Next Article in Journal
Abiotic Stress-Induced Leaf Senescence: Regulatory Mechanisms and Application
Previous Article in Journal
Molecular Mechanisms of Migraine: Nitric Oxide Synthase and Neuropeptides
Previous Article in Special Issue
Dectin-1-Independent Macrophage Phagocytosis of Mycobacterium abscessus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Macrophages in Connective Tissue Disease-Associated Interstitial Lung Disease: Focusing on Molecular Mechanisms and Potential Treatment Strategies

1
Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
2
School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
3
Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
4
Department of Nursing, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
5
Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
6
Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
7
Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 801, Taiwan
8
Laboratory Diagnosis of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
9
Department of Kinesiology, Health and Leisure Studies, National University of Kaohsiung, Kaohsiung 811, Taiwan
10
Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(15), 11995; https://doi.org/10.3390/ijms241511995
Submission received: 20 April 2023 / Revised: 24 July 2023 / Accepted: 25 July 2023 / Published: 26 July 2023
(This article belongs to the Special Issue Macrophages in Respiratory Diseases)

Abstract

:
Connective tissue disease-associated interstitial lung disease (CTD-ILD) is a severe manifestation of CTD that leads to significant morbidity and mortality. Clinically, ILD can occur in diverse CTDs. Pathologically, CTD-ILD is characterized by various histologic patterns, such as nonspecific interstitial pneumonia, organizing pneumonia, and usual interstitial pneumonia. Abnormal immune system responses have traditionally been instrumental in its pathophysiology, and various changes in immune cells have been described, especially in macrophages. This article first briefly overviews the epidemiology, clinical characteristics, impacts, and histopathologic changes associated with CTD-ILD. Next, it summarizes the roles of various signaling pathways in macrophages or products of macrophages in ILD, helped by insights gained from animal models. In the following sections, this review returns to studies of macrophages in CTD-ILD in humans for an overall picture of the current understanding. Finally, we direct attention to potential therapies targeting macrophages in CTD-ILD in investigation or in clinical trials, as well as the future directions regarding macrophages in the context of CTD-ILD. Although the field of macrophages in CTD-ILD is still in its infancy, several lines of evidence suggest the potential of this area.

1. Introduction

Interstitial lung diseases (ILDs) comprise a heterogeneous group of multiple entities characterized by damage to the lung parenchyma due to varying degrees of inflammation and fibrosis [1]. ILDs arise from a broad spectrum of known or unknown etiologies and inflict a substantial burden on patients and health care systems [2]. Connective tissue disease (CTD), also known as collagen vascular disease [3], is a common cause associated with ILD [4], with 15% of ILDs arising from CTD [5]. CTD includes several disease entities, such as dermatomyositis (DM), microscopic polyangiitis (MPA), rheumatoid arthritis (RA), and systemic sclerosis (SSc). Corresponding to CTD-associated ILDs (CTD-ILDs) are DM-associated ILD (DM-ILD), MPA-associated ILD (MPA-ILD), RA-associated ILD (RA-ILD), and SSc-associated ILD (SSc-ILD). Pathologically, CTD-ILD can be classified based on respective histopathologic patterns, including nonspecific interstitial pneumonia (NSIP), organizing pneumonia (OP), and usual interstitial pneumonia (UIP) [6]. In practice, CTD-ILD is usually diagnosed through multidisciplinary discussion (MDD) including review of the clinical features, imaging, serology, and pathology by pulmonologists, radiologists, rheumatologists, and pathologists [7].
Clinically, different CTD-ILDs have distinct epidemiologies, pathologic changes, comorbidities, and prognoses (Figure 1). The incidence of DM-ILD is 1011/105 [8]; it is characterized by OP, NSIP, and UIP [9] and associated with palmar papules and myocardial involvement [10,11], and its mortality rate is as high as 24.62% [12]. The estimated incidence of MPA-ILD is 1.4/105 [13]; it is characterized by UIP and NSIP [14] and associated with decreased renal function [15], and its mortality rate is up to 39% [16]. The estimated incidence of RA-ILD is 3.7/105 [17]; it is presented as UIP [18] and associated with increased asthma, chronic obstructive pulmonary disease (COPD), diabetes, and heart disease [19,20], and its mortality rate is 8.08% [17]. The incidence of SSc-ILD is 1364/105 [21]; it is characterized by NSIP and UIP [14] and is associated with COPD, gastroesophageal reflux disease (GERD), hypertension, and skin disorders [22], and it has high mortality rate (39%) [23]. Overall, in CTD, the presence of ILD could lead to increased morbidity and mortality [24].
Although the mechanisms contributing to ILD in the context of CTDs are not known in detail, the prevailing hypothesis suggests the involvement of abnormal inflammation and subsequent exaggerated fibrotic reactions [1]. Interestingly, macrophages not only coordinate inflammation process but also mediate the formation of fibrosis. Given these characteristics of macrophages, it makes sense that macrophages play critical roles in the pathogenesis of CTD-ILD. The roles of macrophages have been suggested in numerous studies. For example, macrophages in the lungs of SSc-ILD upregulated profibrotic factors that promoted lung fibroblast activation [25]. Another example is DM-ILD. Previous studies reported that ferritin was produced by macrophages [26] and correlated with lung function parameters and survival [27]. IL-18 was also secreted by macrophages [28], elevated in DM-ILD compared with DM without ILD [29], and correlated with the severity of pulmonary disease [30]. Together, these studies point to the importance of macrophages in the development and progression of CTD-ILD.
Macrophages are versatile cells that exhibit high plasticity according to crosstalk between macrophages and the tissue microenvironment. Historically, analogous to the Th1/Th2 dichotomy, studies have characterized macrophages as classically activated (M1) or alternatively activated (M2) according to the activation status. Although the dichotomous inflammatory M1/repair M2 model cannot fully represent the current understanding of macrophage biology in ILD, studies on fibrosis usually build upon this classification scheme. As one of the most versatile cell types with diverse immunity functions, macrophages not only initiate an inflammatory response after injury but are also involved in the injury resolution and repair. In other words, in the airway and lung microenvironment, macrophages orchestrate the development and establishment of ILD. In this process, macrophage products and/or macrophages themselves are implicated at each key step. Work utilizing ILD patient samples provided several pieces of evidence of altered macrophages in ILD, whose functional importance was further confirmed in murine models [31].
Radiologically, CTD-ILD is characterized by various patterns of fibrosis using high-resolution computed tomography (HRCT) in the lung bases of patients with CTD, such as RA and SSc [32]. Most of our understanding of CTD-ILD comes from studies on pulmonary fibrosis (PF) in animal models. Therefore, we will first review the current knowledge of macrophages in the ILD/PF of animal models and then review recent advances in the molecular biology of CTD-ILD in the literature.

2. Roles of Signaling Pathways of Macrophages in ILD/PF in Animal Models

Regarding the relationship between macrophages and PF, several studies support the vital roles of macrophages in the pathogenesis of lung fibrosis. Macrophages in lung tissue samples from patients with fibrosis are higher compared to controls without fibrosis [33]. Macrophages play a mechanistic role in exacerbating lung fibrosis [34]. On the other hand, the ablation of macrophages was shown to significantly decrease fibroblasts and collagen deposition, ameliorating lung fibrosis [35]. Macrophages participate in both the inflammation and tissue repair phases of PF through various mechanisms, as detailed below.
(1) Caspase pathway: Studies show that MCU in PF macrophages leads to augmented expression of carnitine palmitoyltransferase 1A (CPT1A). By inducing Bcl-2 expression, CPT1A decreases caspase-3 activity, which attenuates macrophage apoptosis and thereby facilitates lung fibrosis progression (Table 1). The deletion of Bcl-2 in macrophages protects mice from developing PF [36].
(2) CCR2 axis: The CCL2/CCR2 axis is a major regulator of monocyte trafficking and plays an essential role in PF. CCR2 deficiency suppresses macrophage infiltration and reduces macrophage-derived MMP-2 and MMP-9 production, which decreases lung extracellular matrix content in the lungs (Table 1) and protects mice from PF [37].
(3) CCR4 axis: CCR4 promotes tissue injury through the induction of the M1 macrophage phenotype. In the absence of CCR4, macrophages upregulate the scavenging receptor D6, which attenuates inflammation and tissue injury (Table 1) and protects mice from lung fibrosis [38].
(4) CD204 axis: Collagen type I monomers stimulate macrophages to induce CD204 expression. In the context of elevated CD204, PF macrophages show hyperreactivity to stimulation with collagen type I monomers, resulting in exacerbated CCL18 secretion [39]. CCL18 triggers collagen production by fibroblasts. However, through β2-integrins and scavenger receptors, macrophages in the vicinity bind to collagen type I and increase CCL18 production. These reactions generate a feed-forward loop of augmented, ceaseless macrophage activation and unrestricted collagen production by fibroblasts (Table 1) [40].
(5) Glycolysis: Macrophages from fibrotic lungs assume the elevated expression of the glucose transporter GLUT1 [41], which augments the glycolysis required for the profibrotic profile in macrophages from fibrotic lungs (Table 1) [42].
(6) GSK pathway: TRIB3 is significantly upregulated in the macrophages of patients with PF. The TRIB3-GSK-3β interaction inhibits A20 activity and stabilizes C/EBPβ to induce macrophage activation, which triggers the transformation of lung fibroblasts into myofibroblasts, driving lung fibrosis (Table 1). Consequently, the macrophage-specific knockout of TRIB3 suppresses fibrotic changes in the lungs [43].
(7) HIF pathway: Macrophages of fibrotic lungs show increased HIF1A, which upregulates the ADORA2B receptor on macrophages. ADORA2B contributes to macrophage differentiation and the production of profibrotic mediators (Table 1), facilitating fibrosis in the lung [44].
(8) Itaconate axis: In the murine PF model, aconitate decarboxylase 1 (ACOD1) decarboxylates cis-aconitate to itaconate, and itaconate suppresses fibroblast proliferation and profibrotic activity, thereby limiting the severity of PF (Table 1). ACOD1 deficiency in macrophages induces profibrotic gene expression and worsens lung fibrosis [45].
(9) Macrophage migration: In the process of lung fibrosis, macrophages migrate into or within the lung to orchestrate and amplify fibrosis. In humans, monocyte migration is enhanced in PF. PLXNC1 is underexpressed and Syt7 is overexpressed in PF, and PLXNC1 suppresses Syt7-driven macrophage migration (Table 1). The underexpression or absence of PLXNC1 exacerbates macrophage migration and aggravates experimentally induced lung fibrosis. Conversely, restoring PLXNC1 in macrophages is sufficient to attenuate fibrosis [46].
(10) MAPK signaling: Human fibrotic lung exhibits augmented FOXM1 expression in macrophages. FOXM1 activates DUSP1 and inhibits the p38 MAPK signaling, therefore ameliorating lung fibrosis (Table 1). In agreement with this, macrophage-specific FOXM1 knockout mice develop severe PF. On the other hand, the transfer of FOXM1-expressing monocytes protects FOXM1-deficient mice against lung fibrosis [47].
(11) STAT6 signaling: Cu,Zn-superoxide dismutase (Cu,Zn-SOD) is expressed in macrophages [48]. Cu,Zn-SOD-mediated H2O2 generation activates STAT6 (Table 1) [49]. In the same way, macrophages isolated from fibrotic lungs have elevated Gab2, which increases STAT6 activation (Table 1) [50]. SART1 in macrophages facilitates the STAT6 signaling axis (Table 1) [51]. S1PR2 increases STAT6 activation (Table 1) [52]. These changes all accelerate STAT6 signaling and subsequent M2 polarization, triggering the development of PF [49,50,51,52].
(12) TGF-β signaling: There are several ways to modulate macrophage TGF-β signaling in PF. For example, Akt-mediated reactive oxygen species (ROS) production induces mitophagy and contributes to macrophage apoptosis resistance, which results in upregulated transforming growth factor-β (TGF-β) [53]. Similarly, C/EBP homologous protein (CHOP) decreases the expression of SOCS1 and SOCS3, thereby enhancing STAT6/PPARγ signaling, which is essential for TGF-β production (Table 1) [54]. Another example is MBD2. MBD2 selectively binds to the SH2-containing inositol 5′-phosphatase (SHIP) promoter in macrophages, by which MBD2 represses SHIP expression and enhances PI3K/Akt signaling to promote the macrophage M2 program and the production of downstream TGF-β [55]. In the same way, FBXW7 is an E3 ubiquitin ligase, whose expression in the macrophages of pulmonary tissue fibrosis mice is markedly decreased, and the deficiency of macrophage FBXW7 promotes the recruitment and accumulation of phagocytes, increases the K48-linked polyubiquitination and proteasome degradation of c-Jun, and downregulates the expression of TGF-β (Table 1) [56]. When TGF-β is increased through these pathways, enhanced fibroblast differentiation and fibrosis ensues [53].
(13) TNF pathway: STAT1 is expressed in lung macrophages [57]. STAT1 induces ICAM-1 and downstream TNF from macrophages and the subsequent infiltration of inflammatory cells and eventual fibrosis (Table 1) [58]. In agreement with this, STAT1 inhibition ameliorates collagen deposition in the lungs in an animal model [59]. In the same way, CD300c2 enhances high-mobility group box protein-1 (HMGB-1)-induced macrophage activation to produce tumor necrosis factor (TNF), which is a leukocyte chemoattractants, resulting in the accumulation of augmented immune cells, inflammation, and the aggravation of lung fibrosis (Table 1) [60].
Table 1. Signaling pathways of macrophages in ILD/PF in animal models. “🡪”—lead to, “↓”—decrease, “↑”—increase.
Table 1. Signaling pathways of macrophages in ILD/PF in animal models. “🡪”—lead to, “↓”—decrease, “↑”—increase.
Signaling PathwaysResultsReference
Caspase pathway
 MCU ↑ CPT1A 🡪 ↑ Bcl-2 🡪 ↓ caspase-3 activity ↓ macrophage apoptosis 🡪 ↑ fibrosis[36]
CCR2 axis
 CCR2 ↑ macrophage infiltration 🡪 ↑ MMP-2/MMP-9 ↑ extracellular matrix deposition[37]
CCR4 axis
 CCR4 ↑ inflammation and tissue injury↑ fibrosis[38]
CD204 axis
 collagen type I monomers ↑ CD204↑ CCL18 🡪 ↑ collagen production 🡪 ↑ CCL18[39,40]
Glycolysis
 GLUT1 ↑ glycolysis↑ profibrotic macrophages[42]
GSK pathway
 TRIB3-GSK-3β interaction ↓ A20 activity and stabilizes C/EBPβ 🡪 macrophage activation ↑ fibroblasts transform to myofibroblasts[43]
HIF pathway
 HIF1A ↑ ADORA2B 🡪 ↑ macrophage differentiation↑ profibrotic mediators[44]
Itaconate axis
 ACOD1 🡪 ↑ itaconate↓ fibroblast proliferation and profibrotic activity[45]
Macrophage migration
 PLXNC1↓ macrophage migration[46]
MAPK signaling
 FOXM1 activates DUSP1 🡪 ↓ p38 MAPK signaling ↓ fibrosis[47]
STAT6 signaling
 Cu,Zn-SOD ↑ H2O2 🡪 activates STAT6 ↑ M2 polarization[49]
 Gab2 🡪 activates STAT6 [50]
 SART1 🡪 ↑ activates STAT6 [51]
 S1PR2 🡪 ↑ activates STAT6 [52]
TGF-β signaling
Akt ↑ ROS 🡪 ↑ mitophagy ↑ apoptosis resistance 🡪 ↑ TGF-β↑ fibroblast differentiation[53]
CHOP ↓ SOCS1/SOCS3 🡪 ↑ STAT6/PPARγ signaling 🡪 ↑ TGF-β [54]
MBD2 ↓ SHIP 🡪 ↑ PI3K/Akt signaling 🡪 ↑ M2 polarization 🡪 ↑ TGF-β [55]
FBXW7 ↓ phagocyte recruitment ↑ c-Jun degradation 🡪 ↓ TGF-β [56]
TNF pathway
 STAT1 🡪 ↑ ICAM-1 🡪 ↑ TNF↑ inflammatory cells infiltration 🡪 ↑ fibrosis[58,59]
 CD300c2 🡪 ↑ macrophage activation🡪 ↑ TNF [60]

3. Roles of Macrophage-Derived Secretory Proteins/microRNAs in ILD/PF in Animal Models

In addition to macrophages themselves, macrophage-derived secretory proteins and microRNAs also execute various biological functions to modulate the lung fibrotic process.

3.1. Macrophage-Derived Secretory Proteins/microRNAs That Aggravate ILD/PF in Animal Models

(1) ADAM17: ADAM17 in association with membrane-bound IL-6Rα (mIL-6Rα) is increased in macrophages in fibrotic lungs. ADAM17 promotes the shedding of mIL-6Rα from the membranes of activated macrophages, thereby initiating IL-6 trans-signaling. IL-6 trans-signaling enhances fibroblast proliferation and extracellular matrix (ECM) production (Table 2) and the neutralization of IL-6 trans-signaling attenuates PF [61].
(2) AT1R: Macrophages secrete a considerable number of exosomes bearing AT1R, which are taken up by fibroblasts and result in higher levels of AT1R, the activation of the profibrotic TGF-β/Smad2/Smad3 pathway, the production of α-collagen I, and augmented Ang II secretion by fibroblasts. Interestingly, Ang II increases the number of macrophage exosomes and AT1R secretion, leading to a positive feedback between Ang II and exosome production involved in lung fibrosis (Table 2) [62].
(3) CCL6: CCL6, also called C10, is expressed in the macrophages and also attracts macrophages. Upregulated CCL6 accounts for the increased susceptibility to PF in the mice model. The neutralization of CCL6 attenuates subsequent lung fibrosis (Table 2) [63].
(4) Fibronectin: Compared to normal macrophages, PF macrophages produce more fibronectin [64]. Fibronectin transform fibroblasts to myofibroblasts, which lead to a local accumulation of extracellular matrix and hence the development of fibrosis in the lungs (Table 2) [65].
(5) Galectin-3: Galectin-3 is produced by macrophages and the levels of galectin-3 in CTD-ILD patients are higher than in control patients. Galectin-3 induces the production of TNF-α in macrophages, and galectin-3 expression in macrophages is also induced by TNF-α, creating a self-perpetuating cycle between TNF-α and galectin-3 to support inflammation. In addition, galectin-3 stimulates fibroblasts to induce proliferation (Table 2). All these changes promote lung fibrosis [66].
(6) IGF-I: Macrophages secrete IGF-I, which is increased in PF patients. Macrophage-derived IGF-I activates the prosurvival kinase Akt, which protects myofibroblasts from apoptosis, contributing to fibrosis (Table 2) [67]. Correspondingly, decreasing IGF-I ameliorates the severity of lung fibrosis [37].
(7) IL-1β: IL-1β induces inflammation with leukocyte influx and upregulates myofibroblast differentiation, resulting in augmented pulmonary collagen deposition (Table 2) and lung fibrosis [68,69].
(8) MIP-1α: Macrophages secrete MIP-1α, which promotes leukocyte accumulation [70]. On the contrary, anti-MIP-1α antibodies significantly reduce pulmonary mononuclear phagocyte accumulation and ensue fibrosis (Table 2) [37].
(9) miR-328: Macrophage-derived miR-328 inhibits the family with sequence similarity 13 member A (FAM13A). This induces lung fibroblast proliferation and the overexpression of collagen 1A, collagen 3A, and alpha-smooth muscle actin (α-SMA), which aggravate fibrosis (Table 2) [71]. On the other hand, silencing miR-328 in M2 macrophages alleviates the progression of lung tissue fibrosis [72].
(10) NTN1: NTN1+ macrophages also accumulate in fibrotic lungs. Through interaction with DCC, macrophage-derived NTN1 impacts adrenergic nerve remodeling, which is required for lung fibrosis (Table 2) [73].
(11) PDGF: Macrophages from animal models of PF secrete PDGF, which is a potent mitogen for fibroblasts, facilitating lung fibrosis (Table 2) [74].
(12) S100A4: M2-polarized macrophages secrete S100A4, which stimulates the proliferation and activation of fibroblasts and is increased in PF (Table 2). The inhibition of S100A4 reduces histological evidence of lung fibrosis [75].
(13) TGF-β: In the PF model, macrophages secrete TGF-β [76]. TGF-β increases PAI-1 expression in macrophages via Smad3, leading to extracellular stroma accumulation. These abnormalities favor lung fibrosis [77].
(14) Wnt: Wnt expression is noted in macrophages [78]. Wnt/β-catenin signaling promotes the differentiation of infiltrated monocyte-macrophage populations that contribute to lung fibrosis (Table 2). Therefore, the loss of the Wnt coreceptor low density lipoprotein receptor-related protein 5 (LRP5) attenuates the lung fibrotic process [79].
Table 2. Macrophage-derived secretory proteins/microRNAs in ILD/PF in animal models. “🡪”—lead to, “↓”—decrease, “↑”—increase.
Table 2. Macrophage-derived secretory proteins/microRNAs in ILD/PF in animal models. “🡪”—lead to, “↓”—decrease, “↑”—increase.
MoleculesCharacteristicsSignaling PathwayReference
Macrophage-derived secretory proteins/microRNAs that aggravate ILD/PF
ADAM17Protein↑ shedding of mIL-6Rα 🡪 ↑ IL-6 trans-signaling 🡪 ↑ fibroblast proliferation and ECM production [61]
AT1RProteinAT1R taken up by fibroblasts 🡪 ↑ profibrotic
↑ TGF-β/Smad2/Smad3 pathway, ↑ α-collagen I production, ↑ Ang II secretion by fibroblasts 🡪 ↑ macrophage exosomes and AT1R in exosomes
[62]
CCL6Protein↑ fibrosis[63]
FibronectinProtein↑ myofibroblasts 🡪 ↑ extracellular matrix deposition[64,65]
Galectin-3Protein↑ TNF-α 🡪 ↑ galectin-3 🡪 ↑ inflammation, ↑ fibroblast proliferation[66]
IGF-IProteinactivates Akt 🡪 ↓ myofibroblast apoptosis[67]
IL-1βProtein↑ leukocyte influx, ↑ myofibroblasts 🡪 ↑ collagen deposition[68,69]
MIP-1αProtein↑ leukocyte accumulation 🡪 ↑ fibrosis[37,70]
miR-328microRNA↓ FAM13A 🡪 ↑ fibroblast proliferation and ↑ collagen 1A, ↑ collagen 3A, ↑ α-SMA[71]
NTN1Proteininteracts with DCC 🡪 impacts adrenergic nerve remodeling 🡪 ↑ fibrosis[73]
PDGFProteina mitogen for fibroblasts[74]
S100A4Protein↑ fibroblast proliferation and activation[75]
TGF-βProtein↑ PAI-1 🡪 ↑ extracellular matrix[77]
WntProtein↑ differentiation of monocyte-macrophage 🡪 ↑ fibrosis[79]
Macrophage-derived secretory proteins/microRNAs that ameliorate ILD/PF
Pentraxin-2 (serum amyloid P) [80]Protein↓ M2 differentiation[81]
miR-142-3pmicroRNA↓ TGFβ-R1 🡪 ↓ profibrotic genes[82]

3.2. Macrophage-Derived Secretory Proteins/microRNAs That Ameliorate ILD/PF in Animal Models

(1) Pentraxin-2: Pentraxin-2, also called serum amyloid P (SAP) [80], reduces M2 differentiation and subsequent TGF-β-mediated lung fibrosis (Table 2) [81].
(2) miR-142-3p: Macrophages secrete antifibrotic miR-142-3p, which reduces the expression of transforming growth factor β receptor 1 (TGFβ-R1) and downstream profibrotic genes in PF (Table 2) [82].

4. Roles of Macrophages in CTD-ILD

The evidence mentioned above suggests that macrophages actively participate in the pathogenesis of ILD/PF. In theory, macrophages could also analogously take part in the course of CTD-ILD. This is supported by several observations.

4.1. Macrophages in Dermatomyositis (DM)-Associated ILD (DM-ILD)

Although there are no animal models that directly address the molecular mechanisms through which macrophages contribute to DM-ILD, several studies provide indirect evidence implicating macrophages as the main player in DM-ILD. Pathological analyses show that the infiltration of CD163-positive macrophages into the alveolar spaces is more severe in fatal DM-ILD than in DM-ILD survivors [83], and increased serum CD163 levels are associated with a higher mortality rate in DM-ILD [84]. Similarly, CD206-positive macrophages accumulate more densely in fatal cases of DM-ILD than in DM-ILD survivors, and elevated serum CD206 levels are associated with a higher mortality rate in DM-ILD [84]. Additionally, macrophages express resistin, which is significantly higher in DM-ILD than in DM patients without ILD. Compared with chronic ILD, resistin levels are significantly elevated in rapidly progressive ILD. Acting via nuclear factor kappa B (NFκB) signaling, resistin upregulates IL-1, IL-6, and TNF-α in human monocytes. These proinflammatory cytokines induce resistin expression in macrophages (Figure 2), triggering a self-sustained loop for lung injury [85]. Together, these studies highlight the contribution of macrophages to DM-ILD.

4.2. Macrophages in Microscopic Polyangiitis (MPA)-Associated ILD (MPA-ILD)

In MPA-ILD, macrophage polarization is skewed toward M2, which recruits fibroblasts and transforms them into myofibroblasts, culminating in the formation of lung fibrosis [86]. In the process of M2 polarization, the infiltrated macrophages produce CCL2, which recruits more macrophages to infiltrate the alveolar spaces. The recruited macrophages secrete CCL2, promoting a vicious cycle (Figure 2). In addition, in concert with PDGF secreted by vascular endothelial cells, CCL2 promotes myofibroblast differentiation and ECM production (Figure 2), leading to lung fibrosis [87]. Collectively, these findings implicate macrophages in the pathogenesis of MPA-ILD.

4.3. Macrophages in Rheumatoid Arthritis (RA)-Associated ILD (RA-ILD)

In RA-ILD, autoimmune/complement/interferon cascade genes are altered in macrophages, which might contribute to profibrotic inflammatory lung responses [88]. Several molecules produced by macrophages have been highlighted in the literature (Figure 2).
In RA-ILD, macrophages produce higher IL-6, which promotes the expansion of GM-CSF-producing T cells. GM-CSF is a strong inducer of neutrophil infiltration into the lung and contributes to the progression of RA-ILD in an animal model (Figure 2) [89]. SDC2 in macrophages activates CD148 in fibroblasts, inhibits PI3K/Akt signaling, downregulates PAD2 in fibroblasts, and attenuates collagen production by fibroblasts (Figure 2). The overexpression of SDC2 in alveolar macrophages decreases collagen deposition in the lungs and protects mice from RA-ILD [90]. The augmented secretion of TNF by alveolar macrophages from RA-ILD has been noted [91]. TNF induces an inflammatory phase that is predominated by cellular infiltration to the pulmonary tissue, which subsequently transitions to a fibrotic phase that constitutes the “irreversible” process of collagen deposition in the pulmonary parenchyma, resulting in RA-ILD in a murine model (Figure 2) [92].

4.4. Macrophages in Systemic Sclerosis (SSc)-Associated ILD (SSc-ILD)

In SSc-ILD, macrophages express profibrotic factors that promote the differentiation, migration, and activation of fibroblasts, suggesting the involvement of macrophages in SSc-ILD [25]. Currently, there are several known mechanisms that macrophages utilize to modulate SSc-ILD (Figure 2). (1) Monocytes from SSc-ILD patients reveal a profibrotic phenotype characterized by the expression of CD163 and the enhanced secretion of CCL18 [93], which promotes collagen production (Figure 2) [40]. (2) At the same time, SSc-ILD macrophages release significantly more fibronectin, which leads to fibroblast proliferation and subsequent collagen deposition (Figure 2) [64,93]. (3) Immunologically, immune complexes (ICs) activate human monocytes to secrete M-CSF and IL-6, which in turn induce osteopontin (OPN) from monocytes. Next, OPN facilitates fibroblast migration to areas adjacent to the fibrotic niche, thereby aiding lung fibrosis progression (Figure 2) [94]. (4) Meanwhile, PLXNC1 expression is reduced in SSc-ILD macrophages, which could lead to the excess migration of macrophages (Figure 2), amplifying fibrosis [46]. (5) Moreover, the response gene to complement 32 (RGC32) is abundantly expressed in macrophages, activates NFκB signaling, and promotes inflammatory gene expression by binding to their promoters. RGC32 deficiency in mice impairs the polarization of classically activated macrophages, attenuates the expression of inflammatory mediators in macrophages, including the fibrosis inducers iNOS and IL-1β, which are regulated by NFκB, and significantly ameliorates lung fibrosis (Figure 2) [95]. (6) YKL-40 is elevated in SSc-ILD. Macrophages produce YKL-40, which exerts promitogenic effects on lung fibroblasts (Figure 2) [96]. These findings all support the central roles of macrophages in SSc-ILD.

5. Targeting Macrophages in CTD-ILD

It is hoped that, apart from detailing our current understanding of CTD-ILD pathogenesis, this knowledge could be leveraged to create therapeutic strategies to combat CTD-ILD. However, similar to the understanding of disease pathogenesis, most therapeutic strategies that target macrophages in CTD-ILD originated from studies targeting macrophages in PF. Therefore, in the following section, we review therapeutic strategies to target macrophages in preclinical or clinical models of PF or CTD-ILD (Figure 3):
(1) ABT-199: ABT-199, an inhibitor of Bcl-2, is a potential therapeutic agent that prevents fibrosis progression. Bcl-2 decreases caspase-3 activity, mediating the apoptosis resistance of macrophages, which facilitates lung fibrosis (Figure 3). Mice have lung fibrosis resolution when treated with ABT-199 after fibrosis is established [36].
(2) Anti-IL-33: IL-33 is induced in macrophages during PF. Acting through ST2, IL-33 polarizes M2 macrophages to increase IL-13 and TGF-β1 production. This activates fibroblasts and stimulates fibroblast proliferation. Therefore, IL-33 enhances collagen synthesis by fibroblasts and amplifies the fibrosis of the lung. Accordingly, an anti-IL-33 antibody reduces lung fibrosis (Figure 3) [97].
(3) Clevudine: Clevudine is a purine nucleoside analog indicated for hepatitis B virus (HBV) treatment. Interestingly, clevudine can inhibit the PI3K/Akt signaling and therefore suppresses M2 differentiation. Correspondingly, epithelial-to-mesenchymal transition (EMT) and M2-induced myofibroblast activation are alleviated (Figure 3), thus preventing profibrotic cytokine secretion and downstream collagen deposition, with a concomitant attenuation of fibrotic processes in the lungs [98].
(4) Leflunomide: Leflunomide inhibits Cu,Zn-SOD-mediated H2O2 generation. Consequently, Jmjd3 expression is reduced, preventing macrophage M2 polarization and thereby attenuating lung tissue fibrosis (Figure 3) [49].
(5) GW4869: Macrophages secrete exosomes carrying AT1R, which are taken up by fibroblasts, activating collagen synthesis. GW4869, by inhibiting exosome secretion, attenuates fibrosis in a mouse model (Figure 3) [62].
(6) IL-10: IL-10 suppresses TGF-β production in macrophages and subsequently decreases collagen production in fibroblasts (Figure 3), diminishing the intensity of pulmonary tissue fibrosis [99].
(7) Methyl palmitate: Methyl palmitate inhibits the phosphorylation of inhibitory kappa B alpha (IκBα), resulting in decreased TNF-α and increased IL-10 production by macrophages (Figure 3). These changes ameliorate lung inflammation and fibrosis [100].
(8) Microcystin-leucine arginine (LR): The binding of microcystin-LR to glucose-regulated protein 78 kDa (GRP78) suppresses endoplasmic reticulum unfolded protein response (UPR) signaling, which inhibits M2 macrophage polarization (Figure 3) and leads to attenuated EMT and fibroblast–myofibroblast transition (FMT). As a result, microcystin-LR exerts a therapeutic effect on lung fibrosis [101].
(9) Niclosamide: S100A4 is secreted by M2 macrophages to enhance collagen production in lung fibroblasts. The inhibition of S100A4 production with niclosamide reduces collagen deposition and attenuates lung fibrosis (Figure 3) [75].
(10) Nintedanib: By inhibiting CSF1R, nintedanib blocks the M2 differentiation of monocytes (Figure 3), which reduces macrophage activation and ameliorates fibrosis in a CTD-ILD model [102].
(11) Pirfenidone: Pirfenidone has an indirect inhibitory effect on fibroblast proliferation by suppressing macrophage polarization toward the M2 phenotype (Figure 3), which is therefore helpful for lung fibrosis [103].
(12) PRI-724: β-catenin signaling promotes the TGF-β expression of macrophages, which contributes to collagen synthesis in fibroblasts. As a result, the β-catenin pathway inhibitor PRI-724 inhibits the production of TGF-β by alveolar macrophages and ameliorates collagen deposition in fibrotic lungs (Figure 3) [79,104].
(13) Recombinant human pentraxin-2 (rhPTX-2): Pentraxin-2, also called serum amyloid P (SAP), reduces M2 differentiation and subsequent lung fibrosis (Figure 3) [80,81]. In line with this, rhPTX-2 prevents the deterioration of lung function in PF patients [105].
(14) Schisandra: Schisandra decreases TGF-β, reduces the levels of Smad3 and Smad4, and upregulates Smad7, suppressing M2 macrophage polarization (Figure 3). Accordingly, Schisandra exerts protective effects on lung fibrosis [106].
(15) Ruxolitinib: In CTD-ILD, ruxolitinib, which inhibits JAK/STAT, prevents the upregulation of proinflammatory molecules (CXCL10, IFI44, NOS2, and TNF-α) and profibrotic molecules (ARG1, CHI3L3, FLT1, IL4Ra, and RETNLA) (Figure 3). Ruxolitinib concomitantly reduces M2 polarization and the mononuclear pulmonary infiltrate, leading to significantly reduced CTD-ILD in an animal model [107].
(16) Tacrolimus: In the animal model of PF, tacrolimus inhibits signaling of JAK/STAT pathways and suppresses M2 polarization and M2-induced myofibroblast activation, thereby alleviating PF progression [108].

6. Investigational Agents in Preclinical and Clinical Trials of CTD-ILD

In practice, there is no standard treatment protocol for CTD-ILD. However, considering the autoimmune nature of CTDs, immunomodulatory drugs are the cornerstones in the pharmacologic treatment of CTD-ILD [109]. Considering these uncertainties, therapies that address the ILD component of CTD are currently a field of active research [110]. According to clinicaltrials.gov (accessed on 25 March 2023), 39 clinical trials exploring the therapeutic effects of various agents for CTD-ILD have been planned or are already underway or completed (Table 3). Of the 16 therapeutic agents that act on non-macrophage cells, three (Cyclophosphamide, Mycophenolate Mofetil, and Tadalafil) exhibit therapeutic effects. Of the four therapeutic agents which act on macrophages, three (Nintedanib, Pirfenidone, and Tacrolimus) exhibit therapeutic effects, which is borderline higher than the percentage of agents which act on non-macrophage cells (p = 0.06 by Fisher’s Exact Test). However, most available clinical trials examined agents which act on non-macrophage cells (25 clinical trials) rather than agents that act on macrophage cells (14 clinical trials). Therefore, it is hoped that future clinical trials will focus more on macrophage-directed agents.

7. Concluding Remarks and Future Directions

Despite the significant morbidities and mortalities caused by CTD-ILD, publications detailing how macrophages contribute to ILD in the context of CTD are scarce. More research is necessary to elucidate the distinct roles of specific lung macrophage populations in the CTD-ILD, with the aim of providing new insights into the development of macrophage-directed therapeutic targets and diagnostic tools [31]. Current knowledge is mostly extrapolated from findings on animal models of PF. However, well-established mouse macrophage counterparts might not always be available in humans. A humanized mouse model in which mice are transplanted with alveolar organoids and hematopoietic stem and progenitor cells could be exploited to overcome these issues [69]. Blocking macrophage molecular mechanisms, boosting macrophage signaling pathways, or combinations of these may prove fruitful and provide opportunities to relieve the paucity of proven effective treatments [137] and meet the clinical needs of CTD-ILD. The literature shows that aiming at specific cell types is a viable choice for several disease entities [138,139,140], especially CTDs [141,142]. Several systemic reviews also suggest the therapeutic potential of targeting specific cells in CTD-ILD [143,144]. Moreover, the pathogenesis of CTD-ILD involves other cell types (such as lymphocytes and fibroblasts) whose crosstalk with macrophages requires further characterization [74]. Deciphering these interactions could be another direction of research. We hope that our efforts to elucidate the pathophysiology of ILD macrophages in the context of CTD will offer new insights into the development of macrophage-directed therapeutic approaches for this group of debilitating diseases.

Author Contributions

Conceptualization, C.-C.T., W.-T.L. and J.-H.Y.; investigation, K.-Y.C., P.-Y.W. and C.-Y.Y.; data curation, Y.-W.S., W.-Y.S., C.-C.W., T.-T.O., W.-C.T., C.-J.C., S.-C.L. and S.-J.C.; writing—original draft preparation, C.-C.T. and K.-Y.C.; writing—review and editing, C.-C.T., W.-T.L. and J.-H.Y.; supervision, W.-T.L. and J.-H.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Spagnolo, P.; Distler, O.; Ryerson, C.J.; Tzouvelekis, A.; Lee, J.S.; Bonella, F.; Bouros, D.; Hoffmann-Vold, A.M.; Crestani, B.; Matteson, E.L. Mechanisms of progressive fibrosis in connective tissue disease (CTD)-associated interstitial lung diseases (ILDs). Ann. Rheum. Dis. 2021, 80, 143–150. [Google Scholar] [CrossRef] [PubMed]
  2. Silva, M.; Fernandes, A.; Pereira, A.R.; Madanelo, S.; Clemêncio, T.; Ferreira, P.G. Awareness towards the main ILD among primary care physicians. Multidiscip. Respir. Med. 2022, 17, 848. [Google Scholar] [CrossRef] [PubMed]
  3. Fisseler-Eckhoff, A.; Märker-Hermann, E. Interstitial lung disease associated with connective tissue disease. Pathologe 2021, 42, 4–10. [Google Scholar] [CrossRef] [PubMed]
  4. Yoo, H.; Hino, T.; Han, J.; Franks, T.J.; Im, Y.; Hatabu, H.; Chung, M.P.; Lee, K.S. Connective tissue disease-related interstitial lung disease (CTD-ILD) and interstitial lung abnormality (ILA): Evolving concept of CT findings, pathology and management. Eur. J. Radiol. Open 2021, 8, 100311. [Google Scholar] [CrossRef]
  5. Ahmed, S.; Handa, R. Management of Connective Tissue Disease-related Interstitial Lung Disease. Curr. Pulmonol. Rep. 2022, 11, 86–98. [Google Scholar] [CrossRef]
  6. Mena-Vázquez, N.; Godoy-Navarrete, F.J.; Lisbona-Montañez, J.M.; Redondo-Rodriguez, R.; Manrique-Arija, S.; Rioja, J.; Mucientes, A.; Ruiz-Limón, P.; Garcia-Studer, A.; Ortiz-Márquez, F.; et al. Inflammatory Biomarkers in the Diagnosis and Prognosis of Rheumatoid Arthritis-Associated Interstitial Lung Disease. Int. J. Mol. Sci. 2023, 24, 6800. [Google Scholar] [CrossRef]
  7. Dsouza, K.G.; Alexander, A.S.; Watts, J.R.; Kulkarni, T. Management of interstitial lung disease in patients with autoimmune disease-related interstitial lung disease. Multidiscip. Respir. Med. 2023, 18, 890. [Google Scholar] [CrossRef]
  8. Ng, K.H.; Chen, D.Y.; Lin, C.H.; Chao, W.C.; Chen, Y.M.; Chen, Y.H.; Huang, W.N.; Hsieh, T.Y.; Lai, K.L.; Tang, K.T.; et al. Risk of interstitial lung disease in patients with newly diagnosed systemic autoimmune rheumatic disease: A nationwide, population-based cohort study. Semin. Arthritis Rheum. 2020, 50, 840–845. [Google Scholar] [CrossRef]
  9. Chen, F.; Wang, J.; Zhang, P.; Zuo, Y.; Ye, L.; Wang, G.; Shu, X. Interstitial Lung Disease in Dermatomyositis Without Myositis-Specific and Myositis-Associated Autoantibodies: Study of a Series of 72 Patients from a Single Cohort. Front. Immunol. 2022, 13, 879266. [Google Scholar] [CrossRef]
  10. Lu, J.; Liu, C.; Zhou, X.; Tang, J.; Liu, S.; Tang, M.; Li, M.; Zhu, L. Palmar erythema and palmar papules as predictors for dermatomyositis-related acute/subacute interstitial lung disease: A retrospective study. Rheumatology 2021, 61, 413–421. [Google Scholar] [CrossRef]
  11. Weng, C.; Ding, Z.; Zhou, Y.; Yang, Q.; Xue, L.; Zhang, L.; Wang, G.; Liu, Z. Clinical Characteristics of Dermatomyositis with Interstitial Lung Disease: A Retrospective Case-Control Study. Rheumatol. Ther. 2023, 10, 635–648. [Google Scholar] [CrossRef]
  12. Wang, Q.; Gao, C.; Zhang, C.; Yao, M.; Liang, W.; Sun, W.; Zheng, Z. Tumor markers are associated with rapidly progressive interstitial lung disease in adult-dermatomyositis. Clin. Rheumatol. 2022, 41, 1731–1739. [Google Scholar] [CrossRef]
  13. Pérez, N.; Gargiulo, M.L.Á.; Suarez, L.; Khoury, M.; Gómez, G. Clinical characteristics and prognostic factors in an Argentinian cohort with ANCA-associated vasculitis. Medicina 2021, 81, 198–207. [Google Scholar]
  14. Sweis, J.J.G.; Sweis, N.W.G.; Alnaimat, F.; Jansz, J.; Liao, T.E.; Alsakaty, A.; Azam, A.; Elmergawy, H.; Hanson, H.A.; Ascoli, C.; et al. Immune-mediated lung diseases: A narrative review. Front. Med. 2023, 10, 1160755. [Google Scholar] [CrossRef]
  15. Matsuda, S.; Kotani, T.; Suzuka, T.; Kiboshi, T.; Fukui, K.; Wakama, M.; Ishida, T.; Fujiki, Y.; Shiba, H.; Nagai, K.; et al. Evaluation of poor prognostic factors of respiratory related death in microscopic polyangiitis complicated by interstitial lung disease. Sci. Rep. 2021, 11, 1490. [Google Scholar] [CrossRef]
  16. Takakuwa, Y.; Yamasaki, Y.; Matsushita, H.; Kiyokawa, T.; Mizushima, M.; Tonooka, K.; Nagafuchi, H.; Matsuoka, S.; Ooka, S.; Kawahata, K. Long-term survival, causes of death, and prognostic factors for mortality in patients with microscopic polyangiitis and those with anti-neutrophil cytoplasmic antibody-positive interstitial lung disease: A single-center retrospective study. Int. J. Rheum. Dis. 2023, 26, 446–453. [Google Scholar] [CrossRef]
  17. Fidler, L.; Widdifield, J.; Fisher, J.H.; Shapera, S.; Gershon, A.S. Rheumatoid arthritis associated interstitial lung disease: Trends in epidemiology and mortality in Ontario from 2000 to 2018. Respir. Med. 2023, in press. [Google Scholar] [CrossRef]
  18. Ng, K.H.; Chen, D.Y.; Lin, C.H.; Chao, W.C.; Chen, H.H. Analysis of risk factors of mortality in rheumatoid arthritis patients with interstitial lung disease: A nationwide, population-based cohort study in Taiwan. RMD Open 2022, 8, e002343. [Google Scholar] [CrossRef]
  19. Sparks, J.A.; Jin, Y.; Cho, S.K.; Vine, S.; Desai, R.; Doyle, T.J.; Kim, S.C. Prevalence, incidence and cause-specific mortality of rheumatoid arthritis-associated interstitial lung disease among older rheumatoid arthritis patients. Rheumatology 2021, 60, 3689–3698. [Google Scholar] [CrossRef]
  20. Reid, P.; Guler, S.A. Mortality Trends in Rheumatoid Arthritis: Zooming in on Interstitial Lung Disease. Ann. Am. Thorac. Soc. 2021, 18, 1953–1954. [Google Scholar] [CrossRef]
  21. Panagopoulos, P.; Goules, A.; Hoffmann-Vold, A.M.; Matteson, E.L.; Tzioufas, A. Natural history and screening of interstitial lung disease in systemic autoimmune rheumatic disorders. Ther. Adv. Musculoskelet. Dis. 2021, 13, 1759720X211037519. [Google Scholar] [CrossRef] [PubMed]
  22. Li, Q.; Wallace, L.; Patnaik, P.; Alves, M.; Gahlemann, M.; Kohlbrenner, V.; Raabe, C.; Wang, J.R.; Garry, E.M. Disease frequency, patient characteristics, comorbidity outcomes and immunosuppressive therapy in systemic sclerosis and systemic sclerosis-associated interstitial lung disease: A US cohort study. Rheumatology 2021, 60, 1915–1925. [Google Scholar] [CrossRef] [PubMed]
  23. Hoffmann-Vold, A.M.; Fretheim, H.; Halse, A.K.; Seip, M.; Bitter, H.; Wallenius, M.; Garen, T.; Salberg, A.; Brunborg, C.; Midtvedt, Ø.; et al. Tracking Impact of Interstitial Lung Disease in Systemic Sclerosis in a Complete Nationwide Cohort. Am. J. Respir. Crit. Care Med. 2019, 200, 1258–1266. [Google Scholar] [CrossRef]
  24. Hyldgaard, C.; Bendstrup, E.; Pedersen, A.B.; Pedersen, L.; Ellingsen, T. Interstitial Lung Disease in Connective Tissue Diseases: Survival Patterns in a Population-Based Cohort. J. Clin. Med. 2021, 10, 4830. [Google Scholar] [CrossRef] [PubMed]
  25. Chen, S.; Saeed, A.F.U.H.; Liu, Q.; Jiang, Q.; Xu, H.; Xiao, G.G.; Rao, L.; Duo, Y. Macrophages in immunoregulation and therapeutics. Signal Transduct. Target. Ther. 2023, 8, 207. [Google Scholar] [CrossRef]
  26. Moreira, A.C.; Silva, T.; Mesquita, G.; Gomes, A.C.; Bento, C.M.; Neves, J.V.; Rodrigues, D.F.; Rodrigues, P.N.; Almeida, A.A.; Santambrogio, P.; et al. H-Ferritin Produced by Myeloid Cells Is Released to the Circulation and Plays a Major Role in Liver Iron Distribution during Infection. Int. J. Mol. Sci. 2021, 23, 269. [Google Scholar] [CrossRef]
  27. Enomoto, N. Pathological Roles of Pulmonary Cells in Acute Lung Injury: Lessons from Clinical Practice. Int. J. Mol. Sci. 2022, 23, 15027. [Google Scholar] [CrossRef]
  28. Hofbauer, D.; Mougiakakos, D.; Broggini, L.; Zaiss, M.; Büttner-Herold, M.; Bach, C.; Spriewald, B.; Neumann, F.; Bisht, S.; Nolting, J.; et al. β2-microglobulin triggers NLRP3 inflammasome activation in tumor-associated macrophages to promote multiple myeloma progression. Immunity 2021, 54, 1772–1787.e9. [Google Scholar] [CrossRef]
  29. Chen, J.; Zhang, R.; Xie, M.; Luan, C.; Li, X. Transcriptome Sequencing Identifies PLAUR as an Important Player in Patients with Dermatomyositis-Associated Interstitial Lung Disease. Front. Genet. 2021, 12, 784215. [Google Scholar] [CrossRef]
  30. Yang, Y.; Yin, G.; Hao, J.; Xie, Q.; Liu, Y. Serum Interleukin-18 Level is Associated with Disease Activity and Interstitial Lung Disease in Patients with Dermatomyositis. Arch. Rheumatol. 2017, 32, 181–188. [Google Scholar] [CrossRef]
  31. Byrne, A.J.; Maher, T.M.; Lloyd, C.M. Pulmonary Macrophages: A New Therapeutic Pathway in Fibrosing Lung Disease? Trends Mol. Med. 2016, 22, 303–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Koo, B.S.; Park, K.Y.; Lee, H.J.; Kim, H.J.; Ahn, H.S.; Yim, S.Y.; Jun, J.B. Effect of combined pulmonary fibrosis and emphysema on patients with connective tissue diseases and systemic sclerosis: A systematic review and meta-analysis. Arthritis Res. Ther. 2021, 23, 100. [Google Scholar] [CrossRef] [PubMed]
  33. Misharin, A.V.; Morales-Nebreda, L.; Reyfman, P.A.; Cuda, C.M.; Walter, J.M.; McQuattie-Pimentel, A.C.; Chen, C.I.; Anekalla, K.R.; Joshi, N.; Williams, K.J.N.; et al. Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. J. Exp. Med. 2017, 214, 2387–2404. [Google Scholar] [CrossRef]
  34. Aegerter, H.; Lambrecht, B.N.; Jakubzick, C.V. Biology of lung macrophages in health and disease. Immunity 2022, 55, 1564–1580. [Google Scholar] [CrossRef] [PubMed]
  35. Aran, D.; Looney, A.P.; Liu, L.; Wu, E.; Fong, V.; Hsu, A.; Chak, S.; Naikawadi, R.P.; Wolters, P.J.; Abate, A.R.; et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 2019, 20, 163–172. [Google Scholar] [CrossRef] [PubMed]
  36. Gu, L.; Surolia, R.; Larson-Casey, J.L.; He, C.; Davis, D.; Kang, J.; Antony, V.B.; Carter, A.B. Targeting Cpt1a-Bcl-2 interaction modulates apoptosis resistance and fibrotic remodeling. Cell Death Differ. 2022, 29, 118–132. [Google Scholar] [CrossRef]
  37. Ishida, Y.; Kuninaka, Y.; Mukaida, N.; Kondo, T. Immune Mechanisms of Pulmonary Fibrosis with Bleomycin. Int. J. Mol. Sci. 2023, 24, 3149. [Google Scholar] [CrossRef]
  38. Liu, S.; Liu, C.; Wang, Q.; Min, J. CC Chemokines in Idiopathic Pulmonary Fibrosis: Pathogenic Role and Therapeutic Potential. Biomolecules 2023, 13, 333. [Google Scholar] [CrossRef]
  39. Stahl, M.; Schupp, J.; Jäger, B.; Schmid, M.; Zissel, G.; Müller-Quernheim, J.; Prasse, A. Lung collagens perpetuate pulmonary fibrosis via CD204 and M2 macrophage activation. PLoS ONE 2013, 8, e81382. [Google Scholar] [CrossRef] [Green Version]
  40. Zhang, L.; Wang, Y.; Wu, G.; Xiong, W.; Gu, W.; Wang, C.Y. Macrophages: Friend or foe in idiopathic pulmonary fibrosis? Respir. Res. 2018, 19, 170. [Google Scholar]
  41. Li, J.; Zhai, X.; Sun, X.; Cao, S.; Yuan, Q.; Wang, J. Metabolic reprogramming of pulmonary fibrosis. Front. Pharmacol. 2022, 13, 1031890. [Google Scholar] [CrossRef]
  42. Xie, N.; Cui, H.; Ge, J.; Banerjee, S.; Guo, S.; Dubey, S.; Abraham, E.; Liu, R.M.; Liu, G. Metabolic characterization and RNA profiling reveal glycolytic dependence of profibrotic phenotype of alveolar macrophages in lung fibrosis. Am. J. Physiol. Lung Cell Mol. Physiol. 2017, 313, L834–L844. [Google Scholar] [CrossRef] [Green Version]
  43. Liu, S.; Lv, X.; Wei, X.; Liu, C.; Li, Q.; Min, J.; Hua, F.; Zhang, X.; Li, K.; Li, P.; et al. TRIB3–GSK-3β interaction promotes lung fibrosis and serves as a potential therapeutic target. Acta Pharm. Sin. B 2021, 11, 3105–3119. [Google Scholar] [CrossRef]
  44. Philip, K.; Mills, T.W.; Davies, J.; Chen, N.Y.; Karmouty-Quintana, H.; Luo, F.; Molina, J.G.; Amione-Guerra, J.; Sinha, N.; Guha, A.; et al. HIF1A up-regulates the ADORA2B receptor on alternatively activated macrophages and contributes to pulmonary fibrosis. FASEB J. 2017, 31, 4745–4758. [Google Scholar] [CrossRef] [Green Version]
  45. Ogger, P.P.; Albers, G.J.; Hewitt, R.J.; O’Sullivan, B.J.; Powell, J.E.; Calamita, E.; Ghai, P.; Walker, S.A.; McErlean, P.; Saunders, P.; et al. Itaconate controls the severity of pulmonary fibrosis. Sci. Immunol. 2020, 5, eabc1884. [Google Scholar] [CrossRef]
  46. Peng, X.; Moore, M.; Mathur, A.; Zhou, Y.; Sun, H.; Gan, Y.; Herazo-Maya, J.D.; Kaminski, N.; Hu, X.; Pan, H.; et al. Plexin C1 deficiency permits synaptotagmin 7-mediated macrophage migration and enhances mammalian lung fibrosis. FASEB J. 2016, 30, 4056–4070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Goda, C.; Balli, D.; Black, M.; Milewski, D.; Le, T.; Ustiyan, V.; Ren, X.; Kalinichenko, V.V.; Kalin, T.V. Loss of FOXM1 in macrophages promotes pulmonary fibrosis by activating p38 MAPK signaling pathway. PLoS Genet. 2020, 16, e1008692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. He, C.; Ryan, A.J.; Murthy, S.; Carter, A.B. Accelerated development of pulmonary fibrosis via Cu,Zn-superoxide dismutase-induced alternative activation of macrophages. J. Biol. Chem. 2013, 288, 20745–20757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. He, C.; Larson-Casey, J.L.; Gu, L.; Ryan, A.J.; Murthy, S.; Carter, A.B. Cu,Zn-Superoxide Dismutase-Mediated Redox Regulation of Jumonji Domain Containing 3 Modulates Macrophage Polarization and Pulmonary Fibrosis. Am. J. Respir. Cell Mol. Biol. 2016, 55, 58–71. [Google Scholar] [CrossRef] [Green Version]
  50. Guo, X.; Li, T.; Xu, Y.; Xu, X.; Zhu, Z.; Zhang, Y.; Xu, J.; Xu, K.; Cheng, H.; Zhang, X.; et al. Increased levels of Gab1 and Gab2 adaptor proteins skew interleukin-4 (IL-4) signaling toward M2 macrophage-driven pulmonary fibrosis in mice. J. Biol. Chem. 2017, 292, 14003–14015. [Google Scholar] [CrossRef] [Green Version]
  51. Pan, T.; Zhou, Q.; Miao, K.; Zhang, L.; Wu, G.; Yu, J.; Xu, Y.; Xiong, W.; Li, Y.; Wang, Y. Suppressing Sart1 to modulate macrophage polarization by siRNA-loaded liposomes: A promising therapeutic strategy for pulmonary fibrosis. Theranostics 2021, 11, 1192–1206. [Google Scholar] [CrossRef] [PubMed]
  52. Zhao, J.; Okamoto, Y.; Asano, Y.; Ishimaru, K.; Aki, S.; Yoshioka, K.; Takuwa, N.; Wada, T.; Inagaki, Y.; Takahashi, C.; et al. Sphingosine-1-phosphate receptor-2 facilitates pulmonary fibrosis through potentiating IL-13 pathway in macrophages. PLoS ONE 2018, 13, e0197604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Larson-Casey, J.L.; Deshane, J.S.; Ryan, A.J.; Thannickal, V.J.; Carter, A.B. Macrophage Akt1 Kinase-Mediated Mitophagy Modulates Apoptosis Resistance and Pulmonary Fibrosis. Immunity 2016, 44, 582–596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Yao, Y.; Wang, Y.; Zhang, Z.; He, L.; Zhu, J.; Zhang, M.; He, X.; Cheng, Z.; Ao, Q.; Cao, Y.; et al. Chop Deficiency Protects Mice Against Bleomycin-induced Pulmonary Fibrosis by Attenuating M2 Macrophage Production. Mol. Ther. 2016, 24, 915–925. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, Y.; Zhang, L.; Wu, G.R.; Zhou, Q.; Yue, H.; Rao, L.Z.; Yuan, T.; Mo, B.; Wang, F.X.; Chen, L.M.; et al. MBD2 serves as a viable target against pulmonary fibrosis by inhibiting macrophage M2 program. Sci. Adv. 2021, 7, eabb6075. [Google Scholar] [CrossRef] [PubMed]
  56. He, J.; Du, Y.; Li, G.; Xiao, P.; Sun, X.; Song, W.; Lai, L.; Xia, M.; Zhang, J.; Wang, Q. Myeloid Fbxw7 Prevents Pulmonary Fibrosis by Suppressing TGF-β Production. Front. Immunol. 2021, 12, 760138. [Google Scholar] [CrossRef] [PubMed]
  57. Huo, R.; Guo, Q.; Hu, J.; Li, N.; Gao, R.; Mi, L.; Zhang, Z.; Liu, H.; Guo, Z.; Zhao, H.; et al. Therapeutic Potential of Janus Kinase Inhibitors for the Management of Interstitial Lung Disease. Drug Des. Dev. Ther. 2022, 16, 991–998. [Google Scholar] [CrossRef]
  58. Fan, X.; Wang, Z. STAT1 antisense oligonucleotides attenuate the proinflammatory cytokine release of alveolar macrophages in bleomycin-induced fibrosis. Cell Mol. Immunol. 2005, 2, 211–217. [Google Scholar]
  59. Le, T.K.; Paris, C.; Khan, K.S.; Robson, F.; Ng, W.L.; Rocchi, P. Nucleic Acid-Based Technologies Targeting Coronaviruses. Trends Biochem. Sci. 2021, 46, 351–365. [Google Scholar] [CrossRef]
  60. Nakazawa, Y.; Ohtsuka, S.; Nakahashi-Oda, C.; Shibuya, A. Cutting Edge: Involvement of the Immunoreceptor CD300c2 on Alveolar Macrophages in Bleomycin-Induced Lung Fibrosis. J. Immunol. 2019, 203, 3107–3111. [Google Scholar] [CrossRef]
  61. Cardoneanu, A.; Burlui, A.M.; Macovei, L.A.; Bratoiu, I.; Richter, P.; Rezus, E. Targeting Systemic Sclerosis from Pathogenic Mechanisms to Clinical Manifestations: Why IL-6? Biomedicines 2022, 10, 318. [Google Scholar] [CrossRef]
  62. Sun, N.N.; Zhang, Y.; Huang, W.H.; Zheng, B.J.; Jin, S.Y.; Li, X.; Meng, Y. Macrophage exosomes transfer angiotensin II type 1 receptor to lung fibroblasts mediating bleomycin-induced pulmonary fibrosis. Chin. Med. J. 2021, 134, 2175–2185. [Google Scholar] [CrossRef]
  63. Chen, Y.; Hao, X.; Li, M.; Tian, Z.; Cheng, M. UGRP1-modulated MARCO+ alveolar macrophages contribute to age-related lung fibrosis. Immun. Ageing 2023, 20, 14. [Google Scholar] [CrossRef]
  64. Durairaj, P.; Venkatesan, S.; Narayanan, V.; Babu, M. Protective effects of curcumin on bleomycin-induced changes in lung glycoproteins. Mol. Cell Biochem. 2020, 469, 159–167. [Google Scholar] [CrossRef]
  65. Suka, M.; Kido, T.; Yoshioka, W.; Hachisuka, E.; Okoshi, H.; Yamauchi, T.; Hano, H.; Okano, T.; Yokoyama, M.; Yanagisawa, H. Single intratracheal administration of cross-linked water-soluble acrylic acid polymer causes acute alveolo-interstitial inflammation and the subsequent fibrotic formation possibly via the TGF-β1 pathway in the lung of rats. Toxicology 2021, 448, 152647. [Google Scholar] [CrossRef]
  66. Sciacchitano, S.; Lavra, L.; Morgante, A.; Ulivieri, A.; Magi, F.; De Francesco, G.P.; Bellotti, C.; Salehi, L.B.; Ricci, A. Galectin-3: One Molecule for an Alphabet of Diseases, from A to Z. Int. J. Mol. Sci. 2018, 19, 379. [Google Scholar] [CrossRef] [Green Version]
  67. Wang, Z.; Li, W.; Guo, Q.; Wang, Y.; Ma, L.; Zhang, X. Insulin-Like Growth Factor-1 Signaling in Lung Development and Inflammatory Lung Diseases. Biomed. Res. Int. 2018, 2018, 6057589. [Google Scholar] [CrossRef] [Green Version]
  68. Artlett, C.M. The Mechanism and Regulation of the NLRP3 Inflammasome during Fibrosis. Biomolecules 2022, 12, 634. [Google Scholar] [CrossRef]
  69. Ogawa, T.; Shichino, S.; Ueha, S.; Matsushima, K. Macrophages in lung fibrosis. Int. Immunol. 2021, 33, 665–671. [Google Scholar] [CrossRef]
  70. Liu, G.; Zhai, H.; Zhang, T.; Li, S.; Li, N.; Chen, J.; Gu, M.; Qin, Z.; Liu, X. New therapeutic strategies for IPF: Based on the “phagocytosis-secretion-immunization” network regulation mechanism of pulmonary macrophages. Biomed. Pharmacother. 2019, 118, 109230. [Google Scholar] [CrossRef]
  71. Ramírez-Hernández, A.A.; Velázquez-Enríquez, J.M.; Santos-Álvarez, J.C.; López-Martínez, A.; Reyes-Jiménez, E.; Carrasco-Torres, G.; González-García, K.; Vásquez-Garzón, V.R.; Baltierrez-Hoyos, R. The Role of Extracellular Vesicles in Idiopathic Pulmonary Fibrosis Progression: An Approach on Their Therapeutics Potential. Cells 2022, 11, 630. [Google Scholar] [CrossRef] [PubMed]
  72. Yao, M.Y.; Zhang, W.H.; Ma, W.T.; Liu, Q.H.; Xing, L.H.; Zhao, G.F. microRNA-328 in exosomes derived from M2 macrophages exerts a promotive effect on the progression of pulmonary fibrosis via FAM13A in a rat model. Exp. Mol. Med. 2019, 51, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Gao, R.; Peng, X.; Perry, C.; Sun, H.; Ntokou, A.; Ryu, C.; Gomez, J.L.; Reeves, B.C.; Walia, A.; Kaminski, N.; et al. Macrophage-derived netrin-1 drives adrenergic nerve-associated lung fibrosis. J. Clin. Investig. 2021, 131, e136542. [Google Scholar] [CrossRef] [PubMed]
  74. Gu, Y.; Lawrence, T.; Mohamed, R.; Liang, Y.; Yahaya, B.H. The emerging roles of interstitial macrophages in pulmonary fibrosis: A perspective from scRNA-seq analyses. Front. Immunol. 2022, 13, 923235. [Google Scholar] [CrossRef]
  75. Zhang, W.; Ohno, S.; Steer, B.; Klee, S.; Staab-Weijnitz, C.A.; Wagner, D.; Lehmann, M.; Stoeger, T.; Königshoff, M.; Adler, H. S100a4 Is Secreted by Alternatively Activated Alveolar Macrophages and Promotes Activation of Lung Fibroblasts in Pulmonary Fibrosis. Front. Immunol. 2018, 9, 1216. [Google Scholar] [CrossRef] [Green Version]
  76. Singh, A.; Chakraborty, S.; Wong, S.W.; Hefner, N.A.; Stuart, A.; Qadir, A.S.; Mukhopadhyay, A.; Bachmaier, K.; Shin, J.W.; Rehman, J.; et al. Nanoparticle targeting of de novo profibrotic macrophages mitigates lung fibrosis. Proc. Natl. Acad. Sci. USA 2022, 119, e2121098119. [Google Scholar] [CrossRef]
  77. Han, S.; Lu, Q.; Liu, X. Advances in cellular senescence in idiopathic pulmonary fibrosis (Review). Exp. Ther. Med. 2023, 25, 145. [Google Scholar] [CrossRef]
  78. Hou, J.; Shi, J.; Chen, L.; Lv, Z.; Chen, X.; Cao, H.; Xiang, Z.; Han, X. M2 macrophages promote myofibroblast differentiation of LR-MSCs and are associated with pulmonary fibrogenesis. Cell Commun. Signal. 2018, 16, 89. [Google Scholar] [CrossRef] [Green Version]
  79. Sennello, J.A.; Misharin, A.V.; Flozak, A.S.; Berdnikovs, S.; Cheresh, P.; Varga, J.; Kamp, D.W.; Budinger, G.R.; Gottardi, C.J.; Lam, A.P. Lrp5/β-Catenin Signaling Controls Lung Macrophage Differentiation and Inhibits Resolution of Fibrosis. Am. J. Respir. Cell Mol. Biol. 2017, 56, 191–201. [Google Scholar] [CrossRef] [Green Version]
  80. Glass, D.S.; Grossfeld, D.; Renna, H.A.; Agarwala, P.; Spiegler, P.; DeLeon, J.; Reiss, A.B. Idiopathic pulmonary fibrosis: Current and future treatment. Clin. Respir. J. 2022, 16, 84–96. [Google Scholar] [CrossRef]
  81. Maghsadi, Z.; Azadmehr, A.; Moghadamnia, A.A.; Feizi, F.; Hamidi, N. N-Acetylcysteine attenuated pulmonary fibrosis induced by bleomycin. Res. Pharm. Sci. 2023, 18, 177–184. [Google Scholar]
  82. Guiot, J.; Cambier, M.; Boeckx, A.; Henket, M.; Nivelles, O.; Gester, F.; Louis, E.; Malaise, M.; Dequiedt, F.; Louis, R.; et al. Macrophage-derived exosomes attenuate fibrosis in airway epithelial cells through delivery of antifibrotic miR-142-3p. Thorax 2020, 75, 870–881. [Google Scholar] [CrossRef]
  83. Enomoto, Y.; Suzuki, Y.; Hozumi, H.; Mori, K.; Kono, M.; Karayama, M.; Furuhashi, K.; Fujisawa, T.; Enomoto, N.; Nakamura, Y.; et al. Clinical significance of soluble CD163 in polymyositis-related or dermatomyositis-related interstitial lung disease. Arthritis Res. Ther. 2017, 19, 9. [Google Scholar] [CrossRef] [Green Version]
  84. Horiike, Y.; Suzuki, Y.; Fujisawa, T.; Yasui, H.; Karayama, M.; Hozumi, H.; Furuhashi, K.; Enomoto, N.; Nakamura, Y.; Inui, N.; et al. Successful classification of macrophage-mannose receptor CD206 in severity of anti-MDA5 antibody positive dermatomyositis associated ILD. Rheumatology 2019, 58, 2143–2152. [Google Scholar] [CrossRef]
  85. Ye, L.; Zuo, Y.; Chen, F.; Xu, Y.; Zhang, P.; Yang, H.; Lin, S.; Peng, Q.; Wang, G.; Shu, X. Resistin Expression Is Associated with Interstitial Lung Disease in Dermatomyositis. Front. Med. 2022, 9, 903887. [Google Scholar] [CrossRef]
  86. Matsuda, S.; Kotani, T.; Kuwabara, H.; Suzuka, T.; Kiboshi, T.; Wada, Y.; Ishida, T.; Fujiki, Y.; Shiba, H.; Hata, K.; et al. Association of M2 macrophages, Th2, and B cells with pathomechanism in microscopic polyangiitis complicated by interstitial lung disease. J. Rheumatol. 2022, 49, 913–921. [Google Scholar] [CrossRef]
  87. Matsuda, S.; Kotani, T.; Kuwabara, H.; Suzuka, T.; Kiboshi, T.; Fukui, K.; Ishida, T.; Fujiki, Y.; Shiba, H.; Hata, K.; et al. CCL2 produced by CD68+/CD163+ macrophages as a promising clinical biomarker of microscopic polyangiitis-interstitial lung disease. Rheumatology 2021, 60, 4643–4653. [Google Scholar] [CrossRef]
  88. Gaurav, R.; Mikuls, T.R.; Thiele, G.M.; Nelson, A.J.; Niu, M.; Guda, C.; Eudy, J.D.; Barry, A.E.; Wyatt, T.A.; Romberger, D.J.; et al. High-throughput analysis of lung immune cells in a combined murine model of agriculture dust-triggered airway inflammation with rheumatoid arthritis. PLoS ONE 2021, 16, e0240707. [Google Scholar] [CrossRef]
  89. Shiomi, A.; Usui, T.; Ishikawa, Y.; Shimizu, M.; Murakami, K.; Mimori, T. GM-CSF but not IL-17 is critical for the development of severe interstitial lung disease in SKG mice. J. Immunol. 2014, 193, 849–859. [Google Scholar] [CrossRef] [Green Version]
  90. Tsoyi, K.; Esposito, A.J.; Sun, B.; Bowen, R.G.; Xiong, K.; Poli, F.; Cardenas, R.; Chu, S.G.; Liang, X.; Ryter, S.W.; et al. Syndecan-2 regulates PAD2 to exert antifibrotic effects on RA-ILD fibroblasts. Sci. Rep. 2022, 12, 2847. [Google Scholar] [CrossRef]
  91. Xiong, L.; Ye, H.; Ma, W.L. Animal models of rheumatoid arthritis-associated interstitial lung disease. Immun. Inflamm. Dis. 2021, 9, 37–47. [Google Scholar] [CrossRef] [PubMed]
  92. Wu, E.K.; Henkes, Z.I.; McGowan, B.; Bell, R.D.; Velez, M.J.; Livingstone, A.M.; Ritchlin, C.T.; Schwarz, E.M.; Rahimi, H. TNF-Induced Interstitial Lung Disease in a Murine Arthritis Model: Accumulation of Activated Monocytes, Conventional Dendritic Cells, and CD21+/CD23− B Cell Follicles Is Prevented with Anti-TNF Therapy. J. Immunol. 2019, 203, 2837–2849. [Google Scholar] [CrossRef] [PubMed]
  93. Rudnik, M.; Hukara, A.; Kocherova, I.; Jordan, S.; Schniering, J.; Milleret, V.; Ehrbar, M.; Klingel, K.; Feghali-Bostwick, C.; Distler, O.; et al. Elevated Fibronectin Levels in Profibrotic CD14+ Monocytes and CD14+ Macrophages in Systemic Sclerosis. Front. Immunol. 2021, 12, 642891. [Google Scholar] [CrossRef] [PubMed]
  94. Gao, X.; Jia, G.; Guttman, A.; DePianto, D.J.; Morshead, K.B.; Sun, K.H.; Ramamoorthi, N.; Vander Heiden, J.A.; Modrusan, Z.; Wolters, P.J.; et al. Osteopontin Links Myeloid Activation and Disease Progression in Systemic Sclerosis. Cell Rep. Med. 2020, 1, 100140. [Google Scholar] [CrossRef]
  95. Sun, C.; Chen, S.Y. RGC32 Promotes Bleomycin-Induced Systemic Sclerosis in a Murine Disease Model by Modulating Classically Activated Macrophage Function. J. Immunol. 2018, 200, 2777–2785. [Google Scholar] [CrossRef] [Green Version]
  96. Bonhomme, O.; André, B.; Gester, F.; de Seny, D.; Moermans, C.; Struman, I.; Louis, R.; Malaise, M.; Guiot, J. Biomarkers in systemic sclerosis-associated interstitial lung disease: Review of the literature. Rheumatology 2019, 58, 1534–1546. [Google Scholar] [CrossRef] [Green Version]
  97. Li, D.; Guabiraba, R.; Besnard, A.G.; Komai-Koma, M.; Jabir, M.S.; Zhang, L.; Graham, G.J.; Kurowska-Stolarska, M.; Liew, F.Y.; McSharry, C.; et al. IL-33 promotes ST2-dependent lung fibrosis by the induction of alternatively activated macrophages and innate lymphoid cells in mice. J. Allergy Clin. Immunol. 2014, 134, 1422–1432.e11. [Google Scholar] [CrossRef]
  98. Li, S.; Gao, S.; Jiang, Q.; Liang, Q.; Luan, J.; Zhang, R.; Zhang, F.; Ruan, H.; Li, X.; Zhou, H.; et al. Clevudine attenuates bleomycin-induced early pulmonary fibrosis via regulating M2 macrophage polarization. Int. Immunopharmacol. 2021, 101, 108271. [Google Scholar] [CrossRef]
  99. Nakagome, K.; Dohi, M.; Okunishi, K.; Tanaka, R.; Miyazaki, J.; Yamamoto, K. In Vivo IL-10 gene delivery attenuates bleomycin induced pulmonary fibrosis by inhibiting the production and activation of TGF-beta in the lung. Thorax 2006, 61, 886–894. [Google Scholar] [CrossRef] [Green Version]
  100. El-Demerdash, E. Anti-inflammatory and antifibrotic effects of methyl palmitate. Toxicol. Appl. Pharmacol. 2011, 254, 238–244. [Google Scholar] [CrossRef]
  101. Wang, J.; Xu, L.; Xiang, Z.; Ren, Y.; Zheng, X.; Zhao, Q.; Zhou, Q.; Zhou, Y.; Wang, Y. Microcystin-LR ameliorates pulmonary fibrosis via modulating CD206+ M2-like macrophage polarization. Cell Death Dis. 2020, 11, 136. [Google Scholar] [CrossRef] [Green Version]
  102. Huang, J.; Maier, C.; Zhang, Y.; Soare, A.; Dees, C.; Beyer, C.; Harre, U.; Chen, C.W.; Distler, O.; Schett, G.; et al. Nintedanib inhibits macrophage activation and ameliorates vascular and fibrotic manifestations in the Fra2 mouse model of systemic sclerosis. Ann. Rheum. Dis. 2017, 76, 1941–1948. [Google Scholar] [CrossRef]
  103. Toda, M.; Mizuguchi, S.; Minamiyama, Y.; Yamamoto-Oka, H.; Aota, T.; Kubo, S.; Nishiyama, N.; Shibata, T.; Takemura, S. Pirfenidone suppresses polarization to M2 phenotype macrophages and the fibrogenic activity of rat lung fibroblasts. J. Clin. Biochem. Nutr. 2018, 63, 58–65. [Google Scholar] [CrossRef] [Green Version]
  104. Okazaki, H.; Sato, S.; Koyama, K.; Morizumi, S.; Abe, S.; Azuma, M.; Chen, Y.; Goto, H.; Aono, Y.; Ogawa, H.; et al. The novel inhibitor PRI-724 for Wnt/β-catenin/CBP signaling ameliorates bleomycin-induced pulmonary fibrosis in mice. Exp. Lung Res. 2019, 45, 188–199. [Google Scholar] [CrossRef]
  105. Raghu, G.; Hamblin, M.J.; Brown, A.W.; Golden, J.A.; Ho, L.A.; Wijsenbeek, M.S.; Vasakova, M.; Pesci, A.; Antin-Ozerkis, D.E.; Meyer, K.C.; et al. Long-term evaluation of the safety and efficacy of recombinant human pentraxin-2 (rhPTX-2) in patients with idiopathic pulmonary fibrosis (IPF): An open-label extension study. Respir. Res. 2022, 23, 129. [Google Scholar] [CrossRef]
  106. Guo, Z.; Li, S.; Zhang, N.; Kang, Q.; Zhai, H. Schisandra Inhibit Bleomycin-Induced Idiopathic Pulmonary Fibrosis in Rats via Suppressing M2 Macrophage Polarization. Biomed. Res. Int. 2020, 2020, 5137349. [Google Scholar] [CrossRef]
  107. Lescoat, A.; Lelong, M.; Jeljeli, M.; Piquet-Pellorce, C.; Morzadec, C.; Ballerie, A.; Jouneau, S.; Jego, P.; Vernhet, L.; Batteux, F.; et al. Combined anti-fibrotic and anti-inflammatory properties of JAK-inhibitors on macrophages in vitro and in vivo: Perspectives for scleroderma-associated interstitial lung disease. Biochem. Pharmacol. 2020, 178, 114103. [Google Scholar] [CrossRef]
  108. Liu, B.; Jiang, Q.; Chen, R.; Gao, S.; Xia, Q.; Zhu, J.; Zhang, F.; Shao, C.; Liu, X.; Li, X.; et al. Tacrolimus ameliorates bleomycin-induced pulmonary fibrosis by inhibiting M2 macrophage polarization via JAK2/STAT3 signaling. Int. Immunopharmacol. 2022, 113, 109424. [Google Scholar] [CrossRef]
  109. Boutel, M.; Boutou, A.; Pitsiou, G.; Garyfallos, A.; Dimitroulas, T. Efficacy and Safety of Nintedanib in Patients with Connective Tissue Disease-Interstitial Lung Disease (CTD-ILD): A Real-World Single Center Experience. Diagnostics 2023, 13, 1221. [Google Scholar] [CrossRef]
  110. Kamenova, A.; Tzouvelekis, A.; Margaritopoulos, G.A. Recent advances in the treatment of systemic sclerosis associated interstitial lung disease. Front. Med. 2023, 10, 1155771. [Google Scholar] [CrossRef]
  111. Tardella, M.; Di Carlo, M.; Carotti, M.; Giovagnoni, A.; Salaffi, F. Abatacept in rheumatoid arthritis-associated interstitial lung disease: Short-term outcomes and predictors of progression. Clin. Rheumatol. 2021, 40, 4861–4867. [Google Scholar] [CrossRef] [PubMed]
  112. Khanna, D.; Tashkin, D.P.; Wells, A.U.; Seibold, J.R.; Wax, S.; Vazquez-Mateo, C.; Fleuranceau-Morel, P.; Damian, D.; Denton, C.P. STRATUS: A Phase II Study of Abituzumab in Patients With Systemic Sclerosis-associated Interstitial Lung Disease. J. Rheumatol. 2021, 48, 1295–1298. [Google Scholar] [CrossRef]
  113. Ll Wilkinson, M.G.; Deakin, C.T.; Papadopoulou, C.; Eleftheriou, D.; Wedderburn, L.R. JAK inhibitors: A potential treatment for JDM in the context of the role of interferon-driven pathology. Pediatr. Rheumatol. Online J. 2021, 19, 146. [Google Scholar] [CrossRef] [PubMed]
  114. Mwangi, J.; Litteken, C.; Gorthi, R.; Attoti, Y.; Atluri, R. Belimumab in the Treatment of Connective Tissue Disease-Associated Interstitial Lung Disease: Case Report and Literature Review. Cureus 2021, 13, e19218. [Google Scholar] [CrossRef] [PubMed]
  115. Penke, L.R.K.; Speth, J.; Wettlaufer, S.; Draijer, C.; Peters-Golden, M. Bortezomib Inhibits Lung Fibrosis and Fibroblast Activation without Proteasome Inhibition. Am. J. Respir. Cell Mol. Biol. 2022, 66, 23–37. [Google Scholar] [CrossRef]
  116. Giacomelli, R.; Liakouli, V.; Berardicurti, O.; Ruscitti, P.; Di Benedetto, P.; Carubbi, F.; Guggino, G.; Di Bartolomeo, S.; Ciccia, F.; Triolo, G.; et al. Interstitial lung disease in systemic sclerosis: Current and future treatment. Rheumatol. Int. 2017, 37, 853–863. [Google Scholar] [CrossRef]
  117. Harari, S.; Wells, A.U.; Wuyts, W.A.; Nathan, S.D.; Kirchgaessler, K.U.; Bengus, M.; Behr, J. The 6-min walk test as a primary end-point in interstitial lung disease. Eur. Respir. Rev. 2022, 31, 220087. [Google Scholar] [CrossRef]
  118. Vonk, M.C. Is there still a role for cyclophosphamide in the treatment of systemic sclerosis? J. Scleroderma Relat. Disord. 2021, 6, 117–122. [Google Scholar] [CrossRef]
  119. Kim, G.H.J.; Tashkin, D.P.; Lo, P.; Brown, M.S.; Volkmann, E.R.; Gjertson, D.W.; Khanna, D.; Elashoff, R.M.; Tseng, C.H.; Roth, M.D.; et al. Using Transitional Changes on High-Resolution Computed Tomography to Monitor the Impact of Cyclophosphamide or Mycophenolate Mofetil on Systemic Sclerosis-Related Interstitial Lung Disease. Arthritis Rheumatol. 2020, 72, 316–325. [Google Scholar] [CrossRef]
  120. Eapen, M.S.; Gaikwad, A.V.; Thompson, I.E.; Lu, W.; Myers, S.; Sharma, P.; Sohal, S.S. The effectiveness of immunosuppressive cyclosporin in attenuating the progression of interstitial lung diseases. J. Thorac. Dis. 2019, 11, S1139–S1142. [Google Scholar] [CrossRef]
  121. Chen, I.C.; Liu, Y.C.; Wu, Y.H.; Lo, S.H.; Dai, Z.K.; Hsu, J.H.; Tseng, Y.H. Evaluation of Proteasome Inhibitors in the Treatment of Idiopathic Pulmonary Fibrosis. Cells 2022, 11, 1543. [Google Scholar] [CrossRef]
  122. Assassi, S.; Volkmann, E.R.; Zheng, W.J.; Wang, X.; Wilhalme, H.; Lyons, M.A.; Roth, M.D.; Tashkin, D.P. Peripheral blood gene expression profiling shows predictive significance for response to mycophenolate in systemic sclerosis-related interstitial lung disease. Ann. Rheum. Dis. 2022, 81, 854–860. [Google Scholar] [CrossRef]
  123. Naidu, G.S.R.S.; Sharma, S.K.; Adarsh, M.B.; Dhir, V.; Sinha, A.; Dhooria, S.; Jain, S. Effect of mycophenolate mofetil (MMF) on systemic sclerosis-related interstitial lung disease with mildly impaired lung function: A double-blind, placebo-controlled, randomized trial. Rheumatol. Int. 2020, 40, 207–216. [Google Scholar] [CrossRef]
  124. Kulshrestha, R.; Pandey, A.; Jaggi, A.; Bansal, S. Beneficial effects of N-acetylcysteine on protease-antiprotease balance in attenuating bleomycin-induced pulmonary fibrosis in rats. Iran. J. Basic. Med. Sci. 2020, 23, 396–405. [Google Scholar]
  125. Denton, C.P.; Goh, N.S.; Humphries, S.M.; Maher, T.M.; Spiera, R.; Devaraj, A.; Ho, L.; Stock, C.; Erhardt, E.; Alves, M.; et al. Extent of fibrosis and lung function decline in patients with systemic sclerosis and interstitial lung disease: Data from the SENSCIS trial. Rheumatology 2023, 62, 1870–1876. [Google Scholar] [CrossRef] [PubMed]
  126. Solomon, J.J.; Danoff, S.K.; Woodhead, F.A.; Hurwitz, S.; Maurer, R.; Glaspole, I.; Dellaripa, P.F.; Gooptu, B.; Vassallo, R.; Cox, P.G.; et al. Safety, tolerability, and efficacy of pirfenidone in patients with rheumatoid arthritis-associated interstitial lung disease: A randomised, double-blind, placebo-controlled, phase 2 study. Lancet Respir. Med. 2023, 11, 87–96. [Google Scholar] [CrossRef]
  127. Hsu, V.M.; Denton, C.P.; Domsic, R.T.; Furst, D.E.; Rischmueller, M.; Stanislav, M.; Steen, V.D.; Distler, J.H.W.; Korish, S.; Cooper, A.; et al. Pomalidomide in Patients with Interstitial Lung Disease due to Systemic Sclerosis: A Phase II, Multicenter, Randomized, Double-blind, Placebo-controlled, Parallel-group Study. J. Rheumatol. 2018, 45, 405–410. [Google Scholar] [CrossRef]
  128. Available online: https://www.prometheusbiosciences.com/pipeline/pra023/tl1a/ (accessed on 30 March 2023).
  129. Maher, T.M.; Tudor, V.A.; Saunders, P.; Gibbons, M.A.; Fletcher, S.V.; Denton, C.P.; Hoyles, R.K.; Parfrey, H.; Renzoni, E.A.; Kokosi, M.; et al. Rituximab versus intravenous cyclophosphamide in patients with connective tissue disease-associated interstitial lung disease in the UK (RECITAL): A double-blind, double-dummy, randomised, controlled, phase 2b trial. Lancet Respir. Med. 2023, 11, 45–54. [Google Scholar] [CrossRef]
  130. Takada, K.; Katada, Y.; Ito, S.; Hayashi, T.; Kishi, J.; Itoh, K.; Yamashita, H.; Hirakata, M.; Kawahata, K.; Kawakami, A.; et al. Impact of adding tacrolimus to initial treatment of interstitial pneumonitis in polymyositis/dermatomyositis: A single-arm clinical trial. Rheumatology 2020, 59, 1084–1093. [Google Scholar] [CrossRef] [Green Version]
  131. Kuwana, M.; Wakasugi, N.; Furuya, T.; Uno, S.; Suda, T. Tacrolimus in Patients With Interstitial Pneumonia Associated With Polymyositis or Dermatomyositis: Interim Report of Postmarketing Surveillance in Japan. J. Rheumatol. 2022, 49, 707–718. [Google Scholar] [CrossRef]
  132. Mansour, S.M.; El-Abhar, H.S.; Soubh, A.A. MiR-200a inversely correlates with Hedgehog and TGF-β canonical/non-canonical trajectories to orchestrate the anti-fibrotic effect of Tadalafil in a bleomycin-induced pulmonary fibrosis model. Inflammopharmacology 2021, 29, 167–182. [Google Scholar] [CrossRef] [PubMed]
  133. Parida, J.; Nath, A.; Neyaz, Z.; Agarwal, V. A double blind randomized control trial of oral tadalafil in interstitial lung disease of scleroderma [abstract]. Arthritis Rheum. 2014, 66, S739. [Google Scholar]
  134. Kalyoncu, U.; Bilgin, E.; Erden, A.; Satış, H.; Tufan, A.; Tekgöz, E.; Ateş, A.; Coşkun, B.N.; Yağız, B.; Küçükşahin, O.; et al. Efficacy and safety of tofacitinib in rheumatoid arthritis-associated interstitial lung disease: TReasure real-life data. Clin. Exp. Rheumatol. 2022, 40, 2071–2077. [Google Scholar] [PubMed]
  135. Harrison, C. Itch receptor OSMR attracts industry. Nat. Biotechnol. 2022, 40, 1306. [Google Scholar] [CrossRef]
  136. Yaseen, B.; Lopez, H.; Taki, Z.; Zafar, S.; Rosario, H.; Abdi, B.A.; Vigneswaran, S.; Xing, F.; Arumalla, N.; Black, S.; et al. Interleukin-31 promotes pathogenic mechanisms underlying skin and lung fibrosis in scleroderma. Rheumatology 2020, 59, 2625–2636. [Google Scholar] [CrossRef]
  137. Erre, G.L.; Sebastiani, M.; Manfredi, A.; Gerratana, E.; Atzeni, F.; Passiu, G.; Mangoni, A.A. Antifibrotic drugs in connective tissue disease-related interstitial lung disease (CTD-ILD): From mechanistic insights to therapeutic applications. Drugs Context 2021, 10, 2020-8-6. [Google Scholar] [CrossRef]
  138. Faust, A.; Bäumer, N.; Schlütermann, A.; Becht, M.; Greune, L.; Geyer, C.; Rüter, C.; Margeta, R.; Wittmann, L.; Dersch, P.; et al. Tumor-Cell-Specific Targeting of Ibrutinib: Introducing Electrostatic Antibody-Inhibitor Conjugates (AiCs). Angew. Chem. Int. Ed. Engl. 2022, 61, e202109769. [Google Scholar] [CrossRef]
  139. Gearty, S.V.; Dündar, F.; Zumbo, P.; Espinosa-Carrasco, G.; Shakiba, M.; Sanchez-Rivera, F.J.; Socci, N.D.; Trivedi, P.; Lowe, S.W.; Lauer, P.; et al. An autoimmune stem-like CD8 T cell population drives type 1 diabetes. Nature 2022, 602, 156–161. [Google Scholar] [CrossRef]
  140. Nie, Y.; Zhao, L.; Zhang, X. B Cell Aberrance in Lupus: The Ringleader and the Solution. Clin. Rev. Allergy Immunol. 2022, 62, 301–323. [Google Scholar] [CrossRef]
  141. Bohdziewicz, A.; Pawlik, K.K.; Maciejewska, M.; Sikora, M.; Alda-Malicka, R.; Czuwara, J.; Rudnicka, L. Future Treatment Options in Systemic Sclerosis-Potential Targets and Ongoing Clinical Trials. J. Clin. Med. 2022, 11, 1310. [Google Scholar] [CrossRef]
  142. Radziszewska, A.; Moulder, Z.; Jury, E.C.; Ciurtin, C. CD8+ T Cell Phenotype and Function in Childhood and Adult-Onset Connective Tissue Disease. Int. J. Mol. Sci. 2022, 23, 11431. [Google Scholar] [CrossRef]
  143. Xu, L.; Wang, F.; Luo, F. Rituximab for the treatment of connective tissue disease-associated interstitial lung disease: A systematic review and meta-analysis. Front. Pharmacol. 2022, 13, 1019915. [Google Scholar] [CrossRef]
  144. Bellan, M.; Patrucco, F.; Barone-Adesi, F.; Gavelli, F.; Castello, L.M.; Nerviani, A.; Andreoli, L.; Cavagna, L.; Pirisi, M.; Sainaghi, P.P. Targeting CD20 in the treatment of interstitial lung diseases related to connective tissue diseases: A systematic review. Autoimmun. Rev. 2020, 19, 102453. [Google Scholar] [CrossRef]
Figure 1. Incidence, histopathology features, morbidities, and mortalities of individual CTD-ILD discussed in this review. The figure summarizes incidence, histopathology features, morbidities, and mortalities of CTD-ILD discussed in this review. (1) The incidence of DM-ILD is 1011/105. Histopathological changes associated with DM-ILD are organizing pneumonia (OP), nonspecific interstitial pneumonia (NSIP), and usual interstitial pneumonia (UIP). Patients have increased palmar papules and myocardial involvement, with mortality rate of up to 24.62%. (2) The incidence of MPA-ILD is around 1.4/105. The pathological patterns are often UIP and NSIP. It is associated with decreased renal function and has a mortality rate of up to 39%. (3) The estimated incidence of RA-ILD is 3.7/105. It is characterized by UIP. The patients usually suffer from increased asthma, chronic obstructive pulmonary disease (COPD), diabetes, and heart disease, and the mortality rate is 8.08%. (4) The incidence of SSc-ILD is 1364/105. It is characterized by NSIP and UIP, accompanied by COPD, gastroesophageal reflux disease (GERD), hypertension, and skin disorders and a mortality rate of about 39%. Red arrow—increase; blue arrow—decrease.
Figure 1. Incidence, histopathology features, morbidities, and mortalities of individual CTD-ILD discussed in this review. The figure summarizes incidence, histopathology features, morbidities, and mortalities of CTD-ILD discussed in this review. (1) The incidence of DM-ILD is 1011/105. Histopathological changes associated with DM-ILD are organizing pneumonia (OP), nonspecific interstitial pneumonia (NSIP), and usual interstitial pneumonia (UIP). Patients have increased palmar papules and myocardial involvement, with mortality rate of up to 24.62%. (2) The incidence of MPA-ILD is around 1.4/105. The pathological patterns are often UIP and NSIP. It is associated with decreased renal function and has a mortality rate of up to 39%. (3) The estimated incidence of RA-ILD is 3.7/105. It is characterized by UIP. The patients usually suffer from increased asthma, chronic obstructive pulmonary disease (COPD), diabetes, and heart disease, and the mortality rate is 8.08%. (4) The incidence of SSc-ILD is 1364/105. It is characterized by NSIP and UIP, accompanied by COPD, gastroesophageal reflux disease (GERD), hypertension, and skin disorders and a mortality rate of about 39%. Red arrow—increase; blue arrow—decrease.
Ijms 24 11995 g001
Figure 2. Roles of macrophages in CTD-ILD. The figure depicts mechanisms through which macrophages modulate CTD-ILD. In DM-ILD, macrophages express resistin. Resistin activates the NFκB signaling pathway, increasing the secretion of IL-1, IL-6, and TNF-α. This triggers the expression of resistin in macrophages, creating a self-perpetuating reaction to mediate the inflammatory pathogenesis of DM-ILD. In MPA-ILD, macrophages secrete CCL2, which attracts more macrophages to infiltrate. The recruited macrophages produce CCL2, initiating a self-propagating response. Meanwhile, CCL2 stimulates fibroblast differentiation into myofibroblasts and generates ECM, leading to exaggerated lung fibrosis. In RA-ILD, macrophages are implicated in three ways: (1) Exaggerated IL-6 secretion by macrophages triggers the proliferation of GM-CSF-producing T cells, and GM-CSF recruits neutrophil infiltration into the lung, leading to RA-ILD progression. (2) Macrophage-derived SDC2 interferes with PI3K/Akt signaling, decreases PAD2 in fibroblasts, and reduces collagen deposition in the lungs. (3) Macrophages produce high levels of TNF, which leads to an inflammatory phase predominated by cellular infiltration into the lungs and a subsequent shift to a fibrotic phase with irreversible collagen deposition. In SSc-ILD, six pathways are activated by macrophages to drive SSc-ILD: (1) Macrophages produce increased CCL18, which facilitates collagen production. (2) Macrophages release increased fibronectin, which induces fibroblast proliferation and collagen deposition. (3) Immune complexes stimulate the production of M-CSF and IL-6, which further induce OPN from monocytes. In turn, OPN triggers fibroblast migration, driving fibrosis progression. (4) SSc-ILD shows decreased PLXNC1 expression in macrophages, which boosts macrophage migration. (5) RGC32 promotes iNOS/IL-1β-directed inflammation through NFκB signaling and subsequently enhances the fibrotic response. (6) Macrophage-derived YKL-40, which is increased in SSc-ILD, promotes fibroblast proliferation. Red arrow—increase; Blue arrow—decrease; Black arrow—lead to.
Figure 2. Roles of macrophages in CTD-ILD. The figure depicts mechanisms through which macrophages modulate CTD-ILD. In DM-ILD, macrophages express resistin. Resistin activates the NFκB signaling pathway, increasing the secretion of IL-1, IL-6, and TNF-α. This triggers the expression of resistin in macrophages, creating a self-perpetuating reaction to mediate the inflammatory pathogenesis of DM-ILD. In MPA-ILD, macrophages secrete CCL2, which attracts more macrophages to infiltrate. The recruited macrophages produce CCL2, initiating a self-propagating response. Meanwhile, CCL2 stimulates fibroblast differentiation into myofibroblasts and generates ECM, leading to exaggerated lung fibrosis. In RA-ILD, macrophages are implicated in three ways: (1) Exaggerated IL-6 secretion by macrophages triggers the proliferation of GM-CSF-producing T cells, and GM-CSF recruits neutrophil infiltration into the lung, leading to RA-ILD progression. (2) Macrophage-derived SDC2 interferes with PI3K/Akt signaling, decreases PAD2 in fibroblasts, and reduces collagen deposition in the lungs. (3) Macrophages produce high levels of TNF, which leads to an inflammatory phase predominated by cellular infiltration into the lungs and a subsequent shift to a fibrotic phase with irreversible collagen deposition. In SSc-ILD, six pathways are activated by macrophages to drive SSc-ILD: (1) Macrophages produce increased CCL18, which facilitates collagen production. (2) Macrophages release increased fibronectin, which induces fibroblast proliferation and collagen deposition. (3) Immune complexes stimulate the production of M-CSF and IL-6, which further induce OPN from monocytes. In turn, OPN triggers fibroblast migration, driving fibrosis progression. (4) SSc-ILD shows decreased PLXNC1 expression in macrophages, which boosts macrophage migration. (5) RGC32 promotes iNOS/IL-1β-directed inflammation through NFκB signaling and subsequently enhances the fibrotic response. (6) Macrophage-derived YKL-40, which is increased in SSc-ILD, promotes fibroblast proliferation. Red arrow—increase; Blue arrow—decrease; Black arrow—lead to.
Ijms 24 11995 g002
Figure 3. Potential agents to target macrophages in CTD-ILD. The figure depicts potential strategies to target macrophages in the context of CTD-ILD according to the literature. (1) ABT-199 inhibits Bcl-2. As a result, ABT-199 augments caspase-3 activity and enhances macrophage apoptosis, counteracting lung fibrosis. (2) Anti-IL-33 blocks IL-33 action on ST2 and thereby downregulates IL-13 and TGF-β1 production, which decreases collagen synthesis by fibroblasts. (3) Clevudine, as an inhibitor of the PI3K/Akt signaling pathway, prevents M2 polarization and the subsequent fibrotic response of the lungs. (4) By inhibiting Cu,Zn-SOD-mediated H2O2 generation, leflunomide downregulates Jmjd3 expression, lowers M2 polarization, and diminishes lung fibrosis. (5) GW4869, which eliminates the secretion of AT1R-carrying exosomes from macrophages, decreases collagen synthesis in fibroblasts and ameliorates lung fibrosis. (6) IL-10 curbs TGF-β production in macrophages and prevents collagen deposition by fibroblasts. (7) Methyl palmitate blocks IκBα phosphorylation, reducing TNF-α and boosting IL-10 expression in macrophages, which inhibit lung inflammation and fibrosis. (8) After binding to GRP78, microcystin-LR interferes with UPR signaling, subsequently preventing M2 macrophage polarization and leading to attenuated lung fibrosis. (9) Niclosamide interferes with S100A4 production by macrophages, which ameliorates collagen production by fibroblasts. These changes culminate in reducing the fibrosis of the lungs. (10) Nintedanib limits M2 differentiation by blocking CSF1R and consequently improves fibrosis in a CTD-ILD model. (11) Pirfenidone acts on macrophages to block M2 polarization and, as a result, suppresses fibroblast proliferation and restricts lung fibrosis. (12) The β-catenin pathway inhibitor PRI-724, which blocks β-catenin signaling, lowers TGF-β production in macrophages and contributes to mitigated collagen production by fibroblasts. (13) rhPTX-2 prevents M2 differentiation and avoids PF deterioration. (14) Schisandra reduces TGF-β and downstream Smad3/Smad4 while stimulating Smad7 production. These signaling events disrupt M2 polarization and thereby improving lung fibrosis. (15) Ruxolitinib and (16) Tacrolimus, by inhibiting JAK/STAT signaling, suppress M2 polarizations and hence improve lung fibrosis in an animal model. Red arrow—increase; blue arrow—decrease; cross symbol—block; dashed line—act on.
Figure 3. Potential agents to target macrophages in CTD-ILD. The figure depicts potential strategies to target macrophages in the context of CTD-ILD according to the literature. (1) ABT-199 inhibits Bcl-2. As a result, ABT-199 augments caspase-3 activity and enhances macrophage apoptosis, counteracting lung fibrosis. (2) Anti-IL-33 blocks IL-33 action on ST2 and thereby downregulates IL-13 and TGF-β1 production, which decreases collagen synthesis by fibroblasts. (3) Clevudine, as an inhibitor of the PI3K/Akt signaling pathway, prevents M2 polarization and the subsequent fibrotic response of the lungs. (4) By inhibiting Cu,Zn-SOD-mediated H2O2 generation, leflunomide downregulates Jmjd3 expression, lowers M2 polarization, and diminishes lung fibrosis. (5) GW4869, which eliminates the secretion of AT1R-carrying exosomes from macrophages, decreases collagen synthesis in fibroblasts and ameliorates lung fibrosis. (6) IL-10 curbs TGF-β production in macrophages and prevents collagen deposition by fibroblasts. (7) Methyl palmitate blocks IκBα phosphorylation, reducing TNF-α and boosting IL-10 expression in macrophages, which inhibit lung inflammation and fibrosis. (8) After binding to GRP78, microcystin-LR interferes with UPR signaling, subsequently preventing M2 macrophage polarization and leading to attenuated lung fibrosis. (9) Niclosamide interferes with S100A4 production by macrophages, which ameliorates collagen production by fibroblasts. These changes culminate in reducing the fibrosis of the lungs. (10) Nintedanib limits M2 differentiation by blocking CSF1R and consequently improves fibrosis in a CTD-ILD model. (11) Pirfenidone acts on macrophages to block M2 polarization and, as a result, suppresses fibroblast proliferation and restricts lung fibrosis. (12) The β-catenin pathway inhibitor PRI-724, which blocks β-catenin signaling, lowers TGF-β production in macrophages and contributes to mitigated collagen production by fibroblasts. (13) rhPTX-2 prevents M2 differentiation and avoids PF deterioration. (14) Schisandra reduces TGF-β and downstream Smad3/Smad4 while stimulating Smad7 production. These signaling events disrupt M2 polarization and thereby improving lung fibrosis. (15) Ruxolitinib and (16) Tacrolimus, by inhibiting JAK/STAT signaling, suppress M2 polarizations and hence improve lung fibrosis in an animal model. Red arrow—increase; blue arrow—decrease; cross symbol—block; dashed line—act on.
Ijms 24 11995 g003
Table 3. Efficacy of investigational agents in clinical trials of CTD-ILD in clinicaltrials.gov.
Table 3. Efficacy of investigational agents in clinical trials of CTD-ILD in clinicaltrials.gov.
AgentsTargeted CellsDiseasesStudyOutcome a
AbataceptT cells [111]Antisynthetase syndrome-associated ILDNCT03215927Active
AbataceptT cells [111]RA-ILDNCT03084419Unknown
AbituzumabEpithelial cells [112]SSc-ILDNCT02745145Terminated due to slow enrollment [112]
BasiliximabLymphocytes [113]DM-ILDNCT03192657Unknown
BelimumabB cells [114]SSc-ILDNCT05878717Not yet recruiting
BortezomibFibroblasts [115]SSc-ILDNCT02370693Completed
BosentanFibroblasts [116]SSc-ILDNCT00070590Ineffective [117]
BosentanFibroblasts [116]SSc-ILDNCT00319033Completed
CyclophosphamideLymphocytes [118]Antisynthetase syndrome-associated ILDNCT03770663Recruiting
CyclophosphamideLymphocytes [118]SSc-ILDNCT00883129Improve [119]
CyclophosphamideLymphocytes [118]SSc-ILDNCT01570764Completed
Cyclosporin AT cells [120]Sjogren’s syndrome-associated ILDNCT02370550Unknown
IxazomibFibroblasts [121]SSc-ILDNCT04837131Recruiting
Mycophenolate mofetilLymphocytes [122]Myositis-associated ILDNCT05129410Recruiting
Mycophenolate mofetilLymphocytes [122]SSc-ILDNCT00883129Improve [119]
Mycophenolate mofetilLymphocytes [122]SSc-ILDNCT02896205Ineffective [123]
Mycophenolate mofetilLymphocytes [122]SSc-ILDNCT05785065Not yet recruiting
N-acetylcysteineMacrophages [124]CTD-ILDNCT01424033Terminated due to departure of principal investigator.
NintedanibMacrophagesMyositis-associated ILDNCT05335278Recruiting
NintedanibMacrophagesMyositis-associated ILDNCT05799755Not yet recruiting
NintedanibMacrophagesSSc-ILDNCT02597933Effective [125]
NintedanibMacrophagesSSc-ILDNCT03313180Completed
PirfenidoneMacrophagesCTD-ILDNCT04928586Recruiting
PirfenidoneMacrophagesCTD-ILDNCT05505409Recruiting
PirfenidoneMacrophagesDM-ILDNCT02821689Unknown
PirfenidoneMacrophagesDM-ILDNCT03857854Unknown
PirfenidoneMacrophagesRA-ILDNCT02808871Slow FVC decline [126]
PirfenidoneMacrophagesSSc-ILDNCT03221257Completed
PirfenidoneMacrophagesSSc-ILDNCT03856853Unknown
PomalidomideT cells [127]SSc-ILDNCT01559129Discontinued [127]
PRA023Fibroblasts [128]SSc-ILDNCT05270668Recruiting
RituximabB cells [129]CTD-ILDNCT01862926Not superior to cyclophosphamide [129]
RituximabB cells [129]RA-ILDNCT00578565Inconclusive
TacrolimusMacrophagesMyositis-associated ILDNCT00504348Improve [130]
TacrolimusMacrophagesMyositis-associated ILDNCT02159651Improve [131]
TadalafilFibroblasts [132]SSc-ILDNCT01553981Improve [133]
TofacitinibMyeloid derived suppressor cells [134]RA-ILDNCT04311567Recruiting
TofacitinibMyeloid derived suppressor cells [134]RA-ILDNCT05246293Recruiting
VixarelimabFibroblasts [135,136]SSc-ILDNCT05785624Recruiting
a Recorded at clinicaltrials.gov or in the literature. FVC: functional vital capacity. Unknown: no result has been posted, or the recruitment status is unknown.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tseng, C.-C.; Sung, Y.-W.; Chen, K.-Y.; Wang, P.-Y.; Yen, C.-Y.; Sung, W.-Y.; Wu, C.-C.; Ou, T.-T.; Tsai, W.-C.; Liao, W.-T.; et al. The Role of Macrophages in Connective Tissue Disease-Associated Interstitial Lung Disease: Focusing on Molecular Mechanisms and Potential Treatment Strategies. Int. J. Mol. Sci. 2023, 24, 11995. https://doi.org/10.3390/ijms241511995

AMA Style

Tseng C-C, Sung Y-W, Chen K-Y, Wang P-Y, Yen C-Y, Sung W-Y, Wu C-C, Ou T-T, Tsai W-C, Liao W-T, et al. The Role of Macrophages in Connective Tissue Disease-Associated Interstitial Lung Disease: Focusing on Molecular Mechanisms and Potential Treatment Strategies. International Journal of Molecular Sciences. 2023; 24(15):11995. https://doi.org/10.3390/ijms241511995

Chicago/Turabian Style

Tseng, Chia-Chun, Ya-Wen Sung, Kuan-Yu Chen, Pin-Yi Wang, Chang-Yi Yen, Wan-Yu Sung, Cheng-Chin Wu, Tsan-Teng Ou, Wen-Chan Tsai, Wei-Ting Liao, and et al. 2023. "The Role of Macrophages in Connective Tissue Disease-Associated Interstitial Lung Disease: Focusing on Molecular Mechanisms and Potential Treatment Strategies" International Journal of Molecular Sciences 24, no. 15: 11995. https://doi.org/10.3390/ijms241511995

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop