This AOP is licensed under the BY-SA license. This license allows reusers to distribute, remix, adapt, and build upon the material in any medium or format, so long as attribution is given to the creator. The license allows for commercial use. If you remix, adapt, or build upon the material, you must license the modified material under identical terms.

AOP: 144

Title

A descriptive phrase which references both the Molecular Initiating Event and Adverse Outcome.It should take the form “MIE leading to AO”. For example, “Aromatase inhibition leading to reproductive dysfunction” where Aromatase inhibition is the MIE and reproductive dysfunction the AO. In cases where the MIE is unknown or undefined, the earliest known KE in the chain (i.e., furthest upstream) should be used in lieu of the MIE and it should be made clear that the stated event is a KE and not the MIE.  More help

Endocytic lysosomal uptake leading to liver fibrosis

Short name
A name that succinctly summarises the information from the title. This name should not exceed 90 characters. More help
lysosomal uptake induced liver fibrosis

Graphical Representation

A graphical representation of the AOP.This graphic should list all KEs in sequence, including the MIE (if known) and AO, and the pair-wise relationships (links or KERs) between those KEs. More help
Click to download graphical representation template Explore AOP in a Third Party Tool

Authors

The names and affiliations of the individual(s)/organisation(s) that created/developed the AOP. More help

Marina Kuburic 

Kirsten Gerloff 

Brigitte Landesmann° 

° F3 Chemical Safety and Alternative Methods Unit incorporating EURL ECVAM

Directorate F – Health, Consumers and Reference Materials

Joint Research Centre, European Commission

marinakuburic22(at)gmail.com

Kirsten-Britta.Gerloff(at)yahoo.de

brigittelandesmann(at)gmail.com

Point of Contact

The user responsible for managing the AOP entry in the AOP-KB and controlling write access to the page by defining the contributors as described in the next section.   More help
Marina Kuburic   (email point of contact)

Contributors

Users with write access to the AOP page.  Entries in this field are controlled by the Point of Contact. More help
  • Brigitte Landesmann
  • Marina Kuburic
  • Kirsten Gerloff

Coaches

This field is used to identify coaches who supported the development of the AOP.Each coach selected must be a registered author. More help

Status

Provides users with information concerning how actively the AOP page is being developed, what type of use or input the authors feel comfortable with given the current level of development, and whether it is part of the OECD AOP Development Workplan and has been reviewed and/or endorsed. OECD Status - Tracks the level of review/endorsement the AOP has been subjected to. OECD Project Number - Project number is designated and updated by the OECD. SAAOP Status - Status managed and updated by SAAOP curators. More help
Handbook Version OECD status OECD project
v1.0 EAGMST Under Review 1.47
This AOP was last modified on April 29, 2023 16:02

Revision dates for related pages

Page Revision Date/Time
N/A, Mitochondrial dysfunction 1 March 14, 2024 11:12
Cell injury/death July 15, 2022 09:46
Endocytotic lysosomal uptake December 05, 2018 08:00
Disruption, Lysosome November 12, 2018 08:23
Increased Pro-inflammatory mediators March 21, 2023 15:50
Leukocyte recruitment/activation December 01, 2017 09:33
Activation, Stellate cells November 10, 2019 05:25
Accumulation, Collagen May 17, 2023 15:55
N/A, Liver fibrosis December 05, 2018 08:29
endocytosis leads to Disruption, Lysosome November 12, 2018 10:41
Disruption, Lysosome leads to N/A, Mitochondrial dysfunction 1 November 12, 2018 10:47
N/A, Mitochondrial dysfunction 1 leads to Cell injury/death November 29, 2016 20:08
Cell injury/death leads to Increased pro-inflammatory mediators November 12, 2018 10:52
Increased pro-inflammatory mediators leads to Leukocyte recruitment/activation November 12, 2018 10:55
Leukocyte recruitment/activation leads to Activation, Stellate cells November 12, 2018 10:58
Activation, Stellate cells leads to Accumulation, Collagen December 05, 2018 08:51
Accumulation, Collagen leads to N/A, Liver fibrosis December 05, 2018 08:52
nanoparticles December 21, 2016 09:40
ROS December 21, 2016 09:40
o-methyl-serine dodecylamide hydrochloride (MSDH) December 21, 2016 09:43
3-aminopropanal December 21, 2016 09:44
artesunate December 21, 2016 09:44
Chloroquine bis(phosphate) December 04, 2018 04:25
Norfloxacin December 04, 2018 04:26
Ciprofloxacin December 04, 2018 04:26

Abstract

A concise and informative summation of the AOP under development that can stand-alone from the AOP page. The aim is to capture the highlights of the AOP and its potential scientific and regulatory relevance. More help

Hepatotoxicity is known to be an important endpoint of regulatory concern; it has been one of the frequent reasons for pharmacovigilance safety reports and human health risk assessments. Liver fibrosis, in particular, is a health problem resulting from chronic or repeated-dose chemical exposure and it is considered as an adverse outcome of regulatory interest. Liver fibrosis is a long and complex process involving various hepatic cell types, molecular mediators, receptors and signaling pathways. It occurs as a result of the imbalance between collagen deposition and destruction but also changes in the extracellular matrix composition (ECM). Due to this complexity appropriate cell model is currently unavailable.

The current AOP links endocytic lysosomal uptake and the formation of liver fibrosis. The molecular initiating event (MIE) is endocytic lysosomal uptake of chemicals, leading to lysosomal disruption, the first key event (KE). Lysosomal disruption induces mitochondrial dysfunction, which leads to cell injury and both apoptosis and necrosis. Lysosomal disruption, mitochondrial dysfunction, and cell injury/death present KEs on the cellular level. Cell death releases damage-associated molecular patterns (DAMPs) which leads to increased production of pro-inflammatory mediators, the next KE along the path. Inflammatory mediators attract and activate leukocytes, which present the next KE. Activated leukocytes through molecular mediators activate hepatic stellate cells, which increases α-SMA in them. This KE increases the amount of collagen I and III, which causes its accumulation. Collagen accumulation presents KE at the tissue level and leads to adverse outcome (AO) - liver fibrosis, which changes the normal functioning of the whole organ. There is also an important on-going process present throughout the pathway, which is connected with different KEs- oxidative stress. It is not classified as individual KE, but described in KEs and KERs related to it.

The value of this AOP is that it might support chemical risk assessment by identifying upstream biomarkers for adverse outcome, even though the adequate cell model is not available. This systematic organization of existing knowledge, but also of present uncertainties can facilitate regulatory processes, but also indicate the need for the new testing methods.

The AOP endocytic lysosomal uptake to liver fibrosis has high biological plausibility, supported with empirical evidence. However, quantitative data and temporal sequences between KEs are currently lacking and further efforts are necessary for their provision, but where temporal sequences between KEs are available, they are presented.

AOP Development Strategy

Context

Used to provide background information for AOP reviewers and users that is considered helpful in understanding the biology underlying the AOP and the motivation for its development.The background should NOT provide an overview of the AOP, its KEs or KERs, which are captured in more detail below. More help

Liver fibrosis is currently an important health problem potentially leading in its progressive form to cirrhosis and as such presents a significant economic burden (Lim and Kim, 2008). The only therapy for chronic liver failure is liver transplantation, with 5.500 liver transplantations in Europe on a yearly basis, costing up to €100.000 (approximately US$110.000) the first year (Safadi and Friedman, 2002) and estimated mean cost of US$163.438 in United States (van der Hilst et al., 2008). There are constant research attempts for new therapeutic strategies, but so far without success. AOP concept presents an alternative approach which organizing mechanistic toxicological knowledge can lead to the prevention of liver fibrosis.

The use and possible applications of nanomaterials (NMs) are in constant increase, for example in food, food-related products or cosmetics. Therefore, the safety of NMs systemically taken up in the body needs to be ensured. The liver is known to be one of the main target organs for ingested NMs, but inhaled particles can also reach the liver upon clearance from the lung (Johnston et al., 2010; Cui et al., 2011; Geraets et al., 2012). In vivo experiments on gavaged or injected (intraperitoneal or intravenously) TiO2 suggest a wide range of adverse effects on the liver: an increase in general serum markers for liver damage such as Alanine Aminotransferase or Aspartate Aminotransferase (Liu et al., 2009; Duan et al., 2010), an increase in inflammatory markers such as pro-inflammatory cytokines and/or infiltration of inflammatory cells (Ma et al., 2009; Cui et al., 2011; Kermanizadeh, 2012), an increase of markers for oxidative stress (Liu et al., 2010; Soliman et al., 2013), apoptosis, necrosis and also fibrosis (Chen et al., 2009; Alarifi et al., 2013). Oral NM administration appeared to induce overall milder adverse effects than systemic administration, most likely due to the typically limited absorption of NMs in the GI tract. Thus, it is important to keep in mind that the route of exposure and the size of the NM, play an important role whether these reach the liver, and to which extent they're accumulated (Kermanizadeh, 2014).

Once a chemical is taken up by a cell, it is transported into the lysosome. In the lysosome, the acidic environment can enhance the solubility of a NM, or it remains in the initial nano form.  Both situations can induce toxicity, causing lysosomal swelling, followed by lysosomal damage and the release of pro-apoptotic proteins (Wang et al., 2013; Cho et al., 2011; Cho et al., 2012). But not only NMs cause lysosomal damage: fluoroquinolones (Ouedraogo et al., 2000), lysosomotropic detergents such as o-methyl-serine dodecylamide hydrochloride (Villamil Giraldo et al., 2016), artesunate (Yang et al., 2014), chloroquine (Ashoor et al., 2013) can do the same, and Reactive Oxygen Species (ROS) such as H2O2 can amplify this effect (Repnik et al., 2012).  The amount of lysosomal enzymes released into the cytosol regulates the cell death pathway: controlled increased permeability of lysosomal membrane, caused by limited level of stress, plays a vital role in the induction of apoptosis, whereas massive lysosomal rupture, caused by high-stress levels, leads to necrosis (Bursch, 2001; Guicciardi et al., 2004). Lysosomes are known to trigger mitochondrial-mediated cell death by the release of cathepsins into the cytosol (Repnik et al., 2012). At the same time, however, lysosomes themselves are a source of ROS, which can lead to damage of the mitochondrial membrane (Wang et al., 2013; Kubota et al., 2010). Cell death further leads to inflammation (Faouzi et al., 2001), which activates hepatic stellate cells and induce them to secrete collagen (Casini et al., 1997). It is established that collagen accumulation is a prephase of liver fibrosis (Bataller and Brenner, 2005; Lee and Friedman, 2011).

Overall, the connection between lysosomal and mitochondrial damage with liver inflammation and further on with fibrosis, is well known, regardless if triggered by chemicals, proteins or NMs (reviewed in Malhi and Gores, 2008; Kong et al., 2014). Therefore, due to its high importance, it is described extensively in the current AOP.

Strategy

Provides a description of the approaches to the identification, screening and quality assessment of the data relevant to identification of the key events and key event relationships included in the AOP or AOP network.This information is important as a basis to support the objective/envisaged application of the AOP by the regulatory community and to facilitate the reuse of its components.  Suggested content includes a rationale for and description of the scope and focus of the data search and identification strategy/ies including the nature of preliminary scoping and/or expert input, the overall literature screening strategy and more focused literature surveys to identify additional information (including e.g., key search terms, databases and time period searched, any tools used). More help

Summary of the AOP

This section is for information that describes the overall AOP.The information described in section 1 is entered on the upper portion of an AOP page within the AOP-Wiki. This is where some background information may be provided, the structure of the AOP is described, and the KEs and KERs are listed. More help

Events:

Molecular Initiating Events (MIE)
An MIE is a specialised KE that represents the beginning (point of interaction between a prototypical stressor and the biological system) of an AOP. More help
Key Events (KE)
A measurable event within a specific biological level of organisation. More help
Adverse Outcomes (AO)
An AO is a specialized KE that represents the end (an adverse outcome of regulatory significance) of an AOP. More help
Type Event ID Title Short name
MIE 1539 Endocytotic lysosomal uptake endocytosis
KE 898 Disruption, Lysosome Disruption, Lysosome
KE 177 N/A, Mitochondrial dysfunction 1 N/A, Mitochondrial dysfunction 1
KE 55 Cell injury/death Cell injury/death
KE 1493 Increased Pro-inflammatory mediators Increased pro-inflammatory mediators
KE 1494 Leukocyte recruitment/activation Leukocyte recruitment/activation
KE 265 Activation, Stellate cells Activation, Stellate cells
KE 68 Accumulation, Collagen Accumulation, Collagen
AO 344 N/A, Liver fibrosis N/A, Liver fibrosis

Relationships Between Two Key Events (Including MIEs and AOs)

This table summarizes all of the KERs of the AOP and is populated in the AOP-Wiki as KERs are added to the AOP.Each table entry acts as a link to the individual KER description page. More help

Network View

This network graphic is automatically generated based on the information provided in the MIE(s), KEs, AO(s), KERs and Weight of Evidence (WoE) summary tables. The width of the edges representing the KERs is determined by its WoE confidence level, with thicker lines representing higher degrees of confidence. This network view also shows which KEs are shared with other AOPs. More help

Prototypical Stressors

A structured data field that can be used to identify one or more “prototypical” stressors that act through this AOP. Prototypical stressors are stressors for which responses at multiple key events have been well documented. More help

Life Stage Applicability

The life stage for which the AOP is known to be applicable. More help
Life stage Evidence
Not Otherwise Specified

Taxonomic Applicability

Latin or common names of a species or broader taxonomic grouping (e.g., class, order, family) can be selected.In many cases, individual species identified in these structured fields will be those for which the strongest evidence used in constructing the AOP was available. More help
Term Scientific Term Evidence Link
human Homo sapiens High NCBI
mouse Mus musculus High NCBI
rat Rattus norvegicus High NCBI

Sex Applicability

The sex for which the AOP is known to be applicable. More help
Sex Evidence
Unspecific

Overall Assessment of the AOP

Addressess the relevant biological domain of applicability (i.e., in terms of taxa, sex, life stage, etc.) and Weight of Evidence (WoE) for the overall AOP as a basis to consider appropriate regulatory application (e.g., priority setting, testing strategies or risk assessment). More help

Assessment of the Weight-of-Evidence supporting the AOP

Concordance of dose-response relationships

The present AOP is a purely qualitative description of the pathway. The present literature does not provide quantitative information on dose-response relationships.

Temporal concordance among the key events and adverse outcome

There is empirical evidence to support that a change in KEup leads to a change in the respective KEdown, leading to the AO.

Strength, consistency, and specificity of association of adverse outcome and initiating event

There is strong empirical evidence on the link between MIE and AO that has been described.

Biological plausibility, coherence, and consistency of the experimental evidence

There is high biological plausibility in the description of the AOP and its components. The available data describing the AOP are logic, coherent and consistent with current biological knowledge.

Uncertainties, inconsistencies and data gaps

The present AOP description is plausible but entirely qualitative, and the addition of quantitative data on dose-response relationship and temporal scale would improve its applicability. Also, though there is strong empirical evidence supporting this AOP, at certain KERs we found some inconsistencies that need to be further investigated. Existing uncertainties and inconsistencies are stated in each KER description, but here we will describe the most important ones.

Repnik and colleagues detected that after exposure to LLOMe, cathepsins remain in lysosomes and are being degraded there which is in contradiction with most of the previous studies (Repnik et al., 2017). There is strong empirical evidence that incubation of cathepsin B with mitochondria and cytosolic factors increase mitochondrial permeabilization, but in some studies pharmacological inhibition of cathepsins or knockout of genes coding for cathepsins failed to prevent mitochondrial membrane permeabilization and cell death, suggesting that other lysosomal proteases might be responsible for Bid cleavage (Reiners et al., 2002; Boya et al., 2003).

The histochemical analysis of the hippocampus of rats treated with domoic acid for 15 days has revealed no presence of apoptotic bodies and no Fluoro-Jade B positive cells (Schwarz et al., 2014).

The inflammatory role of HMGB-1 is still not completely clear. There are many studies that confirm its pro-inflammatory activity, but in some experiments, highly purified HMGB-1 had little pro-inflammatory activity (Rouhiainen et al., 2007), while in another injection of recombinant HMGB-1 into infarcted heart muscle in vivo stimulated regeneration and repair (Limana et al., 2005).

Lloyd and colleagues found that several chemokines can stimulate the adherence of peripheral blood lymphocytes to ICAM-1 coated slides (Loyd et al., 1996). However, by using a parallel plate flow chamber, another study failed to observe such an effect (Carr et al., 1996).

Domain of Applicability

Addressess the relevant biological domain(s) of applicability in terms of sex, life-stage, taxa, and other aspects of biological context. More help

Life Stage Applicability

The described AOP is a general mechanism, that furthermore can be considered as not being limited to only the liver as the target organ. Also, to current knowledge, this AOP is not limited to a specific life stage.

Taxonomic Applicability

Most of the work performed to elaborate parts of this AOP was done using murine or human cells and cell lines, human blood samples or tissues, or mouse models. Examples include

mouse: Narumi et al., 1992; Fahy et al., 2001; Kagedal et al., 2001; Faouzi et al., 2001; Werneburg et al., 2002; Chen et al., 2007; Seki et al., 2007; Leung et al., 2008; Dalton et al., 2009; Lee et al., 2009 Gäbele et al., 2009; Nan et al., 2013; Pradere et al., 2013; McHedlidze et al., 2014; Chang et al., 2014;

rat: Rockey et al., 1992;George et al., 1999; Reeves et al., 2000; Luckey and Petersen, 2001; Duffield et al., 2005; Imamura et al., 2005; Natajaran et al., 2006; Liedtke et al., 2011; Li et al., 2012; Jung et al., 2015

human: Bleul et al., 1996; Miyamoto et al., 2000; Andersson et al., 2000; Yamada et al., 2001;  Scaffidi et al., 2002; Safadi and Friedman, 2002; Boya et al., 2003; Cirman et al., 2004; Bataller and Brenner, 2005; Bell et al., 2006; Rockey and Friedman, 2006; Friedman, 2008; Lim and Kim, 2008; Winklhofer and Haass, 2010; Clarke et al., 2010; Hamacher-Brady et al., 2011; Santibañez et al., 2011; Lee and Friedman, 2011; Wang et al., 2013; Loos et al., 2014; Decaris et al., 2015; Sun et al., 2015;

Sex Applicability

As described above, the AOP is widely applicable, therefore no specific sex applicability is known at this point.

Essentiality of the Key Events

The essentiality of KEs can only be assessed relative to the impact of manipulation of a given KE (e.g., experimentally blocking or exacerbating the event) on the downstream sequence of KEs defined for the AOP. Consequently, evidence supporting essentiality is assembled on the AOP page, rather than on the independent KE pages that are meant to stand-alone as modular units without reference to other KEs in the sequence. The nature of experimental evidence that is relevant to assessing essentiality relates to the impact on downstream KEs and the AO if upstream KEs are prevented or modified. This includes: Direct evidence: directly measured experimental support that blocking or preventing a KE prevents or impacts downstream KEs in the pathway in the expected fashion. Indirect evidence: evidence that modulation or attenuation in the magnitude of impact on a specific KE (increased effect or decreased effect) is associated with corresponding changes (increases or decreases) in the magnitude or frequency of one or more downstream KEs. More help

The essentiality of almost all of the KEs in this AOP was rated high as there is much experimental evidence that the blocking of one KE prevents the next downstream KE and therefore the whole AOP. The essentiality of KE Mitochondrial dysfunction was rated as moderate, as there are two pathways to apoptosis (the next downstream KE), intrinsic and extrinsic, and only intrinsic pathway includes mitochondrial dysfunction (reviewed in Elmore, 2007).

Some of the evidence for the essentiality of KEs is listed below.

Exposure of cells to ammonium chloride prior to exposure to sphingosine resulted in the formation of NH3, which entered into lysosomes and became protonated and increased the pH of the organelle. This exposure prevented the accumulation of sphingosine in the lysosome and provided protection against its lysosomolytic and apoptosis-inducing effects (Kagedal et al., 2001).

Inhibition of lysosomal membrane permeabilization (LMP) with Baf A1 – that inhibits lysosomal vacuolar H+ ATPase, prevented mitochondrial membrane permeabilization (MMP) - the next downstream KE, while inhibition of MMP in Bax/Bak double knocks out cells didn't prevent LMP (Boya et al., 2003).

Faouzi and colleagues showed that the inhibition of apoptosis causes inhibition of the inflammatory response (Faouzi et al., 2001), while other study showed that an inhibitor of apoptosis is blocking the release of HMGB-1 mediator specifically (Bell et al., 2006).  

A human CXC chemokine antagonist, growth-related oncogene GROα(8-73), inhibited calcium mobilization induced by MIP-2, and the pretreatment of mice with this antagonist inhibited, in a dose-dependent manner, the influx of neutrophils induced by MIP-2, TNF-α, LPS and IL-1β (McColl and Clark-Lewis, 1999).

There is strong evidence that the blockade of TGF-β alone is sufficient to completely block experimental fibrogenesis in the liver (reviewed in Gressner et al., 2002). The activation of HSCs can be partially blocked by anti-TGF-β antibodies (Zimmermann et al., 2010) and blocked by overexpression of Smad7, a natural antagonist of TGF-β signaling (Dooley et al., 2003).  Macrophage depletion led to a significant reduction in the number of HSCs (Duffield et al., 2005).

Evidence Assessment

Addressess the biological plausibility, empirical support, and quantitative understanding from each KER in an AOP. More help

Summary Table Provide an overall summary of the weight of evidence based on the evaluations of the individual linkages from the Key Event Relationship pages.

Support for essentiality of KEs

MIE Endocytic lysosomal uptake

Essentiality of the MIE is high.

Exposure of cells to NH3 prevented the accumulation of sphingosine, known lysosomotropic agent, in the lysosomes and provided protection against its lysosomolytic and apoptotic effects (Kagedal et al., 2001).

KE1 Lysosomal disruption

Essentiality of the KE1 is high.

Inhibition of LMP prevented MMP, the next downstream KE, and the entire pathway (Boya et al., 2003). The treatment of the cells with cathepsins inhibitors decreased MMP and prevented apoptosis (Roberg et al., 1999; Kagedal et al., 2001).

KE2 Mitochondrial dysfunction

Essentiality of the KE2 is moderate.

There are two apoptotic pathways, intrinsic and extrinsic, and only intrinsic pathway includes mitochondrial dysfunction (reviewed in Elmore, 2007).

KE3 Cell death/injury

Essentiality of the KE3 is high.

The inhibition of apoptosis causes blockage of the release of inflammatory mediators and inhibition of the inflammatory response (Faouzi et al., 2001; Bell et al., 2006).

KE4 Increased inflammatory mediators

Essentiality of the KE4 is high.

Chemokine antagonist can prevent the influx of neutrophils (McColl and Clark-Lewis, 1999).

KE5 Leukocyte recruitment/ activation

Essentiality of the KE5 is high.

Macrophage depletion leads to a significant reduction in the number of HSCs, the next downstream KE (Duffield et al., 2005). After bile duct ligation, Kupffer cell-depleted mice showed almost complete suppression of HSC activation and fibrosis (Seki et al., 2007).

KE6 HSC activation

Essentiality of the KE6 is high.

Experimental inhibition of HSC activation prevents fibrosis (Friedman, 2002; Anan et al., 2006; Kisseleva and Brenner, 2008; Son et al., 2009).

KE7 Collagen accumulation

Essentiality of the KE7 is high.

The continuing imbalance between the deposition and degradation of ECM is a pre-requisite of liver fibrosis (Lee and Friedman, 2011).

AO Liver fibrosis

It is generally accepted that any chronic and repeated liver injury can result in myofibroblast activation, leading to hepatic fibrosis and cirrhosis (Jaeschke, 2002; Lee William, 2003; Ramachandran and Kakar, 2009).

Support for Biological Plausibility of KERs

MIE->KE1

Biological plausibility of the KER between MIE and KE1 is high.

Trapping of lysosomotropic agents accumulates substances inside of the lysosomes, increases the volume of these organelles, and big lysosomes are more prone to rupture (Ono et al., 2003).

KE1->KE2

Biological plausibility of the KER between KE1 and KE2 is high.

In the last decade, there is a growing body of evidence about the strong functional link between lysosomes and mitochondria that play an important role in physiology and pathology (e.g. Todkar et al., 2017). The evidence also showed a link between lysosomal and mitochondrial damage, and that lysosomal damage precedes mitochondrial injury (e.g. Droga-Mazovec et al., 2008; Ghosh et al., 2011).

KE2->KE3

Biological plausibility of the KER between KE2 and KE3 is high.

There is functional mechanistic understanding supporting this relationship between KE2 and KE3, involving ROS formation, ATP depletion and apoptogenic factors (Richter et al., 1996; Leist et al., 1997; Nicotera et al., 1998; Brenner and Mak, 2009; Lu et al., 2014; Zhou et al., 2015).

KE3->KE4

Biological plausibility of the KER between KE3 and KE4 is high.

The dead cells can secrete inflammatory mediators that trigger the infiltration of immune cells. However, if this becomes chronic it has the potential to enhance tissue damage and ultimately induce fibrosis (Jaeschke, 2002; Cullen et al., 2013).

KE4->KE5

Biological plausibility of the KER between KE4 and KE5 is high.

There is much evidence that application of chemokines attracts leukocytes to a specific site in different species (Beck et al., 1997; Lee et al., 2000; Fahy et al., 2001; Nikiforou et al., 2016).

KE5->KE6

Biological plausibility of the KER between KE5 and KE6 is high.

The recruitment of immune cells from the circulation into the injured tissue is the key mechanism during fibrogenesis in the liver (Heymann and Tacke, 2016).

KE6->KE7

Biological plausibility of the KER between KE6 and KE7 is high.

The functional relationship between stellate cells activation and collagen accumulation is consistent with established biological knowledge (Milani et al., 1994; Benyon and Arthur; 2001; Safadi and Friedman, 2002; Kershenobich Stalnikowitz and Weisssbrod , 2003; Bataller and Brenner, 2005; Kolios et al., 2006; ; Guo and Friedman, 2007; Li, Jing-Ting et al., 2008; López-Novoa and Nieto, 2009; Lee und Friedman 2011).

KE7->AO

Biological plausibility of the KER between KE7 and AO is high.

By definition, liver fibrosis is the excessive accumulation of ECM proteins that are produced by HSCs. The KER between this KE and the AO is undisputed (Poynard et al., 1997; Bataller and Brenner, 2005; Rockey and Friedman, 2006; Brancatelli et al., 2009; Lee and Friedman, 2011).

Empirical Support for KERs

MIE->KE1

Empirical support of the KER between MIE and KE1 is high.

Even the accumulation of substances that are physiologically present in lysosomes, such as pro-cathepsins, can lead to lysosomal dysfunction (Jung et al., 2015). Sphingosine is a lysosomotropic agent that accumulates within the lysosomes, where it permeabilizes the membrane via a detergent mechanism and provokes the release of lysosomal enzymes (Kagedal et al., 2001). NM captured in lysosomes can cause lysosomal swelling, disruption and the release of pro-apoptotic proteins (Cho et al., 2011; Cho et al., 2012; Wang et al., 2013).

KE1->KE2

Empirical support of the KER between KE1 and KE2 is high.

LMP is detected a couple of hours earlier than MMP, after exposure to ciprofloxacine, norfloxacine and hydroxychloroquine, and cells with MMP are sub-ensemble of the group of cells with LMP (Boya et al., 2003).

When isolated mitochondria are incubated with purified cathepsin B in the presence of cytolic extracts, a release of cytochrome c from mitochondria is detected (Guicciardi et al., 2000). The microinjection of cathepsin D to the cell causes cytochrome c release, caspases activation, and apoptosis (Roberg et al., 2002).

Bid protein needs to be cleaved in order to cause cytochrome c release (Luo et al., 1998; Gross et al., 1999; Stoka et al., 2001).

Incubation of Bax with isolated mitochondria resulted in cytochrome c release while Bcl-xl inhibits this release (Jurgensmeier et al., 1998).

KE2->KE3

Empirical support of the KER between KE2 and KE3 is moderate.

Mice injected intraperitoneally with DomA have shown an increase of the TUNEL positive cells in the hippocampus, decreased indicators of mitochondria function and elevated ROS levels (Lu et al., 2012, Wu et al., 2012). The incidence of downstream KE (cell death) is higher than the incidence of downstream KE (mitochondrial dysfunction).

In vitro studies (Giordano et al., 2007; 2009) suggest that both KEs are affected by the same dose of DomA and that the incidence of KE down (cell death) is higher than the incidence of KE up (mitochondrial dysfunction), with KE up happening earlier than KE down.

KE3->KE4

Empirical support of the KER between KE3 and KE4 is high.

During the apoptosis of Jurkat cells treated with various agents, HMGB-1 was released into the media (Bell et al., 2006), which was found to activate leukocytes and stimulate the production of pro-inflammatory mediators in vitro (Li et al., 2004; Zimmermann et al., 2004). ATP can also stimulate the production of pro-inflammatory cytokines from macrophages (Ferrari et al., 1997; Ferrari et al., 2006).

Neutralization or genetic deficiency of IL-1 inhibited inflammation responses to injected dead cells (Chen et al., 2007; Kono et al., 2010). Injection into mice of a variety of other dead cell types that genetically lack both IL-1α and IL- 1β stimulated an inflammatory response that was equivalent to that of wildtype necrotic cells (Kono et al., 2010). This implicates that IL-1 that is driving the sterile inflammatory response in many cases is not coming directly from the dead cell but is produced by cells in the host upon recognition of cell death.

KE4->KE5

Empirical support of the KER between KE4 and KE5 is high.

The exposure of mice to FliCind strain S. Typhimurium triggered a significant neutrophil influx in the spleen of wild-type mice, but not of Il1b−/−Il18−/− mice (Jorgensen et al., 2016).

It was shown that exposure of cells to IL-1β, TNF-α, and IFN-γ resulted in the induction of RANTES mRNA and protein (Ortiz et al., 1996; Miyamoto et al., 2000). Intradermal injection of RANTES induces a potent T-lymphocyte and eosinophils recruitment (Fahy et al., 2001; Beck et al., 1997).  Intradermal administration of MIP-1a resulted in the accumulation of monocytes, lymphocytes, eosinophils, and the recruitment of neutrophils (Lee et al., 2000).

The number of white blood cells, monocytes, and neutrophils were increased in cord blood after 6 days of IL-1α exposure (Nikiforou et al., 2016).

KE5->KE6

Empirical support of the KER between KE5 and KE6 is moderate.

Karlmark et al., 2009 found that intrahepatic CD11bF4/80 monocyte-derived cells and liver resident macrophages produce TGF-β1 and thereby directly activate HSCs. It was proven that the treatment of cultured hepatic cells with TGF-β1 increased type I pro-collagen mRNA levels (Czaja et al., 1989).

KE6->KE7

Empirical support of the KER between KE6 and KE7 is moderate.

It is difficult to stimulate sufficient collagen production and its incorporation into a pericellular matrix in vitro. Because of that analytical methods have focused on the measurement of pro-collagen secreted into culture medium or measurement of α-smooth muscle actin (α-SMA) expression, a marker of fibroblast activation. In primary culture, HSCs from normal liver begin to express α-SMA coincident with culture-induced activation (Rockey et al., 1992; Chen and Raghunath, 2009).

KE7->AO

Empirical support of the KER between KE7 and AO is high.

There is a smooth transition from collagen accumulation to liver fibrosis without a definite threshold with plenty in vivo evidence (Poynard et al., 1997; Bataller and Brenner, 2005; Rockey and Friedman, 2006; Brancatelli et al., 2009; Lee and Friedman, 2011).

Known Modulating Factors

Modulating factors (MFs) may alter the shape of the response-response function that describes the quantitative relationship between two KES, thus having an impact on the progression of the pathway or the severity of the AO.The evidence supporting the influence of various modulating factors is assembled within the individual KERs. More help

Quantitative Understanding

Optional field to provide quantitative weight of evidence descriptors.  More help

The quantitative understanding of the AOP is low, as there is a lack of quantitative data on the dose-response relationship. Further research efforts should be made in this direction to improve the quantitative understanding of the present AOP.

Considerations for Potential Applications of the AOP (optional)

Addressess potential applications of an AOP to support regulatory decision-making.This may include, for example, possible utility for test guideline development or refinement, development of integrated testing and assessment approaches, development of (Q)SARs / or chemical profilers to facilitate the grouping of chemicals for subsequent read-across, screening level hazard assessments or even risk assessment. More help

References

List of the literature that was cited for this AOP. More help

Alarifi S, Ali D, Al-Doaiss AA, Ali BA, Ahmed M, Al-Khedhairy AA. Histologic and apoptotic changes induced by titanium dioxide nanoparticles in the livers of rats. Int. J. Nanomedicine (2013) 8:3937–43.

Anan A, Baskin-Bey ES, Bronk SF, Werneburg NW, Shah VH, Gores GJ. Proteasome inhibition induces hepatic stellate cell apoptosis. Hepatology (2006) 43(2): 335-344.

Andersson U, Wang H, Palmblad K, Aveberger AC, Bloom O, Erlandsson-Harris H, Janson A, Kokkola R, Zhang M, Yang H, Tracey KJ. High mobility group 1 protein (HMG-1) stimulates proinflammatory cytokine synthesis in human monocytes. J. Exp. Med. (2000) 192:565–70.

Ashoor R, Yafawi R, Jessen B, Lu S. The Contribution of Lysosomotropism to Autophagy Perturbation. (2013) PLoS ONE 8(11): e82481.

Bataller R, Brenner DA. Liver Fibrosis. J.Clin. Invest. (2005) 115(2):209-218.

Beck LA, Dalke S, Leiferman KM, Bickel CA, Hamilton R, Rosen H, Bochner BS, Schleimer RP. Cutaneous injection of RANTES causes eosinophil recruitment: comparison of nonallergic and allergic human subjects. J. Immunol. (1997) 159:2962–2972.

Bell CW, Jiang W, Reich CF, Pisetsky DS. The extracellular release of HMGB1 during apoptotic cell death.  Am. J. Physiol. Cell. Physiol. (2006) 291(6): C1318–C1325.

Benyon RC, Arthur MJ (2001), Extracellular matrix degradation and the role of stellate cells. Semin. Liver Dis. (2001) 21(3):373-384.

Bleul CC, Fuhlbrigge RC, Casasnovas JM, Aiuti A, Springer TA. A highly efficacious lymphocyte chemoattractant, stromal cell-derived factor 1 (SDF-1). J. Exp. Med. (1996) 184(3): 1101–1109.

Boya P, Andreau K, Poncet D, Zamzami N, Perfettini JL, Metivier D, Ojcius DM, Jäättelä M, Kroemer G. Lysosomal membrane permeabilization induces cell death in a mitochondrion-dependent fashion. J. Exp. Med. (2003) 197:1323–1334.

Brancatelli G, Baron RL, Federle MP, Sparacia G, Pealer K. Focal confluent fibrosis in cirrhotic liver: natural history studied with serial CT. AJR Am. J. Roentgenol. (2009) 192 (5): 1341-1347.

Brenner D, Mak TW. Mitochondrial cell death effectors.  Curr. Opin. Cell. Biol. (2009) 21(6): 871–877.

Bursch W. The autophagosomal-lysosomal compartment in programmed cell death. Cell Death Differ. (2001) 8(6):569-81

Carr MW, Alon R, Springer TA. The C-C chemokine MCP-1 differentially modulates the avidity of beta 1 and beta 2 integrins on T lymphocytes. Immunity (1996) 4: 179–187.

Chang W, Yang M, Song L, Shen K, Wang H, Gao X, Li M, Niu W, Qin X. Isolation and culture of hepatic stellate cells from mouse liver. Acta Biochim. Biophys. Sin. (Shanghai). (2014) 46(4):291-8.

Chaudhry Q, Scotter M, Blackburn J, Ross B, Boxall A, Castle L, Aitken R, Watkins R. Applications and implications of nanotechnologies for the food sector. Food Addit. Contam. Part A. Chem. Anal. Control Expo. Risk. Assess. (2008) 25:241–58.

Chen CJ, Kono H, Golenbock D, Reed G, Akira S, Rock KL. Identification of a key pathway required for the sterile inflammatory response triggered by dying cells. Nat. Med. (2007) 13:851–856.

Chen J, Dong X, Zhao J, Tang G. In vivo acute toxicity of titanium dioxide nanoparticles to mice after intraperitioneal injection. J. Appl. Toxicol. (2009) 29:330–7.

Chen C, Raghunath M. Focus on collagen: in vitro systems to study fibrogenesis and antifibrosis state of the art. Fibrogenesis Tissue Repair (2009) 15 (2):7.

Cho W-S, Duffin R, Howie SEM, Scotton CJ, Wallace WAH, Macnee W, Bradley M, Megson IL, Donaldson K. Progressive severe lung injury by zinc oxide nanoparticles; the role of Zn2+ dissolution inside lysosomes. Part. Fibre Toxicol. (2011) 8:27.

Cho W-S, Duffin R, Thielbeer F, Bradley M, Megson IL, MacNee W, Poland CA, Tran CL, Donaldson K. Zeta potential and solubility to toxic ions as mechanisms of lung inflammation caused by metal/metal oxide nanoparticles. Toxicol. Sci. (2012) 126:469–477.

Cirman T, Orešić K, Mazovec GD, Turk V, Reed JC, Myers RM, Salvesen GS, Turk B. Selective disruption of lysosomes in HeLa cells triggers apoptosis mediated by cleavage of Bid by multiple papain-like lysosomal cathepsins, J. Biol. Chem.  (2004) 279: 3578–3587.

Clarke MCH, Talib S, Fig NL, Bennet MR. Vascular Smooth Muscle Cell Apoptosis Induces Interleukin-1–Directed Inflammation, Effects of Hyperlipidemia-Mediated Inhibition of Phagocytosis. Circ. Res. (2010) 106(2): 363–372.

Cui Y, Liu H, Zhou M, Duan Y, Li N, Gong X, Hu R, Hong M, Hong F. Signaling pathway of inflammatory responses in the mouse liver caused by TiO2 nanoparticles. J. Biomed. Mater. Res. A. (2011) 96:221–9.

Cullen SP, Henry CM, Kearney CJ, Logue SE, Feoktistova M, Tynan GA, Lavelle EC, Leverkus M, Martin SJ. Fas/CD95-induced chemokines can serve as "find-me" signals for apoptotic cells. Mol. Cell. (2013) 49(6):1034-48.

Czaja MJ, Weiner FR, Flanders KC, Giambrone MA, Wind R, Biempica L, Zern MA. (1989), In vitro and in vivo association of transforming growth factor-beta 1 with hepatic fibrosis. J. Cell. Biol. (1989) 108(6): 2477-2482.

Dalton SR, Lee SM, King RN, Nanji AA, Kharbanda KK, Casey CA, McVicker BL. Carbon tetrachloride-induced liver damage in asialoglycoprotein receptor-deficient mice. Biochem. Pharmacol. (2009) 77(7): 1283-1290.

Decaris ML, Emson CL, Li K, Gatmaitan M, Luo F, Cattin J, Nakamura C, Holmes WE, Angel TE, Peters MG, Turner SM, Hellerstein MK. Turnover rates of hepatic collagen and circulating collagen- associated proteins in humans with chronic liver disease. PLoS One (2015) 10 (4) e0123311.

Dekkers S, Krystek P, Peters RJB, Lankveld DPK, Bokkers BGH, van Hoeven-Arentzen PH, Bouwmeester H, Oomen AG. Presence and risks of nanosilica in food products. Nanotoxicology (2011) 5:393–405.

Duan Y, Liu J, Ma L, Li N, Liu H, Wang J, Zheng L, Liu C, Wang X, Zhao X, Yan J, Wang S, Wang H, Zhang X, Hong F. Toxicological characteristics of nanoparticulate anatase titanium dioxide in mice. Biomaterials (2010) 31:894–9.

Duffield JS, Forbes SJ, Constandinou CM, Clay S, Partolina M, Vuthoori S, Wu S, Lang R, Iredale JP. Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J. Clin. Invest. (2005) 115:56-65.

Dooley S, Hamzavi J, Breitkopf K, Wiercinska E, Said HM, Lorenzen J, Ten Dijke P, Gressner AM. Smad7 prevents activation of hepatic stellate cells and liver fibrosis in rats. Gastroenterology (2003) 125: 178–191.

Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. (2007) 35(4):495-516.

Fahy O, Porte H, Sénéchal S, Vorng H, McEuen AR, Buckley MG, Walls AF, Wallaert B, Tonnel AB, Tsicopoulos A. Chemokine-induced cutaneous inflammatory cell infiltration in a model of Hu-PBMC-SCID mice grafted with human skin. Am. J. Pathol. (2001) 158(3):1053-63.

Faouzi S, Burckhardt BE, Hanson JC, Campe CB, Schrum LW, Rippe RA, Maher JJ. Anti-Fas induces hepatic chemokines and promotes inflammation by an NF-B-independent, caspase-3-dependent pathway. J. Biol. Chem. (2001) 276:49077-49082.

Ferrari D, Chiozzi P, Falzoni S, Dal Suino M, Melchiorri L, Baricordi OR, Di Virgilio F. Extracellular ATP triggers IL- 1 beta release by activating the purinergic P2Z receptor of human macrophages. J. Immunol. (1997) 159:1451–1458.

Ferrari D, Pizzirani C, Adinolfi E, Lemoli RM, Curti A, Idzko M, Panther E, Di Virgilio F. The P2X7 receptor: a key player in IL-1 processing and release. J. Immunol. (2006) 176:3877–3883.

Friedman SL. Hepatic Fibrosis-Role of Hepatic Stellate Cell Activation, Med. Gen. Med.(2002)  4(3):27.

Friedman SL. Mechanisms of Hepatic Fibrogenesis. Gastroenterology (2008)  134(6): 1655–1669.

Gäbele E, Froh M, Arteel GE, Uesugi T, Hellerbrand C, Schölmerich J, Brenner DA, Thurman RG, Rippeb RA. TNFalpha is required for cholestasis-induced liver fibrosis in the mouse. Biochem. Biophys. Res. Commun. (2008) 378(3):348-53.

George J, Roulot D, Koteliansky VE, Bissell DM. In vivo inhibition of rat stellate cell activation by soluble transforming growth factor beta type II receptor: a potential new therapy for hepatic fibrosis. Proc. Natl. Acad. Sci. (1999) 96(22): 12719-12724.

Geraets L, Oomen AG, Schroeter JD, Coleman VA, Cassee FR. Tissue distribution of inhaled micro- and nano-sized cerium oxide particles in rats: results from a 28-day exposure study. Toxicol. Sci. (2012) 127:463–73.

Giordano G, White CC, Mohar I, Kavanagh TJ, Costa LG. Glutathione levels modulate domoic acid-induced apoptosis in mouse cerebellar granule cells. Toxicol. Sci. (2007) 100: 433-444.

Giordano G, Li L, White CC, Farin FM, Wilkerson HW, Kavanagh TJ, Costa LG. Muscarinic receptors prevent oxidative stress-mediated apoptosis induced by domoic acid in mouse cerebellar granule cells. J. Neurochem. (2009) 109: 525-538.

Gressner AM, Weiskirchen R, Breitkopf K, Dooley S. Roles of TGF-β in hepatic fibrosis. Front. Biosci. (2002) 7: d793–807.

Gross A, Yin XM, Wang K, Wei MC, Jockel J, Milliman C, Erdjument-Bromage H, Tempst P, Korsmeyer SJ. Caspase cleaved BID targets mitochondrial and is required for cytochrome c release, while BCL-XL prevents this release but not tumor necrosis factor-R1/Fas death. J. Biol. Chem. (1999) 274: 1156 -1163.

Guicciardi ME,  Deussing J, Miyoshi H, Bronk SF, Svingen PA, Peters C, Kaufmann SH, Gores GJ. Cathepsin B contributes to TNF-alpha-mediated hepatocyte apoptosis by promoting mitochondrial release of cytochrome c. J. Clin. Invest. (2000) 106:1127–1137.

Guicciardi ME, Leist M, Gores GJ. Lysosomes in cell death. Oncogene. (2004) 23(16):2881-90.

Guo J, Friedman SL. Hepatic fibrogenesis. Semin. Liver. Dis. (2007) 27(4): 413-426.

Hamacher-Brady A, Stein HA, Turschner S, Toegel I, Mora R, Jennewein N, Efferth T, Eils R, Brady NR. Artesunate activates mitochondrial apoptosis in breast cancer cells via iron-catalyzed lysosomal reactive oxygen species production. J. Biol. Chem. (2011) 286(8):6587-601.

Hamdy N, El-Demerdash E. New therapeutic aspect for carvedilol: antifibrotic effects of carvedilol in chronic carbon tetrachloride-induced liver damage. Toxicol. Appl. Pharmacol. (2012) 261(3): 292-299.

Heymann F, Tacke F. Immunology in the liver–from homeostasis to disease. Nat. Rev. Gastroenterol. Hepatol. (2016) 13: 88–110.

Imamura M, Ogawa T, Sasaguri Y, Chayama K, Ueno H. Suppression of macrophage infiltration inhibits activation of hepatic stellate cells and liver fibrogenesis in rats. Gastroenterology (2005) 128:138-146.

Jaeschke H. Inflammation in response to hepatocellular apoptosis. Hepatology. (2002) 35(4):964-966.

Jaeschke H, Gores GJ, Cederbaum AI, Hinson JA, Pessayre D, Lemasters JJ. Mechanisms of hepatotoxicity.  Toxicological Sciences (2002) 65(2): 166–176.

Johnston HJ, Hutchison G, Christensen FM, Peters S, Hankin S, Stone V. A review of the in vivo and in vitro toxicity of silver and gold particulates: particle attributes and biological mechanisms responsible for the observed toxicity. Crit. Rev. Toxicol. (2010) 40:328–46.

Jorgensen I, Lopez JP, Laufer SA, Miao EA. IL-1β, IL-18, and eicosanoids promote neutrophil recruitment to pore-induced intracellular traps following pyroptosis. Eur. J. Immunol. (2016) 46(12): 2761–2766.

Jung M, Lee J, Seo H-Y, Lim JS, Kim E-K. Cathepsin Inhibition-Induced Lysosomal Dysfunction Enhances Pancreatic Beta-Cell Apoptosis in High Glucose. PLoS ONE. (2015) 10(1):e011697.

Jürgensmeier JM, Xie Z, Deveraux Q, Ellerby L, Bredesen D, Reed JC.  Bax directly induces release of cytochrome c from isolated mitochondria. Proc. Natl. Acad. Sci. U S A. (1998) 95(9): 4997–5002.

Kagedal K, Zhao M, Svensson I, Brunk UT. Sphingosine-induced apoptosis is dependent on lysosomal proteases. The Biochemical journal.  (2001) 359: 335-43.

Karlmark KR, Wasmuth HE, Trautwein C, Tacke F. Chemokine-directed immune cell infiltration in acute and chronic liver disease. Expert. Rev. Gastroenterol. Hepatol. (2008) 2:233-242.

Kermanizadeh A: Engineered Nanomaterial Impact in the Liver following Exposure via an Intravenous Route–The Role of Polymorphonuclear Leukocytes and Gene Expression in the Organ. J. Nanomed. Nanotechnol. (2012) 04:1–7.

Kershenobich Stalnikowitz D,  Weisssbrod AB. Liver Fibrosis and Inflammation. A Review, Annals of Hepatology  (2003) 2(4): 159-163.

Kermanizadeh A, Gaiser BK, Johnston H, Brown DM, Stone V. Toxicological effect of engineered nanomaterials on the liver. Br. J. Pharmacol. (2014) 171(17):3980-7.

Kisseleva T, Brenner DA. (2008), Mechanisms of Fibrogenesis. Exp. Biol. Med. (2008) 233(2):109-122.

Kolios G, Valatas V, Kouroumalis E. Role of Kupffer cells in the pathogenesis of liver disease. World J.Gastroenterol. (2006)  12(46): 7413-7420.

Kong XY, Nesset CK, Damme M, Løberg EM, Lübke T, Mæhlen J, Andersson KB, Lorenzo PI, Roos N, Thoresen GH, Rustan AC, Kase ET, Eskild W. Loss of lysosomal membrane protein NCU-G1 in mice results in spontaneous liver fibrosis with accumulation of lipofuscin and iron in Kupffer cells. Disease models & mechanisms (2014) 7(3): 351-62.

Kono H, Karmarkar D, Iwakura Y, Rock KL. Identification of the cellular sensor that stimulates the inflammatory response to sterile cell death. J. Immunol. (2010) 184(8):4470-8.

Kubota C, Torii S, Hou N, Saito N, Yoshimoto Y, Imai H, Takeuchi T. Constitutive reactive oxygen species generation from autophagosome/lysosome in neuronal oxidative toxicity. J. Biol. Chem. (2010) 285(1):667-74.

Lee SC, Brummet ME, Shahabuddin S, Woodworth TG, Georas SN, Leiferman KM, Gilman SC, Stellato C, Gladue RP, Schleimer RP, Beck LA. Cutaneous injection of human subjects with macrophage inflammatory protein-1 alpha induces significant recruitment of neutrophils and monocytes. J. Immunol. (2000) 164: 3392-3401.

Lee William M. Drug- induced hepatotoxicity. N. Engl. J. Med.  (2003) 349(5): 474-485.

Lee PY, Li Y, Kumagai Y, Xu Y,  Weinstein JS, Kellner ES, Nacionales DC, Butfiloski EJ, van Rooijen N, Akira S, Sobel ES, Satoh M, Reeves WH. Type I Interferon Modulates Monocyte Recruitment and Maturation in Chronic Inflammation. Am. J. Pathol. (2009) 175(5): 2023–2033.

Lee UE, Friedman SL. Mechanisms of Hepatic Fibrogenesis. Best Pract. Res. Clin. Gastroenterol. (2011) 25(2): 195-206.

Leist M, Single B, Castoldi AF, Kuhnle S, Nicotera P. Intracellular adenosine triphosphate (ATP) concentration: a switch in the decision between apoptosis and necrosis. J. Exp. Med. (1997) 185: 1481−1486.

Leung TM, Tipoe GL, Liong EC, Lau TY, Fung ML, Nanji AA. Endothelial nitric oxide synthase is a critical factor in experimental liver fibrosis. Int. J. Exp. Pathol. (2008) 89(4):241-50.

Li J, Wang H, Mason JM, Levine J, Yu M, Ulloa L, Czura CJ, Tracey KJ, Yang H. Recombinant HMGB1 with cytokine-stimulating activity. J. Immunol. Methods. (2004) 289:211–23.

Li JT, Liao ZX, Ping J, Xu D, Wang H. Molecular mechanism of hepatic stellate cell activation and antifibrotic therapeutic strategies. J. Gastroenterol. (2008) 43(6):419–428.

Li L, Zongqiang H, Wen L, Mingdao H, JIanghua R, Peng C, Qiangming S. Establishment of a standardized liver fibrosis model with different pathological stages in rats. Gastroenterol. Res. Pract. (2012) Article ID 560345.

Liedtke C, Luedde T, Sauerbruch T, Scholten D, Streetz K, Tacke F, Tolba R, Trautwein C, Trebicka J, Weiskirchen R. Experimental liver fibrosis research: update on animal models, legal issues and translational aspects. Fibrogenesis Tissue Repair (2013) 6(1):19.

Lim YS, Kim WR. The global impact of hepatic fibrosis and end-stage liver disease. Clin. Liver Dis.  (2008) 12:733–746

Limana F, Germani A, Zacheo A, Kajstura J, Di Carlo A, Borsellino G, Leoni O, Palumbo R, Battistini L, Rastaldo R, Müller S, Pompilio G, Anversa P, Bianchi ME, Capogrossi MC. Exogenous high-mobility group box 1 protein induces myocardial regeneration after infarction via enhanced cardiac C-kit+ cell proliferation and differentiation. Circ. Res. (2005) 97(8): e73–e83

Liu H, Ma L, Zhao J, Liu J, Yan J, Ruan J, Hong F. Biochemical toxicity of nano-anatase TiO2 particles in mice. Biol. Trace Elem. Res. (2009) 129:170–80.

Liu H, Ma L, Liu J, Zhao J, Yan J, Hong F. Toxicity of nano-anatase TiO2 to mice: Liver injury, oxidative stress. Toxicol. Environ. Chem. (2010) 92:175–186.

Lloyd A, Oppenheim J, Kelvin D, Taub D. Chemokines regulate T cell adherence to recombinant adhesion molecules and extracellular matrix proteins. J. Immunol. (1996) 156: 932–938.

Loos C, Syrovets T, Musyanovych A, Mailänder V, Landfester K, Nienhaus GU, Simmet T. Functionalized polystyrene nanoparticles as a platform for studying bio-nano interactions. Beilstein. J. Nanotechnol. (2014) 5:2403-12.

López-Novoa JM, Nieto MA. Inflammation and EMT: an alliance towards organ fibrosis and cancer progression. EMBO Mol. Med. (2009) 1(6-7): 303–314.

Lu J, Wu DM, Zheng YL, Hu B, Cheng W, Zhang ZF. Purple sweet potato color attenuates domoic acid-induced cognitive deficits by promoting estrogen receptor-α-mediated mitochondrial biogenesis signaling in mice. Free Radic. Biol. Med. (2012) 52(3): 646-59.

Lu TH, Su CC, Tang FC, Chen CH, Yen CC, Fang KM, Lee KL, Hung DZ, Chen YW. Chloroacetic acid triggers apoptosis in neuronal cells via a reactive oxygen species-induced endoplasmic reticulum stress signaling pathway. Chem. Biol. Interact. (2014) 225: 1-12.

Luckey SW, Petersen DR. Activation of Kupffer cells during the course of carbon tetrachloride-induced liver injury and fibrosis in rats. Exp. Mol. Pathol.  (2001) 71(3): 226-240.

Luo X, Budihardjo I, Zou H, Slaughter C, Wang X. Bid, aBcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell (1998) 94: 481 - 490.

Ma L, Zhao J, Wang J, Liu J, Duan Y, Liu H, Li N, Yan J, Ruan J, Wang H, Hong F. The Acute Liver Injury in Mice Caused by Nano-Anatase TiO2. Nanoscale Res. Lett. (2009) 4:1275–85.

Malhi H, Gores GJ. Cellular and molecular mechanisms of liver injury. Gastroenterology (2008) 134(6):1641-54.

McColl SR, Clark-Lewis C. Inhibition of Murine Neutrophil Recruitment In Vivo by CXC Chemokine Receptor Antagonists. J. Immunol.  (1999)  163(5): 2829-2835.

McHedlidze T, Waldner M, Zopf S, Walker J, Rankin AL, Schuchmann M, Voehringer D, McKenzie AN, Neurath MF, Pflanz S, Wirtz S. Interleukin-33-dependent innate lymphoid cells mediate hepatic fibrosis. Immunity  (2013) 39: 357–371.

Milani, S. et al. Differential expression of matrix-metalloproteinase-1 and -2 genes in normal and fibrotic human liver. Am. J. Pathol. (1994) 144(3): 528-537.

Miyamoto NG, Medburry PS, Hesselgesser J, Boehlk S, Nelson PJ, Krensky AM, Perez HD. Interleukin-1b induction of the chemokine RANTES promoter in the human astrocytoma line CH235 requires both constitutive and inducible transcription factors. J. Neuroimmunol. (2000) 105:78.

Nan YM, Kong LB, Ren WG,Wang RQ, Du JH, Wen-Cong L, Zhao SX, Zhang YG, Wu WJ, Di HL, Li Y, Yu J. Activation of peroxisome proliferator activated receptor alpha ameliorates ethanol mediated liver fibrosis in mice. Lipids Health Dis. (2013) 12:11.

Narumi S, Wyner L, Stoler MH, Tannenbaum CS, Hamilton TA. Tissue-specific expression of murine IP-10 mRNA following systemic treatment with interferon g. J. Leukoc. Biol. (1992) 52:27.

Natajaran SK, Thomas S, Ramamoorthy P, Basivireddy J, Pulimood AB, Ramachandran A, Balasubramanian KA. Oxidative stress in the development of liver cirrhosis: a comparison of two different experimental models. J. Gastroenterol. Hepatol. (2006) 21(6): 947-957.

Nicotera P, Leist M, Ferrando-May E. Intracellular ATP, a switch in the decision between apoptosis and necrosis. Toxicol. Lett. (1998) 102-103: 139-142.

Nikiforou M, Kemp MW, van Gorp RH, Saito M, Newnham JP, Reynaert NL, Janssen LEW, Jobe AH, Kallapur SG, Kramer BW, Wolfs TG. Selective IL-1α exposure to the fetal gut, lung, and chorioamnion/skin causes intestinal inflammatory and developmental changes in fetal sheep. Lab. Invest. (2016) 96(1): 69–80.

Ono K, Kim SO, Han J. Susceptibility of lysosomes to rupture is a determinant for plasma membrane disruption in tumor necrosis factor alpha-induced cell death. Mol. Cell. Biol. (2003) 23: 665-76.

Ortiz BD, Krensky AM, Nelson PJ. Kinetics of transcription factors regulating the RANTES chemokine gene reveal a developmental switch in nuclear events during T-lymphocyte maturation. Mol. Cell. Biol. (1996) 16:202.

Ouedraogo G, Morliere P, Maziere C, Maziere JC, Santus R. Alteration of the Endocytotic Pathway by Photosensitization with Fluoroquinolones. Photochemistry and Photobiology, (2000) 72(4): 458–463.

Poynard T, Bedossa P, Opolon P. Natural history of liver fibrosis progression in patients with chronic hepatitis C. The OBSVIRC, METAVIR, CLINIVIR, and DOSVIRC groups, Lancet. (1997) 349(9055) :825-832.

Pradere JP, Kluwe J, De Minicis S, Jiao JJ, Gwak GY, Dapito DH, Jang MK, Guenther ND, Mederacke I, Friedman R, Dragomir AC, Aloman C, Schwabe RF. Hepatic macrophages but not dendritic cells contribute to liver fibrosis by promoting the survival of activated hepatic stellate cells in mice. Hepatology (2013) 58:1461–1473.

Ramachandran R, Kakar S. Histological patterns in drug-induced liver disease. J. Clin. Pathol. (2009) 62(6): 481-492.

Reeves HL, Dack CL, Peak M, Burt AD, Day CP. Stress-activated protein kinases in the activation of rat hepatic stellate cells in culture. J. Hepatol.  (2000) 32: 465-472.

Reiners J, Caruso J, Mathieu P, Chelladurai B, Yin X-M, Kessel D. Release of cytochrome c and activation of pro-caspase-9 following lysosomal photodamage involves bid cleavage. Cell death and differentiation. (2002) 9(9):934-944.

Repnik U, Stoka V, Turk V, Turk B. Lysosomes and lysosomal cathepsins in cell death. Biochim. Biophys. Acta. (2012) 1824(1):22-33.

Repnik U, Borg Distefano M,  Speth MT, Ng MYW, Progida C, Hoflack B, Gruenberg J, Griffiths G. L-leucyl-L-leucine methyl ester does not release cysteine cathepsins to the cytosol but inactivates them in transiently permeabilized lysosomes. J. Cell Sci. (2017) 130: 3124–3140.

Richter C, Schweizer M, Cossarizza A, Franceschi C. Control of apoptosis by the cellular ATP level. FEBS Lett. (1996) 378: 107-110.

Roberg K, Johansson U, Ollinger K. Lysosomal release of cathepsin D precedes relocation of cytochrome c and loss of mitochondrial transmembrane potential during apoptosis induced by oxidative stress. Free Radic. Biol. Med. (1999) 27(11-12): 1228–1237.

Roberg K, Kagedal K, Ollinger K. Microinjection of cathepsin d induces caspase-dependent apoptosis in fibroblasts. Am. J. Pathol. (2002) 161:89–96.

Rockey DC, Boyles JK, Gabbiani G, Friedman SL. Rat hepatic lipocytes express smooth muscle actin upon activation in vivo and in culture. J. Submicrosc. Cytol. Pathol. (1992) 24(2):193-203.

Rockey DC, Friedman SL. Hepatic fibrosis and cirrhosis, Zakim and Boyer's Hepatology (2006)5:87-109.

Rouhiainen A, Tumova S, Valmu L, Kalkkinen N, Rauvala H. Analysis of proinflammatory activity of highly purified eukaryotic recombinant HMGB1 (amphoterin). J. Leukoc. Biol. (2007) 81:49–58.

Safadi R, Friedman SL. Hepatic fibrosis--role of hepatic stellate cell activation. Med. Gen. Med. (2002) 4(3): 27.

Santibañez JF, Quintanilla M, Bernabeu C. TGF-β/TGF-β receptor system and its role in physiological and pathological conditions. Clin. Sci. (Lond) (2011) 121(6): 233-251.

Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature (2002) 418:191–5.

Schwarz M, Jandová K, Struk I, Marešová D, Pokorný J, Riljak V. Low dose domoic acid influences spontaneous behavior in adult rats. Physiol. Res. (2014) 63: 369-76.

Seki E, De Minicis S, Osterreicher CH, Kluwe J, Osawa Y, Brenner DA, Schwabe RF. TLR4 enhances TGF-beta signaling and hepatic fibrosis. Nat. Med. (2007) 13:1324–32.

Soliman MM, Attia HF, Hussein MM. Protective Effect of N-Acetylcystiene Against Titanium Dioxide Nanoparticles Modulated Immune Responses in Male Albino Rats. Am. J. Immunol. (2013) 9:148–158.

Son G, Hines IN, Lindquist J, Schrum LW, Rippe RA. Inhibition of phosphatidylinositol 3-kinase signaling in hepatic stellate cells blocks the progression of hepatic fibrosis, Hepatology, (2009) 50(5): 1512–1523.

Stoka V, Turk B, Schendel SL, Kim TH, Cirman T, Snipas SL, Ellerby LM, Bredesen D, Freeze H, Abrahamson M, BroÈmme D, Krajewski S, Reed JC, Yin XM, Turk V, Salvesen GS. Lysosomal protease pathways to apoptosis: cleavage of Bid, not pro-caspases, is the most likely route. J. Biol. Chem. (2001) 276: 3149 - 3157.

Sun Y, Zhu D, Wang G, Wang D, Zhou H, Liu X, Jiang M, Liao L, Zhou Z, Hu J. Pro-Inflammatory Cytokine IL-1β Up-Regulates CXC Chemokine Receptor 4 via Notch and ERK Signaling Pathways in Tongue Squamous Cell Carcinoma. PLoS One. (2015) 10(7):e0132677.

Todkar K, Ilamathi HS, Germain M. Mitochondria and Lysosomes: Discovering Bonds. Front Cell Dev Biol. (2017)5:106.

van der Hilst CS, Ijtsma AJ, Slooff MJ, Tenvergert EM. Cost of Liver Transplantation A Systematic Review and Meta-Analysis Comparing the United States With Other OECD Countries. Medical care research and review : MCRR.(2008) 66: 3-22.

Villamil Giraldo AM, Fyrner T, Wennmalm S, Parikh AN, Öllinger K, Ederth T. Spontaneous Vesiculation and pH-Induced Disassembly of a Lysosomotropic Detergent: Impacts on Lysosomotropism and Lysosomal Delivery. Langmuir (2016) 32: 13566−13575.

Wang F, Bexiga MG, Anguissola S, Boya P, Simpson JC, Salvati A, Dawson KA. Time resolved study of cell death mechanisms induced by amine-modified polystyrene nanoparticles. Nanoscale (2013) 5:10868–76.

Werneburg NW, Guicciardi ME, Bronk SF, Gores GJ. Tumor necrosis factor-alpha-associated lysosomal permeabilization is cathepsin B dependent. Am. J. Physiol. Gastrointest. Liver Physiol. (2002) 283:G947–G956.

Winklhofer K, Haass C. Mitochondrial dysfunction in Parkinson's disease. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease (2010) 1802: 29-44.

Wu DM, Lu J, Zheng YL, Zhang YQ, Hu B, Cheng W, Zhang ZF, Li MQ. Small interfering RNA-mediated knockdown of protein kinase C zeta attenuates domoic acid-induced cognitive deficits in mice. Toxicol. Sci. (2012) 128: 209-222.

Yang ND, Tan SH, Ng S1, Shi Y, Zhou J, Tan KS, Wong WS, Shen HM. Artesunate induces cell death in human cancer cells via enhancing lysosomal function and lysosomal degradation of ferritin. J. Biol. Chem. (2014) 289(48):33425-41.

Yamada T, Fujieda S, Yanagi S, Yamamura H, Inatome R, Yamamoto H, Igawa H, Saito H. IL-1 induced chemokine production through the association of Syk with TNF receptor-associated factor-6 in nasal fibroblast lines. J. Immunol. (2001) 167(1):283-8.

Zhou Q, Liu C, Liu W, Zhang H, Zhang R, Liu J, Zhang J, Xu C, Liu L, Huang S, Chen L. Rotenone induction of hydrogen peroxide inhibits mTOR-mediated S6K1 and 4E-BP1/eIF4E pathways, leading to neuronal apoptosis. Toxicol Sci. (2015) 143: 81-96.

Zimmermann K, Volkel D, Pable S, Lindner T, Kramberger F, Bahrami S, Scheiflinger F. Native versus recombinant high-mobility group B1 proteins: functional activity in vitro. Inflammation. (2004) 28:221–9.

Zimmermann HW, Seidler S, Nattermann J, Gassler N, Hellerbrand C, Zernecke A, Tischendorf JJ, Luedde T, Weiskirchen R, Trautwein C, Tacke F. Functional contribution of elevated circulating and hepatic non-classical CD14CD16 monocytes to inflammation and human liver fibrosis. PLoS One (2010) 5:e11049.