1
|
Bessot A, Röhl J, Emmerich M, Klotz A, Ravichandran A, Meinert C, Waugh D, McGovern J, Gunter J, Bock N. ECM-mimicking hydrogel models of human adipose tissue identify deregulated lipid metabolism in the prostate cancer-adipocyte crosstalk under antiandrogen therapy. Mater Today Bio 2025; 30:101424. [PMID: 39866784 PMCID: PMC11764633 DOI: 10.1016/j.mtbio.2024.101424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/28/2025] Open
Abstract
Antiandrogen therapies are effectively used to treat advanced prostate cancer, but eventually cancer adaptation drives unresolved metastatic castration-resistant prostate cancer (mCRPC). Adipose tissue influences metabolic reprogramming in cancer and was proposed as a contributor to therapy resistance. Using extracellular matrix (ECM)-mimicking hydrogel coculture models of human adipocytes and prostate cancer cells, we show that adipocytes from subcutaneous or bone marrow fat have dissimilar responses under the antiandrogen Enzalutamide. We demonstrate that androgen receptor (AR)-dependent cancer cells (LNCaP) are more influenced by human adipocytes than AR-independent cells (C4-2B), with altered lipid metabolism and adipokine secretion. This response changes under Enzalutamide, with increased AR expression and adipogenic and lipogenic genes in cancer cells and decreased lipid content and gene dysregulation associated with insulin resistance in adipocytes. This is in line with the metabolic syndrome that men with mCRPC under Enzalutamide experience. The all-human, all-3D, models presented here provide a significant advance to dissect the role of fat in therapy response for mCRPC.
Collapse
Affiliation(s)
- Agathe Bessot
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
| | - Joan Röhl
- Faculty of Health Sciences and Medicine, Bond University, Robina, QLD, 4226, Australia
| | - Maria Emmerich
- School of Computation, Information and Technology, Technical University of Munich (TUM), Munich, Germany
| | - Anton Klotz
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Akhilandeshwari Ravichandran
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, QUT, Brisbane, QLD 4000, Australia
- Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies (CTET), QUT, Brisbane, QLD 4000, Australia
| | | | - David Waugh
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5005, Australia
| | - Jacqui McGovern
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies (CTET), QUT, Brisbane, QLD 4000, Australia
| | - Jenni Gunter
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
| | - Nathalie Bock
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling, and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia
| |
Collapse
|
2
|
Tanzhu G, Chen L, Ning J, Xue W, Wang C, Xiao G, Yang J, Zhou R. Metastatic brain tumors: from development to cutting-edge treatment. MedComm (Beijing) 2025; 6:e70020. [PMID: 39712454 PMCID: PMC11661909 DOI: 10.1002/mco2.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 12/24/2024] Open
Abstract
Metastatic brain tumors, also called brain metastasis (BM), represent a challenging complication of advanced tumors. Tumors that commonly metastasize to the brain include lung cancer and breast cancer. In recent years, the prognosis for BM patients has improved, and significant advancements have been made in both clinical and preclinical research. This review focuses on BM originating from lung cancer and breast cancer. We briefly overview the history and epidemiology of BM, as well as the current diagnostic and treatment paradigms. Additionally, we summarize multiomics evidence on the mechanisms of tumor occurrence and development in the era of artificial intelligence and discuss the role of the tumor microenvironment. Preclinically, we introduce the establishment of BM models, detailed molecular mechanisms, and cutting-edge treatment methods. BM is primarily treated with a comprehensive approach, including local treatments such as surgery and radiotherapy. For lung cancer, targeted therapy and immunotherapy have shown efficacy, while in breast cancer, monoclonal antibodies, tyrosine kinase inhibitors, and antibody-drug conjugates are effective in BM. Multiomics approaches assist in clinical diagnosis and treatment, revealing the complex mechanisms of BM. Moreover, preclinical agents often need to cross the blood-brain barrier to achieve high intracranial concentrations, including small-molecule inhibitors, nanoparticles, and peptide drugs. Addressing BM is imperative.
Collapse
Affiliation(s)
- Guilong Tanzhu
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Liu Chen
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Jiaoyang Ning
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Wenxiang Xue
- NHC Key Laboratory of RadiobiologySchool of Public HealthJilin UniversityChangchunJilinChina
| | - Ce Wang
- Department of RadiologyChina‐Japan Friendship HospitalBeijingChina
| | - Gang Xiao
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Jie Yang
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
- Department of DermatologyXiangya HospitalCentral South UniversityChangshaChina
| | - Rongrong Zhou
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
- Xiangya Lung Cancer CenterXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan ProvinceChina
| |
Collapse
|
3
|
XIANG X, NAN Y. [Research Progress on the Mechanism and Diagnostic Markers of Bone Metastasis
in Small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:697-703. [PMID: 39492585 PMCID: PMC11534582 DOI: 10.3779/j.issn.1009-3419.2024.106.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Indexed: 11/05/2024]
Abstract
Small cell lung cancer (SCLC) is a type of lung cancer with high malignant degree, rapid transformation, rapid invasion and metastasis, which is prone to early metastasis and poor prognosis. Bone metastases of SCLC occur in three stages: cancer cells proliferate at the primary site, break through local tissues, enter the blood circulation to form circulating tumor cells (CTCs), reach bone tissue through blood circulation, and take root and germinate to form new tumor sites with the support of the bone microenvironment. However, traditional imaging and pathology examinations have disadvantages such as low sensitivity, high cost and difficulty in implementation. Exploratory studies based on blood marker detection as screening and efficacy evaluation of SCLC bone metastases have been reported in recent years. By reviewing the molecular biological mechanism of SCLC bone metastasis formation, this paper found that conventional diagnostic methods such as imaging and pathological biopsy were inadequate in SCLC bone metastasis. The changes in hyaluronic acid, protein biomarkers, non-coding RNA, and biomarkers in liquid biopsy were earlier than the changes in imaging, which had the advantages of simple operation and good repeatability. It provides a new idea and method for the early diagnosis of SCLC bone metastasis, which is worthy of clinical application.
.
Collapse
|
4
|
Xu Y, Benedikt J, Ye L. Hyaluronic Acid Interacting Molecules Mediated Crosstalk between Cancer Cells and Microenvironment from Primary Tumour to Distant Metastasis. Cancers (Basel) 2024; 16:1907. [PMID: 38791985 PMCID: PMC11119954 DOI: 10.3390/cancers16101907] [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: 04/26/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Hyaluronic acid (HA) is a prominent component of the extracellular matrix, and its interactions with HA-interacting molecules (HAIMs) play a critical role in cancer development and disease progression. This review explores the multifaceted role of HAIMs in the context of cancer, focusing on their influence on disease progression by dissecting relevant cellular and molecular mechanisms in tumour cells and the tumour microenvironment. Cancer progression can be profoundly affected by the interactions between HA and HAIMs. They modulate critical processes such as cell adhesion, migration, invasion, and proliferation. The TME serves as a dynamic platform in which HAIMs contribute to the formation of a unique niche. The resulting changes in HA composition profoundly influence the biophysical properties of the TME. These modifications in the TME, in conjunction with HAIMs, impact angiogenesis, immune cell recruitment, and immune evasion. Therefore, understanding the intricate interplay between HAIMs and HA within the cancer context is essential for developing novel therapeutic strategies. Targeting these interactions offers promising avenues for cancer treatment, as they hold the potential to disrupt critical aspects of disease progression and the TME. Further research in this field is imperative for advancing our knowledge and the treatment of cancer.
Collapse
Affiliation(s)
- Yali Xu
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK;
- School of Engineering, Cardiff University, Cardiff CF24 3AA, UK;
| | | | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK;
| |
Collapse
|
5
|
Wu Z, Lu J, Loo A, Ho N, Nguyen D, Cheng PY, Mohammed AI, Cirillo N. Role of CD44 in Chemotherapy Treatment Outcome: A Scoping Review of Clinical Studies. Int J Mol Sci 2024; 25:3141. [PMID: 38542115 PMCID: PMC10970610 DOI: 10.3390/ijms25063141] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 01/31/2025] Open
Abstract
Cluster of differentiation 44 (CD44), a cell surface adhesion molecule overexpressed in cancer stem cells, has been implicated in chemoresistance. This scoping review, following PRISMA-ScR guidelines, systematically identified and evaluated clinical studies on the impact of CD44 expression on chemotherapy treatment outcomes across various cancer types. The search encompassed PubMed (1985-2023) and SCOPUS (1936-2023) databases, yielding a total of 12,659 articles, of which 40 met the inclusion criteria and were included in the qualitative synthesis using a predefined data extraction table. Data collected included the cancer type, sample size, interventions, control, treatment outcome, study type, expression of CD44 variants and isoforms, and effect of CD44 on chemotherapy outcome. Most of the studies demonstrated an association between increased CD44 expression and negative chemotherapeutic outcomes such as shorter overall survival, increased tumor recurrence, and resistance to chemotherapy, indicating a potential role of CD44 upregulation in chemoresistance in cancer patients. However, a subset of studies also reported non-significant relationships or conflicting results. In summary, this scoping review highlighted the breadth of the available literature investigating the clinical association between CD44 and chemotherapeutic outcomes. Further research is required to elucidate this relationship to aid clinicians in managing CD44-positive cancer patients.
Collapse
Affiliation(s)
- Zihao Wu
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Jillian Lu
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Andrew Loo
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Nathan Ho
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Danny Nguyen
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Po Yueh Cheng
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Ali I. Mohammed
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Nicola Cirillo
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
- School of Dentistry, University of Jordan, Amman 11942, Jordan
| |
Collapse
|
6
|
Han Y, Ki CS. Effect of Matrix Stiffness and Hepatocyte Growth Factor on Small Cell Lung Cancer Cells in Decellularized Extracellular Matrix-Based Hydrogels. Macromol Biosci 2024; 24:e2300356. [PMID: 37877161 DOI: 10.1002/mabi.202300356] [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: 08/04/2023] [Revised: 10/13/2023] [Indexed: 10/26/2023]
Abstract
Small cell lung cancer (SCLC) is one of lethal cancers resulting in very low 5-year-survival rate. Although its clinical treatment largely relies on chemotherapy, SCLC cell physiology in three-dimenstional (3D) matrix has been less explored. In this work, the tumor microenvironment is reconstructed with decellularized porcine pulmonary extracellular matrix (dECM) with hyaluronic acid. To modulate matrix stiffness, the methacrylate groups are introduced into both dECM and hyaluronic acid, followed by photocrosslinking with photoinitiator. The stiffness of the resulting dECM-based hydrogel covers the stiffness of normal or cancerous tissue with varying dECM content. The proliferation and cancer stem cell marker expression of encapsulated SCLC cells are promoted in a compliant hydrogel matrix, which has a low shear modulus similar to that of the normal tissue. The hepatocyte growth factor (HGF) that induces SCLC cell invasion and chemoresistance markedly increases invasiveness and gene expression levels of CD44 and Sox2 in the hydrogel matrix. In addition, HGF treatment causes higher resistance against anticancer drugs (cisplatin and paclitaxel) in the 3D microenvironment. These findings indicate that malignant SCLC can be recapitulated in a pulmonary dECM-based matrix.
Collapse
Affiliation(s)
- Yoobin Han
- Department of Agriculture, Forestry and Bioresources, Seoul National University, Seoul, 08826, Republic of Korea
| | - Chang Seok Ki
- Department of Agriculture, Forestry and Bioresources, Seoul National University, Seoul, 08826, Republic of Korea
- Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
7
|
Awodutire PO, Kattan MW, Ilori OS, Ilori OR. An Accelerated Failure Time Model to Predict Cause-Specific Survival and Prognostic Factors of Lung and Bronchus Cancer Patients with at Least Bone or Brain Metastases: Development and Internal Validation Using a SEER-Based Study. Cancers (Basel) 2024; 16:668. [PMID: 38339420 PMCID: PMC10854571 DOI: 10.3390/cancers16030668] [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: 12/21/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND This study addresses the significant challenge of low survival rates in patients with cause-specific lung cancer accompanied by bone or brain metastases. Recognizing the critical need for an effective predictive model, the research aims to establish survival prediction models using both parametric and non-parametric approaches. METHODS Clinical data from lung cancer patients with at least one bone or brain metastasis between 2000 and 2020 from the SEER database were utilized. Four models were constructed: Cox proportional hazard, Weibull accelerated failure time (AFT), log-normal AFT, and Zografos-Balakrishnan log-normal (ZBLN). Independent prognostic factors for cause-specific survival were identified, and model fit was evaluated using Akaike's and Bayesian information criteria. Internal validation assessed predictive accuracy and discriminability through the Harriel Concordance Index (C-index) and calibration plots. RESULTS A total of 20,412 patients were included, with 14,290 (70%) as the training cohort and 6122 (30%) validation. Independent prognostic factors selected for the study were age, race, sex, primary tumor site, disease grade, total malignant tumor in situ, metastases, treatment modality, and histology. Among the accelerated failure time (AFT) models considered, the ZBLN distribution exhibited the most robust model fit for the 3- and 5-year survival, as evidenced by the lowest values of Akaike's information criterion of 6322 and 79,396, and the Bayesian information criterion of 63,495 and 79,396, respectively. This outperformed other AFT and Cox models (AIC = [156,891, 211,125]; BIC = [158,848, 211,287]). Regarding predictive accuracy, the ZBLN AFT model achieved the highest concordance C-index (0.682, 0.667), a better performance than the Cox model (0.669, 0.643). The calibration curves of the ZBLN AFT model demonstrated a high degree of concordance between actual and predicted values. All variables considered in this study demonstrated significance at the 0.05 level for the ZBLN AFT model. However, differences emerged in the significant variations in survival times between subgroups. The study revealed that patients with only bone metastases have a higher chance of survival compared to only brain and those with bone and brain metastases. CONCLUSIONS The study highlights the underutilized but accurate nature of the accelerated failure time model in predicting lung cancer survival and identifying prognostic factors. These findings have implications for individualized clinical decisions, indicating the potential for screening and professional care of lung cancer patients with at least one bone or brain metastasis in the future.
Collapse
Affiliation(s)
| | | | - Oluwatosin Stephen Ilori
- Ladoke Akintola University of Technology Teaching Hospital, Ogbomosho 212102, Nigeria; (O.S.I.); (O.R.I.)
| | - Oluwatosin Ruth Ilori
- Ladoke Akintola University of Technology Teaching Hospital, Ogbomosho 212102, Nigeria; (O.S.I.); (O.R.I.)
| |
Collapse
|
8
|
Cirillo N. The Hyaluronan/CD44 Axis: A Double-Edged Sword in Cancer. Int J Mol Sci 2023; 24:15812. [PMID: 37958796 PMCID: PMC10649834 DOI: 10.3390/ijms242115812] [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: 09/24/2023] [Revised: 10/21/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Hyaluronic acid (HA) receptor CD44 is widely used for identifying cancer stem cells and its activation promotes stemness. Recent evidence shows that overexpression of CD44 is associated with poor prognosis in most human cancers and mediates therapy resistance. For these reasons, in recent years, CD44 has become a treatment target in precision oncology, often via HA-conjugated antineoplastic drugs. Importantly, HA molecules of different sizes have a dual effect and, therefore, may enhance or attenuate the CD44-mediated signaling pathways, as they compete with endogenous HA for binding to the receptors. The magnitude of these effects could be crucial for cancer progression, as well as for driving the inflammatory response in the tumor microenvironment. The increasingly common use of HA-conjugated drugs in oncology, as well as HA-based compounds as adjuvants in cancer treatment, adds further complexity to the understanding of the net effect of hyaluronan-CD44 activation in cancers. In this review, I focus on the significance of CD44 in malignancy and discuss the dichotomous function of the hyaluronan/CD44 axis in cancer progression.
Collapse
Affiliation(s)
- Nicola Cirillo
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia
| |
Collapse
|
9
|
Hao Y, Zhang F, Ma Y, Luo Y, Zhang Y, Yang N, Liu M, Liu H, Li J. Potential biomarkers for the early detection of bone metastases. Front Oncol 2023; 13:1188357. [PMID: 37404755 PMCID: PMC10315674 DOI: 10.3389/fonc.2023.1188357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023] Open
Abstract
The clinical manifestations of bone metastases are diversified while many sites remain asymptomatic at early stage. As the early diagnosis method is not perfect and the early symptoms of tumor bone metastasis are not typical, bone metastasis is not easy to be detected. Therefore, the search for bone metastasis-related markers is effective for timely detection of tumor bone metastases and the development of drugs to inhibit bone metastases. As a result, bone metastases can only be diagnosed when symptoms are found, increasing the risk of developing skeletal-related event (SREs), which significantly impairs the patient's quality of life. Therefore, the early diagnosis of bone metastases is of great importance for the treatment and prognosis of cancer patients. Changes of bone metabolism indexes appear earlier in bone metastases, but the traditional biochemical indexes of bone metabolism lack of specificity and could be interfered by many factors, which limits their application in the study of bone metastases. Some new biomarkers of bone metastases have good diagnostic value, such as proteins, ncRNAs, circulating tumor cells (CTCs). Therefore, this study mainly reviewed the initial diagnostic biomarkers of bone metastases which were expected to provide references for the early detection of bone metastases.
Collapse
Affiliation(s)
- Yang Hao
- Laboratory of Molecular Biology, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Feifan Zhang
- Laboratory of Molecular Biology, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
- Hunan University of Chinese Medicine, Changsha, China
| | - Yan Ma
- Laboratory of Molecular Biology, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
| | - Yage Luo
- Laboratory of Molecular Biology, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
| | - Yongyong Zhang
- Laboratory of Molecular Biology, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
| | - Ning Yang
- Laboratory of Molecular Biology, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
| | - Man Liu
- Laboratory of Molecular Biology, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
| | - Hongjian Liu
- Department of Orthopaedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jitian Li
- Laboratory of Molecular Biology, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
- Henan University of Chinese Medicine, Zhengzhou, China
- Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|