1
|
Elucidation of the Role of FAM210B in Mitochondrial Metabolism and Erythropoiesis. Mol Cell Biol 2022; 42:e0014322. [PMID: 36374104 PMCID: PMC9753634 DOI: 10.1128/mcb.00143-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Mitochondria play essential and specific roles during erythroid differentiation. Recently, FAM210B, encoding a mitochondrial inner membrane protein, has been identified as a novel target of GATA-1, as well as an erythropoietin-inducible gene. While FAM210B protein is involved in regulate mitochondrial metabolism and heme biosynthesis, its detailed function remains unknown. Here, we generated both knockout and knockdown of endogenous FAM210B in human induced pluripotent stem-derived erythroid progenitor (HiDEP) cells using CRISPR/Cas9 methodology. Intriguingly, erythroid differentiation was more pronounced in the FAM210B-depleted cells, and this resulted in increased frequency of orthochromatic erythroblasts and decreased frequencies of basophilic/polychromatic erythroblasts. Comprehensive metabolite analysis and functional analysis indicated that oxygen consumption rates and the NAD (NAD+)/NADH ratio were significantly decreased, while lactate production was significantly increased in FAM210B deletion HiDEP cells, indicating involvement of FAM210B in mitochondrial energy metabolism in erythroblasts. Finally, we purified FAM210B-interacting protein from K562 cells that stably expressed His/biotin-tagged FAM210B. Mass spectrometry analysis of the His/biotin-purified material indicated interactions with multiple subunits of mitochondrial ATP synthases, such as subunit alpha (ATP5A) and beta (ATP5B). Our results suggested that FAM210B contributes prominently to erythroid differentiation by regulating mitochondrial energy metabolism. Our results provide insights into the pathophysiology of dysregulated hematopoiesis.
Collapse
|
2
|
Wang H, Zhang R, Wu X, Chen Y, Ji W, Wang J, Zhang Y, Xia Y, Tang Y, Yuan J. The Wnt Signaling Pathway in Diabetic Nephropathy. Front Cell Dev Biol 2022; 9:701547. [PMID: 35059392 PMCID: PMC8763969 DOI: 10.3389/fcell.2021.701547] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetic nephropathy (DN) is a serious kidney-related complication of both type 1 and type 2 diabetes mellitus (T1DM, T2DM) and the second major cause of end-stage kidney disease. DN can lead to hypertension, edema, and proteinuria. In some cases, DN can even progress to kidney failure, a life-threatening condition. The precise etiology and pathogenesis of DN remain unknown, although multiple factors are believed to be involved. The main pathological manifestations of DN include mesangial expansion, thickening of the glomerular basement membrane, and podocyte injury. Eventually, these pathological manifestations will lead to glomerulosclerosis, thus affecting renal function. There is an urgent need to develop new strategies for the prevention and treatment of DN. Existing evidence shows that the Wnt signaling cascade plays a key role in regulating the development of DN. Previous studies focused on the role of the Wnt canonical signaling pathway in DN. Subsequently, accumulated evidence on the mechanism of the Wnt non-canonical signaling indicated that Wnt/Ca2+ and Wnt/PCP also have essential roles in the progression of DN. In this review, we summarize the specific mechanisms of Wnt signaling in the occurrence and development of DN in podocyte injury, mesangial cell injury, and renal fibrosis. Also, to elucidate the significance of the Wnt canonical pathway in the process of DN, we uncovered evidence supporting that both Wnt/PCP and Wnt/Ca2+ signaling are critical for DN development.
Collapse
Affiliation(s)
- Haiying Wang
- Department of Physiology, Jining Medical University, Jining, China
| | - Ran Zhang
- Basic Medical School, Jining Medical University, Jining, China
| | - Xinjie Wu
- Basic Medical School, Jining Medical University, Jining, China
| | - Yafen Chen
- Basic Medical School, Jining Medical University, Jining, China
| | - Wei Ji
- Basic Medical School, Jining Medical University, Jining, China
| | - Jingsuo Wang
- Basic Medical School, Jining Medical University, Jining, China
| | - Yawen Zhang
- Basic Medical School, Jining Medical University, Jining, China
| | - Yong Xia
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Yiqun Tang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jinxiang Yuan
- Collaborative Innovation Center, Jining Medical University, Jining, China
| |
Collapse
|
3
|
Wang H, An J, He S, Liao C, Wang J, Tuo B. Chloride intracellular channels as novel biomarkers for digestive system tumors (Review). Mol Med Rep 2021; 24:630. [PMID: 34278487 DOI: 10.3892/mmr.2021.12269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 05/19/2021] [Indexed: 11/06/2022] Open
Abstract
Digestive system malignant tumors are common tumors, and the traditional treatment methods for these tumors include surgical resection, radiotherapy, chemotherapy, and molecularly targeted drugs. However, diagnosis remains challenging, and the early detection of postoperative recurrence is complicated. Therefore, it is necessary to explore novel biomarkers to facilitate clinical diagnosis and treatment. Accumulating evidence supports the crucial role of chloride channels in the development of multiple types of cancers. Given that chloride channels are widely expressed and involved in cell proliferation, apoptosis and cell cycle, among other processes, they may serve as a promising diagnostic and therapeutic target. Chloride intracellular channels (CLICs) are a class of chloride channels that are upregulated or downregulated in certain types of cancer. Furthermore, in certain cases, during cell cycle progression, the localization and function of the cytosolic form of the transmembrane proteins of CLICs are also altered, which may provide a key target for cancer therapy. The aim of the present review was to focus on CLICs as biomarkers for digestive system tumors.
Collapse
Affiliation(s)
- Hui Wang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jiaxing An
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Suyu He
- The Fourth Department of the Digestive Disease Center, Suining Central Hospital, Suining, Sichuan 629000, P.R. China
| | - Chengcheng Liao
- Special Key Laboratory of Oral Disease Research, Higher Education Institution in Guizhou Province, School of Stomatology, Zunyi Medical University, Zunyi, Guizhou 563006, P.R. China
| | - Juan Wang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
4
|
Szabo I, Zoratti M, Biasutto L. Targeting mitochondrial ion channels for cancer therapy. Redox Biol 2021; 42:101846. [PMID: 33419703 PMCID: PMC8113036 DOI: 10.1016/j.redox.2020.101846] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Pharmacological targeting of mitochondrial ion channels is emerging as a promising approach to eliminate cancer cells; as most of these channels are differentially expressed and/or regulated in cancer cells in comparison to healthy ones, this strategy may selectively eliminate the former. Perturbation of ion fluxes across the outer and inner membranes is linked to alterations of redox state, membrane potential and bioenergetic efficiency. This leads to indirect modulation of oxidative phosphorylation, which is/may be fundamental for both cancer and cancer stem cell survival. Furthermore, given the crucial contribution of mitochondria to intrinsic apoptosis, modulation of their ion channels leading to cytochrome c release may be of great advantage in case of resistance to drugs triggering apoptotic events upstream of the mitochondrial phase. In the present review, we give an overview of the known mitochondrial ion channels and of their modulators capable of killing cancer cells. In addition, we discuss state-of-the-art strategies using mitochondriotropic drugs or peptide-based approaches allowing a more efficient and selective targeting of mitochondrial ion channel-linked events.
Collapse
Affiliation(s)
- Ildiko Szabo
- Department of Biology, University of Padova, Italy; CNR Institute of Neurosciences, Padova, Italy.
| | | | - Lucia Biasutto
- CNR Institute of Neurosciences, Padova, Italy; Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
5
|
The Effect of Selenium Nanoparticles on the Osteogenic Differentiation of MC3T3-E1 Cells. NANOMATERIALS 2021; 11:nano11020557. [PMID: 33672352 PMCID: PMC7926403 DOI: 10.3390/nano11020557] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) regulate various functions of cells, including cell death, viability, and differentiation, and nanoparticles influence ROS depending on their size and shape. Selenium is known to regulate various physiological functions, such as cell differentiations and anti-inflammatory functions, and plays an important role in the regulation of ROS as an antioxidant. This study aims to investigate the effect of selenium nanoparticles (SeNPs) on the differentiation of osteogenic MC3T3-E1 cells. After fabrication of SeNPs with a size of 25.3 ± 2.6 nm, and confirmation of its oxidase-like activity, SeNPs were added to MC3T3-E1 cells with or without H2O2: 5~20 μg/mL SeNPs recovered cells damaged by 200 μM H2O2 via the intracellular ROS downregulating role of SeNPs, revealed by the ROS staining assay. The increase in osteogenic maturation with SeNPs was gradually investigated by expression of osteogenic genes at 3 and 7 days, Alkaline phosphatase activity staining at 14 days, and Alizarin red S staining at 28 days. Therefore, the role of SeNPs in regulating ROS and their therapeutic effects on the differentiation of MC3T3-E1 cells were determined, leading to possible applications for bone treatment.
Collapse
|
6
|
Wang W, Li X, Xu Y, Guo W, Yu H, Zhang L, Wang Y, Chen X. Acetylation-stabilized chloride intracellular channel 1 exerts a tumor-promoting effect on cervical cancer cells by activating NF-κB. Cell Oncol (Dordr) 2021; 44:557-568. [PMID: 33469837 DOI: 10.1007/s13402-020-00582-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2020] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Cervical cancer remains a major cause of cancer-related death in women, especially in developing countries. Previously, we found that the acetylation levels of chloride intracellular channel 1 (CLIC1) at lysine 131 were increased in cervical cancer tissues using a label-free proteomics approach. The aim of this study was to further determine the role of CLIC1 expression and its acetylation in cervical cancer. METHODS CLIC1 expression and its implications for the prognosis of cervical cancer were analyzed using primary patient samples and cells, and the Gene Expression Profiling Interactive Analysis (GEPIA) database (gepia.cancer-pku.cn). The effect of CLIC1 on cervical cancer cells was evaluated using Cell Counting Kit (CCK)-8, flow cytometry, scratch wound healing, transwell, Western blotting and co-immunoprecipitation (Co-IP) assays. In vivo tumor growth was assessed using mouse xenograft models. RESULTS We found that CLIC1 expression was increased in cervical cancer tissues and cells and that patients with a high CLIC1 expression tended to have a shorter overall survival time. Knockdown of CLIC1 significantly reduced in vitro cervical cancer cell proliferation, migration and invasion, and in vivo tumorigenesis. At the molecular level, we found that nuclear factor kappa B (NF-κB) activity was positively regulated by CLIC1. Pyrrolidine dithiocarbamate (PDTC), an inhibitor of NF-κB, attenuated the tumor-promoting effect of CLIC1. Moreover, we found that CLIC1 acetylation at K131 was upregulated in cervical cancer cells, which stabilized CLIC1 by inhibiting its ubiquitynation. Substitution of K131 inhibited CLIC1 ubiquitynation and promoted in vitro cervical cancer cell proliferation, migration and invasion, and in vivo tumor growth. In addition, we found that acetyltransferase HAT1 was responsible for CLIC1 acetylation at K131. CONCLUSION Our data indicate that CLIC1 acts as a tumor promoter in cervical cancer, suggesting a potential treatment strategy for cervical cancer by regulating CLIC1 expression and/or acetylation.
Collapse
Affiliation(s)
- Wanyue Wang
- School of Basic Medical Sciences, Qiqihar Medical University, Qiqihar, 161006, Heilongjiang, China
| | - Xin Li
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Ye Xu
- Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, Heilongjiang Province, China
| | - Weikang Guo
- Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, Heilongjiang Province, China
| | - Hui Yu
- Department of Cardiopulmonary Function, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Lu Zhang
- Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, Heilongjiang Province, China
| | - Yaoxian Wang
- Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, Heilongjiang Province, China.
| | - Xiuwei Chen
- Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, Heilongjiang Province, China.
| |
Collapse
|
7
|
Brinkhof B, Zhang B, Cui Z, Ye H, Wang H. ALCAM (CD166) as a gene expression marker for human mesenchymal stromal cell characterisation. Gene X 2020; 763S:100031. [PMID: 32550557 PMCID: PMC7285916 DOI: 10.1016/j.gene.2020.100031] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/08/2020] [Indexed: 02/06/2023] Open
Abstract
Background Human mesenchymal stromal cells (MSCs) phenotypically share their positive expression of the International Society for Cell and Gene Therapy (ISCT) markers CD73, CD90 and CD105 with fibroblasts. Fibroblasts are often co-isolated as an unwanted by-product from biopsy and they can rapidly overgrow the MSCs in culture. Indeed, many other surface markers have been proposed, though no unique MSC specific marker has been identified yet. Quantitative PCR (qPCR) is a precise, efficient and rapid method for gene expression analysis. To identify a marker suitable for accurate MSC characterisation, qPCR was exploited. Methods and results Two commercially obtained bone marrow (BM) derived MSCs and an hTERT immortalised BM-MSC line (MSC-TERT) have been cultured for different days and at different oxygen levels before RNA extraction. Together with RNA samples previous extracted from umbilical cord derived MSCs and MSC-TERT cells cultured in 2D or 3D, this heterogeneous sample set was quantitatively analysed for the expression levels of 18 candidate MSC marker genes. The expression levels in MSCs were compared with the expression levels in fibroblasts to verify the differentiation capability of these genes between MSCs and fibroblasts. None of the ISCT markers could differentiate between fibroblasts and MSCs. A total of six other genes (ALCAM, CLIC1, EDIL3, EPHA2, NECTIN2, and TMEM47) were identified as possible biomarkers for accurate identification of MSCs. Conclusion Justified by considerations on expression level, reliability and specificity, Activated-Leukocyte Cell Adhesion Molecule (ALCAM) was the best candidate for improving the biomarker set of MSC identification.
Collapse
Key Words
- (q)PCR, (quantitative) polymerase chain reaction
- AD, adipose
- AF, Amniotic Fluid
- ALCAM, Activated-Leukocyte Cell Adhesion Molecule
- Activated-leukocyte cell adhesion molecule
- BM, bone marrow
- BSG, Basigin
- Biomarker
- CD, cluster of differentiation
- CLIC1, chloride intracellular channel 1
- CLIC4, chloride intracellular channel 4
- Cq, Quantification cycle
- DF, Dermal Fibroblasts
- DP, Dental Pulp
- EDIL3, EGF like repeats and discoidin domains 3
- ENG, Endoglin
- EPHA2, EPH receptor A2
- ER, Endoplasmatic Reticulum
- FACS, Fluorescence Assisted Cell Sorting
- FN1, Fibronectin 1
- IGFBP7, insulin like growth factor binding protein 7
- ISCT, International Society for Cell and Gene Therapy
- ITGA1, integrin subunit alpha 1
- LAMP1, lysosomal associated membrane protein 1
- LRRC59, leucine rich repeat containing 59
- MCAM, melanoma cell adhesion molecule
- MM, Multiple Myeloma
- MPC, Mesenchymal Progenitor Cell
- MSC
- MSC, Mesenchymal Stromal Cells
- NECTIN2, nectin cell adhesion molecule 2
- NK, Natural Killer
- NT5E, 5′-nucleotidase ecto
- OS, Osteosarcoma
- PL, Placenta
- PPIA, peptidylprolyl isomerase A
- PUM1, pumilio RNA binding family member 1
- RM, Regenerative Medicine
- RNA
- RNA-seq, RNA sequencing
- RT, Reverse Transcriptase
- Regenerative medicine
- SEM, Standard Error of the Mean
- TBP, TATA-box binding protein
- TCF, Tissue Culture Plate
- TE, Tissue Engineering
- TFRC, transferrin receptor
- THY1, Thy-1 cell surface antigen
- TLN1, Talin 1
- TMEM47, transmembrane protein 47
- UC, umbilical cord
- YWHAZ, tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta
- cDNA, DNA complementary to RNA
- qPCR
Collapse
Affiliation(s)
- Bas Brinkhof
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Bo Zhang
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Hui Wang
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom.,Oxford Suzhou Centre for Advanced Research, Suzhou Industrial Park, Jiangsu 215123, China
| |
Collapse
|
8
|
Leanza L, Checchetto V, Biasutto L, Rossa A, Costa R, Bachmann M, Zoratti M, Szabo I. Pharmacological modulation of mitochondrial ion channels. Br J Pharmacol 2019; 176:4258-4283. [PMID: 30440086 DOI: 10.1111/bph.14544] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/15/2018] [Accepted: 10/22/2018] [Indexed: 12/17/2022] Open
Abstract
The field of mitochondrial ion channels has undergone a rapid development during the last three decades, due to the molecular identification of some of the channels residing in the outer and inner membranes. Relevant information about the function of these channels in physiological and pathological settings was gained thanks to genetic models for a few, mitochondria-specific channels. However, many ion channels have multiple localizations within the cell, hampering a clear-cut determination of their function by pharmacological means. The present review summarizes our current knowledge about the ins and outs of mitochondrial ion channels, with special focus on the channels that have received much attention in recent years, namely, the voltage-dependent anion channels, the permeability transition pore (also called mitochondrial megachannel), the mitochondrial calcium uniporter and some of the inner membrane-located potassium channels. In addition, possible strategies to overcome the difficulties of specifically targeting mitochondrial channels versus their counterparts active in other membranes are discussed, as well as the possibilities of modulating channel function by small peptides that compete for binding with protein interacting partners. Altogether, these promising tools along with large-scale chemical screenings set up to identify new, specific channel modulators will hopefully allow us to pinpoint the actual function of most mitochondrial ion channels in the near future and to pharmacologically affect important pathologies in which they are involved, such as neurodegeneration, ischaemic damage and cancer. LINKED ARTICLES: This article is part of a themed section on Mitochondrial Pharmacology: Featured Mechanisms and Approaches for Therapy Translation. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.22/issuetoc.
Collapse
Affiliation(s)
- Luigi Leanza
- Department of Biology, University of Padova, Padova, Italy
| | | | - Lucia Biasutto
- CNR Institute of Neurosciences, Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Andrea Rossa
- Department of Chemical Sciences, University of Padova, Padova, Italy
| | - Roberto Costa
- Department of Biology, University of Padova, Padova, Italy
| | | | - Mario Zoratti
- CNR Institute of Neurosciences, Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ildiko Szabo
- Department of Biology, University of Padova, Padova, Italy.,CNR Institute of Neurosciences, Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
9
|
Gururaja Rao S, Ponnalagu D, Patel NJ, Singh H. Three Decades of Chloride Intracellular Channel Proteins: From Organelle to Organ Physiology. CURRENT PROTOCOLS IN PHARMACOLOGY 2018; 80:11.21.1-11.21.17. [PMID: 30040212 PMCID: PMC6060641 DOI: 10.1002/cpph.36] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intracellular organelles are membranous structures central for maintaining cellular physiology and the overall health of the cell. To maintain cellular function, intracellular organelles are required to tightly regulate their ionic homeostasis. Any imbalance in ionic concentrations can disrupt energy production (mitochondria), protein degradation (lysosomes), DNA replication (nucleus), or cellular signaling (endoplasmic reticulum). Ionic homeostasis is also important for volume regulation of intracellular organelles and is maintained by cation and anion channels as well as transporters. One of the major classes of ion channels predominantly localized to intracellular membranes is chloride intracellular channel proteins (CLICs). They are non-canonical ion channels with six homologs in mammals, existing as either soluble or integral membrane protein forms, with dual functions as enzymes and channels. Provided in this overview is a brief introduction to CLICs, and a summary of recent information on their localization, biophysical properties, and physiological roles. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Shubha Gururaja Rao
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Devasena Ponnalagu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Neel J Patel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Harpreet Singh
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Brum AM, van der Leije CS, Schreuders-Koedam M, Verhoeven J, Janssen M, Dekkers DH, Demmers JA, Eijken M, van de Peppel J, van Leeuwen JP, van der Eerden BC. Identification of Chloride Intracellular Channel Protein 3 as a Novel Gene Affecting Human Bone Formation. JBMR Plus 2017; 1:16-26. [PMID: 30283877 PMCID: PMC6124162 DOI: 10.1002/jbm4.10003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/09/2017] [Indexed: 12/14/2022] Open
Abstract
Osteoporosis is a common skeletal disorder characterized by low bone mass leading to increased bone fragility and fracture susceptibility. The bone building cells, osteoblasts, are derived from mesenchymal stromal cells (MSCs); however, with increasing age osteogenic differentiation is diminished and more adipocytes are seen in the bone marrow, suggesting a shift in MSC lineage commitment. Identification of specific factors that stimulate osteoblast differentiation from human MSCs may deliver therapeutic targets to treat osteoporosis. The aim of this study was to identify novel genes involved in osteoblast differentiation of human bone marrow–derived MSCs (hMSCs). We identified the gene chloride intracellular channel protein 3 (CLIC3) to be strongly upregulated during MSC‐derived osteoblast differentiation. Lentiviral overexpression of CLIC3 in hMSCs caused a 60% increase of matrix mineralization. Conversely, knockdown of CLIC3 in hMSCs using two short‐hairpin RNAs (shRNAs) against CLIC3 resulted in a 69% to 76% reduction in CLIC3 mRNA expression, 53% to 37% less alkaline phosphatase (ALP) activity, and 78% to 88% less matrix mineralization compared to scrambled control. Next, we used an in vivo human bone formation model in which hMSCs lentivirally transduced with the CLIC3 overexpression construct were loaded onto a scaffold (hydroxyapatite‐tricalcium‐phosphate), implanted under the skin of NOD‐SCID mice, and analyzed for bone formation 8 weeks later. CLIC3 overexpression led to a 15‐fold increase in bone formation (0.33% versus 5.05% bone area relative to scaffold). Using a Clic3‐His‐tagged pull‐down assay and liquid chromatography–mass spectrometry (LS/MS)‐based proteomics analysis in lysates of osteogenically differentiated hMSCs, we showed that CLIC3 interacts with NIMA‐related kinase 9 (NEK9) and phosphatidylserine synthase 1 (PTDSS1) in vitro, and this finding was supported by immunofluorescent analysis. In addition, inhibition of NEK9 or PTDSS1 gene expression by shRNAs inhibited osteoblast differentiation and mineralization. In conclusion, we successfully identified CLIC3 to be a lineage‐specific gene regulating osteoblast differentiation and bone formation through its interaction with NEK9 and PTDSS1. © The Authors. JBMR Plus is published by Wiley Periodicals, Inc. on behalf of the American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Andrea M Brum
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Cindy S van der Leije
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Marijke Schreuders-Koedam
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Jeroen Verhoeven
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | | | - Dick Hw Dekkers
- Proteomics Center Erasmus University Medical Center Rotterdam The Netherlands
| | - Jeroen Aa Demmers
- Proteomics Center Erasmus University Medical Center Rotterdam The Netherlands
| | | | - Jeroen van de Peppel
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Johannes Ptm van Leeuwen
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Bram Cj van der Eerden
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| |
Collapse
|
11
|
Wang D, Wang H, Gao F, Wang K, Dong F. ClC-3 Promotes Osteogenic Differentiation in MC3T3-E1 Cell After Dynamic Compression. J Cell Biochem 2016; 118:1606-1613. [PMID: 27922190 DOI: 10.1002/jcb.25823] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/02/2016] [Indexed: 11/11/2022]
Abstract
ClC-3 chloride channel has been proved to have a relationship with the expression of osteogenic markers during osteogenesis, persistent static compression can upregulate the expression of ClC-3 and regulate osteodifferentiation in osteoblasts. However, there was no study about the relationship between the expression of ClC-3 and osteodifferentiation after dynamic compression. In this study, we applied dynamic compression on MC3T3-E1 cells to detect the expression of ClC-3, runt-related transcription factor 2 (Runx2), bone morphogenic protein-2 (BMP-2), osteopontin (OPN), nuclear-associated antigen Ki67 (Ki67), and proliferating cell nuclear antigen (PCNA) in biopress system, then we investigated the expression of these genes after dynamic compression with Chlorotoxin (specific ClC-3 chloride channel inhibitor) added. Under transmission electron microscopy, there were more cell surface protrusions, rough surfaced endoplasmic reticulum, mitochondria, Golgi apparatus, abundant glycogen, and lysosomes scattered in the cytoplasm in MC3T3-E1 cells after dynamic compression. The nucleolus was more obvious. We found that ClC-3 was significantly up-regulated after dynamic compression. The compressive force also up-regulated Runx2, BMP-2, and OPN after dynamic compression for 2, 4 and 8 h. The proliferation gene Ki67 and PCNA did not show significantly change after dynamic compression for 8 h. Chlorotoxin did not change the expression of ClC-3 but reduced the expression of Runx2, BMP-2, and OPN after dynamic compression compared with the group without Cltx added. The data from the current study suggested that ClC-3 may promotes osteogenic differentiation in MC3T3-E1 cell after dynamic compression. J. Cell. Biochem. 118: 1606-1613, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Dawei Wang
- Department of Stomatology, Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei, China
| | - Hao Wang
- Department of Stomatology, Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei, China
| | - Feng Gao
- Department of Pathology, Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei, China
| | - Kun Wang
- Department of Stomatology, Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei, China
| | - Fusheng Dong
- Department of Oral and Maxillofacial Surgery, Stomatology Hospital of Hebei Medical University, Shijiazhuang 050017, Hebei, China.,Hebei Key Laboratory of Oral Medicine, Shijiazhuang 050017, Hebei, China
| |
Collapse
|
12
|
Abstract
Ion channels have emerged as regulators of developmental processes. In model organisms and in people with mutations in ion channels, disruption of ion channel function can affect cell proliferation, cell migration, and craniofacial and limb patterning. Alterations of ion channel function affect morphogenesis in fish, frogs, mammals, and flies, demonstrating that ion channels have conserved roles in developmental processes. One model suggests that ion channels affect proliferation and migration through changes in cell volume. However, ion channels have not explicitly been placed in canonical developmental signaling cascades until recently. This review gives examples of ion channels that influence developmental processes, offers a potential underlying molecular mechanism involving bone morphogenetic protein (BMP) signaling, and finally explores exciting possibilities for manipulating ion channels to influence cell fate for regenerative medicine and to impact disease.
Collapse
Affiliation(s)
- Emily Bates
- Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado 80045;
| |
Collapse
|
13
|
Kirschner G, Balla B, Horváth P, Kövesdi A, Lakatos G, Takács I, Nagy Z, Tóbiás B, Árvai K, Kósa JP, Lakatos P. Effects of imatinib and nilotinib on the whole transcriptome of cultured murine osteoblasts. Mol Med Rep 2016; 14:2025-37. [PMID: 27430367 PMCID: PMC4991674 DOI: 10.3892/mmr.2016.5459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/17/2016] [Indexed: 12/11/2022] Open
Abstract
Numerous clinical observations have confirmed that breakpoint cluster region-abelson fusion oncoprotein tyrosine kinase inhibitors used in leukemia treatment alter bone physiology in a complex manner. The aim of the present study was to analyze the whole transcriptome of cultured murine osteoblasts and determine the changes following treatment with imatinib and nilotinib using Sequencing by Oligonucleotide Ligation and Detection next generation RNA sequencing. This study also aimed to identify candidate signaling pathways and network regulators by multivariate Ingenuity Pathway Analysis. Based on the right-tailed Fisher's exact test, significantly altered pathways including upstream regulators were defined for each drug. The correlation between these pathways and bone metabolism was also examined. The preliminary results suggest the two drugs have different mechanisms of action on osteoblasts, and imatinib was shown to have a greater effect on gene expression. Data also indicated the potential role of a number of genes and signaling cascades that may contribute to identifying novel targets for the treatment of metabolic bone diseases.
Collapse
Affiliation(s)
- Gyöngyi Kirschner
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Bernadett Balla
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Péter Horváth
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Andrea Kövesdi
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Gergely Lakatos
- Second Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - István Takács
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Zsolt Nagy
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Bálint Tóbiás
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Kristóf Árvai
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - János Pál Kósa
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Péter Lakatos
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| |
Collapse
|
14
|
Yang JE, Song MS, Shen Y, Ryu PD, Lee SY. The Role of KV7.3 in Regulating Osteoblast Maturation and Mineralization. Int J Mol Sci 2016; 17:407. [PMID: 26999128 PMCID: PMC4813262 DOI: 10.3390/ijms17030407] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 03/02/2016] [Accepted: 03/10/2016] [Indexed: 11/19/2022] Open
Abstract
KCNQ (KV7) channels are voltage-gated potassium (KV) channels, and the function of KV7 channels in muscles, neurons, and sensory cells is well established. We confirmed that overall blockade of KV channels with tetraethylammonium augmented the mineralization of bone-marrow-derived human mesenchymal stem cells during osteogenic differentiation, and we determined that KV7.3 was expressed in MG-63 and Saos-2 cells at the mRNA and protein levels. In addition, functional KV7 currents were detected in MG-63 cells. Inhibition of KV7.3 by linopirdine or XE991 increased the matrix mineralization during osteoblast differentiation. This was confirmed by alkaline phosphatase, osteocalcin, and osterix in MG-63 cells, whereas the expression of Runx2 showed no significant change. The extracellular glutamate secreted by osteoblasts was also measured to investigate its effect on MG-63 osteoblast differentiation. Blockade of KV7.3 promoted the release of glutamate via the phosphorylation of extracellular signal-regulated kinase 1/2-mediated upregulation of synapsin, and induced the deposition of type 1 collagen. However, activation of KV7.3 by flupirtine did not produce notable changes in matrix mineralization during osteoblast differentiation. These results suggest that KV7.3 could be a novel regulator in osteoblast differentiation.
Collapse
Affiliation(s)
- Ji Eun Yang
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea.
| | - Min Seok Song
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea.
| | - Yiming Shen
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea.
| | - Pan Dong Ryu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea.
| | - So Yeong Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea.
| |
Collapse
|
15
|
Brooks MB, Catalfamo JL, MacNguyen R, Tim D, Fancher S, McCardle JA. A TMEM16F point mutation causes an absence of canine platelet TMEM16F and ineffective activation and death-induced phospholipid scrambling. J Thromb Haemost 2015; 13:2240-52. [PMID: 26414452 DOI: 10.1111/jth.13157] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/12/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND TMEM16F is an ion channel and calcium-dependent lipid scramblase that mediates phosphatidylserine (PS) exposure on the plasma membrane. Two disparate disease phenotypes are associated with TMEM16F loss-of-function mutations: a rare bleeding disorder (Scott syndrome) and skeletal malformations due to aberrant bone mineralization in a TMEM16F knockout mouse. We therefore undertook comparative studies of TMEM16F expression in canine Scott syndrome (CSS), an autosomal recessive platelet defect. OBJECTIVES To define anoctamin proteins and scramblase response of CSS platelets and to determine whether TMEM16F is the CSS disease gene. METHODS CSS TMEM16F cDNA and gene were sequenced and mutation detection was performed in CSS pedigrees. Platelet fractions from CSS dogs were isolated for proteomic and immunologic characterization of TMEM16F. Annexin V was used as a flow cytometric marker of induced platelet PS externalization. RESULTS A TMEM16F splice site mutation segregated with the CSS trait and TMEM16F protein was undetectable in CSS platelet membranes; however, a second anoctamin, TMEM16K, was found. Proteomic analyses revealed a network of 32 proteins that differentially cosegregated with platelet plasma membrane TMEM16F. CSS platelets had profoundly impaired scramblase response to pharmacologic and physiologic agents that increase intraplatelet calcium and conditions that induce apoptotic and necrotic cell death. CONCLUSIONS CSS platelets represent a TMEM16F-null mutant model that demonstrates a central role for TMEM16F in mediating platelet PS externalization in response to activating and death signals. Platelet TMEM16F may prove a novel drug target for modulating platelet procoagulant activity and extending platelet life span.
Collapse
Affiliation(s)
- M B Brooks
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - J L Catalfamo
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - R MacNguyen
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - D Tim
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - S Fancher
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - J A McCardle
- Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| |
Collapse
|
16
|
Averaimo S, Gritti M, Barini E, Gasparini L, Mazzanti M. CLIC1 functional expression is required for cAMP-induced neurite elongation in post-natal mouse retinal ganglion cells. J Neurochem 2014; 131:444-56. [PMID: 25060644 DOI: 10.1111/jnc.12832] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 07/14/2014] [Accepted: 07/21/2014] [Indexed: 01/10/2023]
Abstract
During neuronal differentiation, axonal elongation is regulated by both external and intrinsic stimuli, including neurotropic factors, cytoskeleton dynamics, second messengers such as cyclic adenosine monophosphate (cAMP), and neuronal excitability. Chloride intracellular channel 1 (CLIC1) is a cytoplasmic hydrophilic protein that, upon stimulation, dimerizes and translocates to the plasma membrane, where it contributes to increase the membrane chloride conductance. Here, we investigated the expression of CLIC1 in primary hippocampal neurons and retinal ganglion cells (RGCs) and examined how the functional expression of CLIC1 specifically modulates neurite outgrowth of neonatal murine RGCs. Using a combination of electrophysiology and immunohistochemistry, we found that CLIC1 is expressed in hippocampal neurons and RGCs and that the chloride current mediated by CLIC1 is required for maintaining growth cone morphology and sustaining cAMP-stimulated neurite elongation in dissociated immunopurified RGCs. In cultured RGCs, inhibition of CLIC1 ionic current through the pharmacological blocker IAA94 or a specific anti-CLIC1 antibody directed against its extracellular domain prevents the neurite outgrowth induced by cAMP. CLIC1-mediated chloride current, which results from an increased open probability of the channel, is detected only when cAMP is elevated. Inhibition of protein kinase A prevents such current. These results indicate that CLIC1 functional expression is regulated by cAMP via protein kinase A and is required for neurite outgrowth modulation during neuronal differentiation. Using a combination of electrophysiology and immunohistochemistry, we found that the chloride intracellular channel 1 (CLIC1) protein modulates the speed of neurite growth. The chloride current mediated by CLIC1 is essential for maintaining growth cone morphology and is required for sustaining cAMP-stimulated neurite elongation in dissociated immunopurified neurons. The presence of either the CLIC1 current blocker IAA94 or the anti-CLIC1 antibody inhibits neurite growth of Retina Ganglion Cells cultured in the presence of 10 micromolar forskolin for 24 h.
Collapse
|
17
|
Serrano-Candelas E, Farré D, Aranguren-Ibáñez Á, Martínez-Høyer S, Pérez-Riba M. The vertebrate RCAN gene family: novel insights into evolution, structure and regulation. PLoS One 2014; 9:e85539. [PMID: 24465593 PMCID: PMC3896409 DOI: 10.1371/journal.pone.0085539] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 12/04/2013] [Indexed: 12/30/2022] Open
Abstract
Recently there has been much interest in the Regulators of Calcineurin (RCAN) proteins which are important endogenous modulators of the calcineurin-NFATc signalling pathway. They have been shown to have a crucial role in cellular programmes such as the immune response, muscle fibre remodelling and memory, but also in pathological processes such as cardiac hypertrophy and neurodegenerative diseases. In vertebrates, the RCAN family form a functional subfamily of three members RCAN1, RCAN2 and RCAN3 whereas only one RCAN is present in the rest of Eukarya. In addition, RCAN genes have been shown to collocate with RUNX and CLIC genes in ACD clusters (ACD21, ACD6 and ACD1). How the RCAN genes and their clustering in ACDs evolved is still unknown. After analysing RCAN gene family evolution using bioinformatic tools, we propose that the three RCAN vertebrate genes within the ACD clusters, which evolved from single copy genes present in invertebrates and lower eukaryotes, are the result of two rounds of whole genome duplication, followed by a segmental duplication. This evolutionary scenario involves the loss or gain of some RCAN genes during evolution. In addition, we have analysed RCAN gene structure and identified the existence of several characteristic features that can be involved in RCAN evolution and gene expression regulation. These included: several transposable elements, CpG islands in the 5′ region of the genes, the existence of antisense transcripts (NAT) associated with the three human genes, and considerable evidence for bidirectional promoters that regulate RCAN gene expression. Furthermore, we show that the CpG island associated with the RCAN3 gene promoter is unmethylated and transcriptionally active. All these results provide timely new insights into the molecular mechanisms underlying RCAN function and a more in depth knowledge of this gene family whose members are obvious candidates for the development of future therapies.
Collapse
Affiliation(s)
- Eva Serrano-Candelas
- Cancer and Human Molecular Genetics Department, Bellvitge Biomedical Research Institute – IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Domènec Farré
- Biological Aggression and Response Mechanisms Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer – IDIBAPS, Barcelona, Spain
| | - Álvaro Aranguren-Ibáñez
- Cancer and Human Molecular Genetics Department, Bellvitge Biomedical Research Institute – IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Sergio Martínez-Høyer
- Cancer and Human Molecular Genetics Department, Bellvitge Biomedical Research Institute – IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Mercè Pérez-Riba
- Cancer and Human Molecular Genetics Department, Bellvitge Biomedical Research Institute – IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
- * E-mail:
| |
Collapse
|
18
|
Proteome of human stem cells from periodontal ligament and dental pulp. PLoS One 2013; 8:e71101. [PMID: 23940696 PMCID: PMC3733711 DOI: 10.1371/journal.pone.0071101] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 06/25/2013] [Indexed: 12/12/2022] Open
Abstract
Background Many adult tissues contain a population of stem cells with the ability to regenerate structures similar to the microenvironments from which they are derived in vivo and represent a promising therapy for the regeneration of complex tissues in the clinical disorder. Human adult stem cells (SCs) including bone marrow stem cells (BMSCs), dental pulp stem cells (DPSCs) and periodontal ligament stem cells (PDLSCs) have been characterized for their high proliferative potential, expression of characteristic SC-associated markers and for the plasticity to differentiate in different lineage in vitro. Methodology/Principal Findings The aim of this study is to define the molecular features of stem cells from oral tissue by comparing the proteomic profiles obtained with 2-DE followed by MALDI-TOF/TOF of ex-vivo cultured human PDLSCs, DPSCs and BMSCs. Our results showed qualitative similarities in the proteome profiles among the SCs examined including some significant quantitative differences. To enrich the knowledge of oral SCs proteome we performed an analysis in narrow range pH 4–7 and 6–9, and we found that DPSCs vs PDLSCs express differentially regulated proteins that are potentially related to growth, regulation and genesis of neuronal cells, suggesting that SCs derived from oral tissue source populations may possess the potential ability of neuronal differentiation which is very consistent with their neural crest origin. Conclusion/Significance This study identifies some differentially expressed proteins by using comparative analysis between DPSCs and PDLSCs and BMSCs and suggests that stem cells from oral tissue could have a different cell lineage potency compared to BMSCs.
Collapse
|
19
|
Ma PF, Chen JQ, Wang Z, Liu JL, Li BP. Function of chloride intracellular channel 1 in gastric cancer cells. World J Gastroenterol 2012; 18:3070-3080. [PMID: 22791942 PMCID: PMC3386320 DOI: 10.3748/wjg.v18.i24.3070] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Revised: 02/28/2012] [Accepted: 04/09/2012] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the effect of chloride intracellular channel 1 (CLIC1) on the cell proliferation, apoptosis, migration and invasion of gastric cancer cells. METHODS CLIC1 expression was evaluated in human gastric cancer cell lines SGC-7901 and MGC-803 by real time polymerase chain reaction (RT-PCR). Four segments of small interference RNA (siRNA) targeting CLIC1 mRNA and a no-sense control segment were designed by bioinformatics technology. CLIC1 siRNA was selected using Lipofectamine 2000 and transfected transiently into human gastric cancer SGC-7901 and MGC-803 cells. The transfected efficiency was observed under fluorescence microscope. After transfection, mRNA expression of CLIC1 was detected with RT-PCR and Western blotting was used to detect the protein expression. Proliferation was examined by methyl thiazolyl tetrazolium and apoptosis was detected with flow cytometry. Polycarbonate membrane transwell chamber and Matrigel were used for the detection of the changes of invasion and migration of the two cell lines. RESULTS In gastric cancer cell lines SGC-7901 and MGC-803, CLIC1 was obviously expressed and CLIC1 siRNA could effectively suppress the expression of CLIC1 protein and mRNA. Proliferation of cells transfected with CLIC1 siRNA3 was enhanced notably, and the highest proliferation rate was 23.3% (P = 0.002) in SGC-7901 and 35.55% (P = 0.001) in MGC-803 cells at 48 h. The G2/M phase proportion increased, while G0/G1 and S phase proportions decreased. The apoptotic rate of the CLIC1 siRNA3 group obviously decreased in both SGC-7901 cells (62.24%, P = 0.000) and MGC-803 cells (52.67%, P = 0.004). Down-regulation of CLIC1 led to the inhibition of invasion and migration by 54.31% (P = 0.000) and 33.62% (P = 0.001) in SGC-7901 and 40.74% (P = 0.000) and 29.26% (P = 0.002) in MGC-803. However, there was no significant difference between the mock group cells and the negative control group cells. CONCLUSION High CLIC1 expression can efficiently inhibit proliferation and enhance apoptosis, migration and invasion of gastric cancer cells in vitro. CLIC1 might be a promising target for the treatment of gastric cancer.
Collapse
|
20
|
Id Boufker H, Lagneaux L, Fayyad-Kazan H, Badran B, Najar M, Wiedig M, Ghanem G, Laurent G, Body JJ, Journé F. Role of farnesoid X receptor (FXR) in the process of differentiation of bone marrow stromal cells into osteoblasts. Bone 2011; 49:1219-31. [PMID: 21893226 DOI: 10.1016/j.bone.2011.08.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 08/11/2011] [Accepted: 08/15/2011] [Indexed: 12/16/2022]
Abstract
Bone tissue contains bile acids which accumulate from serum and which can be released in large amounts in the bone microenvironment during bone resorption. However, the direct effects of bile acids on bone cells remain largely unexplored. Bile acids have been identified as physiological ligands of the farnesoid X receptor (FXR, NR1H4). In the present study, we have examined the effects of FXR activation/inhibition on the osteoblastic differentiation of human bone marrow stromal cells (BMSC). We first demonstrated the expression of FXR in BMSC and SaOS2 osteoblast-like cells, and observed that FXR activation by chenodeoxycholic acid (CDCA) or by farnesol (FOH) increases the activity of alkaline phosphatase and the calcification of the extracellular matrix. In addition, we observed that FXR agonists are able to stimulate the expression of osteoblast marker genes [bone sialoprotein (BSP), osteocalcin (OC), osteopontin (OPN) and alkaline phosphatase (ALP)] (FXR involvement validated by shRNA-induced gene silencing), as well as the DNA binding activity of the bone transcription factor RUNX2 (EMSA and ChIP assay). Importantly, we observed that nitrogen-containing bisphosphonates (BPs) inhibit the basal osteoblastic differentiation of BMSC, possibly through suppression of endogenous FOH production, independently of their effects on protein prenylation. Likewise, we found that the FXR antagonist guggulsterone (GGS) inhibits ALP activity, calcium deposition, DNA binding of RUNX2, and bone marker expression, indicating that GGS interferes with osteoblastic differentiation. Furthermore, GGS induced the appearance of lipid vesicles in BMSC and stimulated the expression of adipose tissue markers (peroxisome proliferator activated receptor-gamma (PPARγ), adipoQ, leptin and CCAAT/enhancer-binding protein-alpha (C/EBPα)). In conclusion, our data support a new role for FXR in the modulation of osteoblast/adipocyte balance: its activation stimulates RUNX2-mediated osteoblastic differentiation of BMSC, whereas its inhibition leads to an adipocyte-like phenotype.
Collapse
Affiliation(s)
- Hichame Id Boufker
- Laboratoire d'Hématologie Expérimentale, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
KATP channels in mesenchymal stromal stem cells: strong up-regulation of Kir6.2 subunits upon osteogenic differentiation. Tissue Cell 2011; 43:331-6. [PMID: 21820692 DOI: 10.1016/j.tice.2011.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 05/30/2011] [Accepted: 06/06/2011] [Indexed: 01/13/2023]
Abstract
The promising use of mesenchymal stromal cells (MSC) in regenerative technologies accounts for necessity of detailed study of their physiology. Proliferation and differentiation of multipotent cells often involve changes in their metabolic state. In the present study, we analyzed the expression of ATP-sensitive potassium (K(ATP)) channels in MSC and upon in vitro differentiation. K(ATP) channels are present in many cells and regulate a variety of cellular functions by coupling cell metabolism with membrane potential. Kir6.1, Kir6.2 and SUR2A were expressed in undifferentiated MSC, whereas SUR2B and SUR1 were not detected on cDNA and protein level. Upon adipogenic differentiation Kir6.1 and SUR2A showed a significant reduction of the amount of mRNA by 84% and 95%, respectively, whereas Kir6.2 expression was unchanged. Osteogenic differentiation strongly up-regulated Kir6.2 mRNA (28-fold) whereas Kir6.1 and SUR2A showed no significant change in expression. Quantitative Western blot analysis and immunofluorescence staining confirmed the elevated expression of Kir6.2 upon osteogenic differentiation. Taken together, expression changes of K(ATP) channels may contribute to in vitro differentiation of MSC and represent changes in the metabolic state of the developing tissue.
Collapse
|
22
|
Cui XS, Shen XH, Lee CK, Kang YK, Wakayama T, Kim NH. Analysis of proteomic profiling of mouse embryonic stem cells derived from fertilized, parthenogenetic and androgenetic blastocysts. ACTA ACUST UNITED AC 2011. [DOI: 10.4236/scd.2011.11001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
23
|
Osteogenic role of endosomal chloride channels in MC3T3-E1 cells. Mol Cell Biochem 2010; 342:191-9. [DOI: 10.1007/s11010-010-0483-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Accepted: 05/04/2010] [Indexed: 11/28/2022]
|
24
|
Wang H, Mao Y, Zhang B, Wang T, Li F, Fu S, Xue Y, Yang T, Wen X, Ding Y, Duan X. Chloride channel ClC-3 promotion of osteogenic differentiation through Runx2. J Cell Biochem 2010; 111:49-58. [DOI: 10.1002/jcb.22658] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|