20 April 2021, Volume 35 Issue 2
    

  • Select all
    |
  • Editorial
    TC Theoharides, D Lauritano, G Ronconi, V Calvisi, P Conti
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 417-422. https://doi.org/10.23812/Theo_edit
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Infection with SARS-CoV2 leads to COVID-19, the severity of which derives from the host’s immune response, especially the release of a storm of pro-inflammatory cytokines. This coronavirus infects by first binding to the ectoenzyme Angiotensin Converting Enzyme 2 (ACE2), a serine protease acting as the receptor, while another serine protease is necessary for priming the viral spike “S” protein required for entering the cells. Repurposing existing drugs for potential anti-coronavirus activity have failed. As a result, there were intense efforts to rapidly produce ways of providing prophylactic active immunization (vaccines) or abortive passive (convalescent plasma or monoclonal antibodies) neutralizing antibodies. The availability of vaccines for COVID-19 have been largely successful, but many questions still remain unanswered. In spite of the original enthusiasm, clinical studies using convalescent serum or monoclonal antibodies have shown limited benefit. Moreover, the emergence of Long-COVID syndrome in most infected patients necessitates the development of treatment approaches that may prevent viral entry by blocking both serine proteases involved, as with a liposomal blend of the natural flavonoids luteolin and quercetin.

  • Editorial
    P Conti, FE Pregliasco, V Calvisi, Al Caraffa, CE Gallenga, SK Kritas, G Ronconi
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 423-427. https://doi.org/10.23812/Conti_Edit_35_2_1
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Acute severe respiratory syndrome coronavirus-2 (SARS-CoV-2) infection causes coronavirus disease-2019 (COVID-19) which is associated with inflammation, thrombosis edema, hemorrhage, intra-alveolar fibrin deposition, and vascular and pulmonary damage. In COVID-19, the coronavirus activates macrophages by inducing the generation of pro-inflammatory cytokines [interleukin (IL)-1, IL-6, IL-18 and TNF] that can damage endothelial cells, activate platelets and neutrophils to produce thromboxane A2 (TxA2), and mediate thrombus generation. In severe cases, all these phenomena can lead to patient death. The binding of SARS-CoV-2 to the Toll Like Receptor (TLR) results in the release of pro-IL-1β that is cleaved by caspase-1, followed by the production of active mature IL-1β which is the most important cytokine in causing fever and inflammation. Its activation in COVID-19 can cause a "cytokine storm" with serious biological and clinical consequences. Blockade of IL-1 with inhibitory and anti-inflammatory cytokines represents a new therapeutic strategy also for COVID-19. Recently, very rare allergic reactions to vaccines have been reported, with phenomena of pulmonary thrombosis. These side effects have raised substantial concern in the population. Highly allergic subjects should therefore be vaccinated under strict medical supervision. COVID-19 has accelerated vaccine therapy but also the use of drugs and monoclonal antibodies (mABs) which have been used in COVID-19 therapy. They are primarily adopted to treat high-risk mild-to-moderate non-hospitalized patients, and it has been noted that the administration of two mABs gave better results. mABs, other than polyclonal plasma antibodies from infected subjects with SARS-CoV-2, are produced in the laboratory and are intended to fight SARS-CoV-2. They bind specifically to the antigenic determinant of the spike protein, inhibiting the pathogenicity of the virus. The most suitable individuals for mAB therapy are people at particular risk, such as the elderly and those with serious chronic diseases including diabetics, hypertension and obesity, including subjects suffering from cardiovascular diseases. These antibodies have a well-predetermined target, they bind mainly to the protein S (formed by the S1A, B, C and D subtypes), located on the viral surface, and to the S2 protein that acts as a fuser between the virus and the cell membrane. Since mABs are derived from a single splenic immune cell, they are identical and form a cell clone which can neutralize SARS-CoV-2 by binding to the epitope of the virus. However, this COVID-19 therapy may cause several side effects such as mild pain, bleeding, bruising of the skin, soreness, swelling, thrombotic-type episodes, arterial hypertension, changes in heart activity, slowed bone marrow activity, impaired renal function, diarrhea, fatigue, nausea, vomiting, allergic reaction, fever, and possible subsequent infection may occur at the site of injection. In conclusion, the studies promoting mAB therapy in COVID-19 are very promising but the results are not yet definitive and more investigations are needed to certify both their good neutralizing effects of SARS-CoV-2, and to eliminate, or at least mitigate, the harmful side effects.

  • Review
    G Dipalma, AD Inchingolo, F Inchingolo, IA Charitos, M Di Cosola, AP Cazzolla
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 429-440. https://doi.org/10.23812/20-747-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    This paper aims to describe the biochemical interactions between teeth and the oral environment that occur during the caries process, hence it covers all the steps related to physico-chemical reactions, including the most up-to-date theoretical basis in the clinical application for the prevention and treatment of caries. The terms ‘demineralization’ and ‘remineralization’ that characterise this process were analysed, as well as the role of the microbiota in its interaction with the hard surface of the teeth. The biochemical mechanisms that lead to the onset of carious lesions and those that occur during the healing and repair of such lesions are listed.

  • Article
    R Saggini, GP Anastasi, S Battilomo, P Maietta Latessa, G Costanzo, F Di Carlo, F Festa, G Giardinelli, F Macrì, L Mastropasqua, M Nardone, G Nicoletti, A Orsini, M Pastorelli, R Pellegrino, M Trevisan, L La Mesa, AM Le Pera, MG Privitera, G Ralli, C Villani, B Bricot, RG Davidson, A Hassdenteufel, ZM Hawamdeh, GJ Kleinrensink, E Matheron, VL Stoev, S Stratulat, S Tiron, P Villeneuve, T Paolucci, RG Bellomo
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 441-456. https://doi.org/10.23812/20-743-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Good fundamentals of posture and balance are essential for the efficient performance of both simple daily tasks and more complex movement patterns. In particular, postural balance is the ability to keep the body in equilibrium and to regain balance after the shift of body segments: postural control mechanisms of integration of the visual, vestibular and foot afferential channels contribute to this. This document provides recommendations based on scientific evidence, clinical practice, and consensus between experts concerning the prevention, diagnosis, and treatment of postural dysfunction at the three stages of life as the developmental age, adult age, and old age > 65 years and follows the "National Guidelines on Classification and Measuring of Posture and its Dysfunctions" per the Italian Ministry of Health (December 2017). The paper answers four main questions: i) "Which measures can be adopted to prevent postural dysfunctions?" ii) "What can we do in order to make a correct diagnosis of postural dysfunction?" iii) "What are the correct treatment programs for postural dysfunctions?" iv) Which professional competencies and experiences are useful for preventing, diagnosing and treating postural dysfunctions? By the Consensus of the Experts and the scientific evidence, emerge that the approach to postural dysfunctions requires a multidisciplinary and interdisciplinary team. Furthermore, rehabilitation treatment interventions must be specific to the age groups that have been indicated, to consider the integration of the main systems and subsystems of postural control that change with age.

  • Article
    AM Shamseldeen, M Hamzawy, NA Mahmoud, L Rashed, SS Kamar, IA Harb, N Sharawy
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 457-471. https://doi.org/10.23812/20-693-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Diet pattern is an emerging risk factor for renal disease. The mechanism by which high-fat high fructose (western) diet mediates renal injury is not yet fully understood. The objective of the present study was to investigate the relationship between endoplasmic reticulum (ER) stress and autophagy in the development of renal impairment and aggravation of the inflammatory response. Eighty male rats were randomly divided into four groups as follows: a standard diet-fed (ConD), a high-fat high fructose diet fed (HFHF-V), ConD fed and orally supplemented with vitamin E (ConD-E), and HFHF fed and orally supplemented vitamin E (HFHF-E). After 12 weeks, either lipopolysaccharide (LPS) or saline was injected. We found that upregulation of endoplasmic reticulum stress-related proteins rendered the cells susceptible to injury induced by dysbiosis and microbiota-derived metabolites. A downregulation of autophagy and upregulation of caspase-12 resulted in the loss of intestinal integrity and renal tubular injury. Maintained ER stress also increased the inflammatory response to LPS. In contrast, vitamin E effectively ameliorated ER stress and promoted autophagy to protect intestinal and renal tissues. Our results provide insight into the influences of sustained ER stress activation and autophagy inhibition on the development of renal injury, which may contribute also to the enhanced inflammatory response.

  • Article
    ZB Li, X Chen, XJ Yi
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 473-484. https://doi.org/10.23812/20-570-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Exosomes are involved in a range of processes in lung cancer such as cell proliferation, metastasis, and angiogenesis. Tumor-derived exosomes participate in the formation and progression of lung cancer by delivering functional biomolecules, including microRNAs (miRNA). The purpose of the present study was to determine the role of lung cancer cell-derived exosomal miR-210 in the proliferation and invasion of lung cancer cells and its underlying mechanism. Initially, exosomes were isolated from A549 cells and characterized by transmission electron microscopy and assessment of exosomal marker expression. RT-qPCR determined that miR-210 expression was elevated in exosomes as well as lung cancer cells. As reflected by dual-luciferase reporter assay, miR-210 negatively regulated RUNX3 expression. Following loss- and gain- function assay, it was found that miR-210 inhibition suppressed biological properties of A549 and H460 cells, which could be reversed by the silencing of RUNX3. miR-210 elevation induced the p-PI3K/PI3K and p-AKT/AKT levels, suggesting the activation of PI3K/AKT signaling pathway. Collectively, exosomal miR-210 targeted and negatively regulated RUNX3 expression to promote malignant properties of lung cancer cells by potentiating PI3K/AKT signaling pathway.

  • Article
    M Pietruczuk, L Kraszula, M Kupczyk, P Kuna, M Eusebio
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 485-494. https://doi.org/10.23812/20-689-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Adhesion molecules, as such, play essential roles in T-cell transendothelial extravasation during inflammation. A better understanding of the mechanisms underlying this process may be of value in the management of asthma. The present study employed Magnetic-Activated Cell Sorting (MACS) to isolate human CD8+ T lymphocytes from peripheral blood of asthma patients and controls. The cells were flow cytometrically assessed to evaluate surface expression of an adhesion molecule, L-selectin (CD62L) on the surface of CD8FoxP3-/bright T cell subsets and its response to inflammatory cytokines. We showed that CD8+CD28+TCRαβ+CD62LhighFoxP3bright T cells were deficient in blood of some asthma patients but abundant in others. After co-stimulation of CD8+ T cells with anti-CD3/CD28 in combination with IL-2 and IL-10 or TGF-β, the frequencies of CD8+CD28+TCRα/β+CD62Lhigh T cells in the group of patients were lower than at baseline. Our data indicate that L-selectin expression is regulated by inflammatory cytokines. Overall, these data reveal that asthma phenotypes may be further stratified into micro subtypes with distinct cellular and molecular characteristics, supporting the concept of asthma endotypes.

  • Article
    Y Guan, JQ Chen, XY Li, SN Jiang
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 495-504. https://doi.org/10.23812/20-500-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Lipopolysaccharide (LPS) plays an important role in tumor suppression by activating macrophages. After macrophages activation, a trail of cytokines was secreted, including IL-1β. Previous studies reported that the anti-tumor function of IL-1β is concentration-dependent, and increasing the level of IL-1β will enhance its anti-tumor effect. Cytolysin A (ClyA), a member of the protein family called pore-forming toxins (PFTs), is secreted by Gram-negative bacteria, which has a potential role in enhancing the secretion of IL-1β. In this study, the function of Cytolysin A was evaluated by investigating its ability to induce innate immune responses in macrophages and the signaling pathway(s) involved in LPS-induced production of IL-1β. The production of IL-1β was highly enhanced when the macrophages were treated with LPS and ClyA together. The production of IL-1β was regulated by TLR4-MyD88-IL-1β pathway and NLRP3-ASC-Caspase1-IL1β pathway. By treating the colon cancer cell line CT26 with the conditioned medium, the proliferation of CT26 cells was inhibited and the apoptosis of CT26 cells was increased. In conclusion, this study indicated that ClyA enhances the production of IL-1β induced by LPS in human macrophages. The proliferation of CT26 cells was inhibited and the apoptosis was increased when being treated with the macrophage-conditioned media, which provides a feasible treatment for colon tumor.

  • Article
    HB Zheng, M Wu, G Zhang, KL Chen
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 505-516. https://doi.org/10.23812/20-688-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    The present study aimed to screen abnormally expressed microRNAs (miRs/miRNAs) in patients with postmenopausal osteoporosis (POP) and explore their mechanisms via functional verification. Bone marrow mesenchymal stem cells (BMSCs) were extracted from healthy controls and patients with POP. Differences in osteogenic differentiation and proliferation of human BMSCs were compared between the two groups using Cell Counting Kit-8 (CCK-8) assay and alizarin red staining. A rat model of POP was established. Compared with patients with POP, human BMSCs in healthy controls had significantly enhanced viability at 24, 36, 48 and 72 h. The results of alizarin red staining revealed that the deposition of calcium minerals in human BMSCs were significantly lower in patients with POP. Based on miRNA microarray and reverse transcription-quantitative polymerase chain reaction (PCR) results, the expression levels of miR-7010 and miR-467c decreased, while miR-132 and miR-182 expression increased in the human BMSCs of patients with POP. Alizarin red staining showed that miR-182 markedly suppressed the osteogenic differentiation of primary rat BMSCs in rats. Western blotting and immunofluorescence assay revealed that miR-182 inhibited the expression of osteogenesis markers runt-related transcription factor 2, osterix and actinin-associated LIM protein. The results of the luciferase reporter assay showed that Smad1 is the direct target of miR-182. In rat primary BMSCs, Smad1 overexpression abolished the inhibitory effect of miR-182 on osteogenesis, indicating that miR-182 inhibits osteogenic differentiation of primary rat BMSCs in rats by targeting Smad1. Finally, in vivo experimental results showed that the biomechanical characteristics of bone tissues in POP rats were significantly enhanced by miR-182 inhibition, while they were significantly weakened by miR-182 overexpression. MiR-182 inhibits osteogenic differentiation of rat BMSCs, thus aggravating POP in rats.

  • Article
    H. Ashraf, S. Nawaz, M.Z. Yousaf, Z. Rashid, A.R. Awan, M. Wasim, M. Tayyab, S. Saeed, S. Firyal
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 517-524.
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Foot rot is a necrotic, infectious, and debilitating disease of hooves in ruminants caused by theconcordant effect of Dichelobacter nodosus and Fusobacterium necrophorum. This work aimed foridentification and prevalence of leukotoxin gene, lktA, variant strains of F. necrophorum in cow foot rotlesions. A total of twenty samples were collected from the feet of foot rot-affected cattle. Bacterial DNA wasextracted and amplification of lktA was performed through polymerase chain reaction (PCR) followedby direct DNA sequencing. The sequencing data was subjected to homology search, multiple sequencealignment and phylogenetic tree reconstruction using respective software for taxonomic characterization.This study revealed the presence of JKS-F3, JKS-F2 and JKS-F1 in the foot rot-affected cattle in the localisolates of Punjab, Pakistan, with high prevalence of F. necrophorum JKS-F3 strain (80%) with a lesionscore of 3 and 4. The 3D structure of protein was performed using comparative homology modeling withI-TASSER. The accuracy of the predicted 3D structure was checked using Ramachandran plot analysisand showed that 85.4% amino acids were in the favored region i.e., extended conformation of β-sheetsare clustered in the right hand-region. These findings suggest that early and accurate identification andcharacterization of the causative agent of foot rot is a preliminary requirement to control the infection

  • Article
    P Cong, C Yi, X Wang, Y Peng
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 525-535. https://doi.org/10.23812/20-554-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    This study aimed to construct a vector lentivirus carrying the Smo gene and transfect pancreatic cancer cells positive for CD24CD44 surface antibody and detect the infectivity. A lentivirus carrying a specific Smo fragment was designed and synthesized, and its functionality was tested. An overexpression group, inhibitory group, and negative control group were used for subsequent experimental research and comparison. A virus was successfully designed and produced. The best viral load was the 1X106 TU virus, where the cell growth and fluorescence effect of culture wells with polybrene dilution were the best. These are the transfection conditions and transfection param-eters for subsequent experiments. This plasmid was detected with a flag antibody by Western blot. The result was that it had a large specific 250kD band, and the membrane protein was overexpressed successfully. The expression results of Smo in five groups of cells after virus transfection detected by RT-PCR: blank group were 1.0038±0.0344, CON238 negative group: 1.0276±0.2944d, CON077 negative group: 0.8793±0.0402; LV-SMO15570-2 overexpres-sion group: 2.7479±0.8308, and LV-SMO-RNAi37304-1 inhibition group: 0.2386±0.0481. There were differences among the overexpression group and inhibition group with the other three groups. Homogeneity of variance: Bartlett F = 4.3530, P = 0.0016 < 0.05, heterogeneous. K-W test: cc2 = 10.9905* P = 0.0267, and there was a statisti-cally significant difference. The designed virus achieved the goal requirements. An sRNA fragment was designed for the key gene Smo of the Hh signaling pathway, and a vector lentivirus carrying this fragment was successfully constructed. The expression of Smo was analyzed after transfecting SW1990CD24CD44 positive cells, suggesting that the function of the RNA fragment designed for the key gene Smo in this experiment was successful.

  • Article
    BZ Li, HH Bai, FW Tan, YB Gao, J He
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 537-546. https://doi.org/10.23812/20-683-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    The prognosis of lung cancer remains poor due to the limited biomarker selection for treating patients with optimal chemotherapy. The aim of this study is to discover and identify new biomarkers with the value of predicting chemotherapy responses in a lung adenocarcinoma (AD) specimen. In this study, six pairs of pre-treatment fresh primary lung AD-cancer tumors with varied chemotherapy responses were used to discover new biomarkers by two-dimensional difference gel electrophoresis (2D DIGE). Among the matched protein spots, 19 were up-regulated and 18 were down-regulated in chemo-sensitive tumors versus chemo-resistant tissues. These differentially expressed proteins could be divided into five classes: redox regulation protein, the cytoskeletal protein, cell metabolism enzymes or proteins, apoptosis, signal transduction mediated molecules, and other functional proteins. Proteins of interest, including PRDX2, PRDX6, and Gelsolin, were differentially expressed in chemo-sensitive tumors versus chemo-resistant tissues and these observations were validated by immunohistochemistry in 92 formalin-fixed and paraffin-embedded (FFPE) specimens. Our results demonstrated that PRDX6 protein expression was closely related to tumor response (cc2 = 5.57, P < 0.05), whereas no relationship of PRDX2 and Gelsolin were obtained with tumor response (cc2 = 0.51 P > 0.05, cc2 = 0.41 P > 0.05). This tissue proteomics study provides evidence that PRDX6 may be regarded as a predictive biomarker for poor chemotherapy response, which can be helpful in guiding pretreatment protocols.

  • Article
    YX Qi, XJ Su, LL Wei, J Zhang
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 547-557. https://doi.org/10.23812/20-646-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    The aim of this study was to investigate the effect of erythropoietin (EPO) on the apoptosis of retinal ganglion cells (RGCs) induced by high glucose and its mechanism. Rat primary RGCs were extracted to establish high glucose-induced apoptosis models using a 30 mM high-glucose medium. Then flow cytometry, cell counting kit-8 (CCK-8) assay and Western blotting assay were performed to detect the effects of high-, medium- and low-dose EPO on the apoptosis of RGCs induced by high glucose. Next, the molecular mechanism by which EPO suppressed the high glucose-induced apoptosis of RGCs was explored via gene array assay and bioinformatics analysis. The results and mechanism of bioinformatics analysis were verified by Western blotting assay. Finally, the small interfering ribonucleic acid (siRNA) experiment was applied to knock down tyrosine-protein phosphatase non-receptor type 1 (PTPN1) and PTPN11 to verify their roles in the inhibition of EPO on the apoptosis of RGCs triggered by high glucose. Flow cytometry-Annexin V/propidium iodide (PI) staining and CCK-8 assay confirmed that the high-, medium- and low-dose EPO inhibited the apoptosis of RGCs induced by high glucose in a dose-dependent manner (P<0.05). Subsequently, Western blotting assay results manifested that the high-, medium- and low-dose EPO reduced the expression levels of apoptosis-related proteins active-cysteinyl aspartate specific proteinase 3 (Caspase 3) and active- Caspase 9 in a dose-dependent manner (P<0.05). Moreover, according to gene array assay and bioinformatics analysis results, the c-Jun N-terminal kinase (JNK) signaling pathway, PTPN1 and PTPN11 might exert crucial effects in the inhibition of EPO on the apoptosis of RGCs induced by high glucose. Western blotting assay results also demonstrated that, compared with the high-glucose treatment, the high-dose EPO treatment decreased the protein expression level of phosphorylated (p)-JNK1/JNK but increased the protein expression levels of PTPN1 and PTPN11 (P<0.05). Moreover, flow cytometry-Annexin V/PI staining and CCK-8 assay results revealed that in EPO-treated cells, knocking down PTPN1 and PTPN11 significantly reversed the protective effect of EPO against high glucose-induced retinal ganglion cell apoptosis (P<0.05). Lastly, Western blotting assay illustrated that knocking down PTPN1 and PTPN11 significantly abolished the inhibition of high-dose EPO on the JNK signaling pathway. EPO may suppress the JNK signaling pathway by raising the expression levels of PTPN1 and PTPN11, so as to inhibit the apoptosis of RGCs triggered by high glucose.

  • Article
    QZ Li, LP Sun, HY Shi, Y Chen, H Shen
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 559-569. https://doi.org/10.23812/20-504-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    This study aimed to evaluate the anti-tumor effect of a new generation of protease inhibitor, oprozomib (OPZ), used alone and in combination with cisplatin, also called CDDP, on cervical cancer. Five different types of cervical cancer cell lines - HeLa, Caski, HeLa-CDDP, C33a, and SiHa - and one nontransformed cervical cell line - HaCaT -were treated with OPZ alone or in combination with cisplatin. The inhibitory effects of OPZ and cisplatin on the proliferation of cervical cancer cells were then analyzed using cytotoxicity tests, flow cytometry, and Western blotting. It was found that OPZ alone or in combination with cisplatin can reduce the proliferation of the five types of cancer cells by enhancing the lysis of caspase-3 and PARP and inducing cancer cell apoptosis. In the combined treatment, OPZ was found to inhibit the degradation of inhibitory factor κB alpha induced by cisplatin, thereby inhibiting the activation of NF-κB, which causes cisplatin resistance, and enhancing the sensitivity of the tumor cells to cisplatin. Moreover, OPZ promoted the phosphorylation of the apoptosis signaling pathway JNK that was activated by cisplatin, thereby inducing tumor cell apoptosis. These findings provide a theoretical basis for the clinical use of OPZ alone and in combination with cisplatin in the treatment of cervical cancer.

  • Article
    C Yang, N Chen, XL Tang, XH Qian, CP Cai
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 571-581. https://doi.org/10.23812/20-615-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Both interleukin (IL)-33 and IL-25 induce Th2-type cytokine production by various cell types, suggesting that they may contribute to development of allergic disorders, however, the immunomodulatory effects of IL-33 and IL-25 in ovalbumin (OVA)-induced allergic rhinitis (AR) remain unclear. In the present study, anti-IL-33 and anti-IL-25 Abs were administrated intranasally during rechallenge in OVA-induced AR. Immunomodulatory effects were evaluated by measuring nasal rubbing, sneezing occurrence, serum OVA-specific antibodies, Th2 immune responses, neutrophil, eosinophil and mast cell recruitment into the nasal mucosa. We found that treatment with anti-IL-33 Ab markedly reduced nasal rubbing, sneezing events, Th2 immune responses, serum OVA-specific IgE and IgG1 levels, mucosal neutrophil, eosinophil and mast cell infiltration. In contrast, the effect of IL-25 antagonism was limited to attenuating the Th2 immune responses, and neutrophil and eosinophil infiltration. These observations indicate that IL-33 and IL-25 play a pathogenic role in an established AR mouse model, with a greater contribution of IL-33 than IL-25. Our findings suggest that IL-33 neutralization may be a potential approach for treatment of AR.

  • Letter
    F Tao, C Gu, N Li, Y Ying, LF Cao, QF Xiao, D Ni, YB Zhuang, Q Zhang
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 583-591. https://doi.org/10.23812/20-664-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 gefitinib-resistant 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.

  • Article
    LK Yu, J Zhang, ZY Sun, CL Ruan, H Li, XJ Ruan
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 593-603. https://doi.org/10.23812/21-40-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Autologous chondrocyte (CH) transplantation is a novel strategy to treat post-traumatic osteoarthritis (PTOA). In this study, an in vitro coculture model was used to explore the effects of interleukin (IL)-10 overexpressed CHs on degenerated CHs. The original CHs were isolated from the patients' knee joint cartilage and pretreated with IL-1β to get degenerated CHs. Moreoer, CHs were transfected with a lentivirus vector to overexpress IL-10. After coculture with the degenerated CHs, the apoptosis, collagen X, IL-6, and TNF-α of original CHs were increased, and the collagen II and IL-10 were decreased compared to the separated culture condition. Coculture with original CHs did not alleviate the degeneration of the IL-1β-pretreated CHs. However, coculture with the IL-10-overexpressed CHs rescued the proliferation, collagen II, aggrecan, SOX9, and IL-10 expression, and suppressed the apoptosis, collagen X, RUnx2, IL-6, and TNF-α levels in the IL-1β pretreated CHs. Additionally, the IL-10-overexpressed CHs also maintained a healthy state when cocultured with the degenerated CHs. Therefore, transplanting the IL-10-overexpressed CHs in the treatment of PTOA would obtain a more durable and visible effect in alleviating the CH degeneration.

  • Article
    ZH Liu, DD Qi, X Li, SQ Zhang, Y Zhao, LX Fu, LY Lu
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 605-615. https://doi.org/10.23812/20-391-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs) is vital for bone formation, and its dysfunction is linked to osteoporosis (OP). In this work, we explored the function of long non-coding RNA (lncRNA) small nucleolar RNA host gene 14 (SNHG14) in regulating osteogenic differentiation of hBMSCs. In the present study, the expression of SNHG14 in hBMSCs obtained from OP patients was measured by quantitative real-time polymerase chain reaction (qRT-PCR). SNHG14 was over-expressed or knocked down in hBMSCs, and the expression levels of OP-related genes (ALP, OCN, and OPN) in hBMSCs were detected by qRT-PCR and Western blot. StarBase database and miRanda database were used to predict the binding sites between SNHG14 and miR-185-5p, and between miR-185-5p and 3'UTR of WNT1 inducible signaling pathway protein 2 (WISP2), respectively. Luciferase reporter gene assay was used to validate the binding relationship between SNHG14 and miR-185-5p, and miR-185-5p and 3'UTR of WISP2, respectively. Here, we report that SNHG14 was significantly down-regulated in hBMSCs obtained from patients with OP. Overexpression of SNHG14 promoted osteogenic differentiation, while knockdown of SNHG14 worked oppositely. Mechanistically, miR-185-5p was demonstrated to be a target of SNHG14, and could reverse the function of SNHG14. Additionally, WISP2 was identified as a target gene of miR-185-5p in hBMSCs and could be indirectly regulated by SNHG14. Taken together, down-regulation of SNHG14 in hBMSCs accelerated the progression of OP via regulating miR-185-5p/WISP2 axis.

  • Article
    YH Li, MF Lv, MS Lu, JP Bi
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 617-627. https://doi.org/10.23812/20-638-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    The present study aims to explore the function of human bone marrow mesenchymal stem cell (BMSC)-derived exosomal micro ribonucleic acid (miR)-338-3p in hepatocellular carcinoma (HCC) and further investigate its effect on HCC cell functions. Firstly, BMSCs were co-cultured with HCC cells, and BMSC-derived exosomes were identified. Next, Transwell assay and methyl thiazolyl tetrazolium (MTT) experiments were carried out to detect the effects of miR-338-3p and E26 transformation specific-1 (ETS1) on the viability, invasion, migration, and apoptosis of HCC cells through the exosomes derived from BMSCs. Furthermore, the targeting relationship between miR-338-3p and EST1 was verified via bioinformatics study and dual-luciferase reporter gene analysis. Additionally, Western blotting (WB) was carried out to measure the expression levels of EST1 and other proteins in HCC cells. It was found that BMSCs inhibited HCC cell proliferation, invasion and migration, and induced cell apoptosis, while the inhibitors of exosomes played the opposite roles. In addition, the up-regulation of exosomal miR-338-3p or the silencing of EST1 restrained HCC cell proliferation, invasion and migration, and induced cell apoptosis. In conclusion, BMSC-derived exosomal miR-338-3p delays the development of HCC by down-regulating EST1, providing a new promising treatment target for HCC.

  • Article
    M Chen, MH Li, N Zhang, WW Sun, H Wang, YA Wang, Y Zhao, W Wei
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 629-640. https://doi.org/10.23812/20-537-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Rheumatoid arthritis (RA) is characterized by inflammation of the synovial membrane, accompanied by hyperplasia and neo-angiogenesis, which promote local inflammation. Macrophage-derived exosomes have been reported to enhance inflammation and the immune response. In the present study, we identified a novel exosomal microRNA (miR)-103a, which aids in the regulation of inflammation and angiogenesis in mice with RA, and attempted to identify the underlying mechanism. Initially, a mouse model of RA was established. Thereafter, exosomes were isolated from macrophage RAW264.7 cells and evaluated through transmission electron microscopy and nanoparticle tracking analysis. After prediction and verification of the target genes of miR-103a, RT-qPCR was used to assess miR-103a and HNF4A expression in mice with RA. High expression of miR-103a and low expression of HNF4A were observed in mice with RA, thus, miR-103a was found to target and downregulate HNF4A. Exosomal miR-103a promoted inflammation and angiogenesis in mice with RA which was accompanied by an increase in the levels of factors associated with inflammation and angiogenesis. However, an opposite trend was observed upon HNF4A elevation. Exosomal miR-103a was also found to activate the JAK/STAT3 signaling pathway. In conclusion, exosomal miR-103a inhibited the expression of HNF4A to activate the JAK/STAT3 signaling pathway, thereby exacerbating RA in mice.

  • Article
    L Li, J Bao, M Wang, B Chen, B Luo, F Yan
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 641-655. https://doi.org/10.23812/20-628-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Previous studies have shown that high-fat diet (HFD) may aggravate periodontitis, however the underlining mechanism remains to be further clarified. This study aims to explore whether HFD promotes periodontitis by inducing periodontal microbiota dysbiosis or stem cell dysfunction. A high-fat diet was given to four-week-old male Sprague-Dawley rats for 12 weeks. Periodontitis was induced during the latter 4 weeks. At the end of the 12th week, samples were collected after euthanasia. Maxillae were harvested for histological or microbial analysis. The microbial 16S rRNA gene sequencing was performed with the Illumina MiSeq platform. The data was analyzed through RDP Classifier against the SILVA database. The mandible molars were harvested for isolating periodontal ligament stem cells (PDLSCs). The protein level of p27, p21, and p16, which are negative regulators of the cell cycle, in PDLSCs were detected. Markers of osteogenic differentiation and pro-inflammatory mediators were detected by real-time polymerase chain reaction. Activation of pro-inflammatory signaling pathways was detected by Western blotting. We found that HFD significantly increased ligature-induced alveolar bone loss. HFD resulted in a less diverse periodontal microbiota, with increased proportions of Lactococcus, Bacillus, Alloprevotella, Carnobacterium, and Exiguobacterium and decreased proportion of Nitrospira. HFD increased the protein levels of p27, p16, and p21, and upregulated the expression of osteogenic biomarkers, IL-1β and IL-10 with the ERK1/2 signaling pathway activated in PDLSCs.

  • Letter
    G Di Giacomo, N de Gasperis
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 657-661. https://doi.org/10.23812/20-457-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    N Yao, J Xie, ZY Li, J Zheng, A Jiang, YF Wang, MW Liu
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 663-667. https://doi.org/10.23812/21-34-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    A Conti, GJ Burastero, BJ Hales, D Breda, M Alessio, SE Burastero
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 669-676. https://doi.org/10.23812/21-07-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    ZL Sun, MH Chen, YN Guo, ZQ Liu
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 677-682. https://doi.org/10.23812/20-731-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    A Varricchio, I La Mantia, FP Brunese, G Ciprandi
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 683-686. https://doi.org/10.23812/21-28-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    C Sun, QT Shi, HC Liang
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 687-692. https://doi.org/10.23812/20-735-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    JZ Li, XH Jin, FM Zhang, QR Guo
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 693-697. https://doi.org/10.23812/21-03-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    P Amori, G Vitiello, T Lotti
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 699-702. https://doi.org/10.23812/20-609-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    YX Hou, CL Min, LJ Ma
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 703-708. https://doi.org/10.23812/20-742-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    D Liu, H Zhang, D Liu, T Peng
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 709-715. https://doi.org/10.23812/20-590-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    M Ferrillo, M Migliario, C Curci, A Roccuzzo, M Invernizzi, A de Sire
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 717-723. https://doi.org/10.23812/21-106-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    R Pulcini, F Avolio, B Sinjari, I Robuffo, V Flati, L Pignatelli, S Martinotti, E Toniato
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 725-728. https://doi.org/10.23812/20-556-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    W Arancio, A Ahmed, C Basset, A Jurjus, I Miletich, A Leone
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 729-733. https://doi.org/10.23812/21-12-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    MF Zhu, LX Chen, F Gao, DY Sheng, MJ Xu, JR Zhang, WX Qian
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 735-738. https://doi.org/10.23812/20-627-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    F Rinaldi, A Trink, A Sparavigna, D Pinto
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 739-744. https://doi.org/10.23812/21-33-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    S Wang, YC Wang, KL Dong
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 745-750. https://doi.org/10.23812/20-704-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    D Cavagnetto, M Carossa, A Deregibus, M Lacilla, A Albera, P Ceruti, S Carossa
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 751-756. https://doi.org/10.23812/20-655-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    L Ricciardi, F Furci, S Gangemi
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 757-760. https://doi.org/10.23812/20-566-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    RS Almeida, RI Ferreira, D Aa Galdino, I Ggf Mariano, NF Silveira, PL Almeida, M Ap Andrade
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 761-765. https://doi.org/10.23812/20-584-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    G Barassi, M Supplizi, L Prosperi, G Irace, A Younes, M Della Rovere, A Rabini, A Colombo, A Di Iorio
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 767-773. https://doi.org/10.23812/21-37-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    R Rauso, GF Nicoletti, R Fragola, N Zerbinati, E Nikolli, P Bove, G Tartaro
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 775-777. https://doi.org/10.23812/21-118-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    K Lian, J Cui, L Li, Y Li, QH Zhang
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 779-781. https://doi.org/10.23812/20-522-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    IZ Sadiq, A Muhammad, F Sadiq Abubakar, MN Shuaibu
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 783-786. https://doi.org/10.23812/20-582-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    G Laganà, A Malara, A Koumoulis, M Tepedino, N Venza, P Cozza
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 787-790. https://doi.org/10.23812/20-578-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    N Logoluso, R Ciliberto, I Morelli, GM Peretti, V Pellegrini
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 791-795. https://doi.org/10.23812/20-708-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    L Xiao, Y Yang, G Li
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 797-801. https://doi.org/10.23812/20-569-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    LM Zhang, L Chen, YJ Chen, MW Liu
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 803-809. https://doi.org/10.23812/20-699-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    C Huang, YQ Xia, L Xiao, J Huang, ZM Zhu
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 811-817. https://doi.org/10.23812/20-626-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    KH Tong, J Qian
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 819-826. https://doi.org/10.23812/20-667-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    D Bizzoca, MT Rocchetti, S Scacco, F Taurino, G Vicenti, F Spadaccino, L Moretti, E Ranieri, L Gesualdo, F Moretti, G Peretti, G Banfi, B Moretti, A Gnoni
    Journal of Biological Regulators and Homeostatic Agents. 2021, 35(2): 827-832. https://doi.org/10.23812/20-706-L
    Download PDF ( ) HTML ( )     Knowledge map   Save