Human CellPub Date : 2022-11-01Epub Date: 2022-09-14DOI: 10.1007/s13577-022-00775-5
Borong Zhou, Zhongchao Mai, Ying Ye, Yanan Song, Miao Zhang, Xinlin Yang, Wei Xia, Xiaofeng Qiu
{"title":"The role of PYCR1 in inhibiting 5-fluorouracil-induced ferroptosis and apoptosis through SLC25A10 in colorectal cancer.","authors":"Borong Zhou, Zhongchao Mai, Ying Ye, Yanan Song, Miao Zhang, Xinlin Yang, Wei Xia, Xiaofeng Qiu","doi":"10.1007/s13577-022-00775-5","DOIUrl":"10.1007/s13577-022-00775-5","url":null,"abstract":"<p><p>Although PYCR1 is a well-recognized oncogenic gene for malignant tumors, the causal relationship of its expression with malignant growth and cytotoxic chemotherapeutics remains unclear. Therefore, this study aimed to clarify the role of PYCR1 and its interaction with SLC25A10 in a chemotherapeutic agent 5-fluorouracil (5-FU)'s toxicity to colorectal cancer cells. PYCR1 and SLC25A10 expressions were detected in The Cancer Genome Atlas database and colon adenocarcinoma (COAD) clinical samples. PYCR1 upregulation was associated with SLC25A10 expression and poor prognosis, and its high expression indicated decreased survival rates in patients with COAD. PYCR1 overexpression inhibited lipid reactive oxygen species production and promoted SLC25A10 expression in colorectal cancer cells. PYCR1 silencing enhanced the antitumor effects of 5-FU. Ferroptosis inhibitor deferoxamine suppressed the antitumor effects of PYCR1 silencing, whereas ferroptosis inducer erastin inhibited the protumor effects of PYCR1 overexpression. SLC25A10 overexpression reversed the antitumor effects of PYCR1 silencing in vitro and inhibited the antitumor effects of erastin in vivo. Therefore, PYCR1 is an oncogenic gene that promotes colorectal tumor growth and desensitizes colorectal cancer cells to 5-FU cytotoxicity by preventing apoptosis and ferroptosis.</p>","PeriodicalId":13228,"journal":{"name":"Human Cell","volume":"35 6","pages":"1900-1911"},"PeriodicalIF":4.3,"publicationDate":"2022-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40357103","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Human CellPub Date : 2022-09-01Epub Date: 2022-07-07DOI: 10.1007/s13577-022-00739-9
Jin Song, Xiaolong Xu, Shasha He, Ning Wang, Yunjing Bai, Bo Li, Shengsheng Zhang
{"title":"Exosomal hsa_circ_0017252 attenuates the development of gastric cancer via inhibiting macrophage M2 polarization.","authors":"Jin Song, Xiaolong Xu, Shasha He, Ning Wang, Yunjing Bai, Bo Li, Shengsheng Zhang","doi":"10.1007/s13577-022-00739-9","DOIUrl":"https://doi.org/10.1007/s13577-022-00739-9","url":null,"abstract":"<p><p>Gastric cancer (GC) is an aggressive malignant tumor of the digestive system, with high morbidity rates. We previously demonstrated that miR-17-5p can modify tumorigenesis in GC. In addition, other studies have shown that circRNAs can regulate GC progression by sponging various miRNAs. However, the association between circRNAs and miR-17-5p in GC has not yet been explored. Hence, this study aimed to explore the possible interactions between various circRNAs and miR-17-5p using a dual-luciferase assay. CCK-8 was used to determine cell viability, and a Transwell assay was used to measure cell invasion and migration. Gene expression was assessed using quantitative reverse transcription PCR (RT-qPCR), and exosomes were identified using transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). Annexin V/PI staining was also used to detect cell apoptosis. These investigations collectively revealed that miR-17-5p is a target of the circRNA hsa_circ_0017252 and hsa_circ_0017252 is significantly downregulated in GC tissues. In addition, the overexpression of hsa_circ_0017252 inhibited GC cell migration by sponging of miR-17-5p, and GC cell-secreted exosomal hsa_circ_0017252 effectively inhibited macrophage M2-like polarization, which in turn suppressed GC cell invasion. Notably, exosomes containing hsa_circ_0017252 also suppressed GC tumor growth in vivo. Thus, our data suggest that the overexpression of hsa_circ_0017252 suppresses GC malignancy by sponging miR-17-5p. In addition, exosomal hsa_circ_0017252 excreted from GC cells attenuated GC progression by suppressing macrophage M2-like polarization. These findings improve our basic understanding of GC and open a novel avenue for developing more effective GC treatments.</p>","PeriodicalId":13228,"journal":{"name":"Human Cell","volume":"35 5","pages":"1499-1511"},"PeriodicalIF":4.3,"publicationDate":"2022-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40478089","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Human CellPub Date : 2022-09-01Epub Date: 2022-07-06DOI: 10.1007/s13577-022-00744-y
Sophie Rae, Cathy Spillane, Gordon Blackshields, Stephen F Madden, Joanne Keenan, Britta Stordal
{"title":"The EMT-activator ZEB1 is unrelated to platinum drug resistance in ovarian cancer but is predictive of survival.","authors":"Sophie Rae, Cathy Spillane, Gordon Blackshields, Stephen F Madden, Joanne Keenan, Britta Stordal","doi":"10.1007/s13577-022-00744-y","DOIUrl":"https://doi.org/10.1007/s13577-022-00744-y","url":null,"abstract":"<p><p>The IGROVCDDP cisplatin-resistant ovarian cancer cell line is an unusual model, as it is also cross-resistant to paclitaxel. IGROVCDDP, therefore, models the resistance phenotype of serous ovarian cancer patients who have failed frontline platinum/taxane chemotherapy. IGROVCDDP has also undergone epithelial-mesenchymal transition (EMT). We aim to determine if alterations in EMT-related genes are related to or independent from the drug-resistance phenotypes. EMT gene and protein markers, invasion, motility and morphology were investigated in IGROVCDDP and its parent drug-sensitive cell line IGROV-1. ZEB1 was investigated by qPCR, Western blotting and siRNA knockdown. ZEB1 was also investigated in publicly available ovarian cancer gene-expression datasets. IGROVCDDP cells have decreased protein levels of epithelial marker E-cadherin (6.18-fold, p = 1.58e-04) and higher levels of mesenchymal markers vimentin (2.47-fold, p = 4.43e-03), N-cadherin (4.35-fold, p = 4.76e-03) and ZEB1 (3.43-fold, p = 0.04). IGROVCDDP have a spindle-like morphology consistent with EMT. Knockdown of ZEB1 in IGROVCDDP does not lead to cisplatin sensitivity but shows a reversal of EMT-gene signalling and an increase in cell circularity. High ZEB1 gene expression (HR = 1.31, n = 2051, p = 1.31e-05) is a marker of poor overall survival in high-grade serous ovarian-cancer patients. In contrast, ZEB1 is not predictive of overall survival in high-grade serous ovarian-cancer patients known to be treated with platinum chemotherapy. The increased expression of ZEB1 in IGROVCDDP appears to be independent of the drug-resistance phenotypes. ZEB1 has the potential to be used as biomarker of overall prognosis in ovarian-cancer patients but not of platinum/taxane chemoresistance.</p>","PeriodicalId":13228,"journal":{"name":"Human Cell","volume":"35 5","pages":"1547-1559"},"PeriodicalIF":4.3,"publicationDate":"2022-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9374625/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40565138","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Transcription factor p8 regulates autophagy in response to disulfiram via PI3K/mTOR/p70S6K signaling pathway in pancreatic cancer cells.","authors":"Zhangyu Yao, Xiang Li, Jun Gao, Yutao Wang, Linmei Xiao, Xinxia Chang, Fangzhou Liu, Zhenqing Feng, Xiao Zhang","doi":"10.1007/s13577-022-00731-3","DOIUrl":"10.1007/s13577-022-00731-3","url":null,"abstract":"<p><p>Disulfiram (DSF), which is an inhibitor of aldehyde dehydrogenase (ALDH) and approved by the FDA for the treatment of alcoholism previously, has been repurposed for use as a cancer treatment because of its potent effect in preclinical studies. In this study, we found that disulfiram forms potent complexes with copper (DSF/Cu) inhibited cell proliferation, induced apoptosis in human pancreatic cancer cells, which was detected by flow cytometry and western blotting. Meanwhile, autophagy and autophagic flux also clearly observed by transmission electron microscopy, confocal microscopy and flow cytometry. Our results also showed that DSF/Cu induced transcription factor p8 upregulation and PI3K/mTOR signaling pathway activation detected by real-time PCR and western blotting. Additionally, suppression of p8 inactivated the mTOR signaling pathway and autophagic flux maintained. Furthermore, mechanism study indicated that autophagy induced by DSF/Cu was regulated by p8 and was related to PI3K/mTOR/p70S6K signaling pathway in pancreatic cancer cells. Our findings provide insights into the role of p8 in regulating autophagy induced by DSF/Cu effects in pancreatic cancer cells.</p>","PeriodicalId":13228,"journal":{"name":"Human Cell","volume":"35 5","pages":"1464-1474"},"PeriodicalIF":4.3,"publicationDate":"2022-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40396067","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Human CellPub Date : 2022-09-01Epub Date: 2022-06-25DOI: 10.1007/s13577-022-00732-2
Yang Sun, Yikun Ju, Bairong Fang
{"title":"Exosomes from human adipose-derived mesenchymal stromal/stem cells accelerate angiogenesis in wound healing: implication of the EGR-1/lncRNA-SENCR/DKC1/VEGF-A axis.","authors":"Yang Sun, Yikun Ju, Bairong Fang","doi":"10.1007/s13577-022-00732-2","DOIUrl":"https://doi.org/10.1007/s13577-022-00732-2","url":null,"abstract":"<p><p>Exosomes (Exos) extracted from human adipose mesenchymal stromal/stem cells (hAD-MSCs) have been reported as therapeutic tools for tissue repair, but how they regulate angiogenesis of endothelial cells remains unknown. In this study, hAD-MSCs were isolated, and early growth response factor-1, Smooth muscle and endothelial cell enriched migration/differentiation-associated long-noncoding RNA (lncRNA-SENCR), and vascular endothelial growth factor-A (VEGF-A) overexpression or knockdown was achieved. Exos extracted from hAD-MSCs (hADSC-Exos) were co-cultured with human umbilical vein endothelial cells (HUVECs) to detect the effects of EGR-1, lncRNA-SENCR, and VEGF-A on angiogenesis and the relationships between EGR-1, lncRNA-SENCR, Dyskerin pseudouridine synthase 1 (DKC1), and VEGF-A. An in vivo experiment verified the effect of hADSC-Exos on the wound healing process. hADSC-Exos substantially promoted the proliferation, migration, and angiogenesis of HUVECs, which could be reversed by short-hairpin RNA SENCR (shSENCR) transfection. hADSC-Exos had elevated expression of EGR-1, which bound to the lncRNA-SENCR promoter. The suppressive effect of Exo-shEGR1 on HUVECs was counteracted by SENCR overexpression. LncRNA-SENCR was shown to interact with DKC1. Overexpression of DKC1 or lncRNA-SENCR maintained stable VEGF-A expression. Overexpression of VEGF-A reversed the suppressive effect of shSENCR on HUVECs. Consistent results were obtained in mice in vivo. Overall, hADSC-Exo EGR-1 upregulates lncRNA-SENCR expression to activate the DKC1/VEGF-A axis, facilitating the wound-healing process by increasing angiogenesis.</p>","PeriodicalId":13228,"journal":{"name":"Human Cell","volume":"35 5","pages":"1375-1390"},"PeriodicalIF":4.3,"publicationDate":"2022-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40396790","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Retraction Note to: Long non-coding RNA MIAT promotes the growth of melanoma via targeting miR-150.","authors":"Lifei Zhu, Yexiao Wang, Chaoying Yang, Yanchang Li, Zhixin Zheng, Liangcai Wu, Hui Zhou","doi":"10.1007/s13577-022-00742-0","DOIUrl":"https://doi.org/10.1007/s13577-022-00742-0","url":null,"abstract":"","PeriodicalId":13228,"journal":{"name":"Human Cell","volume":"35 5","pages":"1631"},"PeriodicalIF":4.3,"publicationDate":"2022-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40402269","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Human CellPub Date : 2022-09-01Epub Date: 2022-06-29DOI: 10.1007/s13577-022-00738-w
Yidan Chen, Ying Su, Feng Wang
{"title":"The Piezo1 ion channel in glaucoma: a new perspective on mechanical stress.","authors":"Yidan Chen, Ying Su, Feng Wang","doi":"10.1007/s13577-022-00738-w","DOIUrl":"https://doi.org/10.1007/s13577-022-00738-w","url":null,"abstract":"<p><p>Glaucomatous optic nerve damage caused by pathological intraocular pressure elevation is irreversible, and its course is often difficult to control. This group of eye diseases is closely related to biomechanics, and the correlation between glaucoma pathogenesis and mechanical stimulation has been studied in recent decades. The nonselective cation channel Piezo1, the most important known mechanical stress sensor, is a transmembrane protein widely expressed in various cell types. Piezo1 has been detected throughout the eye, and the close relationship between Piezo1 and glaucoma is being confirmed. Pathological changes in glaucoma occur in both the anterior and posterior segments of the eye, and it is of great interest for researchers to determine whether Piezo1 plays a role in these changes and how it functions. The elucidation of the mechanisms of Piezo1 action in nonocular tissues and the reported roles of similar mechanically activated ion channels in glaucoma will provide an appropriate basis for further investigation. From a new perspective, this review provides a detailed description of the current progress in elucidating the role of Piezo1 in glaucoma, including relevant questions and assumptions, the remaining challenging research directions and mechanism-related therapeutic potential.</p>","PeriodicalId":13228,"journal":{"name":"Human Cell","volume":"35 5","pages":"1307-1322"},"PeriodicalIF":4.3,"publicationDate":"2022-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40408893","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Human CellPub Date : 2022-09-01Epub Date: 2022-07-30DOI: 10.1007/s13577-022-00743-z
Khloud Fakiha
{"title":"Adipose stromal vascular fraction: a promising treatment for severe burn injury.","authors":"Khloud Fakiha","doi":"10.1007/s13577-022-00743-z","DOIUrl":"https://doi.org/10.1007/s13577-022-00743-z","url":null,"abstract":"<p><p>Thermal skin burn injury affects both adults and children globally. Severe burn injury affects a patient's life psychologically, cosmetically, and socially. The pathophysiology of burn injury is well known. Due to the complexity of burn pathophysiology, the development of specific treatment aiding in tissue regeneration is required. Treatment of burn injury depends on burn severity, size of the burn and availability of donor site. Burn healing requires biochemical and cellular events to ensure better cell response to biochemical signals of the healing process. This led to the consideration of using cell therapy for severe burn injury. Adult mesenchymal stem cells have become a therapeutic option because of their ability for self-renewal and differentiation. Adipose stromal vascular fraction (SVF), isolated from adipose tissues, is a heterogeneous cell population that contains adipose-derived stromal/stem cells (ADSC), stromal, endothelial, hematopoietic and pericytic lineages. SVF isolation has advantages over other types of cells; such as heterogeneity of cells, lower invasive extraction procedure, high yield of cells, and fast and easy isolation. Therefore, SVF has many characteristics that enable them to be a therapeutic option for burn treatment. Studies have been conducted mostly in animal models to investigate their therapeutic potential for burn injury. They can be used alone or in combination with other treatment options. Treatment with both ADSCs and/or SVF enhances burn healing through increasing re-epithelization, angiogenesis and decreasing inflammation and scar formation. Research needs to be conducted for a better understanding of the SVF mechanism in burn healing and to optimize current techniques for enhanced treatment outcomes.</p>","PeriodicalId":13228,"journal":{"name":"Human Cell","volume":"35 5","pages":"1323-1337"},"PeriodicalIF":4.3,"publicationDate":"2022-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40559255","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Human CellPub Date : 2022-09-01Epub Date: 2022-07-16DOI: 10.1007/s13577-022-00746-w
Guiyu Song, Feng Jin
{"title":"RhoGDI1 interacts with PHLDA2, suppresses the proliferation, migration, and invasion of trophoblast cells, and participates in the pathogenesis of preeclampsia.","authors":"Guiyu Song, Feng Jin","doi":"10.1007/s13577-022-00746-w","DOIUrl":"https://doi.org/10.1007/s13577-022-00746-w","url":null,"abstract":"<p><p>Preeclampsia (PE) is a pregnancy-associated disease, which is the major cause of mortality on maternity and perinatal infants. It is hypothesized that PE is a consequence of the dysfunction of the trophoblast cells. Pleckstrin homology-like domain, family A, member 2 (PHLDA2) was shown to inhibit the proliferation, migration, and invasion of trophoblast cells in our previous studies. However, the mechanism by which PHLDA2 affects trophoblast cell function has not been clarified. In the current study, co-immunoprecipitation (Co-IP) with mass spectroscopy analysis was used to explore the proteins that interacted with PHLDA2. A total of 291 candidate proteins were found to be associated with PHLDA2. The interaction between PHLDA2 and Rho guanine nucleotide dissociation inhibitor (RhoGDI) 1 was identified by Co-IP and immunofluorescence staining. Western blot analysis indicated that overexpression of PHLDA2 resulted in upregulation of the RhoGDI1 protein levels, which were stabilized in the presence of cycloheximide. Similarly, overexpression of RhoGDI1 promoted PHLDA2 expression and its stability. Furthermore, pull-down and Co-IP results indicated that PHLDA2 repressed the activity of Rho guanosine triphosphate hydrolase family proteins by regulating RhoGDI1 expression. In addition, RhoGDI1 expression was upregulated in the placental tissues of patients with PE. The effects of the suppression of PHLDA2 expression on proliferation, migration, and invasion of trophoblast cells were partly abrogated following knockdown of RhoGDI1. Taken together, the data indicated that RhoGDI1 mediated regulation of PHLDA2 on the biological behavior of trophoblast cells and may participate in the pathophysiology of PE.</p>","PeriodicalId":13228,"journal":{"name":"Human Cell","volume":"35 5","pages":"1440-1452"},"PeriodicalIF":4.3,"publicationDate":"2022-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40620832","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Human CellPub Date : 2022-09-01Epub Date: 2022-07-07DOI: 10.1007/s13577-022-00741-1
Wen Liu, Guojuan Li, Jing Li, Wei Chen
{"title":"Long noncoding RNA TRG-AS1 protects against glucocorticoid-induced osteoporosis in a rat model by regulating miR-802-mediated CAB39/AMPK/SIRT-1/NF-κB axis.","authors":"Wen Liu, Guojuan Li, Jing Li, Wei Chen","doi":"10.1007/s13577-022-00741-1","DOIUrl":"https://doi.org/10.1007/s13577-022-00741-1","url":null,"abstract":"<p><p>The long-term treatment of glucocorticoids is a common cause of osteoporosis (OP). This study concentrated on inquiring into the regulatory role and potential mechanisms of TRG-AS1 on dexamethasone (Dex)-induced OP in rats. We adopted Dex to treat rat osteoblasts and rats to simulate in-vitro and in-vivo OP models, respectively. Gain-of-function assays of TRG-AS1, miR-802 and CAB39 were constructed in rat osteoblasts to make certain the influence of TRG-AS1, miR-802 and CAB39 on differentiation, proliferation and apoptosis of rat osteoblasts. TRG-AS1 and CAB39 were down-regulated in the Dex-induced OP model in rats, in contrast to miR-802. Overexpression of TRG-AS1 restrained Dex-induced inhibition of osteogenic differentiation, promoted CAB39/AMPK/SIRT-1 and inhibited NF-κB, while overexpression of miR-802 bridled the inhibitory effect of TRG-AS1 on OP. miR-802 was targeted by TRG-AS1, and inhibited CAB39. Inhibition of either AMPK or SIRT-1 abated the osteogenic differentiation-promoting effect of CAB39. Animal experiments displayed that overexpressing TRG-AS1 alleviated Dex-induced OP in rats. In conclusion, up-regulation of TRG-AS1 protected against glucocorticoid-induced OP in rats by modulating the miR-802-mediated CAB39/AMPK/SIRT-1/NF-κB axis.</p>","PeriodicalId":13228,"journal":{"name":"Human Cell","volume":"35 5","pages":"1424-1439"},"PeriodicalIF":4.3,"publicationDate":"2022-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40565137","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}