Open Access

Interleukin‑6 signalling as a valuable cornerstone for molecular medicine (Review)

  • Authors:
    • Maria Trovato
    • Salvatore Sciacchitano
    • Alessio Facciolà
    • Andrea Valenti
    • Giuseppa Visalli
    • Angela Di Pietro
  • View Affiliations

  • Published online on: April 16, 2021     https://doi.org/10.3892/ijmm.2021.4940
  • Article Number: 107
  • Copyright: © Trovato et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The biological abilities of interleukin‑6 (IL‑6) have been under investigation for nearly 40 years. IL‑6 works through an interaction with the complex peptide IL‑6 receptor (IL‑6R). IL‑6 is built with four α‑chain nanostructures, while two different chains, IL‑6Rα (gp80) and gp130/IL6β (gp130), are included in IL‑6R. The three‑dimensional shapes of the six chains composing the IL‑6/IL‑6R complex are the basis for the nanomolecular roles of IL‑6 signalling. Genes, pseudogenes and competitive endogenous RNAs of IL‑6 have been identified. In the present review, the roles played by miRNA in the post‑transcriptional regulation of IL‑6 expression are evaluated. mRNAs are absorbed via the ‘sponge’ effect to dynamically balance mRNA levels and this has been assessed with regard to IL‑6 transcription efficiency. According to current knowledge on molecular and nanomolecular structures involved in active IL‑6 signalling, two different IL‑6 models have been proposed. IL‑6 mainly has functions in inflammatory processes, as well as in cognitive activities. Furthermore, the abnormal production of IL‑6 has been found in patients with severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2; also known as COVID‑19). In the present review, both inflammatory and cognitive IL‑6 models were analysed by evaluating the cytological and histological locations of IL‑6 signalling. The goal of this review was to illustrate the roles of the classic and trans‑signalling IL‑6 pathways in endocrine glands such as the thyroid and in the central nervous system. Specifically, autoimmune thyroid diseases, disorders of cognitive processes and SARS‑CoV‑2 virus infection have been examined to determine the contribution of IL‑6 to these disease states.

1. Introduction

Interleukin-6 (IL-6) was first identified as a factor derived from T-helper type 2 (Th2) lymphocytes >40 years ago (1). On the basis of the biological abilities of IL-6 to stimulate B-cell differentiation, the interleukin was categorised among the B-cell stimulating factors (BSFs) and B-cell differentiation factors (BCDFs) (2). As a member of the BSFs, IL-6 was named BSF-2 and grouped together with BSF1 and interleukin-4 (2). IL-6 was included in the group of BCDFs due to its capacity to stimulate the secretion of IgM and IgG in B cells (3). After the nomenclature meeting held in New York at the end of 1988, BCDF/BSF-2 was finally referred to as IL-6 (4), as the biochemical properties of this factor showed an isoelectric point between 5 and 6 (2).

Over the last 40 years, several molecular features of IL-6 have been identified. Furthermore, new abilities of IL-6 have prompted its use as a target in medical practice for infective and cancerous diseases, including COVID-19. The present review highlights the current knowledge on the molecular and nanomolecular structures involved in active IL-6 signalling. By examining both inflammatory and cognitive IL-6 models, new properties of the IL-6 cytokine have been evaluated. Specifically, the cytological and histological locations of IL-6 signalling have been analysed together with serum concentrations of IL-6 in order to distinguish between the classic and trans-signalling IL-6 pathways.

2. Three-dimensional shapes of chains involved in nanomolecular IL-6 signalling

Molecular analysis reveals that the human IL-6 gene is localized on the short arm of chromosome 7 (5). Depending on the genetic approach, IL-6 has been mapped to either the p21 or p15.3 region of the chromosome (6). By expanding from 22,725,889 to 22,732,002 base pairs, four introns and five exons were cloned in both human and mouse IL-6 genes (https://ghr.nlm.nih.gov/gene/IL6#location) (7). A polymorphic locus, Rs1800796, has been identified in the IL-6 promoter region (8). The recognition of genetic mutations in this genomic sequence has been used to assess human cancer risk (8).

In the 212-amino acid human IL-6 glycoprotein, 28 amino acids are linked to peptide signalling (https://www.genecards.org). The molecular mass of IL-6 is 23,718 Da, ranging between 21 and 30 kDa (https://www.genecards.org) (9-11).

The IL-6 topology is composed of a secondary structure that includes helicoidal motives related to four long α-chains. Via a bundle model, these α-chains are structured in three-dimensions to achieve the tertiary shape (9,12-14).

The bundle helicoidal complex is also observed also in a number of human cytokines such as IL-11, oncostatin M (OSM), ciliary neurotrophic (CNTF), leukemic inhibitory (LIF), cardiotrophin-like factor 1 (CT-1), erythropoietin, granulocyte colony-stimulating factor, IL-12, growth hormone, prolactin, IL-10, interferon and leptin (12). However, despite these factors adopting structures similar to the IL-6 bundle prototype model, they show little identity with the IL-6 amino acid sequence (12). By contrast, a viral homolog to the human IL-6 protein has been identified in herpesvirus 8 associated with Kaposi's sarcoma (15). This amino acid sequence is capable of arranging itself in a bundle helicoidal structure. This association is the reason that the viral protein is named viral IL-6 (15).

IL-6 works through interaction with the complex peptide IL-6 receptor (IL-6R). This receptor includes two transmembrane glycoproteins referred to as the IL-6Rα and gp130/IL6ß (gp130) subunits (16,17). Two different active genes contribute to the generation of human IL-6R.

Originally, the IL-6Rα gene was mapped on the long arm of chromosome 1 in the q21.3 cytoband (18) (Fig. 1). By counting 13 exons, this was cloned in 64,258 bases (https://www. genecards.org). The gene that codes for Gp130 is located on chromosome 5 at band q11 (19) (Fig. 1).

The IL-6Rα transcript encodes a modular glycoprotein made up of one α-chain with a size of 80 kDa, also known as IL6Q, gp80, CD126, IL6RA, IL6RQ, IL-6RA or IL-6R-1 (18). Specifically, IL-6Rα acts by binding the IL-6 ligand; however, this activity is not enough to transduce any signal (18). By contrast, the gp130 glycoprotein chain is unable to directly bind IL-6, but is capable of IL-6 signal transduction (19).

The assembly of IL-6 with its respective receptors occurs through a unique two-phase process. First, the four α-chains of IL-6 capture the α-chain of IL-6R with a dissociation constant (Kd) of ~1 Nm (20) (Fig. 2). In this stage, IL-6, composed of a dimeric structure, does not perform any signalling activity (21,22). The next assembly step is the construction of a hexameric cluster, where the complex of five α-chains binds gp130 with a Kd of ~10 pM (17,20) (Fig. 2). The previous binding of IL-6 with IL-6R occurs with lower affinity than that with the complex and gp130. In this second stage, the IL-6 complex is composed of a trimeric structure that, like in the first step, does not perform any signalling. To begin signalling, the IL-6/IL-6Rα/gp130 trimer proceeds through a homodimerization process that forms a hexameric complex (22).

These data suggest that the transition from low to high affinity IL-6 binding occurs due to phenomena pertaining to nanoparticle (NP) spheres. In fact, the geometric shape of the complex becomes crucial for the efficiency of binding at the nanometre scale (21,23). The five α-chains of the IL-6/IL-6Rα complex are only suitable for binding in a pentameric orientation. The hexamer is the competent form for energetic transition leading to the dimerization of the IL-6/IL-6Rα/gp130 cluster. Notably, the pentameric complex of α-chains, corresponding with the dimeric form of the IL-6/IL-6Rα structure, may appear either in the serum or in cellular compartments (Fig. 2). By contrast, the clusters of IL-6/IL-6Rα/gp130 are closed in cellular structures. The IL-6/IL-6Rα/gp130 complex is found in both the non-signalling trimer and signalling hexamer forms (Fig. 2) (23).

The trimeric model of IL-6 signalling is replicated in several other cytokines, including IL-11, LIF, OSM, CNTF and CT-1. This is due to their ability to bind gp130 on the cellular surface to elicit signal transduction (24). For these reasons, the physiological responses of these cytokines could occur simultaneously. These cytokines have been included in the group of L-6-type cytokine receptor mediators (24-26).

There are two main ideas that are inspired by the assessment of the nanomolecular structure of IL-6 signalling: Firstly, the nanomolecular shapes of the IL-6 system are largely independent of genetic composition. Therefore, genetic investigation has to be associated with nanomolecular evidence to completely track the physiological and pathological signals of IL-6. Secondly, the efficacy of IL-6 therapeutic targets is also dependent on the geometric shape of IL-6 signalling structures. Therefore, prior to determining the clinical benefits of IL-6 therapy, the nanomolecular conformations of IL-6 signalling have to be estimated.

3. Genes, pseudogenes and competitive endogenous RNA (ceRNA) for molecular control of IL-6 signalling

In contrast to the parental genes composing IL-6R, a distinct pseudogene was demonstrated for IL-6Rα and the gp130 gene (https://www.genecards.org) (19) (Fig. 2). These pseudogenes share much of their sequences with their corresponding parental genes (19). Conversely, pseudogene transcripts are equivalent to non-coding RNA or to antisense RNA and therefore are unable to produce biologically active proteins (27).

The IL-6Rα pseudogene, IL-6RP1, is found on the long arm of chromosome 9 at the locus q22.2 (Fig. 2). A repetition of gp130 sequence, IL-6STP1, is detected on the short arm of chromosome 17 and assigned at the p11 region (Fig. 2); this corresponds with a gp130 non-transcribed pseudogene (19).

IL6-STP1 transcripts have been shown to be involved in microbial defence processes (28). Via activation of the IL-6 family cytokines, IL6-STP1 stimulates inflammatory cells to secrete acute-phase proteins (APP), including fibrinogen, a1-antitrypsin and hepcidin (28). In a mouse polymicrobial infection model, removal of IL-6STP1 caused inhibition of APP production associated with the dysregulation of the inflammatory response and an increase in mortality (28).

Pseudogene RNAs are part of an intricate network of competitive endogenous RNAs (ceRNAs) where all non-coding RNAs are assigned to two large groups: microRNAs (miRNAs) and long non-coding RNAs (27,29,30). miRNAs have <200 nucleotides, while long non-coding RNAs are >200 molecular bases. However, by using the same code, the full set of ceRNAs competes with parental mRNA (31).

ceRNAs absorb mRNA as a 'sponge' to dynamically balance mRNA levels for protein transcription efficiency (27,32). As a consequence, the ceRNA matrix serves to regulate protein expression (27,32). It is likely that ceRNAs mimic an ancient anti-viral defence biomechanism that appeared during the evolutionary scale of eukaryotic species such as those in plants (33). For these reasons, ceRNAs are the cornerstone of recent molecular strategies that use 'silencing' genes to test cancerous biological aggressiveness, as well as to develop innovative molecular therapeutic approaches for cancer (27,34-37).

Data on the role of miRNA in the post-transcriptional regulation of IL-6 expression are gradually increasing (38). In line with the algorithms of miRanda, MicroCosm v5 and the TargetScan v7.1 database (http://multimir.org), 15 miRNAs profiles have been recorded to have potential involvement with IL-6 expression (38,39) (Table I).

Table I

Data source for mioRNA profiles involved in the regulation of interleukin 6 expression.

Table I

Data source for mioRNA profiles involved in the regulation of interleukin 6 expression.

miRNA(Refs.)
hsa-let-7a(38)
hsa-let-7d(38)
hsa-let-7e(38)
hsa-let-7f(38)
hsa-let-7g(38,39)
hsa-let-7i(38)
hsa-miR-23a(38)
hsa-miR-23b(38)
hsa-miR-26a(38)
hsa-miR-26b(38)
hsa-miR-126(38)
hsa-miR-132(38)
hsa-miR-155(38)
hsa-miR-142-3p(38)
hsa-miR-146-a(38)

[i] hsa, Homo sapiens; let, lethal; miR/miRNA, microRNA.

In non-cancerous cellular models of polymorphonuclear leukocytes (PMNs) taken from cord blood and adult blood, post-transcriptional regulation of IL-6 expression was demonstrated by lethal 7g (let-7g) and miR-142-3p modulation (38). The let-7g gene has been located on chromosome 3 at loci p21 (40,41). This is a genomic region involved in carcinogenic processes of the lung (40,41). miR-142-3p has been associated with colorectal cancer and has been recorded on chromosome 17 at loci q22 (41). Let-7g and miR-142-3p have been found to be related with IL-6 expression, as both downregulate the production of IL-6 mRNA as well as protein (38). The role of miR-142-3p with regard to the endogenous expression of IL-6 has been previously predicted by mouse models (42).

In a cancerous cellular model and in normal controls, IL-6 overexpression was combined with decreases in let-7c, let-7d, let-7f, let-7g and mir-98 (43). Furthermore, all of these miRNAs were expressed in higher concentrations in the normal controls than in the cancerous cells (43). Finally, the let precursor let-7c exhibited a similar effect to let-7g in normal PMNs, as let-7c downregulated the mRNA and protein expression of IL-6 in the cancerous cells and controls (38,43).

In summary, the ceRNAs involved in IL-6 signalling play roles in the regulation of IL-6 expression in physiological processes and even in cancerous transformation of cells.

4. Inflammatory and cognitive models of IL-6 cytokines

Since the identification of IL-6 in stromal, epithelial and muscle cells, new roles and functions have been attributed to this cytokine. It is clear that IL-6 could operate through paracrine, autocrine and endocrine mechanisms (44,45). As long as IL-6 signalling is detected in the endocrine and nervous systems, the model of function for the IL-6 inflammatory cytokine is changed; IL-6 may have roles in the regulation of endocrine secretion and in nervous impulse propagation (46,47).

IL-6 inflammatory model

IL-6 controls the inflammatory response primarily through orchestration of pro-inflammatory and anti-inflammatory effects (48,49). This is due to the activation of two different IL-6 pathways. The first is known as classic signalling, which operates in support of anti-inflammatory effects. Gp80 and gp130 are triggered through serum-derived free IL-6 in the cellular compartment (49). This pathway is dependent on cellular expression of the IL-6R components and the concentration of free IL-6 in the serum (Fig. 1) (49). The second trans-signalling pathway, promotes pro-inflammatory activities via IL-6 (49). In the serum compartment, free IL-6 recruits gp80 and the gp80/IL-6 complex activates cellular gp130 (Fig. 1) (49). Therefore, during trans-signalling with IL-6, a balance is maintained between the amount of serum gp80/IL-6 complex and the cellular expression of gp130 (49).

Expression of IL-6 associated with cognate receptor IL-6-Rα has been detected in cancerous and autoimmune endocrine diseases of the thyroid such as Grave's disease (GD) and Hashimoto's thyroiditis (HT) (50-53). In ex vivo pathological tissue, thyroid follicular cells exhibited intracellular IL-6 and IL-6-Rα. Greater immunoexpression of IL-6 and IL-6R were reported in cancerous follicular cells, as well as in HT and GD cases with high lymphoid infiltration (50-53). Simultaneous expression of receptor and ligand was not observed in normal follicular thyroid cells (50-53). To characterize classic and trans-signalling IL-6 in HT patients, free IL-6 and bound gp80/IL-6 complex levels were measured in the serum (48,49). Both IL-6 pathways appeared to be involved in the development and progression of HT. This was due to an increase in bound gp80/IL-6 in the serum of HT patients compared with that of healthy controls (48). Furthermore, free IL-6 was a candidate for an early diagnostic marker for the development of autoimmune disease in the HIV-seropositive (HIV+) population (49). This was due to a high concentration of free IL-6 in the serum, which was correlated with the occurrence of autoimmune disease in HIV+ subjects.

The inflammatory IL-6 model is frequently used to explain IL-6 modulation of the response to toxicity of nanomaterials (54). In vitro and in vivo studies have underlined the pro-inflammatory effects of several natural and synthetic NPs. Due to their capacity for internalization, inhalation of NPs actives several pathways that, in addition to causing apoptosis, fibrosis, genotoxicity and tumourigenesis, also causes strong inflammation at the lung level and beyond (55-59). Briefly, following phagocytosis by macrophages, the NPs trigger the response of other immune cells. The macrophages also release the inflammasome NLRP3, a multiprotein complex whose activation is prompted by numerous different signals, including pathogen-associated molecular patterns and danger-associated molecular patterns (60,61). NLRP3 is activated by reactive oxygen species (ROS) overproduction and the inflammatory cascade continues since NLRP3, in turn, induces the expression of IL-6, IL-1β and TNF-α genes (62).

In the central nervous system, the NP induction of microglia activation causes the onset and progression of chronic brain inflammation, leading to a loss of neuronal cells and an increase in white matter abnormalities; this is associated with an increased risk for autism spectrum disorders, a lower IQ in children, neurodegenerative diseases, such as Parkinson's disease (PD), Alzheimer's disease and multiple sclerosis, and strokes (63). As demonstrated by Visalli et al (57) in the differentiated SH-SY5Y neuronal model, the exposure to synthetic NPs significantly increased transcript levels of IL-6, IL-1β and TNF-α, confirmed by the measurement of cytokine levels detected in the cell supernatants. The role of neuroinflammation and microglia activation in neurotoxicity was detected in an in vivo study using cortical stereotactic injection of carbon-based NPs into the mouse brain (64). According to Bussy et al (65) the brain region-specific sensitivity to NP exposure is most likely related to the number of microglial cells in the different brain regions.

IL-6 cognitive model

Two main sources have contributed to set the cognitive model of IL-6 cytokines: The cellular distribution and histological localization of IL-6/IL-6R signalling in brain tissue; and the different neurological responses to IL-6 serum concentrations due to activation of either the classic or trans-signalling pathway (66,67).

Normal neurons and microglia are able to secrete IL-6 polypeptide, as well as transcribe genomic IL-6-Rα mRNA (66). IL-6 has been implicated in the pathogenesis and cure of PD. Environmental exposure to pesticides also causes degeneration of dopaminergic neurons by activation of inflammatory cytokines such as IL-6 (68,69). The alteration of dopaminergic transmission produces a complex symptomology due to impairment of motor and cognitive performance. Several new medicinal plant extracts and phytochemicals, such as ellagic acid, are potentially suitable for use to alleviate PD symptoms due of their ability to decrease the anti-inflammatory activities of cytokines in cellular models (70,71)

Conversely, IL-6-Rα sequences have uniquely been detected in microglia and remain undetected in oligodendrocytes and astrocytes (66,72). In human and rat brain tissues, IL-6 was mainly localized in the hippocampal region (67,73,74). Several reports have associated the grey matter of the human brain with cognitive processes such as memory consolidation and learning, appearance of depression, post-traumatic stress and childhood maltreatment disorders (74-76). Under physiological conditions, the left hippocampus showed an association between decreased grey matter volume and an increase in serum IL-6 (74). An increase in right hippocampal volumes involving the head, parahippocampal gyrus and dorsal parts of the amygdala were associated with the IL-6 polymorphism rs1800795 (76).

In neurons and oligodendrocytes, the IL-6 response was mediated by trans-signalling (66). In microglia, the IL-6 classic and trans-signalling pathways were observed (67). In target brain cells neurons, IL-6 trans-signalling promoted neuronal degeneration (77,78). By contrast, the regeneration of neural tissue was mediated through IL-6 classic signalling via the involvement of microglia cells (66,78).

In summary, the classic and trans-signaling pathways are the basis of two models, the inflammatory and cognitive models, with cellular expression and serum levels of IL-6 used to distinguish between them.

5. Role of IL-6 in COVID-19

IL-6 is a cytokine with a number of different functions that plays a role in the host acute response to inflammation; it modulates host defence through numerous different mechanisms and actions directed towards monocytes, B cells and controlling homeostasis between Th1 and Th2 activity (75-79).

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2; also known as COVID-19) primarily attacks airway and alveolar epithelial cells, vascular endothelial cells and macrophages in the lung, where there is expression of its host target receptor, angiotensin-converting enzyme 2 (80). Since IL-6 is relevant during infection that affects the mucosal sites, particularly at the upper and lower respiratory tract levels, it represents one of most important cytokines involved in the host response to SARS-CoV-2 infection (81). Severe COVID-19-induced pneumonia is marked by hyperactivation of the immune system and especially by excessive production of IL-6 (82,83). In this regard, several studies have revealed a strong correlation between high systemic levels of IL-6 and respiratory failure in severely affected patients with COVID-19 (84). According to these studies, a 2.9-fold higher mean IL-6 serum concentration was observed in patients with complicated COVID-19 compared with that in patients presenting with non-complicated disease. The strong correlation between IL-6 and COVID-19 has led to the consideration of serum IL-6 levels as potential diagnostic markers, disease severity stratification indicators and prognostic indexes (85,86). Moderately elevated IL-6 levels (>80 pg/ml) were, in fact, sufficient to identify COVID-19-infected patients and to predict progression towards respiratory failure. In addition, IL-6 has emerged as the most significant predictor of mortality in patients with COVID-19 (87).

The pathophysiological role of IL-6 in COVID-19 is well documented by several studies (83,88). IL-6 is generally considered the key driver of the hyperinflammatory process in COVID-19; it exerts effects on numerous different cellular targets that express a functional IL-6R, including T cells, B cells, vascular endothelial cells, monocytes and hepatocytes (79). By means of these actions on such a wide array of cellular targets, IL-6 mediates key effects on cellular immunity, exerting, at the same time, pro-inflammatory and anti-inflammatory functions (83,88).

The relevance of IL-6 is also demonstrated by the numerous studies that have explored its potential utility as a potential therapeutic target (89-93). The efficacy of a number of targeted monoclonal antibodies, directed against IL-6 or its receptor, is currently under investigation in several different countries. Numerous interventional clinical trials are, in fact, currently ongoing, using drugs that target IL-6, such as tocilizumab (Actemra) (38 interventional clinical trials, none of them yet with final and published results; https://www.clinicaltrials.gov/ct2/results?cond=COVID-19+and+tocilizumab&age_v=&gndr==&type=Intr&rslt==&Search=Apply; accessed on 28 march 2021), sarilumab (Kevzara) (9 interventional clinical trials, none of them yet with final and published results; https://www.clinicaltrials.gov/ct2/results?recrs=&cond=COVID-19+and+Sarilumab&term==&cntry=&state==&city=&dist=; accessed on 28 March 2021), siltuximab (Sylvant) (1 interventional clinical trial; ClinicalTrials.gov identifier, NCT04329650) and clazakizumab (formerly ALD518 and BMS-945429) (6 interventional clinical trials, none of them yet with final and published results; https://www.clinicaltrials.gov/ct2/results?cond=COVID-19+and+Clazakizumab&age_v=&gndr=&type=Intr&rslt=&Search=Apply; accessed on 28 March 2021). These drugs act by inhibiting both classical and trans IL-6 pathways, and represent promising therapeutic options for the treatment of the most severe forms of COVID-19 (94-103).

6. Conclusion

In the present review, the molecular and nanomolecular structures involved in active IL-6 signalling were examined. Specially, the review reported that energetic transition from low to high affinity of IL-6 binding has to occur at the nanometre scale through changes of geometric orientation by conversion from the pentamer to hexamer shape.

The role played by miRNA in the post-transcriptional regulation of IL-6 expression has been evaluated through genes, pseudogenes and ceRNA of IL-6.

In addition, classic and trans-signaling pathways were analysed to evaluate the role of IL-6 in inflammatory and cognitive processes through anatomical localization and serum levels of active compounds in both pathways. Finally, the analysis of the pathogenic, diagnostic, prognostic and therapeutic roles of IL-6 in SARS-CoV-2 infection clearly demonstrates the central role of IL-6 in the ongoing global COVID-19 pandemic.

Availability of data and materials

Not applicable.

Authors' contributions

MT, SS, AF, AV, GV and AP provided substantial contributions to the conception or design of the work, or the acquisition, analysis or interpretation of data. MT, SS, GV and AD were rewponsible for drafting the work or revising it critically for important intellectual content. All authors revised the manuscript and provided important intellectual contributions. All authors have read and approved the final manuscript. All authors agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

No funding was received.

References

1 

Schimpl A and Wecker E: Replacement of T cell function by a T cell product. Nat New Biol. 237:15–17. 1972. View Article : Google Scholar : PubMed/NCBI

2 

Hirano T: Revisiting the 1986 molecular cloning of interleukin 6. Front Immunol. 5:4562014. View Article : Google Scholar : PubMed/NCBI

3 

Hirano T, Taga T, Nakano N, Yasukawa K, Kashiwamura S, Shimizu K, Nakajima K, Pyun KH and Kishimoto T: Purification to homogeneity and characterization of human B-cell differentiation factor (BCDF or BSFp-2). Proc Natl Acad Sci USA. 82:5490–5494. 1985. View Article : Google Scholar : PubMed/NCBI

4 

Sehgal PB, Grieninger G and Tosato G: Regulation of the acute phase and immune responses: Interleukin-6. Ann N Y Acad Sci. 557:1–583. 1989.

5 

Sehgal PB, Zilberstein A, Ruggieri RM, May LT, Ferguson-Smith A, Slate DL, Revel M and Ruddle FH: Human chromosome 7 carries the beta 2 interferon gene. Proc Natl Acad Sci USA. 83:5219–5222. 1986. View Article : Google Scholar : PubMed/NCBI

6 

Sutherland GR, Baker E, Callen DF, Hyland VJ, Wong G, Clark S, Jones SS, Eglinton LK, Shannon MF, Lopez AF, et al: Interleukin 4 is at 5q31 and interleukin 6 is at 7p15. Hum Genet. 79:335–337. 1988. View Article : Google Scholar : PubMed/NCBI

7 

Somers W, Stahl M and Seehra JS: 1.9 A crystal structure of interleukin 6: Implications for a novel mode of receptor dimerization and signaling. EMBO J. 16:989–997. 1997. View Article : Google Scholar : PubMed/NCBI

8 

Zhou L, Zheng Y, Tian T, Liu K, Wang M, Lin S, Deng Y, Dai C, Xu P, Hao Q, et al: Associations of interleukin-6 gene polymorphisms with cancer risk: Evidence based on 49,408 cancer cases and 61,790 controls. Gene. 670:136–147. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Simpson RJ, Hammacher A, Smith DK, Matthews JM and Ward LD: Interleukin-6: Structure-function relationships. Protein Sci. 6:929–955. 1997. View Article : Google Scholar : PubMed/NCBI

10 

Hirano T: Interleukin 6 and its receptor: Ten years later. Int Rev Immunol. 16:249–284. 1998. View Article : Google Scholar : PubMed/NCBI

11 

Hunter CA and Jones SA: IL-6 as a keystone cytokine in health and disease. Nat Immunol. 16:448–457. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Sumikawa H, Fukuhara K, Suzuki E, Matsuo Y and Nishikawa K: Tertiary structural models for human interleukin-6 and evaluation by a sequence-structure compatibility method and NMR experimental information. FEBS Lett. 404:234–240. 1997. View Article : Google Scholar : PubMed/NCBI

13 

Heinrich PC, Castell JV and Andus T: Interleukin-6 and the acute phase response. Biochem J. 265:621–636. 1990. View Article : Google Scholar : PubMed/NCBI

14 

Bazan JF: Structural design and molecular evolution of a cytokine receptor superfamily. Proc Natl Acad Sci USA. 87:6934–6938. 1990. View Article : Google Scholar : PubMed/NCBI

15 

Sakakibara S and Tosato G: Viral Interleukin-6: Role in Kaposi's sarcoma-associated herpesvirus: Associated malignancies. J Interferon Cytokine Res. 31:791–801. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Yamasaki K, Taga T, Hirata Y, Yawata H, Kawanishi Y, Seed B, Taniguchi T, Hirano T and Kishimoto T: Cloning and expression of the human interleukin-6 (BSF-2/IFN beta 2) receptor. Science. 241:825–828. 1988. View Article : Google Scholar : PubMed/NCBI

17 

Hibi M, Murakami M, Saito M, Hirano T, Taga T and Kishimoto T: Molecular cloning and expression of an IL-6 signal transducer, gp130. Cell. 63:1149–1157. 1990. View Article : Google Scholar : PubMed/NCBI

18 

Kluck PM, Wiegant J, Jansen RP, Bolk MW, Raap AK, Willemze R and Landegent JE: The human interleukin-6 receptor alpha chain gene is localized on chromosome 1 band q21. Hum Genet. 90:542–544. 1993. View Article : Google Scholar : PubMed/NCBI

19 

Rodriguez C, Grosgeorge J, Nguyen VC, Gaudray P and Theillet C: Human gp130 transducer chain gene (IL6ST) is localized to chromosome band 5q11 and possesses a pseudogene on chromosome band 17p11. Cytogenet Cell Genet. 70:64–67. 1995. View Article : Google Scholar : PubMed/NCBI

20 

Taga T, Hibi M, Hirata Y, Yamasaki K, Yasukawa K, Matsuda T, Hirano T and Kishimoto T: Interleukin-6 triggers the association of its receptor with a possible signal transducer, gp130. Cell. 58:573–581. 1989. View Article : Google Scholar : PubMed/NCBI

21 

Boulanger MJ, Chow DC, Brevnova EE and Garcia KC: Hexameric structure and assembly of the interleukin-6/IL-6 alpha-receptor/gp130 complex. Science. 300:2101–2104. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Lacroix M, Rousseau F, Guilhot F, Malinge P, Magistrelli G, Herren S, Jones SA, Jones GW, Scheller J, Lissilaa R, et al: Novel insights into interleukin 6 (IL-6) Cis- and trans-signaling pathways by differentially manipulating the assembly of the IL-6 signaling complex. J Biol Chem. 290:26943–26953. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Trovato MC, Andronico D, Sciacchitano S, Ruggeri RM, Picerno I, Di Pietro A and Visalli G: Nanostructures: Between natural environment and medical practice. Rev Environ Health. 33:295–307. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Weidle UH, Klostermann S, Eggle D and Krüger A: Interleukin 6/interleukin 6 receptor interaction and its role as a therapeutic target for treatment of cachexia and cancer. Cancer Genomics Proteomics. 7:287–302. 2010.PubMed/NCBI

25 

Heinrich PC, Behrmann I, Haan S, Hermanns HM, Müller-Newen G and Schaper F: Principles of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem J. 374:1–20. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Scheller J, Chalaris A, Schmidt-Arras D and Rose-John S: The pro- and anti-inflammatory properties of the cytokine inter-leukin-6. Biochim Biophys Acta. 1813:878–888. 2011. View Article : Google Scholar : PubMed/NCBI

27 

An Y, Furber KL and Ji S: Pseudogenes regulate parental gene expression via ceRNA network. J Cell Mol Med. 21:185–192. 2017. View Article : Google Scholar

28 

Kuscuoglu D, Janciauskiene S, Hamesch K, Haybaeck J, Trautwein C and Strnad P: Liver-master and servant of serum proteome. J Hepatol. 69:512–524. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Tuck AC and Tollervey D: A transcriptome-wide atlas of RNP composition reveals diverse classes of mRNAs and lncRNAs. Cell. 154:996–1009. 2013. View Article : Google Scholar : PubMed/NCBI

30 

St Laurent G, Wahlestedt C and Kapranov P: The landscape of long noncoding RNA classification. Trends Genet. 31:239–251. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Pink RC, Wicks K, Caley DP, Punch EK, Jacobs L and Carter DR: Pseudogenes: Pseudo-functional or key regulators in health and disease? RNA. 17:792–798. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Salmena L, Poliseno L, Tay Y, Kats L and Pandolfi PP: A ceRNA hypothesis: The Rosetta stone of a hidden RNA language? Cell. 146:353–358. 2011. View Article : Google Scholar : PubMed/NCBI

33 

van Rij RP and Andino R: The silent treatment: RNAi as a defense against virus infection in mammals. Trends Biotechnol. 24:186–193. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Lavra L, Ulivieri A, Dominici R, Trovato MC, Bartolazzi A, Soddu S and Sciacchitano S: Analysis of the role of p53 and galectin-3 in proliferation and apoptosis of thyroid carcinoma cell lines by specific RNA interference experiments. Biomed Pharmacother. 60:4912006. View Article : Google Scholar

35 

Cecchinelli B, Lavra L, Rinaldo C, Iacovelli S, Gurtner A, Gasbarri A, Ulivieri A, Del Prete F, Trovato M, Piaggio G, et al: Repression of the antiapoptotic molecule Galectin-3 by homeodomain-interacting protein kinase 2-activated p53 is required for p53-Induced apoptosis. Mol Cell Biol. 26:4746–4757. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Bautista RR, Gómez AO, Miranda AH, Dehesa AZ, Villarreal-Garza C, Ávila-Moreno F and Arrieta O: Correction to: Long non-coding RNAs: Implications in targeted diagnoses, prognosis, and improved therapeutic strategies in human non- and triple-negative breast cancer. Clin Epigenetics. 10:1062018. View Article : Google Scholar : PubMed/NCBI

37 

Hosseinahli N, Aghapour M, Duijf PHG and Baradaran B: Treating cancer with microRNA replacement therapy: A literature review. J Cell Physiol. 233:5574–5588. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Huang HC, Yu HR, Hsu TY, Chen IL, Huang HC, Chang JC and Yang KD: MicroRNA-142-3p and let-7g negatively regulates augmented IL-6 production in neonatal polymorphonuclear leukocytes. Int J Biol Sci. 13:690–700. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Liao YC, Wang YS, Guo YC, Lin WL, Chang MH and Juo SH: Let-7g improves multiple endothelial functions through targeting transforming growth factor-beta and SIRT-1 signaling. J Am Coll Cardiol. 63:1685–1694. 2014. View Article : Google Scholar

40 

Johnson CD, Esquela-Kerscher A, Stefani G, Byrom M, Kelnar K, Ovcharenko D, Wilson M, Wang X, Shelton J, Shingara J, et al: The let-7 MicroRNA represses cell proliferation pathways in human cells. Cancer Res. 67:7713–7722. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Gao X, Xu W, Lu T, Zhou J, Ge X and Hua D: MicroRNA-142-3p promotes cellular invasion of colorectal cancer cells by activation of RAC1. Technol Cancer Res Treat. 17:15330338187905082018. View Article : Google Scholar : PubMed/NCBI

42 

Sun Y, Varambally S, Maher CA, Cao Q, Chockley P, Toubai T, Malter C, Nieves E, Tawara I, Wang Y, et al: Targeting of microRNA-142-3p in dendritic cells regulates endotoxin-induced mortality. Blood. 117:6172–6183. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Sung SY, Liao CH, Wu HP, Hsiao WC, Wu IH, Jinpu Yu, Lin SH and Hsieh CL: Loss of let-7 microRNA upregulates IL-6 in bone marrow-derived mesenchymal stem cells triggering a reactive stromal response to prostate cancer. PLoS One. 8:e716372013. View Article : Google Scholar : PubMed/NCBI

44 

Selzner N, Selzner M, Odermatt B, Tian Y, Van Rooijen N and Clavien PA: ICAM-1 triggers liver regeneration through leukocyte recruitment and Kupffer cell-dependent release of TNF-alpha/IL-6 in mice. Gastroenterology. 124:692–700. 2003. View Article : Google Scholar : PubMed/NCBI

45 

Yoshiya S, Shirabe K, Imai D, Toshima T, Yamashita Y, Ikegami T, Okano S, Yoshizumi T, Kawanaka H and Maehara Y: Blockade of the apelin-APJ system promotes mouse liver regeneration by activating Kupffer cells after partial hepatectomy. J Gastroenterol. 50:573–582. 2015. View Article : Google Scholar

46 

Kishimoto T: Interleukin-6: From basic science to medicine-40 years in immunology. Annu Rev Immunol. 23:1–21. 2005. View Article : Google Scholar

47 

Papanicolaou DA and Vgontzas AN: Interleukin-6: The endocrine cytokine. J Clin Endocrinol Metab. 85:1331–1333. 2000. View Article : Google Scholar : PubMed/NCBI

48 

Ruggeri RM, Sciacchitano S, Vitale A, Cardelli P, Galletti M, Vitarelli E, Barresi G, Benvenga S, Trimarchi F and Trovato M: Serum hepatocyte growth factor is increased in hashimoto's thyroiditis whether or not associated with nodular goiter as compared with healthy non goitrous individuals. J Endocrinol Invest. 32:465–469. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Trovato M, Ruggeri RM, Sciacchitano S, Vicchio TM, Picerno I, Pellicanò G, Valenti A and Visalli G: Serum interleukin-6 levels are increased in HIV-infected patients that develop autoimmune disease during long-term follow-up. Immunobiology. 223:264–268. 2018. View Article : Google Scholar

50 

Ruggeri RM, Villari D, Simone A, Scarfi R, Attard M, Orlandi F, Barresi G, Trimarchi F, Trovato M and Benvenga S: Co-expression of interleukin-6 (IL-6) and interleukin-6 receptor (IL-6R) in thyroid nodules is associated with co-expression of CD30 ligand/CD30 receptor. J Endocrinol Invest. 25:959–966. 2002. View Article : Google Scholar

51 

Trovato M, Grosso M, Vitarelli E, Ruggeri RM, Alesci S, Trimarchi F, Barresi G and Benvenga S: Distinctive expression of STAT3 in papillary thyroid carcinomas and a subset of follicular adenomas. Histol Histopathol. 18:393–399. 2003.PubMed/NCBI

52 

Ruggeri RM, Barresi G, Sciacchitano S, Trimarchi F, Benvenga S and Trovato M: Immunoexpression of the CD30 ligand/CD30 and IL-6/IL-6R signals in thyroid autoimmune diseases. Histol Histopathol. 21:249–256. 2006.

53 

Trovato M: A historical excursus of diagnostic methods for Hashimoto thyroiditis and Graves' disease. Gazz Med Ital Arch Sci Med. 179:479–485. 2020. View Article : Google Scholar

54 

Elsabahy M and Wooley KL: Cytokines as biomarkers of nanoparticle immunotoxicity. Chem Soc Rev. 42:5552–5576. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Visalli G, Baluce B, Bertuccio M, Picerno I and Di Pietro A: Mitochondrial-Mediated apoptosis pathway in alveolar epithelial cells exposed to the metals in Combustion-Generated particulate matter. J Toxicol Environ Health A. 78:697–709. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Visalli G, Facciolà A, Iannazzo D, Piperno A, Pistone A and Di Pietro A: The role of the iron catalyst in the toxicity of multi-walled carbon nanotubes (MWCNTs). J Trace Elem Med Biol. 43:153–160. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Visalli G, Currò M, Iannazzo D, Pistone A, Pruiti Ciarello M, Acri G, Testagrossa B, Bertuccio MP, Squeri R and Di Pietro A: In vitro assessment of neurotoxicity and neuroinflammation of homemade MWCNTs. Environ Toxicol Pharmacol. 56:121–128. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Visalli G, Facciolà A, Currò M, Laganà P, La Fauci V, Iannazzo D, Pistone A and Di Pietro A: Mitochondrial impairment induced by sub-chronic exposure to multi-walled carbon nanotubes. Int J Environ Res Public Health. 16:7922019. View Article : Google Scholar :

59 

Facciolà A, Visalli G, La Maestra S, Ceccarelli M, D'Aleo F, Nunnari G, Pellicanò GF and Di Pietro A: Carbon nanotubes and central nervous system: Environmental risks, toxicological aspects and future perspectives. Environ Toxicol Pharmacol. 65:23–30. 2019. View Article : Google Scholar

60 

Palomäki J, Välimäki E, Sund J, Vippola M, Clausen PA, Jensen KA, Savolainen K, Matikainen S and Alenius H: Long, needle-like carbon nanotubes and asbestos activate the NLRP3 inflammasome through a similar mechanism. ACS Nano. 5:6861–6870. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Neagu M, Constantin C, Popescu ID, Zipeto D, Tzanakakis G, Nikitovic D, Fenga C, Stratakis CA, Spandidos DA and Tsatsakis AM: Inflammation and metabolism in cancer cell-mitochondria key player. Front Oncol. 9:3482019. View Article : Google Scholar : PubMed/NCBI

62 

Arnoldussen YJ, Skogstad A, Skaug V, Kasem M, Haugen A, Benker N, Weinbruch S, Apte RN and Zienolddiny S: Involvement of IL-1 genes in the cellular responses to carbon nanotube exposure. Cytokine. 73:128–137. 2015. View Article : Google Scholar : PubMed/NCBI

63 

Migliore L, Uboldi C, Di Bucchianico S and Coppedè F: Nanomaterials and neurodegeneration. Environ Mol Mutagen. 56:149–170. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Bardi G, Nunes A, Gherardini L, Bates K, Al-Jamal KT, Gaillard C, Prato M, Bianco A, Pizzorusso T and Kostarelos K: Functionalized carbon nanotubes in the brain: Cellular internalization and neuroinflammatory responses. PLoS One. 8:e809642013. View Article : Google Scholar : PubMed/NCBI

65 

Bussy C, Al-Jamal KT, Boczkowski J, Lanone S, Prato M, Bianco A and Kostarelos K: Microglia determine brain region-specific neurotoxic responses to chemically functionalized carbon nanotubes. ACS Nano. 9:7815–7830. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Rothaug M, Becker-Pauly C and Rose-John S: The role of inter-leukin-6 signaling in nervous tissue. Biochim Biophys Acta. 1863:1218–1227. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Hu J, Feng X, Valdearcos M, Lutrin D, Uchida Y, Koliwad SK and Maze M: Interleukin-6 is both necessary and sufficient to produce perioperative neurocognitive disorder in mice. Br J Anaesth. 120:537–545. 2018. View Article : Google Scholar : PubMed/NCBI

68 

Fenga C, Gangemi S, Di Salvatore V, Falzone L and Libra M: Immunological effects of occupational exposure to lead (Review). Mol Med Rep. 15:3355–3360. 2017. View Article : Google Scholar : PubMed/NCBI

69 

Gangemi S, Gofita E, Costa C, Teodoro M, Briguglio G, Nikitovic D, Tzanakakis G, Tsatsakis AM, Wilks MF, Spandidos DA and Fenga C: Occupational and environmental exposure to pesticides and cytokine pathways in chronic diseases (Review). Int J Mol Med. 38:1012–1020. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Shahpiri Z, Bahramsoltani R, Hosein Farzaei M, Farzaei F and Rahimi R: Phytochemicals as future drugs for Parkinson's disease: A comprehensive review. Rev Neurosci. 27:651–668. 2016. View Article : Google Scholar : PubMed/NCBI

71 

Ardah MT, Bharathan G, Kitada T and Haque ME: Ellagic acid prevents dopamine neuron degeneration from oxidative stress and neuroinflammation in MPTP Model of Parkinson's disease. Biomolecules. 10:15192020. View Article : Google Scholar

72 

Gadient RA and Otten U: Expression of interleukin-6 (IL-6) and interleukin-6 receptor (IL-6R) mRNAs in rat brain during postnatal development. Brain Res. 637:10–14. 1994. View Article : Google Scholar : PubMed/NCBI

73 

Marsland AL, Gianaros PJ, Abramowitch SM, Manuck SB and Hariri AR: Interleukin-6 covaries inversely with hippocampal grey matter volume in middle-aged adults. Biol Psychiatry. 64:484–490. 2008. View Article : Google Scholar : PubMed/NCBI

74 

MacQueen GM, Campbell S, McEwen BS, Macdonald K, Amano S, Joffe RT, Nahmias C and Young LT: Course of illness, hippocampal function, and hippocampal volume in major depression. Proc Natl Acad Sci USA. 100:1387–1392. 2003. View Article : Google Scholar : PubMed/NCBI

75 

Baune BT, Konrad C, Grotegerd D, Suslow T, Birosova E, Ohrmann P, Bauer J, Arolt V, Heindel W, Domschke K, et al: Interleukin-6 gene (IL-6): A possible role in brain morphology in the healthy adult brain. J Neuroinflammation. 9:1252012. View Article : Google Scholar : PubMed/NCBI

76 

Campbell IL, Abraham CR, Masliah E, Kemper P, Inglis JD, Oldstone MB and Mucke L: Neurologic disease induced in transgenic mice by cerebral overexpression of interleukin 6. Proc Natl Acad Sci USA. 90:10061–10065. 1993. View Article : Google Scholar : PubMed/NCBI

77 

Campbell IL, Erta M, Lim SL, Frausto R, May U, Rose-John S, Scheller J and Hidalgo J: Trans-signaling is a dominant mechanism for the pathogenic actions of interleukin-6 in the brain. J Neurosci. 34:2503–2513. 2014. View Article : Google Scholar : PubMed/NCBI

78 

Chucair-Elliott AJ, Conrady C, Zheng M, Kroll CM, Lane TE and Carr DJ: Microglia-induced IL-6 protects against neuronal loss following HSV-1 infection of neural progenitor cells. Glia. 62:1418–1434. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Chomarat P, Banchereau J, Davoust J and Palucka AK: IL-6 switches the differentiation of monocytes from dendritic cells to macrophages. Nat Immunol. 1:510–514. 2000. View Article : Google Scholar

80 

Urashima M, Chauhan D, Hatziyanni M, Ogata A, Hollenbaugh D, Aruffo A and Anderson KC: CD40 ligand triggers interleukin-6 mediated B cell differentiation. Leuk Res. 20:507–515. 1996. View Article : Google Scholar : PubMed/NCBI

81 

Yang R, Masters AR, Fortner KA, Champagne DP, Yanguas-Casás N, Silberger DJ, Weaver CT, Haynes L and Rincon M: IL-6 promotes the differentiation of a subset of naive CD8+ T cells into IL-21-producing B helper CD8+ T cells. J Exp Med. 213:2281–2291. 2016. View Article : Google Scholar : PubMed/NCBI

82 

Diehl S and Rincón M: The two faces of IL-6 on Th1/Th2 differentiation. Mol Immunol. 39:531–536. 2002. View Article : Google Scholar : PubMed/NCBI

83 

Gubernatorova EO, Gorshkova EA, Polinova AI and Drutskaya MS: IL-6: Relevance for immunopathology of SARS-CoV-2. Cytokine Growth Factor Rev. 53:13–24. 2020. View Article : Google Scholar : PubMed/NCBI

84 

Xu H, Zhong L, Deng J, Peng J, Dan H, Zeng X, Li T and Chen Q: High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int J Oral Sci. 12:82020. View Article : Google Scholar : PubMed/NCBI

85 

Rose-John S, Winthrop K and Calabrese L: The role of IL-6 in host defence against infections: Immunobiology and clinical implications. Nat Rev Rheumatol. 13:399–409. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, Wang T, Zhang X, Chen H, Yu H, et al: Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 130:2620–2629. 2020. View Article : Google Scholar : PubMed/NCBI

87 

Ruan Q, Yang K, Wang W, Jiang L and Song J: Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 46:846–848. 2020. View Article : Google Scholar : PubMed/NCBI

88 

Abbasifard M and Khorramdelazad H: The bio-mission of interleukin-6 in the pathogenesis of COVID-19: A brief look at potential therapeutic tactics. Life Sci. 257:1180972020. View Article : Google Scholar : PubMed/NCBI

89 

Coomes EA and Haghbayan H: Interleukin-6 in Covid-19: A systematic review and meta-analysis. Rev Med Virol. 30:1–9. 2020. View Article : Google Scholar : PubMed/NCBI

90 

Zhang J, Hao Y, Ou W, Ming F, Liang G, Qian Y, Cai Q, Dong S, Hu S, Wang W and Wei S: Serum interleukin-6 is an indicator for severity in 901 patients with SARS-CoV-2 infection: A cohort study. J Transl Med. 18:4062020. View Article : Google Scholar : PubMed/NCBI

91 

Herold T, Jurinovic V, Arnreich C, Hellmuth JC, von Bergwelt-Baildon M, Klein M and Weinberger T: Level of IL-6 predicts respiratory failure in hospitalized symptomatic COVID-19 patients. medRxiv. https://doi.org/10.1101/2020.04.01.20047381. Accessed April 27, 2020.

92 

Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, Xiang J, Wang Y, Song B, Gu X, et al: Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: A retrospective cohort study. Lancet. 395:1054–1062. 2020. View Article : Google Scholar : PubMed/NCBI

93 

Atal S and Fatima Z: IL-6 Inhibitors in the treatment of serious COVID-19: A promising therapy? Pharmaceut Med. 34:223–231. 2020.PubMed/NCBI

94 

Xu X, Han M, Li T, Sun W, Wang D, Fu B, Zhou Y, Zheng X, Yang Y, Li X, et al: Effective treatment of severe COVID-19 patients with tocilizumab. Proc Natl Acad Sci USA. 117:10970–10975. 2020. View Article : Google Scholar : PubMed/NCBI

95 

Fu B, Xu X and Wei H: Why tocilizumab could be an effective treatment for severe COVID-19? J Transl Med. 18:1642020. View Article : Google Scholar : PubMed/NCBI

96 

Toniati P, Piva S, Cattalini M, Garrafa E, Regola F, Castelli F, Franceschini F, Airò P, Bazzani C, Beindorf EA, et al: Tocilizumab for the treatment of severe COVID-19 pneumonia with hyperinflammatory syndrome and acute respiratory failure: A single center study of 100 patients in Brescia, Italy. Autoimmun Rev. 19:1025682020. View Article : Google Scholar : PubMed/NCBI

97 

Issa N, Dumery M, Guisset O, Mourissoux G, Bonnet F and Camou F: Feasibility of tocilizumab in ICU patients with COVID-19. J Med Virol. 93:46–47. 2021. View Article : Google Scholar

98 

Alattar R, Ibrahim TBH, Shaar SH, Abdalla S, Shukri K, Daghfal JN, Khatib MY, Aboukamar M, Abukhattab M, Alsoub HA, et al: Tocilizumab for the treatment of severe coronavirus disease 2019. J Med Virol. 92:2042–2049. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Della-Torre E, Campochiaro C, Cavalli G, De Luca G, Napolitano A, La Marca S, Boffini N, Da Prat V, Di Terlizzi G, Lanzillotta M, et al: Interleukin-6 blockade with sarilumab in severe COVID-19 pneumonia with systemic hyperinflammation: An open-label cohort study. Ann Rheum Dis. 79:1277–1285. 2020. View Article : Google Scholar : PubMed/NCBI

100 

Benucci M, Giannasi G, Cecchini P, Gobbi FL, Damiani A, Grossi V, Infantino M and Manfredi M: COVID-19 pneumonia treated with sarilumab: A clinical series of eight patients. J Med Virol. 92:2368–2370. 2020. View Article : Google Scholar : PubMed/NCBI

101 

Palanques-Pastor T, López-Briz E and Poveda Andrés JL: Involvement of interleukin 6 in SARS-CoV-2 infection: Siltuximab as a therapeutic option against COVID-19. Eur J Hosp Pharm. 27:297–298. 2020. View Article : Google Scholar : PubMed/NCBI

102 

Gritti G, Raimondi F, Ripamonti D, Riva I, Landi F, Alborghetti L, Frigeni M, Damiani M, Micò C, Fagiuoli S, et al: Use of siltuximab in patients with COVID-19 pneumonia requiring ventilatory support. medRxiv. https://doi.org/10.1101/2020.04.01.20048561.

103 

Vaidya G, Czer LSC, Kobashigawa J, Kittleson M, Patel J, Chang D, Kransdorf E, Shikhare A, Tran H, Vo A, et al: Successful treatment of severe COVID-19 pneumonia with clazakizumab in a heart transplant recipient: A case report. Transplant Proc. 52:2711–2714. 2020. View Article : Google Scholar : PubMed/NCBI

104 

Stelzer G, Rosen R, Plaschkes I, Zimmerman S, Twik M, Fishilevich S, Stein TI, Nudel R, Lieder I, Mazor Y, et al: The GeneCards suite: From gene data mining to disease genome sequence analysis. Curr Protoc Bioinformatics. 54:1.30.1–1.30.33. 2016. View Article : Google Scholar

105 

Hunt SE, McLaren W, Gil L, Thormann A, Schuilenburg H, Sheppard D, Parton A, Armean IM, Trevanion SJ, Flicek P and Cunningham F: Ensembl variation resources. Database (Oxford). 2018:bay1192018. View Article : Google Scholar

Related Articles

Journal Cover

June-2021
Volume 47 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Trovato M, Sciacchitano S, Facciolà A, Valenti A, Visalli G and Di Pietro A: Interleukin‑6 signalling as a valuable cornerstone for molecular medicine (Review). Int J Mol Med 47: 107, 2021
APA
Trovato, M., Sciacchitano, S., Facciolà, A., Valenti, A., Visalli, G., & Di Pietro, A. (2021). Interleukin‑6 signalling as a valuable cornerstone for molecular medicine (Review). International Journal of Molecular Medicine, 47, 107. https://doi.org/10.3892/ijmm.2021.4940
MLA
Trovato, M., Sciacchitano, S., Facciolà, A., Valenti, A., Visalli, G., Di Pietro, A."Interleukin‑6 signalling as a valuable cornerstone for molecular medicine (Review)". International Journal of Molecular Medicine 47.6 (2021): 107.
Chicago
Trovato, M., Sciacchitano, S., Facciolà, A., Valenti, A., Visalli, G., Di Pietro, A."Interleukin‑6 signalling as a valuable cornerstone for molecular medicine (Review)". International Journal of Molecular Medicine 47, no. 6 (2021): 107. https://doi.org/10.3892/ijmm.2021.4940