Abstract
The major concern of pharmacology about cytokines has originated from plentiful data showing association between gross changes in their production and pathophysiological processes. Despite the enigmatic role of cytokines in diseases, a number of them have become a subject of cytokine and anti-cytokine immunotherapies. Production of cytokines can be influenced by many endogenous and exogenous stimuli including drugs. Cells of the immune system, such as macrophages and lymphocytes, are richly endowed with receptors for the mediators of physiological functions, such as biogenic amines, adenosine, prostanoids, steroids, etc. Drugs, agonists or antagonists of these receptors can directly or indirectly up- and down-regulate secretion of cytokines and expression of cytokine receptors. Vice versa, cytokines interfere with drug pharmacokinetics and pharmacodynamics through the interactions with cytochrome P450 and multiple drug resistance proteins. The aim of the review is to encourage more intensive studies in these fields of cytokine pharmacology. It also outlines major areas of searching promising candidates for immunotherapeutic interventions.
Keywords: cytokines, immunotherapy, immunopharmacology, immunotoxicology
Cytokines and cytokine network
The story of cytokines may be dated back to the mid of the last century, to the discovery of interferon (IFN) (Isaacs and Lindenmann, 1957). The superfamily of cytokines, mostly small- to medium-sized polypeptides or glycoproteins (6–30 kDa) with multiple regulatory homeostatic functions, has expanded dramatically during the last few years. Presently, about 200 cytokines are recognized. In contrast to hormones, cytokines are usually produced by many cell types, and act in autocrine and paracrine modes of fashion. The typical feature of most cytokines is a low or no constitutive production and transient expression following inducing stimuli. A number of feedback mechanisms influence synergistically or antagonistically production and activity of individual members of the cytokine network. The biological effects of several cytokines are often overlapping (redundancy), and individual cytokines possess multiple regulatory functions (pleiotropy).
Widely accepted criterium of cytokine categorization is the production of cytokines in dependence on distinct lineages of T helper (Th) cells (Table 1). The Th1 cells produce IFN-γ, interleukin (IL)-2, IL-15 and lymphotoxin [tumour necrosis factor (TNF)-β]. The Th2 cells produce IL-4, IL-5, IL-6, IL-9 and IL-13 (Mosmann and Coffman, 1989; Romagnani, 1994). Both Th1 and Th2 cells produce IL-10 (O'Garra and Vieira, 2007). Conventional definition of Th1 and Th2 cells depends strictly on the ability to secrete IFN-γ and IL-4 respectively. Mature Th0 cells secrete the both cytokine types. The Th1 cytokines inhibit growth of intracellular pathogens (viruses, bacteria, fungi) and tumour cells. They enhance the delayed type hypersensitivity, phagocytosis, oxidative burst, inflammatory reactions and expression of class I and II MHC molecules. The Th2 cytokines inhibit growth of extracellular parasites (helminths) and suppress phagocytosis. They augment B cell proliferation, drive antibody production and switch IgG to IgE class of antibodies. They are associated with the development of allergic and related IgE-mediated diseases.
Table 1.
Cytokine |
Major cell producers |
Receptors* | Signalling* | |
---|---|---|---|---|
T cells | Other cell types | |||
IFN-γ | Th1 | CD8+ T cells (Su et al., 1998), natural killer cells (Perussia, 1991), B cells (Pang et al., 1992), macrophages (Markus et al., 1998), keratinocytes (Gröne, 2002), astrocytoma cells (Nitta et al., 1994) | IFN-γRI; IFN-γRII | Jak1, Jak2/STAT1 |
IL-2 | Th1 | Thymocytes (Kroemer and Wick, 1989) | IL-2Rα; IL-2Rβ; γc | Jak1, Jak3/STAT3, STAT5 |
IL-15 | Th1 | Macrophages, monocytes, bone marrow stromal cells (Gosselin et al., 1999), epithelial cells, fibroblasts, muscle cells (Yanagita et al., 2002), keratinocytes (Gröne, 2002) | IL-15Rα; IL-2Rβ; γc | Jak1, Jak3/STAT3, STAT5 |
TNF-β (LT-α) | Th1 | B cells, natural killer cells (Gray et al., 1984), keratinocytes, mast cells (Middel et al., 2000) | TNFR1, TNFR2 | NF-κB |
IL-4 | Th2 | Basophils (Brunner et al., 1993), eosinophils (Moqbel et al., 1995), mast cells (Petray et al., 1993), natural killer cells (Hameg et al., 1999) | IL-4Rα; IL-13Rα; γc | Jak1, Jak3/STAT6 |
IL-5 | Th2 | Natural killer cells (Warren et al., 1995), eosinophils, mast cells (Cousins et al., 1994); Lorentz et al., 1999), basophils (Ying et al., 1995), endothelial cells (Möhle et al., 1997), epithelial cells (Salvi et al., 1999) | IL-5Rα; βc | Jak2/STAT1, STAT3, STAT5 |
IL-6 | Th2 | B cells (Yin, 1990), macrophages (Lee Kim Lee et al., 2007), mast cells (Song et al., 1999), fibroblasts (Defilippi et al., 1987), keratinocytes (Gröne, 2002), endothelial cells (Shalaby et al., 1989), osteoblasts (Miwa et al., 1999) | IL-6R; gp130 | Jak1, Jak2, Tyk2/STAT1, STAT3, STAT5 |
IL-9 | Th2 | Eosinophils, basophils (Shimbara et al., 2000) | IL-9R, γc | Jak1, Jak3/STAT1, STAT3, STAT5 |
IL-13 | Th2 | Natural killer cells (McDermott et al., 2005), basophils (Li et al., 1996), mast cells (Kelly-Welch et al., 2003) | IL-13Rα; IL-4Rα; γc | Jak1, Jak2, Tyk2/STAT6 |
IL-17 | Th17 | CD8+ T cells (Shin et al., 1998), eosinophils (Molet et al., 2001), neutrophils (Ferretti et al., 2003) | IL-17R | Jak1, Jak2, Jak3, Tyk2/STAT1, STAT2, STAT3, STAT4 |
IL-22 | Th17 | Natural killer cells (Wolk and Sabat, 2006) | IL-22R1, IL-10Rβ | Jak1, Tyk2/STAT1, STAT3, STAT5 |
IL-10 | Tr1 | B cells (O'Garra et al., 1992), macrophages and monocytes (de Waal Malefyt et al., 1991), keratinocytes (Gröne, 2002), glial cells (Mizuno et al., 1994; Williams et al., 1996) | IL-10Rα; IL-10Rβ | Jak1, Tyk2/STAT1, STAT3 |
TGF-β | Tr3 (Th3) | Macrophages (Assoian et al., 1987), neutrophils, platelets (Schindler et al., 1998), microglial cells (Chao et al., 1995) | TGF-βRI, TGF-βRII | Smad |
Major references for receptors and signalling: (Bach et al., 1997; Silvennoinen et al., 1997; Imada and Leonard, 2000; Seidel et al., 2000; Lejeune et al., 2002) and references in the text.
It should be emphasized that production of Th1 and Th2 cytokines is not restricted to the Th1 and Th2 lymphocytes respectively. They are produced by many cell types (Table 1). This fact is reflected in the frequently used terms ‘Th1 immune response’ and ‘Th2 immune response’ describing changes in expression of these cytokines irrespective of the cell source.
Currently, there is a growing interest in recently revealed T cell clones exhibiting different cytokine profile from that of Th1 and Th2 cells. They include a third subset of Th cells (Th17) and T regulatory cells (Treg).
Th17 cells secrete IL-17, IL-17F, IL-22 and IL-25 as signature cytokines. They also produce IL-6 and TNF-α, and some of them IFN-γ. The Th17 immunity is an attractive therapeutic target because it is protective against extracellular bacteria and fungi. On the other hand, it may contribute to the development of allergic responses (IL-25), inflammation and autoimmune disorders (e.g. arthritis) (Annunziato et al., 2007; Kaiko et al., 2008).
There are several subsets of Treg cells. Their major function is maintaining self-tolerance via inhibition of effector T cells. The regulatory T cells type 1 (Tr1) are producers of large amounts of IL-10. They also secrete IFN-γ, IL-5 and low to moderate levels of transforming growth factor-β (TGF-β) and IL-2. The regulatory cells of the Tr3 subset (also called Th3 cells) produce preferentially high amounts of TGF-β and low amounts of IL-10. The regulatory cytokines IL-10 and TGF-β suppress pathological immune responses occurring in autoimmune diseases and transplantation. Both TGF-β and IL-10 negatively regulate production of Th1 and Th2 cytokines. Their overexpression may impair the immune mechanisms directed against pathogens and tumour antigens (Levings et al., 2001; Liu and Leung, 2006; Roncarolo et al., 2006). IL-10 possesses antifibrotic activities and may be valuable as a therapeutic cytokine for patients with liver cirrhosis (Rachmawati et al., 2007). The anti-inflammatory effects of IL-10 have led to its use in hyper-inflammatory states such as psoriasis, organ transplantation and Crohn's disease (Spellberg and Edwards, 2001). TGF-β is a multifunctional polypeptide that stimulates synthesis of many components of extracellular matrix, such as collagens, fibronectin and proteoglycans. Increased production of TGF-β1 is associated with normal reparative as well as pathological fibrotic processes in many organs (Gharaee-Kermani and Phan, 2001).
Immune functions of cytokines result from coordinated action of T cells, macrophages and dendritic cells and depend largely on their recruitment to the sites of infection, inflammation and other pathological lesions. The trafficking of immune cells to the sites of infection and inflammation is under tight control of a special class of cytokines with chemoattractant properties (chemokines). They target many cell types (Table 2). Chemokines are classified into four groups, depending on the relative position of the first N-terminal cystein residues. In the CC family (β-chemokines), the first two cysteins are adjacent; in the CXC family (α-chemokines), they are intervened by one amino acid (Table 2). In the CX3C family (δ-chemokines), the first two cysteins are separated by three amino acids. The C family (γ-chemokines) contains only two of the four conserved cysteins. Chemokines are produced by almost all cells. Their typical feature is binding to multiple chemokine receptors that are expressed on many cell types (Table 3). The inducible chemokines are stimulated by Th1 cytokines (IFN-γ, IL-2) and by pro-inflammatory cytokines (IL-1, TNF-α). The Th2 cytokines (IL-4) and Treg cytokines (IL-10, TGF-β) down-regulate secretion of chemokines (Adams and Lloyd, 1997).
Table 2.
Chemokine |
Target cells |
|||||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
B | T | NK | Dc | Mo | Ba | Eo | Ne | Fi | En | Ke | Mc | Sc | ||
CCL2 | MCP-1: monocyte chemotactic protein-1 | |||||||||||||
CCL3 | MIP-1α: macrophage inflammatory protein-1α | |||||||||||||
CCL4 | MIP-1β: macrophage inflammatory protein-1β | |||||||||||||
CCL5 | RANTES: regulated upon activation, normal T cell expressed and secreted | |||||||||||||
CCL7 | MCP-3: monocyte chemotactic protein-3 | |||||||||||||
CCL8 | MCP-2: monocyte chemotactic protein-2 | |||||||||||||
CCL11 | Eotaxin | |||||||||||||
CCL13 | MCP-4: monocyte chemotactic protein-4 | |||||||||||||
CXCL1 | Gro-α: growth-related oncogen-α | |||||||||||||
CXCL4 | PF-4: platelet factor-4 | |||||||||||||
CXCL5 | ENA-78: epithelial derived neutrophil attractant | |||||||||||||
CXCL6 | GCP-2: granulocyte chemotactic protein-2 | |||||||||||||
CXCL7 | NAP-2: platelet basic protein | |||||||||||||
CXCL8 | IL-8: interleukin-8 | |||||||||||||
CXCL9 | MIG: monokine induced by γ-interferon | |||||||||||||
CXCL10 | IP-10: interferon-inducible protein-10 | |||||||||||||
CXCL12 | SDF-1α/β: stromal cell-derived factor-1 |
References supporting the Table: (Howard et al., 1996; Adams and Lloyd, 1997; Saunders and Tarby, 1999).
B, B cells; Ba, basophiles; Dc, dendritic cells; Eo, eosinophils; En, endothelial cells; Fi, fibroblsts; Ke, keratinocytes; Mc, mast cells; Mo, monocytes; Ne, neutrophils; NK, natural killer cells; Sc, stem cells; T, T cells.
Table 3.
Receptor | Cells expressing chemokine receptors |
Chemokine ligands of receptors* |
|||||||||||||||||||||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
CCL subfamily | CXCL subfamily | ||||||||||||||||||||||||||||||
a | b | C | d | e | f | g | h | i | j | k | n | o | p | 2 | 3 | 4 | 5 | 7 | 8 | 11 | 13 | 1 | 4 | 5 | 6 | 7 | 8 | 9 | 10 | 12 | |
CCR1 | |||||||||||||||||||||||||||||||
CCR2 | |||||||||||||||||||||||||||||||
CCR3 | |||||||||||||||||||||||||||||||
CCR4 | |||||||||||||||||||||||||||||||
CCR5 | |||||||||||||||||||||||||||||||
CCR6 | |||||||||||||||||||||||||||||||
CCR7 | |||||||||||||||||||||||||||||||
CCR8 | |||||||||||||||||||||||||||||||
CCR9 | |||||||||||||||||||||||||||||||
CCR10 | |||||||||||||||||||||||||||||||
CXCR1 | |||||||||||||||||||||||||||||||
CXCR2 | |||||||||||||||||||||||||||||||
CXCR3 | |||||||||||||||||||||||||||||||
CXCR4 | |||||||||||||||||||||||||||||||
CXCR5 | |||||||||||||||||||||||||||||||
CXCR6 |
The acronyms and names of chemokines are shown in Table 2.
The CCR6 receptor binds with chemokine CCL20/MIP-3α. The CCR7 receptor binds with chemokines CCL19/MIP-3β and CCL21/SLC. The CCR10 receptor binds with CCL27/CTACK and CCL28/MEC chemokines. The CXRC5 receptor binds with CXCL13/BCA-1. The CXCR6 receptor binds with CXCL16/small inducible cytokine B6 chemokine. Major references to support the table: (Devalaraja and Richmond, 1999; Ebnet and Vestweber, 1999; Mantovani, 1999; Schwarz and Wells, 1999; Rossi and Zlotnik, 2000; Owen, 2001; Onuffer and Horuk, 2002).
a, T cells; b, B cells; c, natural killer cells; d, macrophages; e, monocytes; f, astrocytes; g, dendritic cells; h, basophils; i, eosinophils; j, neutrophils; k, mast cells; n, fibroblast; o, endothelial cells; p, platelets.
Many of the chemokine–chemokine receptor interactions have been identified to be associated with disease conditions, including allergic diseases, asthma, atherosclerosis, multiple sclerosis, psoriasis, rheumatoid arthritis, inflammatory bowel diseases, cancer and HIV infection. Both chemokines and chemokine receptors have become most attractive targets for drug development (Onuffer and Horuk, 2002).
Cytokines act within the cytokine network in all agonistic, antagonistic and synergistic manners. It is not surprising therefore that their biological effects of potential pharmacological interest are not necessarily bound to the typical T cell cytokine phenotypes (Table 4).
Table 4.
Activity | Cytokines |
---|---|
Anti-infectious | Th1: IFN-γ, IL-2, IL-15; Th2: IL-4, -13; others: IFN-α/β/λ, TNF-α, IL-1, -12, -18, GM-, M-, G-CSF; chemokines: MCP-1/CCL2, MIP-1α/CCL3, RANTES/CCL5, MCP-3/CCL7 |
Anti-tumour | Th1: IFN-γ, IL-2, -15; Th2: IL-4, -6, -13; Tr1: IL-10; others: IFN-α/β, TNF-α, IL-12, -18, -23, -24, GM-CSF |
Inflammatory | Th1: IFN-γ; Th2: IL-6; others: TNF-α, IL-1, -12, -18, -19, -22; chemokines: MCP-1/CCL2, MIP-1α/CCL3, RANTES/CCL5, eotaxin/CCL11, MIG/CXCL9, IP-10/CXCL10, I-TAC/CXCL11, Fractalkine/CX3L21 |
Anti-inflammatory | Th2: IL-4, -6, -13; Tr1: IL-10; Th3: TGF-β; others: IL-11, -25, -32 |
Anti-allergic | Th1: IFN-γ; Tr1: IL-10 |
Anti-fibrotic | Th1: IFN-γ; Tr1: IL-10; others: IFN-α/β |
Angiostatic | Th1: IFN-γ, Th2: IL-4, -13; others: IFN-α/β, IL-11, -12; chemokines: PF-4/CXCL4, MIG/CXCL9, IP-10/CXCL10, I-TAC/CXCL11, BCA-1/CXCL13, SLC/CCL21 |
Angiogenic | Th2: IL-6; Th17: IL-17; Th3: TGF-β; others: TNF-α, IL-1, G-CSF, GM-CSF; chemokines: Gro-α, -β, -γ/ CXCL1, 2, 3, ENA-78/CXCL5, GCP-2/CXCL6, NAP-2/CXCL7, IL-8/CXCL8, SDF-1/CXCL12, I-309/CCL1, MCP-1/CCL2, Fractalkine/CX3CL1 |
Hepatoprotective | Th1: IL-15; others: IFN-λ, IL-22 |
Wound healing | Th3: TGF-β; others: TNF-α, IL-1, GM-CSF, FGF, KGFS, ECGF; chemokines: Gro-α/CXCL-1, ENA-78/CXCL5, IL-8/CXCL-8, MCP-1/CCL2, MIP-1α/ CCL3 |
Major references: (Rossi and Zlotnik, 2000; Xing and Wang, 2000), and references in the text.
Cytokine receptors and signalling
Cytokines act through the binding with cytokine receptors that can be grouped in several distinct families on basis of the structural features (Pugh-Humphreys and Thomson, 1998; Vilček, 2003). The cytokine receptors are usually composed of several subunits. For example, the low-affinity receptor for IL-2 (IL-2R) contains only the α subunit (IL-2Rα), while the intermediate affinity IL-2R is composed of the βc and γc subunits (IL-2Rβ, γc/IL-2Rγ respectively). The high-affinity IL-2R contains all three subunits. The receptor subunits are shared by many cytokine receptors. The γc subunit (‘common cytokine receptor γ chain’, initially known as IL-2Rγ) is a component of the receptors for cytokines IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21. These T-cell growth factors thus exhibit overlapping activities. The ‘common cytokine receptor β chain’ (the βc subunit) is shared by receptors for cytokines IL-3, IL-5 and granulocyte-macrophage colony stimulating factor (GM-CSF). The gp130 subunit is shared by IL-6 and IL-11 cytokine receptors.
Ligation of cytokines to the extracellular domains of the membrane-bound cytokine receptor subunits results in homo- or hetero-dimerization of individual subunits. The intracellular domains of the receptor then mediate the signal transduction cascades downstream of cytokine receptors. The signalling pathways depend largely on discrete families of tyrosine kinases.
One of the most important mechanisms of cytokine signalling is the Jak (Janus kinase)-STAT (signal transducer and activator of transcription) pathway (Fujii, 2007). It is used mainly by cytokines that bind with cytokine receptors lacking the intrinsic kinase activity [IFNα/β, IFN-γ, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-17, IL-20, IL-21, IL-22, granulocyte colony stimulating factor (G-CSF), GM-CSF]. The members of the Jak family of cytoplasmic tyrosine kinases (Jak1, Jak2, Jak3, Tyk2) activate the transcription factors of the STAT family (STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b, STAT6) through phosphorylation on a single tyrosine. The STATs form dimers that bind with the promoter sites of target genes. There is an overlapping linkage of different cytokine receptors to a specific array of Jaks and STATs (Liu et al., 1998; Heim, 1999). It is presumed that cytokine specificity is achieved through the integration of the Jak-STAT transactivating pathway with other mechanisms of cytokine signal transduction and transcription (Silvennoinen et al., 1997; Imada and Leonard, 2000).
Only several cytokine receptors contain the kinase activity motifs in their own cytoplasmic domains. The ligand-receptor complexes can thus directly bind and phosphorylate the downstream intracellular signalling substrates. This is an intrinsic property of the receptors for the TGF superfamily of polypeptide growth factors including TGF-β. They signal through the hetero-tetrameric complexes of the type I and type II receptors exhibiting the serine-threonine kinase activity in their intracellular domains. Efficient signalling by certain members of the TGF superfamily further depends on participation of additional co-receptors such as betaglycan (TGF-β type III receptor). The type II receptors phosphorylate the type I receptors; the latter activate the Smad signal transduction pathway. The heteromeric Smad complexes translocate into the nucleus. They bind to DNA directly or indirectly and regulate gene expression (Wrana et al., 1994; Zhu and Burgess, 2001; Miyazawa et al., 2002).
Chemokines are ligands of the rhodopsin family of G protein-coupled seven-helix transmembrane receptors (Devalaraja and Richmond, 1999; Schwarz and Wells, 1999; Onuffer and Horuk, 2002). There are at least 10 distinct β-chemokine (CCR) and seven α-chemokine (CXCR) receptors, which bind β-chemokines (CCL) and α-chemokines CXCL) respectively. Each chemokine has affinity to various chemokine receptors. These receptors often show overlapping ligand specificities (Table 3). The receptor downstream signalling involves multiple pathways including mitogen-activated protein kinases and tyrosine and serine-threonine kinases. Important role is played by mobilization of intracellular Ca2+ following activation of phospholipase C.
In addition to the membrane-bound cytokine receptors, a number of them exist in a soluble form (Pugh-Humphreys and Thomson, 1998; Levine, 2004). Soluble cytokine receptors usually function as natural antagonists for the biological actions of the respective cytokines.
Cytokines in diseases
Gross alterations in cytokine production have been observed in human diseases of different etiological background. They include acute inflammation: wound healing (Gharaee-Kermani and Phan, 2001), chronic inflammation: rheumatoid arthritis (McInnes and Schett, 2007), atherosclerosis (Kleemann et al., 2008), asthma and allergy (Pease, 2006; Holgate and Polosa, 2008), chronic obstructive pulmonary disease (de Boer, 2005), autoimmunity: multiple slerosis (Szczuciński and Losy, 2007), lupus erythematosus (Lit et al., 2007), neurodegeneration: Alzheimer's disease (Tarkowski, 2002), Parkinson's disease (Sawada et al., 2006) and neoplasia (Oppenheim et al., 1997).
Many diseases have been suggested to be associated with a shift in levels of Th1/Th2 cytokines. A typical disease with the shift towards the Th2 immune response is tuberculosis caused by Mycobacterium tuberculosis. Accordingly, the Th1 cytokine IFN-γ has proved useful in the treatment of drug-resistant pulmonary disease (Suárez-Méndez et al., 2004). Insufficient production of Th1 cytokines is associated with increased susceptibility to infections and tumours (Xing and Wang, 2000). The Th1 cytokines IFN-γ, IL-2, TNF-α and TNF-β are effective to eliminate cancer cells whereas the Th2 cytokines are inhibitory on Th1-mediated anti-cancer effects (Gorelik et al., 1994; Takeuchi et al., 1997).
It is believed that restoration of the balance of the Th1/Th2 immune response may be a key to successful immunotherapeutic interventions (Murreille and Leo, 1998). However, a number of findings suggest that this concept requires a thorough re-evaluation as its general implication for human diseases suffers from considerable simplification (Kidd, 2003). The polarized production of Th1 and Th2 cytokines represents extremes in a cytokine spectrum. Intermediate patterns are frequently observed (Openshaw et al., 1995). Inconsistent findings may result at least partially from methodological problems. One of them is the timing of cytokine assays. The Th1 and Th2 cytokines have been observed to be sequentially up-regulated in the atopic asthmatic sensitized state. The airway smooth muscle cells exhibit an early increased mRNA expression of the Th2 cytokines (IL-5), later followed by enhanced mRNA expression of the Th1 cytokines (IFN-γ, IL-2, IL-12) and their receptors (Hakonarson et al., 1999). Similarly, while enhanced levels of Th2 cytokine IL-4 are observed in patients at early stage of rheumatoid arthritis, the Th1 cell clones producing IFN-γ and IL-2 dominate the advanced stages of the disease (Gerli et al., 2002). Conflicting data have been reported for the Th1/Th2 immune response in AIDS. Some data demonstrate a massive switch from Th1 to Th2 cytokines in HIV-infected patients (Clerici and Shearer, 1992). However, this has not been confirmed by other findings (Graziosi et al., 1994).
There are other sound reasons requiring revision of the Th1/Th2 cytokine dichotomy as a disease biomarker. It has been obsrved that diseases consensually considered as Th1 or Th2 diseases may coexist in the same patient. Asthma, a ‘Th1-disease’, has been found to occur more frequently in patients who suffer concomitantly from the ‘Th2-diseases’, such as celiac disease, insulin-dependent diabetes mellitus or rheumatoid arthritis, than in patients without these diseases (Kero et al., 2001). The evaluation of the role of Th1 and Th2 cytokines in lupus erythematosus and other autoimmune diseases has revealed that both classes of cytokines can modify the disease (Theofilopoulos et al., 2001). Although the pathogenesis of atopic disorders, such as allergen-induced asthma, anaphylaxis and rhinoconjunctivities, is associated with Th2 cytokines (IL-4, IL-5, IL-13), the conventional immunotherapy down-regulating these factors has shown only partial therapeutic effectiveness (Lewis, 2002).
Altogether, it seems unlikely that the dichotomous Th1 versus Th2 disease patterns can sufficiently characterize the disease complexity. The investigations on the role of cytokines in diseases remain still incomplete and much effort is needed to achieve more reliable views in this field. Undoubtedly, the models need substantial accomplishments, adding to the existing knowledge novel data on the biological functions of other cytokines including chemokines.
Cytokine and anti-cytokine therapies
It is often unclear whether the disease-associated changes in cytokine production are just an epiphenomenon or an etiological principle of the disease. Should it be a causal factor, it remains to be elucidated which of the cytokines or a group of cytokines can be regarded as disease-relevant targets. Despite the enigmatic role of cytokines in disease etiology, both cytokine and anti-cytokine therapies have been adopted in clinical practice (Table 5). The immunotherapeutic strategies have proved to be useful in various diseases, including hepatitis B and hepatitis C (IFN-α), chronic granulomatous disease (IFN-γ), chronic obstructive pulmonary disease (anti-TNF-α), multiple sclerosis (IFN-β, IL-10, anti-IL-12), rheumatoid arthritis (IL-10, IL-11, anti-TNF-α, anti-IL-1, anti-IL-6), asthma (anti-IL-4, anti-IL-5, anti-IL-13), psoriasis (IL-10, IL-11, anti-IL-12, anti-TNF-α), Crohn's disease (IL-10, IL-11, anti-IFN-γ, anti-TNF-α, anti-IL-1), ulcerative colitis (IL-10, IL-11), cancer (IL-2) and melanoma (anti-IL-8), scleroderma (anti-TGF-β) and others.
Table 5.
Cyokines | Pharmaceutical products | Disease targets |
---|---|---|
IFN-α | •Recombinant IFN-α2a: roferon; peginterferon α2a •Recombinant IFN-α2b: IntronA; peginterferon α2b; albinterferon •Others: wellferon (IFN-αn1); alferon (IFN-αn3) | Hepatitis B (+lamivudin), hepatitis C (+ribavirine) (Cooksley et al., 2003; Clark, 2007) |
IFN-β | •Recombinant IFN-β1a: avonex; rebif •Recombinant IFN-β1b: betaseron; berlex •Chemically modified IFN-β: soluferon | Multiple sclerosis (Bermel and Rudick, 2007) |
IFN-γ | •Recombinant IFN-γ1b: actimmune •Humanized anti-IFN-γ Ab: fontolizumab | Chronic granulomatous disease (actimmune) (Marciano et al., 2004); Crohn's disease (fontolizumab) (Hommes et al., 2006) |
TNF-α | •Fully human anti-TNF-α mAb: adalimumab; golimumab •Mouse/human chimeric IgG1anti-TNF-α mAb: infliximab •Mouse F(ab')2 anti-TNF-α fragment: afelimomab •Humanized mouse anti-TNF-α Ab: CDP-571 •Analogue of TNF-αR: etanercept •Recombinant human sTNFR p55: onercept | Rheumatoid arthritis, psoriasis, Crohn's disease, ankylosing spondylitis, chronic obstructive pulmonary disease, sepsis (afelimomab), juvenile idiopathic arthritis, asthma (etanercept) (Vincent, 2000; Danese et al., 2006; Fan and Leong, 2007; Zhou et al., 2007) |
G-CSF | •Recombinant G-CSF: filgrastim; lenograstim; pegfilgrastim | Febrile neutropenia, bone-marrow transplantation (Cheng et al., 2007) |
GM-CSF | •Recombinant human GM-CSF: sargramostim; molgramostim | Neutropenia after chemotherapy (Waller, 2007) |
TGF-β1 | •Human anti-TGF mAbs: CAT-192 (metelimumab); CAT-152 | Scleroderma, prevention of scarring induced by glaucoma surgery (Pinkas and Teicher, 2006) |
IL-1 | •Recombinant IL-1ra (an IL-1RI-binding molekule): anakirna Human IL-1R1-Fc IgG1 fusion protein: IL-1 Trap | Rheumatoid arthritis, juvenile idiopathic arthritis, Still's disease, Crohn's disease (Dinarello, 2005; Fan and Leong, 2007) |
IL-2 | •Recombinant IL-2: aldesleukin; telecleukin; proleukin •Human/mouse chimeric anti-IL-2Rα mAb: basiliximab •Humanized anti-IL-2Rα mAb: daclizumab | Metastatic renal cell carcinoma (aldesleukin, telecleukin) (Schmidinger et al., 2004; Akaza et al., 2006); renal transplantation (basiliximab, daclizumab) (Swiatecka-Urban, 2003) |
IL-4 | •Soluble recombinant human IL-4R: altrakincept | Asthma (Hendeles et al., 2004) |
IL-5 | •Humanized anti IL-5 mAb: mepolizumab | Asthma (Leckie et al., 2000) |
IL-6 | •Humanized anti-IL6R mAb: atlizumab; tocilizumab | Rheumatoid arthritis (Genovese, 2005; Lipsky, 2006) |
IL-8 | •Fully human anti-IL-8 Ab: ABX-IL8 | Melanoma (Huang et al., 2002) |
IL-10 | •Recombinant human IL-10: ilodecakin | Crohn's disease, rheumatoid arthritis, psoriasis, ulcerative colitis, multiple sclerosis (Marshall, 1999) |
IL-11 | •Recombinant human IL-11: oprelvekin | Thrombocytopenia, ulcerative colitis, psoriasis, rheumatoid arthritis, Crohn's disease (Sitaraman and Gewirtz, 2001) |
IL-12 | •Humanized anti-IL-12 Ab: ABT-874 | Crohn's disease (suggested) (Sandborn, 2004) |
IL-12 + IL-23 | •mAb against p40 subunit of IL-12 and IL-23: ustekinumab | Psoriasis, psoriatic arthritis, multiple sclerosis, Crohn's disease (Wittig, 2007) |
IL-13 | •Humanized anti-IL-13 IgG1 mAb: Ab-01; Ab-02 | Asthma (preclinical studies) (Vugmeyster et al., 2008) |
EPO | •Recombinant erythropoietin: epoetin alfa | Anaemia (Voravud and Sriuranpong, 2005) |
The effectiveness of the immunotherapies may vary, however. No evidence of efficacy has been found in some cases. For example, the human anti-TGF-β Ab (CAT-192) has remained ineffective to treat the cutaneous systemic sclerosis (Denton et al., 2007). The therapeutic effectiveness of IL-2, widely used in oncology, is generally considered significant but rather low (Bambust et al., 2007). IL-2 therapy produces overall response rates of 15–20% in patients with metastatic renal cell carcinoma, but it is associated with serious toxicities affecting all vital organ systems (Dutcher, 2002). It is uncertain whether the dose and combination of IL-2 (aldesleukin) with other agents substantially influence the treatment of renal cell carcinoma (Schmidinger et al., 2004). Recombinant IFN-α2 has beneficial effects in about 30% of patients with well-compensated chronic hepatitis C (Perry and Wilde, 1998). A mainstay in multiple sclerosis treatment is IFN-β. It decreases the progression of disability in multiple sclerosis patients by 30% and reduces relapse rate by 30–50% (Clerico et al., 2007). Highly appreciated is the prophylactic, long-lasting effectiveness of IFN-γ to reduce infections in patients with chronic granulomatous disease (Marciano et al., 2004).
It is supposed that poor efficacy of cytokine and anti-cytokine therapies may be in part due to unfavourable bioavailability and pharmacokinetic profile of the agents. In order to improve these parameters, great attention is paid to pharmaceutical modifications of drugs. One of the most frequently applied approaches is the pegylation of cytokines and anti-cytokine monoclonal antibodies (mAb). It has been successfully applied to IFN-α (peginterferon α2a, peginterferon α2b), G-CSF (pegfilgrastim), mAbs against TNF-α (certolizumab pegol) (Blick and Curran, 2007). Pegylated human IL-1 receptor antagonist (IL-1ra) is under development (Yu et al., 2007). Solubility and bioavailability of IFN-β has increased up to sixfold after replacement of hydrophobic amino acids and cystein by hydrophilic serine in the protein structure (soluferon) (Eisle and von Henneicke, 2007). Another possible variant of pharmaceutical form of cytokine drugs is a binding of cytokines to high-molecular-weight proteins. Fusion of IFNα-2b to human albumin (albinterferon) improves the anti-HCV activity of IFN and extends its elimination half-time as compared with pegylated IFN. Reduced dosing frequency and improved tolerability and compliance have thus been achieved (Subramanian et al., 2007). The cytokine-fusion platform can facilitate a specific tissue targeting of cytokines. For example, the therapeutic potential of IL-10 to treat liver cirrhosis may be enhanced through its fusion with manose-6-phosphate that binds to specific receptors on activated hepatic stellate cells (Rachmawati et al., 2007).
However, it remains unlikely that the therapeutic usage of any individual cytokine can provide complete resolution of the disease. The major limitation is the pleiotropic nature of cytokines and integrated alterations within the cytokine network in diseased organism. The enhancement of efficacy of immunotherapeutic treatments may therefore lead only through more complex and novel strategies.
A promising approach how to overcome the drawbacks of systemic administration of cytokines, that is, to enhance therapeutic effectiveness of cytokines and decrease their toxicity, might be a cytokine gene therapy. In principle, cytokine genes in viral vectors are transduced into cells (e.g. tumour cells, fibroblasts, macrophages) or tissues. Cytokines are then produced locally, at the sites of injury. Preclinical studies have confirmed a proof-of-principle in animal models of disease, using various cytokines (Hao and Shan, 2006). The IFN-α gene therapy has proved effective in a mouse model of human superficial bladder cancer (Adam et al., 2007). Administration of adenoviruses genetically manipulated to express IL-4 or IL-13 cytokine genes results in antiangiogenic effects in adjuvant-induced arthritis in rats (Haas et al., 2006; Haas et al., 2007). The field is now at the early stage of moving towards human trials (Loisel-Meyer et al., 2008).
It should be recognized that any cytokine is probably a double-edge sword meaning both beneficial and detrimental effects to human health.
For example, enhanced levels of pro-inflammatory cytokines, such as IFN-γ, TNF-α, TNF-β, IL-1, IL-6 and IL-12, have been suggested to play a major role in the development of tissue damage in autoimmune diseases (La Cava and Sarvetnick, 1999). The IFN-γ treatment of patients with multiple sclerosis may induce exacerbations of the disease (Panitch et al., 1987). Increased production of IFN-γ has been found to precede clinical attack of multiple sclerosis (Beck et al., 1988). Also IFN-α is an important inducer of autoimmunity. Nearly 20% of patients with malignant tumours and receiving long-term treatment with IFN-α eventually manifest an autoimmune disease, including systemic lupus erythematosus (Rönnblom et al., 2006). A frequent problem seen with IFN-α (not IFN-γ) therapy is induction of thyroid autoantibodies (Ioannou and Isenberg, 2000).
Enhanced levels of IFNs, IL-1, IL-6 and TNF-α are associated with the development of serious episodes of depression (Corcos et al., 2002). IL-2 is a major producer of vascular leak syndrome (Gallagher et al., 2007). Although IL-1 does not produce vascular leak syndrome, vasodilation and hypotension are the dose-limiting toxicities of IL-1 (Oppenheim et al., 1997). Systemic IL-1 markedly enhances ischaemic brain injury via release of neutrophils into circulation, neutrophil adhesion to injured cerebrovasculature and central nervous system invasion, and cell death via activation of matrix metalloproteinase-9. IL-1 can promote metastatic spread as observed in tumour-bearing mice (Bani et al., 1991). Tumour progression is also associated with chemokine monocyte chemoattractant protein (MCP)-1/CCL2, a key factor attracting macrophages to tumours. It has been shown that low to intermediate levels of the chemokine contribute to melanoma development (Gazzaniga et al., 2007). IL-6 is known to have protective effects on survival of neurons. On the other hand, it may be associated with degeneration and cell death in neurological disorders such as Alzheimer's disease (Gadient and Otten, 1997). One of the many biological activities of IL-6 is up-regulation of the hepcidin. This peptide hormone in turn inhibits the supply of iron into plasma. The cumulative deficit of iron is then manifested as anaemia of inflammation, known as anaemia of chronic disease (Ganz and Nemeth, 2006). The immunosuppressive effects of IL-10, promising in the treatment of autoimmunity, are supposed to be one of the mechanisms that contribute to the escape of tumour cells from the local immunosurveilance (Kim et al., 1995). IL-17 is essential in anti-microbial defence of the host, but it promotes bone destruction in arthritis, and augments the activity of osteoclastogenic cytokines TNF-α and IL-1β (Yu and Gaffen, 2008). It enhances angiogenesis and increases in vivo growth of human non-small cell lung cancer (Numasaki et al., 2005).
Prospective candidates for drug development
Chemokines and chemokine receptors
A number of compounds, which are able to antagonize the chemokine receptors, have been suggested as promising drug candidates (Onuffer and Horuk, 2002; Houshmand and Zlotnik, 2003).
The important targets for drug development are chemokine receptors CXCR1 and CXCR2, which bind many CXCL chemokines including IL-8/CXCL8. They are involved in etiopathogenesis of diseases, such as sepsis, atherosclerosis, rheumatoid arthritis, psoriasis and chronic obstructive pulmomary disease. A non-competitive allosteric blocker of these receptor is benzeneacetamide reparixin (syn. repertaxin), which reduces the IL-8/CXCL8-mediated adhesion of polymorphonuclear cells. It is currently clinically investigated for the use in the prevention of ischaemia/reperfusion injury in organ transplantation (Casilli et al., 2005). The function of CXCR1 and CXCR2 receptors is also efficiently antagonized by 3,4-diamino-2,5-thiadiazole-1-oxides (Biju et al., 2008).
The chemokine receptor CXCR3 is involved in rheumatoid arthritis, multiple sclerosis, psoriasis and allograft rejection. The CXCR3 ligands are chemokines MIG/CXCL9, IP-10/CXCL10 and I-TAC/CXCL11. Several compounds of different chemical classes that potently antagonize the action of IP-10/CXCL10 and I-TAC/CXCL11 at the human CXCR3 receptor have recently been discovered (VUF10472, VUF10085/, VUF5834, VUF10132, TAK-779) (Verzijl et al., 2008). Novel di-substituted cyclohexanes, effective in nanomolar concentration, are antagonists of CCR2 receptor (Cherney et al., 2008). A series of bipiperidinyl carboxylic acid amides have proved to be potent and selective antagonists of the chemokine receptor CCR4. They might be useful in asthma, allergy, diabetes and cancer (Kuhn et al., 2007). A promising candidate for treatment of HIV-1 infection is the CCR5 receptor antagonist vicroviroc, an analogue of pyrimidine, 5-({4-[(3S)-4-{2-methoxy-1-[4-(trifluoromethyl)phenyl]ethyl}-3-methylpiperazin-1-yl]-4-methylpiperidin-1-yl}carbonyl)-4,6-dimethylpyrimidine (Strizki et al., 2005). Another co-receptor of HIV entry in cells is the chemokine receptor CXRC4. A number of new antagonists of CXCR4 have been identified. The most attractive of them are bicyclam derivatives (Grande et al., 2008).
There are only a few compounds known to directly inhibit synthesis of chemokines. One of them is bindarit, 2-methyl-2-[[1-(phenylmethyl)-1H-indazol-3yl]methoxy]propanoic acid. It selectively inhibits production of the monocyte chemotactic proteins MCP-1/CCL2, MCP-2/CCL8 and MCP-3/CCL7. This effect along with inhibition of TNF-α is a plausible explanation for therapeutically promising anti-inflammatory effects of bindarit in experimental models of pancreatitis, arthritis, lupus nephritis and colitis (Bhatia et al., 2008; Mirolo et al., 2008). The sub-antimicrobial doses of macrolide antibiotics (clarithromycin, roxithromycin, azithromycin, erythromycin) have been found effective in treatment of asthma, diffuse panbronchiolitis, inflammatory bowel disease and arthritis. It has been suggested that beneficial effects may be due to the suppression of cytokines, including chemokines IL-8/CXCL8 and MIP-1α/CCL3 (Shinkai et al., 2008).
Agonists of toll-like receptors
A special class of agents with prevailing stimulatory effects on production of IFNs are ligands of toll-like receptors (TLRs). The TLRs belong to a superfamily of pattern-recognition receptors playing a crucial role in the detection of molecular patterns of extracellular and intracellular pathogens. So far, 10 members of TLR family have been revealed in humans. The endosomally localized TLR9 recognizes unmethylated CpG (cytosine-phosphate-guanine dinucleotide) motifs of bacterial and viral DNA. This leads ultimately to rapid activation of innate immune responses. A number of phosphorothioate-modified oligodeoxynucleotides with immunostimulatory sequences (CpG ODNs) have been synthesized and used in clinical trials I–III. They target diseases such as hepatitis B, hepatitis C, influenza, anthrax, asthma, allergy, non-Hodgkins lymphoma, melanoma and refractory solid tumours. The agonists of the TLR9 are major activators of type 1 IFNs (IFNα/β). They can produce other cytokines, for example, IFN-γ, IL-6, IL-10 and IL-1ra, as well (Verthelyi et al., 2001; Dalpke et al., 2002; Kandimalla et al., 2005; Krieg, 2006). Therapeutic potential is also possessed by agonists of other TLRs, such as imidazoquinoline derivatives imiquimod and resiquimod. These agents act through the TLR7 and TLR8. Imiquimod was initially developed as an antiviral agent, and has been approved as a widely used immune response modifier for topical treatment of external genital warts, actinic keratoses and superficial basal cell carcinomas (Gaspari, 2007). Imidazoquinoline derivatives are inducers of IFNs IFN-α and IFN-γ. They also activate secretion of other cytokines (IL-1β, IL-6, TNF-α, IL-12p40) and chemokines (IL-8/CXCL8, MIP-1α/CCL3 and MCP-1/CCL2) (Gupta et al., 2004; Thomsen et al., 2004). On the other hand, imiquimod inhibits production of Th2 cytokines IL-4 and IL-5 (Wagner et al., 1999). TLR7 can be activated by certain guanosine analogues, such as 7-thia-8-oxoguanosine and 7-deazaguanosine. They stimulate secretion of IFN-α/β, IFN-γ, IL-6, IL-12 and TNF-α (Lee et al., 2003).
Suppressors of cytokine signalling
Except of having physiological roles in developing organism (Kile and Alexander, 2001), members of the cytokine signalling (SOCS) family of intracellular proteins (SOCS1–7 and CIS) possess crucial roles in the negative regulation of activity of many cytokines. They act through association with Jak-STAT signalling pathways (Yasukawa et al., 2000; Krebs and Hilton, 2001). SOCSs are rapidly induced by various cytokines, for example, IL-1β, IL-6, IFN-γ (Dogusan et al., 2000), IFN-α (Wang et al., 2000) and IL-10 (Cassatella et al., 1999).
The expression of SOCSs can be modulated by various physiological mediators. SOCS-1 is up-regulated by thrombopoietin (Wang et al., 2000). Phosphorylation of SOCS-3 is induced by insulin (Peraldi et al., 2001), up-regulated by growth hormone while it is down-regulated by glucocorticoids (Paul et al., 2000). SOCS proteins play an important role in differentiation of Th1, Th2, Th17 and Treg cells. They are thus involved in diseases of immune etiopathogenesis (Yoshimura et al., 2007). SOCS3 expression is tightly correlated with pathology of asthma and atopic dermatitis. It has been suggested as a new therapeutic target for the development of antiallergic drugs (Seki et al., 2003). The SOCS3 therapy may be useful in the treatment of cancer (He et al., 2003), the SOCS1 therapy in type 1 diabetes (Flodström-Tullberg et al., 2003). Both SOCS1 and SOCS3 have been suggested as factors involved in the resistance to IFN therapy of hepatitis C infection (Vlotides et al., 2004; Imanaka et al., 2005).
Both positive and negative pharmacological manipulation of SOCS proteins would be desirable, although there are no special drugs available in this field. A promising approach is the development of SH2 domain inhibitors targeting protein tyrosine kinase signalling pathways involved in mechanism of SOCS action (Machida and Mayer, 2005). Interestingly, acetylsalicylic acid inhibits the STAT6 signalling pathways mediated by IL-4 and IL-13. This may be a mechanism of beneficial effects of aspirin and salicylate treatment of allergic diseases, including asthma (Perez et al., 2002). The synthetic cannabinoid derivative PRS-211,092 devoid of psychotropic activity stimulates expression of SOCS1 and SOCS3, cytokines IL-6 and IL-10, and inhibits production of IFN-γ, TNF-α, IL-1β, IL-2 and MCP-1/CCL2 (Lavon et al., 2003).
Inhibitors of cytokine maturation
Certain cytokines are produced in a form of precursors that must be further processed to become mature cytokines. The process of maturation is mediated by various endogenous factors. For example, the caspase-1 is important in a chain of proteolytic transformations of IL-1β precursor (pro-IL-1β) to mature IL-1β (Beuscher et al., 1990). It also participates in the processing of the IL-18 precursor (pro-IL-18) to fully active IL-18 (Shtrichman and Samuel, 2001). Caspase 3 is involved in the final synthesis of immunomodulatory IL-16 protein from the 80 kD precursor, pro-IL-16 (Zhang et al., 1998). The regulatory cytokine TGF-β is formed as prepro- and pro-TGF-β precursors, requiring activation via a cascade of processes including proteolytic activation (Li et al., 2006). TNF-α is cleaved from the 26 kDa membrane-bound molecule to the active 17 kDa form. This is done by the TNF-α-converting enzyme (TACE; also referred to as a disintegrin and metalloproteinase 17, ADAM17) (Tsuji et al., 2002). The development of specific agents that would target these processes could be of interest.
So far several inhibitors of TACE were revealed. Marimastat, N-[2,2-dimethyl-1-(methylcarbamoyl)propyl]-2-[hydroxy-(hydroxycarbamoyl)methyl]-4-methyl-pentanamide, almost completely inhibits the lipopolysaccharide-induced soluble TNF-α production (Tsuji et al., 2002). TACE has become a validated therapeutic target for the development of oral TNF-α inhibitors. Very potent is a novel agent TMI-1, (2R, 3S)-2-([[4-(2-butynyloxy)phenyl]sulfonyl]amino)-N,3-dihydroxybutanamide. It inhibits the spontaneous release of TNF-α in human synovium tissue explants from patients with rheumatoid arthritis. TMI-1 effectively reduces adjuvant-induced arthritis in rats (Zhang et al., 2004).
Natural compounds – botanicals
Natural products, notably composite plant products (botanicals) have long been recognized as anti-cancer supportive remedies (Diwanay et al., 2004), and anti-infectious agents (Cowan, 1999) including HIV (Asres et al., 2005). Their beneficial effects may be at least partially mediated by multiple interventions with cytokine expression (Spelman et al., 2006). Herbal and other natural products thus represent a rich source of potential drugs including immunomodulatory agents. A huge effort is presently being done to reconcile the healing experience of traditional medicines with Western medical practice and research (Patwardhan and Gautam, 2005). The search for new therapeutic means is greatly facilitated by recent extensive progress in phytochemistry, analytical biochemistry, biochemistry and bio-analytical methods allowing isolation and identification of the bioactive principles in botanicals (Gullo et al., 2006). These approaches should hopefully overcome the problems inherent to herbal medicines, that is, standardization of the chemical content, their possible contamination with heavy metals, quality of the raw material, etc. (Khan, 2006). The following few examples should be taken as an un-exhaustive demonstration of the vast range of immune activities that many chemically identified compounds, isolated from the original herbal remedies, may provide.
Many flavonol compounds, such as kaempferol, quercetin and, more potently, fisetin, luteolin and apigenin, have been revealed to substantially inhibit production of Th2 cytokines IL-4, IL-5 and IL-13 (not IL-6 and IL-8/CXCL8) by human basophils (Higa et al., 2003; Hirano et al., 2004).
Steroidal lactones isolated from Withania somnifera (withanolides) have been reported to induce the Th1 (IFN-γ, IL-2), but not Th2 (IL-4) cytokines (Bani et al., 2006). Sesquiterpene lactone of guianolide-type thapsigargin from Thapsia garganica is a potent inducer of IFN-γ by human peripheral blood mononuclear cells (Kmoníčkováet al., 2008). Selective affinity to the Th1 immune response is characteristic for naphthopyran derivatives isolated from Eleuthrine americana. While isoleutherin activates production of IFN-γ and IL-2 in CD4+ Th mouse cells, eleutherinol inhibits the both (Hong et al., 2008).
Another inducer of IFN-γ is melanin that is present in many botanicals (e.g. Nigella sativa) traditionally used as immune enhancers. It also stimulates production of IL-6, and TNF-α by human peripheral blood mononuclear cells (El-Obeid et al., 2006). However, opposite effects, that is, inhibition of TNF-α, IL-1β, IL-6 and IL-10 (but not GM-CSF), have been observed with synthetic melanin in lipopolysaccharide-stimulated human peripheral blood mononuclear cells (Mohagheghpour et al., 2000).
Botanical agents, such as proanthocyanidins from Vitis vinifera, silymarin from Silybum marianum and polyphenols from Camelia sinensis, are inducers of IL-12 and suppressors of IL-10. These effects have been suggested as a plausible mechanism of chemoprotection against UV-induced immune suppression and photocarcinogenesis (Katiyar, 2007).
A number of natural compounds, including polyphenols (e.g. resveratrol, quercetin, luteolin, hesperetin, kaempferol, scopoletin, aucubin, nardostachin, honokiol), alkaloids (lycorine), terpenes (acanthoic acid, tanshinone), sterols (guggulsterol) and other chemical classes, have been revealed as inhibitors of production of TNF-α, IL-1β and IL-6 (Paul et al., 2006). These pro-inflammatory cytokines also are down-regulated by a polyphenol curcumin from Curcuma longa (Gonzales and Orlando, 2008). The mechanism of action is obviously inhibition of the TLR4 signalling induced by lipopolysaccharide (Youn et al., 2006).
Potent inhibitors of TGF-β production are emodin6-methyl-1,3,8-trihydroxyanthraquinone from Rheum emodi (Wang et al., 2007) and magnolol, 4-allyl-2-(5-allyl-2-hydroxyphenyl)phenol from Magnolia officinalis (Kim et al., 2007).
Fungal metabolites isolated from mycelia of Verticimonosporium ellipticus, bis-thiodiketopiperazines (emestrins) and cytochalasins are potent antagonists of chemokine receptor CCR2, a receptor for MCP-1/CCL2. The compounds are thus interesting agents for treatment of inflammatory processes associated with rheumatoid arthritis, multiple sclerosis and atherosclerosis (Herath et al., 2005).
A special position among natural products is possessed by a widely used coffee (caffeine) and relatively widely abused cannabis (cannabinoids). Tetrahydrocanabinol (THC), the major active constituent of marijuana, is recognized for its general immune suppressive activity. THC inhibits production of TGF-β (Gardner et al., 2002), TNF-α, GM-CSF, chemokines IL-8/CXCL8, MIP-1α/CCL3, MIP-1β/CCL4 and RANTES/CCL5 (Srivastava SrivastavaB et al., 1998), and Th1 cytokines IFN-γ, IL-12 (Newton et al., 2004) and IL-2 (Condie et al., 1996). However, cannabinoids inhibit the Th2 immune response as well. In vivo administration of cannabinol or THC attenuates the elevation of IL-4, IL-5 and IL-13 steady-state mRNA expression elicited by ovalbumin challenge in the mouse lungs. These data suggest that cannabinoids might be beneficial in the treatment of allergic airway disease (Jan et al., 2003). The THC-induced inhibition of cytokines probably does not depend on CB1 or CB2 cannabinoid receptors (Puffenbarger et al., 2000). The major non-psychoactive cannabinoid in marijuana, cannabidiol suppresses secretion of all IFN-γ, IL-2 and IL-4 (Jan et al., 2007).
The plant cannabinoids cannabidiol, cannabigerol, cannabichromene, cannabidiol acid and THC have been reported to supress tumour growth, the most effective being cannabidiol (Ligresti et al., 2006). However, there also are opposite findings. The immune inhibitory cytokines IL-10 and TGF-β have been found up-regulated at both the tumour site and in the spleens of THC-treated mice. It has been suggested that the THC may promote the tumour growth by inhibiting antitumour immunity through the CB2 receptor-mediated cytokine-dependent pathway (Zhu et al., 2000).
The alkylamides (alkamides) from Echinacea, dodeca-2E,4E,8Z,10Z-tetraenoic acid isobutylamide and dodeca-2E,4E-dienoic acid isobutylamide, bind to the CB2 receptor more strongly than the endogenous cannabinoids. Similar to the endogenous cannabinoid anandamide, they up-regulate constitutive IL-6 expression in human whole blood and inhibit TNF-α, IL-1β and IL-12p70 expression (Raduner et al., 2006).
Caffeine possesses prominent cytokine-inhibitory effects. It suppresses production of all Th1 (IL-2, IFN-γ), Th2 (IL-4, IL-5) and T-regulatory (IL-10) cytokines (Horrigan et al., 2006).
Probiotics
Probiotics are defined as ‘live organisms which when consumed in adequate numbers confer a health benefit on the host’ (Guarner and Schaafsma, 1998). Probiotics are typically lactic acid bacteria selected from the gut flora that are resistant to stomach acid and bile. They are mainly used for prevention and therapy of various gastrointestinal diseases (Damaskos and Kolios, 2008). The example of an effective probiotic therapy is the treatment of diarrhea caused by rotavirus infection in children (Saavedra, 2000). Lactobacilli have been demonstrated beneficial in reducing the urinary tract infections and bacterial vaginosis (Hoesl and Altwein, 2005), atopic dermatitis in children (Kalliomäki et al., 2003), and pouchitis associated with colon surgery in ulcerative colitis (Cottone et al., 2006).
Bacterial probiotic strains enhance the Th1 immune response in inducing secretion of IFN-γ both in vitro (Shida et al., 2006) and in vivo in humans (Meyer et al., 2007). Probiotics substantially elevate cytokines supporting the Th1 immune response, such as IL-12, IL-18 and pro-inflammatory cytokines TNF-α and IL-1 (Miettinen et al., 1998; Cross et al., 2004). The type I IFNs (IFN-α/β) have remained uninfluenced after Lactobacillus casei strain Shirota, or Escherichia coli Nissle and E. coli 2282 (Cross et al., 2004). The Th2 cytokine IL-4 was either uninfluenced in humans in vivo (Meyer et al., 2007) or, together with IL-5, down-regulated by oligodeoxynucleotide BL07S from a probiotic strain of Bifidobacterium longum in a murine model (Takahashi et al., 2006). Production of another Th2 cytokine IL-6 by small intestinal epithelial cells was enhanced by a milk fermented with Lactobacillus helveticus R389 (Vinderola et al., 2007), and by other types of lactobacilli, Lactobacillus rhamnosus E509, E522 and Lactobacillus bulgaricus E585 in human peripheral blood mononuclear cells (Miettinen et al., 1998). The secretion of the regulatory and immunosuppressive cytokine IL-10 was activated by many types of probiotic bacteria (Shida et al., 2006; Meyer et al., 2007). On the other hand, no changes in production of the regulatory cytokine TGF-β were observed (Cross et al., 2004). Comensal bacteria with probiotic properties, Bifidobacterium infantis and Lactobacillus salivarius, stimulated secretion of IL-10 and TNF-α, and decreased secretion of IL-8/CXCL8 in human intestinal epithelial cells (O'Hara et al., 2006).
Active principles of probiotics are poorly understood. Unfortunately, the controlled clinical studies in this area are still sparse, and newly performed ones are urgently needed.
Modulation of cytokine secretion by physiological mediators
Endogenous neurotransmitters, second messengers and other mediators of physiological functions are important regulators of cytokine production. Special reviews have been devoted to the effects of adenosine (Abbracchio and Ceruti, 2007; Haskóet al., 2007), α- and β-adrenergics (Haskó and Szabó, 1998), histamine (Packard and Khan, 2003), serotonin and nicotine (Cloëz-Tayarani and Changeux, 2007), cannabinoids (Klein et al., 2003; Massi et al., 2006), melatonin (Carrillo-Vico et al., 2005) and glucocorticoids (Elenkov, 2004).
The effects may be due to the widespread expression of the respective receptors on the cells of immune system. The dopamine D2 and D3 receptor subtypes have been detected in lymphocytes (Ghosh et al., 2003). Macrophages contain both α- and β-adrenoceptors (Miles et al., 1996). Human peripheral blood mononuclear cells express mRNAs for serotonin receptor types/subtypes 5-HT1A, 5-HT1B, 5-HT1E, 5-HT2A, 5-HT3, 5-HT4, 5-HT6 (Yang et al., 2006). Dendritic cells express the mRNA for all H1, H2 and H3 histamine receptors (Idzko et al., 2002). Cholinergic muscarinic receptor subtypes M2, M3, M4 and M5 have been identified in human peripheral blood lymphocytes (Tayebati et al., 2001; Ricci et al., 2008). Immune cells also express the cannabinoid CB2 receptor (Galiegue et al., 1995), and opioid µ3, κ and δ receptors (Wybran et al., 1979; Wick et al., 1996; Welters et al., 2000). All A1, A2A, A2B and A3 adenosine receptors are present on monocytes and macrophages (Haskóet al., 2007). Prostanoid receptors EP1, EP2, EP3 and EP4 are expressed on human macrophages (Ying et al., 2004; Ratcliffe et al., 2007). Intracellular receptors binding glucocorticoids are present in macrophages and monocytes (Werb et al., 1978). Direct modulation of immune cells by oestrogen is facilitated by the presence of oestrogen receptors in all dendritic cells, macrophages and B lymphocytes (Nalbandian and Kovats, 2005).
Both stimulatory and inhibitory effects of distinct endogenous receptor ligands on cytokine production have been observed. The characteristic feature of activation of adenosine receptors is the inhibition of production of Th1 cytokines (IFN-γ, IL-2) and TNF-α, and stimulation of IL-10. Similar effects are produced by prostaglandins and β2-adrenoceptor agonists (Zídek, 1999). Data on regulation of cytokine secretion by other physiological mediators are largely conflicting. Activation of melatonin receptors may be ineffective (Di Stefano and Paulesu, 1994), stimulatory (Liu et al., 2001) or inhibitory (Raghavendra et al., 2001) with respect to the TNF-α secretion. Production of IL-10 has been reported both stimulated (Raghavendra et al., 2001) and inhibited (Kühlwein and Irwin, 2001) by melatonin. Whereas suppression of TNF-α and up-regulation of IL-10 is usually produced by glucocorticoids, no effects on production of these cytokines have also been observed (Miyazaki et al., 2000; Richards et al., 2000).
The inconsistencies may arise from a variety of reasons. Obviously, the origin and type of cells are of a paramount importance. In dependence on the clone of human T cells, histamine does not influence, inhibit or enhance production of IFN-γ (Krouwels et al., 1998). The strongly inhibited expression of IFN-γ and IL-2 genes by histamine has been found preceded by their activation early during immune stimulation (Arad et al., 1996). Isoproterenol, the agonist of β-adrenergic receptors, inhibits the lipopolysaccharide-induced production of IL-6 by rat renal macrophages (Nakamura et al., 1999). In contrast, it up-regulates the IL-6 production by rat thymic epithelial cells (von Patay et al., 1998). Cannabinoids have been found unable to change serum levels of IL-10 in multiple sclerosis patients (Katona et al., 2005), while cannabidiol inhibits IL-10 in mouse peritoneal macrophages (Sacerdote et al., 2005). It should also be distinguished whether the results have been obtained from experiments in vitro, ex vivo or in vitro. Despite of many experimental findings, clear-cut conclusions cannot be drawn in many cases, and mechanisms of the action remain to be elucidated.
Interaction of cytokines with drug pharmacodynamics and pharmacokinetics
Interference of cytokines with cytochrome P450
The mixed-function oxidase system of cytochromes P450 (CYP) is responsible for the metabolism of a wide variety of drugs and their metabolites as well as for biosynthesis of endogenous compounds such as steroid hormones. The study of interactions between cytokines and drug metabolism was initiated by findings showing that many bacteria and their immune-active products can influence drug metabolism. Depression was observed after the treatment of animals with Freund's complete adjuvant (Whitehouse and Beck, 1973), Bacillus Calmette-Guérin (Farquhar et al., 1976), and Corynebacterium parvum (Giampietri et al., 1981). It was soon found that the effect resulted from enhanced production of cytokines (Descotes, 1985).
Effects of Th1 cytokines
It has been suggested that depression of CYP activities may be a common property of all IFN inducers. Changes in production of IFN-γ and/or other cytokines are tightly associated with down-regulation of CYPs and other enzymes resulting in altered bioactivation and detoxication of drugs (Prandota, 2005). The IFN-γ alone (Singh et al., 1982) and IFN-γ-inducing agents, such as tilorone (Leeson et al., 1976) and polyriboinosinic acid : polyribocytidylic acid (polyI : C) (Robbins and Mannering, 1984), depress the in vivo activity of the CYP system. The CYP3A1 and CYP3A2 mRNA, and CYP2C11 proteins have been found reduced by recombinant IFN-γ in cultured rat hepatocytes (Tapner et al., 1996). The type I IFN (IFN-α; peginterferon alfa-2a) decreases the clearance of theophylline. The inhibition of the de novo synthesis of human CYP1A2 has been suggested as a plausible explanation of this effect (Perry and Jarvis, 2001). IFN-α/β produced by polyI : C augment the rate of loss of CYP1A1 and CYP1A2 in rat liver (Delaporte and Renton, 1997). The decrease in activity of CYP1A2 is associated with occurrence of side effects in patients treated with IFN-α2b (Islam et al., 2002). In variance with these data, chronic administration of IFN-α in patients with hepatitis C has not been found to change the in vivo activities of CYP1A2 and CYP3A (Pageaux et al., 1998). IL-2 decreases the total CYP content and the mRNAs and proteins of CYP2C11 and CYP3A in cultured rat hepatocytes (Tinel et al., 1999). IL-2 monotherapy may be associated with decreased total CYP and monooxygenase activities in patients with hepatic metastases (Elkahwaji et al., 1999).
Effects of Th2 cytokines
IL-4 has been found to increase fivefold the expression of CYP2E1 mRNA in primary human hepatocyte cultures (Abdel-Razzak et al., 1993). IL-6 can down-regulate rat (Muntané-Relat et al., 1995) and human (Jover et al., 2002) CYP3A4 activity, and protein content of CYP1A2, CYP2C11, CYP2B1/2 and CYP3A2 in cultured rat hepatocytes (Carlson and Billings, 1996).
Effects of Treg cytokines
TGF-β1 seems to specifically down-regulate the CYP1 enzymes (CYP1A1, CYP1A2). Constitutive expression of other CYP forms (CYP3A1, CYP2B1/2, CYP2E1) remains unaffected by TGF-β in both humans and rats (Abdel-Razzak et al., 1994; Müller et al., 2000). IL-10 has been found to inhibit CYP4F expression, while IL-1β, IL-6 and TNF-α produce a general inductive response of this enzyme in cultured rat hepatocytes (Kalsotra et al., 2007). IL-10 given to human volunteers significantly decreases CYP3A while no significant changes in CYP1A2 and CYP2D6 activities have been observed (Gorski et al., 2000).
Effects of other cytokines
TNF-α can enhance induction of CYP1B1. On the other hand, it simultaneously suppresses the CYP1A1 expression in rat liver epithelial cells. The CYP1B1 induction has been suggested to be associated with enhanced genotoxic effects of carcinogenic polycyclic aromatic hydrocarbons (Umannováet al., 2008). The pro-inflammatory cytokines TNF-α and IL-1 down-regulate the hepatocyte CYP1A2 in sepsis (Crawford et al., 2004; Wu et al., 2006). TNF-α also decreases protein levels of CYP2B1/2 and CYP3A2 in rat hepatocytes (Carlson and Billings, 1996). It decreases protein levels of CYP2C11 and CYP3A2 in rat liver. The expression of CYP2A1 and CYP2C6 remains unchanged (Nadin et al., 1995). IL-1β has been found to antagonize polycyclic aromatic hydrocarbon-induced CYP1A gene expression (Abdel-Razzak et al., 1994), and to depress expression of CYP2B6, CYP2C9 and CYP3A4 in human hepatocytes (Assenat et al., 2004). IL-1β treatment of rat hepatocytes decreases levels of protein content of CYP1A2, CYP2C11, CYP2B1/2 and CYP3A2 (Carlson and Billings, 1996). The impairment of CYP synthesis after IL-1 has been suggested to be due to the suppression of hepatic haeme pool (Kihara et al., 1999).
Mixtures of cytokines are usually more effective than single cytokines to alter expression of CYPs and their activities. The down-regulatory effect on human hepatocyte CYP2B6 is produced by a cocktail of IL-1/TNF-α/IFN-γ (Aitken et al., 2008). Mixture of TNF-α and IL-6 decreases the total CYP, CYP2E1, CYP3A2 and CYP2C11 protein contents, and inhibits activities of CYP2E1, CYP3A2 and CYP2G11 in rat liver microsomes (Vuppugalla et al., 2003).
The mechanisms of the interaction of cytokines with CYP protein and activity are not well understood. The prevailing suppressive effects of cytokines on cytochrome P450 metabolizing system can be mediated by cytokine-induced NO, although not necessarily so (Carlson and Billings, 1996; Aitken et al., 2008).
The effects of cytokines on CYP protein content and/or CYP activities are summarized in Table 6. It can be concluded that cytokine-mediated alterations in total CYP content and activities of individual CYP enzymes may be reflected in changes in drug metabolism. These changes can contribute to frequently encountered variability in drug response and augment the risk of adverse drug effects in patients.
Table 6.
CYP isoform | Cytokines | ||||||||
---|---|---|---|---|---|---|---|---|---|
IFN-γ | IFN-α | TNF-α | TGF-β1 | IL-1β | IL-2 | IL-4 | IL-6 | IL-10 | |
1A1 | |||||||||
1A2 | |||||||||
1B1 | |||||||||
2A1 | |||||||||
2B1/2 | |||||||||
2B6 | |||||||||
2C11 | |||||||||
2C6 | |||||||||
2C9 | |||||||||
2D6 | |||||||||
2E1 | |||||||||
3A | |||||||||
3A1 | |||||||||
3A2 | |||||||||
3A4 | |||||||||
4F | |||||||||
CYP total |
CYP isoform is: decraesed, increased, not changed.
References to support the table are cited in section ‘Interference of cytokines with cytochrome P450’.
Interference of cytokines with P-glycoprotein
Cytokines have been shown to interfere with the intestinal efflux system. One of the important factors of this system is the multidrug resistance (MDR)-associated P-glycoprotein (known as P-gp, MDR1 or ABCB1). IFN-γ dose- and time-dependently reduces cellular uptake of cyclosporine A (but not methotrexate) in human intestinal cells. The effect is associated with activation of P-gp. The activation of efflux system is probably due to IFN-γ-activated nitric oxide production (Dixit et al., 2005). The ABCB1 gene encoding for P-gp has been found stimulated by IFN-γ also in human macrophages (Puddu et al., 1999). In contrast, cytokines TNF-α (Belliard et al., 2004), IL-1 (Sukhai et al., 2001), IL-2 (Belliard et al., 2002), IL-4 (Tambur et al., 1998) and IL-6 (Hartmann et al., 2001) reduce activity of P-gp. TNF-α plays a pivotal role in the down-regulation of P-gp by endotoxin (Miyoshi et al., 2005). Cytokines may also influence the cerebral and hepatic expression of P-gp (Fernandez et al., 2004).
Interestingly, P-gp is involved in the transmembrane transport of cytokines (e.g. IL-1β, IL-2, IL-4 and IFN-γ) out of the cells (Mizutani et al., 2008).
Conclusions
The cytokine compartment of the immune system has evolved phylogenetically to ensure homeostasis of organisms. Dysbalance in cytokine production is associated with numerous diseases. Both cytokine and anti-cytokine immunotherapies have proved to provide beneficial therapeutic effects. Novel therapeutic strategies targeting the cytokine network are needed to enhance the effectiveness of present immunotherapeutic regimens. Drugs with more specific effects on secretion of cytokines are needed. Studies of prospective drug candidates of both natural and synthetic origin require more complex analysis of the effects within the cytokine network. Possible impact of manipulation of cytokine secretion on pharmacokinetic and pharmacodynamic behaviour of drugs should be more systematically evaluated.
Acknowledgments
This work was supported by grants no. 305/08/0535 (GACR) and 1M0508.
Glossary
Abbreviations:
- Ab
antibody
- BCA-1/CXCL13
B cell attracting chemokine-1
- ENA-78/CXCL5
Epithelial-cell derived neutrophil activating factor-78
- GCP-2/CXCL6
Granulocyte chemoattractant protein-2
- G-CSF
granulocyte colony stimulating factor
- GM-CSF
granulocyte-macrophage colony stimulating factor
- Gro-α, -β, -γ/CXCL1, 2, 3
Growth-regulated oncogene-
- IFN
interferon
- IL
interleukin
- IP-10/CXCL10
interferon-inducible protein-10
- I-TAC/CXCL11
interferon-inducible T cell α-chemoattractant
- LT-α
Lymphotoxin-α
- MCP-1/CCL2
monocyte chemoattractant protein-1
- MCP-3/CCL7
Monocyte chemoattractant protein-3
- M-CSF
Macrophage colony stimulating factor
- MIG/CXCL9
Monokine induced by γ-interferon
- MIP-1α/CCL3
Macrophage inflammatory protein-1 alpha
- MIP-1β/CCL4
Macrophage inflammatory protein-1 beta
- NAP-2/CXCL7
Neutrophil-activating protein-2
- PF-4/CXCL4
Platelet factor-4
- RANTES/CCL5
Regulated upon activation, normal T cell expressed and secreted
- SDF-1/CXCL12
Stromal cell-derived factor-1
- SLC/CCL21
Secondary lymphoid-tissue chemokine
- TGF-β
transforming growth factor-β
- TNF-α
tumour necrosis factor-α
Conflict of interest
The authors state no conflict of interest.
References
- Abbracchio MP, Ceruti S. P1 receptors and cytokine secretion. Purinergic Signal. 2007;3:13–25. doi: 10.1007/s11302-006-9033-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Abdel-Razzak Z, Loyer P, Fautrel A, Gautier JC, Corcos L, Turlin B, et al. Cytokines down-regulate expression of major cytochrome P-450 enzymes in adult human hepatocytes in primary culture. Mol Pharmacol. 1993;44:707–715. [PubMed] [Google Scholar]
- Abdel-Razzak Z, Corcos L, Fautrel A, Campion JP, Guillouzo A. Transforming growth factor-beta 1 down-regulates basal and polycyclic aromatic hydrocarbon-induced cytochromes P-450 1A1 and 1A2 in adult human hepatocytes in primary culture. Mol Pharmacol. 1994;46:1100–1110. [PubMed] [Google Scholar]
- Adam L, Black PC, Kassouf W, Eve B, McConkey D, Munsell MF, et al. Adenoviral mediated interferon-alpha 2b gene therapy suppresses the pro-angiogenic effect of vascular endothelial growth factor in superficial bladder cancer. J Urol. 2007;177:1900–1906. doi: 10.1016/j.juro.2007.01.003. [DOI] [PubMed] [Google Scholar]
- Adams DH, Lloyd AR. Chemokines: leucocyte recruitment and activation cytokines. Lancet. 1997;349:490–495. doi: 10.1016/s0140-6736(96)07524-1. [DOI] [PubMed] [Google Scholar]
- Aitken AE, Lee CM, Morgan ET. Roles of nitric oxide in inflammatory downregulation of human cytochromes P450. Free Radic Biol Med. 2008;44:1161–1168. doi: 10.1016/j.freeradbiomed.2007.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Akaza H, Tsukamoto T, Onishi T, Miki T, Kinouchi T, Naito S. A low-dose combination therapy of interleukin-2 and interferon-α is effective for lung metastasis of renal cell carcinoma: a multicener open study. Int J Clin Oncol. 2006;11:434–440. doi: 10.1007/s10147-006-0596-z. [DOI] [PubMed] [Google Scholar]
- Annunziato F, Cosmi L, Santarlasci V, Maggi L, Liotta F, Mazzinghi B, et al. Phenotypic and functional features of human Th17 cells. J Exp Med. 2007;204:1849–1861. doi: 10.1084/jem.20070663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arad G, Nussinovich R, Na'amad M, Kaempfer R. Dual control of human interleukin-2 and interferon-g gene expression by histamine: activation and suppression. Cell Immunol. 1996;170:149–155. doi: 10.1006/cimm.1996.0145. [DOI] [PubMed] [Google Scholar]
- Asres K, Seyoum A, Veeresham C, Bucar F, Gibbons S. Naturally derived anti-HIV agents. Phytother Res. 2005;19:557–581. doi: 10.1002/ptr.1629. [DOI] [PubMed] [Google Scholar]
- Assenat E, Gerbal-Chaloin S, Larrey D, Saric J, Fabre JM, Maurel P, et al. Interleukin 1β inhibits CAR-induced expression of hepatic genes involved in drug and bilirubin clearance. Hepatology. 2004;40:951–960. doi: 10.1002/hep.20387. [DOI] [PubMed] [Google Scholar]
- Assoian RK, Fleurdelys BE, Stevenson HC, Miller PJ, Madtes DK, Raines EW, et al. Expression and secretion of by β transforming growth factor by activated human macrophages. Proc Natl Acad Sci U S A. 1987;84:6020–6024. doi: 10.1073/pnas.84.17.6020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bach EA, Aguet M, Schreiber RD. The IFNγ receptor: a paradigm for cytokine receptor signaling. Annu Rev Immunol. 1997;15:563–591. doi: 10.1146/annurev.immunol.15.1.563. [DOI] [PubMed] [Google Scholar]
- Bambust I, Van Aelst F, Joosens E, Schallier D, Rezaei Kalantari H, Paulus R, et al. A Belgian registry of interleukin-2 administration for treatment of metastatic renal cell cancer and confrontation with literature data. Acta Clin Belg. 2007;62:223–229. doi: 10.1179/acb.2007.036. [DOI] [PubMed] [Google Scholar]
- Bani MR, Garofalo A, Scanziani E, Giavazzi R. Effect of interleukin-1-β on metastasis formation in different tumor systems. J Natl Cancer Inst. 1991;83:119–123. doi: 10.1093/jnci/83.2.119. [DOI] [PubMed] [Google Scholar]
- Bani S, Gautam M, Sheikh FA, Khan B, Satti NK, Suri KA, et al. Selective Th1 up-regulating activity of Withania somnifera aqueous extract in an experimental system using flow cytometry. J Ethnopharmacol. 2006;107:107–115. doi: 10.1016/j.jep.2006.02.016. [DOI] [PubMed] [Google Scholar]
- Beck J, Rondot P, Catinot L, Falcoff E, Kirchner H, Wietzerbin J. Increased production of interferon gamma and tumor necrosis factor precedes clinical manifestation in multiple sclerosis: do cytokines trigger off exacerbations? Acta Neurol Scand. 1988;78:318–323. doi: 10.1111/j.1600-0404.1988.tb03663.x. [DOI] [PubMed] [Google Scholar]
- Belliard AM, Lacour B, Farinotti R, Leroy C. Effect of tumor necrosis factor-alpha and interferon-gamma on intestinal P-glycoprotein expression, activity, and localization in Caco-2 cells. J Pharm Sci. 2004;93:1524–1536. doi: 10.1002/jps.20072. [DOI] [PubMed] [Google Scholar]
- Belliard AM, Tardivel S, Farinotti R, Lacour B, Leroy C. Effect of hr-IL2 treatment on intestinal P-glycoprotein expression and activity in Caco-2 cells. J Pharm Pharmacol. 2002;54:1103–1109. doi: 10.1211/002235702320266262. [DOI] [PubMed] [Google Scholar]
- Bermel RA, Rudick RA. Interferon-β treatment for multiple sclerosis. Neurotherapeutics. 2007;4:633–646. doi: 10.1016/j.nurt.2007.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beuscher HU, Günther C, Röllinghoff M. IL-1β is secreted by activated murine macrophages as biologically inactive precursor. J Immunol. 1990;144:2179–2183. [PubMed] [Google Scholar]
- Bhatia M, Landolfi C, Basta F, Bovi G, Ramnath RD, de Joannon AC, et al. Treatment with bindarit, an inhibitor of MCP-1 synthesis, protects mice against trinitrobenzene sulfonic acid-induced colitis. Inflamm Res. 2008;57:464–471. doi: 10.1007/s00011-008-7210-y. [DOI] [PubMed] [Google Scholar]
- Biju P, Taveras A, Yu Y, Zheng J, Chao J, Rindgen D, et al. 3,4-Diamino-2,5-thiadiazole-1-oxides as potent CXCR2/CXCR1 antagonists. Bioorg Med Chem Lett. 2008;18:228–231. doi: 10.1016/j.bmcl.2007.10.094. [DOI] [PubMed] [Google Scholar]
- Blick SK, Curran MP. Certolizumab pegol: in Crohn's disease. BioDrugs. 2007;21:195–201. doi: 10.2165/00063030-200721030-00006. [DOI] [PubMed] [Google Scholar]
- de Boer WI. Perspectives for cytokine antagonist therapy in COPD. Drug Discov Today. 2005;10:93–106. doi: 10.1016/S1359-6446(04)03300-8. [DOI] [PubMed] [Google Scholar]
- Brunner T, Heusser CH, Dahinden CA. Human peripheral blood basophils primed by interleukin-3 (IL-3) produce IL-4 in response to immunoglobulin E receptor stimulation. J Exp Med. 1993;177:605–611. doi: 10.1084/jem.177.3.605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carlson TJ, Billings RE. Role of nitric oxide in the cytokine-mediated regulation of cytochrome P-450. Mol Pharmacol. 1996;49:796–801. [PubMed] [Google Scholar]
- Carrillo-Vico A, Guerrero JM, Lardone PJ, Reiter RJ. A review of the multiple actions of melatonin on the immune system. Endocrine. 2005;27:189–200. doi: 10.1385/ENDO:27:2:189. [DOI] [PubMed] [Google Scholar]
- Casilli F, Bianchini A, Gloaguen I, Biordi L, Alesse E, Festuccia C, et al. Inhibition of interleukin-8 (CXCL8/IL-8) responses by repertaxin, a new inhibitor of the chemokine receptors CXCR1 and CXCR2. Biochem Pharmacol. 2005;69:385–394. doi: 10.1016/j.bcp.2004.10.007. [DOI] [PubMed] [Google Scholar]
- Cassatella MA, Gasperini S, Bovolenta C, Calzetti F, Vollebregt M, Scapini P, et al. Interleukin-10 (IL-10) selectively enhances CIS3/SOCS3 mRNA expression in human neutrophils: evidence for an IL-10-induced pathway that is independent of STAT protein activation. Blood. 1999;94:2880–2889. [PubMed] [Google Scholar]
- Chao CC, Hu S, Sheng WS, Tsang M, Peterson PK. Tumor necrosis factor-α mediates the release of bioactive transforming growth factor-β in microglial cell cultures. Clin Immunol Immunoptahol. 1995;77:358–365. doi: 10.1006/clin.1995.1163. [DOI] [PubMed] [Google Scholar]
- Cheng AC, Stephens DP, Currie BJ. Granulocyte-colony stimulating factor (G-CSF) as an adjunct to antibiotics in the treatment of pneumonia in adluts. Cochrane Database Syst Rev. 2007;2:CD004400. doi: 10.1002/14651858.CD004400.pub3. [DOI] [PubMed] [Google Scholar]
- Cherney RJ, Mo R, Meyer DT, Nelson DJ, Lo YC, Yang G, et al. Discovery of disubstituted cyclohexanes as a new class of CC chemokine receptor 2 antagonists. J Med Chem. 2008;51:721–724. doi: 10.1021/jm701488f. [DOI] [PubMed] [Google Scholar]
- Clark R. Pegasys® (peginterferon alfa-2a [40 kDa]) Drugs Cont. 2007;3:65–83. [Google Scholar]
- Clerici M, Shearer GM. A Th1 to Th2 switch is a critical step in the etiology of HIV infection. Immunol Today. 1992;14:107–111. doi: 10.1016/0167-5699(93)90208-3. [DOI] [PubMed] [Google Scholar]
- Clerico M, Contessa G, Durelli L. Interferon-β1a for the treatment of multiple sclerosis. Expert Opin Biol Ther. 2007;7:535–542. doi: 10.1517/14712598.7.4.535. [DOI] [PubMed] [Google Scholar]
- Cloëz-Tayarani I, Changeux JP. Nicotine and serotonin in immune regulation and inflammatory processes: a perspective. J Leukoc Biol. 2007;81:599–606. doi: 10.1189/jlb.0906544. [DOI] [PubMed] [Google Scholar]
- Condie RC, Herring A, Koh WS, Lee M, Kaminski NE. Cannabinoid inhibition of adenylate-mediated signal transduction and IL-2 expression in the murine T-cell line, EL4.IL-2. J Biol Chem. 1996;271:13175–13183. doi: 10.1074/jbc.271.22.13175. [DOI] [PubMed] [Google Scholar]
- Cooksley WG, Piratvisuth T, Lee SD, Mahachai V, Chao YC, Tanwandee T, et al. Peginterferon alpha-2a (40 kDa): an advance in the treatment of hepatitis B e antigen-positive chronic hepatitis B. J Viral Hepat. 2003;10:298–305. doi: 10.1046/j.1365-2893.2003.00450.x. [DOI] [PubMed] [Google Scholar]
- Corcos M, Guilbaud O, Hjalmarsson L, Chambry J, Jeammet P. Cytokines and depression: an analogic approach. Biomed Pharmacother. 2002;56:105–110. doi: 10.1016/s0753-3322(01)00156-1. [DOI] [PubMed] [Google Scholar]
- Cottone M, Orlando A, Modesto I. Postoperative maintenance therapy for inflammatory bowel disease. Curr Opin Gastroenterol. 2006;22:377–381. doi: 10.1097/01.mog.0000231811.95525.7c. [DOI] [PubMed] [Google Scholar]
- Cousins DJ, Staynov DZ, Lee TH. Regulation of interleukin-5 and granulocyte-macrophage colony-stimulating factor expression. Am J Respir Crit Care Med. 1994;150:S50–S53. doi: 10.1164/ajrccm/150.5_Pt_2.S50. [DOI] [PubMed] [Google Scholar]
- Cowan MM. Plant products as antimicrobial agents. Clin Microbiol Rev. 1999;12:564–582. doi: 10.1128/cmr.12.4.564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crawford JH, Yang S, Zhou M, Simms HH, Wang P. Down-regulation of hepatic CYP1A2 plays an important role in inflammatory responses in sepsis. Crit Care Med. 2004;32:502–508. doi: 10.1097/01.CCM.0000109453.57709.E2. [DOI] [PubMed] [Google Scholar]
- Cross ML, Ganner A, Teilab D, Fray LM. Patterns of cytokine induction by gram-positive and gra-negative probiotic bacteria. FEMS Immunol Med Microbiol. 2004;42:173–180. doi: 10.1016/j.femsim.2004.04.001. [DOI] [PubMed] [Google Scholar]
- Dalpke A, Zimmermann S, Heeg K. CpG DNA in the prevention and treatment of infections. BioDrugs. 2002;16:419–431. doi: 10.2165/00063030-200216060-00003. [DOI] [PubMed] [Google Scholar]
- Damaskos D, Kolios G. Probiotics and prebiotics in inflammatory bowel disease: microflora ‘on the scope. Br J Clin Pharmacol. 2008;65:453–467. doi: 10.1111/j.1365-2125.2008.03096.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Danese S, Semeraro S, Armuzzi A, Papa A, Gasbarrini A. Biological therapies for inflammatory bowel disease: research drives clinics. Mini Rev Med Chem. 2006;6:771–784. doi: 10.2174/138955706777698624. [DOI] [PubMed] [Google Scholar]
- Defilippi P, Poupart P, Tavernier J, Fiers W, Content J. Induction and regulation of mRNA encoding 26-kDa protein in human cell lines treated with recombinant human tumor necrosis factor. Proc Natl Acad Sci U S A. 1987;84:4557–4561. doi: 10.1073/pnas.84.13.4557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Delaporte E, Renton KW. Cytochrome P4501A1 and cytochrome P4501A2 are downregulated at both transcriptional and post-trenascriptional levels by conditions resulting in interferon-alpha/beta induction. Life Sci. 1997;60:787–796. doi: 10.1016/s0024-3205(97)00006-4. [DOI] [PubMed] [Google Scholar]
- Denton CP, Merkel PA, Furst DE, Khanna D, Emery P, Hsu VM, et al. Recombinant human anti-transforming growth factor beta1 antibody therapy in systemic sclerosis: a multicenter, randomized, placebo-controlled phase I/II trial of CAT-192. Arthritis Rheum. 2007;56:323–333. doi: 10.1002/art.22289. [DOI] [PubMed] [Google Scholar]
- Descotes J. Immunomodulating agents and hepatic drug-metabolizing enzymes. Drug Metab Rev. 1985;16:175–184. doi: 10.3109/03602538508991434. [DOI] [PubMed] [Google Scholar]
- Devalaraja MN, Richmond A. Multiple chemotactic factors: fine control or redundancy? TIPS. 1999;20:151–156. doi: 10.1016/s0165-6147(99)01342-5. [DOI] [PubMed] [Google Scholar]
- Di Stefano A, Paulesu L. Inhibitory effect of melatonin on production of IFN gamma or TNF alpha in peripheral blood mononuclear cells of some blood donors. J Pineal Res. 1994;17:164–169. doi: 10.1111/j.1600-079x.1994.tb00128.x. [DOI] [PubMed] [Google Scholar]
- Dinarello CA. Blocking IL-1 in systemic inflammation. J Exp Med. 2005;201:1355–1359. doi: 10.1084/jem.20050640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diwanay S, Chitre D, Patwardhan B. Immunoprotection by botanical drugs in cancer chemotherapy. J Ethnopharmacol. 2004;90:49–55. doi: 10.1016/j.jep.2003.09.023. [DOI] [PubMed] [Google Scholar]
- Dixit SG, Zingarelli B, Buckley DJ, Buckley AR, Pauletti GM. Nitric oxide mediates increased P-glycoprotein activity in interferon-γ-stimulated human intestinal cells. Am J Physiol Gastrointest Liver Physiol. 2005;288:G533–G540. doi: 10.1152/ajpgi.00248.2004. [DOI] [PubMed] [Google Scholar]
- Dogusan Z, Hooghe-Peters EL, Berus D, Velkeniers B, Hooghe R. Expression of SOCS genes in normal and leukemic human leukocytes stimulated by prolactin, growth hormone and cytokines. J Neuroimmunol. 2000;109:34–39. doi: 10.1016/s0165-5728(00)00300-3. [DOI] [PubMed] [Google Scholar]
- Dutcher J. Current status of interleukin-2 therapy for metastatic renal cell carcinoma and metastatic melanoma. Oncology (Williston Park) 2002;16(Suppl.)(13):4–10. [PubMed] [Google Scholar]
- Ebnet K, Vestweber D. Molecular mechanisms that control leukocyte extravasation: the selectins and the chemokines. Histochem Cell Biol. 1999;112:1–23. doi: 10.1007/s004180050387. [DOI] [PubMed] [Google Scholar]
- Eisle B, von Henneicke H-H. Soluferon®– a 2nd generation interferon-β with increased bioavailability (Abstract) J Interferon Cytokine Res. 2007;27:703. [Google Scholar]
- El-Obeid A, Al-Harbi S, Al-Jomah N, Hassib A. Herbal melanin modulates tumor necrosis factor alpha (TNF-alpha), interleukin 6 (IL-6) and vascular endothelial growth factor (VEGF) production. Phytomedicine. 2006;13:324–333. doi: 10.1016/j.phymed.2005.03.007. [DOI] [PubMed] [Google Scholar]
- Elenkov IJ. Glucocorticoids and the Th1/Th2 balance. Ann N Y Acad Sci. 2004;1024:138–146. doi: 10.1196/annals.1321.010. [DOI] [PubMed] [Google Scholar]
- Elkahwaji J, Robin MA, Berson A, Tinel M, Lettéron P, Labbe G, et al. Decrease in hepatic cytochrome P450 after interleukin-2 immunotherapy. Biochem Pharmacol. 1999;57:951–954. doi: 10.1016/s0006-2952(98)00372-4. [DOI] [PubMed] [Google Scholar]
- Fan P-T, Leong K-H. The use of biological agents in the treatment of rheumatoid arthritis. Ann Acad Med Singapore. 2007;36:128–134. [PubMed] [Google Scholar]
- Farquhar D, Loo TL, Gutterman JU, Hersh EM, Luna MA. Inhibition of drug-metabolizing enzymes in the rat after Bacillus Calmette-Guérin treatment. Biochem Pharmacol. 1976;25:1529–1535. doi: 10.1016/0006-2952(76)90072-1. [DOI] [PubMed] [Google Scholar]
- Fernandez C, Buyse M, German-Fattal M, Gimenez F. Influence of the pro-inflammatory cytokines on P-glycoprotein expression and functionality. J Pharm Pharmaceut Sci. 2004;7:359–371. [PubMed] [Google Scholar]
- Ferretti S, Bonneau O, Dubois GR, Jones CE, Trifilieff A. IL-17, produced by lymphocytes and neutrophils, is necessary for lipopolysaccharide-induced airway neutrophilia: IL-15 as a possible trigger. J Immunol. 2003;170:2106–2112. doi: 10.4049/jimmunol.170.4.2106. [DOI] [PubMed] [Google Scholar]
- Flodström-Tullberg M, Yadav D, Hägerkvist R, Tsai D, Secrest P, Stotland A, et al. Target cell expression of suppressor of cytokine signaling-1 prevents diabetes in the NOD mouse. Diabetes. 2003;52:2696–2700. doi: 10.2337/diabetes.52.11.2696. [DOI] [PubMed] [Google Scholar]
- Fujii H. Mechanisms of signal transduction from receptors of type I and type II cytokines. J Immunotoxicol. 2007;4:69–76. doi: 10.1080/15476910601154779. [DOI] [PubMed] [Google Scholar]
- Gadient RA, Otten UH. Interleukin-6 (IL-6) – a molecule with both beneficial and destructive potentials. Progr Neurobiol. 1997;52:379–390. doi: 10.1016/s0301-0082(97)00021-x. [DOI] [PubMed] [Google Scholar]
- Galiegue S, Mary S, Marchand J, Dussossoy D, Carriere D, Carayon P, et al. Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations. Eur J Biochem. 1995;232:54–61. doi: 10.1111/j.1432-1033.1995.tb20780.x. [DOI] [PubMed] [Google Scholar]
- Gallagher DC, Bhatt RS, Parikh SM, Patel P, Seery V, McDermott DF, et al. Angiopoietin 2 is a potential mediator of high-dose interleukin 2-induced vascular leak. Clin Cancer Res. 2007;13:2115–2120. doi: 10.1158/1078-0432.CCR-06-2509. [DOI] [PubMed] [Google Scholar]
- Ganz T, Nemeth E. Regulation of iron acquisition and iron distribution in mammals. Biochim Biophys Acta. 2006;1763:690–699. doi: 10.1016/j.bbamcr.2006.03.014. [DOI] [PubMed] [Google Scholar]
- Gardner B, Zu LX, Sharma S, Liu Q, Makriyannis A, Tashkin DP, et al. Autocrine and paracrine regulation of lymphocyte CB2 receptor expression by TGF-β. Biochem Biophys Res Commun. 2002;290:91–96. doi: 10.1006/bbrc.2001.6179. [DOI] [PubMed] [Google Scholar]
- Gaspari AA. Mechanism of action and other potential roles of an immune response modifier. Cutis. 2007;79(Suppl.)(4):36–45. [PubMed] [Google Scholar]
- Gazzaniga S, Bravo AI, Guglielmotti A, van Rooijen N, Maschi F, Vecchi A, et al. Targeting tumor-associated macrophages and inhibition of MCP-1 reduce angiogenesis and tumor growth in a human melanoma xenograft. J Invest Dermatol. 2007;127:2031–2041. doi: 10.1038/sj.jid.5700827. [DOI] [PubMed] [Google Scholar]
- Genovese MC. Biologic therapic in clinical development for the treatment of rheumatoid arthritis. J Clin Rheumatol. 2005;11(Suppl.)(3):S45–S54. doi: 10.1097/01.rhu.0000166625.65114.5f. [DOI] [PubMed] [Google Scholar]
- Gerli R, Bistoni O, Russano A, Fiorucci S, Borgato L, Cesarotti ME, et al. In vivo activated T cells in rheumatoid synovitis. Analysis of Th1- and Th2-type cytokine production at clonal level in different stages of disease. Clin Exp Immunol. 2002;129:549–555. doi: 10.1046/j.1365-2249.2002.01913.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gharaee-Kermani M, Phan SH. Role of cytokines and cytokine therapy in wound healing and fibrotic diseases. Curr Pharm Des. 2001;7:1083–1103. doi: 10.2174/1381612013397573. [DOI] [PubMed] [Google Scholar]
- Ghosh MC, Mondal AC, Basu S, Banerjee S, Majumder J, Bhattacharya D, et al. Dopamine inhibits cytokine release and expression of tyrosine kinases, Lck and Fyn in activated T cells. Int Immunopharmacol. 2003;3:1019–1026. doi: 10.1016/S1567-5769(03)00100-0. [DOI] [PubMed] [Google Scholar]
- Giampietri A, Puccetti P, Contessa AR. Depression of hepatic biotransformations by chemical immunoadjuvants. J Immunopharmacol. 1981;3:251–264. doi: 10.3109/08923978109026430. [DOI] [PubMed] [Google Scholar]
- Gonzales AM, Orlando RA. Curcumin and resveratrol inhibit nuclear factor-kappaB-mediated cytokine expression in adipocytes. Nutr Metab. 2008;12:17–29. doi: 10.1186/1743-7075-5-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorelik L, Prokhorova A, Mokyr MB. Low-dose melpharan-induced shift in the production of a Th2-type cytokine to a Th1-type cytokine in mice bearing a large MOPC-315 tumor. Cancer Immunol Immunother. 1994;39:117–126. [PubMed] [Google Scholar]
- Gorski JC, Hall SD, Becker P, Affrime MB, Cutler DL, Haehner-Daniels B. In vivo effects of interleukin-10 on human cytochrome P450 activity. Clin Pharmacol Ther. 2000;67:32–43. doi: 10.1067/mcp.2000.103860. [DOI] [PubMed] [Google Scholar]
- Gosselin J, Tomolu A, Gallo RC, Flamand L. Interleukin-15 as an activator of natural killer cell-mediated antiviral response. Blood. 1999;94:4210–4219. [PubMed] [Google Scholar]
- Grande F, Garofalo A, Neamati N. Small molecules anti-HIV therapeutics targeting CXCR4. Curr Pharm Des. 2008;14:385–404. doi: 10.2174/138161208783497714. [DOI] [PubMed] [Google Scholar]
- Graziosi C, Pantaleo G, Gantt KR, Fortin JP, Demarest JF, Cohen OJ, et al. Lack of evidence for the dichotomy of Th1 and Th2 predominance in HIV-infected individuals. Science. 1994;265:248–252. doi: 10.1126/science.8023143. [DOI] [PubMed] [Google Scholar]
- Gray PW, Aggarwal BB, Benton CV, Bringman TS, Henzel WJ, Jarrett JA, et al. Cloning and expression of cDNA for human lymphotoxin, a lymphokine with tumour necrosis activity. Nature. 1984;312:721–724. doi: 10.1038/312721a0. [DOI] [PubMed] [Google Scholar]
- Gröne A. Keratinocytes and cytokines. Vet Immunol Immunopathol. 2002;88:1–12. doi: 10.1016/s0165-2427(02)00136-8. [DOI] [PubMed] [Google Scholar]
- Guarner F, Schaafsma GJ. Probiotics. Int J Food Microbiol. 1998;39:237–238. doi: 10.1016/s0168-1605(97)00136-0. [DOI] [PubMed] [Google Scholar]
- Gullo VP, McAlpine J, Lam KS, Baker D, Petersen F. Drug discovery from natural products. J Ind Microbiol Biotechnol. 2006;33:523–531. doi: 10.1007/s10295-006-0107-2. [DOI] [PubMed] [Google Scholar]
- Gupta AK, Cherman AM, Tyring SK. Viral and nonviral uses of imiquimod: a review. J Cutan Med Surg. 2004;8:338–352. doi: 10.1007/s10227-005-0023-5. [DOI] [PubMed] [Google Scholar]
- Haas CS, Amin MA, Allen BB, Ruth JH, Haines GK, III, Woods JM, Koch AE. Inhibition of angiogenesis by interleukin-4 gene therapy in rat adjuvant-induced arthritis. Arthritis Rheum. 2006;54:2402–2414. doi: 10.1002/art.22034. [DOI] [PubMed] [Google Scholar]
- Haas CS, Amin MA, Ruth JH, Allen BL, Ahmed S, Pakozdi A, et al. In vivo inhibition of angiogenesis by interleukin-13 gene therapy in a rat model of rheumatoid arthritis. Arthritis Rheum. 2007;56:2535–2548. doi: 10.1002/art.22823. [DOI] [PubMed] [Google Scholar]
- Hakonarson H, Maskeri N, Carter C, Grunstein MM. Regulation of TH1- and TH2-type cytokine expression and action in atopic asthmatic sensitized airway smooth muscle. J Clin Invest. 1999;103:1077–1087. doi: 10.1172/JCI5809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hameg A, Gouarin C, Gombert JM, Hong SM, VanKaer L, Bach JF, et al. IL−7 up-regulates IL-4 production by splenic NK1.1+ and NK1.1- MHC class I-like/CD1-dependent CD4+ T cells. J Immunol. 1999;162:7067–7074. [PubMed] [Google Scholar]
- Hao J-S, Shan B-E. Immune enhancement and anti-tumour activity of IL-23. Cancer Immunol Immunother. 2006;55:1426–1431. doi: 10.1007/s00262-006-0171-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hartmann G, Kim H, Piquette-Miller M. Regulation of the hepatic multidrug resistance gene expression by endotoxin and inflammatory cytokines in mice. Int Immunopharmac. 2001;1:189–199. doi: 10.1016/s0162-3109(00)00271-x. [DOI] [PubMed] [Google Scholar]
- Haskó G, Szabó C. Regulation of cytokine and chemokine production by transmitters and co-transmitters of the autonomic nervous system. Biochem Pharmacol. 1998;56:1079–1087. doi: 10.1016/s0006-2952(98)00153-1. [DOI] [PubMed] [Google Scholar]
- Haskó G, Pacher P, Deitch EA, Vizi ES. Shaping of monocyte and macrophage function by adenosine. Pharmacol Ther. 2007;113:264–275. doi: 10.1016/j.pharmthera.2006.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He B, You L, Uematsu K, Zang K, Xu Z, Lee AY, et al. SOCS-3 is frequently silenced by hypermethylation and suppresses cell growth in human lung cancer. Proc Natl Acad Sci U S A. 2003;100:14133–14138. doi: 10.1073/pnas.2232790100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heim MH. The Jak-STAT pathway: cytokine signalling from the receptor to the nucleus. J Recept Signal Transduct Res. 1999;19:75–120. doi: 10.3109/10799899909036638. [DOI] [PubMed] [Google Scholar]
- Hendeles L, Asmus M, Chesrown S. Evaluation of cytokine modulators for asthma. Paediatr Respir Rev. 2004;5(Suppl. A):S107–S112. doi: 10.1016/s1526-0542(04)90020-6. [DOI] [PubMed] [Google Scholar]
- Herath KB, Jayasuriya H, Ondeyka JG, Polishook JD, Bills GF, Dombrowski AW, et al. Isolation and structures of novel fungal metabolites as chemokine receptor (CCR2) antagonists. J Antibiot (Tokyo) 2005;58:686–694. doi: 10.1038/ja.2005.94. [DOI] [PubMed] [Google Scholar]
- Higa S, Hirano T, Kotani M, Matsumoto M, Fujita A, Suemura M, et al. Fisetin, a flavonol, inhibits TH2-type cytokine production by activated human basophils. J Allergy Clin Immunol. 2003;111:1299–1306. doi: 10.1067/mai.2003.1456. [DOI] [PubMed] [Google Scholar]
- Hirano T, Higa S, Arimitsu J, Naka T, Shima Y, Ohshima S, et al. Flavonoids such as luteolin, fisetin and apigenin are inhibitors of interleukin-4 and interleukin-13 production by activated human basophils. Int Arch Allergy Immunol. 2004;134:135–140. doi: 10.1159/000078498. [DOI] [PubMed] [Google Scholar]
- Hoesl CE, Altwein JE. The probiotic approach: an alternative treatment option in urology. Eur Urol. 2005;47:288–296. doi: 10.1016/j.eururo.2004.09.011. [DOI] [PubMed] [Google Scholar]
- Holgate ST, Polosa R. Treatment strategies for allergy and athma. Nature Rev. 2008;8:218–230. doi: 10.1038/nri2262. [DOI] [PubMed] [Google Scholar]
- Hommes DW, Mikhajlova TL, Stoinov S, Stimac D, Vucelic B, Lonovics J, et al. Fontolizumab, a humanised anti-interferon gamma antibody, demonstrates safety and clinical activity in patients with moderate to severe Crohn's disease. Gut. 2006;55:1131–1137. doi: 10.1136/gut.2005.079392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hong J-H, Yu ES, Han A-R, Nam J-W, Seo E-K, Hwang ES. Isoleutherin and eleutherinol, naturally occuring selective modulators of Th cell-mediated immune responses. Biochem Biophys Res Commun. 2008;371:278–282. doi: 10.1016/j.bbrc.2008.04.060. [DOI] [PubMed] [Google Scholar]
- Horrigan LA, Kelly JP, Connor TJ. Immunomodulatory effects of caffeine: friend or foe? Pharmacol Ther. 2006;111:877–892. doi: 10.1016/j.pharmthera.2006.02.002. [DOI] [PubMed] [Google Scholar]
- Houshmand P, Zlotnik A. Therapeutic applications in the chemokine superfamily. Curr Opin Chem Biol. 2003;7:457–460. doi: 10.1016/s1367-5931(03)00086-3. [DOI] [PubMed] [Google Scholar]
- Howard OM, Ben-Baruch A, Oppenheim JJ. Chemokines: progress toward identifying molecular targets for therapeutic agents. Trends Biotechnol. 1996;14:46–51. doi: 10.1016/0167-7799(96)80920-6. [DOI] [PubMed] [Google Scholar]
- Huang S, Mills L, Mian B, Tellez C, McCarty M, Yang XD, et al. Fully humanized neutralizing antibodies to interleukin-8 (ABX-IL8) inhibit angiogenesis, tumor growth, and metastasis of human melanoma. Am J Pathol. 2002;161:125–134. doi: 10.1016/S0002-9440(10)64164-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Idzko M, la Sala A, Ferrari D, Panther E, Herouy Y, Dichmann S, et al. Expression and function of histamine receptors in human monocyte-derived dendritic cells. J Allergy Clin Immunol. 2002;109:839–846. doi: 10.1067/mai.2002.124044. [DOI] [PubMed] [Google Scholar]
- Imada K, Leonard WJ. The Jak-STAT pathway. Mol Immunol. 2000;37:1–11. doi: 10.1016/s0161-5890(00)00018-3. [DOI] [PubMed] [Google Scholar]
- Imanaka K, Tamura S, Fukui K, Ito N, Kiso S, Imai Y, et al. Enhanced expression of suppressor of cytokine signalling-1 in the liver of chronic hepatitis C: possible involvement in resistance to interferon therapy. J Viral Hepat. 2005;12:130–138. doi: 10.1111/j.1365-2893.2005.00576.x. [DOI] [PubMed] [Google Scholar]
- Ioannou Y, Isenberg DA. Current evidence for the induction of autoimmune rheumatic manifestations by cytokine therapy. Arthritis Rheum. 2000;43:1431–1442. doi: 10.1002/1529-0131(200007)43:7<1431::AID-ANR3>3.0.CO;2-E. [DOI] [PubMed] [Google Scholar]
- Isaacs A, Lindenmann J. Virus interference. I. The interferon. Proc R Soc Lond B Biol Sci. 1957;147:258–264. [PubMed] [Google Scholar]
- Islam M, Frye RF, Richards TJ, Sbeitan I, Donnelly SS, Glue P, et al. Differential effect of IFNα–2b on the cytochrome P450 enzyme system: a potential basis of IFN toxicity and its modulation by other drugs. Clin Cancer Res. 2002;8:2480–2487. [PubMed] [Google Scholar]
- Jan TR, Farraj AK, Harkema JR, Kaminski NE. Attenuation of the ovalbumin-induced allergic airway response by cannabinoid treatment in A/J mice. Toxicol Appl Pharmacol. 2003;188:24–35. doi: 10.1016/s0041-008x(03)00010-3. [DOI] [PubMed] [Google Scholar]
- Jan TR, Su ST, Wu HY, Liao MH. Suppressive effects of cannabidiol on antigen-specific antibody production and functional activity of splenocytes in ovalbumin-sensitized BALB/c mice. Int Immunopharmacol. 2007;7:773–780. doi: 10.1016/j.intimp.2007.01.015. [DOI] [PubMed] [Google Scholar]
- Jover R, Bort R, Gómez-Lechón MJ, Castell JV. Down-regulation of human CYP3A4 by the inflammatory signal interleukin-6: molecular mechanism and transcription factors involved. FASEB J. 2002;16:1799–1801. doi: 10.1096/fj.02-0195fje. [DOI] [PubMed] [Google Scholar]
- Kaiko GE, Horvat JC, Beagley KW, Hansbro PM. Immunological decision-making: how does the immune system decode to mount a helper T-cell response? Immunology. 2008;122:326–338. doi: 10.1111/j.1365-2567.2007.02719.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalliomäki M, Salminen S, Poussa T, Arvilommi H, Isolauri E. Probiotics and prevention of atopic disease: 4-year follow-up of a randomised placebo-controlled trial. Lancet. 2003;361:1869–1871. doi: 10.1016/S0140-6736(03)13490-3. [DOI] [PubMed] [Google Scholar]
- Kalsotra A, Anakk S, Brommer CL, Kikuta Y, Morgan ET, Strobel HW. Catalytic characterization and cytokine mediated regulation of cytochrome P450 4Fs in rat hepatocytes. Arch Biochem Biophys. 2007;461:104–112. doi: 10.1016/j.abb.2007.02.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kandimalla ER, Bhagat L, Li Y, Yu D, Wang D, Cong YP, et al. Immunomodulatory oligonucleotides containing a cytosine-phosphate-2′-deoxy-7-deazaguanosine motif as potent toll-like receptor 9 agonists. Proc Natl Acad Sci U S A. 2005;102:6925–6930. doi: 10.1073/pnas.0501729102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Katiyar SK. UV-induced immune suppression and photocarcinogenesis: chemoprevention by dietary botanical agents. Cancer Lett. 2007;255:1–11. doi: 10.1016/j.canlet.2007.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Katona S, Kaminski E, Sanders H, Zajicek J. Cannabinoid influence on cytokine profile in multiple sclerosis. Clin Exp Immunol. 2005;140:580–585. doi: 10.1111/j.1365-2249.2005.02803.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelly-Welch AE, Hanson EM, Boothby MR, Keegan AD. Interleukin-4 and interleukin-13 signaling connections maps. Science. 2003;300:1527–1528. doi: 10.1126/science.1085458. [DOI] [PubMed] [Google Scholar]
- Kero J, Gissler M, Hemminki E, Isolauri E. Could TH1 and TH2 diseases coexist? Evaluation of asthma incidence in children with coeliac disease, type 1 diabetes, or rheumatoid arthritis: a register study. J Allergy Clin Immunol. 2001;108:781–783. doi: 10.1067/mai.2001.119557. [DOI] [PubMed] [Google Scholar]
- Khan IA. Issues related to botanicals. Life Sci. 2006;78:2033–2038. doi: 10.1016/j.lfs.2005.12.019. [DOI] [PubMed] [Google Scholar]
- Kidd P. Th1/Th2 balance: the hypothesis, its limitations, and implications for health and disease. Altern Med Rev. 2003;8:223–246. [PubMed] [Google Scholar]
- Kihara T, Umesue I, Soeda S, Toda A, Ono N, Shigematsu H, et al. Hepatic heme metabolism in rats with fever induced by interleukin 1beta. Res Commun Mol Pathol Pharmacol. 1999;104:115–126. [PubMed] [Google Scholar]
- Kile BT, Alexander WS. The suppressors of cytokine signalling (SOCS) Cell Mol Life Sci. 2001;58:1627–1635. doi: 10.1007/PL00000801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim J, Modlin RL, Moy RL, Dubinett SM, McHugh T, Nickoloff BJ, et al. IL-10 production in cutaneous basal and squamous cell carcinomas. A mechanism for evading the local T cell immune response. J Immunol. 1995;155:2240–2247. [PubMed] [Google Scholar]
- Kim YS, Jung DH, Kim NH, Lee YM, Kim JS. Effect of magnolol on TGF-beta1 and fibronectin expression in human retinal pigment epithelial cells under diabetic conditions. Eur J Pharmacol. 2007;562:12–19. doi: 10.1016/j.ejphar.2007.01.048. [DOI] [PubMed] [Google Scholar]
- Kleemann R, Zadelaar S, Kooistra T. Cytokines and atherosclerosis: a comprehensive review of studies in mice. Cardiovasc Res. 2008;79:360–376. doi: 10.1093/cvr/cvn120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klein TW, Newton C, Larsen K, Lu L, Perkins I, Nong L, et al. The cannabinoid system and immune modulation. J Leukoc Biol. 2003;74:486–496. doi: 10.1189/jlb.0303101. [DOI] [PubMed] [Google Scholar]
- Kmonícková E, Melkusová P, Harmatha J, Vokác K, Farghali H, Zídek Z. Inhibitor of sarco-endoplasmic reticulum Ca2+-ATPase thapsigargin stimulates production of nitric oxide and secretion of interferon-gamma. Eur J Pharm. 2008;588:85–92. doi: 10.1016/j.ejphar.2008.03.037. [DOI] [PubMed] [Google Scholar]
- Krebs DL, Hilton DJ. SOCS proteins: negative regulators of cytokine signaling. Stem Cells. 2001;19:378–387. doi: 10.1634/stemcells.19-5-378. [DOI] [PubMed] [Google Scholar]
- Krieg AM. Therapeutic potential of Toll-like receptor 9 activation. Nat Rev Drug Disc. 2006;5:471–484. doi: 10.1038/nrd2059. [DOI] [PubMed] [Google Scholar]
- Kroemer G, Wick G. The role of interleukin 2 in autoimmunity. Immunol Today 1989. 1989;10:246–251. doi: 10.1016/0167-5699(89)90262-4. [DOI] [PubMed] [Google Scholar]
- Krouwels FH, Hol BEA, Lutter R, Bruinier B, Bast A, Jansen HM, et al. Histamine affects interleukin-4, interleukin-5, and interferon-γ production by human T cell clones from the airways and blood. Am J Respir Cell Mol Biol. 1998;18:721–730. doi: 10.1165/ajrcmb.18.5.2909. [DOI] [PubMed] [Google Scholar]
- Kühlwein E, Irwin M. Melatonin modulation of lymphocyte proliferation and Th1/Th2 cytokine expression. J Neuroimmunol. 2001;117:51–57. doi: 10.1016/s0165-5728(01)00325-3. [DOI] [PubMed] [Google Scholar]
- Kuhn CF, Bazin M, Philippe L, Zhang J, Tylaska L, Miret J, et al. Bipiperidinyl carboxylic acid amides as potent, selective, and functionally active CCR4 antagonists. Chem Biol Drug Des. 2007;70:268–272. doi: 10.1111/j.1747-0285.2007.00551.x. [DOI] [PubMed] [Google Scholar]
- La Cava A, Sarvetnick N. The role of cytokines in autoimmunity. Curr Dir Autoimmun. 1999;1:56–71. doi: 10.1159/000060495. [DOI] [PubMed] [Google Scholar]
- Lavon I, Sheinin T, Meilin S, Biton E, Weksler A, Efroni G, et al. A novel synthetic cannabinoid derivative inhibits inflammatory liver damage via negative cytokine regulation. Mol Pharmacol. 2003;64:1334–1341. doi: 10.1124/mol.64.6.1334. [DOI] [PubMed] [Google Scholar]
- Leckie MJ, ten Brinke A, Khan J, Diamant Z, O'Connor BJ, Walls CM, et al. Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet. 2000;356:2144–2148. doi: 10.1016/s0140-6736(00)03496-6. [DOI] [PubMed] [Google Scholar]
- Lee J, Chuang T-H, Redecke V, She L, Pitha PM, Carson DA, et al. Molecular basis for the stimulatory activity of guanine nucleoside analogs: Activation of Toll-like receptor 7. Proc Natl Acad Sci U S A. 2003;100:6646–6651. doi: 10.1073/pnas.0631696100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee JY, Kim JY, Lee YG, Shin WC, Chun T, Rhee MH, et al. Hydroquinine, a reactive metabolite of benzene, reduces macrophage-mediated immune responses. Mol Cells. 2007;23:198–206. [PubMed] [Google Scholar]
- Leeson GA, Biedenbach SA, Chan KY, Gibson JP, Wright GJ. Decrease in the activity of the drug-metabolizing enzymes of rat liver following the administration of tilorone hydrochloride. Drug Metab Dispos. 1976;4:232–238. [PubMed] [Google Scholar]
- Lejeune D, Dumoutier L, Constantinescu S, Kruijer W, Schuringa JJ, Renauld JC. Interleukin-22 (IL-22) activates the JAK/STAT, ERK, JNK, and p38 MAP kinase pathways in a rat hepatoma cell line. Pathways that are shared with and distinct from IL-10. J Biol Chem. 2002;277:33676–33682. doi: 10.1074/jbc.M204204200. [DOI] [PubMed] [Google Scholar]
- Levine SJ. Mechanisms of soluble cytokine receptor generation. J Immunol. 2004;173:5343–5348. doi: 10.4049/jimmunol.173.9.5343. [DOI] [PubMed] [Google Scholar]
- Levings MK, Sangregorio R, Galbiati F, Squadrone S, de Waal Malefyt R, Roncarolo M-G. IFN-α and IL-10 induce the differentiation of human type 1 T regulatory cells. J Immunol. 2001;166:5530–5539. doi: 10.4049/jimmunol.166.9.5530. [DOI] [PubMed] [Google Scholar]
- Lewis DB. Allergy immunotherapy and inhibition of Th2 immune responses: a sufficient strategy? Curr Opin Immunol. 2002;14:644–651. doi: 10.1016/s0952-7915(02)00388-6. [DOI] [PubMed] [Google Scholar]
- Li H, Sim TC, Alam R. IL-13 released by and localized in human basophils. J Immunol. 1996;156:4833–4838. [PubMed] [Google Scholar]
- Li MO, Wan YY, Sanjabi S, Robertson A-KL, Flavell RA. Transforming growth factor-β regulation of immune responses. Annu Rev Immunol. 2006;24:99–146. doi: 10.1146/annurev.immunol.24.021605.090737. [DOI] [PubMed] [Google Scholar]
- Ligresti A, Moriello AS, Starowicz K, Matias I, Pisanti S, De Petrocellis L, et al. Antitumor activity of plant cannabinoids with emphasis on the effect of cannabidiol on human breast carcinoma. J Pharmacol Exp Ther. 2006;318:1375–1387. doi: 10.1124/jpet.106.105247. [DOI] [PubMed] [Google Scholar]
- Lipsky PE. Interleukin-6 and rheumatic diseases. Arthr Res Ther. 2006;8(Suppl.)(2):S4–S8. doi: 10.1186/ar1918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lit LC, Wong CK, Li EK, Tam LS, Lam CW, Lo YM. Elevated gene expression of Th1/Th2 associated transcription factors is correlated with disease activity in patients with systemic lupus erythematosus. J Rheumatol. 2007;34:89–96. [PubMed] [Google Scholar]
- Liu F, Ng TB, Fung MC. Pineal indoles stimulate the gene expression of immunomodulating cytokines. J Neural Transm. 2001;108:397–405. doi: 10.1007/s007020170061. [DOI] [PubMed] [Google Scholar]
- Liu H, Leung BP. CD4+CD25+ regulatory T cells in health and disease. Clin Exp Pharmacol Physiol. 2006;33:519–524. doi: 10.1111/j.1440-1681.2006.04401.x. [DOI] [PubMed] [Google Scholar]
- Liu KD, Gaffen SL, Goldsmith MA. JAK/STAT signaling by cytokine receptors. Curr Opin Immunol. 1998;10:271–278. doi: 10.1016/s0952-7915(98)80165-9. [DOI] [PubMed] [Google Scholar]
- Loisel-Meyer S, Foley R, Medin JA. Immuno-gene therapy approaches for cancer: from in vitro studies to clinical trials. Front Biosci. 2008;13:3202–3214. doi: 10.2741/2921. [DOI] [PubMed] [Google Scholar]
- Lorentz A, Schwenberg S, Mierke C, Manns MP, Bischoff SC. Human intestinal mast cells produce IL-5 in vitro upon IgE receptor cross-linking and in vivo in the course of intestinal inflammatory disease. Eur J Immunol. 1999;29:1496–1503. doi: 10.1002/(SICI)1521-4141(199905)29:05<1496::AID-IMMU1496>3.0.CO;2-5. [DOI] [PubMed] [Google Scholar]
- Machida K, Mayer BJ. The SH2 domain: versatile signaling module and pharmaceutical target. Biochim Biophys Acta. 2005;1747:1–25. doi: 10.1016/j.bbapap.2004.10.005. [DOI] [PubMed] [Google Scholar]
- Mantovani A. The chemokine system: redundancy for robust outputs. Immunol Today. 1999;20:254–257. doi: 10.1016/s0167-5699(99)01469-3. [DOI] [PubMed] [Google Scholar]
- Marciano BE, Wesley R, De Carlo ES, Anderson VL, Barnhart LA, Darnell D, et al. Long-term interferon-γ therapy for patients with chronic granulomatous disease. Clin Infect Dis. 2004;39:692–699. doi: 10.1086/422993. [DOI] [PubMed] [Google Scholar]
- Markus M, Moisés M, Klaus E, Manuel M. Murine macrophages secrete interferon? upon combined stimulation with interleukin (IL)-12 and IL-18: a novel pathway of autocrine macrophage activation. J Exp Med. 1998;187:2103–2109. doi: 10.1084/jem.187.12.2103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marshall JK. Ilodecakin. Schering-Plough Corp. IDrugs. 1999;2:1045–1058. [PubMed] [Google Scholar]
- Massi P, Vaccani A, Paloralo D. Cannabinoids, immune system and cytokine network. Curr Pharm Des. 2006;12:3135–3146. doi: 10.2174/138161206777947425. [DOI] [PubMed] [Google Scholar]
- McDermott JR, Humphreys NE, Forman SP, Donaldson DD, Grencis RK. Intraepithelial NK cell-derived IL-13 induces intestinal pathology associated with nematode infection. J Immunol. 2005;175:3207–3213. doi: 10.4049/jimmunol.175.5.3207. [DOI] [PubMed] [Google Scholar]
- McInnes IB, Schett G. Cytokines in the pathogenesis of rheumatoid arthritis. Nat Rev Immunol. 2007;7:429–442. doi: 10.1038/nri2094. [DOI] [PubMed] [Google Scholar]
- Meyer AL, Elmadfa I, Herbacek I, Micksche M. Probiotic, as well as conventional yogurt, can enhance the stimulated production of proinflammatory cytokines. J Hum Nutr Diet. 2007;20:590–598. doi: 10.1111/j.1365-277X.2007.00807.x. [DOI] [PubMed] [Google Scholar]
- Middel P, Lippert U, Hummel KM, Bertsch HP, Artuc M, Schweyer S, et al. Expression of lymphotoxin-alpha by keratinocytes: a further mediator for the lichenoid reaction. Pathobiology. 2000;68:291–300. doi: 10.1159/000055940. [DOI] [PubMed] [Google Scholar]
- Miettinen M, Matikainen S, Vuopio-Varkila J, Pirhonen J, Varkila K, Kurimoto M, et al. Lactobacilli and streptococci induce interleukin-12 (IL-12), IL-18, and gamma interferon production in human peripheral blood mononuclear cells. Infect Immun. 1998;66:6058–6062. doi: 10.1128/iai.66.12.6058-6062.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miles BA, Lafuse WP, Zwilling BS. Binding of alpha-adrenergic receptors stimulates the anti-mycrobacterial activity of murine peritoneal macrophages. J Neuroimmunol. 1996;71:19–24. doi: 10.1016/s0165-5728(96)00113-0. [DOI] [PubMed] [Google Scholar]
- Mirolo M, Fabbri M, Sironi M, Vecchi A, Guglielmotti A, Mangano G, et al. Impact of the anti-inflammatory agent bindarit on the chemokinome: selective inhibition of the monocyte chemotactic proteins. Eur Cytokine Netw. 2008;19:119–122. doi: 10.1684/ecn.2008.0133. [DOI] [PubMed] [Google Scholar]
- Miwa M, Kozawa O, Tokuda H, Uematsu T. Mitogen-activated protein (MAP) kinases are involved in interleukin-1 (IL-1)-induced IL-6 synthesis in osteoblasts: modulation not of p38 MAP kinase, but of p42/p44 MAP kinase by IL-1-activated protein kinase C. Endocrinology. 1999;140:5120–5125. doi: 10.1210/endo.140.11.7123. [DOI] [PubMed] [Google Scholar]
- Miyazaki Y, Yokozeki H, Awad S, Igawa K, Minatohara K, Satoh T, et al. Glucocorticoids augment the chemically induced production of gene expression of interleukin-1β through NF-κB and AP-1 activation in murine epidermal cells. J Invest Dermatol. 2000;115:746–752. doi: 10.1046/j.1523-1747.2000.00101.x. [DOI] [PubMed] [Google Scholar]
- Miyazawa K, Shinozaki M, Hara T, Furuya T, Miyazono K. Two major Smad pathways in TGF-β superfamily signalling. Genes Cells. 2002;7:1191–1204. doi: 10.1046/j.1365-2443.2002.00599.x. [DOI] [PubMed] [Google Scholar]
- Miyoshi M, Nadai M, Nitta A, Ueyama J, Shimizu A, Takagi K, et al. Role of tumor necrosis factor-alpha in down-regulation of hepatic cytochrome P450 and P-glycoprotein by endotoxin. Eur J Pharmacol. 2005;507:229–237. doi: 10.1016/j.ejphar.2004.11.035. [DOI] [PubMed] [Google Scholar]
- Mizuno T, Sawada M, Marunouchi T, Suzumura A. Production of interleukin-10 by mouse glial cells in culture. Biochem Biophys Res Commun. 1994;205:1907–1915. doi: 10.1006/bbrc.1994.2893. [DOI] [PubMed] [Google Scholar]
- Mizutani T, Masuda M, Nakai E, Furumiya K, Togawa H, Nakamura Y, et al. Genuine functions of P-glycoprotein (ABCB1) Curr Drug Metab. 2008;9:167–174. doi: 10.2174/138920008783571756. [DOI] [PubMed] [Google Scholar]
- Mohagheghpour N, Waleh N, Garger SJ, Dousman L, Grill LK, Tusé D. Synthetic melanin suppresses production of proinflammatory cytokines. Cell Immunol. 2000;199:25–36. doi: 10.1006/cimm.1999.1599. [DOI] [PubMed] [Google Scholar]
- Möhle R, Salemi P, Moore MA, Rafii S. Expression of interleukin-5 by human bone marrow microvascular endothelial cells: implications for the regulation of eosinophilopoiesis in vivo. Br J Haematol. 1997;99:732–738. doi: 10.1046/j.1365-2141.1997.4713279.x. [DOI] [PubMed] [Google Scholar]
- Molet S, Hamid Q, Davoine F, Nutku E, Taha R, Pagé N, et al. IL-17 is increased in asthmatic airways and induces human bronchial fibroblasts to produce cytokines. J Allergy Clin Immunol. 2001;108:420–438. doi: 10.1067/mai.2001.117929. [DOI] [PubMed] [Google Scholar]
- Moqbel R, Ying S, Barkans J, Newman TM, Kimmitt P, Wakelin M, et al. Identification of messenger RNA for IL-4 in human eosinophils with granule localization and release of the translated product. J Immunol. 1995;155:4939–4947. [PubMed] [Google Scholar]
- Mosmann TR, Coffman RL. Th1 and Th2 cells: different patterns of lymphokine secretion lead to different functional properties. Annu Rev Immunol. 1989;7:145–174. doi: 10.1146/annurev.iy.07.040189.001045. [DOI] [PubMed] [Google Scholar]
- Müller GF, Döhr O, El-Bahay C, Kahl R, Abel J. Effect of transforming growth factor-beta1 on cytochrome P450 expression: inhibition of CYP1 mRNA and protein expression in primary rat hepatocytes. Arch Toxicol. 2000;74:145–152. doi: 10.1007/s002040050667. [DOI] [PubMed] [Google Scholar]
- Muntané-Relat J, Ourlin JC, Domergue J, Maurel P. Differential effects of cytokines on the inducible expression of CYP1A1, CYP1A2, and CYP3A4 in human hepatocytes in primary culture. Hepatology. 1995;22:1143–1153. [PubMed] [Google Scholar]
- Murreille E, Leo O. Revisiting the Th1/Th2 paradigm. Scand J Immunol. 1998;47:1–9. doi: 10.1111/j.1365-3083.1998-47-1.00383.x. [DOI] [PubMed] [Google Scholar]
- Nadin L, Butler AM, Farrell GC, Murray M. Pretranslational down-regulation of cytochromes P450 2C11 and 3A2 in male rat liver by tumor necrosis factor alpha. Gastroenterology. 1995;109:198–205. doi: 10.1016/0016-5085(95)90285-6. [DOI] [PubMed] [Google Scholar]
- Nakamura A, Johns EJ, Imaizumi A, Yanagawa Y, Kohsaka T. Effect of β2-adrenoceptor activation and angiotensin II on tumour necrosis factor and interleukin 6 gene transcription in the rat renal resident macrophage cells. Cytokine. 1999;11:759–765. doi: 10.1006/cyto.1999.0488. [DOI] [PubMed] [Google Scholar]
- Nalbandian G, Kovats S. Understanding sex biases in immunity: effects of estrogen on the differentiation and function of antigen-presenting cells. Immunol Res. 2005;31:91–106. doi: 10.1385/IR:31:2:091. [DOI] [PubMed] [Google Scholar]
- Newton CA, Lu T, Nazian SJ, Perkins I, Friedman H, Klein TW. The THC-induced suppression of Th1 polarization in response to Legionella pneumophila infection is not mediated by increases in corticosterone and PGE2. J Leukoc Biol. 2004;76:854–861. doi: 10.1189/jlb.0304184. [DOI] [PubMed] [Google Scholar]
- Nitta T, Ebato M, Sato K, Okumura K. Expression of tumor necrosis factor-α,, -β and interferon-γ genes within human neuroglial tumor cells and brain specimens. Cytokine. 1994;6:171–180. doi: 10.1016/1043-4666(94)90039-6. [DOI] [PubMed] [Google Scholar]
- Numasaki M, Watanabe M, Suzuki T, Takahashi H, Nakamura A, McAllister F, et al. IL-17 enhances the net angiogenic activity and in vivo growth of human non-small cell lung cancer in SCID mice through promoting CXCR-2-dependent angiogenesis. J Immunol. 2005;175:6177–6189. doi: 10.4049/jimmunol.175.9.6177. [DOI] [PubMed] [Google Scholar]
- O'Garra A, Vieira P. TH1 cells control themselves by producing interleukin-10. Nat Rev Immunol. 2007;7:425–428. doi: 10.1038/nri2097. [DOI] [PubMed] [Google Scholar]
- O'Garra A, Chang R, Hastings R, Go N, Haughton G, Howard M. Ly-1 B (B-1) cells are the main source of B cell-derived interleukin-10. Eur J Immunol. 1992;22:711–717. doi: 10.1002/eji.1830220314. [DOI] [PubMed] [Google Scholar]
- O'Hara AM, O'Regan P, Fanning A, O'Mahony C, Macsharry J, Lyons A, et al. Functional modulation of human intestinal epithelial cell responses by Bifidobacterium infantis and Lactobacillus salivarius. Immunology. 2006;118:202–215. doi: 10.1111/j.1365-2567.2006.02358.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Onuffer JJ, Horuk R. Chemokines, chemokine receptors and small-molecule antagonists: recent developments. TIPS. 2002;23:459–467. doi: 10.1016/s0165-6147(02)02064-3. [DOI] [PubMed] [Google Scholar]
- Openshaw P, Murphy EE, Hosken NA, Maino V, Davis K, Murphy K, et al. Heterogenity of intracellular cytokine synthesis at the single-cell level in polarized T helper 1 and T helper 2 popoulations. J Exp Med. 1995;182:1357–1367. doi: 10.1084/jem.182.5.1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oppenheim JJ, Murphy WJ, Chertov O, Schirrmacher V, Wang JM. Prospects for cytokine and chemokine biotherapy. Clin Cancer Res. 1997;3:2682–2686. [PubMed] [Google Scholar]
- Owen C. Chemokine receptors in airway disease: which receptors to target? Pulm Pharmac Therap. 2001;14:193–202. doi: 10.1006/pupt.2001.0281. [DOI] [PubMed] [Google Scholar]
- Packard KA, Khan MM. Effects of histamine on Th1/Th2 cytokine balance. Int Immunopharmac. 2003;3:909–920. doi: 10.1016/S1567-5769(02)00235-7. [DOI] [PubMed] [Google Scholar]
- Pageaux GP, Ie Bricquir Y, Berthou F, Bressot N, Picot MC, Blanc F, et al. Effects of interferon-alpha on cytochrome P-450 isoforms 1A2 and 3A activities in patients with chronic hepatitis C. Eur J Gastroenterol Hepatol. 1998;10:491–495. doi: 10.1097/00042737-199806000-00010. [DOI] [PubMed] [Google Scholar]
- Pang Y, Norihisa Y, Benjamin D, Kantor RRS, Young HA. Interferon-γ-gene expression in human B-cell lines: induction by interleukin-2, protein kinase C activators, and possible effect of hypomethylation on gene regulation. Blood. 1992;80:724–732. [PubMed] [Google Scholar]
- Panitch HS, Hirsch RL, Haley AS, Johnson KP. Exacerbations of multiple sclerosis in patients treated with gamma interferon. Lancet. 1987;1:893–895. doi: 10.1016/s0140-6736(87)92863-7. [DOI] [PubMed] [Google Scholar]
- Patwardhan B, Gautam M. Botanical immunomodulators: scope and opportunities. Drug Discov Today. 2005;10:495–502. doi: 10.1016/S1359-6446(04)03357-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paul AT, Gohil VM, Bhutani KK. Modulating TNF-α signaling with natural products. Drug Discov Today. 2006;11:725–732. doi: 10.1016/j.drudis.2006.06.002. [DOI] [PubMed] [Google Scholar]
- Paul C, Seiliez I, Thissen JP, LeCam A. Regulation of expression of the rat SOCS-3 gene in hepatocytes by growth hormone, interleukin-6 and glucocorticoids. Eur J Biochem. 2000;267:5849–5857. doi: 10.1046/j.1432-1327.2000.01395.x. [DOI] [PubMed] [Google Scholar]
- Pease JE. Asthma, allergy and chemokines. Curr Drug Targ. 2006;7:3–12. doi: 10.2174/138945006775270204. [DOI] [PubMed] [Google Scholar]
- Peraldi P, Filloux C, Emanuelli B, Hilton DJ, Van Obberghen E. Insulin induces suppressor of cytokine signaling-3 tyrosine phosphorylation through janus-activated kinase. J Biol Chem. 2001;276:24614–24620. doi: 10.1074/jbc.M102209200. [DOI] [PubMed] [Google Scholar]
- Perez GM, Melo M, Keegan AD, Zamorano J. Aspirin and salicylates inhibit the IL-4- and IL-13-induced activation of STAT6. J Immunol. 2002;168:1428–1434. doi: 10.4049/jimmunol.168.3.1428. [DOI] [PubMed] [Google Scholar]
- Perry CM, Jarvis B. Peginterferon-alpha-2a (40 kD): a review of its use in the management of chronic hepatitis C. Drugs. 2001;61:2263–2288. doi: 10.2165/00003495-200161150-00013. [DOI] [PubMed] [Google Scholar]
- Perry CM, Wilde MI. Interferon-alpha-2a: a review of its use in chronic hepatitis C. BioDrugs. 1998;10:65–89. doi: 10.2165/00063030-199810010-00005. [DOI] [PubMed] [Google Scholar]
- Perussia B. Lymphokine-activated killer cells, natural killer cells and cytokines. Curr Opin Immunol. 1991;3:49–55. doi: 10.1016/0952-7915(91)90076-d. [DOI] [PubMed] [Google Scholar]
- Petray PB, Rottenberg ME, Bertot G, Corral RS, Diaz A, Orn A, et al. Effect of anti-γ-interferon and anti-interleukin-4 administration on the resistance of mice against infection with reticulotropic and myotropic strains of Trypanosoma cruzi. Immunol Lett. 1993;35:77–80. doi: 10.1016/0165-2478(93)90151-q. [DOI] [PubMed] [Google Scholar]
- Pinkas J, Teicher BA. TGF-β in cancer and as a therapeutic target. Biochem Pharmacol. 2006;72:523–529. doi: 10.1016/j.bcp.2006.03.004. [DOI] [PubMed] [Google Scholar]
- Prandota J. Important role of proinflammatory cytokines/other endogenous substances in drug-induced hepatotoxicity: depression of drug metabolism during infections/inflammation states, and genetic polymorphisms of drug-metabolizing enzymes/cytokines may markedly contribute to this pathology. Am J Ther. 2005;12:254–261. [PubMed] [Google Scholar]
- Puddu P, Fais S, Luciani F, Gherardi G, Dupuis ML, Romagnoli G, et al. Interferon-gamma up-regulates expression and activity of P-glycoprotein in human peripheral blood monocyte-derived macrophages. Lab Invest. 1999;79:1299–1309. [PubMed] [Google Scholar]
- Puffenbarger RA, Boothe AC, Cabral GA. Cannabinoids inhibit LPS-inducible cytokine mRNA expression in rat microglial cells. Glia. 2000;29:58–69. [PubMed] [Google Scholar]
- Pugh-Humphreys RGP, Thomson AW. Cytokines and their receptors as potential therapeutic targets. In: Thomson AW, editor. The Cytokine Handbook. 3rd. San Diego: Academic Press; 1998. pp. 885–951. [Google Scholar]
- Raduner S, Majewska A, Chen JZ, Xie XQ, Hamon J, Faller B, et al. Alkylamides from Echinacea are a new class of cannabinomimetics. Cannabinoid type 2 receptor-dependent and -independent immunomodulatory effects. J Biol Chem. 2006;281:14192–14206. doi: 10.1074/jbc.M601074200. [DOI] [PubMed] [Google Scholar]
- Raghavendra V, Singh V, Kulkarni SK, Agrewala JN. Melatonin enhances Th2 cell mediated immune responses: lack of sensitivity to reversal by naltrexone or benzodiazepine receptor antagonists. Mol Cell Biochem. 2001;221:57–62. doi: 10.1023/a:1010968611716. [DOI] [PubMed] [Google Scholar]
- Rachmawati H, Reker-Smit C, Lub-de Hooge MN, van Loenen-Weemaes A, Poelstra K, Beljaars L. Chemical modification of interleukin-10 with mannose 6-phosphate groups yields a liver-selective cytokine. Drug Metab Dispos. 2007;35:814–821. doi: 10.1124/dmd.106.013490. [DOI] [PubMed] [Google Scholar]
- Ratcliffe MJ, Walding A, Shelton PA, Flaherty A, Dougall IG. Activation of E-prostanoid4 and E-prostanoid2 receptors inhibits TNF-alpha release from human alveolar macrophages. Eur Respir J. 2007;29:986–994. doi: 10.1183/09031936.00131606. [DOI] [PubMed] [Google Scholar]
- Ricci A, Mariotta S, Amenta F, Tayebati SK, Terzano C. Changes in muscarinic cholinergic receptor expression in human peripheral blood lymphocytes in allergic rhinitis patients. Pulm Pharmacol Ther. 2008;21:79–87. doi: 10.1016/j.pupt.2006.12.006. [DOI] [PubMed] [Google Scholar]
- Richards DF, Fernandez M, Caulfield J, Hawrylowicz CM. Glucocorticoids drive human CD8+ T cell differentiation towards a phenotype with high IL-10 and reduced IL-4, IL-5 and IL-13 production. Eur J Immunol. 2000;30:2344–2354. doi: 10.1002/1521-4141(2000)30:8<2344::AID-IMMU2344>3.0.CO;2-7. [DOI] [PubMed] [Google Scholar]
- Robbins MS, Mannering GJ. Effects of the interferon inducing agents tilorone and polyriboinosinic acid . polyribocytidylic acid (poly IC) on the hepatic monooxygenase systems of pregnant and fetal rats. Biochem Pharmacol. 1984;33:1213–1222. doi: 10.1016/0006-2952(84)90172-2. [DOI] [PubMed] [Google Scholar]
- Romagnani S. Lymphokine production by human T cells in disease states. Annu Rev Immunol. 1994;12:227–257. doi: 10.1146/annurev.iy.12.040194.001303. [DOI] [PubMed] [Google Scholar]
- Roncarolo MG, Gregori S, Battaglia M, Bacchetta R, Fleischhauer K, Levings MK. Interleukin-10-secreting type 1 regulatory T cells in rodents and humans. Immunol Rev. 2006;212:28–50. doi: 10.1111/j.0105-2896.2006.00420.x. [DOI] [PubMed] [Google Scholar]
- Rönnblom L, Eloranta ML, Alm GV. The type I interferon system in systemic lupus erythematosus. Arthritis Rheum. 2006;54:408–420. doi: 10.1002/art.21571. [DOI] [PubMed] [Google Scholar]
- Rossi D, Zlotnik A. The biology of chemokines and their receptors. Annu Rev Immunol. 2000;18:217–242. doi: 10.1146/annurev.immunol.18.1.217. [DOI] [PubMed] [Google Scholar]
- Saavedra J. Probiotics and infectious diarrhea. Am J Gastroenterol 95. 2000;1(Suppl.):S16–S18. doi: 10.1016/s0002-9270(99)00811-4. [DOI] [PubMed] [Google Scholar]
- Sacerdote P, Martucci C, Vaccani A, Bariselli F, Panerai AE, Colombo A, et al. The nonpsychoactive component of marijuana cannabidiol modulates chemotaxis and IL-10 and IL-12 production of murine macrophages both in vivo and in vitro. J Neuroimmunol. 2005;159:97–105. doi: 10.1016/j.jneuroim.2004.10.003. [DOI] [PubMed] [Google Scholar]
- Salvi S, Semper A, Blomberg A, Holloway J, Jaffar Z, Papi A, et al. Interleukin-5 production by human airway epithelial cells. Am J Respir Cell Mol Biol. 1999;20:984–991. doi: 10.1165/ajrcmb.20.5.3463. [DOI] [PubMed] [Google Scholar]
- Sandborn WJ. How future tumor necrosis factor antagonists and other compounds will meet the remaining challenges in Crohn's disease. Rev Gastroenterol Disord. 2004;4(Suppl.)(3):S25–S33. [PubMed] [Google Scholar]
- Saunders J, Tarby CM. Opportunities for novel therapeutic agents acting at chemokine receptors. Drug Discov Today. 1999;4:80–91. doi: 10.1016/s1359-6446(98)01280-x. [DOI] [PubMed] [Google Scholar]
- Sawada M, Imamura K, Nagatsu T. Role of cytokines in inflammatory process in Parkinson's disease. J Neural Transm. 2006;70(Suppl.):373–381. doi: 10.1007/978-3-211-45295-0_57. [DOI] [PubMed] [Google Scholar]
- Schindler H, Diefenbach A, Rollinghoff M, Bogdan C. IFN-γ inhibits the production of latent transforming growth factor-β1 by mouse inflammatory macrophages. Eur J Immunol. 1998;28:1181–1188. doi: 10.1002/(SICI)1521-4141(199804)28:04<1181::AID-IMMU1181>3.0.CO;2-O. [DOI] [PubMed] [Google Scholar]
- Schmidinger M, Hejna M, Zielinski CC. Aldesleukin in advanced renal cell carcinoma. Exp Rev Anticancer Ther. 2004;4:957–980. doi: 10.1586/14737140.4.6.957. [DOI] [PubMed] [Google Scholar]
- Schwarz MK, Wells TNC. Interfering with chemokine networks – the hope for new therapeutics. Curr Opin Chem Biol. 1999;3:407–417. doi: 10.1016/S1367-5931(99)80061-1. [DOI] [PubMed] [Google Scholar]
- Seidel HM, Lamb P, Rosen J. Pharmaceutical intervention in the JAK/STAT signaling pathway. Oncogene. 2000;19:2645–2656. doi: 10.1038/sj.onc.1203550. [DOI] [PubMed] [Google Scholar]
- Seki Y, Inoue H, Nagata N, Hayashi K, Fukuyama S, Matsumoto K, et al. SOCS-3 regulates onset and maintenance of TH2-mediated allergic responses. Nat Med. 2003;9:1047–1054. doi: 10.1038/nm896. [DOI] [PubMed] [Google Scholar]
- Shalaby MR, Waage A, Espevik T. Cytokine regulation of interleukin 6 production by human endothelial cells. Cell Immunol. 1989;121:372–382. doi: 10.1016/0008-8749(89)90036-1. [DOI] [PubMed] [Google Scholar]
- Shida K, Suzuki T, Kiyoshima-Shibata J, Shimada S, Nanno M. Essential roles of monocytes in stimulating human peripheral blood mononuclear cells with Lactobacillus casei to produce cytokines and augment natural killer cell activity. Clin Vaccine Immunol. 2006;13:997–1003. doi: 10.1128/CVI.00076-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shimbara A, Christodoulopoulos P, Soussi-Gounni A, Olivenstein R, Nakamura Y, Levitt RC, et al. IL-9 and its receptor in allergic and nonallergic lung disease: increased expression in asthma. J Allergy Clin Immunol. 2000;105:108–115. doi: 10.1016/s0091-6749(00)90185-4. [DOI] [PubMed] [Google Scholar]
- Shin HC, Benbernou N, Fekkar H, Esnault S, Guenounou M. Regulation of IL-17, IFN-γ and IL-10 in human CD8+ T cells by cyclic AMP-dependent signal transduction pathway. Cytokine. 1998;10:841–850. doi: 10.1006/cyto.1998.0375. [DOI] [PubMed] [Google Scholar]
- Shinkai M, Henke MO, Rubin BK. Macrolide antibiotics as immunomodulatory medications: proposed mechanisms of action. Pharmacol Ther. 2008;117:393–405. doi: 10.1016/j.pharmthera.2007.11.001. [DOI] [PubMed] [Google Scholar]
- Shtrichman R, Samuel CE. The role of gamma interferon in antimicrobial immunity. Curr Opin Microbiol. 2001;4:251–259. doi: 10.1016/s1369-5274(00)00199-5. [DOI] [PubMed] [Google Scholar]
- Silvennoinen O, Saharinen P, Paukku K, Takaluoma K, Kovanen P. Cytokine receptor signal transduction through Jak tyrosine kinases and Stat transcription factors. APMIS. 1997;105:497–509. doi: 10.1111/j.1699-0463.1997.tb05047.x. [DOI] [PubMed] [Google Scholar]
- Singh G, Renton KW, Stebbing N. Homogeneous interferon from E. coli depresses hepatic cytochrome P-450 and drug biotransformation. Biochem Biophys Res Commun. 1982;106:1256–1261. doi: 10.1016/0006-291x(82)91247-5. [DOI] [PubMed] [Google Scholar]
- Sitaraman SV, Gewirtz AT. Oprelvekin. Genetics Institute. Curr Opin Investig Drugs. 2001;2:1395–1400. [PubMed] [Google Scholar]
- Song JS, HaleemSmith H, Arudchandran R, Gomez J, Scott PM, Mill JF, et al. Tyrosine phosphorylation of Vav stimulates IL-6 production in mast cells by a Rac/c-Jun N-terminal kinase-dependent pathway. J Immunol. 1999;163:802–810. [PubMed] [Google Scholar]
- Spellberg B, Edwards JEJ. Type 1/type 2 immunity in infectious diseases. Clin Infect Dis. 2001;32:76–102. doi: 10.1086/317537. [DOI] [PubMed] [Google Scholar]
- Spelman K, Burns J, Nichols D, Winters N, Ottersberg S, Tenborg M. Modulation of cytokine expression by traditional medicines: a review of herbal immunomodulators. Altern Med Rev. 2006;11:128–150. [PubMed] [Google Scholar]
- Srivastava MD, Srivastava BI, Brouhard B. Delta9 tetrahydrocannabinol and cannabidiol alter cytokine production by human immune cells. Immunopharmacology. 1998;40:179–185. doi: 10.1016/s0162-3109(98)00041-1. [DOI] [PubMed] [Google Scholar]
- Strizki JM, Tremblay C, Xu S, Wojcik L, Wagner N, Gonsiorek W, et al. Discovery and characterization of vicriviroc (SCH 417690), a CCR5 antagonist with potent activity against human immunodeficiency virus type 1. Antimicrob Agents Chemother. 2005;49:4911–4919. doi: 10.1128/AAC.49.12.4911-4919.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Su HC, Cousens LP, Fast LD, Slifka MK, Bungiro RD, Ahmed R, et al. CD4+ and CD8+ T cell interactions in IFN-gamma and IL-4 responses to viral infections: requirements for IL-12. J Immunol. 1998;160:5007–5017. [PubMed] [Google Scholar]
- Suárez-Méndez R, García-García I, Fernández-Olivera N, Valdés-Quintana M, Milanés-Virelles MT, Carbonell D, et al. Adjuvant interferon gamma in patients with drug - resistant pulmonary tuberculosis: a pilot study. BMC Infect Dis. 2004;4:44–51. doi: 10.1186/1471-2334-4-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Subramanian GM, Fiscella M, Lamousé-Smith A, Zeuzem S, McHutchison JG. Albinterferon alpha-2b: a genetic fusion protein for the treatment of chronic hepatitis C. Nat Biotechnol. 2007;25:1411–1419. doi: 10.1038/nbt1364. [DOI] [PubMed] [Google Scholar]
- Sukhai M, Yong A, Pak A, Piquette-Miller M. Decreased expression of P-glycoprotein in interleukin-1beta and interleukin-6 treated rat hepatocytes. Inflamm Res. 2001;50:362–370. doi: 10.1007/PL00000257. [DOI] [PubMed] [Google Scholar]
- Swiatecka-Urban A. Anti-interleukin-2 receptor antibodies for the prevention of rejection in pediatric renal transplant patients: current status. Paediatr Drugs. 2003;5:699–716. doi: 10.2165/00148581-200305100-00005. [DOI] [PubMed] [Google Scholar]
- Szczuciński A, Losy J. Chemokines and chemokine receptors in multiple sclerosis. Potential targets for new therapies. Acta Neurol Scand. 2007;115:137–146. doi: 10.1111/j.1600-0404.2006.00749.x. [DOI] [PubMed] [Google Scholar]
- Takahashi N, Kitazawa H, Iwabuchi N, Xiao JZ, Miyaji K, Iwatsuki K, et al. Immunostimulatory oligodeoxynucleotide from Bifidobacterium longum suppresses Th2 immune responses in a murine model. Clin Exp Immunol. 2006;145:130–138. doi: 10.1111/j.1365-2249.2006.03111.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takeuchi T, Ueki T, Sasaki Y, Kajiwara T, Li B, Moriyama N, et al. Th2-like response and antitumor effect of anti-interleukin-4 mAb in mice bearing renal cell carcinoma. Cancer Immunol Immunother. 1997;43:375–381. doi: 10.1007/s002620050347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tambur AR, Markham PN, Gebel HM. IL-4 inhibits P-glycoprotein in normal and malignant NK cells. Hum Immunol. 1998;59:483–487. doi: 10.1016/s0198-8859(98)00042-1. [DOI] [PubMed] [Google Scholar]
- Tapner M, Liddle C, Goodwin B, George J, Farrell GC. Interferon gamma down-regulates cytochrome P450 3A genes in primary cultures of well-differentiated rat hepatocytes. Hepatology. 1996;24:367–373. doi: 10.1002/hep.510240213. [DOI] [PubMed] [Google Scholar]
- Tarkowski E. Cytokines in dementias. Curr Drug Targets Inflamm Allergy. 2002;1:193–200. doi: 10.2174/1568010023344670. [DOI] [PubMed] [Google Scholar]
- Tayebati SK, Amenta F, Amici S, El-Assouad D, Gallai V, Ricci A, et al. Peripheral blood lymphocytes muscarinic cholinergic receptor subtypes in Alzheimer's disease: a marker of cholinergic dysfunction? J Neuroimmunol. 2001;121:126–131. doi: 10.1016/s0165-5728(01)00435-0. [DOI] [PubMed] [Google Scholar]
- Theofilopoulos AN, Koundouris S, Kono DH, Lawson BR. The role of IFN-γ in systemic lupus erythematosus: a challenge to the Th1/Th2 paradigm in autoimmunity. Arhritis Res. 2001;3:136–141. doi: 10.1186/ar290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomsen LL, Topley P, Daly MG, Brett SJ, Tite JP. Imiquimod and resiquimod in a mouse model: adjuvants for DNA vaccination by particle-mediated immunotherapeutic delivery. Vaccine. 2004;22:1799–1809. doi: 10.1016/j.vaccine.2003.09.052. [DOI] [PubMed] [Google Scholar]
- Tinel M, Elkahwaji J, Robin MA, Fardel N, Descatoire V, Haouzi D, et al. Interleukin-2 overexpresses c-myc and down-regulates cytochrome P-450 in rat hepatocytes. J Pharmacol Exp Ther. 1999;289:649–655. [PubMed] [Google Scholar]
- Tsuji F, Oki K, Okahara A, Suhara H, Yamanouchi T, Sasano M, et al. Differential effects between marimastat, a TNF-alpha converting enzyme inhibitor, and anti-TNF-alpha antibody on murine models for sepsis and arthritis. Cytokine. 2002;17:294–300. doi: 10.1006/cyto.2002.1015. [DOI] [PubMed] [Google Scholar]
- Umannová L, Machala M, Topinka J, Nováková Z, Milcová A, Kozubík A, et al. Tumor necrosis factor-alpha potentiates genotoxic effects of benzo[a]pyrene in rat liver epithelial cells through upregulation of cytochrome P450 1B1 expression. Mutat Res. 2008;640:162–169. doi: 10.1016/j.mrfmmm.2008.02.001. [DOI] [PubMed] [Google Scholar]
- Verthelyi D, Ishii KJ, Gursel M, Takeshita F, Klinman DM. Human peripheral blood cells differentially recognize and respond to two distinst CpG motifs. J Immunol. 2001;166:2372–2377. doi: 10.4049/jimmunol.166.4.2372. [DOI] [PubMed] [Google Scholar]
- Verzijl D, Storelli S, Scholten DJ, Bosch L, Reinhart TA, Streblow DN, et al. Noncompetitive antagonism and inverse agonism as mechanism of action of nonpeptidergic antagonists at primate and rodent CXCR3 chemokine receptors. J Pharmacol Exp Ther. 2008;325:544–555. doi: 10.1124/jpet.107.134783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vilček J. The cytokines: an overview. In: Thomson AW, Lotze MT, editors. The Cytokine Handbook. 4th edn. Vol. 1. Amsterdam: Academic Press; 2003. pp. 3–18. [Google Scholar]
- Vincent JL. Afelimomab. Int J Clin Pract. 2000;54:190–193. [PubMed] [Google Scholar]
- Vinderola G, Matar C, Palacios J, Perdigón G. Mucosal immunomodulation by the non-bacterial fraction of milk fermented by Lactobacillus helveticus R389. Int J Food Microbiol. 2007;115:180–186. doi: 10.1016/j.ijfoodmicro.2006.10.020. [DOI] [PubMed] [Google Scholar]
- Vlotides G, Sörensen AS, Kopp F, Zitzmann K, Cengic N, Brand S, et al. SOCS-1 and SOCS-3 inhibit IFN-alpha-induced expression of the antiviral proteins 2,5-OAS and MxA. Biochem Biophys Res Commun. 2004;320:1007–1014. doi: 10.1016/j.bbrc.2004.06.051. [DOI] [PubMed] [Google Scholar]
- von Patay B, Loppnow H, Feindt J, Kurz B, Mentlein R. Catecholamines and lipopolysaccharide synergistically induce the release of interleukin-6 from thymic epithelial cells. J Neuroimmunol. 1998;86:182–189. doi: 10.1016/s0165-5728(98)00051-4. [DOI] [PubMed] [Google Scholar]
- Voravud N, Sriuranpong V. Clinical benefits of epoetin alfa (Eprex) 10 000 units subcutaneously thrice weekly in Thai cancer patients with anemia receiving chemotherapy. J Med Assoc Thailand. 2005;88:607–612. [PubMed] [Google Scholar]
- Vugmeyster Y, Szklut P, Tchistiakova L, Abraham W, Kasaian M, Xu X. Preclinical pharmacokinetics, interspecies scaling, and tissue distribution of humanized monoclonal anti-IL-13 antibodies with different IL-13 neutralization mechanisms. Int Immunopharmacol. 2008;8:477–483. doi: 10.1016/j.intimp.2007.12.004. [DOI] [PubMed] [Google Scholar]
- Vuppugalla R, Shah RB, Chimalakonda AP, Fisher CW, Mehvar R. Microsomal cytochrome P450 levels and activities of isolated rat livers perfused with albumin. Pharm Res. 2003;20:81–88. doi: 10.1023/a:1022202926073. [DOI] [PubMed] [Google Scholar]
- de Waal Malefyt R, Abrams J, Bennett B, Figdor CG, de Vries J. Interleukin 10 (IL-10) inhibits cytokine synthesis by human monocytes: an autoregulatory role of IL-10-produced by monocytes. J Exp Med. 1991;174:1209–1220. doi: 10.1084/jem.174.5.1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wagner TL, Ahonen CL, Couture AM, Gibson SJ, Miller RL, Smith RM, et al. Modulation of TH1 and TH2 cytokine production with the immune response modifiers, R-848 and imiquimod. Cell Immunol. 1999;191:10–19. doi: 10.1006/cimm.1998.1406. [DOI] [PubMed] [Google Scholar]
- Waller EK. The role of sargramostim (rhGM-CSF) as immunotherapy. Oncologist. 2007;12(Suppl.)(2):22–26. doi: 10.1634/theoncologist.12-S2-22. [DOI] [PubMed] [Google Scholar]
- Wang CH, Gao ZQ, Ye B, Cai JT, Xie CG, Qian KD, et al. Effect of emodin on pancreatic fibrosis in rats. World J Gastroenterol. 2007;13:378–382. doi: 10.3748/wjg.v13.i3.378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Q, Miyakawa Y, Fox N, Kaushansky K. Interferon-α directly represses megakaryopoiesis by inhibiting thrombopoietin-induced signaling through induction of SOCS-1. Blood. 2000;96:2093–2099. [PubMed] [Google Scholar]
- Warren HS, Kinnear BF, Phillips JH, Lanier LL. Production of IL-5 by human NK cells and regulation of IL-5 secretion by IL-4, IL-10, and IL-12. J Immunol. 1995;154:5144–5152. [PubMed] [Google Scholar]
- Welters ID, Menzebach A, Goumon Y, Langefeld TW, Teschemacher H, Hempelmann G, et al. Morphine suppresses complement receptor expression, phagocytosis, and respiratory burst in neutrophils by a nitric oxide and µ3 opiate receptor-dependent mechanism. J Neuroimmunol. 2000;111:139–145. doi: 10.1016/s0165-5728(00)00401-x. [DOI] [PubMed] [Google Scholar]
- Werb Z, Foley R, Munck A. Interaction of glucocorticoid receptors on monocytes and macrophages. J Exp Med. 1978;147:1684–1694. doi: 10.1084/jem.147.6.1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whitehouse MW, Beck FJ. Impaired drug metabolism in rats with adjuvant-induced arthritis: a brief review. Drug Metab Dispos. 1973;1:251–255. [PubMed] [Google Scholar]
- Wick MJ, Minnerath SR, Roy S, Ramakrishan S, Loh HH. Differential expression of opioid receptor genes in human lymphoid cell lines and peripheral blood lymphocytes. J Neuroimmunol. 1996;64:29–36. doi: 10.1016/0165-5728(95)00144-1. [DOI] [PubMed] [Google Scholar]
- Williams K, Dooley N, Ulvestad E, Becher B, Antel JP. IL-10 production by adult human derived microglial cells. Neurochem Int. 1996;29:55–64. doi: 10.1016/0197-0186(95)00138-7. [DOI] [PubMed] [Google Scholar]
- Wittig BM. Drug evaluation: CNTO-1275, a mAb against IL-12/IL-23p40 for the potential treatment of inflammatory diseases. Curr Opin Investig Drugs. 2007;8:947–954. [PubMed] [Google Scholar]
- Wolk K, Sabat R. Interleukin-22: a novel T- and NK-cell derived cytokine that regulates the biology of tissue cells. Cytokine Growth Factor Rev. 2006;17:367–380. doi: 10.1016/j.cytogfr.2006.09.001. [DOI] [PubMed] [Google Scholar]
- Wrana JL, Attisano L, Wieser R, Ventura F, Massagué J. Mechanism of activation of TGF-β receptor. Nature. 1994;370:341–347. doi: 10.1038/370341a0. [DOI] [PubMed] [Google Scholar]
- Wu R, Cui X, Dong W, Zhou M, Simms HH, Wang P. Suppression of hepatocyte CYP1A2 expression by Kupffer cells via AhR pathway: the central role of proinflammatory cytokines. Int J Mol Med. 2006;18:339–346. [PubMed] [Google Scholar]
- Wybran J, Appelboom T, Famaey J-P, Govaerts A. Suggestive evidence for receptors for morphine and methionine-enkephalin on normal human blood T lymphocytes. J Immunol. 1979;123:1068–1070. [PubMed] [Google Scholar]
- Xing Z, Wang J. Consideration of cytokines as therapeutics agents or targets. Curr Pharmaceut Design. 2000;6:599–611. doi: 10.2174/1381612003400623. [DOI] [PubMed] [Google Scholar]
- Yanagita M, Shimabukuro Y, Nozaki T, Yoshimura N, Watanabe J, Koide H, et al. IL-15 up-regulates iNOS expression and NO production by gingival epithelial cells. Biochem Biophys Res Commun 2002. 2002;297:329–334. doi: 10.1016/s0006-291x(02)02201-5. [DOI] [PubMed] [Google Scholar]
- Yang GB, Qiu CL, Zhao H, Liu Q, Shao Y. Expression of mRNA for multiple serotonin (5-HT) receptor types/subtypes by the peripheral blood mononuclear cells of rhesus macaques. J Neuroimmunol. 2006;178:24–29. doi: 10.1016/j.jneuroim.2006.05.016. [DOI] [PubMed] [Google Scholar]
- Yasukawa H, Sasaki A, Yoshimura A. Negative regulation of cytokine signaling pathways. Annu Rev Immunol. 2000;18:143–164. doi: 10.1146/annurev.immunol.18.1.143. [DOI] [PubMed] [Google Scholar]
- Yin TG. Characterization of IL-6 production by B-cells. Immunol Invest. 1990;19:413–420. doi: 10.3109/08820139009052969. [DOI] [PubMed] [Google Scholar]
- Ying S, Durham SR, Corrigan CJ, Hamid Q, Kay AB. Phenotype of cells expressing mRNA for TH2-type (interleukin 4 and interleukin 5) and TH1-type (interleukin 2 and interferon gamma) cytokines in bronchoalveolar lavage and bronchial biopsies from atopic asthmatic and normal control subjects. Am J Respir Cell Mol Biol. 1995;12:477–487. doi: 10.1165/ajrcmb.12.5.7742012. [DOI] [PubMed] [Google Scholar]
- Ying S, O'Connor BJ, Meng Q, Woodman N, Greenaway S, Wong H, et al. Expression of prostaglandin E2 receptor subtypes on cells in sputum from patients with asthma and controls: effect of allergen inhalational challenge. J Allergy Clin Immunol. 2004;114:1309–1316. doi: 10.1016/j.jaci.2004.08.034. [DOI] [PubMed] [Google Scholar]
- Yoshimura A, Naka T, Kubo K. SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol. 2007;7:454–465. doi: 10.1038/nri2093. [DOI] [PubMed] [Google Scholar]
- Youn HS, Saitoh SI, Miyake K, Hwang DH. Inhibition of homodimerization of Toll-like receptor 4 by curcumin. Biochem Pharmacol. 2006;72:62–69. doi: 10.1016/j.bcp.2006.03.022. [DOI] [PubMed] [Google Scholar]
- Yu JJ, Gaffen SL. Interleukin-17: a novel inflammatory cytokine that brdges innate and adaptive immunity. Front Biosci. 2008;13:170–177. doi: 10.2741/2667. [DOI] [PubMed] [Google Scholar]
- Yu P, Zheng C, Chen J, Zhang G, Liu Y, Suo X, et al. Investigation on PEGylation strategy of recombinant human interleukin-1 receptor antagonist. Bioorg Med Chem. 2007;15:5396–5405. doi: 10.1016/j.bmc.2007.05.061. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Center DM, Wu DM, Cruikshank WW, Yuan J, Andrews DW, et al. Processing and activation of pro-interleukin-16 by caspase-3. J Biol Chem. 1998;273:1144–1149. doi: 10.1074/jbc.273.2.1144. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Hegen M, Xu J, Keith JCJ, Jin G, Du X, et al. Characterization of (2R, 3S)-2-([[4-(2-butynyloxy)phenyl]sulfonyl]amino)-N,3-dihydroxybutanamide, a potent and selective inhibitor of TNF-alpha converting enzyme. Int Immunopharmacol. 2004;4:1845–1857. doi: 10.1016/j.intimp.2004.08.003. [DOI] [PubMed] [Google Scholar]
- Zhou H, Jang H, Fleischmann RM, Bouman-Thio E, Xu Z, Marini JC, et al. Pharmacokinetics and safety of golimumab, a fully human anti-TNF-alpha monoclonal antibody, in subjects with rheumatoid arthritis. J Clin Pharmacol. 2007;47:383–396. doi: 10.1177/0091270006298188. [DOI] [PubMed] [Google Scholar]
- Zhu HJ, Burgess AW. Regulation of transforming growth factor-β signaling. Mol Cell Biol Res Commun. 2001;4:321–330. doi: 10.1006/mcbr.2001.0301. [DOI] [PubMed] [Google Scholar]
- Zhu LX, Sharma S, Stolina M, Gardner B, Roth MD, Tashkin DP, et al. Delta-9-tetrahydrocannabinol inhibits antitumor immunity by a CB2 receptor-mediated, cytokine-dependent pathway. J Immunol. 2000;165:373–380. doi: 10.4049/jimmunol.165.1.373. [DOI] [PubMed] [Google Scholar]
- Zídek Z. Adenosine – cyclic AMP pathways and cytokine expression. Eur Cytokine Netw. 1999;10:319–328. [PubMed] [Google Scholar]