3-L1 Cells Suppresses Adipogenesis via Dysfunction of D-Prostanoid Receptor P1 and P2. Life (Basel) 2023; 13:life13020370. [PMID: 36836727 PMCID: PMC9963520 DOI: 10.3390/life13020370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/20/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
We previously reported that the addition of prostaglandin, (PG)D2, and its chemically stable analog, 11-deoxy-11-methylene-PGD2 (11d-11m-PGD2), during the maturation phase of 3T3-L1 cells promotes adipogenesis. In the present study, we aimed to elucidate the effects of the addition of PGD2 or 11d-11m-PGD2 to 3T3-L1 cells during the differentiation phase on adipogenesis. We found that both PGD2 and 11d-11m-PGD2 suppressed adipogenesis through the downregulation of peroxisome proliferator-activated receptor gamma (PPARγ) expression. However, the latter suppressed adipogenesis more potently than PGD2, most likely because of its higher resistance to spontaneous transformation into PGJ2 derivatives. In addition, this anti-adipogenic effect was attenuated by the coexistence of an IP receptor agonist, suggesting that the effect depends on the intensity of the signaling from the IP receptor. The D-prostanoid receptors 1 (DP1) and 2 (DP2, also known as a chemoattractant receptor-homologous molecule expressed on Th2 cells) are receptors for PGD2. The inhibitory effects of PGD2 and 11d-11m-PGD2 on adipogenesis were slightly attenuated by a DP2 agonist. Furthermore, the addition of PGD2 and 11d-11m-PGD2 during the differentiation phase reduced the DP1 and DP2 expression during the maturation phase. Overall, these results indicated that the addition of PGD2 or 11d-11m-PGD2 during the differentiation phase suppresses adipogenesis via the dysfunction of DP1 and DP2. Therefore, unidentified receptor(s) for both molecules may be involved in the suppression of adipogenesis.
Collapse
Affiliation(s)
- Michael N. N. Nartey
- The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
- Council for Scientific and Industrial Research-Animal Research Institute, Achimota, Accra P.O. Box AH20, Ghana
| | - Mitsuo Jisaka
- The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
- Department of Life Science and Biotechnology, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Institute of Agricultural and Life Sciences, Academic Assembly, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Correspondence:
| | - Pinky Karim Syeda
- Department of Life Science and Biotechnology, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
| | - Kohji Nishimura
- The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
- Department of Life Science and Biotechnology, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Institute of Agricultural and Life Sciences, Academic Assembly, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Interdisciplinary Center for Science Research, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
| | - Hidehisa Shimizu
- The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
- Department of Life Science and Biotechnology, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Institute of Agricultural and Life Sciences, Academic Assembly, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Interdisciplinary Center for Science Research, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
| | - Kazushige Yokota
- The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
- Department of Life Science and Biotechnology, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Institute of Agricultural and Life Sciences, Academic Assembly, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
| |
Collapse
|
4
|
Rumman M, Dhawan J. PTPRU, a quiescence-induced receptor tyrosine phosphatase negatively regulates osteogenic differentiation of human mesenchymal stem cells. Biochem Biophys Res Commun 2022; 636:41-49. [DOI: 10.1016/j.bbrc.2022.10.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/18/2022] [Indexed: 11/02/2022]
|
5
|
Monocytic myeloid-derived suppressive cells mitigate over-adipogenesis of bone marrow microenvironment in aplastic anemia by inhibiting CD8 + T cells. Cell Death Dis 2022; 13:620. [PMID: 35851002 PMCID: PMC9293984 DOI: 10.1038/s41419-022-05080-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 01/21/2023]
Abstract
Aplastic anemia (AA) is a blood disorder resulted from over-activated T-cell related hematopoietic failure, with the characterization of hypocellularity and enhanced adipogenic differentiation of mesenchymal stroma cells (MSCs) in bone marrow (BM). However, little is known about the relationship between immune imbalance and polarized adipogenic abnormity of BM microenvironment in this disease entity. In the present study, we differentiated BM-MSCs into osteoblastic or adipogenic lineages to mimic the osteo-adipogenic differentiation. Activated CD8+ T cells and interferon-γ (IFN-γ) were found to stimulate adipogenesis of BM-MSCs either in vitro or in vivo of AA mouse model. Interestingly, myeloid-derived suppressive cells (MDSCs), one of the immune-regulating populations, were decreased within BM of AA mice. We found that it was not CD11b+Ly6G+Ly6C- granulocytic-MDSCs (gMDSCs) but CD11b+Ly6G-Ly6C+ monocytic-MDSCs (mMDSCs) inhibiting both T cell proliferation and IFN-γ production via inducible nitric oxide synthetase (iNOS) pathway. Single-cell RNA-sequencing (scRNA-seq) of AA- and mMDSCs-treated murine BM cells revealed that mMDSCs transfusion could reconstitute BM hematopoietic progenitors by inhibiting T cells population and signature cytokines and decreasing immature Adipo-Cxcl12-abundant reticular cells within BM. Multi-injection of mMDSCs into AA mice reduced intra-BM T cells infiltration and suppressed BM adipogenesis, which subsequently restored the intra-BM immune balance and eventually prevented pancytopenia and hypo-hematopoiesis. In conclusion, adoptive transfusion of mMDSCs might be a novel immune-regulating strategy to treat AA, accounting for not only restoring the intra-BM immune balance but also improving stroma's multi-differentiating microenvironment.
Collapse
|
6
|
Protein tyrosine phosphatases in skeletal development and diseases. Bone Res 2022; 10:10. [PMID: 35091552 PMCID: PMC8799702 DOI: 10.1038/s41413-021-00181-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/29/2021] [Accepted: 09/14/2021] [Indexed: 12/24/2022] Open
Abstract
Skeletal development and homeostasis in mammals are modulated by finely coordinated processes of migration, proliferation, differentiation, and death of skeletogenic cells originating from the mesoderm and neural crest. Numerous molecular mechanisms are involved in these regulatory processes, one of which is protein posttranslational modifications, particularly protein tyrosine phosphorylation (PYP). PYP occurs mainly through the action of protein tyrosine kinases (PTKs), modifying protein enzymatic activity, changing its cellular localization, and aiding in the assembly or disassembly of protein signaling complexes. Under physiological conditions, PYP is balanced by the coordinated action of PTKs and protein tyrosine phosphatases (PTPs). Dysregulation of PYP can cause genetic, metabolic, developmental, and oncogenic skeletal diseases. Although PYP is a reversible biochemical process, in contrast to PTKs, little is known about how this equilibrium is modulated by PTPs in the skeletal system. Whole-genome sequencing has revealed a large and diverse superfamily of PTP genes (over 100 members) in humans, which can be further divided into cysteine (Cys)-, aspartic acid (Asp)-, and histidine (His)-based PTPs. Here, we review current knowledge about the functions and regulatory mechanisms of 28 PTPs involved in skeletal development and diseases; 27 of them belong to class I and II Cys-based PTPs, and the other is an Asp-based PTP. Recent progress in analyzing animal models that harbor various mutations in these PTPs and future research directions are also discussed. Our literature review indicates that PTPs are as crucial as PTKs in supporting skeletal development and homeostasis.
Collapse
|
7
|
Integrated network pharmacology and cellular assay for the investigation of an anti-obesity effect of 6-shogaol. Food Chem 2021; 374:131755. [PMID: 34883426 DOI: 10.1016/j.foodchem.2021.131755] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/11/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022]
Abstract
This study explored the anti-obesity effect of 6-shogaol and the underlying mechanisms by using Network pharmacology for the prediction and verification of molecular targets and pathways of 6-shogaol against obesity. Furthermore, the results were verified by molecular docking and cell experiments. A total of 86 core targets of 6-shogaol towards obesity were identified. Among them, AKT1 and PIK3CA were confirmed by using the molecular docking. In 3T3-L1 preadipocyte model, 6-shogaol significantly inhibited proliferation and differentiation, reducing the accumulation of lipid droplets. Compared with the control group, the inhibition rates of 6-shogaol on TG and TC were 90.8% and 40.0%, respectively. Additionally, 6-shogaol down-regulated the expression of PPAR-γ and C/EBP-α, while it decreased the phosphorylation of IRS-1, PI3K and AKT. This study, for the first time, confirmed the effect of 6-shogaol on improving obesity through PI3K/AKT pathway. An anti-obesity bioactivity study was further recommended for the development of novel anti-obesity products.
Collapse
|
8
|
Comparison of the effects of co-transplantation of bone marrow hematopoietic stem cells and thymic multipotent stromal cells on the immune system of mice depending on methods. EUREKA: LIFE SCIENCES 2021. [DOI: 10.21303/2504-5695.2021.001993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Physical interaction of multipotent stromal cells (MSCs) and hematopoietic stem cells (HSCs) is a modern approach to effective and focused changes in the properties of HSCs. Resulting of those contact interaction is significant activation of cells with following immune system restoration.
The purpose of the study is to investigate the effect of co-transplantation of bone marrow hematopoietic stem cells (HSCs) and thymic multipotent stromal cells (MSCs) separately and as a union of cells on regeneration of the murine immune system, damaged by cyclophosphamide.
MSCs were obtained from thymuses of C57BL mice using explant technique. Bone marrow cells (BMCs) were obtained by flushing out the femur with a nutrient medium. BMCs were cocultivated for 2 hours on the monolayer of thymus-derived MSCs. The immune deficiency of mice was modelled by the treatment with cyclophosphamide (CP). After that, the cells were co-transplanted in two methods (separately into different the retroorbital sinus and as a union after co-cultivation) and the parameters of the immune system were evaluated. It was shown, that separate co-transplantation of BMCs and thymus-derived MSCs is associated with the restoration of the number of bone marrow cells, thymus, spleen and lymph nodes with an increase in the proliferation index of lymph node cells by 1.4 times compared to control. It normalized the previous reduced concentration of hemoglobin and hematocrit in the blood. Co-transplantation had a suppressive effect on the blast transformation reaction, induced by phytohemagglutinin, by 4.3 times, but showed a stimulating effect on DTHR response by 1.6 times compared to control.
Co-transplantation of the union of BMCs and MSCs is associated with the restoration of the number of bone marrow cells, spleen and lymph nodes. The level of spontaneous apoptosis of lymph node cells significantly increased by 3.3 times compared to control. It had not effect on hematological parameters, but is activated to impact the immune system. Thus, as a result of cells union administration showed normalization of the bactericidal activity of peritoneal macrophages, unlike the separate co-transplantation. This cells graft had a suppressive effect on the number of antibody-producing cells in the spleen by 4.2 times compared to control.
Previous co-cultivation and contact interaction of cells change the properties of cell graft. The effect of co-transplantation of BMCs and thymic MSCs is not a simple additive effect of cells. It is acquiring the features typical to certain cell types, and the expression of new characteristics. We assume this phenomenon as a result development of complex cells cooperative processes in vivo and in vitro
Collapse
|
9
|
Park TJ, Park A, Kim J, Kim JY, Han BS, Oh KJ, Lee EW, Lee SC, Bae KH, Kim WK. Myonectin inhibits adipogenesis in 3T3-L1 preadipocytes by regulating p38 MAPK pathway. BMB Rep 2021. [PMID: 33407993 PMCID: PMC7907746 DOI: 10.5483/bmbrep.2021.54.2.262] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In current times, obesity is a major health problem closely associated with metabolic disease such as diabetes, dyslipidemia, and cardiovascular disease. The direct cause of obesity is known as an abnormal increase in fat cell size and the adipocyte pool. Hyperplasia, the increase in number of adipocytes, results from adipogenesis in which preadipocytes differentiate into mature adipocytes. Adipogenesis is regulated by local and systemic cues that alter transduction pathways and subsequent control of adipogenic transcription factors. Therefore, the regulation of adipogenesis is an important target for preventing obesity. Myonectin, a member of the CTRP family, is a type of myokine released by skeletal muscle cells. Although several studies have shown that myonectin is associated with lipid metabolism, the role of myonectin during adipogenesis is not known. Here, we demonstrate the role of myonectin during adipocyte differentiation of 3T3-L1 cells. We found that myonectin inhibits the adipogenesis of 3T3-L1 preadipocytes with a reduction in the expression of adipogenic transcription factors such as C/EBPα, β and PPARγ. Furthermore, we show that myonectin has an inhibitory effect on adipogenesis through the regulation of the p38 MAPK pathway and CHOP. These findings suggest that myonectin may be a novel therapeutic target for the prevention of obesity.
Collapse
Affiliation(s)
- Tae-Jun Park
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Anna Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Jaehoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jeong-Yoon Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Baek Soo Han
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Eun Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| |
Collapse
|
10
|
Stanford SM, Collins M, Diaz MA, Holmes ZJ, Gries P, Bliss MR, Lodi A, Zhang V, Tiziani S, Bottini N. The low molecular weight protein tyrosine phosphatase promotes adipogenesis and subcutaneous adipocyte hypertrophy. J Cell Physiol 2021; 236:6630-6642. [PMID: 33615467 DOI: 10.1002/jcp.30307] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/29/2020] [Accepted: 01/15/2021] [Indexed: 12/28/2022]
Abstract
Obesity is a major contributing factor to the pathogenesis of Type 2 diabetes. Multiple human genetics studies suggest that high activity of the low molecular weight protein tyrosine phosphatase (LMPTP) promotes metabolic syndrome in obesity. We reported that LMPTP is a critical promoter of insulin resistance in obesity by regulating liver insulin receptor signaling and that inhibition of LMPTP reverses obesity-associated diabetes in mice. Since LMPTP is expressed in adipose tissue but little is known about its function, here we examined the role of LMPTP in adipocyte biology. Using conditional knockout mice, we found that selective deletion of LMPTP in adipocytes impaired obesity-induced subcutaneous adipocyte hypertrophy. We assessed the role of LMPTP in adipogenesis in vitro, and found that LMPTP deletion or knockdown substantially impaired differentiation of primary preadipocytes and 3T3-L1 cells into adipocytes, respectively. Inhibition of LMPTP in 3T3-L1 preadipocytes also reduced adipogenesis and expression of proadipogenic transcription factors peroxisome proliferator activated receptor gamma (PPARγ) and CCAAT/enhancer-binding protein alpha. Inhibition of LMPTP increased basal phosphorylation of platelet-derived growth factor receptor alpha (PDGFRα) on activation motif residue Y849 in 3T3-L1, resulting in increased activation of the mitogen-associated protein kinases p38 and c-Jun N-terminal kinase and increased PPARγ phosphorylation on inhibitory residue S82. Analysis of the metabolome of differentiating 3T3-L1 cells suggested that LMPTP inhibition decreased cell glucose utilization while enhancing mitochondrial respiration and nucleotide synthesis. In summary, we report a novel role for LMPTP as a key driver of adipocyte differentiation via control of PDGFRα signaling.
Collapse
Affiliation(s)
- Stephanie M Stanford
- Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Meghan Collins
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, Texas, USA.,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Michael A Diaz
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Zachary J Holmes
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Paul Gries
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, Texas, USA.,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Matthew R Bliss
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Alessia Lodi
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, Texas, USA.,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Vida Zhang
- Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Stefano Tiziani
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, Texas, USA.,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA.,Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Nunzio Bottini
- Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| |
Collapse
|
11
|
Kaartinen MT, Arora M, Heinonen S, Rissanen A, Kaprio J, Pietiläinen KH. Transglutaminases and Obesity in Humans: Association of F13A1 to Adipocyte Hypertrophy and Adipose Tissue Immune Response. Int J Mol Sci 2020; 21:E8289. [PMID: 33167412 PMCID: PMC7663854 DOI: 10.3390/ijms21218289] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 12/11/2022] Open
Abstract
Transglutaminases TG2 and FXIII-A have recently been linked to adipose tissue biology and obesity, however, human studies for TG family members in adipocytes have not been conducted. In this study, we investigated the association of TGM family members to acquired weight gain in a rare set of monozygotic (MZ) twins discordant for body weight, i.e., heavy-lean twin pairs. We report that F13A1 is the only TGM family member showing significantly altered, higher expression in adipose tissue of the heavier twin. Our previous work linked adipocyte F13A1 to increased weight, body fat mass, adipocyte size, and pro-inflammatory pathways. Here, we explored further the link of F13A1 to adipocyte size in the MZ twins via a previously conducted TWA study that was further mined for genes that specifically associate to hypertrophic adipocytes. We report that differential expression of F13A1 (ΔHeavy-Lean) associated with 47 genes which were linked via gene enrichment analysis to immune response, leucocyte and neutrophil activation, as well as cytokine response and signaling. Our work brings further support to the role of F13A1 in the human adipose tissue pathology, suggesting a role in the cascade that links hypertrophic adipocytes with inflammation.
Collapse
Affiliation(s)
- Mari T. Kaartinen
- Faculty of Medicine (Experimental Medicine), McGill University, Montreal, QC H3A 0J7, Canada;
- Faculty of Dentistry (Biomedical Sciences), McGill University, Montreal, QC H3A 0J7, Canada
| | - Mansi Arora
- Faculty of Medicine (Experimental Medicine), McGill University, Montreal, QC H3A 0J7, Canada;
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (S.H.); (A.R.); (K.H.P.)
| | - Aila Rissanen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (S.H.); (A.R.); (K.H.P.)
| | - Jaakko Kaprio
- Department of Public Health, University of Helsinki, 00100 Helsinki, Finland;
| | - Kirsi H. Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (S.H.); (A.R.); (K.H.P.)
- Abdominal Center, Obesity Center, Endocrinology, University of Helsinki and Helsinki University Central Hospital, 00014 Helsinki, Finland
| |
Collapse
|
12
|
Brave H, MacLoughlin R. State of the Art Review of Cell Therapy in the Treatment of Lung Disease, and the Potential for Aerosol Delivery. Int J Mol Sci 2020; 21:E6435. [PMID: 32899381 PMCID: PMC7503246 DOI: 10.3390/ijms21176435] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023] Open
Abstract
Respiratory and pulmonary diseases are among the leading causes of death globally. Despite tremendous advancements, there are no effective pharmacological therapies capable of curing diseases such as COPD (chronic obstructive pulmonary disease), ARDS (acute respiratory distress syndrome), and COVID-19. Novel and innovative therapies such as advanced therapy medicinal products (ATMPs) are still in early development. However, they have exhibited significant potential preclinically and clinically. There are several longitudinal studies published, primarily focusing on the use of cell therapies for respiratory diseases due to their anti-inflammatory and reparative properties, thereby hinting that they have the capability of reducing mortality and improving the quality of life for patients. The primary objective of this paper is to set out a state of the art review on the use of aerosolized MSCs and their potential to treat these incurable diseases. This review will examine selected respiratory and pulmonary diseases, present an overview of the therapeutic potential of cell therapy and finally provide insight into potential routes of administration, with a focus on aerosol-mediated ATMP delivery.
Collapse
Affiliation(s)
- Hosanna Brave
- College of Medicine, Nursing & Health Sciences, National University of Ireland, H91 TK33 Galway, Ireland;
| | - Ronan MacLoughlin
- Department of Chemistry, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
- Aerogen Ltd. Galway Business Park, H91 HE94 Galway, Ireland
| |
Collapse
|
13
|
Parrillo L, Spinelli R, Longo M, Desiderio A, Mirra P, Nigro C, Fiory F, Hedjazifar S, Mutarelli M, Carissimo A, Formisano P, Miele C, Smith U, Raciti GA, Beguinot F. Altered PTPRD DNA methylation associates with restricted adipogenesis in healthy first-degree relatives of Type 2 diabetes subjects. Epigenomics 2020; 12:873-888. [PMID: 32483983 DOI: 10.2217/epi-2019-0267] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: First-degree relatives (FDR) of individuals with Type 2 diabetes (T2D) feature restricted adipogenesis, which render them more vulnerable to T2D. Epigenetics may contribute to these abnormalities. Methods: FDR pre-adipocyte Methylome and Transcriptome were investigated by MeDIP- and RNA-Seq, respectively. Results: Methylome analysis revealed 2841 differentially methylated regions (DMR) in FDR. Most DMR localized into gene-body and were hypomethylated. The strongest hypomethylation signal was identified in an intronic-DMR at the PTPRD gene. PTPRD hypomethylation in FDR was confirmed by bisulphite sequencing and was responsible for its upregulation. Interestingly, Ptprd-overexpression in 3T3-L1 pre-adipocytes inhibited adipogenesis. Notably, the validated PTPRD-associated DMR was significantly hypomethylated in peripheral blood leukocytes from the same FDR individuals. Finally, PTPRD methylation pattern was also replicated in obese individuals. Conclusion: Our findings indicated a previously unrecognized role of PTPRD in restraining adipogenesis. This abnormality may contribute to increase FDR proclivity toward T2D.
Collapse
Affiliation(s)
- Luca Parrillo
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Rosa Spinelli
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Michele Longo
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Antonella Desiderio
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Paola Mirra
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Cecilia Nigro
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Francesca Fiory
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Shahram Hedjazifar
- Lundberg Laboratory for Diabetes Research, Department of Molecular & Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 41345, Sweden
| | | | | | - Pietro Formisano
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Claudia Miele
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Ulf Smith
- Lundberg Laboratory for Diabetes Research, Department of Molecular & Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 41345, Sweden
| | - Gregory Alexander Raciti
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| | - Francesco Beguinot
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine - Federico II University of Naples, 80131, Italy
| |
Collapse
|
14
|
PTPN21 Overexpression Promotes Osteogenic and Adipogenic Differentiation of Bone Marrow-Derived Mesenchymal Stem Cells but Inhibits the Immunosuppressive Function. Stem Cells Int 2019; 2019:4686132. [PMID: 31885609 PMCID: PMC6907062 DOI: 10.1155/2019/4686132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 08/05/2019] [Accepted: 10/04/2019] [Indexed: 12/18/2022] Open
Abstract
Protein tyrosine phosphatases (PTPs) act as key regulators in various cellular processes such as proliferation, differentiation, and migration. Our previous research demonstrated that non-receptor-typed PTP21 (PTPN21), a member of the PTP family, played a critical role in the proliferation, cell cycle, and chemosensitivity of acute lymphoblastic leukemia cells. However, the role of PTPN21 in the bone marrow microenvironment has not yet been elucidated. In the study, we explored the effects of PTPN21 on human bone marrow-derived mesenchymal stem cells (BM-MSCs) via lentiviral-mediated overexpression and knock-down of PTPN21 in vitro. Overexpressing PTPN21 in BM-MSCs inhibited the proliferation through arresting cell cycle at the G0 phase but rendered them a higher osteogenic and adipogenic differentiation potential. In addition, overexpressing PTPN21 in BM-MSCs increased their senescence levels through upregulation of P21 and P53 and dramatically changed the levels of crosstalk with their typical target cells including immunocytes, tumor cells, and vascular endothelial cells. BM-MSCs overexpressing PTPN21 had an impaired immunosuppressive function and an increased capacity of recruiting tumor cells and vascular endothelial cells in a chemotaxis transwell coculture system. Collectively, our data suggested that PTPN21 acted as a pleiotropic factor in modulating the function of human BM-MSCs.
Collapse
|
15
|
Kazi T, Niibe I, Nishikawa A, Matsuzaki T. Optimal stimulation toward the dermal papilla lineage can be promoted by combined use of osteogenic and adipogenic inducers. FEBS Open Bio 2019; 10:197-210. [PMID: 31730301 PMCID: PMC6996385 DOI: 10.1002/2211-5463.12763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 04/01/2019] [Accepted: 11/13/2019] [Indexed: 11/22/2022] Open
Abstract
Dermal papilla cells (DPCs) play crucial roles in hair regeneration, but they readily lose their hair‐forming ability during in vitro culture. Although the formation of spheroids partially restores the ability, shrinkage of the spheroids makes it difficult to maintain cellular viability. To address this problem, we stimulated DPCs with factors known to induce adipogenic and/or osteogenic differentiation, because DPCs share unique gene expression profiles with adipocytes and osteocytes. We isolated DPCs from versican (vcan)–GFP mice, in which GFP is expressed under the control of a vcan promoter, which is strongly active in DPCs of anagen hair follicles. GFP fluorescence was most intense when the spheroids were made from DPCs cultured in a half‐diluted combination of adipogenic and osteogenic media (CAO1/2), a Dulbecco’s modified Eagle’s medium‐based medium that contains 10% FBS, 275 nm dexamethasone, 2.5 mm β‐glycerol phosphate, 12.5 µg·mL−1 ascorbic acid, 0.125 µm isobutylmethylxanthine and 2.5 ng·mL−1 insulin. The dose of each additive used was less than the optimal dose for adipogenic or osteogenic differentiation, and shrinkage of the spheroids was avoided through the addition of fibroblast growth factor 2 and platelet‐derived growth factor‐AA to CAO1/2. In addition, the gene and protein expression of vcan, osteopontin, alkaline phosphatase and α‐smooth muscle actin in the spheroids were augmented to levels similar to those of the intact dermal papillae, which exhibited restored hair‐forming activity. In conclusion, a combination of certain adipogenic and osteogenic inducers, together with fibroblast growth factor 2 and platelet‐derived growth factor‐AA, can promote differentiation toward the DPC lineage.
Collapse
Affiliation(s)
- Taheruzzaman Kazi
- Bioscience and Biotechnology, The United Graduate School of Agricultural Sciences, Tottori University, Japan
| | - Ichitaro Niibe
- Department of Biological Science, Faculty of Life and Environment Science, Shimane University, Japan
| | - Akio Nishikawa
- Bioscience and Biotechnology, The United Graduate School of Agricultural Sciences, Tottori University, Japan.,Department of Biological Science, Faculty of Life and Environment Science, Shimane University, Japan
| | - Takashi Matsuzaki
- Bioscience and Biotechnology, The United Graduate School of Agricultural Sciences, Tottori University, Japan.,Department of Biological Science, Faculty of Life and Environment Science, Shimane University, Japan
| |
Collapse
|
16
|
Ramazzotti G, Ratti S, Fiume R, Follo MY, Billi AM, Rusciano I, Owusu Obeng E, Manzoli L, Cocco L, Faenza I. Phosphoinositide 3 Kinase Signaling in Human Stem Cells from Reprogramming to Differentiation: A Tale in Cytoplasmic and Nuclear Compartments. Int J Mol Sci 2019; 20:ijms20082026. [PMID: 31022972 PMCID: PMC6514809 DOI: 10.3390/ijms20082026] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/19/2019] [Accepted: 04/21/2019] [Indexed: 12/11/2022] Open
Abstract
Stem cells are undifferentiated cells that can give rise to several different cell types and can self-renew. Given their ability to differentiate into different lineages, stem cells retain huge therapeutic potential for regenerative medicine. Therefore, the understanding of the signaling pathways involved in stem cell pluripotency maintenance and differentiation has a paramount importance in order to understand these biological processes and to develop therapeutic strategies. In this review, we focus on phosphoinositide 3 kinase (PI3K) since its signaling pathway regulates many cellular processes, such as cell growth, proliferation, survival, and cellular transformation. Precisely, in human stem cells, the PI3K cascade is involved in different processes from pluripotency and induced pluripotent stem cell (iPSC) reprogramming to mesenchymal and oral mesenchymal differentiation, through different and interconnected mechanisms.
Collapse
Affiliation(s)
- Giulia Ramazzotti
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Stefano Ratti
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Roberta Fiume
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Matilde Yung Follo
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Anna Maria Billi
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Isabella Rusciano
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Eric Owusu Obeng
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Lucia Manzoli
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Lucio Cocco
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Irene Faenza
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| |
Collapse
|
17
|
Lu HY, Zeng H, Zhang L, Porres JM, Cheng WH. Fecal fermentation products of common bean-derived fiber inhibit C/EBPα and PPARγ expression and lipid accumulation but stimulate PPARδ and UCP2 expression in the adipogenesis of 3T3-L1 cells. J Nutr Biochem 2018; 60:9-15. [DOI: 10.1016/j.jnutbio.2018.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/22/2018] [Accepted: 06/06/2018] [Indexed: 12/19/2022]
|
18
|
Smith SR, Schaaf K, Rajabalee N, Wagner F, Duverger A, Kutsch O, Sun J. The phosphatase PPM1A controls monocyte-to-macrophage differentiation. Sci Rep 2018; 8:902. [PMID: 29343725 PMCID: PMC5772551 DOI: 10.1038/s41598-017-18832-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/18/2017] [Indexed: 02/06/2023] Open
Abstract
Differentiation of circulating monocytes into tissue-bound or tissue-resident macrophages is a critical regulatory process affecting host defense and inflammation. However, the regulatory signaling pathways that control the differentiation of monocytes into specific and distinct functional macrophage subsets are poorly understood. Herein, we demonstrate that monocyte-to-macrophage differentiation is controlled by the Protein Phosphatase, Mg2+/Mn2+-dependent 1A (PPM1A). Genetic manipulation experiments demonstrated that overexpression of PPM1A attenuated the macrophage differentiation program, while knockdown of PPM1A expression accelerated the ability of monocytes to differentiate into macrophages. We identify imiquimod and Pam3CSK4 as two Toll-like receptor agonists that induce PPM1A expression, and show that increased expression of PPM1A at the onset of differentiation impairs cellular adherence, reduces expression of inflammatory (M1) macrophage-specific markers, and inhibits the production of inflammatory cytokines. Our findings reveal PPM1A as a negative threshold regulator of M1-type monocyte-to-macrophage differentiation, establishing it as a key phosphatase that orchestrates this program.
Collapse
Affiliation(s)
- Samuel R Smith
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kaitlyn Schaaf
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nusrah Rajabalee
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Frederic Wagner
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alexandra Duverger
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Olaf Kutsch
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jim Sun
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
19
|
Recent advances in understanding the role of protein-tyrosine phosphatases in development and disease. Dev Biol 2017; 428:283-292. [PMID: 28728679 DOI: 10.1016/j.ydbio.2017.03.023] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 01/15/2023]
Abstract
Protein-tyrosine phosphatases (PTPs) remove phosphate groups from tyrosine residues, and thereby propagate or inhibit signal transduction, and hence influence cellular processes such as cell proliferation and differentiation. The importance of tightly controlled PTP activity is reflected by the numerous mechanisms employed by the cell to control PTP activity, including a variety of post-translational modifications, and restricted subcellular localization. This review highlights the strides made in the last decade and discusses the important role of PTPs in key aspects of embryonic development: the regulation of stem cell self-renewal and differentiation, gastrulation and somitogenesis during early embryonic development, osteogenesis, and angiogenesis. The tentative importance of PTPs in these processes is highlighted by the diseases that present upon aberrant activity.
Collapse
|
20
|
Sri Devi S, Ashokkumar N. Citral, a Monoterpene Inhibits Adipogenesis Through Modulation of Adipogenic Transcription Factors in 3T3-L1 Cells. Indian J Clin Biochem 2017; 33:414-421. [PMID: 30319187 DOI: 10.1007/s12291-017-0692-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/31/2017] [Indexed: 12/26/2022]
Abstract
Obesity is considered as a major global human health problem which significantly increases the risk for development of type 2 diabetes. Citral, a bioactive compound widely found in a variety of foods that are consumed daily. In this study, we investigated the inhibitory effect of citral against adipogenic genes in 3T3-L1 cells. The mouse fibroblast 3T3-L1 pre-adipocytes were differentiated into adipocytes using adipogenic cocktail (5 g/ml insulin, 0.5 mM isobutylmethylxanthine and 10 M dexamethasone). Differentiation of adipocytes was evaluated by assessing triglyceride accumulation assay and cell viability by MTT assay. The PI3K/AKT signaling, adipogenic specific transcription factors (PPARγ, SREBP-1c, FAS and CPD) and inflammatory genes (TNF-α, IL-6 and MCP-1) were analyzed by western blotting and reverse transcriptase PCR in differentiated 3T3-L1 cell lines. In this study, triglyceride accumulation was increased in adipogenic cocktail induced 3T3-L1 cells, whereas treatment of citral significantly decreased levels of triglyceride accumulation in concentration dependent manner. Further, MTT assay shows that there was no reduction of cell viability during the differentiation of 3T3-L1 cells. The differentiated 3T3-L1 cell significantly increases the expression of PI3K/AKT, adipogenic transcription factors (PPARγ, SREBP-1c, FAS and CPD) and inflammatory biomarkers (TNF-α, IL-6 and MCP-1). Conversely, cells were treated with citral significantly suppress the expression of PI3K/AKT, PPARγ, SREBP-1c, FAS, CPD, TNF-α, IL-6 and MCP-1 in dose dependent manner. Thus, citral exhibits beneficial effects to inhibit adipogenesis in 3T3-L1 adipocytes through the modulation of adipogenic transcription factors and inflammatory markers.
Collapse
Affiliation(s)
- Subramaniam Sri Devi
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalai Nagar, Chidambaram, Tamil Nadu 608 002 India
| | - Natarajan Ashokkumar
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalai Nagar, Chidambaram, Tamil Nadu 608 002 India
| |
Collapse
|
21
|
Trohatou O, Zagoura D, Orfanos NK, Pappa KI, Marinos E, Anagnou NP, Roubelakis MG. miR-26a Mediates Adipogenesis of Amniotic Fluid Mesenchymal Stem/Stromal Cells via PTEN, Cyclin E1, and CDK6. Stem Cells Dev 2017; 26:482-494. [PMID: 28068868 DOI: 10.1089/scd.2016.0203] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Recent findings indicate that microRNAs (miRNAs) are critical for the regulatory network of adipogenesis in human mesenchymal stem/stromal cells (MSCs). Fetal MSCs derived from amniotic fluid (AF-MSCs) represent a population of multipotent stem cells characterized by a wide range of differentiation properties that can be applied in cell-based therapies. In this study, miRNA microarray analysis was performed to assess miRNA expression in terminal differentiated AF-MSCs into adipocyte-like cells (AL cells). MiR-26a was identified in high expression levels in AL cells indicating a critical role in the process of adipogenesis. Overexpression of miR-26a in AF-MSCs led to significant induction of their adipogenic differentiation properties that were altered after miR-26a inhibition. We have demonstrated that miR-26a regulates adipogenesis through direct inhibition of PTEN, which in turn promotes activation of Akt pathway. Also, miR-26a modulates cell cycle during adipogenesis by interacting with Cyclin E1 and CDK6. These results point to the regulatory role of miR-26a and its target genes PTEN, Cyclin E1, and CDK6 in adipogenic differentiation of AF-MSCs, providing a basis for understanding the mechanisms of fat cell development and obesity.
Collapse
Affiliation(s)
- Ourania Trohatou
- 1 Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens , Athens, Greece .,2 Cell and Gene Therapy Laboratory, Centre of Basic Research II, Biomedical Research Foundation of the Academy of Athens (BRFAA) , Athens, Greece
| | - Dimitra Zagoura
- 1 Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens , Athens, Greece .,2 Cell and Gene Therapy Laboratory, Centre of Basic Research II, Biomedical Research Foundation of the Academy of Athens (BRFAA) , Athens, Greece
| | - Nikos K Orfanos
- 2 Cell and Gene Therapy Laboratory, Centre of Basic Research II, Biomedical Research Foundation of the Academy of Athens (BRFAA) , Athens, Greece
| | - Kalliopi I Pappa
- 3 First Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens , Athens, Greece
| | - Evangelos Marinos
- 1 Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens , Athens, Greece
| | - Nicholas P Anagnou
- 1 Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens , Athens, Greece .,2 Cell and Gene Therapy Laboratory, Centre of Basic Research II, Biomedical Research Foundation of the Academy of Athens (BRFAA) , Athens, Greece
| | - Maria G Roubelakis
- 1 Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens , Athens, Greece .,2 Cell and Gene Therapy Laboratory, Centre of Basic Research II, Biomedical Research Foundation of the Academy of Athens (BRFAA) , Athens, Greece
| |
Collapse
|
22
|
Kim WK, Oh KJ, Choi HR, Park A, Han BS, Chi SW, Kim SJ, Bae KH, Lee SC. MAP kinase phosphatase 3 inhibits brown adipocyte differentiation via regulation of Erk phosphorylation. Mol Cell Endocrinol 2015; 416:70-6. [PMID: 26325440 DOI: 10.1016/j.mce.2015.08.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/10/2015] [Accepted: 08/25/2015] [Indexed: 12/29/2022]
Abstract
Brown fat has been highlight as a new therapeutic target for treatment of obesity and diabetes. However, molecular mechanism underlying brown adipogenesis are not fully understood. Here, we identified that MAP kinase phosphatase 3 (MKP3) has a novel role as regulator of brown adipocyte differentiation. The expression of MKP3 was significantly decreased during the early stage(s) of brown adipocyte differentiation in HIB-1B cells and primary cells. Ectopic expression of MKP3 led to reduced brown adipocyte differentiation, whereas depletion of MKP3 significantly enhanced the differentiation of primary brown preadipocytes. Consistently, we found an increased brown adipocyte differentiation in MKP3-null MEF cells. These inhibitory effects of MKP3 could be resulted via the temporal regulation of Erk activation. In recent, it was reported that MKP3 deficient mice are resistant to diet-induced obesity, and display enhanced energy expenditure. Taken together, we suggest that MKP3 could be an important factor in the regulation of brown adipocyte differentiation.
Collapse
Affiliation(s)
- Won Kon Kim
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea; Department of Functional Genomics, University of Science and Technology of Korea, Daejeon, 305-806, Republic of Korea
| | - Kyoung-Jin Oh
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea
| | - Hye-Ryung Choi
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea
| | - Anna Park
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea
| | - Baek Soo Han
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea; Department of Functional Genomics, University of Science and Technology of Korea, Daejeon, 305-806, Republic of Korea
| | - Seung-Wook Chi
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea
| | - Seung Jun Kim
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea
| | - Kwang-Hee Bae
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea; Department of Functional Genomics, University of Science and Technology of Korea, Daejeon, 305-806, Republic of Korea.
| | - Sang Chul Lee
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea; Department of Functional Genomics, University of Science and Technology of Korea, Daejeon, 305-806, Republic of Korea.
| |
Collapse
|
23
|
White PJ, Mitchell PL, Schwab M, Trottier J, Kang JX, Barbier O, Marette A. Transgenic ω-3 PUFA enrichment alters morphology and gene expression profile in adipose tissue of obese mice: Potential role for protectins. Metabolism 2015; 64:666-76. [PMID: 25726444 DOI: 10.1016/j.metabol.2015.01.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 01/12/2015] [Accepted: 01/28/2015] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Dietary administration of ω-3 polyunsaturated fatty acids (PUFA) is often associated with altered adipose tissue (AT) morphology and/or function in obese mice. Yet, it is unclear whether this is an indirect consequence of reduced weight gain or results from direct actions of ω-3 PUFA. Here we studied the AT of high fat (HF)-fed fat-1 transgenic mice that convert endogenous ω-6 to ω-3 PUFA while maintaining equivalent fat accretion as their wild-type (WT) counterparts. MATERIALS AND METHODS Adipocyte size profiling, Affymetrix microarray pathway analysis, qPCR and protectin identification and analysis were performed in epididymal AT from hemizygous fat-1(+/-) mice and their wild type littermates that had been fed a HF diet for 8weeks from 6weeks of age. RESULTS Despite equivalent fat pad mass, we found that epididymal AT from HF-fed transgenic animals possesses fewer large and very large but more mid-size adipocytes compared to WT mice. In order to better understand the underlying mechanisms contributing to the observed alteration in adipocyte size we performed an Affymetrix microarray. Pathway analysis of these data highlighted adipogenesis, cholesterol biosynthesis, insulin signaling, prostaglandin synthesis/regulation and small ligand GPCRs as points where differentially expressed genes were significantly overrepresented. Observed changes were confirmed for four candidate genes: Cnr1, Cnr2, Faah and Pparg by qPCR. Finally we demonstrated that protectin DX is present in AT and that protectin DX and protectin D1 promote comparable PPARγ transcriptional activity. CONCLUSIONS These data provide unprecedented evidence that ω-3 PUFA coordinately regulate AT gene expression programs in a manner that is independent of restriction of weight gain or fat accrual and highlight an important influence of ω-3 PUFA on adipogenesis. Furthermore we provide primary evidence suggesting that protectins likely contribute to these effects via their influence on PPARγ.
Collapse
Affiliation(s)
- Phillip J White
- Department of Medicine, Québec Heart and Lung Institute, Université Laval, Ste-Foy, Québec, Canada and Laval University Hospital Research Center, Metabolism, Vascular and Renal Health Axis, Ste-Foy, Québec, Canada
| | - Patricia L Mitchell
- Department of Medicine, Québec Heart and Lung Institute, Université Laval, Ste-Foy, Québec, Canada and Laval University Hospital Research Center, Metabolism, Vascular and Renal Health Axis, Ste-Foy, Québec, Canada
| | - Michael Schwab
- Department of Medicine, Québec Heart and Lung Institute, Université Laval, Ste-Foy, Québec, Canada and Laval University Hospital Research Center, Metabolism, Vascular and Renal Health Axis, Ste-Foy, Québec, Canada
| | - Jocelyn Trottier
- Laboratory of Molecular Pharmacology, CHU-Québec Research Centre and the Faculty of Pharmacy, Laval University, Québec, Canada
| | - Jing X Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Olivier Barbier
- Laboratory of Molecular Pharmacology, CHU-Québec Research Centre and the Faculty of Pharmacy, Laval University, Québec, Canada
| | - André Marette
- Department of Medicine, Québec Heart and Lung Institute, Université Laval, Ste-Foy, Québec, Canada and Laval University Hospital Research Center, Metabolism, Vascular and Renal Health Axis, Ste-Foy, Québec, Canada.
| |
Collapse
|
24
|
Choudhery MS, Badowski M, Muise A, Harris DT. Effect of mild heat stress on the proliferative and differentiative ability of human mesenchymal stromal cells. Cytotherapy 2015; 17:359-368. [PMID: 25536863 DOI: 10.1016/j.jcyt.2014.11.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 10/30/2014] [Accepted: 11/03/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) are an attractive candidate for autologous cell therapy, but regenerative potential can be compromised with extensive in vitro cell passaging. Development of viable cell therapies must address the effect of in vitro passaging to maintain overall functionality of expanded MSCs. METHODS We examined the effect of repeated mild heat shock on the proliferation and differentiation capability of human adipose-derived MSCs. Adipose tissue MSCs were characterized by means of fluorescence activated cell sorting analysis for expression of CD3, CD14, CD19, CD34, CD44, CD45, CD73, CD90 and CD105. Similarly, the expression of SIRT-1, p16(INK4a) and p21 was determined by means of polymerase chain reaction. Measurements of population doubling, doubling time and superoxide dismutase activity were also determined. Differentiation of expanded MSCs into bone and adipose were analyzed qualitatively and quantitatively. RESULTS The strategy led to an increase in expression of SIRT-1 concomitant with enhanced viability, proliferation and delayed senescence. The stressed MSCs showed better differentiation into osteoblasts and adipocytes. CONCLUSIONS The results indicate that mild heat shock could be used to maintain MSC proliferative and differentiation potential.
Collapse
Affiliation(s)
- Mahmood S Choudhery
- Tissue Engineering and Regenerative Medicine Laboratory, Advanced Research Center in Biomedical Sciences, King Edward Medical University, Lahore, Pakistan; Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Michael Badowski
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Angela Muise
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - David T Harris
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona, USA.
| |
Collapse
|
25
|
Kim EY, Kim WK, Oh KJ, Han BS, Lee SC, Bae KH. Recent advances in proteomic studies of adipose tissues and adipocytes. Int J Mol Sci 2015; 16:4581-99. [PMID: 25734986 PMCID: PMC4394436 DOI: 10.3390/ijms16034581] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 12/29/2014] [Accepted: 02/16/2015] [Indexed: 12/27/2022] Open
Abstract
Obesity is a chronic disease that is associated with significantly increased levels of risk of a number of metabolic disorders. Despite these enhanced health risks, the worldwide prevalence of obesity has increased dramatically over the past few decades. Obesity is caused by the accumulation of an abnormal amount of body fat in adipose tissue, which is composed mostly of adipocytes. Thus, a deeper understanding of the regulation mechanism of adipose tissue and/or adipocytes can provide a clue for overcoming obesity-related metabolic diseases. In this review, we describe recent advances in the study of adipose tissue and/or adipocytes, focusing on proteomic approaches. In addition, we suggest future research directions for proteomic studies which may lead to novel treatments of obesity and obesity-related diseases.
Collapse
Affiliation(s)
- Eun Young Kim
- Functional Genomics Research Center, KRIBB, Daejeon 305-806, Korea.
| | - Won Kon Kim
- Functional Genomics Research Center, KRIBB, Daejeon 305-806, Korea.
- Department of Functional Genomics, University of Science and Technology of Korea, Daejeon 305-806, Korea.
| | - Kyoung-Jin Oh
- Functional Genomics Research Center, KRIBB, Daejeon 305-806, Korea.
| | - Baek Soo Han
- Functional Genomics Research Center, KRIBB, Daejeon 305-806, Korea.
- Department of Functional Genomics, University of Science and Technology of Korea, Daejeon 305-806, Korea.
| | - Sang Chul Lee
- Functional Genomics Research Center, KRIBB, Daejeon 305-806, Korea.
- Department of Functional Genomics, University of Science and Technology of Korea, Daejeon 305-806, Korea.
| | - Kwang-Hee Bae
- Functional Genomics Research Center, KRIBB, Daejeon 305-806, Korea.
- Department of Functional Genomics, University of Science and Technology of Korea, Daejeon 305-806, Korea.
| |
Collapse
|
26
|
Choudhery MS, Badowski M, Muise A, Pierce J, Harris DT. Subcutaneous Adipose Tissue-Derived Stem Cell Utility Is Independent of Anatomical Harvest Site. Biores Open Access 2015; 4:131-145. [PMID: 26309790 PMCID: PMC4497709 DOI: 10.1089/biores.2014.0059] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
One of the challenges for tissue engineering and regenerative medicine is to obtain suitably large cell numbers for therapy. Mesenchymal stem cells (MSCs) can easily be expanded in vitro to obtain large numbers of cells, but this approach may induce cellular senescence. The characteristics of cells are dependent on variables like age, body mass index (BMI), and disease conditions, however, and in the case of adipose tissue-derived stem cells (ASCs), anatomical harvest site is also an important variable that can affect the regenerative potential of isolated cells. We therefore had kept the parameters (age, BMI, disease conditions) constant in this study to specifically assess influence of anatomical sites of individual donors on utility of ASCs. Adipose tissue was obtained from multiple anatomical sites in individual donors, and viability and nucleated cell yield were determined. MSC frequency was enumerated using colony forming unit assay and cells were characterized by flow cytometry. Growth characteristics were determined by long-term population doubling analysis of each sample. Finally, MSCs were induced to undergo adipogenic, osteogenic, and chondrogenic differentiation. To validate the findings, these results were compared with similar single harvest sites from multiple individual patients. The results of the current study indicated that MSCs obtained from multiple harvest sites in a single donor have similar morphology and phenotype. All adipose depots in a single donor exhibited similar MSC yield, viability, frequency, and growth characteristics. Equivalent differentiation capacity into osteocytes, adipocytes, and chondrocytes was also observed. On the basis of results, we conclude that it is acceptable to combine MSCs obtained from various anatomical locations in a single donor to obtain suitably large cell numbers required for therapy, avoiding in vitro senescence and lengthy and expensive in vitro culturing and expansion steps.
Collapse
Affiliation(s)
- Mahmood S. Choudhery
- Tissue Engineering and Regenerative Medicine Laboratory, Advance Research Center of Biomedical Sciences, King Edward Medical University, Lahore, Pakistan
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Michael Badowski
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Angela Muise
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - John Pierce
- Aesthetic Surgery of Tucson, Tucson, Arizona
| | - David T. Harris
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| |
Collapse
|
27
|
Dubreuil V, Sap J, Harroch S. Protein tyrosine phosphatase regulation of stem and progenitor cell biology. Semin Cell Dev Biol 2015; 37:82-9. [DOI: 10.1016/j.semcdb.2014.09.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 09/11/2014] [Accepted: 09/15/2014] [Indexed: 12/18/2022]
|
28
|
Kim JW, Kim EY, Kim SY, Byun SK, Lee D, Oh KJ, Kim WK, Han BS, Chi SW, Lee SC, Bae KH. Identification of DNA aptamers toward epithelial cell adhesion molecule via cell-SELEX. Mol Cells 2014; 37:742-6. [PMID: 25266702 PMCID: PMC4213765 DOI: 10.14348/molcells.2014.0208] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 08/12/2014] [Accepted: 08/18/2014] [Indexed: 02/07/2023] Open
Abstract
The epithelial cell adhesion molecule (EpCAM, also known as CD326) is a transmembrane glycoprotein that is specifically detected in most adenocarcinomas and cancer stem cells. In this study, we performed a Cell systematic evolution of ligands by exponential enrichment (SELEX) experiment to isolate the aptamers against EpCAM. After seven round of Cell SELEX, we identified several aptamer candidates. Among the selected aptamers, EP166 specifically binds to cells expressing EpCAM with an equilibrium dissociation constant (Kd) in a micromolar range. On the other hand, it did not bind to negative control cells. Moreover, EP166 binds to J1ES cells, a mouse embryonic stem cell line. Therefore, the isolated aptamers against EpCAM could be used as a stem cell marker or in other applications in both stem cell and cancer studies.
Collapse
Affiliation(s)
- Ji Won Kim
- Research Center for Integrated Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806,
Korea
- Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon 305-806,
Korea
| | - Eun Young Kim
- Research Center for Integrated Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806,
Korea
| | - Sun Young Kim
- Research Center for Integrated Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806,
Korea
| | - Sang Kyung Byun
- Research Center for Integrated Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806,
Korea
- Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon 305-806,
Korea
| | - Dasom Lee
- Research Center for Integrated Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806,
Korea
- Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon 305-806,
Korea
| | - Kyoung-Jin Oh
- Research Center for Integrated Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806,
Korea
| | - Won Kon Kim
- Research Center for Integrated Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806,
Korea
| | - Baek Soo Han
- Research Center for Integrated Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806,
Korea
- Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon 305-806,
Korea
| | - Seung-Wook Chi
- Medical Proteomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806,
Korea
| | - Sang Chul Lee
- Research Center for Integrated Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806,
Korea
- Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon 305-806,
Korea
| | - Kwang-Hee Bae
- Research Center for Integrated Cellulomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806,
Korea
- Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon 305-806,
Korea
| |
Collapse
|
29
|
Zhang Z, Han Y, Song J, Luo R, Jin X, Mu D, Su S, Ji X, Ren YF, Liu H. Interferon-γ regulates the function of mesenchymal stem cells from oral lichen planus via indoleamine 2,3-dioxygenase activity. J Oral Pathol Med 2014; 44:15-27. [PMID: 25212102 DOI: 10.1111/jop.12224] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2014] [Indexed: 12/27/2022]
Abstract
BACKGROUND Little is known about mesenchymal stem cells (MSCs) in normal or inflammatory oral mucosal tissues, such as in oral lichen planus (OLP). Our objectives were to identify, isolate, and characterize MSCs from normal human oral mucosa and OLP lesions, and to evaluate indoleamine 2,3 dioxygenase (IDO) activity in mediating immunomodulation of MSCs from these tissues. METHODS Expressions of MSCs-related markers were examined in isolated cells by flow cytometry. Self-renewal and multilineage differentiations were studied to characterize these MSCs. Interferon-γ (IFN-γ), IDO, and STRO-1 were assessed by immunofluorescence. MSCs from oral mucosa and OLP or IFN-γ-pretreated MSCs were co-cultured with allogeneic mixed lymphocyte reaction assays (MLR). Proliferation and apoptosis of MLR or MSCs were detected by CCK8 and the annexin V-FITC apoptosis detection kit, respectively. IDO expression and activity were measured by real-time PCR, Western blotting, and high-performance liquid chromatography. RESULTS Isolated cells from oral mucosa and OLP expressed MSC-related markers STRO-1, CD105, and CD90 but were absent for hematopoietic stem cell markers CD34. Besides, they all showed self-renewal and multilineage differentiation capacities. MSCs in OLP presented STRO-1/IDO+ phenotype by immunofluorescence. MSCs and IFN-γ-pretreated MSCs could inhibit lymphocyte proliferation via IDO activity, but not via cell apoptosis. Long-term IFN-γ could also inhibit MSC proliferation via IDO activity. CONCLUSIONS Mesenchymal stem cells can be isolated from human oral mucosa and OLP tissues. Besides self-renewal and multilineage differentiation properties, these cells may participate in immunomodulation mediated by IFN-γ via IDO activity in human OLP.
Collapse
Affiliation(s)
- Zhihui Zhang
- School and Hospital of Stomatology, Peking University, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Mixed effects of long-term frozen storage on cord tissue stem cells. Cytotherapy 2014; 16:1313-21. [DOI: 10.1016/j.jcyt.2014.05.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 05/05/2014] [Accepted: 05/28/2014] [Indexed: 01/22/2023]
|
31
|
Choi HR, Kim WK, Park A, Jung H, Han BS, Lee SC, Bae KH. Protein tyrosine phosphatase profiling studies during brown adipogenic differentiation of mouse primary brown preadipocytes. BMB Rep 2014; 46:539-43. [PMID: 24152912 PMCID: PMC4133841 DOI: 10.5483/bmbrep.2013.46.11.058] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 03/16/2013] [Accepted: 03/31/2013] [Indexed: 11/20/2022] Open
Abstract
There is a correlation between obesity and the amount of brown adipose tissue; however, the molecular mechanism of brown adipogenic differentiation has not been as extensively studied. In this study, we performed a protein tyrosine phosphatase (PTP) profiling analysis during the brown adipogenic differentiation of mouse primary brown preadipocytes. Several PTPs, including PTPRF, PTPRZ, and DUSP12 showing differential expression patterns were identified. In the case of DUSP12, the expression level is dramatically downregulated during brown adipogenesis. The ectopic expression of DUSP12 using a retroviral expression system induces the suppression of adipogenic differentiation, whereas a catalytic inactive DUSP12 mutant showed no effect on differentiation. These results suggest that DUSP12 is involved in brown adipogenic differentiation and may be used as a target protein for the treatment or prevention of obesity by the regulation of brown adipogenic differentiation.
Collapse
Affiliation(s)
- Hye-Ryung Choi
- Research Center for Integrated Cellulomics, KRIBB; Department of Functional Genomics, University of Science and Technology (UST), Daejeon 305-806, Korea
| | | | | | | | | | | | | |
Collapse
|
32
|
Kim EY, Kim JW, Kim WK, Han BS, Park SG, Chung BH, Lee SC, Bae KH. Selection of aptamers for mature white adipocytes by cell SELEX using flow cytometry. PLoS One 2014; 9:e97747. [PMID: 24844710 PMCID: PMC4028271 DOI: 10.1371/journal.pone.0097747] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/23/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Adipose tissue, mainly composed of adipocytes, plays an important role in metabolism by regulating energy homeostasis. Obesity is primarily caused by an abundance of adipose tissue. Therefore, specific targeting of adipose tissue is critical during the treatment of obesity, and plays a major role in overcoming it. However, the knowledge of cell-surface markers specific to adipocytes is limited. METHODS AND RESULTS We applied the CELL SELEX (Systematic Evolution of Ligands by EXponential enrichment) method using flow cytometry to isolate molecular probes for specific recognition of adipocytes. The aptamer library, a mixture of FITC-tagged single-stranded random DNAs, is used as a source for acquiring molecular probes. With the increasing number of selection cycles, there was a steady increase in the fluorescence intensity toward mature adipocytes. Through 12 rounds of SELEX, enriched aptamers showing specific recognition toward mature 3T3-L1 adipocyte cells were isolated. Among these, two aptamers (MA-33 and 91) were able to selectively bind to mature adipocytes with an equilibrium dissociation constant (Kd) in the nanomolar range. These aptamers did not bind to preadipocytes or other cell lines (such as HeLa, HEK-293, or C2C12 cells). Additionally, it was confirmed that MA-33 and 91 can distinguish between mature primary white and primary brown adipocytes. CONCLUSIONS These selected aptamers have the potential to be applied as markers for detecting mature white adipocytes and monitoring adipogenesis, and could emerge as an important tool in the treatment of obesity.
Collapse
Affiliation(s)
- Eun Young Kim
- Research Center for Integrated Cellulomics, KRIBB, Daejeon, Republic of Korea
| | - Ji Won Kim
- Research Center for Integrated Cellulomics, KRIBB, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, Republic of Korea
| | - Won Kon Kim
- Research Center for Integrated Cellulomics, KRIBB, Daejeon, Republic of Korea
| | - Baek Soo Han
- Research Center for Integrated Cellulomics, KRIBB, Daejeon, Republic of Korea
| | - Sung Goo Park
- Medical Proteomics Research Center, KRIBB, Daejeon, Republic of Korea
| | - Bong Hyun Chung
- BioNanotechnology Research Center, Bioconvergence Research Institute, KRIBB, Daejeon, Republic of Korea
| | - Sang Chul Lee
- Research Center for Integrated Cellulomics, KRIBB, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, Republic of Korea
| | - Kwang-Hee Bae
- Research Center for Integrated Cellulomics, KRIBB, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, Republic of Korea
| |
Collapse
|
33
|
Borck G, de Vries L, Wu HJ, Smirin-Yosef P, Nürnberg G, Lagovsky I, Ishida LH, Thierry P, Wieczorek D, Nürnberg P, Foley J, Kubisch C, Basel-Vanagaite L. Homozygous truncating PTPRF mutation causes athelia. Hum Genet 2014; 133:1041-7. [PMID: 24781087 DOI: 10.1007/s00439-014-1445-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 04/09/2014] [Indexed: 12/11/2022]
Abstract
Athelia is a very rare entity that is defined by the absence of the nipple-areola complex. It can affect either sex and is mostly part of syndromes including other congenital or ectodermal anomalies, such as limb-mammary syndrome, scalp-ear-nipple syndrome, or ectodermal dysplasias. Here, we report on three children from two branches of an extended consanguineous Israeli Arab family, a girl and two boys, who presented with a spectrum of nipple anomalies ranging from unilateral hypothelia to bilateral athelia but no other consistently associated anomalies except a characteristic eyebrow shape. Using homozygosity mapping after single nucleotide polymorphism (SNP) array genotyping and candidate gene sequencing we identified a homozygous frameshift mutation in PTPRF as the likely cause of nipple anomalies in this family. PTPRF encodes a receptor-type protein phosphatase that localizes to adherens junctions and may be involved in the regulation of epithelial cell-cell contacts, peptide growth factor signaling, and the canonical Wnt pathway. Together with previous reports on female mutant Ptprf mice, which have a lactation defect, and disruption of one allele of PTPRF by a balanced translocation in a woman with amastia, our results indicate a key role for PTPRF in the development of the nipple-areola region.
Collapse
Affiliation(s)
- Guntram Borck
- Institute of Human Genetics, University of Ulm, Ulm, Germany,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Choudhery MS, Badowski M, Muise A, Pierce J, Harris DT. Cryopreservation of whole adipose tissue for future use in regenerative medicine. J Surg Res 2014; 187:24-35. [PMID: 24268882 DOI: 10.1016/j.jss.2013.09.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 08/13/2013] [Accepted: 09/18/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND Human adipose tissue (AT) is an ideal stem cell source for autologous cell-based therapies. The preferred setting for tissue engineering and regenerative medicine applications is the availability of clinically acceptable off-the-shelf cells and cell products. As AT is not always available for use, cryopreserved tissue represents an alternative approach. The aim of the present study was to compare the different properties of mesenchymal stem cells (MSCs) isolated from cryopreserved AT. We have measured cell recovery, viability, phenotype, proliferative potential, and differentiation into mesenchymal (adipogenic, osteogenic, chondrogenic) and nonmesenchymal (neuron-like cells) lineages. MATERIALS AND METHODS AT (n = 10) was harvested from donors and either processed fresh or cryopreserved in liquid nitrogen dewars. Both fresh and thawed tissues were enzymatically digested. MSCs were analyzed by fluorescence-activated cell sorting for CD3, CD14, CD19, CD34, CD44, CD45, CD73, CD90, and CD105 expression. Growth characteristics of both groups were investigated for population doublings, doubling time, saturation density, and plating efficiency. MSCs derived from fresh and thawed tissues were assessed for differentiation potential both qualitatively and quantitatively. RESULTS Adherent cells from fresh and thawed tissues displayed similar fibroblastic morphology. Cryopreservation did not alter expression of phenotypic markers. Similarly, the proliferative potential of MSCs was not compromised by cryopreservation. Furthermore, cryopreservation did not alter the differentiation capability of MSCs as determined with histochemistry, immunofluorescence, and real time reverse transcriptase-polymerase chain reaction. CONCLUSIONS We conclude that human AT could be successfully cryopreserved for future clinical application and the recovered MSCs were equivalent in functionality to the freshly processed MSCs.
Collapse
Affiliation(s)
- Mahmood S Choudhery
- Department of Pathology, Advanced Centre of Research in Biomedical Sciences, King Edward Medical University, Lahore, Pakistan; Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Michael Badowski
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Angela Muise
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - John Pierce
- Aesthetic Surgery of Tucson, Tucson, Arizona
| | - David T Harris
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona.
| |
Collapse
|
35
|
Choudhery MS, Badowski M, Muise A, Pierce J, Harris DT. Donor age negatively impacts adipose tissue-derived mesenchymal stem cell expansion and differentiation. J Transl Med 2014; 12:8. [PMID: 24397850 PMCID: PMC3895760 DOI: 10.1186/1479-5876-12-8] [Citation(s) in RCA: 370] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 12/03/2013] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Human adipose tissue is an ideal autologous source of mesenchymal stem cells (MSCs) for various regenerative medicine and tissue engineering strategies. Aged patients are one of the primary target populations for many promising applications. It has long been known that advanced age is negatively correlated with an organism's reparative and regenerative potential, but little and conflicting information is available about the effects of age on the quality of human adipose tissue derived MSCs (hAT-MSCs). METHODS To study the influence of age, the expansion and in vitro differentiation potential of hAT-MSCs from young (<30 years), adult (35-50 years) and aged (>60 years) individuals were investigated. MSCs were characterized for expression of the genes p16(INK4a) and p21 along with measurements of population doublings (PD), superoxide dismutase (SOD) activity, cellular senescence and differentiation potential. RESULTS Aged MSCs displayed senescent features when compared with cells isolated from young donors, concomitant with reduced viability and proliferation. These features were also associated with significantly reduced differentiation potential in aged MSCs compared to young MSCs. CONCLUSIONS In conclusion, advancing age negatively impacts stem cell function and such age related alterations may be detrimental for successful stem cell therapies.
Collapse
Affiliation(s)
- Mahmood S Choudhery
- Advanced Centre of Research in Biomedical Sciences, King Edward Medical University, Lahore, Pakistan
- Department of Immunobiology, College of Medicine, The University of Arizona, PO Box 245221, 85724, Tucson, AZ, USA
| | - Michael Badowski
- Department of Immunobiology, College of Medicine, The University of Arizona, PO Box 245221, 85724, Tucson, AZ, USA
| | - Angela Muise
- Department of Immunobiology, College of Medicine, The University of Arizona, PO Box 245221, 85724, Tucson, AZ, USA
| | - John Pierce
- Aesthetic Surgery of Tucson, Tucson, AZ, USA
| | - David T Harris
- Department of Immunobiology, College of Medicine, The University of Arizona, PO Box 245221, 85724, Tucson, AZ, USA
| |
Collapse
|
36
|
Gatta V, D'Aurora M, Lanuti P, Pierdomenico L, Sperduti S, Palka G, Gesi M, Marchisio M, Miscia S, Stuppia L. Gene expression modifications in Wharton's Jelly mesenchymal stem cells promoted by prolonged in vitro culturing. BMC Genomics 2013; 14:635. [PMID: 24053474 PMCID: PMC3849041 DOI: 10.1186/1471-2164-14-635] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 09/17/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND It has been demonstrated that the umbilical cord matrix, represented by the Wharton's Jelly (WJ), contains a great number of mesenchymal stem cells (MSCs), characterized by the expression of specific MSCs markers, shared by both human and animal models. The easy access to massive WJ amount makes it an attractive source of MSCs for cell-based therapies. However, as in other stem cell models, a deeper investigation of WJ-derived MSCs (WJ-MSCs) biological properties, probably modulated by their prolonged expansion and fast growth abilities, is required before their use in clinical settings. In this context, in order to analyze specific gene expression modifications occurring in WJ-MSCs, along with their culture prolongation, we investigated the transcriptomic profiles of WJ-MSCs after 4 and 12 passages of in vitro expansion by microarray analysis. RESULTS Hierarchical clustering analysis of the data set originated from a total of 6 experiments revealed that in vitro expansion of WJ-MSCs up to 12 passages promote selective over-expression of 157 genes and down-regulation of 440 genes compared to the 4th passage. IPA software analysis of the biological functions related to the identified sets of genes disclosed several transcripts related to inflammatory and cell stress response, cell proliferation and maturation, and apoptosis. CONCLUSIONS Taken together, these modifications may lead to an impairment of both cell expansion ability and resistance to apoptosis, two hallmarks of aging cells. In conclusion, results provided by the present study suggest the need to develop novel culture protocols able to preserve stem cell plasticity.
Collapse
Affiliation(s)
- Valentina Gatta
- Department of Medicine and Aging Science, School of Medicine and Health Sciences, University "G, d'Annunzio" Chieti-Pescara, via dei Vestini 31, 66013, Chieti, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Choi HR, Kim WK, Kim EY, Han BS, Min JK, Chi SW, Park SG, Bae KH, Lee SC. Dual-specificity phosphatase 10 controls brown adipocyte differentiation by modulating the phosphorylation of p38 mitogen-activated protein kinase. PLoS One 2013; 8:e72340. [PMID: 23977283 PMCID: PMC3748012 DOI: 10.1371/journal.pone.0072340] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 07/09/2013] [Indexed: 11/23/2022] Open
Abstract
Background Brown adipocytes play an important role in regulating the balance of energy, and as such, there is a strong correlation between obesity and the amount of brown adipose tissue. Although the molecular mechanism underlying white adipocyte differentiation has been well characterized, brown adipocyte differentiation has not been studied extensively. Here, we investigate the potential role of dual-specificity phosphatase 10 (DUSP10) in brown adipocyte differentiation using primary brown preadipocytes. Methods and Results The expression of DUSP10 increased continuously after the brown adipocyte differentiation of mouse primary brown preadipocytes, whereas the phosphorylation of p38 was significantly upregulated at an early stage of differentiation followed by steep downregulation. The overexpression of DUSP10 induced a decrease in the level of p38 phosphorylation, resulting in lower lipid accumulation than that in cells overexpressing the inactive mutant DUSP10. The expression levels of several brown adipocyte markers such as PGC-1α, UCP1, and PRDM16 were also significantly reduced upon the ectopic expression of DUSP10. Furthermore, decreased mitochondrial DNA content was detected in cells expressing DUSP10. The results obtained upon treatment with the p38 inhibitor, SB203580, clearly indicated that the phosphorylation of p38 at an early stage is important in brown adipocyte differentiation. The effect of the p38 inhibitor was partially recovered by DUSP10 knockdown using RNAi. Conclusions These results suggest that p38 phosphorylation is controlled by DUSP10 expression. Furthermore, p38 phosphorylation at an early stage is critical in brown adipocyte differentiation. Thus, the regulation of DUSP10 activity affects the efficiency of brown adipogenesis. Consequently, DUSP10 can be used as a novel target protein for the regulation of obesity.
Collapse
Affiliation(s)
- Hye-Ryung Choi
- Research Center for Integrated Cellulomics, KRIBB, Daejeon, Republic of Korea
| | - Won Kon Kim
- Research Center for Integrated Cellulomics, KRIBB, Daejeon, Republic of Korea
| | - Eun Young Kim
- Research Center for Integrated Cellulomics, KRIBB, Daejeon, Republic of Korea
| | - Baek Soo Han
- Research Center for Integrated Cellulomics, KRIBB, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Jeong-Ki Min
- Research Center for Integrated Cellulomics, KRIBB, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Seung-Wook Chi
- Biomedical Proteomics Research Center, KRIBB, Daejeon, Republic of Korea
| | - Sung Goo Park
- Biomedical Proteomics Research Center, KRIBB, Daejeon, Republic of Korea
| | - Kwang-Hee Bae
- Research Center for Integrated Cellulomics, KRIBB, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon, Republic of Korea
- * E-mail: (KHB); (SCL)
| | - Sang Chul Lee
- Research Center for Integrated Cellulomics, KRIBB, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon, Republic of Korea
- * E-mail: (KHB); (SCL)
| |
Collapse
|
38
|
Chen J, Crawford R, Chen C, Xiao Y. The key regulatory roles of the PI3K/Akt signaling pathway in the functionalities of mesenchymal stem cells and applications in tissue regeneration. TISSUE ENGINEERING PART B-REVIEWS 2013; 19:516-28. [PMID: 23651329 DOI: 10.1089/ten.teb.2012.0672] [Citation(s) in RCA: 182] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into various cell types and have been widely used in tissue engineering application. In tissue engineering, a scaffold, MSCs and growth factors are used as essential components and their interactions have been regarded to be important for regeneration of tissues. A critical problem for MSCs in tissue engineering is their low survival ability and functionality. Most MSCs are going to be apoptotic after transplantation. Therefore, increasing MSC survival ability and functionalities is the key for potential applications of MSCs. Several approaches have been studied to increase MSC tissue forming capacity including application of growth factors, overexpression of stem cell regulatory genes, and improvement of biomaterials for scaffolds. The effects of these approaches on MSCs have been associated with activation of the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. The pathway plays central regulatory roles in MSC survival, proliferation, migration, angiogenesis, cytokine production, and differentiation. In this review, we summarize and discuss the literatures related to the roles of the PI3K/Akt pathway in the functionalities of MSCs and the involvement of the pathway in biomaterials-increased MSC functionalities. Biomaterials have been modified in their properties and surface structure and loaded with growth factors to increase MSC functionalities. Several studies demonstrated that the biomaterials-increased MSC functionalities are mediated by the activation of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Jiezhong Chen
- 1 Institute of Health and Biomedical Innovation, Queensland University of Technology , Brisbane, Australia
| | | | | | | |
Collapse
|
39
|
Bae KH, Kim WK, Lee SC. Involvement of protein tyrosine phosphatases in adipogenesis: new anti-obesity targets? BMB Rep 2013; 45:700-6. [PMID: 23261055 PMCID: PMC4133817 DOI: 10.5483/bmbrep.2012.45.12.235] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Obesity is a worldwide epidemic as well as being a major risk factor for diabetes, cardiovascular diseases and several types of cancers. Obesity is mainly due to the overgrowth of adipose tissue arising from an imbalance between energy intake and energy expenditure. Adipose tissue, primarily composed of adipocytes, plays a key role in maintaining whole body energy homeostasis. In view of the treatment of obesity and obesity-related diseases, it is critical to understand the detailed signal transduction mechanisms of adipogenic differentiation. Adipogenic differentiation is tightly regulated by many key signal cascades, including insulin signaling. These signal cascades generally transfer or amplify the signal by using serial tyrosine phosphorylations. Thus, protein tyrosine kinases and protein tyrosine phosphatases are closely related to adipogenic differentiation. Compared to protein tyrosine kinases, protein tyrosine phosphatases have received little attention in adipogenic differentiation. This review aims to highlight the involvement of protein tyrosine phosphatases in adipogenic differentiation and the possibility of protein tyrosine phosphatases as drugs to target obesity. [BMB Reports 2012; 45(12): 700-706]
Collapse
Affiliation(s)
- Kwang-Hee Bae
- Medical Proteomics Research Center, KRIBB, Daejeon 305-806, Korea.
| | | | | |
Collapse
|
40
|
Kim DM, Choi HR, Park A, Shin SM, Bae KH, Lee SC, Kim IC, Kim WK. Retinoic acid inhibits adipogenesis via activation of Wnt signaling pathway in 3T3-L1 preadipocytes. Biochem Biophys Res Commun 2013; 434:455-9. [PMID: 23583383 DOI: 10.1016/j.bbrc.2013.03.095] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 03/27/2013] [Indexed: 01/29/2023]
Abstract
Although retinoic acid (RA) is well known to inhibit the differentiation of 3T3-L1 cells into adipocytes both in vivo and in vitro, its molecular mechanism is not fully understood. In this report, we investigate the inhibitory mechanism of adipocyte differentiation by RA in 3T3-L1 cells. Because both RA and Wnt are known to inhibit adipogenesis at a common step involving the inhibition of PPAR-γ expression, we focused on the crosstalk between these two signaling pathways. We found that RA treatment resulted in a dramatic inhibition of adipogenesis, especially at an early phase of differentiation, and led to increased β-catenin protein expression. Moreover, RA enhances the transcriptional activity of β-catenin as well as Wnt gene expression during adipogenesis. Taken together, the present study demonstrated that Wnt/β-catenin signaling may be associated with the RA-induced suppression of adipogenesis and may cooperatively inhibit adipocyte differentiation.
Collapse
Affiliation(s)
- Dong Min Kim
- Department of Medical System Engineering, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 500-712, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Choudhery MS, Badowski M, Muise A, Harris DT. Comparison of human mesenchymal stem cells derived from adipose and cord tissue. Cytotherapy 2013; 15:330-343. [PMID: 23318344 DOI: 10.1016/j.jcyt.2012.11.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 11/17/2012] [Indexed: 02/08/2023]
Abstract
BACKGROUND AIMS Stem cell therapies can provide an alternative approach for repair and regeneration of tissues and organs. Mesenchymal stem cells (MSCs) are promising candidates for cell-based therapies. Although bone marrow-derived MSCs have multi-lineage differentiation potential, bone marrow is not an optimal source because of the isolation process and low yield. The goal of this study was to investigate comparatively for the first time the in vitro regenerative potential of human MSCs from two other sources: umbilical cord tissue and adipose tissue. METHODS Cells from each tissue were isolated with 100% efficiency and characterized by fluorescence activated cell sorting (FACS) analysis for CD3, CD14, CD19, CD34, CD44, CD45, CD73, CD90 and CD105. Growth characteristics were investigated by population doublings, saturation density and plating efficiency. MSCs derived from both types of tissues were assessed for differentiation potential qualitatively and quantitatively. RESULTS FACS analysis showed no differences in expression of CD3, CD14, CD19, CD34, CD44, CD45, CD73, CD90 and CD105 between cord tissue MSCs (CT-MSCs) and adipose tissue MSCs (AT-MSCs). CT-MSCs showed more proliferative potential than AT-MSCs. When cultured in low numbers to determine colony-forming units (CFUs), CT-MSCs showed less CFUs than AT-MSCs. Cells from both sources efficiently differentiated into adipose, bone, cartilage and neuronal structures as determined with histochemistry, immunofluorescence and real-time reverse transcriptase polymerase chain reaction. CONCLUSIONS MSCs can easily be obtained from umbilical cord and adipose tissues, and it appears that both tissues are suitable sources of stem cells for potential use in regenerative medicine.
Collapse
Affiliation(s)
- Mahmood Saba Choudhery
- National Centre of Excellence in Molecular Biology, The Punjab University, Lahore, Pakistan
| | | | | | | |
Collapse
|
42
|
Abstract
Fyn is a tyrosine kinase with multiple roles in a variety of cellular processes. Here we report that Fyn is a new kinase involved in adipocyte differentiation. Elevated Fyn protein is detected specifically in the adipocytes of obese mice. Moreover, Fyn expression increases progressively in 3T3-L1 cells during in vitro adipogenesis, which correlates with its kinase activity. Inhibition of Fyn by either genetic or pharmacological manipulation restrains the 3T3-L1 preadipocytes from fully differentiating into mature adipocytes. Mechanistically, Fyn regulates the activity of the adipogenic transcription factor signal transducer and activator of transcription 5a (STAT5a) through enhancing its interaction with the GTPase phosphoinositide 3-kinase enhancer A (PIKE-A). The STAT5a activity is therefore reduced in Fyn- or PIKE-ablated adipose tissues, leading to diminished expression of adipogenic markers and adipocyte differentiation. Our data thus demonstrate a novel functional interaction between Fyn, PIKE-A, and STAT5a in mediating adipogenesis.
Collapse
|
43
|
Abstract
The post-translational modification of protein by acetylation has been emerging as a prevalent modification in enzymes that catalyze intermediary metabolism. However, the dynamics of protein acetylation during adipocyte differentiation that involves a major shift in cellular metabolism is not known. In this study, we investigated the temporal changes in acetylation during adipocyte differentiation. Almost all acetylated proteins identified showed a sequential change in acetylation during the differentiation process. While the majority of the acetylated proteins showed a sequential upregulation during adipocyte differentiation, in a few proteins a sequential downregulation of protein acetylation was also observed. Our findings suggest that a wide-ranging temporal change in protein acetylation occurs during adipocyte differentiation including differentially expressed proteins signifying an important role in adipocyte differentiation.
Collapse
|
44
|
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: 0.6] [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
|
45
|
Kim EY, Kim WK, Kang HJ, Kim JH, Chung SJ, Seo YS, Park SG, Lee SC, Bae KH. Acetylation of malate dehydrogenase 1 promotes adipogenic differentiation via activating its enzymatic activity. J Lipid Res 2012; 53:1864-76. [PMID: 22693256 DOI: 10.1194/jlr.m026567] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Acetylation is one of the most crucial post-translational modifications that affect protein function. Protein lysine acetylation is catalyzed by acetyltransferases, and acetyl-CoA functions as the source of the acetyl group. Additionally, acetyl-CoA plays critical roles in maintaining the balance between carbohydrate metabolism and fatty acid synthesis. Here, we sought to determine whether lysine acetylation is an important process for adipocyte differentiation. Based on an analysis of the acetylome during adipogenesis, various proteins displaying significant quantitative changes were identified by LC-MS/MS. Of these identified proteins, we focused on malate dehydrogenase 1 (MDH1). The acetylation level of MDH1 was increased up to 6-fold at the late stage of adipogenesis. Moreover, overexpression of MDH1 in 3T3-L1 preadipocytes induced a significant increase in the number of cells undergoing adipogenesis. The introduction of mutations to putative lysine acetylation sites showed a significant loss of the ability of cells to undergo adipogenic differentiation. Furthermore, the acetylation of MDH1 dramatically enhanced its enzymatic activity and subsequently increased the intracellular levels of NADPH. These results clearly suggest that adipogenic differentiation may be regulated by the acetylation of MDH1 and that the acetylation of MDH1 is one of the cross-talk mechanisms between adipogenesis and the intracellular energy level.
Collapse
Affiliation(s)
- Eun Young Kim
- Department of Biological Sciences, KAIST, Daejeon 305-701, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Liu XF, Bera TK, Kahue C, Escobar T, Fei Z, Raciti GA, Pastan I. ANKRD26 and its interacting partners TRIO, GPS2, HMMR and DIPA regulate adipogenesis in 3T3-L1 cells. PLoS One 2012; 7:e38130. [PMID: 22666460 PMCID: PMC3364200 DOI: 10.1371/journal.pone.0038130] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 05/04/2012] [Indexed: 01/04/2023] Open
Abstract
Partial inactivation of the Ankyrin repeat domain 26 (Ankrd26) gene causes obesity and diabetes in mice and increases spontaneous and induced adipogenesis in mouse embryonic fibroblasts. However, it is not yet known how the Ankrd26 protein carries out its biological functions. We identified by yeast two-hybrid and immunoprecipitation assays the triple functional domain protein (TRIO), the G protein pathway suppressor 2 (GPS2), the delta-interacting protein A (DIPA) and the hyaluronan-mediated motility receptor (HMMR) as ANKRD26 interacting partners. Adipogenesis of 3T3-L1 cells was increased by selective down-regulation of Ankrd26, Trio, Gps2, Hmmr and Dipa. Furthermore, GPS2 and DIPA, which are normally located in the nucleus, were translocated to the cytoplasm, when the C-terminus of ANKRD26 was introduced into these cells. These findings provide biochemical evidence that ANKRD26, TRIO, GPS2 and HMMR are novel and important regulators of adipogenesis and identify new targets for the modulation of adipogenesis.
Collapse
Affiliation(s)
- Xiu-Fen Liu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tapan K. Bera
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Charissa Kahue
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thelma Escobar
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Zhaoliang Fei
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gregory A. Raciti
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
47
|
Kim WK, Choi HR, Park SG, Ko Y, Bae KH, Lee SC. Myostatin inhibits brown adipocyte differentiation via regulation of Smad3-mediated β-catenin stabilization. Int J Biochem Cell Biol 2011; 44:327-34. [PMID: 22094186 DOI: 10.1016/j.biocel.2011.11.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 11/02/2011] [Accepted: 11/02/2011] [Indexed: 12/17/2022]
Abstract
Brown adipocytes play an important role in regulating energy balance, and there is a good correlation between obesity and the amount of brown adipose tissue. Although the molecular mechanism of white adipocyte differentiation has been well characterized, brown adipogenesis has not been studied extensively. Moreover, extracellular factors that regulate brown adipogenic differentiation are not fully understood. Here, we assessed the mechanism of the regulatory action of myostatin in brown adipogenic differentiation using primary brown preadipocytes. Our results clearly showed that differentiation of brown adipocytes was significantly inhibited by myostatin treatment. In addition, myostatin-induced suppression of brown adipogenesis was observed during the early phase of differentiation. Myostatin induced the phosphorylation of Smad3, which led to increased β-catenin stabilization. These effects were blocked by treatment with a Smad3 inhibitor. Expression of brown adipocyte-related genes, such as PPAR-γ, UCP-1, PGC-1α, and PRDM16, were dramatically down-regulated by treatment with myostatin, and further down-regulated by co-treatment with a β-catenin activator. Taken together, the present study demonstrated that myostatin is a potent negative regulator of brown adipogenic differentiation by modulation of Smad3-induced β-catenin stabilization. Our findings suggest that myostatin could be used as an extracellular factor in the control of brown adipocyte differentiation.
Collapse
Affiliation(s)
- Won Kon Kim
- Medical Proteomics Research Center, KRIBB, Daejeon 305-806, Republic of Korea
| | | | | | | | | | | |
Collapse
|
48
|
Kim WK, Jung H, Kim EY, Kim DH, Cho YS, Park BC, Park SG, Ko Y, Bae KH, Lee SC. RPTPμ tyrosine phosphatase promotes adipogenic differentiation via modulation of p120 catenin phosphorylation. Mol Biol Cell 2011; 22:4883-91. [PMID: 21998202 PMCID: PMC3237630 DOI: 10.1091/mbc.e11-03-0175] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Adipocyte differentiation can be regulated by the combined activity of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs). In particular, PTPs act as key regulators in differentiation-associated signaling pathways. We recently found that receptor-type PTPμ (RPTPμ) expression is markedly increased during the adipogenic differentiation of 3T3-L1 preadipocytes and mesenchymal stem cells. Here, we investigate the functional roles of RPTPμ and the mechanism of its involvement in the regulation of signal transduction during adipogenesis of 3T3-L1 cells. Depletion of endogenous RPTPμ by RNA interference significantly inhibited adipogenic differentiation, whereas RPTPμ overexpression led to an increase in adipogenic differentiation. Ectopic expression of p120 catenin suppressed adipocyte differentiation, and the decrease in adipogenesis by p120 catenin was recovered by introducing RPTPμ. Moreover, RPTPμ induced a decrease in the cytoplasmic p120 catenin expression by reducing its tyrosine phosphorylation level, consequently leading to enhanced translocation of Glut-4 to the plasma membrane. On the basis of these results, we propose that RPTPμ acts as a positive regulator of adipogenesis by modulating the cytoplasmic p120 catenin level. Our data conclusively demonstrate that differentiation into adipocytes is controlled by RPTPμ, supporting the utility of RPTPμ and p120 catenin as novel target proteins for the treatment of obesity.
Collapse
Affiliation(s)
- Won Kon Kim
- Medical Proteomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ma QY, Zhang XN, Jiang H, Wang ZQ, Zhang HJ, Xue LQ, Chen MD, Song HD. Mimecan in pituitary corticotroph cells may regulate ACTH secretion and the HPAA. Mol Cell Endocrinol 2011; 341:71-7. [PMID: 21664248 DOI: 10.1016/j.mce.2011.05.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2010] [Revised: 04/15/2011] [Accepted: 05/12/2011] [Indexed: 11/26/2022]
Abstract
Mimecan is a protein of unknown function that is expressed in the pituitary tissues of mouse and human. In this study, we observed the function of mimecan on the proopiomelanocortin (POMC) gene in the pituitary and the hypothalamo-pituitary-adrenal axis (HPAA). Incubating pituitary corticotroph AtT-20 cells with recombinant mimecan protein stimulated adrenocorticotrophic hormone (ACTH) secretion without significantly up-regulating POMC gene expression. In addition, pituitary corticotroph AtT-20 cell corticotropin-releasing hormone receptor 1 (CRHR1) gene expression was induced by mimecan. Interestingly, long-term mimecan overexpression in corticotroph cells increased CRHR1 mRNA levels while slightly decreasing POMC mRNA expression and ACTH secretion. Using mimecan knockout mice, we found that, although the serum ACTH concentration was not significantly different between wild type and mimecan knockout mice under basal conditions, the serum ACTH level was relatively lower in mimecan knockout mice after treatment with corticotropin-releasing hormone (CRH). Meanwhile, we observed that POMC and CRHR1 gene expression decreased in primary cultured knockout mouse pituitary cells compared with wild type cells. Taken together, these data suggest that mimecan expressed in pituitary corticotroph cells mainly regulates ACTH secretion in the pituitary and coordinates the HPAA.
Collapse
Affiliation(s)
- Qin-Yun Ma
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Kim WK, Bae KH, Choi HR, Kim DH, Choi KS, Cho YS, Kim HD, Park SG, Park BC, Ko Y, Lee SC. Leukocyte common antigen-related (LAR) tyrosine phosphatase positively regulates osteoblast differentiation by modulating extracellular signal-regulated kinase (ERK) activation. Mol Cells 2010; 30:335-40. [PMID: 20811813 DOI: 10.1007/s10059-010-0123-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 06/28/2010] [Accepted: 06/29/2010] [Indexed: 11/30/2022] Open
Abstract
Protein tyrosine phosphatases (PTPs) are pivotal regulators of key cellular functions, including cell growth, differentiation, and adhesion. Previously, we reported that leukocyte common antigen-related (LAR) tyrosine phosphatase promotes osteoblast differentiation in MC3T3-E1 preosteoblast cells. In the present study, the mechanism of the regulatory action of LAR on osteoblast differentiation was investigated. The mineralization of extracellular matrix and calcium accumulation in MC3T3-E1 cells were markedly enhanced by LAR overexpression, and these effects were further increased by treatment with a MEK inhibitor. In addition, LAR overexpression dramatically reduced extracellular signal-regulated kinase (Erk) activation during osteoblast differentiation. In contrast, a marginal effect of the inactive LAR mutant on Erk activation was detected. Expression of osteoblast-related genes such as ALP, BSP, DLX5, OCN, and RUNX2, was increased by LAR overexpression during osteoblast differentiation. On the basis of these results, we propose that LAR functions as a positive regulator of osteoblast differentiation by modulating ERK activation. Therefore, LAR phosphatase could be used as a novel regulatory target protein in many bone-associated diseases, including osteoporosis.
Collapse
Affiliation(s)
- Won Kon Kim
- Medical Proteomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|