1
|
Cao R, Li H, Liu G, Yan P, Zhang J, Chen Y, Duan X, Zhao Y, Lei Y, Liu C, Guan H, Xing F, Li Y, Wang K, Kong N, Tian R, Yang P. Aging and autophagic phenotypic changes in bone marrow mesenchymal stem cells in glucocorticoid-induced osteonecrosis. Int Immunopharmacol 2025; 152:114389. [PMID: 40073811 DOI: 10.1016/j.intimp.2025.114389] [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: 08/19/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Glucocorticoid (GC) overuse is the main cause of osteonecrosis of the femoral head (ONFH). The dysfunction of bone marrow mesenchymal stem cells (BMSCs) plays an important role in ONFH pathogenesis. Physiological concentrations of GCs can induce the osteogenic differentiation of BMSCs; however, intervention with high concentrations of GC may lead to changes in aging and autophagy in certain cell types. METHODS We generated an ONFH mouse model by injecting C57BL/6 J mice with MPS. BMSCs were harvested from the femora and tibiae of mice and were analyzed for osteogenesis, adipogenesis, senescence, and cell proliferation. In vitro, BMSCs were treated with different concentrations of GC for 48 h, followed by functional analyses to identify differentially expressed genes (DEGs) associated with ONFH. Additionally, various bioinformatics analyses were performed to identify differentially expressed genes in ONFH. RESULTS BMSCs from ONFH mice showed signs of aging, as indicated by increased SA-β-gal positive cells (4.4-fold) and upregulated p53 (2.6-fold) and p21 (2.0-fold) protein expression. It is also accompanied by changes in osteogenic/lipogenic differentiation ability. Bioinformatics analysis further verified these findings. High-dose GC stimulation significantly induced cellular senescence of BMSCs, as indicated by an increase in SA-β-gal positive cells (6.2-fold) and a decrease in autophagy levels. GC stimulation changes the differentiation fate of BMSCs. CONCLUSIONS Our results indicated that GC-induced ONFH was associated with changes in aging and autophagy in BMSCs. GC not only directly affected the osteogenic differentiation of BMSCs but also indirectly affected their differentiation fate through aging and autophagy changes.
Collapse
Affiliation(s)
- Ruomu Cao
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Heng Li
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Guanzhi Liu
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Peng Yan
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiewen Zhang
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yang Chen
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xudong Duan
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yiwei Zhao
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yutian Lei
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chenkun Liu
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huanshuai Guan
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fangze Xing
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yiyang Li
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kunzheng Wang
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ning Kong
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Run Tian
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Pei Yang
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
2
|
Bai C, Tian B, Zhang M, Qin Q, Shi X, Yang X, Gao X, Zhou X, Shan H, Bai J. Targeting NAMPT-OPA1 for treatment of senile osteoporosis. Aging Cell 2025; 24:e14400. [PMID: 39543818 PMCID: PMC11896342 DOI: 10.1111/acel.14400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024] Open
Abstract
Senescence of bone marrow mesenchymal stem cells (BMSCs) impairs their stemness and osteogenic differentiation, which is the principal cause of senile osteoporosis (SOP). Imbalances in nicotinamide phosphoribosyltransferase (NAMPT) homeostasis have been linked to aging and various diseases. Herein, reduction of NAMPT and impaired osteogenesis were observed in BMSCs from aged human and mouse. Knockdown of Nampt in BMSCs promotes lipogenic differentiation and increases age-related bone loss. Overexpression of Nampt ameliorates the senescence-associated (SA) phenotypes in BMSCs derived from aged mice, as well as promoting osteogenic potential. Mechanistically, NAMPT inhibits BMSCs senescence by facilitating OPA1 expression, which is essential for mitochondrial dynamics. The defect of NAMPT reduced mitochondrial membrane potential, interfered with mitochondrial fusion,and increased SA protein and phenotypes. More importantly, we have confirmed that P7C3, the NAMPT activator, is a novel strategy for reducing SOP bone loss. P7C3 treatment significantly prevents BMSCs senescence by improving mitochondrial function through the NAMPT-OPA1 signaling axis. Taken together, these results reveal that NAMPT is a regulator of BMSCs senescence and osteogenic differentiation. P7C3 is a novel molecule drug to prevent the pathological progression of SOP.
Collapse
Affiliation(s)
- Chao‐wen Bai
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Bo Tian
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Ming‐chao Zhang
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Qin Qin
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Xin Shi
- Institution of NeuroscienceSoochow UniversitySuzhouJiangsuChina
| | - Xi Yang
- Suzhou Medical CollegeSoochow UniversitySuzhouJiangsuChina
| | - Xiang Gao
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Xiao‐zhong Zhou
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Hua‐jian Shan
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Jin‐yu Bai
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|
3
|
Zhang D, Zhu T, Bai J, Chen C, Wen J, Zhou Y, Guan X. Melatonin alleviates senile osteoporosis by regulating autophagy and enhancing fracture healing in aged mice. Bone Joint Res 2025; 14:97-110. [PMID: 39912870 PMCID: PMC11801226 DOI: 10.1302/2046-3758.142.bjr-2024-0112.r2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2025] Open
Abstract
Aims In our previous research, we have found that melatonin (MEL) affects the osteoporotic process. By balancing bone remoulding, autophagy is involved in age-related bone loss. However, as a regulator of autophagy, whether MEL influences senile osteoporosis via regulating autophagy remains unclear. Methods Cellular, radiological, and histopathological evaluations were performed on 36 16-month-old male C57BL6/L mice or aged bone marrow-derived mesenchymal stem cells. A MEL-gelatin methacrylamide system was constructed to aid osteoporotic fracture healing. Results In this study, we found that bone loss, low level of MEL, and decreased autophagy coexisted in aged C57BL6/L mice. A physiological (low, 10 nM but not 100 nM) concentration of MEL restored bone loss, transformed the cytokine framework, and increased the autophagic level in aged mice, whereas inhibition of autophagy unfavourably reduced the positive effects of MEL on bone mass. The autophagy-conducted increased osteogenic lineage commitment and extracellular matrix mineralization, but not matrix synthesis of aged bone marrow-derived mesenchymal stem cells, was responsible for MEL anabolic effects on bone. PIK3C-AKT-MTOR signal was tested to be a main pathway that is involved in MEL-induced autophagy. Conclusion Our data suggest that the application of MEL can restore degenerative osteogenesis of aged bone marrow-derived mesenchymal stem cells, and has the potential to regain bone mass in aged mice through activating autophagy via the PIK3C-AKT-MTOR pathway. MEL therefore may serve as a potential clinical therapy to treat senile osteoporosis.
Collapse
Affiliation(s)
- Denghui Zhang
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Tianer Zhu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Jingyao Bai
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Chunchun Chen
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Junru Wen
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Yi Zhou
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Xiaoxu Guan
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
4
|
Zhang Y, Zhou L, Fu Q, Liu Z. FOXG1 promotes osteogenesis of bone marrow-derived mesenchymal stem cells by activating autophagy through regulating USP14. Commun Biol 2025; 8:59. [PMID: 39814826 PMCID: PMC11735862 DOI: 10.1038/s42003-024-07429-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 12/19/2024] [Indexed: 01/18/2025] Open
Abstract
The osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) is key for bone formation, and its imbalance leads to osteoporosis. Forkhead Box Protein G1 (FOXG1) is associated with osteogenesis, however, the effect of FOXG1 on osteogenesis of BMSCs and ovariectomy (OVX)-induced bone loss is unknown. In our study, FOXG1 expression in BMSCs increases after osteogenic induction. FOXG1 overexpression significantly increases osteoblast marker expression ALP activity, and calcium deposition, while the opposite results are observed in FOXG1 knockdown BMSCs, suggesting that FOXG1 promotes osteogenic differentiation. Additionally, autophagy promotes the differentiation process in BMSCs. We find that FOXG1 induces autophagy, and osteogenic differentiation is blocked via inhibiting FOXG1-caused autophagy, indicating that FOXG1 accelerates osteogenic differentiation via inducing autophagy. Eight-week-old female C57BL/6J mice are used in OVX models, FOXG1 overexpression decreases bone loss by increasing bone formation. Moreover, FOXG1 overexpression suppresses osteoclast differentiation. Mechanically, FOXG1 transcriptionally represses ubiquitin-specific protease14 (USP14) via binding to the USP14 promoter. USP14 overexpression prevents the promoting effect of FOXG1 on osteogenic differentiation in BMSCs. Therefore, our findings suggest that FOXG1 promotes BMSC osteogenic differentiation and inhibits osteoclast differentiation, eventually blocking OVX-induced bone loss, which may provide a promising approach for osteoporosis treatment.
Collapse
Affiliation(s)
- Yiqi Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Long Zhou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qin Fu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ziyun Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
5
|
Behzadi P, Cuevas RA, Crane A, Wendling AA, Chu CC, Moorhead WJ, Wong R, Brown M, Tamakloe J, Suresh S, Salehi P, Jaffe IZ, Kuipers AL, Lukashova L, Verdelis K, St Hilaire C. Rapamycin Reduces Arterial Mineral Density and Promotes Beneficial Vascular Remodeling in a Murine Model of Severe Medial Arterial Calcification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.01.606196. [PMID: 39149364 PMCID: PMC11326142 DOI: 10.1101/2024.08.01.606196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Peripheral artery disease (PAD) is the narrowing of the arteries that carry blood to the lower extremities. PAD has been traditionally associated with atherosclerosis. However, recent studies have found that medial arterial calcification (MAC) is the primary cause of chronic limb ischemia below the knee. MAC involves calcification of the elastic fibers surrounding smooth muscle cells (SMCs) in arteries. Matrix GLA protein (MGP) inhibits vascular calcification by binding circulating calcium and preventing hydroxyapatite crystal deposition, while also modulating osteogenic signaling by blocking BMP-2 activation of RUNX2. Mgp -/- mice develop severe MAC and die around 8 weeks after birth due to aortic rupture or heart failure. We previously discovered a rare genetic disease Arterial Calcification due to Deficiency in CD73 (ACDC) in which patients present with extensive MAC in their lower extremity arteries. Using a patient-specific induced pluripotent stem cell model we found that rapamycin inhibited calcification. Here we investigated whether rapamycin could reduce MAC in vivo using the Mgp -/- murine model. Mgp +/+ and Mgp -/- mice received 5mg/kg rapamycin or vehicle. Calcification content was assessed via microCT, and vascular morphology and extracellular matrix content assessed histologically. Immunostaining and western blot analysis were used to examine SMC phenotype and extracellular matrix content. Rapamycin prolonged Mgp -/- mice lifespan, decreased mineral density in the arteries, maintained SMC contractile phenotype, and improved vessel structure, however, calcification volume was unchanged. Mgp -/- mice with SMC-specific deletion of Raptor or Rictor, did not recapitulate treatment with rapamycin. These findings suggest rapamycin promotes beneficial vascular remodeling in vessels with MAC.
Collapse
Affiliation(s)
- Parya Behzadi
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rolando A Cuevas
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alex Crane
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrew A Wendling
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Claire C Chu
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William J Moorhead
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ryan Wong
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark Brown
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joshua Tamakloe
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Swathi Suresh
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Payam Salehi
- CardioVascular Center, Vascular Surgery, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111-1800, USA
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111-1800, USA
| | - Allison L Kuipers
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lyudmila Lukashova
- Departments of Endodontics and Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Konstantinos Verdelis
- Departments of Endodontics and Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cynthia St Hilaire
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
6
|
Xia TS, Xu SY, Lai LY, Jiang YP, Wang NN, Xin HL. Bitter acids from Humulus lupulus L. alleviate D-galactose induced osteoblastic senescence and bone loss via regulating AKT/mTOR-mediated autophagy. J Food Drug Anal 2024; 32:506-519. [PMID: 39752859 PMCID: PMC11698591 DOI: 10.38212/2224-6614.3508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/13/2024] [Indexed: 01/07/2025] Open
Abstract
Bitter acids (BA) are main component of Humulus lupulus L. (hops). They are known for beer brewing and have various biological and pharmacological properties, especially the bone-protective effect confirmed by our previous in vivo study. Here we aimed to elucidate the anti-senior osteoporosis (SOP) effect of BA on osteoblasts and explore its underlying mechanism. In vitro SOP model was established by D-galactose (D-gal) injured osteoblasts, and the bone formation markers and apoptosis level were measured. mCherry-EGFP-LC3 adenovirus infection and autophagic markers including beclin1 and LC3 proteins were detected to investigate the autophagy level in osteoblasts. To further verify whether BA play the bone-protective role through regulating autophagy, the autophagy inhibitor 3-MA was used, and the cell proliferation, ALP activity, bone mineralization, apoptosis rate and SA-β-gal staining areas were measured. Finally, the protein expressions of AKT/mTOR signaling pathway were detected by Western blotting, and AKT agonist SC79 and mTOR agonist MHY1485 were used to further study the mechanism of BA on AKT/mTOR-mediated autophagy. The results showed that BA stimulated osteoblastic differentiation and inhibited apoptosis proteins Bcl-2/Bax in D-gal-treated osteoblasts. BA also increased the expression of autophagic markers beclin1 and LC3-II/LC3-I in D-gal-treated osteoblasts. mCherry-EGFP-LC3 autophagic double fluorescent adenovirus showed BA promoted the generation of autolysosomes and autophagosomes in D-gal-injured osteoblasts, indicating that BA might prevent osteoblastic bone loss through activating autophagy. Autophagy inhibitor 3-MA was used to further verify whether BA played the bone-protective role via regulating autophagy. The results revealed the promotion effects of BA on proliferation, ALP activity, and mineralized nodule formation in D-gal-injured osteoblasts were eliminated after autophagy blocking with 3-MA, and the inhibitory effects of BA on apoptosis rate and SA-β-gal staining areas were also eliminated. Moreover, BA reduced the phosphorylation levels of AKT, mTOR, p70S6K, and 4EBP in AKT/mTOR pathway, and the promotion of BA on the autophagic markers was blocked after the activation of AKT and mTOR by SC79 and MHY1485. In conclusion, it was the first time to demonstrate that BA improved cell activities and bone formation in aging osteoblasts, and revealed the mechanism of BA against SOP in osteoblasts was activating AKT/mTOR-mediated autophagy.
Collapse
Affiliation(s)
- Tian-Shuang Xia
- School of Pharmacy, Naval Medical University, Shanghai, 200433,
China
| | - Sheng-Yan Xu
- School of Pharmacy, Naval Medical University, Shanghai, 200433,
China
| | - Li-Yong Lai
- School of Pharmacy, Naval Medical University, Shanghai, 200433,
China
| | - Yi-Ping Jiang
- School of Pharmacy, Naval Medical University, Shanghai, 200433,
China
| | - Na-Ni Wang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007,
China
| | - Hai-Liang Xin
- School of Pharmacy, Naval Medical University, Shanghai, 200433,
China
| |
Collapse
|
7
|
Chen R, Yang C, Yang F, Yang A, Xiao H, Peng B, Chen C, Geng B, Xia Y. Targeting the mTOR-Autophagy Axis: Unveiling Therapeutic Potentials in Osteoporosis. Biomolecules 2024; 14:1452. [PMID: 39595628 PMCID: PMC11591800 DOI: 10.3390/biom14111452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Osteoporosis (OP) is a widespread age-related disorder marked by decreased bone density and increased fracture risk, presenting a significant public health challenge. Central to the development and progression of OP is the dysregulation of the mechanistic target of the rapamycin (mTOR)-signaling pathway, which plays a critical role in cellular processes including autophagy, growth, and proliferation. The mTOR-autophagy axis is emerging as a promising therapeutic target due to its regulatory capacity in bone metabolism and homeostasis. This review aims to (1) elucidate the role of mTOR signaling in bone metabolism and its dysregulation in OP, (2) explore the interplay between mTOR and autophagy in the context of bone cell activity, and (3) assess the therapeutic potential of targeting the mTOR pathway with modulators as innovative strategies for OP treatment. By examining the interactions among autophagy, mTOR, and OP, including insights from various types of OP and the impact on different bone cells, this review underscores the complexity of mTOR's role in bone health. Despite advances, significant gaps remain in understanding the detailed mechanisms of mTOR's effects on autophagy and bone cell function, highlighting the need for comprehensive clinical trials to establish the efficacy and safety of mTOR inhibitors in OP management. Future research directions include clarifying mTOR's molecular interactions with bone metabolism and investigating the combined benefits of mTOR modulation with other therapeutic approaches. Addressing these challenges is crucial for developing more effective treatments and improving outcomes for individuals with OP, thereby unveiling the therapeutic potentials of targeting the mTOR-autophagy axis in this prevalent disease.
Collapse
Affiliation(s)
- Rongjin Chen
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
- Department of Orthopedics, Tianshui Hand and Foot Surgery Hospital, Tianshui 741000, China
| | - Chenhui Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
- Department of Orthopedics, Tianshui Hand and Foot Surgery Hospital, Tianshui 741000, China
| | - Fei Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Ao Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Hefang Xiao
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Bo Peng
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Changshun Chen
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Bin Geng
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Yayi Xia
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
8
|
Nie J, Ma S, Wu L, Li Y, Cao J, Li M, Mei P, Cooper PR, Li A, Pei D. SEC31a-ATG9a Interaction Mediates the Recruitment of COPII Vesicles for Autophagosome Formation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405127. [PMID: 39361436 PMCID: PMC11600210 DOI: 10.1002/advs.202405127] [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: 05/11/2024] [Revised: 07/28/2024] [Indexed: 10/05/2024]
Abstract
Autophagy plays an important role in determining stem-cell differentiation. During the osteogenic differentiation of mesenchymal stem cells (MSCs), autophagosome formation is upregulated but the reason is unknown. A long-standing quest in the autophagy field is to find the membrane origin of autophagosomes. In this study, cytoplasmic coat protein complex II (COPII) vesicles, endoplasmic reticulum-derived vesicles responsible for the transport of storage proteins to the Golgi, are demonstrated to be a critical source of osteoblastic autophagosomal membrane. A significant correlation between the number of COPII vesicle and the autophagy level is identified in the rat bone tissues. Disruption of COPII vesicles restrained osteogenesis and decreased the number and size of autophagosomes. SEC31a (an outer coat protein of COPII vesicle) is found to be vital to regulate COPII vesicle-dependent autophagosome formation via interacting with ATG9a of autophagosomal seed vesicles. The interference of Sec31a inhibited autophagosome formation and osteogenesis in vitro and in vivo. These results identified a novel mechanism of autophagosome formation in osteogenic differentiation of stem cells and identified SEC31a as a critical protein that mediates the interplay between COPII and ATG9a vesicles. These findings broaden the understanding of the regulatory mechanism in the osteogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Jiaming Nie
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Shaoyang Ma
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Linyue Wu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Ye Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Jiao Cao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Meng Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Peter Mei
- Department of Oral SciencesFaculty of DentistryUniversity of OtagoDunedin9016New Zealand
| | - Paul R. Cooper
- Department of Oral SciencesFaculty of DentistryUniversity of OtagoDunedin9016New Zealand
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Dandan Pei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| |
Collapse
|
9
|
Zhao Y, Liu J, Zhang Y, Liang M, Li R, Song Y, Wang Y. Mir-381-3p aggravates ovariectomy-induced osteoporosis by inhibiting osteogenic differentiation through targeting KLF5/Wnt/β-catenin signaling pathway. J Orthop Surg Res 2024; 19:480. [PMID: 39152444 PMCID: PMC11330013 DOI: 10.1186/s13018-024-04992-6] [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: 07/01/2024] [Accepted: 08/11/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Increasing evidence shows the pivotal significance of miRNAs in the pathogenesis of osteoporosis. miR-381-3p has been identified as an inhibitor of osteogenesis. This study explored the role and mechanism of miR-381-3p in postmenopausal osteoporosis (PMOP), the most common type of osteoporosis. METHODS Bilateral ovariectomy (OVX) rat model was established and miR-381-3p antagomir was administrated through the tail vein in vivo. The pathological changes in rats were assessed through the evaluation of serum bone turnover markers (BALP, PINP, and CTX-1), hematoxylin and eosin (H&E) staining, as well as the expression of osteoblast differentiation biomarkers. Moreover, isolated bone marrow mesenchymal stem cells from OVX-induced rats (OVX-BMMSCs) were utilized to explore the impact of miR-381-3p on osteoblast differentiation. In addition, the target gene and downstream pathway of miR-381-3p were further investigated both in vivo and in vitro. RESULTS miR-381-3p expression was elevated, whereas KLF5 was suppressed in OVX rats. miR-381-3p antagomir decreased serum levels of bone turnover markers, improved trabecular separation, promoted osteoblast differentiation biomarker expression in OVX rats. ALP activity and mineralization were suppressed, and levels of osteoblast differentiation biomarkers were impeded after miR-381-3p overexpression during osteoblast differentiation of OVX-BMMSCs. While contrasting results were found after inhibition of miR-381-3p. miR-381-3p targets KLF5, negatively affecting its expression as well as its downstream Wnt/β-catenin pathway, both in vivo and in vitro. Silencing of KLF5 restored Wnt/β-catenin activation induced by miR-381-3p antagomir. CONCLUSION miR-381-3p aggravates PMOP by inhibiting osteogenic differentiation through targeting KLF5/Wnt/β-catenin pathway. miR-381-3p appears to be a promising candidate for therapeutic intervention in PMOP.
Collapse
Affiliation(s)
- Yingwei Zhao
- Department of Orthopedic surgery, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China
- Department of Orthopedic surgery, The Fifth Hospital of Harbin, Harbin, Heilongjiang, 150036, China
| | - Jingsong Liu
- Department of Orthopedic surgery, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China
| | - Yubo Zhang
- Department of Orthopedic surgery, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China
| | - Min Liang
- Department of Orthopedic surgery, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China
| | - Rui Li
- Department of Orthopedic surgery, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China
- Spinal Surgery Department, BinZhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Yindong Song
- Department of Orthopedic surgery, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China
- The LiWan Central Hospital of Guang Zhou, Guangzhou, Guangdong, 510000, China
| | - Yansong Wang
- Department of Orthopedic surgery, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China.
| |
Collapse
|
10
|
Rong Y, Liang X, Jiang K, Jia H, Li H, Lu B, Li G. Global Trends in Research of Programmed Cell Death in Osteoporosis: A Bibliometric and Visualized Analysis (2000-2023). Orthop Surg 2024; 16:1783-1800. [PMID: 38923347 PMCID: PMC11293941 DOI: 10.1111/os.14133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Osteoporosis (OP) is a systemic metabolic bone disease that is characterized by decreased bone mineral density and microstructural damage to bone tissue. Recent studies have demonstrated significant advances in the research of programmed cell death (PCD) in OP. However, there is no bibliometric analysis in this research field. This study searched the Web of Science Core Collection (WoSCC) database for literature related to OP and PCD from 2000 to 2023. This study used VOSviewers 1.6.20, the "bibliometrix" R package, and CiteSpace (6.2.R3) for bibliometric and visualization analysis. A total of 2905 articles from 80 countries were included, with China and the United States leading the way. The number of publications related to PCD in OP is increasing year by year. The main research institutions are Shanghai Jiao Tong University, Chinese Medical University, Southern Medical University, Zhejiang University, and Soochow University. Bone is the most popular journal in the field of PCD in OP, and the Journal of Bone and Mineral Research is the most co-cited journal. These publications come from 14,801 authors, with Liu Zong-Ping, Yang Lei, Manolagas Stavros C, Zhang Wei, and Zhao Hong-Yan having published the most papers. Ronald S. Weinstein was co-cited most often. Oxidative stress and autophagy are the current research hot spots for PCD in OP. This bibliometric study provides the first comprehensive summary of trends and developments in PCD research in OP. This information identifies the most recent research frontiers and hot directions, which will provide a definitive reference for scholars studying PCD in OP.
Collapse
Affiliation(s)
- Yi‐fa Rong
- The First College of Clinical MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Xue‐Zhen Liang
- The First College of Clinical MedicineShandong University of Traditional Chinese MedicineJinanChina
- Orthopaedic MicrosurgeryAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| | - Kai Jiang
- The First College of Clinical MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Hai‐Feng Jia
- The First College of Clinical MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Han‐Zheng Li
- The First College of Clinical MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Bo‐Wen Lu
- The First College of Clinical MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Gang Li
- Orthopaedic MicrosurgeryAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
11
|
He Y, Liu Y, Li R, Xiang A, Chen X, Yu Q, Su P. The role of autophagy/lipophagy in the response of osteoblastic cells to hyperlipidemia (Review). Exp Ther Med 2024; 28:328. [PMID: 38979020 PMCID: PMC11229398 DOI: 10.3892/etm.2024.12617] [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: 01/12/2024] [Accepted: 04/10/2024] [Indexed: 07/10/2024] Open
Abstract
There has been interest in the connection between cardiovascular diseases and osteoporosis, both of which share hyperlipidemia as a common pathological basis. Osteoporosis is a progressive metabolic bone disease characterized by reduced bone mass, deteriorated bone microstructure, increased bone fragility and heightened risk of bone fractures. Dysfunction of osteoblastic cells, vital for bone formation, is induced by excessive internalization of lipids under hyperlipidemic conditions, forming the crux of hyperlipidemia-associated osteoporosis. Autophagy, a process fundamental to cell self-regulation, serves a critical role in osteoblastic cell function and bone formation. When activated by lipids, lipophagy inhibits osteoblastic cell differentiation in response to elevated lipid concentrations, resulting in reduced bone mass and osteoporosis. However, an in-depth understanding of the precise roles and mechanisms of lipophagy in the regulation of osteoblastic cell function is required. Study of the molecular mechanisms governing osteoblastic cell response to excessive lipids can result in a clearer understanding of osteoporosis; therefore, potential strategies for preventing hyperlipidemia-induced osteoporosis can be developed. The present review discusses recent progress in elucidating the molecular mechanisms of lipophagy in the regulation of osteoblastic cell function, offering insights into hyperlipidemia-induced osteoporosis.
Collapse
Affiliation(s)
- Yizhang He
- Shaanxi Provincial Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Yantong Liu
- School of Basic Medical Sciences, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Ran Li
- Shaanxi Provincial Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Aoqi Xiang
- Shaanxi Provincial Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Xiaochang Chen
- Shaanxi Provincial Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Qi Yu
- Shaanxi Provincial Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Peihong Su
- Shaanxi Provincial Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| |
Collapse
|
12
|
Pan L, Liu J, Liu C, Guo L, Yang S. Intermittent pulses of methylprednisolone with low-dose prednisone attenuate lupus symptoms in B6.MRL-Fas lpr/J mice with fewer glucocorticoid side effects. Biomed Pharmacother 2024; 177:117138. [PMID: 39018878 DOI: 10.1016/j.biopha.2024.117138] [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: 05/10/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024] Open
Abstract
Glucocorticoids (GCs) are potent anti-inflammatory and immunosuppressant medications and remain the cornerstone of systemic lupus erythematosus (SLE) therapy. However, ongoing exposure to GCs has the potential to elicit multiple adverse effects. Considering the irreplaceability of GCs in SLE therapy, it is important to explore the optimal regimen of GCs. Here, we compared the long-term efficacy and safety of pulsed and oral GC therapy in a lupus-prone mouse model. Mice were grouped using a randomized block design. We monitored survival rates, proteinuria, serum autoantibodies, and complement 3 (C3) levels up to 28 weeks of age, and assessed renal damage, bone quality, lipid deposition in the liver and marrow, glucose metabolic parameters, and levels of hormones of the hypothalamic-pituitary-adrenal (HPA) axis. Finally, we explored the mechanisms underlying the superior efficacy of the pulse regimen over oral prednisone regimen. We found that both GC regimens alleviated the poor survival rate, proteinuria, and glomerulonephritis, while also reducing serum autoantibodies and increasing the level of C3. The pulsed GC regimen showed less resistance to insulin, less suppression of the HPA axis, less bone loss, and less bone marrow fat deposition than the oral GC regimen. Additionally, GC-induced leucine zipper (GILZ) was significantly overexpressed in the GC pulse group. These results suggest that the GC pulse regimen ameliorated symptoms in lupus-prone mice, with fewer side effects, which may be related to GILZ overexpression. Our findings offer a potentially promising GC treatment option for SLE.
Collapse
Affiliation(s)
- Lu Pan
- Department of Pediatric Rheumatology, Immunology & Allergy, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; The Child Health Clinical Research Center of Jilin Province, China
| | - Jinxiang Liu
- Department of Pediatric Rheumatology, Immunology & Allergy, Children's Medical Center, The First Hospital of Jilin University, Changchun, China
| | - Congcong Liu
- Department of Pediatric Rheumatology, Immunology & Allergy, Children's Medical Center, The First Hospital of Jilin University, Changchun, China
| | - Lishuang Guo
- Department of Pediatric Rheumatology, Immunology & Allergy, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; The Child Health Clinical Research Center of Jilin Province, China
| | - Sirui Yang
- Department of Pediatric Rheumatology, Immunology & Allergy, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; The Child Health Clinical Research Center of Jilin Province, China.
| |
Collapse
|
13
|
Krakowian D, Lesiak M, Auguściak-Duma A, Witecka J, Kusz D, Sieroń AL, Gawron K. Analysis of the TID-I and TID-L Splice Variants' Expression Profile under In Vitro Differentiation of Human Mesenchymal Bone Marrow Cells into Osteoblasts. Cells 2024; 13:1021. [PMID: 38920651 PMCID: PMC11201664 DOI: 10.3390/cells13121021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/27/2024] Open
Abstract
Bone formation is a complex process regulated by a variety of pathways that are not yet fully understood. One of the proteins involved in multiple osteogenic pathways is TID (DNAJA3). The aim of this work was to study the association of TID with osteogenesis. Therefore, the expression profiles of the TID splice variants (TID-L, TID-I) and their protein products were analyzed during the proliferation and differentiation of bone marrow mesenchymal stromal cells (B-MSCs) into osteoblasts. As the reference, the hFOB1.19 cell line was used. The phenotype of B-MSCs was confirmed by the presence of CD73, CD90, and CD105 surface antigens on ~97% of cells. The osteoblast phenotype was confirmed by increased alkaline phosphatase activity, calcium deposition, and expression of ALPL and SPP1. The effect of silencing the TID gene on the expression of ALPL and SPP1 was also investigated. The TID proteins and the expression of TID splice variants were detected. After differentiation, the expression of TID-L and TID-I increased 5-fold and 3.7-fold, respectively, while their silencing resulted in increased expression of SPP1. Three days after transfection, the expression of SPP1 increased 7.6-fold and 5.6-fold in B-MSCs and differentiating cells, respectively. Our preliminary study demonstrated that the expression of TID-L and TID-I changes under differentiation of B-MSCs into osteoblasts and may influence the expression of SPP1. However, for better understanding the functional association of these results with the relevant osteogenic pathways, further studies are needed.
Collapse
Affiliation(s)
- Daniel Krakowian
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
- Toxicology Research Group, Łukasiewicz Research Network—Institute of Industrial Organic Chemistry Branch Pszczyna, 43-200 Pszczyna, Poland
| | - Marta Lesiak
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| | - Aleksandra Auguściak-Duma
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| | - Joanna Witecka
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
- Department of Parasitology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland
| | - Damian Kusz
- Department of Orthopaedics and Traumatology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| | - Aleksander L. Sieroń
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| | - Katarzyna Gawron
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| |
Collapse
|
14
|
Zhou H, Lin X, Feng S, Zhu S, Zhou H, Chen H, Youwu H, Wang Z, Wang R, Shao X, Wang J. Metformin mitigates adipogenesis of fibro-adipogenic progenitors after rotator cuff tears via activating mTOR/ULK1-mediated autophagy. Am J Physiol Cell Physiol 2024; 326:C1590-C1603. [PMID: 38586878 PMCID: PMC11371331 DOI: 10.1152/ajpcell.00034.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/09/2024]
Abstract
Muscular fatty infiltration is a common issue after rotator cuff tears (RCTs), which impair shoulder function. Females suffer a higher prevalence and a more severe degree of muscular fatty infiltration after RCT when compared with males, with the underlying mechanisms remaining unclear. Fibro-adipogenic progenitors (FAPs) are the primary source of muscular fatty infiltration following RCT. Our findings disclose that gender-specific disparities in muscular fatty infiltration are linked to mTOR/ULK1-mediated autophagy of FAPs. Decreased autophagic activity contributes to adipogenic differentiation in female FAPs after RCT. Furthermore, metformin could enhance mTOR/ULK1-mediated autophagic processes of FAPs, thereby alleviating fatty infiltration and improving shoulder functionality after RCT. Together, our study reveals that gender differences in muscular fatty infiltration arise from distinct autophagic activities. Metformin could be a promising noninvasive intervention to ameliorate muscular fatty infiltration of RCT.NEW & NOTEWORTHY The current study demonstrated that gender-specific disparities in muscular fatty infiltration are attributed to mTOR/ULK1-mediated autophagy of FAPs. Decreased autophagic activity contributes to adipogenic differentiation in female FAPs after RCT. Moreover, metformin could enhance mTOR/ULK1-mediated autophagic processes of FAPs, thereby alleviating fatty infiltration and improving shoulder functionality after RCT. Therefore, metformin could be a promising noninvasive intervention to ameliorate muscular fatty infiltration of RCT.
Collapse
Affiliation(s)
- Hao Zhou
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xingzuan Lin
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Shujing Feng
- Department of Sports Medicine, School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Siyuan Zhu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Han Zhou
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Huifang Chen
- Department of Sports Medicine, School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China
| | - He Youwu
- Department of Hand Plastic Surgery, The First People's Hospital of Linping District, Hangzhou, People's Republic of China
| | - Zekai Wang
- Department of Life Science, University of Toronto, Toronto, Ontario, Canada
| | - Ru Wang
- Department of Sports Medicine, School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Xiexiang Shao
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jianhua Wang
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
15
|
Xiao HX, Yu L, Xia Y, Chen K, Li WM, Ge GR, Zhang W, Zhang Q, Zhang HT, Geng DC. Sinomenine increases osteogenesis in mice with ovariectomy-induced bone loss by modulating autophagy. World J Stem Cells 2024; 16:486-498. [PMID: 38817333 PMCID: PMC11135257 DOI: 10.4252/wjsc.v16.i5.486] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/01/2024] [Accepted: 04/07/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND A decreased autophagic capacity of bone marrow mesenchymal stromal cells (BMSCs) has been suggested to be an important cause of decreased osteogenic differentiation. A pharmacological increase in autophagy of BMSCs is a potential therapeutic option to increase osteoblast viability and ameliorate osteoporosis. AIM To explore the effects of sinomenine (SIN) on the osteogenic differentiation of BMSCs and the underlying mechanisms. METHODS For in vitro experiments, BMSCs were extracted from sham-treated mice and ovariectomized mice, and the levels of autophagy markers and osteogenic differentiation were examined after treatment with the appropriate concentrations of SIN and the autophagy inhibitor 3-methyladenine. In vivo, the therapeutic effect of SIN was verified by establishing an ovariectomy-induced mouse model and by morphological and histological assays of the mouse femur. RESULTS SIN reduced the levels of AKT and mammalian target of the rapamycin (mTOR) phosphorylation in the phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR signaling pathway, inhibited mTOR activity, and increased autophagy ability of BMSCs, thereby promoting the osteogenic differentiation of BMSCs and effectively alleviating bone loss in ovariectomized mice in vivo. CONCLUSION The Chinese medicine SIN has potential for the treatment of various types of osteoporosis, bone homeostasis disorders, and autophagy-related diseases.
Collapse
Affiliation(s)
- Hai-Xiang Xiao
- Department of Orthopedics, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Centre of Soochow University, Suzhou 215006, Jiangsu Province, China
- Department of Orthopedics, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang 214500, Jiangsu Province, China
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Lei Yu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yu Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Kai Chen
- Department of Orthopedics, Hai'an People's Hospital, Hai'an 226600, Jiangsu Province, China
| | - Wen-Ming Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Gao-Ran Ge
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Wei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Qing Zhang
- Department of Orthopedics, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an Second People's Hospital, Xuzhou 223002, Jiangsu Province, China
| | - Hong-Tao Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - De-Chun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China.
| |
Collapse
|
16
|
Wei Y, Zheng Z, Zhang Y, Sun J, Xu S, Di X, Ding X, Ding G. Regulation of mesenchymal stem cell differentiation by autophagy. Open Med (Wars) 2024; 19:20240968. [PMID: 38799254 PMCID: PMC11117459 DOI: 10.1515/med-2024-0968] [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: 11/22/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/29/2024] Open
Abstract
Autophagy, a process that isolates intracellular components and fuses them with lysosomes for degradation, plays an important cytoprotective role by eliminating harmful intracellular substances and maintaining cellular homeostasis. Mesenchymal stem cells (MSCs) are multipotent progenitor cells with the capacity for self-renewal that can give rise to a subset of tissues and therefore have potential in regenerative medicine. However, a variety of variables influence the biological activity of MSCs following their proliferation and transplantation in vitro. The regulation of autophagy in MSCs represents a possible mechanism that influences MSC differentiation properties under the right microenvironment, affecting their regenerative and therapeutic potential. However, a deeper understanding of exactly how autophagy is mobilized to function as well as clarifying the mechanisms by which autophagy promotes MSCs differentiation is still needed. Here, we review the current literature on the complex link between MSCs differentiation and autophagy induced by various extracellular or intracellular stimuli and the molecular targets that influence MSCs lineage determination, which may highlight the potential regulation of autophagy on MSCs' therapeutic capacity, and provide a broader perspective on the clinical application of MSCs in the treatment of a wide range of diseases.
Collapse
Affiliation(s)
- Yanan Wei
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Zejun Zheng
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Ying Zhang
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Jinmeng Sun
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Shuangshuang Xu
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Xinsheng Di
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Xiaoling Ding
- Clinical Competency Training Center, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Gang Ding
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, Shandong, China
| |
Collapse
|
17
|
Yang J, Pei Q, Wu X, Dai X, Li X, Pan J, Wang B. Stress reduction through cortical bone thickening improves bone mechanical behavior in adult female Beclin-1 +/- mice. Front Bioeng Biotechnol 2024; 12:1357686. [PMID: 38600946 PMCID: PMC11004267 DOI: 10.3389/fbioe.2024.1357686] [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: 12/18/2023] [Accepted: 03/13/2024] [Indexed: 04/12/2024] Open
Abstract
Fragility fractures, which are more prevalent in women, may be significantly influenced by autophagy due to altered bone turnover. As an essential mediator of autophagy, Beclin-1 modulates bone homeostasis by regulating osteoclast and chondrocyte differentiation, however, the alteration in the local bone mechanical environment in female Beclin-1+/- mice remains unclear. In this study, our aim is to investigate the biomechanical behavior of femurs from seven-month-old female wild-type (WT) and Beclin-1+/- mice under peak physiological load, using finite element analysis on micro-CT images. Micro-CT imaging analyses revealed femoral cortical thickening in Beclin-1+/- female mice compared to WT. Three-point bending test demonstrated a 63.94% increase in whole-bone strength and a 61.18% increase in stiffness for female Beclin-1+/- murine femurs, indicating improved biomechanical integrity. After conducting finite element analysis, Beclin-1+/- mice exhibited a 26.99% reduction in von Mises stress and a 31.62% reduction in maximum principal strain in the femoral midshaft, as well as a 36.64% decrease of von Mises stress in the distal femurs, compared to WT mice. Subsequently, the strength-safety factor was determined using an empirical formula, revealing that Beclin-1+/- mice exhibited significantly higher minimum safety factors in both the midshaft and distal regions compared to WT mice. In summary, considering the increased response of bone adaptation to mechanical loading in female Beclin-1+/- mice, our findings indicate that increasing cortical bone thickness significantly improves bone biomechanical behavior by effectively reducing stress and strain within the femoral shaft.
Collapse
Affiliation(s)
- Jiaojiao Yang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Institute of Life Sciences, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Qilin Pei
- Institute of Life Sciences, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Xingfan Wu
- Institute of Life Sciences, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Xin Dai
- Institute of Life Sciences, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Xi Li
- Institute of Life Sciences, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Jun Pan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Bin Wang
- Institute of Life Sciences, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
18
|
Du X, Zang C, Wang Q. Cyclin A1 (CCNA1) inhibits osteoporosis by suppressing transforming growth factor-beta (TGF-beta) pathway in osteoblasts. BMC Musculoskelet Disord 2024; 25:206. [PMID: 38454404 PMCID: PMC10919014 DOI: 10.1186/s12891-024-07303-6] [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: 07/19/2023] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Osteoporosis is a genetic disease caused by the imbalance between osteoblast-led bone formation and osteoclast-induced bone resorption. However, further gene-related pathogenesis remains to be elucidated. METHODS The aberrant expressed genes in osteoporosis was identified by analyzing the microarray profile GSE100609. Serum samples of patients with osteoporosis and normal group were collected, and the mRNA expression of candidate genes was detected by quantitative real-time polymerase chain reaction (qRT-PCR). The mouse cranial osteoblast MC3T3-E1 cells were treated with dexamethasone (DEX) to mimic osteoporosis in vitro. Alizarin Red staining and alkaline phosphatase (ALP) staining methods were combined to measure matrix mineralization deposition of MC3T3-E1 cells. Meanwhile, the expression of osteogenesis related genes including alkaline phosphatase (ALP), osteocalcin (OCN), osteopontin (OPN), Osterix, and bone morphogenetic protein 2 (BMP2) were evaluated by qRT-PCR and western blotting methods. Then the effects of candidate genes on regulating impede bone loss caused by ovariectomy (OVX) in mice were studied. RESULTS Cyclin A1 (CCNA1) was found to be significantly upregulated in serum of osteoporosis patients and the osteoporosis model cells, which was in line with the bioinformatic analysis. The osteogenic differentiation ability of MC3T3-E1 cells was inhibited by DEX treatment, which was manifested by decreased Alizarin Red staining intensity, ALP staining intensity, and expression levels of ALP, OCN, OPN, Osterix, and BMP2. The effects of CCNA1 inhibition on regulating osteogenesis were opposite to that of DEX. Then, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis demonstrated that genes negatively associated with CCNA1 were enriched in the TGF-beta signaling pathway. Inhibitor of TGF-beta signaling pathway partly reversed osteogenesis induced by suppressed CCNA1. Furthermore, suppressed CCNA1 relieved bone mass of OVX mice in vivo. CONCLUSION Downregulation of CCNA1 could activate TGF-beta signaling pathway and promote bone formation, thus playing a role in treatment of osteoporosis.
Collapse
Affiliation(s)
- Xiao Du
- Department of Orthopedics, Beijing Geriatric Hospital, No.118 Hot Spring Road, Haidian District 100095, Beijing, China
| | - Chuanyi Zang
- Department of Orthopedics, Beijing Geriatric Hospital, No.118 Hot Spring Road, Haidian District 100095, Beijing, China
| | - Qinglei Wang
- Department of Orthopedics, Beijing Geriatric Hospital, No.118 Hot Spring Road, Haidian District 100095, Beijing, China.
| |
Collapse
|
19
|
Cheng W, Fu Y, Lin Z, Huang M, Chen Y, Hu Y, Lin Q, Yu B, Liu G. Lipoteichoic acid restrains macrophage senescence via β-catenin/FOXO1/REDD1 pathway in age-related osteoporosis. Aging Cell 2024; 23:e14072. [PMID: 38126583 PMCID: PMC10928565 DOI: 10.1111/acel.14072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Osteoporosis and its related fractures are common causes of morbidity and mortality in older adults, but its underlying molecular and cellular mechanisms remain largely unknown. In this study, we found that lipoteichoic acid (LTA) treatment could ameliorate age-related bone degeneration and attenuate intramedullary macrophage senescence. FOXO1 signaling, which was downregulated and deactivated in aging macrophages, played a key role in the process. Blocking FOXO1 signaling caused decreased REDD1 expression and increased phosphorylation level of mTOR, a major driver of aging, as well as aggravated bone loss and deteriorated macrophage senescence. Moreover, LTA elevated FOXO1 signaling through β-catenin pathway while β-catenin inhibition significantly suppressed FOXO1 signaling, promoted senescence-related protein expression, and accelerated bone degeneration and macrophage senescence. Our findings indicated that β-catenin/FOXO1/REDD1 signaling plays a physiologically significant role that protecting macrophages from senescence during aging.
Collapse
Affiliation(s)
- Weike Cheng
- Department of OrthopaedicsNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative MedicineNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yong Fu
- Department of OrthopaedicsNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative MedicineNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Zexin Lin
- Department of OrthopaedicsNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative MedicineNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Mouzhang Huang
- Department of OrthopaedicsNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative MedicineNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yingqi Chen
- Department of OrthopaedicsNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative MedicineNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yanjun Hu
- Department of OrthopaedicsNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative MedicineNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Qingrong Lin
- Department of OrthopaedicsNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative MedicineNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Bin Yu
- Department of OrthopaedicsNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative MedicineNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Guanqiao Liu
- Department of OrthopaedicsNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative MedicineNanfang Hospital, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
20
|
Zhu L, Wang J, Wu Z, Chen S, He Y, Jiang Y, Luo G, Wu Z, Li Y, Xie J, Zou S, Zhou C. AFF4 regulates osteogenic potential of human periodontal ligament stem cells via mTOR-ULK1-autophagy axis. Cell Prolif 2024; 57:e13546. [PMID: 37731335 PMCID: PMC10849782 DOI: 10.1111/cpr.13546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/23/2023] [Accepted: 08/30/2023] [Indexed: 09/22/2023] Open
Abstract
Scaffold protein AF4/FMR2 family member 4 (AFF4) has been found to play a role in osteogenic commitment of stem cells. However, function of AFF4 in human periodontal ligament stem cells (hPDLSCs) has not been studied yet. This present study aims to investigate the biological effect of AFF4 on osteogenic differentiation of hPDLSCs and potential mechanistic pathway. First, AFF4 expression profile was evaluated in conditions of periodontitis and osteogenic differentiation of hPDLSCs by immunohistochemical staining, western blot and qRT-PCR. Next, si-RNA mediated knockdown and lentiviral transduction mediated overexpression of AFF4 were adopted to explore impact of AFF4 on osteogenic capacity of hPDLSCs. Then, possible mechanistic pathway was identified. At last, pharmacological agonist of autophagy, rapamycin, was utilized to affirm the role of autophagy in AFF4-regulated osteogenesis of hPDLSCs. First, AFF4 expressions were significantly lower in inflamed periodontal tissues and lipopolysaccharides-treated hPDLSCs than controls, and were up-regulated during osteogenic differentiation of hPDLSCs. Next, osteogenic potential of hPDLSCs was impaired by AFF4 knockdown and potentiated by AFF4 overexpression. Moreover, AFF4 was found to positively regulate autophagic activity in hPDLSCs. At last, rapamycin treatment was shown to be able to partly restore AFF4 knockdown-suppressed osteogenic differentiation. Our study demonstrates that AFF4 regulates osteogenic potential of hPDLSCs via targeting autophagic activity. The involvement of AFF4 in periodontal homeostasis was identified for the first time.
Collapse
Affiliation(s)
- Li Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Jiahe Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Zuping Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouChina
| | - Sirui Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Yuying He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Yukun Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Guowen Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Zhuoxuan Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Yuyu Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Jing Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Shujuan Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
- Department of Orthodontics, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
- Department of Pediatric Dentistry, West China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
21
|
Huang X, He W, Fan S, Li H, Ye G. IGF2BP3-mediated enhanced stability of MYLK represses MSC adipogenesis and alleviates obesity and insulin resistance in HFD mice. Cell Mol Life Sci 2024; 81:17. [PMID: 38196046 PMCID: PMC10776757 DOI: 10.1007/s00018-023-05076-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 01/11/2024]
Abstract
Mesenchymal stem cells (MSCs) hold immense potential as multipotent stem cells and serve as a primary source of adipocytes. The process of MSC adipogenesis plays a crucial role in maintaining systemic metabolic homeostasis and has garnered significant attention in tissue bioengineering. N6-methyladenosine (m6A), the most prevalent RNA modification, is known to regulate cell fate and disease. However, the precise involvement of m6A readers in MSC adipogenesis remains unclear. In this study, we investigated the impact of IGF2BP3, a prominent m6A reader, on MSC adipogenesis. Our findings revealed a decrease in IGF2BP3 expression during the natural adipogenic differentiation of MSCs. Furthermore, IGF2BP3 was found to repress MSC adipogenesis by augmenting the levels of MYLK, a calcium/calmodulin-dependent kinase. Mechanistically, IGF2BP3 interacted with MYLK mRNA in an m6A-dependent manner, extending its half-life and subsequently inhibiting the phosphorylation of the ERK1/2 pathway, thereby impeding the adipogenic differentiation of MSCs. Additionally, we successfully achieved the overexpression of IGF2BP3 through intraperitoneal injection of adeno-associated virus serotype Rec2, which specifically targeted adipose tissue. This intervention resulted in reduced body weight and improved insulin resistance in high-fat diet mice. Overall, our study provides novel insights into the role of IGF2BP3 in MSC adipogenesis, shedding light on adipocyte-related disorders and presenting potential targets for related biomedical applications.
Collapse
Affiliation(s)
- Xiuji Huang
- Department of Respiratory and Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, People's Republic of China
| | - Wuhui He
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People's Republic of China
| | - Shuai Fan
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, People's Republic of China
| | - Hui Li
- Department of Respiratory and Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, People's Republic of China.
| | - Guiwen Ye
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, People's Republic of China.
| |
Collapse
|
22
|
Yang S, Gao J, Chen M, Sun Y, Qiao X, Mao H, Guo L, Yu Y, Yang D. Let-7a promotes periodontal bone regeneration of bone marrow mesenchymal stem cell aggregates via the Fas/FasL-autophagy pathway. J Cell Mol Med 2023; 27:4056-4068. [PMID: 37855249 PMCID: PMC10746947 DOI: 10.1111/jcmm.17988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/29/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023] Open
Abstract
Periodontal bone regeneration using bone marrow mesenchymal stem cell (BMMSC) transplantation is a promising method; however, the method for osteogenic differentiation of BMMSCs needs to be improved. In this research, we sought to identify the roles of let-7a in the osteogenesis of BMMSCs and to provide a potential method for periodontal bone regeneration. Our previous study revealed that Fas/FasL is a target of let-7a. In this study, we demonstrated that let-7a overexpression significantly enhanced BMMSC-CAs osteogenesis both in vitro and in vivo. Mechanistically, upregulation of Fas/FasL using the rfas/rfaslg plasmid obstructed the osteogenesis of BMMSCs by inhibiting autophagy. Furthermore, we confirmed that overexpression of let-7a activated autophagy and alleviated the inhibited osteogenesis by the autophagy inhibitor 3-MA and the rfas/rfaslg plasmid of BMMSCs. In general, our findings showed that let-7a promoted the osteogenesis of BMMSCs through the Fas/FasL-autophagy pathway, suggesting that the application of let-7a in BMMSC-CAs based periodontal bone regeneration could be a promising strategy.
Collapse
Affiliation(s)
- Shiyao Yang
- Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Stomatological Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| | - Jing Gao
- Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Stomatological Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| | - Meng Chen
- Department of Oral and Maxillofacial SurgeryDaping Hospital, Army Medical UniversityChongqingChina
| | - Yuting Sun
- Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Stomatological Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| | - Xin Qiao
- Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
| | - Hongchen Mao
- Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Stomatological Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| | - Li Guo
- Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Stomatological Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| | - Yang Yu
- Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Stomatological Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| | - Deqin Yang
- Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Stomatological Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| |
Collapse
|
23
|
Li W, Zhang Q, Gu R, Zeng L, Liu H. Platelet factor 4 induces bone loss by inhibiting the integrin α5-FAK-ERK pathway. Animal Model Exp Med 2023; 6:573-584. [PMID: 37565509 PMCID: PMC10757219 DOI: 10.1002/ame2.12342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/19/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND The effect of platelet factor 4 (PF4) on bone marrow mesenchymal stem cells (BMMSCs) and osteoporosis is poorly understood. Therefore, this study aimed to evaluate the effects of PF4-triggered bone destruction in mice and determine the underlying mechanism. METHODS First, in vitro cell proliferation and cell cycle of BMMSCs were assessed using a CCK8 assay and flow cytometry, respectively. Osteogenic differentiation was confirmed using staining and quantification of alkaline phosphatase and Alizarin Red S. Next, an osteoporotic mouse model was established by performing bilateral ovariectomy (OVX). Furthermore, the PF4 concentrations were obtained using enzyme-linked immunosorbent assay. The bone microarchitecture of the femur was evaluated using microCT and histological analyses. Finally, the key regulators of osteogenesis and pathways were investigated using quantitative real-time polymerase chain reaction and Western blotting. RESULTS Human PF4 widely and moderately decreased the cell proliferation and osteogenic differentiation ability of BMMSCs. Furthermore, the levels of PF4 in the serum and bone marrow were generally increased, whereas bone microarchitecture deteriorated due to OVX. Moreover, in vivo mouse PF4 supplementation triggered bone deterioration of the femur. In addition, several key regulators of osteogenesis were downregulated, and the integrin α5-focal adhesion kinase-extracellular signal-regulated kinase (ITGA5-FAK-ERK) pathway was inhibited due to PF4 supplementation. CONCLUSIONS PF4 may be attributed to OVX-induced bone loss triggered by the suppression of bone formation in vivo and alleviate BMMSC osteogenic differentiation by inhibiting the ITGA5-FAK-ERK pathway.
Collapse
Affiliation(s)
- Wei Li
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of StomatologyPeking UniversityBeijingChina
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial RegionsChinese Academy of Medical SciencesBeijingChina
| | - Qiwei Zhang
- Department of Orthopedics, Beijing Hospital and National Center of Gerontology and Institute of Geriatrics MedicineChinese Academy of Medical SciencesBeijingChina
- Department of OrthopedicsBeijing Eden HospitalBeijingChina
| | - Ranli Gu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of StomatologyPeking UniversityBeijingChina
| | - Lijun Zeng
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of StomatologyPeking UniversityBeijingChina
| | - Hao Liu
- The Central Laboratory, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, National Health Commission Key Laboratory of Digital Technology of StomatologyPeking UniversityBeijingChina
| |
Collapse
|
24
|
Chen Q, Yang Z, Sun X, Long R, Shen J, Wang Z. Inokosterone activates the BMP2 to promote the osteogenic differentiation of bone marrow mesenchymal stem cells and improve bone loss in ovariectomized rats. Biochem Biophys Res Commun 2023; 682:349-358. [PMID: 37839103 DOI: 10.1016/j.bbrc.2023.10.032] [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: 07/21/2023] [Revised: 09/21/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023]
Abstract
Evidence suggests that enhancing the osteogenic ability of bone marrow-derived mesenchymal stem cells (BMSCs) may be beneficial in the fight against osteoporosis (OP) effects. Inokosterone (IS) is a major active constituent of Achyranthis bidentatae radix (ABR), which stimulates osteogenic differentiation of mouse embryonic osteoblasts. This study aims to investigate effect of IS on OP using osteogenic differentiated BMSCs and ovariectomy (OVX)-induced OP rats. The BMSCs were treated with 50, 100, or 200 mg/L IS and OP rats were given 2 or 4 mg/kg of IS by gavage. Cell viability, the osteogenic differentiation marker protein expression level, and mineralization were observed. This study proved that IS improved cell viability, osteogenic differentiation, and cellular mineralization in BMSCs and raised expression levels of bone morphogenetic protein-2 (BMP2), Smad1, runt-related transcription factor 2 (RUNX2), collagen I, ALP, and OCN. By BMP2 knockdown/overexpression, this study also proved the BMP2 signaling pathway activation is a potential biological mechanism of IS to improve osteogenic differentiation and mineralization in osteogenic differentiated BMSCs. In OVX-induced OP rats, IS was observed to antagonize bone loss, improve osteogenic differentiation marker protein expression levels, and activate BMP-2, smad1, and RUNX2. These findings provide scientific support for further investigation of the biological mechanisms of IS in ameliorating OP.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311200, China
| | - Zhihua Yang
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311200, China
| | - Xiangyi Sun
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311200, China
| | - Ruchao Long
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311200, China
| | - Jianwei Shen
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311200, China
| | - Zhen Wang
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311200, China.
| |
Collapse
|
25
|
Wang R, Wang Y, Niu Y, He D, Jin S, Li Z, Zhu L, Chen L, Wu X, Ding C, Wu T, Shi X, Zhang H, Li C, Wang X, Xie Z, Li W, Liu Y. Deep Learning-Predicted Dihydroartemisinin Rescues Osteoporosis by Maintaining Mesenchymal Stem Cell Stemness through Activating Histone 3 Lys 9 Acetylation. ACS CENTRAL SCIENCE 2023; 9:1927-1943. [PMID: 37901168 PMCID: PMC10604014 DOI: 10.1021/acscentsci.3c00794] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Indexed: 10/31/2023]
Abstract
Maintaining the stemness of bone marrow mesenchymal stem cells (BMMSCs) is crucial for bone homeostasis and regeneration. However, in vitro expansion and bone diseases impair BMMSC stemness, limiting its functionality in bone tissue engineering. Using a deep learning-based efficacy prediction system and bone tissue sequencing, we identify a natural small-molecule compound, dihydroartemisinin (DHA), that maintains BMMSC stemness and enhances bone regeneration. During long-term in vitro expansion, DHA preserves BMMSC stemness characteristics, including its self-renewal ability and unbiased differentiation. In an osteoporosis mouse model, oral administration of DHA restores the femur trabecular structure, bone density, and BMMSC stemness in situ. Mechanistically, DHA maintains BMMSC stemness by promoting histone 3 lysine 9 acetylation via GCN5 activation both in vivo and in vitro. Furthermore, the bone-targeted delivery of DHA by mesoporous silica nanoparticles improves its therapeutic efficacy in osteoporosis. Collectively, DHA could be a promising therapeutic agent for treating osteoporosis by maintaining BMMSC stemness.
Collapse
Affiliation(s)
- Ruoxi Wang
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Yu Wang
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Yuting Niu
- Central
Laboratory, National Center for Stomatology & National Clinical
Research Center for Oral Diseases & National Engineering Laboratory
for Digital and Material Technology of Stomatology & Beijing Key
Laboratory of Digital Stomatology & Research Center of Engineering
and Technology for Computerized Dentistry Ministry of Health &
NMPA Key Laboratory for Dental Materials & Translational Research
Center for Orocraniofacial Stem Cells and Systemic Health, Central
Laboratory, Peking University School and
Hospital for Stomatology, Beijing 100081, China
| | - Danqing He
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Shanshan Jin
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Zixin Li
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Lisha Zhu
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Liyuan Chen
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Xiaolan Wu
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Chengye Ding
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Tianhao Wu
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Xinmeng Shi
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - He Zhang
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Chang Li
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Xin Wang
- Peking
University International Cancer Institute, Health Science Center, Peking University, Beijing 100083, China
| | - Zhengwei Xie
- Peking
University International Cancer Institute, Health Science Center, Peking University, Beijing 100083, China
| | - Weiran Li
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| | - Yan Liu
- Laboratory
of Biomimetic Nanomaterials, Department of Orthodontics & National
Center for Stomatology & National Clinical Research Center for
Oral Diseases & National Engineering Laboratory for Digital and
Material Technology of Stomatology & Beijing Key Laboratory of
Digital Stomatology & Research Center of Engineering and Technology
for Computerized Dentistry Ministry of Health & NMPA Key Laboratory
for Dental Materials & Translational Research Center for Orocraniofacial
Stem Cells and Systemic Health, Peking University
School and Hospital for Stomatology, Beijing 100081, China
| |
Collapse
|
26
|
Zhang Q, Lei X, Wang F, He X, Liu L, Hou Y, Liu Y, Jin F, Chen C, Li B. ERK1-mediated immunomodulation of mesenchymal stem cells ameliorates inflammatory disorders. iScience 2023; 26:107868. [PMID: 37790278 PMCID: PMC10543658 DOI: 10.1016/j.isci.2023.107868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/03/2023] [Accepted: 09/06/2023] [Indexed: 10/05/2023] Open
Abstract
Immune system disorders, especially T cell disorders, are important therapeutic targets of mesenchymal stem cells (MSCs) in many autoimmune diseases (ADs). Although extracellular regulated protein kinases (ERKs) play a role in MSC therapy by promoting T cell apoptosis, the mechanism remains unclear. Our findings indicate that ERK1-/- bone marrow MSCs (BMMSCs), but not ERK2-/- BMMSCs, failed to promote T cell apoptosis due to incapacity to activate the ETS2/AURKA/NF-κB/Fas/MCP-1 cascade. Moreover, ERK1-/- BMMSCs were unable to upregulate regulatory T cells and suppress T helper 17 cells. Licochalcone A (LA), which promotes ERK pathway activation, enhanced the therapeutic efficacy of MSC therapy in ulcerative colitis and collagen-induced arthritis mice. Our findings suggest that ERK1, but not ERK2, plays a crucial role in regulating T cells in MSCs. LA-treated MSCs provide a strategy to improve the efficacy of MSC-based treatments for ADs.
Collapse
Affiliation(s)
- Qing Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
- Shannxi Clinical Research Center for Oral Diseases & Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Xiao Lei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Shannxi Clinical Research Center for Oral Diseases & Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Fang Wang
- Department of Blood Purification, General Hospital of Central Theater Command of PLA, 68 Huangpu Road, Wuhan, Hubei 430010, China
| | - Xiaoning He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Lu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Shannxi Clinical Research Center for Oral Diseases & Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Yuxia Hou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| | - Yuan Liu
- The Affiliated Northwest Women’s and Children’s Hospital of Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi 710061, China
| | - Fang Jin
- Shannxi Clinical Research Center for Oral Diseases & Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bei Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| |
Collapse
|
27
|
Zhang H, Yang G, Li J, Xiao L, Guo C, Wang Y. Impaired autophagy activity-induced abnormal differentiation of bone marrow stem cells is related to adolescent idiopathic scoliosis osteopenia. Chin Med J (Engl) 2023; 136:2077-2085. [PMID: 36728938 PMCID: PMC10476821 DOI: 10.1097/cm9.0000000000002165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Osteopenia has been well documented in adolescent idiopathic scoliosis (AIS). Bone marrow stem cells (BMSCs) are a crucial regulator of bone homeostasis. Our previous study revealed a decreased osteogenic ability of BMSCs in AIS-related osteopenia, but the underlying mechanism of this phenomenon remains unclear. METHODS A total of 22 AIS patients and 18 age-matched controls were recruited for this study. Anthropometry and bone mass were measured in all participants. Bone marrow blood was collected for BMSC isolation and culture. Osteogenic and adipogenic induction were performed to observe the differences in the differentiation of BMSCs between the AIS-related osteopenia group and the control group. Furthermore, a total RNA was extracted from isolated BMSCs to perform RNA sequencing and subsequent analysis. RESULTS A lower osteogenic capacity and increased adipogenic capacity of BMSCs in AIS-related osteopenia were revealed. Differences in mRNA expression levels between the AIS-related osteopenia group and the control group were identified, including differences in the expression of LRRC17 , DCLK1 , PCDH7 , TSPAN5 , NHSL2 , and CPT1B . Kyoto Encyclopedia of Genes and Genomes enrichment analyses revealed several biological processes involved in the regulation of autophagy and mitophagy. The Western blotting results of autophagy markers in BMSCs suggested impaired autophagic activity in BMSCs in the AIS-related osteopenia group. CONCLUSION Our study revealed that BMSCs from AIS-related osteopenia patients have lower autophagic activity, which may be related to the lower osteogenic capacity and higher adipogenic capacity of BMSCs and consequently lead to the lower bone mass in AIS patients.
Collapse
Affiliation(s)
- Hongqi Zhang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central-South University, Changsha, Hunan 410008, China
| | - Guanteng Yang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central-South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorder, Xiangya Hospital, Central-South University, Changsha, Hunan 410008, China
| | - Jiong Li
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central-South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorder, Xiangya Hospital, Central-South University, Changsha, Hunan 410008, China
| | - Lige Xiao
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central-South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorder, Xiangya Hospital, Central-South University, Changsha, Hunan 410008, China
| | - Chaofeng Guo
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central-South University, Changsha, Hunan 410008, China
| | - Yuxiang Wang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central-South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorder, Xiangya Hospital, Central-South University, Changsha, Hunan 410008, China
| |
Collapse
|
28
|
Ge J, Yu YJ, Li JY, Li MY, Xia SM, Xue K, Wang SY, Yang C. Activating Wnt/β-catenin signaling by autophagic degradation of APC contributes to the osteoblast differentiation effect of soy isoflavone on osteoporotic mesenchymal stem cells. Acta Pharmacol Sin 2023; 44:1841-1855. [PMID: 36973541 PMCID: PMC10462682 DOI: 10.1038/s41401-023-01066-x] [Citation(s) in RCA: 3] [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: 10/29/2022] [Accepted: 02/17/2023] [Indexed: 03/29/2023]
Abstract
The functional role of autophagy in regulating differentiation of bone marrow mesenchymal stem cells (MSCs) has been studied extensively, but the underlying mechanism remains largely unknown. The Wnt/β-catenin signaling pathway plays a pivotal role in the initiation of osteoblast differentiation of mesenchymal progenitor cells, and the stability of core protein β-catenin is tightly controlled by the APC/Axin/GSK-3β/Ck1α complex. Here we showed that genistein, a predominant soy isoflavone, stimulated osteoblast differentiation of MSCs in vivo and in vitro. Female rats were subjected to bilateral ovariectomy (OVX); four weeks after surgery the rats were orally administered genistein (50 mg·kg-1·d-1) for 8 weeks. The results showed that genistein administration significantly suppressed the bone loss and bone-fat imbalance, and stimulated bone formation in OVX rats. In vitro, genistein (10 nM) markedly activated autophagy and Wnt/β-catenin signaling pathway, and stimulated osteoblast differentiation in OVX-MSCs. Furthermore, we found that genistein promoted autophagic degradation of adenomatous polyposis coli (APC), thus initiated β-catenin-driven osteoblast differentiation. Notably, genistein activated autophagy through transcription factor EB (TFEB) rather than mammalian target of rapamycin (mTOR). These findings unveil the mechanism of how autophagy regulates osteogenesis in OVX-MSCs, which expands our understanding that such interplay could be employed as a useful therapeutic strategy for treating postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Jing Ge
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China
| | - Ye-Jia Yu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China
| | - Jia-Yi Li
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China
| | - Meng-Yu Li
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China
| | - Si-Mo Xia
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China
| | - Ke Xue
- Department of Pastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Shao-Yi Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China.
| | - Chi Yang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China.
| |
Collapse
|
29
|
Ma Z, Chen L, Wang Y, Zhang S, Zheng J, Luo Y, Wang C, Zeng H, Xue L, Tan Z, Wang D. Novel insights of EZH2-mediated epigenetic modifications in degenerative musculoskeletal diseases. Ageing Res Rev 2023; 90:102034. [PMID: 37597667 DOI: 10.1016/j.arr.2023.102034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 07/06/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
Degenerative musculoskeletal diseases (Osteoporosis, Osteoarthritis, Degenerative Spinal Disease and Sarcopenia) are pathological conditions that affect the function and pain of tissues such as bone, cartilage, and muscles, and are closely associated with ageing and long-term degeneration. Enhancer of zeste homolog 2 (EZH2), an important epigenetic regulator, regulates gene expression mainly through the PRC2-dependent trimethylation of histone H3 at lysine 27 (H3K27me3). Increasing evidence suggests that EZH2 is involved in several biological processes closely related to degenerative musculoskeletal diseases, such as osteogenic-adipogenic differentiation of bone marrow mesenchymal stem cells, osteoclast activation, chondrocyte functional status, and satellite cell proliferation and differentiation, mainly through epigenetic regulation (H3K27me3). Therefore, the synthesis and elucidation of the role of EZH2 in degenerative musculoskeletal diseases have attracted increasing attention. In addition, although EZH2 inhibitors have been approved for clinical use, whether they can be repurposed for the treatment of degenerative musculoskeletal diseases needs to be considered. Here, we reviewed the role of EZH2 in the development of degenerative musculoskeletal diseases and brought forward prospects of its pharmacological inhibitors in the improvement of the treatment of the diseases.
Collapse
Affiliation(s)
- Zetao Ma
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, People's Republic of China
| | - Lei Chen
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, People's Republic of China; Shantou University Medical College, Shantou 515031, People's Republic of China
| | - Yushun Wang
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, People's Republic of China
| | - Sheng Zhang
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, People's Republic of China
| | - Jianrui Zheng
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, People's Republic of China
| | - Yuhong Luo
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, People's Republic of China
| | - Chao Wang
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, People's Republic of China
| | - Hui Zeng
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, People's Republic of China
| | - Lixiang Xue
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, People's Republic of China.
| | - Zhen Tan
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, People's Republic of China.
| | - Deli Wang
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, People's Republic of China.
| |
Collapse
|
30
|
Ma C, Yu R, Li J, Chao J, Liu P. Targeting proteostasis network in osteoporosis: Pathological mechanisms and therapeutic implications. Ageing Res Rev 2023; 90:102024. [PMID: 37532006 DOI: 10.1016/j.arr.2023.102024] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/11/2023] [Accepted: 07/28/2023] [Indexed: 08/04/2023]
Abstract
As the most common bone disease, osteoporosis (OP) increases bone fragility and makes patients more vulnerable to the threat of osteoporotic fractures. With the ageing population in today's society, OP has become a huge and growing public health problem. Unfortunately, the clear pathogenesis of OP is still under exploration, and effective interventions are still scarce. Therefore, exploring new targets for pharmacological interventions to develop promising therapeutic drugs for OP is of great clinical value. Previous studies have shown that normal bone remodeling depends on proteostasis, whereas loss of proteostasis during ageing leads to the dysfunctional proteostasis network (PN) that fails to maintain bone homeostasis. Nevertheless, only a few studies have revealed the pathophysiological relationship between bone metabolism and a single component of PN, yet the role of PN as a whole in the pathogenesis of OP is still under investigation. This review comprehensively summarized the role of PN in the pathogenesis of OP and further discussed the potential of PN as innovative drug targets for the therapy of OP.
Collapse
Affiliation(s)
- Cong Ma
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China; Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ronghui Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Junhong Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiashuo Chao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Ping Liu
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China.
| |
Collapse
|
31
|
An F, Wang X, Wang C, Liu Y, Sun B, Zhang J, Gao P, Yan C. Research progress on the role of lncRNA-miRNA networks in regulating adipogenic and osteogenic differentiation of bone marrow mesenchymal stem cells in osteoporosis. Front Endocrinol (Lausanne) 2023; 14:1210627. [PMID: 37645421 PMCID: PMC10461560 DOI: 10.3389/fendo.2023.1210627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/26/2023] [Indexed: 08/31/2023] Open
Abstract
Osteoporosis (OP) is characterized by a decrease in osteoblasts and an increase in adipocytes in the bone marrow compartment, alongside abnormal bone/fat differentiation, which ultimately results in imbalanced bone homeostasis. Bone marrow mesenchymal stem cells (BMSCs) can differentiate into osteoblasts and adipocytes to maintain bone homeostasis. Several studies have shown that lncRNAs are competitive endogenous RNAs that form a lncRNA-miRNA network by targeting miRNA for the regulation of bone/fat differentiation in BMSCs; this mechanism is closely related to the corresponding treatment of OP and is important in the development of novel OP-targeted therapies. However, by reviewing the current literature, it became clear that there are limited summaries discussing the effects of the lncRNA-miRNA network on osteogenic/adipogenic differentiation in BMSCs. Therefore, this article provides a review of the current literature to explore the impact of the lncRNA-miRNA network on the osteogenic/adipogenic differentiation of BMSCs, with the aim of providing a new theoretical basis for the treatment of OP.
Collapse
Affiliation(s)
- Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xiaxia Wang
- School of Tradional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Chunmei Wang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Ying Liu
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Bai Sun
- School of Tradional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Jie Zhang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Peng Gao
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Chunlu Yan
- School of Tradional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
32
|
Liu XP, Li JQ, Li RY, Cao GL, Feng YB, Zhang W. Loss of N-acetylglucosaminyl transferase V is involved in the impaired osteogenic differentiation of bone marrow mesenchymal stem cells. Exp Anim 2023; 72:413-424. [PMID: 37019682 PMCID: PMC10435351 DOI: 10.1538/expanim.22-0129] [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: 09/20/2022] [Accepted: 03/30/2023] [Indexed: 04/07/2023] Open
Abstract
The imbalance of bone resorption and bone formation causes osteoporosis (OP), a common skeletal disorder. Decreased osteogenic activity was found in the bone marrow cultures from N-acetylglucosaminyl transferase V (MGAT5)-deficient mice. We hypothesized that MGAT5 was associated with osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and involved in the pathological mechanisms of osteoporosis. To test this hypothesis, the mRNA and protein expression levels of MGAT5 were determined in bone tissues of ovariectomized (OVX) mice, a well-established OP model, and the role of MGAT5 in osteogenic activity was investigated in murine BMSCs. As expected, being accompanied by the loss of bone mass density and osteogenic markers (runt-related transcription factor 2, osteocalcin and osterix), a reduced expression of MGAT5 in vertebrae and femur tissues were found in OP mice. In vitro, knockdown of Mgat5 inhibited the osteogenic differentiation potential of BMSCs, as evidenced by the decreased expressions of osteogenic markers and less alkaline phosphatase and alizarin red S staining. Mechanically, knockdown of Mgat5 suppressed the nuclear translocation of β-catenin, thereby downregulating the expressions of downstream genes c-myc and axis inhibition protein 2, which were also associated with osteogenic differentiation. In addition, Mgat5 knockdown inhibited bone morphogenetic protein (BMP)/transforming growth factor (TGF)-β signaling pathway. In conclusion, MGAT5 may modulate the osteogenic differentiation of BMSCs via the β-catenin, BMP type 2 (BMP2) and TGF-β signals and involved in the process of OP.
Collapse
Affiliation(s)
- Xiao-Po Liu
- Department of Spinal Surgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Shijiazhuang 050051, Hebei, P.R. China
- Department of Orthopedics, Tangshan Gongren Hospital, No. 27, Wenhua Road, Tangshan 063000, Hebei, P.R. China
| | - Jia-Qi Li
- Department of Spinal Surgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Shijiazhuang 050051, Hebei, P.R. China
| | - Ruo-Yu Li
- Department of Spinal Surgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Shijiazhuang 050051, Hebei, P.R. China
| | - Guo-Long Cao
- Department of Orthopedics, Tangshan Gongren Hospital, No. 27, Wenhua Road, Tangshan 063000, Hebei, P.R. China
| | - Yun-Bo Feng
- Department of Orthopedics, Tangshan Gongren Hospital, No. 27, Wenhua Road, Tangshan 063000, Hebei, P.R. China
| | - Wei Zhang
- Department of Spinal Surgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Shijiazhuang 050051, Hebei, P.R. China
| |
Collapse
|
33
|
Li H, Hu S, Wu R, Zhou H, Zhang K, Li K, Lin W, Shi Q, Chen H, Lv S. 11β-Hydroxysteroid Dehydrogenase Type 1 Facilitates Osteoporosis by Turning on Osteoclastogenesis through Hippo Signaling. Int J Biol Sci 2023; 19:3628-3639. [PMID: 37496992 PMCID: PMC10367550 DOI: 10.7150/ijbs.82933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 06/08/2023] [Indexed: 07/28/2023] Open
Abstract
11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) is a key enzyme that transform cortisone to cortisol, which activates the endogenous glucocorticoid function. 11β-HSD1 has been observed to regulate skeletal metabolism, specifically within osteoblasts. However, the function of 11β-HSD1 in osteoclasts has not been elucidated. In this study, we observed increased 11β-HSD1 expression in osteoclasts within an osteoporotic mice model (ovariectomized mice). Then, 11β-HSD1 global knock-out or knock-in mice were employed to demonstrate its function in manipulating bone metabolism, showing significant bone volume decrease in 11β-HSD1 knock-in mice. Furthermore, specifically knock out 11β-HSD1 in osteoclasts, by crossing cathepsin-cre mice with 11β-HSD1flox/flox mice, presented significant protecting effect of skeleton when they underwent ovariectomy surgery. In vitro experiments showed the endogenous high expression of 11β-HSD1 lead to osteoclast formation and maturation. Meanwhile, we found 11β-HSD1 facilitated mature osteoclasts formation inhibited bone formation coupled H type vessel (CD31hiEmcnhi) growth through reduction of PDFG-BB secretion. Finally, transcriptome sequencing of 11β-HSD1 knock in osteoclast progenitor cells indicated the Hippo pathway1 was mostly enriched. Then, by suppression of YAP expression in Hippo signaling, we observed the redundant of osteoclasts formation even in 11β-HSD1 high expression conditions. In conclusion, our study demonstrated the role of 11β-HSD1 in facilitating osteoclasts formation and maturation through the Hippo signaling, which is a new therapeutic target to manage osteoporosis.
Collapse
Affiliation(s)
- Hanwen Li
- Department of Geriatric Endocrinology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing 210029, Jiangsu, China
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, China
- Department of Orthopedic, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Sihan Hu
- Department of Orthopedic, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Runze Wu
- Department of Endocrinology, Changshu No.2 People's Hospital, Changshu 215500, Jiangsu province, China
| | - Hongyou Zhou
- Department of Orthopedic, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Kai Zhang
- Department of Orthopedic, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ke Li
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Wenzheng Lin
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Qin Shi
- Department of Orthopedic, First Affiliated Hospital of Soochow University, Suzhou, China
- Orthopedic Institute of Soochow University, Suzhou, China
| | - Hao Chen
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Shan Lv
- Department of Geriatric Endocrinology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing 210029, Jiangsu, China
| |
Collapse
|
34
|
Ballesteros J, Rivas D, Duque G. The Role of the Kynurenine Pathway in the Pathophysiology of Frailty, Sarcopenia, and Osteoporosis. Nutrients 2023; 15:3132. [PMID: 37513550 PMCID: PMC10383689 DOI: 10.3390/nu15143132] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Tryptophan is an essential nutrient required to generate vitamin B3 (niacin), which is mainly involved in energy metabolism and DNA production. Alterations in tryptophan metabolism could have significant effects on aging and musculoskeletal health. The kynurenine pathway, essential in tryptophan catabolism, is modulated by inflammatory factors that are increased in older persons, a process known as inflammaging. Osteoporosis, sarcopenia, osteosarcopenia, and frailty have also been linked with chronically increased levels of inflammatory factors. Due to the disruption of the kynurenine pathway by chronic inflammation and/or changes in the gut microbiota, serum levels of toxic metabolites are increased and are associated with the pathophysiology of those conditions. In contrast, anabolic products of this pathway, such as picolinic acid, have demonstrated a positive effect on skeletal muscle and bone. In addition, physical activity can modulate this pathway by promoting the secretion of anabolic kynurenines. According to the evidence collected, kynurenines could have a promising role as biomarkers for osteoporosis sarcopenia, osteosarcopenia, and frailty in older persons. In addition, some of these metabolites could become important targets for developing new pharmacological treatments for these conditions.
Collapse
Affiliation(s)
- Juan Ballesteros
- Servicio de Geriatría, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Daniel Rivas
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Gustavo Duque
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Dr. Joseph Kaufmann Chair in Geriatric Medicine, Faculty of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
35
|
Liu F, Yuan L, Li L, Yang J, Liu J, Chen Y, Zhang J, Lu Y, Yuan Y, Cheng J. S-sulfhydration of SIRT3 combats BMSC senescence and ameliorates osteoporosis via stabilizing heterochromatic and mitochondrial homeostasis. Pharmacol Res 2023; 192:106788. [PMID: 37146925 DOI: 10.1016/j.phrs.2023.106788] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 04/23/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
Senescence of bone marrow mesenchymal stem cells (BMSCs) is one of the leading causes of osteoporosis. SIRT3, an essential NAD-dependent histone deacetylase, is highly correlated with BMSC senescence-mediated bone degradation and mitochondrial/heterochromatic disturbance. S-sulfhydration of cysteine residues favorably enhances SIRT3 activity by forming persulfides. Nevertheless, the underlying molecular mechanism of SIRT3 S-sulfhydration on mitochondrial/heterochromatic homeostasis involved in BMSC senescence remains unknown. Here, we demonstrated that CBS and CSE, endogenous hydrogen sulfide synthases, are downregulated with BMSC senescence. Exogenous H2S donor NaHS-mediated SIRT3 augmentation rescued the senescent phenotypes of BMSCs. Conversely, SIRT3 deletion accelerated oxidative stress-induced BMSC senescence through mitochondrial dysfunction and the detachment of the heterochromatic protein H3K9me3 from the nuclear envelope protein Lamin B1. H2S-mediated SIRT3 S-sulfhydration modification rescued the disorganized heterochromatin and fragmented mitochondria induced by the S-sulfhydration inhibitor dithiothreitol, thus leading to elevated osteogenic capacity and preventing BMSC senescence. The antisenescence effect of S-sulfhydration modification on BMSCs was abolished when the CXXC sites of the SIRT3 zinc finger motif were mutated. In vivo, aged mice-derived BMSCs pretreated with NaHS were orthotopically transplanted to the ovariectomy-induced osteoporotic mice, and we proved that SIRT3 ameliorates bone loss by inhibiting BMSC senescence. Overall, our study for the first time indicates a novel role of SIRT3 S-sulfhydration in stabilizing heterochromatin and mitochondrial homeostasis in counteracting BMSC senescence, providing a potential target for the treatment of degenerative bone diseases.
Collapse
Affiliation(s)
- Fei Liu
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Endocrinology & Metabolism and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Longhui Yuan
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lan Li
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingchao Yang
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingping Liu
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Younan Chen
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Endocrinology & Metabolism and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Zhang
- Core Facility of West China Hospital, Sichuan University, Chengdu P.R. China
| | - Yanrong Lu
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yujia Yuan
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Jingqiu Cheng
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Endocrinology & Metabolism and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Zhang J, Liu S, He Z, Liu H, Liu Y, Hu P, Li Z, Xu J, Luo E. Adiponectin overexpression promotes fracture healing through regulating the osteogenesis and adipogenesis balance in osteoporotic mice. J Bone Miner Metab 2023:10.1007/s00774-023-01420-3. [PMID: 37036534 DOI: 10.1007/s00774-023-01420-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023]
Abstract
INTRODUCTION Osteoporosis invariably manifests as loss of bone, which is replaced by adipose tissue; this can easily lead to fractures, accompanied by delayed and poor healing. Adiponectin (APN) balances osteogenesis and adipogenesis in bone marrow mesenchymal stem cells (BMSCs). Therefore, this study explored whether adiponectin promotes bone fracture healing by regulating the balance between osteogenesis and adipogenesis. MATERIALS AND METHODS We used adenovirus overexpression vectors carrying APN (Ad-APN-GFP) to treat ovariectomized (OVX) mouse BMSCs and osteoporotic bone fractures to investigate the role of APN in bone microenvironment metabolism in osteoporotic fractures. We subsequently established an OVX mice and bone fracture model using Ad-APN-GFP treatment to investigate whether APN could promote bone fracture healing in osteoporotic mice. RESULTS The experimental results showed that APN is a critical molecule in diverse differentiation directions in OVX mouse BMSCs, with pro-osteogenesis and anti-adipogenesis properties. Importantly, our study revealed that Ad-APN-GFP treatment facilitates bone generation and healing around the osteoporotic fracture ends. Moreover, we identified that Sirt1 and Wnt signaling were closely related to the pro-osteogenesis and anti-adipogenesis commitment of APN in OVX mouse BMSCs and femoral tissues. CONCLUSION We demonstrated that APN overexpression facilitates bone fracture healing in osteoporosis. Furthermore, APN overexpression promoted bone formation in OVX mouse BMSCs and bone fracture ends by regulating the balance between osteogenesis and adipogenesis both in vitro and in vivo.
Collapse
Affiliation(s)
- Ju Zhang
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Shibo Liu
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ze He
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Hanghang Liu
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yao Liu
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Pei Hu
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhongming Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Jiazhuang Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - En Luo
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
37
|
Zhu Y, Yang K, Cheng Y, Liu Y, Gu R, Liu X, Liu H, Zhang X, Liu Y. Apoptotic Vesicles Regulate Bone Metabolism via the miR1324/SNX14/SMAD1/5 Signaling Axis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205813. [PMID: 36670083 DOI: 10.1002/smll.202205813] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/17/2022] [Indexed: 06/17/2023]
Abstract
Mesenchymal stem cells (MSCs) are widely used in the treatment of diseases. After their in vivo application, MSCs undergo apoptosis and release apoptotic vesicles (apoVs). This study investigates the role of apoVs derived from human bone marrow mesenchymal stem cells (hBMMSCs) in bone metabolism and the molecular mechanism of the observed effects. The results show that apoVs can promote osteogenesis and inhibit osteoclast formation in vitro and in vivo. ApoVs may therefore attenuate the bone loss caused by primary and secondary osteoporosis and stimulate bone regeneration in areas of bone defect. The mechanisms responsible for apoV-induced bone regeneration include the release of miR1324, which inhibit expression of the target gene Sorting Nexin 14 (SNX14) and thus activate the SMAD1/5 pathway in target cells. Given that MSC-derived apoVs are easily obtained and stored, with low risks of immunological rejection and neoplastic transformation, The findings suggest a novel therapeutic strategy to treat bone loss, including via cell-free approaches to bone tissue engineering.
Collapse
Affiliation(s)
- Yuan Zhu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
- National Center of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Kunkun Yang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
| | - Yawen Cheng
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
| | - Yaoshan Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
| | - Ranli Gu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
| | - Xuenan Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
| | - Hao Liu
- The Central Laboratory, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
- National Center of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, 100081, China
- National Center of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| |
Collapse
|
38
|
Zhang Y, Zhou L, Fu Q, Liu Z. ANKRD1 activates the Wnt signaling pathway by modulating CAV3 expression and thus promotes BMSC osteogenic differentiation and bone formation in ovariectomized mice. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166693. [PMID: 36958710 DOI: 10.1016/j.bbadis.2023.166693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/16/2023] [Accepted: 03/15/2023] [Indexed: 03/25/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) are considered promising materials for treating bone diseases such as osteoporosis (OP). This research explored the functions and molecular mechanism of ankyrin repeat domain 1 (ANKRD1) in BMSC osteogenesis. An OP model in mice was established by bilateral ovariectomy. Manipulation of ANKRD1 expression in BMSCs or femurs was achieved by lentivirus infection. Increased ANKRD1 expression was observed in BMSCs during osteogenic induction. Silencing of ANKRD1 impaired the osteogenesis of BMSCs, as shown by the decreased alkaline phosphatase (ALP) activity, osteogenic gene (Runx2, Col1a1, Bglap, and Spp1) expression, and mineralized formation. ANKRD1-mediated promotion of osteogenesis was also reproduced in mouse MC3T3-E1 preosteoblastic cells. Activation of Wnt/β-catenin signaling, a well-known osteogenic stimulus, was also impaired in ANKRD1-silenced BMSCs. Overexpression of ANKRD1 resulted in the opposite effects on osteogenesis and Wnt/β-catenin signaling. Mechanistic studies revealed that ANKRD1 modulated caveolin-3 (CAV3) expression by reducing CAV3 ubiquitination, and the knockdown of CAV3 impaired the functions of ANKRD1. Additionally, a very low level of ANKRD1 was observed in the BMSCs from OP mice. Rescue of ANKRD1 significantly restored osteogenic differentiation and Wnt signaling activation in BMSCs from ovariectomized mice. The results of micro-CT, H&E staining, and IHC staining showed that ANKRD1 also promoted bone formation and Wnt activation and ameliorated pathological alterations in the femurs of OP mice. Collectively, this study demonstrated that ANKRD1 plays an important role in regulating the osteogenic differentiation of BMSCs and is a promising target for the treatment of OP and other bone diseases.
Collapse
Affiliation(s)
- Yiqi Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Long Zhou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Qin Fu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Ziyun Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
39
|
Autophagy mediates osteoporotic bone regeneration induced by micro-/nano-structured modification on hydroxyapatite bioceramics. ENGINEERED REGENERATION 2023. [DOI: 10.1016/j.engreg.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023] Open
|
40
|
Li W, Jiang WS, Su YR, Tu KW, Zou L, Liao CR, Wu Q, Wang ZH, Zhong ZM, Chen JT, Zhu SY. PINK1/Parkin-mediated mitophagy inhibits osteoblast apoptosis induced by advanced oxidation protein products. Cell Death Dis 2023; 14:88. [PMID: 36750550 PMCID: PMC9905061 DOI: 10.1038/s41419-023-05595-5+10.1038/s41419-020-2298-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2024]
Abstract
Osteoblast apoptosis plays an important role in age-related bone loss and osteoporosis. Our previous study revealed that advanced oxidation protein products (AOPPs) could induce nicotinamide adenine dinucleotide phosphate oxidase (NOX)-derived reactive oxygen species (ROS) production, cause mitochondrial membrane potential (ΔΨm) depolarization, trigger the mitochondria-dependent intrinsic apoptosis pathway, and lead to osteoblast apoptosis and ultimately osteopenia and bone microstructural destruction. In this study, we found that AOPPs also induced mitochondrial ROS (mtROS) generation in osteoblastic MC3T3-E1 cells, which was closely related to NOX-derived ROS, and aggravated the oxidative stress condition, thereby further promoting apoptosis. Removing excessive ROS and damaged mitochondria is the key factor in reversing AOPP-induced apoptosis. Here, by in vitro studies, we showed that rapamycin further activated PINK1/Parkin-mediated mitophagy in AOPP-stimulated MC3T3-E1 cells and significantly alleviated AOPP-induced cell apoptosis by eliminating ROS and damaged mitochondria. Our in vivo studies revealed that PINK1/Parkin-mediated mitophagy could decrease the plasma AOPP concentration and inhibit AOPP-induced osteoblast apoptosis, thus ameliorating AOPP accumulation-related bone loss, bone microstructural destruction and bone mineral density (BMD) loss. Together, our study indicated that therapeutic strategies aimed at upregulating osteoblast mitophagy and preserving mitochondrial function might have potential for treating age-related osteoporosis.
Collapse
Affiliation(s)
- Wei Li
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wang-Sheng Jiang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ya-Ru Su
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ke-Wu Tu
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lin Zou
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Orthopedics, Nanfang Hospital Taihe Branch, Southern Medical University, Guangzhou, China
| | - Cong-Rui Liao
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Wu
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zi-Han Wang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhao-Ming Zhong
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jian-Ting Chen
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Si-Yuan Zhu
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
41
|
Li W, Jiang WS, Su YR, Tu KW, Zou L, Liao CR, Wu Q, Wang ZH, Zhong ZM, Chen JT, Zhu SY. PINK1/Parkin-mediated mitophagy inhibits osteoblast apoptosis induced by advanced oxidation protein products. Cell Death Dis 2023; 14:88. [PMID: 36750550 PMCID: PMC9905061 DOI: 10.1038/s41419-023-05595-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 02/09/2023]
Abstract
Osteoblast apoptosis plays an important role in age-related bone loss and osteoporosis. Our previous study revealed that advanced oxidation protein products (AOPPs) could induce nicotinamide adenine dinucleotide phosphate oxidase (NOX)-derived reactive oxygen species (ROS) production, cause mitochondrial membrane potential (ΔΨm) depolarization, trigger the mitochondria-dependent intrinsic apoptosis pathway, and lead to osteoblast apoptosis and ultimately osteopenia and bone microstructural destruction. In this study, we found that AOPPs also induced mitochondrial ROS (mtROS) generation in osteoblastic MC3T3-E1 cells, which was closely related to NOX-derived ROS, and aggravated the oxidative stress condition, thereby further promoting apoptosis. Removing excessive ROS and damaged mitochondria is the key factor in reversing AOPP-induced apoptosis. Here, by in vitro studies, we showed that rapamycin further activated PINK1/Parkin-mediated mitophagy in AOPP-stimulated MC3T3-E1 cells and significantly alleviated AOPP-induced cell apoptosis by eliminating ROS and damaged mitochondria. Our in vivo studies revealed that PINK1/Parkin-mediated mitophagy could decrease the plasma AOPP concentration and inhibit AOPP-induced osteoblast apoptosis, thus ameliorating AOPP accumulation-related bone loss, bone microstructural destruction and bone mineral density (BMD) loss. Together, our study indicated that therapeutic strategies aimed at upregulating osteoblast mitophagy and preserving mitochondrial function might have potential for treating age-related osteoporosis.
Collapse
Affiliation(s)
- Wei Li
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wang-Sheng Jiang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ya-Ru Su
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ke-Wu Tu
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lin Zou
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Orthopedics, Nanfang Hospital Taihe Branch, Southern Medical University, Guangzhou, China
| | - Cong-Rui Liao
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Wu
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zi-Han Wang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhao-Ming Zhong
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jian-Ting Chen
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Si-Yuan Zhu
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
42
|
Li W, Jiang WS, Su YR, Tu KW, Zou L, Liao CR, Wu Q, Wang ZH, Zhong ZM, Chen JT, Zhu SY. PINK1/Parkin-mediated mitophagy inhibits osteoblast apoptosis induced by advanced oxidation protein products. Cell Death Dis 2023; 14:88. [PMID: 36750550 PMCID: PMC9905061 DOI: 10.1038/s41419-023-05595-5 10.1038/s41419-020-2298-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 07/11/2023]
Abstract
Osteoblast apoptosis plays an important role in age-related bone loss and osteoporosis. Our previous study revealed that advanced oxidation protein products (AOPPs) could induce nicotinamide adenine dinucleotide phosphate oxidase (NOX)-derived reactive oxygen species (ROS) production, cause mitochondrial membrane potential (ΔΨm) depolarization, trigger the mitochondria-dependent intrinsic apoptosis pathway, and lead to osteoblast apoptosis and ultimately osteopenia and bone microstructural destruction. In this study, we found that AOPPs also induced mitochondrial ROS (mtROS) generation in osteoblastic MC3T3-E1 cells, which was closely related to NOX-derived ROS, and aggravated the oxidative stress condition, thereby further promoting apoptosis. Removing excessive ROS and damaged mitochondria is the key factor in reversing AOPP-induced apoptosis. Here, by in vitro studies, we showed that rapamycin further activated PINK1/Parkin-mediated mitophagy in AOPP-stimulated MC3T3-E1 cells and significantly alleviated AOPP-induced cell apoptosis by eliminating ROS and damaged mitochondria. Our in vivo studies revealed that PINK1/Parkin-mediated mitophagy could decrease the plasma AOPP concentration and inhibit AOPP-induced osteoblast apoptosis, thus ameliorating AOPP accumulation-related bone loss, bone microstructural destruction and bone mineral density (BMD) loss. Together, our study indicated that therapeutic strategies aimed at upregulating osteoblast mitophagy and preserving mitochondrial function might have potential for treating age-related osteoporosis.
Collapse
Affiliation(s)
- Wei Li
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wang-Sheng Jiang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ya-Ru Su
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ke-Wu Tu
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lin Zou
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Orthopedics, Nanfang Hospital Taihe Branch, Southern Medical University, Guangzhou, China
| | - Cong-Rui Liao
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Wu
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zi-Han Wang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhao-Ming Zhong
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jian-Ting Chen
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Si-Yuan Zhu
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
43
|
Song Q, Zhou D, Du J, Li T, He X, Wang J, Chao A, Yu B, Shan C. Andrographis paniculata ameliorates estrogen deficiency-related osteoporosis by directing bone marrow mesenchymal stem cell fate. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:52. [PMID: 36819520 PMCID: PMC9929840 DOI: 10.21037/atm-22-1121] [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: 02/15/2022] [Accepted: 01/10/2023] [Indexed: 01/31/2023]
Abstract
Background Although Andrographis paniculata (AP) exhibits various biological functions such as anticancer, anti-inflammatory, antimalarial, antimicrobial, antioxidant, cardioprotective and immunomodulatory, its role in estrogen deficiency-related osteoporosis remains unclear. Methods Ovariectomy (OVX)-induced estrogen deficiency-related osteoporotic mouse models and sham mouse models were established using 8-week-old female C57BL/6J mice. Micro-computed tomography (µCT) scanning was performed to assess the skeletal phenotype. The differentiation potential of bone marrow mesenchymal stem cells (BMSCs) from the OVX and sham groups was assessed by osteogenic or adipogenic induction medium in vitro. To verify the effects of AP, alizarin red S (ARS) staining, alkaline phosphatase (ALP) staining and oil red O (ORO) staining, reverse transcription assay and quantitative real-time polymerase chain reaction were applied to detect the lineage differentiation ability of BMSCs. Results µCT scanning showed that AP treatment attenuated the osteoporotic phenotype in OVX-induced estrogen deficiency-related osteoporotic mice. The results of ARS staining, ALP staining, ORO staining and quantitative real-time polymerase chain reaction indicated that BMSCs from OVX-induced osteoporotic mice displayed a significant reduction in osteogenic differentiation and an increase in adipogenic differentiation, which could be reversed by AP treatment. Conclusions Our findings suggested that AP regulated the differentiation potential of BMSCs and ameliorated the development of estrogen deficiency-related osteoporosis, which might be an effective therapeutic method for estrogen deficiency-related osteoporosis.
Collapse
Affiliation(s)
- Qian Song
- Department of Osteo-internal Medicine, Tianjin Hospital, Tianjin, China
| | - Dongming Zhou
- Department of Traumatic Orthopedics, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia Autonomous Region, China
| | - Jianyang Du
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tao Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shenshan Central Hospital, Shanwei, China
| | - Xueyu He
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shenshan Central Hospital, Shanwei, China
| | - Jianxiong Wang
- Department of Endocrinology, Tianjin Hospital, Tianjin, China
| | - Aijun Chao
- Department of Osteo-internal Medicine, Tianjin Hospital, Tianjin, China
| | - Bingbing Yu
- Department of Orthopedics, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China;,Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chunyan Shan
- Department of Nephrology, Tianjin Medical University, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin, China
| |
Collapse
|
44
|
Zeng C, Wang S, Chen F, Wang Z, Li J, Xie Z, Ma M, Wang P, Shen H, Wu Y. Alpinetin alleviates osteoporosis by promoting osteogenic differentiation in BMSCs by triggering autophagy via PKA/mTOR/ULK1 signaling. Phytother Res 2023; 37:252-270. [PMID: 36104214 PMCID: PMC10087978 DOI: 10.1002/ptr.7610] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 01/19/2023]
Abstract
Osteoporosis, a systemic bone disease that is characterized by a reduction in bone mass and destruction of bone microstructure, is becoming a serious problem worldwide. Bone marrow mesenchymal stem cells (BMSCs) can differentiate into bone-forming osteoblasts, and play an important role in maintaining homeostasis of bone metabolism, thus being a potential therapeutic target for osteoporosis. Although the phytochemical alpinetin (APT) has been reported to possess a variety of pharmacological activities, it is still unclear whether APT can influence the osteogenic differentiation of on BMSCs and if it can improve osteoporosis. In this study, we found that APT treatment was able to enhance osteogenic differentiation levels of human BMSCs in vitro and mouse ones in vivo as revealed by multiple osteogenic markers including increased alkaline phosphatase activity and osteocalcin expression. Mechanistically, the protein kinase A (PKA)/mTOR/ULK1 signaling was involved in the action of APT to enhance the osteogenic differentiation of BMSCs. In addition, oral administration of APT significantly mitigated the bone loss in a dexamethasone-induced mouse model of osteoporosis through strengthening PKA signaling and autophagy. Altogether, these data demonstrate that APT promotes osteogenic differentiation in BMSCs by augmenting the PKA/mTOR/ULK1 autophagy signaling, highlighting its potential therapeutic application for treating osteoporotic diseases.
Collapse
Affiliation(s)
- Chenying Zeng
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Shan Wang
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Fenglei Chen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Ziming Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Jinteng Li
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Zhongyu Xie
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Mengjun Ma
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Peng Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Huiyong Shen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yanfeng Wu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| |
Collapse
|
45
|
Wang X, Yu F, Ye L. Epigenetic control of mesenchymal stem cells orchestrates bone regeneration. Front Endocrinol (Lausanne) 2023; 14:1126787. [PMID: 36950693 PMCID: PMC10025550 DOI: 10.3389/fendo.2023.1126787] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/17/2023] [Indexed: 03/08/2023] Open
Abstract
Recent studies have revealed the vital role of MSCs in bone regeneration. In both self-healing bone regeneration processes and biomaterial-induced healing of bone defects beyond the critical size, MSCs show several functions, including osteogenic differentiation and thus providing seed cells. However, adverse factors such as drug intake and body senescence can significantly affect the functions of MSCs in bone regeneration. Currently, several modalities have been developed to regulate MSCs' phenotype and promote the bone regeneration process. Epigenetic regulation has received much attention because of its heritable nature. Indeed, epigenetic regulation of MSCs is involved in the pathogenesis of a variety of disorders of bone metabolism. Moreover, studies using epigenetic regulation to treat diseases are also being reported. At the same time, the effects of epigenetic regulation on MSCs are yet to be fully understood. This review focuses on recent advances in the effects of epigenetic regulation on osteogenic differentiation, proliferation, and cellular senescence in MSCs. We intend to illustrate how epigenetic regulation of MSCs orchestrates the process of bone regeneration.
Collapse
Affiliation(s)
- Xiaofeng Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fanyuan Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Fanyuan Yu, ; Ling Ye,
| | - Ling Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Fanyuan Yu, ; Ling Ye,
| |
Collapse
|
46
|
Zhang W, Tao KT, Lin J, Liu P, Guan Z, Deng J, Wang D, Zeng H. The Role of m6A in Osteoporosis and the Differentiation of Mesenchymal Stem Cells into Osteoblasts and Adipocytes. Curr Stem Cell Res Ther 2023; 18:339-346. [PMID: 35733319 DOI: 10.2174/1574888x17666220621155341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/07/2022] [Accepted: 04/15/2022] [Indexed: 11/22/2022]
Abstract
Osteoporosis is a systemic disease in which bone mass decreases, leading to an increased risk of bone fragility and fracture. The occurrence of osteoporosis is believed to be related to the disruption of the differentiation of mesenchymal stem cells into osteoblasts and adipocytes. N6-adenylate methylation (m6A) modification is the most common type of chemical RNA modification and refers to a methylation modification formed by the nitrogen atom at position 6 of adenine (A), which is catalyzed by a methyltransferase. The main roles of m6A are the post-transcriptional level regulation of the stability, localization, transportation, splicing, and translation of RNA; these are key elements of various biological activities, including osteoporosis and the differentiation of mesenchymal stem cells into osteoblasts and adipocytes. The main focus of this review is the role of m6A in these two biological processes.
Collapse
Affiliation(s)
- Weifei Zhang
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, P.R. China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, P.R. China
| | - Ke Tao Tao
- Arthritis Clinical and Research Center, Peking University People's Hospital, Beijing 100044, China
| | - Jianjing Lin
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, P.R. China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, P.R. China
- Arthritis Clinical and Research Center, Peking University People's Hospital, Beijing 100044, China
| | - Peng Liu
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, P.R. China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, P.R. China
| | - Zhiping Guan
- Department of Spinal Surgery, Peking University Shenzhen Hospital, Lianhua Road, Shenzhen, 518000 Guangdong, China
| | - Jiapeng Deng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, P.R. China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, P.R. China
| | - Deli Wang
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, P.R. China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, P.R. China
| | - Hui Zeng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, P.R. China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, P.R. China
| |
Collapse
|
47
|
Xing L, Li Y, Li W, Liu R, Geng Y, Ma W, Qiao Y, Li J, Lv Y, Fang Y, Xu P. Expression of RUNX2/LAPTM5 in the Induction of MC3T3-e1 Mineralization and Its Possible Relationship with Autophagy. Tissue Eng Regen Med 2022; 19:1223-1235. [PMID: 36121636 PMCID: PMC9679133 DOI: 10.1007/s13770-022-00477-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/13/2022] [Accepted: 06/27/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The study aims to correlate osteogenesis with autophagy during the mineralization induction of MC3T3-e1 through exploring the expression of runt-related transcription factor 2 (RUNX2)/lysosomal-associated transmembrane protein 5 (LAMPT5). METHODS The induction of mineralization in MC3T3-e1 was followed by detecting the expressions of osteogenesis-related indexes such as RUNX2, alkaline phosphatase (ALP), osteocalcin (OCN), and LAPTM5 using RT-qPCR and Western blot from 0 to 14 days. Transmission electron microscope was utilised in visualizing the alterations of autophagosomes, which was followed by immunofluorescence detecting the subcellular localization of autophagy-related index sequestosome 1 (P62) and microtubule-associated protein 1 light 3 (LC3) protein and scrutinising the expression of P62 mRNA and P62 and LC3 proteins. RESULTS Induction of MC3T3-e1 mineralization demonstrated an increased expression of osteogenesis-related indicators such as RUNX2, ALP, OCN, and LAPTM5 (p < 0.05), as evident from the results of RT-qPCR and Western blot. Meanwhile, the expression of autophagosomes increased one day after mineralization induction and then experienced a gradual decline, and enhanced expression of LC3 protein was noted on days 1-2 of mineralization induction but was then followed by a corresponding reduce. In contrast, a continuous increase was reported in the expression of P62 mRNA and protein, respectively (p < 0.05). Up- and down-regulating RUNX2/LAPTM5 expression alone confirmed the aforementioned results. CONCLUSION It was therefore proposed that RUNX2 may be responsible for an early increase and then a gradual decrease in LAPTM5-mediated autophagy through the regulation of its high expression. Meanwhile, increased LAPTM5 expression in osteogenic mineralization presumed that RUNX2/LAPTM5 promoted autophagy and osteogenic expression, which may play a bridging role in the regulation of autophagy and osteogenesis.
Collapse
Affiliation(s)
- Lei Xing
- Department of Dental Implantology, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510150, China
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yanqin Li
- South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Wenhao Li
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Rong Liu
- Department of Dental Implantology, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510150, China
| | - Yuanming Geng
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weiqun Ma
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yu Qiao
- Department of Dental Implantology, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510150, China
| | - Jianwen Li
- Department of Dental Implantology, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510150, China
| | - Yingtao Lv
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Ying Fang
- Department of Dental Implantology, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510150, China.
| | - Pingping Xu
- Stomatological Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
48
|
Wu H, Lv Y, Wei F, Li C, Ge W, Du W. Comparative analysis of anti-osteoporosis efficacy in Radix Dipsaci before and after processing with salt based on spectrum-effect relationship. J Pharm Biomed Anal 2022; 221:115078. [DOI: 10.1016/j.jpba.2022.115078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/24/2022]
|
49
|
Autophagy Regulates Osteogenic Differentiation of Human Periodontal Ligament Stem Cells Induced by Orthodontic Tension. Stem Cells Int 2022; 2022:2983862. [PMID: 36248255 PMCID: PMC9553533 DOI: 10.1155/2022/2983862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/24/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022] Open
Abstract
Tooth movement is the core of orthodontics. Osteogenesis of the tension side under orthodontic force has great significance on tooth movement and stability, which involves complex mechanical and biological signal transduction. However, the mechanism remains unclear. Through in vitro cell studies, we observed the increased expression levels of osteogenesis-related factors and autophagy-related factors during the osteogenic differentiation of mesenchymal stem cells induced by orthodontic force. The change trend of autophagy-related factors and osteogenesis-related factors is similar, which indicates the involvement of autophagy in osteogenesis. In the study of autophagy-related gene ATG7 silenced cells, the expression level of autophagy was significantly inhibited, and the expression level of osteogenesis-related factors also decreased accordingly. Through drug regulation, we observed that the increase of autophagy level could effectively promote osteogenic differentiation, while the decrease of the autophagy level inhibited this process to some extent. Therefore, autophagy plays an important role in the osteogenic differentiation of mesenchymal stem cells induced by orthodontic force, which provides a novel idea useful for orthodontic treatment in promoting periodontal tissue remodeling and accelerating tooth movement.
Collapse
|
50
|
Sardar A, Gautam S, Sinha S, Rai D, Tripathi AK, Dhaniya G, Mishra PR, Trivedi R. Nanoparticles of naturally occurring PPAR-γ inhibitor betulinic acid ameliorates bone marrow adiposity and pathological bone loss in ovariectomized rats via Wnt/β-catenin pathway. Life Sci 2022; 309:121020. [PMID: 36191680 DOI: 10.1016/j.lfs.2022.121020] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 11/28/2022]
Abstract
AIMS Postmenopausal osteoporosis is one of the world's biggest yet unnoticed health issues. After ovariectomy, declined estrogen level significantly contributes to the elevation of bone marrow adiposity and bone loss leading to osteoporosis. Therapeutics to prevent osteoporosis addressing various aspects are now in short supply. In this study we made an approach to synthesize nanoparticles of naturally occurring PPAR-γ inhibitor, betulinic acid (BA/NPs) and tested the same in altered bone metabolisms developed after ovariectomy. MAIN METHODS The osteogenic efficacy of BA/NPs was established in human and rat primary osteoblast cells using qRT-PCR and immunoblot analysis. Furthermore, lineage allocations of multipotent bone marrow stromal cells were evaluated. Various aspects of altered bone metabolism after ovariectomy such as bone marrow adiposity and pathological bone loss were evaluated using μCT and histological assessments. KEY FINDINGS BA/NPs exert potential osteogenic efficacy by modulating RUNX2 and BMP2. Mechanistically BA/NPs regulate osteoblastogenesis through Wnt/β-catenin signaling. Further, BA/NPs showed the potential to inhibit the differentiation of multipotent BMSCs towards adipogenesis while favouring the osteogenic lineage. In the in vivo study, increased bone marrow adiposity was reduced in ovariectomized rats after BA/NPs treatment as assessed by histology and μCT analysis. Loss of bone mineral density as a hallmark of pathological bone loss was also abrogated by BA/NPs. SIGNIFICANCE Our findings imply that BA/NPs could be used further as a viable drug lead to counteract various pathophysiological challenges after menopause.
Collapse
Affiliation(s)
- Anirban Sardar
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shalini Gautam
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shradha Sinha
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Divya Rai
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | | | - Geeta Dhaniya
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Ritu Trivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|