Next Article in Journal
Bone Tissue and the Nervous System: What Do They Have in Common?
Previous Article in Journal
Natural Infection of Pomegranate (Punica Granatum) by Apple Dimple Fruit Viroid
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Multiple Roles of Periostin in Non-Neoplastic Disease

1
College of Life Science, Northwest University, Xi’an 710069, China
2
Department of Otorhinolaryngology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710032, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2023, 12(1), 50; https://doi.org/10.3390/cells12010050
Submission received: 12 November 2022 / Revised: 5 December 2022 / Accepted: 20 December 2022 / Published: 22 December 2022
(This article belongs to the Section Cell Motility and Adhesion)

Abstract

:
Periostin, identified as a matricellular protein and an ECM protein, plays a central role in non-neoplastic diseases. Periostin and its variants have been considered to be normally involved in the progression of most non-neoplastic diseases, including brain injury, ocular diseases, chronic rhinosinusitis, allergic rhinitis, dental diseases, atopic dermatitis, scleroderma, eosinophilic esophagitis, asthma, cardiovascular diseases, lung diseases, liver diseases, chronic kidney diseases, inflammatory bowel disease, and osteoarthrosis. Periostin interacts with protein receptors and transduces signals primarily through the PI3K/Akt and FAK two channels as well as other pathways to elicit tissue remodeling, fibrosis, inflammation, wound healing, repair, angiogenesis, tissue regeneration, bone formation, barrier, and vascular calcification. This review comprehensively integrates the multiple roles of periostin and its variants in non-neoplastic diseases, proposes the utility of periostin as a biological biomarker, and provides potential drug-developing strategies for targeting periostin.

1. Introduction

Periostin (encoded by the Postn gene) was first recognized in 1993 from a mouse osteoblastic cell line, a secreted molecule containing no transmembrane domain, and was initially known as osteoblast-specific factor-2(OSF-2) [1]. It was renamed periostin in 1999 due to its preferential expression in the periosteum and periodontal ligament in adult mice reported by Horiuchi et al. Periostin used as a regulator promotes the adhesion and differentiation of osteoblasts [2].
At approximately 90 kDa, periostin as an N-glycoprotein contains 23 exons exhibiting an NH2-terminal secretory signal peptide, accompanied by a highly conserved cysteine-rich EMI domain, which engages in the formation of multimers through cysteine disulfide bonds [3,4], four consecutive and homologous tandem Fasciclin I (FAS1) domains binding to integrins (αvβ1, αvβ3, αvβ5, αMβ2, α6β4, α5β1) and a COOH-terminal hydrophilic domain as an alternatively spliced region consisting of exons 15–23 (Figure 1) [5,6,7,8,9]. Periostin-integrin interactions lead to the activation of signaling pathways (Table 1; Figure 2). Apart from interacting with integrin receptors, it also binds other ECM proteins, for example, collagens, fibronectin, tenascin C, or heparin [4,10]. Periostin can form 11 splice variants (Figure 3). The expression pattern of periostin splicing variants has been reported in cerebral ischemia, asthma, MI, IPF, retinal ischemia, pIBD, joint, and serum (Table 2). Comparability of mouse and human periostin amino acid is 89.2% overall and 90.1% in a mature condition. Mouse and human periostin are respectively located on chromosome 3 and chromosome 13q.
Periostin is commonly overexpressed in human tissues during pathological processes. Periostin, as a matricellular protein and an ECM protein, exerts different roles in tissue development and progression of diseases, including brain injury, ocular diseases, chronic rhinosinusitis, allergic rhinitis, dental diseases, atopic dermatitis, scleroderma, eosinophilic esophagitis, asthma, cardiac diseases, lung diseases, liver diseases, chronic kidney diseases, inflammatory bowel disease, and osteoarthrosis. In a normal physiological situation, periostin is beneficial in mediating teeth development, maintaining the integrity of periodontal ligament (PDL) in postnatal teeth enamel formation, and mediating bone remodeling after orthodontic movement [119,120]; periostin promotes migration of mesenchymal cells in an αvβ3- and β1-based Rho/PI3K signaling mechanism during valve maturation [10]. Besides, during pathogenesis, the roles of periostin are more extensive, including tissue remodeling, fibrosis, inflammation, wound healing, repair, angiogenesis, tissue regeneration, bone formation, barrier, and vascular calcification; this makes it different from other ECM proteins.
Periostin assists in modulating the ECM network [9]. Periostin/BMP-1/LOX cascade assisted in collagen cross-linking [121]. During abnormal scar formation, periostin stimulated the secretion of TGF-β1 via the RhoA/ROCK signaling pathway in human dermal fibroblasts (HDFs), yielding a vicious circle [122].
Herein, we have sufficient knowledge of its stimulators, repressors, expression levels, expression patterns, and roles, as well as periostin-involved signaling pathways, the potential of its guiding choice for medicine, and the serviceability of it as a prospective marker in various diseases. Developing drugs based upon periostin-involved functions or certain periostin isoforms-mediated distinctive roles is beneficial in relieving diseases.

2. Early Brain Injury (EBI) and Cerebral Ischemia

The periostin was upregulated in neurons and capillary endothelial cells in the cerebral cortex at 24 h post-subarachnoid hemorrhage (SAH) and initiated BBB disruption, possibly via p38/ERK/MMP-9 signaling pathways and induction of tenascin-C [11].
Following transient cerebral ischemia, isoform 2 minimized the area of cerebral infarction displaying a neuroprotective role with phosphorylation of Akt [111]. Greater serum periostin levels were related to a larger cerebral infarction area and more serious neurological defects at 6-28 days following ischemia [14]. Toll-like receptor 4 (TLR4) selective blockade-IAXO-102 and clarithromycin inhibited BBB disruption and periostin expression [12,13].

3. Cardiovascular Diseases

3.1. Myocardial Infarction (MI)

Ang II evidently increased periostin through Ras/p38 MAPK (mitogen-activated protein kinase)/CREB and ERK/TGF-β1 pathways in myocytes and fibroblasts [123]. Detection of human tissue specimens reflected prominently high periostin expression in ischemic and reperfused tissue, as well as no expression in healthy myocardium [59]. The lineage analyses of mice verified that periostin-expressing CFs mainly derived from a mass of TCF21+ cells [124]. After MI, TGF-β1, mechanical pressure, and Cyclic AMP response element-binding protein 1 (CREB) stimulated cardiac fibroblasts, thereby augmenting ECM deposition, development of collagenous scar and cardiac remodeling, and release of periostin [125]. TGF-β1 upregulated periostin levels in CFs and vascular smooth muscle cells (VSMCs) employing Smad signaling pathways [126,127]. Periostin showed minimal levels under miR-203-3p-binding circumstances restricting cardiomyocytes apoptosis. However, the complex of periostin, miR-203-3p, and small nucleolar RNA host gene 8 (Snhg8) mediated neonatal mouse cardiomyocytes (NMCMs) apoptosis after hypoxia-treated NMCMs, contributing to acute myocardial infarction [66]. Treatment of MI with cardiac mesenchymal stem cells (MSCs) marked by Nestin demonstrated a greater effect on cardiac healing than bone marrow-derived MSCs (NesbmMSCs), which results from part involvement of periostin-induced M2 macrophage polarization [65]. In a rat MI model, Yoshiaki Taniyama et al. discovered four periostin isoforms, including isoforms 1, 2, 5, and 6. Isoform 1 decreased the attachment of fibroblasts and myocytes as well as facilitated myocyte death, leading to ventricular dilation and tissue remodeling. Blockade of exon 17 as prior target assists in protesting cardiac remodeling, diminishing fibrosis, ameliorating ejection fraction, and cardiac function eight weeks after MI [114]. Isoform 6 can mediate the migration of activated fibroblasts and the healing of impaired tissue by αv/FAK/AKT cascade [59]. The inhibition of periostin by valsartan might have an improved effect on cardiac remodeling after MI [57].

3.2. Cardiomyocyte Regeneration

Release of periostin facilitated cardiomyocyte regeneration and angiogenesis by interacting with αvβ1, αvβ3, or αvβ5 integrins on myocytes and vascular endothelial cells to activate the PI3K-Akt pathways after MI. The treatment of animals with periostin patches (lacking the N-terminal signal peptide and C-terminal region) not only perfected cardiac fraction and ejection fraction but also contained fibrosis after MI [5]. Periostin eased inflammation and induced reentry of the cardiomyocytes cycle via TNF-α/NF-κB signaling transduction in conjunction with a declining caspase 7 activity [67]. Periostin ablation hindered myocardial regeneration by suppressing the PI3K/AKT/cyclin D1 transmission [68]. Another work in a mouse model of overexpressed full-length periostin indicated that periostin did not speed up the DNA synthesis of cardiomyocytes [128]. Further studies are needed to clarify these issues.

3.3. Heart Failure

In diabetic rat hearts, periostin is noticeably overexpressed relative to healthy controls [79]. In the experimental autoimmune myocarditis (EAM) rats model, periostin was spotted in macrophages and fibroblasts. It elicited cardiac fibrosis, likely by recruiting immune cells [60]. A recent examination of atrial appendages from atrial fibrillation (AF) patients suggested a clear association between periostin levels of atrial tissues and deteriorated heart failure, as well as lessened ejection fraction [129]. MiR-30a and fibromodulin (FMOD) tempted the descent of periostin levels and the decrease of atrial fibrosis [61]. GSN, silencing P2Y1R, and slit2-Robo1 pathways inversely initiated periostin release, tempting fibrosis [62]. Periostin prompted pyroptosis by triggering the NLRP3/caspase-1 pathway during myocardial ischemia-reperfusion injury (MIRI) [56].
Valsartan and simvastatin (SIM) hindered periostin expression and alleviated pathologic remodeling [63,79]. Targeting of diabetic animals with the antioxidant resveratrol limited myofibroblast activation and downregulated the expression of periostin via suppressing ERK/TGF-β signaling [69].

3.4. Valvular Heart Disease (VHD)

Periostin expression intensively goes up in valvular interstitial cells (VICs) of the mitral valve, compared to wild-type mice. The mitral valve biopsies of male patients going through prosthetic surgery detected a pronounced enhancement in periostin in the ventricular [130]. Besides, periostin was firmly upregulated in the infiltrated inflammatory cells and myofibroblasts within patients with atherosclerotic or rheumatic valves. Meanwhile, massive periostin in the valve leaflet brings about extensive production of matrix metalloproteinase-2 (MMP-2) and MMP-9, leading to severe fibrosis in atherosclerotic and rheumatic VHD [64]. Periostin also prompted the osteogenic potential of aortic valve calcification [131].

3.5. Hypertension and Vascular Calcification

Atrial natriuretic peptide (ANP) inhibited periostin expression in the VSMCs and cardiac fibroblasts [70], but oxidative stress contributes to periostin production [132]. The increase in periostin augmented the differentiation and migration of VSMCs [133].
In a hyperlipidemia-associated model of rats, periostin upregulation caused calcium deposits through the successive inhibition of p53 and SLC7A11 in VSMCs [71]. Additionally, plasma periostin levels were positively connected with the Agatston score in patients with coronary artery calcification (CAC). Periostin promoted glycolysis and mitochondrial malfunction as well as contained peroxisome proliferation-activated receptor γ(PPARγ) in VSMCs, thereby provoking arterial calcification [58].

4. Ocular Diseases

IL-13 obviously stimulated periostin in conjunctival fibroblasts and, to a much smaller extent, in conjunctival epithelial cells. The recruitment of eosinophils and Th2 cytokines expression, including CCL5, IL-4, and IL-13, were restricted in periostin-deleted AC mice [15]. The concentration of tear periostin is heavier among patients with atopic keratoconjunctivitis (AKC) relative to healthy controls. Tear periostin levels had an infinitely positive association with complications of AKC by acting on corneal or conjunctival epithelial cells [17]. Tear periostin was decreased by treating with tacrolimus or betamethasone along with ameliorative clinical traits in the majority of patients with AKC [16].
Periostin upregulation may assist in scleral remodeling in myopia [18]. It was also manifestly increased in the vitreous of patients with proliferative vitreoretinal diseases, such as proliferative vitreoretinopathy (PVR) and proliferative diabetic retinopathy. It was colocalized with α-SMA and M2 macrophage markers in the retinal fibrovascular membrane (FVM). The inhibition of it decreased retinal FVM formation [134]. Another study of diabetic retinopathy patients uncovered a positive correlation between serum periostin with continuous retinopathy and FVM formation [19]. Expression levels of isoforms 1, 2, and 5 are increased when the preretinal pathological neovascularization (NV) reaches the peak; they may be specific periostin splice variants for preretinal pathological NV in retinal ischemia [116].

5. Dental Diseases

During early periodontitis, Wnt5a/CaMKII/Periostin axis mediated collagen and bone formation, maintaining periodontal stabilization [135]. Applying gingivectomy to a rat model presented that periostin promotes ECM generation, as well as increases the formation of fibronectin and collagen via β1/FAK (focal adhesion kinase)/JNK propagation during wound healing. Periostin is not related to myofibroblast differentiation accounting for lessened scar generation [31]. By introducing an excisional palatal model, periostin mRNA and protein expression were upregulated, and it is correlated with fibronectin generation, transition to myofibroblast, and attachment of macrophages to the wound region. Periostin modulated palatal healing via the integrinβ1/RhoA pathway [31].
IL-4 and IL-13 evidently stimulated periostin expression in the human PDL (hPDL). HPDL cells displayed increased proliferation and migration and no significant difference in the generation of inflammatory cytokines under periostin stimulation [34,136]. TNF-α/periostin/JNK promoted the adhesion and osteogenic differentiation competence of human periodontal ligament stem cells (PDLSCs) [34,35].
GCF periostin levels are degressive with the activity and severity of periodontal disease, suggesting its beneficial role in maintaining the function of normal periodontal tissue [32,33]. Salivary periostin levels are positively linked to gingival inflammation and aggressive periodontitis (AgP) severity [33].

6. Chronic Rhinosinusitis (CRS) and Allergic Rhinitis (AR)

Among patients of CRS with nasal polyposis (CRSwNP), expression of the periostin gene seemed to be notably upregulated in nasal polyps than in normal sinus mucosa [137]. Mi1on’ski et al. revealed upregulation of periostin in non-polyp and polyp tissue of patients with CRS compared with patients without CRS [138].
CRSwNP and AR were taken for Th2-dominant inflammatory diseases. Higher periostin levels were related to increased basement membrane thickness, subepithelial fibrosis, and eosinophilia among patients undergoing surgery for CRS [20]. Periostin-induced tissue remodeling by activating the Src/AKT/mTOR signaling pathway and inducing myofibroblasts differentiation and expression of ECM or by enhancing the mRNA expression of MMP-3, MMP-7, MMP-8, and MMP-9 in fibroblasts and MMP-9 in epithelial cells in CRS. Additionally, IgE enhanced the periostin expression by a cultured human mast cell line (LAD2 mast cells), thereby leading to epithelial cells secreting thymic stromal lymphopoietin (TSLP) by binding to integrin, in turn activating mast cells to produce IL-5 [24]. Glucocorticoids (GCs) eased CRS by restricting the increase of periostin [28]. Previous studies thought that tissue periostin expression has evident relation with IL-5 and IL-13 levels among patients with CRSwNP [139,140].
A controversial finding demonstrated that the complete absence of periostin might result in mast cell attachment and polyp-like signs in CRSwNP [25]. Serum periostin was higher in patients with CRS than in controls [141]. Periostin levels of nasal lavage fluids (NLF) might function as a reliable marker involved in CRS [27]. Application of omalizumab, mepolizumab, methylprednisolone, and doxycycline into CRSwNP subjects arrested periostin production and inflammatory responses. Doxycycline decreased nasal periostin levels (p = 0.084), leading to the less frequent onset of asthma and reduced relapse of nasal polyps [22].
In an ovalbumin-treated murine model of AR, periostin knockout mice appeared to have lesser eosinophils, lower nasal symptom scores, and minimal nasal remodeling than controls [21]. Serum periostin serves to estimate the clinical responses to sublingual immunotherapy (SLIT) within house dust mite (HDM)-induced AR subjects [23]. Asarum heterotropoides (AH) and Angelica gigas extract (AG-Ex) interfered with periostin release in HNEpCs (human nasal epithelial cells) and alleviated AR symptoms [29,30]. The treatment of AR with nasal neurectomy pronouncedly reduced NLF periostin value [142].

7. Asthma

Allergic asthma is mostly a Th2-involved heterogeneous inflammation attended by eosinophilia, airway hyperresponsiveness (AHR), and excessive mucus secretion from goblet cells. IL-13/IL-4 have been found to induce periostin expression in bronchial epithelial cells and lung fibroblasts. Periostin isoforms 6, 7, and 8 are evidently expressed in lung fibroblasts [112]. MiR-185-5p negatively modulates mRNA and protein expression of periostin within airway cells and sputum periostin concentration [143]. MiR-221-3p provoked airway eosinophilic inflammation by suppressing CXCL17 expression and subsequently upregulating CCL24, CCL26, and periostin expression in HDM-stimulated mice [144].
A previous investigation into the aspergillus fumigatus antigen-challenged mice model supported that periostin serves a beneficial role in protesting AHR, serum IgE levels, and outcome of peribronchial fibrosis by intensifying TGF-β-mediated Treg differentiation [51]. Besides, another mouse model revealed that periostin suppressed mucus production of goblet cells and increased airflow by checking the expression of Gob5 and Muc5ac [52]. The roles of periostin absence in goblet cell metaplasia (GCM) were involved in at least two pathological mechanisms: direct impacts on differentiation of airway epithelial cells to goblet cells and indirect influences by changing the number of DC-derived cytokines acting on T cells.
A study in HDM-challenged mice offered the opposite effect. Periostin-expressing dendritic cells (DCs) from HDM-challenged wild-type mice kept asthma-like features and IL-13 responses after transferring into periostin null mice [43]. Application of anti-periostin antibody OC-20 weakened the AHR, IgE response, IL-13 responses, and DNA synthesis of T cells incubated with periostin-positive DCs. Periostin-overexpressed epithelial cells manifested that release of TGF-β in epithelial cells is attributed to a signaling pathway involving periostin/MMP-2, MMP-9, resulting in collagen Ⅰ production of airway fibroblasts. The process fuels the matrix stiffening [47]. The crosstalk of periostin and TSLP is an exquisitely driving factor for asthma [145]. In asthma patients, Kanemitsu et al. concluded that the accumulation of periostin in bronchial subepithelium was manifestly linked to the descent of FEV1 [146]. Anti-αMβ2 (specifically to periostin isoforms 1 and 8) and anti-ADAM8 blockers contained adhesion and migration of IL-5-stimulated eosinophils into periostin [113,147,148].
Serum periostin is linked to type 2 biomarkers, including eosinophilia, IgE concentration, and the fraction of NO (FeNO) inhalation, IL-4, and TSLP [149]. High serum periostin levels in patients receiving corticosteroids had prominent relation with the decline of pulmonary function tests and the increase of airflow limitation [46]. Both plasma periostin and saliva periostin levels had the advantage of early diagnosis of asthma [49]. Exhaled breath condensate (EBC) periostin levels seemed to reflect the emergence of CRS in asthma [54]. Sputum periostin levels offer an accurate diagnosis of serious asthma with continuous airflow limitation compared with mild-to-moderate asthma [55].
Lebrikizumab as an anti-IL-13 antibody was available to improve the function of the lung [150]. Periostin levels were strikingly correlated positively with the efficacy of these drugs, which included anti-IL-13 Ab-tralokinumab and dupilumab (common receptor of IL-4 and IL-13) [44,45]. The addition of dupilumab lowers serum periostin expression in AD, asthma, CRSwNP, and EE [26]. In addition to this, the effective therapeutic response of anti-IgE Ab omalizumab in asthma patients was dependent on high serum periostin [48]. Both hydroprednisone therapy and glucocorticoid-induced transcript 1 (GLCCI1) overexpression repressed the airway remodeling in asthma mice model via suppressing IL-13/periostin/TGF-β1 axis [50]. Clarithromycin can alleviate asthma by arresting periostin generation [53].

8. Lung Diseases

8.1. Pulmonary Fibrosis (PF)

Periostin is overexpressed in the lungs of patients with idiopathic pulmonary fibrosis (IPF). It was produced by fibroblasts and promoted their proliferation [151,152]. Nance et al. proposed that periostin mRNA was relatively lacking exon 21 in IPF samples compared to controls [115]. The absence of low-density lipoprotein receptor-related protein 1 (LRP1) prominently irritates the JNK/c-Jun/Fra-2 signaling pathway leading to the induction of α-SMA and periostin expression in human lung fibroblasts (hLF), tempting fibrosis of the lung [75]. Periostin furthered the recruitment of neutrophils and macrophages or myofibroblasts differentiation, accelerating pulmonary fibrosis [72,76]. The crosstalk of TGF-β and periostin also participated in the process of PF [73]. Serum monomeric periostin and EBC periostin both served as possible biomarkers to monitor IPF progression [74]. Moreover, serum periostin was also linked to fibrogenesis in COVID-19 [153]. Periostin of bronchoalveolar lavage fluid (BALF) might exaggerate the onset of eosinophilic pneumonia (EP), IPF, and COVID-19 [79,80]. The siRNA and antisense oligonucleotide targeting periostin, OC-20, and antibodies targeting αv integrin prevented lung fibrosis [151,154,155].

8.2. Pulmonary Hypertension (PH)

In ascending aortic constriction (AAC)-treated PH model, kcnk3-mutated rats presented greater expression of IL-6 and periostin in lung and heart as well as the lower extent of lung ctnnd1 mRNA levels, aggravating pulmonary and heart remodeling as well as lung vascular edema [77]. The feedback cycle between HIF-1α and periostin magnified PH by intensifying the proangiogenic role [78].

9. Atopic Dermatitis (AD)

Characteristics of AD include type 2 immune response, dermal fibrosis, barrier malfunction, and itch. Histamine and TNF superfamily member 14 (TNFSF14) upregulated periostin levels [156,157], which mediated the crosstalk of epithelial/mesenchymal. There exist two potential mechanisms to interpret it: first, IL-4/IL-13 tempts periostin secretion in fibroblasts. Periostin applies to keratinocytes via activating αv-mediated NF-κB signaling accompanied by the release of TSLP, which differentiates or stimulates DCs, developing a vicious cycle of type 2 inflammatory responses. Second, IL-1α and periostin are separately released by keratinocytes and fibroblasts, and their unity applies to fibroblasts by transducing the NF-κB pathway. Activated fibroblasts generate IL-6, contributing to the growth of keratinocytes. In addition, the cross-link of immune cells/non-immune cells with the help of periostin also accounted for the pathological mechanism of allergy. Periostin generated by fibroblasts amplifies adhesion, O2 emergence, and TGF-β release in eosinophils. Activated eosinophils, in turn, lead to periostin generation in fibroblasts. Another paper uncovered the crosstalk of epithelial/sensory neurons. i.e., keratinocytes-derived TSLP directly targets TRPA1+ sensory neurons, irritating skin itch [36]. Signaling transmission of activating αvβ3/TRPV1/TRPA1/NPPB (natriuretic polypeptide B) in sensory neurons is involved in the periostin-mediated itch mechanism. The TSLP-periostin vicious loop also augmented inflammation and itch, creating ever-terrible circumstances: Keratinocytes secreted TSLP unlocking inflammatory response, and then TSLP back triggered the release of keratinocytes-derived periostin by means of TSLPR/JAK/STAT signaling propagation. In turn, periostin reciprocally stimulates the production of keratinocytes-derived TSLP. IL-13/STAT6/periostin/IL-24/STAT3 signaling transmission in keratinocytes sped up the inflammation process by incurring epidermal barrier malfunction [37].
The concentration of serum periostin rests on the grade of clinical severity of AD. It is related to other type 2 biomarkers―LDH and eosinophils, but not with IgE. Thus, monitoring it is of great help for the diagnoses and therapies of AD patients [158]. The blocking antibodies directed toward αv delayed AD progression [159]. By introducing dupilumab drugs, clinical outcomes were improved, and serum periostin evidently decreased [160]. Antioxidant cinnamaldehyde stimulated the NRF2/HMOX1 pathway and alleviated IL-13 and TGF-β1 mediated production of ROS, subsequently downregulating periostin in dermal fibroblasts. It may benefit in treating systemic fibrotic diseases [38].

10. Scleroderma

Periostin was upregulated in the skin of patients with scleroderma. The bleomycin-treated periostin−/− mice showed reduced skin fibrosis followed by the descent of α-SMA+ myofibroblasts. However, recombinant mouse periostin resulted in the generation of collagen1α1 in myofibroblasts via the αv/PI3K/AKT signal axis [39]. Yamaguchi et al. discovered that periostin was colocalized with α-SMA+ myofibroblasts [161] and platelet endothelial cell adhesion molecule-1+ endothelial cells. Elevated serum periostin levels were associated with the severity of skin sclerosis. Crenolanib is an effective medication for diminishing skin and heart fibrosis by inhibiting periostin expression [40].

11. Eosinophilic Esophagitis (EE)

IL-13 and TGF-β stimulated periostin release in primary esophageal fibroblasts. Periostin was manifestly overproduced in the esophageal papillae and correlated positively with esophageal eosinophil amounts among patients with EE. The migration of eosinophils to the esophagus is due to the specific interaction of αM with periostin [41]. Elevated serum periostin levels were positively associated with IL-13 levels and may be used as a biomarker in EE in the presence of anti-IL-13 treatment [42].

12. Liver Diseases

In a mice model of dexamethasone (DEX)-treated fatty liver, DEX induced a higher degree of periostin expression in white adipose tissues, driving liver steatosis in a systemic organ-mediated fashion [81]. Periostin increased hepatic fibrosis and hepatic steatosis by inhibiting peroxisome proliferator-activated receptor-α(PPAR-α) expression [82]. Antisense oligonucleotides (ASOs) targeting periostin lowered hepatic steatosis in conjunction with reduced expression of α-SMA, collagen I, and other fibrotic markers and increased expression levels of PPAR-α. Another literature depicted that the periostin/α6β4/JNK/c-Jun prevented the binding of RORα to PPAR-α, suppressing PPAR-α expression and contributing to hepatosteatosis [83]. Periostin is mainly observed in activated hepatic stellate cells (HSCs). Periostin tempted liver fibrosis by activating LOX and lysyl oxidase-like (LOXL) in chronic liver disease via the αvβ3/PI3K/Smad2/3 signaling pathway [85]. Periostin deletion devastated angiogenesis in the process of liver regeneration [86]. Serum periostin is forcefully correlated with higher nonalcoholic fatty liver disease (NAFLD) [84].

13. Chronic Kidney Disease (CKD)

Periostin is overexpressed in a variety of kidney diseases. It is mainly presented in the glomerulus, renal arteries, tubular cells, and interstitial area. For healthy donors, periostin is found in the vascular pole of the glomerulus and around Bowman’s capsule. Some opposite evidence confirmed that periostin has no expression in healthy kidney specimens. Periostin upregulation contributed to the fibrosis of CKD disease by inducing the FAK/p38/ERK pathway and expression of collagen I [92]. Periostin strengthened fibrosis and apoptosis in tubular epithelial cells by activating the phosphorylated-p38 MAPK pathway, facilitated vascular calcification through αvβ3/Wnt/β-catenin signaling, and accelerated inflammatory reaction by activating the β3/FAK/AKT pathway under NF-κB medication or mTOR complex 1 (mTORC1)-mediated inhibition of autophagy in CKD [87,88,93,94]. Additionally, periostin/αv/ILK (integrin-linked kinase) and periostin/αvβ3/AKT/mTOR signaling pathways both aggravated the growth of cyst epithelial cells in autosomal dominant polycystic kidney disease (ADPKD) [5,89]. In contrast to the above reporter, periostin served beneficial roles in renal repair, such as driving the proliferation of tubular cells via binding to integrin-β1 as well as the polarization of macrophage embodying pro-reparative characteristic following acute kidney injury (AKI) [95].
In hypertensive nephropathy, periostin correlated positively with creatinine and proteinuria. Losartan deterred periostin synthesis leading to lower renal fibrosis [90]. In diabetic renal disease, elevated urine periostin content was accompanied by the emergence of albuminuria [162]. Moreover, serum periostin could estimate diabetic disease stages [97].
Periostin advanced the proliferation of mouse mesangial cells (MMCs) to augment renal malfunction in Immunoglobulin A nephropathy (IgAN) [96]. Urine periostin concentration correlated with tissue fibrosis in biopsy-proven IgA nephropathy subjects [163]. During the progression of UUO, mechanical stress as an initiating signal increased periostin accumulation in collecting duct cells. Subsequently, periostin advanced the production of proinflammatory factor MCP-1 that mediated macrophage infiltration, and then TGF-β secreted by infiltrating cells induced periostin production and strengthened the phenotype change of tubular epithelial cells [92]. After 5/6 nephrectomy, periostin which was detected in the distal tubule (DT) epithelial cell, drove the expression of fibroblast-specific protein-1 (FSP-1) and MMP-9 in distal collecting tubular cells [91]. Platelet-derived growth factor-BB (PDGF-BB) stimulated the PI3K/AKT/periostin signaling cascade, driving the expression of fibronectin and proliferation in MMCs in lupus nephritis [164].

14. Inflammatory Bowel Disease (IBD)

Periostin and αv integrin are more strongly presented in the colon tissues of UC (Ulcerative colitis) patients than in healthy colonic mucosa. Periostin accumulation occurred in pericryptal fibroblasts [165]. Introducing recombinant periostin elicits colitis in periostin-absence mice, and the blocking antibody specific to periostin obviously mitigates intestinal inflammatory disease. TNF-α stimulates the expression of periostin mRNA in intestinal epithelial cells (IECs). Periostin induced IL-8 expression and magnified NF-κB activity in IECs. Meanwhile, the combination of periostin with TNF-α synergistically reinforced IL-8 levels via interaction with integrin αv [98]. The pIBD patients presented elevated peri-cryptal staining compared to controls, but the expression pattern of periostin isoforms showed no significance. Thus, certain specific periostin isoforms and changes in periostin-binding molecule expression levels in the peri-cryptal ring might account for enhanced pericryptal periostin rings in pediatric IBD (pIBD) patients. Great plasma levels of the periostin during the period of pIBD remission may participate in mucosal healing and tissue repair [99]. Another literature on Crohn’s disease (CD) ascertained the cut-off levels of serum periostin in adult patients to serve to diagnose CD and forecast the activity status of CD [100].

15. Osteoarthrosis

15.1. Rheumatoid Arthritis (RA)

In a mouse model of mocking arthritis, periostin loss mice appeared to have a higher degree of inflammation. In RA remission, serum periostin embodied high extent of levels [104], increasing the risk of fragility fractures.

15.2. Osteoarthritis (OA)

Normal articular chondrocytes highly expressed isoforms 1 and 5, and anterior cruciate ligament (ACL) progenitor cells overexpressed isoforms 3, 4, 6, 7, and 8. ACL progenitor cells that highly expressed total periostin, not isoform 1, showed higher cell adhesion than articular chondrocytes that expressed lower total periostin [117]. Mechanical pressure, as the primary reason, initiates and fuels inflammatory responses of OA. The cDNA array analysis revealed that periostin is at maximal levels in the cartilage of OA than controls. The periostin-positive signal was detected in chondrocytes, periphery matrices close to the degraded region, fibrotic cartilage, and tissue of subchondral bone. The application of periostin into isolated human chondrocytes might provoke a high expression of IL-6 and IL-8 accompanied by the sufficient expression of MMP-1, MMP-3, MMP-13, and nitric oxide synthase-2(NOS2) in an NF-κB-activated mechanism [105]. Periostin accelerated cartilage denaturation in Wnt/β-catenin/MMP-13/ADAMTS4- or discoidin domain receptor-1(DDR1)/Akt/Wnt/β-catenin/MMP-13-dependent mechanism [106]. It also had a contributory effect on MMP-2 and MMP-3 expression in OA synoviocytes. Synovial fluid (SF) periostin was positively associated with the progression of OA [109].

15.3. Ankylosing Spondylitis (AS)

Periostin was secreted by osteoblasts in AS. Serum periostin was higher under high inflammatory factors, disease severity, and low radiographic injury conditions [166].

15.4. Osteoporosis

Periostin lowers sclerostin levels, followed by the activation of the LRP5/Wnt/β-catenin cascade, boosting gene transcription within osteoblasts to induce bone formation [101]. In huRANKL-overexpressed mice, cathepsin K (Ctsk) limited bone formation and increased bone fragility by preventing periostin generation, which offers an underlying mechanism for osteoporosis in PMW [167]. The 17β-E2/periostin/Wnt/β-catenin pathway can enhance the osteogenesis of bone marrow stromal cells (BMSCs) in ovariectomized (OVX) rats, thereby decreasing osteoporosis [102]. Periostin also reinforced the osteogenic competence of bone marrow skeletal stem cells in an ILK/Akt/GSK-3β-activated manner [103]. Serum periostin is related inversely to bone mineral density (BMD) in Chinese postmenopausal women (PMW) [168]. Cathepsin K-generated periostin (K-Postn) predominantly reflected fracture of Caucasian PMW with primary hyperparathyroidism (PHPT) [110].

15.5. Developmental Dysplasia of the Hip (DDH)

In chondrocytes, periostin upregulated IL-6 and MMP-3 levels based on the integrin-FAK-Src-NF-κB pathway. Meanwhile, it limited the production of Col2a1 and Acan. Then, IL-6/STAT3/periostin and MMP-3, as a vicious feedback loop, augmented hip dislocation-induced acetabular cartilage denaturation [107].

15.6. Intervertebral Disc Degeneration (IVDD)

Periostin accelerated nucleus pulposus cells (NPCs) apoptosis and intervertebral disc denaturation via the Wnt/β-catenin pathway [108].

16. Conclusions

Periostin exerts an integral role in the crosstalk between tumor cells and tumor microenvironments, cell and matrix, physiological function, and pathological function. Although the substantial data proved its significance in tissue remodeling, fibrosis, inflammation, wound healing, repair, and vascular calcification mediated by diverse signaling pathways, there were still a few works that determined its protective roles in ameliorating CRS and asthma, promoting the regeneration of myocardium and liver as well as renal repair, and maintaining periodontal stabilization, these discrepancies are most probably due to differences in animal models. In addition, different roles caused by disparate diseases are likely attributed to different locations, cell types that respond, and pathologic processes of these diseases. Periostin serves functions by diverse signaling pathways such as FAK, Src, NF-κB, p38, ERK, mTOR, JNK, PI3K, Akt, Smad2/3, MAPK, Wnt/β-catenin, to name just a few. Thus, the application of therapies based on periostin function is of great account and creates a favorable outlook for subsequent clinical studies.
It should be noted that loss of the αv integrin as a way of blocking periostin gives play to the majority of undesirable accidents such as prenatal death, colitis, wasting, and autoimmunity [169,170]. We still need to make significant efforts to boost the development of precision medicine through current knowledge and continuous explorations on exact and detailed mechanisms of periostin-involved diseases, despite the journey being full of challenges.
Furthermore, periostin exhibited the potential of acting as a clinically relevant and serviceable biomarker to aid in the diagnosis, speculate on the progression and activity of the disease, inform on prognosis, and direct choice for therapeutic approaches of disease. Periostin, as an attractive and available biomarker for inflammatory diseases, is presently garnering extensive attention. Nevertheless, it must be noticed that the flaw of periostin as a biomarker is that basal expression levels of serum periostin are held high in childhood until bone development halts [171]. Another issue is that periostin isoforms (1 or 2, 3, 4, 5, and 6) and IgA form a complex in serum, which possibly influences the measuring of serum periostin [118].
Different tissues are characterized by heterogeneous expression profiles of periostin isoforms. Currently, the pathological roles of only several periostin isoforms have been displayed, and the functions of each encoded isoform have not been entirely exposed. Further exploration is needed to analyze the functional property of each coding isoform. Moreover, it is of great urgency to develop emerging drugs on the basis of the stimulators or inhibitors affecting periostin expression, periostin itself, the periostin-involved receptors and signaling pathways, or certain periostin isoforms-mediated channels.

Author Contributions

All authors contributed to the study’s conception and design. Article collection and writing of the first draft were performed by L.Y., Y.C. and T.G. The manuscript was revised by K.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Natural Science Foundation of China (grant numbers No. 81871258 to K.B.).

Data Availability Statement

All cited articles in the current study are available in the public database.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

OSF-2Osteoblast-specific factor-2
FAS1Fasciclin I
HDFsHuman dermal fibroblasts
PDLPeriodontal ligament
FAKFocal adhesion kinase
GCFGingival crevicular fluid
AgPAggressive periodontitis
CRSChronic rhinosinusitis
ARAllergic rhinitis
CRSwNPCRS with nasal polyposis
GCsGlucocorticoids
CTComputed tomography
TSTPThymic stromal lymphopoietin
NLFNasal lavage fluids
SLITSublingual immunotherapy
AHAsarum heterotropoides
HNEpCsHuman nasal epithelial cells
AG-ExAngelica gigas extract
CFsCardiac fibroblasts
MIMyocardial infarction
MAPKMitogen-activated protein kinase
CREBCyclic AMP response element-binding protein 1
VSMCsVascular smooth muscle cells
LOXLysyl oxidase
BMP-1Bone morphogenic protein-1
Snhg8Small nucleolar RNA host gene 8
NMCMsNeonatal mouse cardiomyocytes
MSCsMesenchymal stem cells
EAMExperimental autoimmune myocarditis
AFAtrial fibrillation
GSNGelsolin
FMODFibromodulin
MIRIMyocardial ischemia-reperfusion injury
SIMSimvastatin
VHDValvular heart disease
VICsValvular interstitial cells
MMP-2Matrix metalloproteinase-2
ANPAtrial natriuretic peptide
AHRAirway hyperresponsiveness
Th2T-helper type 2
GCMGoblet cell metaplasia
HDMHouse dust mite
DCsDendritic cells
EBCExhaled breath condensate
GLCCI1Glucocorticoid-induced transcript 1
EEEosinophilic esophagitis
ADAtopic dermatitis
TNFSF14TNF superfamily member 14
NPPBNatriuretic polypeptide B
BPBullous pemphigoid
SDStasis dermatitis
IBDInflammatory bowel disease
UCUlcerative colitis
CDCrohn’s disease
pIBDPediatric IBD
IECsIntestinal epithelial cells
PHPulmonary hypertension
AACAscending aortic constriction
RARheumatoid Arthritis
OAOsteoarthritis
ACLAnterior cruciate ligament
NOS2Nitric oxide synthase-2
DDR1Discoidin domain receptor-1
SFSynovial fluid
ASAnkylosing Spondylitis
CtskCathepsin K
BMSCsBone marrow stromal cells
OVXOvariectomized
BMDBone mineral density
PMWPostmenopausal women
K-PostnCathepsin K-generated periostin
PHPTPrimary hyperparathyroidism
DDHDevelopmental dysplasia of the hip
IVDDIntervertebral disc degeneration
NPCsNucleus pulposus cells
ACAtopic conjunctivitis
AKCAtopic keratoconjunctivitis
FADSFacial atopic dermatitis with scratching
PVRProliferative vitreoretinopathy
FVMFibrovascular membrane
NVNeovascularization
IPFIdiopathic pulmonary fibrosis
LRP1Low-density lipoprotein receptor-related protein 1
hLFHuman lung fibroblasts
BALFBronchoalveolar lavage fluid
EPEosinophilic pneumonia
CACCoronary artery calcification
PPARPeroxisome proliferation-activated receptor
EBIEarly Brain Injury
BBBBlood-brain barrier
SAHSubarachnoid hemorrhage
TLR4Toll-like receptor 4
DEXDexamethasone
ASOsAntisense oligonucleotides
HSCsHepatic stellate cells
LOXLLysyl oxidase-like
NAFLDNonalcoholic fatty liver disease
CKDChronic kidney disease
UIRIUnilateral ischemia-reperfusion injury
AKIAcute kidney injury
mTORC1mTOR complex-1
ILKIntegrin-linked kinase
ADPKDAutosomal dominant polycystic kidney disease
PAPeriostin-binding DNA aptamer
IFTAInterstitial fibrosis/tubular atrophy
MCsMesangial cells
IgANImmunoglobulin A nephropathy
DTDistal tubule
FSP-1Fibroblast specific protein-1
RAASRenin-angiotensin-aldosterone system
PDGF-BBPlatelet-derived growth factor-BB
PCNAProliferating cell nuclear antigen
MMCsMouse mesangial cells

References

  1. Takeshita, S.; Kikuno, R.; Tezuka, K.; Amann, E. Osteoblast-specific factor 2: Cloning of a putative bone adhesion protein with homology with the insect protein fasciclin I. Biochem. J. 1993, 294, 271–278. [Google Scholar] [CrossRef] [Green Version]
  2. Horiuchi, K.; Amizuka, N.; Takeshita, S.; Takamatsu, H.; Katsuura, M.; Ozawa, H.; Toyama, Y.; Bonewald, L.F.; Kudo, A. Identification and characterization of a novel protein, periostin, with restricted expression to periosteum and periodontal ligament and increased expression by transforming growth factor beta. J. Bone Miner. Res. 1999, 14, 1239–1249. [Google Scholar] [CrossRef]
  3. Doliana, R.; Bot, S.; Bonaldo, P.; Colombatti, A. EMI, a novel cysteine-rich domain of EMILINs and other extracellular proteins, interacts with the gC1q domains and participates in multimerization. FEBS Lett. 2000, 484, 164–168. [Google Scholar] [CrossRef] [PubMed]
  4. Kii, I.; Nishiyama, T.; Li, M.; Matsumoto, K.; Saito, M.; Amizuka, N.; Kudo, A. Incorporation of tenascin-C into the extracellular matrix by periostin underlies an extracellular meshwork architecture. J. Biol. Chem. 2010, 285, 2028–2039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Kühn, B.; del Monte, F.; Hajjar, R.J.; Chang, Y.S.; Lebeche, D.; Arab, S.; Keating, M.T. Periostin induces proliferation of differentiated cardiomyocytes and promotes cardiac repair. Nat. Med. 2007, 13, 962–969. [Google Scholar] [CrossRef] [PubMed]
  6. Utispan, K.; Sonongbua, J.; Thuwajit, P.; Chau-In, S.; Pairojkul, C.; Wongkham, S.; Thuwajit, C. Periostin activates integrin α5β1 through a PI3K/AKT-dependent pathway in invasion of cholangiocarcinoma. Int. J. Oncol. 2012, 41, 1110–1118. [Google Scholar] [CrossRef] [Green Version]
  7. Lu, Y.; Liu, X.; Jiao, Y.; Xiong, X.; Wang, E.; Wang, X.; Zhang, Z.; Zhang, H.; Pan, L.; Guan, Y.; et al. Periostin promotes liver steatosis and hypertriglyceridemia through downregulation of PPARα. J. Clin. Invest. 2014, 124, 3501–3513. [Google Scholar] [CrossRef] [Green Version]
  8. Mosher, D.F.; Johansson, M.W.; Gillis, M.E.; Annis, D.S. Periostin and TGF-β-induced protein: Two peas in a pod? Crit. Rev. Biochem. Mol. Biol. 2015, 50, 427–439. [Google Scholar] [CrossRef]
  9. Ratajczak-Wielgomas, K.; Kmiecik, A.; Dziegiel, P. Role of Periostin Expression in Non-Small Cell Lung Cancer: Periostin Silencing Inhibits the Migration and Invasion of Lung Cancer Cells via Regulation of MMP-2 Expression. Int. J. Mol. Sci. 2022, 23, 1240. [Google Scholar] [CrossRef]
  10. Norris, R.A.; Moreno-Rodriguez, R.A.; Sugi, Y.; Hoffman, S.; Amos, J.; Hart, M.M.; Potts, J.D.; Goodwin, R.L.; Markwald, R.R. Periostin regulates atrioventricular valve maturation. Dev. Biol. 2008, 316, 200–213. [Google Scholar] [CrossRef]
  11. Liu, L.; Kawakita, F.; Fujimoto, M.; Nakano, F.; Imanaka-Yoshida, K.; Yoshida, T.; Suzuki, H. Role of Periostin in Early Brain Injury After Subarachnoid Hemorrhage in Mice. Stroke 2017, 48, 1108–1111. [Google Scholar] [CrossRef] [PubMed]
  12. Okada, T.; Kawakita, F.; Nishikawa, H.; Nakano, F.; Liu, L.; Suzuki, H. Selective Toll-Like Receptor 4 Antagonists Prevent Acute Blood-Brain Barrier Disruption After Subarachnoid Hemorrhage in Mice. Mol. Neurobiol. 2019, 56, 976–985. [Google Scholar] [CrossRef] [PubMed]
  13. Kanamaru, H.; Kawakita, F.; Nishikawa, H.; Nakano, F.; Asada, R.; Suzuki, H. Clarithromycin Ameliorates Early Brain Injury After Subarachnoid Hemorrhage via Suppressing Periostin-Related Pathways in Mice. Neurotherapeutics 2021, 18, 1880–1890. [Google Scholar] [CrossRef]
  14. Ambrosius, W.; Michalak, S.; Kazmierski, R.; Lukasik, M.; Andrzejewska, N.; Kozubski, W. The Association between Serum Matricellular Protein: Secreted Protein Acidic and Rich in Cysteine-Like 1 Levels and Ischemic Stroke Severity. J. Stroke Cerebrovasc. Dis. 2018, 27, 682–685. [Google Scholar] [CrossRef] [PubMed]
  15. Asada, Y.; Okano, M.; Ishida, W.; Iwamoto, S.; Fukuda, K.; Hirakata, T.; Tada, N.; Fukushima, A.; Ebihara, N.; Kudo, A.; et al. Periostin deletion suppresses late-phase response in mouse experimental allergic conjunctivitis. Allergol. Int. 2019, 68, 233–239. [Google Scholar] [CrossRef] [PubMed]
  16. Nunomura, S.; Kitajima, I.; Nanri, Y.; Kitajima, M.; Ejiri, N.; Lai, I.S.; Okada, N.; Izuhara, K. The FADS mouse: A novel mouse model of atopic keratoconjunctivitis. J. Allergy Clin. Immunol. 2021, 148, 1596–1602. [Google Scholar] [CrossRef] [PubMed]
  17. Fujishima, H.; Okada, N.; Matsumoto, K.; Fukagawa, K.; Igarashi, A.; Matsuda, A.; Ono, J.; Ohta, S.; Mukai, H.; Yoshikawa, M.; et al. The usefulness of measuring tear periostin for the diagnosis and management of ocular allergic diseases. J. Allergy Clin. Immunol. 2016, 138, 459–467.e452. [Google Scholar] [CrossRef] [Green Version]
  18. Jiang, B.; Shi, C.S. Dynamic changes of periostin and collagen I in the sclera during progressive myopia in guinea pigs. Arq. Bras. Oftalmol. 2020, 83, 190–195. [Google Scholar] [CrossRef]
  19. Indumathi, A.; Senthilkumar, G.P.; Jayashree, K.; Ramesh Babu, K. Assessment of circulating fibrotic proteins (periostin and tenascin -C) In Type 2 diabetes mellitus patients with and without retinopathy. Endocrine 2022, 76, 570–577. [Google Scholar] [CrossRef]
  20. Ebenezer, J.A.; Christensen, J.M.; Oliver, B.G.; Oliver, R.A.; Tjin, G.; Ho, J.; Habib, A.R.; Rimmer, J.; Sacks, R.; Harvey, R.J. Periostin as a marker of mucosal remodelling in chronic rhinosinusitis. Rhinology 2017, 55, 234–241. [Google Scholar] [CrossRef]
  21. Hur, D.G.; Khalmuratova, R.; Ahn, S.K.; Ha, Y.S.; Min, Y.G. Roles of periostin in symptom manifestation and airway remodeling in a murine model of allergic rhinitis. Allergy Asthma. Immunol. Res. 2012, 4, 222–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. De Schryver, E.; Derycke, L.; Calus, L.; Holtappels, G.; Hellings, P.W.; Van Zele, T.; Bachert, C.; Gevaert, P. The effect of systemic treatments on periostin expression reflects their interference with the eosinophilic inflammation in chronic rhinosinusitis with nasal polyps. Rhinology 2017, 55, 152–160. [Google Scholar] [CrossRef] [PubMed]
  23. Hoshino, M.; Akitsu, K.; Kubota, K.; Ohtawa, J. Serum Periostin as a Biomarker for Predicting Clinical Response to House Dust Mite Sublingual Immunotherapy in Allergic Rhinitis. J. Allergy Clin. Immunol. Pract. 2021, 9, 1864–1870. [Google Scholar] [CrossRef] [PubMed]
  24. Du, K.; Wang, M.; Zhang, N.; Yu, P.; Wang, P.; Li, Y.; Wang, X.; Zhang, L.; Bachert, C. Involvement of the extracellular matrix proteins periostin and tenascin C in nasal polyp remodeling by regulating the expression of MMPs. Clin. Transl. Allergy 2021, 11, e12059. [Google Scholar] [CrossRef] [PubMed]
  25. Kim, S.W.; Kim, J.H.; Jung, M.H.; Hur, D.G.; Lee, H.K.; Jeon, S.Y.; Kim, D.W. Periostin may play a protective role in the development of eosinophilic chronic rhinosinusitis with nasal polyps in a mouse model. Laryngoscope 2013, 123, 1075–1081. [Google Scholar] [CrossRef]
  26. Hamilton, J.D.; Harel, S.; Swanson, B.N.; Brian, W.; Chen, Z.; Rice, M.S.; Amin, N.; Ardeleanu, M.; Radin, A.; Shumel, B.; et al. Dupilumab suppresses type 2 inflammatory biomarkers across multiple atopic, allergic diseases. Clin. Exp. Allergy 2021, 51, 915–931. [Google Scholar] [CrossRef]
  27. Chung, Y.W.; Cha, J.; Han, S.; Chen, Y.; Gucek, M.; Cho, H.J.; Nakahira, K.; Choi, A.M.K.; Ryu, J.H.; Yoon, J.H. Apolipoprotein E and Periostin Are Potential Biomarkers of Nasal Mucosal Inflammation. A Parallel Approach of In Vitro and In Vivo Secretomes. Am. J. Respir. Cell Mol. Biol. 2020, 62, 23–34. [Google Scholar] [CrossRef]
  28. Yang, H.W.; Park, J.H.; Shin, J.M.; Lee, H.M. Glucocorticoids ameliorate periostin-induced tissue remodeling in chronic rhinosinusitis with nasal polyps. Clin. Exp. Allergy 2018. [Google Scholar] [CrossRef]
  29. Choi, S.; Jung, M.A.; Hwang, Y.H.; Pyun, B.J.; Lee, J.Y.; Jung, D.H.; Ji, K.Y.; Kim, T. Anti-allergic effects of Asarum heterotropoides on an ovalbumin-induced allergic rhinitis murine model. Biomed. Pharmacother. 2021, 141, 111944. [Google Scholar] [CrossRef] [PubMed]
  30. Ji, K.Y.; Jung, D.H.; Pyun, B.J.; Kim, Y.J.; Lee, J.Y.; Choi, S.; Jung, M.A.; Song, K.H.; Kim, T. Angelica gigas extract ameliorates allergic rhinitis in an ovalbumin-induced mouse model by inhibiting Th2 cell activation. Phytomedicine 2021, 93, 153789. [Google Scholar] [CrossRef]
  31. Kim, S.S.; Nikoloudaki, G.E.; Michelsons, S.; Creber, K.; Hamilton, D.W. Fibronectin synthesis, but not α-smooth muscle expression, is regulated by periostin in gingival healing through FAK/JNK signaling. Sci. Rep. 2019, 9, 2708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Balli, U.; Keles, Z.P.; Avci, B.; Guler, S.; Cetinkaya, B.O.; Keles, G.C. Assessment of periostin levels in serum and gingival crevicular fluid of patients with periodontal disease. J. Periodontal. Res. 2015, 50, 707–713. [Google Scholar] [CrossRef]
  33. Aral, C.A.; Köseoğlu, S.; Sağlam, M.; Pekbağrıyanık, T.; Savran, L. Gingival Crevicular Fluid and Salivary Periostin Levels in Non-Smoker Subjects With Chronic and Aggressive Periodontitis : Periostin Levels in Chronic and Aggressive Periodontitis. Inflammation 2016, 39, 986–993. [Google Scholar] [CrossRef] [PubMed]
  34. Padial-Molina, M.; Volk, S.L.; Rios, H.F. Periostin increases migration and proliferation of human periodontal ligament fibroblasts challenged by tumor necrosis factor -α and Porphyromonas gingivalis lipopolysaccharides. J. Periodontal. Res. 2014, 49, 405–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Tang, Y.; Liu, L.; Wang, P.; Chen, D.; Wu, Z.; Tang, C. Periostin promotes migration and osteogenic differentiation of human periodontal ligament mesenchymal stem cells via the Jun amino-terminal kinases (JNK) pathway under inflammatory conditions. Cell Prolif. 2017, 50, e12369. [Google Scholar] [CrossRef] [Green Version]
  36. Wilson, S.R.; Thé, L.; Batia, L.M.; Beattie, K.; Katibah, G.E.; McClain, S.P.; Pellegrino, M.; Estandian, D.M.; Bautista, D.M. The epithelial cell-derived atopic dermatitis cytokine TSLP activates neurons to induce itch. Cell 2013, 155, 285–295. [Google Scholar] [CrossRef] [Green Version]
  37. Mitamura, Y.; Nunomura, S.; Nanri, Y.; Ogawa, M.; Yoshihara, T.; Masuoka, M.; Tsuji, G.; Nakahara, T.; Hashimoto-Hachiya, A.; Conway, S.J.; et al. The IL-13/periostin/IL-24 pathway causes epidermal barrier dysfunction in allergic skin inflammation. Allergy 2018, 73, 1881–1891. [Google Scholar] [CrossRef]
  38. Uchi, H.; Yasumatsu, M.; Morino-Koga, S.; Mitoma, C.; Furue, M. Inhibition of aryl hydrocarbon receptor signaling and induction of NRF2-mediated antioxidant activity by cinnamaldehyde in human keratinocytes. J. Dermatol. Sci. 2017, 85, 36–43. [Google Scholar] [CrossRef]
  39. Yang, L.; Serada, S.; Fujimoto, M.; Terao, M.; Kotobuki, Y.; Kitaba, S.; Matsui, S.; Kudo, A.; Naka, T.; Murota, H.; et al. Periostin facilitates skin sclerosis via PI3K/Akt dependent mechanism in a mouse model of scleroderma. PLoS ONE 2012, 7, e41994. [Google Scholar] [CrossRef] [Green Version]
  40. Makino, K.; Makino, T.; Stawski, L.; Mantero, J.C.; Lafyatis, R.; Simms, R.; Trojanowska, M. Blockade of PDGF Receptors by Crenolanib Has Therapeutic Effect in Patient Fibroblasts and in Preclinical Models of Systemic Sclerosis. J. Invest. Dermatol. 2017, 137, 1671–1681. [Google Scholar] [CrossRef]
  41. Vimalathas, P.; Farris, A.; Letner, D.; Deshpande, V.; Yajnik, V.; Shreffler, W.; Garber, J. Integrin αM activation and upregulation on esophageal eosinophils and periostin-mediated eosinophil survival in eosinophilic esophagitis. Immunol. Cell Biol. 2018, 96, 426–438. [Google Scholar] [CrossRef] [PubMed]
  42. Dellon, E.S.; Higgins, L.L.; Beitia, R.; Rusin, S.; Woosley, J.T.; Veerappan, R.; Selitsky, S.R.; Parker, J.S.; Genta, R.M.; Lash, R.H.; et al. Prospective assessment of serum periostin as a biomarker for diagnosis and monitoring of eosinophilic oesophagitis. Aliment. Pharmacol. Ther. 2016, 44, 189–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Bentley, J.K.; Chen, Q.; Hong, J.Y.; Popova, A.P.; Lei, J.; Moore, B.B.; Hershenson, M.B. Periostin is required for maximal airways inflammation and hyperresponsiveness in mice. J. Allergy Clin. Immunol. 2014, 134, 1433–1442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Gallagher, A.; Edwards, M.; Nair, P.; Drew, S.; Vyas, A.; Sharma, R.; Marsden, P.A.; Wang, R.; Evans, D.J. Anti-interleukin-13 and anti-interleukin-4 agents versus placebo, anti-interleukin-5 or anti-immunoglobulin-E agents, for people with asthma. Cochrane Database Syst. Rev. 2021, 10, Cd012929. [Google Scholar] [CrossRef]
  45. Brightling, C.E.; Chanez, P.; Leigh, R.; O’Byrne, P.M.; Korn, S.; She, D.; May, R.D.; Streicher, K.; Ranade, K.; Piper, E. Efficacy and safety of tralokinumab in patients with severe uncontrolled asthma: A randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Respir. Med. 2015, 3, 692–701. [Google Scholar] [CrossRef]
  46. Kanemitsu, Y.; Matsumoto, H.; Izuhara, K.; Tohda, Y.; Kita, H.; Horiguchi, T.; Kuwabara, K.; Tomii, K.; Otsuka, K.; Fujimura, M.; et al. Increased periostin associates with greater airflow limitation in patients receiving inhaled corticosteroids. J. Allergy Clin. Immunol. 2013, 132, 305–312.e303. [Google Scholar] [CrossRef] [Green Version]
  47. Sidhu, S.S.; Yuan, S.; Innes, A.L.; Kerr, S.; Woodruff, P.G.; Hou, L.; Muller, S.J.; Fahy, J.V. Roles of epithelial cell-derived periostin in TGF-beta activation, collagen production, and collagen gel elasticity in asthma. Proc. Natl. Acad. Sci. USA 2010, 107, 14170–14175. [Google Scholar] [CrossRef] [Green Version]
  48. Tajiri, T.; Matsumoto, H.; Gon, Y.; Ito, R.; Hashimoto, S.; Izuhara, K.; Suzukawa, M.; Ohta, K.; Ono, J.; Ohta, S.; et al. Utility of serum periostin and free IgE levels in evaluating responsiveness to omalizumab in patients with severe asthma. Allergy 2016, 71, 1472–1479. [Google Scholar] [CrossRef]
  49. Hachim, M.Y.; Elemam, N.M.; Ramakrishnan, R.K.; Hachim, I.Y.; Salameh, L.; Mahboub, B.; Al Heialy, S.; Halwani, R.; Hamoudi, R.; Hamid, Q. Confounding Patient Factors Affecting the Proper Interpretation of the Periostin Level as a Biomarker in Asthma Development. J. Asthma. Allergy 2020, 13, 23–37. [Google Scholar] [CrossRef] [Green Version]
  50. Xun, Q.; Kuang, J.; Yang, Q.; Wang, W.; Zhu, G. Glucocorticoid induced transcript 1 represses airway remodeling of asthmatic mouse via inhibiting IL-13/periostin/TGF-β1 signaling. Int. Immunopharmacol. 2021, 97, 107637. [Google Scholar] [CrossRef]
  51. Gordon, E.D.; Sidhu, S.S.; Wang, Z.E.; Woodruff, P.G.; Yuan, S.; Solon, M.C.; Conway, S.J.; Huang, X.; Locksley, R.M.; Fahy, J.V. A protective role for periostin and TGF-β in IgE-mediated allergy and airway hyperresponsiveness. Clin. Exp. Allergy 2012, 42, 144–155. [Google Scholar] [CrossRef] [PubMed]
  52. Sehra, S.; Yao, W.; Nguyen, E.T.; Ahyi, A.N.; Tuana, F.M.; Ahlfeld, S.K.; Snider, P.; Tepper, R.S.; Petrache, I.; Conway, S.J.; et al. Periostin regulates goblet cell metaplasia in a model of allergic airway inflammation. J. Immunol. 2011, 186, 4959–4966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Komiya, K.; Ohta, S.; Arima, K.; Ogawa, M.; Suzuki, S.; Mitamura, Y.; Nunomura, S.; Nanri, Y.; Yoshihara, T.; Kawaguchi, A.; et al. Clarithromycin attenuates IL-13-induced periostin production in human lung fibroblasts. Respir. Res. 2017, 18, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Wardzyńska, A.; Makowska, J.S.; Pawełczyk, M.; Piechota-Polańczyk, A.; Kurowski, M.; Kowalski, M.L. Periostin in Exhaled Breath Condensate and in Serum of Asthmatic Patients: Relationship to Upper and Lower Airway Disease. Allergy Asthma. Immunol. Res. 2017, 9, 126–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Refaat, M.M.; El Sayed, E.; Abd El-Fattah, W.; Elbanna, A.H.; Sayed, H.M.E. Relationship between sputum periostin level and inflammatory asthma phenotypes in Egyptian patients. J. Asthma. 2021, 58, 1285–1291. [Google Scholar] [CrossRef]
  56. Yao, L.; Song, J.; Meng, X.W.; Ge, J.Y.; Du, B.X.; Yu, J.; Ji, F.H. Periostin aggravates NLRP3 inflammasome-mediated pyroptosis in myocardial ischemia-reperfusion injury. Mol. Cell Probes. 2020, 53, 101596. [Google Scholar] [CrossRef]
  57. Landry, N.M.; Cohen, S.; Dixon, I.M.C. Periostin in cardiovascular disease and development: A tale of two distinct roles. Basic Res. Cardiol. 2018, 113, 1. [Google Scholar] [CrossRef]
  58. Zhu, Y.; Ji, J.J.; Wang, X.D.; Sun, X.J.; Li, M.; Wei, Q.; Ren, L.Q.; Liu, N.F. Periostin promotes arterial calcification through PPARγ-related glucose metabolism reprogramming. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H2222–H2239. [Google Scholar] [CrossRef]
  59. Shimazaki, M.; Nakamura, K.; Kii, I.; Kashima, T.; Amizuka, N.; Li, M.; Saito, M.; Fukuda, K.; Nishiyama, T.; Kitajima, S.; et al. Periostin is essential for cardiac healing after acute myocardial infarction. J. Exp. Med. 2008, 205, 295–303. [Google Scholar] [CrossRef]
  60. Choi, Y.; Oh, H.; Ahn, M.; Kang, T.; Chun, J.; Shin, T.; Kim, J. Immunohistochemical analysis of periostin in the hearts of Lewis rats with experimental autoimmune myocarditis. J. Vet. Med. Sci. 2020, 82, 1545–1550. [Google Scholar] [CrossRef]
  61. Yuan, C.T.; Li, X.X.; Cheng, Q.J.; Wang, Y.H.; Wang, J.H.; Liu, C.L. MiR-30a regulates the atrial fibrillation-induced myocardial fibrosis by targeting snail 1. Int. J. Clin. Exp. Pathol. 2015, 8, 15527–15536. [Google Scholar] [PubMed]
  62. Jana, S.; Aujla, P.; Hu, M.; Kilic, T.; Zhabyeyev, P.; McCulloch, C.A.; Oudit, G.Y.; Kassiri, Z. Gelsolin is an important mediator of Angiotensin II-induced activation of cardiac fibroblasts and fibrosis. FASEB J. 2021, 35, e21932. [Google Scholar] [CrossRef] [PubMed]
  63. Kuo, H.F.; Hsieh, C.C.; Wang, S.C.; Chang, C.Y.; Hung, C.H.; Kuo, P.L.; Liu, Y.R.; Li, C.Y.; Liu, P.L. Simvastatin Attenuates Cardiac Fibrosis via Regulation of Cardiomyocyte-Derived Exosome Secretion. J. Clin. Med. 2019, 8, 794. [Google Scholar] [CrossRef] [Green Version]
  64. Hakuno, D.; Kimura, N.; Yoshioka, M.; Mukai, M.; Kimura, T.; Okada, Y.; Yozu, R.; Shukunami, C.; Hiraki, Y.; Kudo, A.; et al. Periostin advances atherosclerotic and rheumatic cardiac valve degeneration by inducing angiogenesis and MMP production in humans and rodents. J. Clin. Invest. 2010, 120, 2292–2306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Liao, Y.; Li, G.; Zhang, X.; Huang, W.; Xie, D.; Dai, G.; Zhu, S.; Lu, D.; Zhang, Z.; Lin, J.; et al. Cardiac Nestin(+) Mesenchymal Stromal Cells Enhance Healing of Ischemic Heart through Periostin-Mediated M2 Macrophage Polarization. Mol. Ther. 2020, 28, 855–873. [Google Scholar] [CrossRef]
  66. Hu, Y.; Wang, X.; Ding, F.; Liu, C.; Wang, S.; Feng, T.; Meng, S. Periostin renders cardiomyocytes vulnerable to acute myocardial infarction via pro-apoptosis. ESC Heart Fail. 2022, 9, 977–987. [Google Scholar] [CrossRef]
  67. Sözmen, M.; Devrim, A.K.; Kabak, Y.B.; Devrim, T. Periostin alters transcriptional profile in a rat model of isoproterenol-induced cardiotoxicity. Hum. Exp. Toxicol. 2019, 38, 255–266. [Google Scholar] [CrossRef]
  68. Chen, Z.; Xie, J.; Hao, H.; Lin, H.; Wang, L.; Zhang, Y.; Chen, L.; Cao, S.; Huang, X.; Liao, W.; et al. Ablation of periostin inhibits post-infarction myocardial regeneration in neonatal mice mediated by the phosphatidylinositol 3 kinase/glycogen synthase kinase 3β/cyclin D1 signalling pathway. Cardiovasc. Res. 2017, 113, 620–632. [Google Scholar] [CrossRef] [Green Version]
  69. Wu, H.; Li, G.N.; Xie, J.; Li, R.; Chen, Q.H.; Chen, J.Z.; Wei, Z.H.; Kang, L.N.; Xu, B. Resveratrol ameliorates myocardial fibrosis by inhibiting ROS/ERK/TGF-β/periostin pathway in STZ-induced diabetic mice. BMC Cardiovasc. Disord. 2016, 16, 5. [Google Scholar] [CrossRef] [Green Version]
  70. Wang, D.; Oparil, S.; Feng, J.A.; Li, P.; Perry, G.; Chen, L.B.; Dai, M.; John, S.W.; Chen, Y.F. Effects of pressure overload on extracellular matrix expression in the heart of the atrial natriuretic peptide-null mouse. Hypertension 2003, 42, 88–95. [Google Scholar] [CrossRef]
  71. Ma, W.Q.; Sun, X.J.; Zhu, Y.; Liu, N.F. Metformin attenuates hyperlipidaemia-associated vascular calcification through anti-ferroptotic effects. Free Radic. Biol. Med. 2021, 165, 229–242. [Google Scholar] [CrossRef] [PubMed]
  72. Uchida, M.; Shiraishi, H.; Ohta, S.; Arima, K.; Taniguchi, K.; Suzuki, S.; Okamoto, M.; Ahlfeld, S.K.; Ohshima, K.; Kato, S.; et al. Periostin, a matricellular protein, plays a role in the induction of chemokines in pulmonary fibrosis. Am. J. Respir. Cell Mol. Biol. 2012, 46, 677–686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Nanri, Y.; Nunomura, S.; Terasaki, Y.; Yoshihara, T.; Hirano, Y.; Yokosaki, Y.; Yamaguchi, Y.; Feghali-Bostwick, C.; Ajito, K.; Murakami, S.; et al. Cross-Talk between Transforming Growth Factor-β and Periostin Can Be Targeted for Pulmonary Fibrosis. Am. J. Respir. Cell Mol. Biol. 2020, 62, 204–216. [Google Scholar] [CrossRef]
  74. Carpagnano, G.E.; Soccio, P.; Scioscia, G.; Palladino, G.P.; Foschino Barbaro, M.P.; Lacedonia, D. The Potential Role of Airways Periostin in the Clinical Practice of Patients Affected by Idiopathic Pulmonary Fibrosis. Rejuvenation Res. 2021, 24, 302–306. [Google Scholar] [CrossRef] [PubMed]
  75. Schnieder, J.; Mamazhakypov, A.; Birnhuber, A.; Wilhelm, J.; Kwapiszewska, G.; Ruppert, C.; Markart, P.; Wujak, L.; Rubio, K.; Barreto, G.; et al. Loss of LRP1 promotes acquisition of contractile-myofibroblast phenotype and release of active TGF-β1 from ECM stores. Matrix Biol. 2020, 88, 69–88. [Google Scholar] [CrossRef]
  76. Ashley, S.L.; Wilke, C.A.; Kim, K.K.; Moore, B.B. Periostin regulates fibrocyte function to promote myofibroblast differentiation and lung fibrosis. Mucosal. Immunol. 2017, 10, 341–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Lambert, M.; Mendes-Ferreira, P.; Ghigna, M.R.; LeRibeuz, H.; Adão, R.; Boet, A.; Capuano, V.; Rucker-Martin, C.; Brás-Silva, C.; Quarck, R.; et al. Kcnk3 dysfunction exaggerates the development of pulmonary hypertension induced by left ventricular pressure overload. Cardiovasc. Res. 2021, 117, 2474–2488. [Google Scholar] [CrossRef] [PubMed]
  78. Nie, X.; Shen, C.; Tan, J.; Wu, Z.; Wang, W.; Chen, Y.; Dai, Y.; Yang, X.; Ye, S.; Chen, J.; et al. Periostin: A Potential Therapeutic Target For Pulmonary Hypertension? Circ. Res. 2020, 127, 1138–1152. [Google Scholar] [CrossRef]
  79. Guan, J.; Liu, W.Q.; Xing, M.Q.; Shi, Y.; Tan, X.Y.; Jiang, C.Q.; Dai, H.Y. Elevated expression of periostin in diabetic cardiomyopathy and the effect of valsartan. BMC Cardiovasc. Disord. 2015, 15, 90. [Google Scholar] [CrossRef] [Green Version]
  80. Zeng, H.L.; Chen, D.; Yan, J.; Yang, Q.; Han, Q.Q.; Li, S.S.; Cheng, L. Proteomic characteristics of bronchoalveolar lavage fluid in critical COVID-19 patients. Febs. J. 2021, 288, 5190–5200. [Google Scholar] [CrossRef]
  81. Wan, J.; Shan, Y.; Song, X.; Chen, S.; Lu, X.; Jin, J.; Su, Q.; Liu, B.; Sun, W.; Li, B. Adipocyte-derived Periostin mediates glucocorticoid-induced hepatosteatosis in mice. Mol. Metab. 2020, 31, 24–35. [Google Scholar] [CrossRef]
  82. Kobayashi, T.; Kanno, K.; Nguyen, P.T.; Sugiyama, A.; Kawahara, A.; Otani, Y.; Kishikawa, N.; Ito, M.; Tazuma, S. Periostin antisense oligonucleotide prevents hepatic steatosis and fibrosis in a mouse model of non-alcoholic steatohepatitis. J. Gastroenterol. Hepatol. 2020, 35, 2140–2150. [Google Scholar] [CrossRef]
  83. Wu, T.; Wu, S.; Ouyang, G. Periostin: A new extracellular regulator of obesity-induced hepatosteatosis. Cell Metab. 2014, 20, 562–564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Zhu, J.Z.; Zhu, H.T.; Dai, Y.N.; Li, C.X.; Fang, Z.Y.; Zhao, D.J.; Wan, X.Y.; Wang, Y.M.; Wang, F.; Yu, C.H.; et al. Serum periostin is a potential biomarker for non-alcoholic fatty liver disease: A case-control study. Endocrine 2016, 51, 91–100. [Google Scholar] [CrossRef] [PubMed]
  85. Kumar, P.; Smith, T.; Raeman, R.; Chopyk, D.M.; Brink, H.; Liu, Y.; Sulchek, T.; Anania, F.A. Periostin promotes liver fibrogenesis by activating lysyl oxidase in hepatic stellate cells. J. Biol. Chem. 2018, 293, 12781–12792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Wu, T.; Huang, J.; Wu, S.; Huang, Z.; Chen, X.; Liu, Y.; Cui, D.; Song, G.; Luo, Q.; Liu, F.; et al. Deficiency of periostin impairs liver regeneration in mice after partial hepatectomy. Matrix Biol. 2018, 66, 81–92. [Google Scholar] [CrossRef]
  87. Bian, X.; Bai, Y.; Su, X.; Zhao, G.; Sun, G.; Li, D. Knockdown of periostin attenuates 5/6 nephrectomy-induced intrarenal renin-angiotensin system activation, fibrosis, and inflammation in rats. J. Cell Physiol. 2019, 234, 22857–22873. [Google Scholar] [CrossRef]
  88. Prakoura, N.; Kavvadas, P.; Kormann, R.; Dussaule, J.C.; Chadjichristos, C.E.; Chatziantoniou, C. NFκB-Induced Periostin Activates Integrin-β3 Signaling to Promote Renal Injury in GN. J. Am. Soc. Nephrol. 2017, 28, 1475–1490. [Google Scholar] [CrossRef] [Green Version]
  89. Zhang, Y.; Reif, G.; Wallace, D.P. Extracellular matrix, integrins, and focal adhesion signaling in polycystic kidney disease. Cell Signal. 2020, 72, 109646. [Google Scholar] [CrossRef]
  90. Guerrot, D.; Dussaule, J.C.; Mael-Ainin, M.; Xu-Dubois, Y.C.; Rondeau, E.; Chatziantoniou, C.; Placier, S. Identification of periostin as a critical marker of progression/reversal of hypertensive nephropathy. PLoS ONE 2012, 7, e31974. [Google Scholar] [CrossRef]
  91. Satirapoj, B.; Wang, Y.; Chamberlin, M.P.; Dai, T.; LaPage, J.; Phillips, L.; Nast, C.C.; Adler, S.G. Periostin: Novel tissue and urinary biomarker of progressive renal injury induces a coordinated mesenchymal phenotype in tubular cells. Nephrol. Dial. Transplant. 2012, 27, 2702–2711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Mael-Ainin, M.; Abed, A.; Conway, S.J.; Dussaule, J.C.; Chatziantoniou, C. Inhibition of periostin expression protects against the development of renal inflammation and fibrosis. J. Am. Soc. Nephrol. 2014, 25, 1724–1736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. An, J.N.; Yang, S.H.; Kim, Y.C.; Hwang, J.H.; Park, J.Y.; Kim, D.K.; Kim, J.H.; Kim, D.W.; Hur, D.G.; Oh, Y.K.; et al. Periostin induces kidney fibrosis after acute kidney injury via the p38 MAPK pathway. Am. J. Physiol. Renal. Physiol. 2019, 316, F426–F437. [Google Scholar] [CrossRef] [PubMed]
  94. Alesutan, I.; Henze, L.A.; Boehme, B.; Luong, T.T.D.; Zickler, D.; Pieske, B.; Eckardt, K.U.; Pasch, A.; Voelkl, J. Periostin Augments Vascular Smooth Muscle Cell Calcification via β-Catenin Signaling. Biomolecules 2022, 12, 1157. [Google Scholar] [CrossRef]
  95. Kormann, R.; Kavvadas, P.; Placier, S.; Vandermeersch, S.; Dorison, A.; Dussaule, J.C.; Chadjichristos, C.E.; Prakoura, N.; Chatziantoniou, C. Periostin Promotes Cell Proliferation and Macrophage Polarization to Drive Repair after AKI. J. Am. Soc. Nephrol. 2020, 31, 85–100. [Google Scholar] [CrossRef]
  96. Wu, J.; Lin, Q.; Li, S.; Shao, X.; Zhu, X.; Zhang, M.; Zhou, W.; Ni, Z. Periostin Contributes to Immunoglobulin a Nephropathy by Promoting the Proliferation of Mesangial Cells: A Weighted Gene Correlation Network Analysis. Front. Genet. 2020, 11, 595757. [Google Scholar] [CrossRef]
  97. El-Dawla, N.M.Q.; Sallam, A.M.; El-Hefnawy, M.H.; El-Mesallamy, H.O. E-cadherin and periostin in early detection and progression of diabetic nephropathy: Epithelial-to-mesenchymal transition. Clin. Exp. Nephrol. 2019, 23, 1050–1057. [Google Scholar] [CrossRef]
  98. Koh, S.J.; Choi, Y.; Kim, B.G.; Lee, K.L.; Kim, D.W.; Kim, J.H.; Kim, J.W.; Kim, J.S. Matricellular Protein Periostin Mediates Intestinal Inflammation through the Activation of Nuclear Factor κB Signaling. PLoS ONE 2016, 11, e0149652. [Google Scholar] [CrossRef]
  99. Coelho, T.; Sonnenberg-Riethmacher, E.; Gao, Y.; Mossotto, E.; Khojanazarov, A.; Griffin, A.; Mukanova, S.; Ashimkhanova, A.; Haggarty, R.; Borissenko, A.; et al. Expression profile of the matricellular protein periostin in paediatric inflammatory bowel disease. Sci. Rep. 2021, 11, 6194. [Google Scholar] [CrossRef]
  100. Keskin, M.; Topkaç, A. The Predictive Value of Periostin to Diagnose Crohn’s Disease. Turk. J. Gastroenterol. 2022, 33, 127–135. [Google Scholar] [CrossRef]
  101. Bonnet, N.; Standley, K.N.; Bianchi, E.N.; Stadelmann, V.; Foti, M.; Conway, S.J.; Ferrari, S.L. The matricellular protein periostin is required for sost inhibition and the anabolic response to mechanical loading and physical activity. J. Biol. Chem. 2009, 284, 35939–35950. [Google Scholar] [CrossRef] [Green Version]
  102. Li, C.; Li, X.; Wang, X.; Miao, P.; Liu, J.; Li, C.; Li, D.; Zhou, W.; Jin, Z.; Cao, M. Periostin Mediates Oestrogen-Induced Osteogenic Differentiation of Bone Marrow Stromal Cells in Ovariectomised Rats. Biomed. Res. Int. 2020, 2020, 9405909. [Google Scholar] [CrossRef]
  103. Liu, S.; Jin, Z.; Cao, M.; Hao, D.; Li, C.; Li, D.; Zhou, W. Periostin regulates osteogenesis of mesenchymal stem cells from ovariectomized rats through actions on the ILK/Akt/GSK-3β Axis. Genet. Mol. Biol. 2021, 44, e20200461. [Google Scholar] [CrossRef] [PubMed]
  104. Kerschan-Schindl, K.; Ebenbichler, G.; Föeger-Samwald, U.; Leiss, H.; Gesslbauer, C.; Herceg, M.; Stummvoll, G.; Marculescu, R.; Crevenna, R.; Pietschmann, P. Rheumatoid arthritis in remission : Decreased myostatin and increased serum levels of periostin. Wien. Klin. Wochenschr. 2019, 131, 1–7. [Google Scholar] [CrossRef] [Green Version]
  105. Chijimatsu, R.; Kunugiza, Y.; Taniyama, Y.; Nakamura, N.; Tomita, T.; Yoshikawa, H. Expression and pathological effects of periostin in human osteoarthritis cartilage. BMC Musculoskelet. Disord. 2015, 16, 215. [Google Scholar] [CrossRef] [Green Version]
  106. Han, T.; Mignatti, P.; Abramson, S.B.; Attur, M. Periostin interaction with discoidin domain receptor-1 (DDR1) promotes cartilage degeneration. PLoS ONE 2020, 15, e0231501. [Google Scholar] [CrossRef]
  107. Nakamura, Y.; Saitou, M.; Komura, S.; Matsumoto, K.; Ogawa, H.; Miyagawa, T.; Saitou, T.; Imamura, T.; Imai, Y.; Takayanagi, H.; et al. Reduced dynamic loads due to hip dislocation induce acetabular cartilage degeneration by IL-6 and MMP3 via the STAT3/periostin/NF-κB axis. Sci. Rep. 2022, 12, 12207. [Google Scholar] [CrossRef]
  108. Zhu, D.; Wang, Z.; Zhang, G.; Ma, C.; Qiu, X.; Wang, Y.; Liu, M.; Guo, X.; Chen, H.; Deng, Q.; et al. Periostin promotes nucleus pulposus cells apoptosis by activating the Wnt/β-catenin signaling pathway. FASEB J. 2022, 36, e22369. [Google Scholar] [CrossRef] [PubMed]
  109. Tajika, Y.; Moue, T.; Ishikawa, S.; Asano, K.; Okumo, T.; Takagi, H.; Hisamitsu, T. Influence of Periostin on Synoviocytes in Knee Osteoarthritis. In Vivo 2017, 31, 69–77. [Google Scholar] [CrossRef] [Green Version]
  110. Pepe, J.; Bonnet, N.; Cipriani, C.; Ferrara, C.; Rossi, M.; De Martino, V.; Colangelo, L.; Sonato, C.; Cecchetti, V.; Ferrari, S.; et al. Higher serum levels of a cathepsin K-generated periostin fragment are associated with fractures in postmenopausal women with primary hyperparathyroidism: A pilot study. Osteoporos. Int. 2021, 32, 2365–2369. [Google Scholar] [CrossRef]
  111. Shimamura, M.; Taniyama, Y.; Katsuragi, N.; Koibuchi, N.; Kyutoku, M.; Sato, N.; Allahtavakoli, M.; Wakayama, K.; Nakagami, H.; Morishita, R. Role of central nervous system periostin in cerebral ischemia. Stroke 2012, 43, 1108–1114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Takayama, G.; Arima, K.; Kanaji, T.; Toda, S.; Tanaka, H.; Shoji, S.; McKenzie, A.N.; Nagai, H.; Hotokebuchi, T.; Izuhara, K. Periostin: A novel component of subepithelial fibrosis of bronchial asthma downstream of IL-4 and IL-13 signals. J. Allergy Clin. Immunol. 2006, 118, 98–104. [Google Scholar] [CrossRef] [PubMed]
  113. Johansson, M.W.; Annis, D.S.; Mosher, D.F. α(M)β(2) integrin-mediated adhesion and motility of IL-5-stimulated eosinophils on periostin. Am. J. Respir. Cell Mol. Biol. 2013, 48, 503–510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Taniyama, Y.; Katsuragi, N.; Sanada, F.; Azuma, J.; Iekushi, K.; Koibuchi, N.; Okayama, K.; Ikeda-Iwabu, Y.; Muratsu, J.; Otsu, R.; et al. Selective Blockade of Periostin Exon 17 Preserves Cardiac Performance in Acute Myocardial Infarction. Hypertension 2016, 67, 356–361. [Google Scholar] [CrossRef]
  115. Nance, T.; Smith, K.S.; Anaya, V.; Richardson, R.; Ho, L.; Pala, M.; Mostafavi, S.; Battle, A.; Feghali-Bostwick, C.; Rosen, G.; et al. Transcriptome analysis reveals differential splicing events in IPF lung tissue. PLoS ONE 2014, 9, e92111. [Google Scholar] [CrossRef] [Green Version]
  116. Nakama, T.; Yoshida, S.; Ishikawa, K.; Kobayashi, Y.; Abe, T.; Kiyonari, H.; Shioi, G.; Katsuragi, N.; Ishibashi, T.; Morishita, R.; et al. Different roles played by periostin splice variants in retinal neovascularization. Exp. Eye Res. 2016, 153, 133–140. [Google Scholar] [CrossRef] [PubMed]
  117. Cai, L.; Brophy, R.H.; Tycksen, E.D.; Duan, X.; Nunley, R.M.; Rai, M.F. Distinct expression pattern of periostin splice variants in chondrocytes and ligament progenitor cells. FASEB J. 2019, 33, 8386–8405. [Google Scholar] [CrossRef] [PubMed]
  118. Ono, J.; Takai, M.; Kamei, A.; Nunomura, S.; Nanri, Y.; Yoshihara, T.; Ohta, S.; Yasuda, K.; Conway, S.J.; Yokosaki, Y.; et al. Periostin forms a functional complex with IgA in human serum. Allergol. Int. 2020, 69, 111–120. [Google Scholar] [CrossRef] [PubMed]
  119. Norris, R.A.; Damon, B.; Mironov, V.; Kasyanov, V.; Ramamurthi, A.; Moreno-Rodriguez, R.; Trusk, T.; Potts, J.D.; Goodwin, R.L.; Davis, J.; et al. Periostin regulates collagen fibrillogenesis and the biomechanical properties of connective tissues. J. Cell Biochem. 2007, 101, 695–711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Choi, J.W.; Arai, C.; Ishikawa, M.; Shimoda, S.; Nakamura, Y. Fiber system degradation, and periostin and connective tissue growth factor level reduction, in the periodontal ligament of teeth in the absence of masticatory load. J. Periodontal. Res. 2011, 46, 513–521. [Google Scholar] [CrossRef] [PubMed]
  121. Maruhashi, T.; Kii, I.; Saito, M.; Kudo, A. Interaction between periostin and BMP-1 promotes proteolytic activation of lysyl oxidase. J. Biol. Chem. 2010, 285, 13294–13303. [Google Scholar] [CrossRef] [Green Version]
  122. Maeda, D.; Kubo, T.; Kiya, K.; Kawai, K.; Matsuzaki, S.; Kobayashi, D.; Fujiwara, T.; Katayama, T.; Hosokawa, K. Periostin is induced by IL-4/IL-13 in dermal fibroblasts and promotes RhoA/ROCK pathway-mediated TGF-β1 secretion in abnormal scar formation. J. Plast. Surg. Hand Surg. 2019, 53, 288–294. [Google Scholar] [CrossRef] [PubMed]
  123. Li, L.; Fan, D.; Wang, C.; Wang, J.Y.; Cui, X.B.; Wu, D.; Zhou, Y.; Wu, L.L. Angiotensin II increases periostin expression via Ras/p38 MAPK/CREB and ERK1/2/TGF-β1 pathways in cardiac fibroblasts. Cardiovasc. Res. 2011, 91, 80–89. [Google Scholar] [CrossRef]
  124. Kanisicak, O.; Khalil, H.; Ivey, M.J.; Karch, J.; Maliken, B.D.; Correll, R.N.; Brody, M.J.; SC, J.L.; Aronow, B.J.; Tallquist, M.D.; et al. Genetic lineage tracing defines myofibroblast origin and function in the injured heart. Nat. Commun. 2016, 7, 12260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Xue, K.; Chen, S.; Chai, J.; Yan, W.; Zhu, X.; Dai, H.; Wang, W. Upregulation of Periostin Through CREB Participates in Myocardial Infarction-induced Myocardial Fibrosis. J. Cardiovasc. Pharmacol. 2022, 79, 687–697. [Google Scholar] [CrossRef] [PubMed]
  126. Ma, Y.; Iyer, R.P.; Jung, M.; Czubryt, M.P.; Lindsey, M.L. Cardiac Fibroblast Activation Post-Myocardial Infarction: Current Knowledge Gaps. Trends Pharmacol. Sci. 2017, 38, 448–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Snider, P.; Hinton, R.B.; Moreno-Rodriguez, R.A.; Wang, J.; Rogers, R.; Lindsley, A.; Li, F.; Ingram, D.A.; Menick, D.; Field, L.; et al. Periostin is required for maturation and extracellular matrix stabilization of noncardiomyocyte lineages of the heart. Circ. Res. 2008, 102, 752–760. [Google Scholar] [CrossRef]
  128. Drasutiene, A.; Chitwood, W.R.; Rucinskas, K.; Aidietis, A. Reoperative, transapical off-pump neochordae implantation for recurrent degenerative mitral regurgitation resulting from a newly ruptured native chord. Eur. J. Cardiothorac. Surg. 2020, 58, 648–650. [Google Scholar] [CrossRef]
  129. Wu, H.; Xie, J.; Li, G.N.; Chen, Q.H.; Li, R.; Zhang, X.L.; Kang, L.N.; Xu, B. Possible involvement of TGF-β/periostin in fibrosis of right atrial appendages in patients with atrial fibrillation. Int. J. Clin. Exp. Pathol. 2015, 8, 6859–6869. [Google Scholar] [PubMed]
  130. Horne, T.E.; VandeKopple, M.; Sauls, K.; Koenig, S.N.; Anstine, L.J.; Garg, V.; Norris, R.A.; Lincoln, J. Dynamic Heterogeneity of the Heart Valve Interstitial Cell Population in Mitral Valve Health and Disease. J. Cardiovasc. Dev. Dis. 2015, 2, 214–232. [Google Scholar] [CrossRef] [PubMed]
  131. Tkatchenko, T.V.; Moreno-Rodriguez, R.A.; Conway, S.J.; Molkentin, J.D.; Markwald, R.R.; Tkatchenko, A.V. Lack of periostin leads to suppression of Notch1 signaling and calcific aortic valve disease. Physiol. Genomics. 2009, 39, 160–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Wu, H.; Chen, L.; Xie, J.; Li, R.; Li, G.N.; Chen, Q.H.; Zhang, X.L.; Kang, L.N.; Xu, B. Periostin expression induced by oxidative stress contributes to myocardial fibrosis in a rat model of high salt-induced hypertension. Mol. Med. Rep. 2016, 14, 776–782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Lindner, V.; Wang, Q.; Conley, B.A.; Friesel, R.E.; Vary, C.P. Vascular injury induces expression of periostin: Implications for vascular cell differentiation and migration. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 77–83. [Google Scholar] [CrossRef] [PubMed]
  134. Yoshida, S.; Nakama, T.; Ishikawa, K.; Nakao, S.; Sonoda, K.H.; Ishibashi, T. Periostin in vitreoretinal diseases. Cell Mol. Life Sci. 2017, 74, 4329–4337. [Google Scholar] [CrossRef]
  135. Qian, L.; Shujuan, G.; Ping, H.; Li, L.; Weiwei, S.; Yafei, W.; Weidong, T. Wnt5a up-regulates Periostin through CaMKII pathway to influence periodontal tissue destruction in early periodontitis. J. Mol. Histol. 2021, 52, 555–566. [Google Scholar] [CrossRef]
  136. Nakajima, M.; Honda, T.; Miyauchi, S.; Yamazaki, K. Th2 cytokines efficiently stimulate periostin production in gingival fibroblasts but periostin does not induce an inflammatory response in gingival epithelial cells. Arch. Oral. Biol. 2014, 59, 93–101. [Google Scholar] [CrossRef]
  137. Platt, M.P.; Soler, Z.M.; Kao, S.Y.; Metson, R.; Stankovic, K.M. Topographic gene expression in the sinonasal cavity of patients with chronic sinusitis with polyps. Otolaryngol. Head Neck Surg. 2011, 145, 171–175. [Google Scholar] [CrossRef] [Green Version]
  138. Miłoński, J.; Zielińska-Bliźniewska, H.; Przybyłowska, K.; Pietkiewicz, P.; Korzycka-Zaborowska, B.; Majsterek, I.; Olszewski, J. Significance of CYCLOOXYGENASE-2(COX-2), PERIOSTIN (POSTN) and INTERLEUKIN-4(IL-4) gene expression in the pathogenesis of chronic rhinosinusitis with nasal polyps. Eur. Arch. Otorhinolaryngol. 2015, 272, 3715–3720. [Google Scholar] [CrossRef] [Green Version]
  139. Wang, M.; Wang, X.; Zhang, N.; Wang, H.; Li, Y.; Fan, E.; Zhang, L.; Zhang, L.; Bachert, C. Association of periostin expression with eosinophilic inflammation in nasal polyps. J. Allergy Clin. Immunol. 2015, 136, 1700–1703.e1709. [Google Scholar] [CrossRef] [Green Version]
  140. Seshadri, S.; Lu, X.; Purkey, M.R.; Homma, T.; Choi, A.W.; Carter, R.; Suh, L.; Norton, J.; Harris, K.E.; Conley, D.B.; et al. Increased expression of the epithelial anion transporter pendrin/SLC26A4 in nasal polyps of patients with chronic rhinosinusitis. J. Allergy Clin. Immunol. 2015, 136, 1548–1558.e1547. [Google Scholar] [CrossRef]
  141. Maxfield, A.Z.; Landegger, L.D.; Brook, C.D.; Lehmann, A.E.; Campbell, A.P.; Bergmark, R.W.; Stankovic, K.M.; Metson, R. Periostin as a Biomarker for Nasal Polyps in Chronic Rhinosinusitis. Otolaryngol. Head Neck Surg. 2018, 158, 181–186. [Google Scholar] [CrossRef] [PubMed]
  142. Wang, L.; Chen, M.; Xu, M. Effect of posterior nasal neurectomy on the suppression of allergic rhinitis. Am. J. Otolaryngol. 2020, 41, 102410. [Google Scholar] [CrossRef] [PubMed]
  143. Rodrigo-Muñoz, J.M.; Cañas, J.A.; Sastre, B.; Gil-Martínez, M.; García Latorre, R.; Sastre, J.; Del Pozo, V. Role of miR-185-5p as modulator of periostin synthesis and smooth muscle contraction in asthma. J. Cell Physiol. 2022, 237, 1498–1508. [Google Scholar] [CrossRef] [PubMed]
  144. Zhang, K.; Liang, Y.; Feng, Y.; Wu, W.; Zhang, H.; He, J.; Hu, Q.; Zhao, J.; Xu, Y.; Liu, Z.; et al. Decreased epithelial and sputum miR-221-3p associates with airway eosinophilic inflammation and CXCL17 expression in asthma. Am. J. Physiol. Lung. Cell Mol. Physiol. 2018, 315, L253–L264. [Google Scholar] [CrossRef] [PubMed]
  145. Nejman-Gryz, P.; Górska, K.; Paplińska-Goryca, M.; Proboszcz, M.; Krenke, R. Periostin and Thymic Stromal Lymphopoietin-Potential Crosstalk in Obstructive Airway Diseases. J. Clin. Med. 2020, 9, 3667. [Google Scholar] [CrossRef]
  146. Kanemitsu, Y.; Ito, I.; Niimi, A.; Izuhara, K.; Ohta, S.; Ono, J.; Iwata, T.; Matsumoto, H.; Mishima, M. Osteopontin and periostin are associated with a 20-year decline of pulmonary function in patients with asthma. Am. J. Respir. Crit. Care Med. 2014, 190, 472–474. [Google Scholar] [CrossRef]
  147. Barretto, K.T.; Swanson, C.M.; Nguyen, C.L.; Annis, D.S.; Esnault, S.J.; Mosher, D.F.; Johansson, M.W. Control of cytokine-driven eosinophil migratory behavior by TGF-beta-induced protein (TGFBI) and periostin. PLoS ONE 2018, 13, e0201320. [Google Scholar] [CrossRef]
  148. Johansson, M.W.; Khanna, M.; Bortnov, V.; Annis, D.S.; Nguyen, C.L.; Mosher, D.F. IL-5-stimulated eosinophils adherent to periostin undergo stereotypic morphological changes and ADAM8-dependent migration. Clin. Exp. Allergy 2017, 47, 1263–1274. [Google Scholar] [CrossRef]
  149. Izuhara, K.; Nunomura, S.; Nanri, Y.; Ogawa, M.; Ono, J.; Mitamura, Y.; Yoshihara, T. Periostin in inflammation and allergy. Cell Mol. Life Sci. 2017, 74, 4293–4303. [Google Scholar] [CrossRef]
  150. Corren, J.; Lemanske, R.F.; Hanania, N.A.; Korenblat, P.E.; Parsey, M.V.; Arron, J.R.; Harris, J.M.; Scheerens, H.; Wu, L.C.; Su, Z.; et al. Lebrikizumab treatment in adults with asthma. N. Engl. J. Med. 2011, 365, 1088–1098. [Google Scholar] [CrossRef]
  151. Naik, P.K.; Bozyk, P.D.; Bentley, J.K.; Popova, A.P.; Birch, C.M.; Wilke, C.A.; Fry, C.D.; White, E.S.; Sisson, T.H.; Tayob, N.; et al. Periostin promotes fibrosis and predicts progression in patients with idiopathic pulmonary fibrosis. Am. J. Physiol. Lung. Cell Mol. Physiol. 2012, 303, L1046–L1056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Yoshihara, T.; Nanri, Y.; Nunomura, S.; Yamaguchi, Y.; Feghali-Bostwick, C.; Ajito, K.; Murakami, S.; Mawatari, M.; Izuhara, K. Periostin plays a critical role in the cell cycle in lung fibroblasts. Respir. Res. 2020, 21, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Safont, B.; Tarraso, J.; Rodriguez-Borja, E.; Fernández-Fabrellas, E.; Sancho-Chust, J.N.; Molina, V.; Lopez-Ramirez, C.; Lope-Martinez, A.; Cabanes, L.; Andreu, A.L.; et al. Lung Function, Radiological Findings and Biomarkers of Fibrogenesis in a Cohort of COVID-19 Patients Six Months After Hospital Discharge. Arch. Bronconeumol. 2022, 58, 142–149. [Google Scholar] [CrossRef]
  154. Tomaru, A.; Kobayashi, T.; Hinneh, J.A.; Baffour Tonto, P.; D’Alessandro-Gabazza, C.N.; Fujimoto, H.; Fujiwara, K.; Takahashi, Y.; Ohnishi, M.; Yasuma, T.; et al. Oligonucleotide-targeting periostin ameliorates pulmonary fibrosis. Gene Ther. 2017, 24, 706–716. [Google Scholar] [CrossRef]
  155. Hatipoglu, O.F.; Uctepe, E.; Opoku, G.; Wake, H.; Ikemura, K.; Ohtsuki, T.; Inagaki, J.; Gunduz, M.; Gunduz, E.; Watanabe, S.; et al. Osteopontin silencing attenuates bleomycin-induced murine pulmonary fibrosis by regulating epithelial-mesenchymal transition. Biomed. Pharmacother. 2021, 139, 111633. [Google Scholar] [CrossRef] [PubMed]
  156. Yang, L.; Murota, H.; Serada, S.; Fujimoto, M.; Kudo, A.; Naka, T.; Katayama, I. Histamine contributes to tissue remodeling via periostin expression. J. Invest. Dermatol. 2014, 134, 2105–2113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Herro, R.; Shui, J.W.; Zahner, S.; Sidler, D.; Kawakami, Y.; Kawakami, T.; Tamada, K.; Kronenberg, M.; Croft, M. LIGHT-HVEM signaling in keratinocytes controls development of dermatitis. J. Exp. Med. 2018, 215, 415–422. [Google Scholar] [CrossRef] [PubMed]
  158. Kou, K.; Okawa, T.; Yamaguchi, Y.; Ono, J.; Inoue, Y.; Kohno, M.; Matsukura, S.; Kambara, T.; Ohta, S.; Izuhara, K.; et al. Periostin levels correlate with disease severity and chronicity in patients with atopic dermatitis. Br. J. Dermatol. 2014, 171, 283–291. [Google Scholar] [CrossRef]
  159. Masuoka, M.; Shiraishi, H.; Ohta, S.; Suzuki, S.; Arima, K.; Aoki, S.; Toda, S.; Inagaki, N.; Kurihara, Y.; Hayashida, S.; et al. Periostin promotes chronic allergic inflammation in response to Th2 cytokines. J. Clin. Invest. 2012, 122, 2590–2600. [Google Scholar] [CrossRef] [Green Version]
  160. Ariëns, L.F.M.; van der Schaft, J.; Bakker, D.S.; Balak, D.; Romeijn, M.L.E.; Kouwenhoven, T.; Kamsteeg, M.; Giovannone, B.; Drylewicz, J.; van Amerongen, C.C.A.; et al. Dupilumab is very effective in a large cohort of difficult-to-treat adult atopic dermatitis patients: First clinical and biomarker results from the BioDay registry. Allergy 2020, 75, 116–126. [Google Scholar] [CrossRef]
  161. Yamaguchi, Y.; Ono, J.; Masuoka, M.; Ohta, S.; Izuhara, K.; Ikezawa, Z.; Aihara, M.; Takahashi, K. Serum periostin levels are correlated with progressive skin sclerosis in patients with systemic sclerosis. Br. J. Dermatol. 2013, 168, 717–725. [Google Scholar] [CrossRef] [PubMed]
  162. Satirapoj, B.; Tassanasorn, S.; Charoenpitakchai, M.; Supasyndh, O. Periostin as a tissue and urinary biomarker of renal injury in type 2 diabetes mellitus. PLoS ONE 2015, 10, e0124055. [Google Scholar] [CrossRef] [PubMed]
  163. Hwang, J.H.; Lee, J.P.; Kim, C.T.; Yang, S.H.; Kim, J.H.; An, J.N.; Moon, K.C.; Lee, H.; Oh, Y.K.; Joo, K.W.; et al. Urinary Periostin Excretion Predicts Renal Outcome in IgA Nephropathy. Am. J. Nephrol. 2016, 44, 481–492. [Google Scholar] [CrossRef] [PubMed]
  164. Zhao, X.; Hao, J.; Duan, H.; Rong, Z.; Li, F. Phosphoinositide 3-kinase/protein kinase B/periostin mediated platelet-derived growth factor-induced cell proliferation and extracellular matrix production in lupus nephritis. Exp. Biol. Med. 2017, 242, 160–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Kikuchi, Y.; Kashima, T.G.; Nishiyama, T.; Shimazu, K.; Morishita, Y.; Shimazaki, M.; Kii, I.; Horie, H.; Nagai, H.; Kudo, A.; et al. Periostin is expressed in pericryptal fibroblasts and cancer-associated fibroblasts in the colon. J. Histochem. Cytochem. 2008, 56, 753–764. [Google Scholar] [CrossRef] [Green Version]
  166. Sakellariou, G.T.; Anastasilakis, A.D.; Bisbinas, I.; Oikonomou, D.; Gerou, S.; Polyzos, S.A.; Sayegh, F.E. Circulating periostin levels in patients with AS: Association with clinical and radiographic variables, inflammatory markers and molecules involved in bone formation. Rheumatology 2015, 54, 908–914. [Google Scholar] [CrossRef] [Green Version]
  167. Bonnet, N.; Douni, E.; Perréard Lopreno, G.; Besse, M.; Biver, E.; Ferrari, S. RANKL-Induced Increase in Cathepsin K Levels Restricts Cortical Expansion in a Periostin-Dependent Fashion: A Potential New Mechanism of Bone Fragility. J. Bone Miner. Res. 2021, 36, 1636–1645. [Google Scholar] [CrossRef]
  168. Guo, Y.M.; Cheng, J.H.; Zhang, H.; He, J.W.; Yue, H.; Hu, W.W.; Gu, J.M.; Hu, Y.Q.; Fu, W.Z.; Wang, C.; et al. Serum Periostin Level and Genetic Polymorphisms Are Associated with Vertebral Fracture in Chinese Postmenopausal Women. Genes 2022, 13, 439. [Google Scholar] [CrossRef]
  169. Bader, B.L.; Rayburn, H.; Crowley, D.; Hynes, R.O. Extensive vasculogenesis, angiogenesis, and organogenesis precede lethality in mice lacking all alpha v integrins. Cell 1998, 95, 507–519. [Google Scholar] [CrossRef] [Green Version]
  170. Lacy-Hulbert, A.; Smith, A.M.; Tissire, H.; Barry, M.; Crowley, D.; Bronson, R.T.; Roes, J.T.; Savill, J.S.; Hynes, R.O. Ulcerative colitis and autoimmunity induced by loss of myeloid alphav integrins. Proc. Natl. Acad. Sci. USA 2007, 104, 15823–15828. [Google Scholar] [CrossRef]
  171. Inoue, T.; Akashi, K.; Watanabe, M.; Ikeda, Y.; Ashizuka, S.; Motoki, T.; Suzuki, R.; Sagara, N.; Yanagida, N.; Sato, S.; et al. Periostin as a biomarker for the diagnosis of pediatric asthma. Pediatr. Allergy Immunol. 2016, 27, 521–526. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of modular structural domains of periostin and its interaction with different receptors, as well as its stimulators and repressors.
Figure 1. Schematic representation of modular structural domains of periostin and its interaction with different receptors, as well as its stimulators and repressors.
Cells 12 00050 g001
Figure 2. Periostin-integrins interaction and activation of downstream signaling pathways. The FAS1 domain interacts with integrin receptors to activate different and overlapping signaling pathways, which modulate the progression of non-neoplastic diseases under pathological status. (Shared signaling pathways of integrin αv are shown in the box with a dotted line.)
Figure 2. Periostin-integrins interaction and activation of downstream signaling pathways. The FAS1 domain interacts with integrin receptors to activate different and overlapping signaling pathways, which modulate the progression of non-neoplastic diseases under pathological status. (Shared signaling pathways of integrin αv are shown in the box with a dotted line.)
Cells 12 00050 g002
Figure 3. Sequencing of periostin splice variants.
Figure 3. Sequencing of periostin splice variants.
Cells 12 00050 g003
Table 1. Expression (upregulation or downregulation ↓) and roles of periostin, the periostin-involved signaling pathways, therapies based on periostin, and potential disease biomarkers in disease progression.
Table 1. Expression (upregulation or downregulation ↓) and roles of periostin, the periostin-involved signaling pathways, therapies based on periostin, and potential disease biomarkers in disease progression.
Tissues/
Diseases
Expression
of
Periostin
Roles of
Periostin
ReferencePeriostin-
Involved
Downstream Signaling Pathways
ReferenceTherapies Based on
Periostin
ReferencePotential
Disease
Biomarkers
Reference
EBI and cerebral ischemiaBBB disruption[11]p38/ERK/MMP-9[11]IAXO-102
Clarithromycin
[12]
[13]
Serum
periostin
[14]
Ocular diseasesInflammation[15] Betamethasone; tacrolimus[16]Tear periostin[17]
Tissue
remodeling
[18]Serum
periostin
[19]
CRS and ARInflammation[20,21] Omalizumab; mepolizumab; methylprednisolone; doxycycline[22]Serum
periostin
[23]
Tissue
remodeling
[21,24]Src/AKT/mTOR[24]
Protective role[25] Dupilumab[26]NLF periostin[27]
GCs[28]
AH[29]
AG-Ex[30]
PeriodontitisTissue
remodeling
[31] GCF periostin[32,33]
Wound healing[31]β1/FAK/JNK; β1/RhoA[31]
Bone formation[34,35] Saliva periostin[33]
ADInflammation[36]αv/NF-κB/TSLP; NF-κB/IL-6[36]Dupilumab[26]
Itch[36]αvβ3/TRPV1/TRPA1/NPPB; TSLP/TSLPR/JAK/STAT[36]
Epidermal barrier malfunction[37]IL-24/STAT3[37]Cinnamaldehyde[38]
SclerodermaSkin fibrosis [39]αv/PI3K/AKT/collagen1α1[39]Crenolanib[40]Serum
periostin
[40]
EEInflammation[41]αM[41]Dupilumab[26]Serum
periostin
[42]
AsthmaInflammation[43] Tralokinumab; dupilumab[44,45]Serum
periostin
[46]
Tissue
remodeling
[47]Omalizumab[48]Plasma
periostin
[49]
Hydroprednisone[50]Saliva periostin[49]
Protective roles[51,52]Clarithromycin[53]EBC periostin[54]
Sputum
periostin
[55]
Cardiovascular diseases (MI, heart failure, VHD, hypertension, and vascular calcification)Inflammation[56]Periostin/NLRP3/caspase-1[56]Valsartan[57]Plasma
periostin
[58]
Tissue
remodeling
[59,60,61,62,63,64]
Wound healing[59,65]Simvastatin[63]
Cardiomyocytes apoptosis[66]
Myocardial
regeneration
[5,67,68]αvβ1/αvβ3/αvβ5/PI3K/Akt; TNF-α/NF-κB; PI3K/AKT/cyclin D1[5,67,68]Resveratrol[69]
Angiogenesis[5]
Vascular calcification[70,71]Crenolanib[40]
Lung diseases (PF, EP, COVID-19, and PH)Inflammation[72,73] Serum
monomeric periostin
[74]
Tissue
remodeling
[72,75,76,77]EBC periostin[74]
Angiogenesis[78]BALF periostin[79,80]
Liver diseaseLiver steatosis[81,82,83]α6β4/JNK/c-Jun[83] Serum
periostin
[84]
Hepatic fibrosis[82,85]αvβ3/PI3K/Smad2/3/LOX/LOXL[85]
Liver
regeneration and angiogenesis
[86]
CKDInflammation[87,88]αv/ILK; β3/FAK/AKT; αVβ3/AKT/mTOR[5,88,89]Losartan[90]Urine periostin[91]
Renal fibrosis[87,92,93]FAK/p38/ERK; p38 MAPK[92,93]
Vascular
calcification
[94]αvβ3/Wnt/β-catenin[94]
Renal repair[95]
Renal
malfunction
[96]Serum
periostin
[97]
IBDInflammation[98]NF-κB; αv/IL-8[98] Plasma
periostin
[99]
Serum
periostin
[100]
Osteoarthrosis (RA, OA, AS, osteoporosis, DDH, and IVD
D)
↓ in RA and osteoporosisBone formation[101,102,103]Inhibition of sclerostin/LRP5/Wnt, β-catenin; Wnt/β-catenin; ILK/Akt/GSK-3β;[101,102,103] Serum
periostin
[104]
↑ in OA, AS, DDH, and IVDDInflammation[105,106,107,108]NF-κB/IL-6/8; Wnt/β-catenin/MMP-13/ADAMTS4; DDR1/Akt/Wnt/β-catenin/MMP-13; αvβ3/FAK/Src/NF-κB/IL-6/STAT3[105,106,107,108] SF periostin[109]
K-Postn[110]
Table 2. The expression and roles of periostin isoforms in tissues/diseases.
Table 2. The expression and roles of periostin isoforms in tissues/diseases.
Tissues/DiseasesCertain Periostin
Variants Expressed in Tissues/Diseases
Roles of the Periostin VariantsReference
Cerebral ischemiaIsoform 2Isoform 2
Minimizing the area of
cerebral infarction via phosphorylation of Akt
[111]
AsthmaIsoforms 6, 7, and 8Isoform 8
Promoting the eosinophil adhesion under IL-5 stimulation αMβ2]
[112,113]
MIIsoforms 1, 2, 5, and 6Isoform 1
Decreasing the attachment of fibroblasts and myocytes as well as facilitating myocytes death leading to ventricular dilation and tissue remodeling
Isoform 6
Contributing to the migration of activated fibroblasts and healing of impaired tissue via the αv/FAK/AKT signaling pathway
[114]
IPFAll periostin variants lacking exon 21-[115]
Retinal ischemiaIsoforms 1, 2, and 5Isoforms 1, 2, and 5
Promoting preretinal
pathological NV
[116]
pIBDIsoforms 2, 6, 7, and 8-[99]
JointArticular chondrocytes highly expressed isoforms1 and 5, and anterior cruciate ligament(ACL) progenitor cells overexpressed isoforms 3, 4, 6, 7, and 8-[117]
SerumAt least five isoforms, including 1 or 2, 3, 4, 5, 6At least five isoforms, including 1 or 2, 3, 4, 5, 6
Forming complex with IgA
[118]
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

Yang, L.; Guo, T.; Chen, Y.; Bian, K. The Multiple Roles of Periostin in Non-Neoplastic Disease. Cells 2023, 12, 50. https://doi.org/10.3390/cells12010050

AMA Style

Yang L, Guo T, Chen Y, Bian K. The Multiple Roles of Periostin in Non-Neoplastic Disease. Cells. 2023; 12(1):50. https://doi.org/10.3390/cells12010050

Chicago/Turabian Style

Yang, Lina, Tongtong Guo, Yuanyuan Chen, and Ka Bian. 2023. "The Multiple Roles of Periostin in Non-Neoplastic Disease" Cells 12, no. 1: 50. https://doi.org/10.3390/cells12010050

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