51
|
Ye H, Lu M, Tu C, Min L. Necroptosis in the sarcoma immune microenvironment: From biology to therapy. Int Immunopharmacol 2023; 122:110603. [PMID: 37467689 DOI: 10.1016/j.intimp.2023.110603] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/23/2023] [Accepted: 07/02/2023] [Indexed: 07/21/2023]
Abstract
Apoptosis resistance remains a major obstacle to treatment failure in sarcoma. Necroptosis is a caspase-independent programmed cell death, investigated as a novel strategy to eradicate anti-apoptotic tumor cells. The process is mediated by the receptor-interacting proteins kinase family and mixed lineage kinase domain-like proteins, which is morphologically similar to necrosis. Recent studies suggest that necroptosis in the tumor microenvironment has pro- or anti-tumor effects on immune response and cancer development. Necroptosis-related molecules display a remarkable value in prognosis prediction and therapeutic response evaluation of sarcoma. Furthermore, the induction of tumor necroptosis has been explored as a feasible therapeutic strategy against sarcoma and to synergize with immunotherapy. This review discusses the dual roles of necroptosis in the immune microenvironment and tumor progression, and explores the potential of necroptosis as a new target for sarcoma treatment.
Collapse
Affiliation(s)
- Huali Ye
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Minxun Lu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Chongqi Tu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Li Min
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
52
|
Garella R, Bernacchioni C, Chellini F, Tani A, Palmieri F, Parigi M, Guasti D, Cassioli E, Castellini G, Ricca V, Bani D, Sassoli C, Donati C, Squecco R. Adiponectin Modulates Smooth Muscle Cell Morpho-Functional Properties in Murine Gastric Fundus via Sphingosine Kinase 2 Activation. Life (Basel) 2023; 13:1812. [PMID: 37763216 PMCID: PMC10532860 DOI: 10.3390/life13091812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/31/2023] [Accepted: 08/15/2023] [Indexed: 09/29/2023] Open
Abstract
Adipokines are peptide hormones produced by the adipose tissue involved in several biological functions. Among adipokines, adiponectin (ADPN) has antidiabetic and anti-inflammatory properties. It can also modulate food intake at central and peripheral levels, acting on hypothalamus and facilitating gastric relaxation. ADPN exerts its action interacting with two distinct membrane receptors and triggering some well-defined signaling cascades. The ceramidase activity of ADPN receptor has been reported in many tissues: it converts ceramide into sphingosine. In turn, sphingosine kinase (SK) phosphorylates it into sphingosine-1 phosphate (S1P), a crucial mediator of many cellular processes including contractility. Using a multidisciplinary approach that combined biochemical, electrophysiological and morphological investigations, we explored for the first time the possible role of S1P metabolism in mediating ADPN effects on the murine gastric fundus muscle layer. By using a specific pharmacological inhibitor of SK2, we showed that ADPN affects smooth muscle cell membrane properties and contractile machinery via SK2 activation in gastric fundus, adding a piece of knowledge to the action mechanisms of this hormone. These findings help to identify ADPN and its receptors as new therapeutic targets or as possible prognostic markers for diseases with altered energy balance and for pathologies with fat mass content alterations.
Collapse
Affiliation(s)
- Rachele Garella
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (F.P.); (R.S.)
| | - Caterina Bernacchioni
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | - Flaminia Chellini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Alessia Tani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Francesco Palmieri
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (F.P.); (R.S.)
| | - Martina Parigi
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Daniele Guasti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Emanuele Cassioli
- Psychiatry Unit, Department of Health Sciences, University of Florence, 50134 Florence, Italy; (E.C.); (G.C.); (V.R.)
| | - Giovanni Castellini
- Psychiatry Unit, Department of Health Sciences, University of Florence, 50134 Florence, Italy; (E.C.); (G.C.); (V.R.)
| | - Valdo Ricca
- Psychiatry Unit, Department of Health Sciences, University of Florence, 50134 Florence, Italy; (E.C.); (G.C.); (V.R.)
| | - Daniele Bani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Chiara Sassoli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | - Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (F.P.); (R.S.)
| |
Collapse
|
53
|
Mannheimer JD, Tawa G, Gerhold D, Braisted J, Sayers CM, McEachron TA, Meltzer P, Mazcko C, Beck JA, LeBlanc AK. Transcriptional profiling of canine osteosarcoma identifies prognostic gene expression signatures with translational value for humans. Commun Biol 2023; 6:856. [PMID: 37591946 PMCID: PMC10435536 DOI: 10.1038/s42003-023-05208-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 08/03/2023] [Indexed: 08/19/2023] Open
Abstract
Canine osteosarcoma is increasingly recognized as an informative model for human osteosarcoma. Here we show in one of the largest clinically annotated canine osteosarcoma transcriptional datasets that two previously reported, as well as de novo gene signatures devised through single sample Gene Set Enrichment Analysis (ssGSEA), have prognostic utility in both human and canine patients. Shared molecular pathway alterations are seen in immune cell signaling and activation including TH1 and TH2 signaling, interferon signaling, and inflammatory responses. Virtual cell sorting to estimate immune cell populations within canine and human tumors showed similar trends, predominantly for macrophages and CD8+ T cells. Immunohistochemical staining verified the increased presence of immune cells in tumors exhibiting immune gene enrichment. Collectively these findings further validate naturally occurring osteosarcoma of the pet dog as a translationally relevant patient model for humans and improve our understanding of the immunologic and genomic landscape of the disease in both species.
Collapse
Affiliation(s)
- Joshua D Mannheimer
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gregory Tawa
- Division of Preclinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - David Gerhold
- Division of Preclinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - John Braisted
- Division of Preclinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Carly M Sayers
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Troy A McEachron
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paul Meltzer
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jessica A Beck
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
54
|
Jackson KJ, Sullivan CD, Zimel MN, Wustrack RL. Surgical Site Infection Is Not Associated with 1-Year Progression-Free Survival After Endoprosthetic Reconstruction for Lower-Extremity Osteosarcoma: A Secondary Analysis of PARITY Study Data. J Bone Joint Surg Am 2023; 105:49-56. [PMID: 37466580 DOI: 10.2106/jbjs.22.01077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
BACKGROUND Although there is evidence suggesting that postoperative infection confers a survival benefit in osteosarcoma treated with resection and endoprosthetic reconstruction, there have been no prospective studies to date to support these findings. This secondary analysis of Prophylactic Antibiotic Regimens in Tumor Surgery (PARITY) study data examines the relationship between surgical site infection (SSI) and disease progression within 12 months after limb salvage surgery. METHODS The PARITY trial was an international, multicenter, prospective randomized controlled trial of 604 patients who underwent resection of a lower-extremity bone tumor and endoprosthetic reconstruction. Our primary outcome was progression-free survival (PFS) at 1 year following surgery among the patients with osteosarcoma. Subgroup analyses by disease stage at presentation and infection severity were also performed. Cox proportional hazard models were employed to examine the association between clinical and tumor characteristics, SSI, and PFS. Kaplan-Meier analysis was used to determine the effect of SSI on PFS. RESULTS The 274 PARITY patients with osteosarcoma were included in this secondary analysis. Thirty-two (11.7%) of the patients presented with metastasis at baseline; 53 (19.3%) of the patients developed an SSI. There was no difference in 1-year PFS between patients with and without SSI. There was no decreased risk of disease progression at 1 year in patients with localized disease at baseline who developed an SSI (hazard ratio [HR] = 1.21; 95% confidence interval [CI] = 0.64 to 2.28). Infection was associated with increased disease progression at 1 year in patients with baseline metastases (HR = 4.26; 95% CI = 1.11 to 16.3). CONCLUSIONS No positive association was detected between postoperative infection and PFS at 1 year following surgery in this secondary analysis of prospective data. However, this analysis suggests infection could be a risk factor for early disease progression in patients with baseline metastases, and future investigations may better elucidate the association between disease burden and the host immune response to advance immunotherapeutic strategies for osteosarcoma. LEVEL OF EVIDENCE Prognostic Level II. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Kristopher J Jackson
- Department of Medicine, Division of Prevention Science, University of California San Francisco, San Francisco, California
| | - Camille D Sullivan
- Orthopaedic Surgery, University of California San Francisco, San Francisco, California
| | - Melissa N Zimel
- Orthopaedic Surgery, University of California San Francisco, San Francisco, California
| | - Rosanna L Wustrack
- Orthopaedic Surgery, University of California San Francisco, San Francisco, California
| |
Collapse
|
55
|
Han X, Yin M, Gong C, Zhang C, Zhu G, Hu M, Tan K, Jiang L, Wang G, Li L. A1BG-AS1 promotes the biological functions of osteosarcoma cells via regulating the microRNA-148a-3p/USP22 axis and stabilizing the expression of SIRT1 through deubiquitinase function. Expert Opin Ther Targets 2023; 27:1017-1029. [PMID: 37747800 DOI: 10.1080/14728222.2023.2263908] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 09/24/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND The study aims to explore the role of A1BG antisense RNA 1 (A1BG-AS1), microRNA (miR)-148a-3p and ubiquitin-specific protease 22 (USP22) on osteosarcoma (OS) cell growth. RESEARCH DESIGN & METHODS A1BG-AS1, miR-148a-3p, USP22, and silent information regulator 2 homolog 1 (SIRT1) levels in OS tissues and cells were determined. The effects of A1BG-AS1, miR-148a-3p, and USP22 on the biological functions of OS cells were examined by functional assays. In vivo assay was conducted to observe the effect of A1BG-AS1 on OS growth in vitro. The relationship of A1BG-AS1, miR-148a-3p, and USP22 was analyzed by bioinformatics analysis, RNA-fluorescence in situ hybridization, luciferase activity, and RNA binding protein immunoprecipitation assays. The relation between USP22 and SIRT1 was evaluated by immunoprecipitation. RESULTS A1BG-AS1 and USP22 were highly expressed, and miR-148a-3p was lowly expressed in OS tissues and cells. Down-regulation of A1BG-AS1 and USP22 or up-regulation of miR-148a-3p impaired the malignant behaviors of OS cells. A1BG-AS1 sponged miR-148a-3p, and miR-148a-3p targeted USP22, thereby inhibiting USP22 expression. Up-regulating USP22 reversed the A1BG-AS1 suppression-induced phenotypic inhibition of OS cells. USP22 affected the biological functions of OS cells by deubiquitinating SIRT1. CONCLUSION A1BG-AS1 facilitates the biological functions of OS cells via mediating the miR-148a-3p/USP22 axis.
Collapse
Affiliation(s)
- Xiuxin Han
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Mengfan Yin
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
- Department of Orthopedic Surgery, Tianjin Fifth Central Hospital, Tianjin, China
| | - Chen Gong
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - Chao Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Genbao Zhu
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - Mengxue Hu
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - Kemeng Tan
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - La Jiang
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - Guowen Wang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Lili Li
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| |
Collapse
|
56
|
Pires SF, Barros JSD, Costa SSD, Carmo GBD, Scliar MDO, Lengert AVH, Boldrini É, Silva SRMD, Vidal DO, Maschietto M, Krepischi ACV. Analysis of the Mutational Landscape of Osteosarcomas Identifies Genes Related to Metastasis and Prognosis and Disrupted Biological Pathways of Immune Response and Bone Development. Int J Mol Sci 2023; 24:10463. [PMID: 37445641 DOI: 10.3390/ijms241310463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 07/15/2023] Open
Abstract
Osteosarcoma (OS) is the most prevalent type of bone tumor, but slow progress has been achieved in disentangling the full set of genomic events involved in its initiation and progression. We assessed by NGS the mutational spectrum of 28 primary OSs from Brazilian patients, and identified 445 potentially deleterious SNVs/indels and 1176 copy number alterations (CNAs). TP53 was the most recurrently mutated gene, with an overall rate of ~60%, considering SNVs/indels and CNAs. The most frequent CNAs (~60%) were gains at 1q21.2q21.3, 6p21.1, and 8q13.3q24.22, and losses at 10q26 and 13q14.3q21.1. Seven cases presented CNA patterns reminiscent of complex events (chromothripsis and chromoanasynthesis). Putative RB1 and TP53 germline variants were found in five samples associated with metastasis at diagnosis along with complex genomic patterns of CNAs. PTPRQ, KNL1, ZFHX4, and DMD alterations were prevalent in metastatic or deceased patients, being potentially indicative of poor prognosis. TNFRSF11B, involved in skeletal system development and maintenance, emerged as a candidate for osteosarcomagenesis due to its biological function and a high frequency of copy number gains. A protein-protein network enrichment highlighted biological pathways involved in immunity and bone development. Our findings reinforced the high genomic OS instability and heterogeneity, and led to the identification of novel disrupted genes deserving further evaluation as biomarkers due to their association with poor outcomes.
Collapse
Affiliation(s)
- Sara Ferreira Pires
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo 05508-090, Brazil
| | - Juliana Sobral de Barros
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo 05508-090, Brazil
| | - Silvia Souza da Costa
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo 05508-090, Brazil
| | - Gabriel Bandeira do Carmo
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo 05508-090, Brazil
| | - Marília de Oliveira Scliar
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo 05508-090, Brazil
| | | | - Érica Boldrini
- Barretos Children's Cancer Hospital, Barretos 14784-400, Brazil
| | | | - Daniel Onofre Vidal
- Molecular Oncology Research Center (CPOM), Barretos Cancer Hospital, Barretos 14784-384, Brazil
| | - Mariana Maschietto
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-884, Brazil
| | - Ana Cristina Victorino Krepischi
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo 05508-090, Brazil
| |
Collapse
|
57
|
Gong D, Zhao Q, Liu J, Zhao S, Yi C, Lv J, Yu H, Bian E, Tian D. Identification of a novel MYC target gene set signature for predicting the prognosis of osteosarcoma patients. Front Oncol 2023; 13:1169430. [PMID: 37342196 PMCID: PMC10277635 DOI: 10.3389/fonc.2023.1169430] [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: 02/19/2023] [Accepted: 05/04/2023] [Indexed: 06/22/2023] Open
Abstract
Osteosarcoma is a primary malignant tumor found mainly in teenagers and young adults. Patients have very little long-term survival. MYC controls tumor initiation and progression by regulating the expression of its target genes; thus, constructing a risk signature of osteosarcoma MYC target gene set will benefit the evaluation of both treatment and prognosis. In this paper, we used GEO data to download the ChIP-seq data of MYC to obtain the MYC target gene. Then, a risk signature consisting of 10 MYC target genes was developed using Cox regression analysis. The signature indicates that patients in the high-risk group performed poorly. After that, we verified it in the GSE21257 dataset. In addition, the difference in tumor immune function among the low- and high-risk populations was compared by single sample gene enrichment analysis. Immunotherapy and prediction of response to the anticancer drug have shown that the risk signature of the MYC target gene set was positively correlated with immune checkpoint response and drug sensitivity. Functional analysis has demonstrated that these genes are enriched in malignant tumors. Finally, STX10 was selected for functional experimentation. STX10 silence has limited osteosarcoma cell migration, invasion, and proliferation. Therefore, these findings indicated that the MYC target gene set risk signature could be used as a potential therapeutic target and prognostic indicator in patients with osteosarcoma.
Collapse
Affiliation(s)
- Deliang Gong
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qingzhong Zhao
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jun Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shibing Zhao
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chengfeng Yi
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jianwei Lv
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hang Yu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Erbao Bian
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dasheng Tian
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
58
|
Tang Z, Feng H, Shu L, Guo M, Qi B, Pu L, Shi H, Ren J, Li C. Identification of two novel lipid metabolism-related long non-coding RNAs (SNHG17 and LINC00837) as potential signatures for osteosarcoma prognosis and precise treatment. BMC Med Genomics 2023; 16:115. [PMID: 37231440 DOI: 10.1186/s12920-023-01553-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023] Open
Abstract
OBJECTIVE Dysregulated lipid metabolism enhances the development and advancement of many cancers, including osteosarcoma (OS); however, the underlying mechanisms are still largely unknown. Therefore, this investigation aimed to elucidate novel potential lipid metabolism-related long non-coding RNAs (lncRNAs) that regulate OS development and provide novel signatures for its prognosis and precise treatment. MATERIALS AND METHODS The GEO datasets (GSE12865 and GSE16091) were downloaded and analyzed using R software packages. Immunohistochemistry (IHC) was used to evaluate protein levels in OS tissues while real-time qPCR was used to measure lncRNA levels, and MTT assays were used to assess OS cell viability. RESULTS Two lipid metabolism-associated lncRNAs (LM-lncRNAs), small nucleolar RNA host gene 17 (SNHG17) and LINC00837, were identified as efficient and independent prognostic indicators for OS. In addition, further experiments confirmed that SNHG17 and LINC00837 were significantly elevated in OS tissues and cells than para-cancerous counterparts. Knockdown of SNHG17 and LINC00837 synergistically suppressed the viability of OS cells, whereas overexpression of the two lncRNAs promoted OS cell proliferation. Moreover, bioinformatics analysis was conducted to construct six novel SNHG17-microRNA-mRNA competing endogenous RNA (ceRNA) networks, and three lipid metabolism-associated genes (MIF, VDAC2, and CSNK2A2) were found to be abnormally upregulated in OS tissues, suggesting that they were potential effector genes of SNHG17. CONCLUSION In summary, SNHG17 and LINC00837 were found to promote OS cell malignancy, suggesting their use as ideal biomarkers for OS prognosis and treatment.
Collapse
Affiliation(s)
- Zhifang Tang
- Clinical Medical College of Dali University, Dali, Yunnan, 671000, China
| | - Hanzhen Feng
- Clinical Medical College of Dali University, Dali, Yunnan, 671000, China
| | - Longjun Shu
- Department of Orthopedics, The First People's Hospital of Dali City, Yunnan, 671000, Dali, China
| | - Minzheng Guo
- Department of Orthopedics, Kunming Medical University, Kunming, Yunnan, China
| | - Baochuang Qi
- Department of Orthopedics, Kunming Medical University, Kunming, Yunnan, China
| | - Luqiao Pu
- Department of Orthopedics, The 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Hongxin Shi
- Clinical Medical College of Dali University, Dali, Yunnan, 671000, China
| | - Junxiao Ren
- Department of Orthopedics, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Chuan Li
- Department of Orthopedics, The 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China.
| |
Collapse
|
59
|
Reale A, Krutzke L, Cadamuro M, Vitiello A, von Einem J, Kochanek S, Palù G, Parolin C, Calistri A. Human Monocytes Are Suitable Carriers for the Delivery of Oncolytic Herpes Simplex Virus Type 1 In Vitro and in a Chicken Embryo Chorioallantoic Membrane Model of Cancer. Int J Mol Sci 2023; 24:ijms24119255. [PMID: 37298206 DOI: 10.3390/ijms24119255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Oncolytic viruses (OVs) are promising therapeutics for tumors with a poor prognosis. An OV based on herpes simplex virus type 1 (oHSV-1), talimogene laherparepvec (T-VEC), has been recently approved by the Food and Drug Administration (FDA) and by the European Medicines Agency (EMA) for the treatment of unresectable melanoma. T-VEC, like most OVs, is administered via intratumoral injection, underlining the unresolved problem of the systemic delivery of the oncolytic agent for the treatment of metastases and deep-seated tumors. To address this drawback, cells with a tropism for tumors can be loaded ex vivo with OVs and used as carriers for systemic oncolytic virotherapy. Here, we evaluated human monocytes as carrier cells for a prototype oHSV-1 with a similar genetic backbone as T-VEC. Many tumors specifically recruit monocytes from the bloodstream, and autologous monocytes can be obtained from peripheral blood. We demonstrate here that oHSV-1-loaded primary human monocytes migrated in vitro towards epithelial cancer cells of different origin. Moreover, human monocytic leukemia cells selectively delivered oHSV-1 to human head-and-neck xenograft tumors grown on the chorioallantoic membrane (CAM) of fertilized chicken eggs after intravascular injection. Thus, our work shows that monocytes are promising carriers for the delivery of oHSV-1s in vivo, deserving further investigation in animal models.
Collapse
Affiliation(s)
- Alberto Reale
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Lea Krutzke
- Department of Gene Therapy, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Adriana Vitiello
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Jens von Einem
- Institute of Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Stefan Kochanek
- Department of Gene Therapy, Ulm University Medical Center, 89081 Ulm, Germany
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Cristina Parolin
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| |
Collapse
|
60
|
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
|
61
|
Júnior RFDA, Lira GA, Schomann T, Cavalcante RS, Vilar NF, de Paula RCM, Gomes RF, Chung CK, Jorquera-Cordero C, Vepris O, Chan AB, Cruz LJ. Retinoic acid-loaded PLGA nanocarriers targeting cell cholesterol potentialize the antitumour effect of PD-L1 antibody by preventing epithelial-mesenchymal transition mediated by M2-TAM in colorectal cancer. Transl Oncol 2023; 31:101647. [PMID: 36857852 PMCID: PMC9989692 DOI: 10.1016/j.tranon.2023.101647] [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: 08/06/2022] [Revised: 11/30/2022] [Accepted: 02/20/2023] [Indexed: 03/03/2023] Open
Abstract
Tumour-associated macrophages (TAMs) often promote cancer progression through immunosuppression in the tumour microenvironment (TME). However, the signalling pathways crosstalk responsible for this mechanism remain unclear. The aim of our study was to investigate whether the interaction between TAMs and colorectal cancer cells could be down-regulated by nanoparticles (NPs) loaded with retinoic acid (RA) and coated with cholesterol (CHO), in combination with an anti-PD-L1 immune checkpoint inhibitor. Tumours were evaluated by qRT-PCR and immunohistochemistry from allographic tumour growth model. In addition, human tumours were evaluated by Tissue Microarray (TMA) and immunohistochemistry. Complementary analysis of epithelial-mesenchymal transition, cell migration, and macrophage polarisation were evaluated in vitro. We showed that the IL-10R/IL-10 axis is involved in overstimulation of the STAT3 pathway as well as downregulation of the NF-κB signalling pathway, which supports a loop of immunosuppressive cytokines that induces the M2-TAM phenotype. Furthermore, our combined findings suggest that the upregulation of STAT3/NF-κB pathways crosstalk mediated by immunosuppressive cytokines, such as IL-10/PD-L1/TGF-β, via M2-TAMs in the TME, leads to immunosuppression and epithelial-mesenchymal-transition of the colorectal cancer for stimulating Vimentin, CXCL12 and CD163 in the primary tumours. Importantly, NPs holding RA and coated with CHO in combination with anti-PD-L1 were more efficient in blocking this signalling pathway. These results contribute to our understanding of the immunological mechanisms, especially the re-educating of TAMs, and provide a novel management strategy for aggressive colorectal cancers using anti-PD-L1-conjugated nanocarriers.
Collapse
Affiliation(s)
- Raimundo Fernandes de Araújo Júnior
- Cancer and Inflammation Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte Natal, RN 59072-970, Brazil; Post-Graduation Programme in Structural and Functional Biology, Federal University of Rio Grande do Norte, Natal, RN 59072-970, Brazil; Post-Graduation Programme in Health Science, Federal University of Rio Grande do Norte, Natal, RN 59072-970, Brazil; Percuros B.V., Leiden, CL 2333, the Netherlands; Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, ZA 2333, the Netherlands.
| | - George A Lira
- Cancer and Inflammation Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte Natal, RN 59072-970, Brazil; Post-Graduation Programme in Health Science, Federal University of Rio Grande do Norte, Natal, RN 59072-970, Brazil; Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, ZA 2333, the Netherlands; League Against Cancer from Rio Grande do Norte, Advanced Oncology Center, Natal 59075-740, Brazil
| | - Timo Schomann
- Percuros B.V., Leiden, CL 2333, the Netherlands; Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, ZA 2333, the Netherlands
| | - Rômulo S Cavalcante
- Cancer and Inflammation Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte Natal, RN 59072-970, Brazil; Post-Graduation Programme in Health Science, Federal University of Rio Grande do Norte, Natal, RN 59072-970, Brazil
| | - Natalia Feitosa Vilar
- Cancer and Inflammation Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte Natal, RN 59072-970, Brazil
| | | | - Raelle Ferreira Gomes
- Post-Graduation Programme in Chemistry, Federal University of Ceará, Fortaleza, CE 60440-900, Brazil
| | - Chih Kit Chung
- Percuros B.V., Leiden, CL 2333, the Netherlands; Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, ZA 2333, the Netherlands; JeNaCell GmbH, Winzerlaer Straße 2, Jena 07745, Germany
| | - Carla Jorquera-Cordero
- Percuros B.V., Leiden, CL 2333, the Netherlands; Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, CX 3584, the Netherlands
| | - Olena Vepris
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, ZA 2333, the Netherlands
| | - Alan B Chan
- Percuros B.V., Leiden, CL 2333, the Netherlands
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, ZA 2333, the Netherlands
| |
Collapse
|
62
|
Thomas DD, Lacinski RA, Lindsey BA. Single-cell RNA-seq reveals intratumoral heterogeneity in osteosarcoma patients: A review. J Bone Oncol 2023; 39:100475. [PMID: 37034356 PMCID: PMC10074210 DOI: 10.1016/j.jbo.2023.100475] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/08/2023] [Accepted: 03/15/2023] [Indexed: 03/22/2023] Open
Abstract
While primary bone malignancies make up just 0.2% of all cancers, osteosarcoma (OS) is the third most common cancer in adolescents. Due to its highly complex and heterogeneous tumor microenvironment (TME), OS has proven difficult to treat. There has been little to no improvement in therapy for this disease over the last 40 years. Even the recent success of immunotherapies in other blood-borne and solid malignancies has not translated to OS. With frequent recurrence and lung metastases continuing to pose a challenge in the clinic, recent advancements in molecular profiling, such as single-cell RNA sequencing (scRNA-seq), have proven useful in identifying novel biomarkers of OS tumors while providing new insight into this TME that could potentially lead to new therapeutic options. This review combines the analyses of over 150,000 cells from 18 lesions ranging from primary, recurrent, and metastatic OS lesions, revealing distinct cellular populations and gene signatures that exist between them. Here, we detail these previous findings and ultimately convey the intratumoral heterogeneity that exists within OS tumor specimens.
Collapse
Affiliation(s)
- Dylan D. Thomas
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Ryan A. Lacinski
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Brock A. Lindsey
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, United States
- Cancer Institute, West Virginia University School of Medicine, Morgantown, WV, United States
| |
Collapse
|
63
|
Yin P, Xu J, Sun X, Liu T, Chen L, Hong N. Intravoxel incoherent motion and dynamic contrast-enhanced magnetic resonance imaging for neoadjuvant chemotherapy response evaluation in patients with osteosarcoma. Eur J Radiol 2023; 162:110790. [PMID: 36963332 DOI: 10.1016/j.ejrad.2023.110790] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/26/2023]
Abstract
OBJECTIVES This study aims to explore the role of quantitative intravoxel incoherent motion (IVIM) and dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) parameters in characterizing changes in osteosarcoma (OS) patients receiving neoadjuvant chemotherapy (NACT). MATERIAL AND METHODS Twenty-seven patients with histologically proven OS were examined prospectively and divided into good-response group (n = 14) and poor-response group (n = 13). IVIM and DCE-MRI sequences were performed at baseline (pre-NACT) and after three cycles of NACT (post-NACT). Apparent diffusion coefficient (ADC) and IVIM bi-exponential model parameters, including diffusion coefficient (D-Bi), perfusion coefficient (D*-Bi), and perfusion fraction (f-Bi), were evaluated. DCE-MRI parameters, including quantitative parameters (volume transfer constant [Ktrans], elimination rate constant [Kep], and extravascular extracellular space volume ratio [Ve]) and semi-quantitative parameters (initial area under the gadolinium curve [IAUGC] and contrast enhancement rate [CER]), were also measured. RESULTS D-Bi, D*-Bi, and f-Bi post-NACT and ΔD-Bi were statistically different between the good- and poor-response groups (Z1 = - 3.348, Z2 = - 2.572, Z3 = - 2.378, t = 2.235, P < 0.05). ADC, f-Bi, Ktrans, IAUGC, Kep, and CER post-NACT were statistically different from those at pre-NACT (P < 0.05). The receiver operating characteristic curve showed that f-Bi post-NACT had the best performance among all parameters, with area under the curve of 0.769, sensitivity of 1, and specificity of 0.538. The correlation analysis showed that the efficacy of NACT was negatively correlated with D-Bi, D*-Bi post-NACT, and ΔD-Bi (r1 = - 0.530, r2 = - 0.411, r3 = - 0.434, P1 = 0.008, P2 = 0.046, P3 = 0.034) and significantly positively correlated with f-Bi post-NACT (r = 0.482, P = 0.017). CONCLUSIONS The IVIM quantitative parameters D-Bi, D*-Bi, and f-Bi post-NACT and ΔD-Bi could be used as noninvasive imaging biomarkers for early response assessment of NACT in OS.
Collapse
Affiliation(s)
- Ping Yin
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen Nandajie, Xicheng District, Beijing 100044, PR China
| | - Jie Xu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing 100044, PR China
| | - Xin Sun
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing 100044, PR China
| | - Tao Liu
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen Nandajie, Xicheng District, Beijing 100044, PR China
| | - Lei Chen
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen Nandajie, Xicheng District, Beijing 100044, PR China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, 11 Xizhimen Nandajie, Xicheng District, Beijing 100044, PR China.
| |
Collapse
|
64
|
The mechanism of VCP-mediated metastasis of osteosarcoma based on cell autophagy and the EMT pathway. Clin Transl Oncol 2023; 25:653-661. [PMID: 36284060 DOI: 10.1007/s12094-022-02972-y] [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: 07/10/2022] [Accepted: 09/27/2022] [Indexed: 10/31/2022]
Abstract
OBJECTIVE Study of the molecular mechanisms of metastasis is still the research focus for osteosarcoma (OS) prevention. This study investigates the mechanism of valosin-containing protein (VCP) promoting OS metastasis in vitro through autophagy and epithelial-mesenchymal transition (EMT). METHODS Different cell lines of osteosarcoma (143B and MG63) were adopted in this study. The level of VCP expression in osteosarcoma cells was changed, and the level of autophagy and the progression of the epithelial-mesenchymal transition (EMT) were observed. Then autophagy and EMT in OS cells were changed artificially, and proliferation and migration ability were observed. RESULTS The expression of LC3II/I was decreased, but the insolubilized P62 protein expression was increased in the VCP inhibiting group and the autophagy inhibitor treatment group. Simultaneously, E-cadherin protein expression increased while N-cadherin protein expression decreased in the VCP inhibiting group but increased in the TGF-β1 treatment group. In addition, suppressing VCP can cause a decrease in Transforming Growth Factor β1 (TGF-β1), smad2, smad3, phosphorylated smad2 (p-smad2), and phosphorylated smad3 (p-smad3). Autophagy inhibitors and agonists have no significant effect on the migration and invasion of OS cells but can significantly affect the ability of cells to resist anoikis. EMT inhibitors and agonists have a proportional effect on the migration and invasion of OS cells. CONCLUSION VCP is likely to promote the migration and invasion of OS cells by inducing EMT, possibly via TGF-β1/smad2/3 signaling pathway. In this process, VCP-mediated autophagy may contribute to successful distant metastasis of tumor cells indirectly.
Collapse
|
65
|
Li S, Que Y, Yang R, He P, Xu S, Hu Y. Construction of Osteosarcoma Diagnosis Model by Random Forest and Artificial Neural Network. J Pers Med 2023; 13:jpm13030447. [PMID: 36983630 PMCID: PMC10056981 DOI: 10.3390/jpm13030447] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/19/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Osteosarcoma accounts for 28% of primary bone malignancies in adults and up to 56% in children and adolescents (<20 years). However, early diagnosis and treatment are still inadequate, and new improvements are still needed. Missed diagnoses exist due to fewer traditional diagnostic methods, and clinical symptoms are often already present before diagnosis. This study aimed to develop novel and efficient predictive models for the diagnosis of osteosarcoma and to identify potential targets for exploring osteosarcoma markers. First, osteosarcoma and normal tissue expression microarray datasets were downloaded from the Gene Expression Omnibus (GEO). Then we screened the differentially expressed genes (DEGs) in the osteosarcoma and normal groups in the training group. Next, in order to explore the biologically relevant role of DEGs, Metascape and enrichment analyses were also performed on DEGs. The “randomForest” and “neuralnet” packages in R software were used to select representative genes and construct diagnostic models for osteosarcoma. The next step is to validate the model of the artificial neural network. Then, we performed an immune infiltration analysis by using the training set data. Finally, we constructed a prognostic model using representative genes for prognostic analysis. The copy number of osteosarcoma was also analyzed. A random forest classifier identified nine representative genes (ANK1, TGFBR3, RSF21, HSPB8, ITGA7, RHD, AASS, GREM2, NFASC). HSPB8, RHD, AASS, and NFASC were genes we identified that have not been previously reported to be associated with osteosarcoma. The osteosarcoma diagnostic model we constructed has good performance with areas under the curves (AUCs) of 1 and 0.987 in the training and validation groups, respectively. This study opens new horizons for the early diagnosis of osteosarcoma and provides representative markers for the future treatment of osteosarcoma. This is the first study to pioneer the establishment of a genetic diagnosis model for osteosarcoma and advance the development of osteosarcoma diagnosis and treatment.
Collapse
|
66
|
TREM2 as a Prognostic Biomarker for Osteosarcoma Microenvironment Remodeling. JOURNAL OF ONCOLOGY 2023; 2023:3677789. [PMID: 36844870 PMCID: PMC9957636 DOI: 10.1155/2023/3677789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/23/2022] [Accepted: 11/24/2022] [Indexed: 02/19/2023]
Abstract
The tumor microenvironment (TME) acts as a crucial role in the occurrence and development of osteosarcoma (OS). Despite this, the mechanism controlling the components of immunity and stroma in the tumor microenvironment remains a mystery. To conduct this study, we download and collate transcriptome data from the TARGET database, whose full name is Therapeutically Applicable Research to Generate Effective Treatments, as well as available clinical information of OS. The CIBERSORT and ESTIMATE methodology are used to acquire the proportions of components of immunity and stroma and tumor-infiltrating immune cells (TICs). Protein-protein interaction (PPI) networks and Cox regression analysis are used to select differentially expressed genes (DEGs). A prognostic biomarker is determined by intersecting univariate COX and PPI results, which lead to the finding of Triggering receptor expressed on myeloid cells-2 (TREM2). Based on the next analysis, TREM2 expression is positively correlated with OS survival time. Immune function-related genes have enrichment in the group with high expression of TREM2, according to gene set enrichment analysis (GSEA). The percentage of TICs by CIBERSORT methodology revealed that the expression of TREM2 is positively associated with follicular helper T cells, CD8-positive T cells, and M2 macrophages and negatively correlated with plasma cells, M0 macrophages, and naive CD4-positive T cells. All results suggest a possible integral role of TREM2 in the immune-related events of TME. Therefore, TREM2 may be a potential indicator of remodeling of TME in osteosarcoma, which is useful and helpful in predicting the clinical prognostic outcome of OS patients and provide a unique perspective for immunotherapy for OS.
Collapse
|
67
|
Macrophage Repolarization as a Therapeutic Strategy for Osteosarcoma. Int J Mol Sci 2023; 24:ijms24032858. [PMID: 36769180 PMCID: PMC9917837 DOI: 10.3390/ijms24032858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Macrophages are versatile immune cells and can adapt to both external stimuli and their surrounding environment. Macrophages are categorized into two major categories; M1 macrophages release pro-inflammatory cytokines and produce protective responses that lead to antimicrobial or antitumor activity. M2 or tumor-associated macrophages (TAM) release anti-inflammatory cytokines that support tumor growth, invasion capacity, and metastatic potential. Since macrophages can be re-polarized from an M2 to an M1 phenotype with a variety of strategies, this has emerged as an innovative anti-cancer approach. Osteosarcoma (OS) is a kind of bone cancer and consists of a complex niche, and immunotherapy is not very effective. Therefore, immediate attention to new strategies is required. We incorporated the recent studies that have used M2-M1 repolarization strategies in the aspect of treating OS cancer.
Collapse
|
68
|
Targets Involved in the Anti-Cancer Activity of Quercetin in Breast, Colorectal and Liver Neoplasms. Int J Mol Sci 2023; 24:ijms24032952. [PMID: 36769274 PMCID: PMC9918234 DOI: 10.3390/ijms24032952] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Phytochemicals have long been effective partners in the fight against several diseases, including cancer. Among these, flavonoids are valuable allies for both cancer prevention and therapy since they are known to influence a large panel of tumor-related processes. Particularly, it was revealed that quercetin, one of the most common flavonoids, controls apoptosis and inhibits migration and proliferation, events essential for the development of cancer. In this review, we collected the evidence on the anti-cancer activity of quercetin exploring the network of interactions between this flavonol and the proteins responsible for cancer onset and progression focusing on breast, colorectal and liver cancers, owing to their high worldwide incidence. Moreover, quercetin proved to be also a potentiating agent able to push further the anti-cancer activity of common employed anti-neoplastic agents, thus allowing to lower their dosages and, above all, to sensitize again resistant cancer cells. Finally, novel approaches to delivery systems can enhance quercetin's pharmacokinetics, thus boosting its great potentiality even further. Overall, quercetin has a lot of promise, given its multi-target potentiality; thus, more research is strongly encouraged to properly define its pharmaco-toxicological profile and evaluate its potential for usage in adjuvant and chemoprevention therapy.
Collapse
|
69
|
Zhang Y, Chen Y, Chen C, Guo H, Zhou C, Wang H, Liu Z. PITX1 suppresses osteosarcoma metastasis through exosomal LINC00662-mediated M2 macrophage polarization. Clin Exp Metastasis 2023; 40:79-93. [PMID: 36334221 PMCID: PMC9898340 DOI: 10.1007/s10585-022-10192-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 10/24/2022] [Indexed: 11/08/2022]
Abstract
Paired-like homeodomain transcription factor 1 (PITX1) is frequently downregulated in cancers, including osteosarcoma (OS). However, its role in OS remains unknown. Therefore, we aimed to explore the functions and potential mechanisms of PITX1 in OS malignant progression. Elevated PITX1 suppressed OS cell proliferation and migration, based on transwell, proliferation, and colony formation assays. Pathway enrichment analysis of differentially-expressed genes between PITX1-overexpressing and control OS cells indicated that PITX1 expression was associated with the FAK/Src and PI3k/Akt signaling pathways. Mechanistically, ubiquitination assays and rescue experiments showed that PITX1 interacted with transcription factor STAT3, leading to decreased STAT3 transcriptional activity, which repressed the expression of LINC00662. Specific knockdown of LINC00662 reduced the tumor growth and invasion of OS cells induced by downregulated PITX1. Moreover, exosomal LINC00662, derived from PITX1 knockdown OS cell lines activated M2 macrophages in cell co-culture assays. M2 macrophage secreted several cytokines, among which CCL22 was found to cause OS cell EMT. Collectively, our data indicate that PITX1 suppresses OS cell proliferation and metastasis by downregulating LINC00662. Moreover, LINC00662 can be packaged into OS cell-derived exosomes to mediate M2 macrophage polarization to promote OS metastasis via CCL22.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, Guangdong, China
| | - Yelong Chen
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, 515041, Guangdong, China
| | - Chuangzhen Chen
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, Guangdong, China
| | - Huancheng Guo
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, 515041, Guangdong, China
| | - Chunbin Zhou
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, 515041, Guangdong, China
| | - Hu Wang
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, 515041, Guangdong, China
| | - Zhaoyong Liu
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, 515041, Guangdong, China
| |
Collapse
|
70
|
[miR-125b-5p inhibits proliferation and migration of osteosarcoma cells by negatively regulating RAB3D expression]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:68-75. [PMID: 36856212 DOI: 10.12122/j.issn.1673-4254.2023.01.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
OBJECTIVE To investigate the inhibitory effect of miR-125b-5p on proliferation and migration of osteosarcoma and the role of RAB3D in mediating this effect. METHODS The expression level of miR-125b-5p was detected by qRT-PCR in a normal bone cell line (hFOB1.19) and in two osteosarcoma OS cell lines (MG63 and HOS). A miR-125b-5p mimic or inhibitor was transfected in the osteosarcoma cell lines via liposome and the changes in cell proliferation and migration were detected with EDU and Transwell experiments. Bioinformatic analysis was conducted for predicting the target gene of miR-125b-5p, and the expression level of RAB3D in hFOB1.19, MG63, and HOS cells was detected by Western blotting. In the two osteosarcoma cell lines transfected with miR-125b-5p mimic or inhibitor, the expression levels of RAB3D mRNA and protein in osteosarcoma cells were examined with qRT-PCR and Western blotting. The effects of RAB3D overexpression, RAB3D knockdown, or overexpression of both miR-125b-5p and RAB3D on the proliferation and migration of cells were assessed using EDU and Transwell experiments. RESULTS The two osteosarcoma cell lines had significantly lower expression levels of miR-125b-5p (P < 0.05). Bioinformatic analysis predicted that RAB3D was a possible target gene regulated by miR-125b-5p. In osteosarcoma cells, overexpression of miR-125b-5p significantly lowered the expression of RAB3D protein (P < 0.05); inhibiting miR-125b-5p expression significantly decreased RAB3D expression only at the protein level (P < 0.05) without obviously affecting its mRNA level. Modulation of miR-125b-5p and RAB3D levels produced opposite effects on proliferation and migration of osteosarcoma cells, and in cells with overexpression of both miR-125b-5p and RAB3D, the effect of RAB3D on cell proliferation and migration was blocked by miR-125b-5p overexpression (P < 0.05). CONCLUSION Overexpression of miR-125b-5p inhibits the proliferation and migration of osteosarcoma cells by regulating the expression of RAB3D at the post-transcriptional level.
Collapse
|
71
|
Hou C, Lu M, Lei Z, Dai S, Chen W, Du S, Jin Q, Zhou Z, Li H. HMGB1 Positive Feedback Loop Between Cancer Cells and Tumor-Associated Macrophages Promotes Osteosarcoma Migration and Invasion. J Transl Med 2023; 103:100054. [PMID: 36801636 DOI: 10.1016/j.labinv.2022.100054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 10/11/2022] [Accepted: 12/03/2022] [Indexed: 01/11/2023] Open
Abstract
Numerous studies have demonstrated the key roles of tumor-associated macrophages (TAMs) in osteosarcoma metastasis. Higher levels of high mobility group box 1 (HMGB1) promote osteosarcoma progression. However, whether HMGB1 is involved in the polarization of M2 macrophages into M1 macrophages in osteosarcoma remains largely unknown. Here, HMGB1 and CD206 mRNA expression levels were measured by a quantitative reverse transcription-polymerase chain reaction in osteosarcoma tissues and cells. HMGB1 and receptor for advanced glycation end products (RAGE) protein expression levels were measured by western blotting. Osteosarcoma migration was measured using transwell and wound-healing assays, while a transwell assay determined osteosarcoma invasion. Macrophage subtypes were detected using flow cytometry. HMGB1 expression levels were aberrantly enhanced in osteosarcoma tissues compared with normal tissues and were positively correlated with AJCC III and IV stages, lymph node metastasis, and distant metastasis. Silencing HMGB1 inhibited the migration, invasion, and epithelial-mesenchymal transition (EMT) of osteosarcoma cells. Furthermore, reduced HMGB1 expression levels in conditioned media derived from osteosarcoma cells induced the polarization of M2 TAMs to M1 TAMs. In addition, silencing HMGB1 inhibited the liver and lung metastasis of tumors and reduced the expression levels of HMGB1, CD163, and CD206 in vivo. HMGB1 was found to regulate macrophage polarization through RAGE. Polarized M2 macrophages induced osteosarcoma migration and invasion, activating HMGB1 expression in osteosarcoma cells to form a positive feedback loop. In conclusion, HMGB1 and M2 macrophages enhanced osteosarcoma migration, invasion, and EMT through positive feedback regulation. These findings reveal the significance of tumor cell and TAM interactions in the metastatic microenvironment.
Collapse
Affiliation(s)
- Changhe Hou
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ming Lu
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zixiong Lei
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Shuangwu Dai
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Wei Chen
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Shaohua Du
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Qinglin Jin
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zhongxin Zhou
- Department of Vascular Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| | - Haomiao Li
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
72
|
Zeng J, Peng Y, Wang D, Ayesha K, Chen S. The interaction between osteosarcoma and other cells in the bone microenvironment: From mechanism to clinical applications. Front Cell Dev Biol 2023; 11:1123065. [PMID: 37206921 PMCID: PMC10189553 DOI: 10.3389/fcell.2023.1123065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/10/2023] [Indexed: 05/21/2023] Open
Abstract
Osteosarcoma is a primary bone tumor with a high mortality rate. The event-free survival rate has not improved significantly in the past 30 years, which brings a heavy burden to patients and society. The high heterogeneity of osteosarcoma leads to the lack of specific targets and poor therapeutic effect. Tumor microenvironment is the focus of current research, and osteosarcoma is closely related to bone microenvironment. Many soluble factors and extracellular matrix secreted by many cells in the bone microenvironment have been shown to affect the occurrence, proliferation, invasion and metastasis of osteosarcoma through a variety of signaling pathways. Therefore, targeting other cells in the bone microenvironment may improve the prognosis of osteosarcoma. The mechanism by which osteosarcoma interacts with other cells in the bone microenvironment has been extensively investigated, but currently developed drugs targeting the bone microenvironment have poor efficacy. Therefore, we review the regulatory effects of major cells and physical and chemical properties in the bone microenvironment on osteosarcoma, focusing on their complex interactions, potential therapeutic strategies and clinical applications, to deepen our understanding of osteosarcoma and the bone microenvironment and provide reference for future treatment. Targeting other cells in the bone microenvironment may provide potential targets for the development of clinical drugs for osteosarcoma and may improve the prognosis of osteosarcoma.
Collapse
Affiliation(s)
- Jin Zeng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yi Peng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Dong Wang
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Khan Ayesha
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Shijie Chen
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
- *Correspondence: Shijie Chen,
| |
Collapse
|
73
|
Ye H, Tan L, Tu C, Min L. Exosomes in sarcoma: Prospects for clinical applications. Crit Rev Oncol Hematol 2023; 181:103895. [PMID: 36481305 DOI: 10.1016/j.critrevonc.2022.103895] [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: 09/02/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Sarcoma is a group of rare and heterogeneous mesenchymal tumors, prone to late diagnosis and poor prognosis. Exosomes are cell-derived small extracellular vesicles found in most body fluids and contain nucleic acids, proteins, lipids, and other molecules. Qualitative and quantitative changes of exosomes and the contents are associated with sarcoma progression, exhibiting their potential as biomarkers. Exosomes possess the capacity of evading immune responses, bioactivity for trafficking, tumor tropism, and lesion residence. Thus, exosomes could be engineered as tumor-specific vehicles in drugs and RNA delivery systems. Exosomes might also serve as therapeutic targets in targeted therapy and immunotherapy and be involved in chemotherapy resistance. Here, we provide a comprehensive summary of exosome applications in liquid biopsy-based diagnosis and explore their implications in the delivery system, targeted therapy, and chemotherapy resistance of sarcoma. Moreover, challenges in exosome clinical applications are raised and some future research directions are proposed.
Collapse
Affiliation(s)
- Huali Ye
- West China Hospital, West China School of Medicine, Sichuan University, Guoxue Xiang No. 37, Chengdu 610041, Sichuan, People's Republic of China
| | - Linyun Tan
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Guoxue Xiang No. 37, Chengdu 610041, Sichuan, People's Republic of China
| | - Chongqi Tu
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Guoxue Xiang No. 37, Chengdu 610041, Sichuan, People's Republic of China
| | - Li Min
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Guoxue Xiang No. 37, Chengdu 610041, Sichuan, People's Republic of China.
| |
Collapse
|
74
|
Wang J, Gong M, Xiong Z, Zhao Y, Xing D. ADAM19 and TUBB1 Correlate with Tumor Infiltrating Immune Cells and Predicts Prognosis in Osteosarcoma. Comb Chem High Throughput Screen 2023; 26:135-148. [PMID: 35388751 DOI: 10.2174/1386207325666220406112305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/19/2022] [Accepted: 01/29/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Osteosarcoma is the most common type of primary malignant bone tumor. INTRODUCTION This study aimed to explore potential key prognostic genes and their roles in osteosarcoma. METHODS Three microarray datasets for osteosarcoma were downloaded from the GEO database. Differentially expressed genes (DEGs) were screened by the Limma package. Functional enrichment analysis was performed based on DAVID, GeneMANIA, and Metascape databases. Prognostic value of DEGs was elevated by survival analysis. CIBERSORT was used to assess the infiltrating abundance of 22 immune cells, followed by the Pearson correlation analysis between immune cells and prognosis-related genes. Gene set enrichment analysis and drug-gene interactions prediction were performed for prognosis-related genes. RESULTS A total of 8 common up-regulated DEGs and 13 common down-regulated DEGs were screened in the GSE36001 and GSE56001 datasets. Enrichment analysis showed these DEGs were implicated in platelet activation, SMAD protein phosphorylation, lymphocyte/leukocyte/T cells activation, and cell migration. Survival analysis indicated that elevated expression of ADAM19 and TUBB1 were associated with a favorable prognosis. CIBERSORT algorithm revealed the higher infiltrating level of CD8 T cells, macrophages M0, and M2 in osteosarcoma. ADAM19 expression positively correlated with naïve B cells and negatively correlated with activated dendritic cells infiltrating abundance. TUBB1 expression positively correlated with gamma delta T cells while negatively correlated with helper follicular T cells infiltrating abundance. A total of 56 drugs were found to target TUBB1. CONCLUSION ADAM19 and TUBB1 could be prognostic biomarkers in osteosarcoma. Both their expression correlates with tumor infiltrating immune cells. TUBB1 was a multi-drug target that might be a therapeutic target in osteosarcoma.
Collapse
Affiliation(s)
- Jun Wang
- Department of Orthopedics and Trauma, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Mingzhi Gong
- Department of Orthopedics and Trauma, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Zhenggang Xiong
- Department of Orthopedics and Trauma, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Yangyang Zhao
- Department of Orthopedics and Trauma, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Deguo Xing
- Department of Orthopedics and Trauma, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| |
Collapse
|
75
|
Current State of Immunotherapy and Mechanisms of Immune Evasion in Ewing Sarcoma and Osteosarcoma. Cancers (Basel) 2022; 15:cancers15010272. [PMID: 36612267 PMCID: PMC9818129 DOI: 10.3390/cancers15010272] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
We argue here that in many ways, Ewing sarcoma (EwS) is a unique tumor entity and yet, it shares many commonalities with other immunologically cold solid malignancies. From the historical perspective, EwS, osteosarcoma (OS) and other bone and soft-tissue sarcomas were the first types of tumors treated with the immunotherapy approach: more than 100 years ago American surgeon William B. Coley injected his patients with a mixture of heat-inactivated bacteria, achieving survival rates apparently higher than with surgery alone. In contrast to OS which exhibits recurrent somatic copy-number alterations, EwS possesses one of the lowest mutation rates among cancers, being driven by a single oncogenic fusion protein, most frequently EWS-FLI1. In spite these differences, both EwS and OS are allied with immune tolerance and low immunogenicity. We discuss here the potential mechanisms of immune escape in these tumors, including low representation of tumor-specific antigens, low expression levels of MHC-I antigen-presenting molecules, accumulation of immunosuppressive M2 macrophages and myeloid proinflammatory cells, and release of extracellular vesicles (EVs) which are capable of reprogramming host cells in the tumor microenvironment and systemic circulation. We also discuss the vulnerabilities of EwS and OS and potential novel strategies for their targeting.
Collapse
|
76
|
Zhou Y, Jin Q, Chang J, Zhao Z, Sun C. Long non-coding RNA ZMIZ1-AS1 promotes osteosarcoma progression by stabilization of ZMIZ1. Cell Biol Toxicol 2022; 38:1013-1026. [PMID: 34508303 DOI: 10.1007/s10565-021-09641-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/09/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND Osteosarcomas (OS) are frequent primary sarcomas of the bone in children and adolescents. The long non-coding RNAs (lncRNAs) can affect the progression of many cancers by their sense transcripts. The present study was designed to probe the role of ZMIZ1-AS1 and the downstream pathway in OS progression. METHODS Cell proliferation, invasion, and migration were detected by colony formation, transwell, and wound healing assays. The binding of SOX2 or MYC protein with ZMIZ1-AS1 promoter was explored by ChIP assay and dual-luciferase reporter assay. Interaction between PTBP1 protein and ZMIZ1-AS1 (or ZMIZ1 mRNA) was detected by RIP assay. RESULTS SOX2 and MYC are the downstream effectors of the Hippo pathway and transcriptionally activated ZMIZ1-AS1. Compared to the controls, OS tissues and cells contained higher ZMIZ1-AS1 expression. Silencing of ZMIZ1-AS1 repressed OS cell viability, proliferation, migration, and invasion. Our findings further showed that ZMIZ1-AS1 recruits RNA-binding protein PTBP1 to stabilize ZMIZ1 mRNA. PTBP1 or ZMIZ1 overexpression rescues the suppressive effects of silenced ZMIZ1-AS1 on OS cellular processes. Importantly, ZMIZ1-AS1 promotes OS growth in vivo by stabilization of ZMIZ1. CONCLUSIONS Long non-coding RNA ZMIZ1-AS1 promotes OS progression by stabilization of ZMIZ1. The Hippo pathway is inactivated in osteosarcoma. Transcriptional factors SOX2 and MYC downstream the Hippo pathway induce the upregulation of ZMIZ1-AS1 in osteosarcoma. ZMIZ1-AS1 recruits RNA binding protein PTBP1 that stabilizes ZMIZ1, the sense transcript of ZMIZ1-AS1. ZMIZ1-AS1 promotes osteosarcoma cell viability, proliferation, migration, and invasion by ZMIZ1 in a PTBP1 dependent manner.
Collapse
Affiliation(s)
- Yichi Zhou
- Department of Orthopedics, CR & WISCO General Hospital, Wuhan, 430000, Hubei, China
| | - Qi Jin
- Department of Orthopedics, CR & WISCO General Hospital, Wuhan, 430000, Hubei, China
| | - Jianzhong Chang
- Department of Orthopedics, CR & WISCO General Hospital, Wuhan, 430000, Hubei, China
| | - Zufa Zhao
- Department of Orthopedics, CR & WISCO General Hospital, Wuhan, 430000, Hubei, China
| | - Chengjun Sun
- Department of Orthopedics, CR & WISCO General Hospital, Wuhan, 430000, Hubei, China.
| |
Collapse
|
77
|
Liu Z, Zhang L, Zhong Y. Characterization of osteosarcoma subtypes mediated by macrophage-related genes and creation and validation of a risk score system to quantitatively assess the prognosis of osteosarcoma and reflect the tumor microenvironment. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1318. [PMID: 36660647 PMCID: PMC9843337 DOI: 10.21037/atm-22-5613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
Background Macrophages are the main immune components in the microenvironment of osteosarcoma. The treatment strategy centered on macrophages has become a hot topic to improve cancer treatment. However, the research on the role of macrophages in the treatment of osteosarcoma is still in its infancy. Methods The data of osteosarcoma samples were downloaded from the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) and GSE21257 datasets, and the macrophage enrichment fraction of osteosarcoma samples in TARGET was calculated by single-sample gene set enrichment analysis (ssGSEA) method to screen macrophage-related genes for consensus clustering. Differential expression analysis, univariable Cox, and least absolute shrinkage and selection operator (LASSO) regression were conducted to select reliable predictors and create a risk score system. The GSE21257 dataset was used as a verification set to verify the accuracy of risk score system. Results We identified 2 osteosarcoma clusters mediated by 22 macrophage score-related genes, namely cluster 1 (C1) and cluster 2 (C2). Compared with C2, C1 had a significant advantage in prognosis, and the degree of immune cell infiltration in tumor microenvironment (TME) was significantly higher, the expression of immune checkpoint molecules was significantly enhanced, and the Tumor Immune Dysfunction and Exclusion (TIDE) score was also significantly down-regulated. A robust risk score system was presented and validated, which demonstrated accuracy and independence in assessing the risk of death of osteosarcoma. The risk score system could also monitor TME infiltration in osteosarcoma samples and showed a close relationship with osteosarcoma biology, including metastasis and immunity. Conclusions We identified 2 types of clusters mediated by macrophage-related genes and helped to analyze the cluster suitable for immunotherapy. A new prognostic risk score system was created to quantitatively evaluate the prognosis and TME of osteosarcoma, and to provide a new entry point for the design of personalized treatment.
Collapse
Affiliation(s)
- Zhe Liu
- Department of Orthopedics, Jiangxi Cancer Hospital, Nanchang, China
| | - Lei Zhang
- Department of Orthopedics, Jiangxi Cancer Hospital, Nanchang, China
| | - Yun Zhong
- Department of Lymphohematology and Oncology, Jiangxi Cancer Hospital, Nanchang, China
| |
Collapse
|
78
|
Xia Y, Wang D, Piao Y, Chen M, Wang D, Jiang Z, Liu B. Modulation of immunosuppressive cells and noncoding RNAs as immunotherapy in osteosarcoma. Front Immunol 2022; 13:1025532. [PMID: 36457998 PMCID: PMC9705758 DOI: 10.3389/fimmu.2022.1025532] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/03/2022] [Indexed: 07/21/2023] Open
Abstract
The most common bone cancer is osteosarcoma (OS), which mostly affects children and teenagers. Early surgical resection combined with chemotherapy significantly improves the prognosis of patients with OS. Existing chemotherapies have poor efficacy in individuals with distant metastases or inoperable resection, and these patients may respond better to novel immunotherapies. Immune escape, which is mediated by immunosuppressive cells in the tumour microenvironment (TME), is a major cause of poor OS prognosis and a primary target of immunotherapy. Myeloid-derived suppressor cells, regulatory T cells, and tumour-associated macrophages are the main immunosuppressor cells, which can regulate tumorigenesis and growth on a variety of levels through the interaction in the TME. The proliferation, migration, invasion, and epithelial-mesenchymal transition of OS cells can all be impacted by the expression of non-coding RNAs (ncRNAs), which can also influence how immunosuppressive cells work and support immune suppression in TME. Interferon, checkpoint inhibitors, cancer vaccines, and engineered chimeric antigen receptor (CAR-T) T cells for OS have all been developed using information from studies on the metabolic properties of immunosuppressive cells in TME and ncRNAs in OS cells. This review summarizes the regulatory effect of ncRNAs on OS cells as well as the metabolic heterogeneity of immunosuppressive cells in the context of OS immunotherapies.
Collapse
Affiliation(s)
- Yidan Xia
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Yuting Piao
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| | - Minqi Chen
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| | - Duo Wang
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| | - Ziping Jiang
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| | - Bin Liu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
79
|
Fan L, Zhong Z, Lin Y, Li J. Non-coding RNAs as potential biomarkers in osteosarcoma. Front Genet 2022; 13:1028477. [PMID: 36338952 PMCID: PMC9627036 DOI: 10.3389/fgene.2022.1028477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/07/2022] [Indexed: 11/24/2022] Open
Abstract
Osteosarcoma (OS) is a primary solid malignant tumor that occurs most frequently in the metaphysis of long bones. More likely to happen to children and adolescents. OS has high mortality and disability rate. However, the etiology and pathogenesis of OS have not been fully understood till now. Due to the lack of effective biomarkers, OS cannot be precisely detected in the early stage. With the application of next-generation and high-throughput sequencing, more and more abnormally expressed non-coding RNAs(ncRNAs) have been identified in OS. Growing evidences have suggested the ncRNAs, such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), have played an important role in the tumorigenesis and progression of OS. Thus, they can be served as novel biomarkers for diagnosis, treatment and prognosis. This review summarized the application of ncRNA as biomarkers in OS in detail, and discussed the limitation and future improvement of the potential biomarkers.
Collapse
Affiliation(s)
- Lijuan Fan
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, Henan, China
- Luoyang Postgraduate Training Department, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zhenhao Zhong
- Department of Spinal Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Yubo Lin
- School of Clinical Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Jitian Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, Henan, China
- Luoyang Postgraduate Training Department, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The First College for Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- *Correspondence: Jitian Li,
| |
Collapse
|
80
|
Identification of Two Novel Immune Subtypes Characterized by Distinct Prognosis and Tumor Microenvironment in Osteosarcoma. J Immunol Res 2022; 2022:2181525. [PMID: 36254197 PMCID: PMC9569206 DOI: 10.1155/2022/2181525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/02/2022] [Indexed: 11/18/2022] Open
Abstract
Osteosarcoma is a kind of primary malignant tumor of bone. In recent years, its therapeutic effect and prognostic survival are dissatisfactory. The tumor immune microenvironment (TIME) reflects immune status of patients, but it is little known in osteosarcoma. Therefore, this study attempts to conduct a comprehensive analysis to explore TIME of osteosarcoma and identify TIME-related subtypes for clinical management and treatment. We successfully established two novel tumor immune infiltration clusters (TIIC) which are characterized by difference of microenvironment and immune-related biological processes. High tumor immune infiltration cluster (H-TIIC) subtypes with higher immune infiltration score shows a better overall survival. Further, the two immune subtypes are shown to differ in immunotherapy and chemotherapy. The results would be helpful for clinical decision in osteosarcoma.
Collapse
|
81
|
Zhou H, Gan M, Jin X, Dai M, Wang Y, Lei Y, Lin Z, Ming J. miR‑382 inhibits breast cancer progression and metastasis by affecting the M2 polarization of tumor‑associated macrophages by targeting PGC‑1α. Int J Oncol 2022; 61:126. [PMID: 36069230 PMCID: PMC9477106 DOI: 10.3892/ijo.2022.5416] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 08/19/2022] [Indexed: 11/06/2022] Open
Abstract
Macrophages are principal immune cells with a high plasticity in the human body that can differentiate under different conditions in the tumor microenvironment to adopt two polarized phenotypes with opposite functions. Therefore, converting macrophages from the immunosuppressive phenotype (M2) to the inflammatory phenotype (M1) is considered a promising therapeutic strategy for cancer. However, the molecular mechanisms underlying this conversion process have not yet been completely elucidated. In recent years, microRNAs (miRNAs or miRs) have been shown to play key roles in regulating macrophage polarization through their ability to modulate gene expression. In the present study, it was found that miR‑382 expression was significantly downregulated in tumor‑associated macrophages (TAMs) and M2‑polarized macrophages in breast cancer. In vitro, macrophage polarization toward the M2 phenotype and M2‑type cytokine release were inhibited by transfection with miR‑382‑overexpressing lentivirus. Similarly, the overexpression of miR‑382 inhibited the ability of TAMs to promote the malignant behaviors of breast cancer cells. In addition, peroxisome proliferator‑activated receptor γ coactivator‑1α (PGC‑1α) was identified as the downstream target of miR‑382 and it was found that PGC‑1α affected macrophage polarization by altering the metabolic status. The ectopic expression of PGC‑1α restored the phenotype and cytokine secretion of miR‑382‑overexpressing macrophages. Furthermore, PGC‑1α expression reversed the miR‑382‑induced changes in the metabolic state of TAMs and the effects of TAMs on breast cancer cells. Of note, the in vivo growth and metastasis of 4T1 cells were inhibited by miR‑382‑overexpressing TAMs. Taken together, the results of the present study suggest that miR‑382 may alter the metabolic status of macrophages by targeting PGC‑1α, thereby decreasing the proportion of TAMs with the M2 phenotype, and inhibiting the progression and metastasis of breast cancer.
Collapse
Affiliation(s)
- Hua Zhou
- Department of Breast and Thyroid Surgery, The Affiliated Shapingba Hospital of Chongqing University, Chongqing 400030, P.R. China
| | - Mingyu Gan
- Shanxi Medical University, Taiyuan, Shanxi 030607, P.R. China
| | - Xin Jin
- Department of Critical Care Medicine, The Affiliated Fuling Hospital of Chongqing University, Chongqing 408099, P.R. China
| | - Meng Dai
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yuanyuan Wang
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Youyang Lei
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Zijing Lin
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jia Ming
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
82
|
Zu D, Dong Q, Chen S, Chen Y, Yao J, Zou Y, Lin J, Fang B, Wu B. miRNA-331-3p Affects the Proliferation, Metastasis, and Invasion of Osteosarcoma through SOCS1/JAK2/STAT3. JOURNAL OF ONCOLOGY 2022; 2022:6459029. [PMID: 36199788 PMCID: PMC9529391 DOI: 10.1155/2022/6459029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/18/2022]
Abstract
MicroRNAs (miRNAs) are regulatory small noncoding RNAs that play a key role in several types of cancer. It has been reported that miR-331-3p is involved in the development and progression of various cancers, but there are few reports regarding osteosarcoma (OS). The public GEO database was used to analyze the survival difference of miR-331-3p in OS organizations. The level of cell proliferation assay was assessed by CCK-8 and colony formation. First, transwell and wound-healing assays were used to detect the transfer and invasion ability of miR-331-3p in OS. Second, TargetScan, miRDBmiR, TarBase, and dual-luciferase reporter gene assays were used to determine SOCS1 as a targeted regulator. Third, Western blot and immunohistochemistry were used to detect the expression of protein levels. Finally, a mouse model of subcutaneously transplantable tumors is used to evaluate the proliferation of OS in vivo. The low expression of miR-331-3p was negatively correlated with the overall survival of OS patients. Overexpression of miR-331-3p significantly inhibited cell proliferation, metastasis, and invasion. Moreover, miR-331-3p affected the occurrence and development of osteosarcoma by targeting the SOCS1/JAK2/STAT3 signaling pathway. Therefore, miR-331-3p reduces the expression of SOCS1 by combining with its 3'UTR, thereby activating the JAK2/STAT3 signaling pathway to regulate the progression of OS. This provides a new theoretical basis for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Dan Zu
- Central Laboratory, The Central Hospital, Shaoxing University, Shaoxing 312030, China
| | - Qi Dong
- Department of Spine Surgery, The Central Hospital, Shaoxing University, Shaoxing 312030, China
| | - Sunfang Chen
- Department of Spine Surgery, The Central Hospital, Shaoxing University, Shaoxing 312030, China
| | - Yongde Chen
- Central Laboratory, The Central Hospital, Shaoxing University, Shaoxing 312030, China
| | - Jun Yao
- Department of Spine Surgery, The Central Hospital, Shaoxing University, Shaoxing 312030, China
| | - Yubin Zou
- Department of Spine Surgery, The Central Hospital, Shaoxing University, Shaoxing 312030, China
| | - Jiawei Lin
- Department of Spine Surgery, The Central Hospital, Shaoxing University, Shaoxing 312030, China
| | - Bin Fang
- Department of Spine Surgery, The Central Hospital, Shaoxing University, Shaoxing 312030, China
| | - Bing Wu
- Department of Spine Surgery, The Central Hospital, Shaoxing University, Shaoxing 312030, China
| |
Collapse
|
83
|
Hayashi K, Nogawa D, Kobayashi M, Asakawa A, Ohata Y, Kitagawa S, Kubota K, Takahashi H, Yamada M, Oda G, Nakagawa T, Uetake H, Onishi I, Kinowaki Y, Kurata M, Kitagawa M, Yamamoto K. Quantitative high-throughput analysis of tumor infiltrating lymphocytes in breast cancer. Front Oncol 2022; 12:901591. [PMID: 36132149 PMCID: PMC9484474 DOI: 10.3389/fonc.2022.901591] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/22/2022] [Indexed: 12/19/2022] Open
Abstract
In breast cancer (BC), the development of cancer immunotherapy including immune checkpoint inhibitors has progressed. Tumor infiltrating lymphocytes (TILs) is one of the important factors for an immune response between tumor cells and immune cells in the tumor microenvironment, and the presence of TILs has been identified as predictors of response to chemotherapy. However, because complex mechanisms underlies the crosstalk between immune cells and cancer cells, the relationship between immune profiles in the tumor microenvironment and the efficacy of the immune checkpoint blocked has been unclear. Moreover, in many cases of breast cancer, the quantitative analysis of TILs and immuno-modification markers in a single tissue section are not studied. Therefore, we quantified detailed subsets of tumor infiltrating lymphocytes (TILs) from BC tissues and compared among BC subtypes. The TILs of BC tissues from 86 patients were classified using multiplex immunohistochemistry and an artificial intelligence-based analysis system based on T-cell subset markers, immunomodification markers, and the localization of TILs. The levels of CD4/PD1 and CD8/PD1 double-positive stromal TILs were significantly lower in the HER2- BC subtype (p <0.01 and p <0.05, respectively). In triple-negative breast cancer (TNBC), single marker-positive intratumoral TILs did not affect prognosis, however CD4/PDL1, CD8/PD1, and CD8/PDL1 double-positive TILs were significantly associated with TNBC recurrence (p<0.05, p<0.01, and p<0.001, respectively). TIL profiles differed among different BC subtypes, suggesting that the localization of TILs and their tumor-specific subsets influence the BC microenvironment.
Collapse
Affiliation(s)
- Kumiko Hayashi
- Department of Specialized Surgery, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daichi Nogawa
- Department of Comprehensive Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Maki Kobayashi
- Molecular Pathology Group, Translational Research Department, Daiichisankyo RD Novare, Tokyo, Japan
| | - Ayaka Asakawa
- Department of Respiratory Medicine, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yae Ohata
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Shota Kitagawa
- Department of Respiratory Medicine, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuishi Kubota
- Department of Translational Science, Daiichi Sankyo, Inc., Basking Ridge, NJ, United States
| | - Hisashi Takahashi
- Molecular Pathology Group, Translational Research Department, Daiichisankyo RD Novare, Tokyo, Japan
| | - Miyuki Yamada
- Molecular Pathology Group, Translational Research Department, Daiichisankyo RD Novare, Tokyo, Japan
| | - Goshi Oda
- Department of Specialized Surgery, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tsuyoshi Nakagawa
- Department of Specialized Surgery, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Uetake
- Department of Specialized Surgery, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Iichiroh Onishi
- Department of Comprehensive Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuko Kinowaki
- Department of Comprehensive Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Morito Kurata
- Department of Comprehensive Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masanobu Kitagawa
- Department of Comprehensive Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kouhei Yamamoto
- Department of Comprehensive Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Human Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
84
|
Zhang Y, Li L, Tu Y, Feng Z, Li Y, Xiong J. Role of STX6 as a prognostic factor associated with immune infiltration in hepatocellular carcinoma. Oncol Lett 2022; 24:371. [PMID: 36238841 PMCID: PMC9494627 DOI: 10.3892/ol.2022.13491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/11/2022] [Indexed: 11/27/2022] Open
Abstract
Syntaxin 6 (STX6), a soluble N-ethylmaleimide-sensitive factor-activating receptor protein, has formed an increasing part of cancer research. However, to the best of our knowledge, the role of STX6 in hepatocellular carcinoma (HCC) is still unclear. In the present study, data from multiple bioinformatics databases, including The Cancer Genome Atlas, Gene Expression Omnibus, Kaplan-Meier plotter, Tumor Immune Estimation Resource (TIMER) and Gene Expression Profiling Integrative Analysis (GEPIA2), and immunohistochemistry (IHC) were utilized to assess the role of STX6 in HCC. The results demonstrated that STX6 expression was upregulated in HCC tissues compared with normal tissues. STX6 expression was significantly associated with tumor size, Edmondson grade and α-fetoprotein (AFP) level. Furthermore, survival analysis demonstrated that high STX6 expression was significantly associated with poor prognosis in patients with HCC. Furthermore, assessment of the immune infiltrates demonstrated that CD163 expression was positively correlated with the STX6 level when analyzed using the TIMER and GEPIA2 databases. IHC results further demonstrated this association. Furthermore, compared with the typically used AFP, STX6 could have an improved diagnostic value in the diagnosis of HCC. In conclusion, STX6 expression was not only positively associated with poor prognosis but may also be involved in the immune inflammatory reaction in HCC. STX6 may become a potential therapeutic and diagnosis maker for patients with HCC.
Collapse
Affiliation(s)
- Yang Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Leyan Li
- Laboratory of Digestive Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yi Tu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zongfeng Feng
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yong Li
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jianbo Xiong
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
85
|
Sirikaew N, Pruksakorn D, Chaiyawat P, Chutipongtanate S. Mass Spectrometric-Based Proteomics for Biomarker Discovery in Osteosarcoma: Current Status and Future Direction. Int J Mol Sci 2022; 23:ijms23179741. [PMID: 36077137 PMCID: PMC9456544 DOI: 10.3390/ijms23179741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Due to a lack of novel therapies and biomarkers, the clinical outcomes of osteosarcoma patients have not significantly improved for decades. The advancement of mass spectrometry (MS), peptide quantification, and downstream pathway analysis enables the investigation of protein profiles across a wide range of input materials, from cell culture to long-term archived clinical specimens. This can provide insight into osteosarcoma biology and identify candidate biomarkers for diagnosis, prognosis, and stratification of chemotherapy response. In this review, we provide an overview of proteomics studies of osteosarcoma, indicate potential biomarkers that might be promising therapeutic targets, and discuss the challenges and opportunities of mass spectrometric-based proteomics in future osteosarcoma research.
Collapse
Affiliation(s)
- Nutnicha Sirikaew
- Musculoskeletal Science and Translational Research (MSTR) Center, Department of Orthopedics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Dumnoensun Pruksakorn
- Musculoskeletal Science and Translational Research (MSTR) Center, Department of Orthopedics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Parunya Chaiyawat
- Musculoskeletal Science and Translational Research (MSTR) Center, Department of Orthopedics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence: (P.C.); (S.C.)
| | - Somchai Chutipongtanate
- Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Correspondence: (P.C.); (S.C.)
| |
Collapse
|
86
|
Wen Y, Tang F, Tu C, Hornicek F, Duan Z, Min L. Immune checkpoints in osteosarcoma: Recent advances and therapeutic potential. Cancer Lett 2022; 547:215887. [PMID: 35995141 DOI: 10.1016/j.canlet.2022.215887] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 11/02/2022]
Abstract
Osteosarcoma is the most common primary malignant bone tumor and is associated with a high risk of recurrence and distant metastasis. Effective treatment for osteosarcoma, especially advanced osteosarcoma, has stagnated over the past four decades. The advent of immune checkpoint inhibitor (ICI) has transformed the treatment paradigm for multiple malignant tumor types and indicated a potential therapeutic strategy for osteosarcoma. In this review, we discuss recent advances in immune checkpoints, including programmed cell death protein-1 (PD-1), programmed cell death protein ligand-1 (PD-L1), and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), and their related ICIs for osteosarcoma treatment. We present the main existing mechanisms of resistance to ICIs therapy in osteosarcoma. Moreover, we summarize the current strategies for improving the efficacy of ICIs in osteosarcoma and address the potential predictive biomarkers of ICIs treatment in osteosarcoma.
Collapse
Affiliation(s)
- Yang Wen
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Guoxue Xiang No. 37, Chengdu, 610041, Sichuan, People's Republic of China
| | - Fan Tang
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Guoxue Xiang No. 37, Chengdu, 610041, Sichuan, People's Republic of China
| | - Chongqi Tu
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Guoxue Xiang No. 37, Chengdu, 610041, Sichuan, People's Republic of China
| | - Francis Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedics, Sylvester Comprehensive Cancer Center, the University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopaedics, Sylvester Comprehensive Cancer Center, the University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Li Min
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Guoxue Xiang No. 37, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
87
|
m6A-Related lncRNAs Predict Overall Survival of Patients and Regulate the Tumor Immune Microenvironment in Osteosarcoma. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:9315283. [PMID: 35978902 PMCID: PMC9377863 DOI: 10.1155/2022/9315283] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/20/2022] [Accepted: 07/07/2022] [Indexed: 11/26/2022]
Abstract
Background m6A-related lncRNAs have demonstrated great potential tumor diagnostic and therapeutic targets. The goal of this work was to find m6A-regulated lncRNAs in osteosarcoma patients. Method The Cancer Genome Atlas (TCGA) database was used to retrieve RNA sequencing and medical information from osteosarcoma sufferers. The Pearson's correlation test was used to identify the m6A-related lncRNAs. A risk model was built using univariate and multivariable Cox regression analysis. Kaplan–Meier survival analysis and receiver functional requirements were used to assess the risk model's performance (ROC). By using the CIBERSORT method, the associations between the relative risks and different immune cell infiltration were investigated. Lastly, the bioactivities of high-risk and low-risk subgroups were investigated using Gene Set Enrichment Analysis (GSEA). Result A total of 531 m6A-related lncRNAs were obtained from TCGA. Seven lncRNAs have demonstrated prognostic values. A total of 88 OS patients were separated into cluster 1, cluster 2, and cluster 3. The overall survival rate of OS patients in cluster 3 was more favorable than that of those in cluster 1 and cluster 2. The average Stromal score was much higher in cluster 1 than in cluster 2 and cluster 3 (P < 0.05). The expression levels of lncRNAs used in the construction of the risk prediction model in the high-risk group were generally lower than those in the low-risk group. Analysis of patient survival indicated that the survival of the low-risk group was higher than that of the high-risk group (P < 0.0001) and the area under the curve (AUC) of the ROC curve was 0.719. Using the CIBERSORT algorithm, the results revealed that Macrophages M0, Macrophages M2, and T cells CD4 memory resting accounted for a large proportion of immune cell infiltration. By GSEA analysis, our results implied that the high-risk group was mainly involved in unfolded protein response, DNA repair signaling, and epithelial-mesenchymal transition signaling pathway and glycolysis pathway; meanwhile, the low-risk group was mainly involved in estrogen response early and KRAS signaling pathway. Conclusion Our investigation showed that m6A-related lncRNAs remained tightly connected to the immunological microenvironment of osteosarcoma tumors, potentially influencing carcinogenesis and development. The immune microenvironment and immune-related biochemical pathways can be changed by regulating the transcription of M6A modulators or lncRNAs. In addition, we looked for risk-related signaling of m6A-related lncRNAs in osteosarcomas and built and validated the risk prediction system. The findings of our current analysis will facilitate the assessment of outcomes and the development of immunotherapies for sufferers of osteosarcomas.
Collapse
|
88
|
Liu W, Hao Y, Tian X, Jiang J, Qiu Q. The Role of NR4A1 in the Pathophysiology of Osteosarcoma: A Comprehensive Bioinformatics Analysis of the Single-Cell RNA Sequencing Dataset. Front Oncol 2022; 12:879288. [PMID: 35965537 PMCID: PMC9371594 DOI: 10.3389/fonc.2022.879288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Osteosarcoma is a kind of aggressive human malignancy, and the prognosis of the patients with osteosarcoma remains low. Studies have demonstrated that the tumor microenvironment plays a key role in regulating osteosarcoma progression. Recent studies have also shown that scRNA-seq plays an essential role in understanding the tumor heterogeneity and distinct subpopulations of tumors. In order to further understand the scRNA-seq data of osteosarcoma tissues, the present study further analyzed the scRNA-seq dataset (GSE152048) and explored the potential role of nuclear receptor-related genes in the pathophysiology of osteosarcoma. In our analysis, we identified 11 cell types in all the osteosarcoma tissues and nuclear receptors (NRs) were distributed in all types of cells. Further stratification analysis showed that NRs were mainly detected in “TIL” and “Osteoblastic” of the metastasis osteosarcoma, in “TIL”, “Myoblast”, “Endothelial”, and “Myeloid” of the primary osteosarcoma, and in “Chondroblastic”, “Osteoblast”, and “Pericyte” of the recurrent osteosarcoma. The NRs were also differentially expressed in different cell types among the metastasis, primary, and recurrent osteosarcoma. Furthermore, several NRs such as NR4A2, NR4A1, and NR3C1 have been found to be differentially expressed in most types of DEGs among metastasis, primary, and recurrent osteosarcoma. A high expression of NR4A1 in the osteosarcoma tissues was significantly correlated with a shorter 5-year overall survival of patients with osteosarcoma. On the other hand, there was no significant association between NR4A2 expression and the 5-year overall survival of patients with osteosarcoma. The expression of NR4A1 was significantly higher in the metastasis osteosarcoma tissues than in the primary osteosarcoma tissues as validated from GSE32981 and GSE154540. The expression of NR4A1 was significantly higher in osteosarcoma tissues from patients with poor chemosensitivity than that from patients with good chemosensitivity as validated from GSE154540. Further analysis of the scRNA-seq data revealed that the percentage of osteoblasts with a high NR4A1 expression was higher in the recurrent osteosarcoma tissues than that with a low NR4A1 expression. In conclusion, the present study may suggest that NR4A1 may be an important prognostic biomarker for osteosarcoma progression. However, further validation studies should be performed to confirm our findings.
Collapse
Affiliation(s)
- Weidong Liu
- Department of Orthopedic, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| | - Yuedong Hao
- Department of Orthopedic, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| | - Xiao Tian
- Department of Orthopedic, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| | - Jing Jiang
- Department of Clinical Laboratory, Nanchang Medical College, Nanchang, China
- *Correspondence: Quanhe Qiu, ; Jing Jiang,
| | - Quanhe Qiu
- Department of Spine Surgery, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
- *Correspondence: Quanhe Qiu, ; Jing Jiang,
| |
Collapse
|
89
|
Zheng B, Song K, Sun L, Gao Y, Qu Y, Ren C, Yan P, Chen W, Guo W, Zhou C, Yue B. Siglec-15-induced autophagy promotes invasion and metastasis of human osteosarcoma cells by activating the epithelial-mesenchymal transition and Beclin-1/ATG14 pathway. Cell Biosci 2022; 12:109. [PMID: 35842729 PMCID: PMC9287887 DOI: 10.1186/s13578-022-00846-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/06/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Pulmonary metastasis is the main cause of poor prognosis in osteosarcoma. Sialic acid-bound immunoglobulin lectin 15 (Siglec-15) has been demonstrated to be obviously correlated with pulmonary metastasis in osteosarcoma patients. However, the effect of Siglec-15 on autophagy in osteosarcoma remains unclear, while the role and mechanism of Siglec-15-related autophagy in lung metastasis also remain unknown. METHODS The expression levels of Siglec-15 and Beclin-1 were detected in osteosarcoma tissues using immunohistochemistry (IHC). The effect of Siglec-15 on metastasis was investigated using Transwell, wound healing and animal experiments with osteosarcoma cells. Corresponding proteins were confirmed using Western blotting when Siglec-15 or Beclin-1 was silenced or overexpressed. Changes in autophagy and the cytoskeleton were detected using immunofluorescence and transmission electron microscopy. RESULTS Siglec-15 and Beclin-1 expression was evaluated both in lung metastases and in patients who presented with pulmonary metastasis of osteosarcoma. Immunoprecipitation experiments revealed that Siglec-15 interacts directly with Beclin-1, an important autophagic protein. Moreover, loss of Siglec-15 distinctly inhibited autophagy and reduced Beclin-1/ATG14 expression. The decreased invasion and migration caused by Siglec-15 silencing could be reversed by Beclin-1 overexpression. Additionally, autophagy can promote the epithelial-mesenchymal transition (EMT) and affect cytoskeletal rearrangement, which was confirmed by overexpression or silencing of Beclin-1. CONCLUSIONS These findings confirmed the role of Siglec-15 in the regulation of autophagy and elaborated the relationship and mechanisms between autophagy and the metastasis of osteosarcoma cells.
Collapse
Affiliation(s)
- Bingxin Zheng
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266000, People's Republic of China
| | - Keliang Song
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266000, People's Republic of China
| | - Lingling Sun
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Yang Gao
- Medical Department, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Yan Qu
- Industrial Investment Department, Haier, Qingdao, People's Republic of China
| | - Chongmin Ren
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266000, People's Republic of China
| | - Peng Yan
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266000, People's Republic of China
| | - Wenfang Chen
- Department of Physiology, Medical College of Qingdao University, Qingdao, People's Republic of China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China
| | - Chuanli Zhou
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266000, People's Republic of China.
| | - Bin Yue
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266000, People's Republic of China.
| |
Collapse
|
90
|
Bioinformatics Analysis Reveals an Association between Autophagy, Prognosis, Tumor Microenvironment, and Immunotherapy in Osteosarcoma. JOURNAL OF ONCOLOGY 2022; 2022:4220331. [PMID: 35874628 PMCID: PMC9303156 DOI: 10.1155/2022/4220331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/13/2022] [Accepted: 06/20/2022] [Indexed: 12/14/2022]
Abstract
Autophagy is a catabolic pathway involved in the regulation of bone homeostasis. We explore clinical correlation of autophagy-related key molecules to establish risk signature for predicting the prognosis, tumor microenvironment (TME), and immunotherapy response of osteosarcoma. Single cell RNA sequencing data from GSE162454 dataset distinguished malignant cells from normal cells in osteosarcoma. Autophagy-related genes (ARGs) were extracted from the established risk signature of the Molecular Signatures Database of Gene Set Enrichment Analysis (GSEA) by univariate Cox and least absolute shrinkage and selection operator (LASSO) Cox regression analysis. Overall survival (OS), TME score, abundance of infiltrating immune cells, and response to immune-checkpoint blockade (ICB) treatment in patients with different risks were compared based on risk score. Nine ARGs were identified and risk signature was constructed. In all osteosarcoma datasets, the OS was significantly longer in the high-risk patients than low-risk onset. Risk signature significantly stratified clinical outcomes, including OS, metastatic status, and survival status. Risk signature was an independent variable for predicting osteosarcoma OS and showed high accuracy. A nomogram based on risk signature and metastases was developed. The calibration curve confirmed the consistency in 1-year, 3-year, and 5-year predicted OS and the actual OS. The risk score was related to 6 kinds of T cells and macrophages, myeloid-derived suppressor cell, natural killer cell, immune score, and stromal score in TME. The risk signature helped in predicting patients' response to anti-PD1/anti-PD-L1 treatment. The ARGs-led risk signature has important value for survival prediction, risk stratification, tumor microenvironment, and immune response evaluation of osteosarcoma.
Collapse
|
91
|
Wang J, Jin J, Chen T, Zhou Q. Curcumol Synergizes with Cisplatin in Osteosarcoma by Inhibiting M2-like Polarization of Tumor-Associated Macrophages. Molecules 2022; 27:molecules27144345. [PMID: 35889217 PMCID: PMC9318016 DOI: 10.3390/molecules27144345] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/26/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Osteosarcoma is the most prevalent bone cancer, and chemotherapy is still an indispensable treatment in its clinical practice. Cisplatin (CDDP) has become the most commonly used agent for osteosarcoma, although the outcomes of CDDP chemotherapy remain unsatisfactory because of frequent resistance. Here, we report on a promising combination therapy where curcumol, a bioactive sesquiterpenoid, enhanced CDDP-induced apoptosis to eradicate osteosarcoma cells, and revealed that M2-like macrophages might be the underlying associated mechanisms. First, we observed that curcumol enhanced the CDDP-mediated inhibition of cell proliferation and augmented the apoptosis in osteosarcoma cell lines. Curcumol contributed to preventing the migration of osteosarcoma cells when combined with CDDP. Moreover, this drug combination showed more potent tumor-growth suppression in the orthotopic transplantation of osteosarcoma K7M2 WT cells. We then estimated chemotherapy-associated drug-resistant genes, including ABCB1, ABCC1 and ABCG2, and found that curcumol significantly reversed the mRNA levels of CDDP-induced ABCB1, ABCC1 and ABCG2 genes in the tumor tissue. Moreover, M2-like macrophages were enriched in osteosarcoma tissues, and were largely decreased after curcumol and CDDP treatment. Taken together, these findings suggest that curcumol inhibits the polarization of M2-like macrophages and could be a promising combination strategy to synergize with CDDP in the osteosarcoma.
Collapse
Affiliation(s)
- Jincheng Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (J.W.); (J.J.)
| | - Jialu Jin
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (J.W.); (J.J.)
- Department of Pharmacy, Hangzhou Medical College, Hangzhou 310053, China;
| | - Ting Chen
- Department of Pharmacy, Hangzhou Medical College, Hangzhou 310053, China;
| | - Qian Zhou
- Department of Pharmacy, Hangzhou Medical College, Hangzhou 310053, China;
- Correspondence:
| |
Collapse
|
92
|
Betlej G, Ząbek T, Lewińska A, Błoniarz D, Rzeszutek I, Wnuk M. RNA 5-methylcytosine status is associated with DNMT2/TRDMT1 nuclear localization in osteosarcoma cell lines. J Bone Oncol 2022; 36:100448. [PMID: 35942470 PMCID: PMC9356272 DOI: 10.1016/j.jbo.2022.100448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/25/2022] Open
Abstract
Selected phenotypic features of three osteosarcoma (OS) cell lines were evaluated. Redox disequilibrium promoted sustained AKT and ERK1/2 activation. Redox imbalance modulated cell death pathways in OS cells. Nuclear levels of TRDMT1 methyltransferase were associated with RNA methylation. A novel marker for predicting therapy response in OS patients is proposed.
Osteosarcoma (OS) is a pediatric malignant bone tumor with unsatisfying improvements in survival rates due to limited understanding of OS biology and potentially druggable targets. The present study aims to better characterize osteosarcoma U-2 OS, SaOS-2, and MG-63 cell lines that are commonly used as in vitro models of OS. We focused on evaluating the differences in cell death pathways, redox equilibrium, the activity of proliferation-related signaling pathways, DNA damage response, telomere maintenance, DNMT2/TRDMT1-based responses and RNA 5-methylcytosine status. SaOS-2 cells were characterized by higher levels of superoxide and nitric oxide that promoted AKT and ERK1/2 activation thus modulating cell death pathways. OS cell lines also differed in the levels and localization of DNA repair regulator DNMT2/TRDMT1. SaOS-2 cells possessed the lowest levels of total, cytoplasmic and nuclear DNMT2/TRDMT1, whereas in MG-63 cells, the highest levels of nuclear DNMT2/TRDMT1 were associated with the most pronounced status of RNA 5-methylcytosine. In silico analysis revealed potential phosphorylation sites at DNMT2/TRDMT1 that may be related to the regulation of DNMT2/TRDMT1 localization. We postulate that redox homeostasis, proliferation-related pathways and DNMT2/TRDMT1-based effects can be modulated as a part of anti-osteosarcoma strategy reflecting diverse phenotypic features of OS cells.
Collapse
Affiliation(s)
- Gabriela Betlej
- Institute of Physical Culture Studies, College of Medical Sciences, University of Rzeszow, Rzeszow 35-310, Poland
| | - Tomasz Ząbek
- Laboratory of Genomics, National Research Institute of Animal Production, Krakowska 1, Balice 32-083, Poland
| | - Anna Lewińska
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Nature Sciences, University of Rzeszow, Pigonia 1, Rzeszow 35-310, Poland
| | - Dominika Błoniarz
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Nature Sciences, University of Rzeszow, Pigonia 1, Rzeszow 35-310, Poland
| | - Iwona Rzeszutek
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Nature Sciences, University of Rzeszow, Pigonia 1, Rzeszow 35-310, Poland
- Corresponding authors.
| | - Maciej Wnuk
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Nature Sciences, University of Rzeszow, Pigonia 1, Rzeszow 35-310, Poland
- Corresponding authors.
| |
Collapse
|
93
|
Xie W, Chang W, Wang X, Liu F, Wang X, Yuan D, Zhang Y. Allicin Inhibits Osteosarcoma Growth by Promoting Oxidative Stress and Autophagy via the Inactivation of the lncRNA MALAT1-miR-376a-Wnt/ β-Catenin Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4857814. [PMID: 35783190 PMCID: PMC9249524 DOI: 10.1155/2022/4857814] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 12/29/2022]
Abstract
Allicin, an organic sulfur compound extracted from the bulb of Allium sativum, can potentially prevent various tumors. Our previous study found that allicin can effectively suppress the proliferation of osteosarcoma cells. However, the molecular mechanisms have not been illustrated. In this study, Saos-2 and U2OS osteosarcoma cells were used to investigate the underlying mechanisms. A series of experiments were carried out to authenticate the anticancer property of allicin. Knockdown of lncRNA MALAT1 inhibited the proliferation, invasion and migration and promoted apoptosis of osteosarcoma cells. Knockdown of miR-376a increased the proliferation, invasion, and migration and dropped apoptosis of osteosarcoma cells. Furthermore, knockdown of miR-376a reversed the influences of MALAT1 silencing in osteosarcoma cells. Based on our data, MALAT1 could downregulate the expression of miR-376a, subsequently accelerating osteosarcoma. Moreover, oxidative stress and autophagy were identified as the potential key pathway of allicin. Allicin inhibited osteosarcoma growth and promoted oxidative stress and autophagy via MALATI-miR-376a. We also found that allicin promotes oxidative stress and autophagy to inhibit osteosarcoma growth by inhibiting the Wnt/β-catenin pathway in vivo and in vitro. All data showed that allicin promotes oxidative stress and autophagy of osteosarcoma via the MALATI-miR-376a-Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Wenpeng Xie
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Wenjie Chang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Xiaole Wang
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Fei Liu
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Xu Wang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Daotong Yuan
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Yongkui Zhang
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
- Shandong Fupai Pharmaceutical Co., Ltd, Jinan, Shandong, 250000, China
| |
Collapse
|
94
|
Peng Z, Li M, Wang Y, Yang H, Wei W, Liang M, Shi J, Liu R, Li R, Zhang Y, Liu J, Shi X, Wan R, Fu Y, Xie R, Wang Y. Self-Assembling Imageable Silk Hydrogels for the Focal Treatment of Osteosarcoma. Front Cell Dev Biol 2022; 10:698282. [PMID: 35794868 PMCID: PMC9251127 DOI: 10.3389/fcell.2022.698282] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/02/2022] [Indexed: 12/24/2022] Open
Abstract
Background: The standard treatment for osteosarcoma comprises complete surgical resection and neoadjuvant chemotherapy, which may cause serious side effects and partial or total limb loss. Therefore, to avoid the disadvantages of traditional treatment, we developed self-assembling imageable silk hydrogels for osteosarcoma. Methods: We analysed whether iodine induced apoptosis in MG-63 and Saos-2 cells by using CCK-8 and flow cytometry assays and transmission electron microscopy. Western blotting was used to analyse the pathway of iodine-induced apoptosis in osteosarcoma cells. PEG400, silk fibroin solution, polyvinylpyrrolidone iodine (PVP-I), and meglumine diatrizoate (MD) were mixed to produce an imageable hydrogel. A nude mouse model of osteosarcoma was established, and the hydrogel was injected locally into the interior of the osteosarcoma with X-ray guidance. The therapeutic effect and biosafety of the hydrogel were evaluated. Results: Iodine treatment at 18 and 20 µM for 12 h resulted in cell survival rate reduced to 50 ± 2.1% and 50.5 ± 2.7% for MG-63 and Sao-2 cells, respectively (p < 0.01). The proportion of apoptotic cells was significantly higher in the iodine-treatment group than in the control group (p < 0.05), and apoptotic bodies were observed by transmission electron microscopy. Iodine could regulate the death receptor pathway and induce MG-63 and Saos-2 cell apoptosis. The hydrogels were simple to assemble, and gels could be formed within 38 min. A force of less than 50 N was required to inject the gels with a syringe. The hydrogels were readily loaded and led to sustained iodine release over 1 week. The osteosarcoma volume in the PEG-iodine-silk/MD hydrogel group was significantly smaller than that in the other three groups (p < 0.001). Caspase-3 and poly (ADP-ribose) polymerase (PARP) expression levels were significantly higher in the PEG-iodine-silk/MD hydrogel group than in the other three groups (p < 0.001). Haematoxylin and eosin (H&E) staining showed no abnormalities in the heart, liver, spleen, lung, kidney, pancreas or thyroid in any group. Conclusions: Self-assembling imageable silk hydrogels could be injected locally into osteosarcoma tissues with X-ray assistance. With the advantages of good biosafety, low systemic toxicity and minimal invasiveness, self-assembling imageable silk hydrogels provide a promising approach for improving the locoregional control of osteosarcoma.
Collapse
Affiliation(s)
- Zhibin Peng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Ming Li
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Yuan Wang
- Innovation and Entrepreneurship Square, Science and Technology Innovation City, Hi-Tech Zone, Harbin, China
| | - Hongbo Yang
- Department of Orthopedic Surgery, Affiliated Hospital of Chifeng University, Chifeng University, Chifeng, China
| | - Wei Wei
- Department of Orthopedic Surgery, Harbin 242 Hospital, Harbin, China
| | - Min Liang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Jianhui Shi
- Department of Orthopedic Surgery, Heilongjiang Provincial Hospital, Harbin, China
| | - Ruixuan Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Rui Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Yubo Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Jingsong Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Xu Shi
- Department of Orthopedic Surgery, Harbin 242 Hospital, Harbin, China
| | - Ran Wan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Yao Fu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Rui Xie
- Department of Digestive Internal Medicine and Photodynamic Therapy Center, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Yansong Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
- *Correspondence: Yansong Wang,
| |
Collapse
|
95
|
Wang B, Wang X, Li P, Niu X, Liang X, Liu G, Liu Z, Ge H. Osteosarcoma Cell-Derived Exosomal ELFN1-AS1 Mediates Macrophage M2 Polarization via Sponging miR-138-5p and miR-1291 to Promote the Tumorgenesis of Osteosarcoma. Front Oncol 2022; 12:881022. [PMID: 35785218 PMCID: PMC9248260 DOI: 10.3389/fonc.2022.881022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/08/2022] [Indexed: 12/28/2022] Open
Abstract
BackgroundExosomes play an important role in cell-cell communication by transferring genetic materials such as long non-coding RNAs (lncRNAs) between cancer cells and tumor-associated macrophages (TAMs) in the tumor microenvironment (TME). Recent studies revealed that lncRNA ELFN1-AS1 could function as an oncogene in many human cancers. However, the role of extracellular lncRNA ELFN1-AS1 in cell-to-cell communication of osteosarcoma (OS) has not been fully investigated.MethodsFunctional studies, including CCK-8, EdU staining and transwell assay were performed to investigate the role of ELFN1-AS1 in the progression of OS. 143B via xenograft mouse model was established to assess the role of ELFN1-AS1 in vivo. In addition, transmission electron microscopy (TEM) and real-time quantitative PCR (RT-qPCR) assay were used to verify the existence of exosomal ELFN1-AS1.ResultsThe level of ELFN1-AS1 was markedly upregulated in patients with advanced OS and in OS cells. In addition, overexpression of ELFN1-AS1 significantly promoted the proliferation, migration and invasion of OS cells, while knockdown of ELFN1-AS1 exhibited the opposite effects. Meanwhile, ELFN1-AS1 could be transferred from OS cells to macrophages via exosomes. Exosomal ELFN1-AS1 from 143B cells was able to promote macrophage M2 polarization, and M2 macrophage in return facilitated OS progression. Mechanistically, overexpression of ELFN1-AS1 upregulated CREB1 level via sponging miR-138-5p and miR-1291 in macrophage via.ConclusionOS cell-derived exosomal ELFN1-AS1 was able to induce macrophage M2 polarization via sponging miR-138-5p and miR-1291, and M2 macrophage notably facilitated the progression of OS. These data suggested that ELFN1-AS1 might serve as a potential therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Bangmin Wang
- Department of Bone Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xin Wang
- Department of Bone Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Po Li
- Department of Bone Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xiaoying Niu
- Department of Bone Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xiaoxiao Liang
- Department of Bone Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Guancong Liu
- Department of Bone Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Zhiyong Liu
- Department of Bone Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Hong Ge
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- *Correspondence: Hong Ge,
| |
Collapse
|
96
|
Yao W, Hou J, Liu G, Wu F, Yan Q, Guo L, Wang C. LncRNA CBR3-AS1 promotes osteosarcoma progression through the network of miR-140-5p/DDX54-NUCKS1-mTOR signaling pathway. Mol Ther Oncolytics 2022; 25:189-200. [PMID: 35592388 PMCID: PMC9092395 DOI: 10.1016/j.omto.2022.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 03/04/2022] [Indexed: 02/07/2023] Open
Abstract
Long noncoding RNA (lncRNA) CBR3-AS1 (termed as CBR3-AS1) has been reported to be upregulated in several cancers including osteosarcoma. Its positive impact on the proliferation, migration, and invasion of osteosarcoma cells has been unveiled; nevertheless, whether it also affects the stemness and epithelial-mesenchymal transition (EMT) of osteosarcoma cells is unclear. The purpose for this study was to explore the effects of CBR3-AS1 on the stemness and EMT of osteosarcoma cells as well as its underlying mechanism. qRT-PCR and western blot were applied to detect target gene expression. Function assays were conducted to evaluate the effect of genes on the stemness and EMT of osteosarcoma cells. Mechanism assays were done to verify the association among different genes. In vivo assays were also performed. The obtained data showed that CBR3-AS1 demonstrated a high expression in osteosarcoma cells. CBR3-AS1 could promote stemness and EMT of osteosarcoma cells as well as osteosarcoma tumor growth. Mechanically, CBR3-AS1 sponged miR-140-5p and recruited DDX54 to upregulate NUCKS1, thus activating the mTOR signaling pathway. Furthermore, NUCKS1 could facilitate stemness and EMT of osteosarcoma cells. In summary, this study reveals that CBR3-AS1 exerts an oncogenic role in osteosarcoma through modulating the network of the miR-140-5p/DDX54-NUCKS1-mTOR signaling pathway.
Collapse
Affiliation(s)
- Weitao Yao
- Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), 127 Dongming Road, Zhengzhou City, Henan Province, 450000, China
| | - Jingyu Hou
- Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), 127 Dongming Road, Zhengzhou City, Henan Province, 450000, China
| | - Guoqing Liu
- Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), 127 Dongming Road, Zhengzhou City, Henan Province, 450000, China
| | - Fangxing Wu
- Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), 127 Dongming Road, Zhengzhou City, Henan Province, 450000, China
| | - Qiang Yan
- Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), 127 Dongming Road, Zhengzhou City, Henan Province, 450000, China
| | - Liangyu Guo
- Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), 127 Dongming Road, Zhengzhou City, Henan Province, 450000, China
| | - Chuchu Wang
- School of Life Science, Zhengzhou University, 100 Science Avenue, Zhengzhou City, Henan Province, 450001, China
| |
Collapse
|
97
|
Feleke M, Feng W, Rothzerg E, Song D, Wei Q, Kõks S, Wood D, Liu Y, Xu J. Single-cell RNA-seq identification of four differentially expressed survival-related genes by a TARGET: Osteosarcoma database analysis. Exp Biol Med (Maywood) 2022; 247:921-930. [PMID: 35285281 PMCID: PMC9189571 DOI: 10.1177/15353702221080131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/23/2022] [Indexed: 09/05/2023] Open
Abstract
Osteosarcoma (OS) differentially expressed genes (DEGs) have been predicted using the data portal of the Therapeutically Applicable Research to Generate Effective Treatments (TARGET). In this study, we sought to identify cell types that specially express key DEGs (MUC1, COL13A1, JAG2, and KAZALD1) in each of the nine identified cell populations derived from tissues of OS tumors with single-cell RNA-sequencing data. Gene expression levels were pairwise compared between cell clusters and a p value < 0.05 was considered differentially expressed. It was revealed that MUC1 is expressed at high levels in osteoblastic OS cells followed by carcinoma-associated fibroblasts (CAFs) and plasmocytes, respectively. COL13A1 is highly expressed in osteoblastic OS cells, CAFs, and endothelial cells (ECs), respectively. The KAZALD1 gene is expressed in CAFs and osteoblastic OS cells at high levels, but at very low levels in plasmocytes, osteoclasts, NK/T, myeloid cells 1, myeloid cells 2, ECs, and B cells. JAG2 is expressed at significantly high levels in ECs and osteoblastic OS cells, and at relatively lower levels in all other cell types. Interestingly, LSAMP, as an established gene in the development of OS shows high expression in osteoblastic OS cells and CAFs but low in other cells such as osteoclasts. Our findings here highlight the heterogeneity of OS cells and cell-type-dependent DEGs which have potential as therapeutic targets in OS.
Collapse
Affiliation(s)
- Mesalie Feleke
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Wenyu Feng
- Department of Orthopaedics, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Emel Rothzerg
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Dezhi Song
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning 530021, China
| | - Qingjun Wei
- Department of Orthopaedics, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia
| | - David Wood
- Medical School, The University of Western Australia, Perth, WA 6009, Australia
| | - Yun Liu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Department of Orthopaedics, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
98
|
Wu R, Dou X, Li H, Sun Z, Li H, Shen Y, Weng W, Min J. Identification of Cell Subpopulations and Interactive Signaling Pathways From a Single-Cell RNA Sequencing Dataset in Osteosarcoma: A Comprehensive Bioinformatics Analysis. Front Oncol 2022; 12:853979. [PMID: 35515114 PMCID: PMC9066489 DOI: 10.3389/fonc.2022.853979] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/03/2022] [Indexed: 11/21/2022] Open
Abstract
Osteosarcoma is a type of highly aggressive bone tumor arising from primitive cells of mesenchymal origin in adults and is associated with a high rate of tumor relapse. However, there is an urgent need to clarify the molecular mechanisms underlying osteosarcoma development. The present study performed integrated bioinformatics analysis in a single-cell RNA sequencing dataset and explored the potential interactive signaling pathways associated with osteosarcoma development. Single-cell transcriptomic analysis of osteosarcoma tissues was performed by using the Seurat R package, the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of differentially expressed genes was performed by using the clusterProfiler R package, and the cell–cell interaction analysis was performed by using the CellPhoneDB package. Our results showed that 11 clustered cell types were identified across 11 osteosarcoma tissues, with cell types including “osteoblastic”, “myeloid”, “osteoblastic_proli”, “osteoclast”, and “tumor-infiltrating lymphocytes (TILs)” as the main types. The DEGs between different cell types from primary, metastatic, and recurrent osteosarcomas were mainly enriched in the GO terms including “negative regulation of hydrolase activity”, “regulation of peptidase activity”, “regulation of binding”, “negative regulation of proteolysis”, and “negative regulation of peptidase activity” and in the KEGG pathways including “transcriptional misregulation in cancer”, “cellular senescence”, “apoptosis”, “FoxO signaling pathway”, “cell cycle”, “NF-kappa B signaling pathway”, “p53 signaling pathway”, “pentose phosphate pathway”, and “protein export”. For the cell–cell communication network analysis, the different interaction profiles between cell types were detected among primary, metastatic, and recurrent osteosarcomas. Further exploration of the KEGG pathway revealed that these ligand/receptor interactions may be associated with the NF-κB signaling pathway and its interacted mediators. In conclusion, the present study for the first time explored the scRNA-seq dataset in osteosarcoma, and our results revealed the 11 clustered cell types and demonstrated the novel cell–cell interactions among different cell types in primary, metastatic, and recurrent osteosarcomas. The NF-κB signaling pathway may play a key role in regulating the TME of osteosarcoma. The present study may provide new insights into understanding the molecular mechanisms of osteosarcoma pathophysiology.
Collapse
Affiliation(s)
- Rong Wu
- Department of Orthopaedics, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Xiaojie Dou
- Department of Orthopaedics, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Haidong Li
- Department of Orthopaedics, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Zhenguo Sun
- Department of Orthopaedics, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Heng Li
- Department of Orthopaedics, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Yuxin Shen
- Department of Orthopaedics, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Wei Weng
- Department of Orthopaedics, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Jikang Min
- Department of Orthopaedics, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou University, Huzhou, China
| |
Collapse
|
99
|
Molecular Pathways Involved in the Anti-Cancer Activity of Flavonols: A Focus on Myricetin and Kaempferol. Int J Mol Sci 2022; 23:ijms23084411. [PMID: 35457229 PMCID: PMC9026553 DOI: 10.3390/ijms23084411] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/04/2022] [Accepted: 04/14/2022] [Indexed: 12/22/2022] Open
Abstract
Natural compounds have always represented valuable allies in the battle against several illnesses, particularly cancer. In this field, flavonoids are known to modulate a wide panel of mechanisms involved in tumorigenesis, thus rendering them worthy candidates for both cancer prevention and treatment. In particular, it was reported that flavonoids regulate apoptosis, as well as hamper migration and proliferation, crucial events for the progression of cancer. In this review, we collect recent evidence concerning the anti-cancer properties of the flavonols myricetin and kaempferol, discussing their mechanisms of action to give a thorough overview of their noteworthy capabilities, which are comparable to those of their most famous analogue, namely quercetin. On the whole, these flavonols possess great potential, and hence further study is highly advised to allow a proper definition of their pharmaco-toxicological profile and assess their potential use in protocols of chemoprevention and adjuvant therapies.
Collapse
|
100
|
Sulfated alginate oligosaccharide exerts antitumor activity and autophagy induction by inactivating MEK1/ERK/mTOR signaling in a KSR1-dependent manner in osteosarcoma. Oncogenesis 2022; 11:16. [PMID: 35418575 PMCID: PMC9008062 DOI: 10.1038/s41389-022-00390-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/12/2022] [Accepted: 03/18/2022] [Indexed: 11/08/2022] Open
Abstract
Alginate oligosaccharide (AOS) has the function to inhibit tumor progression and the sulfated modification can enhance the antitumor activity. To date, the function and mechanism of sulfated AOS (AOS-SO4) in tumors remain largely elusive. We prepared AOS by the enzymatic degradation of alginate, collected AOS-SO4 by sulfating following the canonical procedure. Using these materials, in vitro assays showed that both AOS and AOS-SO4 elicited antitumor effects in osteosarcoma cells. Sulfated modification significantly enhanced the antitumor activity. In addition, AOS-SO4 had obvious effects on cell cycle arrest, apoptosis, and autophagy induction in vitro and in vivo. Mechanistically, we observed that AOS-SO4 treatment triggered proapoptotic autophagy by inhibiting MEK1/ERK/mTOR signaling. The ERK activator reversed AOS-SO4-induced autophagy. More importantly, we found that KSR1 interacted with MEK1 and functioned as a positive regulator of MEK1 protein in osteosarcoma cells. High KSR1 expression was significantly associated with poor survival in osteosarcoma patients. Together, these results suggest that AOS-SO4 has a better antitumor effect in osteosarcoma by inhibiting MEK1/ERK/mTOR signaling, which is KSR1-dependent; thus, AOS-SO4 can be a new potential therapeutic candidate for the treatment of osteosarcoma.
Collapse
|