Next Article in Journal
Special Issue “Migraine and Headache in Children and Adolescents”
Next Article in Special Issue
Circulating Neurovascular Guidance Molecules and Their Relationship with Peripheral Microvascular Impairment in Systemic Sclerosis
Previous Article in Journal
Biogas Production Potential of Thermophilic Anaerobic Biodegradation of Organic Waste by a Microbial Consortium Identified with Metagenomics
Previous Article in Special Issue
Genetic Association between TNFA Polymorphisms (rs1799964 and rs361525) and Susceptibility to Cancer in Systemic Sclerosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Animal Models of Systemic Sclerosis: Using Nailfold Capillaroscopy as a Potential Tool to Evaluate Microcirculation and Microangiopathy: A Narrative Review

by
Angélica Mandujano
1,* and
Melissa Golubov
2
1
Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City 04960, Mexico
2
Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
*
Author to whom correspondence should be addressed.
Life 2022, 12(5), 703; https://doi.org/10.3390/life12050703
Submission received: 7 April 2022 / Revised: 5 May 2022 / Accepted: 6 May 2022 / Published: 8 May 2022

Abstract

:
Systemic sclerosis (SSc) is an autoimmune disease with three pathogenic hallmarks, i.e., inflammation, vasculopathy, and fibrosis. A wide plethora of animal models have been developed to address the complex pathophysiology and for the development of possible anti-fibrotic treatments. However, no current model comprises all three pathological mechanisms of the disease. To highlight the lack of a complete model, a review of some of the most widely used animal models for SSc was performed. In addition, to date, no model has accomplished the recreation of primary or secondary Raynaud’s phenomenon, a key feature in SSc. In humans, nailfold capillaroscopy (NFC) has been used to evaluate secondary Raynaud’s phenomenon and microvasculature changes in SSc. Being a non-invasive technique, it is widely used both in clinical studies and as a tool for clinical evaluation. Because of this, its potential use in animal models has been neglected. We evaluated NFC in guinea pigs to investigate the possibility of applying this technique to study microcirculation in the nailfold of animal models and in the future, development of an animal model for Raynaud’s phenomenon. The applications are not only to elucidate the pathophysiological mechanisms of vasculopathy but can also be used in the development of novel treatment options.

1. Introduction

Systemic sclerosis (SSc) is a chronic autoimmune disease with a wide range of clinical manifestations. The classic pathogenic triad includes the activation of innate and adaptive immune responses resulting in inflammation, vasculopathy, and fibrosis [1,2]. From a pathophysiological point of view, it is not completely clear whether tissue damage and cell activation are initiated as a result of altered fibroblast activation or due to endothelial cell damage, as both have been implicated.
Vascular features are of the utmost importance, not only because they are almost always present, but also because they are clues to suspect diagnosis in the early course of the disease, long before the appearance of non-vascular manifestations [3,4]. Although vascular damage and endothelial cell apoptosis present quite early [2,5] and there is a close relationship between vasculopathy and fibrosis, the critical events that lead to persistent and uncontrolled fibroblast activation and deposition of extracellular matrix proteins have not been fully elucidated. In this sense, it is necessary to elucidate the relationship between alteration of endothelial cells, fibroblasts, and immune activation in order to disrupt the chain of damage that leads to development of the disease. Despite this, it is clear that vascular manifestations play a central role in the disease. However, there is still little knowledge about the pathophysiology and crucial factors that induce endothelial damage and about the central targets that could induce vascular damage, which could be later used to improve current therapeutic strategies.
Within the vascular manifestations, Raynaud’s phenomenon (RP) is present in ~95% of patients [3,4]. It is a clinical feature that may guide diagnosis, especially if it is accompanied with nailfold capillaroscopy abnormalities. Other clinical features associated with persistent digital ischemia include digital pitting scars, digital ulcers (in around 50% of SSc patients at some point of the disease), and gangrene, all of which are an import cause of morbidity in these patients [3,4]. Furthermore, using videomicroscopy, sublingual abnormalities have been associated with vasculopathy. In addition, some capillary abnormalities correlate with nailfold capillary findings [6]. Unlike other clinical manifestations such as pulmonary arterial hypertension (PAH), interstitial lung disease or scleroderma renal crisis [7], RP and nailfold capillaroscopy abnormalities have not been correlated with specific autoantibodies.
SSc has a very complex pathophysiology in which the interaction of several environmental [8,9,10,11], hereditary [12,13,14,15], and epigenetic modifications [11,16,17,18,19,20] have been involved. Although there are many mediators that have been shown to be involved in the disease, it has not been sufficient for the development of effective disease-modifying therapies, to the point that it remains one of the most fatal rheumatologic diseases [21]. This is due largely to the lack of a single animal model that represents the wide range of manifestations of human SSc. Current animal models recapitulate one or more aspects of SSc [22], but never all, and additionally lack the variety of vascular features that occur in SSc beyond PAH. In fact, there is no appropriate animal model for RP [23], which has limited the study of primary and secondary RP, a very common manifestation, in the context of SSc and other autoimmune diseases. The aim of this narrative review is to highlight the lack of an animal model for RP that resembles SSc vascular manifestations and to propose nailfold capillaroscopy (NFC) as a feasible tool for the study of microcirculation in animal models of SSc as an additional method for the evaluation of vasculopathy. Application of NFC to animal models could potentially contribute to the development of an RP model.

2. Methods

We performed a literature search in PubMed and MEDLINE complete/EBSCO databases. Articles chosen were limited to those written in English and there was no date limit. We used key words such as systemic sclerosis, scleroderma, animal models, fibrosis, vasculopathy RP, and nailfold capillaroscopy. Review articles about animal models of systemic sclerosis, original articles on animal models development, and clinical studies related to targets described in animal models were included.

3. Results

3.1. Animal Models Induced by Administration of Exogenous Agents

There are animal models that use fibrosis-inducing agents such as subcutaneous bleomycin for skin fibrosis [23] or intratracheal administration for pulmonary fibrosis [24,25]. The bleomycin-induced model is one of the most widely used models to study both skin fibrosis [26,27,28] and pulmonary fibrosis [29,30,31]. One of the most striking features in this model is that fibrosis resolves over time with discontinuation of treatment. Although it is an appropriate model for skin fibrosis (and lung fibrosis), studies with this model have not reported the characteristic vasculopathy of SSc. Only Yamamoto et al. found vascular wall thickening in the deep dermis [32]. A different model described by Servettaz and collaborators (2009) was induced by generation of reactive oxygen species (ROS) with prooxidative agents (hydroxyl radicals, hypochlorous acid, peroxynitrites, and superoxide anions). Interestingly, they found that either hypochlorite or hydroxyl radicals can induce systemic disease, cutaneous and lung fibrosis, and anti-topoisomerase I autoantibodies; while those treated with peroxynitrites showed skin fibrosis and anti-centromere protein B antibodies (CENP-B). Another remarkable finding was a reduction in the lumen diameter and intimal fibrosis with intima-media thickening in small renal arteries in mice treated with either hydroxyl radicals or hypochlorous acid [33]. Skin and lung vasculopathy, as well as lung and skin fibrosis, has been ratified by treating this model with hypochlorous acid [34].
Another model is induced by multiple intraperitoneal injections of vinyl chloride, where activation of microchimeric cells leads to dermal and organ fibrosis [35,36,37].
Exogenous subcutaneous injections of transforming growth factor-β TGF-β, connective tissue growth factor (CTGF), and basic fibroblast growth factor (FGF) induce fibrosis in murine models. TGF-β administration induces only transient fibrosis; however, CTGF and FGF lead to persistent fibrosis. It is noteworthy that multiple injections and the use of a combination of the above factors are required to develop lasting fibrosis [32,36]. These and other models are described in greater depth in Table 1.

3.2. Natural or Induced Mutant Models Associated with SSc-like Disease

Tsk1/+ is a spontaneous autosomal dominant mutation of the B10.-D2(58N)/Sn mice strain, affecting fibrillin function [38,39]. It is a tandem duplication within the fibrillin-1 gene [39]. Heterozygous Tsk1/+ mice develop SSc-like features, with important differences regarding involvement of the lung and skin [32,38,39,40]. Tsk2 is generated by administering ethylnitrosourea, which causes an autosomal dominant mutation in chromosome 1 in the offspring of a 101/h strain mice [32,39]. Long et al. reported that Tsk2/+ mice carry a gain-of-function missense point mutation in the procollagen III amino terminal propeptide segment of Col3α1 [41]. Heterozygous mice showed tight skin as seen in TsK1, but with no lung involvement [42].
Of the animal models, the UCD 200 and 206 chicken lines are among the most relevant because they manifest all the hallmarks that most closely resemble human SSc. In the UCD 200 model, endothelial cell apoptosis was identified as an early pathogenic event [5]. A more detailed description of these models is found in Table 1.

3.3. Genetically Engineered Mice Models

Genetically modified models are a powerful methodology to investigate the pathogenesis and therapeutic targets of human diseases. For SSc, there is a mice strain overexpressing the fibroblast kinase-deficient type II TGF-β receptor (TβRΙΙΔk-fib mice). This represents a loss-of-function mutation, which paradoxically develops progressive skin fibrosis especially in the lower back, lung, and gut in a proportion of cases [22,32,39,43]. This same strain showed medial thickening and mild PAH; however, following administration of a VEGF inhibitor, it developed more severe PAH [44]. A type I TGF-β receptor transgenic model also showed upregulation of the TGF-β pathway by fibroblast upregulation of the receptor [38,39].
A fibrillin-1 (Fbn1)-target knock-in mutant mice displayed several features of a systemic sclerosis-like disease, including skin fibrosis, immune cell activation, and autoimmunity with antinuclear and anti-topoisomerase I antibodies, but did not develop microvascular changes [22].
Transgenic mice expressing Fra-2 (Fos related antigen 2) show apoptosis of dermal endothelial cells, rarefication of skin capillaries, as well as skin and lung fibrosis, the latter preceded by obliteration of pulmonary arteries and perivascular inflammation [22,45]. Fli-1 (Friend leukemia integration 1) and kruppel-like factor 5 deficient mice, both with a heterozygous deficiency, develop skin fibrosis, obliterative vasculopathy, and autoantibodies [20]. Mice with urokinase-type plasminogen receptor (uPAR) deletion develop skin and lung fibrosis, as well as microvascular endothelial cell apoptosis with reduced dermis and myocardium capillary density [46,47]. Sirt-3 knockout is another model of PAH and systemic fibrosis [22].
A transgenic endothelin-1 mouse model was generated by transferring the natural promoter into germinal mice; this transgene was expressed mainly in the brain, lung, and kidney. Animals showed interstitial fibrosis of the kidneys and glomerulosclerosis but surprisingly, despite its powerful vasoconstrictor effect, systemic hypertension was absent. This renal fibrosis caused a decrease in the glomerular filtration rate that led to fatal kidney disease. However, instead of developing hypertension, the vascular transgene expression was related to an increase in the media/lumen ratio of intrarenal arteries [48]. In addition, in a subsequent study, chronic expression of endothelin-1 was related to progressive pulmonary fibrosis associated with inflammatory cell infiltration, but interestingly, did not result in significant pulmonary hypertension [49]. In a similar gain-of-function approach, the effects of increased PDGFRα activation limited to cells that normally express the receptor was investigated. Increased PDGFRα signaling led to increased extracellular matrix deposition and hyperplasia of connective tissue with a phenotype similar to SSc. Skin tightening was related to excessive connective tissue deposition and fibrosis of muscle, heart, kidney, and intestine with infiltrates of activated fibroblasts [50]. In addition, in an SSc animal model induced by hypochlorous acid, the phosphorylation of PDGFRβ, which reflects receptor activation, was higher in fibrotic skin [51].
In order to address the possible role of caveolin-1 in caveolae function and signal transduction, Drab and coworkers developed a caveolin-1 knockout mouse (Cav-1). This knockout mouse resulted in the absence of caveolae in endothelial and epithelial cells of lungs and all tissues evaluated including the heart, kidney, and adipose tissue. Interestingly, it was found that this model developed marked pulmonary fibrosis [52]. Increased accumulation of dermal collagen fibers extending to the deeper dermis and around skin appendages was later reported [53]. Studies on pulmonary fibroblasts have shown that TGF-β negatively regulates the expression of caveolin-1. On the contrary, the use of an adenovirus to overexpress caveolin-1 significantly decreased ECM production induced by TGF-β. Disruption of TGF-β signaling by caveolin-1 has been explained by the suppression of phosphorylation of smad-2 in pulmonary fibroblasts [54] and smad-3 in dermal fibroblasts [55]. In cell cultures, the use of caveolin-1 has shown that by decreasing the phosphorylation of Smad-2, it disrupts its interaction with Smad-4 and prevents nuclear translocation of Smad-2. Inhibition of Smad-2 is due to the interaction of the caveolin-1 scaffolding domain with TβRI, which leads to inhibition of the receptor’s kinase activity, thus causing suppression of TGF-β signaling [56].
The MRL/lpr strain is used as a model of systemic autoimmunity, but an MRL/lpr deficient for IFN-γ mice has also been proposed as as a model of SSc. This model lacks the IFN-γ receptor (MRL/lprγR−/−) and with the absence of the IFN-γ antifibrotic response, results in dermal and organ fibrosis and vascular damage [57].
Another interesting model of severe fibrosis in the rat lung is due to the activation of the TGF-β pathway by the Ad-TGFβ1223/225 adenovirus construct; however this is only a pulmonary fibrosis model [58].

3.4. Animal Models with Manipulation of the Immune System

Chronic sclerodermatous graft-versus-host disease (GVHD) resembles fibrotic changes of SSc. There are a couple of models that use this approach. Hamilton and coworkers produced an animal model for both acute and chronic forms of GVHD. For this purpose, they administered a lethal dose of γ radiation in C57BL/6J mice (recipients), and later these mice received bone marrow and spleen cells from LP/J (donors) mice. Those with chronic GVHD develop fibrotic changes in the skin, primaily in the ears, which includes fibrotic bands that restricted extremity mobility. Some of them presented diffuse skin thickening that led to loss of all of their fur. Histologically, the latter was characterized by dermal fibrosis [59]. Another model was described by Claman et al. in 1985 [60]. In this model, spleen and bone marrow cells from B10.D2 mice were transplanted into sub-lethally γ-irradiated BALB/c mice that differed in minor histocompatibility antigens [61]. Skin and lung fibrosis was observed in transplanted animals compared to animal controls. The last model was developed by Ruzek and coworkers. It was induced by spleen cell transplantation from B10.D2 mice to BALB/c recombination-activating gene 2 (RAG-2)-deficient targeted mice, therefore lacking mature T and B cells. They found dermal thickening primarily in extremities and to a lesser degree in the back and abdominal skin. In this model, there was a significant progressive decrease in the lumen diameter of the skin and kidney blood vessels [37,62].
Following a different approach, the administration of autoantibodies has been used to generate animal models. An example is the one induced by administration of human topoisomererase-1 autoantibodies with Freund´s adjuvant. In the same way, the transfer of anti-endothelin receptor type (ETAR) and anti-angiotensin receptor type-1 (AT1R) autoantibodies was associated with the development of obliterative vasculopathy [63].

3.5. Other Genetically Engineered Animal Models That Are Not Fully Characterized

There are other animal models of fibrosis including conditional peroxisome proliferator-activated receptor-γ (PPARγ)-deficient mice, relaxin knockout mouse, phosphatase and tensin homolog (PTEN) knockout mice, and those with overexpression of Wnt10b [38,63], which develop skin fibrosis. These models point to a relationship between these targets and fibrotic mechanisms. Other animal models have explored loss-of-function of different targets to evaluate their effect on fibrosis models induced by fibrotic agents (i.e., bleomycin). These include early growth response protein-1 (Erg-1)-deficient mice, macrophage chemoattractant protein-1 (MCP-1)-deficient mice, and microsomal prostaglandin E2 sythase-1 (mPGES-1)-deficient mice, in which a decrease in fibrosis has been observed [38,64]. All of these models have provided evidence on the mechanisms of fibrosis. However, it is not clear whether they present the three SSc hallmarks, namely, inflammation and autoimmunity, vasculopathy, and fibrosis, all of which participate in the complex pathophysiology of SSc. A description of the main animal models used for SSc can be found in Table 1.
Table 1. Representative animal models of systemic sclerosis. The most relevant characteristics of each model are highlighted.
Table 1. Representative animal models of systemic sclerosis. The most relevant characteristics of each model are highlighted.
ModelTargetFunctionMechanismFibrosisVasculopathyInflammationAutoimmunityClinical EvidenceReferences
Bleomycin-mediated (various mice strains with different susceptibility, C3H/He and B10.A being the most susceptibleNon-specific targetNot applicableTGF-β, collagen, and ECM synthesis upregulation
EC and FB activation
IL-4, IL-13, and CCL2 upregulation
Oxidative stress and activation of NLRP3 inflammasome resulting in collagen synthesis
STAT4 has also been involved
Dermal sclerosis
Lung fibrosis
Vascular wall thickening (in deep dermis) has been reportedPresentTopo I, anti-U1 RNP, anti-histone autoantibodies
Autoantibody that cross-reacts with gastric mucosa
Case reports of SSc-like syndrome in patients with antitumoral bleomycin [22,32,37,38,39,63,64,65,66]
ROS-induced:
Hydroxyl radicals or hypochlorous acid induced (Several strains)
Peroxynitrites induced
Involvement of oxidized topo I and oxidative stress Hydrogen peroxide production by endothelium, monocytes and fibroblastsInvolvement of AOPP, increased synthesis of collagen driven by ROS
Activation of ADAM17/NOTCH, PDGFR, and VEGFRs is involved
Skin fibrosis
Lung fibrosis
Renal involvement
Renal vasculopathy Present Topo I High serum levels of AOPP in dcSSc patients and lung fibrosis vs. controls [33,34,38,40,63,64,67]
TGF-β and CTGF-induced TGF-β and CTGF signalingTGF-β, collagen type I, and CCL2 upregulation TGF-β and CTGF play a role in inducing granulation tissue and fibrosis and increasing synthesis and remodelation of ECM proteinsSkin fibrosis Not reportedPresentNot reportedSee TβRΙΙΔk-fib[32,43]
Angiotensin-II-inducedAngiotensin II receptor signaling activationUpregulation of CTGF, TGF-β, and endothelial-to-mesenchymal transition
MMP-12 has been related to fibrosis and vasculopathy
Administered angiotensin II induces vascular constriction, dermal fibrosis, and inflammation through activation of the TGF-β pathway. Increases fibrocytes and myofibroblasts in skinSkin fibrosisPresent, including cardiovascular remodelingPresentNot reported High levels of Angiotensin II in serum of dcSSc patients.
A subgroup of cases presented elevated angiotensin II signaling
[38,40,63,68]
Tsk1/+Fibrillin-1 geneFibrillin is a structural protein of ECM. It has a direct interaction with latent TGF-β binding protein. It is thought that the failure of sequestration of the large latent complex drives an increase in the activation of TGFβ Hyper-response to IL-4, MCP-3, and TGF-β with increasing synthesis of type I collagen
B cell activation resulted from activation of CD19 signaling
Hypodermal fibrosis
Emphysema-like disease with little fibrosis
Abnormal vascular tone and cardiomyopathyAbsent Topo I and fibrillin-1 autoantibodies Cases of human stiff skin syndrome associated with Fbn1 mutation
Association of Fibrillin-1 SNP haplotypes with SSc
in Choctaw Indians and Japanese populations
[32,37,38,40,69,70,71,72]
Tsk2/+Col3α1Collagen III synthesisGain-of-function mutation that results in increased synthesis of type I and III collagen Dermal fibrosisAbsentPresent Antinuclear, topo I/Scl70, anti-centromere, and anti-dsDNA antibodies [22,32,38,41,73]
UCD-200 and 206 chickens TGFBR1, IGFBP3, EXOC2/IRF4, CCR8, and SOCS1Associated genes play a role in either SSc or autoimmunitySpontaneously develops vascular damage, fibrosis and inflammation. Skin fibrosis
Lung fibrosis
Internal organ fibrosis
Vasculopathy
Intimal proliferation with narrowing of arterioles and capillaries and cell infiltration
Present Polyarthritis
Antinuclear antibodies, AECA, aCL antibodies, and rheumatoid factor
Not reported[5,32,37,38,63]
Kinase-deficient TGFβRII model:
TβRΙΙΔk-fib
Type II TGF-β receptorBinding of the ligand to TβRII drives the phosphorylation of serine residues within the type I receptor (TβRI) to initiate TGF-downstream signaling Paradoxical activation of TGF-β signaling
It is postulated that accessory proteins and a nonsignaling type I receptor can modulate TβRII function
Skin fibrosis
Pulmonary fibrosis
Fibrotic cardiomyopathy
Generalized vascular remodeling Not present Not present Several lines of research support the ling of link between TGF-β and fibrotic disease and systemic sclerosis[22,32,38,39,40,43,74,75,76]
TBRI(CA); Cre-ER miceExpression of a constitutionally active TGF-β1 type I receptorTGF-β signaling activationIncreased collagen synthesisProgressive and generalized dermal fibrosis
Myocardial fibrosis
Altered aortic dynamics
Thickening of blood vessel walls in small arteries of the lung and kidney
Increased levels of von Willebrand factor
Altered endothelin-1 signaling
AbsentNot reportedSee TβRΙΙΔk-fib[38,39,63,77]
Fbn-1 mutations Knock-in mouse:
SSS- associated change W1572C (WC/WC, WC/+)
D1545E
(DE/+) (Homozygous was lethal)
FBN1 geneSee fibrillarin function in Tsk1 modelExcessive elastin, collagen, and microfibrillar aggregates in dermal
TGF-β upregulation
Skin fibrosisNot reportedPresentTopo ISee Tsk1 clinical evidence[22,78]
Fra-2 Tg miceFra-2 overexpressionA member of the Fos family of transcription factors, a downstream mediator of TGF-β and PDGF. Induced by cellular stress and controls apoptosis, inflammation, healing and proliferation.Upregulation of type I collagen in dermal fibroblasts, EC apoptosis and epithelial-to-mesenchymal transition, PDGF signaling activationSkin fibrosis
Pulmonary fibrosis
Vasculopathy
Decrease in the number of capillaries
EC apoptosis, pulmonary arterial occlusion
PresentNot presentFra-2 expression is elevated in SSc dermal fibroblasts, EC, and pneumocyte epithelial cells [38,40,63]
Fli1ΔCTA/ΔCTAFli1Fli1 has roles in hematopoiesis and vasculogenesis, serves as a transcriptional repressor through its CTA domain, inhibits collagen genes, and is a negative regulator of ECMUpregulation of dermal fibrillar collagen and Fli1 protein levelsSkin fibrosisPresent. Includes increases vascular permeability, similar to the one observed in SScNot reportedNot reportedFli1 proteins are reduced in dermal fibroblasts, EC, and SMC of SSc patients [18,38,40]
Fli1 ECKO
Conditional deletion of Fli1 in EC
Fli1Fli1 is a transcription factor expressed in EC and hematopoietic cells. It participates in the regulation of development and differentiation of EC and vasculature Loss of endothelial integrity
Fli-1 depletion has an effect on expression of genes that regulate vascular integrity
AbsentPresent, irregular diameter and disorganization of the dermal vascular networkAbsentNot reportedSee Fli1 mutated mice[40,63]
Fli1-KLF5-KOFli1+KLF5Fli1 is a potent repressor of type I collagen gene
KLF5 has a potentially synergic effect with Fli1
Activation of both canonical and non-canonical TGF-β signaling
Upregulation of ECM genes and CTGF
Skin fibrosis
Lung fibrosis
Obliterative vasculopathy with vascular stenosis, loss and bushy skin capillaries Progressive obliteration of pulmonary arterioles PresentAntinuclear antibodiesDownregulation of KLF5 in skin samples and SSc fibroblasts
Fli1 repression in SSc fibroblast and EC
[20,22]
Sirt3-deficient miceSirt3Class III histone deacetylases that play a key role in maintenance of mitochondrial integrityDeficiency induces spontaneous multiorgan fibrosis as the mice age, accompanied by oxidative stress and mitochondrial damageMultiorgan fibrosis including in the lungs and heartPAHNot reportedNot reportedMarked suppression of Sirt1 has been found in skin biopsies and explanted fibroblasts from SSc patients in two reports[22,79]
Endothelin-1 TgET-1 overexpression Potent vasoconstrictor, regulation of blood flow in microvascular beds, sodium and water reabsorption
Stimulates proliferation and collagen synthesis in normal fibroblast
Fibrogenic effect including: increased pulmonary matrix synthesis with chronic inflammation, excessive collagen production of SSc fibroblasts enhanced by TGF-βRenal fibrosis
Lung fibrosis (chronic overexpression)
Increased media/lumen ratio of intrarenal arteriesPresentNot reportedHigher serum levels and overproduction of endothelin-1 in SSc patients and in primary RP. Levels were associated with skin score and disease duration
Effectiveness of endothelin-1 blockade in pulmonary hypertension and RP treatment
[38,48,49,80,81,82,83]
PDGFR-α hyperactivationPDGFRα conditional overexpressionPDGFs play an important role in the development and maintenance of connective tissue, they are potent mitogens and chemoattractants of mesenchymal cells, they exert their biological effects through binding of PDGFRα/βActivation of cellular programs that generate connective tissue
Connective tissue hyperplasia and increased extracellular matrix deposition
Skin fibrosis
Internal organ fibrosis
Not reportedNot reportedNot reportedIncreased levels of PDGF and PDGFR in skin and lung samples of SSc patients
Presence of stimulatory autoantibodies directed toward PDGFR
More common and intense PDGFRβ inmunoreactivity in small vessels of SSc-associated PAH compared to idiopathic PAH
[51,84,85,86,87]
Cav-1−/− C57B1/6KO miceCaveolin-1It is an integral membrane protein and a structural component of caveolae. It modulates the activity of caveolae and disrupts TGF-β signalingUpregulation of TGF-β and ECM proteins
ROS, HIF1α y NFκB signaling pathways are activated
Thickening of alveolar septa with uncontrolled hyperproliferation of angioblastic cells and fibrosis
Skin fibrosis
Vascular system disfunction
Increased accumulation of collagen fibers around blood vessels of the deep dermis
PresentNot reportedReduced levels of Cav-1 in skin and dermal fibroblasts from SSc patients and in lung samples from ILD-SSc patients
Reduction of Cav-1 in lung fibroblasts and lung tissue in bleomycin-
induced IPF
[52,53,54,56]
Hematopoietic cell transplantation:
Scl-GVHD
LP/J/ C57BL/6J
B10.D2/BALB/c mice
RAG-2 KO mice
Non-specific targetTGF-β upregulation, increased type I collage, chemokines, and other growth factors Fibroblast, T cells, and other leucocytes involved Skin fibrosis
Lung fibrosis (not all models)
Involvement of internal organs (kidney, small intestine)
(not all models)
Vasculopathy
Renal crisis
PresentAutoantibodiesScl-GVHD after hematopoietic cell transplantation[32,38,39,61,62,63,72,88]
Topo I and CFA-induced SSc DNA topoisomeraseRelaxation of supercoiled DNAUpregulation of TGF-β, IL-17 and IL-6Skin fibrosis
Pulmonary fibrosis
Not reportedPresentTopo I Several studies reported the presence of autoantibodies including disease-specific autoantibodies[38,40,63,89]
AECA: anti-endothelial cell autoantibodies, aCL: anticardiolipin antibodies, AOPP: advanced oxidation protein products, Cav-1: caveolin-1, CFA: Complete Freund´s Adjuvant, dcSSC: diffuse cutaneous systemic sclerosis, EC: endothelial cell apoptosis, ECM: extracellular matrix, ET-1: endothelin-1, DE/+: heterozygous D1545E mice, KO: knockout, HIF1α: hypoxia inducible factor 1α, PAH, ILD: interstitial lung disease, IPF: idiopathic pulmonary fibrosis, pulmonary arterial hypertension, MCP-3: monocyte-chemotactic protein-3, NFκB: nuclear factor κB, PDGF: platelet derived growth factor, PDGFR: platelet derived growth factor receptor, ROS: reactive oxygen species, RP: Raynaud´s phenomenon, Tg-mice: transgenic mice, TGF-β: transforming growth factor β, Topo I: anti-DNA topoisomerase I, SSc: systemic sclerosis, Sirt3: sirtuin 3, Scl-GVHD: sclerodermatous graft-versus-host-disease, SMC: smooth, muscle cell, SNP: single nucleotide polymorphism, VEGFR: vascular endothelial growth factors receptor, WC: W1572C mice, WC/WC: homozygous mice, WC/+: heterozygous mice.
Most models, as other authors have pointed out, have used rodents (mainly mice), given the homology to the human genome and other technical and genetically engineered advantages. However, to date, no animal model has been able to recreate proliferative obliterative vasculopathy of nailfold capillaries as observed in SSc patients, or at least it does not seem to have been previously evaluated.

3.6. Nailfold Capillaroscopy in the Guinea Pig

In order to design and propose an animal model that resembles RP (primary or secondary) with abnormal nailfold capillaroscopic findings such as the ones observed in SSc and other autoimmune diseases, it must first be elucidated whether animal nail fold capillaroscopy can be observed. For this purpose, the nailfold (n = 1) of the guinea pig (Cavia pocellus) was investigated by nailfold capillaroscopy (NFC). NFC was performed with a stereoscope VE-S7 with a magnification of 55× (Velab Co., Pharr, TX, USA) and a capillaroscope model JH-1004C with a total magnification of 380× to obtain images recorded with the software MicroCirculationXP (Jiansu Jiahua Electronic Instrument Co., Xuzhou, China). NFC was performed at room temperature on the front and hind limbs of the specimen. The excess hairs near the nailfold were trimmed, and immersion oil was applied to the nailfold. First, a panoramic view of the nailfold was carried out with the stereoscope at a magnification of 55×. Subsequently, the capillaries were observed at a total magnification of 380× with the capillaroscope. Figure 1 shows representative images of the capillaries of the nailfold of the right hind limb of the specimen at 380× (Figure 1C,D). The nailfold capillaries could be observed, and showed a very similar anatomical pattern to the human nailfold at 380× (Figure 1E,F). The number of capillaries and capillary flow could also be observed, and the diameters of the capillaries were measurable. No pharmacological or invasive procedures were performed prior to NFC in the guinea pig. The specimen was kindly lent by Dr. Ernesto López from a private animal care facility.
In normal NFC, the capillaries are usually homogeneously arranged and present a hairpin or “U” shape. They are distributed with their major axis parallel to the skin surface in a pattern described as a “comb-like structure”. However, due to the variability within a population, tortuous and crossed capillaries can be observed with high prevalence, and to a lesser degree, other shapes such as ectatic, bushy or bizarre loops can be observed. The structure and shape are best seen in the most distal row of capillaries. The normal density of capillaries is between 9 and 14 per mm, and the normal diameter of the afferent limbs ranges between 6 and 19 μm and 8 and 20 μm for efferent limbs. The average length of nailfold capillaries can be between 200 and 500 μm, but some of them may seem shorter. A large network of vessels away from the distal row of hairpin capillaries corresponds to the subpapillary venular plexus (SPVP) and is visible in a proportion of subjects more frequently in the 5th finger [90].
NFC parameters in the guinea pig were similar to the values for a healthy human NFC, with 13 capillaries per mm, an afferent limb diameter of 7 μm, efferent limb diameter of 9 μm, and apical limb diameter of 8 μm. The average length was 166 μm, and SPVP could also be observed. This pattern, in general, seems very similar to a human NFC. It is of note in the guinea pig NFC that just one line of hairpin-shaped capillaries seems to be observed distal to SPVP (Figure 1).
Clinically, the observation of capillaries by NFC has gained great relevance in several diseases such as Diabetes Mellitus and hypertension [91,92,93,94,95], but especially in autoimmune rheumatic diseases including diagnosis and as a potential prognostic tool for SSc [95,96,97]. Being a non-invasive, simple and relatively low-cost technique, its main use in clinical practice and in research has been with human subjects. However, its potential use in animal models for the research of the pathophysiology of microangiopathic disease has been missed.
Once we observed that NFC is indeed possible in the guinea pig, NFC in a larger number of wild type guinea pigs and murine models is planned to describe the anatomy of nailfold microstructure under “normal” conditions, as well as in known animal models of SSc for both fibrosis and vascular damage. The need for further validation of anatomical characteristics of this model with a larger sample should be noted, as well as the need to evaluate its application to previously described mice models of SSc.
Once the anatomical characteristics of the models have been described, the goal is to develop an RP model by targeting known mechanisms of RP pathophysiology of primary and secondary RP, such as endothelin-1 and vascular endothelial growth factor (VEGF), among others. The development of these models has, as can be inferred, application in other pathways involved in the disease. Additionally, they may allow development of novel therapies for the disease, especially for vascular features. Although at present there is an vast therapeutic arsenal for vascular manifestations, it does not cover all needs, can be ineffective, and has a high rate of therapeutic failure [3]. In previous work, the possible relationship between capillary abnormalities and leukotrienes was shown. New studies are required to investigate and corroborate this relationship. Animal models that use exogenous agents and gain-or-loss-of-function transgenesis targeting leukotriene pathways and its interaction with other mediators are critical to study the pathophysiology of microangiopathy in greater depth. There is great interest by several study groups, including the authors, in the role of leukotrienes in the pathophysiology of SSc because of its biological activity, which corresponds to the pathogenic triad of SSc [98,99,100,101]. Assessment of microangiopathy in NFC of SSc animal models or RP models could enable research of leukotriene pathways, their interactions with other mediators (i.e., TGF-β, Hypoxia inducible factor 1α, among others), and the usefulness of antileukotriene therapy and other pathogenic mediators or potential antifibrotic therapies in SSc.

4. Conclusions

Currently, there are no animal models that simultaneously present all three main pathological mechanisms of SSc or models that recreate RP. The potential use of NFC to evaluate microcirculation in current models has not been explored. We showed that NFC can be observed in the nailfold of the guinea pig, but could be also applicable to murine models, and is morphologically very similar to human NFC. This encourages the development of RP animal models that could be used for the study of microvascular diseases, including SSc, other autoimmune, and chronic diseases.

Author Contributions

All authors participated in the development of this article. A.M. Conceptualization, methodology, data analysis, writing—original draft preparation, review and editing. M.G. Conceptualization, data analysis and writing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Informed Consent Statement

Written informed consent has been obtained from the healthy volunteer to publish this paper.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank Ernesto López for lending us a guinea pig specimen.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Varga, J.; Abraham, D. Systemic sclerosis: A prototypic multisystem fibrotic disorder. J. Clin. Investig. 2007, 117, 557–567. [Google Scholar] [CrossRef] [PubMed]
  2. Gabrielli, A.; Avvedimento, E.V.; Krieg, T. Scleroderma. N. Engl. J. Med. 2009, 360, 1989–2003. [Google Scholar] [CrossRef]
  3. Hughes, M.; Allanore, Y.; Chung, L.; Pauling, J.D.; Denton, C.P.; Matucci-Cerinic, M. Raynaud phenomenon and digital ulcers in systemic sclerosis. Nat. Rev. Rheumatol. 2020, 16, 208–221. [Google Scholar] [CrossRef] [PubMed]
  4. Cutolo, M.; Soldano, S.; Smith, V. Pathophysiology of systemic sclerosis: Current understanding and new insights. Expert Rev. Clin. Immunol. 2019, 15, 753–764. [Google Scholar] [CrossRef] [PubMed]
  5. Wick, G.; Andersson, L.; Hala, K.; Gershwin, M.E.; Selmi, C.; Erf, G.F.; Lamont, S.J.; Sgonc, R. Avian models with spontaneous autoimmune diseases. Adv. Immunol. 2006, 92, 71–117. [Google Scholar] [CrossRef] [Green Version]
  6. Radic, M.; Thomas, J.; McMillan, S.; Frech, T. Does sublingual microscopy correlate with nailfold videocapillaroscopy in systemic sclerosis? Clin. Rheumatol. 2021, 40, 2263–2266. [Google Scholar] [CrossRef]
  7. Medsger, T.A. Natural history of systemic sclerosis and the assessment of disease activity, severity, functional status, and psychologic well-being. Rheum. Dis. Clin. N. Am. 2003, 29, 255–273. [Google Scholar] [CrossRef]
  8. Muryoi, T.; Kasturi, K.N.; Kafina, M.J.; Cram, D.S.; Harrison, L.C.; Sasaki, T.; Bona, C.A. Antitopoisomerase I monoclonal autoantibodies from scleroderma patients and tight skin mouse interact with similar epitopes. J. Exp. Med. 1992, 175, 1103–1109. [Google Scholar] [CrossRef]
  9. Patel, S.; Morrisroe, K.; Proudman, S.; Hansen, D.; Sahhar, J.; Sim, M.R.; Ngian, G.S.; Walker, J.; Strickland, G.; Wilson, M.; et al. Occupational silica exposure in an Australian systemic sclerosis cohort. Rheumatology 2020, 59, 3900–3905. [Google Scholar] [CrossRef]
  10. Marie, I. Systemic sclerosis and exposure to heavy metals. Autoimmun. Rev. 2019, 18, 62–72. [Google Scholar] [CrossRef]
  11. Maurer, B.; Stanczyk, J.; Jüngel, A.; Akhmetshina, A.; Trenkmann, M.; Brock, M.; Kowal-Bielecka, O.; Gay, R.E.; Michel, B.A.; Distler, J.H.; et al. MicroRNA-29, a key regulator of collagen expression in systemic sclerosis. Arthritis Rheum. 2010, 62, 1733–1743. [Google Scholar] [CrossRef] [PubMed]
  12. Bossini-Castillo, L.; Villanueva-Martin, G.; Kerick, M.; Acosta-Herrera, M.; López-Isac, E.; Simeón, C.P.; Ortego-Centeno, N.; Assassi, S.; Hunzelmann, N.; Gabrielli, A.; et al. Genomic Risk Score impact on susceptibility to systemic sclerosis. Ann. Rheum. Dis. 2021, 80, 118–127. [Google Scholar] [CrossRef] [PubMed]
  13. Feghali-Bostwick, C.; Medsger, T.A.; Wright, T.M. Analysis of systemic sclerosis in twins reveals low concordance for disease and high concordance for the presence of antinuclear antibodies. Arthritis Rheum. 2003, 48, 1956–1963. [Google Scholar] [CrossRef]
  14. Frech, T.; Khanna, D.; Markewitz, B.; Mineau, G.; Pimentel, R.; Sawitzke, A. Heritability of vasculopathy, autoimmune disease, and fibrosis in systemic sclerosis: A population-based study. Arthritis Rheum. 2010, 62, 2109–2116. [Google Scholar] [CrossRef] [Green Version]
  15. Arnett, F.C.; Gourh, P.; Shete, S.; Ahn, C.W.; Honey, R.E.; Agarwal, S.K.; Tan, F.K.; McNearney, T.; Fischbach, M.; Fritzler, M.J.; et al. Major histocompatibility complex (MHC) class II alleles, haplotypes and epitopes which confer susceptibility or protection in systemic sclerosis: Analyses in 1300 Caucasian, African-American and Hispanic cases and 1000 controls. Ann. Rheum. Dis. 2010, 69, 822–827. [Google Scholar] [CrossRef] [Green Version]
  16. Ghosh, A.K.; Bhattacharyya, S.; Lafyatis, R.; Farina, G.; Yu, J.; Thimmapaya, B.; Wei, J.; Varga, J. p300 is elevated in systemic sclerosis and its expression is positively regulated by TGF-Œ≤: Epigenetic feed-forward amplification of fibrosis. J. Investig. Derm. 2013, 133, 1302–1310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Altorok, N.; Tsou, P.S.; Coit, P.; Khanna, D.; Sawalha, A.H. Genome-wide DNA methylation analysis in dermal fibroblasts from patients with diffuse and limited systemic sclerosis reveals common and subset-specific DNA methylation aberrancies. Ann. Rheum. Dis. 2015, 74, 1612–1620. [Google Scholar] [CrossRef] [PubMed]
  18. Wang, Y.; Fan, P.S.; Kahaleh, B. Association between enhanced type I collagen expression and epigenetic repression of the FLI1 gene in scleroderma fibroblasts. Arthritis Rheum. 2006, 54, 2271–2279. [Google Scholar] [CrossRef]
  19. Wang, Y.Y.; Wang, Q.; Sun, X.H.; Liu, R.Z.; Shu, Y.; Kanekura, T.; Huang, J.H.; Li, Y.P.; Wang, J.C.; Zhao, M.; et al. DNA hypermethylation of the forkhead box protein 3 (FOXP3) promoter in CD4+ T cells of patients with systemic sclerosis. Br. J. Derm. 2014, 171, 39–47. [Google Scholar] [CrossRef]
  20. Noda, S.; Asano, Y.; Nishimura, S.; Taniguchi, T.; Fujiu, K.; Manabe, I.; Nakamura, K.; Yamashita, T.; Saigusa, R.; Akamata, K.; et al. Simultaneous downregulation of KLF5 and Fli1 is a key feature underlying systemic sclerosis. Nat. Commun. 2014, 5, 5797. [Google Scholar] [CrossRef] [Green Version]
  21. 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] [PubMed]
  22. Marangoni, R.G.; Varga, J.; Tourtellotte, W.G. Animal models of scleroderma: Recent progress. Curr. Opin. Rheumatol. 2016, 28, 561–570. [Google Scholar] [CrossRef] [PubMed]
  23. Fardoun, M.M.; Nassif, J.; Issa, K.; Baydoun, E.; Eid, A.H. Raynaud’s Phenomenon: A Brief Review of the Underlying Mechanisms. Front. Pharm. 2016, 7, 438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zhang, K.; Flanders, K.C.; Phan, S.H. Cellular localization of transforming growth factor-beta expression in bleomycin-induced pulmonary fibrosis. Am. J. Pathol. 1995, 147, 352–361. [Google Scholar] [PubMed]
  25. Degryse, A.L.; Tanjore, H.; Xu, X.C.; Polosukhin, V.V.; Jones, B.R.; McMahon, F.B.; Gleaves, L.A.; Blackwell, T.S.; Lawson, W.E. Repetitive intratracheal bleomycin models several features of idiopathic pulmonary fibrosis. Am. J. Physiol. Lung Cell Mol. Physiol. 2010, 299, L442–L452. [Google Scholar] [CrossRef] [PubMed]
  26. Yamamoto, T.; Takagawa, S.; Katayama, I.; Yamazaki, K.; Hamazaki, Y.; Shinkai, H.; Nishioka, K. Animal model of sclerotic skin. I: Local injections of bleomycin induce sclerotic skin mimicking scleroderma. J. Investig. Derm. 1999, 112, 456–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Yoshizaki, A.; Iwata, Y.; Komura, K.; Ogawa, F.; Hara, T.; Muroi, E.; Takenaka, M.; Shimizu, K.; Hasegawa, M.; Fujimoto, M.; et al. CD19 regulates skin and lung fibrosis via Toll-like receptor signaling in a model of bleomycin-induced scleroderma. Am. J. Pathol. 2008, 172, 1650–1663. [Google Scholar] [CrossRef] [Green Version]
  28. Yamamoto, T.; Nishioka, K. Cellular and molecular mechanisms of bleomycin-induced murine scleroderma: Current update and future perspective. Exp. Derm. 2005, 14, 81–95. [Google Scholar] [CrossRef] [Green Version]
  29. Adamson, I.Y.; Bowden, D.H. The pathogenesis of bleomycin-induced pulmonary fibrosis in mice. Am. J. Pathol. 1974, 77, 185–197. [Google Scholar]
  30. Szapiel, S.V.; Elson, N.A.; Fulmer, J.D.; Hunninghake, G.W.; Crystal, R.G. Bleomycin-induced interstitial pulmonary disease in the nude, athymic mouse. Am. Rev. Respir. Dis. 1979, 120, 893–899. [Google Scholar] [CrossRef]
  31. Belperio, J.A.; Dy, M.; Burdick, M.D.; Xue, Y.Y.; Li, K.; Elias, J.A.; Keane, M.P. Interaction of IL-13 and C10 in the pathogenesis of bleomycin-induced pulmonary fibrosis. Am. J. Respir. Cell Mol. Biol. 2002, 27, 419–427. [Google Scholar] [CrossRef] [PubMed]
  32. Yamamoto, T. Animal model of systemic sclerosis. J. Derm. 2010, 37, 26–41. [Google Scholar] [CrossRef] [PubMed]
  33. Servettaz, A.; Goulvestre, C.; Kavian, N.; Nicco, C.; Guilpain, P.; Chéreau, C.; Vuiblet, V.; Guillevin, L.; Mouthon, L.; Weill, B.; et al. Selective oxidation of DNA topoisomerase 1 induces systemic sclerosis in the mouse. J. Immunol. 2009, 182, 5855–5864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Meng, M.; Tan, J.; Chen, W.; Du, Q.; Xie, B.; Wang, N.; Zhu, H.; Wang, K. The Fibrosis and Immunological Features of Hypochlorous Acid Induced Mouse Model of Systemic Sclerosis. Front. Immunol. 2019, 10, 1861. [Google Scholar] [CrossRef] [Green Version]
  35. Christner, P.J.; Artlett, C.M.; Conway, R.F.; Jiménez, S.A. Increased numbers of microchimeric cells of fetal origin are associated with dermal fibrosis in mice following injection of vinyl chloride. Arthritis Rheum. 2000, 43, 2598–2605. [Google Scholar] [CrossRef]
  36. Denton, C.P.; Abraham, D.J. Transgenic analysis of scleroderma: Understanding key pathogenic events in vivo. Autoimmun. Rev. 2004, 3, 285–293. [Google Scholar] [CrossRef]
  37. Rogai, V.; Lories, R.J.; Guiducci, S.; Luyten, F.P.; Matucci Cerinic, M. Animal models in systemic sclerosis. Clin. Exp. Rheumatol. 2008, 26, 941–946. [Google Scholar]
  38. Artlett, C.M. Animal models of systemic sclerosis: Their utility and limitations. Open Access Rheumatol. 2014, 6, 65–81. [Google Scholar] [CrossRef] [Green Version]
  39. Beyer, C.; Schett, G.; Distler, O.; Distler, J.H. Animal models of systemic sclerosis: Prospects and limitations. Arthritis Rheum. 2010, 62, 2831–2844. [Google Scholar] [CrossRef]
  40. Asano, Y.; Sato, S. Animal models of scleroderma: Current state and recent development. Curr. Rheumatol. Rep. 2013, 15, 382. [Google Scholar] [CrossRef]
  41. Long, K.B.; Li, Z.; Burgwin, C.M.; Choe, S.G.; Martyanov, V.; Sassi-Gaha, S.; Earl, J.P.; Eutsey, R.A.; Ahmed, A.; Ehrlich, G.D.; et al. The Tsk2/+ mouse fibrotic phenotype is due to a gain-of-function mutation in the PIIINP segment of the Col3a1 gene. J. Investig. Dermatol. 2015, 135, 718–727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Barisic-Dujmovic, T.; Boban, I.; Clark, S.H. Regulation of collagen gene expression in the Tsk2 mouse. J. Cell Physiol. 2008, 215, 464–471. [Google Scholar] [CrossRef] [PubMed]
  43. Denton, C.P.; Zheng, B.; Evans, L.A.; Shi-wen, X.; Ong, V.H.; Fisher, I.; Lazaridis, K.; Abraham, D.J.; Black, C.M.; de Crombrugghe, B. Fibroblast-specific expression of a kinase-deficient type II transforming growth factor beta (TGFbeta) receptor leads to paradoxical activation of TGFbeta signaling pathways with fibrosis in transgenic mice. J. Biol. Chem. 2003, 278, 25109–25119. [Google Scholar] [CrossRef] [Green Version]
  44. Derrett-Smith, E.C.; Dooley, A.; Gilbane, A.J.; Trinder, S.L.; Khan, K.; Baliga, R.; Holmes, A.M.; Hobbs, A.J.; Abraham, D.; Denton, C.P. Endothelial injury in a transforming growth factor β-dependent mouse model of scleroderma induces pulmonary arterial hypertension. Arthritis Rheum. 2013, 65, 2928–2939. [Google Scholar] [CrossRef] [PubMed]
  45. Maurer, B.; Distler, J.H.; Distler, O. The Fra-2 transgenic mouse model of systemic sclerosis. Vasc. Pharm. 2013, 58, 194–201. [Google Scholar] [CrossRef] [PubMed]
  46. Manetti, M.; Rosa, I.; Milia, A.F.; Guiducci, S.; Carmeliet, P.; Ibba-Manneschi, L.; Matucci-Cerinic, M. Inactivation of urokinase-type plasminogen activator receptor (uPAR) gene induces dermal and pulmonary fibrosis and peripheral microvasculopathy in mice: A new model of experimental scleroderma? Ann. Rheum. Dis. 2014, 73, 1700–1709. [Google Scholar] [CrossRef]
  47. Manetti, M.; Rosa, I.; Fazi, M.; Guiducci, S.; Carmeliet, P.; Ibba-Manneschi, L.; Matucci-Cerinic, M. Systemic sclerosis-like histopathological features in the myocardium of uPAR-deficient mice. Ann. Rheum. Dis. 2016, 75, 474–478. [Google Scholar] [CrossRef] [Green Version]
  48. Hocher, B.; Thöne-Reineke, C.; Rohmeiss, P.; Schmager, F.; Slowinski, T.; Burst, V.; Siegmund, F.; Quertermous, T.; Bauer, C.; Neumayer, H.H.; et al. Endothelin-1 transgenic mice develop glomerulosclerosis, interstitial fibrosis, and renal cysts but not hypertension. J. Clin. Investig. 1997, 99, 1380–1389. [Google Scholar] [CrossRef]
  49. Hocher, B.; Schwarz, A.; Fagan, K.A.; Thöne-Reineke, C.; El-Hag, K.; Kusserow, H.; Elitok, S.; Bauer, C.; Neumayer, H.H.; Rodman, D.M.; et al. Pulmonary fibrosis and chronic lung inflammation in ET-1 transgenic mice. Am. J. Respir. Cell Mol. Biol. 2000, 23, 19–26. [Google Scholar] [CrossRef] [Green Version]
  50. Olson, L.E.; Soriano, P. Increased PDGFRalpha activation disrupts connective tissue development and drives systemic fibrosis. Dev. Cell 2009, 16, 303–313. [Google Scholar] [CrossRef] [Green Version]
  51. Kavian, N.; Servettaz, A.; Marut, W.; Nicco, C.; Chéreau, C.; Weill, B.; Batteux, F. Sunitinib inhibits the phosphorylation of platelet-derived growth factor receptor Œ≤ in the skin of mice with scleroderma-like features and prevents the development of the disease. Arthritis Rheum. 2012, 64, 1990–2000. [Google Scholar] [CrossRef] [PubMed]
  52. Drab, M.; Verkade, P.; Elger, M.; Kasper, M.; Lohn, M.; Lauterbach, B.; Menne, J.; Lindschau, C.; Mende, F.; Luft, F.C.; et al. Loss of caveolae, vascular dysfunction, and pulmonary defects in caveolin-1 gene-disrupted mice. Science 2001, 293, 2449–2452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Castello-Cros, R.; Whitaker-Menezes, D.; Molchansky, A.; Purkins, G.; Soslowsky, L.J.; Beason, D.P.; Sotgia, F.; Iozzo, R.V.; Lisanti, M.P. Scleroderma-like properties of skin from caveolin-1-deficient mice: Implications for new treatment strategies in patients with fibrosis and systemic sclerosis. Cell Cycle 2011, 10, 2140–2150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Wang, X.M.; Zhang, Y.; Kim, H.P.; Zhou, Z.; Feghali-Bostwick, C.A.; Liu, F.; Ifedigbo, E.; Xu, X.; Oury, T.D.; Kaminski, N.; et al. Caveolin-1: A critical regulator of lung fibrosis in idiopathic pulmonary fibrosis. J. Exp. Med. 2006, 203, 2895–2906. [Google Scholar] [CrossRef] [PubMed]
  55. Del Galdo, F.; Sotgia, F.; de Almeida, C.J.; Jasmin, J.F.; Musick, M.; Lisanti, M.P.; Jiménez, S.A. Decreased expression of caveolin 1 in patients with systemic sclerosis: Crucial role in the pathogenesis of tissue fibrosis. Arthritis Rheum. 2008, 58, 2854–2865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Razani, B.; Zhang, X.L.; Bitzer, M.; von Gersdorff, G.; Böttinger, E.P.; Lisanti, M.P. Caveolin-1 regulates transforming growth factor (TGF)-beta/SMAD signaling through an interaction with the TGF-beta type I receptor. J. Biol. Chem. 2001, 276, 6727–6738. [Google Scholar] [CrossRef] [Green Version]
  57. Le Hir, M.; Martin, M.; Haas, C. A syndrome resembling human systemic sclerosis (scleroderma) in MRL/lpr mice lacking interferon-gamma (IFN-gamma) receptor (MRL/lprgammaR−/−). Clin. Exp. Immunol. 1999, 115, 281–287. [Google Scholar] [CrossRef]
  58. Sime, P.J.; Xing, Z.; Graham, F.L.; Csaky, K.G.; Gauldie, J. Adenovector-mediated gene transfer of active transforming growth factor-beta1 induces prolonged severe fibrosis in rat lung. J. Clin. Investig. 1997, 100, 768–776. [Google Scholar] [CrossRef] [Green Version]
  59. Hamilton, B.L.; Parkman, R. Acute and chronic graft-versus-host disease induced by minor histocompatibility antigens in mice. Transplantation 1983, 36, 150–155. [Google Scholar] [CrossRef]
  60. Claman, H.N.; Jaffee, B.D.; Huff, J.C.; Clark, R.A. Chronic graft-versus-host disease as a model for scleroderma. II. Mast cell depletion with deposition of immunoglobulins in the skin and fibrosis. Cell Immunol. 1985, 94, 73–84. [Google Scholar] [CrossRef]
  61. McCormick, L.L.; Zhang, Y.; Tootell, E.; Gilliam, A.C. Anti-TGF-beta treatment prevents skin and lung fibrosis in murine sclerodermatous graft-versus-host disease: A model for human scleroderma. J. Immunol. 1999, 163, 5693–5699. [Google Scholar] [PubMed]
  62. Ruzek, M.C.; Jha, S.; Ledbetter, S.; Richards, S.M.; Garman, R.D. A modified model of graft-versus-host-induced systemic sclerosis (scleroderma) exhibits all major aspects of the human disease. Arthritis Rheum. 2004, 50, 1319–1331. [Google Scholar] [CrossRef] [PubMed]
  63. Morin, F.; Kavian, N.; Batteux, F. Animal models of systemic sclerosis. Curr. Pharm. Des. 2015, 21, 2365–2379. [Google Scholar] [CrossRef] [PubMed]
  64. Batteux, F.; Kavian, N.; Servettaz, A. New insights on chemically induced animal models of systemic sclerosis. Curr. Opin. Rheumatol. 2011, 23, 511–518. [Google Scholar] [CrossRef] [PubMed]
  65. Finch, W.R.; Rodnan, G.P.; Buckingham, R.B.; Prince, R.K.; Winkelstein, A. Bleomycin-induced scleroderma. J. Rheumatol. 1980, 7, 651–659. [Google Scholar]
  66. Kerr, L.D.; Spiera, H. Scleroderma in association with the use of bleomycin: A report of 3 cases. J. Rheumatol. 1992, 19, 294–296. [Google Scholar]
  67. Artlett, C.M.; Sassi-Gaha, S.; Rieger, J.L.; Boesteanu, A.C.; Feghali-Bostwick, C.A.; Katsikis, P.D. The inflammasome activating caspase 1 mediates fibrosis and myofibroblast differentiation in systemic sclerosis. Arthritis Rheum. 2011, 63, 3563–3574. [Google Scholar] [CrossRef]
  68. Kawaguchi, Y.; Takagi, K.; Hara, M.; Fukasawa, C.; Sugiura, T.; Nishimagi, E.; Harigai, M.; Kamatani, N. Angiotensin II in the lesional skin of systemic sclerosis patients contributes to tissue fibrosis via angiotensin II type 1 receptors. Arthritis Rheum. 2004, 50, 216–226. [Google Scholar] [CrossRef]
  69. Loeys, B.L.; Gerber, E.E.; Riegert-Johnson, D.; Iqbal, S.; Whiteman, P.; McConnell, V.; Chillakuri, C.R.; Macaya, D.; Coucke, P.J.; De Paepe, A.; et al. Mutations in fibrillin-1 cause congenital scleroderma: Stiff skin syndrome. Sci. Transl. Med. 2010, 2, 23ra20. [Google Scholar] [CrossRef] [Green Version]
  70. Tan, F.K.; Wang, N.; Kuwana, M.; Chakraborty, R.; Bona, C.A.; Milewicz, D.M.; Arnett, F.C. Association of fibrillin 1 single-nucleotide polymorphism haplotypes with systemic sclerosis in Choctaw and Japanese populations. Arthritis Rheum. 2001, 44, 893–901. [Google Scholar] [CrossRef]
  71. Ong, V.H.; Evans, L.A.; Shiwen, X.; Fisher, I.B.; Rajkumar, V.; Abraham, D.J.; Black, C.M.; Denton, C.P. Monocyte chemoattractant protein 3 as a mediator of fibrosis: Overexpression in systemic sclerosis and the type 1 tight-skin mouse. Arthritis Rheum. 2003, 48, 1979–1991. [Google Scholar] [CrossRef] [PubMed]
  72. Christner, P.J.; Jimenez, S.A. Animal models of systemic sclerosis: Insights into systemic sclerosis pathogenesis and potential therapeutic approaches. Curr. Opin. Rheumatol. 2004, 16, 746–752. [Google Scholar] [CrossRef] [PubMed]
  73. Christner, P.J.; Peters, J.; Hawkins, D.; Siracusa, L.D.; Jiménez, S.A. The tight skin 2 mouse. An animal model of scleroderma displaying cutaneous fibrosis and mononuclear cell infiltration. Arthritis Rheum. 1995, 38, 1791–1798. [Google Scholar] [CrossRef] [PubMed]
  74. Varga, J.; Bashey, R.I. Regulation of connective tissue synthesis in systemic sclerosis. Int. Rev. Immunol. 1995, 12, 187–199. [Google Scholar] [CrossRef]
  75. Lafyatis, R. Transforming growth factor β--at the centre of systemic sclerosis. Nat. Rev. Rheumatol. 2014, 10, 706–719. [Google Scholar] [CrossRef]
  76. Martinović Kaliterna, D.; Petrić, M. Biomarkers of skin and lung fibrosis in systemic sclerosis. Expert Rev. Clin. Immunol. 2019, 15, 1215–1223. [Google Scholar] [CrossRef]
  77. Sonnylal, S.; Denton, C.P.; Zheng, B.; Keene, D.R.; He, R.; Adams, H.P.; Vanpelt, C.S.; Geng, Y.J.; Deng, J.M.; Behringer, R.R.; et al. Postnatal induction of transforming growth factor beta signaling in fibroblasts of mice recapitulates clinical, histologic, and biochemical features of scleroderma. Arthritis Rheum. 2007, 56, 334–344. [Google Scholar] [CrossRef]
  78. Gerber, E.E.; Gallo, E.M.; Fontana, S.C.; Davis, E.C.; Wigley, F.M.; Huso, D.L.; Dietz, H.C. Integrin-modulating therapy prevents fibrosis and autoimmunity in mouse models of scleroderma. Nature 2013, 503, 126–130. [Google Scholar] [CrossRef] [Green Version]
  79. Sundaresan, N.R.; Bindu, S.; Pillai, V.B.; Samant, S.; Pan, Y.; Huang, J.Y.; Gupta, M.; Nagalingam, R.S.; Wolfgeher, D.; Verdin, E.; et al. SIRT3 Blocks Aging-Associated Tissue Fibrosis in Mice by Deacetylating and Activating Glycogen Synthase Kinase 3β. Mol. Cell Biol. 2015, 36, 678–692. [Google Scholar] [CrossRef] [Green Version]
  80. Vancheeswaran, R.; Magoulas, T.; Efrat, G.; Wheeler-Jones, C.; Olsen, I.; Penny, R.; Black, C.M. Circulating endothelin-1 levels in systemic sclerosis subsets--A marker of fibrosis or vascular dysfunction? J. Rheumatol. 1994, 21, 1838–1844. [Google Scholar]
  81. Stochmal, A.; Czuwara, J.; Zaremba, M.; Rudnicka, L. Altered serum level of metabolic and endothelial factors in patients with systemic sclerosis. Arch. Dermatol. Res. 2020, 312, 453–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Herrick, A.L. Evidence-based management of Raynaud’s phenomenon. Ther. Adv. Musculoskelet. Dis. 2017, 9, 317–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Kawaguchi, Y.; Suzuki, K.; Hara, M.; Hidaka, T.; Ishizuka, T.; Kawagoe, M.; Nakamura, H. Increased endothelin-1 production in fibroblasts derived from patients with systemic sclerosis. Ann. Rheum. Dis. 1994, 53, 506–510. [Google Scholar] [CrossRef] [PubMed]
  84. Gay, S.; Jones, R.E.; Huang, G.Q.; Gay, R.E. Immunohistologic demonstration of platelet-derived growth factor (PDGF) and sis-oncogene expression in scleroderma. J. Investig. Dermatol. 1989, 92, 301–303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Zheng, X.Y.; Zhang, J.Z.; Tu, P.; Ma, S.Q. Expression of platelet-derived growth factor B-chain and platelet-derived growth factor beta-receptor in fibroblasts of scleroderma. J. Dermatol. Sci. 1998, 18, 90–97. [Google Scholar] [CrossRef] [PubMed]
  86. Ludwicka, A.; Ohba, T.; Trojanowska, M.; Yamakage, A.; Strange, C.; Smith, E.A.; Leroy, E.C.; Sutherland, S.; Silver, R.M. Elevated levels of platelet derived growth factor and transforming growth factor-beta 1 in bronchoalveolar lavage fluid from patients with scleroderma. J. Rheumatol. 1995, 22, 1876–1883. [Google Scholar]
  87. Soria, A.; Cario-André, M.; Lepreux, S.; Rezvani, H.R.; Pasquet, J.M.; Pain, C.; Schaeverbeke, T.; Mahon, F.X.; Taïeb, A. The effect of imatinib (Glivec) on scleroderma and normal dermal fibroblasts: A preclinical study. Dermatology 2008, 216, 109–117. [Google Scholar] [CrossRef]
  88. Yang, H.; Li, Z.T.; Lin, R.; Fan, Z.P.; Huang, F.; Jiang, Q.L.; Zhou, H.S.; Liu, Q.F.; Sun, J. Sclerodermatous chronic graft-versus-host disease after hematopoietic stem cell transplantation: Incidence, clinical characteristics and risk factors. Nan Fang Yi Ke Da Xue Xue Bao 2016, 36, 807–813. [Google Scholar]
  89. Yang, C.; Tang, S.; Zhu, D.; Ding, Y.; Qiao, J. Classical Disease-Specific Autoantibodies in Systemic Sclerosis: Clinical Features, Gene Susceptibility, and Disease Stratification. Front. Med. 2020, 7, 587773. [Google Scholar] [CrossRef]
  90. Allen, J. Capillaroscopy in Healthy Subjects of Different Ages. In Atlas of Capillaroscopy in Rheumatic Disease, 2011st ed.; Cutolo, M., Ed.; Elsevier: Amsterdam, The Netherlands, 2011; pp. 49–53. [Google Scholar]
  91. Bakirci, S.; Celik, E.; Acikgoz, S.B.; Erturk, Z.; Tocoglu, A.G.; Imga, N.N.; Kaya, M.; Tamer, A. The evaluation of nailfold videocapillaroscopy findings in patients with type 2 diabetes with and without diabetic retinopathy. North Clin. Istanb. 2019, 6, 146–150. [Google Scholar] [CrossRef]
  92. Hsu, P.C.; Liao, P.Y.; Chang, H.H.; Chiang, J.Y.; Huang, Y.C.; Lo, L.C. Nailfold capillary abnormalities are associated with type 2 diabetes progression and correlated with peripheral neuropathy. Medicine 2016, 95, e5714. [Google Scholar] [CrossRef] [PubMed]
  93. Kuryliszyn-Moskal, A.; Zarzycki, W.; Dubicki, A.; Moskal, D.; Kosztyła-Hojna, B.; Hryniewicz, A. Clinical usefulness of videocapillaroscopy and selected endothelial cell activation markers in people with Type 1 diabetes mellitus complicated by microangiopathy. Adv. Med. Sci. 2017, 62, 368–373. [Google Scholar] [CrossRef] [PubMed]
  94. Maldonado, G.; Guerrero, R.; Paredes, C.; Ríos, C. Nailfold capillaroscopy in diabetes mellitus. Microvasc. Res. 2017, 112, 41–46. [Google Scholar] [CrossRef] [PubMed]
  95. Gallucci, F.; Russo, R.; Buono, R.; Acampora, R.; Madrid, E.; Uomo, G. Indications and results of videocapillaroscopy in clinical practice. Adv. Med. Sci. 2008, 53, 149–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Cutolo, M.; Pizzorni, C.; Secchi, M.E.; Sulli, A. Capillaroscopy. Best Pract. Res. Clin. Rheumatol. 2008, 22, 1093–1108. [Google Scholar] [CrossRef]
  97. Pavan, T.R.; Bredemeier, M.; Hax, V.; Capobianco, K.G.; da Silva Mendonça Chakr, R.; Xavier, R.M. Capillary loss on nailfold capillary microscopy is associated with mortality in systemic sclerosis. Clin. Rheumatol. 2018, 37, 475–481. [Google Scholar] [CrossRef]
  98. Kowal-Bielecka, O.; Distler, O.; Neidhart, M.; Künzler, P.; Rethage, J.; Nawrath, M.; Carossino, A.; Pap, T.; Müller-Ladner, U.; Michel, B.A.; et al. Evidence of 5-lipoxygenase overexpression in the skin of patients with systemic sclerosis: A newly identified pathway to skin inflammation in systemic sclerosis. Arthritis Rheum. 2001, 44, 1865–1875. [Google Scholar] [CrossRef]
  99. Chwieśko-Minarowska, S.; Kowal, K.; Bielecki, M.; Kowal-Bielecka, O. The role of leukotrienes in the pathogenesis of systemic sclerosis. Folia Histochem. Cytobiol. 2012, 50, 180–185. [Google Scholar] [CrossRef] [Green Version]
  100. Xiao, R.; Yoshida, N.; Higashi, Y.; Lu, Q.J.; Fukushige, T.; Kanzaki, T.; Kanekura, T. Retinoic acids exhibit anti-fibrotic activity through the inhibition of 5-lipoxygenase expression in scleroderma fibroblasts. J. Dermatol. 2011, 38, 345–353. [Google Scholar] [CrossRef]
  101. Liang, M.; Lv, J.; Jiang, Z.; He, H.; Chen, C.; Xiong, Y.; Zhu, X.; Xue, Y.; Yu, Y.; Yang, S.; et al. Promotion of Myofibroblast Differentiation and Tissue Fibrosis by the Leukotriene B. Arthritis Rheumatol. 2020, 72, 1013–1025. [Google Scholar] [CrossRef]
Figure 1. Human and animal NFC. (A) Guinea pig specimen used for NFC. (B) Guinea pig nailfold used for NFC. (C,D) Guinea pig NFC. (E,F) normal human NFC. NFC: nailfold capillaroscopy.
Figure 1. Human and animal NFC. (A) Guinea pig specimen used for NFC. (B) Guinea pig nailfold used for NFC. (C,D) Guinea pig NFC. (E,F) normal human NFC. NFC: nailfold capillaroscopy.
Life 12 00703 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mandujano, A.; Golubov, M. Animal Models of Systemic Sclerosis: Using Nailfold Capillaroscopy as a Potential Tool to Evaluate Microcirculation and Microangiopathy: A Narrative Review. Life 2022, 12, 703. https://doi.org/10.3390/life12050703

AMA Style

Mandujano A, Golubov M. Animal Models of Systemic Sclerosis: Using Nailfold Capillaroscopy as a Potential Tool to Evaluate Microcirculation and Microangiopathy: A Narrative Review. Life. 2022; 12(5):703. https://doi.org/10.3390/life12050703

Chicago/Turabian Style

Mandujano, Angélica, and Melissa Golubov. 2022. "Animal Models of Systemic Sclerosis: Using Nailfold Capillaroscopy as a Potential Tool to Evaluate Microcirculation and Microangiopathy: A Narrative Review" Life 12, no. 5: 703. https://doi.org/10.3390/life12050703

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