20 August 2021, Volume 35 Issue 4
    

  • Select all
    |
  • Editorial
    P Conti, G Ronconi, A Caraffa, C Gallenga, I Frydas, S Kritas, V Calvisi, M Trimarchi
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1197-1200. https://doi.org/10.23812/35-4-EDIT-1
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Mast cells (MCs) are hematopoietic cells developed from bone marrow progenitors in response to the ligand stem cell factor, a trans-membrane tyrosine kinase kit receptor. MCs are located virtually in all vascularized tissues and in proximity to neurons and play a decisive role in both innate and adaptive immune responses. Their activation is involved in oxidative stress correlated with infection and inflammation. Pro-inflammatory cytokines are secreted by MCs after physiologic and psychological stress due to virus infection, including SARS-CoV-2. MCs, along with macrophages and pulmonary alveolar epithelial cells, are the main targets attacked by the coronavirus. COVID-19 induced by SARS-CoV-2 causes inflammatory stress which activates MCs to secrete corticotrophin-releasing hormone (CRH), SP, IL-6, TNF, and IL-1. Toll-like receptor (TLR) virus activation in MCs leads to pro-inflammatory cytokine generation without degranulation, an effect that can be inhibited by IL-10, IL-4, IL-1Ra and IL-37. TLR has the ability to recognize extracellular PAMPs by causing the transcription of NLRP, pro-IL-1, and other pro-inflammatory cytokines. The multi-protein complex, comprising pro-caspase-1, activates caspase-1 which in turn activates pro-IL-1 that is transformed into highly inflammatory mature IL-1. In COVID-19, viral RNA is specifically recognized by TLR, followed by recruiting the signal transfer proteins MyD88, IRAK, IKK and TRAF6 which can activate the NF-κB, resulting in transcription of the pro-inflammatory cytokines IL-1 and TNF, responsible for the “cytokine storm” phenomenon. Meanwhile, a new variant of the coronavirus-19 called C.1.2. has been discovered in the United States in the past few days, the effects of which are unknown, and it is therefore of great concern. Researchers are now testing it on immune cells to see if they react and are comparing it to a delta variant. Thus, from the existing data in biomedical literature, we can conclude that the suppression of pro-inflammatory cytokines in viral infections (including COVID-19) mediated by MCs represents a promising therapy not only in this field of medicine, but also in autoimmune, allergic, and cardiovascular disorders, as well as tumor inflammation where MCs play a key role.

  • Article
    LC Ye, LF Qian, L Liang, LJ Jiang, ZY Che, YH Guo
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1201-1216. https://doi.org/10.23812/21-162-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Osteoporosis (OP) is among the most common frequent chronic metabolic bone diseases in postmenopausal women. Here, the effect and underlying mechanisms of miR-195-5p in OP were investigated both in vivo and in vitro. In this study, the microgravity (MG) environment was simulated in MC3T3-E1 cells, and miR-195-5p overexpression or SMURF1 knockdown model was constructed to test their effects on the proliferation, apoptosis and osteogenic differentiation of MC3T3-E1 cells. Furthermore, an OVX mouse model was constructed in vivo, and adenovirus-loaded miR-195-5p mimics were administered to the mice to overexpress miR-195-5p. HE staining and µCT were adopted to observe pathological changes of femur. The targeted relationship between miR-195-5p and SMURF1 was predicted by bioinformatics analysis and verified by the dual-luciferase reporter assay and RNA immunoprecipitation (RIP) experiment. The results indicated that miR-195-5p was down-regulated in the head of femur of OP mouse model and MC3T3-E1 cells subjected to MG microenvironment. In addition, overexpression of miR-195-5p promoted MC3T3-E1 cell osteogenic differentiation and inhibited apoptosis. Mechanistically, SMURF1 is identified as a target of miR-195-5p, and overexpressing miR-195-5p activates the BMP-2/SMAD/Akt/RUNX2 signal by inhibiting the SMURF1 expression. Moreover, SMURF1 downregulation accelerated the osteogenic differentiation of MC3T3-E1 cells and attenuated MG-mediated apoptosis. In addition, upregulating miR-195-5p reduced osteoporosis in the OVX mouse model, accompanied with SMURF1 downregulation and BMP-2/SMAD/Akt/RUNX2 pathway activation. Collectively, miR-195-5p enhances osteogenic differentiation of osteoclast and relieve OP progression in the mouse model through activation of the BMP-2/SMAD/Akt/RUNX2 axis by targeting SMURF1.

  • Article
    WP Zhu, HG He, HX Zhu, LR Wang, ZH Lin, M Wang, L Wang
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1217-1229. https://doi.org/10.23812/21-173-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    To study the molecular mechanism of interferon-alpha (IFN-α) in the treatment of hepatocellular carcinoma (HCC) and the molecular markers that can predict the therapeutic effect, differentially expressed (DE)-miRNAs, -mRNAs, -lncRNAs, and -circRNAs were screened between 12 samples collected from 4 patients who had not received treatment (control), 4 patients who had received recombinant human interferon a-2b treatment (case1), and 4 patients who had relapsed after receiving recombinant human interferon a-2b treatment (case2). Enrichment analyses were performed to determine the principal functions of the DE-RNAs. We also constructed protein-protein interactions (PPI) and competing endogenous RNA (ceRNA) networks. In addition, a series-cluster analysis was performed to analyze changes in gene expression across different groups of HCC. Furthermore, the expression of the genes were verified in the Cancer Genome Atlas (TCGA) database. A total of 36 union DE-miRNAs, 175 union DE-mRNAs, 65 union DE-lncRNAs, and 52 union DE-circRNAs were obtained between the control vs case1, and case2 vs case1 groups. DE-mRNAs were mainly involved in the mitochondrial inner membrane. DE-circRNAs were mainly enriched in the Golgi apparatus. ceRNA network contained 68 DE-mRNAs, 26 DE-miRNAs, 45 DE-lncRNAs, and 23 DE-circRNAs. A total of 24 DE-miRNAs, 175 DE-mRNAs, 65 DE-lncRNAs, and 52 DE-circRNAs were classified into eight profiles, respectively. A total of 26 genes showed a significant correlation with prognosis of HCC (p < 0.05). Some genes may be used to predict the efficacy of IFN-α in the treatment of HCC. The results may lay a foundation for investigating the different sensitivities of IFN-α in the treatment of HCC.

  • Article
    WZ Wei, B Li, JX Lin, J Zhao, XF Zhang, XQ Wang, Z Lv, J Liu
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1231-1243. https://doi.org/10.23812/21-246-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Disuse osteoporosis (DOP) is one of the major consequences of long space flights. DOP also occurs in patients with spinal cord injuries and prolonged bedridden states that can have a severe impact on human health. Bone marrow mesenchymal stem cells (BMSCs) are multipotent stromal cells that play an important role in bone homeostasis. Long non-coding RNAs (lncRNAs) are involved in regulating osteogenic differentiation of BMSCs, and their abnormal expression might lead to the formation of orthopedic diseases. However, the specific mechanism of DOP has not yet been elucidated. All sequencing data were obtained from Gene Expression Omnibus (GEO) datasets. The limma package of R was applied to identify DEmRNAs and DElncRNAs. Pearson correlation coefficients (PCC) between DElncRNADEmRNA expression levels were calculated. Functional annotation was performed for DEmRNAs coexpressed with DElncRNAs. In addition, the Cytohubba plug-in in Cytoscape was applied to determine the top 10 hub genes. Finally, connectivity map (CMap) analysis was used to identify potential therapeutic drugs for DOP. The gene expression data, GSE100930 and GSE17696, were retrieved from the GEO database. A total of 2,212 differentially expressed mRNAs (DEmRNAs) and 22 differentially expressed lncRNAs (DElncRNAs) were obtained. Gene ontology (GO) functional terms, Kyoto Encyclopedia of Genes, and Genomes (KEGG) pathway enrichment analysis reveal 30 significant GO terms and 13 significant pathways. A coding-non-coding gene co-expression (CNC) network was constructed to study the potential role of hub-DElncRNAs and their co-expressed DEmRNAs in DOP. The lncRNAs, GSNAS1, SNHG12, and EPB41LA4A-AS1, were significant in the CNC network and potential regulators of DOP development. Three bioactive compounds (scoulerine, kinetin riboside, dexanabinol) with potential therapeutic significance for DOP were obtained through the Connectivity Map (CMAP) analysis. Our study revealed a new mechanism for a lineage shift of bone marrow mesenchymal stem cells under microgravity, and linked the function of protein-coding mRNAs with ncRNAs, which may contribute to the development of new therapies for DOP.

  • Article
    XH. Yan, QL. Dong, LP. Zhang, G. Jin
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1255-1264.
    Download PDF ( ) HTML ( )     Knowledge map   Save

    This study aimed to assess the synergistic effects of miR-505-3p and uremic serum on the proliferation of vascular smooth muscle cells (VSMCs) in vitro. Serum samples from uremic patients and healthy controls were collected and treated with 10% serum. Meanwhile, the samples were divided into eight groups: uremic serum, healthy serum, uremic serum + miR-505-3p inhibitor, uremic serum + miR-505-3p negative control (NC), uremic serum + miR-505-3p mimics, healthy serum + miR-505-3p inhibitor, healthy serum + miR-505-3p NC, and healthy serum + miR-505-3p mimics. The proliferation and apoptosis of cells and protein expression of miR-505-3p in these groups were detected. Compared with the healthy serum group, the expression level of miR-505-3p in the uremic serum group was elevated, and the difference was statistically significant (P<0.05). Moreover, the uremic serum + miR-505-3p mimics group had the highest proliferation activity and the lowest rate of apoptosis, and the expression level of caspase-3 was significantly down-regulated (P<0.05), while that of PLK1 was noticeably up-regulated. Inhibition of miR-505-3p significantly reduced uremic serum-mediated proliferation of VSMCs (P<0.05), and the underlying mechanism was found to be associated with the miR-505-3p-caspase-3/PLK1 pathway.

  • Article
    JY. Qiu, J. Yuan
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1265-1277.
    Download PDF ( ) HTML ( )     Knowledge map   Save

    The present study is aimed at investigating the effects of circular RNA circ_0078619 on oral squamous cell carcinoma (OSCC) cell growth, migration, invasion, and apoptosis, as well as its mechanism. The GSE131182 dataset was downloaded from Gene Expression Omnibus (GEO), and GEO2R was adopted to analyze the differentially expressed circular RNAs in OSCC tissues compared with normal tissue samples. Quantitative real-time polymerase chain reaction (qRT-PCR) was conducted for detecting the expressions of circ_0078619, microRNA (miRNA, miR)-142-5p, and Rho-associated coiled-coil containing protein kinase 1 (ROCK1) mRNA. Western blot was adopted to quantify the expression of ROCK1 protein in OSCC cells. Cell counting kit-8 (CCK-8) assay, 5-bromo-2’-deoxyuridine (BrdU) assay, Transwell assay, and flow cytometry were employed to assess cell proliferation, migration, invasion, and apoptosis. Bioinformatics was adopted to predict the targeted relationship between miR-142-5p and circ_0078619 or ROCK1 mRNA 3’UTR, and RNA immunoprecipitation and dual-luciferase reporter gene assays were used for validation. Moreover, Pearson correlation analysis was utilized to analyze the correlation among their expressions. Circ_0078619 expression was elevated in OSCC cells and tissues, and its high expression was strongly associated with advanced TNM (tumor, node, metastases) stage and the poor differentiation of tumor tissue. Knockdown of circ_0078619 repressed OSCC cell growth, migration, and invasion, and promoted cell apoptosis. MiR-142-5p was directly targeted by circ_0078619, which negatively modulated its expression. ROCK1 was a target of miR-142-5p, and ROCK1 expression could be positively and indirectly modulated by circ_0078619. Circ_0078619 and miR-142-5p expressions were negatively correlated, while circ_0078619 and ROCK1 expressions were positively correlated in OSCC tissues. Circ_0078619 promoted OSCC cell growth, migration, and invasion, and suppressed apoptosis by modulating the miR-142-5p/ROCK1 axis.

  • Letter
    JW Zhang, GY Xu, XF Wang, YL Zhao, QR Kong
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1279-1286. https://doi.org/10.23812/20-724-L
    Download PDF ( ) HTML ( )     Knowledge map   Save

    In order to explore the mechanism of gefitinib-acquired resistance in lung cancer, a new biomarker has been developed for early clinical diagnosis and intervention; human NSCLC (Non-Small Cell Lung Cancer) cell lines H292 (denoted as H292S) and PC9 (denoted as PC9S) were used to establish gefitinibresistant NSCLC cell lines H292 and PC9 models. CCK-8 (Cell Counting Kit-8) method was used to test the drug resistance of the cells. circRNAs (circular RNAs) that were differentially expressed before and after resistance were screened by RNA sequencing technology. The effects of circSETD3 overexpression and interference on the sensitivity of gefitinib was observed to analyze the nuclear localization of circSETD3 and verify the interaction between circSETD3-miR-520h-ABCG2. The results showed that the most significant change in differential expression of human NSCLC cell lines before and after drug resistance was hsa_circ_0000567, that is, circSETD3, which is mainly present in the cytoplasm. In H292S and PC9S, compared with the negative control group, the cell proliferation ability of the overexpression group was significantly increased, and the apoptosis ability was significantly decreased. In H292R and PC9R, compared with the negative control group, the proliferation ability of the interference group was significantly decreased, and the apoptosis ability was significantly increased. Overexpression of circSETD3 to H292S and PC9S, the expression of ABCG2 increased significantly. Also, the expression of ABCG2 decreased significantly after transfection with miR-520h mimics. H292R and PC9R interfered with circSETD3, the expression of ABCG2 decreased significantly. Moreover, the expression of ABCG2 increased significantly after transfection with miR-520h inhibitor. In conclusion, circSETD3 can be used as a novel biomarker for lung cancer. It relieves miR-520h degradation of the transporter ABCG2 by down-regulating the miR-520h expression, causing gefitinib to be pumped out of the cell.

  • Letter
    R. Saggini, T. Paolucci, R. Grazia Bellomo, M. Mangone, A. Bernetti, L. Pezzi, M. Damiani, V. Liotti, F. Agostini, C. D’Aurizio
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1287-1291.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    G Vartzelis, A Attilakos, C Tsentidis, I Kalimeraki, D Maritsi, A Marmarinos, A Garoufi
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1293-1297. https://doi.org/10.23812/21-126-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    Y Ren, Y Zhao, W Sun, Y Chen, J Yang, Z Li, X Wu, L Zhao, W Sun, C Lv, N Huang, X Li
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1299-1306. https://doi.org/10.23812/21-171-L
    HTML ( )     Knowledge map   Save
  • Letter
    Y Zhang, A Hou, XR Zhuang, XJ Gao, GX Zhang
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1307-1315. https://doi.org/10.23812/21-36-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    G. Ciprandi, Aragona S.E., Study Group on Urinary Infections
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1317-1321.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    A Petraroli, A de Sire, I Pino, L Moggio, C Marinaro, A Demeco, A Ammendolia
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1323-1329. https://doi.org/10.23812/21-229-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    SP Shao, RJ Zhao, S Lu, LP Wen, JJ Ni, KM Zhu, WD Han
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1331-1336. https://doi.org/10.23812/21-117-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    L. Moretti, A. Notarnicola, A. Ostuni, V. Pesce, G. Maccagnano, M. Coviello, I. Covelli, A. Franchini, F.P. Bianchi, B. Moretti
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1337-1342.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    M. Bednarczyk, E. Fatyga, M. Muc-Wierzgoń, S. Dzięgielewska-Gęsiak, J. Wierzgoń
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1343-1347.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    B. Zucchi, F. Agostini, M. Paoloni, A. Bernetti, M. Mangone, S. Zucchi
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1349-1353.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    YF Lian, XM Jiang, LH Sui, XJ Shi
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1355-1362. https://doi.org/10.23812/21-143-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    J. Wang, P. Zha
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1363-1367.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    FX. Qin, LL. Liu, W. Zhao
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1369-1375.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    G Ciprandi, A Licari, MA Tosca, GL Marseglia
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1377-1381. https://doi.org/10.23812/21-176-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    C-F. Shi, Z-H. Wang, J-C. Li, M-W. Liu
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1383-1388.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    XF Cui, P Lin, J Yu, L Liu, ZY Wang, XJ Tang
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1389-1395. https://doi.org/10.23812/21-148-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    A. Monaco, R. Cattaneo, L. De Simone, P. Smurra, F. Adib, D. Pietropaoli, E. Ortu
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1397-1402.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    MS Cori, PE Ferrara, P Papacci, F Serrao, A Di Polito, A Del Vecchio, I Bastoni, C Di Paola, U Moscato, S Codazza, G Ferriero, G Vento, G Ronconi
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1403-1405. https://doi.org/10.23812/21-199-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    J Wu, CL Han, J Luo, L Wang, HW Zhu, WF Huang, HQ Ruan, GL Liao, SY Li, TQ Gan, L Liang
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1407-1413. https://doi.org/10.23812/21-89-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    HT. Tian, PJ. You, Y. Su, CW. Song, HO. Li, M. Wang
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1415-1420.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    Q Ma, WN Li, HY Liu, HY Zhang, JY Dong, XL Tian
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1431-1436. https://doi.org/10.23812/21-88-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    A. Kulig, J. Strzelczyk, K. Janion, M. Skrzypek, E. Nowakowska-Zajdel
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1437-1443.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    C. Bertoldi
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1445-1449.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    F. Raggini, G. Placella, V. Pace, A. Belluati, V. Salini, P. Antinolfi
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1451-1455.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    S. Passanisi, C. Mignosa, J. Trombatore, P. Di Bella, G.B. Pajno, M. Valenzise
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(4): 1457-1460.
    Download PDF ( ) HTML ( )     Knowledge map   Save