201
|
Abstract
WNTs are extracellular proteins that activate different cell surface receptors linked to canonical and noncanonical WNT signalling pathways. The Wnt genes were originally discovered as important for embryonic development of fruit flies and malignant transformation of mouse mammary cancers. More recently, WNTs have been implicated in a wide spectrum of biological phenomena and diseases. During the last decade, several lines of clinical and preclinical evidence have indicated that WNT signalling is critical for trabecular and cortical bone mass, and this pathway is currently an attractive target for drug development. Based on detailed knowledge of the different WNT signalling pathways, it appears that it might be possible to develop drugs that specifically target cortical and trabecular bone. Neutralization of a bone-specific WNT inhibitor is now being evaluated as a promising anabolic treatment for patients with osteoporosis. Here, we provide the historical background to the discoveries of WNTs, describe the different WNT signalling pathways and summarize the current understanding of how these proteins regulate bone mass by affecting bone formation and resorption.
Collapse
Affiliation(s)
- U H Lerner
- Centre for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Department of Molecular Periodontology, Umeå University, Umeå, Sweden
| | - C Ohlsson
- Centre for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
202
|
Fu C, Xu D, Wang CY, Jin Y, Liu Q, Meng Q, Liu KX, Sun HJ, Liu MZ. Alpha-Lipoic Acid Promotes Osteoblastic Formation in H2
O2
-Treated MC3T3-E1 Cells and Prevents Bone Loss in Ovariectomized Rats. J Cell Physiol 2015; 230:2184-201. [PMID: 25655087 DOI: 10.1002/jcp.24947] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 01/23/2015] [Indexed: 01/09/2023]
Affiliation(s)
- Chao Fu
- Department of Clinical Pharmacology; College of Pharmacy; Dalian Medical University; Dalian China
| | - Dong Xu
- Department of Orthopaedics; First Affiliated Hospital; Dalian Medical University; Dalian China
| | - Chang-Yuan Wang
- Department of Clinical Pharmacology; College of Pharmacy; Dalian Medical University; Dalian China
| | - Yue Jin
- Department of Clinical Pharmacology; College of Pharmacy; Dalian Medical University; Dalian China
| | - Qi Liu
- Department of Clinical Pharmacology; College of Pharmacy; Dalian Medical University; Dalian China
| | - Qiang Meng
- Department of Clinical Pharmacology; College of Pharmacy; Dalian Medical University; Dalian China
| | - Ke-Xin Liu
- Department of Clinical Pharmacology; College of Pharmacy; Dalian Medical University; Dalian China
| | - Hui-Jun Sun
- Department of Clinical Pharmacology; College of Pharmacy; Dalian Medical University; Dalian China
| | - Mo-Zhen Liu
- Department of Orthopaedics; First Affiliated Hospital; Dalian Medical University; Dalian China
| |
Collapse
|
203
|
Xing Y, Chen X, Cao Y, Huang J, Xie X, Wei Y. Expression of Wnt and Notch signaling pathways in inflammatory bowel disease treated with mesenchymal stem cell transplantation: evaluation in a rat model. Stem Cell Res Ther 2015; 6:101. [PMID: 25998108 PMCID: PMC4487973 DOI: 10.1186/s13287-015-0092-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 03/31/2015] [Accepted: 05/11/2015] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION The purpose of this study was to investigate the expression of Wnt and Notch signaling pathway-related genes in inflammatory bowel disease (IBD) treated with mesenchymal stem cell transplantation (MSCT). METHODS TNBS (2,4,6-trinitrobenzene sulfonic acid) was used to establish IBD in a rat model. Mesenchymal stem cells (MSCs) were transplanted via tail vein transfusion. Saline water was used in a control group. The expression of Wnt and Notch main signaling molecules was screened by gene chips and verified by quantitative reverse transcription-polymerase chain reaction in the IBD rat model on day 14 and day 28 after transplantation. RESULTS The IBD rat models were successfully established and MSCs were transplanted into those models. Genome-wide expression profile chips identified a total of 388 differentially expressive genes, of which 191 were upregulated and 197 were downregulated in the MSC-transplanted group in comparison with the IBD control group. Real-time quantitative polymerase chain reaction results showed that the level of Olfm4 mRNA expression in the IBD group (2.54±0.20) was significantly increased compared with the MSCT group (1.39±0.54) and the normal group (1.62±0.25) (P <0.05). The Wnt3a mRNA was more highly expressed in IBD rats (2.92±0.94) and decreased in MSCT rats (0.17±0.63, P <0.05). The expression of GSK-3β mRNA was decreased in the setting of inflammation (0.65±0.04 versus 1.00±0.01 in normal group, P <0.05) but returned to normal levels after MSCT (0.81±0.17). The expression of β-catenin was observed to increase in IBD tissues (1.76±0.44) compared with normal tissues (1.00±0.01, P <0.05), but no difference was found in the MSCT group (1.12±0.36). Wnt11 declined at 14 days and returned to normal levels at 28 days in the IBD group; in comparison, a significantly lower expression was found in MSCT rats. There were no differences in the expression of Fzd3, c-myc, TCF4, and Wnt5a in inflammation, but all of those genes declined after MSCT treatment. CONCLUSIONS The canonical Wnt and Notch signaling pathways are activated in IBD and may be suppressed by stem cell transplantation to differentiate into intestinal epithelium after MSCT. Moreover, the non-canonical Wnt signaling may be inhibited by canonical Wnt signaling in the setting of inflammation and may also be suppressed by MSCT.
Collapse
Affiliation(s)
- Yanfen Xing
- Department of Blood Transfusion, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China. .,Guangdong Key laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China.
| | - Xiaojie Chen
- Department of Blood Transfusion, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China. .,Guangdong Key laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China.
| | - Yanwen Cao
- Guangdong Key laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China. .,Department of Gastroenterology, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China.
| | - Jianyun Huang
- Department of Blood Transfusion, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China. .,Guangdong Key laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China.
| | - Xuhong Xie
- Department of Blood Transfusion, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China. .,Guangdong Key laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China.
| | - Yaming Wei
- Department of Blood Transfusion, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China. .,Guangdong Key laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, No. 1. Panfu Road, Guangzhou, 510180, Guangdong Province, China.
| |
Collapse
|
204
|
Occhetta P, Centola M, Tonnarelli B, Redaelli A, Martin I, Rasponi M. High-Throughput Microfluidic Platform for 3D Cultures of Mesenchymal Stem Cells, Towards Engineering Developmental Processes. Sci Rep 2015; 5:10288. [PMID: 25983217 PMCID: PMC4650750 DOI: 10.1038/srep10288] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 04/09/2015] [Indexed: 01/16/2023] Open
Abstract
The development of in vitro models to screen the effect of different concentrations, combinations and temporal sequences of morpho-regulatory factors on stem/progenitor cells is crucial to investigate and possibly recapitulate developmental processes with adult cells. Here, we designed and validated a microfluidic platform to (i) allow cellular condensation, (ii) culture 3D micromasses of human bone marrow-derived mesenchymal stromal cells (hBM-MSCs) under continuous flow perfusion, and (ii) deliver defined concentrations of morphogens to specific culture units. Condensation of hBM-MSCs was obtained within 3 hours, generating micromasses in uniform sizes (56.2 ± 3.9 μm). As compared to traditional macromass pellet cultures, exposure to morphogens involved in the first phases of embryonic limb development (i.e. Wnt and FGF pathways) yielded more uniform cell response throughout the 3D structures of perfused micromasses (PMMs), and a 34-fold higher percentage of proliferating cells at day 7. The use of a logarithmic serial dilution generator allowed to identify an unexpected concentration of TGFβ3 (0.1 ng/ml) permissive to hBM-MSCs proliferation and inductive to chondrogenesis. This proof-of-principle study supports the described microfluidic system as a tool to investigate processes involved in mesenchymal progenitor cells differentiation, towards a 'developmental engineering' approach for skeletal tissue regeneration.
Collapse
Affiliation(s)
- Paola Occhetta
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Matteo Centola
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Beatrice Tonnarelli
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alberto Redaelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Ivan Martin
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| |
Collapse
|
205
|
Kim JA, Choi HK, Kim TM, Leem SH, Oh IH. Regulation of mesenchymal stromal cells through fine tuning of canonical Wnt signaling. Stem Cell Res 2015; 14:356-68. [PMID: 25863444 DOI: 10.1016/j.scr.2015.02.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 02/23/2015] [Accepted: 02/25/2015] [Indexed: 01/22/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have been extensively utilized for various cell therapeutic trials, but the signals regulating their stromal function remain largely unclear. Here, we show that canonical Wnt signals distinctively regulate MSCs in a biphasic manner depending on signal intensity, i.e., MSCs exhibit proliferation and progenitor self-renewal under low Wnt/β-catenin signaling, whereas they exhibit enhanced osteogenic differentiation with priming to osteoblast-like lineages under high Wnt/β-catenin signaling. Moreover, low or high levels of β-catenin in MSCs distinctly regulated the hematopoietic support of MSCs to promote proliferation or the undifferentiated state of hematopoietic progenitors, respectively. A gene expression study demonstrated that different intracellular levels of β-catenin in MSCs induce distinct transcriptomic changes in subsets of genes belonging to different gene function categories. Different β-catenin levels also induced differences in intracellular levels of the β-catenin co-factors, Tcf-1 and Lef-1. Moreover, nano-scale mass spectrometry of proteins that co-precipitated with β-catenin revealed distinctive spectra of proteins selectively interacting with β-catenin at specific expression levels. Together, these results show that Wnt/β-catenin signals can coax distinct transcription milieu to induce different transcription profiles in MSCs depending on the signal intensity and that fine-tuning of the canonical Wnt signaling intensity can regulate the phase-specific functionality of MSCs.
Collapse
Affiliation(s)
- Jin-A Kim
- Catholic High-Performance Cell Therapy Center & Department of Medical Life Science, Republic of Korea
| | - Hyun-Kyung Choi
- Catholic High-Performance Cell Therapy Center & Department of Medical Life Science, Republic of Korea
| | - Tae-Min Kim
- Center for Cancer Evolution, Medical Research Center, The Catholic University of Korea, Republic of Korea
| | - Sun-Hee Leem
- Dept. of Biological Science, Dong-A University, Busan, Republic of Korea
| | - Il-Hoan Oh
- Catholic High-Performance Cell Therapy Center & Department of Medical Life Science, Republic of Korea.
| |
Collapse
|
206
|
Remote activation of the Wnt/β-catenin signalling pathway using functionalised magnetic particles. PLoS One 2015; 10:e0121761. [PMID: 25781466 PMCID: PMC4363733 DOI: 10.1371/journal.pone.0121761] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 02/11/2015] [Indexed: 01/12/2023] Open
Abstract
Wnt signalling pathways play crucial roles in developmental biology, stem cell fate and tissue patterning and have become an attractive therapeutic target in the fields of tissue engineering and regenerative medicine. Wnt signalling has also been shown to play a role in human Mesenchymal Stem Cell (hMSC) fate, which have shown potential as a cell therapy in bone and cartilage tissue engineering. Previous work has shown that biocompatible magnetic nanoparticles (MNP) can be used to stimulate specific mechanosensitive membrane receptors and ion channels in vitro and in vivo. Using this strategy, we determined the effects of mechano-stimulation of the Wnt Frizzled receptor on Wnt pathway activation in hMSC. Frizzled receptors were tagged using anti-Frizzled functionalised MNP (Fz-MNP). A commercially available oscillating magnetic bioreactor (MICA Biosystems) was used to mechanically stimulate Frizzled receptors remotely. Our results demonstrate that Fz-MNP can activate Wnt/β-catenin signalling at key checkpoints in the signalling pathway. Immunocytochemistry indicated nuclear localisation of the Wnt intracellular messenger β-catenin after treatment with Fz-MNP. A Wnt signalling TCF/LEF responsive luciferase reporter transfected into hMSC was used to assess terminal signal activation at the nucleus. We observed an increase in reporter activity after treatment with Fz-MNP and this effect was enhanced after mechano-stimulation using the magnetic array. Western blot analysis was used to probe the mechanism of signalling activation and indicated that Fz-MNP signal through an LRP independent mechanism. Finally, the gene expression profiles of stress response genes were found to be similar when cells were treated with recombinant Wnt-3A or Fz-MNP. This study provides proof of principle that Wnt signalling and Frizzled receptors are mechanosensitive and can be remotely activated in vitro. Using magnetic nanoparticle technology it may be possible to modulate Wnt signalling pathways and thus control stem cell fate for therapeutic purposes.
Collapse
|
207
|
Gobaa S, Hoehnel S, Lutolf MP. Substrate elasticity modulates the responsiveness of mesenchymal stem cells to commitment cues. Integr Biol (Camb) 2015; 7:1135-42. [PMID: 25749492 DOI: 10.1039/c4ib00176a] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Fate choices of stem cells are regulated in response to a complex array of biochemical and physical signals from their microenvironmental niche. Whereas the molecular composition and the role of mechanical niche cues have been extensively studied, relatively little is known about how both effectors act in concert to modulate stem cell fate. Here we utilized a recently developed artificial niche microarray platform to investigate whether the stiffness of a cell culture substrate influences how niche signaling factors exert their role on adipogenic differentiation of human mesenchymal stem cells (hMSC). We found that substrate stiffness imposes a strictly non-overlapping range of differentiation, highlighting the dominance of physical over the biochemical factors. At a given stiffness, a significant protein-dependent effect on adipogenic differentiation was observed. Furthermore, we show that synergistic interactions between proteins can also be driven by the substrate stiffness. Our results thus highlight the importance of considering the mechanical properties of a target tissue when investigating biochemical niche signals in vitro.
Collapse
Affiliation(s)
- S Gobaa
- Laboratory of Stem Cell Bioengineering (LSCB), Institute of Bioengineering (IBI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | | | | |
Collapse
|
208
|
Viale-Bouroncle S, Klingelhöffer C, Ettl T, Reichert TE, Morsczeck C. A protein kinase A (PKA)/β-catenin pathway sustains the BMP2/DLX3-induced osteogenic differentiation in dental follicle cells (DFCs). Cell Signal 2015; 27:598-605. [PMID: 25530217 DOI: 10.1016/j.cellsig.2014.12.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 11/29/2014] [Accepted: 12/14/2014] [Indexed: 02/08/2023]
Abstract
The directed expression of osteogenic transcription factors via a balanced activation of signaling pathways is an important prerequisite for the development of mineralized tissues. A positive-feedback loop of the BMP2-dependent SMAD signaling pathway and the DLX3 transcription factor (BMP2/DLX3 pathway) directs the osteogenic differentiation of periodontal precursor cells from the dental follicle (DFCs). However, little is known how this BMP2/DLX3 pathway interacts with other crucial signaling pathways such as the WNT/β-catenin signaling pathway. This study investigated the interaction between the BMP2/DLX3 pathway and the WNT pathway during the osteogenic differentiation of DFCs. BMP2 induced the WNT/β-catenin pathway in DFCs and phosphorylates β-catenin via protein kinase A (PKA). Moreover, only BMP2 facilitated the binding of LEF1/SMAD4/β-catenin complex to the DLX3 promoter, while an inducer of the canonical WNT pathway, WNT3A, act as an inhibitor. Although WNT3A inhibits the osteogenic differentiation of DFCs the expression of β-catenin was crucial for both the expression of DLX3 and for the osteogenic differentiation. In conclusion, while the activation of the canonical WNT pathway inhibits the osteogenic differentiation of DFCs, β-catenin sustains the BMP2/DLX3-mediated osteogenic differentiation via the activation of PKA.
Collapse
Affiliation(s)
- S Viale-Bouroncle
- Department of Cranio- and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - C Klingelhöffer
- Department of Cranio- and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - T Ettl
- Department of Cranio- and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - T E Reichert
- Department of Cranio- and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - C Morsczeck
- Department of Cranio- and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
209
|
Narcisi R, Cleary MA, Brama PAJ, Hoogduijn MJ, Tüysüz N, ten Berge D, van Osch GJVM. Long-term expansion, enhanced chondrogenic potential, and suppression of endochondral ossification of adult human MSCs via WNT signaling modulation. Stem Cell Reports 2015; 4:459-72. [PMID: 25733021 PMCID: PMC4375944 DOI: 10.1016/j.stemcr.2015.01.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 01/21/2015] [Accepted: 01/22/2015] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a potential source of chondrogenic cells for the treatment of cartilage disorders, but loss of chondrogenic potential during in vitro expansion and the propensity of cartilage to undergo hypertrophic maturation impede their therapeutic application. Here we report that the signaling protein WNT3A, in combination with FGF2, supports long-term expansion of human bone marrow-derived MSCs. The cells retained their chondrogenic potential and other phenotypic and functional properties of multipotent MSCs, which were gradually lost in the absence of WNT3A. Moreover, we discovered that endogenous WNT signals are the main drivers of the hypertrophic maturation that follows chondrogenic differentiation. Inhibition of WNT signals during differentiation prevented calcification and maintained cartilage properties following implantation in a mouse model. By maintaining potency during expansion and preventing hypertrophic maturation following differentiation, the modulation of WNT signaling removes two major obstacles that impede the clinical application of MSCs in cartilage repair. WNT3A and FGF2 synergistically promote MSC proliferation WNT3A and FGF2 synergistically enhance MSC chondrogenic potential during expansion WNT3A and FGF2 maintain MSC characteristics over multiple passages In vitro WNT signaling modulation leads to stable cartilage formation in vivo
Collapse
Affiliation(s)
- Roberto Narcisi
- Department of Orthopaedics, Erasmus MC, University Medical Center, 3015 CN Rotterdam, the Netherlands
| | - Mairéad A Cleary
- Department of Orthopaedics, Erasmus MC, University Medical Center, 3015 CN Rotterdam, the Netherlands; Section of Veterinary Clinical Sciences, School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Pieter A J Brama
- Section of Veterinary Clinical Sciences, School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Martin J Hoogduijn
- Department of Internal Medicine, Erasmus MC, University Medical Center, 3015 CN Rotterdam, the Netherlands
| | - Nesrin Tüysüz
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Erasmus MC, University Medical Center, 3015 CN Rotterdam, the Netherlands
| | - Derk ten Berge
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Erasmus MC, University Medical Center, 3015 CN Rotterdam, the Netherlands.
| | - Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Center, 3015 CN Rotterdam, the Netherlands; Department of Otorhinolaryngology, Erasmus MC, University Medical Center, 3015 CN Rotterdam, the Netherlands.
| |
Collapse
|
210
|
He Y, Lin F, Chen Y, Tan Z, Bai D, Zhao Q. Overexpression of the Circadian Clock Gene Rev-erbα Affects Murine Bone Mesenchymal Stem Cell Proliferation and Osteogenesis. Stem Cells Dev 2015; 24:1194-204. [PMID: 25539035 DOI: 10.1089/scd.2014.0437] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Bone mesenchymal stem cell (BMSC) age-related changes include decreased osteogenesis and increased adipogenesis. Rev-erbα and the Wnt/β-catenin signaling pathway were known to play important roles in BMSC aging. In this study, we have aimed to elucidate whether Rev-erbα and Wnt/β-catenin signaling interact during BMSC proliferation and osteogenesis. Our results showed that Rev-erbα expression gradually dropped during BMSC osteogenesis, and overexpression of Rev-erbα in BMSCs inhibited cell proliferation and osteogenesis. The inhibition of cell proliferation induced by Rev-erbα overexpression was partially reversed when Wnt/β-catenin signaling was activated. These results suggested that Rev-erbα could promote BMSC aging and may be the negative regulator during the late stage of osteogenesis. The clock gene Rev-erbα and Wnt/β-catenin signaling interact in the regulation of cell proliferation.
Collapse
Affiliation(s)
- Yao He
- 1 Orthodontic Centre, West China College of Stomatology, Sichuan University , Chengdu, China
| | | | | | | | | | | |
Collapse
|
211
|
Jeoung JY, Nam HY, Kwak J, Jin HJ, Lee HJ, Lee BW, Baek JH, Eom JS, Chang EJ, Shin DM, Choi SJ, Kim SW. A decline in Wnt3a signaling is necessary for mesenchymal stem cells to proceed to replicative senescence. Stem Cells Dev 2015; 24:973-82. [PMID: 25437011 DOI: 10.1089/scd.2014.0273] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Umbilical cord blood-derived mesenchymal stem cells are a promising source of cells for regeneration therapy due to their multipotency, high proliferative capacity, relatively noninvasive collection, and ready availability. However, extended cell culture inevitably triggers cellular senescence-the irreversible arrest of cell division-thereby limiting the proliferative lifespan of adult stem cells. Wnt/β-catenin signaling plays a functional role as a key regulator of self-renewal and differentiation in mesenchymal stem cells (MSCs), and thus Wnt/β-catenin signaling and cellular senescence might be closely connected. Here, we show that the expression levels of canonical Wnt families decrease as MSCs age during subculture. Activation of the Wnt pathway by treatment with Wnt3a-conditioned medium or glycogen synthase kinase 3β inhibitors, such as SB-216763 and 6-bromoindirubin-3'-oxime, delays the progression of cellular senescence as shown by the decrease in the senescence effectors p53 and pRb, lowered senescence-associated β-galactosidase activity, and increased telomerase activity. In contrast, suppression of the Wnt pathway by treatment with dickkopf-1 (an antagonist of the Wnt coreceptor) and β-catenin siRNA transfection promotes senescence in MSCs. Interestingly, the magnitude of the response to enhanced Wnt3a/β-catenin signaling appears to depend on the senescent state during extended culture, particularly after multiple passages. These results suggest that Wnt3a signaling might be a predominant factor that could be used to overcome senescence in long-term cultured MSCs by directly intervening in the proliferative capacity and MSC senescence. The functional role of Wnt3a/β-catenin signaling in hedging cellular senescence may allow the development of new approaches for stem cell-based therapies.
Collapse
Affiliation(s)
- Ji Yung Jeoung
- 1 Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Schuh CMAP, Heher P, Weihs AM, Banerjee A, Fuchs C, Gabriel C, Wolbank S, Mittermayr R, Redl H, Rünzler D, Teuschl AH. In vitro extracorporeal shock wave treatment enhances stemness and preserves multipotency of rat and human adipose-derived stem cells. Cytotherapy 2014; 16:1666-78. [DOI: 10.1016/j.jcyt.2014.07.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 07/21/2014] [Accepted: 07/22/2014] [Indexed: 12/11/2022]
|
213
|
Toscani D, Bolzoni M, Accardi F, Aversa F, Giuliani N. The osteoblastic niche in the context of multiple myeloma. Ann N Y Acad Sci 2014; 1335:45-62. [DOI: 10.1111/nyas.12578] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Denise Toscani
- Myeloma Unit, Department of Clinical and Experimental Medicine; University of Parma; Parma Italy
| | - Marina Bolzoni
- Myeloma Unit, Department of Clinical and Experimental Medicine; University of Parma; Parma Italy
| | - Fabrizio Accardi
- Myeloma Unit, Department of Clinical and Experimental Medicine; University of Parma; Parma Italy
| | - Franco Aversa
- Myeloma Unit, Department of Clinical and Experimental Medicine; University of Parma; Parma Italy
| | - Nicola Giuliani
- Myeloma Unit, Department of Clinical and Experimental Medicine; University of Parma; Parma Italy
| |
Collapse
|
214
|
Bulycheva E, Rauner M, Medyouf H, Theurl I, Bornhäuser M, Hofbauer LC, Platzbecker U. Myelodysplasia is in the niche: novel concepts and emerging therapies. Leukemia 2014; 29:259-68. [PMID: 25394715 PMCID: PMC4320287 DOI: 10.1038/leu.2014.325] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 09/25/2014] [Indexed: 12/28/2022]
Abstract
Myelodysplastic syndromes (MDSs) represent clonal disorders mainly of the elderly that are characterized by ineffective hematopoiesis and an increased risk of transformation into acute myeloid leukemia. The pathogenesis of MDS is thought to evolve from accumulation and selection of specific genetic or epigenetic events. Emerging evidence indicates that MDS is not solely a hematopoietic disease but rather affects the entire bone marrow microenvironment, including bone metabolism. Many of these cells, in particular mesenchymal stem and progenitor cells (MSPCs) and osteoblasts, express a number of adhesion molecules and secreted factors that regulate blood regeneration throughout life by contributing to hematopoietic stem and progenitor cell (HSPC) maintenance, self-renewal and differentiation. Several endocrine factors, such as erythropoietin, parathyroid hormone and estrogens, as well as deranged iron metabolism modulate these processes. Thus, interactions between MSPC and HSPC contribute to the pathogenesis of MDS and associated pathologies. A detailed understanding of these mechanisms may help to define novel targets for diagnosis and possibly therapy. In this review, we will discuss the scientific rationale of ‘osteohematology' as an emerging research field in MDS and outline clinical implications.
Collapse
Affiliation(s)
- E Bulycheva
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany
| | - M Rauner
- Medizinische Klinik und Poliklinik III, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany
| | - H Medyouf
- Georg-Speyer-Haus, Institut for Tumor Biology and Experimental Therapy, 60596, Frankfurt am Main, Germany
| | - I Theurl
- Department of Internal Medicine VI, Medical University of Innsbruck, Innsbruck, Austria
| | - M Bornhäuser
- 1] Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany [2] Center for Regenerative Therapies Dresden, Technical University, Dresden, Germany
| | - L C Hofbauer
- 1] Medizinische Klinik und Poliklinik III, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany [2] Center for Regenerative Therapies Dresden, Technical University, Dresden, Germany
| | - U Platzbecker
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany
| |
Collapse
|
215
|
Schönitzer V, Wirtz R, Ulrich V, Berger T, Karl A, Mutschler W, Schieker M, Böcker W. Sox2 Is a Potent Inhibitor of Osteogenic and Adipogenic Differentiation in Human Mesenchymal Stem Cells. Cell Reprogram 2014; 16:355-65. [DOI: 10.1089/cell.2014.0006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Veronika Schönitzer
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University, 80336, Munich, Germany
- These authors contributed equally to this work
| | - Roman Wirtz
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University, 80336, Munich, Germany
- These authors contributed equally to this work
| | - Veronika Ulrich
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University, 80336, Munich, Germany
| | - Tamara Berger
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University, 80336, Munich, Germany
| | - Angelika Karl
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University, 80336, Munich, Germany
| | - Wolf Mutschler
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University, 80336, Munich, Germany
| | - Matthias Schieker
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University, 80336, Munich, Germany
- Current address: Amgen GmbH, 80922, Munich, Germany
| | - Wolfgang Böcker
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University, 80336, Munich, Germany
- Department of Orthopaedic Trauma Surgery, University Hospital, 35385, Giessen, Germany
| |
Collapse
|
216
|
Kasten A, Grüttner C, Kühn JP, Bader R, Pasold J, Frerich B. Comparative in vitro study on magnetic iron oxide nanoparticles for MRI tracking of adipose tissue-derived progenitor cells. PLoS One 2014; 9:e108055. [PMID: 25244560 PMCID: PMC4171509 DOI: 10.1371/journal.pone.0108055] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/16/2014] [Indexed: 02/07/2023] Open
Abstract
Magnetic resonance imaging (MRI) using measurement of the transverse relaxation time (R2*) is to be considered as a promising approach for cell tracking experiments to evaluate the fate of transplanted progenitor cells and develop successful cell therapies for tissue engineering. While the relationship between core composition of nanoparticles and their MRI properties is well studied, little is known about possible effects on progenitor cells. This in vitro study aims at comparing two magnetic iron oxide nanoparticle types, single vs. multi-core nanoparticles, regarding their physico-chemical characteristics, effects on cellular behavior of adipose tissue-derived stem cells (ASC) like differentiation and proliferation as well as their detection and quantification by means of MRI. Quantification of both nanoparticle types revealed a linear correlation between labeling concentration and R2* values. However, according to core composition, different levels of labeling concentrations were needed to achieve comparable R2* values. Cell viability was not altered for all labeling concentrations, whereas the proliferation rate increased with increasing labeling concentrations. Likewise, deposition of lipid droplets as well as matrix calcification revealed to be highly dose-dependent particularly regarding multi-core nanoparticle-labeled cells. Synthesis of cartilage matrix proteins and mRNA expression of collagen type II was also highly dependent on nanoparticle labeling. In general, the differentiation potential was decreased with increasing labeling concentrations. This in vitro study provides the proof of principle for further in vivo tracking experiments of progenitor cells using nanoparticles with different core compositions but also provides striking evidence that combined testing of biological and MRI properties is advisable as improved MRI properties of multi-core nanoparticles may result in altered cell functions.
Collapse
Affiliation(s)
- Annika Kasten
- Department of Oral and Maxillofacial Surgery, Facial Plastic Surgery, Rostock University Medical Center, Rostock, Germany
| | | | - Jens-Peter Kühn
- Department of Radiology and Neuroradiology, Greifswald University Medical Center, Greifswald, Germany
| | - Rainer Bader
- Department of Orthopaedics, Biomechanics and Implant Technology Research Laboratory, Rostock University Medical Center, Rostock, Germany
| | - Juliane Pasold
- Department of Orthopaedics, Biomechanics and Implant Technology Research Laboratory, Rostock University Medical Center, Rostock, Germany
| | - Bernhard Frerich
- Department of Oral and Maxillofacial Surgery, Facial Plastic Surgery, Rostock University Medical Center, Rostock, Germany
- * E-mail:
| |
Collapse
|
217
|
Liedert A, Röntgen V, Schinke T, Benisch P, Ebert R, Jakob F, Klein-Hitpass L, Lennerz JK, Amling M, Ignatius A. Osteoblast-specific Krm2 overexpression and Lrp5 deficiency have different effects on fracture healing in mice. PLoS One 2014; 9:e103250. [PMID: 25061805 PMCID: PMC4111586 DOI: 10.1371/journal.pone.0103250] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 06/28/2014] [Indexed: 01/09/2023] Open
Abstract
The canonical Wnt/β-catenin pathway plays a key role in the regulation of bone remodeling in mice and humans. Two transmembrane proteins that are involved in decreasing the activity of this pathway by binding to extracellular antagonists, such as Dickkopf 1 (Dkk1), are the low-density lipoprotein receptor related protein 5 (Lrp5) and Kremen 2 (Krm2). Lrp 5 deficiency (Lrp5−/−) as well as osteoblast-specific overexpression of Krm2 in mice (Col1a1-Krm2) result in severe osteoporosis occurring at young age. In this study, we analyzed the influence of Lrp5 deficiency and osteoblast-specific overexpression of Krm2 on fracture healing in mice using flexible and semi-rigid fracture fixation. We demonstrated that fracture healing was highly impaired in both mouse genotypes, but that impairment was more severe in Col1a1-Krm2 than in Lrp5−/− mice and particularly evident in mice in which the more flexible fixation was used. Bone formation was more reduced in Col1a1-Krm2 than in Lrp5−/− mice, whereas osteoclast number was similarly increased in both genotypes in comparison with wild-type mice. Using microarray analysis we identified reduced expression of genes mainly involved in osteogenesis that seemed to be responsible for the observed stronger impairment of healing in Col1a1-Krm2 mice. In line with these findings, we detected decreased expression of sphingomyelin phosphodiesterase 3 (Smpd3) and less active β-catenin in the calli of Col1a1-Krm2 mice. Since Krm2 seems to play a significant role in regulating bone formation during fracture healing, antagonizing KRM2 might be a therapeutic option to improve fracture healing under compromised conditions, such as osteoporosis.
Collapse
Affiliation(s)
- Astrid Liedert
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
- * E-mail:
| | - Viktoria Röntgen
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peggy Benisch
- Orthopaedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Regina Ebert
- Orthopaedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Franz Jakob
- Orthopaedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | | | | | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| |
Collapse
|
218
|
Klingberg E, Nurkkala M, Carlsten H, Forsblad-d’Elia H. Biomarkers of Bone Metabolism in Ankylosing Spondylitis in Relation to Osteoproliferation and Osteoporosis. J Rheumatol 2014; 41:1349-56. [DOI: 10.3899/jrheum.131199] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Objective.To identify biomarkers for bone metabolism in patients with ankylosing spondylitis (AS) and to determine the relationship between these biomarkers and disease activity, back mobility, osteoproliferation, and bone mineral density (BMD).Methods.Serum levels of Wingless protein (Wnt-3a), Dickkopf-1 (DKK-1), sclerostin, soluble receptor activator of nuclear factor-κB ligand (sRANKL), and osteoprotegerin were assessed using ELISA. Ankylosing Spondylitis Disease Activity Score-C reactive protein, Bath Ankylosing Spondylitis Disease Activity Index, Bath Ankylosing Spondylitis patient global score, and C-reactive protein (CRP) were used as disease activity measures, and Bath Ankylosing Spondylitis Metrology Index (BASMI) as a measure of spinal mobility. Lateral spine radiographs were scored for chronic AS-related changes (mSASSS). BMD was measured with dual-energy x-ray absorptiometry.Results.Two hundred four patients with AS (NY criteria; 57% men), with a mean age of 50 ± 13 years and disease duration 15 ± 11 years, and 80 age and sex-matched controls were included. The patients with AS had significantly higher serum levels of Wnt-3a (p < 0.001) and lower levels of sclerostin (p = 0.014) and sRANKL (p = 0.047) compared with the controls. High CRP was associated with low sclerostin (rS = −0.21, p = 0.003) and DKK-1 (rS = −0.14, p = 0.045). In multiple linear regression analyses, increasing BASMI and mSASSS were independently associated with older age, male sex, high CRP, and elevated serum levels of Wnt-3a. In addition, mSASSS remained associated with a high number of smoking pack-years after adjusting for age. Low BMD of femoral neck was associated with high mSASSS after adjusting for age.Conclusion.Serum levels of Wnt-3a are elevated in AS and associated with increased BASMI and mSASSS, independent of age, indicating that Wnt-3a could be a biomarker for the osteoproliferative process.
Collapse
|
219
|
Human periodontal ligament derived progenitor cells: effect of STRO-1 cell sorting and Wnt3a treatment on cell behavior. BIOMED RESEARCH INTERNATIONAL 2014; 2014:145423. [PMID: 24864228 PMCID: PMC4020471 DOI: 10.1155/2014/145423] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/08/2014] [Accepted: 04/12/2014] [Indexed: 01/09/2023]
Abstract
OBJECTIVES STRO-1 positive periodontal ligament cells (PDLCs) and unsorted PDLCs have demonstrated potential for periodontal regeneration, but the comparison between unsorted cells and the expanded STRO-1 sorted cells has never been reported. Additionally, Wnt3a is involved in cell proliferation thus may benefit in vitro PDLC expansion. The aim was to evaluate the effect of STRO-1 cell sorting and Wnt3a treatment on cell behavior of human PDLCs (hPDLCs). MATERIALS AND METHODS STRO-1 positive hPDLCs were sorted and the sorted cells were expanded and compared with their unsorted parental cells. Thereafter, hPDLCs were treated with or without Wnt3a and the cell proliferation, self-renewal, and osteogenic differentiation were evaluated. RESULTS No differences were measured between the expanded STRO-1-sorted cells and unsorted parental cells in terms of proliferation, CFU, and mineralization capacity. Wnt3a enhanced the proliferation and self-renewal ability of hPDLCs significantly as displayed by higher DNA content values, a shorter cell population doubling time, and higher expression of the self-renewal gene Oct4. Moreover, Wnt3a promoted the expansion of hPDLCs for 5 passages without affecting cell proliferation, CFU, and osteogenic capacity. CONCLUSIONS Expanded STRO-1-sorted hPDLCs showed no superiority compared to their unsorted parental cells. On the other hand, Wnt3a promotes the efficient hPDLC expansion and retains the self-renewal and osteogenic differentiation capacity.
Collapse
|
220
|
Chen M, Qiao H, Su Z, Li H, Ping Q, Zong L. Emerging therapeutic targets for osteoporosis treatment. Expert Opin Ther Targets 2014; 18:817-31. [PMID: 24766518 DOI: 10.1517/14728222.2014.912632] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION To date, osteoporosis still remains a major public health burden especially for the aging populations. Over the last few decades treatments for osteoporosis have largely focused on anti-resorptive agents represented by bisphosphonates and estrogen therapy that dominated the market. Unsatisfactory efficacy, non-specificity and long-term safety of current therapies necessitate the need for new targets effectively preventing and treating of osteoporosis. AREAS COVERED This review expatiates on the mechanism of osteoporosis occurrence and bone remodeling cycle in detail. New targets of antiresorptive and anabolic agents based on the functions of osteoblasts and osteoclasts as well as associated signaling pathways are outlined. EXPERT OPINION Advanced understanding in the fields of bone remodeling, functions of osteoblasts, osteoclasts and osteocytes associated with osteoporosis occurrence offers the emerging bone-resorptive or bone-formative targets. Currently, molecules involving RANK-RANKL-OPG system and Wnt/β-catenin signaling pathway act as the most promising targets.
Collapse
Affiliation(s)
- Minglei Chen
- China Pharmaceutical University, Key Lab of State Natural Medicine, Department of Pharmaceutics , Nanjing 210009 , PR China +86 25 83271092; +86 25 83271317 ; +86 25 83271092; +86 25 83271317 ; ;
| | | | | | | | | | | |
Collapse
|
221
|
Liu A, Chen S, Cai S, Dong L, Liu L, Yang Y, Guo F, Lu X, He H, Chen Q, Hu S, Qiu H. Wnt5a through noncanonical Wnt/JNK or Wnt/PKC signaling contributes to the differentiation of mesenchymal stem cells into type II alveolar epithelial cells in vitro. PLoS One 2014; 9:e90229. [PMID: 24658098 PMCID: PMC3962348 DOI: 10.1371/journal.pone.0090229] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/27/2014] [Indexed: 01/11/2023] Open
Abstract
The differentiation of mesenchymal stem cells (MSCs) into type II alveolar epithelial (AT II) cells is critical for reepithelization and recovery in acute respiratory distress syndrome (ARDS), and Wnt signaling was considered to be the underlying mechanisms. In our previous study, we found that canonical Wnt pathway promoted the differentiation of MSCs into AT II cells, however the role of the noncanonical Wnt pathway in this process is unclear. It was disclosed in this study that noncanonical Wnt signaling in mouse bone marrow-derived MSCs (mMSCs) was activated during the differentiation of mMSCs into AT II cells in a modified co-culture system with murine lung epithelial-12 cells and small airway growth media. The levels of surfactant protein (SP) C, SPB and SPD, the specific markers of AT II cells, increased in mMSCs when Wnt5a was added to activate noncanonical Wnt signaling, while pretreatment with JNK or PKC inhibitors reversed the promotion of Wnt5a. The differentiation rate of mMSCs also depends on their abilities to accumulate and survive in inflammatory tissue. We found that the Wnt5a supplement promoted the vertical and horizontal migration of mMSCs, ameliorated the cell death and the reduction of Bcl-2/Bax induced by H2O2. The effect of Wnt5a on the migration of mMSCs and their survival after H2O2 exposure were partially inhibited with PKC or JNK blockers. In conclusion, Wnt5a through Wnt/JNK signaling alone or both Wnt/JNK and Wnt/PKC signaling promoted the differentiation of mMSCs into AT II cells and the migration of mMSCs; through Wnt/PKC signaling, Wnt5a increased the survival of mMSCs after H2O2 exposure in vitro.
Collapse
Affiliation(s)
- Airan Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Song Chen
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, P. R. China
| | - Shixia Cai
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Liang Dong
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Le Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Yi Yang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Fengmei Guo
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Xiaomin Lu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Hongli He
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Qihong Chen
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Shuling Hu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Haibo Qiu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P. R. China
- * E-mail:
| |
Collapse
|
222
|
Wnt-dependent osteogenic commitment of bone marrow stromal cells using a novel GSK3β inhibitor. Stem Cell Res 2014; 12:415-27. [DOI: 10.1016/j.scr.2013.10.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 10/02/2013] [Accepted: 10/04/2013] [Indexed: 11/23/2022] Open
|
223
|
Shah N, Morsi Y, Manasseh R. From mechanical stimulation to biological pathways in the regulation of stem cell fate. Cell Biochem Funct 2014; 32:309-25. [PMID: 24574137 DOI: 10.1002/cbf.3027] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 11/28/2013] [Accepted: 01/07/2014] [Indexed: 12/15/2022]
Abstract
Mechanical stimuli are important in directing the fate of stem cells; the effects of mechanical stimuli reported in recent research are reviewed here. Stem cells normally undergo two fundamental processes: proliferation, in which their numbers multiply, and differentiation, in which they transform into the specialized cells needed by the adult organism. Mechanical stimuli are well known to affect both processes of proliferation and differentiation, although the complete pathways relating specific mechanical stimuli to stem cell fate remain to be elucidated. We identified two broad classes of research findings and organized them according to the type of mechanical stress (compressive, tensile or shear) of the stimulus. Firstly, mechanical stress of any type activates stretch-activated channels (SACs) on the cell membrane. Activation of SACs leads to cytoskeletal remodelling and to the expression of genes that regulate the basic growth, survival or apoptosis of the cells and thus regulates proliferation. Secondly, mechanical stress on cells that are physically attached to an extracellular matrix (ECM) initiates remodelling of cell membrane structures called integrins. This second process is highly dependent on the type of mechanical stress applied and result into various biological responses. A further process, the Wnt pathway, is also implicated: crosstalk between the integrin and Wnt pathways regulates the switch from proliferation to differentiation and finally regulates the type of differentiation. Therefore, the stem cell differentiation process involves different signalling molecules and their pathways and most likely depends upon the applied mechanical stimulation.
Collapse
Affiliation(s)
- Nirali Shah
- Faculty of Engineering and Industrial Sciences, Swinburne University of Technology, VIC, Melbourne, Australia
| | | | | |
Collapse
|
224
|
Cai SX, Liu AR, He HL, Chen QH, Yang Y, Guo FM, Huang YZ, Liu L, Qiu HB. Stable Genetic Alterations of β-Catenin and ROR2 Regulate the Wnt Pathway, Affect the Fate of MSCs. J Cell Physiol 2014; 229:791-800. [PMID: 24590964 DOI: 10.1002/jcp.24500] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 10/17/2013] [Indexed: 11/05/2022]
Affiliation(s)
- Shi-Xia Cai
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Ai-Ran Liu
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Hong-Li He
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Qi-Hong Chen
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Yi Yang
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Feng-Mei Guo
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Ying-Zi Huang
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Ling Liu
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Hai-Bo Qiu
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| |
Collapse
|
225
|
Menicanin D, Mrozik KM, Wada N, Marino V, Shi S, Bartold PM, Gronthos S. Periodontal-ligament-derived stem cells exhibit the capacity for long-term survival, self-renewal, and regeneration of multiple tissue types in vivo. Stem Cells Dev 2014; 23:1001-11. [PMID: 24351050 DOI: 10.1089/scd.2013.0490] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Primary periodontal ligament stem cells (PDLSCs) are known to possess multidifferentiation potential and exhibit an immunophenotype similar to that described for bone-marrow-derived mesenchymal stem cells. In the present study, bromo-deoxyuridine (BrdU)-labeled ovine PDLSCs implanted into immunodeficient mice survived after 8 weeks post-transplantation and exhibited the capacity to form bone/cementum-like mineralized tissue, ligament structures similar to Sharpey's fibers with an associated vasculature. To evaluate self-renewal potential, PDLSCs were recovered from harvested primary transplants 8 weeks post-transplantation that exhibit an immunophenotype and multipotential capacity comparable to primary PDLSCs. The re-derived PDLSCs isolated from primary transplants were implanted into secondary ectopic xenogeneic transplants. Histomorphological analysis demonstrated that four out of six donor re-derived PDLSC populations displayed a capacity to survive and form fibrous ligament structures and mineralized tissues associated with vasculature in vivo, although at diminished levels in comparison to primary PDLSCs. Further, the capacity for long-term survival and the potential role of PDLSCs in dental tissue regeneration were determined using an ovine preclinical periodontal defect model. Autologous ex vivo-expanded PDLSCs that were prelabeled with BrdU were seeded onto Gelfoam(®) scaffolds and then transplanted into fenestration defects surgically created in the periodontium of the second premolars. Histological assessment at 8 weeks post-implantation revealed surviving BrdU-positive PDLSCs associated with regenerated periodontium-related tissues, including cementum and bone-like structures. This is the first report to demonstrate the self-renewal capacity of PDLSCs using serial xenogeneic transplants and provides evidence of the long-term survival and tissue contribution of autologous PDLSCs in a preclinical periodontal defect model.
Collapse
Affiliation(s)
- Danijela Menicanin
- 1 Colgate Australian Clinical Dental Research Centre, School of Dentistry, University of Adelaide , Adelaide, Australia
| | | | | | | | | | | | | |
Collapse
|
226
|
Liu S, Zhang E, Yang M, Lu L. Overexpression of Wnt11 promotes chondrogenic differentiation of bone marrow-derived mesenchymal stem cells in synergism with TGF-β. Mol Cell Biochem 2014; 390:123-31. [PMID: 24474615 DOI: 10.1007/s11010-014-1963-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 01/14/2014] [Indexed: 12/19/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs), the most widely used cell source for cartilage tissue engineering, are multipotent cells which have been shown to differentiate into various mesenchyme-lineage cell types including chondrocytes. However, the molecular mechanisms controlling the chondrogenic differentiation of MSCs remain to be fully elucidated. It has been demonstrated that Wnt signaling involves regulating chondrogenesis and MSC differentiation. The aim of the present study was to investigate the role of Wnt11, a member of noncanonical Wnts, in MSCs during chondrogenic differentiation. We observed that overexpression of Wnt11 inhibited proliferation of MSCs and caused a G0/G1 cell cycle arrest. The expression level of chondrogenic markers, aggrecan and Collagen II, was significantly increased in MSCs transduced with Wnt11 as compared with non-transduced cells or MSCs transduced with the empty lentiviral vector. Furthermore, ectopic expression of Wnt11 stimulated gene expression of chondrogenic regulators, SRY-related gene 9, Runt-related transcription factor 2, and Indian hedgehog. Finally, Wnt11 overexpression promoted chondrogenic differentiation of MSCs in synergism with TGF-β. Collectively, these results indicate that Wnt11 plays a crucial role in regulating MSC chondrogenic differentiation.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, 117 North Nanjing Street, 110001, Shenyang, People's Republic of China
| | | | | | | |
Collapse
|
227
|
Biver E, Thouverey C, Magne D, Caverzasio J. Crosstalk between tyrosine kinase receptors, GSK3 and BMP2 signaling during osteoblastic differentiation of human mesenchymal stem cells. Mol Cell Endocrinol 2014; 382:120-130. [PMID: 24060635 DOI: 10.1016/j.mce.2013.09.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 08/16/2013] [Accepted: 09/16/2013] [Indexed: 10/26/2022]
Abstract
Bone morphogenic proteins (BMPs) promote mesenchymal stem cell (MSC) osteogenic differentiation, whereas platelet derived growth factor (PDGF) and fibroblast growth factor (FGF) activate their proliferation through receptors tyrosine kinase (RTK). The effects of PDGF or FGF receptor signaling pathway on BMP2-induced osteoblastic differentiation was investigated in human MSC (HMSC). Inhibition of PDGF or/and FGF receptors enhanced BMP2-induced alkaline phosphatase (ALP) activity, expression of Osterix, ALP and Bone sialoprotein, and matrix calcification. These effects were associated with increased Smad-1 activity, indicating that mitogenic factors interfere with Smad signaling in HMSC differentiation. RTK activate MAPK and inhibit GSK3 through the PI3K/Akt pathway. Biochemical analysis indicated that MAPK JNK and GSK3 especially are potential signaling molecules regulating BMP-induced osteoblastic HMSC differentiation. These observations highlight that the osteogenic effects of BMP2 are modulated by mitogenic factors acting through RTK.
Collapse
Affiliation(s)
- Emmanuel Biver
- Service of Bone Diseases, Department of Internal Medicine Specialties, University Hospital of Geneva, CH-1211 Geneva 14, Switzerland; Pathophysiology of Inflammatory Bone Diseases, PMOI EA4490, Boulogne/Mer, France
| | - Cyril Thouverey
- Service of Bone Diseases, Department of Internal Medicine Specialties, University Hospital of Geneva, CH-1211 Geneva 14, Switzerland
| | - David Magne
- Institut of Molecular and Supramolecular Biochemistry, UMR, CNRS 5246, University of Lyon 1, 69622 Villeurbanne Cedex, France
| | - Joseph Caverzasio
- Service of Bone Diseases, Department of Internal Medicine Specialties, University Hospital of Geneva, CH-1211 Geneva 14, Switzerland.
| |
Collapse
|
228
|
Boyette LB, Tuan RS. Adult Stem Cells and Diseases of Aging. J Clin Med 2014; 3:88-134. [PMID: 24757526 PMCID: PMC3992297 DOI: 10.3390/jcm3010088] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 12/15/2013] [Accepted: 12/17/2013] [Indexed: 02/06/2023] Open
Abstract
Preservation of adult stem cells pools is critical for maintaining tissue homeostasis into old age. Exhaustion of adult stem cell pools as a result of deranged metabolic signaling, premature senescence as a response to oncogenic insults to the somatic genome, and other causes contribute to tissue degeneration with age. Both progeria, an extreme example of early-onset aging, and heritable longevity have provided avenues to study regulation of the aging program and its impact on adult stem cell compartments. In this review, we discuss recent findings concerning the effects of aging on stem cells, contributions of stem cells to age-related pathologies, examples of signaling pathways at work in these processes, and lessons about cellular aging gleaned from the development and refinement of cellular reprogramming technologies. We highlight emerging therapeutic approaches to manipulation of key signaling pathways corrupting or exhausting adult stem cells, as well as other approaches targeted at maintaining robust stem cell pools to extend not only lifespan but healthspan.
Collapse
Affiliation(s)
- Lisa B Boyette
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; ; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; ; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA ; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
229
|
Boyette LB, Creasey OA, Guzik L, Lozito T, Tuan RS. Human bone marrow-derived mesenchymal stem cells display enhanced clonogenicity but impaired differentiation with hypoxic preconditioning. Stem Cells Transl Med 2014; 3:241-54. [PMID: 24436440 DOI: 10.5966/sctm.2013-0079] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stem cells are promising candidate cells for regenerative applications because they possess high proliferative capacity and the potential to differentiate into other cell types. Mesenchymal stem cells (MSCs) are easily sourced but do not retain their proliferative and multilineage differentiative capabilities after prolonged ex vivo propagation. We investigated the use of hypoxia as a preconditioning agent and in differentiating cultures to enhance MSC function. Culture in 5% ambient O(2) consistently enhanced clonogenic potential of primary MSCs from all donors tested. We determined that enhanced clonogenicity was attributable to increased proliferation, increased vascular endothelial growth factor secretion, and increased matrix turnover. Hypoxia did not impact the incidence of cell death. Application of hypoxia to osteogenic cultures resulted in enhanced total mineral deposition, although this effect was detected only in MSCs preconditioned in normoxic conditions. Osteogenesis-associated genes were upregulated in hypoxia, and alkaline phosphatase activity was enhanced. Adipogenic differentiation was inhibited by exposure to hypoxia during differentiation. Chondrogenesis in three-dimensional pellet cultures was inhibited by preconditioning with hypoxia. However, in cultures expanded under normoxia, hypoxia applied during subsequent pellet culture enhanced chondrogenesis. Whereas hypoxic preconditioning appears to be an excellent way to expand a highly clonogenic progenitor pool, our findings suggest that it may blunt the differentiation potential of MSCs, compromising their utility for regenerative tissue engineering. Exposure to hypoxia during differentiation (post-normoxic expansion), however, appears to result in a greater quantity of functional osteoblasts and chondrocytes and ultimately a larger quantity of high-quality differentiated tissue.
Collapse
Affiliation(s)
- Lisa B Boyette
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA; Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, McGowan Institute for Regenerative Medicine, and Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
230
|
Gnanasegaran N, Govindasamy V, Musa S, Kasim NHA. Different isolation methods alter the gene expression profiling of adipose derived stem cells. Int J Med Sci 2014; 11:391-403. [PMID: 24669199 PMCID: PMC3964446 DOI: 10.7150/ijms.7697] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 01/20/2014] [Indexed: 12/22/2022] Open
Abstract
Human adipose stem cells (ASCs) has been in the limelight since its discovery as a suitable source of mesenchymal stem cells (MSCs) in regenerative medicine. Currently, two major techniques are used to isolate ASCs, namely liposuction and tissue biopsy. These two methods are relatively risk-free but the question as to which method could give a more efficient output remains unclear. Thus, this study was carried out to compare and contrast the output generated in regards to growth kinetics, differentiation capabilities in vitro, and gene expression profiling. It was found that ASCs from both isolation methods were comparable in terms of growth kinetics and tri-lineage differentiation. Furthermore, ASCs from both populations were reported as CD44(+), CD73(+), CD90(+), CD166(+), CD34(-), CD45(-) and HLA-DR(-). However, in regards to gene expression, a group of overlapping genes as well as distinct genes were observed. Distinct gene expressions indicated that ASCs (liposuction) has endoderm lineage propensity whereas ASCs (biopsy) has a tendency towards mesoderm/ectoderm lineage. This information suggests involvement in different functional activity in accordance to isolation method. In conclusion, future studies to better understand these gene functions should be carried out in order to contribute in the applicability of each respective cells in regenerative therapy.
Collapse
Affiliation(s)
- Nareshwaran Gnanasegaran
- 1. Regenerative Dentistry Research Group (ReDReG), Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Vijayendran Govindasamy
- 2. Hygieia Innovation Sdn. Bhd, Lot 1G-2G, Lanai Complex No.2, Persiaran Seri Perdana, Persint 10, Federal Territory of Putrajaya, Malaysia
| | - Sabri Musa
- 1. Regenerative Dentistry Research Group (ReDReG), Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia ; 3. Department of Children's Dentistry and Orthodontics, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Hayaty Abu Kasim
- 1. Regenerative Dentistry Research Group (ReDReG), Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia ; 4. Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
231
|
Frith JE, Titmarsh DM, Padmanabhan H, Cooper-White JJ. Microbioreactor array screening of Wnt modulators and microenvironmental factors in osteogenic differentiation of mesenchymal progenitor cells. PLoS One 2013; 8:e82931. [PMID: 24376608 PMCID: PMC3871528 DOI: 10.1371/journal.pone.0082931] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 10/30/2013] [Indexed: 11/19/2022] Open
Abstract
Cellular microenvironmental conditions coordinate to regulate stem cell populations and their differentiation. Mesenchymal precursor cells (MPCs), which have significant potential for a wide range of therapeutic applications, can be expanded or differentiated into osteo- chondro- and adipogenic lineages. The ability to establish, screen, and control aspects of the microenvironment is paramount if we are to elucidate the complex interplay of signaling events that direct cell fate. Whilst modulation of Wnt signaling may be useful to direct osteogenesis in MPCs, there is still significant controversy over how the Wnt signaling pathway influences osteogenesis. In this study, we utilised a full-factorial microbioreactor array (MBA) to rapidly, combinatorially screen several Wnt modulatory compounds (CHIR99021, IWP-4 and IWR-1) and characterise their effects upon osteogenesis. The MBA screening system showed excellent consistency between donors and experimental runs. CHIR99021 (a Wnt agonist) had a profoundly inhibitory effect upon osteogenesis, contrary to expectations, whilst the effects of the IWP-4 and IWR-1 (Wnt antagonists) were confirmed to be inhibitory to osteogenesis, but to a lesser extent than observed for CHIR99021. Importantly, we demonstrated that these results were translatable to standard culture conditions. Using RT-qPCR of osteogenic and Wnt pathway markers, we showed that CHIR exerted its effects via inhibition of ALP and SPP1 expression, even though other osteogenic markers (RUNX2, MSX2, DLX, COL1A1) were upregulated. Lastly, this MBA platform, due to the continuous provision of medium from the first to the last of ten serially connected culture chambers, permitted new insight into the impacts of paracrine signaling on osteogenic differentiation in MPCs, with factors secreted by the MPCs in upstream chambers enhancing the differentiation of cells in downstream chambers. Insights provided by this cell-based assay system will be key to better understanding signaling mechanisms, as well as optimizing MPC growth and differentiation conditions for therapeutic applications.
Collapse
Affiliation(s)
- Jessica E. Frith
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St. Lucia, Queensland, Australia
| | - Drew M. Titmarsh
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St. Lucia, Queensland, Australia
| | - Harish Padmanabhan
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St. Lucia, Queensland, Australia
| | - Justin J. Cooper-White
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St. Lucia, Queensland, Australia
- School of Chemical Engineering, The University of Queensland, St. Lucia, Queensland, Australia
- Materials Science and Engineering Division, CSIRO, Clayton, Victoria, Australia
- * E-mail:
| |
Collapse
|
232
|
Yan XZ, van den Beucken JJJP, Both SK, Yang PS, Jansen JA, Yang F. Biomaterial strategies for stem cell maintenance during in vitro expansion. TISSUE ENGINEERING PART B-REVIEWS 2013; 20:340-54. [PMID: 24168361 DOI: 10.1089/ten.teb.2013.0349] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Stem cells, having the potential for self-renewal and multilineage differentiation, are the building blocks for tissue/organ regeneration. Stem cells can be isolated from various sources but are, in general, available in too small numbers to be used directly for clinical purpose without intermediate expansion procedures in vitro. Although this in vitro expansion of undifferentiated stem cells is necessary, stem cells typically diminish their ability to self-renew and proliferate during passaging. Consequently, maintaining the stemness of stem cells has been recognized as a major challenge in stem cell-based research. This review focuses on the latest developments in maintaining the self-renewal ability of stem cells during in vitro expansion by biomaterial strategies. Further, this review highlights what should be the focus for future studies using stem cells for regenerative applications.
Collapse
Affiliation(s)
- Xiang-Zhen Yan
- 1 Department of Biomaterials, Radboud University Nijmegen Medical Centre , Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|
233
|
Maruotti N, Corrado A, Neve A, Cantatore FP. Systemic effects of Wnt signaling. J Cell Physiol 2013; 228:1428-32. [PMID: 23359342 DOI: 10.1002/jcp.24326] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 01/14/2013] [Indexed: 12/15/2022]
Abstract
Wnt signaling plays a key role in several physiological and pathological aspects. Even if Wnt signal was first described more than 20 years ago, its role in systemic effects, such as angiogenesis and vascular disorders, bone biology, autoimmune diseases, neurological diseases, and neoplastic disorders, was only recently emerged through the use of animal and in vitro models. Moreover, Wnt signaling inhibitors, such as DKK-1, may be advantageously considered targets for the treatment of several diseases, including osteoporosis, vascular diseases, inflammatory diseases, neurological diseases, and cancer. Nevertheless, further studies are required to provide a complete understanding of this complex signaling pathway, and especially of its role in human diseases, considering the possible advantageous effects of Wnt signaling inhibitors on the progression of disease conditions.
Collapse
Affiliation(s)
- Nicola Maruotti
- Department of Rheumatology, University of Foggia Medical School, Foggia, Italy
| | | | | | | |
Collapse
|
234
|
Brown PT, Handorf AM, Jeon WB, Li WJ. Stem cell-based tissue engineering approaches for musculoskeletal regeneration. Curr Pharm Des 2013; 19:3429-45. [PMID: 23432679 DOI: 10.2174/13816128113199990350] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 02/10/2013] [Indexed: 01/01/2023]
Abstract
The field of regenerative medicine and tissue engineering is an ever evolving field that holds promise in treating numerous musculoskeletal diseases and injuries. An important impetus in the development of the field was the discovery and implementation of stem cells. The utilization of mesenchymal stem cells, and later embryonic and induced pluripotent stem cells, opens new arenas for tissue engineering and presents the potential of developing stem cell-based therapies for disease treatment. Multipotent and pluripotent stem cells can produce various lineage tissues, and allow for derivation of a tissue that may be comprised of multiple cell types. As the field grows, the combination of biomaterial scaffolds and bioreactors provides methods to create an environment for stem cells that better represent their microenvironment for new tissue formation. As technologies for the fabrication of biomaterial scaffolds advance, the ability of scaffolds to modulate stem cell behavior advances as well. The composition of scaffolds could be of natural or synthetic materials and could be tailored to enhance cell self-renewal and/or direct cell fates. In addition to biomaterial scaffolds, studies of tissue development and cellular microenvironments have determined other factors, such as growth factors and oxygen tension, that are crucial to the regulation of stem cell activity. The overarching goal of stem cell-based tissue engineering research is to precisely control differentiation of stem cells in culture. In this article, we review current developments in tissue engineering, focusing on several stem cell sources, induction factors including growth factors, oxygen tension, biomaterials, and mechanical stimulation, and the internal and external regulatory mechanisms that govern proliferation and differentiation.
Collapse
Affiliation(s)
- Patrick T Brown
- Wisconsin Institutes of Medical Research, 1111 Highland Ave., Madison, WI 53705, USA
| | | | | | | |
Collapse
|
235
|
Tu B, Peng ZX, Fan QM, Du L, Yan W, Tang TT. Osteosarcoma cells promote the production of pro-tumor cytokines in mesenchymal stem cells by inhibiting their osteogenic differentiation through the TGF-β/Smad2/3 pathway. Exp Cell Res 2013; 320:164-73. [PMID: 24183998 DOI: 10.1016/j.yexcr.2013.10.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 10/15/2013] [Accepted: 10/23/2013] [Indexed: 01/23/2023]
Abstract
Mesenchymal stem cells (MSCs) are among the most important components of the osteosarcoma microenvironment and are reported to promote tumor progression. However, the means by which osteosarcoma cells modulate MSC behavior remains unclear. The aim of this study was to determine the effects of osteosarcoma cells on both the production of pro-tumor cytokines by mesenchymal stem cells (MSCs) and the osteogenic differentiation of MSCs. High level of transforming growth factor-β (TGF-β) was detected in three osteosarcoma cell lines. Conditioned media (CM) from the osteosarcoma cell lines Saos-2 and U2-OS were used to stimulate the cultured MSCs. We found that osteosarcoma cells promoted the production of IL-6 and VEGF in MSCs by inhibiting their osteogenic differentiation. Furthermore, TGF-β in tumor CM was proved to be an important factor. The TGF-β neutralizing antibody antagonized the effects induced by osteosarcoma CM. The inhibition of Smad2/3 by siRNA significantly decreased the production of IL-6 and VEGF in MSCs and induced their osteogenic differentiation. We also found that Smad2/3 enhanced the expression of β-catenin in MSCs by decreasing the level of Dickkopf-1 (DKK1). Although the inhibition of β-catenin did not affect the production of IL-6 or VEGF, or the gene expression of the early osteogenic markers Runx2 and ALP, it did enhance the gene expression of osteocalcin. Taken together, our data indicate that osteosarcoma cells secrete TGF-β to maintain the stemness of MSCs and promote the production of pro-tumor cytokines by these cells.
Collapse
Affiliation(s)
- Bing Tu
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road 639, Shanghai 200011, China
| | | | | | | | | | | |
Collapse
|
236
|
Fakhry M, Hamade E, Badran B, Buchet R, Magne D. Molecular mechanisms of mesenchymal stem cell differentiation towards osteoblasts. World J Stem Cells 2013; 5:136-148. [PMID: 24179602 PMCID: PMC3812518 DOI: 10.4252/wjsc.v5.i4.136] [Citation(s) in RCA: 183] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/01/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023] Open
Abstract
Bone is a dynamic tissue that is constantly renewed by the coordinated action of two cell types, i.e., the bone-resorbing osteoclasts and the bone-forming osteoblasts. However, in some circumstances, bone regeneration exceeds bone self repair capacities. This is notably often the case after bone fractures, osteolytic bone tumor surgery, or osteonecrosis. In this regard, bone tissue engineering with autologous or allogenic mesenchymal stem cells (MSCs) is been widely developed. MSCs can be isolated from bone marrow or other tissues such as adipose tissue or umbilical cord, and can be implanted in bone defects with or without prior amplification and stimulation. However, the outcome of most pre-clinical studies remains relatively disappointing. A better understanding of the successive steps and molecular mechanisms involved in MSC-osteoblastic differentiation appears to be crucial to optimize MSC-bone therapy. In this review, we first present the important growth factors that stimulate osteoblastogenesis. Then we review the main transcription factors that modulate osteoblast differentiation, and the microRNAs (miRs) that inhibit their expression. Finally, we also discuss articles dealing with the use of these factors and miRs in the development of new bone MSC therapy strategies. We particularly focus on the studies using human MSCs, since significant differences exist between osteoblast differentiation mechanisms in humans and mice for instance.
Collapse
|
237
|
Pandurangan AK. Potential targets for prevention of colorectal cancer: a focus on PI3K/Akt/mTOR and Wnt pathways. Asian Pac J Cancer Prev 2013; 14:2201-5. [PMID: 23725112 DOI: 10.7314/apjcp.2013.14.4.2201] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers in many parts of the world. Its development is a multi-step process involving three distinct stages, initiation that alters the molecular message of a normal cell, followed by promotion and progression that ultimately generates a phenotypically altered transformed malignant cell. Reports have suggested an association of the phosphoinositide-3-kinase (PI3K)/Akt pathway with colon tumorigenesis. Activation of Akt signaling and impaired expression of phosphatase and tensin homolog (PTEN) (a negative regulator of Akt) has been reported in 60-70% of human colon cancers and inhibitors of PI3K/Akt signaling have been suggested as potential therapeutic agents. Around 80% of human colon tumors possess mutations in the APC gene and half of the remainder feature β-catenin gene mutations which affect downstream signaling of the PI3K/Akt pathway. In recent years, there has been a great focus in targeting these signaling pathways, with natural and synthetic drugs reducing the tumor burden in different experiment models. In this review we survey the role of PI3K/Akt/mTOR and Wnt signaling in CRC.
Collapse
|
238
|
Li J, Hansen KC, Zhang Y, Dong C, Dinu CZ, Dzieciatkowska M, Pei M. Rejuvenation of chondrogenic potential in a young stem cell microenvironment. Biomaterials 2013; 35:642-53. [PMID: 24148243 DOI: 10.1016/j.biomaterials.2013.09.099] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 09/25/2013] [Indexed: 11/28/2022]
Abstract
Autologous cells suffer from limited cell number and senescence during ex vivo expansion for cartilage repair. Here we found that expansion on extracellular matrix (ECM) deposited by fetal synovium-derived stem cells (SDSCs) (FE) was superior to ECM deposited by adult SDSCs (AE) in promoting cell proliferation and chondrogenic potential. Unique proteins in FE might be responsible for the rejuvenation effect of FE while advantageous proteins in AE might contribute to differentiation more than to proliferation. Compared to AE, the lower elasticity of FE yielded expanded adult SDSCs with lower elasticity which could be responsible for the enhancement of chondrogenic and adipogenic differentiation. MAPK and noncanonical Wnt signals were actively involved in ECM-mediated adult SDSC rejuvenation.
Collapse
Affiliation(s)
- Jingting Li
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, PO Box 9196, One Medical Center Drive, Morgantown, WV 26506, USA; Department of Exercise Physiology, West Virginia University, Morgantown, WV 26506, USA
| | | | | | | | | | | | | |
Collapse
|
239
|
Boudin E, Fijalkowski I, Piters E, Van Hul W. The role of extracellular modulators of canonical Wnt signaling in bone metabolism and diseases. Semin Arthritis Rheum 2013; 43:220-40. [DOI: 10.1016/j.semarthrit.2013.01.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 01/11/2013] [Accepted: 01/16/2013] [Indexed: 12/17/2022]
|
240
|
Activation of Hedgehog signaling by loss of GNAS causes heterotopic ossification. Nat Med 2013; 19:1505-12. [PMID: 24076664 PMCID: PMC3917515 DOI: 10.1038/nm.3314] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 07/19/2013] [Indexed: 12/17/2022]
Abstract
Bone formation is exquisitely controlled in space and time. Heterotopic ossification (HO), the pathologic formation of extra-skeletal bone, occurs as a common complication of trauma or in genetic disorders and can be disabling and lethal. However, the underlying molecular mechanisms are largely unknown. Here we demonstrate that Gαs restricts bone formation to the skeleton by inhibiting Hedgehog (Hh) signaling in mesenchymal progenitor cells. In progressive osseous heteroplasia (POH), a human disease caused by null mutations in GNAS that encodes Gαs, HH signaling is upregulated in ectopic osteoblasts and progenitor cells. Ectopic Hh signaling is sufficient to induce HO, while Hh signaling inhibition blocks HO in animal models. As our previous work has shown that GNAS gain of function mutations upregulate WNT/β-Catenin signaling in fibrous dysplasia (FD), our findings identify Gαs as a critical regulator of osteoblast differentiation by maintaining a balance between two key signaling pathways: Wnt/β-catenin and Hh. HH signaling inhibitors developed for cancer therapy may be repurposed to treat HO and other diseases caused by GNAS inactivation.
Collapse
|
241
|
Effects of constitutive β-catenin activation on vertebral bone growth and remodeling at different postnatal stages in mice. PLoS One 2013; 8:e74093. [PMID: 24066100 PMCID: PMC3774640 DOI: 10.1371/journal.pone.0074093] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 07/26/2013] [Indexed: 11/19/2022] Open
Abstract
Background and Objective The Wnt/β-catenin signaling pathway is essential for controlling bone mass; however, little is known about the variable effects of the constitutive activation of β-catenin (CA-β-catenin) on bone growth and remodeling at different postnatal stages. The goal of the present study was to observe the effects of CA-β-catenin on vertebral bone growth and remodeling in mice at different postnatal stages. In particular, special attention was paid to whether CA-β-catenin has detrimental effects on these processes. Methods Catnblox(ex 3) mice were crossed with mice expressing the TM-inducible Cre fusion protein, which could be activated at designated time points via injection of tamoxifen. β-catenin was stabilized by tamoxifen injection 3 days, and 2, 4, 5, and 7 months after birth, and the effects lasted for one month. Radiographic imaging, micro-computed tomography, immunohistochemistry, and safranin O and tartrate-resistant acid phosphatase staining were employed to observe the effects of CA-β-catenin on vertebral bone growth and remodeling. Results CA-β-catenin in both early (3 days after birth) and late stages (2, 4, 5, and 7 months after birth) increased bone formation and decreased bone resorption, which together increased vertebral bone volume. However, when β-catenin was stabilized in the early stage, vertebral linear growth was retarded, and the mice demonstrated shorter statures. In addition, the newly formed bone was mainly immature and located close to the growth plate. In contrast, when β-catenin was stabilized in the late stage, vertebral linear growth was unaffected, and the newly formed bone was mainly mature and evenly distributed throughout the vertebral body. Conclusions CA-β-catenin in both early and late stages of growth can increase vertebral bone volume, but β-catenin has differential effects on vertebral growth and remodeling when activated at different postnatal stages.
Collapse
|
242
|
Zeng YT, Fu B, Tang GH, Zhang L, Qian YF. Effects of lithium on extraction socket healing in rats assessed with micro-computed tomography. Acta Odontol Scand 2013; 71:1335-40. [PMID: 23351165 DOI: 10.3109/00016357.2013.764004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Lithium is an activator of β-catenin signaling and β-catenin plays an important role in regulating bone formation and remodeling. The purpose of this study was to investigate the effects of lithium on bone repair in tooth extraction sockets in rats. MATERIAL AND METHODS Twenty male Wistar rats were subjected to maxillary left second molar extraction. The animals received a daily injection of lithium chloride (LiCl) or the same dose of sodium chloride (NaCl) starting 7 days before tooth extraction until sacrifice 14 days after extraction. Rats were randomly divided into: (1) a pre-treated group that received LiCl injection from 7 days before to 3 days after tooth extraction; (2) a post-treated group that received LiCl injection starting 4 days after tooth extraction; (3) a continuously treated group that received LiCl injection for the entire 21 days; and (4) a control group that received NaCl injection only. The volume of new bone and the bone density in the extraction socket were quantified by micro-computed tomography. RESULTS The percentage of new bone formation in the extraction socket was as follows: 63.2 ± 13.4% (pre-treated group), 53.9 ± 9.8% (post-treated), 23.8 ± 8.0% (continuously treated) and 37.5 ± 4.2% (control). The difference in percentage was statistically significant between each pair of groups. Pre- and post-treated groups also showed a significant increase in the density of new bone. CONCLUSIONS Lithium enhances bone repair in extraction sockets when delivered before or after tooth extraction. Tooth extraction during lithium treatment may impair bone healing.
Collapse
Affiliation(s)
- Yun Ting Zeng
- Craniofacial Bone Biology and Repair, Shanghai Key Laboratory of Stomatology, Shanghai, PR China
| | | | | | | | | |
Collapse
|
243
|
Kong X, Liu Y, Ye R, Zhu B, Zhu Y, Liu X, Hu C, Luo H, Zhang Y, Ding Y, Jin Y. GSK3β is a checkpoint for TNF-α-mediated impaired osteogenic differentiation of mesenchymal stem cells in inflammatory microenvironments. Biochim Biophys Acta Gen Subj 2013; 1830:5119-29. [PMID: 23911749 DOI: 10.1016/j.bbagen.2013.07.027] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 07/02/2013] [Accepted: 07/24/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND The fate and differentiation of mesenchymal stem cells (MSCs) depend on various microenvironmental cues. In chronic inflammatory bone disease, bone regeneration is inhibited. The present study therefore sought to identify the underlying molecule mechanisms. METHODS We isolated periodontal ligament stem cells (PDLSCs), a new population of MSCs, from the periodontal ligament tissues of periodontitis patients and healthy controls (p-PDLSCs and h-PDLSCs). The secretion of inflammatory cytokines, like TNF-α, IL-1β, IL-6 and IL-8, after LPS stimulation was measured by ELISA. The expressions of p-GSK3β and GSK3β in two types of PDLSCs were detected by Western blot. TOPFlash was used to assay the Tcf/Lef transcriptional activity. Knockdown of GSK3β by siRNA and over-expression of GSK3β by adenoviruses were performed to confirm the role of GSK3β in the impaired osteogenic differentiation of PDLSCs under inflammatory microenvironment. RESULTS We demonstrated that p-PDLSCs displayed impaired osteogenic capacity than h-PDLSCs. Upon inflammatory stimulation, monocytes, but not PDLSCs, released inflammatory cytokines among which TNF-α directly act on PDLSCs and suppressed their osteogenic differentiation. TNF-α induced the phosphorylation of GSK3β, the deactivated form of GSK3β, which increased nuclear β-catenin and Lef-1 accumulation, and eventually reduced the Runx2-associated osteogenesis in PDLSCs. Over-expression of GSK3β rescued osteogenesis in TNF-α-stimulated PDLSCs, whereas inactivation of GSK3β was sufficient to liberate the β-catenin/Lef-1/Runx2 pathway. CONCLUSION GSK3β plays an obligatory role in the TNF-α-mediated inhibition of osteogenesis in MSCs. GENERAL SIGNIFICANCE The strategy to target GSK3β may provide a potential approach to bone regeneration in inflammatory microenvironments.
Collapse
Affiliation(s)
- Xiangwei Kong
- Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China; Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, China; Department of Stomatology, Nanjing Bayi Hospital, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Centola M, Tonnarelli B, Schären S, Glaser N, Barbero A, Martin I. Priming 3D cultures of human mesenchymal stromal cells toward cartilage formation via developmental pathways. Stem Cells Dev 2013; 22:2849-58. [PMID: 23777290 DOI: 10.1089/scd.2013.0216] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The field of regenerative medicine has increasingly recognized the importance to be inspired by developmental processes to identify signaling pathways crucial for 3D organogenesis and tissue regeneration. Here, we aimed at recapitulating the first events occurring during limb development (ie, cell condensation and expansion of an undifferentiated mesenchymal cell population) to prime 3D cultures of human bone marrow-derived mesenchymal stromal/stem cells (hBM-MSC) toward the chondrogenic route. Based on embryonic development studies, we hypothesized that Wnt3a and fibroblast growth factor 2 (FGF2) induce hBM-MSC to proliferate in 3D culture as an undifferentiated pool of progenitors (defined by clonogenic capacity and expression of typical markers), retaining chondrogenic potential upon induction by suitable morphogens. hBM-MSC were responsive to Wnt signaling in 3D pellet culture, as assessed by significant upregulation of main target genes and increase of unphosphorylated β-catenin levels. Wnt3a was able to induce a five-fold increase in the number of proliferating hBM-MSC (6.4% vs. 1.3% in the vehicle condition), although total DNA content of the 3D construct was decreasing over time. Preconditioning with Wnt3a improved transforming growth factor-β1 mediated chondrogenesis (30% more glycosaminoglycans/cell in average). In contrast to developmental and 2D MSC culture models, FGF2 antagonized the Wnt-mediated effects. Interestingly, the CD146⁺ subpopulation was found to be more responsive to Wnt3a. The presented data indicate a possible strategy to prime 3D cultures of hBM-MSC by invoking a "developmental engineering" approach. The study also identifies some opportunities and challenges to cross-fertilize skeletal development models and 3D hBM-MSC culture systems.
Collapse
Affiliation(s)
- Matteo Centola
- Departments of Surgery and Biomedicine, University Hospital Basel, University of Basel , Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
245
|
Knight MN, Hankenson KD. Mesenchymal Stem Cells in Bone Regeneration. Adv Wound Care (New Rochelle) 2013; 2:306-316. [PMID: 24527352 DOI: 10.1089/wound.2012.0420] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Indexed: 01/14/2023] Open
Abstract
SIGNIFICANCE Mesenchymal stem cells (MSCs) play a key role in fracture repair by differentiating to become bone-forming osteoblasts and cartilage-forming chondrocytes. Cartilage then serves as a template for additional bone formation through the process of endochondral ossification. RECENT ADVANCES Endogenous MSCs that contribute to healing are primarily derived from the periosteum, endosteum, and marrow cavity, but also may be contributed from the overlying muscle or through systemic circulation, depending on the type of injury. A variety of growth factor signaling pathways, including BMP, Wnt, and Notch signaling, influence MSC proliferation and differentiation. These MSCs can be therapeutically manipulated to promote differentiation. Furthermore, MSCs can be harvested, cultivated, and delivered to promote bone healing. CRITICAL ISSUES Pharmacologically manipulating the number and differentiation capacity of endogenous MSCs is one potential therapeutic approach to improve healing; however, ideal agents to influence signaling pathways need to be developed and additional therapeutics that activate endogenous MSCs are needed. Whether isolated and purified, MSCs participate directly in the healing process or serve a bystander effect and indirectly influence healing is not well defined. FUTURE DIRECTIONS Studies must focus on better understanding the regulation of endogenous MSCs durings fracture healing. This will reveal novel molecules and pathways to therapeutically target. Similarly, while animal models have demonstrated efficacy in the delivery of MSCs to promote healing, more research is needed to understand ideal donor cells, cultivation methods, and delivery before stem cell therapy approaches can be utilized to repair bone.
Collapse
Affiliation(s)
- M. Noelle Knight
- Veterinary Medical Scientist Training Program, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kurt D. Hankenson
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
246
|
Giuliani N, Lisignoli G, Magnani M, Racano C, Bolzoni M, Dalla Palma B, Spolzino A, Manferdini C, Abati C, Toscani D, Facchini A, Aversa F. New insights into osteogenic and chondrogenic differentiation of human bone marrow mesenchymal stem cells and their potential clinical applications for bone regeneration in pediatric orthopaedics. Stem Cells Int 2013; 2013:312501. [PMID: 23766767 PMCID: PMC3676919 DOI: 10.1155/2013/312501] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 05/08/2013] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) are pluripotent adult stem cells capable of being differentiated into osteoblasts, adipocytes, and chondrocytes. The osteogenic differentiation of hMSCs is regulated either by systemic hormones or by local growth factors able to induce specific intracellular signal pathways that modify the expression and activity of several transcription factors. Runt-related transcription factor 2 (Runx2) and Wnt signaling-related molecules are the major factors critically involved in the osteogenic differentiation process by hMSCs, and SRY-related high-mobility-group (HMG) box transcription factor 9 (SOX9) is involved in the chondrogenic one. hMSCs have generated a great interest in the field of regenerative medicine, particularly in bone regeneration. In this paper, we focused our attention on the molecular mechanisms involved in osteogenic and chondrogenic differentiation of hMSC, and the potential clinical use of hMSCs in osteoarticular pediatric disease characterized by fracture nonunion and pseudarthrosis.
Collapse
Affiliation(s)
- Nicola Giuliani
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Gina Lisignoli
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale e Laboratorio RAMSES, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Marina Magnani
- Paediatric Orthopaedics and Traumatology, Rizzoli Orthopaedic Institute, Via GC Pupilli 1, 40136 Bologna, Italy
| | - Costantina Racano
- Paediatric Orthopaedics and Traumatology, Rizzoli Orthopaedic Institute, Via GC Pupilli 1, 40136 Bologna, Italy
| | - Marina Bolzoni
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Benedetta Dalla Palma
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Angelica Spolzino
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Cristina Manferdini
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale e Laboratorio RAMSES, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Caterina Abati
- Paediatric Orthopaedics and Traumatology, Rizzoli Orthopaedic Institute, Via GC Pupilli 1, 40136 Bologna, Italy
| | - Denise Toscani
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Andrea Facchini
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale e Laboratorio RAMSES, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Franco Aversa
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| |
Collapse
|
247
|
Asumda FZ. Age-associated changes in the ecological niche: implications for mesenchymal stem cell aging. Stem Cell Res Ther 2013; 4:47. [PMID: 23673056 PMCID: PMC3706986 DOI: 10.1186/scrt197] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Adult stem cells are critical for organ-specific regeneration and self-renewal with advancing age. The prospect of being able to reverse tissue-specific post-injury sequelae by harvesting, culturing and transplanting a patient's own stem and progenitor cells is exciting. Mesenchymal stem cells have emerged as a reliable stem cell source for this treatment modality and are currently being tested in numerous ongoing clinical trials. Unfortunately, the fervor over mesenchymal stem cells is mitigated by several lines of evidence suggesting that their efficacy is limited by natural aging. This article discusses the mechanisms and manifestations of age-associated deficiencies in mesenchymal stem cell efficacy. A consideration of recent experimental findings suggests that the ecological niche might be responsible for mesenchymal stem cell aging.
Collapse
Affiliation(s)
- Faizal Z Asumda
- Saint James School of Medicine, 1480 Renaissance Drive, Park Ridge, Chicago, IL, 60068, USA
| |
Collapse
|
248
|
Boddy SL, Chen W, Romero-Guevara R, Kottam L, Bellantuono I, Rivolta MN. Inner ear progenitor cells can be generated in vitro from human bone marrow mesenchymal stem cells. Regen Med 2013; 7:757-67. [PMID: 23164077 DOI: 10.2217/rme.12.58] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM Mouse mesenchymal stem cells (MSCs) can generate sensory neurons and produce inner ear hair cell-like cells. An equivalent source from humans is highly desirable, given their potential application in patient-specific regenerative therapies for deafness. In this study, we explored the ability of human MSCs (hMSCs) to differentiate into otic lineages. MATERIALS & METHODS hMSCs were exposed to culture media conditioned by human fetal auditory stem cells. RESULTS Conditioned media induced the expression of otic progenitor markers PAX8, PAX2, GATA3 and SOX2. After 4 weeks, cells coexpressed ATOH1, MYO7A and POU4F3 (indicators of hair cell lineage) or neuronal markers NEUROG1, POU4F1 and NEFH. Inhibition of WNT signaling prevented differentiation into otic progenitors, while WNT activation partially phenocopied results seen with the conditioned media. CONCLUSION This study demonstrates that hMSCs can be driven to express key genes found in the otic lineages and thereby promotes their status as candidates for regenerative therapies for deafness.
Collapse
Affiliation(s)
- Sarah L Boddy
- Centre for Stem Cell Biology & Department of Biomedical Sciences, University of Sheffield, Alfred Denny Building, Western Bank, Sheffield S10 2TN, UK
| | | | | | | | | | | |
Collapse
|
249
|
Ma B, Landman EBM, Miclea RL, Wit JM, Robanus-Maandag EC, Post JN, Karperien M. WNT signaling and cartilage: of mice and men. Calcif Tissue Int 2013; 92:399-411. [PMID: 23212543 DOI: 10.1007/s00223-012-9675-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 11/03/2012] [Indexed: 01/08/2023]
Abstract
In adult articular cartilage, the extracellular matrix is maintained by a balance between the degradation and the synthesis of matrix components. Chondrocytes that sparsely reside in the matrix and rarely proliferate are the key cellular mediators for cartilage homeostasis. There are indications for the involvement of the WNT signaling pathway in maintaining articular cartilage. Various WNTs are involved in the subsequent stages of chondrocyte differentiation during development, and deregulation of WNT signaling was observed in cartilage degeneration. Even though gene expression and protein synthesis can be activated upon injury, articular cartilage has a limited ability of self-repair and efforts to regenerate articular cartilage have so far not been successful. Because WNT signaling was found to be involved in the development and maintenance of cartilage as well as in the degeneration of cartilage, interfering with this pathway might contribute to improving cartilage regeneration. However, most of the studies on elucidating the role of WNT signaling in these processes were conducted using in vitro or in vivo animal models. Discrepancies have been found in the role of WNT signaling between chondrocytes of mouse and human origin, and extrapolation of results from mouse models to the human situation remains a challenge. Elucidation of detailed WNT signaling functions will provide knowledge to improve cartilage regeneration.
Collapse
Affiliation(s)
- Bin Ma
- Department of Developmental BioEngineering, University of Twente, Drienerlolaan 5, 7522NB, Enschede, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
250
|
Shi Q, Luo S, Jia H, Feng L, Lu X, Zhou L, Cai J. Wnt/β-catenin signaling may be involved with the maturation, but not the differentiation, of insulin-producing cells. Biomed Pharmacother 2013; 67:745-50. [PMID: 23680408 DOI: 10.1016/j.biopha.2013.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 04/08/2013] [Indexed: 10/26/2022] Open
Abstract
Wnt/β-catenin signaling (WNT) has widespread roles during stem cell differentiation. Whether WNT suppresses or promotes insulin-producing cell (IPC) differentiation and function is still not known. In this study, we investigated the role of WNT signaling during human adipose-derived stem cell (hADSC) differentiation into IPCs. Western blot analysis revealed that several key components of WNT were dynamically regulated in a 12-day IPC differentiation assay. Specifically, protein levels of Wnt1, β-catenin, and GSK3β steadily increased from day 0 to day 9 and rapidly decreased by day 12 of differentiation. Similarly, endonuclear β-catenin levels peaked at day 9 and then, fell to pre-differentiation levels. The expression of two WNT pathway targets, TCF-1 and cyclin D1, closely followed the same pattern of regulation, confirming that WNT signaling was transiently activated during IPC differentiation. Interestingly, the inhibition of WNT signaling did not block IPC differentiation; instead, it resulted in the upregulation of IPC-specific markers, including PDX-1, insulin, IRS-1, and IRS-2. Notably, another IPC marker, glucokinase, remained downregulated since it is a direct target of WNT signaling. Next, we examined the effect of maintaining active WNT signaling from day 9 to day 12 of IPC differentiation. Differentiating cells were treated with Wnt1 on day 9, when WNT signaling is typically turned off, and subjected to gene expression analysis on day 12. Remarkably, Wnt1 treatment resulted in reduced expression of IPC-specific markers. Taken together, these data indicate that WNT may not be necessary for IPC differentiation but may be involved in IPC maturation.
Collapse
Affiliation(s)
- Qiping Shi
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, China.
| | | | | | | | | | | | | |
Collapse
|