51
|
Li R, Yan Z, Ye J, Huang H, Wang Z, Wei Q, Wang J, Zhao L, Lu S, Wang X, Tang S, Fan J, Zhang F, Zou Y, Song D, Liao J, Lu M, Liu F, Shi LL, Athiviraham A, Lee MJ, He TC, Zhang Z. The Prodomain-Containing BMP9 Produced from a Stable Line Effectively Regulates the Differentiation of Mesenchymal Stem Cells. Int J Med Sci 2016; 13:8-18. [PMID: 26816490 PMCID: PMC4716815 DOI: 10.7150/ijms.13333] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 11/09/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND BMPs play important roles in regulating stem cell proliferation and differentiation. Using adenovirus-mediated expression of the 14 types of BMPs we demonstrated that BMP9 is one of the most potent BMPs in inducing osteogenic differentiation of mesenchymal stem cells (MSCs), which was undetected in the early studies using recombinant BMP9 proteins. Endogenous BMPs are expressed as a precursor protein that contains an N-terminal signal peptide, a prodomain and a C-terminal mature peptide. Most commercially available recombinant BMP9 proteins are purified from the cells expressing the mature peptide. It is unclear how effectively these recombinant BMP9 proteins functionally recapitulate endogenous BMP9. METHODS A stable cell line expressing the full coding region of mouse BMP9 was established in HEK-293 cells by using the piggyBac transposon system. The biological activities and stability of the conditioned medium generated from the stable line were analyzed. RESULTS The stable HEK-293 line expresses a high level of mouse BMP9. BMP9 conditioned medium (BMP9-cm) was shown to effectively induce osteogenic differentiation of MSCs, to activate BMP-R specific Smad signaling, and to up-regulate downstream target genes in MSCs. The biological activity of BMP9-cm is at least comparable with that induced by AdBMP9 in vitro. Furthermore, BMP9-cm exhibits an excellent stability profile as its biological activity is not affected by long-term storage at -80ºC, repeated thawing cycles, and extended storage at 4ºC. CONCLUSIONS We have established a producer line that stably expresses a high level of active BMP9 protein. Such producer line should be a valuable resource for generating biologically active BMP9 protein for studying BMP9 signaling mechanism and functions.
Collapse
Affiliation(s)
- Ruifang Li
- 1. Department of Neurology, Hubei Zhongshan Hospital, Wuhan, China; 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Zhengjian Yan
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Jixing Ye
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 4. Department of Biomedical Engineering, School of Bioengineering, Chongqing University, Chongqing, China
| | - He Huang
- 5. Ben May Department for Cancer Research, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zhongliang Wang
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Qiang Wei
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Lianggong Zhao
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 6. Department of Orthopaedic Surgery, the Second Affiliated Hospital of Lanzhou University, Lanzhou, China
| | - Shun Lu
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 7. Department of Orthopaedic Surgery, Shandong Provincial Hospital and Shandong University School of Medicine, Jinan, China
| | - Xin Wang
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 8. Department of Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Shengli Tang
- 1. Department of Neurology, Hubei Zhongshan Hospital, Wuhan, China; 9. Department of General Surgery, the Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiaming Fan
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Fugui Zhang
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Yulong Zou
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Dongzhe Song
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 8. Department of Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Junyi Liao
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Minpeng Lu
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Feng Liu
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 3. Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Lewis L Shi
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Aravind Athiviraham
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Michael J Lee
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Tong-Chuan He
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Zhonglin Zhang
- 2. Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA; 9. Department of General Surgery, the Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
52
|
Common mutations in ALK2/ACVR1, a multi-faceted receptor, have roles in distinct pediatric musculoskeletal and neural orphan disorders. Cytokine Growth Factor Rev 2015; 27:93-104. [PMID: 26776312 DOI: 10.1016/j.cytogfr.2015.12.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Activin receptor-like kinase-2 (ALK2), the product of ACVR1, is a member of the type I bone morphogenetic protein (BMP) receptor family. ALK2 exerts key and non-redundant roles in numerous developmental processes, including the specification, growth and morphogenesis of endochondral skeletal elements. There is also strong evidence that BMP signaling plays important roles in determination, differentiation and function of neural cells and tissues. Here we focus on the intriguing discovery that common activating mutations in ALK2 occur in Fibrodysplasia Ossificans Progressiva (FOP) and Diffuse Intrinsic Pontine Gliomas (DIPGs), distinct pediatric disorders of significant severity that are associated with premature death. Pathogenesis and treatment remain elusive for both. We consider recent studies on the nature of the ACVR1 mutations, possible modes of action and targets, and plausible therapeutic measures. Comparisons of the diverse - but genetically interrelated - pathologies of FOP and DIPG will continue to be of major mutual benefit with broad biomedical and clinical relevance.
Collapse
|
53
|
Kienast Y, Jucknischke U, Scheiblich S, Thier M, de Wouters M, Haas A, Lehmann C, Brand V, Bernicke D, Honold K, Lorenz S. Rapid Activation of Bone Morphogenic Protein 9 by Receptor-mediated Displacement of Pro-domains. J Biol Chem 2015; 291:3395-410. [PMID: 26677222 DOI: 10.1074/jbc.m115.680009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Indexed: 12/21/2022] Open
Abstract
By non-covalent association after proteolytic cleavage, the pro-domains modulate the activities of the mature growth factor domains across the transforming growth factor-β family. In the case of bone morphogenic protein 9 (BMP9), however, the pro-domains do not inhibit the bioactivity of the growth factor, and the BMP9·pro-domain complexes have equivalent biological activities as the BMP9 mature ligand dimers. By using real-time surface plasmon resonance, we could demonstrate that either binding of pro-domain-complexed BMP9 to type I receptor activin receptor-like kinase 1 (ALK1), type II receptors, co-receptor endoglin, or to mature BMP9 domain targeting antibodies leads to immediate and complete displacement of the pro-domains from the complex. Vice versa, pro-domain binding by an anti-pro-domain antibody results in release of the mature BMP9 growth factor. Based on these findings, we adjusted ELISA assays to measure the protein levels of different BMP9 variants. Although mature BMP9 and inactive precursor BMP9 protein were directly detectable by ELISA, BMP9·pro-domain complex could only be measured indirectly as dissociated fragments due to displacement of mature growth factor and pro-domains after antibody binding. Our studies provide a model in which BMP9 can be readily activated upon getting into contact with its receptors. This increases the understanding of the underlying biology of BMP9 activation and also provides guidance for ELISA development for the detection of circulating BMP9 variants.
Collapse
Affiliation(s)
- Yvonne Kienast
- From the Roche Pharma Research and Early Development (pRED), Discovery Oncology, Roche Innovation Center Penzberg, 82377 Penzberg, Germany,
| | - Ute Jucknischke
- From the Roche Pharma Research and Early Development (pRED), Discovery Oncology, Roche Innovation Center Penzberg, 82377 Penzberg, Germany
| | - Stefan Scheiblich
- From the Roche Pharma Research and Early Development (pRED), Discovery Oncology, Roche Innovation Center Penzberg, 82377 Penzberg, Germany
| | - Martina Thier
- From the Roche Pharma Research and Early Development (pRED), Translational Technologies and Bioinformatics, Roche Innovation Center, Basel, 4070 Basel, Switzerland, and
| | - Mariana de Wouters
- From the Roche Pharma Research and Early Development (pRED), Translational Technologies and Bioinformatics, Roche Innovation Center, Basel, 4070 Basel, Switzerland, and
| | - Alexander Haas
- From the Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Penzberg, 82377 Penzberg Germany
| | - Christian Lehmann
- From the Roche Pharma Research and Early Development (pRED), Discovery Oncology, Roche Innovation Center Penzberg, 82377 Penzberg, Germany
| | - Verena Brand
- From the Roche Pharma Research and Early Development (pRED)
| | - Dirk Bernicke
- From the Roche Pharma Research and Early Development (pRED), Discovery Oncology, Roche Innovation Center Penzberg, 82377 Penzberg, Germany
| | - Konrad Honold
- From the Roche Pharma Research and Early Development (pRED), Discovery Oncology, Roche Innovation Center Penzberg, 82377 Penzberg, Germany
| | - Stefan Lorenz
- From the Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Penzberg, 82377 Penzberg Germany
| |
Collapse
|
54
|
Borodinsky LN, Belgacem YH. Crosstalk among electrical activity, trophic factors and morphogenetic proteins in the regulation of neurotransmitter phenotype specification. J Chem Neuroanat 2015; 73:3-8. [PMID: 26686293 DOI: 10.1016/j.jchemneu.2015.12.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 11/29/2015] [Accepted: 12/02/2015] [Indexed: 01/11/2023]
Abstract
Morphogenetic proteins are responsible for patterning the embryonic nervous system by enabling cell proliferation that will populate all the neural structures and by specifying neural progenitors that imprint different identities in differentiating neurons. The adoption of specific neurotransmitter phenotypes is crucial for the progression of neuronal differentiation, enabling neurons to connect with each other and with target tissues. Preliminary neurotransmitter specification originates from morphogen-driven neural progenitor specification through the combinatorial expression of transcription factors according to morphogen concentration gradients, which progressively restrict the identity that born neurons adopt. However, neurotransmitter phenotype is not immutable, instead trophic factors released from target tissues and environmental stimuli change expression of neurotransmitter-synthesizing enzymes and specific vesicular transporters modifying neuronal neurotransmitter identity. Here we review studies identifying the mechanisms of catecholaminergic, GABAergic, glutamatergic, cholinergic and serotonergic early specification and of the plasticity of these neurotransmitter phenotypes during development and in the adult nervous system. The emergence of spontaneous electrical activity in developing neurons recruits morphogenetic proteins in the process of neurotransmitter phenotype plasticity, which ultimately equips the nervous system and the whole organism with adaptability for optimal performance in a changing environment.
Collapse
Affiliation(s)
- Laura N Borodinsky
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, 2425 Stockton Blvd, Sacramento, CA 95817, United States.
| | - Yesser H Belgacem
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, 2425 Stockton Blvd, Sacramento, CA 95817, United States
| |
Collapse
|
55
|
Yue W, Li Y, Zhang T, Jiang M, Qian Y, Zhang M, Sheng N, Feng S, Tang K, Yu X, Shu Y, Yue C, Jing N. ESC-Derived Basal Forebrain Cholinergic Neurons Ameliorate the Cognitive Symptoms Associated with Alzheimer's Disease in Mouse Models. Stem Cell Reports 2015; 5:776-790. [PMID: 26489896 PMCID: PMC4649256 DOI: 10.1016/j.stemcr.2015.09.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 09/11/2015] [Accepted: 09/11/2015] [Indexed: 12/16/2022] Open
Abstract
Degeneration of basal forebrain cholinergic neurons (BFCNs) is associated with cognitive impairments of Alzheimer’s disease (AD), implying that BFCNs hold potentials in exploring stem cell-based replacement therapy for AD. However, studies on derivation of BFCNs from embryonic stem cells (ESCs) are limited, and the application of ESC-derived BFCNs remains to be determined. Here, we report on differentiation approaches for directing both mouse and human ESCs into mature BFCNs. These ESC-derived BFCNs exhibit features similar to those of their in vivo counterparts and acquire appropriate functional properties. After transplantation into the basal forebrain of AD model mice, ESC-derived BFCN progenitors predominantly differentiate into mature cholinergic neurons that functionally integrate into the endogenous basal forebrain cholinergic projection system. The AD mice grafted with mouse or human BFCNs exhibit improvements in learning and memory performances. Our findings suggest a promising perspective of ESC-derived BFCNs in the development of stem cell-based therapies for treatment of AD. Mouse and human ESCs differentiate into basal forebrain cholinergic neurons (BFCNs) ESC-derived BFCNs exhibit functional properties in vitro and transplanted in vivo ESC-derived BFCNs functionally integrate into the basal forebrain of AD mice The AD model mice grafted with BFCNs exhibit improvements in cognitive abilities
Collapse
Affiliation(s)
- Wei Yue
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuanyuan Li
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ting Zhang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Man Jiang
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yun Qian
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Min Zhang
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Nengyin Sheng
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Su Feng
- Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Ke Tang
- Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Xiang Yu
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yousheng Shu
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chunmei Yue
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Naihe Jing
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
56
|
González-Núñez M, Riolobos AS, Castellano O, Fuentes-Calvo I, de los Ángeles Sevilla M, Oujo B, Pericacho M, Cruz-Gonzalez I, Pérez-Barriocanal F, ten Dijke P, López-Novoa JM. Heterozygous disruption of activin receptor-like kinase 1 is associated with increased arterial pressure in mice. Dis Model Mech 2015; 8:1427-39. [PMID: 26398936 PMCID: PMC4631783 DOI: 10.1242/dmm.019695] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 08/27/2015] [Indexed: 12/20/2022] Open
Abstract
The activin receptor-like kinase 1 (ALK-1) is a type I cell-surface receptor for the transforming growth factor-β (TGF-β) family of proteins. Hypertension is related to TGF-β1, because increased TGF-β1 expression is correlated with an elevation in arterial pressure (AP) and TGF-β expression is upregulated by the renin-angiotensin-aldosterone system. The purpose of this study was to assess the role of ALK-1 in regulation of AP using Alk1 haploinsufficient mice (Alk1(+/-)). We observed that systolic and diastolic AP were significantly higher in Alk1(+/-) than in Alk1(+/+) mice, and all functional and structural cardiac parameters (echocardiography and electrocardiography) were similar in both groups. Alk1(+/-) mice showed alterations in the circadian rhythm of AP, with higher AP than Alk1(+/+) mice during most of the light period. Higher AP in Alk1(+/-) mice is not a result of a reduction in the NO-dependent vasodilator response or of overactivation of the peripheral renin-angiotensin system. However, intracerebroventricular administration of losartan had a hypotensive effect in Alk1(+/-) and not in Alk1(+/+) mice. Alk1(+/-) mice showed a greater hypotensive response to the β-adrenergic antagonist atenolol and higher concentrations of epinephrine and norepinephrine in plasma than Alk1(+/+) mice. The number of brain cholinergic neurons in the anterior basal forebrain was reduced in Alk1(+/-) mice. Thus, we concluded that the ALK-1 receptor is involved in the control of AP, and the high AP of Alk1(+/-) mice is explained mainly by the sympathetic overactivation shown by these animals, which is probably related to the decreased number of cholinergic neurons.
Collapse
Affiliation(s)
- María González-Núñez
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Unidad de Fisiopatología Renal y Cardiovascular, Instituto 'Reina Sofía' de Investigación Nefrológica, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain
| | - Adela S Riolobos
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain Instituto de Neurociencias de Castilla y León (INCYL), Salamanca 37008, Spain
| | - Orlando Castellano
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain Instituto de Neurociencias de Castilla y León (INCYL), Salamanca 37008, Spain
| | - Isabel Fuentes-Calvo
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Unidad de Fisiopatología Renal y Cardiovascular, Instituto 'Reina Sofía' de Investigación Nefrológica, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain
| | | | - Bárbara Oujo
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Unidad de Fisiopatología Renal y Cardiovascular, Instituto 'Reina Sofía' de Investigación Nefrológica, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain
| | - Miguel Pericacho
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Unidad de Fisiopatología Renal y Cardiovascular, Instituto 'Reina Sofía' de Investigación Nefrológica, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain
| | - Ignacio Cruz-Gonzalez
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain Departamento de Cardiología, Hospital Universitario de Salamanca, Salamanca 37007, Spain
| | - Fernando Pérez-Barriocanal
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Unidad de Fisiopatología Renal y Cardiovascular, Instituto 'Reina Sofía' de Investigación Nefrológica, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain
| | - Peter ten Dijke
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Jose M López-Novoa
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Unidad de Fisiopatología Renal y Cardiovascular, Instituto 'Reina Sofía' de Investigación Nefrológica, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain
| |
Collapse
|
57
|
Modeling the Interaction between β-Amyloid Aggregates and Choline Acetyltransferase Activity and Its Relation with Cholinergic Dysfunction through Two-Enzyme/Two-Compartment Model. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2015; 2015:923762. [PMID: 26413144 PMCID: PMC4568057 DOI: 10.1155/2015/923762] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 07/14/2015] [Indexed: 11/17/2022]
Abstract
The effect of β-amyloid aggregates on activity of choline acetyltransferase (ChAT) which is responsible for synthesizing acetylcholine (ACh) in human brain is investigated through the two-enzyme/two-compartment (2E2C) model where the presynaptic neuron is considered as compartment 1 while both the synaptic cleft and the postsynaptic neuron are considered as compartment 2 through suggesting three different kinetic mechanisms for the inhibition effect. It is found that the incorporation of ChAT inhibition by β-amyloid aggregates into the 2E2C model is able to yield dynamic solutions for concentrations of generated β-amyloid, ACh, choline, acetate, and pH in addition to the rates of ACh synthesis and ACh hydrolysis in compartments 1 and 2. It is observed that ChAT activity needs a high concentration of β-amyloid aggregates production rate. It is found that ChAT activity is reduced significantly when neurons are exposed to high levels of β-amyloid aggregates leading to reduction in levels of ACh which is one of the most significant physiological symptoms of AD. Furthermore, the system of ACh neurocycle is dominated by the oscillatory behavior when ChAT enzyme is completely inhibited by β-amyloid. It is observed that the direct inactivation of ChAT by β-amyloid aggregates may be a probable mechanism contributing to the development of AD.
Collapse
|
58
|
Li L, Dong Q, Wang Y, Feng Q, Zhou P, Ou X, Meng Q, He T, Luo J. Hedgehog signaling is involved in the BMP9-induced osteogenic differentiation of mesenchymal stem cells. Int J Mol Med 2015; 35:1641-50. [PMID: 25872645 DOI: 10.3892/ijmm.2015.2172] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 03/30/2015] [Indexed: 11/05/2022] Open
Abstract
Nonunion is a serious complication of a bone fracture that may occur in any bone of the skeletal system. It occurs when a broken bone fails to heal. Mesenchymal stem cell (MSC)-based tissue engineering technology has been considered an efficient method to improve the healing rate of nonunions. Although previous studies have demonstrated that bone morphogenetic protein 9 (BMP9) is highly capable of promoting the osteogenic differentiation of MSCs, the mechanisms involved remain largely unclear. In the present study, we investigated the possible involvement and the detailed role of Hedgehog (Hh) signaling in the BMP9-induced osteogenic differentiation of MSCs. It was found that BMP9 exerts an effect on Hh signaling in MSCs. The expression levels of early markers of BMP9-induced osteogenic differentiation, such as alkaline phosphatase (ALP) activity, and late markers of osteogenic differentiation, such as matrix mineralization, as well as the expression levels of osteopontin (OPN) and osteocalcin (OCN) were decreased by the Hh signaling inhibitor, cyclopamine, whereas these levels were increased by the Hh signaling agonist, purmorphamine. Furthermore, the BMP9-induced transcriptional activity of Smad1/5/8 and the expression of pivotal osteogenic transcription factors were reduced by cyclopamine, and were increased by purmorphamine. Taken together, our results demonstrate that BMP9 exerts an effect on Hh signaling in MSCs. What is most noteworthy, however, is that the inhibition or enhancement of Hh signaling resulted in the reduction and augmentation of the BMP9-induced osteogenic differentiation of MSCs, respectively, suggesting that Hh signaling is involved and plays a regulatory role in the osteogenic differentiation of MSCs induced by BMP9.
Collapse
Affiliation(s)
- Li Li
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qian Dong
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yufeng Wang
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qiaoling Feng
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Pengfei Zhou
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing 401147, P.R. China
| | - Xinying Ou
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qiurong Meng
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Tongchuan He
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jinyong Luo
- Key Laboratory of Diagnostic Medicine Designated by The Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
59
|
Nanoparticle-mediated growth factor delivery systems: A new way to treat Alzheimer's disease. J Control Release 2015; 206:187-205. [DOI: 10.1016/j.jconrel.2015.03.024] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/19/2015] [Accepted: 03/20/2015] [Indexed: 01/03/2023]
|
60
|
Lauzon MA, Daviau A, Marcos B, Faucheux N. Growth factor treatment to overcome Alzheimer's dysfunctional signaling. Cell Signal 2015; 27:1025-38. [PMID: 25744541 DOI: 10.1016/j.cellsig.2015.02.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 02/16/2015] [Indexed: 10/23/2022]
Abstract
The number of people suffering from Alzheimer's disease (AD) will increase as the world population ages, creating a huge socio-economic burden. The three pathophysiological hallmarks of AD are the cholinergic system dysfunction, the β-amyloid peptide deposition and the Tau protein hyperphosphorylation. Current treatments have only transient effects and each tends to concentrate on a single pathophysiological aspect of AD. This review first provides an overall view of AD in terms of its pathophysiological symptoms and signaling dysfunction. We then examine the therapeutic potential of growth factors (GFs) by showing how they can overcome the dysfunctional cell signaling that occurs in AD. Finally, we discuss new alternatives to GFs that help overcome the problem of brain uptake, such as small peptides, with evidence from some of our unpublished data on human neuronal cell line.
Collapse
Affiliation(s)
- Marc-Antoine Lauzon
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Alex Daviau
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Bernard Marcos
- Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Nathalie Faucheux
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada.
| |
Collapse
|
61
|
Zhu D, Mackenzie NCW, Shanahan CM, Shroff RC, Farquharson C, MacRae VE. BMP-9 regulates the osteoblastic differentiation and calcification of vascular smooth muscle cells through an ALK1 mediated pathway. J Cell Mol Med 2014; 19:165-74. [PMID: 25297851 PMCID: PMC4288360 DOI: 10.1111/jcmm.12373] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 06/13/2014] [Indexed: 01/24/2023] Open
Abstract
The process of vascular calcification shares many similarities with that of physiological skeletal mineralization, and involves the deposition of hydroxyapatite crystals in arteries. However, the cellular mechanisms responsible have yet to be fully explained. Bone morphogenetic protein (BMP-9) has been shown to exert direct effects on both bone development and vascular function. In the present study, we have investigated the role of BMP-9 in vascular smooth muscle cell (VSMC) calcification. Vessel calcification in chronic kidney disease (CKD) begins pre-dialysis, with factors specific to the dialysis milieu triggering accelerated calcification. Intriguingly, BMP-9 was markedly elevated in serum from CKD children on dialysis. Furthermore, in vitro studies revealed that BMP-9 treatment causes a significant increase in VSMC calcium content, alkaline phosphatase (ALP) activity and mRNA expression of osteogenic markers. BMP-9-induced calcium deposition was significantly reduced following treatment with the ALP inhibitor 2,5-Dimethoxy-N-(quinolin-3-yl) benzenesulfonamide confirming the mediatory role of ALP in this process. The inhibition of ALK1 signalling using a soluble chimeric protein significantly reduced calcium deposition and ALP activity, confirming that BMP-9 is a physiological ALK1 ligand. Signal transduction studies revealed that BMP-9 induced Smad2, Smad3 and Smad1/5/8 phosphorylation. As these Smad proteins directly bind to Smad4 to activate target genes, siRNA studies were subsequently undertaken to examine the functional role of Smad4 in VSMC calcification. Smad4-siRNA transfection induced a significant reduction in ALP activity and calcium deposition. These novel data demonstrate that BMP-9 induces VSMC osteogenic differentiation and calcification via ALK1, Smad and ALP dependent mechanisms. This may identify new potential therapeutic strategies for clinical intervention.
Collapse
Affiliation(s)
- Dongxing Zhu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | | | | | | | | | | |
Collapse
|
62
|
Wei Z, Salmon RM, Upton PD, Morrell NW, Li W. Regulation of bone morphogenetic protein 9 (BMP9) by redox-dependent proteolysis. J Biol Chem 2014; 289:31150-9. [PMID: 25237187 PMCID: PMC4223318 DOI: 10.1074/jbc.m114.579771] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BMP9, a member of the TGFβ superfamily, is a homodimer that forms a signaling complex with two type I and two type II receptors. Signaling through high-affinity activin receptor-like kinase 1 (ALK1) in endothelial cells, circulating BMP9 acts as a vascular quiescence factor, maintaining endothelial homeostasis. BMP9 is also the most potent BMP for inducing osteogenic signaling in mesenchymal stem cells in vitro and promoting bone formation in vivo. This activity requires ALK1, the lower affinity type I receptor ALK2, and higher concentrations of BMP9. In adults, BMP9 is constitutively expressed in hepatocytes and secreted into the circulation. Optimum concentrations of BMP9 are essential to maintain the highly specific endothelial-protective function. Factors regulating BMP9 stability and activity remain unknown. Here, we showed by chromatography and a 1.9 Å crystal structure that stable BMP9 dimers could form either with (D-form) or without (M-form) an intermolecular disulfide bond. Although both forms of BMP9 were capable of binding to the prodomain and ALK1, the M-form demonstrated less sustained induction of Smad1/5/8 phosphorylation. The two forms could be converted into each other by changing the redox potential, and this redox switch caused a major alteration in BMP9 stability. The M-form displayed greater susceptibility to redox-dependent cleavage by proteases present in serum. This study provides a mechanism for the regulation of circulating BMP9 concentrations and may provide new rationales for approaches to modify BMP9 levels for therapeutic purposes.
Collapse
Affiliation(s)
- Zhenquan Wei
- From the Department of Medicine, University of Cambridge, School of Clinical Medicine, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Richard M Salmon
- From the Department of Medicine, University of Cambridge, School of Clinical Medicine, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Paul D Upton
- From the Department of Medicine, University of Cambridge, School of Clinical Medicine, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Nicholas W Morrell
- From the Department of Medicine, University of Cambridge, School of Clinical Medicine, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Wei Li
- From the Department of Medicine, University of Cambridge, School of Clinical Medicine, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
63
|
Hu Y, Liang J, Cui H, Wang X, Rong H, Shao B, Cui H. Wharton's jelly mesenchymal stem cells differentiate into retinal progenitor cells. Neural Regen Res 2014; 8:1783-92. [PMID: 25206475 PMCID: PMC4145957 DOI: 10.3969/j.issn.1673-5374.2013.19.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Accepted: 05/05/2013] [Indexed: 01/09/2023] Open
Abstract
Human Wharton's jelly mesenchymal stem cells were isolated from fetal umbilical cord. Cells were cultured in serum-free neural stem cell-conditioned medium or neural stem cell-conditioned medium supplemented with Dkk-1, a Wnt/β catenin pathway antagonist, and LeftyA, a Nodal signaling pathway antagonist to induce differentiation into retinal progenitor cells. Inverted microscopy showed that after induction, the spindle-shaped or fibroblast-like Wharton's jelly mesenchymal stem cells changed into bulbous cells with numerous processes. Immunofluorescent cytochemical ing and reverse-transcription PCR showed positive expression of retinal progenitor cell markers, Pax6 and Rx, as well as weakly down-regulated nestin expression. These results demonstrate that Wharton's jelly mesenchymal stem cells are capable of differentiating into retinal progenitor cells in vitro.
Collapse
Affiliation(s)
- Ying Hu
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University, Harbin 200120, Heilongjiang Province, China ; Department of Ophthalmology, Shanghai East Hospital Affiliated to Tongji University, Shanghai 200120, China
| | - Jun Liang
- Department of Histology and Embryology, Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang Province, China
| | - Hongping Cui
- Department of Ophthalmology, Shanghai East Hospital Affiliated to Tongji University, Shanghai 200120, China
| | - Xinmei Wang
- Department of Ophthalmology, Fourth Affiliated Hospital, Harbin Medical University, Harbin 200120, Heilongjiang Province, China
| | - Hua Rong
- Department of Ophthalmology, Shanghai East Hospital Affiliated to Tongji University, Shanghai 200120, China
| | - Bin Shao
- Department of Head-Neck and Breast Tumor, Mudanjiang Tumor Hospital, Mudanjiang 157009, Heilongjiang Province, China
| | - Hao Cui
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University, Harbin 200120, Heilongjiang Province, China
| |
Collapse
|
64
|
Luan H, Yang L, Liu L, Liu S, Zhao X, Sui H, Wang J, Wang S. Effects of platycodins on liver complications of type 2 diabetes. Mol Med Rep 2014; 10:1597-603. [PMID: 25017203 DOI: 10.3892/mmr.2014.2363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 01/23/2014] [Indexed: 11/06/2022] Open
Abstract
The aim of the current study was to investigate the therapeutic effects and mechanism of platycodin in liver complications of type 2 diabetes. All rats were randomly divided into two groups: The control group (normal diet) and the model group (a high‑fat and high‑sugar diet). The model group was injected with 2% streptozocin (25 mg/kg body weight) through the tail vein following 4 weeks of dieting. After a total of 8 weeks of dieting, fasting blood glucose (FBG) and liver function were examined. The high‑fat and high‑sugar diet was continued in the successful model rats, which were randomly divided into four groups and treated with the following doses of platycodins: The untreated, and 50, 100 and 200 mg/kg body weight/day groups. Platycodins treatment lasted for 12 weeks. Platycodins treatment at a dose of 200 mg/kg body weight/day reduced the FBG, glutamate pyruvate transaminase (GPT), glutamic oxalacetic transaminase, triglycerides, total cholesterol (TC), low‑density lipoprotein (LDL) and liver index levels compared with the untreated group (P<0.05), while the high‑density lipoprotein levels increased (P<0.05). Furthermore, FBG, GPT, TC and LDL levels were returned to the normal level. This dose also increased the expression of BMP‑9 mRNA and BMP‑9 protein, and reduced the expression of Smad‑4 mRNA and Smad‑4 protein. These findings indicate that platycodins can rectify disorders of blood glucose and lipid metabolism, improve liver index and protect liver function in liver complications of type 2 diabetes. The current study suggests that this therapeutic effect is mediated through the BMP‑9/Smad‑4 pathway.
Collapse
Affiliation(s)
- Haiyan Luan
- Department of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Limin Yang
- Department of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Lei Liu
- Department of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Shuang Liu
- Department of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Xiaolian Zhao
- Department of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Hongyu Sui
- Department of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Jingtao Wang
- Department of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Shuqiu Wang
- Department of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| |
Collapse
|
65
|
Mellott TJ, Pender SM, Burke RM, Langley EA, Blusztajn JK. IGF2 ameliorates amyloidosis, increases cholinergic marker expression and raises BMP9 and neurotrophin levels in the hippocampus of the APPswePS1dE9 Alzheimer's disease model mice. PLoS One 2014; 9:e94287. [PMID: 24732467 PMCID: PMC3986048 DOI: 10.1371/journal.pone.0094287] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 03/12/2014] [Indexed: 02/07/2023] Open
Abstract
The development of an effective therapy for Alzheimer's disease (AD) is a major challenge to biomedical sciences. Because much of early AD pathophysiology includes hippocampal abnormalities, a viable treatment strategy might be to use trophic factors that support hippocampal integrity and function. IGF2 is an attractive candidate as it acts in the hippocampus to enhance memory consolidation, stimulate adult neurogenesis and upregulate cholinergic marker expression and acetylcholine (ACh) release. We performed a seven-day intracerebroventricular infusion of IGF2 in transgenic APPswe.PS1dE9 AD model mice that express green fluorescent protein in cholinergic neurons (APP.PS1/CHGFP) and in wild type WT/CHGFP littermates at 6 months of age representing early AD-like disease. IGF2 reduced the number of hippocampal Aβ40- and Aβ42-positive amyloid plaques in APP.PS1/CHGFP mice. Moreover, IGF2 increased hippocampal protein levels of the ACh-synthesizing enzyme, choline acetyltransferase in both WT/CHGFP and APP.PS1/CHGFP mice. The latter effect was likely mediated by increased protein expression of the cholinergic differentiating factor, BMP9, observed in IGF2-treated mice as compared to controls. IGF2 also increased the protein levels of hippocampal NGF, BDNF, NT3 and IGF1 and of doublecortin, a marker of neurogenesis. These data show that IGF2 administration is effective in reversing and preventing several pathophysiologic processes associated with AD and suggest that IGF2 may constitute a therapeutic target for AD.
Collapse
Affiliation(s)
- Tiffany J. Mellott
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Sarah M. Pender
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Rebecca M. Burke
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Erika A. Langley
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jan Krzysztof Blusztajn
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
66
|
Zhao YF, Xu J, Wang WJ, Wang J, He JW, Li L, Dong Q, Xiao Y, Duan XL, Yang X, Liang YW, Song T, Tang M, Zhao D, Luo JY. Activation of JNKs is essential for BMP9-induced osteogenic differentiation of mesenchymal stem cells. BMB Rep 2014; 46:422-7. [PMID: 23977991 PMCID: PMC4133909 DOI: 10.5483/bmbrep.2013.46.8.266] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although BMP9 is highly capable of promoting osteogenic differentiation of mesenchymal stem cell (MSCs), the molecular mechanism involved remains to be fully elucidated. Here, we explore the possible involvement and detail role of JNKs (c-Jun N-terminal kinases) in BMP9-induced osteogenic differentiation of MSCs. It was found that BMP9 stimulated the activation of JNKs in MSCs. BMP9-induced osteogenic differentiation of MSCs was dramatically inhibited by JNKs inhibitor SP600125. Moreover, BMP9-activated Smads signaling was decreased by SP600125 treatment in MSCs. The effects of inhibitor are reproduced with adenoviruses expressing siRNA targeted JNKs. Taken together, our results revealed that JNKs was activated in BMP9-induced osteogenic differentiation of MSCs. What is most noteworthy, however, is that inhibition of JNKs activity resulted in reduction of BMP9-induced osteogenic differentiation of MSCs, implying that activation of JNKs is essential for BMP9 osteoinductive activity. [BMB Reports 2013; 46(8):422-427]
Collapse
Affiliation(s)
- Yan-fang Zhao
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Kuo MMC, Nguyen PH, Jeon YH, Kim S, Yoon SM, Choe S. MB109 as bioactive human bone morphogenetic protein-9 refolded and purified from E. coli inclusion bodies. Microb Cell Fact 2014; 13:29. [PMID: 24559319 PMCID: PMC3936849 DOI: 10.1186/1475-2859-13-29] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/14/2014] [Indexed: 12/25/2022] Open
Abstract
Background The development of chemical refolding of transforming growth factor-beta (TGF-β) superfamily ligands has been instrumental to produce the recombinant proteins for biochemical studies and exploring the potential of protein therapeutics. The osteogenic human bone morphogenetic protein-2 (hBMP-2) and its Drosophila DPP homolog were the early successful cases of refolding into functional form. Despite the similarity in their three dimensional structure and amino acid sequences, several other TGF-β superfamily ligands could not be refolded readily by the same methods. Results Here, we report a comprehensive study on the variables of a rapid-dilution refolding method, including the concentrations of protein, salt, detergent and redox agents, pH, refolding duration and the presence of aggregation suppressors and host-cell contaminants, in order to identify the optimal condition to refold human BMP-9 (hBMP-9). To produce a recombinant form of hBMP-9 in E. coli cells, a synthetic codon-optimized gene was designed to encode the mature domain of hBMP-9 (Ser320 – Arg429) directly behind the first methionine, which we herein referred to as MB109. An effective purification scheme was also developed to purify the refolded MB109 to homogeneity with a final yield of 7.8 mg from 100 mg of chromatography-purified inclusion bodies as a starting material. The chemically refolded MB109 binds to ALK1, ActRIIb and BMPRII receptors with relatively high affinity as compared to other Type I and Type II receptors based on surface plasmon resonance analysis. Smad1-dependent luciferase assay in C2C12 cells shows that the MB109 has an EC50 of 0.61 ng/mL (25 pM), which is nearly the same as hBMP-9. Conclusion MB109 is prone to be refolded as non-functional dimer and higher order multimers in most of the conditions tested, but bioactive MB109 dimer can be refolded with high efficiency in a narrow window, which is strongly dependent on the pH, refolding duration, the presence of aggregation suppressors and the concentrations of protein, salt and detegent. These results add to the current understanding of producing recombinant TGF-β superfamily ligands in the microbial E. coli system. An application of the technique to produce a large number of synthetic TGF-β chimeras for activity screen is also discussed.
Collapse
Affiliation(s)
- Mario Meng-Chiang Kuo
- Protein Engineering Laboratory, joint Center for Biosciences, Songdo Smart Valley, 214 Sondgo-dong, Yeonsu-gu, Incheon 406-840, Korea.
| | | | | | | | | | | |
Collapse
|
68
|
Manolakos E, Vetro A, Garas A, Thomaidis L, Kefalas K, Kitsos G, Ziegler M, Liehr T, Zuffardi O, Papoulidis I. Proximal 10q duplication in a child with severe central hypotonia characterized by array-comparative genomic hybridization: A case report and review of the literature. Exp Ther Med 2014; 7:953-957. [PMID: 24669257 PMCID: PMC3964923 DOI: 10.3892/etm.2014.1520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 01/08/2014] [Indexed: 11/05/2022] Open
Abstract
Proximal 10q duplication is a well-defined but rare genetic syndrome. Duplication of proximal segments of the long arm of chromosome 10 results in a pattern of malformations, which are distinct from those of the more common distal 10q trisomy syndrome. The present study describes the case of a boy with phenotypic abnormalities (severe central hypotonia, mild ataxia, moderate developmental delay and mild dysmorphic features), due to duplication of chromosome region, 10q11.21→q11.22, which was characterized by the array-comparative genomic hybridization (CGH) technique. The phenotypic findings were compared with those in eight additional similar published cases. Major similarities have emerged, suggesting a likely minimal critical region. However, only detailed characterization of additional cases may provide firm conclusions.
Collapse
Affiliation(s)
| | - Annalisa Vetro
- Department of Human and Hereditary Pathology, University of Pavia, Pavia 27100, Italy
| | - Antonios Garas
- Department of Gynecology, Larissa Medical School, University of Thessaly, Larissa 41335, Greece
| | - Loretta Thomaidis
- Developmental Assessment Unit, Second Department of Paediatrics, P&A Kyriakou Children's Hospital, University of Athens, Athens 11527, Greece
| | | | - George Kitsos
- Department of Ophthalmology, University of Ioannina, Ioannina 45110, Greece
| | - Monika Ziegler
- Institute of Human Genetics and Anthropology, Jena University Hospital, Jena D-07743, Germany
| | - Thomas Liehr
- Institute of Human Genetics and Anthropology, Jena University Hospital, Jena D-07743, Germany
| | - Orsetta Zuffardi
- Department of Human and Hereditary Pathology, University of Pavia, Pavia 27100, Italy
| | | |
Collapse
|
69
|
Ren W, Sun X, Wang K, Feng H, Liu Y, Fei C, Wan S, Wang W, Luo J, Shi Q, Tang M, Zuo G, Weng Y, He T, Zhang Y. BMP9 inhibits the bone metastasis of breast cancer cells by downregulating CCN2 (connective tissue growth factor, CTGF) expression. Mol Biol Rep 2014; 41:1373-83. [PMID: 24413988 DOI: 10.1007/s11033-013-2982-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 12/24/2013] [Indexed: 01/14/2023]
Abstract
Bone morphogenetic proteins (BMPs), which belong to the transforming growth factor-β superfamily, regulate a wide range of cellular responses including cell proliferation, differentiation, adhesion, migration, and apoptosis. BMP9, the latest BMP to be discovered, is reportedly expressed in a variety of human carcinoma cell lines, but the role of BMP9 in breast cancer has not been fully clarified. In a previous study, BMP9 was found to inhibit the growth, migration, and invasiveness of MDA-MB-231 breast cancer cells. In the current study, the effect of BMP9 on the bone metastasis of breast cancer cells was investigated. After absent or low expression of BMP9 was detected in the MDA-MB-231 breast cancer cells and breast non-tumor adjacent tissues using Western blot and immunohistochemistry, In our previous study, BMP9 could inhibit the proliferation and invasiveness of breast cancer cells MDA-MB-231 in vitro and in vivo. This paper shows that BMP9 inhibit the bone metastasis of breast cancer cells by activating the BMP/Smad signaling pathway and downregulating connective tissue growth factor (CTGF); however, when CTGF expression was maintained, the inhibitory effect of BMP9 on the MDA-MB-231 cells was abolished. Together, these observations indicate that BMP9 is an important mediator of breast cancer bone metastasis and a potential therapeutic target for treating this deadly disease.
Collapse
Affiliation(s)
- Wei Ren
- Department of General Surgery, The First Affiliated Hospitals of Chongqing Medical University, Chongqing, 400016, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
BMP9 ameliorates amyloidosis and the cholinergic defect in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 2013; 110:19567-72. [PMID: 24218590 DOI: 10.1073/pnas.1319297110] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Bone morphogenetic protein 9 (BMP9) promotes the acquisition of the cholinergic phenotype in basal forebrain cholinergic neurons (BFCN) during development and protects these neurons from cholinergic dedifferentiation following axotomy when administered in vivo. A decline in BFCN function occurs in patients with Alzheimer's disease (AD) and contributes to the AD-associated memory deficits. We infused BMP9 intracerebroventricularly for 7 d in transgenic AD model mice expressing green fluorescent protein specifically in cholinergic neurons (APP.PS1/CHGFP) and in wild-type littermate controls (WT/CHGFP). We used 5-mo-old mice, an age when the AD transgenics display early amyloid deposition and few cholinergic defects, and 10-mo-old mice, by which time these mice exhibit established disease. BMP9 infusion reduced the number of Aβ42-positive amyloid plaques in the hippocampus and cerebral cortex of 5- and 10-mo-old APP.PS1/CHGFP mice and reversed the reductions in choline acetyltransferase protein levels in the hippocampus of 10-mo-old APP.PS1/CHGFP mice. The treatment increased cholinergic fiber density in the hippocampus of both WT/CHGFP and APP.PS1/CHGFP mice at both ages. BMP9 infusion also increased hippocampal levels of neurotrophin 3, insulin-like growth factor 1, and nerve growth factor and of the nerve growth factor receptors, tyrosine kinase receptor A and p75/NGFR, irrespective of the genotype of the mice. These data show that BMP9 administration is effective in reducing the Aβ42 amyloid plaque burden, reversing cholinergic neuron abnormalities, and generating a neurotrophic milieu for BFCN in a mouse model of AD and provide evidence that the BMP9-signaling pathway may constitute a therapeutic target for AD.
Collapse
|
71
|
Wang JH, Liu YZ, Yin LJ, Chen L, Huang J, Liu Y, Zhang RX, Zhou LY, Yang QJ, Luo JY, Zuo GW, Deng ZL, He BC. BMP9 and COX-2 form an important regulatory loop in BMP9-induced osteogenic differentiation of mesenchymal stem cells. Bone 2013; 57:311-21. [PMID: 23981660 DOI: 10.1016/j.bone.2013.08.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 07/03/2013] [Accepted: 08/13/2013] [Indexed: 01/11/2023]
Abstract
Mesenchymal stem cells (MSCs) can self-renew and differentiate into osteogenic, chondrogenic, adipogenic and myogenic lineages. It's reported that bone morphogenetic protein 9 (BMP9) is one of the most potent osteogenic BMPs to initiate the commitment of MSCs to osteoblast lineage. Cyclooxygenase-2 (COX-2) is critical for bone fracture healing and osteogenic differentiation in MSCs. However, the relationship between COX-2 and BMP9 in osteogenesis remains unknown. Herein, we investigate the role of COX-2 in BMP9-induced osteogenesis in MSCs. We demonstrate that COX-2 is up-regulated as a target of BMP9 in MSCs. Both COX-2 inhibitor (NS-398) and COX-2 knockdown siRNAs can effectively decrease alkaline phosphatase (ALP) activities induced by BMP9 in MSCs. NS-398 also down-regulates BMP9-induced expression of osteopontin and osteocalcin, so does the matrix mineralization. The in vivo studies indicate that knockdown of COX-2 attenuates BMP9-induced ectopic bone formation. In perinatal limb culture assay, NS-398 is shown to reduce the hypertropic chondrocyte zone and ossification induced by BMP9. Mechanistically, knockdown of COX-2 significantly inhibits the BMP9 up-regulated expression of Runx2 and Dlx-5 in MSCs, which can be rescued by exogenous expression of COX-2. Furthermore, knockdown of COX-2 apparently reduces BMP9 induced BMPR-Smad reporter activity, the phosphorylation of Smad1/5/8, and the expression of Smad6 and Smad7 in MSCs. NS-398 blocks the expression of BMP9 mediated by BMP9 recombinant adenovirus. Taken together, our findings suggest that COX-2 plays an important role in BMP9 induced osteogenic differentiation in MSCs; BMP9 and COX-2 may form an important regulatory loop to orchestrate the osteogenic differentiation in MSCs.
Collapse
Affiliation(s)
- Jin-Hua Wang
- Chongqing key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, People's Republic of China; The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Wang Y, Hong S, Li M, Zhang J, Bi Y, He Y, Liu X, Nan G, Su Y, Zhu G, Li R, Zhang W, Wang J, Zhang H, Kong Y, Shui W, Wu N, He Y, Chen X, Luu HH, Haydon RC, Shi LL, He TC, Qin J. Noggin resistance contributes to the potent osteogenic capability of BMP9 in mesenchymal stem cells. J Orthop Res 2013; 31:1796-803. [PMID: 23861103 DOI: 10.1002/jor.22427] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 06/07/2013] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitors and can differentiate into osteogenic, chondrogenic, and adipogenic lineages. Bone morphogenetic proteins (BMPs) play important roles in stem cell proliferation and differentiation. We recently demonstrated that BMP9 is a potent but less understood osteogenic factor. We previously found that BMP9-induced ectopic bone formation is not inhibited by BMP3. Here, we investigate the effect of BMP antagonist noggin on BMP9-induced osteogenic differentiation. BMP antagonists noggin, chording, gremlin, follistatin, and BMP3 are highly expressed in MSCs, while noggin and follistatin are lowly expressed in more differentiated pre-osteoblast C2C12 cells. BMP9-induced osteogenic markers and matrix mineralization are not inhibited by noggin, while noggin blunts BMP2, BMP4, BMP6, and BMP7-induced osteogenic markers and mineralization. Likewise, ectopic bone formation by MSCs transduced with BMP9, but not the other four BMPs, is resistant to noggin inhibition. BMP9-induced nuclear translocation of Smad1/5/8 is not affected by noggin, while noggin blocks BMP2-induced activation of Smad1/5/8 in MSCs. Noggin fails to inhibit BMP9-induced expression of downstream targets in MSCs. Thus, our results strongly suggest that BMP9 may effectively overcome noggin inhibition, which should at least in part contribute to BMP9's potent osteogenic capability in MSCs.
Collapse
Affiliation(s)
- Yi Wang
- Stem Cell Biology and Therapy Laboratory of the Key Laboratory for Pediatrics Co-Designated by Chinese Ministry of Education, Department of Pediatric Surgery, The Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Zhang J, Weng Y, Liu X, Wang J, Zhang W, Kim SH, Zhang H, Li R, Kong Y, Chen X, Shui W, Wang N, Zhao C, Wu N, He Y, Nan G, Chen X, Wen S, Zhang H, Deng F, Wan L, Luu HH, Haydon RC, Shi LL, He TC, Shi Q. Endoplasmic reticulum (ER) stress inducible factor cysteine-rich with EGF-like domains 2 (Creld2) is an important mediator of BMP9-regulated osteogenic differentiation of mesenchymal stem cells. PLoS One 2013; 8:e73086. [PMID: 24019898 PMCID: PMC3760886 DOI: 10.1371/journal.pone.0073086] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 07/15/2013] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitors that can undergo osteogenic differentiation under proper stimuli. We demonstrated that BMP9 is one of the most osteogenic BMPs. However, the molecular mechanism underlying BMP9-initiated osteogenic signaling in MSCs remains unclear. Through gene expression profiling analysis we identified several candidate mediators of BMP9 osteogenic signaling. Here, we focus on one such signaling mediator and investigate the functional role of cysteine-rich with EGF-like domains 2 (Creld2) in BMP9-initiated osteogenic signaling. Creld2 was originally identified as an ER stress-inducible factor localized in the ER-Golgi apparatus. Our genomewide expression profiling analysis indicates that Creld2 is among the top up-regulated genes in BMP9-stimulated MSCs. We confirm that Creld2 is up-regulated by BMP9 in MSCs. ChIP analysis indicates that Smad1/5/8 directly binds to the Creld2 promoter in a BMP9-dependent fashion. Exogenous expression of Creld2 in MSCs potentiates BMP9-induced early and late osteogenic markers, and matrix mineralization. Conversely, silencing Creld2 expression inhibits BMP9-induced osteogenic differentiation. In vivo stem cell implantation assay reveals that exogenous Creld2 promotes BMP9-induced ectopic bone formation and matrix mineralization, whereas silencing Creld2 expression diminishes BMP9-induced bone formation and matrix mineralization. We further show that Creld2 is localized in ER and the ER stress inducers potentiate BMP9-induced osteogenic differentiation. Our results strongly suggest that Creld2 may be directly regulated by BMP9 and ER stress response may play an important role in regulating osteogenic differentiation.
Collapse
Affiliation(s)
- Jiye Zhang
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Yaguang Weng
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Xing Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
- Stem Cell Biology and Therapy Laboratory of the Key Laboratory for Pediatrics co-designated by Chinese Ministry of Education and Chongqing Bureau of Education, The Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Jinhua Wang
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Wenwen Zhang
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Stephanie H. Kim
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Hongyu Zhang
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Ruidong Li
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Yuhan Kong
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Xiang Chen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
- Department of Orthopaedic Surgery, The Affiliated Tangdu Hospital of the Fourth Military Medical University, Xi’an, China
| | - Wei Shui
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Ning Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
- School of Laboratory Medicine and the Affiliated Southwest Hospital of the Third Military Medical University, Chongqing, China
| | - Chen Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
- School of Laboratory Medicine and the Affiliated Southwest Hospital of the Third Military Medical University, Chongqing, China
| | - Ningning Wu
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Yunfeng He
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Guoxin Nan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
- Stem Cell Biology and Therapy Laboratory of the Key Laboratory for Pediatrics co-designated by Chinese Ministry of Education and Chongqing Bureau of Education, The Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xian Chen
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Sheng Wen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
- Stem Cell Biology and Therapy Laboratory of the Key Laboratory for Pediatrics co-designated by Chinese Ministry of Education and Chongqing Bureau of Education, The Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Hongmei Zhang
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Fang Deng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
- School of Laboratory Medicine and the Affiliated Southwest Hospital of the Third Military Medical University, Chongqing, China
| | - Lihua Wan
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Lewis L. Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Tong-Chuan He
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
- Stem Cell Biology and Therapy Laboratory of the Key Laboratory for Pediatrics co-designated by Chinese Ministry of Education and Chongqing Bureau of Education, The Children’s Hospital of Chongqing Medical University, Chongqing, China
- * E-mail: (TCH); (QS)
| | - Qiong Shi
- Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
- * E-mail: (TCH); (QS)
| |
Collapse
|
74
|
Lv Z, Yang D, Li J, Hu M, Luo M, Zhan X, Song P, Liu C, Bai H, Li B, Yang Y, Chen Y, Shi Q, Weng Y. Bone morphogenetic protein 9 overexpression reduces osteosarcoma cell migration and invasion. Mol Cells 2013; 36:119-26. [PMID: 23807047 PMCID: PMC3887952 DOI: 10.1007/s10059-013-0043-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/08/2013] [Accepted: 05/24/2013] [Indexed: 01/24/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is known to promote tumor migration and invasion. Bone morphogenetic proteins (BMPs) are members of the TGF-β family expressed in a variety of human carcinoma cell lines. The role of bone morphogenetic protein 9 (BMP9), the most powerful osteogenic factor, in osteosarcoma (OS) progression has not been fully clarified. The expression of BMP9 and its receptors in OS cell lines was analyzed by RT-PCR. We found that BMP9 and its receptors were expressed in OS cell lines. We further investigated the influence of BMP9 on the biological behaviors of OS cells. BMP9 overexpression in the OS cell lines 143B and MG63 inhibited in vitro cell migration and invasion. We further investigated the expression of a panel of cancer-related genes and found that BMP9 overexpression increased the phosphorylation of Smad1/5/8 proteins, increased the expression of ID1, and reduced the expression and activity of matrix metalloproteinase 9 (MMP9) in OS cells. BMP9 silencing induced the opposite effects. We also found that BMP9 may not affect the chemokine (C-X-C motif) ligand 12 (CXCL12)/C-X-C chemokine receptor type 4 (CXCR4) axis to regulate the invasiveness and metastatic capacity of OS cells. Interestingly, CXCR4 was expressed in both 143B and MG63 cells, while CXCL12 was only detected in MG63 cells. Taken together, we hypothesize that BMP9 inhibits the migration and invasiveness of OS cells through a Smad-dependent pathway by downregulating the expression and activity of MMP9.
Collapse
Affiliation(s)
- Zilan Lv
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Dandan Yang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Jie Li
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Min Hu
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Min Luo
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Xiaoqin Zhan
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Peipei Song
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Chen Liu
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Huili Bai
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Baolin Li
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Yang Yang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Yingying Chen
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Qiong Shi
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Yaguang Weng
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| |
Collapse
|
75
|
Beederman M, Lamplot JD, Nan G, Wang J, Liu X, Yin L, Li R, Shui W, Zhang H, Kim SH, Zhang W, Zhang J, Kong Y, Denduluri S, Rogers MR, Pratt A, Haydon RC, Luu HH, Angeles J, Shi LL, He TC. BMP signaling in mesenchymal stem cell differentiation and bone formation. JOURNAL OF BIOMEDICAL SCIENCE AND ENGINEERING 2013; 6:32-52. [PMID: 26819651 PMCID: PMC4725591 DOI: 10.4236/jbise.2013.68a1004] [Citation(s) in RCA: 211] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Bone morphogenetic proteins (BMPs) are members of the TGF-β superfamily and have diverse functions during development and organogenesis. BMPs play a major role in skeletal development and bone formation, and disruptions in BMP signaling cause a variety of skeletal and extraskeletal anomalies. Several knockout models have provided insight into the mechanisms responsible for these phenotypes. Proper bone formation requires the differentiation of osteoblasts from mesenchymal stem cell (MSC) precursors, a process mediated in part by BMP signaling. Multiple BMPs, including BMP2, BMP6, BMP7 and BMP9, promote osteoblastic differentiation of MSCs both in vitro and in vivo. BMP9 is one of the most osteogenic BMPs yet is a poorly characterized member of the BMP family. Several studies demonstrate that the mechanisms controlling BMP9-mediated osteogenesis differ from other osteogenic BMPs, but little is known about these specific mechanisms. Several pathways critical to BMP9-mediated osteogenesis are also important in the differentiation of other cell lineages, including adipocytes and chondrocytes. BMP9 has also demonstrated translational promise in spinal fusion and bone fracture repair. This review will summarize our current knowledge of BMP-mediated osteogenesis, with a focus on BMP9, by presenting recently completed work which may help us to further elucidate these pathways.
Collapse
Affiliation(s)
- Maureen Beederman
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA
| | - Joseph D Lamplot
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA
| | - Guoxin Nan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA; Stem Cell Biology and Therapy Laboratory of the Key Laboratory for Pediatrics Co-Designated by Chinese Ministry of Education, The Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jinhua Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA; The Affiliated Hospitals and the Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Xing Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA; Stem Cell Biology and Therapy Laboratory of the Key Laboratory for Pediatrics Co-Designated by Chinese Ministry of Education, The Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Liangjun Yin
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA; The Affiliated Hospitals and the Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Ruidong Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA; The Affiliated Hospitals and the Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Wei Shui
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA; The Affiliated Hospitals and the Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Hongyu Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA; The Affiliated Hospitals and the Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Stephanie H Kim
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA
| | - Wenwen Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA; The Affiliated Hospitals and the Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Jiye Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA; The Affiliated Hospitals and the Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Yuhan Kong
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA; The Affiliated Hospitals and the Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Sahitya Denduluri
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA
| | - Mary Rose Rogers
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA
| | - Abdullah Pratt
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA
| | - Rex C Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA
| | - Hue H Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA
| | - Jovito Angeles
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA
| | - Lewis L Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, USA; Stem Cell Biology and Therapy Laboratory of the Key Laboratory for Pediatrics Co-Designated by Chinese Ministry of Education, The Children's Hospital of Chongqing Medical University, Chongqing, China; The Affiliated Hospitals and the Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| |
Collapse
|
76
|
Hegarty SV, O'Keeffe GW, Sullivan AM. BMP-Smad 1/5/8 signalling in the development of the nervous system. Prog Neurobiol 2013; 109:28-41. [PMID: 23891815 DOI: 10.1016/j.pneurobio.2013.07.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 07/16/2013] [Accepted: 07/16/2013] [Indexed: 02/07/2023]
Abstract
The transcription factors, Smad1, Smad5 and Smad8, are the pivotal intracellular effectors of the bone morphogenetic protein (BMP) family of proteins. BMPs and their receptors are expressed in the nervous system (NS) throughout its development. This review focuses on the actions of Smad 1/5/8 in the developing NS. The mechanisms by which these Smad proteins regulate the induction of the neuroectoderm, the central nervous system (CNS) primordium, and finally the neural crest, which gives rise to the peripheral nervous system (PNS), are reviewed herein. We describe how, following neural tube closure, the most dorsal aspect of the tube becomes a signalling centre for BMPs, which directs the pattern of the development of the dorsal spinal cord (SC), through the action of Smad1, Smad5 and Smad8. The direct effects of Smad 1/5/8 signalling on the development of neuronal and non-neuronal cells from various neural progenitor cell populations are then described. Finally, this review discusses the neurodevelopmental abnormalities associated with the knockdown of Smad 1/5/8.
Collapse
Affiliation(s)
- Shane V Hegarty
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| | | | | |
Collapse
|
77
|
Liu C, Weng Y, Yuan T, Zhang H, Bai H, Li B, Yang D, Zhang R, He F, Yan S, Zhan X, Shi Q. CXCL12/CXCR4 signal axis plays an important role in mediating bone morphogenetic protein 9-induced osteogenic differentiation of mesenchymal stem cells. Int J Med Sci 2013; 10:1181-92. [PMID: 23935395 PMCID: PMC3739017 DOI: 10.7150/ijms.6657] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 06/11/2013] [Indexed: 12/30/2022] Open
Abstract
Mesenchymal progenitor stem cells (MPCs) are a group of bone marrow stromal progenitor cells processing osteogenic, chondrogenic, adipogenic and myogenic lineages differentiations. Previous studies have demonstrated that bone morphogeneic protein 9(BMP9) is one of the most osteogenic BMPs both in vitro and in vivo, however, the underlying molecular mechanism of osteogenesis induced by BMP9 is needed to be deep explored. Here, we used the recombinant adenoviruses assay to introduce BMP9 into C3H10T1/2 mesenchymal stem cells to elucidate the role of CXCL12/CXCR4 signal axis during BMP9-incuced osteogenic differentiation. The results showed that CXCL12 and CXCR4 expressions were down-regulated at the stage of BMP9-induced osteogenic differentiation, in a dose- and time-dependent. Pretreatment of C3H10T1/2 cells with CXCL12/CXCR4 could significantly affect the early and mid osteogenic markers alkaline phosphatase (ALP), osteocalcin (OCN), the transcription factors of Runx2, Osx, Plzf and Dlx5 expression, through activating the Smad, MAPK signaling pathway. Addition of exogenous CXCL12 did not affect the changes of the late osteogenic marker calcium deposition. Thus, our findings suggest a co-requirement of the CXCL12/CXCR4 signal axis in BMP9-induced the early- and mid-process of osteogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Chen Liu
- 1. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yaguang Weng
- 1. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Taixian Yuan
- 1. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Hong Zhang
- 2. Department of Laboratory Medicine, Jinan Sixth Hospital, Zhangqiu250200, China
| | - Huili Bai
- 1. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Baolin Li
- 1. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Dandan Yang
- 1. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ruyi Zhang
- 1. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Fang He
- 1. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shujuan Yan
- 1. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiaoqin Zhan
- 1. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qiong Shi
- 1. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
78
|
Liu X, Qin J, Luo Q, Bi Y, Zhu G, Jiang W, Kim SH, Li M, Su Y, Nan G, Cui J, Zhang W, Li R, Chen X, Kong Y, Zhang J, Wang J, Rogers MR, Zhang H, Shui W, Zhao C, Wang N, Liang X, Wu N, He Y, Luu HH, Haydon RC, Shi LL, Li T, He TC, Li M. Cross-talk between EGF and BMP9 signalling pathways regulates the osteogenic differentiation of mesenchymal stem cells. J Cell Mol Med 2013; 17:1160-72. [PMID: 23844832 PMCID: PMC4118175 DOI: 10.1111/jcmm.12097] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 05/13/2013] [Accepted: 05/20/2013] [Indexed: 01/13/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitors, which give rise to several lineages, including bone, cartilage and fat. Epidermal growth factor (EGF) stimulates cell growth, proliferation and differentiation. EGF acts by binding with high affinity to epidermal growth factor receptor (EGFR) on the cell surface and stimulating the intrinsic protein tyrosine kinase activity of its receptor, which initiates a signal transduction cascade causing a variety of biochemical changes within the cell and regulating cell proliferation and differentiation. We have identified BMP9 as one of the most osteogenic BMPs in MSCs. In this study, we investigate if EGF signalling cross-talks with BMP9 and regulates BMP9-induced osteogenic differentiation. We find that EGF potentiates BMP9-induced early and late osteogenic markers of MSCs in vitro, which can be effectively blunted by EGFR inhibitors Gefitinib and Erlotinib or receptor tyrosine kinase inhibitors AG-1478 and AG-494 in a dose- and time-dependent manner. Furthermore, EGF significantly augments BMP9-induced bone formation in the cultured mouse foetal limb explants. In vivo stem cell implantation experiment reveals that exogenous expression of EGF in MSCs can effectively potentiate BMP9-induced ectopic bone formation, yielding larger and more mature bone masses. Interestingly, we find that, while EGF can induce BMP9 expression in MSCs, EGFR expression is directly up-regulated by BMP9 through Smad1/5/8 signalling pathway. Thus, the cross-talk between EGF and BMP9 signalling pathways in MSCs may underline their important roles in regulating osteogenic differentiation. Harnessing the synergy between BMP9 and EGF should be beneficial for enhancing osteogenesis in regenerative medicine.
Collapse
Affiliation(s)
- Xing Liu
- Stem Cell Biology and Therapy Laboratory of the Key Laboratory for Pediatrics designated by Chinese Ministry of Education and Chongqing Bureau of Education, Department of Orthopaedic Surgery, The Children's Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
The ALK-1/Smad1 pathway in cardiovascular physiopathology. A new target for therapy? Biochim Biophys Acta Mol Basis Dis 2013; 1832:1492-510. [PMID: 23707512 DOI: 10.1016/j.bbadis.2013.05.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 05/04/2013] [Accepted: 05/13/2013] [Indexed: 01/04/2023]
Abstract
Activin receptor-like kinase-1 or ALK-1 is a type I cell surface receptor for the transforming growth factor-β (TGF-β) family of proteins. The role of ALK-1 in endothelial cells biology and in angiogenesis has been thoroughly studied by many authors. However, it has been recently suggested a possible role of ALK-1 in cardiovascular homeostasis. ALK-1 is not only expressed in endothelial cells but also in smooth muscle cells, myofibroblast, hepatic stellate cells, chondrocytes, monocytes, myoblasts, macrophages or fibroblasts, but its role in these cells have not been deeply analyzed. Due to the function of ALK-1 in these cells, this receptor plays a role in several cardiovascular diseases. Animals with ALK-1 haploinsufficiency and patients with mutations in Acvrl1 (the gene that codifies for ALK-1) develop type-2 Hereditary Hemorrhagic Telangiectasia. Moreover, ALK-1 heterozygous mice develop pulmonary hypertension. Higher levels of ALK-1 have been observed in atherosclerotic plaques, suggesting a possible protector role of this receptor. ALK-1 deficiency is also related to the development of arteriovenous malformations (AVMs). Besides, due to the ability of ALK-1 to regulate cell proliferation and migration, and to modulate extracellular matrix (ECM) protein expression in several cell types, ALK-1 has been now demonstrated to play an important role in cardiovascular remodeling. In this review, we would like to offer a complete vision of the role of ALK-1 in many process related to cardiovascular homeostasis, and the involvement of this protein in the development of cardiovascular diseases, suggesting the possibility of using the ALK-1/smad-1 pathway as a powerful therapeutic target.
Collapse
|
80
|
Song T, Wang W, Xu J, Zhao D, Dong Q, Li L, Yang X, Duan X, Liang Y, Xiao Y, Wang J, He J, Tang M, Wang J, Luo J. Fibroblast growth factor 2 inhibits bone morphogenetic protein 9-induced osteogenic differentiation of mesenchymal stem cells by repressing Smads signaling and subsequently reducing Smads dependent up-regulation of ALK1 and ALK2. Int J Biochem Cell Biol 2013; 45:1639-46. [PMID: 23680673 DOI: 10.1016/j.biocel.2013.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 04/10/2013] [Accepted: 05/06/2013] [Indexed: 12/24/2022]
Abstract
Understanding the interactions between growth factors and bone morphogenic proteins (BMPs) signaling remains a crucial issue to optimize the use of mesenchymal stem cells (MSCs) and BMPs in bone tissue engineering. BMP9 is highly capable of promoting osteogenic differentiation of MSCs. Fibroblast growth factor 2 (FGF2) is abundantly secreted during the healing process of fractures or in surgery bone sites. Herein, we explore the detail effect of FGF2 on BMP9-induced osteogenic differentiation of MSCs. It was found that FGF2 inhibited BMP9-induced osteogenic differentiation by blocking BMP9-induced Smads signaling and subsequently reducing Smads dependent up-regulation of ALK1 and ALK2 in MSCs. This effect was rescued by exogenous expression of ALK1 and ALK2, which are proved to be receptors for BMP9. Our results discovered a clue to explain the mechanism involved in the inhibitory effect of FGF2 on BMP9-induced osteogenic differentiation of MSCs. This crosstalk between FGF2 and BMP9 should be emphasized in the future use of BMP9 in therapeutic purpose of fracture repair.
Collapse
Affiliation(s)
- Tao Song
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 40016, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Abstract
Hypothalamic neural circuits are known to regulate energy homeostasis and feeding behavior, but how these circuits are established during development is not well understood. Here we report that embryonic neural progenitors that express the transcription factor OLIG1 contribute neurons to the ventral hypothalamus including the arcuate nucleus (ARH), a center that regulates feeding behavior. Ablation of bone morphogenetic protein receptor 1a (BMPR1A) in the OLIG1 lineage resulted in hypophagia, hypoglycemia, and weight loss after the second postnatal week with death by week 4. Differentiation and specification of inhibitory hypothalamic neurons contributing to melanocortin and dopaminergic systems were abnormal in the BMPR1A-deficient ARH. Although the hypophagia promoted expression of the orexigenic neuropeptide agouti related protein (AgRP) in the BMPR1A-deficient ARH, there was a profound decrease of AgRP(+) axonal terminals in the mutant ARH targets including dorsomedial and paraventricular hypothalamic nuclei. Projection of AgRP(+) neurons to these nuclei is known to be regulated by leptin. Leptin injection in neonatal mice increased bone morphogenic protein (BMP) signaling in the ventral hypothalamus, and blocking BMP signaling prevented leptin-induced neurite outgrowth in ARH explant cultures. These findings suggest that BMPR1A signaling is critical for postnatal establishment of leptin-responsive orexigenic fibers from ARH to multiple hypothalamic nuclei. More generally these observations indicate that BMPR1A signaling regulates postnatal establishment of OLIG1 lineage-derived ARH neuronal circuits that are critical for leptin-mediated feeding behavior.
Collapse
|
82
|
Fong D, Bisson M, Laberge G, McManus S, Grenier G, Faucheux N, Roux S. Bone morphogenetic protein-9 activates Smad and ERK pathways and supports human osteoclast function and survival in vitro. Cell Signal 2013; 25:717-28. [PMID: 23313128 DOI: 10.1016/j.cellsig.2012.12.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 11/25/2012] [Accepted: 12/10/2012] [Indexed: 12/12/2022]
Abstract
BMP-9 is a potent osteogenic factor; however, its effects on osteoclasts, the bone-resorbing cells, remain unknown. To determine the effects of BMP-9 on osteoclast formation, activity and survival, we used human cord blood monocytes as osteoclast precursors that form multinucleated osteoclasts in the presence of RANKL and M-CSF in long-term cultures. BMP-9 did not affect osteoclast formation, but adding BMP-9 at the end of the culture period significantly increased bone resorption compared to untreated cultures, and reduced both the rate of apoptosis and caspase-9 activity. BMP-9 also significantly downregulated the expression of pro-apoptotic Bid, but only after RANKL and M-CSF, which are both osteoclast survival factors, had been eliminated from the culture medium. To investigate the mechanisms involved in the effects of BMP-9, we first showed that osteoclasts expressed some BMP receptors, including BMPR-IA, BMPR-IB, ALK1, and BMPR-II. We also found that BMP-9 was able to induce the phosphorylation of Smad-1/5/8 and ERK 1/2 proteins, but did not induce p38 phosphorylation. Finally, knocking down the BMPR-II receptor abrogated the BMP-9-induced ERK-signaling, as well as the increase in bone resorption. In conclusion, these results show for the first time that BMP-9 directly affects human osteoclasts, enhancing bone resorption and protecting osteoclasts against apoptosis. BMP-9 signaling in human osteoclasts involves the canonical Smad-1/5/8 pathway, and the ERK pathway.
Collapse
Affiliation(s)
- David Fong
- Division of Rheumatology, Faculty of Medicine, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
83
|
Inhibition of ERK1/2 Kinase Enhances BMP9-induced Osteogenic Differentiation of Mesenchymal Stem Cells*. PROG BIOCHEM BIOPHYS 2013. [DOI: 10.3724/sp.j.1206.2012.00170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
84
|
Liang J, Wu S, Zhao H, Li SL, Liu ZX, Wu J, Zhou L. Human umbilical cord mesenchymal stem cells derived from Wharton's jelly differentiate into cholinergic-like neurons in vitro. Neurosci Lett 2013. [DOI: 10.1016/j.neulet.2012.11.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
85
|
Hu N, Jiang D, Huang E, Liu X, Li R, Liang X, Kim SH, Chen X, Gao JL, Zhang H, Zhang W, Kong YH, Zhang J, Wang J, Shui W, Luo X, Liu B, Cui J, Rogers MR, Shen J, Zhao C, Wang N, Wu N, Luu HH, Haydon RC, He TC, Huang W. BMP9-regulated angiogenic signaling plays an important role in the osteogenic differentiation of mesenchymal progenitor cells. J Cell Sci 2012. [PMID: 23203800 DOI: 10.1242/jcs.114231] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mesenchymal stromal progenitor cells (MSCs) are multipotent progenitors that can be isolated from numerous tissues. MSCs can undergo osteogenic differentiation under proper stimuli. We have recently demonstrated that bone morphogenetic protein 9 (BMP9) is one of the most osteogenic BMPs. As one of the least studied BMPs, BMP9 has been shown to regulate angiogenesis in endothelial cells. However, it is unclear whether BMP9-regulated angiogenic signaling plays any important role in the BMP9-initiated osteogenic pathway in MSCs. Here, we investigate the functional role of hypoxia-inducible factor 1α (HIF1α)-mediated angiogenic signaling in BMP9-regulated osteogenic differentiation of MSCs. We find that BMP9 induces HIF1α expression in MSCs through Smad1/5/8 signaling. Exogenous expression of HIF1α potentiates BMP9-induced osteogenic differentiation of MSCs both in vitro and in vivo. siRNA-mediated silencing of HIF1α or HIF1α inhibitor CAY10585 profoundly blunts BMP9-induced osteogenic signaling in MSCs. HIF1α expression regulated by cobalt-induced hypoxia also recapitulates the synergistic effect between HIF1α and BMP9 in osteogenic differentiation. Mechanistically, HIF1α is shown to exert its synergistic effect with BMP9 by inducing both angiogenic signaling and osteogenic signaling in MSCs. Thus, our findings should not only expand our understanding of the molecular basis behind BMP9-regulated osteoblastic lineage-specific differentiation, but also provide an opportunity to harness the BMP9-induced synergy between osteogenic and angiogenic signaling pathways in regenerative medicine.
Collapse
Affiliation(s)
- Ning Hu
- The First Affiliated Hospital and the Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Bond AM, Bhalala OG, Kessler JA. The dynamic role of bone morphogenetic proteins in neural stem cell fate and maturation. Dev Neurobiol 2012; 72:1068-84. [PMID: 22489086 DOI: 10.1002/dneu.22022] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The bone morphogenetic proteins (BMPs) are a group of powerful morphogens that are critical for development of the nervous system. The effects of BMP signaling on neural stem cells are myriad and dynamic, changing with each stage of development. During early development inhibition of BMP signaling differentiates neuroectoderm from ectoderm, and BMP signaling helps to specify neural crest. Thus modulation of BMP signaling underlies formation of both the central and peripheral nervous systems. BMPs secreted from dorsal structures then form a gradient which helps pattern the dorsal-ventral axis of the developing spinal cord and brain. During forebrain development BMPs sequentially induce neurogenesis and then astrogliogenesis and participate in neurite outgrowth from immature neurons. BMP signaling also plays a critical role in maintaining adult neural stem cell niches in the subventricular zone (SVZ) and subgranular zone (SGZ). BMPs are able to exert such diverse effects through closely regulated temporospatial expression and interaction with other signaling pathways.
Collapse
Affiliation(s)
- Allison M Bond
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | |
Collapse
|
87
|
Li B, Yang Y, Jiang S, Ni B, Chen K, Jiang L. Adenovirus-mediated overexpression of BMP-9 inhibits human osteosarcoma cell growth and migration through downregulation of the PI3K/AKT pathway. Int J Oncol 2012; 41:1809-19. [PMID: 22948234 DOI: 10.3892/ijo.2012.1617] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Accepted: 08/20/2012] [Indexed: 11/06/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are members of the TGF-β superfamily of signaling molecules and have previously been shown to be associated with the biological behavior of osteosarcoma. However, to date the effects and molecular mechanisms of BMP-9 on osteosarcoma progression are unknown. We performed real-time PCR and western blot analysis to characterize the endogenous expression of BMP-9 in osteosarcoma cell lines. We used a recombinant adenovirus expressing BMP-9 (adBMP-9) to infect osteosarcoma cell lines with relatively low endogenous BMP-9 expression to determine the functional relevance of BMP-9 overexpression to osteosarcoma cell growth and migration in vitro and in vivo, and further investigated the expression levels of Ki-67, matrix metallopeptidase-9 (MMP-9), phosphoinositide 3-kinase p85α (PI3Kp85α) and phosphorylated AKT (p-AKT). As a result, osteosarcoma cell proliferation and migration were significantly diminished by adBMP-9, indicated by MTT and wound-healing assays, and cell apoptosis was markedly induced, indicated by Hoechst 33342/PI assay and Annexin V-FITC apoptosis detection. When BMP-9 expression was enhanced, the expression of PI3Kp85α, p-AKT, Ki-67 and MMP-9 was downregulated in osteosarcoma cells. In addition, the tumor volumes in MG-63 and HOS subcutaneous nude mouse models treated with adBMP-9 were significantly smaller compared to those of the ad-GFP group. These results suggested that the enhanced expression of BMP-9 in osteosarcoma cells by adBMP-9 exerted inhibitory effects on growth and migration of osteosarcoma cells possibly via blockade of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Bo Li
- Department of Orthopedic Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200092, PR China
| | | | | | | | | | | |
Collapse
|
88
|
Xu DJ, Zhao YZ, Wang J, He JW, Weng YG, Luo JY. Smads, p38 and ERK1/2 are involved in BMP9-induced osteogenic differentiation of C3H10T1/2 mesenchymal stem cells. BMB Rep 2012; 45:247-52. [PMID: 22531136 DOI: 10.5483/bmbrep.2012.45.4.247] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although previous studies have demonstrated that BMP9 is highly capable of inducing osteogenic differentiation of mesenchymal stem cells, the molecular mechanism involved remains to be fully elucidated. In this study, we showed that BMP9 simultaneously promotes the activation of Smad1/5/8, p38 and ERK1/2 in C3H10T1/2 cells. Knockdown of Smad4 with RNA interference reduced nuclear translocation of Smad1/5/8, and disrupted BMP9-induced osteogenic differentiation. BMP9-induced osteogenic differentiation was blocked by p38 inhibitor SB203580, whereas enhanced by ERK1/2 inhibitor PD98059. SB203580 decreased BMP9-activated Smads singling, and yet PD98059 stimulated Smads singling in C3H10T1/2 cells. The effects of inhibitor were reproduced with adenovirus expressing siRNA targeted p38 and ERK1/2, respectively. Taken together, our findings revealed that Smads, p38 and ERK1/2 are involved in BMP9-induced osteogenic differentiation. Also, it is noteworthy that p38 and ERK1/2 may play opposing regulatory roles in mediating BMP9-induced osteogenic differentiation of C3H10T1/2 cells.
Collapse
Affiliation(s)
- Dao-jing Xu
- Key Laboratory of Diagnostic Medicine, Chinese Ministry of Education, Chongqing Medical University, Chongqing, PR China
| | | | | | | | | | | |
Collapse
|
89
|
Zhao Y, Song T, Wang W, Wang J, He J, Wu N, Tang M, He B, Luo J. P38 and ERK1/2 MAPKs act in opposition to regulate BMP9-induced osteogenic differentiation of mesenchymal progenitor cells. PLoS One 2012; 7:e43383. [PMID: 22912865 PMCID: PMC3422272 DOI: 10.1371/journal.pone.0043383] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Accepted: 07/23/2012] [Indexed: 12/15/2022] Open
Abstract
Although previous studies have demonstrated that BMP9 is highly capable of inducing osteogenic differentiation and bone formation, the precise molecular mechanism involved remains to be fully elucidated. In this current study, we explore the possible involvement and detail effects of p38 and ERK1/2 MAPKs on BMP9-indcued osteogenic differentiation of mesenchymal progenitor cell (MPCs). We find that BMP9 simultaneously stimulates the activation of p38 and ERK1/2 in MPCs. BMP9-induced early osteogenic marker, such as alkaline phosphatase (ALP), and late osteogenic markers, such as matrix mineralization and osteocalcin (OC) are inhibited by p38 inhibitor SB203580, whereas enhanced by ERK1/2 inhibitor PD98059. BMP9-induced activation of Runx2 and Smads signaling are reduced by SB203580, and yet increased by PD98059 in MPCs. The in vitro effects of inhibitors are reproduced with adenoviruses expressing siRNA targeted p38 and ERK1/2, respectively. Using mouse calvarial organ culture and subcutaneous MPCs implantation, we find that inhibition of p38 activity leads to significant decrease in BMP9-induced osteogenic differentiation and bone formation, however, blockage of ERK1/2 results in effective increase in BMP9-indcued osteogenic differentiation in vivo. Together, our results reveal that p38 and ERK1/2 MAPKs are activated in BMP9-induced osteogenic differentiation of MPCs. What is most noteworthy, however, is that p38 and ERK1/2 act in opposition to regulate BMP9-induced osteogenic differentiation of MPCs.
Collapse
Affiliation(s)
- Yingze Zhao
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Tao Song
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Wenjuan Wang
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Jin Wang
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Juanwen He
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Ningning Wu
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Min Tang
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Baicheng He
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Jinyong Luo
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
- * E-mail:
| |
Collapse
|
90
|
Huang E, Zhu G, Jiang W, Yang K, Gao Y, Luo Q, Gao JL, Kim SH, Liu X, Li M, Shi Q, Hu N, Wang L, Liu H, Cui J, Zhang W, Li R, Chen X, Kong YH, Zhang J, Wang J, Shen J, Bi Y, Statz J, He BC, Luo J, Wang H, Xiong F, Luu HH, Haydon RC, Yang L, He TC. Growth hormone synergizes with BMP9 in osteogenic differentiation by activating the JAK/STAT/IGF1 pathway in murine multilineage cells. J Bone Miner Res 2012; 27:1566-75. [PMID: 22467218 DOI: 10.1002/jbmr.1622] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Growth hormone (GH) is usually released by somatotrophs in the anterior pituitary in response to the GH-releasing hormone and plays an important role in skeleton development and postnatal growth. However, it is unclear if extrapituitary GH exerts any effect on murine multilineage cells (MMCs). MMCs are multipotent progenitors that give rise to several lineages, including bone, cartilage, and fat. We have identified bone morphogenic protein 9 (BMP9) as one of the most osteogenic BMPs in MMCs by regulating a distinct set of downstream mediators. In this study, we find that GH is one of the most significantly upregulated genes by BMP9 in mouse MMCs through expression-profiling analysis. We confirm that GH is a direct early target of and upregulated by BMP9 signaling. Exogenous GH synergizes with BMP9 on inducing early and late osteogenic markers in MMCs. Furthermore, BMP9 and GH costimulation leads to a significant expansion of growth plate in cultured limb explants. Although GH alone does not induce de novo bone formation in an ectopic bone formation model, BMP9 and GH costimulated MMCs form more mature bone, which can be inhibited by silencing GH expression. The synergistic osteogenic activity between BMP9 and GH can be significantly blunted by JAK/STAT inhibitors, leading to a decrease in GH-regulated insulin-like growth factor 1 (IGF1) expression in MMCs. Our results strongly suggest that BMP9 may effectively regulate extrapituitary GH expression in MMCs. Thus, it is conceivable that the BMP9-GH-IGF axis may be exploited as an innovative strategy to enhance osteogenesis in regenerative medicine.
Collapse
Affiliation(s)
- Enyi Huang
- School of Bioengineering, Chongqing University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Abstract
Various studies have demonstrated that somatic differentiated cells can be reprogrammed into other differentiated states or into pluripotency, thus showing that the differentiated cellular state is not irreversible. These findings have generated intense interest in the process of reprogramming and in mechanisms that govern the pluripotent state. However, the realization that differentiated cells can be triggered to switch to considerably different lineages also emphasizes that we need to understand how the identity of mature cells is normally maintained. Here we review recent studies on how the differentiated state is controlled at the transcriptional level and discuss how new insights have begun to elucidate mechanisms underlying the stable maintenance of mature cell identities.
Collapse
Affiliation(s)
- Johan Holmberg
- Ludwig Institute for Cancer Research Ltd, BOX 240, Karolinska Institutet, S-171 77 Stockholm, Sweden.
| | | |
Collapse
|
92
|
Jonakait GM, Pratt L, Acevedo G, Ni L. Microglial regulation of cholinergic differentiation in the basal forebrain. Dev Neurobiol 2012; 72:857-64. [DOI: 10.1002/dneu.20969] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
93
|
Cabeza C, Figueroa A, Lazo OM, Galleguillos C, Pissani C, Klein A, Gonzalez-Billault C, Inestrosa NC, Alvarez AR, Zanlungo S, Bronfman FC. Cholinergic abnormalities, endosomal alterations and up-regulation of nerve growth factor signaling in Niemann-Pick type C disease. Mol Neurodegener 2012; 7:11. [PMID: 22458984 PMCID: PMC3395862 DOI: 10.1186/1750-1326-7-11] [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: 08/04/2011] [Accepted: 03/29/2012] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Neurotrophins and their receptors regulate several aspects of the developing and mature nervous system, including neuronal morphology and survival. Neurotrophin receptors are active in signaling endosomes, which are organelles that propagate neurotrophin signaling along neuronal processes. Defects in the Npc1 gene are associated with the accumulation of cholesterol and lipids in late endosomes and lysosomes, leading to neurodegeneration and Niemann-Pick type C (NPC) disease. The aim of this work was to assess whether the endosomal and lysosomal alterations observed in NPC disease disrupt neurotrophin signaling. As models, we used i) NPC1-deficient mice to evaluate the central cholinergic septo-hippocampal pathway and its response to nerve growth factor (NGF) after axotomy and ii) PC12 cells treated with U18666A, a pharmacological cellular model of NPC, stimulated with NGF. RESULTS NPC1-deficient cholinergic cells respond to NGF after axotomy and exhibit increased levels of choline acetyl transferase (ChAT), whose gene is under the control of NGF signaling, compared to wild type cholinergic neurons. This finding was correlated with increased ChAT and phosphorylated Akt in basal forebrain homogenates. In addition, we found that cholinergic neurons from NPC1-deficient mice had disrupted neuronal morphology, suggesting early signs of neurodegeneration. Consistently, PC12 cells treated with U18666A presented a clear NPC cellular phenotype with a prominent endocytic dysfunction that includes an increased size of TrkA-containing endosomes and reduced recycling of the receptor. This result correlates with increased sensitivity to NGF, and, in particular, with up-regulation of the Akt and PLC-γ signaling pathways, increased neurite extension, increased phosphorylation of tau protein and cell death when PC12 cells are differentiated and treated with U18666A. CONCLUSIONS Our results suggest that the NPC cellular phenotype causes neuronal dysfunction through the abnormal up-regulation of survival pathways, which causes the perturbation of signaling cascades and anomalous phosphorylation of the cytoskeleton.
Collapse
Affiliation(s)
- Carolina Cabeza
- Physiology Department, Millennium Nucleus in Regenerative Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Li C, Yang X, He Y, Ye G, Li X, Zhang X, Zhou L, Deng F. Bone morphogenetic protein-9 induces osteogenic differentiation of rat dental follicle stem cells in P38 and ERK1/2 MAPK dependent manner. Int J Med Sci 2012; 9:862-71. [PMID: 23155360 PMCID: PMC3498751 DOI: 10.7150/ijms.5027] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Accepted: 09/17/2012] [Indexed: 11/28/2022] Open
Abstract
Dental follicle stem cells are a group of cells possessing osteogenic, adipogenetic and neurogenic differentiations, but the specific mechanism underlying the multilineage differentiation remains still unclear. Great attention has been paid to bone morphogenetic protein-9 (BMP-9) due to its potent osteogenic activity. In the present study, rat dental follicle stem cells were isolated and purified, and cells of passage 3 underwent adenovirus mediated BMP-9 gene transfection to prepare dental follicle stem cells with stable BMP-9 expression. Detection of alkaline phosphatase (ALP) and calcium deposition showed dental follicle stem cells transfected with BMP-9 gene could significantly promote the osteogenesis. In addition, SB203580 and PD98059 were employed to block the p38 mitogen-activated protein kinase (p38MAPK) and extracellular signal-regulated kinase (ERK1/2), respectively. Detection of ALP and calcium deposition revealed the BMP-9 induced osteogenic differentiation of dental follicle stem cells depended on MAPK signaling pathway.
Collapse
Affiliation(s)
- Conghua Li
- Department of Stomatology, Research Center for Oral Diseases and Biomedicine in Chongqing, Affiliated Stomatological Hospital, Chongqing Medical University, China
| | | | | | | | | | | | | | | |
Collapse
|
95
|
Bidart M, Ricard N, Levet S, Samson M, Mallet C, David L, Subileau M, Tillet E, Feige JJ, Bailly S. BMP9 is produced by hepatocytes and circulates mainly in an active mature form complexed to its prodomain. Cell Mol Life Sci 2012; 69:313-24. [PMID: 21710321 PMCID: PMC11114909 DOI: 10.1007/s00018-011-0751-1] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 05/31/2011] [Accepted: 06/07/2011] [Indexed: 01/29/2023]
Abstract
Bone Morphogenetic Protein 9 (BMP9) has been recently found to be the physiological ligand for the activin receptor-like kinase 1 (ALK1), and to be a major circulating vascular quiescence factor. Moreover, a soluble chimeric ALK1 protein (ALK1-Fc) has recently been developed and showed powerful anti-tumor growth and anti-angiogenic effects. However, not much is known concerning BMP9. This prompted us to investigate the human endogenous sources of this cytokine and to further characterize its circulating form(s) and its function. Analysis of BMP9 expression reveals that BMP9 is produced by hepatocytes and intrahepatic biliary epithelial cells. Gel filtration analysis combined with ELISA and biological assays demonstrate that BMP9 circulates in plasma (1) as an unprocessed inactive form that can be further activated by furin a serine endoprotease, and (2) as a mature and fully active form (composed of the mature form associated with its prodomain). Analysis of BMP9 circulating levels during mouse development demonstrates that BMP9 peaks during the first 3 weeks after birth and then decreases to 2 ng/mL in adulthood. We also show that circulating BMP9 physiologically induces a constitutive Smad1/5/8 phosphorylation in endothelial cells. Taken together, our results argue for the role of BMP9 as a hepatocyte-derived factor, circulating in inactive (40%) and active (60%) forms, the latter constantly activating endothelial cells to maintain them in a resting state.
Collapse
Affiliation(s)
- Marie Bidart
- Unit 1036, Biology of Cancer and Infection, INSERM, 17 rue des Martyrs, 38054 Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, 38041 Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, 38054 Grenoble, France
- Pôle Recherche, Centre Hospitalier Universitaire de Grenoble, 38043 Grenoble, France
| | - Nicolas Ricard
- Unit 1036, Biology of Cancer and Infection, INSERM, 17 rue des Martyrs, 38054 Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, 38041 Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, 38054 Grenoble, France
| | - Sandrine Levet
- Unit 1036, Biology of Cancer and Infection, INSERM, 17 rue des Martyrs, 38054 Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, 38041 Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, 38054 Grenoble, France
| | - Michel Samson
- U620/EA 4427 SeRAIC, INSERM, Université Rennes 1, 35043 Rennes, France
| | - Christine Mallet
- Unit 1036, Biology of Cancer and Infection, INSERM, 17 rue des Martyrs, 38054 Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, 38041 Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, 38054 Grenoble, France
| | - Laurent David
- Unit 1036, Biology of Cancer and Infection, INSERM, 17 rue des Martyrs, 38054 Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, 38041 Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, 38054 Grenoble, France
- Center for Systems Biology, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5 Canada
| | - Mariela Subileau
- Unit 1036, Biology of Cancer and Infection, INSERM, 17 rue des Martyrs, 38054 Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, 38041 Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, 38054 Grenoble, France
| | - Emmanuelle Tillet
- Unit 1036, Biology of Cancer and Infection, INSERM, 17 rue des Martyrs, 38054 Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, 38041 Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, 38054 Grenoble, France
| | - Jean-Jacques Feige
- Unit 1036, Biology of Cancer and Infection, INSERM, 17 rue des Martyrs, 38054 Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, 38041 Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, 38054 Grenoble, France
| | - Sabine Bailly
- Unit 1036, Biology of Cancer and Infection, INSERM, 17 rue des Martyrs, 38054 Grenoble, France
- UJF-Grenoble 1, Biology of Cancer and Infection, 38041 Grenoble, France
- CEA, DSV/iRTSV, Biology of Cancer and Infection, 38054 Grenoble, France
| |
Collapse
|
96
|
ZHAO D, WANG J, LUO JY, LIU YL, WANG H, ZENG ZF, YUAN J. Bone Morphogenetic Protein 9 Regulate Osteogenic Differentiation of C3H10T1/2 Mesenchymal Stem Cells Through p38 Kinase Pathway*. PROG BIOCHEM BIOPHYS 2011. [DOI: 10.3724/sp.j.1206.2011.00200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
97
|
Bissonnette CJ, Lyass L, Bhattacharyya BJ, Belmadani A, Miller RJ, Kessler JA. The controlled generation of functional basal forebrain cholinergic neurons from human embryonic stem cells. Stem Cells 2011; 29:802-11. [PMID: 21381151 DOI: 10.1002/stem.626] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An early substantial loss of basal forebrain cholinergic neurons (BFCN) is a constant feature of Alzheimer's disease and is associated with deficits in spatial learning and memory. The ability to selectively control the differentiation of human embryonic stem cells (hESCs) into BFCN would be a significant step toward a cell replacement therapy. We demonstrate here a method for the derivation of a predominantly pure population of BFCN from hESC cells using diffusible ligands present in the forebrain at developmentally relevant time periods. Overexpression of two relevant human transcription factors in hESC-derived neural progenitors also generates BFCN. These neurons express only those markers characteristic of BFCN, generate action potentials, and form functional cholinergic synapses in murine hippocampal slice cultures. siRNA-mediated knockdown of the transcription factors blocks BFCN generation by the diffusible ligands, clearly demonstrating the factors both necessary and sufficient for the controlled derivation of this neuronal population. The ability to selectively control the differentiation of hESCs into BFCN is a significant step both for understanding mechanisms regulating BFCN lineage commitment and for the development of both cell transplant-mediated therapeutic interventions for Alzheimer's disease and high-throughput screening for agents that promote BFCN survival.
Collapse
Affiliation(s)
- Christopher J Bissonnette
- Department of Neurology, Northwestern University's Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | | | | | | |
Collapse
|
98
|
Use and efficacy of bone morphogenetic proteins in fracture healing. INTERNATIONAL ORTHOPAEDICS 2011; 35:1271-80. [PMID: 21698428 PMCID: PMC3167450 DOI: 10.1007/s00264-011-1301-z] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2011] [Accepted: 06/04/2011] [Indexed: 01/09/2023]
Abstract
Purpose This review evaluates the application of bone morphogenetic proteins (BMPs) in delayed bone repair, aiming at a broad audience from clinicians to scientists. Next to an overview of the role of the different BMPs, their antagonists and their current applications, special attention is focused on new scientific developments improving the effects of BMP-based therapy for bone repair. Methods Publication searches in PubMed and Embase revealed 850 relevant articles on the criteria ‘BMP’ AND ‘bone repair’ (as of May 2011). The abstracts were carefully reviewed and papers were selected according to the content. Results The resulting publications showed that BMP-2 and BMP-7 are clearly the most extensively evaluated BMPs, in general with positive results on bone healing, comparable to the use of unspecific preparations such as autologous bone grafts or platelet-rich plasma. Conclusions Although the efficacy of BMPs as stimulators of bone repair has been demonstrated in model systems and clinical studies, the use of BMPs to enhance fracture healing in the clinical setting is still controversial. Issues such as when, where and how much of which BMP is the most effective and profitable to use still have to be elucidated. But optimisation of the BMP products used in combination with cheaper production methods will inevitably stimulate the clinical use of BMPs for bone fracture healing in the near future.
Collapse
|
99
|
Lopez-Coviella I, Mellott TJ, Schnitzler AC, Blusztajn JK. BMP9 protects septal neurons from axotomy-evoked loss of cholinergic phenotype. PLoS One 2011; 6:e21166. [PMID: 21695154 PMCID: PMC3113905 DOI: 10.1371/journal.pone.0021166] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 05/21/2011] [Indexed: 11/18/2022] Open
Abstract
Background Cholinergic projection from the septum to the hippocampus is crucial for normal cognitive function and degeneration of cells and nerve fibers within the septohippocampal pathway contributes to the pathophysiology of Alzheimer's disease. Bone morphogenetic protein (BMP) 9 is a cholinergic differentiating factor during development both in vivo and in vitro. Methodology/Principal Findings To determine whether BMP9 could protect the adult cholinergic septohippocampal pathway from axotomy-evoked loss of the cholinergic phenotype, we performed unilateral fimbria-fornix transection in mice and treated them with a continuous intracerebroventricular infusion of BMP9 for six days. The number of choline acetyltransferase (CHAT)-positive cells was reduced by 50% in the medial septal nucleus ipsilateral to the lesion as compared to the intact, contralateral side, and BMP9 infusion prevented this loss in a dose-dependent manner. Moreover, BMP9 prevented most of the decline of hippocampal acetylcholine levels ipsilateral to the lesion, and markedly increased CHAT, choline transporter CHT, NGF receptors p75 (NGFR-p75) and TrkA (NTRK1), and NGF protein content in both the lesioned and unlesioned hippocampi. In addition, BMP9 infusion reduced bilaterally hippocampal levels of basic FGF (FGF2) protein. Conclusions/Significance These data indicate that BMP9 administration can prevent lesion-evoked impairment of the cholinergic septohippocampal neurons in adult mice and, by inducing NGF, establishes a trophic environment for these cells.
Collapse
Affiliation(s)
- Ignacio Lopez-Coviella
- Department of Psychiatry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Tiffany J. Mellott
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Aletta C. Schnitzler
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jan K. Blusztajn
- Department of Psychiatry, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
100
|
Crivello NA, Blusztajn JK, Joseph JA, Shukitt-Hale B, Smith DE. Short-term nutritional folate deficiency in rats has a greater effect on choline and acetylcholine metabolism in the peripheral nervous system than in the brain, and this effect escalates with age. Nutr Res 2011; 30:722-30. [PMID: 21056288 DOI: 10.1016/j.nutres.2010.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 09/13/2010] [Accepted: 09/13/2010] [Indexed: 10/18/2022]
Abstract
The hypothesis of this study is that a folate-deficient diet (FD) has a greater effect on cholinergic system in the peripheral nervous system than in the brain, and that this effect escalates with age. It was tested by comparing choline and acetylcholine levels in male Sprague Dawley rats fed either control or folate-deficient diets for 10 weeks, starting at age 4 weeks (the young group) or 9 months (the adult group). Folate-deficient diet consumption resulted in depletion of plasma folate in both age groups. In young folate-deficient rats, liver and lung choline levels were significantly lower than those in the respective controls. No other significant effects of FD on choline and acetylcholine metabolism were found in young rats. In adult rats, FD consumption markedly decreased choline levels in the liver, kidneys, and heart; furthermore, choline levels in the cortex and striatum were moderately elevated, although hippocampal choline levels were not affected. Acetylcholine levels were higher in the heart, cortex, and striatum but lower in the hippocampus in adult folate-deficient rats, as compared to controls. Higher acetylcholine levels in the striatum in adult folate-deficient rats were also associated with higher dopamine release in the striatal slices. Thus, both age groups showed higher cholinergic metabolic sensitivity to FD in the peripheral nervous system than in the brain. However, compensatory abilities appeared to be better in the young group, implicating the adult group as a preferred model for further investigation of folate-choline-acetylcholine interactions and their role in brain plasticity and cognitive functions.
Collapse
Affiliation(s)
- Natalia A Crivello
- Nutrition and Neurocognition Laboratory, Jean Mayer United States Department of Agriculture Human Nutrition Research Center on Aging (JM USDA HNRCA) at Tufts University, Boston, MA 02111, USA.
| | | | | | | | | |
Collapse
|