Skip to main content

Table 1 Animal models of AE-PF

From: Animal models of acute exacerbation of pulmonary fibrosis

Animal species

Fibrotic Agents

Challenging agents of AE

Lung pathology

Advantages

Disadvantages

References

Agent

Dose

Route

Time of administration

Mouse

FITC

γHV-68①

5 × 104 PFU

I.N

D14

Interstitial edema, intraalveolar hemorrhage, alveolar epithelial denudation.

Introduced a novel animal model for herpesvirus-induced AE-PF and offered insights into the underlying mechanisms, including fibrocyte recruitment.

The mortality rate in AE mice did not align consistently with that observed in AE patients.

McMillan et al., 2008 [44]

Mouse

BLM

γHV-68①

5 × 104 PFU

I.N

D14

Focal mononuclear inflammation, diffuse inflammation, stromal collagen deposition.

The model elucidated the underlying mechanism by which increased apoptosis of AEC2 triggers the onset of AE, a key feature of IPF.

The evidence of apoptosis measured by cleaved poly-ADP ribose polymerase was inconclusive.

Ashley et al., 2014 [62]

Mouse

SPC-DTR

Spnâ‘¡

1 × 107 CFU

O.T

D14

---

The model mimicked the exaggerated inflammatory mechanisms seen in AE-IPF.

The establishment of the SPC-DTR fibrosis model requires repeated administration, which may lead to drug resistance.

Knippenberg et al., 2015 [61]

Mouse

BLM

LPSâ‘¢

0.5 mg/kg

O.A

D7

Fibrotic changes, prominent rise in inflammatory cell infiltration.

Assessing the results using chest CT scans and blood gas analysis is more clinically relevant.

Operation-induced AE-IPF occurs rarely in patients, making the practicality of this model poor.

Kimura et al., 2015 [124]

Mouse

BLM

BLMâ‘¢

4 mg/kg

I.T

D21

Hyaline membrane formation, pulmonary edema, a large amount of collagen deposition

The mouse model of AE-IPF was triggered by a non-infectious mechanism.

The core assumption that fibrosis stems from an external trigger rather than an internal genetic or epigenetic response remains unchanged

Wei et al., 2016127

Mouse

BLM

HSV1â‘ 

5 × 105 PFU

I.N

D14

Diffuse alveolar damage, interstitial edema, intraalveolar hemorrhage, alveolar epithelial denudation, hyaline membranes formation, collagen deposition.

This model simulated some of the clinical manifestations, pathological features, and inflammatory mechanisms in AE-IPF patients.

The causal association between endoplasmic reticulum stress and stimulator of interferon genes deficiency needs further investigation.

Qiu et al., 2017 [65]

Mouse

PHMG

CdCl2â‘¢

0.2 Âµg

I.T

D3, D6, D9, D12

Granulomatous inflammation, fibrosis alveolar ducts, foamy macrophage infiltration, bronchioloalveolar epithelial hyperplasia, collagen deposition.

Because CdCl2 is a component of tobacco, this model simulates lung fibrosis that may be caused by tobacco.

The reagents are toxic, and PHMG is not the preferred agent; the timing of CdCl2 administration was too early.

Kim et al. 2018 [128]

Mouse

BLM

Nickel chlorideâ‘¢

5 mg/kg

I.T

D0

Collagen deposition, alveolitis, collapsed alveolar space, inflammatory cells accumulation.

Nickel is heavy metal that can simulate lung fibrosis caused by industrial pollution.

Nickel chloride and BLM were administered simultaneously, which is not consistent with clinical settings.

McElroy et al., 2018 [129]

Mouse

BLM

HSV1â‘ 

5 × 105 PFU

I.N

D21

Alveolar septal congestion, edema, inflammation, alveolar epithelial damage, hyaline membrane formation.

This model uses human HSV1, mirroring AE-IPF symptoms including lung function decline, increased mortality, acute lung injury, and abnormal inflammation pathways.

The elevated levels of IL-17 A and endoplasmic reticulum stress may not be the primary drivers of pulmonary fibrosis.

Chen et al., 2019 [64]

Mouse

Ad TGF-β1

CNDGâ‘¡

1 × 108 CFU

I.T

D5

Significantly increased macrophage, lymphocyte, and neutrophil infiltration, enhanced collagen deposition, increased epithelial cell apoptosis.

The study included validation experiments in both male and female mice.

Animals may mount an immune response; the expression of transgenes may exceed physiological levels.

D’Alessandro-Gabazza et al., 2020 [59]

Mouse

BLM

Spnâ‘¡

1 × 105 CFU

I.T

D14

A significant increase in fibrosis and collagen deposition

This study found that AIM2 inflammasome activation plays a crucial role in worsening lung fibrosis during bacterial infections by connecting it to glucose transporter 1-mediated glycolysis.

Did not report the levels of hydroxyproline in the lung tissues

Cho et al., 2020 [63]

Mouse

Ad TGF-β1

Spnâ‘¡

1 × 107 CFU

O.T

D14

Increased interstitial inflammation, alveolar epithelial hyperplasia, thickened alveolar septa, elevated collagen deposition

This study enhances our understanding of infection-induced lung fibrosis exacerbation in two preclinical mouse models.

Transgene expression was higher than physiologically possible; Possible immune response to viral vectors.

Moyé et al., 2020 [37], Knippenberg et al., 2015 [61]

Mouse

BLM

Corisinâ‘¢

300 Âµg

I.T

D20

Significantly increased Ashcroft score and collagen-stained (trichrome) area.

The study included validation experiments in both male and female mice.

Future studies should assess corisin’s efficacy in inducing AE in various mouse models of pulmonary fibrosis.

D’Alessandro-Gabazza et al., 2022 [60]

Mouse

BLM

NT127â‘¡

1 × 107 CFU

I.T

D7

Increased inflammatory cell infiltration, airway structural damage, collagen deposition.

This is the first report that Gram-negative bacteria have been used to induce AE model of pulmonary fibrosis.

NT127 administrated on day 7 was not in the fibrotic stage; no further evidence of increased interstitial inflammation

Chen et al., 2022 [119]

Mouse

BLM

LPSâ‘¢

1 mg/kg

I.T

D5, D7, D9

Severe inflammation, thicker interalveolar spaces, collapsed alveoli, increased collagen, and severe lung distortion.

Simulates extreme inflammation in AE-IPF.

LPS-induced AE was applied on days 5, 7, and 9 during the inflammatory phase, rather than the fibrotic phase.

Jia et al., 2022 [123]

Rat

BLM

LPSâ‘¢

0.05 or 0.15 mg/kg

I.T

D7

Significant fibrosis, prominent inflammatory cells infiltration and alveolar enlargement

Assessing the results using chest CT scans and blood gas analysis is more clinically relevant. Simulates extreme inflammation in AE-IPF.

LPS-induced AE was monitored at 24 h after administration, which differs from clinical settings where changes in related indicators may occur later.

Miyamoto et al., 2022 [67]

  1. Abbreviations: AE: acute exacerbation; AE-PF: acute exacerbation of pulmonary fibrosis; Ad TGF-β1: adenoviral vector delivery of active transforming growth factor-β1; BLM: bleomycin;CdCl2: cadmium chloride; CNDG: staphylococcus nepalensis strain CNDG; FITC: fluorescein isothiocyanate; HSV1: Herpes Simplex virus 1; γHV-68: γ-Herpesvirus − 68; I.N: intranasally; I.T: intratracheally; LPS: lipopolysaccharide; NT127: non-typeable Haemophilus influenzae strain 127; O.T: orotracheally; O.A: oropharyngeal aspiration; PHMG: polyhmethylene guanidine; SPC-DTR: surfactant protein C-diphteria toxin receptor
  2. â‘  .Virus; â‘¡.Bacteria; â‘¢.Others