20 October 2020, Volume 34 Issue 5
    

  • Select all
    |
  • Editorial
    P. Conti, Al. Caraffa, C.E. Gallenga, R. Ross, S.K. Kritas, I. Frydas, A. Younes, P.Di Emidio, G. Ronconi, E. Toniato
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1623-1627. https://doi.org/10.23812/20-34-4EDIT-65
    Download PDF ( ) HTML ( )     Knowledge map   Save

    IL-1 induces a significant number of metabolic and hematological changes. In experimental animals, IL-1 treatments cause hypotension due to rapid reduction of systemic blood pressure, reduced vascular resistance, increased heart rate and leukocyte aggregations. IL-1 causes endothelial dysfunction, the triggering factor of which may be of a different nature including pathogen infection. This dysfunction, which includes macrophage intervention and increased protein permeability, can be mediated by several factors including cytokines and arachidonic acid products. These effects are caused by the induction of IL-1 in various pathologies, including those caused by pathogenic viral infections, including SARS-CoV-2 which provokes COVID-19. Activation of macrophages by coronavirus-19 leads to the release of pro-inflammatory cytokines, metalloproteinases and other proteolytic enzymes that can cause thrombi formation and severe respiratory dysfunction. Patients with COVID-19, seriously ill and hospitalized in intensive care, present systemic inflammation, intravascular coagulopathy with high risk of thrombotic complications, and venous thromboembolism, effects mostly mediated by IL-1. In these patients the lungs are the most critical target organ as it can present an increase in the degradation products of fibrin, fibrinogen and D-dimer, with organ lesions and respiratory failure. It is well known that IL-1 induces itself and another very important pro-inflammatory cytokine, TNF, which also participates in hemodynamic states, including shock syndrome in COVID-19. Both IL-1 and TNF cause pulmonary edema, thrombosis and bleeding. In addition to hypotension and resistance of systemic blood pressure, IL-1 causes leukopenia and thrombocytopenia. The formation of thrombi is the main complication of the circulatory system and functionality of the organ, and represents an important cause of morbidity and mortality. IL-1 causes platelet vascular thrombogenicity also on non-endothelial cells by stimulating the formation of thromboxane A2 which is released into the inflamed environment. IL-1 is the most important immune molecule in inducing fever, since it is involved in the metabolism of arachidonic acid which increases from vascular endothelial organs of the hypothalamus. The pathogenesis of thrombosis, vascular inflammation and angigenesis involves the mediation of the activation of the prostanoid thromboxane A2 receptor. In 1986, in an interesting article (Conti P, Reale M, Fiore S, Cancelli A, Angeletti PU, Dinarello CA. In vitro enhanced thromboxane B2 release by polymorphonuclear leukocytes and macrophages after treatment with human recombinant interleukin 1. Prostaglandins. 1986 Jul;32(1):111-5), we reported for the first time that IL-1 induces thromboxane B2 (TxB2) releases in activated neutrophils and macrophages. An increase in thromboxane can induce leukocyte aggregation and systemic inflammation, which would account for the dramatic thrombi formation and organ dysfunction. Hence, IL-1 stimulates endothelial cell-leukocyte adhesion, and TxB2 production. All these events are supported by the large increase in neutrophils that adhere to the lung and the decrease in lymphocytes. Therefore, ecosanoids such as TxA2 (detected as TxB2) have a powerful action on vascular inflammation and platelet aggregation, mediating the formation of thrombi. The thrombogenesis that occurs in COVID-19 includes platelet and cell aggregation with clotting abnormalities, and anti-clotting inhibitor agents are used in the prevention and therapy of thrombotic diseases. Prevention of or induction of TxA2 avoids thrombi formation induced by IL-1. However, in some serious vascular events where TxA2 increases significantly, it is difficult to inhibit, therefore, it would be much better to prevent its induction and generation by blocking its inductors including IL-1. The inhibition or lack of formation of IL-1 avoids all the above pathological events which can lead to death of the patient. The treatment of innate immune cells producing IL-1 with IL-1 receptor antagonist (IL-1Ra) can avoid hemodynamic changes, septic shock and organ inflammation by carrying out a new therapeutic efficacy on COVID-19 induced by SARS-CoV-2.

  • Editorial
    P. Conti, Al. Caraffa, G. Tetè, C.E. Gallenga, R. Ross, S.K. Kritas, I. Frydas, A. Younes, P.Di Emidio, G. Ronconi
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1629-1632. https://doi.org/10.23812/20-2EDIT
    Download PDF ( ) HTML ( )     Knowledge map   Save

    SARS-CoV-2 virus is an infectious agent commonly found in certain mammalian animal species and today also in humans. SARS-CoV-2, can cause a pandemic infection with severe acute lung injury respiratory distress syndrome in patients with COVID-19, that can lead to patient death across all ages. The pathology associated with pandemic infection is linked to an over-response of immune cells, including virus-activated macrophages and mast cells (MCs). The local inflammatory response in the lung that occurs after exposure to SARS-CoV-2 is due to a complex network of activated inflammatory innate immune cells and structural lung cells such as bronchial epithelial cells, endothelial cells and fibroblasts. Bronchial epithelial cells and fibroblasts activated by SARS-CoV-2 can result in the up-regulation of pro-inflammatory cytokines and induction of MC differentiation. In addition, endothelial cells which control leukocyte traffic through the expression of adhesion molecules are also able to amplify leukocyte activation by generating interleukin (IL)-1, IL-6 and CXC chemokines. In this pathologic environment, the activation of mast cells (MCs) causes the release of histamine, proteases, cytokines, chemokines and arachidonic acid compounds, such as prostaglandin D2 and leukotrienes, all of which are involved in the inflammatory network. Histamine is stored endogenously within the secretory granules of MCs and is released into the vessels after cell stimulation. Histamine is involved in the expression of chemokine IL-8 and cytokine IL-6, an effect that can be inhibited by histamine receptor antagonists. IL-1 is a pleiotropic cytokine that is mainly active in inflammation and immunity. Alveolar macrophages activated by SARS-CoV-2 through the TLR produce IL-1 which stimulates MCs to produce IL-6. IL-1 in combination with IL-6 leads to excessive inflammation which can be lethal. In an interesting study published several years ago (by E. Vannier et al., 1993), it was found that histamine as well as IL-1 are implicated in the pathogenesis of pulmonary inflammatory reaction, after micorganism immune cell activation. IL-1 in combination with histamine can cause a strong increase of IL-1 levels and, consequently, a higher degree of inflammation. However, it has been reported that histamine alone has no effect on IL-1 production. Furthermore, histamine enhances IL-1-induced IL-6 gene expression and protein synthesis via H2 receptors in peripheral monocytes. Therefore, since MCs are large producers of histamine in inflammatory reactions, this vasoactive amine, by increasing the production of IL-1, can amplify the inflammatory process in the lung infected with SARS-CoV-2. Here, we have proposed for the first time an emerging role for histamine released by MCs which in combination with IL-1 can cause an increase in lung inflammation induced by the viral infection SARS-CoV-2.

  • Editorial
    T.C. Theoharides, P. Conti
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1633-1636. https://doi.org/10.23812/20-EDIT3
    Download PDF ( ) HTML ( )     Knowledge map   Save

    COVID-19 derives from infection with Coronavirus [severe acute respiratory syndrome (SARS)-CoV-2] and is associated with high morbidity and mortality due to release of a storm of pro-inflammatory cytokines and thrombogenic agents resulting in destruction of the lungs. Many reports indicate that a considerable number of patients who are positive for SARS-CoV-2 are asymptomatic or have mild symptoms. However, increasing evidence suggests that many such patients who either recovered from or had mild symptoms after COVID-19 exhibit diffuse, multiorgan, symptoms months after the infection. These symptoms include malaise, myalgias, chest tightness, brain fog and other neuropsychiatric symptoms that were originally reported in children and named Multisystem Inflammatory Syndrome (MIS-C). Now the US Center for Disease Control (CDC) announced the recognition of a similar condition in adults, named Multisystem Inflammatory Syndrome (MIS-A). The symptoms characterizing these conditions are very similar to those associated with Mast Cell Activation Syndrome (MCAS, US ICD-110 code D89.42-idiopathic mast cell activation syndrome). Hence, the possibility of MCAS should be evaluated in any patient with MIS and/or multisystem inflammatory symptoms. In either case, these syndromes should be addressed with liposomal formulation (in olive pomace oil) of the flavone luteolin (e.g. PureLut® or FibroProtek®) together with the antihistamine rupatadine, which also has anti-platelet activating factor (PAF) activity and inhibits mast cells that have been implicated in the pathogenesis of cytokine storms in COVID-19.

  • Article
    R-H. Gu, B. Tan, J. Ma, W-Y. Shen, Y-S. Zuo, L. Shi
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1637-1646. https://doi.org/10.23812/20-34-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    This study aims to investigate the value of the combined detection of carcinoembryonic antigen (CEA), Neuron-specific enolase (NSE) and the level of Interleukin-18 (IL-18) in the serum in the diagnosis of lung cancer. The correlation between these parameters and the expression levels of B-cell lymphoma-2 (Bcl-2) protein were also studied. Eighty patients with lung cancer were included in the lung cancer group. These patients underwent surgery in the Department of Oncology of Huai'an Second People's Hospital between February 2016 and February 2018. During the same period, another 80 patients with benign lung lesions were registered in the benign lesion group and 80 healthy people were enrolled in the control group. Enzyme-linked immunosorbent assay (ELISA) was used to detect the expression levels of CEA, NSE and IL-18. The diagnostic critical value of these factors was used as positive indicator. When CEA, NSE and IL-18 levels were positive at the same time, the combined detection was considered to be positive. WB was used to detect Bcl-2 expression level. We also analyzed the possible correlation between CEA, NSE, IL-18 levels and the Bcl-2 expression levels. The CEA, NSE and IL-18 expression levels in the serum of the lung cancer group were significantly higher than those in the benign lesion and the control groups (p< 0.05). The area under ROC curve for CEA, NSE and IL-18 respectively was 0.770 (0.697-0.843), 0.829 (0.767-0.890), 0.721 (0.642-0.800) (p< 0.001). IL-18 level was negatively correlated with the level of Bcl-2 mRNA in the tissue (r=-0.380, p< 0.001). In conclusion, CEA, NSE and IL-18 have a good auxiliary diagnostic value in patients with lung cancer. The combined detection could improve the sensitivity and specificity of lung can¬cer diagnosis. There was a negative correlation between IL-18 and Bcl-2 levels which suggested a potential inhibitory role of IL-18 on the lung cancer cells apoptosis pathway.

  • Article
    C-R. Chen, H-L. Bi, X. Li, Z-M. Li
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1647-1656. https://doi.org/10.23812/20-169-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    The effects of remifentanil were studied on cerebral ischemia-reperfusion injury (IRI) in rats, and its regulatory effect was explored on the N-methyl D-aspartate receptor subtype 2B (NR2B)/calcium/calmodulin-dependent protein kinase type II subunit alpha (CaMKII) signaling pathway in cerebral tissues. A total of 40 Sprague-Dawley rats were randomly assigned into sham group, model group, low-dose group (remifentanil injected into the caudal veins at 2 μg/kg) and high-dose group (remifentanil injected into the caudal veins at 10 μg/kg). Then, in the model, low-dose and high-dose groups, the rat model of cerebral IRI was established through middle cerebral artery occlusion (namely, ischemia for 1 h and reperfusion for 2 h), while no thread was inserted into the rats in the sham group. Neurological function of rats in each group was evaluated, and the cerebral infarct size was measured using 2,3,5-triphenyltetrazolium chloride (TTC) staining. Moreover, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining was performed to determine the neuronal apoptosis level, and mitochondrial membrane potential was measured via JC-1 assay. Moreover, the reactive oxygen species (ROS) level in neurons and the malondialdehyde (MDA) content in cerebral tissues were determined using 2',7'-dichlorofluorescin diacetate (DCFH-DA) assay and enzyme-linked immunosorbent assay (ELISA), respectively. Finally, the expression levels of apoptosis-associated proteins and the NR2B/CaMKIIα signaling pathway-associated proteins in cerebral tissues were measured by Western blotting. Remifentanil preconditioning substantially decreased the neurological score of rats (p<0.01), cerebral infarct size (p<0.01), neuronal apoptosis level (p<0.01), ROS level in neurons (p<0.01), MDA content (p<0.01) and expression levels of cysteinyl aspartate specific proteinase-3 (Caspase-3), NR2B, phosphorylated CaMKIIα (p-CaMKIIα) and p-cAMP responsive element binding protein (p-CREB) (p<0.01), but it increased the mitochondrial membrane potential (p<0.01) and B-cell lymphoma 2 (Bcl-2)/Bcl-2-associated X protein (Bax) ratio (p<0.01). Remifentanil can repress the NR2B/ CaMKIIα signaling pathway in the neurons of rats with cerebral IRI to decrease the p-CREB expression, ROS level and MDA content in neurons, neuronal apoptosis level and cerebral infarct size, and increase the mitochondrial membrane potential, thereby protecting the neurological function.

  • Article
    H-N. Fan, X-H. Liao, J. Zhang, H-M. Zheng
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1657-1668. https://doi.org/10.23812/20-210-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Infiltration of macrophages is associated with tumor progression and poor prognosis in multiple malignancies, but the underlying mechanisms by which macrophages contribute to colorectal cancer (CRC) have not yet been elucidated. The purpose of this study was to discuss the potential mechanisms of macrophages in CRC. The MTT assay was used to assess cell viability. The expression of the proliferation-related marker PCNA was detected by Western blot analysis. The 10 most important factors (PDGF, VEGF, TNFα, bFGF, IL-8, TGF-β, IFN-γ, SPARC, IL-1β and IL-6) secreted by macrophages were knocked down by RNA interference (RNAi), and the mRNA expression levels of these 10 factors were analyzed by qRT-PCR. The effect of these factors on cell proliferation was assessed by the MTT assay. The miRNAs regulated by IL-1β in CRC cells were identified by miRNA microarray and qRT-PCR analyses. The proliferation ability of miR-28-3p inhibitor on CRC cells was detected by colony formation assay. The association of IL-1β and miR-28-3p expression with the clinicopathological characteristics in patients with CRC was analyzed by TCGA RNA-seq data. As a result, macrophages promoted the proliferation of CRC cells in a time- and number-dependent manner, and these effects were associated with the upregulation of PCNA and the macrophage-secreted cytokine IL-1β, which had the most significant effect on CRC cell proliferation. Furthermore, downregulation of miR-28-3p was induced by IL-1β in CRC cells. The miR-28-3p inhibitor promoted the proliferation in CRC cells. Moreover, upregulation of IL-1β expression or downregulation of miR-28-3p expression was associated with poor survival in patients with CRC. Therefore, these data demonstrated that macrophages promoted CRC cell proliferation via IL-1β-mediated downregulation of miR-28-3p.

  • Article
    Q-Y. Tang, J-X. Wei, S-F. Xue, G-H. Liu, L-X. Fu
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1679-1688. https://doi.org/10.23812/20-252-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Acute lung injury (ALI)/Acute respiratory distress syndrome (ARDS) is a very dangerous disease. The purpose of this study was to investigate the effects of fibrogrowth factor-2 (FGF-2) on lipopolysaccharide (LPS)-induced lung injury and its mechanisms. C57/BL6 mice were used in the study and LPS was used to construct the ALI/ARDS model. In addition, human normal lung epithelial cell line BEAS-2B was cultured to investigate the effect of FGF-2 on the lung and its mechanism of action in vitro. FGF-2 significantly reduced wet/dry weight ratio of mice, the number of cells and inflammatory factors in BALF, and MPO activity in lung tissue. In addition, FGF-2 also reduced the level of oxidative stress in mouse lung tissue. In vitro, FGF-2 effectively reduced LPS-induced inflammatory and apoptotic levels of BEAS-2B cells and increased the activity of the PI3K/Akt signaling pathway. However, LY294002, an inhibitor of the PI3K/Akt signaling pathway, alleviated the protective effect of FGF-2 on lung tissue. Therefore, FGF-2 attenuated inflammation, oxidative stress and apoptosis in alveolar epithelial cells by activating the PI3K/Akt signaling pathway.

  • Article
    H. Grar, W. Dib, H. Gourine, H. Negaoui, BHF. Taleb, A. Louaar, S. Ouldhocine, H. Kaddouri, O. Kheroua, D. Saidi
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1689-1697. https://doi.org/10.23812/20-24-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Increased intestinal permeability due to barrier dysfunction is supposed to cause several gastrointestinal diseases. We have previously demonstrated that a single β-carotene (BC) dose protects against increase in anaphylactic response in β-lactoglobulin (BLG)-sensitized mice with no effect on the epithelial permeability and weak recovery of villi length. Utilizing the same murine ex vivo intestinal model, the aim of this study was to investigate the effect of different BC doses on BLG-mediated intestinal epithelial barrier disturbances. Jejunum was harvested from BLG-sensitized mice pretreated with either one of three different doses of BC (5, 10 and 20 mg/ kg body weight) and mounted on Ussing Chambers. Transepithelial electrical resistance (TER) and short-circuit current (Isc) were recorded as indicators of intestinal epithelial barrier function. Histopathological analysis of the intestine was carried out for the control and experimental mice. TNF-α and IL-6 levels were determined in serum using ELISA, and the analysis of antioxidant activity was performed for reduced glutathione (GSH) and thiobarbituric acid reactive substances (TBARS). BC was capable of enhancing the intestinal barrier function, as indicated by the increased TER and the decreased Isc. Intestinal damage characterized by the shortening of villi and infiltration of intestinal lymphocytes was significantly reversed by BC pretreatment. Such effects of BC were accompanied by a reduction in the levels of IL-6 and TBARS and an increase of GSH. TNF-α levels were reduced only at the lowest BC dose. These findings may encourage the use of BC-based therapies for controlling the breakdown of the intestinal barrier in vivo.

  • Article
    XZ. Wang, SF. Zhang, ZH. Yang, ZW. Ye, J. Liu
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1699-1708. https://doi.org/10.23812/20-23-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Osteosarcoma is the most prevailing malignant bone tumor among adolescents. Punicalagin, a polyphenolic compound extracted from pomegranate, possesses many functions such as anti-oxidation, anti-bacterial, anti-virus, and immunosuppression, which can counter the aggressiveness of a variety of cancers such cervical, ovarian and prostate. This study aimed to investigate the inhibitory effect of punicalagin on the proliferation and metastasis of osteosarcoma cells and its potential regulatory mechanisms. Osteosarcoma cell lines (HOS cells, U2OS cells and MG63 cells) were treated with different doses of punicalagin, and the effects on osteosarcoma cell activity were examined in vitro using cell counting kit-8 (CCK-8), colony formation and apoptosis assays. The mobility, migration and invasion abilities of osteosarcoma cells were detected by wound healing and Transwell assays. NF-κB activity was explored by the NF-κB p65 luciferase reporter assay. Western blot was used to investigate the expressions of downstream proteins. We found that punicalagin inhibited the viability of osteosarcoma cells in vitro in dose-dependent and time-dependent manners and promoted apoptosis. In addition, punicalagin could significantly impede the mobility, migration and invasion abilities of osteosarcoma cells. In terms of mechanism, punicalagin down-regulated the expressions of p65, survivin, XIAP, CIAP2 and other proteins, and suppressed the proliferation and metastasis of osteosarcoma cells by repressing NF-κB signaling pathway. In conclusion, it is concluded that punicalagin restrains the growth and metastasis of osteosarcoma by obstructing the NF-κB signal transduction pathway.

  • Article
    SS. Zhang, XQ. Kan, P. Liu, LZ. Yin, QY. Li, HY. Xu
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1709-1717. https://doi.org/10.23812/20-231-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Preeclampsia (PE) may induce gestational failure, threatening a significant number of pregnant women. Recently, microRNAs (miRNAs) have been reported to participate in PE progression, whereas the precise functions and potential mechanisms of miR-20b in placental trophoblast cells as well as in PE progression remain poorly understood. In the present study, real-time quantitative polymerase chain reaction (RT-qPCR) analysis was used to detect expressions of miR-20b and myeloid cell leukemin- 1(MCL-1) mRNA. Cell viability was investigated by cell counting kit-8(CCK-8) assays. Cell invasion and migration abilities were determined by Transwell assays. Western blot was performed to detect MCL-1 protein expressions. The interaction between miR-20b and MCL-1 was investigated by bioinformatics analysis and luciferase activity assay. The results of the study demonstrated that miR-20b was highly expressed in placental tissues of patients with PE. Moreover, miR-20b overexpression inhibited HTR8/ SVneo cell proliferation, invasion and migration. Furthermore, MCL-1 was targeted by miR-20b, and MCL-1 restoration could partially attenuate the effect of miR-20b on HTR8/SVneo cells. In conclusion, the results indicate that miR-20b may contribute to PE through inhibiting proliferation, invasion and migration of placental trophoblast cells by targeting MCL-1. Therefore, miR-20b may be used as a notable biomarker for the diagnosis, prevention, and treatment of PE. MiR-20b targeting MCL-1 deserves further investigation in order to explore their potential role in PE.

  • Article
    E.-H. Liu, Z.-N. Zheng, C.-X. Xiao, X. Liu, X.-Q. Lin
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1719-1727. https://doi.org/10.23812/20-326-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    The purpose of this study was to investigate the influence of interleukin(IL)-22 on the Janus kinase/ signal transducer and activator of transcription 3 (JAK/STAT3) signaling pathway and sepsis-induced liver injury in rats. A total of 48 Sprague-Dawley rats were randomly divided into sham-operated group (n=12), model group (n=12), low-dose group (n=12) and high-dose group (n=12). Next, rat models of sepsis-induced liver injury were established through cecal ligation and puncture (CLP). At 12 h after surgery, blood was collected by heart puncture to detect liver function of the rats. It was found that the activity of alanine aminotransferase (ALT) and aspartame aminotransferase (AST) and the content of total bilirubin were reduced in low-dose group and high-dose group. Hematoxylin-eosin (HE) staining results revealed that after treatment with IL-22, the liver injury was relieved compared with model group. Moreover, the results of TUNEL staining assay revealed that the apoptosis level of liver cells declined after treatment with IL-22. Enzyme-linked immunosorbent assay (ELISA) results demonstrated that the levels of IL-6 and TNF-α were reduced, while the level of IL-10 was increased after treatment with IL-22. Moreover, it was discovered that the SOD content was overtly elevated in low-dose and high-dose groups compared with that in the model group. Finally, using Western blotting, it was confirmed that in comparison with the model group, the levels of Bcl-2/Bax and JAK/STAT3 signaling pathway-related proteins were markedly raised, while the level of Caspase-3 was decreased in the low-dose and high-dose groups. In conclusion, IL-22 can improve liver function, reduce the apoptosis level of liver cells, the expression of apoptosis-related proteins and the release of inflammatory factors, and alleviate liver injury by activating the JAK/STAT3 signaling pathway.

  • Article
    G.Di Spigna, P. Ladogana, B. Covelli, M. Ricciardone, S. Salzano, D.Spalletti Cernia, I. Mormile, G. Varriale, O. Catapano, G. Spadaro, M. Mormile, L. Postiglione
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1729-1737. https://doi.org/10.23812/20-297-A
    Download PDF ( ) HTML ( )     Knowledge map   Save

    Molecular characterization of IgE reactivity of specific individual components of allergenic extracts is now possible due to the technology of recombinant allergens derived from studies of molecular biology of allergic pathology. The identification of the immunoreactivity to single allergenic components in allergic subjects allows to specifically define her/his allergic profile and obtain the so-termed Component Resolved Diagnosis (CRD). Molecular allergens can be classified into those that induce the respiratory allergic reactivity and those that identify the food-related allergic pathology. It is also essential to identify those molecular allergens whose immunoreactivity is able to connect the two clinical conditions: respiratory symptoms and food allergy symptoms. The present study was conducted on 50 patients with a clinical history of hypersensitivity to pollen and/or allergy and positivity to Skin Prick Test. The sera were analyzed in our laboratories and the panel of recombinant allergens was applied in the case of positivity of the specific IgE. Of the 50 patients enrolled, 31 were selected as positive to 4 main pan-allergen Bet v1, Par j2, Art v1 and Phl p1; among these, 14 subjects showed one allergen-specific IgE towards natural extracts of tested foods even in absence of clinical history. CRD allows for an increased accuracy in allergy diagnosis and prognosis and plays an important role in: a) resolving genuine vs cross-reactive sensitization in poly-sensitized patients, b) assessing, in selected cases, the risk of severe, systemic vs mild, local reactions in food allergy, and c) identifying patients and triggering allergens for specific immunotherapy (ITS). In light of our results, we believe that the transition from a diagnostic based on the use of allergenic extracts to another one based on the use of single allergenic molecules that is able to define the specific allergenic profile of each patient, seems to be able to revolutionize the allergy diagnosis.

  • Letter
    F. Qu, J. Liu, AP. Yang, RL. Tu
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1739-1744. https://doi.org/10.23812/20-54-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    HX. Shi, XF. Sui, DQ. Wang, QT. Han, FQ. Meng, XZ. Zhao
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1745-1748. https://doi.org/10.23812/20-149-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    H. Wang, X. Zhou, B. Zhang, L. Sun
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1749-1755. https://doi.org/10.23812/20-288-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    L. Zhong, P-P. Qiao, B-L. Wang, C. Liu
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1757-1761. https://doi.org/10.23812/20-261-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    XH. Wang, MZ. Teng, Q. Liu, J. Bao, RJ. Zhuang, XY. Wang
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1763-1769. https://doi.org/10.23812/20-229-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    XY. Mu, Q Chen, J Xie
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1771-1777. https://doi.org/10.23812/20-254-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    T. Lei, B. Gong, Q. Meng, M. Zhang
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1779-1785. https://doi.org/10.23812/20-230-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    C-Q. Song, M. Wang, S-M. Zhang, X-Y. Ma
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1782-1792. https://doi.org/10.23812/20-420-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    L. Ye, YY. Pan, FH. Jin, DH. Zhang, QL. Xu, L. Zhang
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1793-1797. https://doi.org/10.23812/20-189-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    R-D. Sui, H-T. Wang, X-Y. Yu, X-Q. Zhang, X-L. Wei, D-Y. Xin, D-W. Han
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1799-1807. https://doi.org/10.23812/20-351-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    QH. Liu, HL. Li, H. Zhang, Z. Li, M. Zhao, TT. Zhang
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1809-1817. https://doi.org/10.23812/20-265-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    CY. Huang, MY. Li, W. Liu, XX. Li, Y. Xu, JY. Li, QQ. Yao, LM. Wang
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1819-1824. https://doi.org/10.23812/20-105-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    W. Chen, X-L. Shi, X-H He, Y-H. Mao, C. Li, N. Dong
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1825-1829. https://doi.org/10.23812/20-428-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    ZG. Sun, ZF. Lang, YD. Mu, J. Li, CX. Xing, L. Yan, S. Li
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1831-1836. https://doi.org/10.23812/20-228-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    R. Sadoughifar, M. Goldust, J. Scala, M. Fioranelli, N.Van Thuong, M. Tirant, A. Salimi, K.M. Lomonosov, T. Lotti
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1837-1838. https://doi.org/10.23812/20-445-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    HM. Wang, ML. Yan
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1839-1845. https://doi.org/10.23812/20-309-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    XL. Wang, JL. Wang, W. Ma, Y. Jiang, JH. Wan, SJ. Jiang, ZK. He, XJ. Wang, ZY. An, XX. Liu, RJ. Cui
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1847-1855. https://doi.org/10.23812/20-311-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    Y. Chen, L. Liu, YY. Xing, Q. Li, GH. Zhao, YY. Lu, JJ. Yang
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1857-1862. https://doi.org/10.23812/20-321-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Article
    J-R. Lin
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1863-1867. https://doi.org/10.23812/20-151-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    Q. Yang, X.-Y. Wei, X.-H. Tang, X.-Y. Chen
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1869-1873. https://doi.org/10.23812/20-163-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    M. Saeed, M.I. Khan, R. Arshad, M.A. Farooq, M.A. Rehman, A. Ishaque
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1875-1878. https://doi.org/10.23812/20-175-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    X. Deng, F. Li, Y. Li, B. Yang, R. Han, C. Tang, F. Fang
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1879-1884. https://doi.org/10.23812/20-336-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    Y-Y. Hu, D-P. Ji, Y-Y. Yu, Y-J. Li, B. Li, G-H. Hou, X-D. Zhang, C-C. Duan, R-B. Hu, J. Xu
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1885-1888. https://doi.org/10.23812/20-352-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    XF. Qu, Z. Xu, X.F. Lin
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1889-1895. https://doi.org/10.23812/20-317-L.
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    WW. Yang, L. Li, S. Tan, ZY. Liu, Y. Jiang
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1897-1900. https://doi.org/10.23812/20-353-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    XH. Liu, CM. Zhang, PQ. Pan, DQ. Zhang, X. Huang, ZH. Ji
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1901-1908. https://doi.org/10.23812/20-376-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    F-Q. Liu, C-P. Liu, M-H. Xu, J. Zhu, W. Yu, L-L. Wang
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1909-1913. https://doi.org/10.23812/20-429-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    WB. Xu, N. Zhang
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1915-1921. https://doi.org/10.23812/20-334-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    XY. Wu, YL. Zhang, HL. Xia, ZM. Guan, ZY. Liu, WX. Wang, Y. Liu
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1923-1928. https://doi.org/10.23812/20-373-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    C. Caruso, S. Colantuono, G. Celi, L. Salerno, G.De Martino, G. Ianiro, C. Casale, E. Nucera, S.Del Giacco, A. Gasbarrini
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1929-1934. https://doi.org/10.23812/20-162-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    E. Ortu, G. Barrucci, G. Aprile, L. Guerrini, D. Pietropaoli, A. Monaco
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1935-1939. https://doi.org/10.23812/20-141-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    R. Patini, G. Saponaro, M. Todaro, P. Doneddu, P. Gallenzi, P.De Angelis, A. Pelosi, M. Furlani, F. Peverini, B. D'Errico, L. Raffaelli, G. Gasparini
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1941-1948. https://doi.org/10.23812/20-327-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    M. Guelfi, R. Fabbrini, MG. Guelfi
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1949-1953. https://doi.org/10.23812/20-238-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    A. Paladini, G. Guzzardi, C. Stanca, B.Del Sette, S. Tettoni, D. Negroni, M. Cernigliaro, A. Galbiati, M. Spinetta, S. Gentilli, A. Carriero
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1955-1957. https://doi.org/10.23812/20-315-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    T. Filardi, S. Morano, S. Tartaglione, M.G. Porpora, C. Tiberti, A. Lenzi, A. Angeloni, E. Anastasi
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1959-1962. https://doi.org/10.23812/20-365-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    A. Saggini, M. Cristinziano, C. Ljoka, S. Salini, P.E. Ferrara, G. Ronconi, A. Orlandi, C. Foti
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1963-1965. https://doi.org/10.23812/20-385-L
    Download PDF ( ) HTML ( )     Knowledge map   Save
  • Letter
    F. Gazzani, C. Pavoni, C. Danesi, G. Gastaldi, R. Lione
    Journal of Biological Regulators and Homeostatic Agents. 2020, 34(5): 1967-1970. https://doi.org/10.23812/20-399-L
    Download PDF ( ) HTML ( )     Knowledge map   Save