1
|
Yang H, Liang C, Luo J, Liu X, Wang W, Zheng K, Luo D, Hou Y, Guo D, Lin D, Zheng X, Li X. Transplantation of Wnt5a-modified Bone Marrow Mesenchymal Stem Cells Promotes Recovery After Spinal Cord Injury via the PI3K/AKT Pathway. Mol Neurobiol 2024; 61:10830-10844. [PMID: 38795301 PMCID: PMC11584464 DOI: 10.1007/s12035-024-04248-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 05/16/2024] [Indexed: 05/27/2024]
Abstract
Spinal cord injury (SCI) is a severe neurological condition that can lead to paralysis or even death. This study explored the potential benefits of bone marrow mesenchymal stem cell (BMSC) transplantation for repairing SCI. BMSCs also differentiate into astrocytes within damaged spinal cord tissues hindering the cell transplantation efficacy, therefore it is crucial to enhance their neuronal differentiation rate to facilitate spinal cord repair. Wnt5a, an upstream protein in the non-classical Wnt signaling pathway, has been implicated in stem cell migration, differentiation, and neurite formation but its role in the neuronal differentiation of BMSCs remains unclear. Thus, this study investigated the role and underlying mechanisms of Wnt5a in promoting neuronal differentiation of BMSCs both in vivo and in vitro. Wnt5a enhanced neuronal differentiation of BMSCs in vitro while reducing astrocyte differentiation. Additionally, high-throughput RNA sequencing revealed a correlation between Wnt5a and phosphoinositide 3-kinase (PI3K)/protein kinase B(AKT) signaling, which was confirmed by the use of the PI3K inhibitor LY294002 to reverse the effects of Wnt5a on BMSC neuronal differentiation. Furthermore, transplantation of Wnt5a-modified BMSCs into SCI rats effectively improved the histomorphology (Hematoxylin and eosin [H&E], Nissl and Luxol Fast Blue [LFB] staining), motor function scores (Footprint test and Basso-Beattie-Bresnahan [BBB]scores)and promoted neuron production, axonal formation, and remodeling of myelin sheaths (microtubule associated protein-2 [MAP-2], growth-associated protein 43 [GAP43], myelin basic protein [MBP]), while reducing astrocyte production (glial fibrillary acidic protein [GFAP]). Therefore, targeting the Wnt5a/PI3K/AKT pathway could enhance BMSC transplantation for SCI treatment.
Collapse
Affiliation(s)
- Haimei Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Chaolun Liang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Department of Orthopedics (Joint Surgery), Guangdong Province Hospital of Chinese Medicine, Zhuhai, 519015, Guangdong, China
| | - Junhua Luo
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Xiuzhen Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Wanshun Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Kunrui Zheng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Dan Luo
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Yu Hou
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Da Guo
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Dingkun Lin
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Xiasheng Zheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China.
| | - Xing Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
2
|
Hsu SH, Chen LR, Chen KH. Primary Osteoporosis Induced by Androgen and Estrogen Deficiency: The Molecular and Cellular Perspective on Pathophysiological Mechanisms and Treatments. Int J Mol Sci 2024; 25:12139. [PMID: 39596206 PMCID: PMC11593909 DOI: 10.3390/ijms252212139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Primary osteoporosis is closely linked to hormone deficiency, which disrupts the balance of bone remodeling. It affects postmenopausal women but also significantly impacts older men. Estrogen can promote the production of osteoprotegerin, a decoy receptor for RANKL, thereby preventing RANKL from activating osteoclasts. Furthermore, estrogen promotes osteoblast survival and function via activation of the Wnt signaling pathway. Likewise, androgens play a critical role in bone metabolism, primarily through their conversion to estrogen in men. Estrogen deficiency accelerates bone resorption through a rise in pro-inflammatory cytokines (IL-1, IL-6, TNF-α) and RANKL, which promote osteoclastogenesis. In the classic genomic pathway, estrogen binds to estrogen receptors in the cytoplasm, forming a complex that migrates to the nucleus and binds to estrogen response elements on DNA, regulating gene transcription. Androgens can be defined as high-affinity ligands for the androgen receptor; their combination can serve as a ligand-inducible transcription factor. Hormone replacement therapy has shown promise but comes with associated risks and side effects. In contrast, the non-genomic pathway involves rapid signaling cascades initiated at the cell membrane, influencing cellular functions without directly altering gene expression. Therefore, the ligand-independent actions and rapid signaling pathways of estrogen and androgen receptors can be harnessed to develop new drugs that provide bone protection without the side effects of traditional hormone therapies. To manage primary osteoporosis, other pharmacological treatments (bisphosphonates, teriparatide, RANKL inhibitors, sclerostin inhibitors, SERMs, and calcitonin salmon) can ameliorate osteoporosis and improve BMD via actions on different pathways. Non-pharmacological treatments include nutritional support and exercise, as well as the dietary intake of antioxidants and natural products. The current study reviews the processes of bone remodeling, hormone actions, hormone receptor status, and therapeutic targets of primary osteoporosis. However, many detailed cellular and molecular mechanisms underlying primary osteoporosis seem complicated and unexplored and warrant further investigation.
Collapse
Affiliation(s)
- Shao-Heng Hsu
- Department of Medical Education, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, New Taipei City 231, Taiwan;
| | - Li-Ru Chen
- Department of Physical Medicine and Rehabilitation, Mackay Memorial Hospital, Taipei 104, Taiwan;
- Department of Mechanical Engineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Kuo-Hu Chen
- Department of Obstetrics and Gynecology, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, New Taipei City 231, Taiwan
- School of Medicine, Tzu-Chi University, Hualien 970, Taiwan
| |
Collapse
|
3
|
Dennhag N, Kahsay A, Nissen I, Nord H, Chermenina M, Liu J, Arner A, Liu JX, Backman LJ, Remeseiro S, von Hofsten J, Pedrosa Domellöf F. fhl2b mediates extraocular muscle protection in zebrafish models of muscular dystrophies and its ectopic expression ameliorates affected body muscles. Nat Commun 2024; 15:1950. [PMID: 38431640 PMCID: PMC10908798 DOI: 10.1038/s41467-024-46187-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
In muscular dystrophies, muscle fibers loose integrity and die, causing significant suffering and premature death. Strikingly, the extraocular muscles (EOMs) are spared, functioning well despite the disease progression. Although EOMs have been shown to differ from body musculature, the mechanisms underlying this inherent resistance to muscle dystrophies remain unknown. Here, we demonstrate important differences in gene expression as a response to muscle dystrophies between the EOMs and trunk muscles in zebrafish via transcriptomic profiling. We show that the LIM-protein Fhl2 is increased in response to the knockout of desmin, plectin and obscurin, cytoskeletal proteins whose knockout causes different muscle dystrophies, and contributes to disease protection of the EOMs. Moreover, we show that ectopic expression of fhl2b can partially rescue the muscle phenotype in the zebrafish Duchenne muscular dystrophy model sapje, significantly improving their survival. Therefore, Fhl2 is a protective agent and a candidate target gene for therapy of muscular dystrophies.
Collapse
Affiliation(s)
- Nils Dennhag
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | - Abraha Kahsay
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | - Itzel Nissen
- Department of Medical and Translational Biology; Section of Molecular Medicine, Umeå University, Umeå, Sweden
- Wallenberg Center for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Hanna Nord
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Maria Chermenina
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | - Jiao Liu
- Div. Thoracic Surgery, Dept. Clinical Sciences, Lund University, Lund, Sweden
- College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Anders Arner
- Div. Thoracic Surgery, Dept. Clinical Sciences, Lund University, Lund, Sweden
| | - Jing-Xia Liu
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Ludvig J Backman
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Silvia Remeseiro
- Department of Medical and Translational Biology; Section of Molecular Medicine, Umeå University, Umeå, Sweden
- Wallenberg Center for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Jonas von Hofsten
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden.
| | - Fatima Pedrosa Domellöf
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden.
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden.
| |
Collapse
|
4
|
Rifai A, Weerasinghe DK, Tilaye GA, Nisbet D, Hodge JM, Pasco JA, Williams LJ, Samarasinghe RM, Williams RJ. Biofabrication of functional bone tissue: defining tissue-engineered scaffolds from nature. Front Bioeng Biotechnol 2023; 11:1185841. [PMID: 37614632 PMCID: PMC10444209 DOI: 10.3389/fbioe.2023.1185841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/24/2023] [Indexed: 08/25/2023] Open
Abstract
Damage to bone leads to pain and loss of movement in the musculoskeletal system. Although bone can regenerate, sometimes it is damaged beyond its innate capacity. Research interest is increasingly turning to tissue engineering (TE) processes to provide a clinical solution for bone defects. Despite the increasing biomimicry of tissue-engineered scaffolds, significant gaps remain in creating the complex bone substitutes, which include the biochemical and physical conditions required to recapitulate bone cells' natural growth, differentiation and maturation. Combining advanced biomaterials with new additive manufacturing technologies allows the development of 3D tissue, capable of forming cell aggregates and organoids based on natural and stimulated cues. Here, we provide an overview of the structure and mechanical properties of natural bone, the role of bone cells, the remodelling process, cytokines and signalling pathways, causes of bone defects and typical treatments and new TE strategies. We highlight processes of selecting biomaterials, cells and growth factors. Finally, we discuss innovative tissue-engineered models that have physiological and anatomical relevance for cancer treatments, injectable stimuli gels, and other therapeutic drug delivery systems. We also review current challenges and prospects of bone TE. Overall, this review serves as guide to understand and develop better tissue-engineered bone designs.
Collapse
Affiliation(s)
- Aaqil Rifai
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - D. Kavindi Weerasinghe
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Gebreselassie Addisu Tilaye
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - David Nisbet
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, VIC, Australia
- Laboratory of Advanced Biomaterials, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Aikenhead Centre for Medical Discovery, St. Vincent’s Hospital, Melbourne, VIC, Australia
| | - Jason M. Hodge
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| | - Julie A. Pasco
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Department of Medicine-Western Health, The University of Melbourne, St Albans, VIC, Australia
| | - Lana J. Williams
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| | - Rasika M. Samarasinghe
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Richard J. Williams
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Aikenhead Centre for Medical Discovery, St. Vincent’s Hospital, Melbourne, VIC, Australia
| |
Collapse
|
5
|
Zhang X, Wang G, Wang W, Ran C, Piao F, Ma Z, Zhang Z, Zheng G, Cao F, Xie H, Cui D, Samuel Okoye C, Yu X, Wang Z, Zhao D. Bone marrow mesenchymal stem cells paracrine TGF-β1 to mediate the biological activity of osteoblasts in bone repair. Cytokine 2023; 164:156139. [PMID: 36738525 DOI: 10.1016/j.cyto.2023.156139] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Bone marrow mesenchymal stem cells (BMSCs) are an important source of seed cells for regenerative medicine and tissue engineering therapy. BMSCs have multiple differentiation potentials and can release paracrine factors to facilitate tissue repair. Although the role of the osteogenic differentiation of BMSCs has been fully confirmed, the function and mechanism of BMSC paracrine factors in bone repair are still largely unclear. This study aimed to determine the roles of transforming growth factor beta-1 (TGF-β1) produced by BMSCs in bone tissue repair. METHODS To confirm our hypothesis, we used a Transwell system to coculture hBMSCs and human osteoblast-like cells without contact, which could not only avoid the interference of the osteogenic differentiation of hBMSCs but also establish the cell-cell relationship between hBMSCs and human osteoblast-like cells and provide stable paracrine substances. In the transwell coculture system, alkaline phosphatase activity, mineralized nodule formation, cell migration and chemotaxis analysis assays were conducted. RESULTS Osteogenesis, migration and chemotaxis of osteoblast-like cells were regulated by BMSCs in a paracrine manner via the upregulation of osteogenic and migration-associated genes. A TGF-β receptor I inhibitor (LY3200882) significantly antagonized BMSC-induced biological activity and related gene expression in osteoblast-like cells. Interestingly, coculture with osteoblast-like cells significantly increased the production of TGF-β1 by BMSCs, and there was potential intercellular communication between BMSCs and osteoblast-like cells. CONCLUSIONS Our findings provide evidence that the biological mechanism of BMSC-produced TGF-β1 promotes bone regeneration and repair, providing a theoretical basis and new directions for the application of BMSC transplantation in the treatment of osteonecrosis and bone injury.
Collapse
Affiliation(s)
- Xiuzhi Zhang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Guangkuo Wang
- Department of Orthopaedics, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, China
| | - Weidan Wang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China.
| | - Chunxiao Ran
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Fengyuan Piao
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Zhijie Ma
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Zhaodong Zhang
- Department of Orthopaedics, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, China
| | - Guoshuang Zheng
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Fang Cao
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Hui Xie
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Daping Cui
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Chukwuemeka Samuel Okoye
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Xiaoming Yu
- School of Material Science and Engineering, Shenyang Ligong University, Shenyang 110159, China
| | - Ziming Wang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Dewei Zhao
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China.
| |
Collapse
|
6
|
Clemente-Olivo MP, Hernández-Quiles M, Sparrius R, van der Stoel MM, Janssen V, Habibe JJ, van den Burg J, Jongejan A, Alcaraz-Sobrevals P, van Es R, Vos H, Kalkhoven E, de Vries CJM. Early adipogenesis is repressed through the newly identified FHL2-NFAT5 signaling complex. Cell Signal 2023; 104:110587. [PMID: 36610523 DOI: 10.1016/j.cellsig.2023.110587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/25/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
The LIM-domain-only protein FHL2 is a modulator of signal transduction and has been shown to direct the differentiation of mesenchymal stem cells towards osteoblast and myocyte phenotypes. We hypothesized that FHL2 may simultaneously interfere with the induction of the adipocyte lineage. Therefore, we investigated the role of FHL2 in adipocyte differentiation. For these studies pre-adipocytes isolated from mouse adipose tissue and the 3T3-L1 (pre)adipocyte cell line were applied. We performed FHL2 gain of function and knockdown experiments followed by extensive RNAseq analyses and phenotypic characterization of the cells by oil-red O (ORO) lipid staining. Through affinity-purification mass spectrometry (AP-MS) novel FHL2 interacting proteins were identified. Here we report that FHL2 is expressed in pre-adipocytes and for accurate adipocyte differentiation, this protein needs to be downregulated during the early stages of adipogenesis. More specifically, constitutive overexpression of FHL2 drastically inhibits adipocyte differentiation in 3T3-L1 cells, which was demonstrated by suppressed activation of the adipogenic gene expression program as shown by RNAseq analyses, and diminished lipid accumulation. Analysis of the protein-protein interactions mediating this repressive activity of FHL2 on adipogenesis revealed the interaction of FHL2 with the Nuclear factor of activated T-cells 5 (NFAT5). NFAT5 is an established inhibitor of adipocyte differentiation and its knockdown rescued the inhibitory effect of FHL2 overexpression on 3T3-L1 differentiation, indicating that these proteins act cooperatively. We present a new regulatory function of FHL2 in early adipocyte differentiation and revealed that FHL2-mediated inhibition of pre-adipocyte differentiation is dependent on its interaction with NFAT5. FHL2 expression increases with aging, which may affect mesenchymal stem cell differentiation, more specifically inhibit adipocyte differentiation.
Collapse
Affiliation(s)
- Maria P Clemente-Olivo
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam, the Netherlands
| | - Miguel Hernández-Quiles
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Rinske Sparrius
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands
| | - Miesje M van der Stoel
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands
| | - Vera Janssen
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands
| | - Jayron J Habibe
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam, the Netherlands
| | - Janny van den Burg
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands
| | - Aldo Jongejan
- Amsterdam UMC location University of Amsterdam, Department of Bioinformatics, Amsterdam, the Netherlands
| | - Paula Alcaraz-Sobrevals
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Robert van Es
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Harmjan Vos
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Eric Kalkhoven
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Carlie J M de Vries
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
7
|
Progress of Wnt Signaling Pathway in Osteoporosis. Biomolecules 2023; 13:biom13030483. [PMID: 36979418 PMCID: PMC10046187 DOI: 10.3390/biom13030483] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Osteoporosis, one of the serious health diseases, involves bone mass loss, bone density diminishing, and degeneration of bone microstructure, which is accompanied by a tendency toward bone fragility and a predisposition to fracture. More than 200 million people worldwide suffer from osteoporosis, and the cost of treating osteoporotic fractures is expected to reach at least $25 billion by 2025. The generation and development of osteoporosis are regulated by genetic factors and regulatory factors such as TGF-β, BMP, and FGF through multiple pathways, including the Wnt signaling pathway, the Notch signaling pathway, and the MAPK signaling pathway. Among them, the Wnt signaling pathway is one of the most important pathways. It is not only involved in bone development and metabolism but also in the differentiation and proliferation of chondrocytes, mesenchymal stem cells, osteoclasts, and osteoblasts. Dkk-1 and SOST are Wnt inhibitory proteins that can inhibit the activation of the canonical Wnt signaling pathway and block the proliferation and differentiation of osteoblasts. Therefore, they may serve as potential targets for the treatment of osteoporosis. In this review, we analyzed the mechanisms of Wnt proteins, β-catenin, and signaling molecules in the process of signal transduction and summarized the relationship between the Wnt signaling pathway and bone-related cells. We hope to attract attention to the role of the Wnt signaling pathway in osteoporosis and offer new perspectives and approaches to making a diagnosis and giving treatment for osteoporosis.
Collapse
|
8
|
Addressing posterior tilt displacement during surgery to lower failure risk of sub-capital Garden types 1 and 2 femoral fractures. Arch Orthop Trauma Surg 2022; 142:1885-1893. [PMID: 33959801 DOI: 10.1007/s00402-021-03900-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/05/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Sub-capital femoral fractures (SCFF) are impacted or non-displaced in Garden types 1 and 2, respectively. Non-surgical treatment is protected weight-bearing combined with physiotherapy and radiographic follow-up in selected patients. Traditionally, in situ pinning is the surgical treatment of choice. The aim of this study was to estimate whether the valgus deformity in Garden types 1 and 2 (AO classification 31B1.1 and 31B1.2) SCFF is a virtual perception of a posterior tilt deformity and if addressing this deformity improves patients' outcomes. MATERIALS AND METHODS The records of 96 patients with Garden Types 1 and 2 SCFF treated in tertiary medical center between 1/2014 and 9/2017 were retrospectively reviewed. They all had preoperative hip joint anteroposterior and lateral radiographic views. 75 patients had additional computed tomography (CT) scans. Femoral head displacement was measured on an anteroposterior and axial radiograph projections and were performed before and after surgery. Preoperative 3D reconstructions were performed for a better fracture characterization, and assessment of the imaging was performed by the first author. RESULTS The average age of the study cohort was 73 years (range 28-96, 68% females). There were 58 right-sided and 38 left-sided fractures. Ninety patients had Type 1 and six patients had Type 2 fractures. The average preoperative posterior tilt was 15 degrees and the average valgus displacement was 10 degrees on plain radiographs compared to 28 degrees and 11 degrees, respectively, on CT scans. Posterior tilt was found with a virtual perception as valgus-impacted fractures. The postoperative posterior tilt was corrected to an average of 3 degrees and the valgus displacement to 5 degrees. CONCLUSION CT provides an accurate modality for measuring femoral head displacement and fracture extent. The posterior tilt displacement should be addressed during surgery to lower failure risk and the need for additional procedures. IRB APPROVAL TLV-0292-15. LEVEL OF EVIDENCE IV.
Collapse
|
9
|
Impact of Treadmill Interval Running on the Appearance of Zinc Finger Protein FHL2 in Bone Marrow Cells in a Rat Model: A Pilot Study. Life (Basel) 2022; 12:life12040528. [PMID: 35455019 PMCID: PMC9029125 DOI: 10.3390/life12040528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
Although the benefits of physical exercise to preserve bone quality are now widely recognized, the intimate mechanisms leading to the underlying cell responses still require further investigations. Interval training running, for instance, appears as a generator of impacts on the skeleton, and particularly on the progenitor cells located in the bone marrow. Therefore, if this kind of stimulus initiates bone cell proliferation and differentiation, the activation of a devoted signaling pathway by mechano-transduction seems likely. This study aimed at investigating the effects of an interval running program on the appearance of the zinc finger protein FHL2 in bone cells and their anatomical location. Twelve 5-week-old male Wistar rats were randomly allocated to one of the following groups (n = 6 per group): sedentary control (SED) or high-intensity interval running (EX, 8 consecutive weeks). FHL2 identification in bone cells was performed by immuno-histochemistry on serial sections of radii. We hypothesized that impacts generated by running could activate, in vivo, a specific signaling pathway, through an integrin-mediated mechano-transductive process, leading to the synthesis of FHL2 in bone marrow cells. Our data demonstrated the systematic appearance of FHL2 (% labeled cells: 7.5%, p < 0.001) in bone marrow obtained from EX rats, whereas no FHL2 was revealed in SED rats. These results suggest that the mechanical impacts generated during high-intensity interval running activate a signaling pathway involving nuclear FHL2, such as that also observed with dexamethasone administration. Consequently, interval running could be proposed as a non-pharmacological strategy to contribute to bone marrow cell osteogenic differentiation.
Collapse
|
10
|
Rocha RA, Fox JM, Genever PG, Hancock Y. Biomolecular phenotyping and heterogeneity assessment of mesenchymal stromal cells using label-free Raman spectroscopy. Sci Rep 2021; 11:4385. [PMID: 33623051 PMCID: PMC7902661 DOI: 10.1038/s41598-021-81991-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/30/2020] [Indexed: 11/09/2022] Open
Abstract
Easy, quantitative measures of biomolecular heterogeneity and high-stratified phenotyping are needed to identify and characterise complex disease processes at the single-cell level, as well as to predict cell fate. Here, we demonstrate how Raman spectroscopy can be used in the difficult-to-assess case of clonal, bone-derived mesenchymal stromal cells (MSCs) to identify MSC lines and group these according to biological function (e.g., differentiation capacity). Biomolecular stratification is achieved using high-precision measures obtained from representative statistical sampling that also enable quantified heterogeneity assessment. Application to primary MSCs and human dermal fibroblasts shows use of these measures as a label-free assay to classify cell sub-types within complex heterogeneous cell populations, thus demonstrating the potential for therapeutic translation, and broad application to the phenotypic characterisation of other cells.
Collapse
Affiliation(s)
- R A Rocha
- Department of Physics, University of York, Heslington, York, YO10 5DD, UK
- Federal University of Technology-Paraná, Campus Dois Vizinhos, Paraná, 85660-000, Brazil
| | - J M Fox
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK
- York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK
| | - P G Genever
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK
- York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK
| | - Y Hancock
- Department of Physics, University of York, Heslington, York, YO10 5DD, UK.
- York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK.
- York Cross-disciplinary Centre for Systems Analysis, University of York, Heslington, York, YO30 5GG, UK.
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, SE19RT, UK.
| |
Collapse
|
11
|
Mi B, Yan C, Xue H, Chen L, Panayi AC, Hu L, Hu Y, Cao F, Sun Y, Zhou W, Xiong Y, Liu G. Inhibition of Circulating miR-194-5p Reverses Osteoporosis through Wnt5a/β-Catenin-Dependent Induction of Osteogenic Differentiation. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 21:814-823. [PMID: 32791453 PMCID: PMC7419275 DOI: 10.1016/j.omtn.2020.07.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/26/2020] [Accepted: 07/20/2020] [Indexed: 01/14/2023]
Abstract
Mesenchymal stem cells (MSCs) critically contribute to bone formation, and proper induction of osteogenic differentiation can lead to an increase in bone mass. In the present study, we reported that an increased miR-194-5p level in plasma is inversely related to the degree of bone formation in osteoporosis patients. We also noted that increased miR-194-5p in the MSCs of ovariectomized (OVX) mice and agomiR-194-5p manipulation of miR-194-5p significantly suppressed bone formation, both in aged and OVX mice. Furthermore, our in vitro study showed that overexpression of miR-194-5p suppresses osteogenic differentiation, as evidenced by the decreased bone formation marker genes and matrix mineralization. The luciferase assay indicated that Wnt family member 5a (Wnt5a) is a target gene of miR-194-5p that positively regulates osteogenic differentiation. Collectively, these data indicated that miR-194-5p inhibition may be a potential strategy for osteoporosis prevention.
Collapse
Affiliation(s)
- Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chenchen Yan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hang Xue
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lang Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Adriana C Panayi
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Liangcong Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yiqiang Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Faqi Cao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yun Sun
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wu Zhou
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuan Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Guohui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
12
|
Ma X, Jia C, Chu M, Fu D, Lei Q, Ding X, Wu X, Guo X, Pei J, Bao P, Yan P, Liang C. Transcriptome and DNA Methylation Analyses of the Molecular Mechanisms Underlying with Longissimus dorsi Muscles at Different Stages of Development in the Polled Yak. Genes (Basel) 2019; 10:genes10120970. [PMID: 31779203 PMCID: PMC6947547 DOI: 10.3390/genes10120970] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 02/04/2023] Open
Abstract
DNA methylation modifications are implicated in many biological processes. As the most common epigenetic mechanism DNA methylation also affects muscle growth and development. The majority of previous studies have focused on different varieties of yak, but little is known about the epigenetic regulation mechanisms in different age groups of animals. The development of muscles in the different stages of yak growth remains unclear. In this study, we selected the longissimus dorsi muscle tissue at three different growth stages of the yak, namely, 90-day-old fetuses (group E), six months old (group M), and three years old (group A). Using RNA-Seq transcriptome sequencing and methyl-RAD whole-genome methylation sequencing technology, changes in gene expression levels and DNA methylation status throughout the genome were investigated during the stages of yak development. Each group was represented by three biological replicates. The intersections of expression patterns of 7694 differentially expressed genes (DEGs) were identified (padj < 0.01, |log2FC| > 1.2) at each of the three developmental periods. Time-series expression profile clustering analysis indicated that the DEGs were significantly arranged into eight clusters which could be divided into two classes (padj < 0.05), class I profiles that were downregulated and class II profiles that were upregulated. Based on this cluster analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that DEGs from class I profiles were significantly (padj < 0.05) enriched in 21 pathways, the most enriched pathway being the Axon guidance signaling pathway. DEGs from the class II profile were significantly enriched in 58 pathways, the pathway most strongly enriched being Metabolic pathway. After establishing the methylation profiles of the whole genomes, and using two groups of comparisons, the three combinations of groups (M-vs.-E, M-vs.-A, A-vs.-E) were found to have 1344, 822, and 420 genes, respectively, that were differentially methylated at CCGG sites and 2282, 3056, and 537 genes, respectively, at CCWGG sites. The two sets of data were integrated and the negative correlations between DEGs and differentially methylated promoters (DMPs) analyzed, which confirmed that TMEM8C, IGF2, CACNA1S and MUSTN1 were methylated in the promoter region and that expression of the modified genes was negatively correlated. Interestingly, these four genes, from what was mentioned above, perform vital roles in yak muscle growth and represent a reference for future genomic and epigenomic studies in muscle development, in addition to enabling marker-assisted selection of growth traits.
Collapse
Affiliation(s)
- Xiaoming Ma
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (X.M.); (C.J.); (M.C.); (D.F.); (Q.L.); (X.D.); (X.W.); (X.G.); (J.P.); (P.B.)
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Congjun Jia
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (X.M.); (C.J.); (M.C.); (D.F.); (Q.L.); (X.D.); (X.W.); (X.G.); (J.P.); (P.B.)
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Min Chu
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (X.M.); (C.J.); (M.C.); (D.F.); (Q.L.); (X.D.); (X.W.); (X.G.); (J.P.); (P.B.)
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Donghai Fu
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (X.M.); (C.J.); (M.C.); (D.F.); (Q.L.); (X.D.); (X.W.); (X.G.); (J.P.); (P.B.)
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Qinhui Lei
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (X.M.); (C.J.); (M.C.); (D.F.); (Q.L.); (X.D.); (X.W.); (X.G.); (J.P.); (P.B.)
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Xuezhi Ding
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (X.M.); (C.J.); (M.C.); (D.F.); (Q.L.); (X.D.); (X.W.); (X.G.); (J.P.); (P.B.)
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Xiaoyun Wu
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (X.M.); (C.J.); (M.C.); (D.F.); (Q.L.); (X.D.); (X.W.); (X.G.); (J.P.); (P.B.)
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Xian Guo
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (X.M.); (C.J.); (M.C.); (D.F.); (Q.L.); (X.D.); (X.W.); (X.G.); (J.P.); (P.B.)
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Jie Pei
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (X.M.); (C.J.); (M.C.); (D.F.); (Q.L.); (X.D.); (X.W.); (X.G.); (J.P.); (P.B.)
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Pengjia Bao
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (X.M.); (C.J.); (M.C.); (D.F.); (Q.L.); (X.D.); (X.W.); (X.G.); (J.P.); (P.B.)
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Ping Yan
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (X.M.); (C.J.); (M.C.); (D.F.); (Q.L.); (X.D.); (X.W.); (X.G.); (J.P.); (P.B.)
- Correspondence: (P.Y.); (C.L.); Tel.: +86-0931-2115288 (P.Y.); +86-0931-2115271 (C.L.)
| | - Chunnian Liang
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Correspondence: (P.Y.); (C.L.); Tel.: +86-0931-2115288 (P.Y.); +86-0931-2115271 (C.L.)
| |
Collapse
|
13
|
Liao YP, Du WM, Hu Y, Li FS, Ma Y, Wang H, Zhu JH, Zhou Y, Li Q, Su YX, He BC. CREB/Wnt10b mediates the effect of COX-2 on promoting BMP9-induced osteogenic differentiation via reducing adipogenic differentiation in mesenchymal stem cells. J Cell Biochem 2018; 120:9572-9587. [PMID: 30525243 DOI: 10.1002/jcb.28234] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 11/15/2018] [Indexed: 01/10/2023]
Abstract
Bone morphogenetic protein 9 (BMP9) is one of the most potent osteogenic factors, which may be a potential candidate for bone tissue engineering. However, the osteogenic capacity of BMP9 still need to be further enhanced. In this study, we determined the effect of Wnt10b on BMP9-induced osteogenic differentiation in mesenchymal stem cell (MSCs) and the possible mechanism underlying this process. We introduced the polymerase chain reaction (PCR), Western blot analysis, histochemical stain, ectopic bone formation, and microcomputed tomography analysis to evaluate the effect of Wnt10b on BMP9-induced osteogenic differentiation. Meanwhile, PCR, Western blot analysis, chromatin immunoprecipitation, and immunoprecipitation were used to analyze the possible relationship between BMP9 and Wnt10b. We found that BMP9 upregulates Wnt10b in C3H10T1/2 cells. Wnt10b increases the osteogenic markers and bone formation induced by BMP9 in C3H10T1/2 cells, and silencing Wnt10b decreases these effects of BMP9. Meanwhile, Wnt10b enhances the level of phosphorylated Smad1/5/8 (p-Smad1/5/8) induced by BMP9, which can be reduced by silencing Wnt10b. On the contrary, Wnt10b inhibits adipogenic markers induced by BMP9, which can be decreased by silencing Wnt10b. Further analysis indicated that BMP9 upregulates cyclooxygenase-2 (COX-2) and phosphorylation of cAMP-responsive element binding (p-CREB) simultaneously. COX-2 potentiates the effect of BMP9 on increasing p-CREB and Wnt10b, while silencing COX-2 decreases these effects. p-CREB interacts with p-Smad1/5/8 to bind the promoter of Wnt10b in C3H10T1/2 cells. Our findings suggested that Wnt10b can promote BMP9-induced osteogenic differentiation in MSCs, which may be mediated through enhancing BMP/Smad signal and reducing adipogenic differentiation; BMP9 may upregulate Wnt10b via the COX-2/p-CREB-dependent manner.
Collapse
Affiliation(s)
- Yun-Peng Liao
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, People's Republic of China
| | - Wei-Min Du
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, People's Republic of China
| | - Ying Hu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, People's Republic of China
| | - Fu-Shu Li
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yan Ma
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, People's Republic of China
| | - Han Wang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, People's Republic of China
| | - Jia-Hui Zhu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, People's Republic of China
| | - Ya Zhou
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, People's Republic of China
| | - Qin Li
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yu-Xi Su
- Department of Orthopedic, Children Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Bai-Cheng He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
14
|
Zhang X, Chen J, Pei X, Wang J, Wan Q, Jiang S, Huang C, Pei X. Enhanced Osseointegration of Porous Titanium Modified with Zeolitic Imidazolate Framework-8. ACS APPLIED MATERIALS & INTERFACES 2017; 9:25171-25183. [PMID: 28696091 DOI: 10.1021/acsami.7b07800] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Xin Zhang
- State
Key Laboratory of Oral Diseases, National Clinical Research
Center for Oral Diseases, §Department of Prosthodontics, West China Hospital of
Stomatology, ⊥Postanesthesia Care Unit, West China Hospital of Stomatology, and ∥College of Chemistry, Sichuan University, Chengdu 610000, P.R. China
| | - Junyu Chen
- State
Key Laboratory of Oral Diseases, National Clinical Research
Center for Oral Diseases, §Department of Prosthodontics, West China Hospital of
Stomatology, ⊥Postanesthesia Care Unit, West China Hospital of Stomatology, and ∥College of Chemistry, Sichuan University, Chengdu 610000, P.R. China
| | - Xiang Pei
- State
Key Laboratory of Oral Diseases, National Clinical Research
Center for Oral Diseases, §Department of Prosthodontics, West China Hospital of
Stomatology, ⊥Postanesthesia Care Unit, West China Hospital of Stomatology, and ∥College of Chemistry, Sichuan University, Chengdu 610000, P.R. China
| | - Jian Wang
- State
Key Laboratory of Oral Diseases, National Clinical Research
Center for Oral Diseases, §Department of Prosthodontics, West China Hospital of
Stomatology, ⊥Postanesthesia Care Unit, West China Hospital of Stomatology, and ∥College of Chemistry, Sichuan University, Chengdu 610000, P.R. China
| | - Qianbing Wan
- State
Key Laboratory of Oral Diseases, National Clinical Research
Center for Oral Diseases, §Department of Prosthodontics, West China Hospital of
Stomatology, ⊥Postanesthesia Care Unit, West China Hospital of Stomatology, and ∥College of Chemistry, Sichuan University, Chengdu 610000, P.R. China
| | - Shaokang Jiang
- State
Key Laboratory of Oral Diseases, National Clinical Research
Center for Oral Diseases, §Department of Prosthodontics, West China Hospital of
Stomatology, ⊥Postanesthesia Care Unit, West China Hospital of Stomatology, and ∥College of Chemistry, Sichuan University, Chengdu 610000, P.R. China
| | - Chao Huang
- State
Key Laboratory of Oral Diseases, National Clinical Research
Center for Oral Diseases, §Department of Prosthodontics, West China Hospital of
Stomatology, ⊥Postanesthesia Care Unit, West China Hospital of Stomatology, and ∥College of Chemistry, Sichuan University, Chengdu 610000, P.R. China
| | - Xibo Pei
- State
Key Laboratory of Oral Diseases, National Clinical Research
Center for Oral Diseases, §Department of Prosthodontics, West China Hospital of
Stomatology, ⊥Postanesthesia Care Unit, West China Hospital of Stomatology, and ∥College of Chemistry, Sichuan University, Chengdu 610000, P.R. China
| |
Collapse
|
15
|
Plikus MV, Guerrero-Juarez CF, Ito M, Li YR, Dedhia PH, Zheng Y, Shao M, Gay DL, Ramos R, Hsi TC, Oh JW, Wang X, Ramirez A, Konopelski SE, Elzein A, Wang A, Supapannachart RJ, Lee HL, Lim CH, Nace A, Guo A, Treffeisen E, Andl T, Ramirez RN, Murad R, Offermanns S, Metzger D, Chambon P, Widgerow AD, Tuan TL, Mortazavi A, Gupta RK, Hamilton BA, Millar SE, Seale P, Pear WS, Lazar MA, Cotsarelis G. Regeneration of fat cells from myofibroblasts during wound healing. Science 2017; 355:748-752. [PMID: 28059714 DOI: 10.1126/science.aai8792] [Citation(s) in RCA: 423] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/19/2016] [Indexed: 12/14/2022]
Abstract
Although regeneration through the reprogramming of one cell lineage to another occurs in fish and amphibians, it has not been observed in mammals. We discovered in the mouse that during wound healing, adipocytes regenerate from myofibroblasts, a cell type thought to be differentiated and nonadipogenic. Myofibroblast reprogramming required neogenic hair follicles, which triggered bone morphogenetic protein (BMP) signaling and then activation of adipocyte transcription factors expressed during development. Overexpression of the BMP antagonist Noggin in hair follicles or deletion of the BMP receptor in myofibroblasts prevented adipocyte formation. Adipocytes formed from human keloid fibroblasts either when treated with BMP or when placed with human hair follicles in vitro. Thus, we identify the myofibroblast as a plastic cell type that may be manipulated to treat scars in humans.
Collapse
Affiliation(s)
- Maksim V Plikus
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA. .,Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Christian F Guerrero-Juarez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Mayumi Ito
- The Ronald O. Perelman Department of Dermatology, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Yun Rose Li
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Priya H Dedhia
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ying Zheng
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mengle Shao
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Denise L Gay
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,INSERM U967, Commissariat à L'énergie Atomique et aux Énergies Alternatives, Institut de Radiobiologie Cellulaire et Moléculaire 92265 Fontenay-aux-Roses Cedex, France
| | - Raul Ramos
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Tsai-Ching Hsi
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Ji Won Oh
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA.,Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Xiaojie Wang
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Amanda Ramirez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Sara E Konopelski
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Arijh Elzein
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Anne Wang
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rarinthip June Supapannachart
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hye-Lim Lee
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Chae Ho Lim
- The Ronald O. Perelman Department of Dermatology, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Arben Nace
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amy Guo
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elsa Treffeisen
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thomas Andl
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 328116, USA
| | - Ricardo N Ramirez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Rabi Murad
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Daniel Metzger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, INSERM U964, Université de Strasbourg, Illkirch 67404, France
| | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, INSERM U964, Institut d'Etudes Avancées de l'Université de Strasbourg, Collège de France, Illkirch 67404, France
| | - Alan D Widgerow
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, Irvine, CA 92868, USA
| | - Tai-Lan Tuan
- The Saban Research Institute of Children's Hospital Los Angeles and Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bruce A Hamilton
- Departments of Medicine and Cellular and Molecular Medicine, Moores Cancer Center and Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sarah E Millar
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patrick Seale
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Warren S Pear
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mitchell A Lazar
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - George Cotsarelis
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
16
|
Cheng X, Wei B, Sun L, Hu X, Liang J, Chen Y. Astragaloside I Stimulates Osteoblast Differentiation Through the Wnt/β-catenin Signaling Pathway. Phytother Res 2016; 30:1680-1688. [PMID: 27397144 DOI: 10.1002/ptr.5674] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/10/2016] [Accepted: 06/04/2016] [Indexed: 11/10/2022]
Abstract
Astragaloside I (As-I), one of the main active ingredients in Astragalus membranaceus, is believed to have osteogenic properties, but this hypothesis has not been investigated in detail. In the present work, the As-I-induced osteogenic effects and its underlying mechanism were studied in MC3T3-E1 cells. The results indicated that the cellular levels of ALP and extracellular matrix calcium increased in a dose-dependent manner by As-I. To clarify the mechanisms involved in this process, the effect of As-I on the key osteogenic-related genes was investigated. We found that As-I stimulated the expression of β-catenin and Runx2 in MC3T3-E1 cells, which play central roles in the Wnt/β-catenin signaling pathway, suggesting that As-I could promote osteoblastic differentiation by regulating the Wnt/β-catenin signaling pathway. Moreover, the osteogenic effect of As-I could be inhibited by DKK-1, which is the classical inhibitor of Wnt/β-catenin-signaling pathway. Furthermore, As-I also increased BMP-2, BGP and OPG/RANKL expression, which are also activated by Wnt/β-catenin signaling pathway. Taken together, our findings show that As-I stimulates osteoblast differentiation through the Wnt/β-catenin signaling pathway, which also activates the BMP pathway and RANK pathway, thus highlighting the As-I for pharmaceutical and medicinal applications such as treating bone disease. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Xun Cheng
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei University, Wuhan, 430062, China
| | - Biaofang Wei
- Department of Orthopaedic, Linyi People's Hospital, Science and Technology Develop Project of Shandong province, Linyi, 276000, China
| | - Lijuan Sun
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei University, Wuhan, 430062, China.
| | - Xiaofang Hu
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei University, Wuhan, 430062, China
| | - Jichao Liang
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei University, Wuhan, 430062, China
| | - Yong Chen
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei University, Wuhan, 430062, China.
| |
Collapse
|
17
|
TLR4 Activation Promotes Bone Marrow MSC Proliferation and Osteogenic Differentiation via Wnt3a and Wnt5a Signaling. PLoS One 2016; 11:e0149876. [PMID: 26930594 PMCID: PMC4773221 DOI: 10.1371/journal.pone.0149876] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 02/06/2016] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) from adult bone marrow maintain their self-renewal ability and the ability to differentiate into osteoblast. Thus, adult bone marrow MSCs play a key role in the regeneration of bone tissue. Previous studies indicated that TLR4 is expressed in MSCs and is critical in regulating the fate decision of MSCs. However, the exact functional role and underlying mechanisms of how TLR4 regulate bone marrow MSC proliferation and differentiation are unclear. Here, we found that activated TLR4 by its ligand LPS promoted the proliferation and osteogenic differentiation of MSCs in vitro. TLR4 activation by LPS also increased cytokine IL-6 and IL-1β production in MSCs. In addition, LPS treatment has no effect on inducing cell death of MSCs. Deletion of TLR4 expression in MSCs completely eliminated the effects of LPS on MSC proliferation, osteogenic differentiation and cytokine production. We also found that the mRNA and protein expression of Wnt3a and Wnt5a, two important factors in regulating MSC fate decision, was upregulated in a TLR4-dependent manner. Silencing Wnt3a with specific siRNA remarkably inhibited TLR4-induced MSC proliferation, while Wnt5a specific siRNA treatment significantly antagonized TLR4-induced MSC osteogenic differentiation. These results together suggested that TLR4 regulates bone marrow MSC proliferation and osteogenic differentiation through Wnt3a and Wnt5a signaling. These finding provide new data to understand the role and the molecular mechanisms of TLR4 in regulating bone marrow MSC functions. These data also provide new insight in developing new therapy in bone regeneration using MSCs by modulating TLR4 and Wnt signaling activity.
Collapse
|
18
|
Comprehensive transcriptomic and proteomic characterization of human mesenchymal stem cells reveals source specific cellular markers. Sci Rep 2016; 6:21507. [PMID: 26857143 PMCID: PMC4746666 DOI: 10.1038/srep21507] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/26/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSC) are multipotent cells with great potential in therapy, reflected by more than 500 MSC-based clinical trials registered with the NIH. MSC are derived from multiple tissues but require invasive harvesting and imply donor-to-donor variability. Embryonic stem cell-derived MSC (ESC-MSC) may provide an alternative, but how similar they are to ex vivo MSC is unknown. Here we performed an in depth characterization of human ESC-MSC, comparing them to human bone marrow-derived MSC (BM-MSC) as well as human embryonic stem cells (hESC) by transcriptomics (RNA-seq) and quantitative proteomics (nanoLC-MS/MS using SILAC). Data integration highlighted and validated a central role of vesicle-mediated transport and exosomes in MSC biology and also demonstrated, through enrichment analysis, their versatility and broad application potential. Particular emphasis was placed on comparing profiles between ESC-MSC and BM-MSC and assessing their equivalency. Data presented here shows that differences between ESC-MSC and BM-MSC are similar in magnitude to those reported for MSC of different origin and the former may thus represent an alternative source for therapeutic applications. Finally, we report an unprecedented coverage of MSC CD markers, as well as membrane associated proteins which may benefit immunofluorescence-based applications and contribute to a refined molecular description of MSC.
Collapse
|
19
|
Wu SM, Shih LH, Lee JY, Shen YJ, Lee HH. Estrogen enhances activity of Wnt signaling during osteogenesis by inducing Fhl1 expression. J Cell Biochem 2016; 116:1419-30. [PMID: 25676585 DOI: 10.1002/jcb.25102] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 01/23/2015] [Indexed: 01/13/2023]
Abstract
Estrogen is a crucial hormone for osteoclast inhibition and for preventing osteoporosis. However, the hormone's role in osteoblast growth and differentiation remains unclear. The complexity of estrogen's role in guiding osteoblast behavior arises partly from crosstalk with other signaling pathways, including Wnt signaling. In this study, we show that the Wnt agonist, LiCl, induced Fhl1 gene expression, which substantially enhanced osteoblast differentiation. Staining with alizarin red revealed that MC3T3-E1 mineralization was enhanced by overexpression of Fhl1. In addition, Fhl1 promoted the expression of the osteogenic markers, Runt-related transcription factor 2 (Runx2), osteocalcin (OCN), and osteopontin (OPN), whereas MC3T3-E1 cells with gene knockdown of Fhl1 exhibited limited mineralization and expression of Runx2, OCN, and OPN. We further demonstrate evidences from quantitative reverse transcription real-time polymerase chain reaction and reporter assay that Fhl1 expression was synergistically stimulated by estrogen (E2) and LiCl, but reduced by the estrogen-receptor inhibitor fulvestrant (ICI 182,780). However, estrogen could not enhance osteogenesis while Fhl1 expression was knocked down. Because estrogen and Wnt signaling frequently interact in developmental processes, we propose that Fhl1 can be an acting molecule mediating both signaling pathways during osteogenesis.
Collapse
Affiliation(s)
- Shao-Min Wu
- Department of Bio-Agricultural Sciences, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Lan-Hsin Shih
- Department of Bio-Agricultural Sciences, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Jing-Yu Lee
- Department of Bio-Agricultural Sciences, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Yi-Jun Shen
- Department of Bio-Agricultural Sciences, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Hu-Hui Lee
- Department of Bio-Agricultural Sciences, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| |
Collapse
|
20
|
Mindaye ST, Surdo JL, Bauer SR, Alterman MA. System-wide survey of proteomic responses of human bone marrow stromal cells (hBMSCs) to in vitro cultivation. Stem Cell Res 2015; 15:655-664. [PMID: 26523674 DOI: 10.1016/j.scr.2015.09.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 09/21/2015] [Accepted: 09/26/2015] [Indexed: 01/12/2023] Open
Abstract
Human bone marrow stromal cells (hBMSCs, also loosely called bone marrow-derived mesenchymal stem cells) are the subject of increasing numbers of clinical trials and laboratory research. Our group recently reported on the optimization of a workflow for a sensitive proteomic study of hBMSCs. Here, we couple this workflow with a label-free protein quantitation method to investigate the molecular responses of hBMSCs to long-term in vitro passaging. We explored the proteomic responses of hBMSCs by assessing the expression levels of proteins at early passage (passage 3, P3) and late passage (P7). We used multiple biological as well as technical replicates to ensure that the detected proteomic changes are repeatable between cultures and thus likely to be biologically relevant. Over 1700 proteins were quantified at three passages and a list of differentially expressed proteins was compiled. Bioinformatics-based network analysis and term enrichment revealed that metabolic pathways are largely altered, where many proteins in the glycolytic, pentose phosphate, and TCA pathways were shown to be largely upregulated in late passages. We also observed significant proteomic alterations in functional categories including apoptosis, and ER-based protein processing and sorting following in vitro cell aging. We posit that the comprehensive map outlined in this report of affected phenotypes as well as the underpinning molecular factors tremendously benefit the effort to uncovering targets that are not just used only to monitor cell fitness but can be employed to slowdown the in vitro aging process in hBMSCs and hence ensure manufacturing of cells with known quality, efficacy and stability.
Collapse
Affiliation(s)
- Samuel T Mindaye
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Jessica Lo Surdo
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Steven R Bauer
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Michail A Alterman
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States.
| |
Collapse
|
21
|
Cancer-Osteoblast Interaction Reduces Sost Expression in Osteoblasts and Up-Regulates lncRNA MALAT1 in Prostate Cancer. MICROARRAYS 2015; 4:503-19. [PMID: 27600237 PMCID: PMC4996404 DOI: 10.3390/microarrays4040503] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/17/2015] [Accepted: 10/22/2015] [Indexed: 12/29/2022]
Abstract
Dynamic interaction between prostate cancer and the bone microenvironment is a major contributor to metastasis of prostate cancer to bone. In this study, we utilized an in vitro co-culture model of PC3 prostate cancer cells and osteoblasts followed by microarray based gene expression profiling to identify previously unrecognized prostate cancer–bone microenvironment interactions. Factors secreted by PC3 cells resulted in the up-regulation of many genes in osteoblasts associated with bone metabolism and cancer metastasis, including Mmp13, Il-6 and Tgfb2, and down-regulation of Wnt inhibitor Sost. To determine whether altered Sost expression in the bone microenvironment has an effect on prostate cancer metastasis, we co-cultured PC3 cells with Sost knockout (SostKO) osteoblasts and wildtype (WT) osteoblasts and identified several genes differentially regulated between PC3-SostKO osteoblast co-cultures and PC3-WT osteoblast co-cultures. Co-culturing PC3 cells with WT osteoblasts up-regulated cancer-associated long noncoding RNA (lncRNA) MALAT1 in PC3 cells. MALAT1 expression was further enhanced when PC3 cells were co-cultured with SostKO osteoblasts and treatment with recombinant Sost down-regulated MALAT1 expression in these cells. Our results suggest that reduced Sost expression in the tumor microenvironment may promote bone metastasis by up-regulating MALAT1 in prostate cancer.
Collapse
|
22
|
Marie PJ. Osteoblast dysfunctions in bone diseases: from cellular and molecular mechanisms to therapeutic strategies. Cell Mol Life Sci 2015; 72:1347-61. [PMID: 25487608 PMCID: PMC11113967 DOI: 10.1007/s00018-014-1801-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 11/13/2014] [Accepted: 12/01/2014] [Indexed: 12/27/2022]
Abstract
Several metabolic, genetic and oncogenic bone diseases are characterized by defective or excessive bone formation. These abnormalities are caused by dysfunctions in the commitment, differentiation or survival of cells of the osteoblast lineage. During the recent years, significant advances have been made in our understanding of the cellular and molecular mechanisms underlying the osteoblast dysfunctions in osteoporosis, skeletal dysplasias and primary bone tumors. This led to suggest novel therapeutic approaches to correct these abnormalities such as the modulation of WNT signaling, the pharmacological modulation of proteasome-mediated protein degradation, the induction of osteoprogenitor cell differentiation, the repression of cancer cell proliferation and the manipulation of epigenetic mechanisms. This article reviews our current understanding of the major cellular and molecular mechanisms inducing osteoblastic cell abnormalities in age-related bone loss, genetic skeletal dysplasias and primary bone tumors, and discusses emerging therapeutic strategies to counteract the osteoblast abnormalities in these disorders of bone formation.
Collapse
Affiliation(s)
- Pierre J Marie
- INSERM UMR-1132, Hôpital Lariboisière, 2 rue Ambroise Paré, 75475, Paris Cedex 10, France,
| |
Collapse
|
23
|
Tian X, Wang Q, Wang X. Four and a Half LIM Domain Protein 2 Enhances Differentiation and Mineralization of Human Dental Pulp Cells. J Endod 2015; 41:513-9. [DOI: 10.1016/j.joen.2014.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 12/03/2014] [Accepted: 12/09/2014] [Indexed: 12/14/2022]
|
24
|
Bouaziz W, Funck-Brentano T, Lin H, Marty C, Ea HK, Hay E, Cohen-Solal M. Loss of sclerostin promotes osteoarthritis in mice via β-catenin-dependent and -independent Wnt pathways. Arthritis Res Ther 2015; 17:24. [PMID: 25656376 PMCID: PMC4355467 DOI: 10.1186/s13075-015-0540-6] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 01/23/2015] [Indexed: 11/21/2022] Open
Abstract
Introduction Sclerostin is a Wnt inhibitor produced by osteocytes that regulates bone formation. Because bone tissue contributes to the development of osteoarthritis (OA), we investigated the role of sclerostin in bone and cartilage in a joint instability model in mice. Methods Ten-week-old SOST-knockout (SOST-KO) and wild-type (WT) mice underwent destabilization of the medial meniscus (DMM). We measured bone volume at the medial femoral condyle and osteophyte volume and determined the OA score and expression of matrix proteins. Primary murine chondrocytes were cultured with Wnt3a and sclerostin to assess the expression of matrix proteins, proteoglycan release and glycosaminoglycan accumulation. Results Sclerostin was expressed in calcified cartilage of WT mice with OA. In SOST-KO mice, cartilage was preserved despite high bone volume. However, SOST-KO mice with DMM had a high OA score, with increased expression of aggrecanases and type X collagen. Moreover, SOST-KO mice with OA showed disrupted anabolic–catabolic balance and cartilage damage. In primary chondrocytes, sclerostin addition abolished Wnt3a-increased expression of a disintegrin and metalloproteinase with thrombospondin motifs, matrix metalloproteinases and type X collagen by inhibiting the canonical Wnt pathway. Moreover, sclerostin inhibited Wnt-phosphorylated c-Jun N-terminal kinase (JNK) and rescued the expression of anabolic genes. Furthermore, sclerostin treatment inhibited both Wnt canonical and non-canonical JNK pathways in chondrocytes, thus preserving metabolism. Conclusion Sclerostin may play an important role in maintaining cartilage integrity in OA. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0540-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wafa Bouaziz
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| | - Thomas Funck-Brentano
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| | - Hilène Lin
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| | - Caroline Marty
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| | - Hang-Korng Ea
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| | - Eric Hay
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| | - Martine Cohen-Solal
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| |
Collapse
|
25
|
Ouji Y, Ishizaka S, Nakamura-Uchiyama F, Okuzaki D, Yoshikawa M. Partial maintenance and long-term expansion of murine skin epithelial stem cells by Wnt-3a in vitro. J Invest Dermatol 2014; 135:1598-1608. [PMID: 25437427 DOI: 10.1038/jid.2014.510] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 11/13/2014] [Accepted: 11/19/2014] [Indexed: 01/02/2023]
Abstract
CD49f(+)CD34(+) cells, a skin epithelial stem cell (EpSC)-rich population, were prepared from adult mouse skin and cultured in the presence of Wnt-3a without feeder cells. CD34 expression was retained in about 10% of the cells, which had proliferated about 1,000-fold by day 10, although completely lost by day 14. CD49f(+)CD34(+) cells sorted on day 10 retained canonical Wnt-responsiveness, proliferated markedly in the presence of Wnt-3a, maintained undifferentiated epithelial cell marker expression, and promoted hair follicle development in vivo. Those were subjected to a second 10-day culture with Wnt-3a and sorted, and then the same procedures were repeated a total of 15 times. CD49f(+)CD34(+) cells obtained from each of those cultures retained the same EpSC characteristics as the original cells. CD34(+) and CD34(-) cells were found to produce Wnt-3a and Wnt/β-catenin inhibitors, respectively. CD34(+) cells resided as small cellular clusters surrounded by a large amount of CD34(-) cells. Furthermore, we found that exogenous Wnt-3a delayed the conversion of CD34(+) cells to CD34(-) cells and induced CD34(-) cells to suppress the production of Wnt/β-catenin inhibitors, likely leading to generation of a microenvironment favorable for maintaining EpSCs. Our results suggest the possibility of partial long-term maintenance of EpSCs in vitro by Wnt-3a.
Collapse
Affiliation(s)
- Yukiteru Ouji
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Japan; Program in Tissue Engineering, Department of Parasitology, Nara Medical University, Kashihara, Japan.
| | - Shigeaki Ishizaka
- Program in Tissue Engineering, Department of Parasitology, Nara Medical University, Kashihara, Japan
| | | | - Daisuke Okuzaki
- DNA-chip Development Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Masahide Yoshikawa
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Japan; Program in Tissue Engineering, Department of Parasitology, Nara Medical University, Kashihara, Japan.
| |
Collapse
|
26
|
FHL2 regulates hematopoietic stem cell functions under stress conditions. Leukemia 2014; 29:615-24. [PMID: 25179730 PMCID: PMC4346553 DOI: 10.1038/leu.2014.254] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 07/16/2014] [Accepted: 08/20/2014] [Indexed: 12/22/2022]
Abstract
FHL2, a member of the four and one half LIM domain protein family, is a critical transcriptional modulator. Here, we identify FHL2 as a critical regulator of hematopoietic stem cells (HSCs) that is essential for maintaining HSC self-renewal under regenerative stress. We find that Fhl2 loss has limited effects on hematopoiesis under homeostatic conditions. In contrast, Fhl2-null chimeric mice reconstituted with Fhl2-null bone marrow cells developed abnormal hematopoiesis with significantly reduced numbers of HSCs, hematopoietic progenitor cells (HPCs), red blood cells and platelets as well as hemoglobin levels. In addition, HSCs displayed a significantly reduced self-renewal capacity and were skewed toward myeloid lineage differentiation. We find that Fhl2 loss reduces both HSC quiescence and survival in response to regenerative stress, probably as a consequence of Fhl2-loss-mediated down-regulation of cyclin dependent kinase (CDK)-inhibitors, including p21(Cip) and p27(Kip1). Interestingly, FHL2 is regulated under control of a tissue specific promoter in hematopoietic cells and it is down-regulated by DNA hypermethylation in the leukemia cell line and primary leukemia cells. Furthermore, we find that down-regulation of FHL2 frequently occurs in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) patients, raising a possibility that FHL2 down-regulation plays a role in the pathogenesis of myeloid malignancies.
Collapse
|
27
|
Lau KHW, Baylink DJ, Zhou XD, Rodriguez D, Bonewald LF, Li Z, Ruffoni D, Müller R, Kesavan C, Sheng MHC. Osteocyte-derived insulin-like growth factor I is essential for determining bone mechanosensitivity. Am J Physiol Endocrinol Metab 2013; 305:E271-81. [PMID: 23715728 DOI: 10.1152/ajpendo.00092.2013] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This study sought to determine whether deficient Igf1 expression in osteocytes would affect loading-induced osteogenic response. Tibias of osteocyte Igf1 conditional knockout (KO) mice (generated by cross-breeding Igf1 floxed mice with Dmp1-Cre transgenic mice) and wild-type (WT) littermates were subjected to four-point bending for 2 wk. Microcomputed tomography confirmed that the size of tibias of conditional mutants was smaller. Loading with an equivalent loading strain increased periosteal woven bone and endosteal lamellar bone formation in WT mice but not in conditional KO mice. Consistent with the lack of an osteogenic response, the loading failed to upregulate expression of early mechanoresponsive genes (Igf1, Cox-2, c-fos) or osteogenic genes (Cbfa-1, and osteocalcin) in conditional KO bones. The lack of osteogenic response was not due to reduced osteocyte density or insufficient loading strain. Deficient osteocyte Igf1 expression reduced the loading-induced upregulation of expression of canonical Wnt signaling genes (Wnt10b, Lrp5, Dkk1, sFrp2). The loading also reduced (by 40%) Sost expression in WT mice, but the loading not only did not reduce but upregulated (~1.5-fold) Sost expression in conditional KO mice. Conditional disruption of Igf1 in osteocytes also abolished the loading-induced increase in the bone β-catenin protein level. These findings suggest an impaired response in the loading-induced upregulation of the Wnt signaling in conditional KO mice. In summary, conditional disruption of Igf1 in osteocytes abolished the loading-induced activation of the Wnt signaling and the corresponding osteogenic response. In conclusion, osteocyte-derived IGF-I plays a key determining role in bone mechanosensitivity.
Collapse
Affiliation(s)
- K-H William Lau
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
LIM-only protein FHL2 activates NF-κB signaling in the control of liver regeneration and hepatocarcinogenesis. Mol Cell Biol 2013; 33:3299-308. [PMID: 23775124 DOI: 10.1128/mcb.00105-13] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Four-and-a-half LIM-only protein 2 (FHL2) is an important mediator in many signaling pathways. In this study, we analyzed the functions of FHL2 in nuclear factor κB (NF-κB) signaling in the liver. We show that FHL2 enhanced tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) activity in transcriptional activation of NF-κB targets by stabilizing the protein. TRAF6 is a binding partner of FHL2 and an important component of the Toll-like receptor-NF-κB pathway. Knockdown of FHL2 in 293-hTLR4/MD2-CD14 cells impaired lipopolysaccharide (LPS)-induced NF-κB activity, which regulates expression of inflammatory cytokines. Indeed, FHL2(-/-) macrophages showed significantly reduced production of TNF and interleukin 6 (IL-6) following LPS stimulation. TNF and IL-6 are the key cytokines that prime liver regeneration after hepatic injury. Following partial hepatectomy, FHL2(-/-) mice exhibited diminished induction of TNF and IL-6 and delayed hepatocyte regeneration. In the liver, NF-κB signaling orchestrates inflammatory cross talk between hepatocytes and hepatic immune cells that promote chemical hepatocarcinogenesis. We found that deficiency of FHL2 reduced susceptibility to diethylnitrosamine-induced hepatocarcinogenesis, correlating with the activator function of FHL2 in NF-κB signaling. Our findings demonstrate FHL2 as a positive regulator of NF-κB activity in liver regeneration and carcinogenesis and highlight the importance of FHL2 in both hepatocytes and hepatic immune cells.
Collapse
|