1
|
Gorjipour F, Bohloolighashghaei S, Sotoudeheian M, Pazoki Toroudi H. Fetal adnexa-derived allogeneic mesenchymal stem cells for cardiac regeneration: the future trend of cell-based therapy for age-related adverse conditions. Hum Cell 2025; 38:61. [PMID: 39998714 DOI: 10.1007/s13577-025-01190-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025]
Abstract
Heart failure is known as the leading cause of mortality and morbidity in adults, not only in USA but worldwide. Since the world's population is aging, the burden of cardiovascular disorders is increasing. Mesenchymal stem/stromal cells (MSCs) from a patient's bone marrow or other tissues have been widely used as the primary source of stem cells for cellular cardiomyoplasty. The incongruencies that exist between various cell-therapy approaches for cardiac diseases could be attributed to variations in cell processing methods, quality of the process, and cell donors. Off-the-shelf preparations of MSCs, enabled by batch processing of the cells and controlled cell processing factories in regulated facilities, may offer opportunities to overcome these problems. In this study, for the first time, we focused on the fetal membranes and childbirth byproducts as a promising source of cells for regenerative medicine. While many studies have described the advantages of cells derived from these organs, their advantage as a source of younger cells has not been sufficiently covered by the literature. Thus, herein, we highlight challenges that may arise from the impairment of the regenerative capacity of MSCs due to donor age and how allograft cells from fetal adnexa can be a promising substitute for the aged patients' stem cells for myocardial regeneration. Moreover, obstacles to the use of off-the-shelf cell-therapy preparations in regenerative medicine are briefly summarized here.
Collapse
Affiliation(s)
- Fazel Gorjipour
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Hamidreza Pazoki Toroudi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Zhao J, Chen A, Wang R, Qiu D, Chen H, Li J, Zhang J, Wang T, Wang Y, Lin Y, Zhou J, Du Y, Yuan H, Zhang Y, Miao D, Wang Y, Jin J. Bmi-1 Epigenetically Orchestrates Osteogenic and Adipogenic Differentiation of Bone Marrow Mesenchymal Stem Cells to Delay Bone Aging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404518. [PMID: 39225325 PMCID: PMC11633582 DOI: 10.1002/advs.202404518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/05/2024] [Indexed: 09/04/2024]
Abstract
With the increase in the aging population, senile osteoporosis (SOP) has become a major global public health concern. Here, it is found that Prx1 and Bmi-1 co-localized in trabecular bone, bone marrow cavity, endosteum, and periosteum. Prx1-driven Bmi-1 knockout in bone-marrow mesenchymal stem cells (BMSCs) reduced bone mass and increased bone marrow adiposity by inhibiting osteoblastic bone formation, promoting osteoclastic bone resorption, downregulating the proliferation and osteogenic differentiation of BMSCs, and upregulating the adipogenic differentiation of BMSCs. However, Prx1-driven Bmi-1 overexpression showed a contrasting phenotype to Prx1-driven Bmi-1 knockout in BMSCs. Regarding mechanism, Bmi-1-RING1B bound to DNMT3A and promoted its ubiquitination and inhibited DNA methylation of Runx2 at the region from 45047012 to 45047313 bp, thus promoting the osteogenic differentiation of BMSCs. Moreover, Bmi-1-EZH2 repressed the transcription of Cebpa by promoting H3K27 trimethylation at the promoter region -1605 to -1596 bp, thus inhibiting the adipogenic differentiation of BMSCs. It is also found that Prx1-driven Bmi-1 overexpression rescued the SOP induced by Prx1-driven Bmi-1 knockout in BMSCs. Thus, Bmi-1 functioned as a hub protein in the epigenetic regulation of BMSCs differentiation to delay bone aging. The Prx1-driven Bmi-1 overexpression in BMSCs can be used as an approach for the translational therapy of SOP.
Collapse
Affiliation(s)
- Jingyu Zhao
- Department of Human AnatomyResearch Centre for Bone and Stem CellsSchool of Basic Medical SciencesKey Laboratory for Aging & DiseaseSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjingJiangsu211166China
| | - Ao Chen
- Department of Human AnatomyResearch Centre for Bone and Stem CellsSchool of Basic Medical SciencesKey Laboratory for Aging & DiseaseSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjingJiangsu211166China
| | - Rong Wang
- Department of Human AnatomyResearch Centre for Bone and Stem CellsSchool of Basic Medical SciencesKey Laboratory for Aging & DiseaseSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjingJiangsu211166China
| | - Dong Qiu
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityState Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral DiseasesJiangsu Province Engineering Research Centre of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsu210029China
| | - Haiyun Chen
- Department of Human AnatomyResearch Centre for Bone and Stem CellsSchool of Basic Medical SciencesKey Laboratory for Aging & DiseaseSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjingJiangsu211166China
| | - Jiyu Li
- Department of Human AnatomyResearch Centre for Bone and Stem CellsSchool of Basic Medical SciencesKey Laboratory for Aging & DiseaseSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjingJiangsu211166China
| | - Jin'ge Zhang
- Department of Human AnatomyResearch Centre for Bone and Stem CellsSchool of Basic Medical SciencesKey Laboratory for Aging & DiseaseSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjingJiangsu211166China
| | - Tianxiao Wang
- School of PharmacyNanjing Medical UniversityNanjingJiangsu211166China
| | - Yue Wang
- Department of Human AnatomyResearch Centre for Bone and Stem CellsSchool of Basic Medical SciencesKey Laboratory for Aging & DiseaseSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjingJiangsu211166China
| | - Yujie Lin
- Department of Human AnatomyResearch Centre for Bone and Stem CellsSchool of Basic Medical SciencesKey Laboratory for Aging & DiseaseSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjingJiangsu211166China
| | - Jiawen Zhou
- Department of Human AnatomyResearch Centre for Bone and Stem CellsSchool of Basic Medical SciencesKey Laboratory for Aging & DiseaseSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjingJiangsu211166China
| | - Yifei Du
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityState Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral DiseasesJiangsu Province Engineering Research Centre of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsu210029China
| | - Hua Yuan
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityState Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral DiseasesJiangsu Province Engineering Research Centre of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsu210029China
| | - Yongjie Zhang
- Department of Human AnatomyResearch Centre for Bone and Stem CellsSchool of Basic Medical SciencesKey Laboratory for Aging & DiseaseSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjingJiangsu211166China
| | - Dengshun Miao
- Department of Human AnatomyResearch Centre for Bone and Stem CellsSchool of Basic Medical SciencesKey Laboratory for Aging & DiseaseSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjingJiangsu211166China
| | - Yuli Wang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityState Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral DiseasesJiangsu Province Engineering Research Centre of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsu210029China
| | - Jianliang Jin
- Department of Human AnatomyResearch Centre for Bone and Stem CellsSchool of Basic Medical SciencesKey Laboratory for Aging & DiseaseSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjingJiangsu211166China
| |
Collapse
|
3
|
Hosoya A, Takebe H, Seki-Kishimoto Y, Noguchi Y, Ninomiya T, Yukita A, Yoshiba N, Washio A, Iijima M, Morotomi T, Kitamura C, Nakamura H. Polycomb protein Bmi1 promotes odontoblast differentiation by accelerating Wnt and BMP signaling pathways. Histochem Cell Biol 2024; 163:11. [PMID: 39589557 DOI: 10.1007/s00418-024-02337-2] [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] [Accepted: 09/29/2024] [Indexed: 11/27/2024]
Abstract
Bmi1 is a polycomb protein localized in stem cells and maintains their stemness. This protein is also reported to regulate the expression of various differentiation genes. In this study, to analyze the role of Bmi1 during dentinogenesis, we examined the immunohistochemical localization of Bmi1 during rat tooth development as well as after cavity preparation. Bmi1 localization was hardly detected in the dental mesenchyme at the bud and cap stages. After the bell stage, however, this protein became detectable in preodontoblasts and early odontoblasts just beginning dentin matrix secretion. As dentin formation progressed, Bmi1 immunoreactivity in the odontoblasts decreased in intensity. After cavity preparation, cells lining the dentin and some pulp cells under the cavity were immunopositive for Bmi1 at 4 days. Odontoblast-like cells forming reparative dentin were immunopositive for Bmi1 at 1 week, whereas their immunoreactivity was not detected after 8 weeks. We further analyzed the function of Bmi1 using KN-3 cells, a dental mesenchymal cell line. Overexpression of Bmi1 in KN-3 cells promoted mineralized tissue formation. In contrast, siRNA knockdown of Bmi1 in KN-3 cells reduced alkaline phosphatase activity and the expression of odontoblast differentiation marker genes such as Runx2, osterix, and osteocalcin. Additionally, KN-3 cells transfected with siRNA against Bmi1 showed reduced nuclear transition of β-catenin and expression of phosphorylated-Smad1/5/8. Taken together, these findings suggest that Bmi1 was localized in the odontoblast-lineage cells in their early differentiation stages. Bmi1 might positively regulate their differentiation by accelerating Wnt and BMP signaling pathways.
Collapse
Affiliation(s)
- Akihiro Hosoya
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan.
| | - Hiroaki Takebe
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan
| | - Yuri Seki-Kishimoto
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293, Japan
| | - Yukiko Noguchi
- Division of Orthodontics and Dentofacial Orthopedics, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| | - Tadashi Ninomiya
- Department of Anatomy, Nihon University School of Dentistry, Tokyo, Japan
| | - Akira Yukita
- Faculty of Education, Shizuoka University, Shizuoka, Japan
| | - Nagako Yoshiba
- Department of Oral Health Science, Course for Oral Life Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ayako Washio
- Division of Endodontics and Restorative Dentistry, Department of Oral Functions, Kyushu Dental University, Fukuoka, Japan
| | - Masahiro Iijima
- Division of Orthodontics and Dentofacial Orthopedics, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| | - Takahiko Morotomi
- Department of Endodontics, School of Dentistry, Aichi Gakuin University, Aichi, Japan
| | - Chiaki Kitamura
- Division of Endodontics and Restorative Dentistry, Department of Oral Functions, Kyushu Dental University, Fukuoka, Japan
| | - Hiroaki Nakamura
- Institute for Oral Science, Matsumoto Dental University, Nagano, Japan
| |
Collapse
|
4
|
Yang C, Qiao W, Xue Q, Goltzman D, Miao D, Dong Z. The senolytic agent ABT263 ameliorates osteoporosis caused by active vitamin D insufficiency through selective clearance of senescent skeletal cells. J Orthop Translat 2024; 49:107-118. [PMID: 39430127 PMCID: PMC11490840 DOI: 10.1016/j.jot.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/05/2024] [Accepted: 08/16/2024] [Indexed: 10/22/2024] Open
Abstract
Background/Objective Active vitamin D insufficiency accelerates the development of osteoporosis, with senescent bone cells and the senescence-associated secretory phenotype (SASP) playing crucial roles. This study aimed to investigate whether the senolytic agent ABT263 could correct osteoporosis caused by active vitamin D insufficiency by selectively clearing senescent cells. Methods Bone marrow mesenchymal stem cells (BM-MSCs) from young and aged mice were treated with ABT263 in vitro, and 1,25(OH)2D-insufficient (Cyp27b1+/-) mice were administered ABT263 in vivo. Cellular, molecular, imaging, and histopathological analyses were performed to compare treated cells and mice with control groups. Results ABT263 induced apoptosis in senescent BM-MSCs by downregulating Bcl2 and upregulating Bax expression. It also induced apoptosis in senescent BM-MSCs from 1,25(OH)2D-insufficient mice. ABT263 administration corrected bone loss caused by 1,25(OH)2D insufficiency by increasing bone density, bone volume, trabecular number, trabecular thickness, and collagen synthesis. It also enhanced osteoblastic bone formation and reduced osteoclastic bone resorption in vivo. ABT263 treatment corrected the impaired osteogenic action of BM-MSCs by promoting their proliferation and osteogenic differentiation. Furthermore, it corrected oxidative stress and DNA damage caused by 1,25(OH)2D insufficiency by increasing SOD-2 and decreasing γ-H2A.X expression. Finally, ABT263 corrected bone cell senescence and SASP caused by 1,25(OH)2D insufficiency by reducing the expression of senescence and SASP-related genes and proteins. Conclusion ABT263 can correct osteoporosis caused by active vitamin D insufficiency by selectively clearing senescent skeletal cells, reducing oxidative stress, DNA damage, and SASP, and promoting bone formation while inhibiting bone resorption. These findings provide new insights into the potential therapeutic application of senolytic agents in the treatment of osteoporosis associated with active vitamin D insufficiency. The translational potential of this article This study highlights the therapeutic potential of ABT263, a senolytic compound, in treating osteoporosis caused by active vitamin D insufficiency. By selectively eliminating senescent bone cells and their associated SASP, ABT263 intervention demonstrates the ability to restore bone homeostasis, prevent further bone loss, and promote bone formation. These findings contribute to the growing body of research supporting the use of senolytic therapies for the prevention and treatment of age-related bone disorders. The translational potential of this study lies in the development of novel therapeutic strategies targeting cellular senescence to combat osteoporosis, particularly in cases where vitamin D insufficiency is a contributing factor. Further clinical studies are warranted to validate the efficacy and safety of ABT263 and other senolytic agents in the treatment of osteoporosis in humans.
Collapse
Affiliation(s)
- Cuicui Yang
- The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Wanxin Qiao
- The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Qi Xue
- The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - David Goltzman
- Calcium Research Laboratory, McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, H4A 3J1, Canada
| | - Dengshun Miao
- The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Zhan Dong
- Department of Orthopedics, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Zhang XF, Wang ZX, Zhang BW, Huang KP, Ren TX, Wang T, Cheng X, Hu P, Xu WH, Li J, Zhang JX, Wang H. TGF-β1-triggered BMI1 and SMAD2 cooperatively regulate miR-191 to modulate bone formation. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102164. [PMID: 38549914 PMCID: PMC10973191 DOI: 10.1016/j.omtn.2024.102164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 03/04/2024] [Indexed: 08/09/2024]
Abstract
Transforming growth factor β 1 (TGF-β1), as the most abundant signaling molecule in bone matrix, is essential for bone homeostasis. However, the signaling transduction of TGF-β1 in the bone-forming microenvironment remains unknown. Here, we showed that microRNA-191 (miR-191) was downregulated during osteogenesis and further decreased by osteo-favoring TGF-β1 in bone marrow mesenchymal stem cells (BMSCs). MiR-191 was lower in bone tissues from children than in those from middle-aged individuals and it was negatively correlated with collagen type I alpha 1 chain (COL1A1). MiR-191 depletion significantly increased osteogenesis and bone formation in vivo. Hydrogels embedded with miR-191-low BMSCs displayed a powerful bone repair effect. Mechanistically, transcription factors BMI1 and SMAD2 coordinately controlled miR-191 level. In detail, BMI1 and pSMAD2 were both upregulated by TGF-β1 under osteogenic condition. SMAD2 activated miR-191 transcription, while BMI1 competed with SMAD2 for binding to miR-191 promoter region, thus disturbing the activation of SMAD2 on miR-191 and reducing miR-191 level. Altogether, our findings reveal that miR-191 regulated by TGF-β1-induced BMI1 and SMAD2 negatively modulated bone formation and regeneration, and inhibition of miR-191 might be therapeutically useful to enhance bone repair in clinic.
Collapse
Affiliation(s)
- Xiao-Fei Zhang
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Zi-Xuan Wang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Bo-Wen Zhang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Kun-Peng Huang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Tian-Xing Ren
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Ting Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xing Cheng
- Health Care Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Ping Hu
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Wei-Hua Xu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Jin Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Jin-Xiang Zhang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| |
Collapse
|
6
|
Liu B, Zhang J, Zhang J, Ji X, Wang R, Gong A, Miao D. Metformin prevents mandibular bone loss in a mouse model of accelerated aging by correcting dysregulated AMPK-mTOR signaling and osteoclast differentiation. J Orthop Translat 2024; 46:129-142. [PMID: 38867742 PMCID: PMC11167204 DOI: 10.1016/j.jot.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/02/2024] [Accepted: 03/08/2024] [Indexed: 06/14/2024] Open
Abstract
Background Age-related mandibular osteoporosis frequently causes loose teeth, difficulty eating, and disfiguration in elders. Bmi1-/- mice displaying accelerated skeletal aging represent a useful model for testing interventions against premature jaw bone loss. As an anti-aging agent, metformin may ameliorate molecular dysfunction driving osteoporosis pathogenesis. We explored the mechanisms of mandibular osteopenia in Bmi1-/- mice and prevention by metformin treatment. Methods Three mouse groups were utilized: wild-type controls, untreated Bmi1-/-, and Bmi1-/- receiving 1 g/kg metformin diet. Mandibular bone phenotype was assessed by X-ray, micro-CT, histology, and immunohistochemistry. AMPK-mTOR pathway analysis, senescence markers, osteoblast and osteoclast gene expression were evaluated in jaw tissue. Osteoclast differentiation capacity and associated signaling molecules were examined in cultured Bmi1-/- bone marrow mononuclear cells ± metformin. Results Bmi1 loss reduced mandible bone density concomitant with decreased AMPK activity, increased mTOR signaling and cellular senescence in jaw tissue versus wild-type controls. This was accompanied by impaired osteoblast function and upregulated osteoclastogenesis markers. Metformin administration normalized AMPK-mTOR balance, oxidative stress and senescence signaling to significantly improve mandibular bone architecture in Bmi1-/- mice. In culture, metformin attenuated excessive osteoclast differentiation from Bmi1-/- marrow precursors by correcting dysregulated AMPK-mTOR-p53 pathway activity and suppressing novel pro-osteoclastogenic factor Stfa1. Conclusions Our study newly demonstrates metformin prevents accelerated jaw bone loss in a premature aging murine model by rectifying molecular dysfunction in cellular energy sensors, redox state, senescence and osteoclastogenesis pathways. Targeting such age-associated mechanisms contributing to osteoporosis pathogenesis may help maintain oral health and aesthetics in the growing elderly population. Translational potential The pronounced mandibular osteopenia exhibited in Bmi1-/- mice represents an accelerated model of jaw bone deterioration observed during human aging. Our finding that metformin preserves mandibular bone integrity in this progeroid model has important clinical implications. As an inexpensive oral medication already widely used to manage diabetes, metformin holds translational promise for mitigating age-related osteoporosis. The mandible is essential for chewing, swallowing, speech and facial structure, but progressively loses bone mass and strength with advancing age, significantly impacting seniors' nutrition, physical function and self-image. Our results suggest metformin's ability to rectify cellular energy imbalance, oxidative stress and osteoclast overactivity may help maintain jaw bone health into old age. Further research is still needed given metformin's multifaceted biology and bone regulation by diverse pathways. However, this preclinical study provides a strong rationale for clinical trials specifically examining mandibular outcomes in elderly subjects receiving standard metformin treatment for diabetes or prediabetes. Determining if metformin supplementation can prevent or delay oral disability and disfigurement from senescent jaw bone loss in the growing aged population represents an important public health priority. In summary, our mechanistic findings in a genetic mouse model indicate metformin merits investigation in rigorous human studies for alleviating morbidity associated with age-related mandibular osteoporosis.
Collapse
Affiliation(s)
- Boyang Liu
- Department of Human Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, PR China
| | - Jiao Zhang
- Department of Plastic Surgery, Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, PR China
| | - Jinge Zhang
- Department of Human Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, PR China
| | - Xiaolei Ji
- Department of Stomatology, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, PR China
| | - Rong Wang
- Department of Human Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, PR China
| | - Aixiu Gong
- Department of Stomatology, Children's Hospital of Nanjing Medical University, Nanjing, PR China
| | - Dengshun Miao
- Department of Human Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, PR China
| |
Collapse
|
7
|
Wu JZ, Zhou C, Liu S, Zhang JX, Yang W, Shi HB, Zhou WZ. TGF-β1 inhibitor P144 protects against benign restenosis after esophageal stenting through TGF-β1/Smads signaling pathway inhibition. Arab J Gastroenterol 2024; 25:214-222. [PMID: 38369402 DOI: 10.1016/j.ajg.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/26/2023] [Accepted: 02/10/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND AND STUDY AIMS Esophageal restenosis is a serious complication after esophageal stent placement, which influences the clinical prognosis of stent implantation and the patient's quality of life. TGF-β1/Smads signaling pathway plays an important role in the development of the eosinophilic esophagitis and scar repair after skin trauma. However, the role of TGF-β1/Smads in the development of esophageal restenosis after esophageal stent placement remains unknown. Our study aimed to investigate whether TGF-β1/Smads plays an important role in the development of esophageal restenosis after esophageal stent, and whether the exogenous TGF-β1 inhibitor supplement could ameliorate the esophageal restenosis after esophageal stent. MATERIAL AND METHODS We established the model of esophageal restenosis after esophageal stenting in rats, and determined the expression levels of TGF-β1/Smads signaling pathway and the relevant markers of fibroblast activation by immunochemistry (IHC), Western Blot and real time qPCR. Those all the indicators were also determined in esophageal fibroblast when exposed to rhTGF-β1 with or without TGF-β1 inhibitor P144. RESULTS The serum level of IL-1β and TNFα were significantly increased in stent implantation group compared to blank control group, and obviously ameliorated when treated with P144. The TGF-β1/Smads signaling pathway and the relevant markers of fibroblast activation were significantly increased in stent implantation group compared to blank control group, and obviously ameliorated when treated with P144. Those all the indicators were significantly increased when exposed to rhTGF-β1, and obviously decreased when treated with P144. CONCLUSIONS TGF-β1 Inhibitor P144 could protect against benign restenosis after esophageal stenting by down-regulating the expression levels of relevant markers of fibroblast activation through TGF-β1/Smads signaling pathway inhibition, and may be used as a novel therapy for benign restenosis after esophageal stenting.
Collapse
Affiliation(s)
- Jun-Zheng Wu
- Department of Interventional Radiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, 321 Zhongshan Road, Gulou District, Nanjing 210029, China
| | - Chun Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Gulou District, Nanjing 210029, China
| | - Sheng Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Gulou District, Nanjing 210029, China
| | - Jin-Xing Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Gulou District, Nanjing 210029, China
| | - Wei Yang
- Department of Interventional Radiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Gulou District, Nanjing 210029, China
| | - Hai-Bin Shi
- Department of Interventional Radiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Gulou District, Nanjing 210029, China
| | - Wei-Zhong Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Gulou District, Nanjing 210029, China.
| |
Collapse
|
8
|
Agh F, Mousavi SH, Aryaeian N, Amiri F, Jalilvand MR, Hasani M, Vahid F, Sepahvand F, Vosugh M. Senescence of bone marrow mesenchymal stem cells in Wistar male rats receiving normal chow/high-calorie diets with/without vitamin D. Biogerontology 2023; 24:801-812. [PMID: 37606875 DOI: 10.1007/s10522-023-10048-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/23/2023] [Indexed: 08/23/2023]
Abstract
Bone marrow mesenchymal stem cells (BM-MSCs) have a momentous function in the composition of the bone marrow microenvironment because of their many valuable properties and abilities, such as immunomodulation and hematopoiesis. The features and actions of MSCs are influenced by senescence, which may be affected by various factors such as nutritional/micronutrients status, e.g., vitamin D. This study aimed to examine the effects of a high-calorie diet (HCD) with/without vitamin D on BM-MSCs senescence. In the first phase, 48 middle-aged rats were fed a normal chow diet (NCD, n = 24) and an HCD (n = 24) for 26 weeks. Afterward, the rats in each group were randomly divided into three equal subgroups. Immediately, eight-rat from each diet group were sacrificed to assess the HCD effects on the first phase measurements. In the second phase, the remaining 4 groups of rats were fed either NCD or HCD with (6 IU/g) or without vitamin D (standard intake: 1 IU/g); in other words, in this phase, the animals were fed (a) NCD, (b) NCD plus vitamin D, (c) HCD, and (d) HCD plus vitamin D for 4 months. BM-MSCs were isolated and evaluated for P16INK4a, P38 MAPK, and Bmi-1 gene expression, reactive oxygen species (ROS) levels, SA-β-gal activity, and cell cycle profile at the end of both phases. After 26 weeks (first phase), the ROS level, SA-β-gal-positive cells, and cells in the G1 phase were significantly higher in HCD-fed rats than in NCD-fed ones (P < 0.05). HCD prescription did not significantly affect cells in the S and G2 phases (p > 0.05). Compared with the NCD-fed animals, P16INK4a and P38 MAPK gene expression were up-regulated in the HCD-fed animals; also, Bmi-1 gene expression was down-regulated (P < 0.05). BM-MSCs from vitamin D-treated rats (second phase) exhibited reduced mRNA levels of P16INK4a and P38 MAPK genes and increased Bmi-1 mRNA levels (all P < 0.05). Vitamin D prescription also declined the percentage of SA-β-gal-positive cells, ROS levels, and the cells in the G1 phase and increased the cells in the S phase in both NCD and HCD-fed animals (P < 0.05). The reduction of the cells in the G2 phase in rats fed with an NCD plus vitamin D was statistically non-significant (P = 0.128) and significant in HCD plus vitamin D rats (P = 0.002). HCD accelerates BM-MSCs senescence, and vitamin D reduces BM-MSCs senescence biomarkers.
Collapse
Affiliation(s)
- Fahimeh Agh
- Saveh University of Medical Sciences, Saveh, Iran
- Student Research Committee, Saveh University of Medical Sciences, Saveh, Iran
| | - Seyed Hadi Mousavi
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| | - Naheed Aryaeian
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| | - Fatemehsadat Amiri
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Jalilvand
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Motahareh Hasani
- Department of Nutrition, School of Public Health, Golestan University of Medical Sciences, Gorgan, Iran
| | - Farhad Vahid
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Fatemeh Sepahvand
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
9
|
Chen H, Cai G, Ruan X, Lu Y, Li G, Chen Z, Guan Z, Zhang H, Sun W, Wang H. Bone-targeted bortezomib increases bone formation within Calvarial trans-sutural distraction osteogenesis. Bone 2023; 169:116677. [PMID: 36646264 DOI: 10.1016/j.bone.2023.116677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 01/01/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023]
Abstract
The high rate of relapse in craniofacial disharmony treatment via trans-sutural distraction osteogenesis (TSDO) is due to the failure to form a stable bone bridge in the suture gap. Bisphosphonates (BP) have a high propensity to localize to hydroxyapatite in the bone matrix and are commonly used as targeting ligands for local delivery of therapeutics into bone microenvironment. Bone-targeted Bortezomib (BP-Btz) is chemosynthetic by linking Btz (Bortezomib) to a BP residue and could target bone tissue to promote osteoblast differentiation and inhibit osteoclastogenesis. Here, suture-derived mesenchymal stem cells (SuSCs) and osteoclasts were treated with Btz and BP-Btz. Aforesaid drugs were injected locally into the sagittal sutures to explore their effects in TSDO. Further, pharmacological properties of BP-Btz in the suture expansion model were assessed by fluorescent BP analogs and levels of total ubiquitinated (Ub)-proteins. The results showed that BP-Btz could stimulate osteogenic differentiation of SuSCs, bind to bone matrix and inhibit osteoclastogenesis. Biological effects of BP-Btz were similar with those of Btz in osteoblast differentiation and osteoclastogenesis inhibition in vitro. Activated bone metabolism were detected after 14 days in the sagittal suture expansion model. Increased osteoid area, remarkably decreased osteoclast surface and enhanced osteogenesis were detected in vivo after treatment with BP-Btz. Green fluorescence signal detection and pharmacodynamic studies revealed that BP-Btz bound to suture edge, released Btz in remodeling conditions, had a higher local concentration and sustained longer than free Btz. This study delineated the clinical potential of bone-targeted Btz conjugate as an efficacious strategy to promote trans-sutural distraction osteogenesis.
Collapse
Affiliation(s)
- Hongyu Chen
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Guanhui Cai
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Xiaolei Ruan
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yahui Lu
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Gen Li
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zhenwei Chen
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zhaolan Guan
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Hengwei Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Wen Sun
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| | - Hua Wang
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
10
|
Peng M, Wu J, Wang W, Liao T, Xu S, Xiao D, He Z, Yang X. Alpha-tocopherol enhances spermatogonial stem cell proliferation and restores mouse spermatogenesis by up-regulating BMI1. Front Nutr 2023; 10:1141964. [PMID: 37139440 PMCID: PMC10150882 DOI: 10.3389/fnut.2023.1141964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/31/2023] [Indexed: 05/05/2023] Open
Abstract
Purpose Spermatogonial stem cells (SSCs) are essential for maintaining reproductive function in males. B-lymphoma Mo-MLV insertion region 1 (BMI1) is a vital transcription repressor that regulates cell proliferation and differentiation. However, little is known about the role of BMI1 in mediating the fate of mammalian SSCs and in male reproduction. This study investigated whether BMI1 is essential for male reproduction and the role of alpha-tocopherol (α-tocopherol), a protective agent for male fertility, as a modulator of BMI1 both in vitro and in vivo. Methods Methyl thiazolyl tetrazolium (MTT) and 5-ethynyl-2'-deoxyuridine (EDU) assays were used to assess the effect of BMI1 on the proliferative ability of the mouse SSC line C18-4. Real-time polymerase chain reaction (PCR), western blotting, and immunofluorescence were applied to investigate changes in the mRNA and protein expression levels of BMI1. Male mice were used to investigate the effect of α-tocopherol and a BMI1 inhibitor on reproduction-associated functionality in vivo. Results Analysis revealed that BMI1 was expressed at high levels in testicular tissues and spermatogonia in mice. The silencing of BMI1 inhibited the proliferation of SSCs and DNA synthesis and enhanced the levels of γ-H2AX. α-tocopherol enhanced the proliferation and DNA synthesis of C18-4 cells, and increased the levels of BMI1. Notably, α-tocopherol rescued the inhibition of cell proliferation and DNA damage in C18-4 cells caused by the silencing of BMI1. Furthermore, α-tocopherol restored sperm count (Ctrl vs. PTC-209, p = 0.0034; Ctrl vs. PTC-209 + α-tocopherol, p = 0.7293) and normalized sperm malformation such as broken heads, irregular heads, lost and curled tails in vivo, as demonstrated by its antagonism with the BMI1 inhibitor PTC-209. Conclusion Analysis demonstrated that α-tocopherol is a potent in vitro and in vivo modulator of BMI1, a transcription factor that plays an important role in in SSC proliferation and spermatogenesis. Our findings identify a new target and strategy for treating male infertility that deserves further pre-clinical investigation.
Collapse
|
11
|
Grunbaum A, Kremer R. Parathyroid hormone-related protein (PTHrP) and malignancy. VITAMINS AND HORMONES 2022; 120:133-177. [PMID: 35953108 DOI: 10.1016/bs.vh.2022.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
PTHrP (parathyroid hormone related protein) is an important mediator of malignancy-related tumor progression and hypercalcemia that shares considerable homology and functionality with parathyroid hormone. In this chapter, we review what has been elucidated to date regarding PTHrP's role in malignancies. Starting with a review of calcium metabolism and regulation, we then summarize the discovery and structure of PTHrP and development of sensitive immunoassays for specific measurement. Subsequently, we explore its role in tumor progression, with emphasis on the primary tumor as well as skeletal and non-osseus metastases. We then consider the clinical implications of PTHrP in cancer before concluding with a discussion of both established and potential treatments for malignancy associated hypercalcemia and bone metastases.
Collapse
Affiliation(s)
- Ami Grunbaum
- Calcium Research Laboratories and Department of Medicine, McGill University and McGill University Health Centre, Montreal, QC, Canada
| | - Richard Kremer
- Calcium Research Laboratories and Department of Medicine, McGill University and McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
12
|
Wagner KD, Wagner N. The Senescence Markers p16INK4A, p14ARF/p19ARF, and p21 in Organ Development and Homeostasis. Cells 2022; 11:cells11121966. [PMID: 35741095 PMCID: PMC9221567 DOI: 10.3390/cells11121966] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023] Open
Abstract
It is widely accepted that senescent cells accumulate with aging. They are characterized by replicative arrest and the release of a myriad of factors commonly called the senescence-associated secretory phenotype. Despite the replicative cell cycle arrest, these cells are metabolically active and functional. The release of SASP factors is mostly thought to cause tissue dysfunction and to induce senescence in surrounding cells. As major markers for aging and senescence, p16INK4, p14ARF/p19ARF, and p21 are established. Importantly, senescence is also implicated in development, cancer, and tissue homeostasis. While many markers of senescence have been identified, none are able to unambiguously identify all senescent cells. However, increased levels of the cyclin-dependent kinase inhibitors p16INK4A and p21 are often used to identify cells with senescence-associated phenotypes. We review here the knowledge of senescence, p16INK4A, p14ARF/p19ARF, and p21 in embryonic and postnatal development and potential functions in pathophysiology and homeostasis. The establishment of senolytic therapies with the ultimate goal to improve healthy aging requires care and detailed knowledge about the involvement of senescence and senescence-associated proteins in developmental processes and homeostatic mechanism. The review contributes to these topics, summarizes open questions, and provides some directions for future research.
Collapse
|
13
|
Li J, Camirand A, Zakikhani M, Sellin K, Guo Y, Luan X, Mihalcioiu C, Kremer R. Parathyroid Hormone-Related Protein Inhibition Blocks Triple-Negative Breast Cancer Expansion in Bone Through Epithelial to Mesenchymal Transition Reversal. JBMR Plus 2022; 6:e10587. [PMID: 35720668 PMCID: PMC9189913 DOI: 10.1002/jbm4.10587] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 11/10/2022] Open
Abstract
Parathyroid hormone-related protein (PTHrP) plays a major role in skeletal metastasis but its action mechanism has not been fully defined. We previously demonstrated the crucial importance of PTHrP in promoting mammary tumor initiation, growth, and metastasis in a mouse model with a mammary epithelium-targeted Pthlh gene ablation. We demonstrate here a novel mechanism for bone invasion involving PTHrP induction of epithelial to mesenchymal transition (EMT) and cancer stem cells (CSCs) regulation. Clustered regularly interspaced short palindromic repeats (CRISPR)-mediated Pthlh gene ablation was used to study EMT markers, phenotype, and invasiveness in two triple-negative breast cancer (TNBC) cell types (established MDA-MB-231 and patient-derived PT-TNBC cells). In vitro, Pthlh ablation in TNBC cells reduced EMT markers, mammosphere-forming ability, and CD44high/CD24low cells ratio. In vivo, cells were injected intratibially into athymic nude mice, and therapeutic treatment with our anti-PTHrP blocking antibody was started 2 weeks after skeletal tumors were established. In vivo, compared to control, lytic bone lesion from Pthlh -ablated cells decreased significantly over 2 weeks by 27% for MDA-MB-231 and by 75% for PT-TNBC-injected mice (p < 0.001). Micro-CT (μCT) analyses also showed that antibody therapy reduced bone lytic volume loss by 52% and 48% for non-ablated MDA-MB-231 and PT-TNBC, respectively (p < 0.05). Antibody therapy reduced skeletal tumor burden by 45% and 87% for non-ablated MDA-MB-231 and PT-TNBC, respectively (p < 0.002) and caused a significant decrease of CSC/EMT markers ALDH1, vimentin, and Slug, and an increase in E-cadherin in bone lesions. We conclude that PTHrP is a targetable EMT molecular driver and suggest that its pharmacological blockade can provide a potential therapeutic approach against established TNBC-derived skeletal lesions. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jiarong Li
- Centre for Translational BiologyMcGill University Health CentreMontréalQCCanada
| | - Anne Camirand
- Centre for Translational BiologyMcGill University Health CentreMontréalQCCanada
| | - Mahvash Zakikhani
- Centre for Translational BiologyMcGill University Health CentreMontréalQCCanada
| | - Karine Sellin
- Centre for Translational BiologyMcGill University Health CentreMontréalQCCanada
| | - Yubo Guo
- Centre for Translational BiologyMcGill University Health CentreMontréalQCCanada
- Third Affiliated HospitalBeijing University of Chinese MedicineBeijingChina
| | - XiaoRui Luan
- Centre for Translational BiologyMcGill University Health CentreMontréalQCCanada
- Department of Genetics, School of MedicineZhejiang UniversityHangzhouChina
| | - Catalin Mihalcioiu
- Centre for Translational BiologyMcGill University Health CentreMontréalQCCanada
| | - Richard Kremer
- Centre for Translational BiologyMcGill University Health CentreMontréalQCCanada
| |
Collapse
|
14
|
Chen A, Li X, Zhao J, Zhou J, Xie C, Chen H, Wang Q, Wang R, Miao D, Li J, Jin J. Chronic alcohol reduces bone mass through inhibiting proliferation and promoting aging of endothelial cells in type-H vessels. Stem Cells Dev 2022; 31:541-554. [DOI: 10.1089/scd.2021.0337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Ao Chen
- Nanjing Medical University, 12461, Research Centre for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing, Jiangsu, China
| | - Xiaoting Li
- Nanjing Medical University, 12461, Department of Nutrition and Food Safety, School of Public Health, Nanjing, Jiangsu, China
| | - Jingyu Zhao
- Nanjing Medical University, 12461, Research Centre for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing, Jiangsu, China
| | - Jiawen Zhou
- Nanjing Medical University, 12461, Research Centre for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing, Jiangsu, China
| | - Chunfeng Xie
- Nanjing Medical University, 12461, Department of Nutrition and Food Safety, School of Public Health, Nanjing, Jiangsu, China
| | - Haiyun Chen
- Nanjing Medical University, 12461, Anti-aging Research Laboratory, Friendship Plastic Surgery Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiuyi Wang
- Nanjing Medical University, 12461, Research Centre for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing, Jiangsu, China
| | - Rong Wang
- Nanjing Medical University, 12461, Research Centre for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing, Jiangsu, China
| | - Dengshun Miao
- Nanjing Medical University, Nanjing, Jiangsu, China, 210029, ,
| | - Jie Li
- Xuzhou Medical University, 38044, Department of Orthopaedics, Xuzhou Central Hospital; The Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jianliang Jin
- Nanjing Medical University, 12461, Nanjing, China, 211166
- No.101,Longmian Avenue,Jiangning DistrictChina
| |
Collapse
|
15
|
Chen H, Zhou J, Chen H, Liang J, Xie C, Gu X, Wang R, Mao Z, Zhang Y, Li Q, Zuo G, Miao D, Jin J. Bmi-1-RING1B prevents GATA4-dependent senescence-associated pathological cardiac hypertrophy by promoting autophagic degradation of GATA4. Clin Transl Med 2022; 12:e574. [PMID: 35390228 PMCID: PMC8989148 DOI: 10.1002/ctm2.574] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 01/05/2023] Open
Abstract
AIMS Senescence-associated pathological cardiac hypertrophy (SA-PCH) is associated with upregulation of foetal genes, fibrosis, senescence-associated secretory phenotype (SASP), cardiac dysfunction and increased morbidity and mortality. Therefore, we conducted experiments to investigate whether GATA4 accumulation induces SA-PCH, and whether Bmi-1-RING1B promotes GATA4 ubiquitination and its selective autophagic degradation to prevent SA-PCH. METHODS AND RESULTS Bmi-1-deficient (Bmi-1-/- ), transgenic Bmi-1 overexpressing (Bmi-1Tg ) and wild-type (WT) mice were infused with angiotensin II (Ang II) to stimulate the development of SA-PCH. Through bioinformatics analysis with RNA sequencing data from cardiac tissues, we found that Bmi-1-RING1B and autophagy are negatively related to SA-PCH. Bmi-1 deficiency promoted GATA4-dependent SA-PCH by increasing GATA4 protein and hypertrophy-related molecules transcribed by GATA4 such as ANP and BNP. Bmi-1 deficiency stimulated NF-κB-p65-dependent SASP, leading to cardiac dysfunction, cardiomyocyte hypertrophy and senescence. Bmi-1 overexpression repressed GATA4-dependent SA-PCH. GATA4 degraded by Bmi-1 was mainly dependent on autophagy rather than proteasome. In human myocardium, p16 positively correlated with ANP and GATA4 and negatively correlated with LC3B, Bmi-1 and RING1B; GATA4 positively correlated with p62 and negatively correlated with Bmi-1 and LC3B. With increased p16 protein levels, ANP-, BNP- and GATA4-positive cells or areas increased; however, LC3B-positive cells or areas decreased in human myocardium. GATA4 is ubiquitinated after combining with Bmi-1-RING1B, which is then recognised by p62, is translocated to autophagosomes to form autophagolysosomes and degraded. Downregulated GATA4 ameliorated SA-PCH and cardiac dysfunction by reducing GATA4-dependent hypertrophy and SASP-related molecules. Bmi-1 combined with RING1B (residues 1-179) and C-terminus of GATA4 (residues 206-443 including zinc finger domains) through residues 1-95, including a RING-HC-finger. RING1B combined with C-terminus of GATA4 through the C-terminus (residues 180-336). Adeno-associated viral vector serotype 9 (AAV9)-cytomegalovirus (CMV)-Bmi-1-RING1B treatment significantly attenuated GATA4-dependent SA-PCH through promoting GATA4 autophagic degradation. CONCLUSIONS Bmi-1-RING1B maintained cardiac function and prevented SA-PCH by promoting selective autophagy for degrading GATA4. TRANSLATIONAL PERSPECTIVE AAV9-CMV-Bmi-1-RING1B could be used for translational gene therapy to ubiquitinate GATA4 and prevent GATA4-dependent SA-PCH. Also, the combined domains between Bmi-1-RING1B and GATA4 in aging cardiomyocytes could be therapeutic targets for identifying stapled peptides in clinical applications to promote the combination of Bmi-1-RING1B with GATA4 and the ubiquitination of GATA4 to prevent SA-PCH and heart failure. We found that degradation of cardiac GATA4 by Bmi-1 was mainly dependent on autophagy rather than proteasome, and autophagy agonists metformin and rapamycin could ameliorate the SA-PCH, suggesting that activation of autophagy with metformin or rapamycin could also be a promising method to prevent SA-PCH.
Collapse
Affiliation(s)
- Haiyun Chen
- The Research Center for AgingAffiliated Friendship Plastic Surgery Hospital of Nanjing Medical UniversityNanjingJiangsu210029China
| | - Jiawen Zhou
- Department of Human AnatomyResearch Centre for Bone and Stem CellsKey Laboratory for Aging & DiseaseThe State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingJiangsu211166China
| | - Hongjie Chen
- Department of Human AnatomyResearch Centre for Bone and Stem CellsKey Laboratory for Aging & DiseaseThe State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingJiangsu211166China
| | - Jialong Liang
- Department of Human AnatomyResearch Centre for Bone and Stem CellsKey Laboratory for Aging & DiseaseThe State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingJiangsu211166China
| | - Chunfeng Xie
- Department of Nutrition and Food SafetySchool of Public HealthNanjing Medical UniversityNanjingJiangsu211166China
| | - Xin Gu
- Department of Human AnatomyResearch Centre for Bone and Stem CellsKey Laboratory for Aging & DiseaseThe State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingJiangsu211166China
| | - Rong Wang
- Department of Human AnatomyResearch Centre for Bone and Stem CellsKey Laboratory for Aging & DiseaseThe State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingJiangsu211166China
| | - Zhiyuan Mao
- Department of Human AnatomyResearch Centre for Bone and Stem CellsKey Laboratory for Aging & DiseaseThe State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingJiangsu211166China
| | - Yongjie Zhang
- Department of Human AnatomyResearch Centre for Bone and Stem CellsKey Laboratory for Aging & DiseaseThe State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingJiangsu211166China
| | - Qing Li
- Department of Science and TechnologyJiangsu Jiankang Vocational CollegeNanjingJiangsu210029China
| | - Guoping Zuo
- Department of Human AnatomyResearch Centre for Bone and Stem CellsKey Laboratory for Aging & DiseaseThe State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingJiangsu211166China
| | - Dengshun Miao
- Department of Human AnatomyResearch Centre for Bone and Stem CellsKey Laboratory for Aging & DiseaseThe State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingJiangsu211166China
- The Research Center for AgingAffiliated Friendship Plastic Surgery Hospital of Nanjing Medical UniversityNanjingJiangsu210029China
| | - Jianliang Jin
- Department of Human AnatomyResearch Centre for Bone and Stem CellsKey Laboratory for Aging & DiseaseThe State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingJiangsu211166China
| |
Collapse
|
16
|
Yin Y, Wang Q, Xie C, Chen H, Jin J, Miao D. Amniotic membrane mesenchymal stem cells-based therapy improves Bmi-1-deficient mandible osteoporosis through stimulating osteoblastic bone formation and inhibiting osteoclastic bone resorption. J Tissue Eng Regen Med 2022; 16:538-549. [PMID: 35319819 DOI: 10.1002/term.3300] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/13/2022] [Accepted: 03/14/2022] [Indexed: 11/10/2022]
Abstract
Mandible osteoporosis with age is characterized by greater fragility and accompanied with abnormal oral function. Mesenchymal stem cell transplantation can ameliorate osteoporosis. Bmi-1 is a transcriptional repressor which is an important regulator of cell cycle, stem cells self-renewal, and cell senescence. Here, we use a new kind of membrane mesenchymal stem cells (MSCs), amniotic membrane mesenchymal stem cells (AMSCs), to explore therapeutic effects on Bmi-1-deficient caused mandible osteoporosis. Phenotypes of mandibles from 5-week-old Bmi-1-deficient mice with AMSCs-based therapy were compared with age-matched Bmi-1-deficient mandibles without AMSCs-based therapy and wild-type mice. Bmi-1-deficient mice without AMSCs-based therapy displayed mandible osteoporosis accompanied with the rising senescence-associated molecules and imbalance redox homeostasis. Results showed that the alveolar bone volume, cortical thickness, type I collagen and osteocalcin immunopositive areas, mRNA expression levels of alkaline phosphatase, superoxide dismutase, gluathione reductase, and protein expression level of Runx2 were all reduced significantly in Bmi-1-/- mandibles. Protein levels of PPARγ, p16, p21, p53, and redox gene levels of Bnip3l, Cdo1, Duox1, and Duox2 were up-regulated in mandibles from vehicle-transplanted Bmi-1-/- mice. Also, osteoclasts were activated in Bmi-1-/- alveolar bone. Transplanted AMSCs migrated into mandibles and improved all the parameters in Bmi-1-/- mandibles with AMSCs-based therapy. These findings indicate that AMSCs-based therapy could rescue mandible osteoporosis induced by Bmi-1 deficiency through stimulating osteoblastic bone formation and inhibiting osteoclastic bone resorption. Our findings implied that AMSCs-based therapy had preventative and therapeutic potential for mandible osteoporosis.
Collapse
Affiliation(s)
- Ying Yin
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, China
| | - Qiujiao Wang
- Department of Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, China.,Department of Stomatology, The Affiliated Jiangyin Hospital of Medical College of Southeast University, Jiangyin, China
| | - Chunfeng Xie
- Department of Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, China
| | - Haiyun Chen
- Department of Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, China
| | - Jianliang Jin
- Department of Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, China
| | - Dengshun Miao
- Department of Anatomy, Histology and Embryology, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, China.,Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
17
|
Bmi1 signaling maintains the plasticity of airway epithelial progenitors in response to persistent silica exposures. Toxicology 2022; 470:153152. [DOI: 10.1016/j.tox.2022.153152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 11/18/2022]
|
18
|
Zhou J, Hou C, Chen H, Qin Z, Miao Z, Zhao J, Wang Q, Cui M, Xie C, Wang R, Li Q, Zuo G, Miao D, Jin J. P16 I NK 4a Deletion Ameliorates Damage of Intestinal Epithelial Barrier and Microbial Dysbiosis in a Stress-Induced Premature Senescence Model of Bmi-1 Deficiency. Front Cell Dev Biol 2021; 9:671564. [PMID: 34712655 PMCID: PMC8545785 DOI: 10.3389/fcell.2021.671564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022] Open
Abstract
This study aimed to determine whether Bmi-1 deficiency leads to intestinal epithelial barrier destruction and microbiota dysfunction, which members of the microbial community alter barrier function with age, and whether p16INK4a deletion could reverse the damage of intestinal epithelial barrier and microbial dysbiosis. Intestines from Bmi-1–deficient (Bmi-1–/–), Bmi-1 and p16INK4a double-knockout (Bmi-1–/–p16INK4a–/–), and wild-type mice were observed for aging and inflammation. Duolink Proximity Ligation Assay, immunoprecipitation, and construction of p16INK4a overexpressed adenovirus and the overexpressed plasmids of full-length, mutant, or truncated fragments for occludin were used for analyzing the interaction between p16INK4a and occludin. High-throughput sequencing of V4 region amplicon of 16S ribosomal RNA was conducted using intestinal microbiota. We found Bmi-1 deficiency destructed barrier structure, barrier function, and tight junction (TJ) in intestinal epithelium; decreased the TJ proteins; increased tumor necrosis factor α (TNF-α)–dependent barrier permeability; and up-regulated proinflammatory level of macrophages induced by intestinal microbial dysbiosis. The transplantation of fecal microbiota from wild-type mice ameliorated TJ in intestinal epithelium of Bmi-1–/– and Bmi-1–/–p16INK4a–/– mice. Harmful bacteria including Desulfovibrio, Helicobacter, and Oscillibacter were at a higher level in Bmi-1–/– mice. More harmful bacteria Desulfovibrio entered the epithelium and promoted macrophages-secreted TNF-α and caused TNF-α–dependent barrier permeability and aging. Accumulated p16INK4a combined with occludin at the 1st–160th residue in cytoplasm of intestinal epithelium cells from Bmi-1–/– mice, which blocked formation of TJ and the repair of intestinal epithelium barrier. P16INK4a deletion could maintain barrier function and microbiota balance in Bmi-1–/– mice through strengthening formation of TJ and decreasing macrophages-secreted TNF-α induced by Desulfovibrio entering the intestinal epithelium. Thus, Bmi-1 maintained intestinal TJ, epithelial barrier function, and microbiota balance through preventing senescence characterized by p16INK4a accumulation. The clearance of p16INK4a-positive cells in aging intestinal epithelium would be a new method for maintaining barrier function and microbiota balance. The residues 1–160 of occludin could be a novel therapeutic target for identifying small molecular antagonistic peptides to prevent the combination of p16INK4a with occludin for protecting TJ.
Collapse
Affiliation(s)
- Jiawen Zhou
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Chenxing Hou
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Haiyun Chen
- Anti-Aging Research Laboratory, Friendship Plastic Surgery Hospital, Nanjing Medical University, Nanjing, China
| | - Ziyue Qin
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Zi'an Miao
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Jingyu Zhao
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Qiuyi Wang
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Min Cui
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Chunfeng Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Rong Wang
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Qing Li
- Department of Science and Technology, Jiangsu Jiankang Vocational College, Nanjing, China
| | - Guoping Zuo
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Dengshun Miao
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Anti-Aging Research Laboratory, Friendship Plastic Surgery Hospital, Nanjing Medical University, Nanjing, China
| | - Jianliang Jin
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
19
|
Takebe H, Irie K, Hosoya A. Localization of Bmi1 in osteoblast-lineage cells during endochondral ossification. Anat Rec (Hoboken) 2021; 305:1112-1118. [PMID: 34101367 DOI: 10.1002/ar.24693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 11/08/2022]
Abstract
Encoded by B cell-specific moloney murine leukemia virus integration site 1, Bmi1 is part of the polycomb group of proteins localized in stem and undifferentiated cells. It regulates the expression of various differentiation genes. However, the regulatory mechanism of skeletal development by Bmi1 remains poorly understood. In this study, we aimed to observe Bmi1 distribution during endochondral ossification processes in rat bone development and fracture healing. Immunoreactivity of Bmi1 was detected in the mesenchymal cell aggregation area at embryonic day (E) 14 and in cells around the center of cartilage primordium at E 16. Subsequently, the calcified bone matrix was formed around the cartilage primordium, and osteoblasts expressing Runt-related transcription factor 2 (Runx2) and Osterix (Osx) showed immunopositivity for Bmi1. At 4 days after bone fracture, the connective tissue around the fractured bone contained Bmi1-positive cells. At 42 days after fracture, osteoblasts along the surface of the new bone revealed Bmi1-, Runx2- and Osx-positive reactions, but the Bmi1 immunoreactivity in osteocytes was less than the Runx2 and Osx immunoreactivities. In conclusion, Bmi1 is localized in the osteoblast-lineage cells in their early differentiation stages, and it might regulate their differentiation during endochondral ossification.
Collapse
Affiliation(s)
- Hiroaki Takebe
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu-cho, Ishikari-gun, Hokkaido, Japan
| | - Kazuharu Irie
- Division of Anatomy, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu-cho, Ishikari-gun, Hokkaido, Japan
| | - Akihiro Hosoya
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu-cho, Ishikari-gun, Hokkaido, Japan
| |
Collapse
|
20
|
Yin Y, Zhou N, Zhang H, Dai X, Lv X, Chen N, Miao D, Hu Q. Bmi1 regulate tooth and mandible development by inhibiting p16 signal pathway. J Cell Mol Med 2021; 25:4195-4203. [PMID: 33745198 PMCID: PMC8093977 DOI: 10.1111/jcmm.16468] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 02/02/2021] [Accepted: 03/05/2021] [Indexed: 12/31/2022] Open
Abstract
To determine whether the deletion of p16 can correct tooth and mandible growth retardation caused by Bmi1 deficiency, we compared the tooth and mandible phenotypes of homozygous p16‐deficient (p16−/−) mice, homozygous Bmi1‐deficient (Bmi1−/−) mice, double homozygous Bmi1 and p16‐deficient (Bmi1−/−p16−/−) mice to those of their wild‐type littermates at 4 weeks of age by radiograph, histochemistry and immunohistochemistry. Results showed that compared to Bmi1−/− mice, the dental mineral density, dental volume and dentin sialoprotein immunopositive areas were increased, whereas the ratio of the predentin area to total dentin area and that of biglycan immunopositive area to dentin area were decreased in Bmi1−/−p16−/− mice. These results indicate that the deletion of p16 can improve tooth development in Bmi1 knockout mice. Compared to Bmi1−/− mice, the mandible mineral density, cortical thickness, alveolar bone volume, osteoblast number and activity, alkaline phosphatase positive area were all increased significantly in Bmi1−/−p16−/− mice. These results indicate that the deletion of p16 can improve mandible growth in Bmi1 knockout mice. Furthermore, the protein expression levels of cyclin D, CDK4 and p53 were increased significantly in p16−/− mice compared with those from wild‐type mice; the protein expression levels of cyclin D and CDK4 were decreased significantly, whereas those of p27 and p53 were increased significantly in Bmi1−/− mice; these parameters were partly rescued in Bmi1−/−p16−/− mice compared with those from Bmi1−/− mice. Therefore, our results indicate that Bmi1 plays roles in regulating tooth and mandible development by inhibiting p16 signal pathway which initiated entry into cell cycle.
Collapse
Affiliation(s)
- Ying Yin
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Anatomy, Histology and Embryology, State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, China
| | - Nan Zhou
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Non-communicable Disease Prevention, Nanjing Municipal Center for Disease Control and Prevention, Nanjing, China
| | - Hui Zhang
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiuliang Dai
- Reproductive Center, Nanjing Medical University Affiliated Changzhou Maternal and Child Health Care Hospital, Changzhou, China
| | - Xianhui Lv
- Department of Anatomy, Histology and Embryology, State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, China
| | - Ning Chen
- Institute of Stomatology, Nanjing Medical University, Nanjing, China
| | - Dengshun Miao
- Department of Anatomy, Histology and Embryology, State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, China.,The Research Center for Aging, Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, China
| | - Qingang Hu
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
21
|
Wen H, He Y, Zhang K, Yang X, Hao D, Jiang Y, He B. Mini-review: Functions and Action Mechanisms of PQQ in Osteoporosis and Neuro Injury. Curr Stem Cell Res Ther 2020; 15:32-36. [PMID: 30526470 DOI: 10.2174/1574888x14666181210165539] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 10/04/2018] [Accepted: 10/12/2018] [Indexed: 11/22/2022]
Abstract
Pyrroloquinoline Quinone (PQQ) is the third coenzyme found after niacinamide and flavone nucleotides and is widely present in microorganisms, plants, animals, and humans. PQQ can stimulate the growth of organisms and is very important for the growth, development and reproduction of animals. Owing to the inherent properties of PQQ as an antioxidant and redox modulator in various systems. In recent years, the role of PQQ in the field of osteoporosis and neuro injury has become a research hotspot. This article mainly discusses the derivatives, distribution of PQQ, in vitro models of osteoporosis and neuro injury, and the research progress of its mechanism of action. It provides new ideas in the study of osteoporosis and neuro injury.
Collapse
Affiliation(s)
- Hao Wen
- Department of Spine Surgery, Honghui Hospital Affiliated to Xi'an Jiaotong University, Xi'an, China.,Yan'an University Medical School, Yan'an, China
| | - Yuan He
- Fifth Hospital of Xi'an, Xi'an , China
| | - Ke Zhang
- Yan'an University Medical School, Yan'an, China
| | - Xiaobin Yang
- Department of Spine Surgery, Honghui Hospital Affiliated to Xi'an Jiaotong University, Xi'an, China
| | - Dingjun Hao
- Department of Spine Surgery, Honghui Hospital Affiliated to Xi'an Jiaotong University, Xi'an, China
| | - Yonghong Jiang
- Department of Spine Surgery, Honghui Hospital Affiliated to Xi'an Jiaotong University, Xi'an, China
| | - Baorong He
- Department of Spine Surgery, Honghui Hospital Affiliated to Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
22
|
Delgado D, Garate A, Sánchez P, Bilbao AM, García Del Caño G, Salles J, Sánchez M. Biological and structural effects after intraosseous infiltrations of age-dependent platelet-rich plasma: An in vivo study. J Orthop Res 2020; 38:1931-1941. [PMID: 32129513 DOI: 10.1002/jor.24646] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 01/09/2020] [Accepted: 02/29/2020] [Indexed: 02/04/2023]
Abstract
Platelet-rich plasma (PRP) is an increasingly widespread treatment for joint pathologies. Its characteristics and administration route are variables that may influence the clinical outcome. The aim of this in vivo study was to analyze in aged rats the biological and structure effects of intraosseous infiltrations of two different types of PRP obtained from young and old donors. During 6 months intraosseous infiltrations were performed and 4 days after the last infiltration, animals were sacrificed, and bones were extracted for micro-computed tomography (micro-CT) and histological analysis. Molecular composition of the PRP of aged donors presented higher levels of proinflammatory molecules. The histological studies showed a greater cellularity of bone marrow in groups treated with PRP. Concerning micro-CT analysis, young PRP showed a better femoral bone structure according to values of percentage of trabecular bone, trabecular space, trabecular density, and subchondral bone plate volume. In summary, this study has demonstrated that intraosseous infiltrations of PRP from young donors prevent from age-related bone degeneration. This treatment could stimulate the biological processes that maintain homeostasis and bone structure and avoid osteoarticular pathologies.
Collapse
Affiliation(s)
- Diego Delgado
- Advanced Biological Therapy Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain
| | - Ane Garate
- Advanced Biological Therapy Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain
| | - Pello Sánchez
- Advanced Biological Therapy Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain
| | - Ane Miren Bilbao
- Arthroscopic Surgery Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain
| | - Gontzal García Del Caño
- Department of Neurosciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Joan Salles
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Mikel Sánchez
- Advanced Biological Therapy Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain.,Arthroscopic Surgery Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain
| |
Collapse
|
23
|
Zhou J, Chen A, Wang Z, Zhang J, Chen H, Zhang H, Wang R, Miao D, Jin J. Bmi-1 determines the stemness of renal stem or progenitor cells. Biochem Biophys Res Commun 2020; 529:1165-1172. [PMID: 32819581 DOI: 10.1016/j.bbrc.2020.06.140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/26/2020] [Indexed: 12/24/2022]
Abstract
Renal stem or progenitor cells (RSCs), labeled with CD24 and CD133, play an important role during the repair of renal injury. Bmi-1 is a critical factor in regulating stemness of adult stem cells or progenitor cells. To investigate whether Bmi-1 determines the stemness of RSCs by inhibiting p16 and p53, and/or maintaining redox balance, RSCs were isolated, cultured and analyzed for stemness characterizations. In RSCs from Bmi-1-deficient (Bmi-1-/-) mice and wild type (WT) littermates, self-renewal, stemness, and expressions of molecules for regulating redox balance and cell cycle progression were compared. Self-renewal of RSCs from Bmi-1 and p16 double-knockout (Bmi-1-/-p16-/-), Bmi-1 and p53 double-knockout (Bmi-1-/-p53-/-) and N-acetylcysteine (NAC)-treated Bmi-1-/- mice were further analyzed for amelioration. Human renal proximal tubular epithelial cells (HK2) were also used for signaling analysis. Our results showed that third-passage RSCs from WT mice had good stemness; Bmi-1 deficiency led to the decreased stemness, and the increased apoptosis for RSCs; NAC treatment or p16/p53 deletion ameliorated the decreased self-renewal of RSCs in Bmi-1 deficiency mice by maintaining redox balance or inhibiting cell cycle arrest respectively; Oxidative stress (OS) could negatively feedback regulate the mRNA expressions of Bmi-1, p16 and p53. In conclusion, Bmi-1 determined the stemness of RSCs through maintaining redox balance and preventing cell cycle arrest. Thus, Bmi-1 signaling molecules would be novel therapeutic targets for maintaining RSCs and hampering the progression of kidney diseases to prevent renal failure.
Collapse
Affiliation(s)
- Jiawen Zhou
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Ao Chen
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Ziyang Wang
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jin'ge Zhang
- School of Nursing, Shanxi Medical University, Jinzhong, Shanxi, 030001, China
| | - Haiyun Chen
- Anti-aging Research Laboratory, Friendship Plastic Surgery Hospital, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Hengzhi Zhang
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Rong Wang
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Dengshun Miao
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jianliang Jin
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
24
|
Gao T, Lin M, Shao B, Zhou Q, Wang Y, Chen X, Zhao D, Dai X, Shen C, Cheng H, Yang S, Li H, Zheng B, Zhong X, Yu J, Chen L, Huang X. BMI1 promotes steroidogenesis through maintaining redox homeostasis in mouse MLTC-1 and primary Leydig cells. Cell Cycle 2020; 19:1884-1898. [PMID: 32594840 PMCID: PMC7469621 DOI: 10.1080/15384101.2020.1779471] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In males, aging is accompanied by decline in serum testosterone levels due to impairment of testicular Leydig cells. The polycomb protein BMI1 has recently been identified as an anti-aging factor. In our previous study, BMI1 null mice showed decreased serum testosterone and Leydig cell population, excessive oxidative stress and p16/p19 signaling activation. However, a cause-and-effect relationship between phenotypes and pathways was not investigated. Here, we used the rescue approach to study the role of oxidative stress or p16/p19 in BMI1-mediated steroidogenesis. Our results revealed that treatment with antioxidant NAC, but not down-regulation of p16/p19, largely rescued cell senescence, DNA damage and steroidogenesis in BMI1-deficient mouse MLTC-1 and primary Leydig cells. Collectively, our study demonstrates that BMI1 orchestrates steroidogenesis mainly through maintaining redox homeostasis, and thus, BMI1 may be a novel and potential therapeutic target for treatment of hypogonadism.
Collapse
Affiliation(s)
- Tingting Gao
- Center of Clinical Reproductive Medicine, The Affiliated Changzhou Matemity and Child Health Care Hospital of Nanjing Medical University , Changzhou, China
| | - Meng Lin
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University , Nanjing, China
| | - Binbin Shao
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital , Nanjing, China
| | - Qiao Zhou
- Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital , Nanjing, China
| | - Yufeng Wang
- Center of Clinical Reproductive Medicine, The Affiliated Changzhou Matemity and Child Health Care Hospital of Nanjing Medical University , Changzhou, China
| | - Xia Chen
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangsu University, Jiangsu University , Zhenjiang, China
| | - Dan Zhao
- Fourth Affiliated Hospital of Jiangsu University , Zhenjiang, China
| | - Xiuliang Dai
- Center of Clinical Reproductive Medicine, The Affiliated Changzhou Matemity and Child Health Care Hospital of Nanjing Medical University , Changzhou, China
| | - Cong Shen
- Center for Reproduction and Genetics, NHC Key Laboratory of Male Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University , Suzhou, China
| | - Hongbo Cheng
- Center for Reproduction and Genetics, NHC Key Laboratory of Male Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University , Suzhou, China
| | - Shenmin Yang
- Center for Reproduction and Genetics, NHC Key Laboratory of Male Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University , Suzhou, China
| | - Hong Li
- Center for Reproduction and Genetics, NHC Key Laboratory of Male Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University , Suzhou, China
| | - Bo Zheng
- Center for Reproduction and Genetics, NHC Key Laboratory of Male Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University , Suzhou, China.,State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University , Suzhou, China
| | - Xingming Zhong
- NHC Key Laboratory of Male Reproduction and Genetics , Guangdong, China.,Department of Reproductive Immunity and Genetics, Family Planning Research Institute of Guangdong Province , Guangdong, China.,Department of Reproductive Immunity and Genetics, Family Planning Special Hospital of Guangdong Province , Guangzhou, China
| | - Jun Yu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangsu University, Jiangsu University , Zhenjiang, China
| | - Li Chen
- Center of Clinical Reproductive Medicine, The Affiliated Changzhou Matemity and Child Health Care Hospital of Nanjing Medical University , Changzhou, China
| | - Xiaoyan Huang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University , Nanjing, China
| |
Collapse
|
25
|
Zhang Q, Li J, Li Y, Che H, Chen Y, Dong J, Xian CJ, Miao D, Wang L, Ren Y. Bmi deficiency causes oxidative stress and intervertebral disc degeneration which can be alleviated by antioxidant treatment. J Cell Mol Med 2020; 24:8950-8961. [PMID: 32583517 PMCID: PMC7417700 DOI: 10.1111/jcmm.15528] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/07/2020] [Accepted: 06/02/2020] [Indexed: 02/01/2023] Open
Abstract
The transcriptional repressor Bmi‐1 is involved in cell‐cycle regulation and cell senescence, the deficiency of which has been shown to cause oxidative stress. This study investigated whether Bmi‐1 deficiency plays a role in promoting disc degeneration and the effect of treatment with antioxidant N‐acetylcysteine (NAC) on intervertebral disc degeneration. Bmi‐1−/− mice were treated with the antioxidant NAC, supplied in drinking water (Bmi‐1−/−+NAC). For in vitro experiments, mouse intervertebral discs were cultured under low oxygen tension and serum‐limiting conditions in the presence of tumour necrosis factor α and interleukin 1β in order to mimic degenerative insult. Disc metabolism parameters in these in vitro and in vivo studies were evaluated by histopathological, immunohistochemical and molecular methods. Bmi‐1−/− mice showed lower collagen Ⅱ and aggrecan levels and higher collagen Ⅹ levels than wild‐type and Bmi‐1−/−+NAC mice. Bmi‐1−/− mice showed significantly lower superoxide dismutase (SOD)‐1, SOD‐2, glutathione peroxidase (GPX)‐1 and GPX‐3 levels than their wild‐type littermates and Bmi‐1−/−+ NAC mice. Relative to Bmi‐1−/− mice, the control and Bmi‐1−/−+NAC mice showed significantly lower p16, p21, and p53 levels. These results demonstrate that Bmi‐1 plays an important role in attenuating intervertebral disc degeneration in mice by inhibiting oxidative stress and cell apoptosis.
Collapse
Affiliation(s)
- Qunhu Zhang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Orthopaedics, Suqian First Hospital, Suqian, Jiangsu, China
| | - Jie Li
- Department of Orthopaedics, Xuzhou Central Hospital, Xuzhou Clinical College of Nanjing Medical University, The Affiliated Xuzhou Hospital of Southeast University, Xuzhou, Jiangsu, China
| | - You Li
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hui Che
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Chen
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianghui Dong
- Department of Hand Surgery, Department of Plastic Reconstructive Surgery, Ningbo No. 6 Hospital, Ningbo, Zhejiang, China.,UniSA Clinical and Health Sciences and UniSA Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Cory J Xian
- UniSA Clinical and Health Sciences and UniSA Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Dengshun Miao
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liping Wang
- Department of Hand Surgery, Department of Plastic Reconstructive Surgery, Ningbo No. 6 Hospital, Ningbo, Zhejiang, China.,UniSA Clinical and Health Sciences and UniSA Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Yongxin Ren
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
26
|
Jacques C, Tesfaye R, Lavaud M, Georges S, Baud’huin M, Lamoureux F, Ory B. Implication of the p53-Related miR-34c, -125b, and -203 in the Osteoblastic Differentiation and the Malignant Transformation of Bone Sarcomas. Cells 2020; 9:cells9040810. [PMID: 32230926 PMCID: PMC7226610 DOI: 10.3390/cells9040810] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 02/07/2023] Open
Abstract
The formation of the skeleton occurs throughout the lives of vertebrates and is achieved through the balanced activities of two kinds of specialized bone cells: the bone-forming osteoblasts and the bone-resorbing osteoclasts. Impairment in the remodeling processes dramatically hampers the proper healing of fractures and can also result in malignant bone diseases such as osteosarcoma. MicroRNAs (miRNAs) are a class of small non-coding single-strand RNAs implicated in the control of various cellular activities such as proliferation, differentiation, and apoptosis. Their post-transcriptional regulatory role confers on them inhibitory functions toward specific target mRNAs. As miRNAs are involved in the differentiation program of precursor cells, it is now well established that this class of molecules also influences bone formation by affecting osteoblastic differentiation and the fate of osteoblasts. In response to various cell signals, the tumor-suppressor protein p53 activates a huge range of genes, whose miRNAs promote genomic-integrity maintenance, cell-cycle arrest, cell senescence, and apoptosis. Here, we review the role of three p53-related miRNAs, miR-34c, -125b, and -203, in the bone-remodeling context and, in particular, in osteoblastic differentiation. The second aim of this study is to deal with the potential implication of these miRNAs in osteosarcoma development and progression.
Collapse
|
27
|
Sun H, Qiao W, Cui M, Yang C, Wang R, Goltzman D, Jin J, Miao D. The Polycomb Protein Bmi1 Plays a Crucial Role in the Prevention of 1,25(OH) 2 D Deficiency-Induced Bone Loss. J Bone Miner Res 2020; 35:583-595. [PMID: 31725940 DOI: 10.1002/jbmr.3921] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/06/2019] [Accepted: 11/11/2019] [Indexed: 12/14/2022]
Abstract
We analyzed the skeletal phenotypes of heterozygous null Cyp27b1 (Cyp27b1+/- ) mice and their wild-type (WT) littermates to determine whether haploinsufficiency of Cyp27b1 accelerated bone loss, and to examine potential mechanisms of such loss. We found that serum 1,25-dihydroxyvitamin D [1,25(OH)2 D] levels were significantly decreased in aging Cyp27b1+/- mice, which displayed an osteoporotic phenotype. This was accompanied by a reduction of expression of the B lymphoma Moloney murine leukemia virus (Mo-MLV) insertion region 1 (Bmi1) at both gene and protein levels. Using chromatin immunoprecipitation (ChIP)-PCR, electrophoretic mobility shift assay (EMSA) and a luciferase reporter assay, we then showed that 1,25(OH)2 D3 upregulated Bmi1 expression at a transcriptional level via the vitamin D receptor (VDR). To determine whether Bmi1 overexpression in mesenchymal stem cells (MSCs) could correct bone loss induced by 1,25(OH)2 D deficiency, we overexpressed Bmi1 in MSCs using Prx1-driven Bmi1 transgenic mice (Bmi1Tg ) mice. We then compared the bone phenotypes of Bmi1Tg mice on a Cyp27b1+/- background, with those of Cyp27b1+/- mice and with those of WT mice, all at 8 months of age. We found that overexpression of Bmi1 in MSCs corrected the bone phenotype of Cyp27b1+/- mice by increasing osteoblastic bone formation, reducing osteoclastic bone resorption, increasing bone volume, and increasing bone mineral density. Bmi1 overexpression in MSCs also corrected 1,25(OH)2 D deficiency-induced oxidative stress and DNA damage, and cellular senescence of Cyp27b1+/- mice by reducing levels of reactive oxygen species (ROS), elevating serum total superoxide dismutase levels, reducing the percentage of γH2 A.X, p16, IL-1β, and TNF-α-positive cells and decreasing γH2A.X, p16, p19, p53, p21, IL-1β, and IL-6 expression levels. Furthermore, 1,25(OH)2 D stimulated the osteogenic differentiation of MSCs, both ex vivo and in vitro, from WT mice but not from Bmi1-/- mice and 1,25(OH)2 D administration in vivo increased osteoblastic bone formation in WT, but not in Bmi1 -/- mice. Our results indicate that Bmi1, a key downstream target of 1,25(OH)2 D, plays a crucial role in preventing bone loss induced by 1,25(OH)2 D deficiency. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Haijian Sun
- State Key Laboratory of Reproductive Medicine, Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, China
| | - Wanxin Qiao
- State Key Laboratory of Reproductive Medicine, Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, China
| | - Min Cui
- State Key Laboratory of Reproductive Medicine, Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, China
| | - Cuicui Yang
- State Key Laboratory of Reproductive Medicine, Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, China
| | - Rong Wang
- State Key Laboratory of Reproductive Medicine, Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, China
| | - David Goltzman
- Calcium Research Laboratory, McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Jianliang Jin
- State Key Laboratory of Reproductive Medicine, Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, China
| | - Dengshun Miao
- State Key Laboratory of Reproductive Medicine, Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, China
| |
Collapse
|
28
|
Wang H, Zhang H, Srinivasan V, Tao J, Sun W, Lin X, Wu T, Boyce BF, Ebetino FH, Boeckman RK, Xing L. Targeting Bortezomib to Bone Increases Its Bone Anabolic Activity and Reduces Systemic Adverse Effects in Mice. J Bone Miner Res 2020; 35:343-356. [PMID: 31610066 PMCID: PMC10587833 DOI: 10.1002/jbmr.3889] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 08/15/2019] [Accepted: 09/07/2019] [Indexed: 12/20/2022]
Abstract
Bortezomib (Btz) is a proteasome inhibitor approved by the FDA to treat multiple myeloma. It also increases bone volume by promoting osteoblast differentiation and inhibiting osteoclastogenesis in mice. However, Btz has severe systemic adverse effects, which would limit its use as a bone anabolic agent. Here, we designed and synthesized a bone-targeted form of Btz by conjugating it to a bisphosphonate (BP) with no antiresorptive activity. We report that BP-Btz inhibited osteoclast formation and bone resorption and stimulated osteoblast differentiation in vitro similar to Btz. In vivo, BP-Btz increased bone volume more effectively than Btz in three mouse models: untreated wild-type mice, mice with ovariectomy, and aged mice with tibial factures. Importantly, BP-Btz had significantly less systemic side effects than Btz, including less thymic cell death, sympathetic nerve damage, and thrombocytopenia, and it improved survival rates in aged mice. Thus, BP-Btz represents a novel anabolic agent to treat conditions, such as postmenopausal and age-related bone loss. Bone targeting is an attractive approach to repurpose approved drugs to treat skeletal diseases. © 2019 American Society for Bone and Mineral Research. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Hua Wang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Institute of Stomatology, Nanjing Medical University, Jiangsu Key Laboratory of Oral Diseases, Nanjing, China
| | - Hengwei Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Venkat Srinivasan
- Department of Chemistry, University of Rochester, Rochester, NY, USA
| | - Jianguo Tao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Wen Sun
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Xi Lin
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Tao Wu
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Bone Disease, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Frank H Ebetino
- Department of Chemistry, University of Rochester, Rochester, NY, USA
- BioVinc, Pasadena, CA, USA
| | - Robert K Boeckman
- Department of Chemistry, University of Rochester, Rochester, NY, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
29
|
Chen G, Zhang Y, Yu S, Sun W, Miao D. Bmi1 Overexpression in Mesenchymal Stem Cells Exerts Antiaging and Antiosteoporosis Effects by Inactivating p16/p19 Signaling and Inhibiting Oxidative Stress. Stem Cells 2019; 37:1200-1211. [PMID: 30895687 PMCID: PMC6851636 DOI: 10.1002/stem.3007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/24/2019] [Accepted: 03/03/2019] [Indexed: 01/19/2023]
Abstract
We previously demonstrated that Bmi1 deficiency leads to osteoporosis phenotype by inhibiting the proliferation and osteogenic differentiation of bone marrow mesenchymal stem cells (MSCs), but it is unclear whether overexpression of Bmi1 in MSCs stimulates skeletal development and rescues Bmi1 deficiency-induced osteoporosis. To answer this question, we constructed transgenic mice (Bmi1Tg ) that overexpressed Bmi1 driven by the Prx1 gene and analyzed their skeletal phenotype differences with that of wild-type littermates. We then hybridized Bmi1Tg to Bmi1-/- mice to generate Bmi1-/- mice overexpressing Bmi1 in MSCs and compared their skeletal phenotypes with those of Bmi1-/- and wild-type mice using imaging, histopathological, immunohistochemical, histomorphometric, cellular, and molecular methods. Bmi1Tg mice exhibited enhanced bone growth and osteoblast formation, including the augmentation of bone size, cortical and trabecular volume, number of osteoblasts, alkaline phosphatase (ALP)-positive and type I collagen-positive areas, number of total colony forming unit fibroblasts (CFU-f) and ALP+ CFU-f, and osteogenic gene expression levels. Consistently, MSC overexpressing Bmi1 in the Bmi1-/- background not only largely reversed Bmi1 systemic deficiency-induced skeletal growth retardation and osteoporosis, but also partially reversed Bmi1 deficiency-induced systemic growth retardation and premature aging. To further explore the mechanism of action of MSCs overexpressing Bmi1 in antiosteoporosis and antiaging, we examined changes in oxidative stress and expression levels of p16 and p19. Our results showed that overexpression of Bmi1 in MSCs inhibited oxidative stress and downregulated p16 and p19. Taken together, the results of this study indicate that overexpression of Bmi1 in MSCs exerts antiaging and antiosteoporosis effects by inactivating p16/p19 signaling and inhibiting oxidative stress. Stem Cells 2019;37:1200-1211.
Collapse
Affiliation(s)
- Guangpei Chen
- Department of Human Anatomy, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China.,The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, People's Republic of China
| | - Ying Zhang
- The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, People's Republic of China.,Department of Anatomy, Histology, and Embryology, Suzhou Health and Technology College, Suzhou, People's Republic of China
| | - Shuxiang Yu
- The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, People's Republic of China
| | - Wen Sun
- The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, People's Republic of China
| | - Dengshun Miao
- The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, People's Republic of China.,The Research Center for Aging, Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
30
|
Wang H, Hu Z, Wu J, Mei Y, Zhang Q, Zhang H, Miao D, Sun W. Sirt1 Promotes Osteogenic Differentiation and Increases Alveolar Bone Mass via Bmi1 Activation in Mice. J Bone Miner Res 2019; 34:1169-1181. [PMID: 30690778 DOI: 10.1002/jbmr.3677] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 12/27/2018] [Accepted: 01/11/2019] [Indexed: 12/16/2022]
Abstract
Sirtuin 1 (Sirt1), a protein deacetylase, is a novel target for bone metabolism. To investigate whether overexpression of Sirt1 in mandibular mesenchymal stem cells (M-MSCs) increased alveolar bone mass in vivo, we generated Sirt1 transgenic mice (Sirt1TG ), with Sirt1 gene expression driven by the Prx1 gene, which represents the mesenchymal lineage. Our results demonstrated that overexpression of Sirt1 in M-MSCs increased the alveolar bone volume in 1-month-old, 9-month-old, and 18-month-old Sirt1TG mice compared with age-matched wild-type (WT) mice, and in ovariectomized Sirt1TG mice compared with ovariectomized WT mice by stimulating M-MSC differentiation into osteoblasts. Treatment with resveratrol, a Sirt1 activator, increased Sirt1 binding with Bmi1 and reduced Bmi1 acetylation in a dose-dependent manner demonstrated in M-MSC cultures. Both treatment with resveratrol in M-MSC cultures and overexpressed Sirt1 in M-MSCs ex vivo cultures increased nuclear translocation of Bmi1. Furthermore, we demonstrated that deletion of Bmi1 blocked the increased alveolar bone volume in Sirt1TG mice. The Sirt1 activator resveratrol inhibited human MSC senescence and promoted their differentiation into osteoblasts, which were associated with upregulating the expression levels of Sirt1 and nuclear translocation of Bmi1. The present results suggested that Sirt1 promotes MSC proliferation and osteogenic differentiation, inhibits MSC senescence to increase alveolar bone volume by promoting the deacetylation and nuclear translocation of Bmi1. Thus, our study elucidated the mechanism by which Sirt1 increases alveolar bone mass, and these findings are important for the clinical application of the Sirt1 activator resveratrol for the promotion of alveolar bone formation and prevention of alveolar bone loss. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Hua Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zixuan Hu
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Jun Wu
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Yukun Mei
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Qian Zhang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Hengwei Zhang
- Center for Musculoskeletal Research (CMSR), Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Dengshun Miao
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Wen Sun
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
The role of sirtuin 1 and its activator, resveratrol in osteoarthritis. Biosci Rep 2019; 39:BSR20190189. [PMID: 30996115 PMCID: PMC6509056 DOI: 10.1042/bsr20190189] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 12/19/2022] Open
Abstract
Osteoarthitis (OA) is the most common aging-related joint pathology; the aging process results in changes to joint tissues that ultimately contribute to the development of OA. Articular chondrocytes exhibit an aging-related decline in their proliferative and synthetic capacity. Sirtuin 1 (SIRT 1), a longevity gene related to many diseases associated with aging, is a nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase and master metabolic regulator. Along with its natural activator resveratrol, SIRT 1 actively participates in the OA pathological progress. SIRT 1 expression in osteoarthritic cartilage decreases in the disease progression of OA; it appears to play a predominantly regulatory role in OA. SIRT 1 can regulate the expression of extracellular matrix (ECM)-related proteins; promote mesenchymal stem cell differentiation; play anti-catabolic, anti-inflammatory, anti-oxidative stress, and anti-apoptosis roles; participate in the autophagic process; and regulate bone homeostasis in OA. Resveratrol can activate SIRT 1 in order to inhibit OA disease progression. In the future, activating SIRT 1 via resveratrol with improved bioavailability may be an appropriate therapeutic approach for OA.
Collapse
|
32
|
Adamik J, Roodman GD, Galson DL. Epigenetic-Based Mechanisms of Osteoblast Suppression in Multiple Myeloma Bone Disease. JBMR Plus 2019; 3:e10183. [PMID: 30918921 PMCID: PMC6419609 DOI: 10.1002/jbm4.10183] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 12/29/2018] [Accepted: 02/03/2019] [Indexed: 12/18/2022] Open
Abstract
Multiple myeloma (MM) bone disease is characterized by the development of osteolytic lesions, which cause severe complications affecting the morbidity, mortality, and treatment of myeloma patients. Myeloma tumors seeded within the bone microenvironment promote hyperactivation of osteoclasts and suppression of osteoblast differentiation. Because of this prolonged suppression of bone marrow stromal cells’ (BMSCs) differentiation into functioning osteoblasts, bone lesions in patients persist even in the absence of active disease. Current antiresorptive therapy provides insufficient bone anabolic effects to reliably repair MM lesions. It has become widely accepted that myeloma‐exposed BMSCs have an altered phenotype with pro‐inflammatory, immune‐modulatory, anti‐osteogenic, and pro‐adipogenic properties. In this review, we focus on the role of epigenetic‐based modalities in the establishment and maintenance of myeloma‐induced suppression of osteogenic commitment of BMSCs. We will focus on recent studies demonstrating the involvement of chromatin‐modifying enzymes in transcriptional repression of osteogenic genes in MM‐BMSCs. We will further address the epigenetic plasticity in the differentiation commitment of osteoprogenitor cells and assess the involvement of chromatin modifiers in MSC‐lineage switching from osteogenic to adipogenic in the context of the inflammatory myeloma microenvironment. Lastly, we will discuss the potential of employing small molecule epigenetic inhibitors currently used in the MM research as therapeutics and bone anabolic agents in the prevention or repair of osteolytic lesions in MM. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Juraj Adamik
- Department of Medicine Division of Hematology/Oncology, UPMC Hillman Cancer Center, The McGowan Institute for Regenerative Medicine University of Pittsburgh Pittsburgh PA USA
| | - G David Roodman
- Department of Medicine Division of Hematology-Oncology Indiana University Indianapolis IN USA.,Richard L Roudebush VA Medical Center Indianapolis IN USA
| | - Deborah L Galson
- Department of Medicine Division of Hematology/Oncology, UPMC Hillman Cancer Center, The McGowan Institute for Regenerative Medicine University of Pittsburgh Pittsburgh PA USA
| |
Collapse
|
33
|
Wang R, Xue X, Wang Y, Zhao H, Zhang Y, Wang H, Miao D. BMI1 Deficiency Results in Female Infertility by Activating p16/p19 Signaling and Increasing Oxidative Stress. Int J Biol Sci 2019; 15:870-881. [PMID: 30906217 PMCID: PMC6429020 DOI: 10.7150/ijbs.30488] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/12/2019] [Indexed: 01/20/2023] Open
Abstract
The polycomb repressor B lymphoma Mo-MLV insertion region 1 (BMI1) is a core composition of polycomb repressive complex 1 (PRC1) and contributes to diverse fundamental cellular processes including cell senescence, apoptosis and proliferation. To investigate the role and mechanism of BMI1 in maintaining normal female reproductive function, we compared the differences in reproductive phenotypes between Bmi1-deficient and wild-type female mice. The Bmi1-deficient female mice were then supplemented with N-acetylcysteine in their drinking water to explore whether antioxidant supplementation could improve reproductive dysfunction caused by BMI1 deficiency. The results revealed that Bmi1 deletion resulted in complete infertility in female mice, estrous cycle disorder, and follicular developmental disorders. The reactive oxygen species levels in the ovarian tissue were increased; the ability of antioxidant enzymes was downregulated; the expression levels of p19 and p53 proteins were significantly upregulated. We also found that oocytes derived from Bmi1-deficient mice could not develop into embryos by in vitro fertilization and in vitro culture of embryos. Furthermore, supplementation with the antioxidant NAC not only improved the reproductive defects caused by Bmi1 deletion, but also largely rescued the ability of Bmi1-deficient oocytes to develop into embryos in vitro. These results indicated that cells lacking Bmi1 resulted in female infertility by activating the p16/p19 signaling pathway, increasing oxidative stress and DNA damage, inhibiting granulosa cell proliferation, and inducing granulosa cell apoptosis. Thus, BMI1 may be a novel potential target for the clinical treatment of female infertility.
Collapse
Affiliation(s)
- Rong Wang
- State Key Laboratory of Reproductive Medicine, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
- The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xian Xue
- State Key Laboratory of Reproductive Medicine, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
- The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yang Wang
- State Key Laboratory of Reproductive Medicine, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haiyang Zhao
- State Key Laboratory of Reproductive Medicine, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuling Zhang
- Department of Ultrasound, Taikang Xianlin Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Hui Wang
- State Key Laboratory of Reproductive Medicine, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dengshun Miao
- State Key Laboratory of Reproductive Medicine, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
- The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
34
|
Katoh-Fukui Y, Baba T, Sato T, Otake H, Nagakui-Noguchi Y, Shindo M, Suyama M, Ohkawa Y, Tsumura H, Morohashi KI, Fukami M. Mouse polycomb group gene Cbx2 promotes osteoblastic but suppresses adipogenic differentiation in postnatal long bones. Bone 2019; 120:219-231. [PMID: 30389610 DOI: 10.1016/j.bone.2018.10.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 10/19/2018] [Accepted: 10/19/2018] [Indexed: 12/29/2022]
Abstract
A set of key developmental genes is essential for skeletal growth from multipotent progenitor cells at weaning. Polycomb group proteins, which regulate such genes contributes to the cell lineage commitment and subsequent differentiation via epigenetic chromatin modification and remodeling. However, it is unclear which cell lineage and gene sets are targeted by polycomb proteins during skeletal growth. We now report that mice deficient in a polycomb group gene Cbx2cterm/cterm exhibited skeletal hypoplasia in the tibia, femur, and cranium. Long bone cavities in these mice contained fewer multipotent mesenchymal stromal cells. RNA-sequencing of bone marrow cells showed downregulation and upregulation of osteoblastic and adipogenic genes, respectively. Furthermore, the expression levels of genes specifically expressed in B-cell precursors were decreased. Forced expression of Cbx2 in Cbx2cterm/cterm bone marrow stromal cell recovered fibroblastic colony formation and suppressed adipogenic differentiation. Collectively, our results suggest that Cbx2 controls the maintenance and adipogenic differentiation of mesenchymal stromal cells in the bone marrow.
Collapse
Affiliation(s)
- Yuko Katoh-Fukui
- Department of Molecular Endocrinology, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan.
| | - Takashi Baba
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Sato
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan; Division of Bioinformatics, Kyushu University, Fukuoka, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Fukuoka, Japan
| | - Hiroyuki Otake
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Miyuki Shindo
- Department of Experimental Animals, National Research Institute of Child Health and Development, Tokyo, Japan
| | - Mikita Suyama
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan; Division of Bioinformatics, Kyushu University, Fukuoka, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Fukuoka, Japan
| | - Yasuyuki Ohkawa
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Fukuoka, Japan; Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hideki Tsumura
- Department of Experimental Animals, National Research Institute of Child Health and Development, Tokyo, Japan
| | - Ken-Ichirou Morohashi
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
35
|
Sun W, Qiao W, Zhou B, Hu Z, Yan Q, Wu J, Wang R, Zhang Q, Miao D. Overexpression of Sirt1 in mesenchymal stem cells protects against bone loss in mice by FOXO3a deacetylation and oxidative stress inhibition. Metabolism 2018; 88:61-71. [PMID: 30318050 DOI: 10.1016/j.metabol.2018.06.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/23/2018] [Accepted: 06/17/2018] [Indexed: 12/29/2022]
Abstract
OBJECTIVE B cell-specific Moloney murine leukemia virus integration site 1 (Bmi-1) deficiency (Bmi-1-/-) leads to an osteoporotic phenotype with a significant downregulation of Sirt1 protein expression. Sirtuin 1 (Sirt1) haploinsufficiency results in a bone loss by decreased bone formation; however, it is unclear whether Sirt1 overexpression in mesenchymal stem cells (MSCs) plays an anti-osteoporotic role. The aim of the study is to identify whether the overexpression of Sirt1 in MSCs could restore skeletal growth retardation and osteoporosis in Bmi-1 deficient mice. METHODS We used our new generated transgenic mouse model that overexpresses Sirt1 in its MSCs (Sirt1TG) to cross with Bmi-1-/- mice to generate Bmi-1-/- mice with Sirt1 overexpression in MSCs, and compared their skeletal metabolism with those of their Bmi-1-/- and wild-type (WT) littermates (6 mice for each genotype) at 4 weeks of age using imaging, histopathological, immunohistochemical, histomorphometric, cellular, and molecular methods. RESULTS The levels of expression for Sirt1 were noticeably higher in the skeletal tissue of Sirt1TG mice than in those of WT mice. In Comparison to WT mice, the body weight and size, skeletal size, bone volume, osteoblast number, alkaline phosphatase and type I collagen positive areas, osteogenic related gene expression levels were all significantly increased in the Sirt1TG mice. Overexpression of Sirt1 in Bmi-1-/- mouse MSCs resulted in a longer lifespan, improved skeletal growth and significantly increased bone mass by stimulating osteoblastic bone formation and inhibiting osteoclastic bone resorption in the Bmi-1-/- mice, although the defects were not completely restored. Furthermore, Sirt1 overexpression in MSCs reduced the acetylation level of FOXO3a (Forkhead box O3a), increasing levels of expression for FOXO3a and SOD2 (Superoxide dismutase 2) in bony tissue, enhanced osteogenesis and reduced osteogenic cell senescence. We also demonstrated that nicotinamide, a Sirt1 inhibitor, blocks the effect of overexpression of Sirt1 in MSCs on osteogenesis and osteogenic cell senescence. CONCLUSIONS Taken together, these results demonstrate that Sirt1 overexpression in MSCs increased the osteoblastic bone formation and partially restores the defects in skeletal growth and osteogenesis in Bmi-1-/- mice by FOXO3a deacetylation and oxidative stress inhibition. Our data support the proposal that Sirt1 is a target for promoting bone formation as an anabolic approach for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Wen Sun
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China; Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Wanxin Qiao
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Bin Zhou
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Zixuan Hu
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Quanquan Yan
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China; Shanghai Lida Polytechnic Institute, Shanghai, China
| | - Jun Wu
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Rong Wang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Qian Zhang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Dengshun Miao
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China; Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
36
|
Wang H, Lv C, Gu Y, Li Q, Xie L, Zhang H, Miao D, Sun W. Overexpressed Sirt1 in MSCs Promotes Dentin Formation in Bmi1-Deficient Mice. J Dent Res 2018; 97:1365-1373. [PMID: 29932801 DOI: 10.1177/0022034518781509] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Sirt1 promotes odontoblastic gene expression in human dental pulp cells, whereas the inhibition of Sirt1 downregulates the expression of those genes. To investigate whether the overexpression of Sirt1 in mesenchymal stem cells (MSCs) driven by Prx1 promoter could rescue the dentin formation defects in Bmi1-deficient (Bmi1-/-) mice, we established the MSCs overexpressing Sirt1 in Bmi1 knockout mice (Sirt1TGBmi1-/-). First, we used Prx1-Cre/ROSAnTnG mice to demonstrate that Prx1 linage cells exist mainly in the pulp horns at 4 wk of age. Second, we found that 4-wk-old Sirt1TG mice had increased tooth volume as compared with wild-type (WT) littermates. The expression level of Sirt1 was significantly higher in dental papilla mesenchymal cells of Sirt1TG mice than WT mice. Furthermore, we demonstrated that the tooth mineralization, dental volume, dentin sialoprotein-immunopositive areas, odontoblastic gene expression, and percentage of proliferating BrdU-positive cells were significantly elevated in the Sirt1TG mice and dramatically reduced in the Bmi1-/- mice versus the WT littermates at 4 wk of age. However, the areas of predentin and the percentage of TUNEL-positive apoptotic cells were significantly reduced in the Sirt1TG mice but dramatically increased in the Bmi1-/- mice as compared with the WT littermates. All these parameters were rescued in the Sirt1TGBmi1-/- mice versus the Bmi1-/- mice. Finally, by using dental papilla mesenchymal cells, we found that the overexpression of Sirt1 rescued the reduced cell proliferation and differentiation and increased the cell apoptosis caused by Bmi1 deficiency, which was associated with increased p53 deacetylation. Therefore, this study indicates that Sirt1 is a potential therapeutic target for promoting dentin formation in an anabolic approach to the treatment of dental developmental defects.
Collapse
Affiliation(s)
- H Wang
- 1 Jiangsu Key Laboratory of Oral Diseases and Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - C Lv
- 1 Jiangsu Key Laboratory of Oral Diseases and Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,2 Department of Stomatology, Taizhou People's Hospital of Jiangsu Province, Taizhou, China
| | - Y Gu
- 1 Jiangsu Key Laboratory of Oral Diseases and Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Q Li
- 1 Jiangsu Key Laboratory of Oral Diseases and Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - L Xie
- 1 Jiangsu Key Laboratory of Oral Diseases and Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - H Zhang
- 3 Center for Musculoskeletal Research, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - D Miao
- 1 Jiangsu Key Laboratory of Oral Diseases and Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - W Sun
- 1 Jiangsu Key Laboratory of Oral Diseases and Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
37
|
Dai X, Zhang Q, Yu Z, Sun W, Wang R, Miao D. Bmi1 Deficient Mice Exhibit Male Infertility. Int J Biol Sci 2018; 14:358-368. [PMID: 29559852 PMCID: PMC5859480 DOI: 10.7150/ijbs.23325] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 01/21/2018] [Indexed: 12/14/2022] Open
Abstract
Previous studies have demonstrated that the polycomb repressor Bmi1 is universally expressed in all types of testicular cells and might regulate the spermatogonia proliferation, however, it is unclear whether Bmi1 plays a critical role in maintaining normal male fertility in vivo. To answer this question, we first confirmed that Bmi1 is universally expressed in all types of testicular cells and found that the gene relative expression levels of Bmi1 in testis were the highest relative to other organs. Then we investigated the role of Bmi1 in maintaining normal male fertility using Bmi1 knockout male mouse model. Our results demonstrated that Bmi1 deficiency resulted in totally male infertility with smaller testis, severe oligospermia and sperm malformation. Mechanistically, decreased serum testosterone levels with down-regulating 3βHSD and 17βHSD expression levels, reduced germ cell proliferation, increased germ cell apoptosis with up-regulating p16, p19, p53 and p21 expression levels, increased reactive oxygen species (ROS) and H2O2 levels with down-regulating gene expression levels of anti-oxidant enzymes, and increased 8-OHdG and γ.H2AX positive cells in testis were observed in Bmi1 deficient mice compared with wild-type mice. These results indicate that Bmi1 deficiency results in male infertility by reducing testosterone syntheses, increasing oxidative stress and DNA damage, activating p16 and p19 signaling pathway, inhibiting germ cell proliferation and inducing germ cell apoptosis and sperm malformation. Thus, Bmi1 may be a novel and potential target for the clinic treatment of male infertility.
Collapse
Affiliation(s)
- Xiuliang Dai
- Department of Reproductive Medicine Center, Affiliated Changzhou Maternity and Child Health Care Hospital, Nanjing Medical University, Changzhou, Jiangsu, China.,State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qian Zhang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhenzhen Yu
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiwei Sun
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rong Wang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dengshun Miao
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
38
|
García-Giménez JL, Rubio-Belmar PA, Peiró-Chova L, Hervás D, González-Rodríguez D, Ibañez-Cabellos JS, Bas-Hermida P, Mena-Mollá S, García-López EM, Pallardó FV, Bas T. Circulating miRNAs as diagnostic biomarkers for adolescent idiopathic scoliosis. Sci Rep 2018; 8:2646. [PMID: 29422531 PMCID: PMC5805715 DOI: 10.1038/s41598-018-21146-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 01/31/2018] [Indexed: 11/09/2022] Open
Abstract
The aetiology of adolescent idiopathic scoliosis (AIS) has been linked to many factors, such as asymmetric growth, neuromuscular condition, bone strength and genetic background. Recently, epigenetic factors have been proposed as contributors of AIS physiopathology, but information about the molecular mechanisms and pathways involved is scarce. Regarding epigenetic factors, microRNAs (miRNAs) are molecules that contribute to gene expression modulation by regulating important cellular pathways. We herein used Next-Generation Sequencing to discover a series of circulating miRNAs detected in the blood samples of AIS patients, which yielded a unique miRNA biomarker signature that diagnoses AIS with high sensitivity and specificity. We propose that these miRNAs participate in the epigenetic control of signalling pathways by regulating osteoblast and osteoclast differentiation, thus modulating the genetic background of AIS patients. Our study yielded two relevant results: 1) evidence for the deregulated miRNAs that participate in osteoblast/osteoclast differentiation mechanisms in AIS; 2) this miRNA-signature can be potentially used as a clinical tool for molecular AIS diagnosis. Using miRNAs as biomarkers for AIS diagnostics is especially relevant since miRNAs can serve for early diagnoses and for evaluating the positive effects of applied therapies to therefore reduce the need of high-risk surgical interventions.
Collapse
Affiliation(s)
- José Luis García-Giménez
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain.
- Instituto de Investigación Sanitaria INCLIVA, Avenida de Menéndez y Pelayo, 4, 46010, Valencia, Spain.
- Dept. Physiology. Faculty of Medicine and Dentistry, University of Valencia, Av/Blasco Ibañez, 15, 46010, Valencia, Spain.
| | - Pedro Antonio Rubio-Belmar
- Instituto de Investigación Sanitaria IISLAFE, Av/Fernando Abril Martorell, 106. Torre A 7, 46026, Valencia, Spain
- Unidad de Raquis. Hospital Universitari i Politècnic La Fe, Av/Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Lorena Peiró-Chova
- Instituto de Investigación Sanitaria INCLIVA, Avenida de Menéndez y Pelayo, 4, 46010, Valencia, Spain
| | - David Hervás
- Unidad de Bioestadística, Instituto de Investigación Sanitaria IISLAFE, Av/Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Daymé González-Rodríguez
- Instituto de Investigación Sanitaria INCLIVA, Avenida de Menéndez y Pelayo, 4, 46010, Valencia, Spain
| | - José Santiago Ibañez-Cabellos
- Instituto de Investigación Sanitaria INCLIVA, Avenida de Menéndez y Pelayo, 4, 46010, Valencia, Spain
- Dept. Physiology. Faculty of Medicine and Dentistry, University of Valencia, Av/Blasco Ibañez, 15, 46010, Valencia, Spain
| | - Paloma Bas-Hermida
- Instituto de Investigación Sanitaria IISLAFE, Av/Fernando Abril Martorell, 106. Torre A 7, 46026, Valencia, Spain
- Unidad de Raquis. Hospital Universitari i Politècnic La Fe, Av/Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Salvador Mena-Mollá
- Dept. Physiology. Faculty of Medicine and Dentistry, University of Valencia, Av/Blasco Ibañez, 15, 46010, Valencia, Spain
| | - Eva María García-López
- Instituto de Investigación Sanitaria INCLIVA, Avenida de Menéndez y Pelayo, 4, 46010, Valencia, Spain
- Dept. Physiology. Faculty of Medicine and Dentistry, University of Valencia, Av/Blasco Ibañez, 15, 46010, Valencia, Spain
| | - Federico V Pallardó
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain
- Instituto de Investigación Sanitaria INCLIVA, Avenida de Menéndez y Pelayo, 4, 46010, Valencia, Spain
- Dept. Physiology. Faculty of Medicine and Dentistry, University of Valencia, Av/Blasco Ibañez, 15, 46010, Valencia, Spain
| | - Teresa Bas
- Instituto de Investigación Sanitaria IISLAFE, Av/Fernando Abril Martorell, 106. Torre A 7, 46026, Valencia, Spain
- Unidad de Raquis. Hospital Universitari i Politècnic La Fe, Av/Fernando Abril Martorell, 106, 46026, Valencia, Spain
| |
Collapse
|
39
|
Wang Y, Zhang H, Yang Z, Miao D, Zhang D. Rho Kinase Inhibitor, Fasudil, Attenuates Contrast‐induced Acute Kidney Injury. Basic Clin Pharmacol Toxicol 2017; 122:278-287. [PMID: 28929640 DOI: 10.1111/bcpt.12895] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/21/2017] [Indexed: 01/10/2023]
Affiliation(s)
- Yiming Wang
- Department of Cardiology First Affiliated Hospital of Nanjing Medical University Nanjing China
- Department of Cardiology Zhenjiang First People's Hospital Zhenjiang China
| | - Hao Zhang
- Department of Cardiology First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Zhijian Yang
- Department of Cardiology First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Dengshun Miao
- Research Center for Bone and Stem Cells Nanjing Medical University Nanjing China
| | - Dingguo Zhang
- Department of Cardiology First Affiliated Hospital of Nanjing Medical University Nanjing China
| |
Collapse
|
40
|
Jin J, Tao J, Gu X, Yu Z, Wang R, Zuo G, Li Q, Lv X, Miao D. P16 INK4a Deletion Ameliorated Renal Tubulointerstitial Injury in a Stress-induced Premature Senescence Model of Bmi-1 Deficiency. Sci Rep 2017; 7:7502. [PMID: 28790310 PMCID: PMC5548892 DOI: 10.1038/s41598-017-06868-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 06/20/2017] [Indexed: 12/16/2022] Open
Abstract
To determine whether p16 INK4a deletion ameliorated renal tubulointerstitial injury by inhibiting a senescence-associated secretory phenotype (SASP) in Bmi-1-deficient (Bmi-1 -/-) mice, renal phenotypes were compared among 5-week-old Bmi-1 and p16 INK4a double-knockout, and Bmi-1 -/- and wild-type mice. Fifth-passage renal interstitial fibroblasts (RIFs) from the three groups were analyzed for senescence and proliferation. The effect of Bmi-1 deficiency on epithelial-to-mesenchymal transition (EMT) was examined in Bmi-1-knockdown human renal proximal tubular epithelial (HK2) cells, which were treated with concentrated conditioned medium (CM) from the fifth-passage renal interstitial fibroblasts (RIFs) of above three group mice or with exogenous TGF-β1. Our results demonstrated that p16 INK4a deletion largely rescued renal aging phenotypes caused by Bmi-1 deficiency, including impaired renal structure and function, decreased proliferation, increased apoptosis, senescence and SASP, DNA damage, NF-κB and TGF-β1/Smad signal activation, inflammatory cell infiltration, and tubulointerstitial fibrosis and tubular atrophy. P16 INK4a deletion also promoted proliferation, reduced senescence and SASP of RIFs and subsequently inhibited EMT of Bmi-1-knockdown HK2 cells. TGF-β1 further induced the EMT of Bmi-1-knockdown HK2 cells. Thus, p16 INK4a positive senescent cells would be a therapeutic target for preventing renal tubulointerstitial injury.
Collapse
MESH Headings
- Acute Kidney Injury/genetics
- Acute Kidney Injury/metabolism
- Acute Kidney Injury/pathology
- Acute Kidney Injury/prevention & control
- Animals
- Cell Line, Transformed
- Cell Proliferation
- Cellular Senescence
- Coculture Techniques
- Culture Media, Conditioned/pharmacology
- Cyclin-Dependent Kinase Inhibitor p16/deficiency
- Cyclin-Dependent Kinase Inhibitor p16/genetics
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Epithelial-Mesenchymal Transition/genetics
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Gene Expression Regulation
- Humans
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Mice
- Mice, Knockout
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Nephritis, Interstitial/genetics
- Nephritis, Interstitial/metabolism
- Nephritis, Interstitial/pathology
- Nephritis, Interstitial/prevention & control
- Polycomb Repressive Complex 1/antagonists & inhibitors
- Polycomb Repressive Complex 1/deficiency
- Polycomb Repressive Complex 1/genetics
- Polycomb Repressive Complex 1/metabolism
- Proto-Oncogene Proteins/deficiency
- Proto-Oncogene Proteins/genetics
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Smad Proteins/genetics
- Smad Proteins/metabolism
- Transforming Growth Factor beta1/genetics
- Transforming Growth Factor beta1/metabolism
- Transforming Growth Factor beta1/pharmacology
Collapse
Affiliation(s)
- Jianliang Jin
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jianguo Tao
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xin Gu
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Zhenzhen Yu
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Rong Wang
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Guoping Zuo
- Laboratory Centre for Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Qing Li
- Department of Science and Technology, Jiangsu Jiankang Vocational College, Nanjing, Jiangsu, 210029, China
| | - Xianhui Lv
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Dengshun Miao
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
41
|
Sabbieti MG, Marchetti L, Censi R, Lacava G, Agas D. Role of PTH in Bone Marrow Niche and HSC Regulation. CURRENT STEM CELL REPORTS 2017. [DOI: 10.1007/s40778-017-0091-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
42
|
Li J, Wang Q, Yang R, Zhang J, Li X, Zhou X, Miao D. BMI-1 Mediates Estrogen-Deficiency-Induced Bone Loss by Inhibiting Reactive Oxygen Species Accumulation and T Cell Activation. J Bone Miner Res 2017; 32:962-973. [PMID: 27943387 DOI: 10.1002/jbmr.3059] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 12/03/2016] [Accepted: 12/08/2016] [Indexed: 11/07/2022]
Abstract
Previous studies have shown that estrogen regulates bone homeostasis through regulatory effects on oxidative stress. However, it is unclear how estrogen deficiency triggers reactive oxygen species (ROS) accumulation. Recent studies provide evidence that the B lymphoma Mo-MLV insertion region 1 (BMI-1) plays a critical role in protection against oxidative stress and that this gene is directly regulated by estrogen via estrogen receptor (ER) at the transcriptional level. In this study, ovariectomized mice were given drinking water with/without antioxidant N-acetyl-cysteine (NAC, 1 mg/mL) supplementation, and compared with each other and with sham mice. Results showed that ovariectomy resulted in bone loss with increased osteoclast surface, increased ROS levels, T cell activation, and increased TNF and RANKL levels in serum and in CD4 T cells; NAC supplementation largely prevented these alterations. BMI-1 expression levels were dramatically downregulated in CD4 T cells from ovariectomized mice. We supplemented drinking water to BMI-1-deficient mice with/without NAC and compared them with each other and with wild-type (WT) mice. We found that BMI-1 deficiency mimicked alterations observed in ovariectomy whereas NAC supplementation reversed all alterations induced by BMI-1 deficiency. Because T cells are critical in mediating ovariectomy-induced bone loss, we further assessed whether BMI-1 overexpression in lymphocytes can protect against estrogen deficiency-induced osteoclastogenesis and bone loss by inhibiting oxidative stress, T cell activation, and RANKL production. When WT and Eμ-BMI-1 transgenic mice with BMI-1 specifically overexpressed in lymphocytes were ovariectomized and compared with each other and with WT sham mice, we found that BMI-1 overexpression in lymphocytes clearly reversed all alterations induced by ovariectomy. Results from this study indicate that estrogen deficiency downregulates BMI-1 and subsequently increases ROS, T cell activation, and RANKL production in T cells, thus enhancing osteoclastogenesis and accelerating bone loss. This study clarifies a novel mechanism regulating estrogen deficiency-induced bone loss. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jinbo Li
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qian Wang
- Department of Human Anatomy, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Renlei Yang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaqi Zhang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xing Li
- Department of Immuno-oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xichao Zhou
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dengshun Miao
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
43
|
Lv X, Yu Z, Xie C, Dai X, Li Q, Miao D, Jin J. Bmi-1 plays a critical role in the protection from acute tubular necrosis by mobilizing renal stem/progenitor cells. Biochem Biophys Res Commun 2016; 482:742-749. [PMID: 27871857 DOI: 10.1016/j.bbrc.2016.11.105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 11/17/2016] [Indexed: 02/06/2023]
Abstract
The regeneration of injured tubular cell occurs primarily from intrinsic renal stem/progenitor cells (RSCs) labeled with CD24 and CD133 after acute tubular necrosis (ATN). Bmi-1 plays a crucial role in regulating self-renewal, differentiation and aging of multiple adult stem cells and progenitor cells. Bmi-1 was rapidly elevated in the induction of adult kidney regeneration by renal injury. To determine whether Bmi-1 maintained mobilization of RSCs in the protection from ATN, glycerol-rhabdomyolysis-induced ATN were performed in wild type (WT) and Bmi-1-deficient (Bmi-1-/-) mice. Their ATN phenotypes were analyzed; CD24 and CD133 double positive (CD24+CD133+) cells were measured; and the levels of serum urea nitrogen (SUN) and serum creatinine (SCr) were detected. We found that CD24+CD133+ RSCs were mobilized in WT ATN mice with the increased expression of Bmi-1; Bmi-1 deficiency led to increased tubular cast formation and necrosis, elevated levels of SUN and SCr, decreased tubular proliferation, and immobilized ratio of RSCs in ATN. These findings indicated that Bmi-1 played a critical role in the protection from ATN by maintaining mobilization of RSCs and would be a novel therapeutic target for preventing the progression of ATN.
Collapse
Affiliation(s)
- Xianhui Lv
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Zhenzhen Yu
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Chunfeng Xie
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiuliang Dai
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Qing Li
- Department of Science and Technology, Jiangsu Jiankang Vocational College, Nanjing, Jiangsu, 210029, China
| | - Dengshun Miao
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jianliang Jin
- Research Centre for Bone and Stem Cells, Department of Human Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
44
|
1, 25-dihydroxy-vitamin D3 with tumor necrosis factor-alpha protects against rheumatoid arthritis by promoting p53 acetylation-mediated apoptosis via Sirt1 in synoviocytes. Cell Death Dis 2016; 7:e2423. [PMID: 27763638 PMCID: PMC5133971 DOI: 10.1038/cddis.2016.300] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/14/2016] [Accepted: 08/25/2016] [Indexed: 01/09/2023]
Abstract
Impaired apoptosis of fibroblast-like synoviocytes (FLSs) causes synovial hyperplasia, facilitating destruction of cartilage and bone in rheumatoid arthritis (RA). Tumor necrosis factor (TNF)-α, a dominant inflammatory mediator in RA pathogenesis, promotes progression of RA symptoms. Prevalence of 1, 25-dihydroxy-vitamin D3 (hereafter termed VD) deficiency is 30-63% in patients with RA. Whether VD leads to apoptosis or enhances TNF-α-mediated apoptosis in FLSs to ameliorate RA is unclear. To determine this, 10-week-old CYP27B1-deficient (CYP27B1-/-) mice with collagen-induced arthritis (CIA) were intraperitoneally treated with 1 μg/kg VD every other day for 9 weeks. RA phenotypes were compared between vehicle-treated CYP27B1-/- and wild-type CIA mice. Human rheumatoid FLS-MH7A cells were treated with Dulbecco's modified Eagle's medium (DMEM) without fetal bovine serum (FBS) for 24 h, then with different concentrations of VD and TNF-α, human vitamin D receptor (VDR) siRNA or the p53 pro-apoptotic inhibitor pifithrin-α. Apoptosis and p53 pro-apoptotic signaling were analyzed. The 19-week-old vehicle-treated CYP27B1-/- CIA mice had increased cumulative arthritis scores and levels of serous rheumatoid factors and C-reactive protein. They had exacerbated articular cartilage and bone destruction, joint space narrowing, joint stiffness, deformity and dysfunction, synovitis and TNF-α secretion, FLS hyperplasia with increased proliferation and decreased apoptosis compared to CIA mice. These RA phenotypes that were aggravated in CIA mice by CYP27B1 deficiency were largely rescued by VD treatment. In vitro, VD with TNF-α treatment upregulated p53 acetylation-mediated apoptosis in MH7A cells by promoting Sirt1 translocation from the nucleus to the cytoplasm. These findings indicated that VD with TNF-α protected against RA by promoting apoptosis of FLSs. The results indicated that clinical administration of VD could be a specific therapy to promote FLS apoptosis and prevent RA progression.
Collapse
|
45
|
Zhou X, Dai X, Wu X, Ji J, Karaplis A, Goltzman D, Yang X, Miao D. Overexpression of Bmi1 in Lymphocytes Stimulates Skeletogenesis by Improving the Osteogenic Microenvironment. Sci Rep 2016; 6:29171. [PMID: 27373231 PMCID: PMC4931581 DOI: 10.1038/srep29171] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 06/15/2016] [Indexed: 12/13/2022] Open
Abstract
To investigate whether overexpression of Bmi1 in lymphocytes can stimulate skeletogenesis by improving the osteogenic microenvironment, we examined the skeletal phenotype of EμBmi1 transgenic mice with overexpression of Bmi1 in lymphocytes. The size of the skeleton, trabecular bone volume and osteoblast number, indices of proliferation and differentiation of bone marrow mesenchymal stem cells (BM-MSCs) were increased significantly, ROS levels were reduced and antioxidative capacity was enhanced in EμBmi1 mice compared to WT mice. In PTHrP1-84 knockin (Pthrp(KI/KI)) mice, the expression levels of Bmi1 are reduced and potentially can mediate the premature osteoporosis observed. We therefore generated a Pthrp(KI/KI) mice overexpressing Bmi1 in lymphocytes and compared them with Pthrp(KI/KI) and WT littermates. Overexpression of Bmi1 in Pthrp(KI/KI) mice resulted in a longer lifespan, increased body weight and improvement in skeletal growth and parameters of osteoblastic bone formation with reduced ROS levels and DNA damage response parameters. Our results demonstrate that overexpression of Bmi1 in lymphocytes can stimulate osteogenesis in vivo and partially rescue defects in skeletal growth and osteogenesis in Pthrp(KI/KI) mice. These studies therefore indicate that overexpression of Bmi1 in lymphocytes can stimulate skeletogenesis by inhibiting oxidative stress and improving the osteogenic microenvironment.
Collapse
Affiliation(s)
- Xichao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Xiuliang Dai
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Xuan Wu
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Ji Ji
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
- Department of Fundamentals of Nursing, School of Nursing, Nanjing Medical University, Nanjing, China
| | - Andrew Karaplis
- The Department of Medicine, McGill University, Montreal, Canada
| | - David Goltzman
- The Department of Medicine, McGill University, Montreal, Canada
| | - Xiangjiao Yang
- The Department of Medicine, McGill University, Montreal, Canada
- Rosalind & Morris Goodman Cancer Research Center, Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Dengshun Miao
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
46
|
Becker M, Potapenko T, Niklaus A, Bieback K, Ho AD, Müller AM. Polycomb Protein BMI1 Regulates Osteogenic Differentiation of Human Adipose Tissue-Derived Mesenchymal Stem Cells Downstream of GSK3. Stem Cells Dev 2016; 25:922-33. [PMID: 27100571 DOI: 10.1089/scd.2015.0277] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Polycomb proteins such as the B lymphoma Mo-MLV insertion region 1 homolog (BMI1) are essential chromatin factors for the self-renewal and differentiation of embryonic and adult stem cells. BMI1 also plays a critical role in osteogenesis as Bmi1-deficient mice display a skeletal phenotype caused by the exhaustion of the mesenchymal stem cell pool. In this study, we have studied the role of BMI1 in the osteogenic differentiation of human adipose tissue-derived mesenchymal stem cells (hASCs). BMI1 protein, but not RNA levels, increases during in vitro osteogenic differentiation of hASCs. Overexpression of BMI1 leads to an osteogenic priming of hASCs under nondifferentiating conditions and enhanced osteogenesis upon differentiation, along with increased BMP2 and WNT11 expressions. Conversely, knockdown of BMI1 expression reduces osteogenic differentiation. Furthermore, our studies indicate that during osteogenic differentiation of hASCs, BMI1 is a downstream target of GSK3 signaling. BMI1, therefore, acts as a pro-osteogenic differentiation factor in hASCs and hence it is a promising target for active modulation of hASC-derived osteogenesis.
Collapse
Affiliation(s)
- Matthias Becker
- 1 Institute for Medical Radiation and Cell Research (MSZ), Center of Experimental Molecular Medicine (ZEMM) , Würzburg, Germany
| | - Tamara Potapenko
- 1 Institute for Medical Radiation and Cell Research (MSZ), Center of Experimental Molecular Medicine (ZEMM) , Würzburg, Germany
| | - Andrea Niklaus
- 1 Institute for Medical Radiation and Cell Research (MSZ), Center of Experimental Molecular Medicine (ZEMM) , Würzburg, Germany
| | - Karen Bieback
- 2 Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg , German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Anthony D Ho
- 3 Department of Internal Medicine V, Heidelberg University Hospital , Heidelberg, Germany
| | - Albrecht M Müller
- 1 Institute for Medical Radiation and Cell Research (MSZ), Center of Experimental Molecular Medicine (ZEMM) , Würzburg, Germany
| |
Collapse
|
47
|
Sun W, Wu J, Huang L, Liu H, Wang R, Karaplis A, Goltzman D, Miao D. PTHrP Nuclear Localization and Carboxyl Terminus Sequences Modulate Dental and Mandibular Development in Part via the Action of p27. Endocrinology 2016; 157:1372-84. [PMID: 26859332 DOI: 10.1210/en.2015-1555] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
To determine whether the action of the PTHrP nuclear localization sequence and C terminus is mediated through p27 in modulating dental and mandibular development, compound mutant mice, which are homozygous for both p27 deletion and the PTHrP1-84 knock-in mutation (p27(-/-)Pthrp(KI/KI)), were generated. Their teeth and mandibular phenotypes were compared with those of p27(-/-), Pthrp(KI/KI), and wild-type mice. At 2 weeks of age, the mandibular mineral density, alveolar bone volume, osteoblast numbers, and dental volume, dentin sialoprotein-immunopositive areas in the first molar were increased significantly in p27(-/-) mice and decreased dramatically in both Pthrp(KI/KI) and p27(-/-) Pthrp(KI/KI) mice compared with wild-type mice; however, these parameters were partly rescued in p27(-/-) Pthrp(KI/KI) mice compared with Pthrp(KI/KI) mice. These data demonstrate that the deletion of p27 in Pthrp(KI/KI) mice can partially rescue defects in dental and mandibular development. Furthermore, we found that deletion of p27 in Pthrp(KI/KI) mice partially corrected the dental and mandibular phenotype by modulating cell cyclin-regulating molecules and antioxidant enzymes. This study therefore indicates that the p27 pathway may function downstream in the action of PTHrP nuclear localization sequence to regulate dental and mandibular development.
Collapse
Affiliation(s)
- Wen Sun
- State Key Laboratory of Reproductive Medicine (W.S., J.W., L.H., H.L., R.W., D.M.), The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China; and Lady Davis Institute for Medical Research (A.K.), Jewish General Hospital, and Calcium Research Laboratory (D.G.), McGill University Health Centre and Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A1
| | - Jun Wu
- State Key Laboratory of Reproductive Medicine (W.S., J.W., L.H., H.L., R.W., D.M.), The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China; and Lady Davis Institute for Medical Research (A.K.), Jewish General Hospital, and Calcium Research Laboratory (D.G.), McGill University Health Centre and Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A1
| | - Linying Huang
- State Key Laboratory of Reproductive Medicine (W.S., J.W., L.H., H.L., R.W., D.M.), The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China; and Lady Davis Institute for Medical Research (A.K.), Jewish General Hospital, and Calcium Research Laboratory (D.G.), McGill University Health Centre and Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A1
| | - Hong Liu
- State Key Laboratory of Reproductive Medicine (W.S., J.W., L.H., H.L., R.W., D.M.), The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China; and Lady Davis Institute for Medical Research (A.K.), Jewish General Hospital, and Calcium Research Laboratory (D.G.), McGill University Health Centre and Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A1
| | - Rong Wang
- State Key Laboratory of Reproductive Medicine (W.S., J.W., L.H., H.L., R.W., D.M.), The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China; and Lady Davis Institute for Medical Research (A.K.), Jewish General Hospital, and Calcium Research Laboratory (D.G.), McGill University Health Centre and Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A1
| | - Andrew Karaplis
- State Key Laboratory of Reproductive Medicine (W.S., J.W., L.H., H.L., R.W., D.M.), The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China; and Lady Davis Institute for Medical Research (A.K.), Jewish General Hospital, and Calcium Research Laboratory (D.G.), McGill University Health Centre and Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A1
| | - David Goltzman
- State Key Laboratory of Reproductive Medicine (W.S., J.W., L.H., H.L., R.W., D.M.), The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China; and Lady Davis Institute for Medical Research (A.K.), Jewish General Hospital, and Calcium Research Laboratory (D.G.), McGill University Health Centre and Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A1
| | - Dengshun Miao
- State Key Laboratory of Reproductive Medicine (W.S., J.W., L.H., H.L., R.W., D.M.), The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China; and Lady Davis Institute for Medical Research (A.K.), Jewish General Hospital, and Calcium Research Laboratory (D.G.), McGill University Health Centre and Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A1
| |
Collapse
|
48
|
Bolomsky A, Schlangen K, Schreiner W, Zojer N, Ludwig H. Targeting of BMI-1 with PTC-209 shows potent anti-myeloma activity and impairs the tumour microenvironment. J Hematol Oncol 2016; 9:17. [PMID: 26935956 PMCID: PMC4776359 DOI: 10.1186/s13045-016-0247-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/23/2016] [Indexed: 11/30/2022] Open
Abstract
Background The polycomb complex protein BMI-1 (BMI-1) is a putative oncogene reported to be overexpressed in multiple myeloma (MM). Silencing of BMI-1 was shown to impair the growth and survival of MM cells. However, therapeutic agents specifically targeting BMI-1 were not available so far. Here, we investigated PTC-209, a novel small molecule inhibitor of BMI-1, for its activity in MM. Methods BMI-1 expression was analysed in human MM cell lines and primary MM cells by using publically available gene expression profiling (GEP) data. The anti-MM activity of PTC-209 was investigated by viability testing, cell cycle analysis, annexin V and 7-AAD staining, quantification of cleaved poly(ADP-ribose) polymerase (PARP), JC-1 as well as colony formation assays. Deregulation of central myeloma growth and survival genes was studied by quantitative PCR and flow cytometry, respectively. In addition, the impact of PTC-209 on in vitro osteoclast, osteoblast and tube formation was analysed. Results We confirmed overexpression of BMI-1 in MM patients by using publically available GEP datasets. Of note, BMI-1 expression was further increased at relapse which translated into significantly shorter overall survival in relapsed/refractory patients treated with bortezomib or dexamethasone. Treatment with PTC-209 significantly decreased viable cell numbers in human MM cell lines, induced a G1 cell cycle arrest, promoted apoptosis and demonstrated synergistic activity with pomalidomide and carfilzomib. The anti-MM activity of PTC-209 was accompanied by a significant decrease of cyclin D1 (CCND1) and v-myc avian myelocytomatosis viral oncogene homolog (MYC) expression as well as upregulation of cyclin-dependent kinase inhibitor 1A (CDKN1A) and cyclin-dependent kinase inhibitor 1B (CDKN1B). We also observed upregulation of NOXA (up to 3.6 ± 1.2-fold induction, P = 0.009) and subsequent downregulation of myeloid cell leukemia 1 (MCL-1) protein levels, which likely mediates the apoptotic effects of PTC-209. Importantly, the anti-MM activity was upheld in the presence of stromal support or myeloma growth factors insulin-like growth factor 1 (IGF-1) and interleukin 6 (IL-6). In the MM microenvironment, PTC-209 impaired tube formation, impaired osteoclast development and decreased osteoblast formation in a dose-dependent manner (P < 0.01 at 1 μM, respectively). The latter might be attributed to an induction of DKK1 and was reversed by concurrent anti-DKK1 antibody treatment. Conclusions We confirmed overexpression of BMI-1 in MM highlighting its role as an attractive drug target and reveal therapeutic targeting of BMI-1 by PTC-209 as a promising novel therapeutic intervention for MM. Electronic supplementary material The online version of this article (doi:10.1186/s13045-016-0247-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Arnold Bolomsky
- Wilhelminen Cancer Research Institute, Department of Medicine I, Wilhelminenspital, Montleartstraße 37, 1160, Vienna, Austria.
| | - Karin Schlangen
- Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Vienna, Austria.
| | - Wolfgang Schreiner
- Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Vienna, Austria.
| | - Niklas Zojer
- Wilhelminen Cancer Research Institute, Department of Medicine I, Wilhelminenspital, Montleartstraße 37, 1160, Vienna, Austria.
| | - Heinz Ludwig
- Wilhelminen Cancer Research Institute, Department of Medicine I, Wilhelminenspital, Montleartstraße 37, 1160, Vienna, Austria.
| |
Collapse
|
49
|
Zhu M, Zhang J, Dong Z, Zhang Y, Wang R, Karaplis A, Goltzman D, Miao D. The p27 Pathway Modulates the Regulation of Skeletal Growth and Osteoblastic Bone Formation by Parathyroid Hormone-Related Peptide. J Bone Miner Res 2015; 30:1969-79. [PMID: 25917430 DOI: 10.1002/jbmr.2544] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 04/15/2015] [Accepted: 04/23/2015] [Indexed: 01/14/2023]
Abstract
Parathyroid hormone-related peptide (PTHrP) 1-84 knock-in mice (Pthrp KI) develop skeletal growth retardation and defective osteoblastic bone formation. To further examine the mechanisms underlying this phenotype, microarray analyses of differential gene expression profiles were performed in long bone extracts from Pthrp KI mice and their wild-type (WT) littermates. We found that the expression levels of p27, p16, and p53 were significantly upregulated in Pthrp KI mice relative to WT littermates. To determine whether p27 was involved in the regulation by PTHrP of skeletal growth and development in vivo, we generated compound mutant mice, which were homozygous for both p27 deletion and the Pthrp KI mutation (p27(-/-) Pthrp KI). We then compared p27(-/-) Pthrp KI mice with p27(-/-), Pthrp KI, and WT littermates. Deletion of p27 in Pthrp KI mice resulted in a longer lifespan, increased body weight, and improvement in skeletal growth. At 2 weeks of age, skeletal parameters, including length of long bones, size of epiphyses, numbers of proliferating cell nuclear antigen (PCNA)-positive chondrocytes, bone mineral density, trabecular bone volume, osteoblast numbers, and alkaline phosphatase (ALP)-, type I collagen-, and osteocalcin-positive bone areas were increased in p27(-/-) mice and reduced in both Pthrp KI and p27(-/-) Pthrp KI mice compared with WT mice; however, these parameters were increased in p27(-/-) Pthrp KI mice compared with Pthrp KI mice. As well, protein expression levels of PTHR, IGF-1, and Bmi-1, and the numbers of total colony-forming unit fibroblastic (CFU-f) and ALP-positive CFU-f were similarly increased in p27(-/-) Pthrp KI mice compared with Pthrp KI mice. Our results demonstrate that deletion of p27 in Pthrp KI mice can partially rescue defects in skeletal growth and osteoblastic bone formation by enhancing endochondral bone formation and osteogenesis. These studies, therefore, indicate that the p27 pathway may function downstream in the action of PTHrP to regulate skeletal growth and development.
Collapse
Affiliation(s)
- Min Zhu
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jing Zhang
- Department of Human Anatomy, Basic Medical College of Nanchang University, Nanchang, People's Republic of China
| | - Zhan Dong
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Ying Zhang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Rong Wang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Andrew Karaplis
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Canada
| | - David Goltzman
- Calcium Research Laboratory, McGill University Health Centre and Department of Medicine, McGill University, Montreal, Canada
| | - Dengshun Miao
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology, and Embryology, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
50
|
Xie C, Jin J, Lv X, Tao J, Wang R, Miao D. Anti-aging Effect of Transplanted Amniotic Membrane Mesenchymal Stem Cells in a Premature Aging Model of Bmi-1 Deficiency. Sci Rep 2015; 5:13975. [PMID: 26370922 PMCID: PMC4570627 DOI: 10.1038/srep13975] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 08/12/2015] [Indexed: 02/01/2023] Open
Abstract
To determine whether transplanted amniotic membrane mesenchymal stem cells (AMSCs) ameliorated the premature senescent phenotype of Bmi-1-deficient mice, postnatal 2-day-old Bmi-1(-/-) mice were injected intraperitoneally with the second-passage AMSCs from amniotic membranes of β-galactosidase (β-gal) transgenic mice or wild-type (WT) mice labeled with DiI. Three reinjections were given, once every seven days. Phenotypes of 5-week-old β-gal(+) AMSC-transplanted or 6-week-old DiI(+) AMSC-transplanted Bmi-1(-/-) mice were compared with vehicle-transplanted Bmi-1(-/-) and WT mice. Vehicle-transplanted Bmi-1(-/-) mice displayed growth retardation and premature aging with decreased cell proliferation and increased cell apoptosis; a decreased ratio and dysmaturity of lymphocytic series; premature osteoporosis with reduced osteogenesis and increased adipogenesis; redox imbalance and DNA damage in multiple organs. Transplanted AMSCs carried Bmi-1 migrated into multiple organs, proliferated and differentiated into multiple tissue cells, promoted growth and delayed senescence in Bmi-1(-/-) transplant recipients. The dysmaturity of lymphocytic series were ameliorated, premature osteoporosis were rescued by promoting osteogenesis and inhibiting adipogenesis, the oxidative stress and DNA damage in multiple organs were inhibited by the AMSC transplantation in Bmi-1(-/-) mice. These findings indicate that AMSC transplantation ameliorated the premature senescent phenotype of Bmi-1-deficient mice and could be a novel therapy to delay aging and prevent aging-associated degenerative diseases.
Collapse
Affiliation(s)
- Chunfeng Xie
- The State Key Laboratory of Reproductive Medicine, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Jianliang Jin
- The State Key Laboratory of Reproductive Medicine, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Xianhui Lv
- The State Key Laboratory of Reproductive Medicine, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Jianguo Tao
- The State Key Laboratory of Reproductive Medicine, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Rong Wang
- The State Key Laboratory of Reproductive Medicine, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Dengshun Miao
- The State Key Laboratory of Reproductive Medicine, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| |
Collapse
|