1
|
Wu W, Yu M, Liu Y, Wang X, Wang L, Wei Q, Feng H, Chen J. Effects of a functional milk powder supplemented with rhizoma drynariae extracts on bone health in growing and ovariectomized rats: More effective for lumbar vertebrae? J Dairy Sci 2025; 108:3480-3498. [PMID: 39892604 DOI: 10.3168/jds.2024-25896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 12/24/2024] [Indexed: 02/04/2025]
Abstract
Bone health is vital through all life stages and has become a growing public health concern. The role of rhizoma drynariae (RD) as a traditional Chinese medicine for bone injury recovery has been widely investigated. Still, there is little research related to RD as a functional component in food to improve calcium bioavailability and bone health. Milk powder is a daily vital food source of dietary calcium. The present study aimed to investigate and compare the effects of a functional milk powder reinforced with RD extracts (RDE) or vitamin D, vitamin K2, and calcium, or both, using Sprague-Dawley rats, with both low calcium growing rat model and low calcium ovariectomized (OVX) rat model. For growing rats, RDE increased bone mineral density (BMD) and improved the microstructure of bone trabecula in the femur and lumbar vertebrae. The levels of serum bone turnover markers C-telopeptide of type Ⅰ collagen (CTX-I), procollagen type Ⅰ amino-terminal peptide (PINP), and osteocalcin (BGP) were increased with RDE supplementation. The tartrate-resistant acid phosphatase staining suggested a decrease in the number of osteoclasts in the femur. Immunohistochemistry showed that the osteoclast-related protein RANKL was downregulated, and the osteogenic-associated proteins RUNX2 and OSX significantly increased in the lumbar vertebrae. Similarly, in OVX rats, RDE improved BMD and microstructure in lumbar vertebrae, and the levels of CTX-I, PINP, and BGP were also increased. In conclusion, the novel functional milk powder supplemented with RDE facilitated osteogenesis and inhibited osteoclast in rats at both stages, especially with lumbar vertebrae in growing rats. Our findings provide new prospects and a more precise target for enhancing bone health by incorporating RDE as a functional component in milk powder.
Collapse
Affiliation(s)
- Wenjing Wu
- Department of Nutrition and Food Safety, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu 610041, China
| | - Mengqi Yu
- Department of Nutrition and Food Safety, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu 610041, China
| | - Yan Liu
- National Center of Technology Innovation for Dairy, Hohhot 010110, China
| | - Xiaoya Wang
- Department of Nutrition and Food Safety, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu 610041, China
| | - Liang Wang
- Department of Nutrition and Food Safety, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu 610041, China
| | - Qijie Wei
- Department of Nutrition and Food Safety, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu 610041, China
| | - Haotian Feng
- National Center of Technology Innovation for Dairy, Hohhot 010110, China.
| | - Jinyao Chen
- Department of Nutrition and Food Safety, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu 610041, China.
| |
Collapse
|
2
|
Xiao T, Shi Y, Ye Y, Wang J, Wang W, Yu H, Yan M, Yu J. Circ-SPATA13 regulates the osteogenic differentiation of human periodontal ligament stem cells through the miR-485-5p_R + 1/BMP7 axis. Cell Signal 2025; 127:111561. [PMID: 39667547 DOI: 10.1016/j.cellsig.2024.111561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/29/2024] [Accepted: 12/08/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Human periodontal ligament stem cells (PDLSCs) are widely available and have strong osteogenic differentiation ability, which makes them promising tools for bone regeneration. Circular RNAs (circRNAs) play a variety of functions in the process of cell differentiation and are potential therapeutic targets. Here, we identified a new circRNA, circ-SPATA13, and found that it was highly positively correlated with the osteogenic differentiation of PDLSCs. Therefore, in this study, we revealed the significance and mechanism of circ-SPATA13 in the osteogenic differentiation of PDLSCs. METHODS PDLSCs were isolated from third molars with incomplete apical development and induced to undergo chondrogenic, adipogenic, or osteogenic differentiation. Surface markers were detected via flow cytometry. Proliferation was assessed with EdU and CCK-8 assays. The circ-SPATA13 and miR-485-5p_R + 1-mediated control of mineral deposition was evaluated through alizarin red and alkaline phosphatase staining. Osteogenesis-related factor expression was detected through western blotting, immunofluorescence, and qRT-PCR. Fluorescence in situ hybridization was used to examine circ-SPATA13 localization within PDLSCs. The relationships among circ-SPATA13, miR-485-5p_R + 1, and BMP7 during PDLSCs osteogenesis were assessed through western blotting, qRT-PCR, dual-luciferase assay, rescue experiment, and bioinformatics approaches. RESULTS Primary PDLSCs expressing mesenchymal stem cell surface markers were isolated. Circ-SPATA13 was identified and found to have no impact on PDLSC proliferation, whereas it was a positive regulator of their osteogenic differentiation, a process which was antagonized by miR-485-5p_R + 1. Dual-luciferase reporter assays revealed that circ-SPATA13 was able to function as a molecular sponge to sequester miR-485-5p_R + 1 within PDLSCs, while this miRNA was able to bind to the 3'-UTR of the target mRNA BMP7. In rescue experiments, circ-SPATA13 was confirmed to regulate the osteogenic differentiation of PDLSCs via this miR-485-5p_R + 1/BMP7 axis. Moreover, in vivo experiments in rats demonstrated that the overexpression of circ-SPATA13 in PDLSCs was associated with the promotion of bone formation in a skull defect model system. CONCLUSION These data supported the osteogenic functions of circ-SPATA13 in PDLSCs. Mechanistically, this circRNA was found to function as a molecular sponge for miR-485-5p_R + 1, in turn targeting BMP7 to promote the osteogenic differentiation of PDLSCs. This circ-SPATA13/miR-485-5p_R + 1/BMP7 axis may be a novel target for treatments promoting PDLSCs osteogenic differentiation.
Collapse
Affiliation(s)
- Tong Xiao
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Yijia Shi
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Yu Ye
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Jing Wang
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Wenmin Wang
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Haowen Yu
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Maoshen Yan
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Jinhua Yu
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
3
|
Kim JH, Schulte AJ, Sarver AL, Lee D, Angelos MG, Frantz AM, Forster CL, O'Brien TD, Cornax I, O'Sullivan MG, Cheng N, Lewellen M, Oseth L, Kumar S, Bullman S, Pedamallu CS, Goyal SM, Meyerson M, Lund TC, Breen M, Lindblad-Toh K, Dickerson EB, Kaufman DS, Modiano JF. Hemangiosarcoma Cells Promote Conserved Host-derived Hematopoietic Expansion. CANCER RESEARCH COMMUNICATIONS 2024; 4:1467-1480. [PMID: 38757809 PMCID: PMC11166094 DOI: 10.1158/2767-9764.crc-23-0441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/29/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024]
Abstract
Hemangiosarcoma and angiosarcoma are soft-tissue sarcomas of blood vessel-forming cells in dogs and humans, respectively. These vasoformative sarcomas are aggressive and highly metastatic, with disorganized, irregular blood-filled vascular spaces. Our objective was to define molecular programs which support the niche that enables progression of canine hemangiosarcoma and human angiosarcoma. Dog-in-mouse hemangiosarcoma xenografts recapitulated the vasoformative and highly angiogenic morphology and molecular characteristics of primary tumors. Blood vessels in the tumors were complex and disorganized, and they were lined by both donor and host cells. In a series of xenografts, we observed that the transplanted hemangiosarcoma cells created exuberant myeloid hyperplasia and gave rise to lymphoproliferative tumors of mouse origin. Our functional analyses indicate that hemangiosarcoma cells generate a microenvironment that supports expansion and differentiation of hematopoietic progenitor populations. Furthermore, gene expression profiling data revealed hemangiosarcoma cells expressed a repertoire of hematopoietic cytokines capable of regulating the surrounding stromal cells. We conclude that canine hemangiosarcomas, and possibly human angiosarcomas, maintain molecular properties that provide hematopoietic support and facilitate stromal reactions, suggesting their potential involvement in promoting the growth of hematopoietic tumors. SIGNIFICANCE We demonstrate that hemangiosarcomas regulate molecular programs supporting hematopoietic expansion and differentiation, providing insights into their potential roles in creating a permissive stromal-immune environment for tumor progression.
Collapse
Affiliation(s)
- Jong Hyuk Kim
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
- University of Florida Health Cancer Center, University of Florida, Gainesville, Florida
- Intelligent Critical Care Center, University of Florida, Gainesville, Florida
- Artificial Intelligence Academic Initiative (AI) Center, University of Florida, Gainesville, Florida
| | - Ashley J. Schulte
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Aaron L. Sarver
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Institute for Health Informatics, University of Minnesota, Minneapolis, Minnesota
| | - Donghee Lee
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Mathew G. Angelos
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
- Department of Medicine (Division of Hematology, Oncology, and Transplantation), Medical School, University of Minnesota, Minneapolis, Minnesota
- Microbiology, Immunology and Cancer Biology (MICaB) Graduate Program, University of Minnesota, Minneapolis, Minnesota
- Department of Medicine, Division of Hematology and Oncology, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Aric M. Frantz
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Capstan Therapeutics, San Diego, California
| | - Colleen L. Forster
- The University of Minnesota Biological Materials Procurement Network (BioNet), University of Minnesota, Minneapolis, Minnesota
| | - Timothy D. O'Brien
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
| | - Ingrid Cornax
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Janssen Research and Development, LLC
| | - M. Gerard O'Sullivan
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
| | - Nuojin Cheng
- School of Mathematics, College of Science and Engineering, University of Minnesota, Minneapolis, Minnesota
- Applied Mathematics, University of Colorado Boulder, Boulder, Colorado
| | - Mitzi Lewellen
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - LeAnn Oseth
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Sunil Kumar
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
| | - Susan Bullman
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Chandra Sekhar Pedamallu
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Sagar M. Goyal
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
| | - Matthew Meyerson
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Troy C. Lund
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
- Cancer Genetics Program, University of North Carolina Lineberger Comprehensive Cancer Center, Raleigh, North Carolina
| | - Kerstin Lindblad-Toh
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Science of Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Erin B. Dickerson
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Dan S. Kaufman
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
- Department of Medicine (Division of Hematology, Oncology, and Transplantation), Medical School, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota
- Division of Regenerative Medicine, Department of Medicine, University of California-San Diego, La Jolla, California
| | - Jaime F. Modiano
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, Minnesota
- Center for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
4
|
Mohammed SA, Abd Elsattar M, Abd-Allah SH, Habashy OY, Abdelghany EMA, Hussein S, Abdullah O. Effect of Bone-Marrow-Derived Mesenchymal Stem Cells on the Healing of Bone Fractures. J Interferon Cytokine Res 2021; 41:336-346. [PMID: 34543130 DOI: 10.1089/jir.2021.0093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
This study was performed to evaluate the effectiveness of mesenchymal stem cells (MSCs) on bone healing and to assess the role of various chemical stimulants and mediators in healing. Forty female mice were randomly assigned to 4 groups (10 mice each) after the induction of fixed fractures: group I: received fixation only; group II: received phosphate-buffered saline (PBS); group III: received intralesion MSCs (IL-MSCs); and group IV: received intraperitoneal MSCs (IP-MSCs). Serum alkaline phosphatase (ALP) levels and the expression of the osteocalcin (OCN), bone morphogenetic protein-2 (BMP-2), and stromal-derived factor-1 (SDF-1) genes were measured. ALP reached baseline level only in IL-MSCs, whereas OCN reached baseline level in MSCs recipients (IL-MSCs and IP-MSCs). BMP-2 significantly increased in MSCs recipients 3 weeks postfracture and increased in all groups 8 weeks postfracture with significant increases in MSC recipients than the fixation and PBS groups. The highest BMP-2 expression was reached in IL-MSC group. MSCs either locally or systemically improves or accelerates the healing of bone fractures with better results obtained after local injection, as shown by biochemical, radiological, and histological findings. MSCs are effective candidates for bone regeneration.
Collapse
Affiliation(s)
- Shuzan Ali Mohammed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Mahasen Abd Elsattar
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Somia Hassan Abd-Allah
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Omnia Youssif Habashy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Eman M A Abdelghany
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Samia Hussein
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Omnia Abdullah
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt
| |
Collapse
|
5
|
Gebara N, Correia Y, Wang K, Bussolati B. Angiogenic Properties of Placenta-Derived Extracellular Vesicles in Normal Pregnancy and in Preeclampsia. Int J Mol Sci 2021; 22:5402. [PMID: 34065595 PMCID: PMC8160914 DOI: 10.3390/ijms22105402] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is one of the main processes that coordinate the biological events leading to a successful pregnancy, and its imbalance characterizes several pregnancy-related diseases, including preeclampsia. Intracellular interactions via extracellular vesicles (EVs) contribute to pregnancy's physiology and pathophysiology, and to the fetal-maternal interaction. The present review outlines the implications of EV-mediated crosstalk in the angiogenic process in healthy pregnancy and its dysregulation in preeclampsia. In particular, the effect of EVs derived from gestational tissues in pro and anti-angiogenic processes in the physiological and pathological setting is described. Moreover, the application of EVs from placental stem cells in the clinical setting is reported.
Collapse
Affiliation(s)
- Natalia Gebara
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| | - Yolanda Correia
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK; (Y.C.); (K.W.)
| | - Keqing Wang
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK; (Y.C.); (K.W.)
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| |
Collapse
|
6
|
Priester C, MacDonald A, Dhar M, Bow A. Examining the Characteristics and Applications of Mesenchymal, Induced Pluripotent, and Embryonic Stem Cells for Tissue Engineering Approaches across the Germ Layers. Pharmaceuticals (Basel) 2020; 13:E344. [PMID: 33114710 PMCID: PMC7692540 DOI: 10.3390/ph13110344] [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: 08/14/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
The field of regenerative medicine utilizes a wide array of technologies and techniques for repairing and restoring function to damaged tissues. Among these, stem cells offer one of the most potent and promising biological tools to facilitate such goals. Implementation of mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs) offer varying advantages based on availability and efficacy in the target tissue. The focus of this review is to discuss characteristics of these three subset stem cell populations and examine their utility in tissue engineering. In particular, the development of therapeutics that utilize cell-based approaches, divided by germinal layer to further assess research targeting specific tissues of the mesoderm, ectoderm, and endoderm. The combinatorial application of MSCs, iPSCs, and ESCs with natural and synthetic scaffold technologies can enhance the reparative capacity and survival of implanted cells. Continued efforts to generate more standardized approaches for these cells may provide improved study-to-study variations on implementation, thereby increasing the clinical translatability of cell-based therapeutics. Coupling clinically translatable research with commercially oriented methods offers the potential to drastically advance medical treatments for multiple diseases and injuries, improving the quality of life for many individuals.
Collapse
Affiliation(s)
- Caitlin Priester
- Department of Animal Science, University of Tennessee, Knoxville, TN 37998, USA;
| | - Amber MacDonald
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Madhu Dhar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Austin Bow
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| |
Collapse
|
7
|
Menger MM, Laschke MW, Orth M, Pohlemann T, Menger MD, Histing T. Vascularization Strategies in the Prevention of Nonunion Formation. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:107-132. [PMID: 32635857 DOI: 10.1089/ten.teb.2020.0111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Delayed healing and nonunion formation are major challenges in orthopedic surgery, which require the development of novel treatment strategies. Vascularization is considered one of the major prerequisites for successful bone healing, providing an adequate nutrient supply and allowing the infiltration of progenitor cells to the fracture site. Hence, during the last decade, a considerable number of studies have focused on the evaluation of vascularization strategies to prevent or to treat nonunion formation. These involve (1) biophysical applications, (2) systemic pharmacological interventions, and (3) tissue engineering, including sophisticated scaffold materials, local growth factor delivery systems, cell-based techniques, and surgical vascularization approaches. Accumulating evidence indicates that in nonunions, these strategies are indeed capable of improving the process of bone healing. The major challenge for the future will now be the translation of these strategies into clinical practice to make them accessible for the majority of patients. If this succeeds, these vascularization strategies may markedly reduce the incidence of nonunion formation. Impact statement Delayed healing and nonunion formation are a major clinical problem in orthopedic surgery. This review provides an overview of vascularization strategies for the prevention and treatment of nonunions. The successful translation of these strategies in clinical practice is of major importance to achieve adequate bone healing.
Collapse
Affiliation(s)
- Maximilian M Menger
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Marcel Orth
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Tim Pohlemann
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Tina Histing
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
8
|
Role of biomechanics in vascularization of tissue-engineered bones. J Biomech 2020; 110:109920. [PMID: 32827778 DOI: 10.1016/j.jbiomech.2020.109920] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 12/23/2022]
Abstract
Biomaterial based reconstruction is still the most commonly employed method of small bone defect reconstruction. Bone tissue-engineered techniques are improving, and adjuncts such as vascularization technologies allow re-evaluation of traditional reconstructive methods for healingofcritical-sized bone defect. Slow infiltration rate of vasculogenesis after cell-seeded scaffold implantation limits the use of clinically relevant large-sized scaffolds. Hence, in vitro vascularization within the tissue-engineered bone before implantation is required to overcome the serious challenge of low cell survival rate after implantation which affects bone tissue regeneration and osseointegration. Mechanobiological interactions between cells and microvascular mechanics regulate biological processes regarding cell behavior. In addition, load-bearing scaffolds demand mechanical stability properties after vascularization to have adequate strength while implanted. With the advent of bioreactors, vascularization has been greatly improved by biomechanical regulation of stem cell differentiation through fluid-induced shear stress and synergizing osteogenic and angiogenic differentiation in multispecies coculture cells. The benefits of vascularization are clear: avoidance of mass transfer limitation and oxygen deprivation, a significant decrease in cell necrosis, and consequently bone development, regeneration and remodeling. Here, we discuss specific techniques to avoid pitfalls and optimize vascularization results of tissue-engineered bone. Cell source, scaffold modifications and bioreactor design, and technique specifics all play a critical role in this new, and rapidly growing method for bone defect reconstruction. Given the crucial importance of long-term survival of vascular network in physiological function of 3D engineered-bone constructs, greater knowledge of vascularization approaches may lead to the development of new strategies towards stabilization of formed vascular structure.
Collapse
|
9
|
Eljarrah A, Gergues M, Pobiarzyn PW, Sandiford OA, Rameshwar P. Therapeutic Potential of Mesenchymal Stem Cells in Immune-Mediated Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:93-108. [PMID: 31898783 DOI: 10.1007/978-3-030-31206-0_5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can self-renew and differentiate into cells of all germ layers. MSCs can be easily attracted to the site of tissue insult with high levels of inflammatory mediators. The general ability of MSCs to migrate at the sites of tissue injury suggested an innate ability for these cells to be involved in baseline tissue repair. The bone marrow is one of the primary sources of MSCs, though they can be ubiquitous. An attractive property of MSCs for clinical application is their ability to cross allogeneic barrier. However, alone, MSCs are not immune suppressive cells. Rather, they can be licensed by the tissue microenvironment to become immune suppressor cells. Immune suppressor functions of MSCs include those that blunt cytotoxicity of natural killer cells, suppression of T-cell proliferation, and "veto" function. MSCs, as third-party cells, suppress the immune response that generally recapitulates graft-versus-host disease (GvHD) responses. Based on the plastic functions of MSCs, these cells have dominated the field of cell-based therapies, such as anti-inflammatory and drug delivery. Here, we focus on the potential use of MSC for immunological disorders such as Crohn's disease and GvHD.
Collapse
Affiliation(s)
- Adam Eljarrah
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Rutgers School of Graduate School at New Jersey Medical School, Newark, NJ, USA
| | - Marina Gergues
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Rutgers School of Graduate School at New Jersey Medical School, Newark, NJ, USA
| | - Piotr W Pobiarzyn
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Rutgers School of Graduate School at New Jersey Medical School, Newark, NJ, USA
| | - Oleta A Sandiford
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Rutgers School of Graduate School at New Jersey Medical School, Newark, NJ, USA
| | - Pranela Rameshwar
- Department of Medicine - Division of Hematology/Oncology, New Jersey Medical School, Rutgers School of Biomedical Health Science, Newark, NJ, USA.
| |
Collapse
|
10
|
Genetically Engineered-MSC Therapies for Non-unions, Delayed Unions and Critical-size Bone Defects. Int J Mol Sci 2019; 20:ijms20143430. [PMID: 31336890 PMCID: PMC6678255 DOI: 10.3390/ijms20143430] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/21/2022] Open
Abstract
The normal bone regeneration process is a complex and coordinated series of events involving different cell types and molecules. However, this process is impaired in critical-size/large bone defects, with non-unions or delayed unions remaining a major clinical problem. Novel strategies are needed to aid the current therapeutic approaches. Mesenchymal stem/stromal cells (MSCs) are able to promote bone regeneration. Their beneficial effects can be improved by modulating the expression levels of specific genes with the purpose of stimulating MSC proliferation, osteogenic differentiation or their immunomodulatory capacity. In this context, the genetic engineering of MSCs is expected to further enhance their pro-regenerative properties and accelerate bone healing. Herein, we review the most promising molecular candidates (protein-coding and non-coding transcripts) and discuss the different methodologies to engineer and deliver MSCs, mainly focusing on in vivo animal studies. Considering the potential of the MSC secretome for bone repair, this topic has also been addressed. Furthermore, the promising results of clinical studies using MSC for bone regeneration are discussed. Finally, we debate the advantages and limitations of using MSCs, or genetically-engineered MSCs, and their potential as promoters of bone fracture regeneration/repair.
Collapse
|
11
|
Zhu H, Kaufman DS. An Improved Method to Produce Clinical-Scale Natural Killer Cells from Human Pluripotent Stem Cells. Methods Mol Biol 2019; 2048:107-119. [PMID: 31396935 DOI: 10.1007/978-1-4939-9728-2_12] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Human natural killer (NK) cell-based adoptive anticancer immunotherapy has gained intense interest with many clinical trials actively recruiting patients to treat a variety of both hematological malignancies and solid tumors. Most of these trials use primary NK cells isolated either from peripheral blood (PB-NK cells) or umbilical cord blood (UCB-NK cells), though these sources require NK cell collection for each patient leading to donor variability and heterogeneity in the NK cell populations. In contrast, NK cells derived human embryonic stem cells (hESC-NK cells) or induced pluripotent stem cells (hiPSC-NK cells) provide more homogeneous cell populations that can be grown at clinical scale, and genetically engineered if needed. These characteristics make hESC-/iPSC-derived NK cells an ideal cell population for developing standardized, "off-the-shelf" immunotherapy products. Additionally, production of NK cells from undifferentiated human pluripotent stem cells enables studies to better define pathways that regulate human NK cell development and function. Our group previously has established a stromal-free, two-stage culture system to derive NK cells from hESC/hiPSC in vitro followed by clinical-scale expansion of these cells using interleukin (IL)-21 expressing artificial antigen-presenting cells. However, prior to differentiation, this method requires single-cell adaptation of hESCs/hiPSCs which takes months. Recently we optimized this method by adapting the mouse embryonic fibroblast-dependent hESC/hiPSC to feeder-free culture conditions. These feeder-free hESCs/hiPSCs are directly used to form embryoid body (EB) to generate hemato-endothelial precursor cells. This new method produces mature, functional NK cells with higher efficiency to enable rapid production of an essentially unlimited number of homogenous NK cells that can be used for standardized, targeted immunotherapy for the treatment of refractory cancers and infectious diseases.
Collapse
Affiliation(s)
- Huang Zhu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, USA.
| | - Dan S Kaufman
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
12
|
Fliefel R, Ehrenfeld M, Otto S. Induced pluripotent stem cells (iPSCs) as a new source of bone in reconstructive surgery: A systematic review and meta-analysis of preclinical studies. J Tissue Eng Regen Med 2018; 12:1780-1797. [DOI: 10.1002/term.2697] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 04/16/2018] [Accepted: 05/03/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Riham Fliefel
- Experimental Surgery and Regenerative Medicine (ExperiMed), Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry; Alexandria University; Alexandria Egypt
| | - Michael Ehrenfeld
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
| | - Sven Otto
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
| |
Collapse
|
13
|
Wang HJ, Zhang Y, Kato S, Nakagawa K, Kimura F, Miyazawa T, Wang JY. HPLC-MS/MS: A potential method to track the in vivo degradation of zein-based biomaterial. J Biomed Mater Res A 2017; 106:606-613. [PMID: 28960906 DOI: 10.1002/jbm.a.36252] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/02/2017] [Accepted: 09/26/2017] [Indexed: 01/01/2023]
Abstract
Given the inadequacies of existing clinic tracking strategies, such as isotopic tracer techniques, one of the major thrusts in protein-based tissue engineering substitutes prior to use in clinic is to develop a safe technique that can effectively track their degradation in vivo. Keeping in view the possible application of a natural polymer, zein as a novel bone substitute, with the advantages of good bio-compatibility, bio-degradability and outstanding mechanical properties, we attempted here to construct a HPLC-MS/MS method to track the in vivo degradation of zein porous scaffold. Histological observation and immunohistochemistry analysis using the intramuscular implantation model of rats clearly indicated that zein porous scaffold has certain osteoinductive ability. More importantly, HPLC-MS/MS detected the changes of amino acids levels in plasma and different organs after the implantation of scaffolds. With these findings, it could be concluded that HPLC-MS/MS might be a potential method to track the in vivo degradation of protein-based tissue engineering substitutes. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 606-613, 2018.
Collapse
Affiliation(s)
- Hua-Jie Wang
- School of Biomedical Engineering, Shanghai Jiaotong University, Dongchuan Road, Shanghai, 200240, China.,Food Biotechnology Innovation Project, New Industry Creation Hatchery Center (NICHe) at Tohoku University, Sendai, 980-0845, Japan
| | - Yue Zhang
- School of Biomedical Engineering, Shanghai Jiaotong University, Dongchuan Road, Shanghai, 200240, China
| | - Shunji Kato
- Food and Biodynamic Chemistry Laboratory, School of Agriculture, Tohoku University, Sendai, 980-0845, Japan
| | - Kiyotaka Nakagawa
- Food and Biodynamic Chemistry Laboratory, School of Agriculture, Tohoku University, Sendai, 980-0845, Japan
| | - Fumiko Kimura
- Food and Biodynamic Chemistry Laboratory, School of Agriculture, Tohoku University, Sendai, 980-0845, Japan
| | - Teruo Miyazawa
- Food Biotechnology Innovation Project, New Industry Creation Hatchery Center (NICHe) at Tohoku University, Sendai, 980-0845, Japan.,Food and Biodynamic Chemistry Laboratory, School of Agriculture, Tohoku University, Sendai, 980-0845, Japan
| | - Jin-Ye Wang
- School of Biomedical Engineering, Shanghai Jiaotong University, Dongchuan Road, Shanghai, 200240, China
| |
Collapse
|
14
|
Csobonyeiova M, Polak S, Zamborsky R, Danisovic L. iPS cell technologies and their prospect for bone regeneration and disease modeling: A mini review. J Adv Res 2017; 8:321-327. [PMID: 28386481 PMCID: PMC5374850 DOI: 10.1016/j.jare.2017.02.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/24/2017] [Accepted: 02/25/2017] [Indexed: 12/27/2022] Open
Abstract
Bone disorders are a group of varied acute and chronic traumatic, degenerative, malignant or congenital conditions affecting the musculoskeletal system. They are prevalent in society and, with an ageing population, the incidence and impact on the population's health is growing. Severe persisting pain and limited mobility are the major symptoms of the disorder that impair the quality of life in affected patients. Current therapies only partially treat the disorders, offering management of symptoms, or temporary replacement with inert materials. However, during the last few years, the options for the treatment of bone disorders have greatly expanded, thanks to the advent of regenerative medicine. Skeletal cell-based regeneration medicine offers promising reparative therapies for patients. Mesenchymal stem (stromal) cells from different tissues have been gradually translated into clinical practice; however, there are a number of limitations. The introduction of reprogramming methods and the subsequent production of induced pluripotent stem cells provides a possibility to create human-specific models of bone disorders. Furthermore, human-induced pluripotent stem cell-based autologous transplantation is considered to be future breakthrough in the field of regenerative medicine. The main goal of the present paper is to review recent applications of induced pluripotent stem cells in bone disease modeling and to discuss possible future therapy options. The present article contributes to the dissemination of scientific and pre-clinical results between physicians, mainly orthopedist and thus supports the translation to clinical practice.
Collapse
Affiliation(s)
- Maria Csobonyeiova
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Stefan Polak
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Radoslav Zamborsky
- Department of Orthopaedics, Faculty of Medicine, Comenius University and Children’s University Hospital, Limbova 1, 831 01 Bratislava, Slovakia
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
- Regenmed Ltd., Medena 29, 811 02 Bratislava, Slovakia
| |
Collapse
|