Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 The altered differentiation and senescence of BMSCs contribute to senile osteoporosis. Transplantation of normal BMSCs, modification of altered differentiation ability of BMSCs, and elimination of senescent BMSCs can effectively treat the senile osteoporo + 1127 word(s) 1127 2020-01-08 04:38:46 |
2 format correct + 5 word(s) 1132 2020-11-01 09:10:37 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Qadir, A.; Liang, S.; Wu, Z.; Chen, Z.; Hu, L.; Qian, A. Senile Osteoporosis by BMSCs. Encyclopedia. Available online: https://encyclopedia.pub/entry/298 (accessed on 20 April 2024).
Qadir A, Liang S, Wu Z, Chen Z, Hu L, Qian A. Senile Osteoporosis by BMSCs. Encyclopedia. Available at: https://encyclopedia.pub/entry/298. Accessed April 20, 2024.
Qadir, Abdul, Shujing Liang, Zixiang Wu, Zhihao Chen, Lifang Hu, Airong Qian. "Senile Osteoporosis by BMSCs" Encyclopedia, https://encyclopedia.pub/entry/298 (accessed April 20, 2024).
Qadir, A., Liang, S., Wu, Z., Chen, Z., Hu, L., & Qian, A. (2020, February 15). Senile Osteoporosis by BMSCs. In Encyclopedia. https://encyclopedia.pub/entry/298
Qadir, Abdul, et al. "Senile Osteoporosis by BMSCs." Encyclopedia. Web. 15 February, 2020.
Senile Osteoporosis by BMSCs
Edit

Senile osteoporosis has become a worldwide bone disease with the aging of the world population. Unlike postmenopausal osteoporosis which is linked to menopause in women, senile osteoporosis is due to aging, hence, affecting both men and women. Evidence has shown that with age increase, bone marrow stromal cells (BMSCs) differentiate into more adipocytes rather than osteoblasts and undergo senescence, which leads to decreased bone formation and contributes to senile osteoporosis. Therefore, modulating BMSCs to stimulate them either differentiate into more osteoblasts than adipocytes or eliminate their senescence will be wonderful strategies for treating senile osteoporosis. Here, the treatment of senile osteoporosis by aiming at BMSCs is introduced.

senile osteoporosis bone marrow stromal cells treatment

1. Introduction

In old ages, bone marrow stromal cells (BMSCs) either differentiate into more adipocytes than osteoblasts or assume senescence, which ultimately results in senile osteoporosis. Therefore, in order to treat senile osteoporosis, it is required to use the strategies in what BMSCs can be stimulated either to differentiate into more osteoblasts than adipocytes or be eliminated their senescence. To date, numerous molecules including parathyroid hormone (PTH 1–84) or only its N-terminal fragment teriparatide (PTH 1–34), bisphosphonates, tetracycline, cationic peptides and antibodies like denosumab and romosozumab have been used in the treatment of senile osteoporosis [1][2][3][4][5]. However, most of them are limited either, due to their severe side effects or inhibition of just bone resorption without decreasing bone regeneration. Therefore, in order to reduce such limitations, there is the need of using cell-based therapy strategy, for which BMSCs can act as an ideal cell source, due to their self-renewing and differentiation ability into various types of cells. In addition, easy isolation with high yields from different tissues, and immunosuppressive and immunoprivileged properties of BMSCs also make them the preferable cell source in cell-based therapies [6].

2. Treatment of Senile Osteoporosis by Aiming at BMSCs

In order to treat senile osteoporosis, several researchers have reported the successful transplantation of BMSCs using animal models. Transplanted BMSCs serve in bone formation either by allocating damaged areas to differentiate into osteoblasts or assume paracrine mode, due to which they secrete specific growth factors to make a favorable environment for the nearby cells to repair the degenerative tissue [7]. Ichioka et al. injected normal allogeneic BMSCs intra bone marrow into the senescence accelerated mouse prone 6 (SAMP6) mice, which naturally prone to senile osteoporosis in their early lives. They demonstrated that the injected normal BMSCs were able to prevent the senile osteoporosis in SAMP6 mice with an increase in trabecular bone mass and decline in bone mineral density (BMD) loss [8]. Takada et al. also treated osteoporosis after it occurred in aged SAMP6 mice by injecting normal allogeneic BMSCs locally into their bone marrow. After the clinical examinations, no signs of senile osteoporosis were found, hence, succeeded in proving their hypothesis [9]. In another experimental procedure, when BMSCs isolated from healthy rats were injected into the bone marrow of femurs of osteoporotic female ovariectomized rats, a quite increase in the bone mass of femur was observed after examination [10]. Similarly, Kiernan et al. also found an increase in bone formation when they injected systemically normal allogeneic BMSCs into the bone marrow of senile osteoporotic mouse model, giving a clue towards their applications against human senile osteoporosis [11].

Certain factors, microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) have also been recognized to play significant roles in treating senile osteoporosis by stimulating BMSCs to differentiate into more osteoblasts than adipocytes. Suppression of ectopic viral integration site‑1 (Evi1) gene through RNA interference in rat BMSCs resulted in increased osteogenesis and decreased adipogenesis, suggesting Evi1 as a potent target for treating osteoporosis [12]. Jing et al. have reported enhancer of zeste homology 2 (EZH2) factor as a competent therapeutic target for enhancing bone formation during osteoporosis as its suppression led to increased osteogenesis rather than adipogenesis [13]. Recently, Zhou et al. uncovered the role of orcinol glucoside (OG), a constituent of traditional Chinese medicine, in promoting bone formation. They reported that OG was able to revert the BMSCs differentiation fashion of more into adipocytes than osteoblasts in old ages through Wnt/β-catenin signaling pathway, thus, may act as a novel therapeutic agent against senile osteoporosis [14]. Li et al. found the increased bone formation and decreased fat accumulation after injecting aptamer-antagomiR-188 into the bone marrow of osteoporotic aged mice. The aptamer-antagomiR-188 actually inhibited miR-188, whose overexpression is actually responsible for reducing osteogenesis and increasing adipogenesis [15]. Let-7, a miRNA family, has also been distinguished to promote osteogenesis and decline adipogenesis in BMSCs [16]. Very recently, Zhao et al. have demonstrated that miR-21 possesses the ability to stimulate the osteogenic differentiation of BMSCs by finding the role of miR-21 inhibitor in inhibiting BMSCs differentiation into osteoblasts [17]. Recently, lncRNA Bmncr was found as key regulator in promoting osteogenesis and inhibiting adipogenesis in mice during aging, suggesting it to be a therapeutic target against senile osteoporosis in future [18]. Chen et al. reported that overexpression of lncRNA XIST led to the inhibition of osteogenic differentiation of BMSCs in 3-week-old Sprague Dawley rats [19], thus, its inhibition through specific inhibitor can revert the phenomenon and can treat the senile osteoporosis. Most recently, Zhu et al. have identified lncRNA HOXA-AS2 as a key positive regulator in causing osteogenesis in BMSCs through NF-κB signaling inactivation [20], which may act as a new therapeutic target against senile osteoporosis.

Different approaches have also been used to eliminate the senescence of BMSCs, and thus, treat senile osteoporosis. Elimination of senescent cells is of much importance regarding bone mass and strength. In order to uncover such importance, Farr et al. used some genetic and pharmacological procedures to eliminate the senescent cells. They found that activating INK-ATTAC caspase 8 in senescent cells or treating senescent cells with JAK inhibitor or senolytics increased bone mass and bone strength in mice with the bone loss [21]. A senolytic drug, ABT263 can also reduce senescence associated factors, hence, can act as a good therapeutic drug against senile osteoporosis [22]. Gao et al. delivered tetramethylpyrazine (TMP) locally into the bone marrow of aging mice with established senescent BMSCs microenvironment and a significant reduction was found in senescent phenotype via modulating Ezh2-H3k27me3, suggesting TMP as a potent local eliminator of senescent BMSCs in age-related bone loss [23]. Sun et al. suppressed the expression of NADPH oxidase, which is mainly involved in ROS formation in BMSCs. They found a significant increase in osteoblasts differentiation of BMSCs. Moreover, they also found an increase in bone formation after treating SAMP6 mice with apocynin for three months, hence, declared apocynin as a competent therapeutic agent against age-related bone loss [24]. More recently, Zhou et al. demonstrated that resveratrol was able to attenuate senescence and promote osteogenic differentiation of BMSCs by inhibiting AMPK activation/ROS inhibition signaling pathway in aged mouse, suggesting resveratrol as a novel therapy against senile osteoporosis, due to its inhibiting effects on ROS formation in BMSCs [25].

References

  1. Cara A. Cipriano; Paul S. Issack; Lisa Shindle; Clément M. L. Werner; David L. Helfet; Joseph M. Lane; Recent Advances Toward the Clinical Application of PTH (1-34) in Fracture Healing. HSS Journal ® 2009, 5, 149-153, 10.1007/s11420-009-9109-8.
  2. Andrew R. Hilgenbrink; Philip S. Low; Folate Receptor-Mediated Drug Targeting: From Therapeutics to Diagnostics. Journal of Pharmaceutical Sciences 2005, 94, 2135-2146, 10.1002/jps.20457.
  3. E Michael Lewiecki; Rajani V Dinavahi; Marise Lazaretti-Castro; Peter R Ebeling; Jonathan D Adachi; Akimitsu Miyauchi; Evelien Gielen; Cassandra E Milmont; Cesar Libanati; Andreas Grauer; One Year of Romosozumab Followed by Two Years of Denosumab Maintains Fracture Risk Reductions: Results of the FRAME Extension Study. Journal of Bone and Mineral Research 2018, 34, 419-428, 10.1002/jbmr.3622.
  4. Federico Mingozzi; Katherine A. High; Erratum: Therapeutic in vivo gene transfer for genetic disease using AAV: progress and challenges. Nature Reviews Genetics 2011, 12, 515-515, 10.1038/nrg3027.
  5. Zia Ur Rehman; Inge S. Zuhorn; Dick Hoekstra; How cationic lipids transfer nucleic acids into cells and across cellular membranes: Recent advances. Journal of Controlled Release 2013, 166, 46-56, 10.1016/j.jconrel.2012.12.014.
  6. Jitrada Phetfong; Tanwarat Sanvoranart; Kuneerat Nartprayut; Natakarn Nimsanor; Kanokwan Seenprachawong; Virapong Prachayasittikul; Aungkura Supokawej; Osteoporosis: the current status of mesenchymal stem cell-based therapy.. Cellular & Molecular Biology Letters 2016, 21, 12, 10.1186/s11658-016-0013-1.
  7. Xinhai Ye; Peng Zhang; Shaobo Xue; Yipin Xu; Jian Tan; Guangpeng Liu; Adipose-derived stem cells alleviate osteoporosis by enchancing osteogenesis and inhibiting adipogenesis in a rabbit model. Cytotherapy 2014, 16, 1643-1655, 10.1016/j.jcyt.2014.07.009.
  8. Naoya Ichioka; Muneo Inaba; Taketohi Kushida; Takashi Esumi; Kazuhiko Takahara; Kayo Inaba; Ryokei Ogawa; Hirokazu Iida; Susumu Ikehara; Prevention of Senile Osteoporosis in SAMP6 Mice by Intrabone Marrow Injection of Allogeneic Bone Marrow Cells. STEM CELLS 2002, 20, 542-551, 10.1634/stemcells.20-6-542.
  9. Gowdhaman Et Al. Gowdhaman Et Al.; Tjprc; Sourabh Jain Et Al. Sourabh Jain Et Al.; Test 1. International Journal of Agricultural Science and Research 2018, 8, 1-15, 10.24247/ijdrdjun20186.
  10. Natália De Melo Ocarino; Jankerle Neves Boeloni; Vanda Jorgetti; Dawidson Assis Gomes; Alfredo Miranda Goes; Rogéria Serakides; Intra-bone marrow injection of mesenchymal stem cells improves the femur bone mass of osteoporotic female rats. Connective Tissue Research 2010, 51, 426-433, 10.3109/03008201003597049.
  11. Jeffrey Kiernan; Sally Hu; Marc D. Grynpas; John E. Davies; William L. Stanford; Systemic Mesenchymal Stromal Cell Transplantation Prevents Functional Bone Loss in a Mouse Model of Age-Related Osteoporosis.. STEM CELLS Translational Medicine 2016, 5, 683-93, 10.5966/sctm.2015-0231.
  12. Qijun An; Dou Wu; Yuehong Ma; Biao Zhou; Qiang Liu; Suppression of Evi1 promotes the osteogenic differentiation and inhibits the adipogenic differentiation of bone marrow-derived mesenchymal stem cells in vitro. International Journal of Molecular Medicine 2015, 36, 1615-1622, 10.3892/ijmm.2015.2385.
  13. Huan Jing; Li Liao; Yulin An; Xiaoxia Su; Shiyu Liu; Yi Shuai; Xinjing Zhang; Yan Jin; Suppression of EZH2 Prevents the Shift of Osteoporotic MSC Fate to Adipocyte and Enhances Bone Formation During Osteoporosis. Molecular Therapy 2016, 24, 217-229, 10.1038/mt.2015.152.
  14. Xinying Zhou; Zezheng Liu; Bin Huang; Huibo Yan; Changsheng Yang; Qingchu Li; Dadi Jin; Orcinol glucoside facilitates the shift of MSC fate to osteoblast and prevents adipogenesis via Wnt/β-catenin signaling pathway.. Drug Design, Development and Therapy 2019, 13, 2703-2713, 10.2147/DDDT.S208458.
  15. Chang-Jun Li; Peng Cheng; Meng-Ke Liang; Yu-Si Chen; Qiong Lu; Jin-Yu Wang; Zhu-Ying Xia; Hou-De Zhou; Xu Cao; Hui Xie; Er-Yuan Liao; Xiang-Hang Luo; MicroRNA-188 regulates age-related switch between osteoblast and adipocyte differentiation.. Journal of Clinical Investigation 2015, 125, 1509-22, 10.1172/JCI77716.
  16. Jianfeng Wei; Hongling Li; Shihua Wang; Tangping Li; Junfen Fan; Xiaolei Liang; Jing Li; Qin Han; Li Zhu; Linyuan Fan; Robert Chunhua Zhao; let-7 Enhances Osteogenesis and Bone Formation While Repressing Adipogenesis of Human Stromal/Mesenchymal Stem Cells by Regulating HMGA2. Stem Cells and Development 2014, 23, 1452-1463, 10.1089/scd.2013.0600.
  17. Zhongfu Zhao; Xiaoguang Li; Dexun Zou; Yongyun Lian; Shaohua Tian; Zhi Dou; Expression of microRNA-21 in osteoporotic patients and its involvement in the regulation of osteogenic differentiation. Experimental and Therapeutic Medicine 2018, 17, 709-714, 10.3892/etm.2018.6998.
  18. Chang-Jun Li; Ye Xiao; Mi Yang; Tian Su; Xi Sun; Qi Guo; Yan Huang; Xiang-Hang Luo; Long noncoding RNA Bmncr regulates mesenchymal stem cell fate during skeletal aging. Journal of Clinical Investigation 2018, 128, 5251-5266, 10.1172/jci99044.
  19. Xi Chen; Lei Yang; Dawei Ge; Weiwei Wang; Zhaowei Yin; Junwei Yan; Xiaojian Cao; Chunzhi Jiang; Shengnai Zheng; Bin Liang; Long non-coding RNA XIST promotes osteoporosis through inhibiting bone marrow mesenchymal stem cell differentiation. Experimental and Therapeutic Medicine 2018, 17, 803-811, 10.3892/etm.2018.7033.
  20. Xinxing Zhu; Jinjin Yu; Jiang Du; Genshen Zhong; Liang Qiao; Juntang Lin; LncRNA HOXA‐AS2 positively regulates osteogenesis of mesenchymal stem cells through inactivating NF‐κB signalling. Journal of Cellular and Molecular Medicine 2018, 23, 1325-1332, 10.1111/jcmm.14034.
  21. Joshua N Farr; Ming Xu; Megan M Weivoda; David G Monroe; Daniel G Fraser; Jennifer L Onken; Brittany A Negley; Jad G Sfeir; Mikolaj B Ogrodnik; Christine M Hachfeld; Nathan K Lebrasseur; Matthew T Drake; Robert J Pignolo; Tamar Pirtskhalava; Tamara Tchkonia; Merry Jo Oursler; James L Kirkland; Sundeep Khosla; Erratum: Corrigendum: Targeting cellular senescence prevents age-related bone loss in mice. Nature Medicine 2017, 23, 1384-1384, 10.1038/nm1117-1384c.
  22. Ha‐Neui Kim; Jianhui Chang; Lijian Shao; Li Han; Srividhya Iyer; Stavros C. Manolagas; Charles A. O'brien; Robert L. Jilka; Daohong Zhou; Maria Almeida; DNA damage and senescence in osteoprogenitors expressing Osx1 may cause their decrease with age. Aging Cell 2017, 16, 693-703, 10.1111/acel.12597.
  23. Bo Gao; Xisheng Lin; Huan Jing; Jing Fan; Chenchen Ji; Qiang Jie; Chao Zheng; Di Wang; Xiaolong Xu; Yaqian Hu; Weiguang Lu; Zhuojing Luo; Liu Yang; Local delivery of tetramethylpyrazine eliminates the senescent phenotype of bone marrow mesenchymal stromal cells and creates an anti‐inflammatory and angiogenic environment in aging mice. Aging Cell 2018, 17, e12741, 10.1111/acel.12741.
  24. Jinlong Sun; Leiguo Ming; Fengqing Shang; Lijuan Shen; Jihua Chen; Yan Jin; Apocynin suppression of NADPH oxidase reverses the aging process in mesenchymal stem cells to promote osteogenesis and increase bone mass. Scientific Reports 2015, 5, 18572, 10.1038/srep18572.
  25. Ting Zhou; Yurong Yan; Chenchen Zhao; Yao Xu; Qiong Wang; Na Xu; Resveratrol improves osteogenic differentiation of senescent bone mesenchymal stem cells through inhibiting endogenous reactive oxygen species production via AMPK activation. Redox Report 2019, 24, 62-69, 10.1080/13510002.2019.1658376.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 849
Revisions: 2 times (View History)
Update Date: 01 Nov 2020
1000/1000