1
|
Xiao X, Zhang M, Qian Y, Wang X, Wu Q. KLF9 regulates osteogenic differentiation of mesenchymal stem cells. J Mol Histol 2024; 55:503-512. [PMID: 38801643 DOI: 10.1007/s10735-024-10204-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 05/16/2024] [Indexed: 05/29/2024]
Abstract
Osteoporosis is a progressive skeletal disease which is characterized by reduced bone mass and degradation of bone microstructure. Mesenchymal stem cells (MSCs) have the potential to inhibit osteoporosis since they are multipotent stem cells that can differentiate into multiple types of cells including osteoblasts. Hence the mechanism of osteogenic differentiation of MSCs deserves comprehensive study. Here we report that KLF9 is a novel regulator in osteogenic differentiation of MSCs. We observed that depletion of KLF9 can largely compromise the osteogenic differentiation ability of MSCs. In addition, we revealed that inhibition of the PI3K-Akt pathway could also affect osteogenic differentiation since KLF9 depletion inhibits PI3K expression. Finally, we discovered that KLF9 expression can be induced by dexamethasone which is an essential component in osteogenic induction medium. Taken together, our study provides new insights into the regulatory role of KLF9 in osteogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Xiaoxiao Xiao
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
- Faculty of Chinese Medicine, Hunan Traditional Chinese Medical College, Zhuzhou, China
| | - Ming Zhang
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yiwei Qian
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Xuepeng Wang
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Qiang Wu
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
2
|
Daponte V, Henke K, Drissi H. Current perspectives on the multiple roles of osteoclasts: Mechanisms of osteoclast-osteoblast communication and potential clinical implications. eLife 2024; 13:e95083. [PMID: 38591777 PMCID: PMC11003748 DOI: 10.7554/elife.95083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/29/2024] [Indexed: 04/10/2024] Open
Abstract
Bone remodeling is a complex process involving the coordinated actions of osteoblasts and osteoclasts to maintain bone homeostasis. While the influence of osteoblasts on osteoclast differentiation is well established, the reciprocal regulation of osteoblasts by osteoclasts has long remained enigmatic. In the past few years, a fascinating new role for osteoclasts has been unveiled in promoting bone formation and facilitating osteoblast migration to the remodeling sites through a number of different mechanisms, including the release of factors from the bone matrix following bone resorption and direct cell-cell interactions. Additionally, considerable evidence has shown that osteoclasts can secrete coupling factors known as clastokines, emphasizing the crucial role of these cells in maintaining bone homeostasis. Due to their osteoprotective function, clastokines hold great promise as potential therapeutic targets for bone diseases. However, despite long-standing work to uncover new clastokines and their effect in vivo, more substantial efforts are still required to decipher the mechanisms and pathways behind their activity in order to translate them into therapies. This comprehensive review provides insights into our evolving understanding of the osteoclast function, highlights the significance of clastokines in bone remodeling, and explores their potential as treatments for bone diseases suggesting future directions for the field.
Collapse
Affiliation(s)
- Valentina Daponte
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
- VA Medical CenterAtlantaUnited States
| | - Katrin Henke
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
- VA Medical CenterAtlantaUnited States
| |
Collapse
|
3
|
Wang F, Ye Y, Zhang Z, Teng W, Sun H, Chai X, Zhou X, Chen J, Mou H, Eloy Y, Jin X, Chen L, Shao Z, Wu Y, Shen Y, Liu A, Lin P, Wang J, Yu X, Ye Z. PDGFR in PDGF-BB/PDGFR Signaling Pathway Does Orchestrates Osteogenesis in a Temporal Manner. RESEARCH (WASHINGTON, D.C.) 2023; 6:0086. [PMID: 37223474 PMCID: PMC10202377 DOI: 10.34133/research.0086] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 02/13/2023] [Indexed: 12/01/2023]
Abstract
Platelet-derived growth factor-BB (PDGF-BB)/platelet-derived growth factor receptor-β (PDGFR-β) pathway is conventionally considered as an important pathway to promote osteogenesis; however, recent study suggested its role during osteogenesis to be controversial. Regarding the differential functions of this pathway during 3 stages of bone healing, we hypothesized that temporal inhibition of PDGF-BB/PDGFR-β pathway could shift the proliferation/differentiation balance of skeletal stem and progenitor cells, toward osteogenic lineage, which leads to improved bone regeneration. We first validated that inhibition of PDGFR-β at late stage of osteogenic induction effectively enhanced differentiation toward osteoblasts. This effect was also replicated in vivo by showing accelerated bone formation when block PDGFR-β pathway at late stage of critical bone defect healing mediated using biomaterials. Further, we found that such PDGFR-β inhibitor-initiated bone healing was also effective in the absence of scaffold implantation when administrated intraperitoneally. Mechanistically, timely inhibition of PDGFR-β blocked extracellular regulated protein kinase 1/2 pathway, which shift proliferation/differentiation balance of skeletal stem and progenitor cell to osteogenic lineage by upregulating osteogenesis-related products of Smad to induce osteogenesis. This study offered updated understanding of the use of PDGFR-β pathway and provides new insight routes of action and novel therapeutic methods in the field of bone repair.
Collapse
Affiliation(s)
- Fangqian Wang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Yuxiao Ye
- School of Material Science and Engineering, University of New South Wales, Sydney 2052, Australia
| | - Zengjie Zhang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Wangsiyuan Teng
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Hangxiang Sun
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Xupeng Chai
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Xingzhi Zhou
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Jiayu Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Haochen Mou
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Yinwang Eloy
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Xiaoqiang Jin
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Liang Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Zhenxuan Shao
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Yan Wu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Yue Shen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - An Liu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Peng Lin
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Jianwei Wang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Xiaohua Yu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Zhaoming Ye
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| |
Collapse
|
4
|
Improvement of Bone Marrow Necrosis by Tyrosine Kinase Inhibitor Substitution in a Pediatric Patient With Philadelphia Chromosome-positive Acute Lymphoblastic Leukemia. J Pediatr Hematol Oncol 2022; 44:e539-e542. [PMID: 33843813 DOI: 10.1097/mph.0000000000002157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/06/2021] [Indexed: 11/26/2022]
Abstract
Bone marrow necrosis (BMN) describes necrosis of the myeloid tissues without cortical bone involvement. Imatinib, a tyrosine kinase inhibitor, can trigger BMN during the treatment of malignant disease. In such cases, it is necessary to reduce imatinib dose or discontinue its administration, which could influence treatment outcomes. Here, we report a 6-year-old boy with Philadelphia chromosome-positive acute lymphoblastic leukemia, who developed BMN in response to imatinib. We replaced imatinib with dasatinib, and necrotic lesions gradually disappeared and were never exacerbated. In Philadelphia chromosome-positive acute lymphoblastic leukemia with BMN, tyrosine kinase inhibitor replacement may allow continued chemotherapy without intensity reduction.
Collapse
|
5
|
Su Q, Xu B, Tian Z, Gong Z. 1,3,5-triazines inhibit osteosarcoma and avert lung metastasis in a patient-derived orthotopic xenograft mouse model with favorable pharmacokinetics. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:295-301. [PMID: 35656180 PMCID: PMC9148403 DOI: 10.22038/ijbms.2022.62705.13873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/23/2022] [Indexed: 11/10/2022]
Abstract
Objectives Osteosarcoma is a major solid malignant tumor of bone, possessing significant burden on healthcare due to non-availability of specific anticancer agents. The current study was conducted to identify novel 1,3,5-triazine derivatives against osteosarcoma. Materials and Methods The compounds were synthesized in a straight-forward two-step reaction and subsequently tested against PI3K and mTOR kinase and anticancer activity against osteosarcoma cells (MG-63, U2-OS, and Saos-2). The effect of the most potent compound was evaluated on apoptosis and cell phase of Saos-2 cells. The pharmacological activity was further established in the patient-derived orthotopic xenograft (PDOX) mouse model. Results The developed compounds 8 (a-f) showed significant inhibitory activities against PI3K, mTOR, and OS cells. Among the tested series, compound 8a showed highly potent PI3K/mTOR inhibitory activity with significant anticancer activity against Saos-2 cells compared with Imatinib as standard. It also induces apoptosis and causes G2/M arrest in Saos-2 cells. Compound 8a significantly improved body weight, reduced tumor volume, and inhibited lung metastasis in athymic nude mice in a PDOX mouse model. It also showed optimal pharmacokinetic parameters in SD rats. Conclusion In summary, 1,3,5-triazine analogs were identified as new PI3K/mTOR inhibitors against osteosarcoma.
Collapse
Affiliation(s)
- Qing Su
- Department of Orthopedic Oncology, Yantai Shan Hospital, Yantai, 264003, China
| | - Baolin Xu
- Department of Orthopedics, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Zhoubin Tian
- Departments of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Ziling Gong
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233, China
| |
Collapse
|
6
|
Orticelli V, Papait A, Vertua E, Bonassi Signoroni P, Romele P, Di Pietro L, Magatti M, Teofili L, Silini AR, Parolini O. Human amniotic mesenchymal stromal cells support the ex vivo expansion of cord blood hematopoietic stem cells. Stem Cells Transl Med 2021; 10:1516-1529. [PMID: 34327849 PMCID: PMC8550705 DOI: 10.1002/sctm.21-0130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/03/2021] [Accepted: 06/06/2021] [Indexed: 12/29/2022] Open
Abstract
Currently, more than 30 000 allogeneic hematopoietic stem cell (HSC) transplantations have been performed for the treatment of hematological and nonhematological diseases using HSC from umbilical cord blood (CB). However, the wide utilization of CB as a source of HSC is limited by the low number of cells recovered. One strategy to expand ex vivo CB‐HSC is represented by the use of bone marrow mesenchymal stromal cells (BM‐MSCs) as a feeder to enhance HSC proliferation while maintaining HSC stemness. Indeed, BM‐MSCs have been recognized as one of the most relevant players in the HSC niche. Thus, it has been hypothesized that they can support the ex vivo expansion of HSC by mimicking the physiological microenvironment present in the hematopoietic niche. Due to the role of placenta in supporting fetal hematopoiesis, MSC derived from the amniotic membrane (hAMSC) of human term placenta could represent an interesting alternative to BM‐MSC as a feeder layer to enhance the proliferation and maintain HSC stemness. Therefore, in this study we investigated if hAMSC could support the ex vivo expansion of HSC and progenitor cells. The capacity of hAMSCs to support the ex vivo expansion of CB‐HSC was evaluated in comparison to the control condition represented by the CB‐CD34+ cells without a feeder layer. The coculture was performed at two different CD34+:MSC ratios (1:2 and 1:8) in both cell‐to‐cell contact and transwell setting. After 7 days, the cells were collected and analyzed for phenotype and functionality. Our results suggest that hAMSCs represent a valuable alternative to BM‐MSC to support: (a) the ex vivo expansion of CB‐HSC in both contact and transwell systems, (b) the colony forming unit ability, and (c) long‐term culture initiating cells ability. Overall, these findings may contribute to address the unmet need of high HSC content in CB units available for transplantation.
Collapse
Affiliation(s)
- Valentina Orticelli
- Dipartimento di Scienze della vita e sanità pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.,IRCCS Fondazione Policlinico Universitario "Agostino Gemelli", Rome, Italy
| | - Andrea Papait
- Dipartimento di Scienze della vita e sanità pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.,Centro di Ricerca E. Menni, Fondazione Poliambulanza, Brescia, Italy
| | - Elsa Vertua
- Centro di Ricerca E. Menni, Fondazione Poliambulanza, Brescia, Italy
| | | | - Pietro Romele
- Centro di Ricerca E. Menni, Fondazione Poliambulanza, Brescia, Italy
| | - Lorena Di Pietro
- Dipartimento di Scienze della vita e sanità pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marta Magatti
- Centro di Ricerca E. Menni, Fondazione Poliambulanza, Brescia, Italy
| | - Luciana Teofili
- IRCCS Fondazione Policlinico Universitario "Agostino Gemelli", Rome, Italy
| | | | - Ornella Parolini
- Dipartimento di Scienze della vita e sanità pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.,IRCCS Fondazione Policlinico Universitario "Agostino Gemelli", Rome, Italy
| |
Collapse
|
7
|
Yin S, Cao Y. Hydrogels for Large-Scale Expansion of Stem Cells. Acta Biomater 2021; 128:1-20. [PMID: 33746032 DOI: 10.1016/j.actbio.2021.03.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 12/18/2022]
Abstract
Stem cells demonstrate considerable promise for various preclinical and clinical applications, including drug screening, disease treatments, and regenerative medicine. Producing high-quality and large amounts of stem cells is in demand for these applications. Despite challenges, as hydrogel-based cell culture technology has developed, tremendous progress has been made in stem cell expansion and directed differentiation. Hydrogels are soft materials with abundant water. Many hydrogel properties, including biodegradability, mechanical strength, and porosity, have been shown to play essential roles in regulating stem cell proliferation and differentiation. The biochemical and physical properties of hydrogels can be specifically tailored to mimic the native microenvironment that various stem cells reside in vivo. A few hydrogel-based systems have been developed for successful stem cell cultures and expansion in vitro. In this review, we summarize various types of hydrogels that have been designed to effectively enhance the proliferation of hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs), respectively. According to each stem cell type's preference, we also discuss strategies for fabricating hydrogels with biochemical and mechanical cues and other characteristics representing microenvironments of stem cells in vivo. STATEMENT OF SIGNIFICANCE: In this review article we summarize current progress on the construction of hydrogel systems for the culture and expansion of various stem cells, including hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs). The Significance includes: (1) Provide detailed discussion on the stem cell niches that should be considered for stem cell in vitro expansion. (2) Summarize various strategies to construct hydrogels that can largely recapture the microenvironment of native stem cells. (3) Suggest a few future directions that can be implemented to improve current in vitro stem cell expansion systems.
Collapse
Affiliation(s)
- Sheng Yin
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China; Chemistry and Biomedicine innovation center, Nanjing University, Nanjing, 210093, China; Institute for Brain Sciences, Nanjing University, Nanjing, 210093, China; Shenzhen Research Institute of Nanjing University, Shenzhen, China, 518057
| | - Yi Cao
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China; Chemistry and Biomedicine innovation center, Nanjing University, Nanjing, 210093, China; Institute for Brain Sciences, Nanjing University, Nanjing, 210093, China; Shenzhen Research Institute of Nanjing University, Shenzhen, China, 518057.
| |
Collapse
|
8
|
Towards Physiologic Culture Approaches to Improve Standard Cultivation of Mesenchymal Stem Cells. Cells 2021; 10:cells10040886. [PMID: 33924517 PMCID: PMC8069108 DOI: 10.3390/cells10040886] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are of great interest for their use in cell-based therapies due to their multipotent differentiation and immunomodulatory capacities. In consequence of limited numbers following their isolation from the donor tissue, MSCs require extensive expansion performed in traditional 2D cell culture setups to reach adequate amounts for therapeutic use. However, prolonged culture of MSCs in vitro has been shown to decrease their differentiation potential and alter their immunomodulatory properties. For that reason, preservation of these physiological characteristics of MSCs throughout their in vitro culture is essential for improving the efficiency of therapeutic and in vitro modeling applications. With this objective in mind, many studies already investigated certain parameters for enhancing current standard MSC culture protocols with regard to the effects of specific culture media components or culture conditions. Although there is a lot of diversity in the final therapeutic uses of the cells, the primary stage of standard isolation and expansion is imperative. Therefore, we want to review on approaches for optimizing standard MSC culture protocols during this essential primary step of in vitro expansion. The reviewed studies investigate and suggest improvements focused on culture media components (amino acids, ascorbic acid, glucose level, growth factors, lipids, platelet lysate, trace elements, serum, and xenogeneic components) as well as culture conditions and processes (hypoxia, cell seeding, and dissociation during passaging), in order to preserve the MSC phenotype and functionality during the primary phase of in vitro culture.
Collapse
|
9
|
Chen J, Shen Y, Shen Z, Cheng L, Zhou S. Tissue engineering of the larynx: A contemporary review. J Clin Lab Anal 2020; 35:e23646. [PMID: 33320365 PMCID: PMC7891509 DOI: 10.1002/jcla.23646] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/17/2022] Open
Abstract
Objective Tissue engineering has been a topic of extensive research in recent years and has been applied to the regeneration and restoration of many organs including the larynx. Currently, research investigating tissue engineering of the larynx is either ongoing or in the preclinical trial stage. Methods A literature search was performed on the Advanced search field of PubMed using the keywords: “(laryncheal tissue engineering) AND (cartilage regeneration OR scaffolds OR stem cells OR biomolecules).” After applying the selection criteria, 65 articles were included in the study. Results The present review focuses on the rapidly expanding field of tissue‐engineered larynx, which aims to provide stem cell–based scaffolds combined with biological active factors such as growth factors for larynx reconstruction and regeneration. The trend in recent studies is to use new techniques for scaffold construction, such as 3D printing, are developed. All of these strategies have been instrumental in guiding optimization of the tissue‐engineered larynx, leading to a level of clinical induction beyond the in vivo animal experimental phase. Conclusions This review summarizes the current progress and outlines the necessary basic components of regenerative laryngeal medicine in preclinical fields. Finally, it considers the design of scaffolds, support of growth factors, and cell therapies toward potential clinical application.
Collapse
Affiliation(s)
- Jingjing Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Lihuili Hospital, Ningbo University, Ningbo, 315040, China.,Department of Otorhinolaryngology- Head and Neck Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Yi Shen
- Department of Otorhinolaryngology-Head and Neck Surgery, Lihuili Hospital, Ningbo University, Ningbo, 315040, China
| | - Zhisen Shen
- Department of Otorhinolaryngology-Head and Neck Surgery, Lihuili Hospital, Ningbo University, Ningbo, 315040, China
| | - Lixin Cheng
- Department of Otorhinolaryngology-Head and Neck Surgery, Lihuili Hospital, Ningbo University, Ningbo, 315040, China
| | - Shuihong Zhou
- Department of Otorhinolaryngology- Head and Neck Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| |
Collapse
|
10
|
Hong HS, Kim S, Jin Y, Son Y. Substance P enhances the therapeutic effect of MSCs by modulating their angiogenic potential. J Cell Mol Med 2020; 24:12560-12571. [PMID: 32985796 PMCID: PMC7687016 DOI: 10.1111/jcmm.15804] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/30/2020] [Accepted: 08/02/2020] [Indexed: 01/01/2023] Open
Abstract
Bone marrow mesenchymal stem cell (MSC) therapy acts through multiple differentiations in damaged tissue or via secretion of paracrine factors, as demonstrated in various inflammatory and ischaemic diseases. However, long‐term ex vivo culture to obtain a sufficient number of cells in MSC transplantation leads to cellular senescence, deficiency of the paracrine potential, and loss of survival rate post‐transplantation. In this study, we evaluated whether supplementation of MSCs with substance P (SP) can improve their therapeutic potential. SP treatment elevated the secretion of paracrine/angiogenic factors, including VEGF, SDF‐1a and PDGF‐BB, from late passage MSCs in vitro. MSCs supplemented with SP accelerated epidermal/dermal regeneration and neovascularization and suppressed inflammation in vivo, compared to MSCs transplanted alone. Importantly, supplementation with SP enabled the incorporation of transplanted human MSCs into the host vasculature as pericytes via PDGF signalling, leading to the direct engagement of transplanted cells in compact vasculature formation. Our results showed that SP is capable of restoring the cellular potential of senescent stem cells, possibly by modulating the generation of paracrine factors from MSCs, which might accelerate MSC‐mediated tissue repair. Thus, SP is anticipated to be a potential beneficial agent in MSC therapy for inflammatory or ischaemic diseases and cutaneous wounds.
Collapse
Affiliation(s)
- Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, Korea.,East-West Medical Research Institute, Kyung Hee University Hospital, Seoul, Korea.,Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Seoul, Korea
| | - Suna Kim
- Department of Genetic Engineering, College of Life Science, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea
| | - Yinji Jin
- Department of Genetic Engineering, College of Life Science, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea
| | - Youngsook Son
- Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Seoul, Korea.,Department of Genetic Engineering, College of Life Science, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea
| |
Collapse
|
11
|
Thanasrisuebwong P, Kiattavorncharoen S, Surarit R, Phruksaniyom C, Ruangsawasdi N. Red and Yellow Injectable Platelet-Rich Fibrin Demonstrated Differential Effects on Periodontal Ligament Stem Cell Proliferation, Migration, and Osteogenic Differentiation. Int J Mol Sci 2020; 21:ijms21145153. [PMID: 32708242 PMCID: PMC7404021 DOI: 10.3390/ijms21145153] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/14/2020] [Accepted: 07/20/2020] [Indexed: 02/07/2023] Open
Abstract
The biological benefits of using two fractions derived from injectable platelet-rich fibrin (i-PRF) in bone regeneration remain unclear. Thus, the current study examined two fractionation protocols producing yellow i-PRF and red i-PRF on periodontal ligament stem cells (PDLSCs). The i-PRF samples from five donors were harvested from two different levels, with and without a buffy coat layer, to obtain red and yellow i-PRF, respectively. The PDLSCs were isolated and characterized before their experimental use. The culture medium in each assay was loaded with 20% of the conditioned medium containing the factors released from the red and yellow i-PRF. Cell proliferation and cell migration were determined with an MTT and trans-well assay, respectively. Osteogenic differentiation was investigated using alkaline phosphatase and Alizarin red staining. The efficiency of both i-PRFs was statistically compared. We found that the factors released from the red i-PRF had a greater effect on cell proliferation and cell migration. Moreover, the factors released from the yellow i-PRF stimulated PDLSC osteogenic differentiation earlier compared with the red i-PRF. These data suggest that the red i-PRF might be suitable for using in bone regeneration because it induced the mobilization and growth of bone regenerative cells without inducing premature mineralization.
Collapse
Affiliation(s)
- Prakan Thanasrisuebwong
- Dental Implant Center, Dental Hospital, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand;
| | - Sirichai Kiattavorncharoen
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand;
| | - Rudee Surarit
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand;
| | - Chareerut Phruksaniyom
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand;
| | - Nisarat Ruangsawasdi
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand;
- Correspondence:
| |
Collapse
|
12
|
A Comprehensive Review of Concentrated Growth Factors and Their Novel Applications in Facial Reconstructive and Regenerative Medicine. Aesthetic Plast Surg 2020; 44:1047-1057. [PMID: 31970453 DOI: 10.1007/s00266-020-01620-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/12/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Concentrated growth factors (CGFs) are the latest generation of platelet concentrates. The objective of developing CGF is to increase therapeutic efficacy. However, few studies have supported the superiority of CGF in composition and efficacy. The reconstruction and regeneration process is complicated and long term, whereas bioactivity of CGF is not durable. The purpose of this review is threefold. The first is to recommend more comparative studies between CGF and other platelet concentrates. The second is to constitute a continuous drug delivery system by combining CGF with other biomaterials. Finally, the novel use of CGF in facial regenerative and reconstructive medicine will be highlighted. METHODS A comprehensive review of literature regarding the use of CGF in facial regenerative and reconstructive medicine was performed. Based on the inclusion and exclusion criteria, a total of 135 articles were included. RESULTS The use of CGF involving facial rejuvenation, cartilage grafting, facial bone defects, facial peripheral nerve injury and wounding is reviewed. The reconstructive and regenerative principles lie in firm fibrin scaffolds and continuous in situ delivery of multiple growth factors. CONCLUSIONS CGF represents an advance in personalized medicine concept. However, the current scientific evidences about the use of CGF are limited. More basic and clinical studies should be conducted to understand the characteristics and clinical application of CGF. LEVEL OF EVIDENCE V This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266.
Collapse
|
13
|
Galland S, Stamenkovic I. Mesenchymal stromal cells in cancer: a review of their immunomodulatory functions and dual effects on tumor progression. J Pathol 2019; 250:555-572. [PMID: 31608444 PMCID: PMC7217065 DOI: 10.1002/path.5357] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/03/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem or stromal cells (MSCs) are pluripotent cells implicated in a broad range of physiological events, including organogenesis and maintenance of tissue homeostasis as well as tissue regeneration and repair. Because their current definition is somewhat loose – based primarily on their ability to differentiate into a variety of mesenchymal tissues, adhere to plastic, and express, or lack, a handful of cell surface markers – MSCs likely encompass several subpopulations, which may have diverse properties. Their diversity may explain, at least in part, the pleiotropic functions that they display in different physiological and pathological settings. In the context of tissue injury, MSCs can respectively promote and attenuate inflammation during the early and late phases of tissue repair. They may thereby act as sensors of the inflammatory response and secrete mediators that boost or temper the response as required by the stage of the reparatory and regenerative process. MSCs are also implicated in regulating tumor development, in which they are increasingly recognized to play a complex role. Thus, MSCs can both promote and constrain tumor progression by directly affecting tumor cells via secreted mediators and cell–cell interactions and by modulating the innate and adaptive immune response. This review summarizes our current understanding of MSC involvement in tumor development and highlights the mechanistic underpinnings of their implication in tumor growth and progression. © 2020 Authors. Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Sabine Galland
- Laboratory of Experimental Pathology, Institute of Pathology, CHUV, Lausanne, Switzerland
| | - Ivan Stamenkovic
- Laboratory of Experimental Pathology, Institute of Pathology, CHUV, Lausanne, Switzerland
| |
Collapse
|
14
|
Kroschwald LM, Tauer JT, Kroschwald SI, Suttorp M, Wiedenfeld A, Beissert S, Bauer A, Rauner M. Imatinib mesylate and nilotinib decrease synthesis of bone matrix in vitro. Oncol Lett 2019; 18:2102-2108. [PMID: 31423283 DOI: 10.3892/ol.2019.10518] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 05/13/2019] [Indexed: 11/06/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs), such as imatinib (IMA) and nilotinib (NIL), are the cornerstone of chronic myeloid leukemia (CML) treatment via the blockade of the oncogenic BCR-ABL1 fusion protein. However, skeletal side effects are commonly observed in pediatric patients receiving long-term treatment with IMA. Additionally, in vitro studies have shown that IMA and NIL alter vitamin D metabolism, which may further impair bone metabolism. To determine whether TKIs directly affect bone cell function, the present study treated the human osteoblastic cell line SaOS-2 with IMA or NIL and assessed effects on their mineralization capacity as well as mRNA expression of receptor activator of nuclear factor κB ligand (RANKL) and osteoprotegerin (OPG), two cytokines that regulate osteoclastogenesis. Both TKIs significantly inhibited mineralization and downregulated osteoblast marker genes, including alkaline phosphatase, osteocalcin, osterix, as well as genes associated with the pro-osteogenic Wnt signaling pathway; NIL was more potent than IMA. In addition, both TKIs increased the RANKL/OPG ratio, which is known to stimulate osteoclastogenesis. The present results suggested that the TKIs IMA and NIL directly inhibited osteoblast differentiation and directly promoted a pro-osteoclastogenic environment through the RANKL-OPG signaling axis. Thus, we propose that future work is required to determine whether the bone health of CML patients undergoing TKI-treatment should be routinely monitored.
Collapse
Affiliation(s)
- Lysann Michaela Kroschwald
- Department of Dermatology, Medical Faculty, Technical University Dresden, D-01307 Dresden, Germany.,Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty, Technical University Dresden, D-01307 Dresden, Germany
| | - Josephine Tabea Tauer
- Department of Pediatrics, Shriners Hospital for Children, McGill University, Montreal QC H4A 0A9, Canada
| | - Sonja Ingrid Kroschwald
- Simon Alberti Group, Max Planck Institute of Molecular Cell Biology and Genetics, D-01307 Dresden, Germany.,Institute for Biochemistry, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Meinolf Suttorp
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, Medical Faculty, Technical University Dresden, D-01307 Dresden, Germany
| | - Anne Wiedenfeld
- Department of Dermatology, Medical Faculty, Technical University Dresden, D-01307 Dresden, Germany
| | - Stefan Beissert
- Department of Dermatology, Medical Faculty, Technical University Dresden, D-01307 Dresden, Germany
| | - Andrea Bauer
- Department of Dermatology, Medical Faculty, Technical University Dresden, D-01307 Dresden, Germany
| | - Martina Rauner
- Department of Medicine III and Center for Healthy Aging, Medical Faculty, Technical University Dresden, D-01307 Dresden, Germany
| |
Collapse
|
15
|
Gelatinous bone marrow transformation and emergence of clonal Philadelphia-negative cytogenetic abnormalities with excess blasts in a patient with chronic myeloid leukemia treated with dasatinib. Anticancer Drugs 2019; 30:416-421. [DOI: 10.1097/cad.0000000000000763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
GM1 Ganglioside Promotes Osteogenic Differentiation of Human Tendon Stem Cells. Stem Cells Int 2018; 2018:4706943. [PMID: 30210549 PMCID: PMC6126069 DOI: 10.1155/2018/4706943] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 07/26/2018] [Indexed: 12/17/2022] Open
Abstract
Gangliosides, the sialic acid-conjugated glycosphingolipids present in the lipid rafts, have been recognized as important regulators of cell proliferation, migration, and apoptosis. Due to their peculiar localization in the cell membrane, they modulate the activity of several key cell receptors, and increasing evidence supports their involvement also in stem cell differentiation. In this context, herein we report the role played by the ganglioside GM1 in the osteogenic differentiation of human tendon stem cells (hTSCs). In particular, we found an increase of GM1 levels during osteogenesis that is instrumental for driving the process. In fact, supplementation of the ganglioside in the medium significantly increased the osteogenic differentiation capability of hTSCs. Mechanistically, we found that GM1 supplementation caused a reduction in the phosphorylation of the platelet-derived growth factor receptor-β (PDGFR-β), which is a known inhibitor of osteogenic commitment. These results were further corroborated by the observation that GM1 supplementation was able to revert the inhibitory effects on osteogenesis when the process was inhibited with exogenous PDGF.
Collapse
|
17
|
Poggi A, Varesano S, Zocchi MR. How to Hit Mesenchymal Stromal Cells and Make the Tumor Microenvironment Immunostimulant Rather Than Immunosuppressive. Front Immunol 2018; 9:262. [PMID: 29515580 PMCID: PMC5825917 DOI: 10.3389/fimmu.2018.00262] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/30/2018] [Indexed: 12/17/2022] Open
Abstract
Experimental evidence indicates that mesenchymal stromal cells (MSCs) may regulate tumor microenvironment (TME). It is conceivable that the interaction with MSC can influence neoplastic cell functional behavior, remodeling TME and generating a tumor cell niche that supports tissue neovascularization, tumor invasion and metastasization. In addition, MSC can release transforming growth factor-beta that is involved in the epithelial-mesenchymal transition of carcinoma cells; this transition is essential to give rise to aggressive tumor cells and favor cancer progression. Also, MSC can both affect the anti-tumor immune response and limit drug availability surrounding tumor cells, thus creating a sort of barrier. This mechanism, in principle, should limit tumor expansion but, on the contrary, often leads to the impairment of the immune system-mediated recognition of tumor cells. Furthermore, the cross-talk between MSC and anti-tumor lymphocytes of the innate and adaptive arms of the immune system strongly drives TME to become immunosuppressive. Indeed, MSC can trigger the generation of several types of regulatory cells which block immune response and eventually impair the elimination of tumor cells. Based on these considerations, it should be possible to favor the anti-tumor immune response acting on TME. First, we will review the molecular mechanisms involved in MSC-mediated regulation of immune response. Second, we will focus on the experimental data supporting that it is possible to convert TME from immunosuppressive to immunostimulant, specifically targeting MSC.
Collapse
Affiliation(s)
- Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, Policlinico San Martino, Genoa, Italy
| | - Serena Varesano
- Molecular Oncology and Angiogenesis Unit, Policlinico San Martino, Genoa, Italy
| | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
18
|
Borriello A, Caldarelli I, Bencivenga D, Stampone E, Perrotta S, Oliva A, Della Ragione F. Tyrosine kinase inhibitors and mesenchymal stromal cells: effects on self-renewal, commitment and functions. Oncotarget 2018; 8:5540-5565. [PMID: 27750212 PMCID: PMC5354929 DOI: 10.18632/oncotarget.12649] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/04/2016] [Indexed: 12/18/2022] Open
Abstract
The hope of selectively targeting cancer cells by therapy and eradicating definitively malignancies is based on the identification of pathways or metabolisms that clearly distinguish “normal” from “transformed” phenotypes. Some tyrosine kinase activities, specifically unregulated and potently activated in malignant cells, might represent important targets of therapy. Consequently, tyrosine kinase inhibitors (TKIs) might be thought as the “vanguard” of molecularly targeted therapy for human neoplasias. Imatinib and the successive generations of inhibitors of Bcr-Abl1 kinase, represent the major successful examples of TKI use in cancer treatment. Other tyrosine kinases have been selected as targets of therapy, but the efficacy of their inhibition, although evident, is less definite. Two major negative effects exist in this therapeutic strategy and are linked to the specificity of the drugs and to the role of the targeted kinase in non-malignant cells. In this review, we will discuss the data available on the TKIs effects on the metabolism and functions of mesenchymal stromal cells (MSCs). MSCs are widely distributed in human tissues and play key physiological roles; nevertheless, they might be responsible for important pathologies. At present, bone marrow (BM) MSCs have been studied in greater detail, for both embryological origins and functions. The available data are evocative of an unexpected degree of complexity and heterogeneity of BM-MSCs. It is conceivable that this grade of intricacy occurs also in MSCs of other organs. Therefore, in perspective, the negative effects of TKIs on MSCs might represent a critical problem in long-term cancer therapies based on such inhibitors.
Collapse
Affiliation(s)
- Adriana Borriello
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Ilaria Caldarelli
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Debora Bencivenga
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Emanuela Stampone
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Silverio Perrotta
- Department of Woman, Child and of General and Specialized Surgery, Second University of Naples, Naples, Italy
| | - Adriana Oliva
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Fulvio Della Ragione
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| |
Collapse
|
19
|
Haque N, Abdullah BJJ, Kasim NHA. Secretome: Pharmaceuticals for Cell-Free Regenerative Therapy. STEM CELL DRUGS - A NEW GENERATION OF BIOPHARMACEUTICALS 2018. [DOI: 10.1007/978-3-319-99328-7_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
20
|
Haque N, Widera D, Abu Kasim NH. Stem Cells from Human Extracted Deciduous Teeth Expanded in Foetal Bovine and Human Sera Express Different Paracrine Factors After Exposure to Freshly Prepared Human Serum. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1084:175-186. [PMID: 30771186 DOI: 10.1007/5584_2018_299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND The response of stem cells to paracrine factors within the host's body plays an important role in the regeneration process after transplantation. The aim of this study was to determine the viability and paracrine factor profile of stem cells from human extracted deciduous teeth (SHED) pre-cultivated in media supplemented with either foetal bovine serum (FBS) or pooled human serum (pHS) in the presence of individual human sera (iHS). METHODS SHED (n = 3) from passage 4 were expanded in FBS (FBS-SHED) or pHS (pHS-SHED) supplemented media until passage 7. During expansion, the proliferation of SHED was determined. Cells at passage 7 were further expanded in human serum from four individual donors (iHS) for 120 h followed by assessment of cell viability and profiling of the secreted paracrine factors. RESULTS Proliferation of SHED was significantly higher (p < 0.05) in pHS supplemented media compared to FBS supplemented media. pHS-SHED also maintained their higher proliferation rate compared to FBS-SHED in the presence of iHS. In iHS supplemented media, FBS-SHED expressed significantly higher levels of SDF-1A (p < 0.05) after 24 h compared to pHS-SHED. Similar results were found for HGF (p < 0.01), LIF (p < 0.05), PDGF-BB (p < 0.05), SDF-1A (p < 0.01), and IL-10 (p < 0.05) when cell culture supernatants from FBS-SHED were profiled 120 h post-incubation. CONCLUSION SHED expanded in pHS instead of FBS have higher proliferative capacity and show an altered secretion profile. Further studies are needed to determine whether these differences could result in better engraftment and regeneration following transplantation.
Collapse
Affiliation(s)
- Nazmul Haque
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Jenjarum, Selangor, Malaysia.,Regenerative Dentistry Research Group, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, Reading, UK
| | - Noor Hayaty Abu Kasim
- Regenerative Dentistry Research Group, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia. .,Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
21
|
Kroschwald L, Suttorp M, Tauer JT, Zimmermann N, Günther C, Bauer A. Off‑target effect of imatinib and nilotinib on human vitamin D3 metabolism. Mol Med Rep 2017; 17:1382-1388. [PMID: 29115640 DOI: 10.3892/mmr.2017.7952] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 10/09/2017] [Indexed: 11/06/2022] Open
Abstract
Prolonged treatment with tyrosine kinase inhibitors (TKI) including imatinib (IMA) or nilotinib (NIL), induces severe disturbances of bone metabolism in patients with chronic myeloid leukaemia. As vitamin D3 (VD3) is involved in the complex cycle of bone remodelling, the present study investigated in vitro, the influence of IMA and NIL on VD3 metabolism i) in HaCaT cells and ii) in cultured outer root sheath keratinocytes (ORS‑KC) from hair follicles of IMA treated children. Cells were incubated in the presence of IMA or NIL. Concomitantly, specific inhibitors were applied to analyze the inhibition of the VD3 processing cytochrome P450 isoenzyme family by TKIs. In vitro, IMA and NIL significantly impaired the production of calcitriol in HaCaT and cultured ORS‑KC cells from hair follicles of IMA treated children. For NIL, this inhibitory effect demonstrated a 4‑fold increase. In HaCaT and ORS‑KC, application of specific CYP450 inhibitors revealed that CYP27B1 was impaired by IMA and NIL leading to an intracellular accumulation of calcidiol. However, during TKI treatment, KC of IMA treated children revealed no differences in calcidiol and calcitriol levels. In conclusion, IMA and NIL interfere with the vitamin D3 cascade due to their metabolism by CYP27B1.
Collapse
Affiliation(s)
- Lysann Kroschwald
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, D‑01307 Dresden, Germany
| | - Meinolf Suttorp
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University Hospital Carl Gustav Carus, TU Dresden, D‑01307 Dresden, Germany
| | - Josephine Tabea Tauer
- Department of Pediatrics, Shriners Hospital for Children, Montréal, QC H4A 0A9, Canada
| | - Nick Zimmermann
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, D‑01307 Dresden, Germany
| | - Claudia Günther
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, D‑01307 Dresden, Germany
| | - Andrea Bauer
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, D‑01307 Dresden, Germany
| |
Collapse
|
22
|
Liang T, Zhu L, Gao W, Gong M, Ren J, Yao H, Wang K, Shi D. Coculture of endothelial progenitor cells and mesenchymal stem cells enhanced their proliferation and angiogenesis through PDGF and Notch signaling. FEBS Open Bio 2017; 7:1722-1736. [PMID: 29123981 PMCID: PMC5666384 DOI: 10.1002/2211-5463.12317] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 08/31/2017] [Accepted: 08/31/2017] [Indexed: 01/06/2023] Open
Abstract
The beneficial effects of combined use of mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs) on tissue repair and regeneration after injury have been demonstrated, but the underlying mechanism remains incompletely understood. This study aimed to investigate the effects of direct contact coculture of human bone marrow‐derived EPCs (hEPCs)/human bone marrow‐derived MSCs (hMSCs) on their proliferation and angiogenic capacities and the underlying mechanism. hEPCs and hMSCs were cocultured in a 2D mixed monolayer or a 3D transwell membrane cell‐to‐cell coculture system. Cell proliferation was determined by Cell Counting Kit‐8. Angiogenic capacity was evaluated by in vitro angiogenesis assay. Platelet‐derived growth factor‐BB (PDGF‐BB), PDGF receptor neutralizing antibody (AB‐PDGFR), and DAPT (a γ‐secretase inhibitor) were used to investigate PDGF and Notch signaling. Cell proliferation was significantly enhanced by hEPCs/hMSCs 3D‐coculture and PDGF‐BB treatment, but inhibited by AB‐PDGFR. Expression of cyclin D1, PDGFR, Notch1, and Hes1 was markedly enhanced by PDGF‐BB but inhibited by DAPT. In vitro angiogenesis assay showed that hEPCs/hMSCs coculture and PDGF‐BB significantly enhanced angiogenic capacity, whereas AB‐PDGFR significantly reduced the angiogenic capacity. PDGF‐BB increased the expression of kinase insert domain receptor (KDR, an endothelial marker) and activated Notch1 signaling in cocultured cells, while DAPT attenuated the promoting effect of PDGF‐BB on KDR expression of hEPCs/hMSCs coculture. hEPCs/hMSCs coculture enhanced their proliferation and angiogenic capacities. PDGF and Notch signaling pathways participated in the promoting effects of hEPCs/hMSCs coculture, and there was crosstalk between these two signaling pathways. Our findings should aid understanding of the mechanism of beneficial effects of hEPCs/hMSCs coculture.
Collapse
Affiliation(s)
- Tangzhao Liang
- Department of Orthopaedic Surgery the Third Affiliated Hospital of Sun Yat-sen University Guangzhou China
| | - Lei Zhu
- Department of Plastic and Reconstructive Surgery the Third Affiliated Hospital of Sun Yat-sen University Guangzhou China
| | - Wenling Gao
- Department of Periodontology Faculty of Dentistry Prince Philip Dental Hospital The University of Hong Kong China
| | - Ming Gong
- Department of Orthopedic Surgery Shenzhen Hospital of Southern Medical University China
| | - Jianhua Ren
- Department of Orthopaedic Surgery the Third Affiliated Hospital of Sun Yat-sen University Guangzhou China
| | - Hui Yao
- Department of Orthopaedic Surgery the Third Affiliated Hospital of Sun Yat-sen University Guangzhou China
| | - Kun Wang
- Department of Orthopaedic Surgery the Third Affiliated Hospital of Sun Yat-sen University Guangzhou China
| | - Dehai Shi
- Department of Orthopaedic Surgery the Third Affiliated Hospital of Sun Yat-sen University Guangzhou China
| |
Collapse
|
23
|
Camorani S, Hill BS, Fontanella R, Greco A, Gramanzini M, Auletta L, Gargiulo S, Albanese S, Lucarelli E, Cerchia L, Zannetti A. Inhibition of Bone Marrow-Derived Mesenchymal Stem Cells Homing Towards Triple-Negative Breast Cancer Microenvironment Using an Anti-PDGFRβ Aptamer. Am J Cancer Res 2017; 7:3595-3607. [PMID: 28912898 PMCID: PMC5596446 DOI: 10.7150/thno.18974] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 06/20/2017] [Indexed: 12/11/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs) are shown to participate in tumor progression by establishing a favorable tumor microenvironment (TME) that promote metastasis through a cytokine networks. However, the mechanism of homing and recruitment of BM-MSCs into tumors and their potential role in malignant tissue progression is poorly understood and controversial. Here we show that BM-MSCs increase aggressiveness of triple-negative breast cancer (TNBC) cell lines evaluated as capability to migrate, invade and acquire stemness markers. Importantly, we demonstrate that the treatment of BM-MSCs with a nuclease-resistant RNA aptamer against platelet-derived growth factor receptor β (PDGFRβ) causes the inhibition of receptor-dependent signaling pathways thus drastically hampering BM-MSC recruitment towards TNBC cell lines and BM-MSCs trans-differentiation into carcinoma-associated fibroblast (CAF)-like cells. Moreover, in vivo molecular imaging analysis demonstrated the aptamer ability to prevent BM-MSCs homing to TNBC xenografts. Collectively, our results indicate the anti-PDGFRβ aptamer as a novel therapeutic tool to interfere with BM-MSCs attraction to TNBC providing the rationale to further explore the aptamer in more complex pre-clinical settings.
Collapse
|
24
|
Aggoune D, Sorel N, Bonnet ML, Goujon JM, Tarte K, Hérault O, Domenech J, Réa D, Legros L, Johnson-Ansa H, Rousselot P, Cayssials E, Guerci-Bresler A, Bennaceur-Griscelli A, Chomel JC, Turhan AG. Bone marrow mesenchymal stromal cell (MSC) gene profiling in chronic myeloid leukemia (CML) patients at diagnosis and in deep molecular response induced by tyrosine kinase inhibitors (TKIs). Leuk Res 2017; 60:94-102. [PMID: 28772207 DOI: 10.1016/j.leukres.2017.07.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/01/2017] [Accepted: 07/25/2017] [Indexed: 01/05/2023]
Abstract
Although it has been well-demonstrated that bone marrow mesenchymal stromal cells (MSCs) from CML patients do not belong to the Ph1-positive clone, there is growing evidence that they could play a role in the leukemogenesis process or the protection of leukemic stem cells from the effects of tyrosine kinase inhibitors (TKIs). The aim of the present study was to identify genes differentially expressed in MSCs isolated from CML patients at diagnosis (CML-MSCs) as compared to MSCs from healthy controls. Using a custom gene-profiling assay, we identified six genes over-expressed in CML-MSCs (BMP1, FOXO3, MET, MITF, NANOG, PDPN), with the two highest levels being documented for PDPN (PODOPLANIN) and NANOG. To determine whether this aberrant signature persisted in patients in deep molecular response induced by TKIs, we analyzed MSCs derived from such patients (MR-MSCs). This analysis showed that, despite the deep molecular responses, BMP1, MET, MITF, NANOG, and PDPN mRNA were upregulated in MR-MSCs. Moreover, BMP1, MITF, and NANOG mRNA expressions in MR-MSCs were found to be intermediate between control MSCs and CML-MSCs. These results suggest that CML-MSCs exhibit an abnormal gene expression pattern which might have been established during the leukemogenic process and persist in patients in deep molecular response.
Collapse
Affiliation(s)
| | - Nathalie Sorel
- INSERM, U935, F-86000 Poitiers, France; CHU de Poitiers, Service de Cancérologie Biologique, F-86021 Poitiers, France
| | | | - Jean-Michel Goujon
- CHU de Poitiers, Service d'Anatomie et cytologie pathologiques, F-86021 Poitiers, France; INSERM, U1082, F-86021 Poitiers, France
| | | | - Olivier Hérault
- CHU de Tours, Service d'Hématologie Biologique, F-37032 Tours, France; CNRS UMR 7292, équipe LNOx, Université François Rabelais, F-37032 Tours, France
| | - Jorge Domenech
- CHU de Tours, Service d'Hématologie Biologique, F-37032 Tours, France; CNRS UMR 7292, équipe LNOx, Université François Rabelais, F-37032 Tours, France
| | - Delphine Réa
- Hôpital Saint Louis, Service d'Hématologie Adulte, F-75000 Paris, France; INSERM, UMRS-1160, IUH-Université Paris Diderot-Paris 7, F-75000 Paris, France
| | - Laurence Legros
- Hôpital l'Archet, Service d'Hématologie Clinique, F-06202 Nice, France
| | | | - Philippe Rousselot
- Centre Hospitalier de Versailles, Service d'Hématologie et Oncologie, F-78150 Le Chesnay, France; EA4340, Université Versailles-Saint Quentin en Yvelines, Université Paris-Saclay, France
| | - Emilie Cayssials
- INSERM, CIC-P 0802, F-86000 Poitiers, France; CHU de Poitiers, Service d'Oncologie Hématologique et Thérapie Cellulaire, F-86000, Poitiers, France
| | | | - Annelise Bennaceur-Griscelli
- Hôpital Paul Brousse, Service d'Hématologie Biologique, F-94800 Villejuif, France; NSERM U935, F-94807 Villejuif, France; Université Paris Sud, F-94270 Le Kremlin-Bicêtre, France
| | - Jean-Claude Chomel
- INSERM, U935, F-86000 Poitiers, France; CHU de Poitiers, Service de Cancérologie Biologique, F-86021 Poitiers, France
| | - Ali G Turhan
- INSERM, U935, F-86000 Poitiers, France; Hôpital Paul Brousse, Service d'Hématologie Biologique, F-94800 Villejuif, France; NSERM U935, F-94807 Villejuif, France; Université Paris Sud, F-94270 Le Kremlin-Bicêtre, France; Hôpital Bicêtre, Service d'Hématologie Biologique, F-94270 Le Kremlin Bicêtre, France.
| |
Collapse
|
25
|
Gelatinous Marrow Transformation Associated with Imatinib: Case Report and Literature Review. Case Rep Hematol 2017; 2017:1950724. [PMID: 28133556 PMCID: PMC5241459 DOI: 10.1155/2017/1950724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 12/14/2016] [Indexed: 11/23/2022] Open
Abstract
Gelatinous marrow transformation (GMT) is a rare condition observed in severe illness or malnutrition, in which the bone marrow contains amorphous “gelatinous” extracellular material, and histopathology demonstrates varied degrees of fat cell atrophy and loss of hematopoietic elements. An association of GMT with imatinib use in chronic myeloid leukemia (CML) has been reported recently. The objective of this study is to describe a case of GMT associated with imatinib use and review the existing similar cases in the literature to identify epidemiological patterns and potential imatinib-induced mechanisms leading to gelatinous conversion.
Collapse
|
26
|
Pooled Human Serum Increases Regenerative Potential of In Vitro Expanded Stem Cells from Human Extracted Deciduous Teeth. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1083:29-44. [PMID: 28730381 DOI: 10.1007/5584_2017_74] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In regenerative therapy, in vitro expansion of stem cells is critical to obtain a significantly higher number of cells for successful engraftment after transplantation. However, stem cells lose its regenerative potential and enter senescence during in vitro expansion. In this study, the influence of foetal bovine serum (FBS) and pooled human serum (pHS) on the proliferation, morphology and migration of stem cells from human extracted deciduous teeth (SHED) was compared. SHED (n = 3) was expanded in KnockOut DMEM supplemented with either pHS (pHS-SM) or FBS (FBS-SM). pHS was prepared using peripheral blood serum of six healthy male adults, aged between 21 and 35 years old. The number of live SHED was significantly higher, from passage 5 to 7, when cultured in pHS-SM compared to those cultured in FBS-SM (p < 0.05). Number of cells having flattened morphology, characteristics of partially differentiated and senescent cells, was significantly lower (p < 0.05) in pHS-SM (3%) compared to those in FBS-SM (7%). Furthermore, migration of SHED in pHS-SM was found to be more directional. The presence of selected ten paracrine factors known for their proliferation and migration potential was detected in all six individual human sera, used to produce pHS, none of which were detected in FBS. Ingenuity Pathway Analysis showed the possible involvement of the 'ephrin receptor signalling pathway' to regulate the proliferation and migration of SHED in pHS-SM. In conclusion, pHS-SM showed significantly higher proliferation rate and could maintain significantly lower number of senescent cells and support directional migration of cells.
Collapse
|
27
|
O’Sullivan S, Tay ML, Lin JM, Bava U, Callon K, Cornish J, Naot D, Grey A. Tyrosine Kinase Inhibitors Regulate OPG through Inhibition of PDGFRβ. PLoS One 2016; 11:e0164727. [PMID: 27737004 PMCID: PMC5063333 DOI: 10.1371/journal.pone.0164727] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 09/29/2016] [Indexed: 01/01/2023] Open
Abstract
Nilotinib and imatinib are tyrosine kinase inhibitors (TKIs) used in the treatment of chronic myeloid leukemia (CML) and gastrointestinal stromal tumors (GIST). In vitro, imatinib and nilotinib inhibit osteoclastogenesis, and in patients they reduce levels of bone resorption. One of the mechanisms that might underlie these effects is an increase in the production of osteoprotegerin (OPG). In the current work we report that platelet-derived growth factor receptor beta (PDGFRβ) signaling regulates OPG production in vitro. In addition, we have shown that TKIs have effects on RANKL signaling through inhibition of the PDGFRβ and other target receptors. These findings have implications for our understanding of the mechanisms by which TKIs affect osteoclastogenesis, and the role of PDGFRβ signaling in regulating osteoclastogenesis. Further studies are indicated to confirm the clinical effects of PDGFRβ-inhibitors and to elaborate the intracellular pathways that underpin these effects.
Collapse
Affiliation(s)
- Susannah O’Sullivan
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
- * E-mail:
| | - Mei Lin Tay
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Jian-Ming Lin
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Usha Bava
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Karen Callon
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Jillian Cornish
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Dorit Naot
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Andrew Grey
- Department of Medicine, University of Auckland, Auckland, New Zealand
| |
Collapse
|
28
|
Samis J, Lee P, Zimmerman D, Arceci RJ, Suttorp M, Hijiya N. Recognizing Endocrinopathies Associated With Tyrosine Kinase Inhibitor Therapy in Children With Chronic Myelogenous Leukemia. Pediatr Blood Cancer 2016; 63:1332-8. [PMID: 27100618 DOI: 10.1002/pbc.26028] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 03/18/2016] [Indexed: 01/19/2023]
Abstract
Side effects of tyrosine kinase inhibitor (TKI) treatment vary in children and adults with chronic myelogenous leukemia (CML). As children have a much longer life expectancy than adults, TKI therapy may continue for decades and with long-term consequences that differ from adults. Children may develop endocrinopathies related to "off-target" effects of TKIs, such as delayed growth, changes in bone metabolism, thyroid abnormalities, and effects on puberty and fertility. These endocrinopathies present additional challenges for pediatric patients with CML. This review critically evaluates the literature on long-term endocrine side effects of TKIs in the pediatric CML population and provides suggested recommendations.
Collapse
Affiliation(s)
- Jill Samis
- Division of Pediatric Endocrinology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Paul Lee
- Division of Pediatric Hematology Oncology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Donald Zimmerman
- Division of Pediatric Endocrinology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Robert J Arceci
- Department of Child Health, The Ron Matricaria Institute of Molecular Medicine at Phoenix Children's Hospital, University of Arizona College of Medicine, Phoenix, Arizona
| | - Meinolf Suttorp
- Pediatric Hematology, Oncology & Stem Cell Transplantation, Department of Pediatrics, Children's Hospital, Technical University of Dresden, Dresden, Germany
| | - Nobuko Hijiya
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Division of Pediatric Hematology Oncology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| |
Collapse
|
29
|
Kirschner G, Balla B, Horváth P, Kövesdi A, Lakatos G, Takács I, Nagy Z, Tóbiás B, Árvai K, Kósa JP, Lakatos P. Effects of imatinib and nilotinib on the whole transcriptome of cultured murine osteoblasts. Mol Med Rep 2016; 14:2025-37. [PMID: 27430367 PMCID: PMC4991674 DOI: 10.3892/mmr.2016.5459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/17/2016] [Indexed: 12/11/2022] Open
Abstract
Numerous clinical observations have confirmed that breakpoint cluster region-abelson fusion oncoprotein tyrosine kinase inhibitors used in leukemia treatment alter bone physiology in a complex manner. The aim of the present study was to analyze the whole transcriptome of cultured murine osteoblasts and determine the changes following treatment with imatinib and nilotinib using Sequencing by Oligonucleotide Ligation and Detection next generation RNA sequencing. This study also aimed to identify candidate signaling pathways and network regulators by multivariate Ingenuity Pathway Analysis. Based on the right-tailed Fisher's exact test, significantly altered pathways including upstream regulators were defined for each drug. The correlation between these pathways and bone metabolism was also examined. The preliminary results suggest the two drugs have different mechanisms of action on osteoblasts, and imatinib was shown to have a greater effect on gene expression. Data also indicated the potential role of a number of genes and signaling cascades that may contribute to identifying novel targets for the treatment of metabolic bone diseases.
Collapse
Affiliation(s)
- Gyöngyi Kirschner
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Bernadett Balla
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Péter Horváth
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Andrea Kövesdi
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Gergely Lakatos
- Second Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - István Takács
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Zsolt Nagy
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Bálint Tóbiás
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Kristóf Árvai
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - János Pál Kósa
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Péter Lakatos
- First Department of Internal Medicine, Semmelweis University, 1083 Budapest, Hungary
| |
Collapse
|
30
|
Escobar CH, Chaparro O. Xeno-Free Extraction, Culture, and Cryopreservation of Human Adipose-Derived Mesenchymal Stem Cells. Stem Cells Transl Med 2016; 5:358-65. [PMID: 26838269 DOI: 10.5966/sctm.2015-0094] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 12/04/2015] [Indexed: 01/27/2023] Open
Abstract
Molecules of animal or bacterial origin, which pose a risk for zoonoses or immune rejection, are commonly used for extraction, culture, and cryopreservation of mesenchymal stem cells. There is no sequential and orderly protocol for producing human adipose-derived stem cells (hASCs) under xeno-free conditions. After standardizing a human platelet lysate (hPL) production protocol, four human adipose tissue samples were processed through explants with fetal bovine serum (FBS)-supplemented or hPL-supplemented media for extracting the adipose-derived stem cells. The cells were cultivated in cell culture medium + hPL (5%) or FBS (10%). The cellular replication rate, immunophenotype, and differentiation potential were evaluated at fourth passage. Cellular viability was evaluated before and after cryopreservation of the cells, with an hPL-based solution compared with an FBS-based solution. The explants cultured in hPL-supplemented media showed earlier and faster hASC proliferation than did those supplemented with FBS. Likewise, cells grown in hPL-supplemented media showed a greater proliferation rate, without losing the immunophenotype. Osteogenic differentiation of xeno-free hASC was higher than the hASC produced in standard conditions. However, adipogenic differentiation was reduced in xeno-free hASC. Finally, the cells cryopreserved in an hPL-based solution showed a higher cellular viability than the cells cryopreserved in an FBS-based. In conclusion, we have developed a complete xeno-free protocol for extracting, culturing, and cryopreserving hASCs that can be safely implemented in clinical studies.
Collapse
Affiliation(s)
- Carlos Hugo Escobar
- Basic Science Department, Medicine School, Fundación Universitaria de Ciencias de la Salud, Bogotá, Colombia Physiology Department, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Orlando Chaparro
- Physiology Department, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
31
|
Wysokinski D, Pawlowska E, Blasiak J. RUNX2: A Master Bone Growth Regulator That May Be Involved in the DNA Damage Response. DNA Cell Biol 2015; 34:305-15. [DOI: 10.1089/dna.2014.2688] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
| | | | - Janusz Blasiak
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
| |
Collapse
|
32
|
MEHLIG LYSANNMICHAELA, GARVE CLAUDIA, TAUER JOSEPHINETABEA, SUTTORP MEINOLF, BAUER ANDREA. Inhibitory effects of imatinib on vitamin D3 synthesis in human keratinocytes. Mol Med Rep 2014; 11:3143-7. [DOI: 10.3892/mmr.2014.3074] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 11/07/2014] [Indexed: 11/06/2022] Open
|
33
|
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]
|
34
|
Yao W, Guan M, Jia J, Dai W, Lay YAE, Amugongo S, Liu R, Olivos D, Saunders M, Lam KS, Nolta J, Olvera D, Ritchie RO, Lane NE. Reversing bone loss by directing mesenchymal stem cells to bone. Stem Cells 2014; 31:2003-14. [PMID: 23818248 DOI: 10.1002/stem.1461] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 02/01/2013] [Accepted: 03/26/2013] [Indexed: 01/16/2023]
Abstract
Bone regeneration by systemic transplantation of mesenchymal stem cells (MSCs) is problematic due to the inability to control the MSCs' commitment, growth, and differentiation into functional osteoblasts on the bone surface. Our research group has developed a method to direct the MSCs to the bone surface by conjugating a synthetic peptidomimetic ligand (LLP2A) that has high affinity for activated α4β1 integrin on the MSC surface, with a bisphosphonates (alendronate) that has high affinity for bone (LLP2A-Ale), to direct the transplanted MSCs to bone. Our in vitro experiments demonstrated that mobilization of LLP2A-Ale to hydroxyapatite accelerated MSC migration that was associated with an increase in the phosphorylation of Akt kinase and osteoblastogenesis. LLP2A-Ale increased the homing of the transplanted MSCs to bone as well as the osteoblast surface, significantly increased the rate of bone formation and restored both trabecular and cortical bone loss induced by estrogen deficiency or advanced age in mice. These results support LLP2A-Ale as a novel therapeutic option to direct the transplanted MSCs to bone for the treatment of established bone loss related to hormone deficiency and aging.
Collapse
Affiliation(s)
- Wei Yao
- Department of Internal Medicine, University of California at Davis Medical Center, Sacramento, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Nataraj V, Kandasamy D, Bakhshi S. Imatinib-induced avascular necrosis of femur in childhood chronic myeloid leukemia. Pediatr Hematol Oncol 2014; 31:268-70. [PMID: 24383853 DOI: 10.3109/08880018.2013.862588] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Vijayamurugan Nataraj
- 1Departments of Medical Oncology, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | | | |
Collapse
|
36
|
Imatinib mesylate exerts anti-proliferative effects on osteosarcoma cells and inhibits the tumour growth in immunocompetent murine models. PLoS One 2014; 9:e90795. [PMID: 24599309 PMCID: PMC3944320 DOI: 10.1371/journal.pone.0090795] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 02/04/2014] [Indexed: 01/11/2023] Open
Abstract
Osteosarcoma is the most common primary malignant bone tumour characterized by osteoid production and/or osteolytic lesions of bone. A lack of response to chemotherapeutic treatments shows the importance of exploring new therapeutic methods. Imatinib mesylate (Gleevec, Novartis Pharma), a tyrosine kinase inhibitor, was originally developed for the treatment of chronic myeloid leukemia. Several studies revealed that imatinib mesylate inhibits osteoclast differentiation through the M-CSFR pathway and activates osteoblast differentiation through PDGFR pathway, two key cells involved in the vicious cycle controlling the tumour development. The present study investigated the in vitro effects of imatinib mesylate on the proliferation, apoptosis, cell cycle, and migration ability of five osteosarcoma cell lines (human: MG-63, HOS; rat: OSRGA; mice: MOS-J, POS-1). Imatinib mesylate was also assessed as a curative and preventive treatment in two syngenic osteosarcoma models: MOS-J (mixed osteoblastic/osteolytic osteosarcoma) and POS-1 (undifferentiated osteosarcoma). Imatinib mesylate exhibited a dose-dependent anti-proliferative effect in all cell lines studied. The drug induced a G0/G1 cell cycle arrest in most cell lines, except for POS-1 and HOS cells that were blocked in the S phase. In addition, imatinib mesylate induced cell death and strongly inhibited osteosarcoma cell migration. In the MOS-J osteosarcoma model, oral administration of imatinib mesylate significantly inhibited the tumour development in both preventive and curative approaches. A phospho-receptor tyrosine kinase array kit revealed that PDGFRα, among 7 other receptors (PDFGFRβ, Axl, RYK, EGFR, EphA2 and 10, IGF1R), appears as one of the main molecular targets for imatinib mesylate. In the light of the present study and the literature, it would be particularly interesting to revisit therapeutic evaluation of imatinib mesylate in osteosarcoma according to the tyrosine-kinase receptor status of patients.
Collapse
|
37
|
Estrada-González PK, Gómez-Ceja L, Montesinos JJ, Mayani H, Chávez-González A, Meillón L, Delgado N, Sánchez-Nava E, Flores-Figueroa E. Decreased frequency, but normal functional integrity of mesenchymal stromal cells derived from untreated and Imatinib-treated chronic myeloid leukemia patients. Leuk Res 2014; 38:594-600. [PMID: 24661629 DOI: 10.1016/j.leukres.2014.02.011] [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: 11/21/2013] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 02/07/2023]
Abstract
In vitro, Imatinib inhibits the proliferation and stimulates the osteogenic and adipogenic differentiation of mesenchymal stromal cells (MSC). However, it is unknown whether Imatinib affects the biology of MSC in vivo. We asked whether MSC from long-term Imatinib-treated CML patients were affected by the in vivo treatment. MSC from untreated and Imatinib-treated patients displayed normal functional properties (i.e. proliferation, immunophenotype, differentiation and hematopoietic supportive capacity) - but a decreased frequency. In vitro, Imatinib lost its effect when discontinued; which suggest that it has a reversible effect on MSC. Therefore it might lose its effect on MSC after discontinuation in vivo.
Collapse
Affiliation(s)
- P K Estrada-González
- Niche and Microenvironment Laboratory, Oncology Research Unit, Oncology Hospital, Mexico; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Coyoacán, México City, Mexico
| | - L Gómez-Ceja
- Niche and Microenvironment Laboratory, Oncology Research Unit, Oncology Hospital, Mexico; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Coyoacán, México City, Mexico
| | - J J Montesinos
- Mesenchymal Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, Mexico
| | - H Mayani
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, Mexico
| | - A Chávez-González
- Leukemic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, Mexico
| | - L Meillón
- Hematology Service, Bernardo Sepulveda Hospital, National Medical Center, IMSS, México City, Mexico
| | - N Delgado
- Hematology Service, Bernardo Sepulveda Hospital, National Medical Center, IMSS, México City, Mexico
| | - E Sánchez-Nava
- Hematology Service, Bernardo Sepulveda Hospital, National Medical Center, IMSS, México City, Mexico
| | - E Flores-Figueroa
- Niche and Microenvironment Laboratory, Oncology Research Unit, Oncology Hospital, Mexico.
| |
Collapse
|
38
|
Sharma SK, Choudhary D, Handoo A, Kharya G, Gupta N, Pavecha P, Chadha R. Gelatinous transformation of bone marrow following the use of dasatinib in a patient with philadelphia chromosome-positive acute lymphoblastic leukemia. Leuk Res Rep 2013; 2:7-8. [PMID: 24371767 DOI: 10.1016/j.lrr.2012.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Indexed: 11/15/2022] Open
Affiliation(s)
- Sanjeev Kumar Sharma
- Department of Hemato-oncology and Bone Marrow Transplant, BL Kapur Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Dharma Choudhary
- Department of Hemato-oncology and Bone Marrow Transplant, BL Kapur Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Anil Handoo
- Department of Hemato-oncology and Bone Marrow Transplant, BL Kapur Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Gaurav Kharya
- Department of Hemato-oncology and Bone Marrow Transplant, BL Kapur Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Nitin Gupta
- Department of Hemato-oncology and Bone Marrow Transplant, BL Kapur Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Punita Pavecha
- Department of Hemato-oncology and Bone Marrow Transplant, BL Kapur Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Ritu Chadha
- Department of Hemato-oncology and Bone Marrow Transplant, BL Kapur Superspeciality Hospital, Pusa Road, New Delhi, India
| |
Collapse
|
39
|
Hu B, El Haj AJ, Dobson J. Receptor-targeted, magneto-mechanical stimulation of osteogenic differentiation of human bone marrow-derived mesenchymal stem cells. Int J Mol Sci 2013; 14:19276-93. [PMID: 24065106 PMCID: PMC3794833 DOI: 10.3390/ijms140919276] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/02/2013] [Accepted: 09/05/2013] [Indexed: 11/16/2022] Open
Abstract
Mechanical cues are employed to promote stem cell differentiation and functional tissue formation in tissue engineering and regenerative medicine. We have developed a Magnetic Force Bioreactor (MFB) that delivers highly targeted local forces to cells at a pico-newton level, utilizing magnetic micro- and nano-particles to target cell surface receptors. In this study, we investigated the effects of magnetically targeting and actuating specific two mechanical-sensitive cell membrane receptors-platelet-derived growth factor receptor α (PDGFRα) and integrin ανβ3. It was found that a higher mineral-to-matrix ratio was obtained after three weeks of magneto-mechanical stimulation coupled with osteogenic medium culture by initially targeting PDGFRα compared with targeting integrin ανβ3 and non-treated controls. Moreover, different initiation sites caused a differentiated response profile when using a 2-day-lagged magneto-mechanical stimulation over culture periods of 7 and 12 days). However, both resulted in statistically higher osteogenic marker genes expression compared with immediate magneto-mechanical stimulation. These results provide insights into important parameters for designing appropriate protocols for ex vivo induced bone formation via magneto-mechanical actuation.
Collapse
Affiliation(s)
- Bin Hu
- Institute of Science and Technology in Medicine, Guy Hilton Research Center, Keele University, Thornburrow Drive, Hartshill, Stoke on Trent, ST4 7QB, UK; E-Mails: (B.H.); (A.J.E.H.)
| | - Alicia J El Haj
- Institute of Science and Technology in Medicine, Guy Hilton Research Center, Keele University, Thornburrow Drive, Hartshill, Stoke on Trent, ST4 7QB, UK; E-Mails: (B.H.); (A.J.E.H.)
| | - Jon Dobson
- J. Crayton Puritt Family Department of Biomedical Engineering, Department of Materials Science and Engineering, Institute of Cell Engineering and Regenerative Medicine (ICERM), University of Florida, Gainesville, FL 32611, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-352-273-9222; Fax: +1-352-273-9221
| |
Collapse
|
40
|
Chen J, Crawford R, Chen C, Xiao Y. The key regulatory roles of the PI3K/Akt signaling pathway in the functionalities of mesenchymal stem cells and applications in tissue regeneration. TISSUE ENGINEERING PART B-REVIEWS 2013; 19:516-28. [PMID: 23651329 DOI: 10.1089/ten.teb.2012.0672] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into various cell types and have been widely used in tissue engineering application. In tissue engineering, a scaffold, MSCs and growth factors are used as essential components and their interactions have been regarded to be important for regeneration of tissues. A critical problem for MSCs in tissue engineering is their low survival ability and functionality. Most MSCs are going to be apoptotic after transplantation. Therefore, increasing MSC survival ability and functionalities is the key for potential applications of MSCs. Several approaches have been studied to increase MSC tissue forming capacity including application of growth factors, overexpression of stem cell regulatory genes, and improvement of biomaterials for scaffolds. The effects of these approaches on MSCs have been associated with activation of the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. The pathway plays central regulatory roles in MSC survival, proliferation, migration, angiogenesis, cytokine production, and differentiation. In this review, we summarize and discuss the literatures related to the roles of the PI3K/Akt pathway in the functionalities of MSCs and the involvement of the pathway in biomaterials-increased MSC functionalities. Biomaterials have been modified in their properties and surface structure and loaded with growth factors to increase MSC functionalities. Several studies demonstrated that the biomaterials-increased MSC functionalities are mediated by the activation of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Jiezhong Chen
- 1 Institute of Health and Biomedical Innovation, Queensland University of Technology , Brisbane, Australia
| | | | | | | |
Collapse
|
41
|
Hoffman MD, Benoit DSW. Agonism of Wnt-β-catenin signalling promotes mesenchymal stem cell (MSC) expansion. J Tissue Eng Regen Med 2013; 9:E13-26. [PMID: 23554411 DOI: 10.1002/term.1736] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 01/09/2013] [Accepted: 01/30/2013] [Indexed: 12/17/2022]
Abstract
Promoting mesenchymal stem cell (MSC) proliferation has numerous applications in stem cell therapies, particularly in the area of regenerative medicine. In order for cell-based regenerative approaches to be realized, MSC proliferation must be achieved in a controlled manner without compromising stem cell differentiation capacities. Here we demonstrate that 6-bromoindirubin-3'-oxime (BIO) increases MSC β-catenin activity 106-fold and stem cell-associated gene expression ~33-fold, respectively, over untreated controls. Subsequently, BIO treatment increases MSC populations 1.8-fold in typical 2D culture conditions, as well as 1.3-fold when encapsulated within hydrogels compared to untreated cells. Furthermore, we demonstrate that BIO treatment does not reduce MSC multipotency where MSCs maintain their ability to differentiate into osteoblasts, chondrocytes and adipocytes using standard conditions. Taken together, our results demonstrate BIO's potential utility as a proliferative agent for cell transplantation and tissue regeneration.
Collapse
Affiliation(s)
- Michael D Hoffman
- Departments of Biomedical Engineering, Chemical Engineering and Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Danielle S W Benoit
- Departments of Biomedical Engineering, Chemical Engineering and Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
42
|
Naylor AJ, Azzam E, Smith S, Croft A, Poyser C, Duffield JS, Huso DL, Gay S, Ospelt C, Cooper MS, Isacke C, Goodyear SR, Rogers MJ, Buckley CD. The mesenchymal stem cell marker CD248 (endosialin) is a negative regulator of bone formation in mice. ACTA ACUST UNITED AC 2013; 64:3334-43. [PMID: 22674221 DOI: 10.1002/art.34556] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE CD248 (tumor endothelial marker 1/endosialin) is found on stromal cells and is highly expressed during malignancy and inflammation. Studies have shown a reduction in inflammatory arthritis in CD248-knockout (CD248(-/-) ) mice. The aim of the present study was to investigate the functional effect of genetic deletion of CD248 on bone mass. METHODS Western blotting, polymerase chain reaction, and immunofluorescence were used to investigate the expression of CD248 in humans and mice. Micro-computed tomography and the 3-point bending test were used to measure bone parameters and mechanical properties of the tibiae of 10-week-old wild-type (WT) or CD248(-/-) mice. Human and mouse primary osteoblasts were cultured in medium containing 10 mM β-glycerophosphate and 50 μg/ml ascorbic acid to induce mineralization, and then treated with platelet-derived growth factor BB (PDGF-BB). The mineral apposition rate in vivo was calculated by identifying newly formed bone via calcein labeling. RESULTS Expression of CD248 was seen in human and mouse osteoblasts, but not osteoclasts. CD248(-/-) mouse tibiae had higher bone mass and superior mechanical properties (increased load required to cause fracture) compared to WT mice. Primary osteoblasts from CD248(-/-) mice induced increased mineralization in vitro and produced increased bone over 7 days in vivo. There was no decrease in bone mineralization and no increase in proliferation of osteoblasts in response to stimulation with PDGF-BB, which could be attributed to a defect in PDGF signal transduction in the CD248(-/-) mice. CONCLUSION There is an unmet clinical need to address rheumatoid arthritis-associated bone loss. Genetic deletion of CD248 in mice results in high bone mass due to increased osteoblast-mediated bone formation, suggesting that targeting CD248 in rheumatoid arthritis may have the effect of increasing bone mass in addition to the previously reported effect of reducing inflammation.
Collapse
|
43
|
Vandyke K, Fitter S, Drew J, Fukumoto S, Schultz CG, Sims NA, Yeung DT, Hughes TP, Zannettino ACW. Prospective histomorphometric and DXA evaluation of bone remodeling in imatinib-treated CML patients: evidence for site-specific skeletal effects. J Clin Endocrinol Metab 2013; 98:67-76. [PMID: 23144472 DOI: 10.1210/jc.2012-2426] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
CONTEXT Imatinib is a tyrosine kinase inhibitor that has been successfully used to treat Philadelphia chromosome-positive chronic myeloid leukemia (CML) and Kit(+) gastrointestinal stromal tumors. We have previously shown that imatinib therapy is associated with an increase in trabecular bone volume. OBJECTIVE In the present study, we performed a prospective analysis of bone indices in imatinib-treated CML patients to determine the mechanism responsible for this altered bone remodeling. DESIGN, PATIENTS, AND INTERVENTION: This study assessed the effects of high-dose (600 mg/d) imatinib on bone parameters in newly diagnosed chronic-phase Philadelphia chromosome-positive CML patients (n = 11) enrolled in the TIDEL II study. At baseline and after 6, 12, and 24 months of treatment, serum markers of bone remodeling were quantitated, dual-energy x-ray absorptiometry analysis of bone mineral density (BMD) was carried out, and a bone biopsy was collected for histological and micro-computed tomography analysis. RESULTS Our studies show that the increase in trabecular bone volume and trabecular thickness after imatinib treatment was associated with a significant decrease in osteoclast numbers, accompanied by a significant decrease in serum levels of a marker of osteoclast activity. In contrast, osteoblast numbers were not altered by up to 24 months of imatinib treatment. Notably, we also found that imatinib caused a site-specific decrease in BMD at the femoral neck. CONCLUSIONS These data suggest that imatinib therapy dysregulates bone remodeling, causing a generalized decrease in osteoclast number and activity that is not counterbalanced by a decrease in osteoblast activity, leading to increased trabecular bone volume. Further long-term investigations are required to determine the causes and consequences of the site-specific decrease in BMD at the femoral neck.
Collapse
MESH Headings
- Absorptiometry, Photon
- Adult
- Aged
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Benzamides
- Bone Density/drug effects
- Bone Remodeling/drug effects
- Bone Remodeling/physiology
- Bone and Bones/diagnostic imaging
- Bone and Bones/drug effects
- Bone and Bones/pathology
- Female
- Femur Neck/diagnostic imaging
- Femur Neck/drug effects
- Femur Neck/pathology
- Forearm/diagnostic imaging
- Forearm/pathology
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnostic imaging
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology
- Lumbar Vertebrae/diagnostic imaging
- Lumbar Vertebrae/drug effects
- Lumbar Vertebrae/pathology
- Male
- Middle Aged
- Organ Specificity/drug effects
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
Collapse
Affiliation(s)
- Kate Vandyke
- Myeloma Research Laboratory, Division of Hematology, Centre for Cancer Biology, SA Pathology, GPO Box 14, Adelaide, SA, Australia 5000
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Fitter S, Vandyke K, Gronthos S, Zannettino ACW. Suppression of PDGF-induced PI3 kinase activity by imatinib promotes adipogenesis and adiponectin secretion. J Mol Endocrinol 2012; 48:229-40. [PMID: 22474082 DOI: 10.1530/jme-12-0003] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Improved glucose and lipid metabolism is a unique side effect of imatinib therapy in some chronic myeloid leukaemia (CML) patients. We recently reported that plasma levels of adiponectin, an important regulator of insulin sensitivity, are elevated following imatinib therapy in CML patients, which could account for these improved metabolic outcomes. Adiponectin is secreted exclusively from adipocytes, suggesting that imatinib modulates adiponectin levels directly, by transcriptional upregulation of adiponectin in pre-existing adipocytes, and/or indirectly, by stimulating adipogenesis. In this report, we have demonstrated that imatinib promotes adipogenic differentiation of human mesenchymal stromal cells (MSCs), which in turn secrete high-molecular-weight adiponectin. Conversely, imatinib does not stimulate adiponectin secretion from mature adipocytes. We hypothesise that inhibition of PDGFRα (PDGFRA) and PDGFRβ (PDGFRB) is the mechanism by which imatinib promotes adipogenesis. Supporting this, functional blocking antibodies to PDGFR promote adipogenesis and adiponectin secretion in MSC cultures. We have shown that imatinib is a potent inhibitor of PDGF-induced PI3 kinase activation and, using a PI3 kinase p110α-specific inhibitor (PIK-75), we have demonstrated that suppression of this pathway recapitulates the effects of imatinib on MSC differentiation. Furthermore, using mitogens that activate the PI3 kinase pathway, or MSCs expressing constitutively activated Akt, we have shown that activation of the PI3 kinase pathway negates the pro-adipogenic effects of imatinib. Taken together, our results suggest that imatinib increases plasma adiponectin levels by promoting adipogenesis through the suppression of PI3 kinase signalling downstream of PDGFR.
Collapse
Affiliation(s)
- Stephen Fitter
- Myeloma Research Laboratory, Bone and Cancer Research Laboratories, Department of Haematology, Institute of Medical and Veterinary Science, Centre for Cancer Biology, SA Pathology, GPO Box 14, Adelaide, South Australia 5000, Australia
| | | | | | | |
Collapse
|
45
|
Miron RJ, Zhang YF. Osteoinduction: a review of old concepts with new standards. J Dent Res 2012; 91:736-44. [PMID: 22318372 DOI: 10.1177/0022034511435260] [Citation(s) in RCA: 223] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Since the discovery of osteoinduction in the early 20th century, innovative biomaterials with osteoinductive potential have emerged. Over the last 50 years, however, our ability to describe biological phenomena accurately has been improved dramatically by advancements in cell and molecular biology. The aim of this review is to divide the osteoinduction phenomenon into 3 principles: (1) mesenchymal cell recruitment, (2) mesenchymal differentiation to bone-forming osteoblasts, and (3) ectopic bone formation in vivo. Furthermore, this review formulates guidelines for in vitro and in vivo experimental testing for accurately defining new biomaterials as osteoinductive. The use of growth factors with osteoinductive potential in periodontal and oral surgery is discussed. These concepts and guidelines aim to guide the future direction of emerging biomaterials in bone regeneration.
Collapse
Affiliation(s)
- R J Miron
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | | |
Collapse
|
46
|
Fekete N, Gadelorge M, Fürst D, Maurer C, Dausend J, Fleury-Cappellesso S, Mailänder V, Lotfi R, Ignatius A, Sensebé L, Bourin P, Schrezenmeier H, Rojewski MT. Platelet lysate from whole blood-derived pooled platelet concentrates and apheresis-derived platelet concentrates for the isolation and expansion of human bone marrow mesenchymal stromal cells: production process, content and identification of active components. Cytotherapy 2012; 14:540-54. [PMID: 22296115 PMCID: PMC3400099 DOI: 10.3109/14653249.2012.655420] [Citation(s) in RCA: 221] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background aims The clinical use of human mesenchymal stromal cells (MSC) requires ex vivo expansion in media containing supplements such as fetal bovine serum or, alternatively, human platelet lysate (PL). Methods Platelet concentrates were frozen, quarantine stored, thawed and sterile filtered to obtain PL. PL content and its effect on fibroblast-colony-forming unit (CFU-F) formation, MSC proliferation and large-scale expansion were studied. Results PL contained high levels of basic fibroblast growth factor (bFGF), soluble CD40L (sCD40L), vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), platelet-derived growth factor AA (PDGF-AA), platelet-derived growth factor AB/BB (PDGF-AB/BB), chemokine (C-C) ligand 5 (CCL5; RANTES) transforming growth factor-β1 (TGF-β1) and chemokine (C-X-C) ligand 1/2/3 (GRO), with low batch-to-batch variability, and most were stable for up to 14 days. Inhibition of PDGF-BB and bFGF decreased MSC proliferation by about 20% and 50%, respectively. The strongest inhibition (about 75%) was observed with a combination of anti-bFGF + anti-PDGF-BB and anti-bFGF + anti-TGF-β1 + anti-PDGF-BB. Interestingly, various combinations of recombinant PDGF-BB, bFGF and TGF-β1 were not sufficient to promote cell proliferation. PL from whole blood-derived pooled platelet concentrates and apheresis platelet concentrates did not differ significantly in their growth-promoting activity on MSC. Conclusions PL enhances MSC proliferation and can be regarded as a safe tool for MSC expansion for clinical purposes. \in particular, PDGF-BB and bFGF are essential components for the growth-promoting effect of PL, but are not sufficient for MSC proliferation.
Collapse
Affiliation(s)
- Natalie Fekete
- Institut für Transfusionsmedizin, Universität Ulm und Institut für Klinische Transfusionsmedizin und Immungenetik Ulm, DRK-Blutspendedienst Baden-Württemberg-Hessen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Jönsson S, Hjorth-Hansen H, Olsson B, Wadenvik H, Sundan A, Standal T. Imatinib inhibits proliferation of human mesenchymal stem cells and promotes early but not late osteoblast differentiation in vitro. J Bone Miner Metab 2012; 30:119-23. [PMID: 21993629 DOI: 10.1007/s00774-011-0323-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 09/04/2011] [Indexed: 12/11/2022]
Abstract
Altered bone metabolism has been reported in patients with chronic myeloid leukemia treated with the tyrosine kinase inhibitor imatinib. Several studies have shown that imatinib inhibits the differentiation and activity of osteoclasts in vitro, whereas the effects of imatinib on osteoblast differentiation are less clear. In this study osteoblast differentiation was induced in human mesenchymal stem cells (hMSCs) by treatment with bone morphogenetic protein 2 in vitro. Imatinib inhibited proliferation of hMSCs in a dose-dependent manner. Even though imatinib promoted early osteoblast differentiation assessed by alkaline phosphate activity, mineralization measured by Alizarin Red staining (ARS) was reduced by imatinib. Moreover, the inhibitory effect of imatinib on mineralization was most prominent at low concentrations of imatinib. When we measured the relative mRNA expression levels of Runx2, we found that Runx2 expression was higher in imatinib-treated (5 μM) cultures at early time points during differentiation. On the other hand, the expression of Osterix late during differentiation was lower in imatinib-treated (5 μM) cultures, corresponding to the ARS results. Thus, the effect of imatinib on osteoblast differentiation is not only dependent on the drug concentration, but indeed also on the maturation stage of the cells. This finding might partly explain why previous studies on the effects of imatinib osteoblast differentiation have shown different results.
Collapse
Affiliation(s)
- Sofia Jönsson
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway.
| | | | | | | | | | | |
Collapse
|
48
|
Borriello A, Caldarelli I, Basile MA, Bencivenga D, Tramontano A, Perrotta S, Ragione FD, Oliva A. The tyrosine kinase inhibitor dasatinib induces a marked adipogenic differentiation of human multipotent mesenchymal stromal cells. PLoS One 2011; 6:e28555. [PMID: 22164306 PMCID: PMC3229607 DOI: 10.1371/journal.pone.0028555] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 11/10/2011] [Indexed: 12/22/2022] Open
Abstract
Background The introduction of specific BCR-ABL inhibitors in chronic myelogenous leukemia therapy has entirely mutated the prognosis of this hematologic cancer from being a fatal disorder to becoming a chronic disease. Due to the probable long lasting treatment with tyrosine-kinase inhibitors (TKIs), the knowledge of their effects on normal cells is of pivotal importance. Design and Methods We investigated the effects of dasatinib treatment on human bone marrow-derived mesenchymal stromal cells (MSCs). Results Our findings demonstrate, for the first time, that dasatinib induces MSCs adipocytic differentiation. Particularly, when the TKI is added to the medium inducing osteogenic differentiation, a high MSCs percentage acquires adipocytic morphology and overexpresses adipocytic specific genes, including PPARγ, CEBPα, LPL and SREBP1c. Dasatinib also inhibits the activity of alkaline phosphatase, an osteogenic marker, and remarkably reduces matrix mineralization. The increase of PPARγ is also confirmed at protein level. The component of osteogenic medium required for dasatinib-induced adipogenesis is dexamethasone. Intriguingly, the increase of adipocytic markers is also observed in MSCs treated with dasatinib alone. The TKI effect is phenotype-specific, since fibroblasts do not undergo adipocytic differentiation or PPARγ increase. Conclusions Our data demonstrate that dasatinib treatment affects bone marrow MSCs commitment and suggest that TKIs therapy might modify normal phenotypes with potential significant negative consequences.
Collapse
Affiliation(s)
- Adriana Borriello
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
| | - Ilaria Caldarelli
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
| | - Maria Assunta Basile
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
| | - Debora Bencivenga
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
| | - Annunziata Tramontano
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
| | - Silverio Perrotta
- Department of Pediatrics, Second University of Naples, Naples, Italy
| | - Fulvio Della Ragione
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
- * E-mail:
| | - Adriana Oliva
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
| |
Collapse
|
49
|
Baldazzi V, Tassi R, Lapini A, Lunghi A, Garofoli E, Caruso S, Carini M, Mazzanti R. Sunitinib-induced hyperparathyroidism: a possible mechanism to altered bone homeostasis. Cancer 2011; 118:3165-72. [PMID: 21956360 DOI: 10.1002/cncr.26435] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 06/01/2011] [Accepted: 06/20/2011] [Indexed: 11/10/2022]
Abstract
BACKGROUND Sunitinib malate is an orally bioavailable tyrosine kinase inhibitor that is active against many tyrosine kinase receptors involving crucial pathways in both healthy tissues and malignant tissues. Because its use in clinical practice is quite recent, many of its possible side effects remain unknown. In this report, the authors describe the incidence of new-onset hyperparathyroidism in a cohort of patients with metastatic renal cell carcinoma who received treatment with sunitinib. METHODS Twenty-six patients who received first-line sunitinib for metastatic renal cell carcinoma were enrolled in this study for a mineral and parathyroid function assessment. Plasma levels of intact parathyroid hormone; serum levels of calcium, phosphorus, 25-hydroxyvitamin D(3), and 1,25-dihydrovitamin D(3); and urinary 24-hour calcium and phosphorus excretion all were measured in each patient. Biochemical evaluations were performed before the beginning of treatment and at the end of each sunitinib treatment period. RESULTS Eighteen of 26 patients (69.2%) developed hyperparathyroidism with normal serum calcium levels, and 6 of them developed hypophosphatemia. Patients presented with a mean elevation of parathyroid hormone after 2.2 cycles of sunitinib. The levels of 25-OH vitamin D(3) were stable over the course of treatment, whereas 1,25-OH vitamin D(3) levels were increased in 5 hyperparathyroid patients. Those who presenting with elevated parathyroid hormone levels had low or undetectable urinary calcium levels. Parathyroid hormone elevation usually persisted but did not progress during long-term therapy with sunitinib. Permanent treatment interruption resulted in a resolution of hyperparathyroidism. CONCLUSIONS Hyperparathyroidism developed in an high percentage of patients on sunitinib. Therefore, the authors concluded that sunitinib may affect parathyroid function and bone mineral homeostasis, possibly resulting in abnormal bone remodeling.
Collapse
Affiliation(s)
- Valentina Baldazzi
- Second Medical Oncology Unit, Careggi University Hospital, Florence University, Florence, Italy
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Paracrine molecules of mesenchymal stem cells for hematopoietic stem cell niche. BONE MARROW RESEARCH 2011; 2011:353878. [PMID: 22046560 PMCID: PMC3196250 DOI: 10.1155/2011/353878] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 07/26/2011] [Accepted: 07/26/2011] [Indexed: 12/24/2022]
Abstract
Hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) are both adult stem cells residing in the bone marrow. MSCs interact with HSCs, they stimulate and enhance the proliferation of HSCs by secreting regulatory molecules and cytokines, providing a specialized microenvironment for controlling the process of hematopoiesis. In this paper we discuss how MSCs contribute to HSC niche, maintain the stemness and proliferation of HSCs, and support HSC transplantation.
Collapse
|