1
|
Zhang L, Pan Y, Pan F, Huang S, Wang F, Zeng Z, Chen H, Tian X. MATN4 as a target gene of HIF-1α promotes the proliferation and metastasis of osteosarcoma. Aging (Albany NY) 2024; 16:10462-10476. [PMID: 38889378 PMCID: PMC11236324 DOI: 10.18632/aging.205941] [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: 08/28/2023] [Accepted: 03/03/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Osteosarcoma is a highly malignant bone tumor that exhibits rapid growth and early metastasis. Hypoxia plays a pivotal role in promoting the proliferation and metastasis of osteosarcoma through a series of molecular events, which are partially mediated and regulated by HIF-1α. However, the regulatory network associated with HIF-1α in osteosarcoma remains limited. Therefore, the objective of this study was to identify critical hypoxia-associated genes and investigate their effects and molecular mechanisms in osteosarcoma cells. METHODS Through bioinformatics analysis, matrilin-4 (MATN4) was identified as a crucial gene associated with hypoxia. The expression of MATN4 and HIF-1α was assessed using immunohistochemistry, RT-qPCR, and western blotting. The proliferative capacity of osteosarcoma cells was assessed through the utilization of CCK-8, EDU staining, and colony formation assays. The effects of MATN4 on the mobility of OS cells were evaluated using wound-healing assays and transwell assays. The interaction between MATN4 and HIF-1α was detected through chromatin immunoprecipitation. RESULTS MATN4 is overexpressed in osteosarcoma tissue and cells, particularly in osteosarcoma cells with high metastatic potential. Knockdown of MATN4 inhibits the proliferation, migration, and invasion abilities of osteosarcoma cells and reverses the promoting effects of hypoxia on these functions. Additionally, HIF-1α binds to MATN4 and upregulates its expression. Interestingly, knockdown of HIF-1α reduces the stimulatory effects of MATN4 overexpression on the proliferation, migration, and invasion of osteosarcoma cells under hypoxic conditions. CONCLUSIONS Taken together, our results suggest that MATN4 is regulated by HIF-1α and confers a more aggressive phenotype on OS cells. This evidence suggests that MATN4 may act as a potential target for OS diagnosis and treatment.
Collapse
Affiliation(s)
- Lu Zhang
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550000, Guizhou, China
| | - Yujie Pan
- Department of Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, Guizhou, China
- Department of Traumatic Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, Guizhou, China
| | - Feng Pan
- Department of Bone and Joint Surgery, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang 550000, Guizhou, China
| | - Songsong Huang
- Department of Pathology, The Afflicted Hospital of Guizhou Medical University, Guiyang 550000, Guizhou, China
| | - Fengyan Wang
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550000, Guizhou, China
| | - Zhirui Zeng
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Guizhou Medical University, Guiyang 550000, China
| | - Houping Chen
- Department of Orthopedics, Guiyang Maternal and Child Health-Care Hospital, Guiyang 550000, China
| | - Xiaobin Tian
- School of Clinical Medicine, Guizhou Medical University, Guiyang 550000, Guizhou, China
- Department of Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, Guizhou, China
| |
Collapse
|
2
|
Perivoliotis K, Ntellas P, Dadouli K, Samara AA, Sotiriou S, Ioannou M, Tepetes K. Microvessel Density (MVD) in Patients with Osteosarcoma: A Systematic Review and Meta-Analysis. Cancer Invest 2024; 42:104-114. [PMID: 38345052 DOI: 10.1080/07357907.2024.2311266] [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: 03/12/2022] [Accepted: 01/24/2024] [Indexed: 02/21/2024]
Abstract
A meta-analysis was designed and conducted to estimate the effect of tumoral microvessel density (MVD) on the survival of patients with osteosarcoma. There was no difference between high and low MVD regarding the overall (OS) and disease-free (DFS) survival. Low MVD tumors displayed a lower DFS at the third year of follow-up. Although primary metastases did not affect the mean MVD measurements, tumors with a good chemotherapy response had a higher MVD value. Although no significant differences between tumoral MVD, OS and DFS were found, good adjuvant therapy responders had a significant higher vascularization pattern.
Collapse
Affiliation(s)
| | - Panagiotis Ntellas
- Department of Pathology, University Hospital of Larissa, Larissa, Greece
| | - Katerina Dadouli
- Postgraduate Programme (MSc): Research Methodology in Biomedicine, Biostatistics and Clinical Bioinformatics at University of Thessaly, Thessaly, Greece
| | - Athina A Samara
- Department of Surgery, University Hospital of Larissa, Larissa, Greece
| | - Sotirios Sotiriou
- Department of Embryology, University Hospital of Larissa, Larissa, Greece
| | - Maria Ioannou
- Department of Pathology, University Hospital of Larissa, Larissa, Greece
| | | |
Collapse
|
3
|
Todosenko N, Khlusov I, Yurova K, Khaziakhmatova O, Litvinova L. Signal Pathways and microRNAs in Osteosarcoma Growth and the Dual Role of Mesenchymal Stem Cells in Oncogenesis. Int J Mol Sci 2023; 24:ijms24108993. [PMID: 37240338 DOI: 10.3390/ijms24108993] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
The major challenges in Osteosarcoma (OS) therapy are its heterogeneity and drug resistance. The development of new therapeutic approaches to overcome the major growth mechanisms of OS is urgently needed. The search for specific molecular targets and promising innovative approaches in OS therapy, including drug delivery methods, is an urgent problem. Modern regenerative medicine focuses on harnessing the potential of mesenchymal stem cells (MSCs) because they have low immunogenicity. MSCs are important cells that have received considerable attention in cancer research. Currently, new cell-based methods for using MSCs in medicine are being actively investigated and tested, especially as carriers for chemotherapeutics, nanoparticles, and photosensitizers. However, despite the inexhaustible regenerative potential and known anticancer properties of MSCs, they may trigger the development and progression of bone tumors. A better understanding of the complex cellular and molecular mechanisms of OS pathogenesis is essential to identify novel molecular effectors involved in oncogenesis. The current review focuses on signaling pathways and miRNAs involved in the development of OS and describes the role of MSCs in oncogenesis and their potential for antitumor cell-based therapy.
Collapse
Affiliation(s)
- Natalia Todosenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Igor Khlusov
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
- Laboratory of Cellular and Microfluidic Technologies, Siberian State Medical University, 2, Moskovskii Trakt, 634050 Tomsk, Russia
| | - Kristina Yurova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Olga Khaziakhmatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Larisa Litvinova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
- Laboratory of Cellular and Microfluidic Technologies, Siberian State Medical University, 2, Moskovskii Trakt, 634050 Tomsk, Russia
| |
Collapse
|
4
|
Zhang W, Lyu P, Andreev D, Jia Y, Zhang F, Bozec A. Hypoxia-immune-related microenvironment prognostic signature for osteosarcoma. Front Cell Dev Biol 2022; 10:974851. [PMID: 36578780 PMCID: PMC9791087 DOI: 10.3389/fcell.2022.974851] [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: 07/08/2022] [Accepted: 11/25/2022] [Indexed: 12/14/2022] Open
Abstract
Introduction: Increasing evidences have shown that hypoxia and the immune microenvironment play vital roles in the development of osteosarcoma. However, reliable gene signatures based on the combination of hypoxia and the immune status for prognostic prediction of osteosarcoma have so far not been identified. Methods: The individual hypoxia and immune status of osteosarcoma patients were identified with transcriptomic profiles of a training cohort from the TARGET database using ssGSEA and ESTIMATE algorithms, respectively. Lasso regression and stepwise Cox regression were performed to develop a hypoxia-immune-based gene signature. An independent cohort from the GEO database was used for external validation. Finally, a nomogram was constructed based on the gene signature and clinical features to improve the risk stratification and to quantify the risk assessment for individual patients. Results: Hypoxia and the immune status were significantly associated with the prognosis of osteosarcoma patients. Seven hypoxia- and immune-related genes (BNIP3, SLC38A5, SLC5A3, CKMT2, S100A3, CXCL11 and PGM1) were identified to be involved in our prognostic signature. In the training cohort, the prognostic signature discriminated high-risk patients with osteosarcoma. The hypoxia-immune-based gene signature proved to be a stable and predictive method as determined in different datasets and subgroups of patients. Furthermore, a nomogram based on the prognostic signature was generated to optimize the risk stratification and to quantify the risk assessment. Similar results were validated in an independent GEO cohort, confirming the stability and reliability of the prognostic signature. Conclusion: The hypoxia-immune-based prognostic signature might contribute to the optimization of risk stratification for survival and personalized management of osteosarcoma patients.
Collapse
|
5
|
Khojastehnezhad MA, Seyedi SMR, Raoufi F, Asoodeh A. Association of hypoxia-inducible factor 1 expressions with prognosis role as a survival prognostic biomarker in the patients with osteosarcoma: a meta-analysis. Expert Rev Mol Diagn 2022; 22:1099-1106. [PMID: 36510847 DOI: 10.1080/14737159.2022.2157719] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Osteosarcoma, the most prevalent primary bone cancer, tends to relapse or metastasize quickly. Hypoxia-inducible factor-1 alpha (HIF-1α) affects tumor metabolism, differentiation, angiogenesis, proliferation, and metastasis. Many studies have investigated the possible inconsistent prognostic value of HIF-1 α. This study evaluated the correlation between HIF-1 α expression and prognosis in osteosarcoma patients. METHODS : A total of 978 patients from 12 studies were followed up. A meta-analysis was conducted on articles investigating HIF-1 α prognostic value in osteosarcoma patients. The authors excluded articles with overlapping data, duplicate data, reviews, case reports, and letters that did not provide original data. Calculation of the hazard ratios (HR) and pooled risk ratios (RR) with corresponding 95% confidence intervals were used to determine the association degree (CIs). RESULTS It was determined that HIF-1 α in osteosarcoma patients had a prognostic value based on the RRs and HRs. The results showed that high HIF-1 α expression was associated with a worse prognosis when compared to low or undetectable HIF-1 α expression. CONCLUSION HIF-1 α overexpression was found to predict poor outcomes in osteosarcomas. The present study suggests that HIF-1α is a useful prognostic biomarker to predict OS in patients with osteosarcoma.
Collapse
Affiliation(s)
| | | | - Farveh Raoufi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ahmad Asoodeh
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
6
|
Chen W, Wu P, Yu F, Luo G, Qing L, Tang J. HIF-1α Regulates Bone Homeostasis and Angiogenesis, Participating in the Occurrence of Bone Metabolic Diseases. Cells 2022; 11:cells11223552. [PMID: 36428981 PMCID: PMC9688488 DOI: 10.3390/cells11223552] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/16/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
In the physiological condition, the skeletal system's bone resorption and formation are in dynamic balance, called bone homeostasis. However, bone homeostasis is destroyed under pathological conditions, leading to the occurrence of bone metabolism diseases. The expression of hypoxia-inducible factor-1α (HIF-1α) is regulated by oxygen concentration. It affects energy metabolism, which plays a vital role in preventing bone metabolic diseases. This review focuses on the HIF-1α pathway and describes in detail the possible mechanism of its involvement in the regulation of bone homeostasis and angiogenesis, as well as the current experimental studies on the use of HIF-1α in the prevention of bone metabolic diseases. HIF-1α/RANKL/Notch1 pathway bidirectionally regulates the differentiation of macrophages into osteoclasts under different conditions. In addition, HIF-1α is also regulated by many factors, including hypoxia, cofactor activity, non-coding RNA, trace elements, etc. As a pivotal pathway for coupling angiogenesis and osteogenesis, HIF-1α has been widely studied in bone metabolic diseases such as bone defect, osteoporosis, osteonecrosis of the femoral head, fracture, and nonunion. The wide application of biomaterials in bone metabolism also provides a reasonable basis for the experimental study of HIF-1α in preventing bone metabolic diseases.
Collapse
|
7
|
Ma J, Guo Z, Yang X, Zhu Y. Exploration of various roles of hypoxia genes in osteosarcoma. Sci Rep 2022; 12:18293. [PMID: 36316355 PMCID: PMC9622735 DOI: 10.1038/s41598-022-17622-0] [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: 11/10/2021] [Accepted: 07/28/2022] [Indexed: 12/31/2022] Open
Abstract
Osteosarcoma is a primary malignant tumor that often metastasizes in orthopedic diseases. Although multi-drug chemotherapy and surgical treatment have significantly improved the survival and prognosis of patients with osteosarcoma, the survival rate is still very low due to frequent metastases in patients with osteosarcoma. In-depth exploration of the relationship between various influencing factors of osteosarcoma is very important for screening promising therapeutic targets. This study used multivariate COX regression analysis to select the hypoxia genes SLC2A1 and FBP1 in patients with osteosarcoma, and used the expression of these two genes to divide the patients with osteosarcoma into high-risk and low-risk groups. Then, we first constructed a prognostic model based on the patient's risk value and compared the survival difference between the high expression group and the low expression group. Second, in the high expression group and the low expression group, compare the differences in tumor invasion and inflammatory gene expression between the two groups of immune cells. Finally, the ferroptosis-related genes with differences between the high expression group and the low expression group were screened, and the correlation between these genes was analyzed. In the high-risk group, immune cells with higher tumor invasiveness, macrophages M0 and immune cells with lower invasiveness included: mast cell resting, regulatory T cells (Tregs) and monocytes. Finally, among genes related to ferroptosis, we found AKR1C2, AKR1C1 and ALOX15 that may be related to hypoxia. These ferroptosis-related genes were discovered for the first time in osteosarcoma. Among them, the hypoxia gene FBP1 is positively correlated with the ferroptosis genes AKR1C1 and ALOX15, and the hypoxia gene SLC2A1 is negatively correlated with the ferroptosis genes AKR1C2, AKR1C1 and ALOX15. This study constructed a prognostic model based on hypoxia-related genes SLC2A1 and FBP1 in patients with osteosarcoma, and explored their correlation with immune cells, inflammatory markers and ferroptosis-related genes. This indicates that SLC2A1 and FBP1 are promising targets for osteosarcoma research.
Collapse
Affiliation(s)
- Jimin Ma
- grid.186775.a0000 0000 9490 772XDepartment of Orthopedics, Fuyang Hospital of Anhui Medical University, 99 Huangshan Road, Fuhe Modern Industrial Park, Yingzhou District, Fuyang, 236000 Anhui Province China
| | - Ziming Guo
- grid.186775.a0000 0000 9490 772XDepartment of Orthopedics, Fuyang Hospital of Anhui Medical University, 99 Huangshan Road, Fuhe Modern Industrial Park, Yingzhou District, Fuyang, 236000 Anhui Province China
| | - Xuefei Yang
- grid.186775.a0000 0000 9490 772XDepartment of Orthopedics, Fuyang Hospital of Anhui Medical University, 99 Huangshan Road, Fuhe Modern Industrial Park, Yingzhou District, Fuyang, 236000 Anhui Province China
| | - Yakun Zhu
- grid.186775.a0000 0000 9490 772XDepartment of Orthopedics, Fuyang Hospital of Anhui Medical University, 99 Huangshan Road, Fuhe Modern Industrial Park, Yingzhou District, Fuyang, 236000 Anhui Province China
| |
Collapse
|
8
|
Pierrevelcin M, Flacher V, Mueller CG, Vauchelles R, Guerin E, Lhermitte B, Pencreach E, Reisch A, Muller Q, Doumard L, Boufenghour W, Klymchenko AS, Foppolo S, Nazon C, Weingertner N, Martin S, Briandet C, Laithier V, Di Marco A, Bund L, Obrecht A, Villa P, Dontenwill M, Entz-Werlé N. Engineering Novel 3D Models to Recreate High-Grade Osteosarcoma and its Immune and Extracellular Matrix Microenvironment. Adv Healthc Mater 2022; 11:e2200195. [PMID: 36057996 DOI: 10.1002/adhm.202200195] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/24/2022] [Indexed: 01/27/2023]
Abstract
Osteosarcoma (OS) is the most common primary bone cancer, where the overall 5-year surviving rate is below 20% in resistant forms. Accelerating cures for those poor outcome patients remains a challenge. Nevertheless, several studies of agents targeting abnormal cancerous pathways have yielded disappointing results when translated into clinic because of the lack of accurate OS preclinical modeling. So, any effort to design preclinical drug testing may consider all inter-, intra-, and extra-tumoral heterogeneities throughout models mimicking extracellular and immune microenvironment. Therefore, the bioengineering of patient-derived models reproducing the OS heterogeneity, the interaction with tumor-associated macrophages (TAMs), and the modulation of oxygen concentrations additionally to recreation of bone scaffold is proposed here. Eight 2D preclinical models mimicking several OS clinical situations and their TAMs in hypoxic conditions are developed first and, subsequently, the paired 3D models faithfully preserving histological and biological characteristics are generated. It is possible to shape reproducibly M2-like macrophages cultured with all OS patient-derived cell lines in both dimensions. The final 3D models pooling all heterogeneity features are providing accurate proliferation and migration data to understand the mechanisms involved in OS and immune cells/biomatrix interactions and sustained such that engineered 3D preclinical systems will improve personalized medicine.
Collapse
Affiliation(s)
- Marina Pierrevelcin
- UMR CNRS 7021, Laboratory of Biomaging and Pathologies, Faculté de Pharmacie, 74 route du Rhin, Illkirch, 67405, France
| | - Vincent Flacher
- CNRS UPR3572, Laboratory I2CT - Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, 2, Allée Konrad Roentgen, Strasbourg, 67084, France
| | - Christopher G Mueller
- CNRS UPR3572, Laboratory I2CT - Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, 2, Allée Konrad Roentgen, Strasbourg, 67084, France
| | - Romain Vauchelles
- UMR CNRS 7021, Laboratory of Biomaging and Pathologies, Faculté de Pharmacie, 74 route du Rhin, Illkirch, 67405, France
| | - Eric Guerin
- Department of Cancer Molecular Genetics, Laboratory of Biochemistry and Molecular Biology, University Hospital of Strasbourg, 1 avenue Molière, Strasbourg, 67098, France
| | - Benoît Lhermitte
- Pathology department, University Hospital of Strasbourg, 1 avenue Molière, Strasbourg, 67098, France
| | - Erwan Pencreach
- Department of Cancer Molecular Genetics, Laboratory of Biochemistry and Molecular Biology, University Hospital of Strasbourg, 1 avenue Molière, Strasbourg, 67098, France
| | - Andreas Reisch
- UMR CNRS 7021, Laboratory of Biomaging and Pathologies, Faculté de Pharmacie, 74 route du Rhin, Illkirch, 67405, France
| | - Quentin Muller
- CNRS UPR3572, Laboratory I2CT - Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, 2, Allée Konrad Roentgen, Strasbourg, 67084, France
| | - Layal Doumard
- CNRS UPR3572, Laboratory I2CT - Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, 2, Allée Konrad Roentgen, Strasbourg, 67084, France
| | - Wacym Boufenghour
- CNRS UPR3572, Laboratory I2CT - Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, 2, Allée Konrad Roentgen, Strasbourg, 67084, France
| | - Andrey S Klymchenko
- UMR CNRS 7021, Laboratory of Biomaging and Pathologies, Faculté de Pharmacie, 74 route du Rhin, Illkirch, 67405, France
| | - Sophie Foppolo
- UMR CNRS 7021, Laboratory of Biomaging and Pathologies, Faculté de Pharmacie, 74 route du Rhin, Illkirch, 67405, France
| | - Charlotte Nazon
- Pediatric Onco-hematology unit, University Hospital of Strasbourg, 1 avenue Molière, Strasbourg, 67098, France
| | - Noelle Weingertner
- Pathology department, University Hospital of Strasbourg, 1 avenue Molière, Strasbourg, 67098, France
| | - Sophie Martin
- UMR CNRS 7021, Laboratory of Biomaging and Pathologies, Faculté de Pharmacie, 74 route du Rhin, Illkirch, 67405, France
| | - Claire Briandet
- Pediatric Onco-hematology unit, Hospital of "Le Bocage"- University Hospital of Dijon, 1 bd Jeanne d'Arc, Dijon, 21079, France
| | - Véronique Laithier
- Pediatric Onco-hematology unit, University Hospital of Besançon, 3, boulevard A. Fleming, Besançon, 25030, France
| | - Antonio Di Marco
- Department of Orthopedic Surgery and Traumatology, University Hospital of Strasbourg, 1 avenue Molière, Strasbourg, 67098, France
| | - Laurent Bund
- Department of Pediatric Surgery, University Hospital of Strasbourg, 1 avenue Molière, Strasbourg, 67098, France
| | - Adeline Obrecht
- PCBIS Plate-forme de chimie biologique intégrative de Strasbourg, UMS 3286 CNRS, University of Strasbourg, Labex Medalis, 300 Bld Sébastien Brant, Illkirch, 67412, France
| | - Pascal Villa
- PCBIS Plate-forme de chimie biologique intégrative de Strasbourg, UMS 3286 CNRS, University of Strasbourg, Labex Medalis, 300 Bld Sébastien Brant, Illkirch, 67412, France
| | - Monique Dontenwill
- UMR CNRS 7021, Laboratory of Biomaging and Pathologies, Faculté de Pharmacie, 74 route du Rhin, Illkirch, 67405, France
| | - Natacha Entz-Werlé
- UMR CNRS 7021, Laboratory of Biomaging and Pathologies, Faculté de Pharmacie, 74 route du Rhin, Illkirch, 67405, France.,Pediatric Onco-hematology unit, University Hospital of Strasbourg, 1 avenue Molière, Strasbourg, 67098, France
| |
Collapse
|
9
|
Nazon C, Pierrevelcin M, Willaume T, Lhermitte B, Weingertner N, Marco AD, Bund L, Vincent F, Bierry G, Gomez-Brouchet A, Redini F, Gaspar N, Dontenwill M, Entz-Werle N. Together Intra-Tumor Hypoxia and Macrophagic Immunity Are Driven Worst Outcome in Pediatric High-Grade Osteosarcomas. Cancers (Basel) 2022; 14:cancers14061482. [PMID: 35326631 PMCID: PMC8945994 DOI: 10.3390/cancers14061482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 12/25/2022] Open
Abstract
Simple Summary Radiological and immunohistochemical data were correlated with the outcome in a retrospective monocentric cohort of 30 pediatric osteosarcomas (OTS). A necrotic volume of more than 50 cm3 at diagnosis was significantly linked to a worse overall survival (OS). Regarding immunohistochemical analyses, an overexpression of hypoxic markers, such as HIF-1α and anhydrase carbonic IX (CAIX), was significantly linked to a worse OS, while pS6-RP hyperexpression was correlated with a better survival. We also featured that CD68 positive cells, representative of macrophagic M1 polarization, were mostly associated with HIF-1α and CAIX hyperexpressions and that M2-like polarization, mostly related to CD163 positivity, was correlated to mTor activation. These findings, involving clinical, radiological and biology data, allowed us to hypothesize a dual signature association ready to use routinely in future protocols. Abstract Background: Osteosarcomas (OTS) represent the most common primary bone cancer diagnosed in adolescents and young adults. Despite remarkable advances, there are no objective molecular or imaging markers able to predict an OTS outcome at diagnosis. Focusing on biomarkers contributing broadly to treatment resistance, we examine the interplay between the tumor-associated macrophages and intra-tumor hypoxia. Methods: Radiological and immunohistochemical (IHC) data were correlated with the outcome in a retrospective and monocentric cohort of 30 pediatric OTS. We studied hypoxic (pS6, phospho-mTor, HIF-1α and carbonic anhydrase IX (CAIX)) and macrophagic (CD68 and CD163) biomarkers. Results: The imaging analyses were based on MRI manual volumetric measures on axial post-contrast T1 weighted images, where, for each tumor, we determined the necrotic volume and its ratio to the entire tumor volume. When they were above 50 cm3 and 20%, respectively, they correlated with a worse overall survival (p = 0.0072 and p = 0.0136, respectively) and event-free survival (p = 0.0059 and p = 0.0143, respectively). IHC assessments enable a significant statistical link between HIF-1α/CAIX hyper-expressions, CD68+ cells and a worse outcome, whereas activation of mTor pathway was linked to a better survival rate and CD163+ cells. Conclusions: This study evidenced the links between hypoxia and immunity in OTS, as their poor outcome may be related to a larger necrotic volume on diagnostic MRI and, in biopsies, to a specific IHC profile.
Collapse
Affiliation(s)
- Charlotte Nazon
- Pediatric Onco-Hematology Unit, University Hospital of Strasbourg, 1 Avenue Molière, CEDEX, 67098 Strasbourg, France; (C.N.); (F.V.)
| | - Marina Pierrevelcin
- CNRS UMR 7021, Laboratory of Bioimaging and Pathologies, Faculty of Pharmacy, 74 Route du Rhin, 67401 Illkirch, France; (M.P.); (B.L.); (M.D.)
| | - Thibault Willaume
- Radiology Department, University Hospital of Strasbourg, 1 Avenue Molière, CEDEX, 67098 Strasbourg, France; (T.W.); (G.B.)
| | - Benoît Lhermitte
- CNRS UMR 7021, Laboratory of Bioimaging and Pathologies, Faculty of Pharmacy, 74 Route du Rhin, 67401 Illkirch, France; (M.P.); (B.L.); (M.D.)
- Pathology Department, University Hospital of Strasbourg, 1 Avenue Molière, CEDEX, 67098 Strasbourg, France;
| | - Noelle Weingertner
- Pathology Department, University Hospital of Strasbourg, 1 Avenue Molière, CEDEX, 67098 Strasbourg, France;
| | - Antonio Di Marco
- Department of Orthopedic Surgery and Traumatology, University Hospital of Strasbourg, 1 Avenue Molière, CEDEX, 67098 Strasbourg, France;
| | - Laurent Bund
- Department of Pediatric Surgery, University Hospital of Strasbourg, 1 Avenue Molière, CEDEX, 67098 Strasbourg, France;
| | - Florence Vincent
- Pediatric Onco-Hematology Unit, University Hospital of Strasbourg, 1 Avenue Molière, CEDEX, 67098 Strasbourg, France; (C.N.); (F.V.)
| | - Guillaume Bierry
- Radiology Department, University Hospital of Strasbourg, 1 Avenue Molière, CEDEX, 67098 Strasbourg, France; (T.W.); (G.B.)
| | - Anne Gomez-Brouchet
- Department of Pathology, University Hospital of Toulouse, 1 Avenue Irène Joliot Curie, 31100 Toulouse, France;
| | - Françoise Redini
- INSERM UMR1238, PHY-OS, Bone Sarcomas and Remodeling of Calcified Tissues, Nantes University, 44000 Nantes, France;
| | - Nathalie Gaspar
- Department of Oncology for Children and Adolescents, Gustave Roussy, 94805 Villejuif, France;
- INSERM U1015, Gustave Roussy, University of Paris-Saclay, 94805 Villejuif, France
- University of Paris-Saclay, 91400 Orsay, France
| | - Monique Dontenwill
- CNRS UMR 7021, Laboratory of Bioimaging and Pathologies, Faculty of Pharmacy, 74 Route du Rhin, 67401 Illkirch, France; (M.P.); (B.L.); (M.D.)
| | - Natacha Entz-Werle
- Pediatric Onco-Hematology Unit, University Hospital of Strasbourg, 1 Avenue Molière, CEDEX, 67098 Strasbourg, France; (C.N.); (F.V.)
- CNRS UMR 7021, Laboratory of Bioimaging and Pathologies, Faculty of Pharmacy, 74 Route du Rhin, 67401 Illkirch, France; (M.P.); (B.L.); (M.D.)
- Correspondence: ; Tel.: +33-3-88-12-83-96; Fax: +33-3-88-12-80-92
| |
Collapse
|
10
|
Gomez-Brouchet A, Illac C, Ledoux A, Fortin PY, de Barros S, Vabre C, Despas F, Peries S, Casaroli C, Bouvier C, Aubert S, de Pinieux G, Larousserie F, Galmiche L, Talmont F, Pitson S, Maddelein ML, Cuvillier O. Sphingosine Kinase-1 Is Overexpressed and Correlates with Hypoxia in Osteosarcoma: Relationship with Clinicopathological Parameters. Cancers (Basel) 2022; 14:cancers14030499. [PMID: 35158767 PMCID: PMC8833796 DOI: 10.3390/cancers14030499] [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: 12/08/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 11/16/2022] Open
Abstract
The Sphingosine kinase-1/Sphingosine 1-Phosphate (SphK1/S1P) signaling pathway is overexpressed in various cancers, and is instrumental for the adaptation to hypoxia in a number of solid tumor models, but no data are available in osteosarcoma. Here we report that SphK1 and the S1P1 receptor are involved in HIF-1α accumulation in hypoxic osteosarcoma cells. FTY720 (Fingolimod), which targets SphK1 and S1P1, prevented HIF-1α accumulation, and also inhibited cell proliferation in both normoxia and hypoxia unlike conventional chemotherapy. In human biopsies, a significant increase of SphK1 activity was observed in cancer compared with normal bones. In all sets of TMA samples (130 cases of osteosarcoma), immunohistochemical analysis showed the hypoxic marker GLUT-1, SphK1 and S1P1 were expressed in tumors. SphK1 correlated with the GLUT-1 suggesting that SphK1 is overexpressed and correlates with intratumoral hypoxia. No correlation was found between GLUT-1 or SphK1 and response to chemotherapy, but a statistical difference was found with increased S1P1 expression in patients with poor response in long bone osteosarcomas. Importantly, multivariate analyses showed that GLUT-1 was associated with an increased risk of death in flat bone, whereas SphK1 and S1P1 were associated with an increased risk of death in long bones.
Collapse
Affiliation(s)
- Anne Gomez-Brouchet
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
- Département d’Anatomie et Cytologie Pathologies, Institut Universitaire du Cancer de Toulouse–Oncopôle (IUCT-O), 31059 Toulouse, France
- Cancer Biobank, Institut Universitaire du Cancer de Toulouse–Oncopôle (IUCT-O), 31059 Toulouse, France;
- Correspondence: (A.G.-B.); (O.C.)
| | - Claire Illac
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
- Département d’Anatomie et Cytologie Pathologies, Institut Universitaire du Cancer de Toulouse–Oncopôle (IUCT-O), 31059 Toulouse, France
| | - Adeline Ledoux
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
| | - Pierre-Yves Fortin
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
| | - Sandra de Barros
- Service de Pharmacologie Clinique, Hôpitaux de Toulouse, 31300 Toulouse, France; (S.d.B.); (C.V.); (F.D.); (S.P.)
| | - Clémentine Vabre
- Service de Pharmacologie Clinique, Hôpitaux de Toulouse, 31300 Toulouse, France; (S.d.B.); (C.V.); (F.D.); (S.P.)
| | - Fabien Despas
- Service de Pharmacologie Clinique, Hôpitaux de Toulouse, 31300 Toulouse, France; (S.d.B.); (C.V.); (F.D.); (S.P.)
| | - Sophie Peries
- Service de Pharmacologie Clinique, Hôpitaux de Toulouse, 31300 Toulouse, France; (S.d.B.); (C.V.); (F.D.); (S.P.)
| | - Christelle Casaroli
- Cancer Biobank, Institut Universitaire du Cancer de Toulouse–Oncopôle (IUCT-O), 31059 Toulouse, France;
| | - Corinne Bouvier
- Department of Pathology, CHU la Timone, 13005 Marseille, France;
| | | | | | - Frédérique Larousserie
- Department of Pathology, AP-HP, Hôpital Cochin, Universiteé Paris Descartes, 75014 Paris, France;
| | - Louise Galmiche
- Centre Hospitalier Universitaire de Nantes Hôtel Dieu, 44000 Nantes, France;
| | - Franck Talmont
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
| | - Stuart Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia;
| | - Marie-Lise Maddelein
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
| | - Olivier Cuvillier
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
- Correspondence: (A.G.-B.); (O.C.)
| |
Collapse
|
11
|
Di Pompo G, Cortini M, Baldini N, Avnet S. Acid Microenvironment in Bone Sarcomas. Cancers (Basel) 2021; 13:cancers13153848. [PMID: 34359749 PMCID: PMC8345667 DOI: 10.3390/cancers13153848] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/24/2021] [Accepted: 07/28/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Although rare, malignant bone sarcomas have devastating clinical implications for the health and survival of young adults and children. To date, efforts to identify the molecular drivers and targets have focused on cancer cells or on the interplay between cancer cells and stromal cells in the tumour microenvironment. On the contrary, in the current literature, the role of the chemical-physical conditions of the tumour microenvironment that may be implicated in sarcoma aggressiveness and progression are poorly reported and discussed. Among these, extracellular acidosis is a well-recognized hallmark of bone sarcomas and promotes cancer growth and dissemination but data presented on this topic are fragmented. Hence, we intended to provide a general and comprehensive overview of the causes and implications of acidosis in bone sarcoma. Abstract In bone sarcomas, extracellular proton accumulation is an intrinsic driver of malignancy. Extracellular acidosis increases stemness, invasion, angiogenesis, metastasis, and resistance to therapy of cancer cells. It reprograms tumour-associated stroma into a protumour phenotype through the release of inflammatory cytokines. It affects bone homeostasis, as extracellular proton accumulation is perceived by acid-sensing ion channels located at the cell membrane of normal bone cells. In bone, acidosis results from the altered glycolytic metabolism of bone cancer cells and the resorption activity of tumour-induced osteoclasts that share the same ecosystem. Proton extrusion activity is mediated by extruders and transporters located at the cell membrane of normal and transformed cells, including vacuolar ATPase and carbonic anhydrase IX, or by the release of highly acidic lysosomes by exocytosis. To date, a number of investigations have focused on the effects of acidosis and its inhibition in bone sarcomas, including studies evaluating the use of photodynamic therapy. In this review, we will discuss the current status of all findings on extracellular acidosis in bone sarcomas, with a specific focus on the characteristics of the bone microenvironment and the acid-targeting therapeutic approaches that are currently being evaluated.
Collapse
Affiliation(s)
- Gemma Di Pompo
- Biomedical Science and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.D.P.); (M.C.); (N.B.)
| | - Margherita Cortini
- Biomedical Science and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.D.P.); (M.C.); (N.B.)
| | - Nicola Baldini
- Biomedical Science and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.D.P.); (M.C.); (N.B.)
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
- Correspondence:
| |
Collapse
|
12
|
Prudowsky ZD, Yustein JT. Recent Insights into Therapy Resistance in Osteosarcoma. Cancers (Basel) 2020; 13:E83. [PMID: 33396725 PMCID: PMC7795058 DOI: 10.3390/cancers13010083] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/22/2022] Open
Abstract
Osteosarcoma, the most common bone malignancy of childhood, has been a challenge to treat and cure. Standard chemotherapy regimens work well for many patients, but there remain minimal options for patients with progressive or resistant disease, as clinical trials over recent decades have failed to significantly improve survival. A better understanding of therapy resistance is necessary to improve current treatments and design new strategies for future treatment options. In this review, we discuss known mechanisms and recent scientific advancements regarding osteosarcoma and its patterns of resistance against chemotherapy, radiation, and other newly-introduced therapeutics.
Collapse
Affiliation(s)
- Zachary D. Prudowsky
- Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center, Houston, TX 77030, USA;
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jason T. Yustein
- Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center, Houston, TX 77030, USA;
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
13
|
Gola C, Iussich S, Noury S, Martano M, Gattino F, Morello E, Martignani E, Maniscalco L, Accornero P, Buracco P, Aresu L, De Maria R. Clinical significance and in vitro cellular regulation of hypoxia mimicry on HIF-1α and downstream genes in canine appendicular osteosarcoma. Vet J 2020; 264:105538. [PMID: 33012439 DOI: 10.1016/j.tvjl.2020.105538] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 12/11/2022]
Abstract
Cellular adaptation to a hypoxic microenvironment is essential for tumour progression and is largely mediated by HIF-1α and hypoxia-regulated factors, including CXCR4, VEGF-A and GLUT-1. In human osteosarcoma, hypoxia is associated with resistance to chemotherapy as well as with metastasis and poor survival, whereas little is known about its role in canine osteosarcoma (cOSA). This study aimed primarily to evaluate the prognostic value of several known hypoxic markers in cOSA. Immunohistochemical analysis for HIF-1α, CXCR4, VEGF-A and GLUT-1 was performed on 56 appendicular OSA samples; correlations with clinicopathological features and outcome was investigated. The second aim was to investigate the in vitro regulation of markers under chemically induced hypoxia (CoCl2). Two primary canine osteosarcoma cell lines were selected, and Western blotting, immunofluorescence and qRT-PCR were used to study protein and gene expression. Dogs with high-grade OSA (35.7%) were more susceptible to the development of metastases (P = 0.047) and showed high HIF-1α protein expression (P = 0.007). Moreover, HIF-1α overexpression (56%) was correlated with a shorter disease-free interval (DFI; P = 0.01), indicating that it is a reliable negative prognostic marker. The in vitro experiments identified an accumulation of HIF-1α in cOSA cells after chemically induced hypoxia, leading to a significant increase in GLUT-1 transcript (P = 0.02). HIF-1α might be a promising prognostic marker, highlighting opportunities for the use of therapeutic strategies targeting the hypoxic microenvironment in cOSA. These results reinforce the role of the dog as a comparative animal model since similar hypoxic mechanisms are reported in human osteosarcoma.
Collapse
Affiliation(s)
- C Gola
- Department of Veterinary Science, University of Turin, Grugliasco (TO), Italy
| | - S Iussich
- Department of Veterinary Science, University of Turin, Grugliasco (TO), Italy
| | - S Noury
- Hassan II Institute of Agronomy and Veterinary Medicine, Rabat, Morocco
| | - M Martano
- Department of Veterinary Science, University of Parma, Parma (PR)
| | - F Gattino
- Department of Veterinary Science, University of Turin, Grugliasco (TO), Italy
| | - E Morello
- Department of Veterinary Science, University of Turin, Grugliasco (TO), Italy
| | - E Martignani
- Department of Veterinary Science, University of Turin, Grugliasco (TO), Italy
| | - L Maniscalco
- Department of Veterinary Science, University of Turin, Grugliasco (TO), Italy
| | - P Accornero
- Department of Veterinary Science, University of Turin, Grugliasco (TO), Italy
| | - P Buracco
- Department of Veterinary Science, University of Turin, Grugliasco (TO), Italy
| | - L Aresu
- Department of Veterinary Science, University of Turin, Grugliasco (TO), Italy
| | - R De Maria
- Department of Veterinary Science, University of Turin, Grugliasco (TO), Italy.
| |
Collapse
|
14
|
Pierrevelcin M, Fuchs Q, Lhermitte B, Messé M, Guérin E, Weingertner N, Martin S, Lelong-Rebel I, Nazon C, Dontenwill M, Entz-Werlé N. Focus on Hypoxia-Related Pathways in Pediatric Osteosarcomas and Their Druggability. Cells 2020; 9:cells9091998. [PMID: 32878021 PMCID: PMC7564372 DOI: 10.3390/cells9091998] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/21/2020] [Accepted: 08/22/2020] [Indexed: 12/17/2022] Open
Abstract
Osteosarcoma is the most frequent primary bone tumor diagnosed during adolescence and young adulthood. It is associated with the worst outcomes in the case of poor response to chemotherapy and in metastatic disease. While no molecular biomarkers are clearly and currently associated with those worse situations, the study of pathways involved in the high level of tumor necrosis and in the immune/metabolic intra-tumor environment seems to be a way to understand these resistant and progressive osteosarcomas. In this review, we provide an updated overview of the role of hypoxia in osteosarcoma oncogenesis, progression and during treatment. We describe the role of normoxic/hypoxic environment in normal tissues, bones and osteosarcomas to understand their role and to estimate their druggability. We focus particularly on the role of intra-tumor hypoxia in osteosarcoma cell resistance to treatments and its impact in its endogenous immune component. Together, these previously published observations conduct us to present potential perspectives on the use of therapies targeting hypoxia pathways. These therapies could afford new treatment approaches in this bone cancer. Nevertheless, to study the osteosarcoma cell druggability, we now need specific in vitro models closely mimicking the tumor, its intra-tumor hypoxia and the immune microenvironment to more accurately predict treatment efficacy and be complementary to mouse models.
Collapse
Affiliation(s)
- Marina Pierrevelcin
- Laboratory of Bioimaging and Pathologies, UMR CNRS 7021, 67405 Illkirch, France; (M.P.); (Q.F.); (B.L.); (M.M.); (S.M.); (I.L.-R.); (M.D.)
| | - Quentin Fuchs
- Laboratory of Bioimaging and Pathologies, UMR CNRS 7021, 67405 Illkirch, France; (M.P.); (Q.F.); (B.L.); (M.M.); (S.M.); (I.L.-R.); (M.D.)
| | - Benoit Lhermitte
- Laboratory of Bioimaging and Pathologies, UMR CNRS 7021, 67405 Illkirch, France; (M.P.); (Q.F.); (B.L.); (M.M.); (S.M.); (I.L.-R.); (M.D.)
- Pathology Department, University Hospital of Strasbourg, 67098 Strasbourg, France;
| | - Melissa Messé
- Laboratory of Bioimaging and Pathologies, UMR CNRS 7021, 67405 Illkirch, France; (M.P.); (Q.F.); (B.L.); (M.M.); (S.M.); (I.L.-R.); (M.D.)
| | - Eric Guérin
- Oncobiology, Laboratory of Biochemistry and Molecular Biology, University Hospital of Strasbourg, 67098 Strasbourg, France;
| | - Noelle Weingertner
- Pathology Department, University Hospital of Strasbourg, 67098 Strasbourg, France;
| | - Sophie Martin
- Laboratory of Bioimaging and Pathologies, UMR CNRS 7021, 67405 Illkirch, France; (M.P.); (Q.F.); (B.L.); (M.M.); (S.M.); (I.L.-R.); (M.D.)
| | - Isabelle Lelong-Rebel
- Laboratory of Bioimaging and Pathologies, UMR CNRS 7021, 67405 Illkirch, France; (M.P.); (Q.F.); (B.L.); (M.M.); (S.M.); (I.L.-R.); (M.D.)
| | - Charlotte Nazon
- Pediatric Oncohematology Unit, University Hospital of Strasbourg, 67098 Strasbourg, France;
| | - Monique Dontenwill
- Laboratory of Bioimaging and Pathologies, UMR CNRS 7021, 67405 Illkirch, France; (M.P.); (Q.F.); (B.L.); (M.M.); (S.M.); (I.L.-R.); (M.D.)
| | - Natacha Entz-Werlé
- Laboratory of Bioimaging and Pathologies, UMR CNRS 7021, 67405 Illkirch, France; (M.P.); (Q.F.); (B.L.); (M.M.); (S.M.); (I.L.-R.); (M.D.)
- Pediatric Oncohematology Unit, University Hospital of Strasbourg, 67098 Strasbourg, France;
- Correspondence: ; Tel.: +33-3-8812-8396; Fax: +33-3-8812-8092
| |
Collapse
|
15
|
Molina ER, Chim LK, Barrios S, Ludwig JA, Mikos AG. Modeling the Tumor Microenvironment and Pathogenic Signaling in Bone Sarcoma. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:249-271. [PMID: 32057288 PMCID: PMC7310212 DOI: 10.1089/ten.teb.2019.0302] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/07/2020] [Indexed: 12/17/2022]
Abstract
Investigations of cancer biology and screening of potential therapeutics for efficacy and safety begin in the preclinical laboratory setting. A staple of most basic research in cancer involves the use of tissue culture plates, on which immortalized cell lines are grown in monolayers. However, this practice has been in use for over six decades and does not account for vital elements of the tumor microenvironment that are thought to aid in initiation, propagation, and ultimately, metastasis of cancer. Furthermore, information gleaned from these techniques does not always translate to animal models or, more crucially, clinical trials in cancer patients. Osteosarcoma (OS) and Ewing sarcoma (ES) are the most common primary tumors of bone, but outcomes for patients with metastatic or recurrent disease have stagnated in recent decades. The unique elements of the bone tumor microenvironment have been shown to play critical roles in the pathogenesis of these tumors and thus should be incorporated in the preclinical models of these diseases. In recent years, the field of tissue engineering has leveraged techniques used in designing scaffolds for regenerative medicine to engineer preclinical tumor models that incorporate spatiotemporal control of physical and biological elements. We herein review the clinical aspects of OS and ES, critical elements present in the sarcoma microenvironment, and engineering approaches to model the bone tumor microenvironment. Impact statement The current paradigm of cancer biology investigation and therapeutic testing relies heavily on monolayer, monoculture methods developed over half a century ago. However, these methods often lack essential hallmarks of the cancer microenvironment that contribute to tumor pathogenesis. Tissue engineers incorporate scaffolds, mechanical forces, cells, and bioactive signals into biological environments to drive cell phenotype. Investigators of bone sarcomas, aggressive tumors that often rob patients of decades of life, have begun to use tissue engineering techniques to devise in vitro models for these diseases. Their efforts highlight how critical elements of the cancer microenvironment directly affect tumor signaling and pathogenesis.
Collapse
Affiliation(s)
- Eric R. Molina
- Department of Bioengineering, Rice University, Houston, Texas
| | - Letitia K. Chim
- Department of Bioengineering, Rice University, Houston, Texas
| | - Sergio Barrios
- Department of Bioengineering, Rice University, Houston, Texas
| | - Joseph A. Ludwig
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | | |
Collapse
|
16
|
Watanabe A, Harimoto N, Yokobori T, Araki K, Kubo N, Igarashi T, Tsukagoshi M, Ishii N, Yamanaka T, Handa T, Oyama T, Higuchi T, Shirabe K. FDG-PET reflects tumor viability on SUV in colorectal cancer liver metastasis. Int J Clin Oncol 2020; 25:322-329. [PMID: 31612350 DOI: 10.1007/s10147-019-01557-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 09/28/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Liver resection is the most effective procedure for colorectal cancer liver metastasis (CRLM); however, early recurrence is an important problem that affects the postoperative prognoses of patients with CRLM. We previously suggested a therapeutic algorithm for CRLM using fluorodeoxyglucose-positron emission tomography (FDG-PET) and revealed the applicability of FDG-PET in predicting the prognosis after liver resection of CRLM. In this study, we assessed the correlation between FDG-PET and biological viability such as proliferation or metabolic activity. METHODS We retrospectively evaluated 61 patients who underwent hepatectomy for CRLM. We assessed hypoxia inducible factor-1α (HIF-1α), pyruvate kinase isozyme M2 (PKM2), glucose transporter 1 (GLUT1), and Ki-67 expression via immunohistochemistry and evaluated the correlation between standardized uptake value (SUV) and these factors. RESULTS High HIF-1α, PKM2, and GLUT1 expression were positively correlated with high SUV expression (P = 0.0444, 0.0296, and 0.0245, respectively). Ki-67 and SUV were also positively correlated (P = 0.00164). HIF-1α expression and PKM2 expression were significantly correlated (P = 0.0430), and PKM2 expression and GLUT1 expression were extremely significantly correlated (P < 0.0001). CONCLUSION SUV reflected tumor proliferation or metabolic factors in CRLM. FDG-PET could be a useful modality for assessing tumor viability and may provide useful information regarding the appropriate treatment strategy for CRLM.
Collapse
Affiliation(s)
- Akira Watanabe
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Norifumi Harimoto
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| | - Takehiko Yokobori
- Research Program for Omics-Based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Kenichiro Araki
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Norio Kubo
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takamichi Igarashi
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Mariko Tsukagoshi
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Norihiro Ishii
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takahiro Yamanaka
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Tadashi Handa
- Department of Diagnostic Pathology, Gunma University, Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Tetsunari Oyama
- Department of Diagnostic Pathology, Gunma University, Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Tetsuya Higuchi
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Ken Shirabe
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
17
|
Li HY, Zhou T, Lin W, Lin S, Zhong H. Association of hypoxia-inducible factor-1α (HIF1α) 1790G/A gene polymorphism with renal cell carcinoma and prostate cancer susceptibility: a meta-analysis. BMC MEDICAL GENETICS 2019; 20:141. [PMID: 31419966 PMCID: PMC6698016 DOI: 10.1186/s12881-019-0874-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND This meta-analysis was performed to evaluate the relationship between hypoxia-inducible factor-1α (HIF1α) 1790G/A gene polymorphism and the susceptibility to renal cell carcinoma (RCC) and prostate cancer (PCa). METHODS Association investigations were identified and included from the Embase, Cochrane Library and PubMed databases on March 1, 2018, and eligible investigations were analyzed by meta-analysis. Odds ratios (OR) were used to express the dichotomous data, and the 95% confidence intervals (CI) were also calculated. RESULTS In this meta-analysis, we found that the AA genotype of HIF1α 1790G/A was positively associated with the risk of RCC in overall populations, Caucasians, but not for Asians. G allele and GG genotype were not associated with the susceptibility of RCC in overall populations, Caucasians, and Asians. The G allele was negatively associated with PCa susceptibility in overall populations, Asians, but not for Caucasians. GG genotype was negatively associated with PCa susceptibility in Asians, but not for overall populations and Caucasians. HIF1α 1790G/A AA genotype was not associated with PCa susceptibility in overall populations of Caucasians or Asians. CONCLUSION AA genotype of HIF1α 1790G/A was positively associated with RCC risk in overall populations and Caucasians. Furthermore, the G allele was negatively associated with prostate cancer susceptibility in overall populations, Asians, and GG genotype was negatively associated with PCa susceptibility in Asians.
Collapse
Affiliation(s)
- Hong-Yan Li
- Department of Nephrology, Huadu District People’s Hospital of Guangzhou, Southern Medical University, Guangzhou, 510800 China
| | - Tianbiao Zhou
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, 515041, No 69 Dongxia Road, Shantou, China
| | - Wenshan Lin
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, 515041, No 69 Dongxia Road, Shantou, China
| | - Shujun Lin
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, 515041, No 69 Dongxia Road, Shantou, China
| | - Hongzhen Zhong
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, 515041, No 69 Dongxia Road, Shantou, China
| |
Collapse
|
18
|
Massimini M, De Maria R, Malatesta D, Romanucci M, D'Anselmo A, Della Salda L. Establishment of three-dimensional canine osteosarcoma cell lines showing vasculogenic mimicry and evaluation of biological properties after treatment with 17-AAG. Vet Comp Oncol 2019; 17:376-384. [PMID: 31006970 DOI: 10.1111/vco.12482] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/11/2019] [Accepted: 04/14/2019] [Indexed: 12/17/2022]
Abstract
Vasculogenic mimicry (VM) is an alternative type of blood perfusion characterized by formation of non-endothelial cell-lined microcirculatory channels and is responsible for aggressive tumour biology and increased tumour-related mortality. VM-correlated genes are associated with vascular endothelial grown factor receptor 1 (VEGFR1), and hypoxia-related (hypoxia inducible factor 1 α-HIF1α) signalling pathways, whose molecules are client proteins of Hsp90 (heat shock protein 90) and are potential therapeutic targets. This pilot study was aimed to investigate vasculogenic mimicry in a three-dimensional (3D) cell culture system of two aggressive canine osteosarcoma (OSA) cell lines (D22 and D17), and to evaluate the response of these cells to 17-AAG (17-N-allylamino-17-demethoxygeldanamycin) treatment in relation to tubular-like structure formation in vitro. Only D17 cell line formed hollow matrix channels in long-term 3D cultures and assumed endothelial morphology, with cells expressing both Hsp90 and VEGFR1, but lacking expression of endothelial marker CD31. 17-AAG treatment inhibited migration of D17 OSA cells, also decreasing VM markers in vitro and inducing a reduction of HIF1α transcript and protein in this cell line. Taken together, these preliminary data indicate that the biological effects of 17-AAG on D17 3D culture and on HIF1α regulation can provide interesting information to translate these findings from the basic research to clinical approach for the treatment of canine OSA as a model in comparative oncology.
Collapse
Affiliation(s)
| | - Raffaella De Maria
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | | | | | - Angela D'Anselmo
- Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | | |
Collapse
|
19
|
Smith CJ, Perfetti TA. In vitro cobalt-stimulated hypoxia-inducible factor-1 overexpression does not correlate with cancer risk from cobalt exposure in humans. TOXICOLOGY RESEARCH AND APPLICATION 2019. [DOI: 10.1177/2397847319850167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Carr J Smith
- Albemarle Corporation, Mobile, AL, USA
- Department of Nurse Anesthesia, Florida State University, Tallahassee, FL, USA
| | | |
Collapse
|
20
|
Hypoxia promotes osteosarcoma cell proliferation and migration through enhancing platelet-derived growth factor-BB/platelet-derived growth factor receptor-β axis. Biochem Biophys Res Commun 2019; 512:360-366. [PMID: 30894277 DOI: 10.1016/j.bbrc.2019.03.040] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 03/07/2019] [Indexed: 01/10/2023]
Abstract
Osteosarcoma is a primary malignant bone tumor, characterized by high therapeutic resistance and poor outcomes, due to unclear pathological mechanisms. It has been shown recently that the platelet-derived growth factor (PDGF)/platelet-derived growth factor receptor (PDGFR) pathway is closely associated with the pathogenesis of osteosarcoma. Hypoxia is a critical hallmark of tumor microenvironment that promotes the malignant phenotype in many solid tumors and a fundamental impediment to effective tumor therapy. In this study, we confirmed that hypoxia is an important feature of osteosarcoma, validated by the positive immunohistochemistry staining of hypoxia marker hypoxia-inducible factor-1α (HIF-1α) and carbonic anhydrase IX (CAIX) in osteosarcoma tissue samples. More importantly, we discovered that hypoxia could transcriptionally upregulate the expression of both PDGF-BB and PDGFR-β in osteosarcoma cells in vitro. Likewise, we also established that hypoxia-induced PDGF-BB is strongly related to the enhanced cell proliferation and migration, by activating AKT, ERK1/2, and STAT3 signaling pathways. Notably, when using an antibody to block the autocrine of PDGF-BB, cell proliferation and migration were partially aborted in hypoxia. Collectively, we demonstrated that the hypoxia-activated PDGF-BB/PDGFR-β axis plays essential roles in osteosarcoma progression. These findings may shed light on the molecular pathogenesis of osteosarcoma, and provide a novel strategy for osteosarcoma treatment by combinational targeting hypoxia and PDGF-BB/PDGFR signaling.
Collapse
|
21
|
Luo D, Ren H, Zhang W, Xian H, Lian K, Liu H. Clinicopathological and prognostic value of hypoxia-inducible factor-1α in patients with bone tumor: a systematic review and meta-analysis. J Orthop Surg Res 2019; 14:56. [PMID: 30782196 PMCID: PMC6381668 DOI: 10.1186/s13018-019-1101-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 02/14/2019] [Indexed: 11/29/2022] Open
Abstract
Background Recently, many studies have shown the role of hypoxia-inducible factor-1α (HIF-1α) expression in the outcome of bone tumor. However, the results remain inconclusive. It is necessary to carry out a meta-analysis of all the current available data to clarify the relationship between HIF-1α and survival or clinicopathological features of bone tumor. Methods PubMed, Cochrane Library, Web of Science, China National Knowledge Internet, and Wanfang databases were used to search the relationship between HIF-1α and bone tumor. Articles investigating clinicopathological and prognostic value of HIF-1α in bone tumor patients were enrolled in this meta-analysis. Overlapping articles, duplicate data, reviews, case reports, and letters without original data were excluded. The pooled risk ratios (RRs) and hazard ratios (HRs) were used to evaluate the clinicopathological and prognostic value of HIF-1α on bone tumor patients, respectively. Results A total of 28 studies including 1443 patients were included in this meta-analysis, which were involved in three different types of bone tumor including 3 chondrosarcomas, 2 giant cell tumors of bone, and 23 osteosarcomas. Our results showed that high expression levels of HIF-1α were associated with poorer OS (overall survival) (HR = 2.61, 95% CI 2.11–3.23, P < 0.001) and shorter DFS (disease-free survival) (HR = 2.02, 95% CI 1.41–2.89, P < 0.001) in bone tumor. In addition, this study also analyzed the role of HIF-1α expression in clinicopathological features, which were closely related with the severity of bone tumor, including differentiation, clinical stage, metastasis, and microvessel density. Our results indicated that HIF-1α overexpression was significantly associated with differentiation (RR = 1.56, 95% CI 1.00–2.43, P = 0.049), clinical stage (RR = 1.75, 95% CI 1.25–2.45, P = 0.001), metastasis (RR = 1.78, 95% CI 1.58–2.00, P < 0.001), and microvessel density (SMD = 2.34, 95% CI 1.35–3.34, P < 0.001) of bone tumor. Conclusions HIF-1α overexpression indicated an unfavorable factor for OS and DFS in bone tumor, suggesting that HIF-1α may serve as a potential prognostic marker for bone tumor. Electronic supplementary material The online version of this article (10.1186/s13018-019-1101-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Deqing Luo
- Department of Orthopaedic Surgery, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, 363000, Fujian Province, China
| | - Hongyue Ren
- Department of Pathology, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, 363000, Fujian Province, China
| | - Wenjiao Zhang
- Department of Orthopaedic Surgery, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, 363000, Fujian Province, China
| | - Hang Xian
- Department of Orthopaedic Surgery, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, 363000, Fujian Province, China
| | - Kejian Lian
- Department of Orthopaedic Surgery, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, 363000, Fujian Province, China
| | - Hui Liu
- Department of Orthopaedic Surgery, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, 363000, Fujian Province, China.
| |
Collapse
|
22
|
Yang Y, Wang J, Li H, Liu L, Yao M, Xiao T. Association between prognosis and SEMA4D/Plexin-B1 expression in various malignancies: A meta-analysis. Medicine (Baltimore) 2019; 98:e13298. [PMID: 30762724 PMCID: PMC6407964 DOI: 10.1097/md.0000000000013298] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION SEMA4D and its high affinity receptor Plexin-B1 showed a promising prognosis prediction for carcinoma patients in recent studies, we performed a meta-analysis to evaluate the prognostic role of them in various malignancies. METHODS A systematic literature search was performed in PubMed, Embase, Web of Science, and CNKI from inception till July 2017. Eligible studies were identified by different reviewers. Hazard ratios (HRs)/related ratios (RRs) and their corresponding 95% confidence intervals (CIs) were extracted to investigate the relevance between malignancies prognosis and SEMA4D/Plexin-B1. RESULTS Around 2638 patients from 14 studies were included in this meta-analysis. High expression of SEMA4D was significantly associated with overall survival (OS) and disease-free survival/progression-free survival/recurrence-free survival (DFS/PFS/RFS) in tumors (respectively, HRos = 2.05, 95%CI: 1.68-2.50, P < .001; HRdfs/pfs/rfs = 1.59, 95%CI = 1.27-1.98, P < .001). However, the relationship between SEMA4D expression and prognosis of breast cancer patients was failed to find (HR = 0.76, 95%CI = 0.32-1.82, P = .539). Plexin-B1 level showed a significant positive correlation both with OS and DFS of Caucasian breast cancer patients (respectively, HRos = 0.56, 95%CI: 0.39-0.79, P = .001; HRdfs = 0.68, 95%CI = 0.51-0.90, P = .008) CONCLUSIONS:: SEMA4D could be a prospective biomarker for prognostic prediction of various malignancies except breast cancer. For Caucasian breast cancer patients, SEMA4D's high affinity receptor Plexin-B1 showed a significant positive correlation with survival.
Collapse
Affiliation(s)
- Yibo Yang
- Department of Sport Surgery and Sport Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)
| | - Jing Wang
- Department of Sport Surgery and Sport Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)
| | - Hui Li
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Lihong Liu
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Maojin Yao
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA
| | - Tao Xiao
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| |
Collapse
|
23
|
Egners A, Rezaei M, Kuzmanov A, Poitz DM, Streichert D, Müller-Reichert T, Wielockx B, Breier G. PHD3 Acts as Tumor Suppressor in Mouse Osteosarcoma and Influences Tumor Vascularization via PDGF-C Signaling. Cancers (Basel) 2018; 10:cancers10120496. [PMID: 30563292 PMCID: PMC6316346 DOI: 10.3390/cancers10120496] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/30/2018] [Accepted: 12/03/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer cell proliferation and insufficient blood supply can lead to the development of hypoxic areas in the tumor tissue. The adaptation to the hypoxic environment is mediated by a transcriptional complex called hypoxia-inducible factor (HIF). HIF protein levels are tightly controlled by oxygen-dependent prolyl hydroxylase domain proteins (PHDs). However, the precise roles of these enzymes in tumor progression and their downstream signaling pathways are not fully characterized. Here, we study PHD3 function in murine experimental osteosarcoma. Unexpectedly, PHD3 silencing in LM8 cells affects neither HIF-1α protein levels, nor the expression of various HIF-1 target genes. Subcutaneous injection of PHD3-silenced tumor cells accelerated tumor progression and was accompanied by dramatic phenotypic changes in the tumor vasculature. Blood vessels in advanced PHD3-silenced tumors were enlarged whereas their density was greatly reduced. Examination of the molecular pathways underlying these alterations revealed that platelet-derived growth factor (PDGF)-C signaling is activated in the vasculature of PHD3-deficient tumors. Silencing of PDGF-C depleted tumor growth, increased vessel density and reduced vessel size. Our data show that PHD3 controls tumor growth and vessel architecture in LM8 osteosarcoma by regulating the PDGF-C pathway, and support the hypothesis that different members of the PHD family exert unique functions in tumors.
Collapse
Affiliation(s)
- Antje Egners
- Department of General, Visceral and Transplantation Surgery, RWTH University Hospital, 52074 Aachen, Germany.
- Department of Pathology, TU Dresden, 01307 Dresden, Germany.
| | - Maryam Rezaei
- Department of Biochemistry, University of Münster, 48149 Münster, Germany.
| | - Aleksandar Kuzmanov
- Department of Dermatology, University Hospital Zurich, CH-8952 Schlieren, Switzerland.
| | - David M Poitz
- Institute for Clinical Chemistry and Laboratory Medicine, TU Dresden, 01307 Dresden, Germany.
| | - Doreen Streichert
- Core Facility Cellular Imaging, Experimental Center, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany.
| | - Thomas Müller-Reichert
- Core Facility Cellular Imaging, Experimental Center, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany.
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, TU Dresden, 01307 Dresden, Germany.
| | - Georg Breier
- Division of Medical Biology, Department of Psychiatry and Psychotherapy, TU Dresden, 01307 Dresden, Germany.
| |
Collapse
|
24
|
Zhang T, Kastrenopoulou A, Larrouture Q, Athanasou NA, Knowles HJ. Angiopoietin-like 4 promotes osteosarcoma cell proliferation and migration and stimulates osteoclastogenesis. BMC Cancer 2018; 18:536. [PMID: 29739381 PMCID: PMC5941625 DOI: 10.1186/s12885-018-4468-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 04/30/2018] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Osteosarcoma is the most common primary bone cancer in children and young adults. It is highly aggressive and patients that present with metastasis have a poor prognosis. Angiopoietin-like 4 (ANGPTL4) drives the progression and metastasis of many solid tumours, but has not been described in osteosarcoma tissue. ANGPTL4 also enhances osteoclast activity, which is required for osteosarcoma growth in bone. We therefore investigated the expression and function of ANGPTL4 in human osteosarcoma tissue and cell lines. METHODS Expression of ANGPTL4 in osteosarcoma tissue microarrays was determined by immunohistochemistry. Hypoxic secretion of ANGPTL4 was tested by ELISA and Western blot. Regulation of ANGPTL4 by hypoxia-inducible factor (HIF) was investigated using isoform specific HIF siRNA (HIF-1α, HIF-2α). Effects of ANGPTL4 on cell proliferation, migration (scratch wound assay), colony formation and osteoblastogenesis were assessed using exogenous ANGPTL4 or cells stably transfected with ANGPTL4. Osteoclastogenic differentiation of CD14+ monocytes was assessed by staining for tartrate-resistant acid phosphatase (TRAP), bone resorption was assessed by lacunar resorption of dentine. RESULTS ANGPTL4 was immunohistochemically detectable in 76/109 cases. ANGPTL4 was induced by hypoxia in 6 osteosarcoma cell lines, under the control of the HIF-1α transcription factor. MG-63 cells transfected with an ANGPTL4 over-expression plasmid exhibited increased proliferation and migration capacity and promoted osteoclastogenesis and osteoclast-mediated bone resorption. Individually the full-length form of ANGPTL4 could increase MG-63 cell proliferation, whereas N-terminal ANGPTL4 mediated the other pro-tumourigenic phenotypes. CONCLUSIONS This study describes a role(s) for ANGPTL4 in osteosarcoma and identifies ANGPTL4 as a treatment target that could potentially reduce tumour progression, inhibit angiogenesis, reduce bone destruction and prevent metastatic events.
Collapse
Affiliation(s)
- T. Zhang
- Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford, OX3 7LD UK
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China
| | - A. Kastrenopoulou
- Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford, OX3 7LD UK
| | - Q. Larrouture
- Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford, OX3 7LD UK
| | - N. A. Athanasou
- Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford, OX3 7LD UK
| | - H. J. Knowles
- Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford, OX3 7LD UK
| |
Collapse
|
25
|
Cortini M, Avnet S, Baldini N. Mesenchymal stroma: Role in osteosarcoma progression. Cancer Lett 2017; 405:90-99. [PMID: 28774797 DOI: 10.1016/j.canlet.2017.07.024] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/19/2017] [Accepted: 07/23/2017] [Indexed: 12/21/2022]
Abstract
The initiation and progression of malignant tumors are supported by their microenvironment: cancer cells per se cannot explain growth and formation of the primary or metastasis, and a combination of proliferating tumor cells, cancer stem cells, immune cells mesenchymal stromal cells and/or cancer-associated fibroblasts all contribute to the tumor bulk. The interaction between these multiple players, under different microenvironmental conditions of biochemical and physical stimuli (i.e. oxygen tension, pH, matrix mechanics), regulates the production and biological activity of several soluble factors, extracellular matrix components, and extracellular vesicles that are needed for growth, maintenance, chemoresistance and metastatization of cancer. In osteosarcoma, a very aggressive cancer of young adults characterized by the extensive need for more effective therapies, this aspect has been only recently explored. In this view, we will discuss the role of stroma, with a particular focus on the mesenchymal stroma, contributing to osteosarcoma progression through inherent features for homing, neovascularization, paracrine cross-feeding, microvesicle secretion, and immune modulation, and also by responding to the changes of the microenvironment that are induced by tumor cells. The most recent advances in the molecular cues triggered by cytokines, soluble factors, and metabolites that are partially beginning to unravel the axis between stromal elements of mesenchymal origin and osteosarcoma cells, will be reviewed providing insights likely to be used for novel therapeutic approaches against sarcomas.
Collapse
Affiliation(s)
- Margherita Cortini
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sofia Avnet
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Nicola Baldini
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
26
|
Radiation-induced inflammatory cascade and its reverberating crosstalks as potential cause of post-radiotherapy second malignancies. Cancer Metastasis Rev 2017; 36:375-393. [DOI: 10.1007/s10555-017-9669-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
27
|
Knowles HJ. Multiple Roles of Angiopoietin-Like 4 in Osteolytic Disease. Front Endocrinol (Lausanne) 2017; 8:80. [PMID: 28458654 PMCID: PMC5394121 DOI: 10.3389/fendo.2017.00080] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 03/30/2017] [Indexed: 12/17/2022] Open
Abstract
Hypoxia and the hypoxia-inducible factor (HIF) transcription factor drive pathological bone loss in conditions including rheumatoid arthritis (RA), osteoarthritis, osteoporosis, primary bone tumours, and bone metastatic cancer. There is therefore considerable interest in determining the function(s) of HIF-induced genes in these pathologies. Angiopoietin-like 4 (ANGPTL4) is an adipose-derived, HIF-1α- and PPARγ-induced gene that was originally discovered as an endocrine and autocrine/paracrine regulator of lipid metabolism. Given the inverse relationship between bone adiposity and fracture risk, ANGPTL4 might be considered a good candidate for mediating the downstream effects of HIF-1α relevant to osteolytic disease. This review will consider the possible roles of ANGPTL4 in regulation of osteoclast-mediated bone resorption, cartilage degradation, angiogenesis, and inflammation, focusing on results obtained in the study of RA. Possible roles in other musculoskeletal pathologies will also be discussed. This will highlight ANGPTL4 as a regulator of multiple disease processes, which could represent a novel therapeutic target in osteolytic musculoskeletal disease.
Collapse
Affiliation(s)
- Helen J. Knowles
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- *Correspondence: Helen J. Knowles,
| |
Collapse
|