Fig 1 - available via license: CC BY
Content may be subject to copyright.
PARK2 is downregulated in osteosarcoma tissues and cell lines, and PARK2 overexpression models are constructed. There are two representative cases comparing OS tissues and adjacent non-tumor tissues detected by immunohistochemistry (a). The expression of PARK2 mRNA in SAOS2, HOS, U2OS, and hFOB1.19 cells (b). The expression level of the protein Parkin was significantly enhanced in HOS and U2OS cell lines after transfection was detected using western blot assays (c) and immunofluorescence assays (d). Magnification, ×200 (a), ×1000 (d). Scale bar, 50 μm (a, d). **P < 0.01, ***P < 0.001 

PARK2 is downregulated in osteosarcoma tissues and cell lines, and PARK2 overexpression models are constructed. There are two representative cases comparing OS tissues and adjacent non-tumor tissues detected by immunohistochemistry (a). The expression of PARK2 mRNA in SAOS2, HOS, U2OS, and hFOB1.19 cells (b). The expression level of the protein Parkin was significantly enhanced in HOS and U2OS cell lines after transfection was detected using western blot assays (c) and immunofluorescence assays (d). Magnification, ×200 (a), ×1000 (d). Scale bar, 50 μm (a, d). **P < 0.01, ***P < 0.001 

Source publication
Article
Full-text available
Osteosarcoma (OS) is the most common primary malignant bone tumor mainly occurring in children and adolescents. In past decades, studies revealed that PARK2 was a vital tumor suppressor gene in many malignant solid tumors. However, the role of PARK2 in OS remains largely unclear. Therefore, we assessed PARK2 expression in OS tissue and adjacent non...

Contexts in source publication

Context 1
... evaluate the role played by PARK2 in OS develop- ment, 46 primary OS tissues and their adjacent non- tumor tissues were studied using PARK2 IHC (Fig. 1a). The results showed that 76% (35/46) of the adjacent non- tumor tissues and 37% (17/46) of the OS tissues expressed the PARK2 protein (P < 0.05). Moreover, the expression of PARK2 was significantly associated with higher tumor stage (P < 0.05, Table 1). Next, we examined PARK2 mRNA expression by real-time PCR in the human OS cell lines ...
Context 2
... < 0.05). Moreover, the expression of PARK2 was significantly associated with higher tumor stage (P < 0.05, Table 1). Next, we examined PARK2 mRNA expression by real-time PCR in the human OS cell lines SAOS2, HOS, and U2OS, and in the normal human osteoblastic cell line hFOB1.19. The level of PARK2 mRNA decreased significantly in the OS cell lines (Fig. 1b). Collectively, these results demonstrate that PARK2 is downregulated in human OS tissues and cells, and it is associated with OS progression, which might play a role in suppressing human OS ...
Context 3
... shows low expression levels in the HOS and U2OS cell lines, which were selected for overexpression models. Transfection rates of PARK2 gene overexpression group (HOS-PARK2 and U2OS-PARK2) and negative control group (HOS-NC and U2OS-NC) were close to 90%, which were further confirmed by western blot and immunofluorescence assay (Fig. 1c, d). The stably trans- fected cells were used to investigate biological functions and potential mechanisms in OS. Cell viability (Fig. 2a) and colony formation assays (Fig. 2b) showed that the PARK2 group significantly inhibited cell growth relative to that in the NC group (P < 0.05). Ki67 staining revealed cell proliferation rates in ...
Context 4
... ± 121.8 mm 3 vs. 1,905 ± 390.8 mm 3 and the tumor weights were 1.552 ± 0.088 g vs. 2.058 ± 0.201 g in the PARK2 and NC groups, respectively (P < 0.05, Fig. 4b, c). Tumor specimens were used for further pathological analysis. IHC detection showed that the PARK2 protein level in the PARK2 group was dramati- cally higher than that in the NC group (Fig. 4c1, ...
Context 5
... and eosin (H&E) staining revealed that the PARK2 group attained fewer hemocytes and vascular structures in the Codman-triangle area than those in the NC group (Fig. 4d3-6). Masson staining showed that the knee joint structure of the PARK2 group was intact; the joint cavity exhibited no obvious narrowing, and the articular surface was smooth ( Fig. 4d7-10). However, in the NC group, the OS cells invaded the articular cavity, the articular cavity narrowed, and the articular cartilage of the tibia was severely damaged (Fig. 4d10). The western blot assay revealed that the PARK2 gene was negatively associated with the expression level of the protein VEGF in the tibial xenografts (P < 0.05; ...
Context 6
... Masson staining showed that the knee joint structure of the PARK2 group was intact; the joint cavity exhibited no obvious narrowing, and the articular surface was smooth ( Fig. 4d7-10). However, in the NC group, the OS cells invaded the articular cavity, the articular cavity narrowed, and the articular cartilage of the tibia was severely damaged (Fig. 4d10). The western blot assay revealed that the PARK2 gene was negatively associated with the expression level of the protein VEGF in the tibial xenografts (P < 0.05; Fig. 4e). These data were detected by an immunofluorescence assay and flow cytometry, respectively. Magnification, ×400 (c). Scale bar, 50 μm (c). *P < 0.05, **P < 0.01.Each ...

Citations

... Guo et al., 2019;Long et al., 2023). Similarly, PARK2 is recognized as a human tumor suppressor gene (Duan et al., 2019;Lei et al., 2018) and is also linked to the development of Parkinson's disease through mutations (Zilocchi et al., 2020). In the context of the LVK breed, it is conceivable that mutations within TANC2, PARK2, and EPHA10 genes could be associated with the domestication process of the goats. ...
... Western blotting studies showed that REC8 can effectively down-regulate the expression of p-STAT3 and VEGF in SH-SY5Y and SK-N-AS cell lines. It has been reported that inhibition of the STAT3 pathway decreases VEGF expression and plays a significant role in NB [20] and in the genesis and progression of numerous other tumors [21,22]. In addition, STAT3 was thought to be the primary transcription factor for the VEGF promoter [23]. ...
Article
Full-text available
Background Neuroblastoma, one of the most prevalent childhood cancers, is often treated with surgery, radiation, and chemotherapy. However, prognosis and survival are still dismal for children with neuroblastoma at high risk. Consequently, it is vital to identify new and effective treatment targets. As a component of the meiotic cohesion complex, REC8 is involved in a wide range of malignancies. The current work assessed the impact of REC8 knockdown on SH-SY5Y and SK-N-AS neuroblastoma cells and delved into the molecular mechanism behind this effect. Methods Knockdown of REC8 using the small interfering (si) RNA technology, and the results were verified by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and western blot. The Cell Counting Kit-8 (CCK-8) was used to examine cell proliferation, while flow cytometry was used to examine cell cycle progression and apoptosis. Analyses of angiogenesis included tube formation experiments. Transwell tests were used to examine cell migration and invasion. Results The data showed that downregulation of the REC8 led to a substantial decrease in cell proliferation by stopping the cell cycle in the G1 phase. REC8 knockdown significantly reduced neuroblastoma cell proliferation, migration, invasion, angiogenesis, induced cell cycle arrest, and enhanced apoptosis. We also discovered that repressing REC8 expression in neuroblastoma cell lines SH-SY5Y and SK-N-AS reduced their ability to activate the STAT3/VEGF signaling pathway. Conclusions Neuroblastoma therapy may benefit from targeting REC8 and its downstream targets.
... Abnormally activated JAK/STAT signaling has been reported to contribute to the development of multiple cancers. The JAK2/ STAT3 pathway was stimulated to participate in the migration, proliferation, and invasion of osteosarcoma cells induced by PARK2 [40]. REX1 overexpression plays a catalytic role in the progression of cervical cancer by upregulating JAK2/STAT3 activity [41]. ...
Article
Full-text available
Liver cancer stem cells (LCSCs) contribute to tumor recurrence and cancer cell proliferation in patients with hepatocellular carcinoma (HCC). METTL3-catalyzed m⁶A modification is relevant to the cancer stem cell (CSC) phenotype, including LCSCs. LCSCs were isolated from MHCC-97H and HepG2 cells through flow cytometry. UALCAN data were used to analyze the expression of METTL3 in liver hepatocellular carcinoma (LIHC) tissues. Loss- and gain-of-function experiments were utilized to assess the biological effects of METTL3 and SOCS3 on the proliferation and stemness phenotypes in vitro and in vivo. The mechanisms underlying the impact of METTL3 were explored using qPCR, MeRIP-qPCR, dual-luciferase reporter, and western blot assays. METTL3 was significantly upregulated in LIHC tissues according to the UALCAN database. METTL3 was highly expressed in LIHC and was significantly correlated with individual cancer stage, tumor grade and lymph node metastasis. Patients with low METTL3 expression had a longer overall survival time based on the data from UALCAN. In addition, the level of METTL3 was enhanced in LCSCs and decreased in non-LCSCs compared to HCC cells. Moreover, overexpression of METTL3 stimulated the proliferation and stemness of LCSCs in vitro and in vivo, while loss of METTL3 impeded it. Bioinformatics analysis combined with validation experiments determined that m⁶A was modified by METTL3-targeting SOCS3 mRNA. METTL3 had side effects regarding the stability of SOCS3 mRNA. SOCS3 overexpression impaired and SOCS3 depletion facilitated the development of LCSCs via the JAK2/STAT3 pathway. Furthermore, METTL3 depletion suppressed proliferation and stemness in LCSCs, which was restored by SOCS3 knockdown or colivelin treatment. We discovered that METTL3 facilitated the stemness and tumorigenicity of LCSCs by modifying SOCS3 mRNA with m⁶A.
... High parkin levels are expected to play a protective role when evaluating tumor growth and evolution. Parkin exhibits a protective function by accumulating in neoplasic cells and degrading excess cyclin E1, which inhibits tumor cell replication [25][26][27][28]. However, the opposite mechanism was also attributed to the presence of high levels of parkin. ...
Article
Pediatric neoplasms represent an important group of childhood diseases. Biomarkers with prognostic function can help to manage this complex process. In this context, the tissue expression of parkin, could be used as a prognostic biomarker for the individual in the main solid pediatric tumors. We aimed to investigate the correlation between the tissue expression of parkin and the clinical-pathological characteristics, and to determine if parkin can be used as a prognostic marker. We assessed immune histochemical analysis of parkin in five solid pediatric tumors. High tissue expression of parkin was associated with positive prognostic factors for astrocytoma's and nephronblastomas, while in medulloblastomas and neuroblastomas; the same underlying aspect was associated with poor prognostic factors. Choroid plexus tumors showed no association. Parkin showed favorable behavior in patients with in astrocytoma's and nephroblastomas. In medulloblastomas and neuroblastomas, results showed the opposite. Research may enable an analysis of the overall behavior of this molecule as a prognostic tool.
... Although PINK1 is not involved superoxide production in BAECs under acute hypoxia, whether PINK1 promotes ECs angiogenesis is unknown, and the potential mechanism remains to be further studied [30]. Unlike PINK1, the downstream Parkin has been reported to inhibit tumor angiogenesis through ubiquitination degradation of HIF-1α at lysine 477 [145], and inhibiting JAK2/STAT3/VEGF pathways in osteosarcoma [146]. In mouse aortic ECs (MAECs), overexpression of Parkin decreased eNOS expression and induced mitochondrial dysfunction by ubiquitination of estrogen-related receptor α (ERRα), independent of autophagy and apoptosis, indicating activation of parkin in ECs might be detrimental in ECs [147]. ...
Article
Full-text available
Endothelial cells (ECs) angiogenesis is the process of sprouting new vessels from the existing ones, playing critical roles in physiological and pathological processes such as wound healing, placentation, ischemia/reperfusion, cardiovascular disease and cancer metastasis. Although mitochondria are not the major sites of energy source in ECs, they function as important biosynthetic and signaling hubs to regulate ECs metabolism and adaptations to local environment, thus affecting ECs migration, proliferation and angiogenic process. The understanding of the importance and potential mechanisms of mitochondrial proteins in regulating ECs metabolism, function and the process of angiogenesis has developed in the past decades. Thus, in this review, we discuss the current understanding of mitochondrial proteins in ECs metabolism, function and angiogeneic signaling, to provide new and therapeutic targets for treatment of diverse cardiovascular and angiogenesis-dependent diseases.
... Thereafter, cells were collected and resuspended in an equal volume of PBS. The cells were incubated with APC annexin V and 7-AAD (LK-AP105-100; Multisciences, Hangzhou, China), and the percentage of apoptotic cells was determined by flow cytometry (Beckman Coulter, USA) [43,44]. ...
Article
Full-text available
Background and aims Chimeric antigen receptor (CAR)-T cell therapy is a novel type of immunotherapy. However, the use of CAR-T cells to treat acute myeloid leukaemia (AML) has limitations. B7-H3 is expressed in several malignancies, including some types of AML cells. However, its expression in normal tissues is low. Therefore, B7-H3 is ideal for targeted AML therapy. Materials and methods First, we constructed B7-H3 CAR that can target B7-H3, and then constructed B7-H3-CAR-T cells in vitro, which were co-incubated with six AML cell lines expressing different levels of B7-H3, respectively. The toxicity and cytokines were detected by flow cytometry. In vivo, AML model was established in B-NSG mice to study the toxicity of B7-H3-CAR T on AML cells. Results In vitro functional tests showed that B7-H3-CAR-T cells were cytotoxic to B7-H3-positive AML tumor cells and had good scavenging effect on B7-H3-expressing AML cell lines, and the cytokine results were consistent. In vivo, B7-H3-CAR-T cells significantly inhibited tumor cell growth in a mouse model of AML, prolonging mouse survival compared with controls. Conclusion B7-H3-CAR-T cells may serve as a novel therapeutic method for the targeted treatment of AML.
... The details of all signaling pathway networks identified were also shown in Figures types; C: chordal graph of ligand-receptor interactions among all cell types). Among the total of 57 signaling pathways, the following signaling pathways were related to osteosarcoma: COLLAGEN (Baumann and Hennet, 2016;Elenjord et al., 2009;Levinson et al., 2002;Yamaguchi et al., 2005), CD99 (Manara et al., 2006;Sciandra et al., 2014;Zucchini et al., 2014), PTN (He et al., 2019;Qin et al., 2022;Sun et al., 2020), MIF (Liu et al., 2014), SPP1(Dalla-Torre et al., 2006;Li et al., 2017), FN1 (Saba et al., 2019;Zhou et al., 2019), LAMININ (Heino and Massague, 1989), FGF (Kurogi et al., 1996;Laulederkind et al., 2000;Li et al., 2019;Xu et al., 2010), VEGF (Ji et al., 2020;Lei et al., 2018;Oda et al., 2006;Tsai et al., 2017;, Frontiers in Genetics frontiersin.org 08 2019), GALECTIN (Gomez-Brouchet et al., 2010;Miao et al., 2014;Park et al., 2015;Zhou et al., 2014), PERIOSTIN Xu et al., 2022), VISFATIN Wang et al., 2019Wang et al., , 2016, ITGB2 (Dai et al., 2018), NOTCH (Jin et al., 2017;Mu et al., 2013;Ongaro et al., 2016;Tanaka et al., 2009;Zhang et al., 2010), IGF (Armakolas et al., 2016;Giatagana et al., 2022; , VWF (Wang et al., 2020), and PDGF (Chen et al., 2009;Egners et al., 2018;Heldin et al., 1986). ...
Article
Full-text available
Osteosarcoma (OS) is a common bone cancer in children and adolescents, and metastasis and recurrence are the major causes of poor treatment outcomes. A better understanding of the tumor microenvironment is required to develop an effective treatment for OS. In this paper, a single-cell RNA sequencing dataset was taken to a systematic genetic analysis, and potential signaling pathways linked with osteosarcoma development were explored. Our findings revealed 25 clusters across 11 osteosarcoma tissues, with 11 cell types including “Chondroblastic cells”, “Osteoblastic cells”, “Myeloid cells”, “Pericytes”, “Fibroblasts”, “Proliferating osteoblastic cells”, “Osteoclasts”, “TILs”, “Endothelial cells”, “Mesenchymal stem cells”, and “Myoblasts”. The results of Cell communication analysis showed 17 potential cellular communication networks including “COLLAGEN signaling pathway network”, “CD99 signaling pathway network”, “PTN signaling pathway network”, “MIF signaling pathway network”, “SPP1 signaling pathway network”, “FN1 signaling pathway network”, “LAMININ signaling pathway network”, “FGF signaling pathway network”, “VEGF signaling pathway network”, “GALECTIN signaling pathway network”, “PERIOSTIN signaling pathway network”, “VISFATIN signaling pathway network”, “ITGB2 signaling pathway network”, “NOTCH signaling pathway network”, “IGF signaling pathway network”, “VWF signaling pathway network”, “PDGF signaling pathway network”. This research may provide novel insights into the pathophysiology of OS’s molecular processes.
... In our research, the results showed miR-375 regulated the expression of JAK2 via repressing its translation in RMECs. STAT3 frequently acts as an important downstream target of JAK2, and the JAK2/STAT3 pathway plays important roles in regulating several physiological processes [24,25]. Inhibition of the JAK2/STAT3 pathway induced by ANGPTL1 represses angiogenesis and metastasis in hepatocellular carcinoma [26]. ...
... Inhibition of the JAK2/STAT3 pathway induced by ANGPTL1 represses angiogenesis and metastasis in hepatocellular carcinoma [26]. VEGF, the critical regulatory factor in angiogenesis, is a transcriptional target of the JAK2/STAT3 pathway [25,27]. The present study proved that the inhibitory roles of miR-375 in proliferation and migration were abolished by the restoration of JAK2 expression in RMECs. ...
Article
Full-text available
Aberrant neovascularization in the retina is an important threat to vision and closely related to several retinal diseases, such as wet form of age-related macular degeneration, diabetic retinopathy, and retinopathy of prematurity. However, the pathogenesis remains largely unknown. MicroRNAs (miRNAs) have been demonstrated to play critical regulatory roles in angiogenesis. Therefore, we aimed to identify the key miRNAs that regulate retinal neovascularization and elucidate the potential underlying mechanisms. In the present study, we performed RNA sequencing of microRNAs in the retina and found that miR-375 was significantly downregulated in the retina of oxygen-induced retinopathy mice. In retinal microvascular endothelial cells (RMECs), overexpression of miR-375 inhibited cell proliferation and angiogenesis. Conversely, inhibition of miR-375 had the opposite effects. Moreover, our results showed that miR-375 negatively regulated the protein expression of JAK2 by inhibiting its translation. The promoting effects of anti-miR-375 on cell proliferation and angiogenesis were attenuated by an inhibitor of STAT3. These results indicate that miR-375 mitigates cell proliferation and angiogenesis, at least in part, through the JAK2/STAT3 pathway in RMECs, which implies an important underlying mechanism of retinal angiogenesis and provides potential therapeutic targets for retinal microangiopathy.
... Tumor-derived angiogenic factors were regulated by various signaling pathways, such as STAT3, ERK, AKT, and NF-κB pathways [17,[27][28][29]. Previous reports have shown that PELP1 regulate the phosphorylation of STAT3 (p-STAT3), and STAT3 regulate the expression of VEGFA to affect the angiogenesis of tumor [30,31]. ...
... Abnormal activation of STAT3 regulates the STAT3/VEGF signaling pathway and promotes VEGF expression [34][35] . VEGF expression can be down-regulated by inhibiting the JAK2/STAT3 pathway [36] . The well-established JAK/STAT pathway inhibitor AG-490, also known as Tyrphostin AG-490, was used in our study to specifically block the phosphorylation and activation of JAK2 and STAT3 [37][38] . ...
Article
Full-text available
Aim: To investigate the effect of leptin on the angiogenesis of RF/6A cells (monkey retinal choroidal endothelial cells) in vitro and test the cellular signaling in the mechanism. Methods: RF/6A cells were cultured in vitro and randomly divided into four groups: normal control, with leptin at 50, 100, 200 ng/mL for cell counting kit-8 (CCK8). RF/6A cell proliferation and migration were examined by Transwell assays, while RF/6A cell tube formation by Matrigel assay. JAK2, p-JAK2, STAT3, and p-STAT3 protein expression was measured by Western blotting. Cells were then divided into the following treatment groups: control, 100 ng/mL leptin and AG-490 (100 ng/mL leptin+10 µmol/L AG-490) for examinations of RF/6A cellular behaviour again. Analysis of differences was carried out using one-way ANOVA and least significant difference (LSD). Results: RF/6A cell proliferation, migration and cell tube formation were promoted significantly by leptin in a dose-dependent manner (P<0.05). Western blotting showed that leptin up-regulated p-JAK2 and p-STAT3 expression levels. Treatment with the JAK/STAT pathway inhibitor, AG-490, decreased leptin-induced p-JAK2 and p-STAT3 expression, and inhibited cell proliferation, migration and cell tube formation induced by leptin (P<0.05). Conclusion: Leptin can promote RF/6A cell angiogenesis in vitro via activation of the JAK2/STAT3 signaling pathway.