1
|
Qi Y, Wang Y, Yuan J, Xu Y, Pan H. Unveiling the therapeutic promise: exploring Lysophosphatidic Acid (LPA) signaling in malignant bone tumors for novel cancer treatments. Lipids Health Dis 2024; 23:204. [PMID: 38943207 PMCID: PMC11212261 DOI: 10.1186/s12944-024-02196-9] [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: 01/31/2024] [Accepted: 06/19/2024] [Indexed: 07/01/2024] Open
Abstract
Malignant bone tumors, including primary bone cancer and metastatic bone tumors, are a significant clinical challenge due to their high frequency of presentation, poor prognosis and lack of effective treatments and therapies. Bone tumors are often accompanied by skeletal complications such as bone destruction and cancer-induced bone pain. However, the mechanisms involved in bone cancer progression, bone metastasis and skeletal complications remain unclear. Lysophosphatidic acid (LPA), an intercellular lipid signaling molecule that exerts a wide range of biological effects mainly through specifically binding to LPA receptors (LPARs), has been found to be present at high levels in the ascites of bone tumor patients. Numerous studies have suggested that LPA plays a role in primary malignant bone tumors, bone metastasis, and skeletal complications. In this review, we summarize the role of LPA signaling in primary bone cancer, bone metastasis and skeletal complications. Modulating LPA signaling may represent a novel avenue for future therapeutic treatments for bone cancer, potentially improving patient prognosis and quality of life.
Collapse
Affiliation(s)
- Yichen Qi
- Huankui Academy, Nanchang University, Nanchang, 330031, China
- Neurological Institute of Jiangxi Province, Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
- Department of Neurology, Xiangya Hospital, Central South University, Jiangxi Hospital, National Regional Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, Jiangxi, 330038, P. R. China
| | - Yukai Wang
- School of Life Sciences, Nanchang University, Nanchang, 330031, China
- Neurological Institute of Jiangxi Province, Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
- Department of Neurology, Xiangya Hospital, Central South University, Jiangxi Hospital, National Regional Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, Jiangxi, 330038, P. R. China
| | - Jinping Yuan
- The First Clinical Medical College, Nanchang University, Nanchang, 330031, China
- Neurological Institute of Jiangxi Province, Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
- Department of Neurology, Xiangya Hospital, Central South University, Jiangxi Hospital, National Regional Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, Jiangxi, 330038, P. R. China
| | - Yufei Xu
- The First Clinical Medical College, Nanchang University, Nanchang, 330031, China
- Neurological Institute of Jiangxi Province, Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
- Department of Neurology, Xiangya Hospital, Central South University, Jiangxi Hospital, National Regional Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, Jiangxi, 330038, P. R. China
| | - Haili Pan
- Neurological Institute of Jiangxi Province, Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China.
- Department of Neurology, Xiangya Hospital, Central South University, Jiangxi Hospital, National Regional Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, Jiangxi, 330038, P. R. China.
| |
Collapse
|
2
|
Li S, Liu G, Hu S. Osteoporosis: interferon-gamma-mediated bone remodeling in osteoimmunology. Front Immunol 2024; 15:1396122. [PMID: 38817601 PMCID: PMC11137183 DOI: 10.3389/fimmu.2024.1396122] [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: 03/05/2024] [Accepted: 04/26/2024] [Indexed: 06/01/2024] Open
Abstract
As the world population ages, osteoporosis, the most common disease of bone metabolism, affects more than 200 million people worldwide. The etiology is an imbalance in bone remodeling process resulting in more significant bone resorption than bone remodeling. With the advent of the osteoimmunology field, the immune system's role in skeletal pathologies is gradually being discovered. The cytokine interferon-gamma (IFN-γ), a member of the interferon family, is an important factor in the etiology and treatment of osteoporosis because it mediates bone remodeling. This review starts with bone remodeling process and includes the cellular and key signaling pathways of bone remodeling. The effects of IFN-γ on osteoblasts, osteoclasts, and bone mass are discussed separately, while the overall effects of IFN-γ on primary and secondary osteoporosis are summarized. The net effect of IFN-γ on bone appears to be highly dependent on the environment, dose, concentration, and stage of cellular differentiation. This review focuses on the mechanisms of bone remodeling and bone immunology, with a comprehensive discussion of the relationship between IFN-γ and osteoporosis. Finding the paradoxical balance of IFN-γ in bone immunology and exploring the potential of its clinical application provide new ideas for the clinical treatment of osteoporosis and drug development.
Collapse
Affiliation(s)
- Siying Li
- The Orthopaedic Center, The First People’s Hospital of Wenling, Taizhou University Affiliated Wenling Hospital, Wenling, Zhejiang, China
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| | - Siwang Hu
- The Orthopaedic Center, The First People’s Hospital of Wenling, Taizhou University Affiliated Wenling Hospital, Wenling, Zhejiang, China
| |
Collapse
|
3
|
Liu Y, Li B, Chen X, Xiong H, Huang C. The effect of immunomodulatory drugs on bone metabolism of patients with multiple myeloma. Expert Rev Hematol 2024; 17:47-54. [PMID: 38319240 DOI: 10.1080/17474086.2024.2316090] [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: 09/27/2023] [Accepted: 02/05/2024] [Indexed: 02/07/2024]
Abstract
INTRODUCTION Immunomodulatory drugs (IMiDs) are widely used in the management of newly diagnosed and relapsed/refractory multiple myeloma patients. These agents show their potential effect on myeloma bone disease (MBD), including inhibition of osteoclasts activity and effects on osteoblasts differentiation. It is unclear whether these effects are direct, which may have an impact on bone formation markers when combined with proteasome inhibitors. AREAS COVERED This review summarizes the available evidence on the role of IMiDs in microenvironment regulation and their potential effects on bone metabolism. The literature search methodology consisted of searching PubMed for basic and clinical trials using medical subject terms. Included articles were screened and evaluated by the coauthors of this review. EXPERT OPINION As a therapeutic option, IMiDs directly affect preosteoblast/osteoclast differentiation. The combination of proteasome inhibitors may counteract the short-term up-regulation of osteogenic activity markers, and therefore intravenous zoledronic acid is recommended, however, obtaining a more significant myeloma response will have a long-term positive impact on myeloma bone disease.
Collapse
Affiliation(s)
- Yang Liu
- Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Clinical Research Institute, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Bo Li
- Department of Intensive Care Unit, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaomin Chen
- Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Clinical Research Institute, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Hao Xiong
- Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Clinical Research Institute, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunlan Huang
- Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Clinical Research Institute, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
4
|
Li Q, Zhang J, Xiao S, Hu M, Cheng J, Yao C, Zhuang Q. The impact of liver fibrosis on the progression of hepatocellular carcinoma via a hypoxia-immune-integrated prognostic model. Int Immunopharmacol 2023; 125:111136. [PMID: 37935090 DOI: 10.1016/j.intimp.2023.111136] [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: 06/30/2023] [Revised: 10/04/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023]
Abstract
The impact of liver fibrosis on the deterioration of hepatocellular carcinoma (HCC) remains controversial. We hope to explore this issue through establishing a fibrosis-hypoxia-glycolysis-immune related prognostic model. Liver fibrosis-related genes from Molecular Signatures Database were used to evaluate the degree of fibrosis in HCC patients from the TCGA database. The patients were divided into two groups using the fibrosis-related expression matrix based on the algorithm uniform manifold approximation and projection (UMAP) and evaluated for fibrosis by UMAP cluster and gene enrichment analysis. Prognostic model was constructed by differential analysis, LASSO, and multivariate regression analysis. Immune-infiltration analysis was performed by CIBERSORT. Quantitative PCR and immunohistochemistry were performed to measure the gene expression levels in HCC patients from our hospital. In 365 HCC patients from the TCGA database, 111 HCC patients with high fibrosis score have a worse prognosis than those with low fibrosis based on 129 genes related to liver fibrosis, which may be caused by the interaction between fibrosis, angiogenesis, hypoxia, glycolysis, inflammatory response, and high immune infiltration. We constructed a Fibrosis-Hypoxia-Glycolysis-Immune Prognostic Model (FHGISig), which could significantly predict disease progression in HCC patients. Furthermore, we revealed a close correlation between FHGISig and immune cell infiltration level as well as immune checkpoints. Finally, PCR results found TFF3 mRNA was significantly lower in cirrhotic HCC patients compared with non-cirrhotic ones. Liver fibrosis is a poor-prognostic factor for HCC, and our FHGISig could significantly predict disease progression, which could also be a potential predictive marker for immunotherapy in HCC patients.
Collapse
Affiliation(s)
- Qianyuan Li
- Department of Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Junbo Zhang
- Transplantation Center, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Sheng Xiao
- Department of Pathology, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Min Hu
- Department of Hematology, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570105, China
| | - Jie Cheng
- Department of Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Chenjiao Yao
- Department of General Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.
| | - Quan Zhuang
- Transplantation Center, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Research Center of National Health Ministry on Transplantation Medicine, Changsha, Hunan 410013, China.
| |
Collapse
|
5
|
Gao YM, Pei Y, Zhao FF, Wang L. Osteoclasts in Osteosarcoma: Mechanisms, Interactions, and Therapeutic Prospects. Cancer Manag Res 2023; 15:1323-1337. [PMID: 38027241 PMCID: PMC10661907 DOI: 10.2147/cmar.s431213] [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: 07/18/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
Osteosarcoma is an extremely malignant tumor, and its pathogenesis is complex and remains incompletely understood. Most cases of osteosarcoma are accompanied by symptoms of bone loss or result in pathological fractures due to weakened bones. Enhancing the survival rate of osteosarcoma patients has proven to be a long-standing challenge. Numerous studies mentioned in this paper, including in-vitro, in-vivo, and in-situ studies have consistently indicated a close association between the symptoms of bone loss associated with osteosarcoma and the presence of osteoclasts. As the sole cells capable of bone resorption, osteoclasts participate in a malignant cycle within the osteosarcoma microenvironment. These cells interact with osteoblasts and osteosarcoma cells, secreting various factors that further influence these cells, disrupting bone homeostasis, and shifting the balance toward bone resorption, thereby promoting the onset and progression of osteosarcoma. Moreover, the interaction between osteoclasts and various other cells types, such as tumor-associated macrophages, myeloid-derived suppressor cells, DCs cells, T cells, and tumor-associated fibroblasts in the osteosarcoma microenvironment plays a crucial role in disease progression. Consequently, understanding the role of osteoclasts in osteosarcoma has sparked significant interest. This review primarily examines the physiological characteristics and functional mechanisms of osteoclasts in osteosarcoma, and briefly discusses potential therapies targeting osteoclasts for osteosarcoma treatment. These studies provide fresh ideas and directions for future research on the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Yi-Ming Gao
- Department of Orthopedic Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
| | - Yan Pei
- Department of Orthopedic Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
| | - Fei-Fei Zhao
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
| | - Ling Wang
- Department of Orthopedic Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
| |
Collapse
|
6
|
Meng C, Jiang B, Liu W, Wang L, Zhao Z, Bai R, Zhao Y. MiR-217 regulates autophagy through OPG/RANKL/RANK in giant cell tumors. J Orthop Surg Res 2023; 18:346. [PMID: 37165403 PMCID: PMC10170763 DOI: 10.1186/s13018-023-03826-1] [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] [Received: 07/21/2022] [Accepted: 05/04/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Increasing evidence suggests that microRNAs (miRNAs) play a crucial role in cancer development and progression. Our previous study showed remarkably lower levels of miR-217 in GCT cells and tissues, and miR-217 re-expression inhibited the occurrence and development of GCT in vitro; however, the associated mechanisms remain unknown. Thus, this study aimed to explore the mechanisms underlying the proliferation inhibitory effect of miR-217 in GCT cells. METHODS The proliferative potential of the GCT cells was measured with an MTT assay and BrdU straining. Changes in GCT cell migration and invasion was assessed by a transwell assay. Finally, Western blot and RT-PCR assays were employed to evaluate OPG/RANKL/RANK signaling pathway-related protein expression. RESULTS The excessive upregulation of miR-217 markedly suppressed GCT cell proliferation and tumorigenesis both in vitro and in vivo. miR-217 overexpression could inhibit the OPG/RANKL/RANK signaling pathway in vitro and in vivo. Furthermore, ALP activity was significantly decreased in GCT cells following miR-217 treatment. Importantly, miR-217 could inhibit autophagy-related protein expression and autophagosome/autolysosome formation in GCT cells and tissues. CONCLUSION These results suggest that miR-217 upregulation could inhibit the occurrence and development of GCT by blocking autophagy. These findings offer an effective therapeutic target to improve the survival rates of patients with CGT in the future.
Collapse
Affiliation(s)
- Chenyang Meng
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010030, China
| | - Boyong Jiang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010030, China
| | - Wanlin Liu
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010030, China
| | | | - Zhenqun Zhao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010030, China
| | - Rui Bai
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010030, China.
| | - Yan Zhao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010030, China.
| |
Collapse
|
7
|
Influence of Chinese Herbal Formula on Bone Characteristics of Cobb Broiler Chickens. Genes (Basel) 2022; 13:genes13101865. [PMID: 36292748 PMCID: PMC9601401 DOI: 10.3390/genes13101865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/04/2022] Open
Abstract
To evaluate the prevention and treatment effect of a Chinese herbal formula (CHF) on the bone disease of Cobb broiler chickens, compare its efficacy with Bisphosphonates (BPs), and provide a theoretical basis for studying the nutritional regulation technology of CHF to improve the bone characteristics of broiler chickens. In this study, 560 one-day-old Cobb broiler chickens were examined for the influence of Chinese herbal formula (CHF) and Bisphosphonates (BPs). Different doses of CHF and BPs were added to the diet, and the 30- and 60-day-old live weight, tibial bone strength, the microstructure of the distal femur cancellous bone, blood biochemical indexes related to bone metabolism, and genes related to bone metabolism were determined and analyzed. The results showed that the live weight of Cobb broilers fed with CHF and BPs in the diet was as follows: The live weight of the CHF group was higher than that of the normal control (NC) group, while the live weight of the BPs group was lower than that of the NC group; the CHF and BPs improved the bone strength of Cobb broilers and increased the elastic modulus, yield strength, and maximum stress of the tibia. CHF and BPs increased the cancellous bone mineral density (BMD), bone tissue ratio (BV/TV), bone surface area tissue volume ratio (BS/TV), bone trabecular thickness (Tb.Th), and bone trabecular number (Tb.N) in the distal femur, and decreased the bone surface area bone volume ratio (BS/BV) and bone trabecular separation (Tb.Sp). Thus, the microstructure of the bone tissue of the distal femur was improved to a certain extent. Both the CHF and the BPs also increased the serum levels of the vitamin D receptor (VDR), osteoprotegerin (OPG), and alkaline phosphatase (ALP), and decreased the content of osteocalcin (OT). Meanwhile, CHF and BPs upregulated the expression of osteogenic genes (BMP-2, OPG, Runx-2) to promote bone formation and downregulated the expression of osteoclastic genes (RANK, RANKL, TNF-α) to inhibit bone resorption, thus affecting bone metabolism. Conclusion: The CHF could improve the skeletal characteristics of Cobb broilers by upregulating the expression of bone-forming-related genes and downregulating the expression of bone-breaking-related genes, thus preventing and controlling skeletal diseases in Cobb broilers. Its effect was comparable to that of BPs. Meanwhile, the CHF-H group achieved the best results in promoting the growth and improvement of the skeletal characteristics of Cobb broilers based on the live weight and skeletal-characteristics-related indexes.
Collapse
|
8
|
Dong HH, Li J, Kang L, Wei Q, Li Y. Simultaneous multiple myeloma and non‑small cell lung carcinoma: A case report and review of the literature. Oncol Lett 2022; 23:195. [PMID: 35572492 PMCID: PMC9100747 DOI: 10.3892/ol.2022.13315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/12/2022] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM) is the second commonest hematologic malignancy. Synchronous presentation of MM and lung cancer is a rare occurrence. The present study reports a case of MM combined with lung cancer and reviews previously reported cases of the co-existence of non-small cell lung carcinoma and MM. At Hebei General Hospital (Shijiazhuang, China), a 52-year-man was diagnosed with MM complicated by lung lesion. Lung computed tomography (CT) showed an increase in lesion density after the second cycle of chemotherapy. The lesion was surgically removed and the patient was diagnosed with non-small cell lung carcinoma by lung biopsy pathology. After the fifth cycle of VDT (bortezomib, dexamethasone and thalidomide), the patient received autologous stem cell transplantation. Immunohistochemical staining for CD38, CD138, CD39, CD203a and TNF-α were positive in both MM and lung cancer; CD73 was only positive in lung cancer. The present study described the rare event of the simultaneous occurrence of MM and lung adenocarcinoma and discussed the potential link between the two tumors. CD38 may play a role in MM and lung cancer by changing the bone marrow microenvironment through adenosine.
Collapse
Affiliation(s)
- Huan-Huan Dong
- Department of Graduate School, Hebei North University, Zhangjiakou, Hebei 075132, P.R. China
| | - Jing Li
- Department of Hematology, Hebei Province Hospital of Chinese Medicine, Shijiazhuang, Hebei 050013, P.R. China
| | - Lin Kang
- Department of Pathology, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Qiang Wei
- Department of Nuclear Medicine, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Yan Li
- Department of Hematology, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
9
|
Wang B, Wang H, Li Y, Song L. Lipid metabolism within the bone micro-environment is closely associated with bone metabolism in physiological and pathophysiological stages. Lipids Health Dis 2022; 21:5. [PMID: 34996476 PMCID: PMC8742318 DOI: 10.1186/s12944-021-01615-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Recent advances in society have resulted in the emergence of both hyperlipidemia and obesity as life-threatening conditions in people with implications for various types of diseases, such as cardiovascular diseases and cancer. This is further complicated by a global rise in the aging population, especially menopausal women, who mostly suffer from overweight and bone loss simultaneously. Interestingly, clinical observations in these women suggest that osteoarthritis may be linked to a higher body mass index (BMI), which has led many to believe that there may be some degree of bone dysfunction associated with conditions such as obesity. It is also common practice in many outpatient settings to encourage patients to control their BMI and lose weight in an attempt to mitigate mechanical stress and thus reduce bone pain and joint dysfunction. Together, studies show that bone is not only a mechanical organ but also a critical component of metabolism, and various endocrine functions, such as calcium metabolism. Numerous studies have demonstrated a relationship between metabolic dysfunction in bone and abnormal lipid metabolism. Previous studies have also regarded obesity as a metabolic disorder. However, the relationship between lipid metabolism and bone metabolism has not been fully elucidated. In this narrative review, the data describing the close relationship between bone and lipid metabolism was summarized and the impact on both the normal physiology and pathophysiology of these tissues was discussed at both the molecular and cellular levels.
Collapse
Affiliation(s)
- Bo Wang
- Department of Orthopedics, The first affiliated Hospital of Army Medical University, Army Medical University, Gaotanyan Street No. 30, Chongqing, 400038, China
| | - Heng Wang
- Department of Orthopedics, The first affiliated Hospital of Army Medical University, Army Medical University, Gaotanyan Street No. 30, Chongqing, 400038, China
- Battalion of Basic Medical Sciences, Army Medical University, Chongqing, 400038, China
| | - Yuancheng Li
- Battalion of Basic Medical Sciences, Army Medical University, Chongqing, 400038, China
| | - Lei Song
- Department of Orthopedics, The first affiliated Hospital of Army Medical University, Army Medical University, Gaotanyan Street No. 30, Chongqing, 400038, China.
| |
Collapse
|
10
|
Colombo MV, Bersini S, Arrigoni C, Gilardi M, Sansoni V, Ragni E, Candiani G, Lombardi G, Moretti M. Engineering the early bone metastatic niche through human vascularized immuno bone minitissues. Biofabrication 2021; 13. [PMID: 33735854 DOI: 10.1088/1758-5090/abefea] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/18/2021] [Indexed: 01/04/2023]
Abstract
Bone metastases occur in 65%-80% advanced breast cancer patients. Although significant progresses have been made in understanding the biological mechanisms driving the bone metastatic cascade, traditional 2Din vitromodels and animal studies are not effectively reproducing breast cancer cells (CCs) interactions with the bone microenvironment and suffer from species-specific differences, respectively. Moreover, simplifiedin vitromodels cannot realistically estimate drug anti-tumoral properties and side effects, hence leading to pre-clinical testing frequent failures. To solve this issue, a 3D metastatic bone minitissue (MBm) is designed with embedded human osteoblasts, osteoclasts, bone-resident macrophages, endothelial cells and breast CCs. This minitissue recapitulates key features of the bone metastatic niche, including the alteration of macrophage polarization and microvascular architecture, along with the induction of CC micrometastases and osteomimicry. The minitissue reflects breast CC organ-specific metastatization to bone compared to a muscle minitissue. Finally, two FDA approved drugs, doxorubicin and rapamycin, have been tested showing that the dose required to impair CC growth is significantly higher in the MBm compared to a simpler CC monoculture minitissue. The MBm allows the investigation of metastasis key biological features and represents a reliable tool to better predict drug effects on the metastatic bone microenvironment.
Collapse
Affiliation(s)
- Maria Vittoria Colombo
- Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland.,Biocompatibility and Cell Culture Laboratory 'BioCell', Department of Chemistry, Materials and Chemical Engineering 'Giulio Natta', Politecnico di Milano, 20133 Milano, Italy
| | - Simone Bersini
- Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland
| | - Chiara Arrigoni
- Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland
| | - Mara Gilardi
- Institute of Pathology, University Hospital of Basel, Basel 4056, Switzerland
| | - Veronica Sansoni
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry and Molecular Biology, 20161 Milano, Italy
| | - Enrico Ragni
- IRCCS Istituto Ortopedico Galeazzi, Orthopedic Biotechnology Lab, 20161 Milano, Italy
| | - Gabriele Candiani
- Biocompatibility and Cell Culture Laboratory 'BioCell', Department of Chemistry, Materials and Chemical Engineering 'Giulio Natta', Politecnico di Milano, 20133 Milano, Italy
| | - Giovanni Lombardi
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry and Molecular Biology, 20161 Milano, Italy.,Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań 61-871, Poland
| | - Matteo Moretti
- Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland.,IRCCS Istituto Ortopedico Galeazzi, Cell and Tissue Engineering Laboratory, 20161 Milano, Italy.,Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| |
Collapse
|
11
|
Kinget L, Roussel E, Lambrechts D, Boeckx B, Vanginderhuysen L, Albersen M, Rodríguez-Antona C, Graña-Castro O, Inglada-Pérez L, Verbiest A, Zucman-Rossi J, Couchy G, Caruso S, Laenen A, Baldewijns M, Beuselinck B. MicroRNAs Possibly Involved in the Development of Bone Metastasis in Clear-Cell Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13071554. [PMID: 33800656 PMCID: PMC8036650 DOI: 10.3390/cancers13071554] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Bone metastases cause substantial morbidity and implicate worse clinical outcomes for clear-cell renal cell carcinoma patients. MicroRNAs are small RNA molecules that modulate gene translation and are involved in the development of cancer and metastasis. We identified six microRNAs that are potentially specifically involved in metastasis to bone, of which two seem protective and four implicate a higher risk. This aids further understanding of the process of metastasizing to bone. Furthermore, these microRNA hold potential for biomarkers or therapeutic targets. Abstract Bone metastasis in clear-cell renal cell carcinoma (ccRCC) leads to substantial morbidity through skeletal related adverse events and implicates worse clinical outcomes. MicroRNAs (miRNA) are small non-protein coding RNA molecules with important regulatory functions in cancer development and metastasis. In this retrospective analysis we present dysregulated miRNA in ccRCC, which are associated with bone metastasis. In particular, miR-23a-3p, miR-27a-3p, miR-20a-5p, and miR-335-3p specifically correlated with the earlier appearance of bone metastasis, compared to metastasis in other organs. In contrast, miR-30b-3p and miR-139-3p were correlated with less occurrence of bone metastasis. These miRNAs are potential biomarkers and attractive targets for miRNA inhibitors or mimics, which could lead to novel therapeutic possibilities for bone targeted treatment in metastatic ccRCC.
Collapse
Affiliation(s)
- Lisa Kinget
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, 3000 Leuven, Belgium; (L.K.); (L.V.); (A.V.)
| | - Eduard Roussel
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (E.R.); (M.A.)
| | - Diether Lambrechts
- Laboratory of Translational Genetics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (D.L.); (B.B.)
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Bram Boeckx
- Laboratory of Translational Genetics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (D.L.); (B.B.)
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Loïc Vanginderhuysen
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, 3000 Leuven, Belgium; (L.K.); (L.V.); (A.V.)
| | - Maarten Albersen
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (E.R.); (M.A.)
| | | | - Osvaldo Graña-Castro
- Centro Nacional de Investigaciones Oncológicas (CNIO), 28040 Madrid, Spain; (C.R.-A.); (O.G.-C.)
| | - Lucía Inglada-Pérez
- Department of Statistics and Operational Research, Faculty of Medicine, Complutense University, 28040 Madrid, Spain;
| | - Annelies Verbiest
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, 3000 Leuven, Belgium; (L.K.); (L.V.); (A.V.)
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, INSERM, Functional Genomics of Solid Tumors Laboratory, Équipe Labellisée Ligue Nationale contre le Cancer, Labex OncoImmunology, F-75006 Paris, France; (J.Z.-R.); (G.C.); (S.C.)
| | - Gabrielle Couchy
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, INSERM, Functional Genomics of Solid Tumors Laboratory, Équipe Labellisée Ligue Nationale contre le Cancer, Labex OncoImmunology, F-75006 Paris, France; (J.Z.-R.); (G.C.); (S.C.)
| | - Stefano Caruso
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, INSERM, Functional Genomics of Solid Tumors Laboratory, Équipe Labellisée Ligue Nationale contre le Cancer, Labex OncoImmunology, F-75006 Paris, France; (J.Z.-R.); (G.C.); (S.C.)
| | | | | | - Benoit Beuselinck
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, 3000 Leuven, Belgium; (L.K.); (L.V.); (A.V.)
- Correspondence: ; Tel.: +32-16-346900
| |
Collapse
|
12
|
Apoptosis-Inducing TNF Superfamily Ligands for Cancer Therapy. Cancers (Basel) 2021; 13:cancers13071543. [PMID: 33801589 PMCID: PMC8036978 DOI: 10.3390/cancers13071543] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is a complex disease with apoptosis evasion as one of its hallmarks; therefore, apoptosis induction in transformed cells seems a promising approach as a cancer treatment. TNF apoptosis-inducing ligands, which are naturally present in the body and possess tumoricidal activity, are attractive candidates. The most studied proteins are TNF-α, FasL, and TNF-related apoptosis-inducing ligand (TRAIL). Over the years, different recombinant TNF family-derived apoptosis-inducing ligands and agonists have been designed. Their stability, specificity, and half-life have been improved because most of the TNF ligands have the disadvantages of having a short half-life and affinity to more than one receptor. Here, we review the outlook on apoptosis-inducing ligands as cancer treatments in diverse preclinical and clinical stages and summarize strategies of overcoming their natural limitations to improve their effectiveness.
Collapse
|
13
|
Characterization of Macrophages and Osteoclasts in the Osteosarcoma Tumor Microenvironment at Diagnosis: New Perspective for Osteosarcoma Treatment? Cancers (Basel) 2021; 13:cancers13030423. [PMID: 33498676 PMCID: PMC7866157 DOI: 10.3390/cancers13030423] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/06/2021] [Accepted: 01/15/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Due to the great genetic instability of osteosarcoma (OS), a recurrent molecular therapeutic target has not been identified to date. Therefore, characterization of the OS tumor microenvironment (TME) might offer new therapeutic perspectives. The OS2006 trial, originally designed to evaluate the impact of zoledronic acid (ZA, osteoclast-inhibitor) addition to conventional OS-therapies, was ended preliminary due to a negative impact on patient survival. Through retrospective biomarker analysis of the unique biological samples collected during the trial, we demonstrate here that ZA not only acts on harmful osteoclasts but also on protective macrophages, clarifying its detrimental effect. By multiplex immunohistochemistry, applied on additional OS biopsies, an important bipotent macrophage-population (CD168+/CD163+), homogenously distributed throughout OS tumor areas, was identified. These bipotent cells might play a determining role in the evolution of OS and offer a novel therapeutic approach. A clear definition of the macrophage populations present at diagnosis could re-enforce therapeutic decisions. Abstract Biological and histopathological techniques identified osteoclasts and macrophages as targets of zoledronic acid (ZA), a therapeutic agent that was detrimental for patients in the French OS2006 trial. Conventional and multiplex immunohistochemistry of microenvironmental and OS cells were performed on biopsies of 124 OS2006 patients and 17 surgical (“OSNew”) biopsies respectively. CSF-1R (common osteoclast/macrophage progenitor) and TRAP (osteoclast activity) levels in serum of 108 patients were correlated to response to chemotherapy and to prognosis. TRAP levels at surgery and at the end of the protocol were significantly lower in ZA+ than ZA− patients (padj = 0.0011; 0.0132). For ZA+-patients, an increase in the CSF-1R level between diagnosis and surgery and a high TRAP level in the serum at biopsy were associated with a better response to chemotherapy (p = 0.0091; p = 0.0251). At diagnosis, high CD163+ was associated with good prognosis, while low TRAP activity was associated with better overall survival in ZA− patients only. Multiplex immunohistochemistry demonstrated remarkable bipotent CD68+/CD163+ macrophages, homogeneously distributed throughout OS regions, aside osteoclasts (CD68+/CD163−) mostly residing in osteolytic territories and osteoid-matrix-associated CD68−/CD163+ macrophages. We demonstrate that ZA not only acts on harmful osteoclasts but also on protective macrophages, and hypothesize that the bipotent CD68+/CD163+ macrophages might present novel therapeutic targets.
Collapse
|
14
|
Deligiorgi MV, Panayiotidis MI, Trafalis DT. Repurposing denosumab in breast cancer beyond prevention of skeletal related events: Could nonclinical data be translated into clinical practice? Expert Rev Clin Pharmacol 2020; 13:1235-1252. [PMID: 33070648 DOI: 10.1080/17512433.2020.1839416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Denosumab is a human monoclonal antibody inhibiting the receptor activator of nuclear factor kappa-B ligand (RANKL). Initially approved as antiosteοporotic agent, denosumab is being currently pursued as a candidate for drug repurposing in oncology, especially breast cancer. AREAS COVERED The present review provides an overview of the therapeutic potential of denosumab in breast cancer beyond prevention of skeletal-related events (SREs), with focus on prevention of carcinogenesis in BRCA mutation carriers and on adjuvant treatment in early breast cancer patients. Study search was conducted on the following electronic databases: PubMed, Google scholar, Scopus.com, ClinicalTrials.gov, and European Union Clinical Trials Register from 2008 until June 2020. EXPERT OPINION Nonclinical data have established links between RANKL signaling and breast cancer initiation and progression, rationalizing exploring the potential bone-independent anticancer role of denosumab beyond SREs prevention. Preclinical and preliminary clinical data show that denosumab may inhibit carcinogenesis in BRCA mutation carriers. Denosumab adjuvant in early breast cancer has been shown, though inconsistently, to provide a disease-free survival benefit for a subgroup of patients. Despite promising results, the incorporation of denosumab in preventive and therapeutic protocols of breast cancer beyond prevention of SREs cannot be endorsed until further research consolidates its efficacy.
Collapse
Affiliation(s)
- Maria V Deligiorgi
- Department of Pharmacology, Clinical Pharmacology Unit, Faculty of Medicine, National and Kapodistrian University of Athens , Athens, Greece
| | - Mihalis I Panayiotidis
- Department of Electron Microscopy & Molecular Pathology, The Cyprus Institute of Neurology & Genetics , Nicosia, Cyprus.,The Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Dimitrios T Trafalis
- Department of Pharmacology, Clinical Pharmacology Unit, Faculty of Medicine, National and Kapodistrian University of Athens , Athens, Greece
| |
Collapse
|
15
|
Qian H, Lei T, Ye Z, Hu Y, Lei P. From the Performance to the Essence: The Biological Mechanisms of How Tantalum Contributes to Osteogenesis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5162524. [PMID: 32802853 PMCID: PMC7403943 DOI: 10.1155/2020/5162524] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022]
Abstract
Despite the brilliant bioactive performance of tantalum as an orthopedic biomaterial verified through laboratory researches and clinical practice in the past decades, scarce evidences about the essential mechanisms of how tantalum contributes to osteogenesis were systematically discussed. Up to now, a few studies have uncovered preliminarily the biological mechanism of tantalum in osteogenic differentiation and osteogenesis; it is of great necessity to map out the panorama through which tantalum contributes to new bone formation. This minireview summarized current advances to demonstrate the probable signaling pathways and underlying molecular cascades through which tantalum orchestrates osteogenesis, which mainly contain Wnt/β-catenin signaling pathway, BMP signaling pathway, TGF-β signaling pathway, and integrin signaling pathway. Limits of subsistent studies and further work are also discussed, providing a novel vision for the study and application of tantalum.
Collapse
Affiliation(s)
- Hu Qian
- Department of Orthopedics, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
- Xiangya School of Medicine, Central South University, 172 Tongzipo Road, Changsha, 410008 Hunan, China
| | - Ting Lei
- Department of Orthopedics, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Zhimin Ye
- Xiangya School of Medicine, Central South University, 172 Tongzipo Road, Changsha, 410008 Hunan, China
| | - Yihe Hu
- Department of Orthopedics, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Pengfei Lei
- Department of Orthopedics, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| |
Collapse
|
16
|
Luan F, Li X, Cheng X, Huangfu L, Han J, Guo T, Du H, Wen X, Ji J. TNFRSF11B activates Wnt/β-catenin signaling and promotes gastric cancer progression. Int J Biol Sci 2020; 16:1956-1971. [PMID: 32398963 PMCID: PMC7211174 DOI: 10.7150/ijbs.43630] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 03/23/2020] [Indexed: 12/24/2022] Open
Abstract
Tumor necrosis factor receptor superfamily member 11B (TNFRSF11B) has been studied to be involved in the development and progression of several human malignancies. However, little is unveiled regarding the complex mechanisms of TNFRSF11B in human gastric cancer (GC). The clinical significance of TNFRSF11B was assessed in 70 and 160 GC tissues using immunohistochemistry method and gene microarray analysis, respectively. The biological function of TNFRSF11B was studied in vitro and in vivo assays. Immunofluorescence assay was used to evaluate the expression of β-catenin in the nucleus. The expression of β-catenin and related protein was determined by Western blot. The interaction between TNFRSF11B and GSK3β was detected by co-immunoprecipitation. We demonstrated that TNFRSF11B was highly expressed in the cytoplasm of GC and associated with the patient poor outcome. Our studies showed that TNFRSF11B in GC cells significantly promoted cell proliferation, migration, invasion in vitro and tumorigenic ability in vitro and in vivo. Meanwhile, TNFRSF11B inhibited GC cell apoptosis. The proportion of nuclear active β-catenin showed positively correlation with TNFRSF11B expression. TNFRSF11B directly combined with GSK-3β upregulating its phosphorylation, and increased expression of β-catenin and its downstream effectors. Collectively, these findings demonstrate that TNFRSF11B promote the aggressive phenotypes of GC cells and activated Wnt/β-catenin signaling. Accordingly, TNFRSF11B had potential as a biomarker and inhibition of TNFRSF11B expression might offer a new therapeutic target for GC patients.
Collapse
Affiliation(s)
- Fengming Luan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of education), Division of gastrointestinal Cancer Translational Research laboratory, Peking University Cancer Hospital & Institute, Beijing, China
- Department of gastrointestinal surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaomei Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of education), Division of gastrointestinal Cancer Translational Research laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaojing Cheng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of education), Division of gastrointestinal Cancer Translational Research laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Longtao Huangfu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of education), Division of gastrointestinal Cancer Translational Research laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jing Han
- Key laboratory of Carcinogenesis and Translational Research (Ministry of education), Division of gastrointestinal Cancer Translational Research laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ting Guo
- Key laboratory of Carcinogenesis and Translational Research (Ministry of education), Division of gastrointestinal Cancer Translational Research laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hong Du
- Key laboratory of Carcinogenesis and Translational Research (Ministry of education), Division of gastrointestinal Cancer Translational Research laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xianzi Wen
- Key laboratory of Carcinogenesis and Translational Research (Ministry of education), Division of gastrointestinal Cancer Translational Research laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiafu Ji
- Key laboratory of Carcinogenesis and Translational Research (Ministry of education), Division of gastrointestinal Cancer Translational Research laboratory, Peking University Cancer Hospital & Institute, Beijing, China
- Department of gastrointestinal surgery, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
17
|
Sun Y, Liang Y, Li Z, Xia N. Liraglutide Promotes Osteoblastic Differentiation in MC3T3-E1 Cells by ERK5 Pathway. Int J Endocrinol 2020; 2020:8821077. [PMID: 33488706 PMCID: PMC7780226 DOI: 10.1155/2020/8821077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/03/2020] [Accepted: 11/24/2020] [Indexed: 01/29/2023] Open
Abstract
Liraglutide is a glucagon-like peptide-1 analogue widely used in the treatment of type 2 diabetes mellitus. However, the effects of liraglutide on osteoblast proliferation and differentiation in MC3T3-E1 cells have not been fully elucidated. In the present study, the promoting effects of liraglutide were investigated in MC3T3-E1 cells. The results indicated that cell viability was affected following the treatment of the cells with different concentrations of liraglutide (0, 10, 100, and 1000 nM) at different time periods of culture (24, 48, and 72 h). Moreover, the activity levels of alkaline phosphatase and the number of mineralized nodules in MC3T3-E1 cells were significantly increased following treatment with 100 nM liraglutide. The mRNA and protein levels of Col-1, OPG, and OCN in MC3T3-E1 cells were also markedly increased following 100 nM liraglutide treatment compared with those of the control group. The expression levels of the ERK5 signaling pathway key proteins (MEK5, p-ERK5, ERK5, and NUR77) were increased following liraglutide treatment in MC3T3-E1 cells, and the gene expression levels of the ERK5 signaling pathway were also elevated. Moreover, the ERK5 inhibitor XMD8-92 significantly decreased the expression levels of p-ERK5 and NUR77 as well as the proliferation of osteoblasts. However, these changes could be rescued by liraglutide to some extent. Therefore, these results revealed that liraglutide may promote osteoblastic differentiation and proliferation in MC3T3-E1 cells via the activation of the ERK5 signaling pathway.
Collapse
Affiliation(s)
- Yue Sun
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yuzhen Liang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zhengming Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Ning Xia
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|