Skip to main content

Main menu

  • Home
  • Current issue
  • Past issues
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • Institutional open access agreements
    • Peer reviewer login
  • Alerts
  • Subscriptions
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

User menu

  • Log in
  • Subscribe
  • Contact Us
  • My Cart

Search

  • Advanced search
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

Login

European Respiratory Society

Advanced Search

  • Home
  • Current issue
  • Past issues
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • Institutional open access agreements
    • Peer reviewer login
  • Alerts
  • Subscriptions

Animal models of silicosis: fishing for new therapeutic targets and treatments

Alicia Martínez-López, Sergio Candel, Sylwia D. Tyrkalska
European Respiratory Review 2023 32: 230078; DOI: 10.1183/16000617.0078-2023
Alicia Martínez-López
1Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, Murcia, Spain
2Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
4These authors contributed equally to this work
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sergio Candel
1Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, Murcia, Spain
2Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
3Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
4These authors contributed equally to this work
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sylwia D. Tyrkalska
1Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, Murcia, Spain
2Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
3Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Sylwia D. Tyrkalska
  • For correspondence: tyrkalska.sylwia@gmail.com
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Silicosis as an occupational lung disease has been present in our lives for centuries. Research studies have already developed and implemented many animal models to study the pathogenesis and molecular basis of the disease and enabled the search for treatments. As all experimental animal models used to date have their advantages and disadvantages, there is a continuous search for a better model, which will not only accelerate basic research, but also contribute to clinical aspects and drug development. We review here, for the first time, the main animal models developed to date to study silicosis and the unique advantages of the zebrafish model that make it an optimal complement to other models. Among the main advantages of zebrafish for modelling human diseases are its ease of husbandry, low maintenance cost, external fertilisation and development, its transparency from early life, and its amenability to chemical and genetic screening. We discuss the use of zebrafish as a model of silicosis, its similarities to other animal models and the characteristics of patients at molecular and clinical levels, and show the current state of the art of inflammatory and fibrotic zebrafish models that could be used in silicosis research.

Tweetable abstract

This review discusses the use of zebrafish as a model of silicosis and its similarities to other animal models, as well as the current state of the art of inflammatory and fibrotic zebrafish models that could be used in silicosis research. https://bit.ly/3XgdDgU

Silicosis and silica crystals

Silicosis is an irreversible occupational lung disease caused by intense and prolonged inhalation of silica dust with crystals up to 10 µm in diameter in various industries, such as cutting, crushing or grinding. It belongs to the group of interstitial lung diseases called pneumoconiosis [1]. Crystallised silicon dioxide (SiO2), which is part of silica dust, is biologically active and the resulting deposition of inorganic solid waste in the bronchi, lymph nodes or pulmonary parenchyma is associated with respiratory dysfunction [2]. SiO2 is found in nature in different forms of quartz as a crystalline deposit. It is also one of the most abundant elements in Earth's crust, accounting for 59%, and is the main constituent of over 95% of all rocks [3]. Silica can occur in two forms, namely crystalline and noncrystalline (amorphous) [4]. Amorphous forms are generally considered to have low toxicity, as there are few studies suggesting adverse health effects [4]. Although inhaled silica crystals are extremely toxic to lung tissue, brief or one-time exposure is unlikely to cause significant health problems in humans [5]. Normally, crystalline particles of different sizes are inhaled together, although larger particles can be captured and expelled by natural defences through the mechanisms of coughing, sneezing and mucus production. Unfortunately, smaller crystalline particles can remain in the lung and accumulate without causing immediate discomfort, ultimately leading to disease [6].

Silicosis is one of the oldest known causes of lung disease, characterised by lung dysfunction, persistent pulmonary inflammation, formation of silicotic nodules and irreversible pulmonary fibrosis leading to respiratory failure and eventually death, with more than 12 900 deaths worldwide in 2019 [5, 7]. The prevalence and incidence of this disease have been rising, especially in developing countries, although it is also a high-profile occupational health problem in developed countries [8, 9]. The duration and intensity of exposure to crystalline silica dust are not the only determinants of its pathogenicity; host factors may also influence the susceptibility of an individual. Smaller particle size may increase the fibrogenicity of the dust. Depending on the severity of the disease, its onset and its speed of progression, silicosis is classified into chronic silicosis, accelerated silicosis, complicated silicosis and acute silicosis. The most common form is chronic silicosis, which occurs after 20 years of exposure to silica dust and is characterised by the presence of silicotic nodules that first develop in the hilar lymph nodes and then in the lungs, reaching 3 mm in diameter [10]. Each nodule has a well-demarcated central area of whorled hyalinised collagen fibres, with aggregates of dust-laden macrophages at the periphery. Over time, these lung nodules become confluent, erasing the intervening normal lung parenchyma and resulting in progressive massive fibrosis.

Accelerated silicosis occurs after 10 years of silica dust exposure and, although the changes are similar to those present in chronic silicosis, the clinical features appear sooner and the nodules develop earlier and are more cellular than fibrotic [11]. Finally, acute silicosis is characterised by different pathology from chronic silicosis, as it shows severe alveolitis and alveolar proteinosis. In addition, alveolar architecture may be preserved in this case, but with minimal fibrosis and rare small silicotic nodules [12, 13]. Patients with acute silicosis may become disabled within months of exposure.

In the early stages of the disease there may be no symptoms, meaning that the patient may be asymptomatic, with no pain and normal spirometric test results, but in the more advanced form of silicosis the person may tire easily with any kind of effort due to breathing difficulties and these symptoms may not appear until years after exposure. Over time, silica dust causes fibrous alterations in the lungs, decreasing respiratory capacity and loss of lung elasticity [14]. If the disease reaches the progressive massive fibrosis stage, exertional dyspnoea may occur [15].

The association between silicosis and tuberculosis has been known for a long time and patients with silicosis show a susceptibility to mycobacterial infections, called silicotuberculosis. The threefold increased risk of incidence is still not well understood. A possible reason may be damage to alveolar macrophages caused by silica crystals, which leads to inhibition of their ability to kill mycobacteria. Even prolonged exposure of silica, without causing silicosis, similarly increases the risk of tuberculosis [16, 17]. In addition to higher susceptibility to mycobacterial infections, patients with silicosis may present chronic bronchitis and airflow limitation, nontuberculous Mycobacterium infection, fungal lung infection, compensatory emphysema and pneumothorax as a pulmonary complication [18]. In addition, an association between silicosis and some autoimmune diseases, such as nephritis, scleroderma and systemic lupus erythematosus, especially in acute or accelerated silicosis, has been demonstrated [19]. Finally, in 1996, the International Agency for Research on Cancer classified silica crystals as a group 1 carcinogen with a strong potential to cause lung cancer in humans [20, 21].

To be diagnosed with silicosis, three key elements must be met. Initially, patient history should reveal exposure to enough silica dust to cause the disease. Next, characteristic radiological features such as chest imaging are necessary to reveal findings compatible with silicosis, usually by radiography or high-resolution computed tomography. There are standard approaches to the recognition and description of chest radiographic changes used in clinical practice. A widely accepted scheme was published by the International Labour Office, which provides a stepwise method for assessment and description of the shape, size, location and profusion of opacities that may have resulted from dust exposure; the patient's radiographs are classified after comparison with standard radiographs [22]. Finally, other conditions such as tuberculosis, sarcoidosis and histoplasmosis need to be excluded in order to determine that there are no other underlying diseases that are more likely to be causing the abnormalities [23]. Lung biopsy is rarely necessary but may be required to distinguish between progressive massive fibrosis and lung cancer. Physical examination adds nothing new but is a standard procedure. Although spirometry has most often been used as the method of first choice to assess pulmonary alterations in patients exposed to silica, it has limited sensitivity in detecting abnormalities before extensive damage occurs [24]. In addition, assessment of alveolar silica crystal load in bronchoalveolar lavage fluid may aid diagnosis.

Apart from the existing direct diagnosis of the disease, the validation of biomarkers by assessing their ability to indicate exposure, effect, disease or susceptibility has been proposed [25]. A large number of biomarkers in plasma or urine with potential to detect silicosis lung disease have already been investigated, including, among others, club cell protein 16 [26], heme oxygenase 1 [27], interleukin-6 [28], copper and zinc [29], selenium [30], and nephonectin [31]. However, it has not yet been possible to establish a single biomarker with the best potential, although it is very clear that there is a need to identify biomarkers to understand the pathogenesis of silicosis and the biological mechanisms underlying its progression.

Recently, exhaled breath condensate (EBC) has been used to assess the respiratory health of silicosis patients. The EBC is collected and then analysed for volatile and nonvolatile macromolecules, including oxidative stress, inflammatory derived biomarkers and other organic compounds [32, 33]. Interestingly, it has been shown that crystalline silica particles can be detected in the EBC of exposed patients [34]. EBC assessment is a noninvasive and highly accurate technique, making it an attractive option for the early detection of silicosis.

Treatment for silicosis

As silicosis is a chronic disease, there is no cure and once the damage is done it cannot be reversed [15]. Currently, the treatment options available are very limited and focus solely on relieving symptoms, preventing further progress of the disease and improving general condition, as well as reducing the risks of associated disorders. These include cessation of exposure to silica dust, cough suppressants, antibiotics for bacterial lung infection, chest physiotherapy to aid bronchial drainage of mucus, oxygen administration to treat hypoxaemia, bronchodilators to facilitate breathing and many others. Unfortunately, to date, pharmacological approaches have not shown a beneficial effect in halting or reversing the progression of fibrosis. The antifibrotic drugs pirfenidone and nintedanib, both currently in phase 2 of clinical trials for silicosis, are used in other fibrotic lung diseases such as idiopathic pulmonary fibrosis and are known to be effective in a wide range of progressive fibrotic lung diseases, by downregulating the production of growth factors and procollagens I and II, or inhibiting both nonreceptor tyrosine kinases (nRTKs) and receptor tyrosine kinases (RTKs), respectively [35–38]. Whole-lung lavage to remove retained dust is being investigated as a therapy for people with moderate-to-severe silicosis [39, 40]. In addition, lung transplantation, which essentially involves replacing damaged lung tissue, may be the most effective treatment. It has already been performed in some patients with advanced severe silicosis, but it is a limited resource. It is associated with serious risks inherent to lung transplant surgery, availability of ideal donor match, the consequences of long-term immunosuppression as well as the high cost of the whole procedure [41].

Several novel experimental approaches have been explored for pulmonary silicosis. Most of them consist of suppressing inflammasome activation, reducing oxidative stress by antioxidants, limiting caspase-1-driven inflammation, restoring the function of the autophagy–lysosomal system or blocking pro-inflammatory cytokines or pro-fibrotic factors [41, 42]. In addition, treatment with microRNA [43, 44], mesenchymal stem cells [45–47] or extracellular vesicles from mesenchymal stem cells [48, 49] has also shown favourable effects. Improvement of silicosis-dependent lung fibrosis has also been observed with agents increasing cyclic adenosine monophosphate (cAMP) or guanosine monophosphate (cGMP), such as inhibitors of phosphodiesterases [50, 51] or for corticosteroids [52–54]. As the importance of metabolic reprogramming of arachidonic acid, especially its derivatives prostaglandin D2 and thromboxane A2, in the progression of silicosis has been demonstrated, blocking its receptors by ramatroban may be a novel potential therapeutic therapy to inhibit the progression of silicosis [55]. In addition, several experimental studies have demonstrated a wide potential of natural plant-based compounds that may influence several of the pathological mechanisms of silicosis and thus may mitigate silica-induced inflammation as well as the process of fibrosis (table 1) [56–64].

View this table:
  • View inline
  • View popup
TABLE 1

Plant-based compounds used in different animal models of silicosis

Several methods are used to prevent exposure to respirable silica crystals and avoid the development of silicosis. First, the implementation of appropriate dust control measures in work environments is essential for prevention of the disease. Next, crystalline dust should be captured at the point of generation by enclosure and extracted by an extractor through ducts to suitable bag filters. In addition to ventilation, water-integrated tools should be used. The use of personal protective equipment as well as personal respirators that provide adequate respiratory protection must be used continuously to ensure protection at all times [65].

Animal models of silicosis

An animal model is a nonhuman species that is widely studied to understand biological process or characteristics, with the expectation that the findings obtained in this organism will allow understanding the action of other individuals. At present, animal models are widely used for the reproduction of all different types of human diseases, allowing the deeper study of physical and pathological mechanisms and the search for new therapies and treatments. Silicosis can occur spontaneously in nature and many cases have been reported in numerous mammalian species such as pigs [66], dogs [67], camels [68], horses [69, 70], nonhuman primates [71], river otters [72] and marsupials [73]. It has also been found in various species of birds like ground dwelling kiwi (Apteryx spp.) [73, 74]. Unfortunately, although this lung disease is common in many different species, not all of them can be used as experimental organisms to model silicosis in the laboratory because of difficulties in handling, manipulation and/or genetic modification, among others.

There are many different animal models used for their resemblance to human lung silicosis, as summarised in table 2; however, recently, the murine model has been widely used. The choice of experimental animal depends on the characteristics of the disease to be studied as well as the economical and organisational capacity of the experimental unit. Importantly, it has been shown that gender influences the severity of lung injury as well as the development of fibrosis, which is why male animals are often chosen for the experiments whenever possible [75, 76]. In addition, studies have shown a direct correlation between the dose and time of exposure to the silica crystalline dust and the severity of the silicotic lung disease [77, 78]. In addition, the presentation and severity of the disease has been shown to depend on the genetic predisposition of the experimental animal chosen for the study [79, 80]. Furthermore, it is important to bear in mind while performing an experiment that the artificial induction of silicosis will also depend on the form of silica and the route of exposure, as well as whether the administration of crystals is single or repeated in the experimental animal [5, 81–83]. It has also been found that the volume of material instilled can influence the severity of the disease, with animals receiving the same dose in a larger volume showing more diffuse inflammation and more severe pathology [84–86]. Finally, when the method of silica administration requires a liquid carrier, PBS is often used in experiments as a control [84, 87] and, when administered in a gaseous state, pure air is used [88].

View this table:
  • View inline
  • View popup
TABLE 2

Animal models of silicosis

Zebrafish models of silicosis

In 1995, the World Health Organization initiated a campaign with the goal of eliminating silicosis from the world by 2030; however, unfortunately, over the years, silicosis remains a major health problem internationally [89]. Therefore, there is a continuous need to generate new tools as well as to implement new experimental animal models for the study of lung silicosis.

Since 1960, zebrafish (Danio rerio) has emerged as an experimental model organism to study vertebrate developmental biology, cancer, toxicology, drug discovery, molecular genetics and, most importantly, it is nowadays widely used in biomedicine to model many different human diseases [90]. As a small tropical freshwater fish, it is ideal for use as an experimental model in the laboratory, as it ensures ease and low cost of husbandry. In addition, its physical and developmental characteristics as a vertebrate and the development and function of zebrafish organs closely resemble those of humans. There is a long list of the advantages of using zebrafish to model lung silicosis in vivo. One very important advantage is that adult female fish can produce up to 200–300 offspring per week, which makes them very suitable for high-throughput genetic studies with high efficiency and robustness. What differentiates them from mammalian models is that their embryonic development is external and the larvae are completely transparent without any feeding requirements for up to 5 days post-fertilisation [91]. During the first weeks of life, all major organ systems are formed and start to function, which can be observed under low-power microscopy due to their transparency [92]. Zebrafish embryos and larvae are relatively small, reaching millimetres in length, allowing hundreds of individuals to fit into a single Petri dish. This makes them more economically profitable at a reasonable cost than maintaining bigger experimental animals that require more space and resources. Importantly, genetic conservation between zebrafish and humans is very high, as they share 70% of all genes and 84% of all human disease genes [93]. The zebrafish genome is fully sequenced and allows for easy genetic modification. Many new genome-editing technologies have emerged and are used for creating new transgenic and mutant lines. However, as a nonmammalian species, zebrafish also have certain disadvantages for modelling human diseases. Most importantly, fish lack some mammalian organs, such as lungs. Hence, modelling lung diseases may seem impossible in this model, but fortunately it has been overcome. For example, the hindbrain ventricle situated in the back of the zebrafish head, a cavity into which immune cells can be easily recruited, mimics the alveolar environment of the human lung. It has already been used to model mycobacterial infections, coronavirus disease 2019 and silicosis in zebrafish [94–97]. In addition, gene duplication can lead to gene subfunctionalisation and neofunctionalisation, and phenotypic characteristics of diseases caused by ortholog genes may be different in zebrafish models.

Hindbrain crystalline silica dust injection

The zebrafish model of silicosis has been established by injecting crystalline silica dust resuspended in sterile PBS into the hindbrain of zebrafish larvae. This neuroepithelium-lined cavity has been used to mimic the alveolar environment that can be found in human lungs and, as a convenient injection site, allows the study of the interaction between silica crystals and host immune cells (figure 1). Normally, the hindbrain lacks macrophages and can be used to assess the recruitment of those cells to the injection site. In the case of neutrophils, although they may be present in the cavity, additional stimuli are able to enforce neutrophil recruitment as well. Because of the rapid development of zebrafish larvae and changes in larval physiology, which basically include shrinking of the hindbrain ventricle over time as well as increased rigidity of the larval epithelium, needle penetration becomes difficult or even impossible. The optimal time for this procedure is around 30–48 h post-fertilisation in zebrafish larvae. In detail, at that age zebrafish larvae need to be dechorionated, anesthetised and placed on a Petri plate to be pulled by the suction device, which is a microprobe called a VAMP (Vacuum-Assisted MicroProbe) [98]. One by one, the larvae can be immobilised and the crystalline silica dust can be injected several times into the hindbrain until the cavity is filled. Hindbrain injection can be done by directly injecting the needle into the cavity (however, this can generate a hole through which the injected liquid and crystals can escape) or it can be performed by penetration through the forebrain [95].

FIGURE 1
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1

Similarities between human pulmonary alveolus and zebrafish hindbrain ventricle.

The zebrafish model of silicosis has shown similar characteristics at the molecular level to those seen in different experimental animal models, as well as in silicotic patients. SiO2 crystals were able to induce both local and systemic inflammation in zebrafish [95]. The crystals induced significant recruitment of neutrophils and macrophages at the injection site, demonstrating that they can be recognised by the zebrafish immune system. The C-X-C motif chemokine ligand 8/C-X-C motif chemokine receptor 2 axis has been shown to be essential for the spreading of the inflammatory response induced by silica crystals. In addition, induction of proinflammatory cytokines and M1 polarisation of macrophages after silica injection was also observed in the experimental zebrafish model. This model has also revealed that silica crystals activate Toll-like receptor and Nlrp3-inflammasome signalling pathways in zebrafish. Moreover, neutrophilia and monocytosis were found in larvae injected with silica crystals, suggesting that SiO2 crystals can activate emergency myelopoiesis. Interestingly, two signalling pathways have been shown to be activated by silica crystals in zebrafish: a Caspa/Gsdme (caspase a/gasdermin e) axis that regulates myeloid cell pyroptosis and inflammation, and a Caspa/Gata1 (gata-binding protein 1) axis that promotes emergency myelopoesis [95].

Fibrotic processes also appear to be present in the zebrafish model of silicosis as a result of silica crystal injection, which start after resolution of inflammation. Thus, 5 days post-injection of silica crystals, the fibrotic markers tgfb1, acta2 (smooth muscle actin alpha 2), col1a1a (type I collagen alpha 1a), col1a2 (type I collagen alpha 2) and fn1a (fibronectin 1a) were significantly elevated at the site of injection, as were some (acta2, col1a2 and fn1a) in the whole body [95]. In addition, a histological study could be performed on silica injected fish, thus adding information about the pathophysiological mechanisms of fibrosis in this model.

Another interesting observation is the lower motility of the larvae injected with silica dust [95]. They remained in the centre of the Petri dish, in contrast to the control larvae, which were very motile and always explored the edges of the dish. In addition, the silica-injected fish showed an impaired escape response to a tactile stimulus, whereas the control larvae instantly reacted by swimming rapidly away when touched on the tail.

It should be noticed that silicotuberculosis can also be easily modelled in zebrafish by combining the already well-established mycobacterial infection model in zebrafish [94] with the new model of silicosis in zebrafish [95].

Inflammatory zebrafish transgenic reporter lines

The zebrafish model of silicosis generated by injecting silica dust into the hindbrain of zebrafish larvae can be used not only in wild-type fish, but also in many different transgenic or mutant lines, which may help to decipher the mechanisms orchestrating the pathology and constitute a unique platform for chemical screening in the future. An important characteristic of zebrafish as an experimental model is the wide variety of transgenic lines expressing fluorescent genes driven by a lineage-specific promoter, which allows real-time imaging of pathophysiological processes in vivo [99]. Of interest for the study of silicosis and its pathophysiology has been the development of stable transgenic markers for macrophages and leukocytes, as well as pro-inflammatory markers that allow simultaneous and real-time fluorescent detection of the processes occurring during the development of the disease. Here, we summarise the main representative zebrafish transgenic lines labelling different cell populations or molecules that can be used for characterisation of inflammatory events in silicosis (table 3).

View this table:
  • View inline
  • View popup
TABLE 3

Zebrafish transgenic lines useful for studying silicosis

The contribution of immune cells to the development of silicosis can be easily assessed. To study neutrophil recruitment, their total number or dispersion after silica crystal injection, the transgenic lines Tg(mpx:eGFP) and Tg(lyz:dsRED) have been generated, in which neutrophils express the fluorescent proteins eGFP or dsRED, respectively, under the control of the specific promoter myeloperoxidase (mpx) or lysozyme (lyz) promoters [100, 101]. Similarly, zebrafish transgenic lines Tg(mpeg1:eGFP) and Tg(mpeg1:mCherry) with the promoter of the macrophage-specific marker mpeg1 (macrophage-expressed gene 1) [102] can be used to study macrophage recruitment or their total number in response to SiO2 as a stimulus. From 3 days post-fertilisation, the downregulation of endogenous mpeg1 gene expression in zebrafish was observed and an additional long-term transgenic line with fluorescent macrophages was generated. Thus, the mfap4 (microfibril-associated protein 4) promoter has been used as a macrophage-specific marker leading to long-term expression of the red fluorescent protein mCherry in the line Tg(mfap4:mCherry-F) [103]. In addition, two other transgenic lines driving eGFP expression in macrophages have been generated, although they are not as widely used as the previous ones. The first uses the promoter of the immunoresponsive gene 1 (irg1), which labels macrophages only after activation Tg(irg1:eGFP) [104]. The other, Tg(fms:eGFP), uses a bacterial artificial chromosome transgene with the promoter of the zebrafish fms gene, encoding the macrophage colony stimulating factor 1 receptor, to label macrophages and skin xanthophores with eGFP [105]. Interestingly, the transgenic line Tg(coro1a:eGFP), with eGFP driven by the promoter of coronin 1a, is expressed in a full repertoire of myeloid cells, as it is highly enriched in leukocytes [106]. In addition, the zebrafish transgenic line Tg(lck:eGFP) can be used to assess the role of T-cells in the pathogenesis of silicosis by expressing eGFP under the control of the T-cell-specific tyrosine kinase (lck) promoter [107].

The production of inflammatory markers can also be tracked in vivo by fluorescent proteins. To date, several transgenic reporter lines have been generated using the promoters of pro-inflammatory molecules that drive the expression of fluorescent proteins. The nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) is considered a master regulator of inflammation and immune responses. The NF-κB reporter system in vivo, consisting of the expression of the fluorescent protein eGFP under the promoter of the master transcription factor NF-κB, can serve as a useful tool to monitor temporal and spatial patterns of activation to stimuli [108]. In fact, this line has been successfully used to demonstrate the induction of NF-κB following silica crystal injection [95].

The pro-inflammatory cytokine interleukin-1β (IL-1β) is a key mediator of the inflammatory response and can provide an important information about the inflammatory status of the studied process. A Tg(il1b:GFP-F) zebrafish line has been generated, in which the expression of farnesylated GFP, which is membrane-targeted, is driven by the il1b promoter. It has been used to study the spatiotemporal behaviour of the cells that produce the il1b transcripts in resting conditions and after stimulus [109]. Similarly to Tg(NFkB:eGFP), Tg(il1b:GFP-F) has been used to show that il1b increases locally after silica dust injection [95].

Another very useful line is the Tg(tnfa:eGFP-F) line, in which the expression of eGFP is driven after tnfa gene promoter stimuli [110]. This line can be used to track the polarisation of macrophages into M1 in vivo and to track Tnfa+ cells upon stimuli [110]. In particular, silica crystal injection into the hindbrain of larvae caused M1 polarisation of macrophages [95].

Fibrosis zebrafish transgenic reporter lines

Zebrafish transgenic lines used to study the process of fibrosis in vivo have been widely used. The promoter of the zebrafish fibrosis marker acta2 has been used to drive the expression of the green and red fluorescent proteins Tg(acta2:eGFP) and Tg(acta2:mCherry), resulting in the labelling of vascular mural cells and visceral smooth muscle cells [111]. Another useful line for modelling fibrosis in zebrafish models of silicosis is Tg(krt19:col1a2-GFP), which express Col1a2 fused to GFP under the promoter krt19, driving expression in the basal layer of epidermal cells, which are cells previously reported to be responsible for deposition of skin collagen I in early stages of zebrafish development [112]. The expression of Col1a2 is restricted to one cell type in this model and fluorescence became apparent within the skin at 2 days post-fertilisation and increased in intensity until 5 days post-fertilisation, being first observed in the dorsal region of the head. GFP–collagen was detected intracellularly in occasional epidermal cells, although it was also apparent as extracellular wispy fibrils adjacent to these cells [112]. Finally, other zebrafish lines could be designed to drive expression of the fluorescent protein with the promoters of other markers of fibrosis, such as Col1a1a, Col1a2, Fn1 and others. These transgenic lines could be used to assess this pathological process in vivo by and for high-throughput antifibrotic drug screening.

Concluding remarks and future directions

To date, silicosis lung disease has no available cure that can alter the course of the disease and no medications that can reverse lung damage. Strategies to effectively combat crystalline silica lung disease are prevention by reducing and controlling exposure to SiO2 dust. The development of drug therapies requires fundamental knowledge of the biology of the disease and the molecular basis of the pathogenesis. However, despite many inconveniences, drug development strategies to deal with lung silicosis can be classified into the following groups that offer many different drug targets, as follows: 1) targets to prevent the onset of inflammation by anti-cytokine therapy; 2) targets to prevent the onset of inflammation by blocking inflammasome activation; 3) targets to prevent the onset of inflammation by agents enhancing an autophagy–lysosomal system; 4) targets to prevent the onset of inflammation by antioxidants; 5) targets to prevent the onset of inflammation by corticosteroids; 6) use of agents that increase cAMP and cGMP; 7) use of agents that influence TGF-β-signalling; 8) use of microRNAs; and 9) targets to prevent the process of fibrosis, called anti-fibrotic drugs [50, 54, 87, 113–124]. With all these possibilities in mind, several treatments are in different stages of development, giving hope to patients with silicosis. In relation to this, the zebrafish is a great model for human health and can act as an intermediary between in vitro assays performed in cells and mammalian in vivo studies to evaluate efficacy, appropriate dose, toxicity and simultaneous assessment of different drugs [125, 126]. Unfortunately, all tests performed with cell-based methods provide limited information on the aspects of absorption, distribution, metabolism, excretion and toxicity of anti-silicotic compounds, whereas in vivo zebrafish assays often allow discovering these pharmacological aspects. The compounds tested must have the ability to be absorbed, reach the target tissue and avoid rapid metabolism and excretion in the in vivo zebrafish model. The great advantage of testing newly discovered compounds in zebrafish is that they can be rapidly tested in mammalian models in vivo with minimal optimisation of pharmacological aspects.

The entire drug discovery process is very time-consuming and the usual timeline for approval of a new therapeutic agent for any given disease can be as long as 10 years. The most promising and time-saving strategy for the search, identification and development of treatment for lung diseases is the use of drugs that have already been approved by the United States Food and Drug Administration (FDA), especially drugs involved in diseases with inflammatory and fibrotic lung processes [127]. This will shorten the path to clinical research, as the identified compounds have already been tested pharmacokinetically and undergone safety trials to be approved for human use. To date, many drug libraries are available for testing in in vitro and in vivo models [128]. Beyond the design of novel drugs and discovery and the testing of already FDA-approved drugs, screening natural products for anti-inflammatory and antifibrotic activity also seems to be a good approach [56]. Herbal compounds may be beneficial for pulmonary silicosis considering their biological relevance, characteristics and diversity. The “COlleCtion of Open NatUral producTs” (COCONUT) has been created, which contains structures and sparse annotations of more than 400 000 nonredundant natural products that can be tested in experimental animal models of human diseases [129]. In conclusion, all of the above approaches can easily be used in zebrafish models of many human diseases, such as the silicosis model, as they have already shown high efficacy in high-throughput screening of repurposed drugs [130–132] and natural or herbal products [133–135], as well as helping to discover specific therapeutic targets in humans.

Footnotes

  • Provenance: Submitted article, peer reviewed.

  • Conflict of interest: All authors have nothing to disclose.

  • Support statement: This work has been funded by Fundación Séneca (Saavedra Fajardo postdoctoral contract 21118/SF/19 to S. Candel), the Spanish Ministry of Science and Innovation (Juan de la Cierva-Incorporación postdoctoral contract to S.D. Tyrkalska) and ZEBER research grant funded by Consejería de Salud de la CARM.

  • Received April 18, 2023.
  • Accepted June 8, 2023.
  • Copyright ©The authors 2023
http://creativecommons.org/licenses/by-nc/4.0/

This version is distributed under the terms of the Creative Commons Attribution Non-Commercial Licence 4.0. For commercial reproduction rights and permissions contact permissions{at}ersnet.org

References

  1. ↵
    1. Delight N,
    2. Sachs H
    . Pneumoconiosis. Treasure Island, FL, StatPearls, 2022.
  2. ↵
    1. Skuland T,
    2. Ovrevik J,
    3. Låg M
    , et al. Role of size and surface area for pro-inflammatory responses to silica nanoparticles in epithelial lung cells: importance of exposure conditions. Toxicol In Vitro 2014; 28: 146–155. doi:10.1016/j.tiv.2013.10.018
    OpenUrl
  3. ↵
    1. Pavan C,
    2. Delle Piane M,
    3. Gullo M, et al.
    The puzzling issue of silica toxicity: are silanols bridging the gaps between surface states and pathogenicity? Part Fibre Toxicol 2019; 16: 32. doi:10.1186/s12989-019-0315-3
    OpenUrl
  4. ↵
    1. Merget R,
    2. Bauer T,
    3. Küpper HU, et al.
    Health hazards due to the inhalation of amorphous silica. Arch Toxicol 2002; 75: 625–634. doi:10.1007/s002040100266
    OpenUrlCrossRefPubMed
  5. ↵
    1. Leung CC,
    2. Yu IT,
    3. Chen W
    . Silicosis. Lancet 2012; 379: 2008–2018. doi:10.1016/S0140-6736(12)60235-9
    OpenUrlCrossRefPubMed
  6. ↵
    1. Rimal B,
    2. Greenberg AK,
    3. Rom WN
    . Basic pathogenetic mechanisms in silicosis: current understanding. Curr Opin Pulm Med 2005; 11: 169–173. doi:10.1097/01.mcp.0000152998.11335.24
    OpenUrlCrossRefPubMed
  7. ↵
    1. Chen S,
    2. Liu M,
    3. Xie F
    . Global and national burden and trends of mortality and disability-adjusted life years for silicosis, from 1990 to 2019: results from the Global Burden of Disease study 2019. BMC Pulm Med 2022; 22: 240. doi:10.1186/s12890-022-02040-9
    OpenUrl
  8. ↵
    1. Reilly MJ,
    2. Timmer SJ,
    3. Rosenman KD
    . The burden of silicosis in Michigan: 1988–2016. Ann Am Thorac Soc 2018; 15: 1404–1410. doi:10.1513/AnnalsATS.201802-117OC
    OpenUrlPubMed
  9. ↵
    1. Knight D,
    2. Ehrlich R,
    3. Fielding K, et al.
    Trends in silicosis prevalence and the healthy worker effect among gold miners in South Africa: a prevalence study with follow up of employment status. BMC Public Health 2015; 15: 1258. doi:10.1186/s12889-015-2566-8
    OpenUrl
  10. ↵
    1. Murray J,
    2. Webster I,
    3. Reid G, et al.
    The relation between fibrosis of hilar lymph glands and the development of parenchymal silicosis. Br J Ind Med 1991; 48: 267–269.
    OpenUrlPubMed
  11. ↵
    1. Hutyrová B,
    2. Smolková P,
    3. Nakládalová M, et al.
    Case of accelerated silicosis in a sandblaster. Ind Health 2015; 53: 178–183. doi:10.2486/indhealth.2013-0032
    OpenUrlPubMed
  12. ↵
    1. Ishii H,
    2. Trapnell BC,
    3. Tazawa R, et al.
    Comparative study of high-resolution CT findings between autoimmune and secondary pulmonary alveolar proteinosis. Chest 2009; 136: 1348–1355. doi:10.1378/chest.09-0097
    OpenUrlCrossRefPubMed
  13. ↵
    1. Duchange L,
    2. Brichet A,
    3. Lamblin C, et al.
    [Acute silicosis. Clinical, radiologic, functional, and cytologic characteristics of the broncho-alveolar fluids. Observations of 6 cases]. Rev Mal Respir 1998; 15: 527–534.
    OpenUrlPubMed
  14. ↵
    1. Li X,
    2. Wang Y,
    3. An G, et al.
    Bone marrow mesenchymal stem cells attenuate silica-induced pulmonary fibrosis via paracrine mechanisms. Toxicol Lett 2017; 270: 96–107. doi:10.1016/j.toxlet.2017.02.016
    OpenUrl
  15. ↵
    1. Wagner GR
    . Asbestosis and silicosis. Lancet 1997; 349: 1311–1315. doi:10.1016/S0140-6736(96)07336-9
    OpenUrlCrossRefPubMed
  16. ↵
    1. Cowie RL
    . The epidemiology of tuberculosis in gold miners with silicosis. Am J Respir Crit Care Med 1994; 150: 1460–1462. doi:10.1164/ajrccm.150.5.7952577
    OpenUrlCrossRefPubMed
  17. ↵
    1. Hnizdo E,
    2. Murray J
    . Risk of pulmonary tuberculosis relative to silicosis and exposure to silica dust in South African gold miners. Occup Environ Med 1998; 55: 496–502. doi:10.1136/oem.55.7.496
    OpenUrlAbstract/FREE Full Text
  18. ↵
    1. Rees D,
    2. Murray J
    . Silica, silicosis and tuberculosis. Int J Tuberc Lung Dis 2007; 11: 474–484.
    OpenUrlPubMed
  19. ↵
    1. Pollard KM
    . Silica, silicosis, and autoimmunity. Front Immunol 2016; 7: 97. doi:10.3389/fimmu.2016.00097
    OpenUrlCrossRefPubMed
  20. ↵
    1. Sato T,
    2. Shimosato T,
    3. Klinman DM
    . Silicosis and lung cancer: current perspectives. Lung Cancer 2018; 9: 91–101. doi:10.2147/LCTT.S156376
    OpenUrl
  21. ↵
    1. Partanen T,
    2. Pukkala E,
    3. Vainio H, et al.
    Increased incidence of lung and skin cancer in Finnish silicotic patients. J Occup Med 1994; 36: 616–622. doi:10.1097/00043764-199412000-00013
    OpenUrlPubMed
  22. ↵
    1. Muszyńska-Graca M,
    2. Dąbkowska B,
    3. Brewczyński PZ
    . [Guidelines for the use of the International Classification of Radiographs of Pneumoconioses of the International Labour Office (ILO): Substantial changes in the current edition]. Med Pr 2016; 67: 833–837. doi:10.13075/mp.5893.00493
    OpenUrl
  23. ↵
    1. Apte SH,
    2. Tan ME,
    3. Lutzky VP, et al.
    Alveolar crystal burden in stone workers with artificial stone silicosis. Respirology 2022; 27: 437–446. doi:10.1111/resp.14229
    OpenUrl
  24. ↵
    1. Chao TP,
    2. Sperandio EF,
    3. Ostolin TLVP, et al.
    Use of cardiopulmonary exercise testing to assess early ventilatory changes related to occupational particulate matter. Braz J Med Biol Res 2018; 51: e6486. doi:10.1590/1414-431x20186486
    OpenUrl
  25. ↵
    1. Schulte PA
    . The use of biomarkers in surveillance, medical screening, and intervention. Mutat Res 2005; 592: 155–163. doi:10.1016/j.mrfmmm.2005.06.019
    OpenUrlCrossRefPubMed
  26. ↵
    1. Naha N,
    2. Muhamed JCJ,
    3. Pagdhune A, et al.
    Club cell protein 16 as a biomarker for early detection of silicosis. Indian J Med Res 2020; 151: 319–325. doi:10.4103/ijmr.IJMR_1799_18
    OpenUrl
  27. ↵
    1. Sato T,
    2. Saito Y,
    3. Inoue S, et al.
    Serum heme oxygenase-1 as a marker of lung function decline in patients with chronic silicosis. J Occup Environ Med 2012; 54: 1461–1466. doi:10.1097/JOM.0b013e3182636e93
    OpenUrl
  28. ↵
    1. Liu SJ,
    2. Wang P,
    3. Jiao J, et al.
    Differential gene expression associated with inflammation in peripheral blood cells of patients with pneumoconiosis. J Occup Health 2016; 58: 373–380. doi:10.1539/joh.16-0021-OA
    OpenUrl
  29. ↵
    1. Beshir S,
    2. Aziz H,
    3. Shaheen W, et al.
    Serum levels of copper, ceruloplasmin and angiotensin converting enzyme among silicotic and non-silicotic workers. Open Access Maced J Med Sci 2015; 3: 467–473. doi:10.3889/oamjms.2015.065
    OpenUrl
  30. ↵
    1. Muzembo BA,
    2. Dumavibhat N,
    3. Ngatu NR, et al.
    Serum selenium and selenoprotein p in patients with silicosis. J Trace Elem Med Biol 2013; 27: 40–44. doi:10.1016/j.jtemb.2012.05.003
    OpenUrl
  31. ↵
    1. Lee S,
    2. Honda M,
    3. Yamamoto S, et al.
    Role of nephronectin in pathophysiology of silicosis. Int J Mol Sci 2019; 20: 2581. doi:10.3390/ijms20102581
    OpenUrl
  32. ↵
    1. Horváth I,
    2. Barnes PJ,
    3. Loukides S, et al.
    A European Respiratory Society technical standard: exhaled biomarkers in lung disease. Eur Respir J 2017; 49: 1600965. doi:10.1183/13993003.00965-2016
    OpenUrlAbstract/FREE Full Text
  33. ↵
    1. Sauni R,
    2. Oksa P,
    3. Lehtimäki L, et al.
    Increased alveolar nitric oxide and systemic inflammation markers in silica-exposed workers. Occup Environ Med 2012; 69: 256–260. doi:10.1136/oemed-2011-100347
    OpenUrlAbstract/FREE Full Text
  34. ↵
    1. Leese E,
    2. Staff JF,
    3. Carolan VA, et al.
    Exhaled breath condensate: a novel matrix for biological monitoring to assess occupational exposure to respirable crystalline silica. Ann Work Expo Health 2017; 61: 902–906. doi:10.1093/annweh/wxx047
    OpenUrl
  35. ↵
    1. Richeldi L,
    2. du Bois RM,
    3. Raghu G, et al.
    Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med 2014; 370: 2071–2082. doi:10.1056/NEJMoa1402584
    OpenUrlCrossRefPubMed
    1. Taniguchi H,
    2. Ebina M,
    3. Kondoh Y, et al.
    Pirfenidone in idiopathic pulmonary fibrosis. Eur Respir J 2010; 35: 821–829. doi:10.1183/09031936.00005209
    OpenUrlAbstract/FREE Full Text
    1. Aimo A,
    2. Spitaleri G,
    3. Nieri D, et al.
    Pirfenidone for idiopathic pulmonary fibrosis and beyond. Card Fail Rev 2022; 8: e12. doi:10.15420/cfr.2021.30
    OpenUrl
  36. ↵
    1. Wollin L,
    2. Wex E,
    3. Pautsch A, et al.
    Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J 2015; 45: 1434–1445. doi:10.1183/09031936.00174914
    OpenUrlAbstract/FREE Full Text
  37. ↵
    1. Wilt JL,
    2. Banks DE,
    3. Weissman DN, et al.
    Reduction of lung dust burden in pneumoconiosis by whole-lung lavage. J Occup Environ Med 1996; 38: 619–624. doi:10.1097/00043764-199606000-00014
    OpenUrlCrossRefPubMed
  38. ↵
    1. Chambers DC,
    2. Apte SH,
    3. Deller D, et al.
    Radiological outcomes of whole lung lavage for artificial stone-associated silicosis. Respirology 2021; 26: 501–503. doi:10.1111/resp.14018
    OpenUrl
  39. ↵
    1. McEwen K,
    2. Brodie L
    . Lung transplantation for silicosis and recovery: an Australian case study. Br J Nurs 2021; 30: 178–183. doi:10.12968/bjon.2021.30.3.178
    OpenUrl
  40. ↵
    1. Lee CM,
    2. Park JW,
    3. Cho WK, et al.
    Modifiers of TGF-β1 effector function as novel therapeutic targets of pulmonary fibrosis. Korean J Intern Med 2014; 29: 281–290. doi:10.3904/kjim.2014.29.3.281
    OpenUrlCrossRefPubMed
  41. ↵
    1. Das S,
    2. Kumar M,
    3. Negi V, et al.
    MicroRNA-326 regulates profibrotic functions of transforming growth factor-β in pulmonary fibrosis. Am J Respir Cell Mol Biol 2014; 50: 882–892. doi:10.1165/rcmb.2013-0195OC
    OpenUrlCrossRefPubMed
  42. ↵
    1. Xu T,
    2. Yan W,
    3. Wu Q, et al.
    MiR-326 inhibits inflammation and promotes autophagy in silica-induced pulmonary fibrosis through targeting TNFSF14 and PTBP1. Chem Res Toxicol 2019; 32: 2192–2203. doi:10.1021/acs.chemrestox.9b00194
    OpenUrl
  43. ↵
    1. Liu WW,
    2. Wang HX,
    3. Yu W, et al.
    Treatment of silicosis with hepatocyte growth factor-modified autologous bone marrow stromal cells: a non-randomized study with follow-up. Genet Mol Res 2015; 14: 10672–10681. doi:10.4238/2015.September.9.7
    OpenUrl
    1. Chen S,
    2. Cui G,
    3. Peng C, et al.
    Transplantation of adipose-derived mesenchymal stem cells attenuates pulmonary fibrosis of silicosis via anti-inflammatory and anti-apoptosis effects in rats. Stem Cell Res Ther 2018; 9: 110. doi:10.1186/s13287-018-0846-9
    OpenUrl
  44. ↵
    1. Li X,
    2. An G,
    3. Wang Y, et al.
    Targeted migration of bone marrow mesenchymal stem cells inhibits silica-induced pulmonary fibrosis in rats. Stem Cell Res Ther 2018; 9: 335. doi:10.1186/s13287-018-1083-y
    OpenUrl
  45. ↵
    1. Choi M,
    2. Ban T,
    3. Rhim T
    . Therapeutic use of stem cell transplantation for cell replacement or cytoprotective effect of microvesicle released from mesenchymal stem cell. Mol Cells 2014; 37: 133–139. doi:10.14348/molcells.2014.2317
    OpenUrlCrossRefPubMed
  46. ↵
    1. Phinney DG,
    2. Di Giuseppe M,
    3. Njah J, et al.
    Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nat Commun 2015; 6: 8472. doi:10.1038/ncomms9472
    OpenUrlCrossRefPubMed
  47. ↵
    1. Abdelaziz RR,
    2. Elkashef WF,
    3. Said E
    . Tadalafil reduces airway hyperactivity and protects against lung and respiratory airways dysfunction in a rat model of silicosis. Int Immunopharmacol 2016; 40: 530–541. doi:10.1016/j.intimp.2016.10.007
    OpenUrl
  48. ↵
    1. Kolb M,
    2. Raghu G,
    3. Wells AU, et al.
    Nintedanib plus sildenafil in patients with idiopathic pulmonary fibrosis. N Engl J Med 2018; 379: 1722–1731. doi:10.1056/NEJMoa1811737
    OpenUrlPubMed
  49. ↵
    1. Barbarin V,
    2. Nihoul A,
    3. Misson P, et al.
    The role of pro- and anti-inflammatory responses in silica-induced lung fibrosis. Respir Res 2005; 6: 112. doi:10.1186/1465-9921-6-112
    OpenUrlCrossRefPubMed
    1. Trentin PG,
    2. Ferreira TP,
    3. Arantes AC, et al.
    Annexin A1 mimetic peptide controls the inflammatory and fibrotic effects of silica particles in mice. Br J Pharmacol 2015; 172: 3058–3071. doi:10.1111/bph.13109
    OpenUrlCrossRefPubMed
  50. ↵
    1. Ferreira TPT,
    2. Lima JGME,
    3. Farias-Filho FA, et al.
    Intranasal flunisolide suppresses pathological alterations caused by silica particles in the lungs of mice. Front Endocrinol 2020; 11: 388. doi:10.3389/fendo.2020.00388
    OpenUrl
  51. ↵
    1. Pang J,
    2. Qi X,
    3. Luo Y, et al.
    Multi-omics study of silicosis reveals the potential therapeutic targets PGD. Theranostics 2021; 11: 2381–2394. doi:10.7150/thno.47627
    OpenUrl
  52. ↵
    1. Adamcakova J,
    2. Mokra D
    . Herbal compounds in the treatment of pulmonary silicosis. Physiol Res 2021; 70: S275–S287. doi:10.33549/physiolres.934817
    OpenUrl
    1. Zhu Z,
    2. Li Q,
    3. Xu C, et al.
    Sodium tanshinone IIA sulfonate attenuates silica-induced pulmonary fibrosis in rats via activation of the Nrf2 and thioredoxin system. Environ Toxicol Pharmacol 2020; 80: 103461. doi:10.1016/j.etap.2020.103461
    OpenUrl
    1. Liu H,
    2. Yu H,
    3. Cao Z, et al.
    Kaempferol modulates autophagy and alleviates silica-induced pulmonary fibrosis. DNA Cell Biol 2019; 38: 1418–1426. doi:10.1089/dna.2019.4941
    OpenUrl
    1. Li N,
    2. Feng F,
    3. Wu K, et al.
    Inhibitory effects of astragaloside IV on silica-induced pulmonary fibrosis via inactivating TGF-β1/Smad3 signaling. Biomed Pharmacother 2019; 119: 109387. doi:10.1016/j.biopha.2019.109387
    OpenUrl
    1. Du S,
    2. Li C,
    3. Lu Y, et al.
    Dioscin alleviates crystalline silica-induced pulmonary inflammation and fibrosis through promoting alveolar macrophage autophagy. Theranostics 2019; 9: 1878–1892. doi:10.7150/thno.29682
    OpenUrl
    1. Li C,
    2. Lu Y,
    3. Du S, et al.
    Dioscin exerts protective effects against crystalline silica-induced pulmonary fibrosis in mice. Theranostics 2017; 7: 4255–4275. doi:10.7150/thno.20270
    OpenUrlCrossRefPubMed
    1. Peng HB,
    2. Wang RX,
    3. Deng HJ, et al.
    Protective effects of oleanolic acid on oxidative stress and the expression of cytokines and collagen by the AKT/NF-κB pathway in silicotic rats. Mol Med Rep 2017; 15: 3121–3128. doi:10.3892/mmr.2017.6402
    OpenUrl
    1. Li S,
    2. Shao L,
    3. Fang J, et al.
    Hesperetin attenuates silica-induced lung injury by reducing oxidative damage and inflammatory response. Exp Ther Med 2021; 21: 297. doi:10.3892/etm.2021.9728
    OpenUrl
  53. ↵
    1. Yang T,
    2. Wang J,
    3. Pang Y, et al.
    Emodin suppresses silica-induced lung fibrosis by promoting Sirt1 signaling via direct contact. Mol Med Rep 2016; 14: 4643–4649. doi:10.3892/mmr.2016.5838
    OpenUrl
  54. ↵
    1. Kulkarni GK
    . Prevention and control of silicosis: a national challenge. Indian J Occup Environ Med 2007; 11: 95–96. doi:10.4103/0019-5278.38456
    OpenUrlPubMed
  55. ↵
    1. Roperto F,
    2. Damiano S,
    3. De Vico G, et al.
    Silicate pneumoconiosis in pigs: optical and scanning electron microscopical investigations with X-ray microanalysis. J Comp Pathol 1994; 110: 227–236. doi:10.1016/S0021-9975(08)80276-9
    OpenUrlPubMed
  56. ↵
    1. Canfield PJ,
    2. Rothwell TL,
    3. Papadimitriou JM, et al.
    Siliceous pneumoconiosis in two dogs. J Comp Pathol 1989; 100: 199–202. doi:10.1016/0021-9975(89)90131-X
    OpenUrlCrossRefPubMed
  57. ↵
    1. Hansen HJ,
    2. Jama FM,
    3. Nilsson C, et al.
    Silicate pneumoconiosis in camels (Camelus dromedarius L). Zentralbl Veterinarmed A 1989; 36: 789–796. doi:10.1111/j.1439-0442.1989.tb00793.x
    OpenUrlPubMed
  58. ↵
    1. Schwartz LW,
    2. Knight HD,
    3. Whittig LD, et al.
    Silicate pneumoconiosis and pulmonary fibrosis in horses from the Monterey-Carmel peninsula. Chest 1981; 80: Suppl. 1: 82–85. doi:10.1378/chest.80.1_Supplement.82S
    OpenUrlCrossRefPubMed
  59. ↵
    1. Arens AM,
    2. Barr B,
    3. Puchalski SM, et al.
    Osteoporosis associated with pulmonary silicosis in an equine bone fragility syndrome. Vet Pathol 2011; 48: 593–615. doi:10.1177/0300985810385151
    OpenUrlPubMed
  60. ↵
    1. Hannothiaux MH,
    2. Scharfman A,
    3. Wastiaux A, et al.
    An attempt to evaluate lung aggression in monkey silicosis: hydrolases, peroxidase and antiproteases activities in serial bronchoalveolar lavages. Eur Respir J 1991; 4: 191–204. doi:10.1183/09031936.93.04020191
    OpenUrlAbstract/FREE Full Text
  61. ↵
    1. Suedmeyer WK,
    2. Johnson G,
    3. Lovell MA
    . Pulmonary silicosis in three North American river otters (Lutra canadensis). J Zoo Wildl Med 1999; 30: 564–572.
    OpenUrlPubMed
  62. ↵
    1. Brambilla C,
    2. Abraham J,
    3. Brambilla E, et al.
    Comparative pathology of silicate pneumoconiosis. Am J Pathol 1979; 96: 149–170.
    OpenUrlPubMed
  63. ↵
    1. Evans MG,
    2. Slocombe RF,
    3. Schwartz LD
    . Pulmonary silicosis in captive ring-necked pheasants: definitive diagnosis by electron probe X-ray microanalysis. Vet Pathol 1988; 25: 239–241. doi:10.1177/030098588802500310
    OpenUrlCrossRefPubMed
  64. ↵
    1. Camp PG,
    2. Dimich-Ward H,
    3. Kennedy SM
    . Women and occupational lung disease: sex differences and gender influences on research and disease outcomes. Clin Chest Med 2004; 25: 269–279. doi:10.1016/j.ccm.2004.01.004
    OpenUrlCrossRefPubMed
  65. ↵
    1. Poinen-Rughooputh S,
    2. Rughooputh MS,
    3. Guo Y, et al.
    Sex-related differences in the risk of silicosis among Chinese pottery workers: a cohort study. J Occup Environ Med 2021; 63: 74–79. doi:10.1097/JOM.0000000000002068
    OpenUrl
  66. ↵
    1. Callis AH,
    2. Sohnle PG,
    3. Mandel GS, et al.
    Kinetics of inflammatory and fibrotic pulmonary changes in a murine model of silicosis. J Lab Clin Med 1985; 105: 547–553.
    OpenUrlPubMed
  67. ↵
    1. Davis GS,
    2. Leslie KO,
    3. Hemenway DR
    . Silicosis in mice: effects of dose, time, and genetic strain. J Environ Pathol Toxicol Oncol 1998; 17: 81–97.
    OpenUrlPubMed
  68. ↵
    1. Mayeux JM,
    2. Escalante GM,
    3. Christy JM, et al.
    Silicosis and silica-induced autoimmunity in the diversity outbred mouse. Front Immunol 2018; 9: 874. doi:10.3389/fimmu.2018.00874
    OpenUrlCrossRef
  69. ↵
    1. Brown JM,
    2. Archer AJ,
    3. Pfau JC, et al.
    Silica accelerated systemic autoimmune disease in lupus-prone New Zealand mixed mice. Clin Exp Immunol 2003; 131: 415–421. doi:10.1046/j.1365-2249.2003.02094.x
    OpenUrlCrossRefPubMed
  70. ↵
    1. Moore BB,
    2. Lawson WE,
    3. Oury TD, et al.
    Animal models of fibrotic lung disease. Am J Respir Cell Mol Biol 2013; 49: 167–179. doi:10.1165/rcmb.2013-0094TR
    OpenUrlCrossRefPubMed
    1. Lakatos HF,
    2. Burgess HA,
    3. Thatcher TH, et al.
    Oropharyngeal aspiration of a silica suspension produces a superior model of silicosis in the mouse when compared to intratracheal instillation. Exp Lung Res 2006; 32: 181–199. doi:10.1080/01902140600817465
    OpenUrlCrossRefPubMed
  71. ↵
    1. Lacher SE,
    2. Johnson C,
    3. Jessop F, et al.
    Murine pulmonary inflammation model: a comparative study of anesthesia and instillation methods. Inhal Toxicol 2010; 22: 77–83. doi:10.3109/08958370902929969
    OpenUrlCrossRefPubMed
  72. ↵
    1. Mayeux JM,
    2. Kono DH,
    3. Pollard KM
    . Development of experimental silicosis in inbred and outbred mice depends on instillation volume. Sci Rep 2019; 9: 14190. doi:10.1038/s41598-019-50725-9
    OpenUrl
    1. Driscoll KE,
    2. Costa DL,
    3. Hatch G, et al.
    Intratracheal instillation as an exposure technique for the evaluation of respiratory tract toxicity: uses and limitations. Toxicol Sci 2000; 55: 24–35. doi:10.1093/toxsci/55.1.24
    OpenUrlCrossRefPubMed
  73. ↵
    1. Helms MN,
    2. Torres-Gonzalez E,
    3. Goodson P, et al.
    Direct tracheal instillation of solutes into mouse lung. J Vis Exp 2010; 42: 1941. doi:10.3791/1941
    OpenUrlPubMed
  74. ↵
    1. Biswas R,
    2. Trout KL,
    3. Jessop F, et al.
    Imipramine blocks acute silicosis in a mouse model. Part Fibre Toxicol 2017; 14: 36. doi:10.1186/s12989-017-0217-1
    OpenUrl
  75. ↵
    1. Li T,
    2. Yang XY,
    3. Xu DJ, et al.
    OC-STAMP overexpression drives lung alveolar epithelial cell type II senescence in silicosis. Oxid Med Cell Longev 2021; 2021: 4158495.
    OpenUrl
  76. ↵
    1. Sharma N,
    2. Kundu D,
    3. Dhaked S, et al.
    Silicosis and silicotuberculosis in India. Bull World Health Organ 2016; 94: 777–778. doi:10.2471/BLT.15.163550
    OpenUrlCrossRefPubMed
  77. ↵
    1. Santoriello C,
    2. Zon LI
    . Hooked! Modeling human disease in zebrafish. J Clin Invest 2012; 122: 2337–2343. doi:10.1172/JCI60434
    OpenUrlCrossRefPubMed
  78. ↵
    1. Garcia-Moreno D,
    2. Tyrkalska SD,
    3. Valera-Pérez A, et al.
    The zebrafish: a research model to understand the evolution of vertebrate immunity. Fish Shellfish Immunol 2019; 90: 215–222. doi:10.1016/j.fsi.2019.04.067
    OpenUrl
  79. ↵
    1. White RM,
    2. Sessa A,
    3. Burke C, et al.
    Transparent adult zebrafish as a tool for in vivo transplantation analysis. Cell Stem Cell 2008; 2: 183–189. doi:10.1016/j.stem.2007.11.002
    OpenUrlCrossRefPubMed
  80. ↵
    1. Howe K,
    2. Clark MD,
    3. Torroja CF, et al.
    The zebrafish reference genome sequence and its relationship to the human genome. Nature 2013; 496: 498–503. doi:10.1038/nature12111
    OpenUrlCrossRefPubMed
  81. ↵
    1. Cambier CJ,
    2. Takaki KK,
    3. Larson RP, et al.
    Mycobacteria manipulate macrophage recruitment through coordinated use of membrane lipids. Nature 2014; 505: 218–222. doi:10.1038/nature12799
    OpenUrlCrossRefPubMed
  82. ↵
    1. Tyrkalska SD,
    2. Pedoto A,
    3. Martínez-López A, et al.
    Silica crystals activate Toll-like receptors and inflammasomes to promote local and systemic immune responses in zebrafish. Dev Comp Immunol 2023; 138: 104523. doi:10.1016/j.dci.2022.104523
    OpenUrl
    1. Tyrkalska SD,
    2. Martínez-López A,
    3. Pedoto A, et al.
    The spike protein of SARS-CoV-2 signals via TLR2 in zebrafish. Dev Comp Immunol 2023; 140: 104626. doi:10.1016/j.dci.2022.104626
    OpenUrl
  83. ↵
    1. Tyrkalska SD,
    2. Martínez-López A,
    3. Arroyo AB, et al.
    Differential proinflammatory activities of spike proteins of SARS-CoV-2 variants of concern. Sci Adv 2022; 8: eabo0732. doi:10.1126/sciadv.abo0732
    OpenUrlCrossRef
  84. ↵
    1. Takaki K,
    2. Davis JM,
    3. Winglee K, et al.
    Evaluation of the pathogenesis and treatment of Mycobacterium marinum infection in zebrafish. Nat Protoc 2013; 8: 1114–1124. doi:10.1038/nprot.2013.068
    OpenUrlCrossRefPubMed
  85. ↵
    1. Choe CP,
    2. Choi SY,
    3. Kee Y, et al.
    Transgenic fluorescent zebrafish lines that have revolutionized biomedical research. Lab Anim Res 2021; 37: 26. doi:10.1186/s42826-021-00103-2
    OpenUrl
  86. ↵
    1. Renshaw SA,
    2. Loynes CA,
    3. Trushell DM, et al.
    A transgenic zebrafish model of neutrophilic inflammation. Blood 2006; 108: 3976–3978. doi:10.1182/blood-2006-05-024075
    OpenUrlAbstract/FREE Full Text
  87. ↵
    1. Hall C,
    2. Flores MV,
    3. Storm T, et al.
    The zebrafish lysozyme C promoter drives myeloid-specific expression in transgenic fish. BMC Dev Biol 2007; 7: 42. doi:10.1186/1471-213X-7-42
    OpenUrlCrossRefPubMed
  88. ↵
    1. Ellett F,
    2. Pase L,
    3. Hayman JW, et al.
    mpeg1 promoter transgenes direct macrophage-lineage expression in zebrafish. Blood 2011; 117: e49–e56. doi:10.1182/blood-2010-10-314120
    OpenUrlAbstract/FREE Full Text
  89. ↵
    1. Walton EM,
    2. Cronan MR,
    3. Beerman RW, et al.
    The macrophage-specific promoter mfap4 allows live, long-term analysis of macrophage behavior during mycobacterial infection in zebrafish. PLoS One 2015; 10: e0138949. doi:10.1371/journal.pone.0138949
    OpenUrlCrossRefPubMed
  90. ↵
    1. Sanderson LE,
    2. Chien AT,
    3. Astin JW, et al.
    An inducible transgene reports activation of macrophages in live zebrafish larvae. Dev Comp Immunol 2015; 53: 63–69. doi:10.1016/j.dci.2015.06.013
    OpenUrlCrossRefPubMed
  91. ↵
    1. Gray C,
    2. Loynes CA,
    3. Whyte MK, et al.
    Simultaneous intravital imaging of macrophage and neutrophil behaviour during inflammation using a novel transgenic zebrafish. Thromb Haemost 2011; 105: 811–819. doi:10.1160/TH10-08-0525
    OpenUrlCrossRefPubMed
  92. ↵
    1. Li L,
    2. Yan B,
    3. Shi YQ, et al.
    Live imaging reveals differing roles of macrophages and neutrophils during zebrafish tail fin regeneration. J Biol Chem 2012; 287: 25353–25360. doi:10.1074/jbc.M112.349126
    OpenUrlAbstract/FREE Full Text
  93. ↵
    1. Langenau DM,
    2. Ferrando AA,
    3. Traver D, et al.
    In vivo tracking of T cell development, ablation, and engraftment in transgenic zebrafish. Proc Natl Acad Sci USA 2004; 101: 7369–7374. doi:10.1073/pnas.0402248101
    OpenUrlAbstract/FREE Full Text
  94. ↵
    1. Kanther M,
    2. Sun X,
    3. Mühlbauer M, et al.
    Microbial colonization induces dynamic temporal and spatial patterns of NF-κB activation in the zebrafish digestive tract. Gastroenterology 2011; 141: 197–207. doi:10.1053/j.gastro.2011.03.042
    OpenUrlCrossRefPubMed
  95. ↵
    1. Nguyen-Chi M,
    2. Phan QT,
    3. Gonzalez C, et al.
    Transient infection of the zebrafish notochord with E. coli induces chronic inflammation. Dis Model Mech 2014; 7: 871–882. doi:10.1242/dmm.014498
    OpenUrlAbstract/FREE Full Text
  96. ↵
    1. Nguyen-Chi M,
    2. Laplace-Builhe B,
    3. Travnickova J, et al.
    Identification of polarized macrophage subsets in zebrafish. Elife 2015; 4: e07288. doi:10.7554/eLife.07288
    OpenUrlCrossRefPubMed
  97. ↵
    1. Whitesell TR,
    2. Kennedy RM,
    3. Carter AD, et al.
    An α-smooth muscle actin (acta2/αsma) zebrafish transgenic line marking vascular mural cells and visceral smooth muscle cells. PLoS One 2014; 9: e90590. doi:10.1371/journal.pone.0090590
    OpenUrlCrossRefPubMed
  98. ↵
    1. Morris JL,
    2. Cross SJ,
    3. Lu Y, et al.
    Live imaging of collagen deposition during skin development and repair in a collagen I–GFP fusion transgenic zebrafish line. Dev Biol 2018; 441: 4–11. doi:10.1016/j.ydbio.2018.06.001
    OpenUrlCrossRefPubMed
  99. ↵
    1. Rosenbloom J,
    2. Macarak E,
    3. Piera-Velazquez S, et al.
    Human fibrotic diseases: current challenges in fibrosis research. Methods Mol Biol 2017; 1627: 1–23. doi:10.1007/978-1-4939-7113-8_1
    OpenUrlCrossRefPubMed
    1. Wollin L,
    2. Maillet I,
    3. Quesniaux V, et al.
    Antifibrotic and anti-inflammatory activity of the tyrosine kinase inhibitor nintedanib in experimental models of lung fibrosis. J Pharmacol Exp Ther 2014; 349: 209–220. doi:10.1124/jpet.113.208223
    OpenUrlAbstract/FREE Full Text
    1. Piguet PF,
    2. Vesin C,
    3. Grau GE, et al.
    Interleukin 1 receptor antagonist (IL-1ra) prevents or cures pulmonary fibrosis elicited in mice by bleomycin or silica. Cytokine 1993; 5: 57–61. doi:10.1016/1043-4666(93)90024-Y
    OpenUrlCrossRefPubMed
    1. Song L,
    2. Weng D,
    3. Dai W, et al.
    Th17 can regulate silica-induced lung inflammation through an IL-1β-dependent mechanism. J Cell Mol Med 2014; 18: 1773–1784. doi:10.1111/jcmm.12341
    OpenUrl
    1. Piguet PF,
    2. Vesin C
    . Treatment by human recombinant soluble TNF receptor of pulmonary fibrosis induced by bleomycin or silica in mice. Eur Respir J 1994; 7: 515–518. doi:10.1183/09031936.94.07030515
    OpenUrlAbstract/FREE Full Text
    1. Zahid A,
    2. Li B,
    3. Kombe AJK, et al.
    Pharmacological inhibitors of the NLRP3 inflammasome. Front Immunol 2019; 10: 2538. doi:10.3389/fimmu.2019.02538
    OpenUrlCrossRef
    1. Caseley EA,
    2. Poulter JA,
    3. Rodrigues F, et al.
    Inflammasome inhibition under physiological and pharmacological conditions. Genes Immun 2020; 21: 211–223. doi:10.1038/s41435-020-0104-x
    OpenUrl
    1. Tan S,
    2. Yang S,
    3. Chen G, et al.
    Trehalose alleviates apoptosis by protecting the autophagy-lysosomal system in alveolar macrophages during human silicosis. Life Sci 2020; 257: 118043. doi:10.1016/j.lfs.2020.118043
    OpenUrl
    1. Sun J,
    2. Song P,
    3. Wang Y, et al.
    Clinical efficacy of acetylcysteine combined with tetrandrine tablets in the treatment of silicosis and the effect on serum IL-6 and TNF-α. Exp Ther Med 2019; 18: 3383–3388. doi:10.3892/etm.2019.7966
    OpenUrl
    1. Zhang J,
    2. Wang Y,
    3. Zhang S, et al.
    Effects of tetrandrine combined with acetylcysteine on exercise tolerance, pulmonary function and serum TNF-β1 and MMP-7 in silicosis patients. Exp Ther Med 2020; 19: 2195–2201. doi:10.3892/etm.2020.8431
    OpenUrl
    1. Qu Y,
    2. Zhang L,
    3. Kang Z, et al.
    Ponatinib ameliorates pulmonary fibrosis by suppressing TGF-β1/Smad3 pathway. Pulm Pharmacol Ther 2015; 34: 1–7. doi:10.1016/j.pupt.2015.07.004
    OpenUrl
  100. ↵
    1. Sun J,
    2. Li Q,
    3. Lian X, et al.
    MicroRNA-29b mediates lung mesenchymal-epithelial transition and prevents lung fibrosis in the silicosis model. Mol Ther Nucleic Acids 2019; 14: 20–31. doi:10.1016/j.omtn.2018.10.017
    OpenUrlCrossRef
  101. ↵
    1. Ding CB,
    2. Zhang JP,
    3. Zhao Y, et al.
    Zebrafish as a potential model organism for drug test against hepatitis C virus. PLoS One 2011; 6: e22921. doi:10.1371/journal.pone.0022921
    OpenUrlCrossRefPubMed
  102. ↵
    1. Cassar S,
    2. Adatto I,
    3. Freeman JL, et al.
    Use of zebrafish in drug discovery toxicology. Chem Res Toxicol 2020; 33: 95–118. doi:10.1021/acs.chemrestox.9b00335
    OpenUrlCrossRef
  103. ↵
    1. Yanagihara T,
    2. Chong SG,
    3. Vierhout M, et al.
    Current models of pulmonary fibrosis for future drug discovery efforts. Expert Opin Drug Discov 2020; 15: 931–941. doi:10.1080/17460441.2020.1755252
    OpenUrl
  104. ↵
    1. Corsello SM,
    2. Bittker JA,
    3. Liu Z, et al.
    The Drug Repurposing Hub: a next-generation drug library and information resource. Nat Med 2017; 23: 405–408. doi:10.1038/nm.4306
    OpenUrlCrossRefPubMed
  105. ↵
    1. Sorokina M,
    2. Steinbeck C
    . Review on natural products databases: where to find data in 2020. J Cheminform 2020; 12: 20. doi:10.1186/s13321-020-00424-9
    OpenUrl
  106. ↵
    1. Ganzen L,
    2. Ko MJ,
    3. Zhang M, et al.
    Drug screening with zebrafish visual behavior identifies carvedilol as a potential treatment for an autosomal dominant form of retinitis pigmentosa. Sci Rep 2021; 11: 11432. doi:10.1038/s41598-021-89482-z
    OpenUrl
    1. Cousin MA,
    2. Ebbert JO,
    3. Wiinamaki AR, et al.
    Larval zebrafish model for FDA-approved drug repositioning for tobacco dependence treatment. PLoS One 2014; 9: e90467. doi:10.1371/journal.pone.0090467
    OpenUrlCrossRefPubMed
  107. ↵
    1. Rovira M,
    2. Huang W,
    3. Yusuff S, et al.
    Chemical screen identifies FDA-approved drugs and target pathways that induce precocious pancreatic endocrine differentiation. Proc Natl Acad Sci USA 2011; 108: 19264–19269. doi:10.1073/pnas.1113081108
    OpenUrlAbstract/FREE Full Text
  108. ↵
    1. Liang F,
    2. Han Y,
    3. Gao H, et al.
    Kaempferol identified by zebrafish assay and fine fractionations strategy from Dysosma versipellis inhibits angiogenesis through VEGF and FGF pathways. Sci Rep 2015; 5: 14468. doi:10.1038/srep14468
    OpenUrl
    1. Brillatz T,
    2. Lauritano C,
    3. Jacmin M, et al.
    Zebrafish-based identification of the antiseizure nucleoside inosine from the marine diatom Skeletonema marinoi. PLoS One 2018; 13: e0196195. doi:10.1371/journal.pone.0196195
    OpenUrl
  109. ↵
    1. Cheng MC,
    2. Lee TH,
    3. Chu YT, et al.
    Melanogenesis inhibitors from the rhizoma of Ligusticum sinense in B16-F10 melanoma cells in vitro and zebrafish in vivo. Int J Mol Sci 2018; 19: 3994. doi:10.3390/ijms19123994
    OpenUrl
    1. Kato K,
    2. Zemskova MA,
    3. Hanss AD, et al.
    Muc1 deficiency exacerbates pulmonary fibrosis in a mouse model of silicosis. Biochem Biophys Res Commun 2017; 493: 1230–1235. doi:10.1016/j.bbrc.2017.09.047
    OpenUrl
    1. Peukert K,
    2. Steinhagen F,
    3. Fox M, et al.
    Tetracycline ameliorates silica-induced pulmonary inflammation and fibrosis via inhibition of caspase-1. Respir Res 2022; 23: 21. doi:10.1186/s12931-022-01937-7
    OpenUrl
    1. Thakur SA,
    2. Beamer CA,
    3. Migliaccio CT, et al.
    Critical role of MARCO in crystalline silica-induced pulmonary inflammation. Toxicol Sci 2009; 108: 462–471. doi:10.1093/toxsci/kfp011
    OpenUrlCrossRefPubMed
    1. Cassel SL,
    2. Eisenbarth SC,
    3. Iyer SS, et al.
    The Nalp3 inflammasome is essential for the development of silicosis. Proc Natl Acad Sci USA 2008; 105: 9035–9040. doi:10.1073/pnas.0803933105
    OpenUrlAbstract/FREE Full Text
    1. Li B,
    2. Mu M,
    3. Sun Q, et al.
    A suitable silicosis mouse model was constructed by repeated inhalation of silica dust via nose. Toxicol Lett 2021; 353: 1–12. doi:10.1016/j.toxlet.2021.09.014
    OpenUrl
    1. Kawasaki H
    . A mechanistic review of silica-induced inhalation toxicity. Inhal Toxicol 2015; 27: 363–377. doi:10.3109/08958378.2015.1066905
    OpenUrl
    1. James DM,
    2. Morris TG,
    3. Marks J
    . Drug treatment of experimental silicosis. Br J Ind Med 1960; 17: 36–40.
    OpenUrlPubMed
    1. Feng F,
    2. Cheng P,
    3. Zhang H, et al.
    The protective role of tanshinone IIA in silicosis rat model via TGF-β1/Smad signaling suppression, NOX4 inhibition and Nrf2/ARE signaling activation. Drug Des Devel Ther 2019; 13: 4275–4290. doi:10.2147/DDDT.S230572
    OpenUrl
    1. Jiao J,
    2. Li L,
    3. Yao W, et al.
    Influence of silica exposure for lung silicosis rat. Dis Markers 2021; 2021: 6268091.
    OpenUrl
    1. Zhu Y,
    2. Yao J,
    3. Duan Y, et al.
    Protein expression profile in rat silicosis model reveals upregulation of PTPN2 and its inhibitory effect on epithelial-mesenchymal transition by dephosphorylation of STAT3. Int J Mol Sci 2020; 21: 1189. doi:10.3390/ijms21041189
    OpenUrl
    1. Sjöstrand M,
    2. Rylander R
    . Enzymes in lung lavage fluid after inhalation exposure to silica dust. Environ Res 1984; 33: 307–311. doi:10.1016/0013-9351(84)90028-8
    OpenUrlPubMed
    1. Dauber JH,
    2. Rossman MD,
    3. Pietra GG, et al.
    Experimental silicosis: morphologic and biochemical abnormalities produced by intratracheal instillation of quartz into guinea pig lungs. Am J Pathol 1980; 101: 595–612.
    OpenUrlPubMed
    1. Marks GS,
    2. Marasas LW
    . Changes in the lung lipids of rabbits and guinea-pigs exposed to the inhalation of silica dust. Br J Ind Med 1960; 17: 31–35.
    OpenUrlPubMed
    1. Schepers GW,
    2. Delahant AB,
    3. Schmidt JG, et al.
    The biological action of Degussa submicron amorphous silica dust (Dow Corning silica). III. Inhalation studies on rabbits. AMA Arch Ind Health 1957; 16: 280–301.
    OpenUrl
    1. Fallon JT
    . Specific tissue reaction to phospholipids: a suggested explanation for the similarity of the lesions of silicosis and pulmonary tubercolosis. Can Med Assoc J 1937; 36: 223–228.
    OpenUrlPubMed
    1. Mosinger M,
    2. Jouglard-Duplay J,
    3. Versino A, et al.
    [On experimental silicosis in the rabbit obtained by the intravenous administration of silica]. Arch Mal Prof 1963; 24: 503–521.
    OpenUrlPubMed
    1. Mosinger M,
    2. Jouglard-Duplay J,
    3. Versino A, et al.
    [Electrophoretic changes in rabbits receiving intravenous silica injections (statistical study)]. Arch Mal Prof 1963; 24: 881–886.
    OpenUrlPubMed
    1. Sato E,
    2. McDonald SA,
    3. Fan Y, et al.
    Analysis of particles from hamster lungs following pulmonary talc exposures: implications for pathogenicity. Part Fibre Toxicol 2020; 17: 20. doi:10.1186/s12989-020-00356-0
    OpenUrl
    1. Beck BD,
    2. Feldman HA,
    3. Brain JD, et al.
    The pulmonary toxicity of talc and granite dust as estimated from an in vivo hamster bioassay. Toxicol Appl Pharmacol 1987; 87: 222–234. doi:10.1016/0041-008X(87)90284-5
    OpenUrlCrossRefPubMed
    1. Bégin R,
    2. Massé S,
    3. Dufresne A
    . Further information on aluminium inhalation in silicosis. Occup Environ Med 1995; 52: 778–780. doi:10.1136/oem.52.11.778
    OpenUrlAbstract/FREE Full Text
    1. Bégin R,
    2. Cantin A,
    3. Massé S, et al.
    Aluminium inhalation in sheep silicosis. Int J Exp Pathol 1993; 74: 299–307.
    OpenUrlPubMed
    1. Bégin R,
    2. Dufresne A,
    3. Cantin A, et al.
    Quartz exposure, retention, and early silicosis in sheep. Exp Lung Res 1989; 15: 409–428. doi:10.3109/01902148909087868
    OpenUrlPubMed
    1. Lesur O,
    2. Melloni B,
    3. Cantin AM, et al.
    Silica-exposed lung fluids have a proliferative activity for type II epithelial cells: a study on human and sheep alveolar fluids. Exp Lung Res 1992; 18: 633–654. doi:10.3109/01902149209031699
    OpenUrlCrossRefPubMed
    1. Van Duffel J,
    2. Meersman F,
    3. Tosetti R, et al.
    [Experimental pulmonary silicosis in the dog]. G Ital Mal Torace 1967; 21: 11–23.
    OpenUrlPubMed
    1. Mosinger M,
    2. Gras A,
    3. Fiorentini H
    . [Experimental demonstrations of silicosis in the dog]. Ann Med Leg Criminol Police Sci Toxicol 1961; 41: 550–551.
    OpenUrl
    1. Hammond E,
    2. Newell JD,
    3. Dilger SK, et al.
    Computed tomography and magnetic resonance imaging for longitudinal characterization of lung structure changes in a Yucatan miniature pig silicosis model. Toxicol Pathol 2016; 44: 373–381. doi:10.1177/0192623315622303
    OpenUrlCrossRefPubMed
PreviousNext
Back to top
View this article with LENS
Vol 32 Issue 169 Table of Contents
European Respiratory Review: 32 (169)
  • Table of Contents
  • Index by author
Email

Thank you for your interest in spreading the word on European Respiratory Society .

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Animal models of silicosis: fishing for new therapeutic targets and treatments
(Your Name) has sent you a message from European Respiratory Society
(Your Name) thought you would like to see the European Respiratory Society web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
Citation Tools
Animal models of silicosis: fishing for new therapeutic targets and treatments
Alicia Martínez-López, Sergio Candel, Sylwia D. Tyrkalska
European Respiratory Review Sep 2023, 32 (169) 230078; DOI: 10.1183/16000617.0078-2023

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Animal models of silicosis: fishing for new therapeutic targets and treatments
Alicia Martínez-López, Sergio Candel, Sylwia D. Tyrkalska
European Respiratory Review Sep 2023, 32 (169) 230078; DOI: 10.1183/16000617.0078-2023
del.icio.us logo Digg logo Reddit logo Technorati logo Twitter logo CiteULike logo Connotea logo Facebook logo Google logo Mendeley logo
Full Text (PDF)

Jump To

  • Article
    • Abstract
    • Tweetable abstract
    • Silicosis and silica crystals
    • Treatment for silicosis
    • Animal models of silicosis
    • Zebrafish models of silicosis
    • Concluding remarks and future directions
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Subjects

  • Epidemiology, occupational and environmental lung disease
  • Lung biology and experimental studies
  • Tweet Widget
  • Facebook Like
  • Google Plus One

More in this TOC Section

  • Neuropsychiatric events associated with montelukast in patients with asthma
  • Methods to assess COPD medications adherence in healthcare databases
  • Sarcoidosis-associated pulmonary fibrosis: joining the dots
Show more Reviews

Related Articles

Navigate

  • Home
  • Current issue
  • Archive

About the ERR

  • Journal information
  • Editorial board
  • Press
  • Permissions and reprints
  • Advertising
  • Sponsorship

The European Respiratory Society

  • Society home
  • myERS
  • Privacy policy
  • Accessibility

ERS publications

  • European Respiratory Journal
  • ERJ Open Research
  • European Respiratory Review
  • Breathe
  • ERS books online
  • ERS Bookshop

Help

  • Feedback

For authors

  • Instructions for authors
  • Publication ethics and malpractice
  • Submit a manuscript

For readers

  • Alerts
  • Subjects
  • RSS

Subscriptions

  • Accessing the ERS publications

Contact us

European Respiratory Society
442 Glossop Road
Sheffield S10 2PX
United Kingdom
Tel: +44 114 2672860
Email: journals@ersnet.org

ISSN

Print ISSN: 0905-9180
Online ISSN: 1600-0617

Copyright © 2023 by the European Respiratory Society