gms | German Medical Science

GMS Current Topics in Otorhinolaryngology - Head and Neck Surgery

Deutsche Gesellschaft für Hals-Nasen-Ohren-Heilkunde, Kopf- und Hals-Chirurgie e.V. (DGHNOKHC)

ISSN 1865-1011

Cellular and molecular mechanisms in environmental and occupational inhalation toxicology

Review Article

Suche in Medline nach

GMS Current Topics in Otorhinolaryngology - Head and Neck Surgery 2004;3:Doc02

Die elektronische Version dieses Artikels ist vollständig und ist verfügbar unter: http://www.egms.de/de/journals/cto/2004-3/cto000002.shtml

Veröffentlicht: 28. Dezember 2004

© 2004 Riechelmann.
Dieser Artikel ist ein Open Access-Artikel und steht unter den Creative Commons Lizenzbedingungen (http://creativecommons.org/licenses/by-nc-nd/3.0/deed.de). Er darf vervielfältigt, verbreitet und öffentlich zugänglich gemacht werden, vorausgesetzt dass Autor und Quelle genannt werden.


Abstract

The central issue of this review are inflammatory changes that take place in the mucous membranes of the respiratory tract as a result of inhaled pollutants. Of particular relevance are dusts, SO2, ozone, aldehydes und volatile organic compounds. Bioorganic pollutants, especially fragments of bacteria and fungi, occur predominantly in indoor dusts. They activate the toll-like/IL-1 receptor and lead to the activation of the transcription factor NF-κB for the release of numerous proinflammatory cytokines. Metals are predominant in ambient air dust particles. They induce the release of reactive oxygen species that cause damage to lipids, proteins and the DNA of the cell. As well as NF-κB, transcription factors that foster proliferation are activated via stress activated protein kinases. Organic compounds such as polycyclic aromatic hydrocarbons and nitroso-compounds of incomplete combustion processes activate additional via the cytosolic arylhydrocarbon receptor for detoxification enzymes. Sulphur dioxide leads to acid stress, and ozone to oxidative stress of the cell. This is accompanied by the release of proinflammatory cytokines via stress activated protein kinases. Aldehydes and volatile organic compounds activate the vanilloid receptor of trigeminal nerve fibres and induce a hyperreactivity of the mucous membrane via the release of nerve growth factors. The mechanisms described work synergistically and lead to a chronic inflammatory reaction of the mucous membranes of the upper respiratory tract that is regularly demonstrable in inhabitants of western industrial nations. It is unclear whether we are dealing here with a physiological inflammation or with an at least partially avoidable result of chronic pollutant exposure.

Keywords: respiratory mucous membrane, toxicology, endotoxins, metals, polycyclic aromatic hydrocarbons, nitrogen monoxide, reactive oxygen species, cell membrane receptors, signal transduction, transcription factors


1. Introduction

Illnesses result from a complex interplay between favourable individual factors (susceptibility), harmful agents (noxae) effecting individuals, additional external factors as well as individual assimilation of these variables [Fig. 1]. Usually one of these factors is at the fore as a cause of the illness, e.g. genetic disposition in the case of hereditary diseases, anatomical variants in some forms of chronic sinusitis, an acute infection due to a defined causative agent or also a psychological conflict situation leading to physical symptoms. A large number of factors are not noticed in the clinic or in practice. Nevertheless they exist.

For example, for a doctor in a pollution free rural area, sinusitis has exactly the same appearance as it would for a doctor in an area with severe air pollution. However, sinusitis clearly occurs more frequently in areas of high pollution than in low pollution areas [1], [2], [3], [4], [5], [6], [7], [8], [9], [10], [11]. In a region with air pollution, environmental pollutants are thus involved in the emergence of sinusitis. However, this is not immediately recognizable and in individual cases cannot be quantified, either. Analogous to this, employees in particular professions suffer more frequently from malignancies of the upper aerodigestive tract than the population average. This difference also remains significant if the effects of alcohol and tobacco consumption are compensated for [12]. Here, too, the job effect on the emergence of the illness is in individual cases not immediately apparent and usually difficult to quantify.

These examples show that environmental and occupational pollutants play a role in disorders of the head and neck. This has been demonstrated for acute and chronic inflammation of the nose and paranasal sinuses, the middle ear, the throat and the larynx, allergies of the respiratory tract, disorders of the sense of smell, of hearing and of balance, as well as head and neck tumours [13]. Only in individual cases are environmental and occupational pollutants the decisive causal factor. Mostly it is a case of cofactors that further the emergence of a disorder. For various poisons, toxicological mechanisms have been examined with classic pharmacological methods. This does not apply to the effect mechanisms of many inhalation pollutants occurring in the environment and workplace. Right up until only a few years ago it was above all the morphological and functional changes that were described in specialist literature, the actual damage mechanisms remained unknown. Here, over the last few years a great deal of new knowledge has been gained through the use of cell biological techniques that are the subject of the present review.


2. Examples of inhaled pollutants relevant to ENT

Those chemical substances that are relevant for diseases of the head and neck are predominantly absorbed via inhalation; we are dealing here with substances transmitted by air. On the one hand they appear as gasses. For the state of a substance at a particular temperature, its volatility is a decisive factor. Boiling point is a simple indicator for volatility. Substances with a boiling point of over 200° are of secondary importance as inhalation gasses but can be absorbed by particles and so gain entry to the respiratory tract. The details of the concentration of gasses result in mass/volume, mostly in µg/m3. Gasses with environmental medical significance are SO2, NOx, ozone and formaldehyde. From an occupational medical point of view, volatile organic compounds (VOC), volatile complex mixtures containing hydrocarbon and irritant gasses are of additional significance.

The second form of pollutants transmitted by air is aerosols. Aerosols are fine dispersions of liquids or solid substances in a gas, frequently in air. Fog is liquid dispersant aerosols, airborne dusts are particulate dispersal aerosols and fumes are aerosols from combustion processes. Aerosol concentrations are also given in mass/volume, typically in µg/m3. Air pollutants in the form of particles have the highest environmental and industrial medical relevance as inhaled noxae [Fig. 2]. Environmental dusts are complex particulate air pollutants from the real environment that emerge, for example, due to combustion processes, as a result of swirling up of dust from the ground, of condensation of gaseous air components or due to the mechanical rubbing off of various materials. Urban dusts are environmental dusts that collect in urban agglomeration zones. Combustion residue is a component of environmental and industrial dusts with particularly toxicological characteristics. They contain, amongst other things, polycyclic aromatic hydrocarbons (PAH), nitroso compounds and aromatic amines. Coal fly ash (CFA) is a product of combustion from hard coal heating with a high level of quartz and aluminium hydroxide, residual oil fly ash (ROFA) derives from from oil heating and has a high metal content. CFA and ROFA can be collected from the exhaust particles of coal- or oil-fired power stations for experimental investigations with appropriate filters. Diesel exhaust particles (DEP) come predominantly from motor vehicle traffic and can be produced for experimental purposes from the exhaust fumes of diesel engines. Bioorganic dusts come from organic materials that owe their origin biological processes. Toxicologically, fragments of bacteria and fungi are of prime importance. The extremely inhomogeneous group of substances includes, for example, livestock dusts, laboratory animal dusts, feedstuff and grain dusts, food dusts, wood dusts as well as dusts containing mites and fungi, for example from biologic waste. Environmental dusts from ambient air, and in particular indoor dusts, also contain bioorganic pollutants (bacteria, fungi pollen, spores, mite excrement) that are related to health disorders. Quartz and asbestos dusts belong to the common problem aerosols in industrial medicine. These are followed by, for example, combustion residue such as welding fumes, bioorganic dusts and cooling and lubrication agents.


3.Toxicokinetics of the upper respiratory tract

Toxicology is concerned with the harmful effects of chemical substances. Effects of radioactive or other radiation, as well as mechanical or thermal effects do not belong to toxicology in the narrow sense [14]. The harmful effect of a substance is dependent upon its concentration and duration of action at the structures affected. These two factors are characterised by absorption, distribution, metabolic conversion and excretion and are the subjects of toxicokinetics. In pharmacological and toxicological texts books, toxicokinetics, main emphases are enteral absorption quota, first pass effect in the liver, distribution volumes as well as renal and biliary elimination. In the case of inhalative absorption, one has to differentiate between two cases. On the one hand it can lead to systemic effects following inhalative absorption, for example with inhalative anaesthesia. In this case, except for enteral absorption, the mechanisms described above apply. Local effects on the mucus membranes of the respiratory tract, however, are frequently in the foreground. These are the subjects of inhalation toxicology. Toxicokinetic factors of particular relevance in inhalation toxicology are listed in table 1 [Tab. 1].

3.1. Inhalability

Gasses can generally be inhaled. Under the influence of gravity gasses with a higher specific weight sink. For this reason the body size (children) or the position of the room (cellar) can influence the capacity of a gas to be inhaled. In the case of dusts, the mechanisms of inhalability are more complex. Dust in general is a disperse distribution of solid substances with particle sizes of below 200 µm [15]. Airborne dusts have a particle size of below 100 µm; larger particles sediment on the ground within a few seconds. Aerosols with a speck or droplet size of over 100 µm (USA: >35 µm) are for this reason not included in the fraction of those that can be inhaled.

3.2. Absorption and deposition of inhaled pollutants

The respiratory toxic effect of inhaled pollutants depends essentially upon the region of the respiratory tract they are either adsorbed or absorbed into. Here what is understood by adsorption is the attachment of a substance to a two-dimensional surface (deposition); what is understood by absorption is the intake into a three-dimensional matrix such as, for example, secretion or tissue. By resorption, absorption into the blood or lymph system is indicated. In the case of gasses, water solubility and reactivity are the main physical characteristics for absorption into the respiratory tract [16]. Gasses easily soluble in water are dissolved into the secretions of the upper respiratory tract and spread their effects here. In so far as they are not spread as aerosol droplets, only a small part of gasses easily soluble in water reach the lower respiratory tract. An overview of the absorption of a number of substances in the form of gas in the upper respiratory tract is provided in table 2 [Tab. 2].

In the case of aerosols, the deposition of particles is dependent upon the diameter, the form of the particles and their surface characteristics. Should one wish to describe the deposition by the particle size alone, then the influence of form and surface characteristics must be compensated for through calculation. To this purpose the particle sizes are converted into equivalent spherical diameters. This so-called aerodynamic diameter corresponds to the diameter of a sphere with the standard mass of 1g/cm3 and of the same sedimentation speed as the dust particle under observation. In which compartment of the respiratory tract the particles are predominantly deposited is dependent upon the aerodynamic diameter. The mass median of the aerodynamic diameter (MMAD) is the aerodynamic diameter that is either exceeded or fallen short of by 50% of the mass (not the amount) of the particle under observation. It is used for a simple characterisation of a dust mixture. Those dust fractions are termed total suspended particles (TSP) whose aerodynamic diameter is either smaller than or equal to 35 µm. This corresponds more or less to the inhalative fraction according to DIN ISO 7708. Fractions of up to 10 µm (particulate matter 10 [pm10]) are regarded as inhalative dust, which corresponds more or less to the thoracic fraction according to DIN ISO 7708. As a rule of thumb, the equation PM10 [µg/m3] = 0,55 x TSP [µg/m3] [17] is valid.

Dust particles characterised as respirable are up to 2•5 µm [pm2.5]. This corresponds with the above norm more or less to the alveolar fraction. Particles < 0•1 µm are characterised as ultrafine particles. The relation between aerodynamic diameter and deposition compartment in the respiratory tract according to DIN ISO 7708 in non-logarithmic presentation follows from illustration 3 [Fig. 3] [18].

A significant factor for the harmful effect of an inhaled agent is first and foremost the concentration of the harmful substance in the air breathed in and the duration of exposure. In addition, the effect of inhaled pollutants is influenced by variable breathing parameters. Thus there are fundamental differences in the harmful effects of inhaled pollutants depending upon whether one is dealing with breathing through the nose or the mouth. The volume of breath and breathing frequency also play a considerable role. The surface of the mucous membrane with which the harmful substances inhaled come into contact is also relevant [19]. Due to this, the harmful effects of inhaled pollutants differ between children and adults and between rest or physical strain.

3.3. Interaction with the respiratory secretion

Respiratory secretion contains water (95%), electrolytes (1%), proteins (1%), phospholipids (including surfactant) and carbohydrates (approx. 1%) [20]. It forms a 10 - 16 µm thick layer above the tissues of the respiratory tract. As first described by Lucas and Douglas in 1934 [21], respiratory secretion consists of two layers. The lower low-viscous periciliar liquid layer (sol phase) is formed predominantly by transepithelial transport of ions and water from ciliated epithelia and brush cells [22]. What are dealt with here are similar and partly identical processes of water and ion transport as are found in the kidney. The highly viscous surface gel phase (mucous layer) shows a high share of integrated glycoproteins (mucins). They are formed predominantly in the submucous glands and the goblet cells. The 200-500 kD large glycoproteins (mucins) have an amino acid framework on which various short chain sugars (fucose, galactose) and amino sugars (N-acetylneuraminic acids, N-acetylglucosamine, N-acetylgalactosamine) are either o-gylcosidic or n-glycosidic attached. Apomucins are peptides that form the central framework of the mucins [23], [24]. The apomucins Muc1, Muc3, Muc4, Muc5A, Muc5B, Muc5AC, Muc7, Muc8 and Muc13 are expressed in the respiratory tract. The genes and their mRNAs are identified in epithelial cells of the submucous glands and in the goblet cells of the respiratory tract [25], [26]. The regulation of the gene expression of different mucins is subject to species differences [27]. As well as this, the mucins Muc1 and Muc4 are involved in cellular signal transduction [28]. Numerous inhaled pollutants stimulate the mucin expression in the respiratory tract and thus contribute to hypersecretion [29], [30]. In the case of the exposure to noxious substances, the constant activation of mucin genes leads to goblet cell hyperplasia, a characteristic feature of chronic inflammation of the respiratory tract [31]. Consecutively, this leads to the dysregulation of mucus composition, with hyperviscous secretion. This impairs the mucocilliary transport and encourages colonization of pathogenic organisms.

In order to gain access to the mucous membrane of the respiratory tract, a noxious substance must overcome the secretion barrier. This process differs with noxious substances in the form of gas or particles. For lipophilic gasses, the watery respiratory secretion is to a large degree impenetrable. An insignificant solution of lipophilic substances in respiratory secretion can be conveyed through phospholipids with amphiphilic properties. Water-soluble gasses go into solution in the respiratory secretion. At the same time, acids frequently form that can damage the epithelia of the respiratory tract. This is the case with SO2 or NO2, for example. Respiratory secretion has a buffer capacity in the range of 6 µmol H+-Ions per pH-level. This buffer capacity is provided to a great extent by mucins [32], [33]. Through the buffer effect of the secretion, the respiratory mucous membrane is protected from acidification caused, for example, by inflammatory processes [34], [35] or by the exposure to noxious substances and an optimal secretion transport in the neutral pH range is provided [36], [37].

Particulate air pollution is firstly adsorbed onto the mucous blanket of the upper respiratory tract and the proximal tracheobronchial tree. The mucous blanket functions from a technical point of view as an adhesion filter [38]. Adsorbed particles release water-soluble components such as salts, metals or proteins that then reach the epithelia of the respiratory tract. In this way, for example, water-soluble allergen proteins also pass into the mucous membrane of the respiratory tract. Through gaps in the mucous layer, however, smaller particles also reach the respiratory epithelia. Here they are phagocytosed mainly by local macrophages and neutrophils and to a lesser extent by epithelial cells [39].

Alongside its function as a mechanical barrier and acid/ base buffer, respiratory secretion can also neutralise a various exogenous and endogenous agents. This protective function is based partly on the activity of mucins. After the addition of mucins to the cell culture medium, the pro-inflammatory effect of environmental dusts on BEAS-2B cells was significantly reduced [40]. The effect of irritants is limited by peptidases resident in secretion. They degrade proinflammatory peptides, such as substance P released from nerve fibres, and thus exert an anti-inflammatory effect. Typical representative of these enzymes resident in secretion are neutral endopeptidases (NEP), angiotensin converting enzyme (ACE) or dipeptidylpeptidase IV (DPPIV) [41], [42]. Reactive oxygen species are neutralised by a variety of antioxidants resident in secretion. These include reduced glutathione, ascorbic acid, uric acid, a tocopherol, transferrin and ceruloplasmin [43], [44], [45]. On the one hand they intercept inhaled oxygen radicals such as ozone, but can also absorb released reactive oxygen species in the bounds of inflammation processes from respiratory cells. Lactoferrin stems from the specific granula of neutrophil granulocytes and is secreted from the seromucous glands of the respiratory tract. As a chelating agent, it has a high iron-binding capacity, but also binds other bivalent transitional metals and can thus reduce the toxicity of inhaled metal compounds. The lactoferrin metal complex, which is less poisonous in comparison to pure metal ion, is absorbed into the respiratory cell via a lactoferrin receptor and added to the cellular metal metabolism [46], [47]. Defensins are microbicide peptides formed above all in neutrophil granulocytes, but also in macrophages and respiratory epithelia. Human respiratory epithelia release β-Defensin 2 within 1-2 hours after contact with mucous forms of pseudomonas aeruginosa. The receptor for this is the toll-like receptor 2 [48], [49], [50].

3.4. Metabolism of foreign substances in mucous membrane of the respiratory tract

If the inhaled pollutants have overcome the secretion barrier, they are metabolized by the cellular detoxification system of the mucous membranes of the respiratory tract. The mucous membrane of the nose and tracheobronchial tree is, after the liver, the most active metabolic organ [51], [52], [53], [54], [55], [56]. If, for example, radioactively marked nitrosamines are applied either intraperitoneal or intravenously to the laboratory animal, they accumulate just as intensively in the mucous membrane of the nose as in the liver [Fig. 4] [57].

Typical phase I and phase II biotransformation processes take place here [58]. The aim of these processes is the increase of the polarity and the water-solubility of xenobiotics so that they can be excreted via the kidneys or bile. Phase I and II enzymes also play a decisive role in the detoxification of toxins associated with tobacco smoke. A dysfunction of these detoxification enzymes, for example through gene polymorphisms, possibly furthers the development of head and neck tumours [59], [60], [61]. Detoxification enzymes in humans are mainly to be found in surface cells of the respiratory epithelia and in the sustentacular cells and Bowman glands of the olfactory epithelium [62].

The phase I enzymes belong predominantly to the group of cytochrome P-450 oxygenases (CYP P-450). The name comes from "pigment 450" as the reduced CYP P-450 shows an absorption band at 450 nm. In humans it is a case of at least 50 related enzymes with broadly overlapping substrate specificity. This enzyme group transmits oxygen atoms and can convert approx. 1 million different substrates. The resulting intermediate product, usually an epoxide, is very reactive and often more harmful than the initial substance [Fig. 5]. It can cause damage to lipids and proteins of the cell and more particularly DNA damage. The intermediate product of phase I biotransformation is conjugated in a second metabolism stage through phase II enzymes with polar molecules such as, for example, glucuronic acids or glutathione to a molecule easily soluble in water and excreted.

Phase I enzymes that were identified in the mucous membrane of the nose are listed in table 3 [Tab. 3]. Amongst others phase II enzymes, UDP glucuronyl transferases and glutathione S transferases were detected in the mucous membrane of the nose. UDP glucuronyl transferases transmit activated glucuronic acids to OH groups in, for example, phenols and alcohols, to COOH groups of organic acids or to NH2 groups of amines or primary amides. Glutathione S transferases transmit glutathione to electrophile centres, for example in epoxides, alkylhalides or chinones [63].

3.5. Elimination of inhaled pollutants

In the the upper respiratory tract and the tracheobronchial tree, the most important elimination mechanism is the mucociliary transport system. Noxious substances entrapped in respiratory secretion are transported at a speed of approx. 5 mm/min towards the throat and then swallowed. The driving force of the mucociliary transport system is the staggered beat activity of the cilia of respiratory epithelia. The cilia bend slightly to the side on the recovery beat, which allows them to carry out this movement entirely within the sol phase. On the active beat they stretch, reach into the surface gel phase with their ends and push it towards the throat [Fig. 6]. The ciliary beat frequency of human nasal mucous membrane is around 10 Hz.

Numerous inhalative pollutants interfere with the mucociliary transport. This results in the inhaled substances being able to remain for a longer time in a respiratory compartment and to accumulate there. In this way, their toxic function can be strengthened [64]. Although not quantifiable, one can assume that disturbances of the mucociliary transport also encourage the development of respiratory infections. The raised tendency for infection in patients with cystic fibrosis and ciliary dyskinesia bears this out.

In contrast to the upper respiratory tracts and the proximal tracheobronchial tree, the terminal sections of the bronchial tree and the alveolar space have no mucociliary transport. Particles that get this far are predominantly absorbed by macrophages and carried away via lymphatic drainage. This process is partly accompanied by a distinctive inflammatory reaction. The various elimination mechanisms result in the upper and lower respiratory tracts reacting differently to particle exposure. Whilst phagocytosis is very well characterised as an elimination mechanism for the lower respiratory tract, in the upper respiratory tract there is only few data for phagocytosis. As recent studies have shown, epithelial cells are also capable of phagocytosis [39], [65], [66], [67]. The significance of this observation for the elimination of inhaled pollutants is, however, still unclear. Further important elimination mechanisms of the upper respiratory tract are coughing, sneezing and profuse hypersecretion with which noxious substances can be expelled or flushed out from a respiratory compartment. It is true that these symptoms are an important expression of irritation syndromes, but they play a secondary role as elimination mechanisms in environmental and industrial medicine.

Resorbed via inhalation, that is to say systemically absorbed, inhaled pollutants are excreted predominantly via the exhalation. This is the basis of occupational threshold values in the air expelled, for example after benzene exposure. In addition, systemically absorbed inhaled pollutants are renally and biliarly eliminated.

3.6. Systemic resorption

Inhaled pollutants can have a harmful local effect on the respiratory tracts (airway toxicity) or on other organs after resorption. Typical examples for the effects of an inhaled agent far from the point of resorption are carbon monoxide poisoning or the narcotic effect of numerous volatile organic compounds such as inhalation anaesthetics. Included among the systemic effects on inhaled pollutants are neurotoxic and especially vestibulotoxic and ototoxic effects from organic solvents and complex mixtures containing hydrocarbons. Worthy of note in this context is the association of cardiovascular mortality and pollution caused by environmental dusts [68]. In a cohort study, the risk of dying within 16 years of diseases of the heart or circulation rose by up to eight percent per 10 µg/m3 pm2.5 [69]. The cause of this is probably systemic effects as a result of the release of inflammation mediators in the lower respiratory tract and alveolar space [68], [70].


4. Mechanisms of inhaled pollutants

The toxicity of a substance is a characteristic specific to it which determines in which way it interacts with living structures. Which harmful effects a substance causes in the organism and via which mechanisms it does it are the subjects of toxicodynamics.

4.1. Changes in respiratory mucous membranes

On the level of tissue, toxic effects of inhaled pollutants are described in detail [Fig. 7]. Numerous environmental pollutants can be observed to lead to an accumulation of neutrophil granulocytes and macrophages in the respiratory mucous membranes, in other words triggering an unspecific inflammatory reaction [71], [72], [73], [74], [75], [76], [77], [78], [79], [80], [81]. Cytokines and chemokines released by cells of the respiratory mucous membrane after contact with the noxious agent play a central role in these inflammatory reactions [82], [83], [84], [85], [86], [87], [88], [89], [90], [91], [92], [93], [94], [95], [96], [97], [98], [99], [100], [101], [102], [103], [104], [105], [106], [107]. In respiratory toxicology, relevant cytokines derive from cells that can be found in the respiratory mucous membrane. Epithelial cells are most common. Their main task is the maintenance of a mechanic barrier between the outside world and the organism. As well as this they form a functional unit of the mucociliary apparatus. Over the past years it has been recognised that human respiratory epithelia can in addition induce an unspecific inflammatory reaction. They are capable of releasing various cytokines and chemokines that further inflammation as a reaction to external stimulus. These include IL-1, IL-6, IL-8, TNFα, G-CSF, RANTES, ENA-78 and GRO [39], [84], [87], [88], [108], [109], [110]. Amongst the numerous effects of these proteins are that endothelial cells express adhesion molecules in their cell membrane in neighbouring capillary vessels. These cell receptors mediate the migration of further inflammatory cells, predominantly of macrophages and neutrophil granulocytes into the tissue. These release cytotoxic substances such as reactive oxygen species (ROS), myeloperoxidase (MPO), prostaglandins (PG) and leukotrienes (LT). Cellular dysfunction with impaired mucociliary transport, disturbances of the cell-cell contact with a break up of tight junctions and the detachment of the cells from the basal membrane (epithelial shedding) all result from the inhales pollutants themselves, as well as the cytokines and the inflammatory mediators secondary released by infiltrating inflammatory cells. The barrier function of the epithelium is significantly disrupted. The epithelial damage introduce repair processes through fibroblasts and proliferating epithelia that result in structural changes in the respiratory mucous membrane with extensive deposition of extracellular matrix proteins such as collagen (remodelling, thickening of the basal membrane). Should the inflammatory stimulus persist, the proliferating cells no longer differentiate to ciliated epithelia but rather to goblet cells (goblet cell hyperplasia) and squamous epithelium (metaplasia). Damage of the protective epithelial layer, released inflammatory mediators and various inhaled agents themselves activate trigeminal nociceptors within the mucous membrane and induce airway hyperreactivity.

4.1.1. Basics of the cell response

These processes are described for various inhaled pollutants in detail. The subject of current research is the question of how inhaled pollutants initially cause the cells of the respiratory mucous membrane to release proinflammatory cytokines, for example. This can first be clarified on a cell-biological level. The airway cell must always be made to raise the synthesis of proteins, in this case cytokines.

To this end, the stimulus must first be recognised by the cell and the signal transmitted from the cell periphery into the cell nucleus [Fig. 8]. This follows from cell receptors and cascade-like activation of intracellular signal proteins. The genetic information for the proteins of the cell is stored in the DNA in the cell nucleus. The conversion of the genetic information of a DNA segment into a target product, mostly a protein, is summarised under the term gene expression. According to the central dogma of cell biology, the path from DNA leads via the RNA to protein. In the cell nucleus, therefore, genes must be activated and transcribed into messenger RNA (mRNA). The influence of the transcription rate of genes (gene regulation) is a fundamental toxicity mechanism on inhaled pollutants. The gene regulation follows from regulatory transcription factors; a group of intracellular proteins that can either promote of silence the transcription of certain genes.

The mRNA must be passed on from the nucleus to the ribosomal protein synthesis apparatus in the cytosol of the cell. Here the translation into a protein takes place using the blueprint of the mRNA. Numerous proteins created in this way are channelled out of the cell. They may serve, for example, as components of the intermediary tissue substance (extracellular matrix ECM), or influence, as messengers, the behaviour of other cells (cytokines) or generate, as transmitters, a nerve action potential in other cells. Other proteins are incorporated into the cell membrane and transmit cell-cell or cell-matrix contacts or serve as cell receptors for the receiving of signals from other cells. The majority of the proteins formed remain in the cell. They carry out structural tasks as components of the cytoskeleton, for example, are involved in cell movement or provide for cellular metabolism, cell division or apoptosis. Also included in the proteins remaining in the cell are the transcription factors and cellular signal transduction molecules that as regulatory proteins together make up approx. 20% of the cell proteins.

Numerous intracellular proteins are involved in the intracellular signal transduction, for example, from a cell membrane receptor to the cell nucleus. Like blood clotting factors, these intracellular signal proteins are functionally connected one behind the other and are activated in the form of a cascade. The reversible protein activation and intracellular signal transduction are basic cell biological mechanisms that are given a central role in the conveyance of the harmful effects of inhaled pollutants.

4.1.2. Protein activation, protein kinases and intracellular signal transduction

In order to fulfil their complex task, cellular proteins must be able to reversibly alter their form and activity (allostery). These reversible changes result from the linking of small molecules to proteins [111]. The coupling of phosphate groups (phosphorylation), methyl groups or acetyl groups [112], nitric oxides (nitrosylation) [113], [114] or oxygen compounds [115] is typical [112]. As a result of the coupling, the protein changes its three-dimensional structure. The reversible change of intracellular proteins results particularly often as protein phosphorylation that is conveyed through enzymatic active proteins, the protein kinases. The phosphate groups are predominantly bound to amino acids that bear free hydroxyl (OH) groups. These include serine, threonine and tyrosine. Serine/threonine kinases are proteins that transmit a phosphate group to serine and/or threonine, tyrosine kinases transmit the phosphate group to tyrosine residue. Protein phosphorylation frequently results in a protein becoming enzymatically active itself and for its part catalyses the coupling of a phosphate group with another protein. This results in intracellular cascades of protein activation. The dephosphorylation and thus frequently the inactivation of proteins results from phosphatases. In the hypothetical resting state of a cell, activation maintains its balance through protein kinases and inactivation through phosphatases. An activation of the cell can thus result from the activation of protein kinases or the inactivation of phosphatases.

Phosphorylation can alter the activity of a protein in many ways. As well as protein kinase activity or phosphatase activity, binding sites for other proteins can be formed and proteins coupled with one and other, the linking to DNA molecules elicited, the permeability for example of ions altered, the enzyme activity in the context of metabolism processes influenced or movement processes performed. As well as the reversible protein activation through the linking of small groups of molecules, there is also irreversible protein activation resulting from the splitting off of protein parts, for example with blood clotting factors, the complementary system or caspases in the context of apoptosis.

Protein kinases and phosphatases are the main functional elements of intracellular signal transduction. Acting in concert, they provide intracellular signal pathways. One example is the mitogen activated protein kinases pathway (MAPK path) that leads from activated growth factor receptors to the activation of the extracellular signal regulated protein kinase 1 and 2 (ERK1/2). If mitogens (e.g. growth factors) bind to a cell membrane receptor, then a tyrosine kinase becomes active on that part of the receptor that projects into the interior of the cell (cytoplasmic domain). Via intermediate steps, a small GTP binding protein (Ras: Ras and Raf1 are the products of genes that in the case of a mutation can encourage tumour formation (protooncogenes). Should Ras or Raf1 proteins be formed through mutation that are constantly active, the cell is placed under constant growth and division pressure.) is activated that then activates the actual MAPK pathway. This consists of three proteins: RAF1 (a MAP-kinase-kinase-kinase or MAP 3 kinase), the MAP-kinase-kinase 1 (Syn MEK1, a MAP-2-kinase) and finally the MAP-kinase ERK1/2 (an MAP-1-kinase, as it were), that activates several transcription factors (e.g. Elk1, c-Myc, c-Fos). These transcription factors enhance the transcription of genes that promote cell growth and division. As well as the pure signal transmission, the MAPK pathway fulfils further functions. An activation threshold is built in, as MEK1 must be phosphorylated in several serine and threonine residues in order for its part to develop kinase activity. In addition the signal is amplified as Raf1 activates numerous MEK1 proteins. The MAP kinases of different signal transduction paths also influence each other, which allows a finely coordinated regulation of the cell response. The development of specific inhibitors of cellular signal transduction molecules is the subject of current pharmacological research.

A further relevant signal transduction pathway in inhalation toxicology is the stress-activated protein kinase (SAPK) path. Two closely related signal transduction pathways are subsumed under this term, i.e. the c Jun N-terminal kinase (JNK path) and the p38 pathway [Fig. 9]. Cellular stressors such as heat, acidity or radiation lead to the activation of substances closely related to Ras, the small GTP binding proteins Rac1 and Cdc42. These activate analogously to the MAPK path SAP-3 kinases and SAP-2 kinases and finally the protein kinases JNK or p38. These two protein kinases activate several transcription factors (e.g. c-Jun, STAT1, STAT3, c-Myc, CREB) that stimulate cell growth and division and that are involved in the expression of pro-inflammatory cytokines. The intracellular signal transduction of toll-like/IL-1 (TIL) receptors proceeds more easily. This receptor has no intrinsic tyrosine kinase activity, but does activate the receptor-associated tyrosine kinases interleukin 1 receptor-associated kinase (IRAK) and TNF receptor-associated factor 6 (TRAF6). These activate the NF-κB inducing kinase (NIK) and the inhibitor of NF-κB kinase (IKK) that activates NF-κB via intermediate steps. With cytokine receptors, the coupling of the ligands leads to the activation of the receptor-associated protein kinases described as Janus kinases (JAK). These directly activate a group of transcription factors described as signal transducer and activator of transcription (STAT). Like NF-κB, they mediate the transcription of genes that promote inflammation. In the most simple of cases, the receptor itself serves as the transcription factor. This is the case with the cytosolic arylhydrocarbon receptor. It is activated via lipophilic organic compounds such as PAH and then passes into the cell nucleus where amongst other things it activates genes for detoxification enzymes.

4.2. Toxicity mechanisms of bioorganic pollutants

The mechanisms of the initial cell activation, that is the perception of the harmful stimulus and the intracellular signal transduction, differ considerably between various inhaled pollutants. Respiratory, toxicologically relevant substances with exemplary character explain this initial cell activation in the following, where first of all particulate bioorganic pollutants are examined.

Dusts contain numerous fragments that are derived from living organisms. It concerns above all cell wall fragments of bacteria and fungi that have adhered to the surface of dust particles. They play a key role for the respiratory toxicity of dusts and are predominantly represented in the insoluble fraction PM2.5-10 [116]. The indicator substance for bioorganic pollutants in dusts are endotoxins. These are lipopolysaccharides (LPS) anchored to the cell wall of gram- bacteria that are composed of the immunologically active lipid A, a nuclear polysaccharide and a specific O-chain. Inhalation of endotoxins leads to the release of TNFα, IL-1β, IL-6 und IL-8 and consecutively to a neutrophil inflammatory reaction of the respiratory mucous membrane of the lower [117], [118], [119], and in higher concentrations also in the upper respiratory tracts [120], [121]. For people suffering from asthma or nasal allergies, these reactions are intensified [122].

Ambient air dusts have lower LPS activities than dusts found indoor. Utah valley dust, an ambient air dust from an industrial agglomeration zone rich in metals, has an endotoxin activity of 0.6 EU/mg [40]. Representative indoor dust from north German households has, according to our measurements, an endotoxin activity of 24 EU/mg dust, whilst dust from south German households with no livestock showed an activity of 30 EU/mg dust [123]. If we assume roughly an average indoor dust concentration of 50 - 250 µg/m3, then our results lie below 50 EU/m3 air. This corresponds to the Dutch limit for indoor endotoxin, i.e.approximately 5 ng/m3 [124]. In Dutch office blocks with sick-building syndrome, on average 250 ng/m3 endotoxin were found [125]. Bioorganic pollutants also play an outstanding role in occupational respiratory toxicology [126]. High LPS concentrations can be found in dust from working animals (stable dusts, on average 100 ng/m3) [127], laboratory animals, feedstuff and grain dusts, food dusts, wood dusts as well as in dusts containing mites and fungi, e.g. from the waste industry (75-575 EU/m3) [86]. In addition, high concentrations of endotoxins of up to 25000 ng/ml [128] occur in cooling and lubricating substances in the metal working industry.

Endotoxins can be neutralised by polymixin B or lipopolysaccharide binding protein. Through neutralisation studies, the endotoxin caused part of the inflammatory reaction can be estimated. The proinflammatory and cytotoxic effects of urban dusts from Zurich and Lugano were almost completely inhibited by polymixin B, and thus can above all be put down to bioorganic pollutants [95]. An at least partial neutralisation of the proinflammatory effects of environmental dusts by LPS neutralising substances has been established in several further studies [94], [116], [129], [130], [131], [132], [133], [134], [135], [136], [137], [138], [139], [140].

In respiratory epithelia, glucans from the cell wall of moulds trigger similar effects to LPS. This could be shown with the help of transcription profiles by DNA microarrays. The microarray technology makes possible the simultaneous recording of changes in transcription of several thousand genes. We have carried out a comparative examination of the effects of the two most widely spread bioorganic pollutants, LPS from gram- bacteria and 1,3-β-glucan from the cell wall of moulds on the respiratory epithelial cell line BEAS-2B. Various cytokines or chemokines released by the cells were also recorded. The reactions of epithelial cells to LPS and 1,3-β-glucan were very similar, both on the mRNA level as well as on the protein level. As well as LPS, further bacterial cell wall components are determined such as lipoteichoic acid, peptidoglycans, FMLP and so-called CpG motives (see 4.2.1. table 5). Along with glycans, mannans and mannanoproteins are also included in the pollution caused by fungi.

4.2.1. Cellular receptors for bioorganic pollution

An crucial initial step in respiratory inflammation through dusts is the phagocytosis of particles that are covered with bioorganic pollutants. By phagocytosis we understand the recognition, absorption and digestion of particle-shaped substances. Among the professional phagocytes are dendritic cells (DC), macrophages, monocytes and neutrophil granulocytes. To a lesser extent other cells, such as respiratory epithelia, are capable of phagocytosis [39]. The first step of phagocytosis is the recognition of the particle to be phagocytosed and its attachment to the cell membrane. This takes place on receptors on the surface of the cell membrane.

Here two cases must be distinguished between [141]. In the first case the bacteria and fungi particles on the particle surface are already known to the immune system and are opsonised by immunoglobulins. In this case the particle binding results via the FC-receptor (FcR) in the cell membrane of the phygocytes [Tab. 4]. FcR for IgG signifies FcγR, for IgA FcαR and for IgE FcεR. The FcγR group is the most significant for phagocytosis, but a various number of subtypes have to be differentiated [Tab. 4]. For IgM there are no Fc receptors. If the particle binds complement or IgM, then particle recognition can result via a complement receptor. The complement receptors CR1-CR5 [Tab. 4] exhibit different cell distribution patterns and functions [142]. If particles are phagocytosed via an Fc or complement receptor, a strong proinflammatory reaction is set off in the phagocyte.

In the second case the particle is unknown to the immune system and not opsonised with immunoglobulin or complement. Parts of the bacteria or fungi on the particles are then recognised by special cell receptors. The cell wall of bacteria and fungi consists of regularly ordered modules of protein, fats and sugars that give their surface its typical pattern. These patterns differ from the surface of animal cells with no cell wall and were named 'pathogen associated molecular patterns' (PAMP) by Janeway and Medzhitov. The PAMPs are recognised by cells of the innate immune system with pattern recognition receptors (PRRs) [Tab. 5]. This includes the toll-like receptors, the mannose receptor and the glucan receptor. As well as surface patterns, structure patterns of proteins or DNA modules of a number of different pathogens are also well known to the phagocytes.

The coupling of a ligand to a toll-like receptor leads to numerous proinflammatory changes in the cell through activation of the transcription factor NF-κB.

4.2.2. Gene regulation, transcription factors and NF-κB

Gene regulation, that is the alteration of the transcription rate of genes through transcription factors, plays a central role in respiratory toxicology. An enzyme complex, the RNA-polymerase II complex (The basic possibility to transcribe a gene into mRNA depends upon the organisational state of the chromatin. Low grade condensed euchromatin can be transcribed whilst high grade condensed heterochromatin is not accessible to the transcription apparatus. This organisational state of the chromatin is transmitted on cell division to the daughter cell. This explains why a basal cell of the respiratory epithelium does not suddenly produce for instance a nerve cell [143].), manages the transcription of DNA into mRNA. Some genes are continuously (constitutively) active. These are household genes that encode, for example, proteins of basic metabolism or the cytoskeleton of the cell. For transcription they require proteins that are referred to as "basal transcription factors". They bind to the promoter region in the DNA strand and bring about the linking of the RNA polymerase II. Basal transcription factors are as a rule present in sufficient numbers and thus do not act in a regulatory manner. Amongst the basal transcription factors are, for example, the TATA binding protein, SP1, and NF1.

In contrast to the constitutively active genes, with inducible genes the transcription can be either enhanced or inhibited; in an extreme case they can be either turned on or off. With these genes, regulatory sequences called responsive elements are built into the DNA strand along with the promoters. The responsive elements that strengthen transcription are called enhancers, those that reduce the transcription rate silencers. They contain nucleotide sequences that arrange the binding of "regulatory transcription factors" to the DNA. Some frequently described regulatory transcription factors and the nucleotide sequences of the responsive element within the DNA strand are presented in table 6 [Tab. 6].

If a regulatory transcription factor links to an enhancing responsive element, it facilitates the binding of the RNA polymerase II complex to the promoter region. In this way the transcription rate of this gene is raised and more mRNA is formed, which frequently, but not always, leads to an increase in the release of the encoded protein.

The several hundred regulatory transcription factors at present known constitute approximately 10% of the amount of protein in a cell. They can be divided up into five groups according to characteristic structural and functional parts - so-called motifs - of the DNA binding domain. These are the helix-turn-helix motif, the zinc finger motif, the leucine zipper motif, the copper fist motif and the basal helix-turn-helix motif. These structural features fit to bays in the DNA molecule [Fig. 10] and link onto them when particular nucleotide sequences can be found there.

Respiratory inflammation caused by inhaled pollutants such as, for example, bioorganic pollutants, are substantially mediated by the transcription factor NF-κB [40], [90], [91], [97], [99], [102], [144], [145], [146], [147], [148], [149], [150], [151], [152], [153], [154]. NF-κB is composed of two proteins of a group of DNA binding proteins of which the combination of proteins p50 und RelA is the most common [Fig. 11]. In resting cells NF-κB is bound to an inhibitor (IκB) present in the cytosol. The binding to the inhibitor prevents the translocation of NF-κB in the cell nucleus. Activation of toll-like receptors that, with regard to the intracellular signal transduction, are very closely related to the IL-1 receptor, leads via phosphorlation of NF-κB inducing kinase (NIK) to the activation of the IκB kinase. The activation of the IκB kinase (IKK) results in phosphorylation of IκB. Following this the NF-κB/IκB complex binds to ubiquitin that supplies it to a proteasome, i.e. a cystosolic protein degradation apparatus. The inhibitor is cleaved from the complex in the Proteasome, exposing an amino acid sequence (nuclear localisation peptide sequence [NLS]) that arranges the transport of NF-κB into the cell nucleus. In the cell nucleus, NF-κB binds to responsive elements with the sequence 5'-GGGGACTTTCC-3', whereby a certain variability is tolerated with the middle nucleotides [155], [156].

NF-κB activates genes for several proinflammatory cytokines [Tab. 7] [157], [158]. As well as this it reinforces the expression of adhesion molecules, immune receptors and acute phase proteins [155]. NF-κB activation also leads to the upregulation of iNOS, cyclooxygenase 2 and the PAF receptor [159] and regulates the transcription of various genes for apoptosis and cell division.

NF-κB is not only involved in the transcription of numerous proinflammatory signal molecules, several intracellular signal transduction pathways from several receptors also converge on NF-κB [157]. As well as the described pathway via the toll-like/IL-1 receptor, NF-κB can also be activated via the TNF receptor. In addition, NF-κB is activated through C5a via a receptor linked to a G protein [146], via bradykinin receptors [147], via arylhydrocarbon receptors [148], via ROS [102], [151], [160] and also via Fc receptors through the activation of the protein kinase C. For its part, NF-κB leads to the release of TNFα and IL-1 resulting in a self -reinforcing control loop. As well as the cytokines, the expression of the inhibitor is also increased, which checks the NF-κB activation as a negative regulatory loop.

4.2.3. Phagocytosis and oxidative burst

After the binding of the bacterial or fungal fragments to Fc, complement, or pattern recognition receptors, the particle is internalised. With this the second phase of the inflammatory cell activation through bioorganic pollutants begins. Pseudopodia push themselves around the particle and engulf it into a phagosome [161], [162]. Through the occupation of the receptor, small GTP binding Rho proteins Rac and CdC42 are activated. Via complex mechanisms, they act as intermediary in the accretion of actin monomers (G actin) to the receptor. Through further lengthening of the actin filaments, pseudopodia are pushed around the particle. In addition, activated tropmyosin-1 is possibly involved in these processes [163], [164], [165]. If the particle is completely internalised, then an oxidative burst occurs. This means the sudden, dramatic increase in reactive oxygen species in the cell within the scope of the phagocytosis processes. In this way, several proteins store together in the wall of the phagosomes to form the NADPH oxidase complex [166] that transmits electrons to molecular oxygen [Fig. 12]. The oxygen species that abruptly results here decompose the internalised particle. The NADPH oxidase complex generates an H+-ion during the electron transmission that is channelled into the phagosome via a proton channel. The acidification in the phagosome speeds up the breakdown of the internalised particle.

At the same time, the enzyme iNOS (inducible nitric oxide synthase) is activated and formed into high concentrations of nitrogen monoxide (NO). This is a very reactive radical that introduces similar harmful effects such as ROS [167], [168], [169], [170]. Professional phagocytes have absorbed and to a large extent decomposed a particle in approximately 30-60 minutes. Here, considerable amounts of ROS can be released that along with the affected cell can even damage neighbouring cells and can induce a local inflammatory reaction. The formation of ROS also leads to the activation of NF-κB, as further described below.

4.2.4. Reactive oxygen species and nitrogen monoxide

Due to the particular significance of reactive oxygen species and nitrogen monoxide in respiratory toxicology, these substances should be looked at in more detail. The physiological generation of energy of the cell through molecular oxygen results primarily in the mitochondrial membrane as the oxidative phosphorlation of ADP to energy-rich ATP. The mediating enzyme is the NADH dehydrogenase complex of the respiratory chain. Reduced, highly reactive oxygen metabolites result as a by-product. These include superoxide or hyperoxide

anion (O2 -), hydrogen peroxide (H2O2) and the particularly reactive hydroxide ion OH that forms in the presence of transition metals and that when dissolved appears as H3 O2 - (The point next to the symbol for the element represents the unpaired electron of the radical.). These reduced O2 derivatives are more reactive than molecular oxygen and for this reason are referred to as reactive oxygen species (ROS) [171]. The term "oxygen radical" is inexact as molecular oxygen itself is a biradical. Aproximately 1 2% of these toxic metabolic by-products can be released during oxidative phosphorylation. Antioxidative enzymes in the cell rapidly break them down. In this way, superoxiddismutase (SOD) reduces the superoxide O2 - to H2O2 and catalases and peroxidases, for example the glutathione peroxidase, H2O2 to H2O. Normally ROS and enzymes which break down ROS are in balance in the cell, however, through an increase in the amount of ROS or through a reduced activity of the ROS degrading enzymes, for example through glutathione depletion after SO2 exposure, the intracellular ROS concentration can increase. This is called oxidative stress.

As well as mitochondria and the phagosome within the scope of the oxidative burst, the smooth endoplasmic reticulum belongs to the intracellular ROS sources: ROS originate here, for example within the scope of the xenobiotics detoxification through cytochrome P-450 oxidases. Among the exogenic ROS sources are transition metals, γ-rays, UV rays, nutritional components and a variety of drugs [171]. ROS leads to damage of various cell components. Among the substances particularly sensitive to ROS are proteins, lipid membranes and DNA. With proteins, ROS leads to protein peroxidation. Aldehydes, ketones, and carbonyls are formed. Proteins modified in this way change their tertiary structure, lose their function and are quickly broken down inside cytosolic proteasomes. In this way a large number of cell functions such as energy generation, maintenance of membrane potential, movement processes, DNA repair and deactivation of signal proteins through phosphatase are impaired [171]. The weak point of the ROS in lipid membranes of the cell is their double bounds in unsaturated fatty acids. During peroxidation, reactive aldehydes, lipid hydroperoxides or cyclic endoperoxides occur that peroxidise further unsaturated fatty acids. A chain reaction occurs that can lead to a peroxidation of all the unsaturated fatty acids of a lipid membrane [171]. The third target structure of ROS is DNA molecules, whereby hydroxide ions have a particularly destructive effect. They cause single and double strand breaks, changes in the DNA bases, loss of purine bases, damage to the sugar backbone and DNA-protein adduct formation. Frequent adducts through ROS are 8-hydroxydeoxyguanosin (8-OhdG), 8-hydroxyadenin, thyminglycol or 5-hydroxymethyluracil [171]. 8-OhdG is a indicator for ROS-induced DNA damage commonly employed in environmental medicine.

In low concentrations, ROS have a stimulating effect of a large number of cell processes and are thus essential for the proper functioning of the cell. Amongst other things they act as intracellular signal molecules that reinforce the transcription of enzymes that break down ROS [115]. As well as this they influence the signal transduction ubiquitously by checking the activity of tyrosin phosphatases, thus reinforcing the tyrosinkinase effect [172]. They are involved in cell growth and cell division, as well as in the modulation of numerous biochemical processes, including the formation of prostaglandins. These positive ROS effects are probably the cause of the so-called "antioxidant paradox". Dietetic supplementation with antioxidants such as vitamin C, tocopheroles, polyphenoles and carotenoids in large studies with several thousand participants has produced no protection against illness, on the contrary, its has resulted in a raised incidence of illness [173], [174]. Concentrations of antioxidants that appear too high may also impair the redox balance and place the cell under reductive stress [171].

Nitrogen monoxide (NO) is a gaseous, very reactive free radical. It is formed through NO synthases (NOS) from the amino acid arginin. Here NADPH and O2 are used. NO is broken down within a few seconds. It functions on the one hand as a cellular signal molecule in, amongst others, nerve cells, endothelial and epithelial cells. For this purpose, NO is continually formed ("constitutively expressed") in low concentrations from the neuronal nNOS or the endothelial eNOS. In neuronal systems it serves as a neurotransmitter, in blood vessels is has a vasodilatory effect (endothelium-derived relaxing factor) and in the bronchial, system it is bronchodilatory.

The function of NO catalysed through iNOS (inducible NOS) in the context of phagocytosis is very different. It is secreted in very high concentrations in the phagolysosome and has a similar effect to ROS [167], [168], [169], [170]. It also leaves the cell and can cause damage in neighbouring cells. In addition, it merges with exhaled air where it serves as a sensitive marker for inflammations of the mucous membrane. NO is also measurable in the nose. It stems almost exclusively from the nasal sinuses and is an exceptional marker for inflammatory alterations of the nasal sinuses [175], [176], [177], [178], [179]. The reaction of NO with superoxides leads to the formation of peroxynitrit, an extremely cytotoxic compound. The release of peroxynitrit into cytosol influences the function of numerous proteins through nitrosylation [113]. Amongst other things it reacts with the amino acid tyrosine to form nitrotyrosine (NO2Tyr). This compound can be called on to confirm the presence of reactive nitrogen species in tissue and secretions [180].

In the foreground of the NO effect, however, is less its short-term increase within the scope of phagocytosis processes, than the long-term increase through the increased supply of the enzyme iNOS. The transcription of iNOS is upregulated through endotoxins and various cytokines such as TNFα, IL-1β and IFN-γ. One important signal transduction pathway takes place via the activation of NF-κB and STAT1 [181]. Continuous NO increase influences the regulation of numerous cell processes and functions as a ubiquitous signal molecule independent of signal transduction pathways by nitrosylsing important cellular molecules [172].

4.3. Toxicity mechanisms of metals

In industrial agglomeration zones in particular, metals play a prominent role in the harmful effects of ambient air dusts [40], [153], [182], [183], [184]. The toxic effect of such dusts on the respiratory system can be almost completely inhibited by chelating agents [40], [84], [145]. The transition metal zinc, copper, iron, nickel and vanadium are at the forefront. Metals are mainly detectable on the soluble fraction of pm2.5 [116]. In small amounts, transition metals are essential for the proper functioning of the cell. The intake of transition metal into the cell results from active transport processes. Because of their toxicity, metals are bound intracellularly to storage proteins (ferritin, metallothionein) [185], [186], [187]. If the cellular metal storage capacity is exceeded, free metal ions occur in the cell. Within the cell, free transition metals transmit an electron to the hydrogen peroxide that is present in small amounts in accordance with the Fenton reaction ( Equation 1 ). In this way a highly reactive hydroxide ion occurs and the cell is placed under oxidative stress [188], [189]. The released ROS lead to damage to proteins, lipids and DNA of the cell. The molecules altered by ROS trigger a stress response in the cell. They activate intracellular signal transduction cascades, whereby the stress activated kinase (SAPK) pathway is of particular significance [190], [191]. The induction of the stress activated kinase pathway through reactive oxygen species is a key mechanism, like metals, but also bioorganic compounds, lead to the release of proinflammatory cytokines. Here, the effects of metals and bioorganic compounds on the formation of ROS are synergistic [192]. Below, the stress response of the cell shall be observed in detail considering the metal-induced oxidative stress as an example.

4.3.1. Stress response of the cell

Changes in the surrounding condition of the cell interior require adaptation processes in the cell that are combined under the term cellular stress response. As well as oxidative stress, the stress response can also be produced by countless stimuli, for example by the withdrawal of nutrients and oxygen, or the heavy strain caused by typical cell functions such as muscle cell contractions. Other typical cell stressors are temporary heat (heat shock, e.g. 5 mins. at 42°C), ischemia/reperfusion, UV rays, DNA damage, hyperosmolarity, changes in pH values, proinflammatory cytokines, vasoactive peptides and also growth and differentiation signals. The varying stress responses are almost as numerous as the stressors themselves. Among these are apoptosis, cell cycle arrest, metabolic activation, release of proinflammatory cytokines and also growth, cell division or differentiation.

The stress activation of cells results from the stress activated protein kinase pathway (SAPK pathway). The activation of the SAPK pathway [Fig. 13] results via different mechanisms [193]. Cytokines and growth factors trigger the SAPK pathway in parallel to the typical signal transduction cascades activated by their specific receptors. Cell stressors, such as UV rays, osmotic stress, acid stress or ROS affecting the cell externally, react initially with parts of the lipid membrane. One typical chemical reaction is the hydrolysis of membrane sphingomyelins to ceramides. These act as second messenger and activate the SAPK pathway. Intracellular-active agents such as ROS or NO, as well as heat shock or toxic metabolites, for example in the breakdown of ethanol, lead to protein denaturations. Through protein denaturation, an indirect activation of the SPAK pathway probably results through suspension of the deactivating phosphatases [194]. Lipid peroxides also possibly bring about the activation of the SAPK pathway [145] in the cell membrane of cell organelles such as 4-hydroxy-2-nonenal (HNE) or malondialdehyde [195]. Ultimately DNA damage also leads via the tyrosine kinase c-Abl to the activation of the SAPK pathway [196], [197].

The SAPK pathway leads to the activation of various transcription factors, whereby activator protein 1 (AP 1) takes on a key role. No direct activation of NF-κB takes place via the SAPK pathway. In several types of cells, ROS can activate NF-κB directly and without the involvement of the SAPK pathway and thus contribute to the release of different cytokines [155], [198]. The mechanisms of this activation are not exactly known [199], [200], [201], [202], [203]. One plausible explanation for this is activation of CBP/p300 via the SAPK pathway, a co-activator of NF-κB which occurs constitutively in small amounts in the nucleus. This phenomenon was observed among others with English environmental dusts and appears to be typical for the release of ROS in respiratory epithelia induced by metals [144], [145], [153].

The biological effects of the activation of the SAPK pathway differ between lymphoid and non-lymphoid cells. In the case of the majority of non-lymphoid cells, cell growth and division are limited and proinflammatory cytokines are released [197]. Thus via the formation of ROS, metals lead to the release of proinflammatory cytokines and chemokines (IL-1, IL-6, IL-8, TNFα) from respiratory epithelia [204]. In T lymphocytes, the SAPK pathway appears to be essential for the differentiation to effector cells and to effector-specific cytokine production [205]. Excessive cellular stress triggers off an apoptosis programme in the cell [206], [207]. Stress activation is, amongst other things, to be observed in with the exposure to asbestos. Asbestos induces oxidative stress in macrophages, possibly through the release of Fe2+. This results in a reduction of anti-oxidative intracellular glutathione and an upregulation of AP-1 [208]. An activation of NF-κB occurs in respiratory epithelia that can be reduced through the prior application of antioxidants [209]. Quartz dusts also lead via the release of reactive oxygen species to the activation of NF-κB and the release of IL-8 from human respiratory epithelia [150].

4.4. Organic compounds and combustion residues

As well bioorganic pollutants and metal, organic compounds, e.g. from combustion processes, also play a considerable proinflammatory role. Organic compounds in urban dusts account for about 20% of the mass at pm10, and about 45% at pm2.5 [206]. From the point of view of industrial medicine, workers in coking plants, the metal working industry, blacktop workers and welders are exposed to combustion residue and smoke to a considerable extent. Present in combustion residue are different groups of substances harmful to health such as metal, polycyclic aromatic hydrocarbons, halogenated hydrocarbons (e.g. dioxin) and nitrosamines. Particulate emissions from diesel engines (diesel exhaust particles or DEP) are very common and of great relevance for environmental medicine. This involves saturated and unsaturated aliphatic hydrocarbons and their substitution products such as aldehydes and ketones, anhydrides and carbon acids. A further considerable component is polycyclic aromatic hydrocarbons [PAH], as well as nitro-, amino- and hydroxo-combinations.

Diesel soot is probably responsible for the association between heavy goods traffic and allergic respiratory disorders that have been shown in numerous studies [210], [211], [212], [213], [214], [215], [216], [217], [218], [219]. As well as allergic respiratory disorders of the upper respiratory tract in residents of areas heavily affected by traffic, non-allergic disorders also occur in large numbers [220]. After many years exposure to diesel soot, chronic inflammatory and degeneratively altered nasal cytologies were found in Swiss customs officers [221]. The PAHs typical for DEP are, however, also found in normal environmental dusts in concentrations that can trigger off inflammatory reaction in the respiratory tract [206]. DEP induce changes in respiratory cells similar to bioorganic pollutants and metals, and thus lead to a release of ROS and in this way induce cellular stress [88], [222]. As a consequence, this results in an activation of the SAPK pathway [222] and the release of proinflammatory cytokines [223], [224].

4.4.1. The arylhydrocarbon receptor

The PAHs contained in the combustion residue, halogenated aromatics and N-substituted polycyclic hydrocarbons function via an additional specific toxicity mechanism. These substances are mostly lipophilic and can pass the cell membrane. Within the cell they bind to a cytosolic receptor, the arylhydrocarbon receptor (AhR) [225], [226]. Activation of the Ah receptor [Fig. 14] leads to its translocation in the cell nucleus the activation of genes that are regulated via the xenobiotic responsive element. Genes for detoxification enzymes such as CYP1A1, CYP1A2 and CYP1B1 and NAD(P)H quinon oxidoreductase and UDP-glucuronosyltransferase are more intensely transcribed and are also traceable as protein [227]. In addition haemoxygenase-1 is expressed that stabilises the cellular redox potential [58], [206], [222]. The AhR brings about proinflammatory impulses through its interaction with NF-κB [228], [229], [230], [231]. Activation of the AhR influences the cell cycle, amongst other things by activation of cyclin-dependent kinase 2 and inhibition of TGF-β [232]. The carcinogenic effect of typical AhR ligands such as benzo(a)pyrene is also transmitted via the AhR. It is no longer detectable in AhR deficient mice [233]. Pei and colleagues describe an upregulation of p53, probably via an analogous NF-κB dependent promoter region in the p53 gene [148]. The AhR also interacts via the nucleus translocation factor ARNT with hypoxia response elements that, for example, regulate the expression of erythropoietin [234]. The AhR is an essential mediator of cytotoxicity transmitted via cigarette smoke [235].

4.5. Toxicity mechanisms of low toxicity particles

Dockery et al. were amongst the first to recognise the special respiratory toxicity of particulate air pollutants [236]. The question has to be answered as to whether the particles themselves are responsible for their respiratory toxicity as a result of physical and mechanical actions, or toxic dust constituents. In order to answer this question, numerous studies were conducted with dusts whose components are generally considered non-toxic. To be correct, these experimental dusts rarely found in the natural environment are termed "low toxic". Most animal experimental studies with low toxicity particles were carried out on the lower respiratory tract. In the lower respiratory tract definite, if comparatively weak inflammatory changes to the mucous membrane are triggered by low toxicity particles of, for example, melamine, polystyrene, titan dioxide or barium sulphate. This inflammatory reaction in the lower respiratory tract is predominantly determined by size and surface characteristics of the particle [237], [238], [239].

The key biological mechanism for the inflammatory reaction through low toxic particles in the lower respiratory tract is its phagocytosis through alveolar macrophages. This cell population comprises 40-50% of the cells in a bronchoalveolar lavage. For a long time it was unclear how macrophages manage to phagocyte particles without special immunological distinguishing features. The major role of scavenger receptors in these processes is now clear to a large extent [240], [241], [242], [243]. Scavenger receptors (SR) are the most primitive form of pattern recognition receptors [244]. They serve the phagocytosis of non-opsonized particles even if they do not have particular structural features. Due to the special collagenous makeup of their extramembranous domains, SR have a "sticky" effect and accordingly demonstrate a distinctive ligand promiscuity. They are to be found predominantly on macrophages but can also found on respiratory epithelia [245]. As well as the phagocytosis of gram+ und gram- bacteria, they convey the pathological effects of pollutants [246]. After their amino acid sequence they are assigned to types A, B and C. To type A receptors belong the groups SR-AI, SR-AII and MARCO. MARCO is to be found above all on lymph node and spleen macrophages. Their occurrence on alveolar macrophages is the subject of debate [242], [243], [247]. Type A SR only induce a weak inflammatory activation. With low toxic particles the phagocytosis is thus only accompanied by a weak inflammatory reaction [248]. This makes the scavenger receptor suitable for the neutral phagocytosis of apoptotic cells seen from the inflammatory aspect [142]. As well as low toxic particles, toxic particles from the environment also undergo phagocytosis via the SR [248]. Research in this area is facilitated by modern detection techniques [249], [250]. The proinflammatory effects with phagocytosis via SR are probably conveyed by a comparatively weak oxidative burst.

Whilst low toxicity dusts can induce an inflammatory reaction in the lower respiratory tracts, the upper respiratory tracts react to low toxic dusts via local reflex mechanisms. In the case of 32 healthy test persons at concentrations of 5000 µg/m3, low toxicity dusts (calcium carbonate with an average particle size of 15 µm) led, depending upon the dosage, to a reactive acceleration of mucociliary transport, an increase of the nasal respiratory resistance and feelings of nasal dryness [251]. These changes came about, however, without a nasal inflammatory reaction being detected. Two explanation present themselves for the lack of nasal inflammatory reaction on exposure in the upper respiratory tracts with respect to low toxicity particles: macrophages as professional phagocytes are considerably more scarce in the upper respiratory tracts than in the lower and the particles deposited in the nose are larger so that they cannot so easily undergo phagocytosis.

4.6. Sulphur dioxide and nitrogen dioxide

Sulphur dioxide is a pungent, colourless gas. It is highly water-soluble. In the upper respiratory tracts inhaled SO2 is almost completely dissolved in the respiratory secretion and reacts with H2O to form sulphurous acid. Less than 5% of the inhaled SO2 reaches the lower respiratory tracts. For this reason, it is mainly disorders of the upper respiratory tracts that occur with high ambient air-SO2 concentrations [8], [252], [253]. Central to this is the increased susceptibility for infections of the upper respiratory tracts. Changes in the lower respiratory tracts can be correlated either only in small measure or not at all with ambient air-SO2 concentrations [254], [255]. Changes in the lower respiratory tracts with comparatively high ambient air concentrations are, however, described [256].

After SO2 exposure, glycoproteins in the secretion first buffer the released protons. In this way the viscosity of the secretion is increased and the mucociliary transport decelerated [257], [258]. If the buffer capacity of the secretion is exhausted, the pH value drops and acid molecules come into contact with the cell and ciliary membrane of the respiratory epithelia. The exposure to protons leads to the damage to the epithelium. The sulphites do not damage the epithelium [259]. Through the slowing down of the ciliary beat the mucociliary transport is further impaired. Ultra-structurally, the development of fine stress fibres can first be observed and then the formation of small blebs in the cell and ciliary membrane (membrane blebs). At higher concentrations an increasing vacuolization can be recognised and then finally a break-up of the tight junctions [260], [Fig. 15].

Analogous ultra-structural changes can be frequently observed when cells are placed under varying degrees of stress, as here in the case of acid stress. It involves active cellular processes as part of this stress response. Due to their load, acid molecules cannot penetrate the lipid membrane, but they denature proteins in the cell membrane [193]. This releases a large number of cellular reactions that lead to the stress response of the cell described above. Thus acid stress activates NF-κB in human tracheal epithelial cells that leads, amongst other things, to an increased expression of IL-1 and the PAF receptor. The raising of the PAF receptor to the surface of the cell increases the adherence of streptococcus pneumoniae to the epithelial cells and thus aids the development of respiratory infections [261].

Another mechanism of the SO2 effect is not based upon the acid activity, but rather on the reaction with cellular glutathione. Glutathione is a considerable factor towards the maintenance of the cellular redox balance. SO2 reacts with glutathione to form S-sulfo-glutathione and depletes cellular glutathione. What follows is a reduction of the activity of glutathione-dependent detoxification enzymes such as glutathione peroxidase or glutathione reductase. This reduces the cellular resistance to oxidative stress [262]. The slightly increased incidence of lung cancer mortality on SO2 exposure is probably to be seen in this context [263], [264], [265].

SO2 works as an irritant on the respiratory tracts. The cause of this is the permissive effect of protons on the Vanilloid receptor on trigeminal fibres of the respiratory mucous membrane [Fig. 16]. These do not activate the receptor themselves, but result in a considerable increase in its sensitivity to numerous stimuli. In this way the receptors, under the influence of protons, are already activated at a temperature of 37°C [266]. Basic mechanisms of irritation of the mucous membrane are more closely described with the volatile organic compounds.

Nitric oxides are predominantly present in the environment in the form of NO2. Nitrogen monoxide reacts in the presence of oxygen in the air to form NO2. It is moderately water-soluble and reaches the lower respiratory tracts at about 50%. Nitric acid is formed in the water of the respiratory secretion following the reaction Equation 2 . In a carefully planned study, it was discovered that the incidence of atopic disorders in childhood does correlate with the NO2 content of ambient air, but not with individual NO2 exposure [212]. NO2 thus serves solely as an indicator substance for traffic congestion. With people it is only high (2000 µg/m3) NO2 concentrations that lead to inflammatory changes in the mucous membrane of the nose [267], whilst with persons suffering from an allergy, significantly lower NO2 concentrations can considerably enhance the allergic response [268], [269].

4.7. Ozone

Ozone is a strong oxidation agent. In urban agglomeration areas it occurs in toxic concentrations with UV light. Ozone is extremely reactive and is retained by reducing substances of respiratory secretion. Surfactant in respiratory secretion contains numerous unsaturated fatty acids that react with ozone to create more stable aldehydes, hydroxyhydroperoxides and endoperoxides. These lipid ozonation products, as well as small amounts of the inhaled ozone itself, penetrate through the respiratory secretion reaching the cell membrane of the cells in the respiratory tract [270], [271]. In the membrane of these cells, ozone and the still reactive lipid ozonation products that developed in the secretion lead in turn to the peroxidation of unsaturated fatty acids, whereby a chain reaction of lipid oxidations can occur. The reactive ozonation products of the membrane lipids [Fig. 17] place the cell under oxidative stress [272]. Numerous further reactive compounds occur including nonanal, hexanal [273], [274] and F2 isoprostane. This substances resembles prostaglandin with similar proinflammatory effects. It also is a good marker of the in vivo lipid peroxidation [275], [276]. As well as this, membrane proteins are denatured. Activation of the SAPK pathway follows with activation of phospholipase A2, C and D, of PI-3 kinase, protein kinase B, NF-κB, STAT-1, upregulation of the inducible NO synthase, to the formation of NO and the release of IL-8 and TNFα from macrophages, neutrophils and respiratory epithelia [272], [277], [278], [279], [280].

After a few hours, the release of the chemokine IL-8 results in airway neutrophilia, the prominent feature in nasal cytologies after ozone exposure. The neutrophil granulocytes increase the epithelial damage induced by ozone [281]. In addition, ozone leads in vitro to the release of neuropeptides from nasal mucous membrane [282]. Mucous membrane irritation is probably not attributable to ozone itself, but rather to ozone oxidation products such as formaldehydes, peroxyacetyl nitrate, peroxybenzol nitrate, acrolein or, for example, ozonation products from terpenes [283].

Long-term or repeated ozone exposure leads to epithelial desquamation, probably as a result of an increased production of the interstitial substance tenascin C [284]. A mucoid degeneration of the mucous membrane with disturbances of mucociliary transport occurs, as well as a reactive proliferation of epithelia [285], [286], [287], [288], [289], [290]. In addition to a proliferation of respiratory epithelia, DNA damage was detected that is typical for ROS [291].

4.8. Aldehydes and volatile organic compounds

Aldehydes and volatile organic compounds are predominantly present indoor. Up to 50 different substances are frequently detectable in a concentration range of 1 µg/m3 each. Indoor formaldehyde frequently reaches concentrations above 50 µg/m3 [292], [293]. Toxicologically, their irritant effect on eyes and the respiratory tract are in the foreground. The Sensory Irritation Test can quantify the irritative effect of a substance [294]. A detailed list of the irritative properties of a variety of substances can be found in Bos et.al. [295]. One of the strongest known irritant agents of relevance for industrial medicine is toluol-2•4-diisocyanate (2•4-TDI). It was recently suggested that predominantly trigeminally transmitted nasal sensory irritants be separated from predominantly vagally transmitted pulmonary irritants. Nasal irritants typically result in an extended breathing rest with delayed expiration, whilst pulmonary irritants result in a breathing rest with delayed inspiration [296]. Whether we are really dealing here with characteristic substance features and receptor interactions, or whether, for example via differing solubility, different respiratory compartments are affected by the same mechanism, is at present not clear.

There are several new findings on the mechanisms of the irritant effect of chemical agents in the upper respiratory tracts and how they lead to neurogenic inflammation and hyperreactivity [297], [298], [299]. Via the respiratory secretion, water-soluble irritant agents gain access to the nerve endings of non-myelinated sensitive nerve fibres whose cell bodies lie in the trigeminal ganglion [300], [301]. Here, numerous irritant agents bind to a non-selective receptor of the axon membrane, the Vanilloid receptor of subtype 1 (VR 1) [302]. Dusts and ozone also activate VR 1 receptors [282], [303], [304]. This can take place anywhere in the nerve fibres pathway, not only at its end point. With the attachment of the ligands to the Vanilloid receptor, a receptor-operated ion channel opens and an influx of Ca2+ und Na+ takes place [Fig. 18]. This leads to membrane depolarisation and to the discharge of a nerve action potential through stress-dependent Na+ channels in the membrane of the nerve fibres. In the trigeminal ganglion, SP and glutamate are released that transmit the irritant conduction towards the centre. In the mesencephalon, protective reflexes such as sneezing or coughing are triggered. However, nerve action potentials does not only run in an orthodromic direction towards the centre, but also in an antidromic line into the periphery where tachykinins such as substance P (SP), neurokinin A or B are released at nerve endings. These tachykinins have proinflammatory effects and project a neurogenic inflammatory reaction of the tissue. They lead to vascular dilation, oedema formation, hypersecretion and to an inflammatory cell influx [301], [305], [306].

SP prefers the NK1 receptor, neurokinin A the NK2 receptor and neurokinin B the NK3 receptor. In the mucous membrane of the upper respiratory tracts, changes induced by tachykinins are projected for the most part via the NK1 receptor [307]. Predominantly, the NK2 receptor causes a bronchial constriction, the NK3 receptor has not yet been positively detected in human respiratory tracts [308]. The NK1 receptor belongs to the G protein-coupled receptors. Ligand binding leads to the activation of phospholipase C and adenylatcyclase [309] that leads to the contraction of smooth muscle cells of the vessel wall. The vasodilative effect thus results not through the effect on the smooth muscle in the vessel wall itself, but rather via the endothelium. In contrast to the smooth vascular muscle cells, the endothelial cells express the NK1 receptor. If these are activated, the endothelial cells then release prostacycline and NO, which secondarily leads to the relaxation of vessel smooth muscle cells (see background information nitrogen monoxide) [310], [311]. At the same time, the activation of the endothelial NK1 receptor leads to contraction of the endothelial cells. Gaps emerge in the vessel wall through which plasma proteins and water can pass over into the tissue [312]. This results in an inflammatory oedema. Glandular hypersecretion is conveyed predominantly by the NK1 receptors onto glandular cells [308].

The neurokinin system and the immune system have a brisk, two-way relationship. Via the NK1 receptor, SP has a predominantly chemotactic effect on neutrophils [313], [314], [315], [316], [317], [318], [319], it enhances their ROS production [320] and inhibits their apoptosis [321]. SP also has a chemotactic effect on T lymphocytes [322], monocytes [322] and dendritic cells [323]. Eosinophils are directly attracted by SP [314] or after previous peptide activation [324]. Secretoneurin, which is released into the respiratory tracts by nociceptive fibres, also has distinctive eosinophil-chemotactic features [325]; it was found in considerable amounts in nasal irrigation and in nasal mucous membrane biopsies of people suffering from allergies [326]. The NK1 receptor is only expressed on mast cells after preliminary treatment with IL-4 or SCF [327]. After this type of priming, however, SP can also bring about a release of histamine [328].

Longer-term exposition to irritants and inflammatory irritants, but also uniquely extreme exposition, can lead to a persistent hyperreactivity of the respiratory tracts [329], [330], [331], [332], [333]. After inhalation of sulphurous acids, guinea pigs develop a respiratory hyperreactivity lasting several weeks. It can be inhibited by a NK1 receptor antagonist [334]. Changes in the nociceptive system of the respiratory tracts form the basis of this persistent hyperreactivity, whereby the density of trigeminal group C fibres in the mucous membrane of respiratory tract apparently increases. Exposition with TDI [60 ppb] over 2 h results within 24 hours in an increased density of trigeminal group C fibres in the nasal mucous membrane, as well as an increase in SP and preprotachykinin in these fibres [300], [335]. Very similar changes take place after exposure to asphalt fumes [336].

The increased density of nociceptive fibres in the mucous membrane after exposure to irritants can probably be traced back to the release of nerve growth factors. NGF leads to the sprouting of nociceptive fibres [337], [338] and consecutively to hyperreactivity of the respiratory tract [339], [340]. As well as a short-term rise in SP after nasal irritation [341], an increased level of the nerve growth factor was measurable in the nasal lavage [335], [342]. As well as nerve fibres themselves, macrophages and epithelial cells contribute to the release of NGF [343], [344]. The signal transduction probably runs via protein kinase C and ERK1/2 [345], [346]. Transgenic mice that release NGF from mucous-producing bronchial epithelia (Clara cells) develop a marked bronchial hyperreactivity [347]. Transgenic mice without the NGF-p75 gene exhibit disturbances in the nerve fibres containing tachykinins and react less to sensory irritants [348]. With these animal models, the significance of nerve fibres containing tachykinins for the proinflammatory effect of ozone was examined. It was shown that the ozone-induced inflammatory reactions with animals deficient in tachykinins was considerable less common than was the case with normal animals, and proceeded considerably more strongly with animals with an overproduction of tachykinins. This would indicate that the irritant effect of ozone or its reaction products contributes significant to its inflammatory effect [349].

An important protective and regulatory function of the respiratory secretion is to be found in the rapid breakdown of biologically active tachykinins through varying amino peptides such as neutral endopeptidase (NEP), angiotensin converting enzyme (ACE) or dipeptidylpeptidase IV (DPPIV) [41]. A result of a lack of these enzymes that break down tachykinins in secretion can contribute greatly to the emergence of respiratory hyperreactivity [42], [350]. The value of oral neurokinin receptor inhibitors must first be proved [351], [352]. As well as their irritative effect, volatile organic compounds also have cytotoxic and genotixic features [81], [353] that in environmentally relevant concentrations, however, are not crucial.

4.9. Synopsis of the mechanisms of inhaled pollutants

Air pollutants lead to inflammatory changes in the mucous membrane of the respiratory tract. Mixtures of numerous toxic substances that can be assigned to several archetypes are inhaled. In the real exposure situation, these agents act in a synchronous or sequential manner and exhibit synergistic effects. Thus one can assume that in the real exposure situation all described cellular toxicity mechanisms are simultaneously activated to a greater or lesser degree.

Ozone and acid gasses such as SO2 denature molecules from the cell membrane. The denatured lipids and proteins activate an intracellular signal transaction cascade, the SAPK pathway. This leads to release of prostaglandins and leukotrienes that promote inflammation, to the activation of transcription factors such as AP-1 that activate genes encoding proteins for cell proliferation and extracellular matrix. As well as this, SAPK activate the transcription factor NF-κB that in turn activates genes for a large number of proteins with proinflammatory effects.

Metals are dissolved from inhaled dust particles and channelled into the cell by metal transporters. Here they bring about the formation of reactive oxygen species and nitrogen monoxide, that is to say highly reactive radicals. For their part, these then directly activate the SAPK pathway and also NF-κB via partly still unexplained mechanisms.

The indicator substance for bioorganic pollutants, mainly fragments of bacteria and fungi, is endotoxin. They bind to toll-like/IL-1 (TIL) receptors. This leads to the activation of NF-κB via intracellular signal molecules. As well as this, TIL receptors convey the phagocytosis of the particles with adherent endotoxins. This leads via the oxidative burst to the release of ROS and NO. Endotoxins also have positive features. They shape the TH1 -preferred immune response and thus work against the emergence of allergies of the respiratory tracts [136].

Organic compounds from combustion processes, for example polycyclic aromatic hydrocarbons (PAH), are lipophilic. They can penetrate the cell membrane and bind to the cytosolic arylhydrocarbon receptor. This moves into the cell nucleus and activates genes for detoxifications enzymes, but also encourages a proliferation and proinflammatory effect via interaction with NF-κB.

Irritant agents often belong chemically to aldehydes and volatile organic compounds. They release substance P (SP) from the nociceptors of the respiratory mucous membrane. SP has a high affinity interaction with the NK1 receptor. Subsequently, the respiratory cell releases nerve growth factor (NGF) that increases the nociceptor density in the respiratory mucous membrane. This contributes to the development of mucous membrane hyperreactivity. The intracellular signal transduction from the NK1 receptor to the activation of the gene for NGF is not conclusively explained, but there is a great deal of evidence that protein kinase C is involved. In addition, the transcription factor C-fos is apparently activated via the ERK1/2 pathway . As well as this, protein kinase C activates the transcription factor NF-κB after the coupling of SP to the NK1 receptor.

[Fig. 19]

These mechanisms induce a chronic neutrophil-skewed inflammatory reaction in the mucous membrane of the upper respiratory tracts. These inflammatory reactions are detectable in practically every inhabitant of western industrial nations and were termed "physiological inflammation" by Niels Mygind, one of the pioneers of modern nasal mucous membrane research [354]. This implies that the steady exposition to toxic substances is providential and must be accepted as physiological. It is at present unclear to what extent ubiquitous nasal exposition to pollutants contributes to the fact that ca. 15% of the population of western industrial nations suffer from chronic disorders of the nose and sinuses [355].


References

1.
von-Mutius E, Sherrill DL, Fritzsch C, Martinez FD, Lebowitz MD. Air pollution and upper respiratory symptoms in children from East Germany. Eur Respir J 1995; 8: 723-8.
2.
Calderon-Garciduenas L, Rodriguez-Alcaraz A, Villarreal-Calderon A, Lyght O, Janszen D, Morgan KT. Nasal epithelium as a sentinel for airborne environmental pollution. Toxicol Sci 1998; 46: 352-64.
3.
Keles N, Ilicali OC, Deger K. Impact of air pollution on prevalence of rhinitis in Istanbul. Arch Environ Health 1999; 54: 48-51.
4.
Keles N, Ilicali C. The impact of outdoor pollution on upper respiratory diseases. Rhinology 1998; 36: 24-7.
5.
Abbey DE, Hwang BL, Burchette RJ, Vancuren T, Mills PK. Estimated long-term ambient concentrations of PM10 and development of respiratory symptoms in a nonsmoking population. Arch Environ Health 1995; 50: 139-52.
6.
Pope CA, Bates D.V., Raizenne M.E. Health effects of particulate air pollution: time for reassessment? Environ Health Pers 1995; 103: 472-780.
7.
Hajat S, Anderson HR, Atkinson RW, Haines A. Effects of air pollution on general practitioner consultations for upper respiratory diseases in London. Occup Environ Med 2002; 59: 294-9.
8.
Jaakkola JJ, Paunio M, Virtanen M, Heinonen OP. Low-level air pollution and upper respiratory infections in children. Am J Public Health 1991; 81: 1060-1063.
9.
Gordian ME, Ozkaynak H, Xue J, Morris SS, Spengler JD. Particulate air pollution and respiratory disease in Anchorage, Alaska. Environ Health Perspect 1996; 104: 290-297.
10.
Ostro BD, Eskeland GS, Sanchez JM, Feyzioglu T. Air pollution and health effects: A study of medical visits among children in Santiago, Chile. Environ Health Perspect 1999; 107: 69-73.
11.
van der Zee SC, Hoek G, Boezen MH, Schouten JP, van Wijnen JH, Brunekreef B. Acute effects of air pollution on respiratory health of 50-70 yr old adults. Eur Respir J 2000; 15: 700-709.
12.
Riechelmann H. Berufliche Verursachung und Bewertung von Mundhöhlen- und Rachentumoren. Laryngorhinootologie 2002; 81: 573-579.
13.
Riechelmann H. Umweltmedizin in der HNO-Heilkunde. Laryngorhinootologie 2000; 79: 100-27.
14.
Oehlmann J, Markert B. Humantoxikologie. Stuttgart: Wissenschaftliche Verlagsgesellschaft mbH, 1997.
15.
Römpp Chemie Lexikon. 9. Auflage ed. Stuttgart, New York: Georg Thieme Verlag, 1995.
16.
Medinsky MA, Bond JA. Sites and mechanisms for uptake of gases and vapors in the respiratory tract. Toxicology 2001; 160: 165-172.
17.
Samet J.M. The relationship between acute and chronic health effects of air pollution. In: Bates D.V., Brain JD, Driscoll KE, Dungworth DL, Grafström R, Harris CC et al., (Hrsg.). Relationships between acute and chronic effects of air pollution. Washington: ILSI Press, 2000: 11-21.
18.
Deutsche Forschungsgemeinschaft: Senatskommission zur Prüfung gesundheitsschädlicher Arbeitsstoffe. MAK- und BAT-Werte-Liste. Weinheim: VCH Verlagsgesellschaft mbH, 1996.
19.
Jarabek AM. Inhalation RfC methodology: dosimetric adjustments and dose-response estimation of noncancer toxicity in the upper respiratory tract. In: Miller FJ (ed.): Nasal toxicity and dosimetry of inhaled xenobiotics. Taylor and Francis, Washington 1995; 301-325.
20.
Kaliner MA, Shelhamer JH, Borson DB, Patow CA, Nadel JA, Marom Z. Resiratory mucus. In: Kaliner MA, Barnes PJ, (Hrsg.). The Airways, neural control in health and disease. New York: Dekker, 1988: 575-593.
21.
Lucas AM, Douglas LC. Principles underlying ciliary activity in the respiratory tract.II.A comparison of nasal clearance in man, monkey and other mammals. Arch of Otolaryngology 1934; 20: 518-541.
22.
Nadel JA, Widdicombe JH, Peatfield AC. Regulation of airway secretion, ion transport, and water movement. In: Fischmann AP, (Hrsg.). The Respiratory System. 1985: 419-45.
23.
Lamblin G, Aubert JP, Perini JM, Klein A, Porchet N, Degand P et al. Human respiratory mucins. Eur Respir J 1992; 5: 247-256.
24.
Kim KC, Hisatsune A, Kim dJ, Miyata T. Pharmacology of airway goblet cell mucin release. J Pharmacol Sci 2003; 92: 301-307.
25.
Sharma P, Dudus L, Nielsen PA, Clausen H, Yankaskas JR, Hollingsworth MA et al. MUC5B and MUC7 are differentially expressed in mucous and serous cells of submucosal glands in human bronchial airways. Am J Respir Cell Mol Biol 1998; 19: 30-37.
26.
Reid CJ, Gould S, Harris A. Developmental expression of mucin genes in the human respiratory tract. Am J Respir Cell and Molecular Biol 1997; 17: 592-598.
27.
Gray T, Koo JS, Nettesheim P. Regulation of mucous differentiation and mucin gene expression in the tracheobronchial epithelium. Toxicology 2001; 160: 35-46.
28.
Carraway KL, Ramsauer VP, Haq B, Carothers Carraway CA. Cell signaling through membrane mucins. Bioessays 2003; 25: 66-71.
29.
Basbaum C, Lemjabbar H, Longphre M, Li D, Gensch E, McNamara N. Control of mucin transcription by diverse injury-induced signaling pathways. Am J Respir Crit Care Med 1999; 160: S44-S48.
30.
Basbaum C, Li D, Gensch E, Gallup M, Lemjabbar H. Mechanisms by which gram-positive bacteria and tobacco smoke stimulate mucin induction through the epidermal growth factor receptor (EGFR). Novartis Found Symp 2002; 248: 171-176.
31.
Rose MC, Piazza FM, Chen YA, Alimam MZ, Bautista MV, Letwin N et al. Model systems for investigating mucin gene expression in airway diseases. J Aerosol Med 2000; 13: 245-261.
32.
Holma B. Influence of buffer capacity and pH-dependant rheological properties of respiratory mucus on health effects due to acid pollution. Sci Tot Environ 1985; 41: 101-123.
33.
Holma B. Effects of inhaled acids on airway mucus and its consequences for health. Environ Health Persp 1989; 79: 109-113.
34.
Buhrmester CC. A study of the hydrogen-ion concentration, nitrogen content and viscosity of nasal secretions. Ann Oto Rhino Laryngol 1933; 42: 1041-57.
35.
Stierna P, Soederlund K, Hultman E. Chronic maxillary sinusitis. Energy metabolism in sinus mucosa and secretion. Acta Otolaryngol 1991; 111: 135-43.
36.
van de Donk H, Zuidema J, Merkus F. The influence of the pH and osmotic pressure upon tracheal ciliary beat frequency as determined with a new photo-electric registration device. Rhinology 1980; 18: 93-104.
37.
Luk CK, Dulfano MJ. Effect of pH, viscosity, and ionic strength on ciliary beating frequency of human bronchial explants. Am Rev Resp Dis Suppl 1982; 4: 244.
38.
Keck T, Leiacker R, Klotz M, Lindemann J, Riechelmann H, Rettinger G. Detection of particles within the nasal airways during respiration. Eur Arch Otorhinolaryngol 2000; 257: 493-7.
39.
Boland S, Baeza-Squiban A, Fournier T, Houcine O, Gendron MC, Chevrier M et al. Diesel exhaust particles are taken up by human airway epithelial cells in vitro and alter cytokine production. Am J Physiol 1999; 276: 604-13.
40.
Kennedy T, Ghio AJ, Reed W, Samet J, Zagorski J, Quay J et al. Copper-dependent inflammation and nuclear factor-kappaB activation by particulate air pollution. Am J Respir Cell Mol Biol 1998; 19: 366-378.
41.
Ohkubo K, Baraniuk JN, Hohman R, Merida M, Hersh LB, Kaliner MA. Aminopeptidase activity in human nasal mucosa. J Allergy Clin Immunol 1998; 102: 741-50.
42.
Grouzmann E, Monod M, Landis B, Wilk S, Brakch N, Nicoucar K et al. Loss of dipeptidylpeptidase IV activity in chronic rhinosinusitis contributes to the neurogenic inflammation induced by substance P in the nasal mucosa. Faseb J 2002; 16: 1132-1134.
43.
van der Vliet A, O'Neill CA, Cross CE, Koostra JM, Volz WG, Halliwell B et al. Determination of low-molecular-mass antioxidant concentrations in human respiratory tract lining fluids. Am J Physiol 1999; 276: L289-L296.
44.
Pacht ER, Davis WB. Role of transferrin and ceruloplasmin in antioxidant activity of lung epithelial lining fluid. J Appl Physiol 1988; 64: 2092-2099.
45.
Blomberg A, Sainsbury C, Rudell B, Frew AJ, Holgate ST, Sandstrom T et al. Nasal cavity lining fluid ascorbic acid concentration increases in healthy human volunteers following short term exposure to diesel exhaust. Free Radic Res 1998; 28: 59-67.
46.
Ghio AJ, Carter JD, Dailey LA, Devlin RB, Samet JM. Respiratory epithelial cells demonstrate lactoferrin receptors that increase after metal exposure. Am J Physiol 1999; 276: L933-L940.
47.
Elsbach P, Weiss J, Levy O. Oxygen-independent antimicrobial systems of phagocytes. In: Gallin JI, Snyderman R, (Hrsg.). Inflammation basic principles and clinical correlates. Philadelphia, PA: Lippincott Williams & Wilkins, 1999: 801-817.
48.
Wang X, Zhang Z, Louboutin JP, Moser C, Weiner DJ, Wilson JM. Airway epithelia regulate expression of human ß-defensin 2 through Toll-like receptor 2. Faseb J 2003; 17: 1727-1729.
49.
van Wetering S, Sterk PJ, Rabe KF, Hiemstra PS. Defensins: key players or bystanders in infection, injury, and repair in the lung? J Allergy Clin Immunol 1999; 104: 1131-8.
50.
Harder J, Meyer-Hoffert U, Teran LM, Schwichtenberg L, Bartels J, Maune S et al. Mucoid pseudomonas aeruginosa, TNF-alpha , and IL-1beta , but Not IL-6, induce human ß -Defensin-2 in respiratory epithelia. Am J Respir Cell Mol Biol 2000; 22: 714-721.
51.
Thornton-Manning JR, Dahl AR. Metabolic capacity of nasal tissue interspecies comparisons of xenobiotic-metabolizing enzymes. Mutat Res 1997; 380: 43-59.
52.
Blanchard KT, Morris JB. Effects of m-xylene on rat nasal cytochrome P450 mixed function oxidase activities. Toxicol Lett 1994; 70: 253-259.
53.
Sarkar MA. Drug metabolism in the nasal mucosa. Pharm Res 1992; 9: 1-9.
54.
Gervasi PG, Longo V, Naldi F, Panattoni G, Ursino F. Xenobiotic-metabolizing enzymes in human respiratory nasal mucosa. Biochem Pharmacol 1991; 41: 177-184.
55.
Dahl AR, Hadley WM. Nasal cavity enzymes involved in xenobiotic metabolism: effects on the toxicity of inhalants. Crit Rev Toxicol 1991; 21: 345-372.
56.
Bogdanffy MS. Biotransformation enzymes in the rodent nasal mucosa: the value of a histochemical approach. Environ Health Perspect 1990; 85: 177-186.
57.
Brittebo EB, Castonguay A, Rafter JJ, Kowalski B, Ahlman M, Brandt I. Metabolism of xenobiotics and steroid hormones in the nasal mucosa. In: Barrow C, (Hrsg.). Toxicology of the nasal passages. Washington, New York, London: Hemisphere Publishing Corporation, 1986: 211-234.
58.
Bonvallot V, Baeza-Squiban A, Baulig A, Brulant S, Boland S, Muzeau F et al. Organic compounds from diesel exhaust particles elicit a proinflammatory response in human airway epithelial cells and induce cytochrome p450 1A1 expression. Am J Respir Cell Mol Biol 2001; 25: 515-21.
59.
Gronau S, Konig-Greger D, Rettinger G, Riechelmann H. GSTM1-Genpolymorphismus bei Patienten mit Kopf-Hals-Tumoren. Laryngorhinootologie 2000; 79: 341-4.
60.
Gronau S, Koenig-Greger D, Jerg M, Riechelmann H. Gene polymorphisms in detoxification enzymes as susceptibility factor for head and neck cancer? Otolaryngol Head Neck Surg 2003; 128: 674-680.
61.
Gronau S, Koenig GD, Jerg M, Riechelmann H. 11Beta-hydroxysteroid dehydrogenase 1 expression in squamous cell carcinomas of the head and neck. Clin Otolaryngol 2002; 27: 453-457.
62.
Ding X, Kaminsky LS. Human extrahepatic cytochromes P450: Function in Xenobiotic Metabolism and Tissue-Selective Chemical Toxicity in the Respiratory and Gastrointestinal Tracts. Annu Rev Pharmacol Toxicol 2003; 43: 149-173.
63.
Willey JC, Crawford JT, Olson G, Hammersley JE, Jenning CA, Weaver L et al. Expression measurement of genes related to cancer susceptibility in human bronchial epithelial cells. In: Bates D.V., Brain JD, Driscoll KE, Dungworth DL, Grafström R, Harris CC et al., (Hrsg.). Relationships between acute and chronic effects of air pollution. Washington: ILSI Press, 2000: 79-96.
64.
Wanner A. Allergic mucociliary dysfunction. Laryngoscope 1983; 93: 68-70.
65.
Scott G, Leopardi S, Parker L, Babiarz L, Seiberg M, Han R. The proteinase-activated receptor-2 mediates phagocytosis in a Rho-dependent manner in human keratinocytes. J Invest Dermatol 2003; 121: 529-541.
66.
Kato T, Yashiro T, Murata Y, Herbert DC, Oshikawa K, Bando M et al. Evidence that exogenous substances can be phagocytized by alveolar epithelial cells and transported into blood capillaries. Cell Tissue Res 2003; 311: 47-51.
67.
Travaglione S, Falzano L, Fabbri A, Stringaro A, Fais S, Fiorentini C. Epithelial cells and expression of the phagocytic marker CD68: scavenging of apoptotic bodies following Rho activation. Toxicol In Vitro 2002; 16: 405-411.
68.
Godleski J.J. Cardiovascular responses to inhaled particles. In: Bates D.V., Brain JD, Driscoll KE, Dungworth DL, Grafström R, Harris CC et al., (Hrsg.). Relationships between acute and chronic effects of air pollution. Washington: ILSI Press, 2000: 142-155.
69.
Pope CA3, Burnett RT, Thun MJ, Calle EE, Krewski D, Ito K et al. Lung cancer, cardiopulmonary mortality, and long-term exposure to fine particulate air pollution. Jama 2002; 287: 1132-41.
70.
Johnson T, Devli R, Ghio AJ, Huang YC, Costa DL, Engle C et al. Cardiopulmonary effects of nebulized residual oil fly ash in anesthetized pigs. In: Bates D.V., Driscoll KE, Dungworth DL, Fabel H, Grafström R, Harkema JR et al., (Hrsg.). Crucial issues in inhalation research - mechanistic, clinical and epidemiologic. Stuttgart: Fraunhofer IRB Verlag, 2002: 199-212.
71.
Bohm GM, Saldiva PH, Pasqualucci CA, Massad E, Martins M, Zin WA et al. Biological effects of air pollution in Sao Paulo and Cubatao. Environ Res 1989; 49: 208-16.
72.
Calderon-Garciduenas L, Rodriguez-Alcaraz A, Garcia R, Sanchez G, Barragan G, Camacho R et al. Human nasal mucosal changes after exposure to urban pollution. Environ Health Perspect 1994; 102: 1074-80.
73.
Calderon-Garciduenas L, Rodriguez-Alcaraz A, Garcia R, Ramirez L, Barragan G. Nasal inflammatory responses in children exposed to a polluted urban atmosphere. J Toxicol Environ Health 1995; 45: 427-37.
74.
Calderon-Garciduenas L, Rodriguez-Alcaraz A, Valencia-Salazar G, Mora-Tascareno A, Garcia R, Osnaya N et al. Nasal biopsies of children exposed to air pollutants. Toxicol Pathol 2001; 29: 558-64.
75.
Calderon-Garciduenas L, Valencia-Salazar G, Rodriguez-Alcaraz A, Gambling TM, Garcia R, Osnaya N et al. Ultrastructural nasal pathology in children chronically and sequentially exposed to air pollutants. Am J Respir Cell Mol Biol 2001; 24: 132-8.
76.
Kreyling WG, Dirscherl P, Ferron GA, Heilmann P, Josten M, Miaskowski U et al. Health effects of sulfur-related environmental air pollution. III. Nonspecific respiratory defense capacities. Inhal Toxicol 1999; 11: 391-422.
77.
Lastbom BL, Camner P. Human experimental studies on the effects of inhaled particles. Scand J Work Environ Health 2000; 26: 43-8.
78.
Mochca-Morales J. Nasal albumin in a population exposed for the first time to urban pollution. Arch Med Res 2000; 31: 409-14.
79.
Moss OR, Gross EA, James RA, Janszen DB, Ross PW, Roberts KC et al. Respiratory tract toxicity in rats exposed to Mexico City air. Res Rep Health Eff Inst 2001; 1-24.
80.
Saldiva PH, King M, Delmonte VL, Macchione M, Parada MA, Daliberto ML et al. Respiratory alterations due to urban air pollution: an experimental study in rats. Environ Res 1992; 57: 19-33.
81.
Shusterman D. Toxicology of nasal irritants. Curr Allergy Asthma Rep 2003; 3: 258-265.
82.
Baeza-Squiban A, Bonvallot V, Boland S, Marano F. Airborne particles evoke an inflammatory response in human airway epithelium. Activation of transcription factors. Cell Biol Toxicol 1999; 15: 375-80.
83.
Calderon-Garciduenas L, Devlin RB, Miller FJ. Respiratory tract pathology and cytokine imbalance in clinically healthy children chronically and sequentially exposed to air pollutants. Med Hypotheses 2000; 55: 373-8.
84.
Carter JD, Ghio AJ, Samet JM, Devlin RB. Cytokine production by human airway epithelial cells after exposure to an air pollution particle is metal-dependent. Toxicol Appl Pharmacol 1997; 146: 180-8.
85.
Devalia JL, Bayram H, Rusznak C, Calderon M, Sapsford RJ, Abdelaziz MA et al. Mechanisms of pollution-induced airway disease: in vitro studies in the upper and lower airways. Allergy 1997; 52: 45-51.
86.
Douwes J, Wouters I, Dubbeld H, van Zwieten L, Steerenberg P, Doekes G et al. Upper airway inflammation assessed by nasal lavage in compost workers: A relation with bio-aerosol exposure. Am J Ind Med 2000; 37: 459-68.
87.
Fujii T, Hayashi S, Hogg JC, Vincent R, van Eeden SF. Particulate matter induces cytokine expression in human bronchial epithelial cells. Am J Respir Cell Mol Biol 2001; 25: 265-271.
88.
Hashimoto S, Gon Y, Takeshita I, Matsumoto K, Jibiki I, Takizawa H et al. Diesel exhaust particles activate p38 MAP kinase to produce interleukin 8 and RANTES by human bronchial epithelial cells and N-acetylcysteine attenuates p38 MAP kinase activation. Am J Respir Crit Care Med 2000; 161: 280-285.
89.
Hirvonen MR, Ruotsalainen M, Roponen M, Hyvarinen A, Husman T, Kosma VM et al. Nitric oxide and proinflammatory cytokines in nasal lavage fluid associated with symptoms and exposure to moldy building microbes. Am J Respir Crit Care Med 1999; 160: 1943-6.
90.
Jimenez LA, Drost EM, Gilmour PS, Rahman I, Antonicelli F, Ritchie H et al. PM(10)-exposed macrophages stimulate a proinflammatory response in lung epithelial cells via TNF-alpha. Am J Physiol Lung Cell Mol Physiol 2002; 282: L237-L248.
91.
Krishna MT, Chauhan AJ, Frew AJ, Holgate ST. Toxicological mechanisms underlying oxidant pollutant-induced airway injury. Rev Environ Health 1998; 13: 59-71.
92.
Long CM, Suh HH, Kobzik L, Catalano PJ, Ning YY, Koutrakis P. A pilot investigation of the relative toxicity of indoor and outdoor fine particles: in vitro effects of endotoxin and other particulate properties. Environ Health Perspect 2001; 109: 1019-26.
93.
Mills PR, Davies RJ, Devalia JL. Airway epithelial cells, cytokines, and pollutants. Am J Respir Crit Care Med 1999; 160: 38-43.
94.
Monn C, Becker S. Cytotoxicity and induction of proinflammatory cytokines from human monocytes exposed to fine (PM2.5) and coarse particles (PM10-2.5) in outdoor and indoor air. Toxicol Appl Pharmacol 1999; 155: 245-52.
95.
Monn C, Naef R, Koller T. Reactions of macrophages exposed to particles <10 microm. Environ Res 2003; 91: 35-44.
96.
Ohtoshi T, Takizawa H, Okazaki H, Kawasaki S, Takeuchi N, Ohta K et al. Diesel exhaust particles stimulate human airway epithelial cells to produce cytokines relevant to airway inflammation in vitro. J Allergy Clin Immunol 1998; 101: 778-85.
97.
Pei XH, Nakanishi Y, Inoue H, Takayama K, Bai F, Hara N. Polycyclic aromatic hydrocarbons induce IL-8 expression through nuclear factor kappaB activation in A549 cell line. Cytokine 2002; 19: 236-241.
98.
Purokivi MK, Hirvonen MR, Randell JT, Roponen MH, Meklin TM, Nevalainen AL et al. Changes in pro-inflammatory cytokines in association with exposure to moisture-damaged building microbes. Eur Respir J 2001; 18: 951-8.
99.
Quay JL, Reed W, Samet J, Devlin RB. Air pollution particles induce IL-6 gene expression in human airway epithelial cells via NF-kappaB activation. Am J Respir Cell Mol Biol 1998; 19: 98-106.
100.
Saraf A, Larsson L, Larsson BM, Larsson K, Palmberg L. House dust induces IL-6 and IL-8 response in A549 epithelial cells. Indoor Air 1999; 9: 219-25.
101.
Schierhorn K, Zhang M, Matthias C, Kunkel G. Influence of ozone and nitrogen dioxide on histamine and interleukin formation in a human nasal mucosa culture system. Am J Respir Cell Mol Biol 1999; 20: 1013-9.
102.
Shukla A, Timblin C, BeruBe K, Gordon T, McKinney W, Driscoll K et al. Inhaled particulate matter causes expression of nuclear factor (NF)-kappaB-related genes and oxidant-dependent NF-kappaB activation in vitro. Am J Respir Cell Mol Biol 2000; 23: 182-187.
103.
Steerenberg PA, Fischer PH, van Bree L, van Loveren H. Nasal lavage biomarkers in air pollution epidemiology. Arch Toxicol Suppl 1997; 19: 207-16.
104.
Takizawa H, Ohtoshi T, Kawasaki S, Abe S, Sugawara I, Nakahara K et al. Diesel exhaust particles activate human bronchial epithelial cells to express inflammatory mediators in the airways: a review. Respirology 2000; 5: 197-203.
105.
Tao F, Kobzik L. Lung macrophage-epithelial cell interactions amplify particle-mediated cytokine release. Am J Respir Cell Mol Biol 2002; 26: 499-505.
106.
van Eeden SF, Tan WC, Suwa T, Mukae H, Terashima T, Fujii T et al. Cytokines involved in the systemic inflammatory response induced by exposure to particulate matter air pollutants (PM(10)). Am J Respir Crit Care Med 2001; 164: 826-830.
107.
Veronesi B, Oortgiesen M, Carter JD, Devlin RB. Particulate matter initiates inflammatory cytokine release by activation of capsaicin and acid receptors in a human bronchial epithelial cell line. Toxicol Appl Pharmacol 1999; 154: 106-15.
108.
Griego SD, Weston CB, Adams JL, Tal-Singer R, Dillon SB. Role of p38 mitogen-activated protein kinase in rhinovirus-induced cytokine production by bronchial epithelial cells. J Immunol 2000; 165: 5211-20.
109.
Altman GB, Altman LC, Luchtel DL, Jabbour AJ, Baker C. Release of RANTES from nasal and bronchial epithelial cells. Cell Biol Toxicol 1997; 13: 205-213.
110.
Bachert C. Die Schleimhaut der oberen Atemwege - Zur Pathophysiologie der Entzündung. Oto-Rhino-Laryngology 1995; Suppl. I: 155-220.
111.
Monod J, Wyman J, Changeux JP. On the nature of allosteric transitions: a plausible model. J Mol Biol 1965; 12: 88-118.
112.
Zhang Y, Reinberg D. Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes Dev 2001; 15: 2343-2360.
113.
Stamler JS, Lamas S, Fang FC. Nitrosylation. The prototypic redox-based signaling mechanism. Cell 2001; 106: 675-683.
114.
Park HS, Huh SH, Kim MS, Lee SH, Choi EJ. Nitric oxide negatively regulates c-Jun N-terminal kinase/stress-activated protein kinase by means of S-nitrosylation. Proc Natl Acad Sci U S A 2000; 97: 14382-14387.
115.
Kim SO, Merchant K, Nudelman R, Beyer WF Jr, Keng T, DeAngelo J et al. OxyR: a molecular code for redox-related signaling. Cell 2002; 109: 383-396.
116.
Huang SL, Cheng WL, Lee CT, Huang HC, Chan CC. Contribution of endotoxin in macrophage cytokine response to ambient particles in vitro. J Toxicol Environ Health A 2002; 65: 1261-1272.
117.
Michel O, Nagy AM, Schroeven M, Duchateau J, Neve J, Fondu P et al. Dose-response relationship to inhaled endotoxin in normal subjects. Am J Respir Crit Care Med 1997; 156: 1157-1164.
118.
Nightingale JA, Rogers DF, Hart LA, Kharitonov SA, Chung KF, Barnes PJ. Effect of inhaled endotoxin on induced sputum in normal, atopic, and atopic asthmatic subjects. Thorax 1998; 53: 563-571.
119.
Sandstrom T, Bjermer L, Rylander R. Lipopolysaccharide (LPS) inhalation in healthy subjects increases neutrophils, lymphocytes and fibronectin levels in bronchoalveolar lavage fluid. Eur Respir J 1992; 5: 992-996.
120.
Danuser B, Rebsamen H, Weber C, Krueger H. Lipopolysaccharide-induced nasal cytokine response: a dose-response evaluation. Eur Arch Otorhinolaryngol 2000; 257: 527-532.
121.
Sigsgaard T, Bonefeld-Jorgensen EC, Kjaergaard SK, Mamas S, Pedersen OF. Cytokine release from the nasal mucosa and whole blood after experimental exposures to organic dusts. Eur Respir J 2000; 16: 140-5.
122.
Peden DB, Tucker K, Murphy P, Newlin-Clapp L, Boehlecke B, Hazucha M et al. Eosinophil influx to the nasal airway after local, low-level LPS challenge in humans. J Allergy Clin Immunol 1999; 104: 388-94.
123.
von-Mutius E, Braun-Fahrlander C, Schierl R, Riedler J, Ehlermann S, Maisch S et al. Exposure to endotoxin or other bacterial components might protect against the development of atopy. Clin Exp Allergy 2000; 30: 1230-4.
124.
Myatt T, Milton D. Endotoxins. In: Spengler JD, Samet JM, McCarthy JF, (Hrsg.). . New York: McGraw-Hill, 2001: 42.1-42.14.
125.
Teeuw KB, Vandenbroucke-Grauls CM, Verhoef J. Airborne gram-negative bacteria and endotoxin in sick building syndrome. A study in Dutch governmental office buildings. Arch Intern Med 1994; 154: 2339-2345.
126.
Nowak D. Klinisch-experimentelle und epidemiologische Untersuchungen zur Wirkung irritativer Berufs- und Umweltnoxen auf den Atemtrakt. Regensburg: S. Roderer Verlag, 1997.
127.
Larsson BM, Larsson K, Malmberg P, Martensson L, Palmberg L. Airway responses in naive subjects to exposure in poultry houses: comparison between cage rearing system and alternative rearing system for laying hens. Am J Ind Med 1999; 35: 142-9.
128.
Laitinen S, Linnainmaa M, Laitinen J, Kiviranta H, Reiman M, Liesivuori J. Endotoxins and IgG antibodies as indicators of occupational exposure to the microbial contaminants of metal-working fluids. Int Arch Occup Environ Health 1999; 72: 443-450.
129.
Becker S, Soukup JM, Gilmour MI, Devlin RB. Stimulation of human and rat alveolar macrophages by urban air particulates: effects on oxidant radical generation and cytokine production. Toxicol Appl Pharmacol 1996; 141: 637-48.
130.
Becker S, Fenton MJ, Soukup JM. Involvement of microbial components and toll-like receptors 2 and 4 in cytokine responses to air pollution particles. Am J Respir Cell Mol Biol 2002; 27: 611-618.
131.
Douwes J, Zuidhof A, Doekes G, van der Zee SC, Wouters I, Boezen MH et al. (1-->3)-beta-D-glucan and endotoxin in house dust and peak flow variability in children. Am J Respir Crit Care Med 2000; 162: 1348-1354.
132.
Fogelmark B, Goto H, Yuasa K, Marchat B, Rylander R. Acute pulmonary toxicity of inhaled beta-1,3-glucan and endotoxin. Agents Actions 1992; 35: 50-6.
133.
Gehring U, Douwes J, Doekes G, Koch A, Bischof W, Fahlbusch B et al. Beta(1-->3)-glucan in house dust of German homes: housing characteristics, occupant behavior, and relations with endotoxins, allergens, and molds. Environ Health Perspect 2001; 109: 139-44.
134.
Imrich A, Ning Y, Kobzik L. Insoluble components of concentrated air particles mediate alveolar macrophage responses in vitro. Toxicol Appl Pharmacol 2000; 167: 140-150.
135.
Khair OA, Davies RJ, Devalia JL. Bacterial-induced release of inflammatory mediators by bronchial epithelial cells. Eur Respir J 1996; 9: 1913-22.
136.
Liu AH. Endotoxin exposure in allergy and asthma: reconciling a paradox. J Allergy Clin Immunol 2002; 109: 379-392.
137.
Long CM, Suh HH, Kobzik L, Catalano PJ, Ning YY, Koutrakis P. A pilot investigation of the relative toxicity of indoor and outdoor fine particles: in vitro effects of endotoxin and other particulate properties. Environ Health Perspect 2001; 109: 1019-1026.
138.
Ning Y, Imrich A, Goldsmith CA, Qin G, Kobzik L. Alveolar macrophage cytokine production in response to air particles in vitro: role of endotoxin. J Toxicol Environ Health A 2000; 59: 165-180.
139.
Park JH, Spiegelman DL, Burge HA, Gold DR, Chew GL, Milton DK. Longitudinal study of dust and airborne endotoxin in the home. Environ Health Perspect 2000; 108: 1023-8.
140.
George D, Schwartz DA. The role of endotoxin in acute and chronic grain dust induced airway disease. In: Bates D.V., Brain JD, Driscoll KE, Dungworth DL, Grafström R, Harris CC et al., (Hrsg.). Relationships between acute and chronic effects of air pollution. Washington: ILSI Press, 2000: 169-184.
141.
Parod RJ, Brain JD. Immune opsonin-independent phagocytosis by pulmonary macrophages. The Journal of Immunology 1986; 136: 2041-2047.
142.
Garcia-Garcia E, Rosales C. Signal transduction during Fc receptor-mediated phagocytosis. J Leukoc Biol 2002; 72: 1092-1108.
143.
Orphanides G, Reinberg D. A unified theory of gene expression. Cell 2002; 108: 439-451.
144.
Jaspers I, Samet JM, Reed W. Arsenite Exposure of Cultured Airway Epithelial Cells Activates kappa B-dependent Interleukin-8 Gene Expression in the Absence of Nuclear Factor-kappa B Nuclear Translocation. Journal of Biological Chemistry 1999; 274: 31025-31033.
145.
Jaspers I, Samet J, Erzurum S, Reed W. Vanadium-Induced kappa B-Dependent Transcription Depends upon Peroxide-Induced Activation of the p38 Mitogen-Activated Protein Kinase. American Journal of Respiratory Cell and Molecular Biology 2000; 23: 95-102.
146.
Hsu MH, Wang M, Browning DD, Mukaida N, Ye RD. NF-{kappa}B Activation Is Required for C5a-Induced Interleukin-8 Gene Expression in Mononuclear Cells. Blood 1999; 93: 3241-3249.
147.
Pan ZK, Ye RD, Christiansen SC, Jagels MA, Bokoch GM, Zuraw BL. Role of the Rho GTPase in bradykinin-stimulated Nuclear Factor-kB activation and IL-1ß gene expression in cultured human epithelial cells. J Immunol 1998; 160: 3038-3045.
148.
Pei XH, Nakanishi Y, Takayama K, Bai F, Hara N. Benzo[a]pyrene Activates the Human p53 Gene through Induction of Nuclear Factor kappa B Activity. Journal of Biological Chemistry 1999; 274: 35240-35246.
149.
Kwon O, Lee E, Moon TC, Jung H, Lin CX, Nam KS et al. Expression of cyclooxygenase-2 and pro-inflammatory cytokines induced by 2,2',4,4',5,5'-hexachlorobiphenyl (PCB 153) in human mast cells requires NF-kappa B activation. Biol Pharm Bull 2002; 25: 1165-1168.
150.
Desaki M, Takizawa H, Kasama T, Kobayashi K, Morita Y, Yamamoto K. Nuclear factor-kappa b activation in silica-induced interleukin 8 production by human bronchial epithelial cells. Cytokine 2000; 12: 1257-1260.
151.
Takizawa H, Ohtoshi T, Kawasaki S, Kohyama T, Desaki M, Kasama T et al. Diesel exhaust particles induce NF-kappa B activation in human bronchial epithelial cells in vitro: importance in cytokine transcription. J Immunol 1999; 162: 4705-11.
152.
Churg A, Wright JL. Airway wall remodeling induced by occupational mineral dusts and air pollutant particles. Chest 2002; 122: 306S-309S.
153.
Jimenez LA, Thompson J, Brown DA, Rahman I, Antonicelli F, Duffin R et al. Activation of NF-kappaB by PM(10) occurs via an iron-mediated mechanism in the absence of IkappaB degradation. Toxicol Appl Pharmacol 2000; 166: 101-10.
154.
Peden DB. Pollutants and asthma: role of air toxics. Environ Health Perspect 2002; 110 Suppl 4: 565-568.
155.
Blackwell TS, Christman JW. The role of Nuclear Factor-kappa B in cytokine gene regulation. American Journal of Respiratory Cell and Molecular Biology 1997; 17: 3-9.
156.
Bureau F, Delhalle S, Bonizzi G, Fievez L, Dogne S, Kirschvink N et al. Mechanisms of persistent NF-kappa B activity in the bronchi of an animal model of asthma. J Immunol 2000; 165: 5822-5830.
157.
Mercurio F, Manning AM. Multiple signals converging on NF-kappaB. Curr Opin Cell Biol 1999; 11: 226-232.
158.
Hanada T, Yoshimura A. Regulation of cytokine signaling and inflammation. Cytokine Growth Factor Rev 2002; 13: 413-421.
159.
Christman JW, Sadikot RT, Blackwell TS. The role of nuclear factor-kappa B in pulmonary diseases. Chest 2000; 117: 1482-1487.
160.
Quay JL, Reed W, Samet J, Devlin RB. Air pollution particles induce IL-6 gene expression in human airway epithelial cells via NF-kappaB activation. Am J Respir Cell Mol Biol 1998; 19: 98-106.
161.
Chimini G, Chavrier P. Function of Rho family proteins in actin dynamics during phagocytosis and engulfment. Nat Cell Biol 2000; 2: E191-E196.
162.
Ridley AJ. Rho GTPases and cell migration. J Cell Sci 2001; 114: 2713-2722.
163.
Houle F, Rousseau S, Morrice N, Luc M, Mongrain S, Turner CE et al. Extracellular signal-regulated kinase mediates phosphorylation of tropomyosin-1 to promote cytoskeleton remodeling in response to oxidative stress: impact on membrane blebbing. Mol Biol Cell 2003; 14: 1418-1432.
164.
Greenberg S. Biology of phagocytosis. In: Gallin JI, Snyderman R, (Hrsg.). Inflammation basic principles and clinical correlates. Philadelphia, PA: Lippincott Williams & Wilkins, 1999: 681-701.
165.
Stossel TT. Mechanical responses of white blood cells. In: Gallin JI, Snyderman R, (Hrsg.). Inflammation basic principles and clinical correlates. Philadelphia, PA: Lippincott Williams & Wilkins, 1999: 661-679.
166.
DeLeo FR, Allen LA, Apicella M, Nauseef WM. NADPH Oxidase Activation and Assembly During Phagocytosis. The Journal of Immunology 1999; 163: 6732-6740
167.
Carreras MC, Riobo NA, Pargament GA, Boveris A, Poderoso JJ. Effects of respiratory burst inhibitors on nitric oxide production by human neutrophils. Free Radic Res 1997; 26: 325-334.
168.
Carreras MC, Pargament GA, Catz SD, Poderoso JJ, Boveris A. Kinetics of nitric oxide and hydrogen peroxide production and formation of peroxynitrite during the respiratory burst of human neutrophils. FEBS Lett 1994; 341: 65-68.
169.
MacMicking J, Xie QW, Nathan C. Nitric oxide and macrophage function. Annu Rev Immunol 1997; 15: 323-350.
170.
Bogdan C, Rollinghoff M, Diefenbach A. The role of nitric oxide in innate immunity. Immunol Rev 2000; 173: 17-26.
171.
Kohen R, Nyska A. Oxidation of biological systems: oxidative stress phenomena, antioxidants, redox reactions, and methods for their quantification. Toxicol Pathol 2002; 30: 620-650.
172.
Nathan C. Specificity of a third kind: reactive oxygen and nitrogen intermediates in cell signaling. J Clin Invest 2003; 111: 769-778.
173.
The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. The Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group. N Engl J Med 1994; 330: 1029-1035.
174.
Paolini M, Abdel-Rahman SZ, Cantelli-Forti G, Legator MS. Chemoprevention or antichemoprevention? A salutary warning From the beta-carotene experience. J Natl Cancer Inst 2001; 93: 1110-1111.
175.
Palm JP, Graf P, Lundberg JO, Alving K. Characterization of exhaled nitric oxide: introducing a new reproducible method for nasal nitric oxide measurements. Eur Respir J 2000; 16: 236-41.
176.
Schedin U, Frostell C, Persson MG, Jakobsson J, Andersson G, Gustafsson LE. Contribution from upper and lower airways to exhaled endogenous nitric oxide in humans. Acta Anaesthesiol Scand 1995; 39: 327-32.
177.
Furukawa K, Harrison DG, Saleh D, Shennib H, Chagnon FP, Glaid A. Expression of nitric oxide synthase in the human nasal mucosa. Am J Respir Crit Care Med 1996; 153: 847-850.
178.
Lundberg JO, Farkas-Szallasi T, Weitzberg E, Rinder J, Lidholm J, Anggaard A et al. High nitric oxide production in human paranasal sinuses. Nat Med 1995; 1: 370-3.
179.
Lundberg JO, Weitzberg E. Nasal nitric oxide in man. Thorax 1999; 54: 947-52.
180.
Haddad IY, Pataki G, Hu P, Galliani C, Beckman JS, Matalon S. Quantitation of nitrotyrosine levels in lung sections of patients and animals with acute lung injury. J Clin Invest 1994; 94: 2407-2413.
181.
Feng X, Guo Z, Nourbakhsh M, Hauser H, Ganster R, Shao L et al. Identification of a negative response element in the human inducible nitric-oxide synthase (hiNOS) promoter: The role of NF-kappa B-repressing factor (NRF) in basal repression of the hiNOS gene. Proc Natl Acad Sci U S A 2002; 99: 14212-14217.
182.
Aust AE, Ball JC, Hu AA, Lighty JS, Smith KR, Straccia AM et al. Particle characteristics responsible for effects on human lung epithelial cells. Res Rep Health Eff Inst 2002; 1-65.
183.
Dreher KL, Jaskot RH, Lehmann JR, Richards JH, McGee JK, Ghio AJ et al. Soluble transition metals mediate residual oil fly ash induced acute lung injury. J Toxicol Environ Health 1997; 50: 285-305.
184.
Frampton MW, Ghio AJ, Samet JM, Carson JL, Carter JD, Devlin RB. Effects of aqueous extracts of PM(10) filters from the Utah valley on human airway epithelial cells. Am J Physiol 1999; 277: 960-7.
185.
Himeno S, Yanagiya T, Enomoto S, Kondo Y, Imura N. Cellular cadmium uptake mediated by the transport system for manganese. Tohoku J Exp Med 2002; 196: 43-50.
186.
Guerinot ML. The ZIP family of metal transporters. Biochim Biophys Acta 2000; 1465: 190-198.
187.
Hubert N, Hentze MW. Previously uncharacterized isoforms of divalent metal transporter (DMT)-1: implications for regulation and cellular function. Proc Natl Acad Sci U S A 2002; 99: 12345-12350.
188.
Ghio AJ, Stonehuerner J, Dailey LA, Carter JD. Metals associated with both the water-soluble and insoluble fractions of an ambient air pollution particle catalyze an oxidative stress. Inhal Toxicol 1999; 11: 37-49.
189.
Stohs SJ, Bagchi D. Oxidative mechanisms in the toxicity of metal ions. Free Radic Biol Med 1995; 18: 321-36.
190.
Samet JM, Graves LM, Quay J, Dailey LA, Devlin RB, Ghio AJ et al. Activation of MAPKs in human bronchial epithelial cells exposed to metals. Am J Physiol 1998; 275: L551-L558.
191.
Bernstam L, Nriagu J. Molecular aspects of arsenic stress. J Toxicol Environ Health B Crit Rev 2000; 3: 293-322.
192.
Zhou YM, Zhong CY, Kennedy IM, Leppert VJ, Pinkerton KE. Oxidative stress and NFkappaB activation in the lungs of rats: a synergistic interaction between soot and iron particles. Toxicol Appl Pharmacol 2003; 190: 157-169.
193.
Meriin AB, Yaglom JA, Gabai VL, Zon L, Ganiatsas S, Mosser DD et al. Protein-damaging stresses activate c-Jun N-terminal kinase via inhibition of its dephosphorylation: a novel pathway controlled by HSP72. Mol Cell Biol 1999; 19: 2547-2555.
194.
Cheng JZ, Sharma R, Yang Y, Singhal SS, Sharma A, Saini MK et al. Accelerated metabolism and exclusion of 4-hydroxynonenal through induction of RLIP76 and hGST5.8 is an early adaptive response of cells to heat and oxidative stress. J Biol Chem 2001; 276: 41213-41223.
195.
Marnett LJ. Oxy radicals, lipid peroxidation and DNA damage. Toxicology 2002; 181-182: 219-222.
196.
Kharbanda S, Pandey P, Yamauchi T, Kumar S, Kaneki M, Kumar V et al. Activation of MEK Kinase 1 by the c-Abl Protein Tyrosine Kinase in Response to DNA Damage. Mol Cell Biol 2000; 20: 4979-4989.
197.
Kyriakis JM, Avruch J. Sounding the alarm: protein kinase cascades activated by stress and inflammation. J Biol Chem 1996; 271: 24313-24316.
198.
Gius D, Botero A, Shah S, Curry HA. Intracellular oxidation/reduction status in the regulation of transcription factors NF-kappaB and AP-1. Toxicol Lett 1999; 106: 93-106.
199.
Schoonbroodt S, Piette J. Oxidative stress interference with the nuclear factor-kappa B activation pathways. Biochem Pharmacol 2000; 60: 1075-1083.
200.
Bowie A, O'Neill LA. Oxidative stress and nuclear factor-kappaB activation: a reassessment of the evidence in the light of recent discoveries. Biochem Pharmacol 2000; 59: 13-23.
201.
Schoonbroodt S, Ferreira V, Best-Belpomme M, Boelaert JR, Legrand-Poels S, Korner M et al. Crucial role of the amino-terminal tyrosine residue 42 and the carboxyl-terminal PEST domain of I kappa B alpha in NF-kappa B activation by an oxidative stress. J Immunol 2000; 164: 4292-4300.
202.
Byun MS, Jeon KI, Choi JW, Shim JY, Jue DM. Dual effect of oxidative stress on NF-kappakB activation in HeLa cells. Exp Mol Med 2002; 34: 332-339.
203.
Chandel NS, Trzyna WC, McClintock DS, Schumacker PT. Role of oxidants in NF-kappa B activation and TNF-alpha gene transcription induced by hypoxia and endotoxin. J Immunol 2000; 165: 1013-1021.
204.
Hetland RB, Refsnes M, Myran T, Johansen BV, Uthus N, Schwarze PE. Mineral and/or metal content as critical determinants of particle-induced release of IL-6 and IL-8 from A549 cells. J Toxicol Environ Health A 2000; 60: 47-65.
205.
Dong C, Davis RJ, Flavell RA. Signaling by the JNK group of MAP kinases. c-jun N-terminal Kinase. J Clin Immunol 2001; 21: 253-257.
206.
Li N, Kim S, Wang M, Froines J, Sioutas C, Nel A. Use of a stratified oxidative stress model to study the biological effects of ambient concentrated and diesel exhaust particulate matter. Inhal Toxicol 2002; 14: 459-486.
207.
Kobayashi SD, Voyich JM, Buhl CL, Stahl RM, DeLeo FR. Global changes in gene expression by human polymorphonuclear leukocytes during receptor-mediated phagocytosis: Cell fate is regulated at the level of gene expression. PNAS 2002; 99: 6901-6906.
208.
Beswick PH. Free radical activity of industrial fibers: role of iron in oxidative stress and activation of transcription factors. Environ Health Perspect 1997; 105S: 1313-1317.
209.
Brown DM, Beswick PH, Donaldson K. Induction of nuclear translocation of NF-kappaB in epithelial cells by respirable mineral fibres. J Pathol 1999; 189: 258-264.
210.
Brunekreef B, Janssen NA, de Hartog J, Harssema H, Knape M, van Vliet P. Air pollution from truck traffic and lung function in children living near motorways. Epidemiology 1997; 8: 298-303.
211.
van Vliet P, Knape M, de Hartog J, Janssen N, Harssema H, Brunekreef B. Motor vehicle exhaust and chronic respiratory symptoms in children living near freeways. Environ Res 1997; 74: 122-132.
212.
Kramer U, Koch T, Ranft U, Ring J, Behrendt H. Traffic-related air pollution is associated with atopy in children living in urban areas. Epidemiology 2000; 11: 64-70.
213.
Nel AE, Diaz-Sanchez D, Ng D, Hiura T, Saxon A. Enhancement of allergic inflammation by the interaction between diesel exhaust particles and the immune system. J Allergy Clin Immunol 1998; 102: 539-54.
214.
Diaz-Sanchez D, Garcia MP, Wang M, Jyrala M, Saxon A. Nasal challenge with diesel exhaust particles can induce sensitization to a neoallergen in the human mucosa. J Allergy Clin Immunol 1999; 104: 1183-8.
215.
Diaz-Sanchez D, Tsien A, Fleming J, Saxon A. Combined diesel exhaust particulate and ragweed allergen challenge markedly enhances human in vivo nasal ragweed-specific IgE and skews cytokine production to a T helper cell 2-type pattern. J Immunol 1997; 158: 2406-13.
216.
Diaz-Sanchez D, Dotson AR, Takenaka H, Saxon A. Diesel exhaust particles induce local IgE production in vivo and alter the pattern of IgE messenger RNA isoforms. J Clin Invest 1994; 94: 1417-25.
217.
Diaz-Sanchez D, Penichet-Garcia M, Saxon A. Diesel exhaust particles directly induce activated mast cells to degranulate and increase histamine levels and symptom severity. J Allergy Clin Immunol 2000; 106: 1140-6.
218.
Diaz-Sanchez D, Proietti L, Polosa R. Diesel fumes and the rising prevalence of atopy: an urban legend? Curr Allergy Asthma Rep 2003; 3: 146-52.
219.
Wang M, Saxon A, Diaz-Sanchez D. Early IL-4 production driving Th2 differentiation in a human in vivo allergic model is mast cell derived. Clin Immunol 1999; 90: 47-54.
220.
Montnemery P, Popovic M, Andersson M, Greiff L, Nyberg P, Lofdahl CG et al. Influence of heavy traffic, city dwelling and socio-economic status on nasal symptoms assessed in a postal population survey. Respir Med 2003; 97: 970-977.
221.
Gluck U, Schutz R, Gebbers JO. Cytopathology of the nasal mucosa in chronic exposure to diesel engine emission: a five-year survey of Swiss customs officers. Environ Health Perspect 2003; 111: 925-929.
222.
Li N, Wang M, Oberley TD, Sempf JM, Nel AE. Comparison of the pro-oxidative and proinflammatory effects of organic diesel exhaust particle chemicals in bronchial epithelial cells and macrophages. J Immunol 2002; 169: 4531-4541.
223.
Diaz-Sanchez D, Tsien A, Casillas A, Dotson AR, Saxon A. Enhanced nasal cytokine production in human beings after in vivo challenge with diesel exhaust particles. J Allergy Clin Immunol 1996; 98: 114-23.
224.
Diaz-Sanchez D, Jyrala M, Ng D, Nel A, Saxon A. In vivo nasal challenge with diesel exhaust particles enhances expression of the CC chemokines rantes, MIP-1alpha, and MCP-3 in humans. Clin Immunol 2000; 97: 140-5.
225.
Meek MD. Ah receptor and estrogen receptor-dependent modulation of gene expression by extracts of diesel exhaust particles. Environ Res 1998; 79: 114-21.
226.
Saeki K, Matsuda T, Kato TA, Yamada K, Mizutani T, Matsui S et al. Activation of the human Ah receptor by aza-polycyclic aromatic hydrocarbons and their halogenated derivatives. Biol Pharm Bull 2003; 26: 448-452.
227.
Gonzalez FJ, Fernandez-Salguero P. The Aryl Hydrocarbon Receptor. Studies Using the AHR-Null Mice. Drug Metab Dispos 1998; 26: 1194-1198.
228.
Tian Y, Rabson AB, Gallo MA. Ah receptor and NF-kappaB interactions: mechanisms and physiological implications. Chem Biol Interact 2002; 141: 97-115.
229.
Puga A, Barnes SJ, Chang C, Zhu H, Nephew KP, Khan SA et al. Activation of transcription factors activator protein-1 and nuclear factor-kappaB by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Biochem Pharmacol 2000; 59: 997-1005.
230.
Kim DW, Gazourian L, Quadri SA, Romieu-Mourez R, Sherr DH, Sonenshein GE. The RelA NF-kappaB subunit and the aryl hydrocarbon receptor (AhR) cooperate to transactivate the c-myc promoter in mammary cells. Oncogene 2000; 19: 5498-5506.
231.
Tian Y, Ke S, Denison M, Rabson AB, Gallo MA. Ah Receptor and NF-kappa B Interactions, a Potential Mechanism for Dioxin Toxicity. Journal of Biological Chemistry 1999; 274: 510-515.
232.
Elizondo G, Fernandez-Salguero P, Sheikh MS, Kim GY, Fornace AJ, Lee KS et al. Altered Cell Cycle Control at the G2/M Phases in Aryl Hydrocarbon Receptor-Null Embryo Fibroblast. Mol Pharmacol 2000; 57: 1056-1063.
233.
Shimizu Y, Nakatsuru Y, Ichinose M, Takahashi Y, Kume H, Mimura J et al. Benzo[a]pyrene carcinogenicity is lost in mice lacking the aryl hydrocarbon receptor. PNAS 2000; 97: 779-782.
234.
Chan WK, Yao G, Gu YZ, Bradfield CA. Cross-talk between the Aryl Hydrocarbon Receptor and Hypoxia Inducible Factor Signaling Pathways. Demonstration of competition and compensation. Journal of Biological Chemistry 1999; 274: 12115-12123.
235.
Dertinger SD, Nazarenko DA, Silverstone AE, Gasiewicz TA. Aryl hydrocarbon receptor signaling plays a significant role in mediating benzo[a]pyrene- and cigarette smoke condensate-induced cytogenetic damage in vivo. Carcinogenesis 2001; 22: 171-7.
236.
Dockery DW, Speizer FE, Stram DO, Ware JH, Spengler JD, Ferris BG, Jr. Effects of inhalable particles on respiratory health of children. Am Rev Respir Dis 1989; 139: 587-594.
237.
Cullen RT, Tran CL, Buchanan D, Davis JM, Searl A, Jones AD et al. Inhalation of poorly soluble particles. I. Differences in inflammatory response and clearance during exposure. Inhal Toxicol 2000; 12: 1089-particulates 111.
238.
Donaldson K. Nonneoplastic lung responses induced in experimental animals by exposure to poorly soluble nonfibrous particles. Inhal Toxicol 2000; 12: 121-39.
239.
Brown DM, Wilson MR, MacNee W, Stone V, Donaldson K. Size-dependent proinflammatory effects of ultrafine polystyrene particles: a role for surface area and oxidative stress in the enhanced activity of ultrafines. Toxicol Appl Pharmacol 2001; 175: 191-9.
240.
Kobzik L. Lung macrophage uptake of unopsonized environmental particulates. Role of scavenger-type receptors. J Immunol 1995; 155: 367-376.
241.
Obot CJ, Morandi MT, Beebe TP, Hamilton RF, Holian A. Surface components of airborne particulate matter induce macrophage apoptosis through scavenger receptors. Toxicol Appl Pharmacol 2002; 184: 98-106.
242.
Palecanda A, Paulauskis J, Al Mutairi E, Imrich A, Qin G, Suzuki H et al. Role of the Scavenger Receptor MARCO in Alveolar Macrophage Binding of Unopsonized Environmental Particles. The Journal of Experimental Medicine 1999; 189: 1497-1506.
243.
Palecanda A, Kobzik L. Receptors for unopsonized particles: the role of alveolar macrophage scavenger receptors. Curr Mol Med 2001; 1: 589-595.
244.
Fraser IP, Ezekowitz RAB. Receptors for microbial products: Carbohydrates. In: Gallin JI, Snyderman R, (Hrsg.). Inflammation basic principles and clinical correlates. Philadelphia, PA: Lippincott Williams & Wilkins, 1999: 515-524.
245.
Stringer B, Imrich A, Kobzik L. Lung epithelial cell (A549) interaction with unopsonized environmental particulates: quantitation of particle-specific binding and IL-8 production. Exp Lung Res 1996; 22: 495-508.
246.
Dunne DW, Resnick D, Greenberg J, Krieger M, Joiner KA. The Type I Macrophage Scavenger Receptor Binds to Gram-Positive Bacteria and Recognizes Lipoteichoic Acid. PNAS 1994; 91: 1863-1867.
247.
Elomaa O, Sankala M, Pikkarainen T, Bergmann U, Tuuttila A, Raatikainen-Ahokas A et al. Structure of the human macrophage MARCO receptor and characterization of its bacteria-binding region. J Biol Chem 1998; 273: 4530-4538.
248.
Goldsmith CA, Frevert C, Imrich A, Sioutas C, Kobzik L. Alveolar macrophage interaction with air pollution particulates. Environ Health Perspect 1997; 105 Suppl 5: 1191-1195.
249.
Stringer B, Imrich A, Kobzik L. Flow cytometric assay of lung macrophage uptake of environmental particulates. Cytometry 1995; 20: 23-32.
250.
Tao F, Palecanda A, Kumar S, Kobzik L. Generation of a Monoclonal Antibody that Blocks Epithelial Binding of Unopsonized Particles. Hybrid Hybridomics 2003; 22: 17-21.
251.
Riechelmann H, Rettinger G, Weschta M, Keck T, Deutschle T. Effects of low-toxicity particulate matter on human nasal function. J Occup Environ Med 2003; 45: 54-60.
252.
Heinrich J. Nonallergic respiratory morbidity improved along with a decline of traditional air pollution levels: a review. Eur Respir J Suppl 2003; 40: 64s-69s.
253.
von Mutius E, Sherrill DL, Fritzsch C, Martinez FD, Lebowitz MD. Air pollution and upper respiratory symptoms in children from East Germany. Eur Respir J 1995; 8: 723-8.
254.
Peters A, Goldstein IF, Beyer U, Franke K, Heinrich J, Dockery DW et al. Acute health effects of exposure to high levels of air pollution in eastern Europe. Am J Epidemiol 1996; 144: 570-81.
255.
Peacock JL, Symonds P, Jackson P, Bremner SA, Scarlett JF, Strachan DP et al. Acute effects of winter air pollution on respiratory function in schoolchildren in southern England. Occup Environ Med 2003; 60: 82-89.
256.
Pikhart H, Bobak M, Gorynski P, Wojtyniak B, Danova J, Celko MA et al. Outdoor sulphur dioxide and respiratory symptoms in Czech and Polish school children: a small-area study (SAVIAH). Small-Area Variation in Air Pollution and Health. Int Arch Occup Environ Health 2001; 74: 574-578.
257.
Knorst MM, Kienast K, Riechelmann H, Muller-Quernheim J, Ferlinz R. Effect of sulfur dioxide on mucociliary activity and ciliary beat frequency in guinea pig trachea. Int Arch Occup Environ Health 1994; 65: 325-8.
258.
Kienast K, Riechelmann H, Knorst M, Schlegel J, Muller-Quernheim J, Schellenberg J et al. An experimental model for the exposure of human ciliated cells to sulfur dioxide at different concentrations. Clin Investig 1994; 72: 215-9.
259.
Gunnison AF, Sellakumar A, Currie D, Snyder EA. Distribution, metabolism and toxicity of inhaled sulfur dioxide and endogenously generated sulfite in the respiratory tract of normal and sulfite oxidase-deficient rats. J Toxicol Environ Health 1987; 21: 141-62.
260.
Riechelmann H, Maurer J, Kienast K, Hafner B, Mann WJ. Respiratory epithelium exposed to sulfur dioxide--functional and ultrastructural alterations. Laryngoscope 1995; 105: 295-299.
261.
Ishizuka S, Yamaya M, Suzuki T, Nakayama K, Kamanaka M, Ida S et al. Acid exposure stimulates the adherence of Streptococcus pneumoniae to cultured human airway epithelial cells: effects on platelet-activating factor receptor expression. Am J Respir Cell Mol Biol 2001; 24: 459-68.
262.
Langley-Evans SC, Phillips GJ, Jackson AA. Sulphur dioxide: a potent glutathione depleting agent. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol 1996; 114: 89-98.
263.
Lee WJ, Teschke K, Kauppinen T, Andersen A, Jappinen P, Szadkowska-Stanczyk I et al. Mortality from lung cancer in workers exposed to sulfur dioxide in the pulp and paper industry. Environ Health Perspect 2002; 110: 991-995.
264.
Nyberg F, Gustavsson P, Jarup L, Bellander T, Berglind N, Jakobsson R et al. Urban air pollution and lung cancer in Stockholm. Epidemiology 2000; 11: 487-495.
265.
Sulfur dioxide and some sulfites, bisulfites and metabisulfites. IARC Monogr Eval Carcinog Risks Hum 1992; 54: 131-188.
266.
Tominaga M, Julius D. Capsaicin receptor in the pain pathway. Jpn J Pharmacol 2000; 83: 20-24.
267.
Blomberg A, Krishna MT, Bocchino V, Biscione GL, Shute JK, Kelly FJ et al. The inflammatory effects of 2 ppm NO2 on the airways of healthy subjects. Am J Respir Crit Care Med 1997; 156: 418-424.
268.
Wang JH, Devalia JL, Duddle JM, Hamilton SA, Davies RJ. Effect of six-hour exposure to nitrogen dioxide on early-phase nasal response to allergen challenge in patients with a history of seasonal allergic rhinitis. J Allergy Clin Immunol 1995; 96: 669-76.
269.
Wang JH, Duddle J, Devalia JL, Davies RJ. Nitrogen dioxide increases eosinophil activation in the early-phase response to nasal allergen provocation. Int Arch Allergy Immunol 1995; 107: 103-5.
270.
Pryor WA, Squadrito GL, Friedman M. The cascade mechanism to explain ozone toxicity: the role of lipid ozonation products. Free Radic Biol Med 1995; 19: 935-941.
271.
Pryor WA, Squadrito GL, Friedman M. A new mechanism for the toxicity of ozone. Toxicol Lett 1995; 83: 287-93.
272.
Nichols BG, Woods JS, Luchtel DL, Corral J, Koenig JQ. Effects of ozone exposure on nuclear factor-kappaB activation and tumor necrosis factor-alpha expression in human nasal epithelial cells. Toxicol Sci 2001; 60: 356-62.
273.
Frampton MW, Pryor WA, Cueto R, Cox C, Morrow PE, Utell MJ. Ozone exposure increases aldehydes in epithelial lining fluid in human lung. Am J Respir Crit Care Med 1999; 159: 1134-1137.
274.
Frampton MW, Pryor WA, Cueto R, Cox C, Morrow PE, Utell MJ. Aldehydes (nonanal and hexanal) in rat and human bronchoalveolar lavage fluid after ozone exposure. Res Rep Health Eff Inst 1999; 1-15.
275.
Mustafa MG. Biochemical basis of ozone toxicity. Free Radic Biol Med 1990; 9: 245-265.
276.
Long NC, Suh J, Morrow JD, Schiestl RH, Murthy GG, Brain JD et al. Ozone causes lipid peroxidation but little antioxidant depletion in exercising and nonexercising hamsters. J Appl Physiol 2001; 91: 1694-1700.
277.
Laskin DL, Fakhrzadeh L, Heck DE, Gerecke D, Laskin JD. Upregulation of phosphoinositide 3-kinase and protein kinase B in alveolar macrophages following ozone inhalation. Role of NF-kappaB and STAT-1 in ozone-induced nitric oxide production and toxicity. Mol Cell Biochem 2002; 234-235: 91-98.
278.
Jaspers I, Flescher E, Chen LC. Ozone-induced IL-8 expression and transcription factor binding in respiratory epithelial cells. Am J Physiol 1997; 272: 504-11.
279.
Haddad EB, Salmon M, Koto H, Barnes PJ, Adcock I, Chung KF. Ozone induction of cytokine-induced neutrophil chemoattractant (CINC) and nuclear factor-kappa b in rat lung: inhibition by corticosteroids. FEBS Lett 1996; 379: 265-8.
280.
Kafoury RM, Pryor WA, Squadrito GL, Salgo MG, Zou X, Friedman M. Lipid ozonation products activate phospholipases A2, C, and D. Toxicol Appl Pharmacol 1998; 150: 338-349.
281.
Cho HY, Hotchkiss JA, Bennett CB, Harkema JR. Neutrophil-dependent and neutrophil-independent alterations in the nasal epithelium of ozone-exposed rats. Am J Respir Crit Care Med 2000; 162: 629-636.
282.
Schierhorn K, Hanf G, Fischer A, Umland B, Olze H, Kunkel G. Ozone-induced release of neuropeptides from human nasal mucosa cells. Int Arch Allergy Immunol 2002; 129: 145-151.
283.
Wilkins CK, Wolkoff P, Clausen PA, Hammer M, Nielsen GD. Upper airway irritation of terpene/ozone oxidation products (TOPS). Dependence on reaction time, relative humidity and initial ozone concentration. Toxicol Lett 2003; 143: 109-114.
284.
Potter-Perigo S, Kaplan ED, Luchtel DL, Baker C, Altman LC, Wight TN. Ozone alters the expression of tenascin-C in cultured primate nasal epithelial cells. Am J Respir Cell Mol Biol 1998; 18: 471-478.
285.
Harkema JR, Hotchkiss JA, Griffith WC. Mucous cell metaplasia in rat nasal epithelium after a 20-month exposure to ozone: a morphometric study of epithelial differentiation. Am J Respir Cell Mol Biol 1997; 16: 521-530.
286.
Harkema JR, Morgan KT, Gross EA, Catalano PJ, Griffith WC. Consequences of prolonged inhalation of ozone on F344/N rats: collaborative studies. Part VII: Effects on the nasal mucociliary apparatus. Res Rep Health Eff Inst 1994; 3-26.
287.
Harkema JR, Catalano PJ, Hotchkiss JA. Consequences of prolonged inhalation of ozone on F344/N rats: collaborative studies. Part XII: Atrophy of bone in nasal turbinates. Res Rep Health Eff Inst 1997; 1-19
288.
Johnson NF, Hotchkiss JA, Harkema JR, Henderson RF. Proliferative responses of rat nasal epithelia to ozone. Toxicol Appl Pharmacol 1990; 103: 143-155.
289.
Harkema JR, Barr EB, Hotchkiss JA. Responses of rat nasal epithelium to short- and long-term exposures of ozone: image analysis of epithelial injury, adaptation and repair. Microsc Res Tech 1997; 36: 276-286.
290.
Harkema JR, Hotchkiss JA, Barr EB, Bennett CB, Gallup M, Lee JK et al. Long-lasting effects of chronic ozone exposure on rat nasal epithelium. Am J Respir Cell Mol Biol 1999; 20: 517-29.
291.
Jorge SA, Menck CF, Sies H, Osborne MR, Phillips DH, Sarasin A et al. Mutagenic fingerprint of ozone in human cells. DNA Repair (Amst) 2002; 1: 369-378.
292.
Godish T. Aldehydes. In: Spengler JD, Samet JM, McCarthy JF, (Hrsg.). Indoor air quality handbook. New York: McGraw-Hill, 2001: 32.1-32.22.
293.
Tucker G. Volatile organic compounds. In: Spengler JD, Samet JM, McCarthy JF, (Hrsg.). Indoor air quality handbook. New York: McGraw-Hill, 2001: 31.1-31.20.
294.
Alarie Y. Sensory irritation of the upper airways by airborne chemicals. Toxicol Appl Pharmacol 1973; 24: 279-297.
295.
Bos PM, Zwart A, Reuzel PG, Bragt PC. Evaluation of the sensory irritation test for the assessment of occupational health risk. Crit Rev Toxicol 1991; 21: 423-450.
296.
Alarie Y, Nielsen GD, Schaper MM. Animal bioasays for evaluation of indoor air quality. In: Spengler JD, Samet JM, McCarthy JF, (Hrsg.). Indoor air quality handbook. New York: McGraw-Hill, 2001: 23.1-23.49.
297.
Severini C, Improta G, Falconieri-Erspamer G, Salvadori S, Erspamer V. The tachykinin peptide family. Pharmacol Rev 2002; 54: 285-322.
298.
Shirasaki H, Asakura K, Narita SI, Kataura A. Expression of substance P (NK1) receptor mRNA in human nose. Acta Otolaryngol 1998; 118: 717-22.
299.
Richardson JD, Vasko MR. Cellular mechanisms of neurogenic inflammation. J Pharmacol Exp Ther 2002; 302: 839-845.
300.
Hunter DD, Satterfield BE, Huang J, Fedan JS, Dey RD. Toluene diisocyanate enhances substance P in sensory neurons innervating the nasal mucosa. Am J Respir Crit Care Med 2000; 161: 543-549.
301.
Morris JB, Stanek J, Gianutsos G. Sensory nerve-mediated immediate nasal responses to inspired acrolein. J Appl Physiol 1999; 87: 1877-1886.
302.
Sasamura T, Kuraishi Y. Peripheral and central actions of capsaicin and VR1 receptor. Jpn J Pharmacol 1999; 80: 275-280.
303.
Veronesi B, Oortgiesen M. Neurogenic inflammation and particulate matter (PM) air pollutants. Neurotoxicology 2001; 22: 795-810.
304.
Wu ZX, Satterfield BE, Dey RD. Substance P released from intrinsic airway neurons contributes to ozone-enhanced airway hyperresponsiveness in ferret trachea. J Appl Physiol 2003; 95: 742-750.
305.
Canning BJ. Potential role of tachykinins in inflammatory diseases. J Allergy Clin Immunol 1997; 99: 579-82.
306.
Hood VC, Cruwys SC, Urban L, Kidd BL. Differential role of neurokinin receptors in human lymphocyte and monocyte chemotaxis. Regul Pept 2000; 96: 17-21.
307.
van der Kleij HP, Kraneveld AD, Redegeld FA, Gerard NP, Morteau O, Nijkamp FP. The tachykinin NK1 receptor is crucial for the development of non-atopic airway inflammation and hyperresponsiveness. Eur J Pharmacol 2003; 476: 249-255.
308.
Phillips JE, Hey JA, Corboz MR. Tachykinin NK3 and NK1 receptor activation elicits secretion from porcine airway submucosal glands. British Journal of Pharmacology 2003; 138: 254-260.
309.
Nakajima Y, Tsuchida K, Negishi M, Ito S, Nakanishi S. Direct linkage of three tachykinin receptors to stimulation of both phosphatidylinositol hydrolysis and cyclic AMP cascades in transfected Chinese hamster ovary cells. J Biol Chem 1992; 267: 2437-2442.
310.
Mechiche H, Koroglu A, Candenas L, Pinto FM, Birembaut P, Bardou M et al. Neurokinins induce relaxation of human pulmonary vessels through stimulation of endothelial NK1 receptors. J Cardiovasc Pharmacol 2003; 41: 343-355.
311.
Pedersen KE, Buckner CK, Meeker SN, Undem BJ. Pharmacological examination of the neurokinin-1 receptor mediating relaxation of human intralobar pulmonary artery. J Pharmacol Exp Ther 2000; 292: 319-325.
312.
Baluk P, Thurston G, Murphy TJ, Bunnett NW, McDonald DM. Neurogenic plasma leakage in mouse airways. Br J Pharmacol 1999; 126: 522-528.
313.
Quinlan KL, Naik SM, Cannon G, Armstrong CA, Bunnett NW, Ansel JC et al. Substance P Activates Coincident NF-AT- and NF-{kappa}B-Dependent Adhesion Molecule Gene Expression in Microvascular Endothelial Cells Through Intracellular Calcium Mobilization. J Immunol 1999; 163: 5656-5665.
314.
Baluk P, Bertrand C, Geppetti P, McDonald DM, Nadel JA. NK1 receptors mediate leukocyte adhesion in neurogenic inflammation in the rat trachea. Am J Physiol 1995; 268: L263-L269.
315.
DeRose V, Robbins RA, Snider RM, Spurzem JR, Thiele GM, Rennard SI et al. Substance P increases neutrophil adhesion to bronchial epithelial cells. J Immunol 1994; 152: 1339-1346.
316.
Kuo HP, Lin HC, Hwang KH, Wang CH, Lu LC. Lipopolysaccharide enhances substance P-mediated neutrophil adherence to epithelial cells and cytokine release. Am J Respir Crit Care Med 2000; 162: 1891-1897.
317.
Tominaga K, Honda K, Akahoshi A, Makino Y, Kawarabayashi T, Takano Y et al. Substance P causes adhesion of neutrophils to endothelial cells via protein kinase C. Biol Pharm Bull 1999; 22: 1242-1245.
318.
Dianzani C, Collino M, Lombardi G, Garbarino G, Fantozzi R. Substance P increases neutrophil adhesion to human umbilical vein endothelial cells. Br J Pharmacol 2003; 139: 1103-1110.
319.
Kahler CM, Pischel A, Kaufmann G, Wiedermann CJ. Influence of neuropeptides on neutrophil adhesion and transmigration through a lung fibroblast barrier in vitro. Exp Lung Res 2001; 27: 25-46.
320.
Tanabe T, Otani H, Bao L, Mikami Y, Yasukura T, Ninomiya T et al. Intracellular signaling pathway of substance P-induced superoxide production in human neutrophils. Eur J Pharmacol 1996; 299: 187-195.
321.
Bockmann S, Seep J, Jonas L. Delay of neutrophil apoptosis by the neuropeptide substance P: involvement of caspase cascade. Peptides 2001; 22: 661-670.
322.
Hood VC, Cruwys SC, Urban L, Kidd BL. Differential role of neurokinin receptors in human lymphocyte and monocyte chemotaxis. Regul Pept 2000; 96: 17-21.
323.
Dunzendorfer S, Kaser A, Meierhofer C, Tilg H, Wiedermann CJ. Cutting edge: peripheral neuropeptides attract immature and arrest mature blood-derived dendritic cells. J Immunol 2001; 166: 2167-2172.
324.
Numao T, Agrawal DK. Neuropeptides modulate human eosinophil chemotaxis. The Journal of Immunology 1992; 149: 3309-3315.
325.
Dunzendorfer S, Schratzberger P, Reinisch N, Kahler CM, Wiedermann CJ. Secretoneurin, a novel neuropeptide, is a potent chemoattractant for human eosinophils. Blood 1998; 91: 1527-1532.
326.
Korsgren M, Erjefalt JS, Hinterholzl J, Fischer-Colbrie R, Ahlstrom Emanuelsson C, Andersson M et al. Neural expression and increased lavage fluid levels of secretoneurin in seasonal allergic rhinitis. Am J Respir Crit Care Med 2003; 200212-1508OC.
327.
van der Kleij HP, Ma D, Redegeld FA, Kraneveld AD, Nijkamp FP, Bienenstock J. Functional expression of neurokinin 1 receptors on mast cells induced by IL-4 and stem cell factor. J Immunol 2003; 171: 2074-2079.
328.
Lilly CM, Hall AE, Rodger IW, Kobzik L, Haley KJ, Drazen JM. Substance P-induced histamine release in tracheally perfused guinea pig lungs. J Appl Physiol 1995; 78: 1234-1241.
329.
Brass DM, Savov JD, Gavett SH, Haykal-Coates N, Schwartz DA. Subchronic endotoxin inhalation causes persistent airway disease. Am J Physiol Lung Cell Mol Physiol 2003; 285: L755-L761.
330.
Dube D, Puruckherr M, Byrd RP, Jr., Roy TM. Reactive airways dysfunction syndrome following metal fume fever. Tenn Med 2002; 95: 236-238.
331.
Quirce S, Gala G, Perez-Camo I, Sanchez-Fernandez C, Pacheco A, Losada E. Irritant-induced asthma: clinical and functional aspects. J Asthma 2000; 37: 267-274.
332.
Leroyer C, Malo JL, Girard D, Dufour JG, Gautrin D. Chronic rhinitis in workers at risk of reactive airways dysfunction syndrome due to exposure to chlorine. Occup Environ Med 1999; 56: 334-338.
333.
Deschamps D, Questel F, Baud FJ, Gervais P, Dally S. Persistent asthma after acute inhalation of organophosphate insecticide. Lancet 1994; 344: 1712.
334.
Teramoto S, Tanaka H, Kaneko S, Abe S. Neurokinin-1 receptor antagonist inhibits short-term sulfuric-acid-induced airway hyperresponsiveness in sensitized guinea pigs. Int Arch Allergy Immunol 2000; 121: 53-6.
335.
Castranova V, Frazer DG, Manley LK, Dey RD. Pulmonary alterations associated with inhalation of occupational and environmental irritants. Int Immunopharmacol 2002; 2: 163-172.
336.
Sikora ER, Stone S, Tomblyn S, Frazer DG, Castranova V, Dey RD. Asphalt exposure enhances neuropeptide levels in sensory neurons projecting to the rat nasal epithelium. J Toxicol Environ Health A 2003; 66: 1015-1027.
337.
Romero MI, Rangappa N, Li L, Lightfoot E, Garry MG, Smith GM. Extensive Sprouting of Sensory Afferents and Hyperalgesia Induced by Conditional Expression of Nerve Growth Factor in the Adult Spinal Cord. J Neurosci 2000; 20: 4435-4445.
338.
Bibel M, Barde YA. Neurotrophins: key regulators of cell fate and cell shape in the vertebrate nervous system. Genes Dev 2000; 14: 2919-2937.
339.
Braun A, Quarcoo D, Schulte-Herbruggen O, Lommatzsch M, Hoyle G, Renz H. Nerve growth factor induces airway hyperresponsiveness in mice. Int Arch Allergy Immunol 2001; 124: 205-207.
340.
de Vries A, Dessing M, Engels F, Henricks P, Nijkamp F. Nerve Growth Factor Induces a Neurokinin-1 Receptor- Mediated Airway Hyperresponsiveness in Guinea Pigs. Am J Respir Crit Care Med 1999; 159: 1541-1544.
341.
Baraniuk JN, Ali M, Yuta A, Fang SY, Naranch K. Hypertonic saline nasal provocation stimulates nociceptive nerves, substance P release, and glandular mucous exocytosis in normal humans. Am J Respir Crit Care Med 1999; 160: 655-62.
342.
Sanico AM, Stanisz AM, Gleeson TD, Bora S, Proud D, Bienenstock J et al. Nerve growth factor expression and release in allergic inflammatory disease of the upper airways. Am J Respir Crit Care Med 2000; 161: 1631-1635.
343.
Kanda N, Watanabe S. Histamine enhances the production of nerve growth factor in human keratinocytes. J Invest Dermatol 2003; 121: 570-577.
344.
Fox AJ, Patel HJ, Barnes PJ, Belvisi MG. Release of nerve growth factor by human pulmonary epithelial cells: role in airway inflammatory diseases. Eur J Pharmacol 2001; 424: 159-162.
345.
Lallemend F, Lefebvre PP, Hans G, Rigo JM, Van De Water TR, Moonen G et al. Substance P protects spiral ganglion neurons from apoptosis via PKC-Ca2+-MAPK/ERK pathways. J Neurochem 2003; 87: 508-521.
346.
Weyer U, Schafer R, Himmler A, Mayer SK, Burger E, Czernilofsky AP et al. Establishment of a cellular assay system for G protein-linked receptors: coupling of human NK2 and 5-HT2 receptors to phospholipase C activates a luciferase reporter gene. Receptors Channels 1993; 1: 193-200.
347.
Hoyle GW, Graham RM, Finkelstein JB, Nguyen KP, Gozal D, Friedman M. Hyperinnervation of the airways in transgenic mice overexpressing nerve growth factor. Am J Respir Cell Mol Biol 1998; 18: 149-157
348.
Lee KF, Li E, Huber LJ, Landis SC, Sharpe AH, Chao MV et al. Targeted mutation of the gene encoding the low affinity NGF receptor p75 leads to deficits in the peripheral sensory nervous system. Cell 1992; 69: 737-749.
349.
Graham RM, Friedman M, Hoyle GW. Sensory Nerves Promote Ozone-induced Lung Inflammation in Mice. Am J Respir Crit Care Med 2001; 164: 307-313.
350.
Tamaoki J, Chiyotani A, Tagaya E, Araake M, Nagai A. Airway hyper-responsiveness to neurokinin A and bradykinin following Mycoplasma pneumoniae infection associated with reduced epithelial neutral endopeptidase. Microbiology 1998; 144 ( Pt 9): 2481-2486.
351.
Rumsey WL, Aharony D, Bialecki RA, Abbott BM, Barthlow HG, Caccese R et al. Pharmacological Characterization of ZD6021: A Novel, Orally Active Antagonist of the Tachykinin Receptors. Journal of Pharmacology And Experimental Therapeutics 2001; 298: 307-315.
352.
Anthes JC, Chapman RW, Richard C, Eckel S, Corboz M, Hey JA et al. SCH 206272: a potent, orally active tachykinin NK(1), NK(2), and NK(3) receptor antagonist. Eur J Pharmacol 2002; 450: 191-202.
353.
Korting HC, Schindler S, Hartinger A, Kerscher M, Angerpointner T, Maibach HI. MTT-assay and neutral red release (NRR)-assay: relative role in the prediction of the irritancy potential of surfactants. Life Sci 1994; 55: 533-540.
354.
Mygind N. Nasal Allergy. 2nd ed. Oxford, London, Edinburgh, Boston, Melbourne: Blackwell Scientific Publications, 1979.
355.
Benson V, Marano MA. Current estimates from the National Health Interview Survey, 1995. Vital Health Stat 10 1998; 1-428.
356.
Bauer S, Kirschning CJ, Hacker H, Redecke V, Hausmann S, Akira S et al. Human TLR9 confers responsiveness to bacterial DNA via species-specific CpG motif recognition. PNAS 2001; 98: 9237-9242.
357.
Andrisani OM. CREB-mediated transcriptional control. Crit Rev Eukaryot Gene Expr 1999; 9: 19-32.
358.
Shaulian E, Karin M. AP-1 as a regulator of cell life and death. Nat Cell Biol 2002; 4: E131-E136.
359.
Horsley V, Pavlath GK. NFAT: ubiquitous regulator of cell differentiation and adaptation. J Cell Biol 2002; 156: 771-774.
360.
Chen F, Castranova V, Shi X, Demers LM. New insights into the role of nuclear factor-kappaB, a ubiquitous transcription factor in the initiation of diseases. Clin Chem 1999; 45: 7-17.