Advances in Clinical and Experimental Medicine

Title abbreviation: Adv Clin Exp Med
5-Year IF – 2.0, IF – 1.9, JCI (2024) – 0.43
Scopus CiteScore – 4.3
Q1 in SJR 2024, SJR score – 0.598, H-index: 49 (SJR)
ICV – 161.00; MNiSW – 70 pts
Initial editorial assessment and first decision within 24 h

ISSN 1899–5276 (print), ISSN 2451-2680 (online)
Periodicity – monthly

Download original text (EN)

Advances in Clinical and Experimental Medicine

Ahead of print

doi: 10.17219/acem/204831

Publication type: review

Language: English

License: Creative Commons Attribution 3.0 Unported (CC BY 3.0)

Download citation:

  • BIBTEX (JabRef, Mendeley)
  • RIS (Papers, Reference Manager, RefWorks, Zotero)

Cite as:


Kozłowska WU, Łomzik J, Kamiński K, Kazimierczyk R. Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) as potential biomarkers for pulmonary arterial hypertension: A review [published online as ahead of print on August 1, 2025]. Adv Clin Exp Med. 2026. doi:10.17219/acem/204831

Matrix metalloproteinases and tissue inhibitors of metalloproteinases as potential biomarkers for pulmonary arterial hypertension: A review

Wiktoria U. Kozłowska1,A,B,C,D,F, Jakub Łomzik1,A,B,C,D,F, Karol Kamiński2,3,A,E,F, Remigiusz Kazimierczyk2,A,D,E,F

1 Students’ Scientific Society, Department of Cardiology, Medical University of Bialystok, Poland

2 Department of Cardiology and Internal Medicine with Cardiac Intensive Care Unit, Medical University of Bialystok, Poland

3 Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Poland

Graphical abstract


Graphical abstracts

Highlights


• Late-stage pulmonary arterial hypertension (PAH) diagnosis delays treatment: Nonspecific symptoms often result in advanced disease at presentation, worsening prognosis.
• Matrix metalloproteinases (MMPs) and tissue inhibitors (TIMPs) as PAH biomarkers: Emerging evidence positions MMP/TIMP profiling for early detection and prognostic assessment.
• MMP-2, MMP-7 and MMP-9 show the strongest prognostic value in PAH: These specific MMPs outperform others in predicting disease progression and patient outcomes.
• Significant biomarker concentration differences distinguish PAH from other pulmonary hypertension types: Multiple studies confirm diagnostic specificity for MMP/TIMP levels.
• Prospective studies are crucial to validate MMP/TIMP profiling in PAH: Ongoing research aims to integrate these biomarkers into clinical diagnostic and prognostic workflows.

Abstract

Pulmonary arterial hypertension (PAH) is a rare and progressive syndrome that is frequently diagnosed at an advanced stage due to the nonspecific nature of its symptoms. Current research aims to identify novel diagnostic tools, including biomarkers, to facilitate earlier detection and differentiation of pulmonary hypertension (PH) subtypes. Matrix metalloproteinases (MMPs) play a critical role in the pathogenesis of PAH through extracellular matrix (ECM) remodeling, with their activity tightly regulated by tissue inhibitors of metalloproteinases (TIMPs). This review summarizes existing studies on the potential of MMPs and TIMPs as biomarkers for PAH. Our analysis highlights significant differences in MMP concentrations between PAH patients and healthy controls. In particular, MMP-2, MMP-7 and MMP-9 exhibit promising prognostic value, which could contribute to risk stratification and support clinical decision-making in the future. However, large-scale, randomized prospective studies involving well-characterized patient cohorts are necessary to confirm their clinical utility and clarify their mechanistic roles in PAH pathogenesis.

Key words: prognosis, biomarkers, pulmonary hypertension, tissue inhibitors of metalloproteinases, metalloproteinases

Introduction

Pulmonary arterial hypertension (PAH) is a progressive disorder marked by dysregulated endothelial-derived vasoactive factors, chronic inflammation and structural remodeling of the pulmonary vasculature.1 These pathological changes ultimately contribute to right ventricular (RV) failure.2 Hemodynamically, PAH is defined by a mean pulmonary arterial pressure (mPAP) >20 mm Hg, pulmonary vascular resistance (PVR) >2 Wood units and pulmonary arterial wedge pressure (PAWP) ≤15 mm Hg, as determined by right heart catheterization (RHC).3, 4

Despite the growing number of available treatment options, the management of PAH remains a significant clinical challenge worldwide. Identifying novel biomarkers is essential for enhancing diagnostic accuracy and recognizing patient subgroups at higher risk of poor outcomes. Among the various molecular pathways implicated in PAH, matrix metalloproteinases (MMPs) and their endogenous inhibitors – tissue inhibitors of metalloproteinases (TIMPs) – represent a promising area of ongoing research. A deeper understanding of these molecules may offer valuable insights into disease pathogenesis and help guide the development of future therapeutic strategies.

Objectives

This review synthesizes current evidence on the role of MMPs and TIMPs in PAH, with a focus on their implications for pathophysiology and their utility as biomarkers of this disease.

Methodology

A comprehensive literature search was conducted using the PubMed and Scopus electronic databases from their inception through July 31, 2024. The search strategy incorporated Medical Subject Headings (MeSH) terms, including “pulmonary arterial hypertension metalloproteinases” and “pulmonary arterial hypertension biomarkers.” A total of 3,231 potentially relevant articles were initially identified. All manuscripts were independently screened by 2 reviewers. Studies were excluded based on the following criteria: 1) not relevant to the selected topic, 2) case reports, 3) non-English language publications, 4) duplicate records, 5) lack of full-text availability, 6) animal studies, and 7) pediatric studies. After applying these exclusion criteria, 22 manuscripts were deemed eligible for inclusion in the analysis.

Interplay between MMPs and TIMPs

Matrix metalloproteinases are a family of zinc-dependent endoproteases that play a central role in the degradation and remodeling of extracellular matrix (ECM) components.5, 6 Their activity is tightly regulated through several mechanisms, including modulation of gene expression,7 activation of proenzymes,8 inhibition by TIMPs,9 subcellular compartmentalization,10 and formation of protein complexes.11 Most MMPs are not constitutively expressed; rather, their transcription is induced in response to pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α), interleukin 1 (IL-1) and various growth factors.12, 13

Additionally, certain MMPs are stored in the granules of inflammatory cells, thereby restricting their enzymatic activity to specific microenvironments.14 Dysregulated MMP expression and activity have been implicated in the pathogenesis of various diseases, particularly cardiovascular disorders.15, 16 These endoproteases are initially synthesized as pre-proMMPs; during translation, the signal peptide is cleaved to produce the latent proMMP form. In this inactive state, the cysteine residue within the propeptide maintains coordination with a Zn2+ ion at the catalytic site through the “cysteine switch” mechanism, thereby preventing enzymatic activity. Activation occurs when this switch is disrupted, typically via proteolytic cleavage by enzymes such as serine proteases, furin endopeptidases, plasmin, or other MMPs.13, 17

The expression and activity of MMPs are tightly regulated by TIMPs.18 The TIMPs are the principal endogenous inhibitors of the MMP family, exhibiting varying inhibitory efficacy against different MMPs, along with distinct patterns of tissue expression and regulatory control.19 This family comprises 4 members – TIMP-1 through TIMP-4.6, 14 Despite their relatively small size, TIMPs serve multiple biochemical and physiological functions, including inhibition of active MMPs, activation of proMMPs, promotion of cell proliferation, matrix binding, and regulation of angiogenesis and apoptosis.14, 20 The TIMPs are distributed across various cellular compartments: TIMP-1, -2, -3, and -4 can localize to the cell surface; TIMP-3 is also found in the ECM; and TIMP-1, -2 and -4 can exist in soluble forms. This spatial diversity enables TIMPs to modulate a broad range of cellular signaling pathways.19

The TIMPs are composed of 2 adjacent domains, each stabilized by 3 intramolecular disulfide bonds. The N-terminal domain, referred to as the “inhibitory domain”, is primarily responsible for MMP inhibition, as it alone is sufficient to suppress MMP activity. The highly conserved core epitope within this domain – centered around the N-terminal strand and characterized by a “Cys-X-Cys” motif – forms the principal interaction site with the catalytic domain of MMPs. This binding involves coordination with the catalytic zinc ion at the MMP active site, effectively blocking the substrate-binding cleft.21, 22 In addition to TIMPs, other molecules, such as α2-macroglobulin and amyloid β precursor protein, have also been identified as inhibitors of MMP activity.23, 24

The balance between MMPs and TIMPs largely influences the ECM composition, which tightly regulates MMP activity.25 Published evidence suggests that MMPs may contribute to the pathophysiology of PAH.26, 27 The early pathological progression of PAH primarily involves endothelial cells (ECs) and smooth muscle cells (SMCs) in the vascular wall, as well as pulmonary artery smooth muscle cells (PASMCs).28, 29 The PASMCs are critically involved in disease advancement.30 The pathological state of PASMCs is associated with PASMC phenotypic switching.28

Several mechanisms have been proposed as potential initiating events in PAH, including increased blood flow and shear stress, hypoxia, inflammation, oxidative stress, and altered bone morphogenetic protein receptor 2 signaling. These factors lead to endothelial/mesenchymal transition, in which ECs adopt a mesenchymal phenotype and express gene profiles characteristic of SMCs.28, 31 In fact, phenotypic switching is an early vascular self-repair mechanism in response to stimulation.1 During this process, ECs lose their intercellular junctions and detach from the intimal monolayer. Subsequently, they migrate into the medial layer, where they undergo dedifferentiation into myofibroblast-like mesenchymal cells.25 These cells exhibit increased secretion of α-smooth muscle actin, collagen, vimentin, and MMPs, as well as serine elastases and lysyl oxidases, while simultaneously showing reduced expression of their endogenous inhibitors, including TIMPs.25, 31, 32 Myofibroblast-like cells contribute to ECM remodeling through enhanced collagen deposition and cross-linking.25

Oxidative stress, exacerbated by inflammation, has also been implicated in MMP dysregulation. Reactive oxygen species (ROS) generated in response to inflammation have been shown to increase MMP secretion while downregulating TIMP expression in SMCs, ECs and fibroblasts. Furthermore, pro-inflammatory cytokines stimulate the recruitment and activation of macrophages and neutrophils, which subsequently secrete MMPs and serine elastases. The degradation products of collagen and elastin generated by heightened proteolytic activity exhibit pro-inflammatory properties, thereby sustaining inflammation through a positive feedback loop.25, 33

Notably, the upregulation of MMP-1 and MMP-9 has been observed, enhancing the migratory capacity of adventitial fibroblasts.34 Deregulation of MMPs has been implicated in multiple pathological processes, including ECs and SMCs migration, hyperplasia, adventitial fibroblast transdifferentiation, increased ECM turnover, and inflammatory cell recruitment. These changes collectively contribute to increased vascular stiffness, preceding abnormal pulmonary arterial pressure and elevated PVR.25 Additionally, ECM degradation products and growth factors further stimulate MMP secretion,25 exacerbating vascular remodeling in small to medium-sized pulmonary vessels, ultimately leading to RV hypertrophy and right-sided heart failure (HF).35

Due to the non-specificity of the symptoms, PAH is frequently diagnosed at a late stage.36 At present, the only routinely used clinical markers that correlate with myocardial stress and provide prognostic information are brain natriuretic peptide and N-terminal pro B-type natriuretic peptide (NT-proBNP).3, 37 However, these are not specific to pulmonary hypertension (PH), as they can be elevated in other heart diseases, showing significant variability.3 Research is now focusing on identifying new diagnostic methods, including biomarkers that allow for prognosis, diagnosis and differentiation of PH subtypes.38 Among the compounds under investigation are MMPs and their inhibitors, which may potentially fulfil this role.

Metalloproteinases

Matrix metalloproteinase 1

Matrix metalloproteinase 1 (MMP-1), a collagenase-1, acts on various substrates, including collagen types I, II, III, VII, VIII, and X, as well as gelatin, aggrecan, casein, nidogen, serpins, versican, perlecan, proteoglycan link protein, and tenascin-C.26, 39 Its expression is stimulated by high glucose levels in cultured ECs and macrophages40 and by C-reactive protein (CRP).41 Pulmonary arterial hypertension is often associated with glucose metabolism disorders,42, 43 and increased concentrations of this molecule in the lungs of PAH patients,42 but unchanged circulating levels have been observed.44 Furthermore, inflammation has been implicated in the pathophysiology of PAH,33 including its role in pulmonary vascular remodeling,45 as reflected by elevated CRP levels in PAH patients.46

Functionally, MMP-1 has been shown to induce the expression of a subset of proangiogenic genes in human microvascular ECs.47 Furthermore, it enhances vascular endothelial growth factor receptor-2 (VEGF-2) expression and ECs proliferation via protease-activated receptor 1 (PAR-1)48 signaling, a mechanism that may also contribute to pulmonary arterial vasoconstriction.49

Elevated serum concentrations of MMP-1 have been demonstrated in PAH patients.27, 50 Additionally, when monocytes were isolated, M1-polarized macrophages from PAH patients, but not from healthy controls, exhibited significantly higher MMP-1 protein levels compared to M0 and M2-polarized macrophages. Immunohistochemical assessment of lung tissue from PAH patients showed notably increased MMP-1 immunoreactivity compared to control samples.27 However, quantitative real-time polymerase chain reaction (qPCR) analysis has not revealed significant differences in MMP-1 transcript expression between cells obtained from lung tissue from patients with idiopathic PAH (IPAH) during lung transplantation and samples from lobectomy of patients with localized lung cancer.51 Similarly, no difference in MMP-1 expression has been observed between pulmonary arteries of IPAH patients and downsized non-tumorous, non-transplanted donor lungs used as controls.52

Matrix metalloproteinase 2

Matrix metalloproteinase 2 (MMP-2), a member of the gelatinase subfamily of MMPs, degrades a broad range of substrates, including collagen types I, IV, V, VII, X, XI, XIV, as well as gelatin, aggrecan, elastin, fibronectin, laminin, nidogen, proteoglycan link protein, and versican.39, 53 This MMP directly affects various cellular functions by modulating the activity of biologically active molecules or interacting with cell surface receptors.54 It is secreted as an inactive proenzyme, which undergoes activation via membrane type 1-matrix metalloproteinase (MT1-MMP). This process is tightly regulated by TIMP-2 at the cell surface at the site where activation is required.55 Several alternative pathways for MT1-MMP-dependent of MMP-2 have been described, involving proteases such as neutrophil elastase, cathepsin G, proteinase-3,56 and plasmin.57 In addition to these pathways, a TIMP-independent activation route has been described, mediated by MT1-MMP and claudin-5.58 Furthermore, the generation of ROS by nicotinamide adenine dinucleotide phosphate oxidase, induced by mechanical stretch, increases MMP-2 messenger RNA (mRNA) expression and pro-MMP-2 release.59

A significantly elevated concentration of MMP-2 has been demonstrated in patients with PAH compared to the control group of healthy volunteers.60 Comparable results were observed in individuals with PH, where 19 out of 36 individuals included in the analysis were diagnosed with IPAH or associated PAH (APAH).61 Plasma circulating levels of MMP-2 were substantially increased in PH patients compared to healthy controls.61 Remodeling biomarkers, including MMP-2, and NT-proBNP, did not show significant differences in concentration between patients with PAH and those with other forms of PH.61 A study investigating patients with PH, including IPAH, PAH associated with connective tissue disease (PAH-CTD), chronic thromboembolic PH (CTEPH), PH due to left heart disease (PH-LHD), and non-PH controls (individuals suspected of PH who underwent RHC and had a mPAP <25 mm Hg), demonstrated that mean plasma circulating levels of MMP-2 were significantly higher in PAH-CTD and PH-LHD patients compared to non-PH controls.62 Furthermore, in PH patients, MMP-2 concentrations exhibited a weak but statistically significant correlation with cardiac output, PAWP and 6-minute walking distance (6MWD). Next, patients with plasma MMP-2 levels in the 1st (below 176 ng/mL) and 2nd quartiles (176–206 ng/mL) had significantly better 5-year survival and time to clinical worsening (TTCW) than those in the 4th quartile (≥246 ng/mL).62 These findings were further confirmed by Arvidsson et al., who linked elevated serum MMP-2 concentrations to an unfavorable prognosis in PAH.63 Increased circulating MMP-2 levels at the time of diagnosis correlated with higher European Society of Cardiology (ESC) and European Respiratory Society (ERS) risk scores, as well as with mean right atrial pressure, NT-proBNP, and 6MWD, indicating worsening right heart function and reduced exercise capacity. MMP-2 was identified as a valuable negative prognostic marker.63

Moreover, Karamanian et al. presented a study based on plasma analyses in PAH patients and healthy volunteers, in which it was shown that MMP-2 circulating levels were elevated in the plasma of non-IPAH patients, but not in the plasma of IPAH patients.64

Studies were also conducted on pulmonary artery tissue from patients undergoing lung transplantation for IPAH and from patients treated by lobectomy for localized lung cancer, who served as controls. In cultured PASMCs derived from patients with IPAH, MMP-2 activity is elevated, attributable to both elevated total MMP-2 expression and a higher proportion of its active form. In situ zymography and immunolocalization showed that MMP-2 was associated with SMCs and elastic fibers. Furthermore, in the arteries of IPAH patients, pronounced gelatinolytic activity and MMP-2 immunostaining were observed along the internal elastic lamina, extending even to its disruption.51 In contrast, the latent MMP-2 levels in IPAH cells remained indistinguishable from those in control cells.51 Conversely, another study reported no significant variation in MMP-2 expression levels in the pulmonary arteries of IPAH patients.52

A notable observation in one study was the elevated concentration of circulating CD34+CD133+ bone marrow-derived proangiogenic precursors in the peripheral blood of IPAH patients compared to healthy controls, with levels correlating with PAP. However, resident endothelial progenitor levels in the pulmonary arteries of IPAH patients were comparable to those in the control group. Colony-forming units of ECs derived from CD34+CD133+ bone marrow precursors of IPAH patients exhibited increased MMP-2 secretion, a greater affinity for angiogenic tube formation, and a tendency to form disorganized cell clusters spontaneously.65

Notably, no significant differences in pro-MMP-2 content were found between PAH patients and those with chronic obstructive pulmonary disease or CTEPH. Additionally, no correlation was observed between pro-MMP-2 levels and PH severity indices.66

Concerning patients with systemic sclerosis-associated PAH (PAH-SSc), MMP-2 concentrations were significantly higher compared to the group of systemic sclerosis (SSc) patients without elevated pulmonary pressure.67 Another study involving 35 SSc patients, of whom 12 had PAH, found that MMP-2 circulating levels were similar in SSc patients with or without PAH.68

Matrix metalloproteinase 3

Matrix metalloproteinase 3 (MMP-3), or stromelysin-1, is involved in the degradation of various ECM components, including matrix proteins, growth factors, proteases, surface receptors, and adhesion molecules. Notably, this metalloproteinase can activate various pro-MMPs, making the synthesis and activation of MMP-3 a fundamental initiating event in the MMP-mediated degradation process.26, 69

Cells derived from IPAH patients demonstrated a marked reduction in MMP-3 production compared to control cells obtained from lung tissue excised during lobectomy for localized lung cancer in 6 individuals. Additionally, immunostaining for MMP-3 was detected exclusively in the media of pulmonary arteries in the control group.51 These findings are further supported by another study,60 which reported significantly lower serum MMP-3 concentrations in PAH patients compared to other subgroups of PH: CTEPH, PH due to HF with preserved ejection fraction (HFpEF-PH), PH due to HF with reduced ejection fraction (HFrEF-PH), and HF without PH (HF-NON-PH).60

Matrix metalloproteinase 7

Matrix metalloproteinase 7 (MMP-7) belongs to the matrilysins, which lack a hemopexin domain and act on many cell surface molecules.26 It is responsible for the degradation of collagen types I, II, III, V, IV, and X, aggrecan, casein, elastin, enactin, laminin, and proteoglycan link protein.39 Matrix metalloproteinase 7 degrades recombinant and native soluble vascular endothelial growth factor receptor-1 (sVEGFR-1), leading to the release of vascular endothelial growth factor (VEGF) from sequestration by sVEGFR-1. In vitro assays demonstrate that MMP-7 promotes VEGF-driven angiogenesis by degrading sVEGFR-1, which otherwise sequesters VEGF and inhibits its activity. Additionally, this metalloproteinase releases VEGF from sVEGFR-1 secreted by human ECs, providing a regulatory mechanism for VEGF bioavailability within the local endothelial microenvironment.70 Elevated concentrations of MMP-7 have been observed in comparison with healthy volunteers without RHC; however, these levels remain lower than those measured in other patient groups (CTEPH, HFpEF-PH, HFrEF-PH, and HF-NON-PH). Receiver operating characteristic (ROC) analysis of plasma MMP-7 concentrations in PAH compared to other PH groups and HF-NON-PH showed an area under the curve (AUC) of 0.75, with a sensitivity of 58.7% and specificity of 83.3%.60 It is noteworthy that plasma levels of MMP-7 increase with age,71 and patients with PAH were generally the oldest among the assessed PH groups. Nevertheless, relatively lower concentrations of MMP-7 were noted compared to other disease groups.60 Arvidsson et al.60 further demonstrated that plasma MMP-7 can differentiate PAH from other causes of dyspnea, including HF with or without PH, as well as from healthy controls, suggesting its potential utility as a novel diagnostic biomarker.

Matrix metalloproteinase 8

Matrix metalloproteinase 8 (MMP-8), a neutrophil-derived collagenase-2, is critically involved in the degradation of ECM components, targeting collagen types I, II, III, V, VII, VIII, and X, as well as gelatin, aggrecan, laminin, and nidogen.39 It is one of the MMPs participating in stem/progenitor cell mobilization and recruitment in blood vessel formation and vascular remodeling.26 Additionally, MMP-8 counteracts pathological mechanobiological feedback by modifying matrix composition and disrupting integrin-β3/FAK and YAP/TAZ-dependent mechanical signaling in PASMCs.72

Dieffenbach et al. conducted a study involving a diverse group of patients, including those with IPAH and APAH, which they categorized into a combined group of patients with PAH, PH, and healthy volunteers. Their findings revealed that the mean plasma concentration of MMP-8 was 18 times higher in PAH patients compared to controls. Furthermore, immunofluorescence staining demonstrated increased MMP-8 expression in PASMCs and the pulmonary endothelium of PAH patients relative to the control group.72

Matrix metalloproteinase 9

Matrix metalloproteinase 9 (MMP-9), a gelatinase-B enzyme, is responsible for the degradation of collagen types IV, V, VII, X, and XIV, fibronectin, laminin, nidogen, proteoglycan link protein, and versican.39 Its secretion is stimulated by high glucose levels.40 Notably, a reduction in the number of NK cells has been observed in PAH lung tissue, accompanied by functional impairments and overexpression of MMP-9.73 Elevated circulating levels of MMP-9 have been detected in the plasma of individuals with PAH,60, 74 suggesting a potential role in pulmonary vascular remodeling. Furthermore, for IPAH, study results are inconsistent, showing both increased levels of MMP-964, 75 and unchanged concentrations.62 Moreover, no statistical correlation was found between plasma MMP-9 levels and hemodynamic parameters or 6MWD, and no statistically meaningful association with survival was detected.62

It is of particular relevance that MMP-9 may be produced by macrophages.76 Chi et al. examined monocytes isolated from PAH patients and healthy donors, differentiating them into M0, M1 or M2 macrophage phenotypes. After differentiation with macrophage colony-stimulating factor into macrophages (M0), MMP-9 mRNA expression was significantly upregulated in PAH patients compared to controls. Similarly, MMP-9 mRNA levels were elevated in M0 and M1 macrophages derived from PAH patients relative to their counterparts in the control group.27

In a subset of patients with PAH related to PAH-CTD and PAH associated with congenital heart disease (PAH-CHD), circulating levels of examined compounds, including MMP-9, were high and did not significantly differ from concentrations in patients with idiopathic, hereditary and anorexigenic PAH. These results suggest that elevated concentrations are markers of disease presence rather than markers of PAH etiology.74 Gene expression profiling of peripheral blood mononuclear cells from patients with IPAH, PAH-SSc, and healthy controls identified MMP-9-related genes, which were further analyzed using RT-PCR. This analysis revealed a clinically significant upregulation of these genes in patients with mild PAH compared to healthy controls, whereas no differences were observed in those with severe PAH.77 Interestingly, circulating MMP-9 concentrations were found to be higher in SSc patients without PAH than in those with PAH involvement.68 Vascular remodeling in CHD-associated APAH and IPAH, as well as in donor lung tissue, was analyzed for mRNA and protein expression patterns. Changes in vascular tissue demonstrated increased mRNA levels of MMP-9 in comparison with adjacent remodeled pulmonary arteries. While plexiform lesions in IPAH and APAH demonstrated only minor differences in remodeling-associated gene regulation, MMP-9 expression was upregulated in APAH compared to adjacent arteries, with a similar but statistically nonsignificant trend observed in IPAH. Importantly, hemodynamic parameters, including mPAP and PVR values measured prior to lung transplantation, had no effect on remodeling-associated gene expression in plexiform lesions.78

Pro-MMP-9 levels in circulating monocytes were found to correlate with key indices of severity of PH-induced cardiac failure but not with PVR or mPAP, suggesting a link between the pro-MMP-9 content of circulating monocytes and HF rather than pulmonary artery remodeling. Subgroup analysis further demonstrated that pro-MMP-9 was lower in patients with HF, defined by a right atrial pressure (Pra) >8 mm Hg and a cardiac index (CI) <2.5 L/min/m2, than in patients without HF.66

Matrix metalloproteinase 10

Matrix metalloproteinase 10 (MMP-10), a stromelysin-2 enzyme, acts on various substrates, including collagen types III, IV, V, gelatin, fibronectin, laminin, nidogen,39 and participates in pro-MMPs proteolysis.26 Increased serum concentrations of MMP-10 have been observed in PAH patients compared to healthy controls, with strong expression detected in the media and adventitia of human pulmonary arteries.27 Notably, MMP-10 expression was significantly reduced in the intima and media, as well as in perivascular tissue, of pulmonary arteries from IPAH patients. To explore the potential prognostic significance of regulated collagens and collagen-processing enzymes, circulating levels of MMP-10 were compared between donors and patients with IPAH. Both groups showed similar concentrations of MMP-10.52

Matrix metalloproteinase 12

Matrix metalloproteinase 12 (MMP-12), a macrophage metalloelastase,39 is involved in macrophage infiltration during inflammation. By facilitating macrophage migration across basement membranes, this enzyme enables their recruitment to inflamed tissues, thereby amplifying and sustaining the inflammatory cascade.79 Notably, circulating MMP-12 concentrations were significantly elevated in patients with PAH compared to the control group.60

Matrix metalloproteinase 19

The catalytic domain of matrix metalloproteinase 19 (MMP-19) exhibits proteolytic activity against a broad spectrum of ECM components, including collagen type IV, laminin, nidogen, large tenascin-C isoform, fibronectin, gelatin type I in vitro. These findings suggest its potential involvement in ECM remodeling.13, 80 Matrix metalloproteinase 19 is inhibited by TIMP-3 and, to a lesser extent, TIMP-1. Its expression was significantly elevated only in the intima + media of IPAH vessels compared to donor vessels.52

Table 1 summarizes the studies discussed above regarding the potential utility of MMPs as biomarkers for PAH. The table includes the PAH subtype and the number of patients enrolled in each study.

Tissue inhibitors of metalloproteinase

Tissue inhibitor of metalloproteinase 1

Tissue inhibitors of metalloproteinase 1 (TIMP-1) have been shown to exhibit multiple functions in regulating biological processes such as cell growth, apoptosis and differentiation.81 Independent of its metalloproteinase inhibitory activity, it exerts these effects through the activation of p38, mitogen-activated protein kinase and c-Jun N-terminal kinase.82 The inhibitor suppresses the activity of most MMPs.83 Furthermore, plasma TIMP-1 correlates with markers of left ventricular diastolic filling and serves as a potential noninvasive marker of fibrosis.84 Notably, its circulating concentrations increase with age and are higher in males, individuals who smoke and those with diabetes.85 While TIMPs exhibit limited specificity towards individual MMPs, TIMP-1 preferentially binds to MMP-9,86 and their levels indicate a positive correlation.87

Higher concentrations of circulating TIMP-1 have been reported in the blood of PAH patients compared to age- and sex-matched healthy controls.62, 74, 88 In a subset of patients with PAH-CTD and PAH-CHD, circulating levels of TIMP-1 were high but did not differ significantly from those in individuals with idiopathic, hereditary and anorexigenic PAH.74 In a study focusing on patients with limited systemic sclerosis (lSSc), increases in TIMP-1 were found compared to healthy controls. It is worth emphasizing that the concentration of this molecule was higher in lSSc patients with PAH (PAH-lSSc) compared to those without PAH, underscoring its relevance as a biomarker for PAH-lSSc.89

An imbalance between MMP and TIMP was identified in cultured PASMCs derived from patients with IPAH through increased expression of TIMP-1. Immunostaining for this compound was observed in pulmonary arteries obtained from both control individuals and patients with IPAH. Furthermore, the same study showed a reduction in MMP-3 expression. This imbalance between MMP-3 and TIMP-1 may be responsible for promoting ECM accumulation.51 Notably, a significant correlation was observed between TIMP-1 and PAWP as well as the 6MWD. TIMP-1 plasma concentrations also increased with the rising New York Heart Association (NYHA) functional class. Moreover, patients with TIMP-1 plasma circulating levels above the median exhibited significantly lower 5-year survival and a higher risk of death compared to those with concentrations below the median. This molecule demonstrates prognostic potential in the described cohort.62 In opposition to previous results, MMP-2/TIMP-1 and MMP-9/TIMP-1 ratios did not correlate with hemodynamic or clinical parameters, such as mPAP, PAWP, CI, or PVR, in IPAH patients.90 Multianalyte profiling analysis revealed increased circulating TIMP-1 levels in both IPAH and non-IPAH patients compared to healthy controls. Variations in inhibitor concentrations were observed across patient subgroups, with the highest levels detected in IPAH patients.64 In contrast to the aforementioned studies, TIMP-1 expression was reduced in the intima and media of pulmonary artery profiles from IPAH patient lungs, and its blood levels did not differ significantly between PAH patients and healthy volunteers.52 In the intima+media of IPAH vessels, collagens (COL4A5, COL14A1 and COL18A1), MMP-19 and a disintegrin and metalloprotease (ADAM) 33 were higher expressed, whereas MMP-10, ADAM17, TIMP1, and TIMP3 were less abundant.

Tissue inhibitor of metalloproteinase 2

Tissue inhibitor of metalloproteinase 2 (TIMP-2) is constitutively expressed in most tissues, but it is not inducible by growth factors.19 This inhibitor exhibits antiangiogenic and antiapoptotic activities.20 In addition to inhibiting most MMPs, it also suppresses a disintegrin and metalloproteinase 12 (ADAM12).91 A key function of TIMP-2 is its participation in the formation of the ternary complex responsible for proMMP-2 activation at the cell surface.92

In IPAH, no significant alterations in TIMP-2 expression were detected in cultured PASMCs; however, increased TIMP-2 transcript expression was observed in PASMCs from IPAH patients compared to controls.51 Moreover, no differences in TIMP-2 expression were found between the lung tissue of patients with IPAH and the control group.52 In a study focused on patients with lSSc, RT-PCR confirmed an increased expression of 9 genes, including TIMP-2, in PAH-lSSc patients. A significant difference in TIMP-2 expression was noted between the PAH-lSSc and lSSc without PAH samples, which persisted between these groups even after excluding patients with extensive pulmonary fibrosis or mildly elevated pulmonary capillary wedge pressure. This suggests that these PAH biomarkers are not primarily caused by pulmonary fibrosis or HF.89

Tissue inhibitor of metalloproteinase 3

Tissue inhibitor of metalloproteinase 3 (TIMP-3) is distinguished by its ability to bind directly to ECM proteins, potentially stabilizing MMP-TIMP complexes within the interstitial space.32 It has the broadest range of substrates, including MMPs,19, 93 as well as several ADAMs: ADAM-17 (TNF-α-converting enzyme),19 ADAM-10, ADAM-12,9 and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTSs): ADAMTS-4 and ADAMTS-5.94 An increase in TIMP-3 levels and the quantity of this protein in lung tissue from patients with IPAH has been observed compared to normal lung tissue. The majority of MMP inhibitor expression in the lungs is likely attributable to SMCs of the bronchi and pulmonary artery, as well as adult pulmonary fibroblasts. TIMP-3 expression has also been documented in lung ECs, ECs from placental microvessels and umbilical veins, and non-adherent blood-derived cell lines.95 In contrast to these findings, significantly lower gene expression of the aforementioned inhibitor was reported in the perivascular tissue of IPAH patients.52 TIMP3 expression was significantly lower in perivascular tissue of IPAH patients.

Tissue inhibitor of metalloproteinase 4

Tissue inhibitor of metalloproteinase 4 (TIMP-4) is the largest identified human MMP inhibitor96 and has been associated with inflammation in cardiovascular disease, suggesting its potential role as a systemic biomarker of vascular inflammatory activity. Beyond its ability to inhibit MMPs, TIMP-4 also suppresses ADAM28.97 Elevated circulating TIMP-4 levels have been observed in patients with increased pulmonary artery systolic pressure measurements on echocardiography among patients with SSc, where pulmonary artery systolic pressure is considered elevated if it reaches or exceeds 40 mm Hg.98

In healthy individuals, plasma TIMP-4 concentrations are not influenced by sex or age.60 Schumann et al. confirmed that plasma TIMP-4 concentrations were significantly elevated in PH patients compared with healthy age- and sex-matched volunteers. The remodeling biomarkers, including TIMP-4, were not statistically significantly different between PAH and other forms of PH.61 The concentration of the metalloproteinase inhibitor was significantly higher in patients with PAH compared to the control group.60 This contradicted findings by Tiede et al., where mean circulating levels of TIMP-4 in plasma were notably lower in patients with IPAH compared with non-PH controls and patients with PH-LHD.62 These discrepancies may be attributable to differences in study populations; the first cited included both IPAH and PAH-SSc patients. Additionally, TIMP-4 exhibited modest yet significant correlations with mPAP, PAWP, PVR, and 6MWD,62 and its elevated concentrations are associated with unfavorable prognosis in PAH.63 No differences in TIMP-4 expression were found in the pulmonary arteries between IPAH patients and the control group.52 Within a subgroup selected from the previously mentioned cohort,62 the MMP2/TIMP-4 ratio was assessed, which showed a notable correlation with mPAP, PVR, estimated glomerular filtration rate (eGFR), and tricuspid annular plane systolic excursion, while MMP9/TIMP-4 showed significant correlation with mPAP and eGFR. Moreover, MMP2/TIMP-4 demonstrated considerable results in the ROC analysis predicting death and cardiovascular events.90

ADAM, ADAMTS and ADAMTSL

The equilibrium between ECM degradation and remodeling is regulated not only by MMPs but also by a disintegrin and metalloproteinases (ADAMs) and ADAMTSs,92 which are proteases closely related to MMPs.99 These molecules carry MMP and disintegrin-like domains, giving them the properties of both proteases and adhesion molecules.100, 101 Crucially, they can also be inhibited by TIMPs.14 ADAMTS8 has been implicated in PAH pathogenesis and RV failure, along with the expansion of PASMCs, ECM remodeling and endothelial dysfunction in an autocrine/paracrine manner.102

In a cohort of patients with IPAH, no significant differences in ADAMTS1 and ADAMTS13 expression were observed when compared to healthy volunteers.52 Upregulation of ADAMTS8 has been demonstrated in areas of α-smooth muscle actin, along with increased levels of this compound in the lungs of patients with PAH. Furthermore, RT-PCR has shown that the ADAMTS8 gene is more strongly expressed in PASMCs from PAH patients (PAH-PASMCs) than in control PASMCs. Notably, no significant difference in the expression of this protease was observed between control pulmonary artery endothelial cells (PAEC) and those from patients with pulmonary arterial hypertension (PAH-PAEC). Moreover, the expression levels were comparable in both PAH-PAEC and PAH-PASMCs.102 These findings indicate that ADAMTS8 upregulation in PASMCs may contribute to the pathogenesis of PAH through PASMCs proliferation and migration, increased MMP activity and mitochondrial dysfunction.103

In patients with IPAH, no differences in ADAMTS13 expression were found compared to healthy controls, although the pulmonary embolism group exhibited slightly lower concentrations. Additionally, there was no correlation with sex or ethnic identity.104 Plasma samples from patients with incidentally diagnosed untreated PAH, as well as those with CTEPH, HFpEF, and a control population with dyspnea or HF without PH (dyspnea/HF-non-PH), were analyzed, alongside samples from 20 healthy controls. In contrast to previous reports, ADAMTS13 concentrations were significantly lower in PAH patients compared to individuals with CTEPH, HFpEF-PH, HFrEF-PH, dyspnea/HF-non-PH, and healthy controls. No differences were observed between subgroups analyzing IPAH and SSc-PAH. Furthermore, a correlation between ADAMTS13 levels and PAWP was established. The study concluded that using a multifactorial logistic regression model, incorporating age- and sex-adjusted ADAMTS13 concentrations, effectively differentiated PAH patients from other dyspnea-related disease groups, achieving AUC of 0.91, with a sensitivity of 87.5% and specificity of 78.4%.105

In a study on ADAMTSL4, Li et al.106 confirmed that plasma ADAMTSL4 protein concentrations were significantly elevated in patients with IPAH and CTEPH compared to healthy controls. It is noteworthy that the AUC for ADAMTSL4 was 0.947 for the PH group (IPAH and CTEPH combined) and 0.981 for the IPAH subgroup. Furthermore, plasma ADAMTSL4 concentrations exhibited a positive correlation with mPAP.

ADAM-10 and ADAM-17 play a critical role in the inflammatory response by promoting the release of pro-inflammatory cytokines, such as TNF-α and IL-6, from peripheral blood macrophages and ECs.107 ADAM33 is involved in promoting angiogenesis, as well as stimulating cell proliferation and differentiation.108 No changes in ADAM10 expression were found in the media or intima of pulmonary arteries in patients with IPAH, but a significant reduction in expression of ADAM17 and upregulation of ADAM33 was observed.52 Additionally, plasma ADAM33 concentrations remained unchanged in IPAH patients compared to controls.52

Table 2 provides a summary of the studies examining the potential role of TIMPs as biomarkers in PAH. It outlines the PAH subtype investigated and the corresponding number of patients included in each study.

Discussion

The described compounds offer promising possibilities for more advanced diagnosis and for assessing prognosis in this challenging condition. Metalloproteinases exert a fundamental role in the pathogenesis of PAH through ECM remodeling. The concentrations of specific MMPs differ significantly between PAH patients and healthy individuals. Particularly, MMP-2, MMP-7 and MMP-9 exhibit considerable prognostic potential, which may help in clinical decisions in the future. Furthermore, TIMP-1 and TIMP-2, when assessed in conjunction with a panel of 7 additional circulating proteins, have been shown to effectively identify PAH patients at high risk of mortality.109 A potential biomarker, it can refer not only to the concentration of a particular molecule but also to their proportion to one another, for instance, MMP-2/TIMP-4 and MMP-9/TIMP-4, which correlate with clinical outcomes. In addition, manuscripts have shown differences in biomarker concentrations between PAH and other causes of PH. These findings, along with comparisons of biomarker concentrations between PAH and other causes of PH, have been drawn from Table 1 and Table 2, which summarize and contrast the referenced studies. Notably, the small sample sizes in many studies, an inherent limitation given the rarity of PAH, present challenges in formulating clinically valid conclusions. Furthermore, a significant proportion of investigations have relied on lung tissue samples, restricting the practical application of potential biomarkers due to the invasive nature of tissue sampling. Future research should prioritize less-invasive biomarkers to facilitate broader clinical implementation.

Phenotypic switching, which plays a unique and pivotal role in the initiation of PAH, is considered a critical early marker of pulmonary vascular remodeling and disease progression.28 Identifying biomarkers capable of capturing the onset of PAH could enable risk stratification and facilitate earlier therapeutic intervention. Emerging therapies, such as sotatercept, have been shown to improve quality of life and potentially extend survival in patients with PAH. A delay of 2 years in initiating sotatercept therapy could result in a loss of 4.1 life years per patient,110 underscoring the essential requirement for early diagnosis and treatment initiation.

Advancements in the understanding of MMPs and their inhibitors have contributed to the identification of novel therapeutic targets for PAH. Lercanidipine, a vasoselective dihydropyridine calcium channel blocker, has demonstrated potential clinical benefits in PAH by reducing serum MMP-9 levels and improving survival without affecting proMMP-2 activity or TIMP-1 levels.111 Meanwhile, the endogenous elastase inhibitor – elafin – has been postulated to exert protective effects against PAH by suppressing MMP-9 activity. Experimental models of PAH have consistently shown increased elastase activity, and elafin is now progressing through clinical evaluation, with initial trials assessing its safety and tolerability in healthy volunteers.112

Limitations

Our analysis was limited to 2 databases, which may have restricted the number of studies identified. However, PubMed and Scopus are the 2 largest databases primarily focused on medical research, ensuring broad coverage of relevant literature. The screening process was conducted independently by 2 reviewers, who subsequently compared the included studies, thereby minimizing the risk of selection bias. All studies meeting the inclusion criteria were considered, which resulted in variability in sample sizes and PAH subtypes included. These differences were systematically presented in tables, allowing for a more objective comparison of the reported findings.

Conclusions

These findings highlight the pivotal role of MMP-related pathways in the pathophysiology of PAH and suggest that targeted modulation of these mechanisms may offer promising therapeutic avenues. However, to determine the clinical applicability of MMP-targeted therapies and their potential to alter disease progression, rigorous evaluation through prospective, controlled trials is essential. Large-scale studies are required to validate these initial observations, with particular emphasis on the standardization of biomarker measurement protocols – including sample collection, processing and analytical methods – to ensure reproducibility and comparability across research settings. As our understanding of PAH deepens, the integration of validated biomarker assessments into clinical practice may enhance patient outcomes through more precise, personalized therapeutic strategies.

Use of AI and AI-assisted technology

Not applicable.

Tables


Table 1. Summary of research on matrix metalloproteinase (MMP) concentrations in pulmonary arterial hypertension (PAH)

Molecule

Studied groups

Sample

Groups size

Change in concentration

Reference

MMP-1

PAH

plasma

lung tissue

nPAH = 39

nHealthyControl = 30

Increase in PAH compared to the control.

Chi et al., 202227

IPAH

PAH-SSc

plasma

lung tissue

nIPAH = 27

nPAH-SSc = 27

nHealthyControl = 6

Increase in IPAH and PAH-SSc compared to the control.

Mickael et al., 202250

IPAH

lung tissue

nIPAH = 5

nLungCancerControl = 6

No difference between groups.

Lepetit et al., 200551

IPAH

lung tissue

nIPAH = 20

nLungDonorControl = 22

No difference between groups.

Hoffmann et al., 201552

MMP-2

IPAH

lung tissue

nIPAH = 5

nLungCancerControl = 6

Increase in IPAH compared to the control.

Lepetit et al., 200551

IPAH

lung tissue

nIPAH = 20

nLungDonorControl = 22

No difference between groups.

Hoffmann et al., 201552

PAH

CTEPH

HFpEF-PH

HFrEF-PH

plasma

nPAH = 48

nCTEPH = 20

nHFpEF-PH = 33

nHFrEF-PH = 36

nHF-NON-PH = 15

nHealthyControl = 20

Increase in PAH and PH groups (CTEPH, HFpEF-PH, HFrEF-PH) compared to the control.

Arvidsson et al., 201960

IPAH

APAH

group 2 PH

group 3 PH

CTEPH

plasma

nIPAH = 16

nAPAH = 3

nGroup2PH = 2

nGroup3PH = 4

nCTEPH = 11

nHealthyControl = 44

Increase in PH groups (IPAH, APAH, group 2 and 3 PH, CTEPH) compared to the control.

Schumann et al., 201061

IPAH

PAH-CTD

CTEPH

PH-LHD

plasma

nIPAH = 42

nPAH-CTD = 42

nCTEPH = 43

nPH-LHD = 33

nNon-PHControl = 44

Increase in PAH-CTD and PH-LHD compared to the non-PH control.

Tiede et al., 201662

IPAH

HPAH

PAH-SSc

PPHTN

DPAH

PAH-CHD

plasma

nIPAH = 58

nHPAH = 5

nPAH-SSc = 15

nSLE-PAH = 3

nPPHTN = 13

nDPAH = 6

nPAH-CHD = 5

nHealthyControl = 51

Increase in PAH groups (without IPAH) compared to the control.

Karamanian et al., 201464

PAH-SSc

plasma

nPAH-SSc = 77

nSSc-non-PHControl = 80

Increase in PAH-SSc compared to the control.

Bauer et al., 202167

PAH-SSc

plasma

nPAH-SSc = 12

nSSc-non-PAH = 23

No difference between groups.

Giannelli et al., 200568

Pro-MMP-2

IPAH

PPHTN

PAH-CHD

anorexigen associated PAH

PH-COPD

CTEPH

lung tissue

nIPAH = 2

nPPHTN = 1

nPAH-CHD = 2

nAnorexigen associatedPAH = 3

nPH-COPD = 6

nCPTE = 3

No difference between groups.

Cantini-Salignac et al., 200666

MMP-3

IPAH

lung tissue

nIPAH = 5

nLungCancerControl = 6

Decrease in IPAH compared to the control.

Lepetit et al., 200551

PAH

CTEPH

HFpEF-PH

HFrEF-PH

plasma

nPAH = 48

nCTEPH = 20

nHFpEF-PH = 33

nHFrEF-PH = 36

nHF-NON-PH = 15

nHealthyControl = 20

Decrease in PAH compared to PH groups (CTEPH, HFpEF-PH, HFrEF-PH).

Arvidsson et al., 201960

MMP-7

IPAH

lung tissue

nIPAH = 20

nLungDonorControl = 22

No difference between groups.

Hoffmann et al., 201552

PAH

CTEPH

HFpEF-PH

HFrEF-PH

plasma

nPAH = 48

nCTEPH = 20

nHFpEF-PH = 33

nHFrEF-PH = 36

nHF-NON-PH = 15

nHealthyControl = 20

Increase in PH groups (PAH, CTEPH, HFpEF-PH, HFrEF-PH) compared to the control.

Arvidsson et al., 201960

MMP-8

IPAH

APAH

group 2 PH

group 3 PH

group 4 PH

plasma

lung tissue

nIPAH = 10

nAPAH = 7

nGroup2PH = 1

nGroup3PH = 19

nGroup4PH = 2

nHealthyControl = 12

Increase in PAH (IPAH, APAH) compared to the PH groups (group 2, 3, 4 PH) and the control.

Dieffenbach et al., 202172

MMP-9

PAH

lung tissue

nPAH = 39

nHealthyControl = 30

Increase in PAH compared to the control.

Chi et al., 202227

PAH

CTEPH

HFpEF-PH

HFrEF-PH

plasma

nPAH = 48

nCTEPH = 20

nHFpEF-PH = 33

nHFrEF-PH = 36

nHF-NON-PH = 15

nHealthyControl = 20

Increase in PH groups (PAH, CTEPH, HFpEF-PH, HFrEF-PH) compared to the control.

Arvidsson et al., 201960

IPAH

PAH-CTD

CTEPH

PH-LHD

plasma

nIPAH = 42

nPAH-CTD = 42

nCTEPH = 43

nPH-LHD = 33

nNon-PHControl = 44

No difference between groups.

Tiede et al., 201662

IPAH

HPAH

PAH-SSc

PPHTN

DPAH

PAH-CHD

plasma

nIPAH = 58

nHPAH = 5

nPAH-SSc = 15

nSLE-PAH = 3

nPPHTN = 13

nDPAH = 6

nPAH-CHD = 5

nHealthyControl = 51

Increase in PAH groups (IPAH, HPAH, PAH-SSc, PPHTN, DPAH, PAH-CHD) compared to the control.

Karamanian et al., 201464

PAH-SSc

plasma

nPAH-SSc = 12

nSSc-non-PAH = 23

Decrease in PAH-SSc compared to the SSc-non-PAH.

Giannelli et al., 200568

IPAH

anorexigen associated PAH

HPAH

plasma

nPAH = 68

nHealthyControl = 37

Increase in PAH (IPAH, HPAH, Anorexigen associated PAH) compared to the control.

Safdar et al., 201474

IPAH

CVD-PAH

plasma

nIPAH = 42

nCVD-PAH = 2

Increase in IPAH compared to the manufacturer -supplied reference.

Hiremath et al., 201075

IPAH

PAH-SSc

plasma

nIPAH = 9

nPAH-SSc = 10

nHealthyControl = 5

Increase in mild PAH (WHO I & II IPAH and PAH-SSc) compared to the control.

Grigoryev et al., 200877

Pro-MMP-9

IPAH

PPHTN

PAH-CHD

anorexigen associated PAH

PH-COPD

CPTE

lung tissue

nIPAH = 2

nPPHTN = 1

nPAH-CHD = 2

nAnorexigenassociatedPAH = 3

nPH-COPD = 6

nCPTE = 3

No difference between groups.

Cantini-Salignac et al., 200666

MMP-10

PAH

lung tissue

plasma

nPAH = 39

nHealthyControl = 30

Increase in PAH compared to the control.

Chi et al., 202227

IPAH

plasma

nIPAH = 40

nLungDonorControl = 40

No difference between groups.

Hoffmann et al., 201552

IPAH

lung tissue

nIPAH = 20

nLungDonorControl = 22

Decrease in IPAH compared to the control.

Hoffmann et al., 201552

MMP-12

PAH

CTEPH

HFpEF-PH

HFrEF-PH

plasma

nPAH = 48

nCTEPH = 20

nHFpEF-PH = 33

nHFrEF-PH = 36

nHF-NON-PH = 15

nHealthyControl = 20

Increase in PH groups (PAH, CTEPH, HFpEF-PH, HFrEF-PH) compared to the control.

Arvidsson et al., 201960

MMP-19

IPAH

lung tissue

nIPAH = 20

nLungDonorControl = 22

Increase in IPAH compared to the control.

Hoffmann et al., 201552

APAH – associated pulmonary arterial hypertension; PAH-CHD – congenital heart disease pulmonary arterial hypertension; CPTE – chronic pulmonary thromboembolism; CTEPH – chronic thromboembolic pulmonary hypertension; CVD-PAH – collagen vascular disease pulmonary arterial hypertension; DPAH – drug- and toxin-associated pulmonary arterial hypertension; HF-NON-PH – heart failure without pulmonary hypertension; HFpEF-PH – pulmonary hypertension due to heart failure with preserved ejection fraction; HFrEF-PH – pulmonary hypertension due to heart failure with reduced ejection fraction; HPAH – heritable pulmonary arterial hypertension; IPAH – idiopathic pulmonary arterial hypertension; PAH-CTD – pulmonary arterial hypertension associated with connective tissue diseases; PAH-SSc – systemic sclerosis-associated pulmonary arterial hypertension; PH – pulmonary hypertension; PH-COPD – chronic obstructive pulmonary disease-associated pulmonary hypertension; PH-LHD – pulmonary hypertension due to left heart disease; PPHTN – portopulmonary hypertension; SLE-PAH – systemic lupus erythematosus-associated pulmonary arterial hypertension.
Table 2. Summary of research on tissue inhibitor of metalloproteinases (TIMPs), A disintegrin and metalloproteinase (ADAM) and ADAM with thrombospondin motifs (ADAMTS) concentrations in pulmonary arterial hypertension (PAH)

Molecule

Studied groups

Sample

Group size

Change in concentration

Reference

TIMP-1

IPAH

lung tissue

nIPAH = 5

nLungCancerControl = 6

Increase in IPAH compared to the control.

Lepetit et al., 200551

IPAH

plasma

nIPAH = 40

nLungDonorControls = 40

No difference between groups.

Hoffmann et al., 201552

IPAH

lung tissue

nIPAH = 20

nLungDonorControls = 22

Decrease in IPAH compared to the control.

Hoffmann et al., 201552

IPAH

PAH-CTD

CTEPH

PH-LHD

plasma

nIPAH = 42

nPAH-CTD = 42

nCTEPH = 43

nPH-LHD = 33

nNon-PHControl = 44

Increase in PAH-CTD compared to the CTEPH and control.

Tiede et al., 201662

IPAH

HPAH

PAH-SSc

PPHTN

DPAH

PAH-CHD

plasma

nIPAH = 58

nHPAH = 5

nPAH-SSc = 15

nSLE-PAH = 3

nPPHTN = 13

nDPAH = 6

nPAH-CHD = 5

nHealthyControl = 51

Increase in PAH groups (IPAH, HPAH, PAH-SSc, PPHTN, DPAH, PAH-CHD) compared to the control.

Karamanian et al., 201464

IPAH

anorexigen-associated PAH

HPAH

plasma

nPAH = 68

nHealthyControl = 37

Increase in PAH (IPAH, HPAH, anorexigen associated PAH) compared to the control.

Safdar et al., 201474

IPAH

CTD-PAH

plasma

nIPAH = 12

nCTD-PAH = 3

nHealthyControl = 10

Increase in PAH (IPAH, CTD-PAH) compared to the control.

Fenster et al., 201688

PAH-lSSc

plasma

n-PAH-lSSc = 15

nlSSc-noPAH = 21

nHealthyControl = 10

Increase in PAH-lSSc compared to the lSSc-noPAH and control.

Pendergrass et al., 201089

TIMP-2

IPAH

lung tissue

nIPAH = 5

nLungCancerControl = 6

No difference between groups.

Lepetit et al., 200551

IPAH

lung tissue

nIPAH = 20

nLungDonorControls = 22

No difference between groups.

Hoffmann et al., 201552

PAH-lSSc

plasma

nPAH-lSSc = 15

nlSSc-noPAH = 21

nHealthyControl = 10

Increase in PAH-lSSc compared to the lSSc-noPAH and control.

Pendergrass et al., 201089

TIMP-3

IPAH

lung tissue

nIPAH = 20

nLungDonorControls = 22

Decrease in IPAH compared to the control.

Hoffmann et al., 201552

IPAH

lung tissue

nIPAH = 4

nHealthyControl = 4

Increase in IPAH compared to the control.

Edgar et al., 200695

TIMP-4

IPAH

lung tissue

nIPAH = 20

nLungDonorControls = 22

No difference between groups.

Hoffmann et al., 201552

PAH

CTEPH

HFpEF-PH

HFrEF-PH

plasma

nPAH = 48

nCTEPH = 20

nHFpEF-PH = 33

nHFrEF-PH = 36

nHF-NON-PH = 15

nHealthyControl = 20

Increase in PAH and PH groups (CTEPH, HFpEF-PH, HFrEF-PH) compared to the control.

Arvidsson et al., 201960

IPAH

APAH

group 2 PH

group 3 PH

CTEPH

plasma

nIPAH = 16

nAPAH = 3

nGroup2PH = 2

nGroup3PH = 4

nCTEPH = 11

nHealthyControl = 44

Increased in PH groups (IPAH, APAH, group 2 and 3 PH, CTEPH) compared to the control.

Schumann et al., 201061

IPAH

PAH-CTD

CTEPH

PH-LHD

plasma

nIPAH = 42

nPAH-CTD = 42

nCTEPH = 43

nPH-LHD = 33

nNon-PHControl = 44

Decrease in IPAH compared to the non-PH control and PH-LHD.

Tiede et al., 201662

ADAMTS1

IPAH

lung tissue

nIPAH = 20

nLungDonorControls = 22

No difference between groups.

Hoffmann et al., 201552

ADAMTS8

PAH

lung tissue

nPAH = 6

nControlPASMCs = 5

Increase in PAH compared to the control.

Omura et al., 2019102

ADAMTS13

IPAH

lung tissue

nIPAH = 20

nLungDonorControls = 22

No difference between groups.

Hoffmann et al., 201552

CTEPH

CTED

IPAH

PE

plasma

nCTEPH = 208

nCTED = 35

nIPAH = 30

nPE = 28

nHealthyControl = 68

No difference between IPAH and the control.

Newnham et al., 2019104

IPAH

FPAH

PAH-SSc

PAH-CTD

CTEPH

HFpEF-PH

HFrEF-PH

plasma

nIPAH = 21

nFPAH = 2

nPAH-SSc = 21

nPAH-CTD = 4

nCTEPH = 20

nHFpEF-PH = 31

nHFrEF-PH = 36

nHFrEF = 8

nHFpEF = 7

nHealthyControl = 20

Decrease in PAH (IPAH, FPAH, PAH-SSc, PAH-CTD) compared to the CTEPH, HFpEF-PH, HFrEF-PH and control.

Ahmed et al., 2021105

ADAMTSL4

IPAH

plasma

nIPAH = 8

nHealthyControl = 9

Increase in IPAH compared to the control.

Li et al., 2023106

ADAM10

IPAH

lung tissue

nIPAH = 20

nLungDonorControls = 22

No difference between groups.

Hoffmann et al., 201552

ADAM17

IPAH

lung tissue

nIPAH = 20

nLungDonorControls = 22

Decrease in IPAH compared to the control.

Hoffmann et al., 201552

ADAM33

IPAH

lung tissue

nIPAH = 20

nLungDonorControls = 22

Increase in IPAH compared to the control.

Hoffmann et al., 201552

IPAH

plasma

nIPAH = 40

nLungDonorControls = 40

No difference between groups.

Hoffmann et al., 201552

APAH – associated pulmonary arterial hypertension; PAH-CHD – congenital heart disease pulmonary arterial hypertension; CPTE – chronic pulmonary thromboembolism; CTEPH – chronic thromboembolic pulmonary hypertension; CVD-PAH – collagen vascular disease pulmonary arterial hypertension; DPAH – drug- and toxin-associated pulmonary arterial hypertension; HF-NON-PH – heart failure without pulmonary hypertension; HFpEF-PH – pulmonary hypertension due to heart failure with preserved ejection fraction; HFrEF-PH – pulmonary hypertension due to heart failure with reduced ejection fraction; HPAH – heritable pulmonary arterial hypertension; IPAH – idiopathic pulmonary arterial hypertension; PAH-CTD – pulmonary arterial hypertension associated with connective tissue diseases; PAH-SSc – systemic sclerosis-associated pulmonary arterial hypertension; PH – pulmonary hypertension; PH-COPD – chronic obstructive pulmonary disease-associated pulmonary hypertension; PH-LHD – pulmonary hypertension due to left heart disease; PPHTN – portopulmonary hypertension; SLE-PAH – systemic lupus erythematosus-associated pulmonary arterial hypertension.

References (112)

  1. Schermuly RT, Ghofrani HA, Wilkins MR, Grimminger F. Mechanisms of disease: Pulmonary arterial hypertension. Nat Rev Cardiol. 2011;8(8):443–455. doi:10.1038/nrcardio.2011.87
  2. Naeije R, Richter MJ, Rubin LJ. The physiological basis of pulmonary arterial hypertension. Eur Respir J. 2022;59(6):2102334. doi:10.1183/13993003.02334-2021
  3. Humbert M, Kovacs G, Hoeper MM, et al. 2022 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Heart J. 2022;43(38):3618–3731. doi:10.1093/eurheartj/ehac237
  4. Maron BA. Revised definition of pulmonary hypertension and approach to management: A clinical primer. JAHA. 2023;12(8):e029024. doi:10.1161/JAHA.122.029024
  5. Löffek S, Schilling O, Franzke CW. Biological role of matrix metalloproteinases: A critical balance. Eur Respir J. 2011;38(1):191–208. doi:10.1183/09031936.00146510
  6. Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases: Structure, function, and biochemistry. Circ Res. 2003;92(8):827–839. doi:10.1161/01.RES.0000070112.80711.3D
  7. Yan C, Boyd DD. Regulation of matrix metalloproteinase gene expression. J Cell Physiol. 2007;211(1):19–26. doi:10.1002/jcp.20948
  8. Murphy G, Stanton H, Cowell S, et al. Mechanisms for pro matrix metalloproteinase activation. APMIS. 1999;107(1–6):38–44. doi:10.1111/j.1699-0463.1999.tb01524.x
  9. Ramnath N, Creaven PJ. Matrix metalloproteinase inhibitors. Curr Oncol Rep. 2004;6(2):96–102. doi:10.1007/s11912-004-0020-7
  10. Mannello F, Medda V. Nuclear localization of matrix metalloproteinases. Prog Histochem Cytochem. 2012;47(1):27–58. doi:10.1016/j.proghi.2011.12.002
  11. Hadler-Olsen E, Fadnes B, Sylte I, Uhlin-Hansen L, Winberg J. Regulation of matrix metalloproteinase activity in health and disease. FEBS J. 2011;278(1):28–45. doi:10.1111/j.1742-4658.2010.07920.x
  12. Zitka O, Kukacka J, Krizkov S, et al. Matrix metalloproteinases. Curr Med Chem. 2010;17(31):3751–3768. doi:10.2174/092986710793213724
  13. Simões G, Pereira T, Caseiro A. Matrix metaloproteinases in vascular pathology. Microvasc Res. 2022;143:104398. doi:10.1016/j.mvr.2022.104398
  14. Tallant C, Marrero A, Gomis-Rüth FX. Matrix metalloproteinases: Fold and function of their catalytic domains. Biochim Biophys Acta Mol Cell Res. 2010;1803(1):20–28. doi:10.1016/j.bbamcr.2009.04.003
  15. Yabluchanskiy A, Ma Y, Iyer RP, Hall ME, Lindsey ML. Matrix metalloproteinase-9: Many shades of function in cardiovascular disease. Physiology. 2013;28(6):391–403. doi:10.1152/physiol.00029.2013
  16. Tayebjee MH, Nadar S, Blann AD, Gareth Beevers D, MacFadyen RJ, Lip GYH. Matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 in hypertension and their relationship to cardiovascular risk and treatment: A substudy of the Anglo-Scandinavian Cardiac Outcomes Trial (ASCOT). Am J Hypertens. 2004;17(9):764–769. doi:10.1016/j.amjhyper.2004.05.019
  17. Laronha H, Caldeira J. Structure and function of human matrix metalloproteinases. Cells. 2020;9(5):1076. doi:10.3390/cells9051076
  18. Cui N, Hu M, Khalil RA. Biochemical and biological attributes of matrix metalloproteinases. Prog Mol Biol Transl Sci. 2017;147:1–73. doi:10.1016/bs.pmbts.2017.02.005
  19. Cabral-Pacheco GA, Garza-Veloz I, Castruita-De La Rosa C, et al. The roles of matrix metalloproteinases and their inhibitors in human diseases. Int J Mol Sci. 2020;21(24):9739. doi:10.3390/ijms21249739
  20. Costanzo L, Soto B, Meier R, Geraghty P. The biology and function of tissue inhibitor of metalloproteinase 2 in the lungs. Pulm Med. 2022;2022:3632764. doi:10.1155/2022/3632764
  21. Raeeszadeh-Sarmazdeh M, Do L, Hritz B. Metalloproteinases and their inhibitors: Potential for the development of new therapeutics. Cells. 2020;9(5):1313. doi:10.3390/cells9051313
  22. Li K, Tay FR, Yiu CKY. The past, present and future perspectives of matrix metalloproteinase inhibitors. Pharmacol Ther. 2020;207:107465. doi:10.1016/j.pharmthera.2019.107465
  23. Wang X, Khalil RA. Matrix metalloproteinases, vascular remodeling, and vascular disease. Adv Pharmacol. 2018;81:241–330. doix:10.1016/bs.apha.2017.08.002
  24. Hopps E, Lo Presti R, Caimi G. Matrix metalloproteases in arterial hypertension and their trend after antihypertensive treatment. Kidney Blood Press Res. 2017;42(2):347–357. doi:10.1159/000477785
  25. Thenappan T, Chan SY, Weir EK. Role of extracellular matrix in the pathogenesis of pulmonary arterial hypertension. Am J Physiol Heart Circ Physiol. 2018;315(5):H1322–H1331. doi:10.1152/ajpheart.00136.2018
  26. Chen Q, Jin M, Yang F, Zhu J, Xiao Q, Zhang L. Matrix metalloproteinases: Inflammatory regulators of cell behaviors in vascular formation and remodeling. Mediators Inflamm. 2013;2013:928315. doi:10.1155/2013/928315
  27. Chi PL, Cheng CC, Hung CC, et al. MMP-10 from M1 macrophages promotes pulmonary vascular remodeling and pulmonary arterial hypertension. Int J Biol Sci. 2022;18(1):331–348. doi:10.7150/ijbs.66472
  28. Ma B, Cao Y, Qin J, Chen Z, Hu G, Li Q. Pulmonary artery smooth muscle cell phenotypic switching: A key event in the early stage of pulmonary artery hypertension. Drug Discov Today. 2023;28(5):103559. doi:10.1016/j.drudis.2023.103559
  29. Zhang D, Gou Z, Qu Y, Su X. Understanding how methyltransferase-like 3 functions in lung diseases: From pathogenesis to clinical application. Biomed Pharmacother. 2024;179:117421. doi:10.1016/j.biopha.2024.117421
  30. Gao Y, Chen T, Raj JU. Endothelial and smooth muscle cell interactions in the pathobiology of pulmonary hypertension. Am J Respir Cell Mol Biol. 2016;54(4):451–460. doi:10.1165/rcmb.2015-0323TR
  31. Stenmark KR, Frid M, Perros F. Endothelial-to-mesenchymal transition: An evolving paradigm and a promising therapeutic target in PAH. Circulation. 2016;133(18):1734–1737. doi:10.1161/CIRCULATIONAHA.116.022479
  32. Chelladurai P, Seeger W, Pullamsetti SS. Matrix metalloproteinases and their inhibitors in pulmonary hypertension. Eur Respir J. 2012;40(3):766–782. doi:10.1183/09031936.00209911
  33. Kumar R, Graham B. How does inflammation contribute to pulmonary hypertension? Eur Respir J. 2018;51(1):1702403. doi:10.1183/13993003.02403-2017
  34. Devendran A, Kar S, Bailey R, Trivieri MG. The role of bone morphogenetic protein receptor type 2 (BMPR2) and the prospects of utilizing induced pluripotent stem cells (iPSCs) in pulmonary arterial hypertension disease modeling. Cells. 2022;11(23):3823. doi:10.3390/cells11233823
  35. Humbert M, Sitbon O, Chaouat A, et al. Survival in patients with idiopathic, familial, and anorexigen-associated pulmonary arterial hypertension in the modern management era. Circulation. 2010;122(2):156–163. doi:10.1161/CIRCULATIONAHA.109.911818
  36. Humbert M, Gerry Coghlan J, Khanna D. Early detection and management of pulmonary arterial hypertension. Eur Respir Rev. 2012;21(126):306–312. doi:10.1183/09059180.00005112
  37. Foris V, Kovacs G, Tscherner M, Olschewski A, Olschewski H. Biomarkers in pulmonary hypertension. Chest. 2013;144(1):274–283. doi:10.1378/chest.12-1246
  38. Kazimierczyk R, Błaszczak P, Kowal K, et al. The significance of diminished sTWEAK and P-selectin content in platelets of patients with pulmonary arterial hypertension. Cytokine. 2018;107:52–58. doi:10.1016/j.cyto.2017.11.014
  39. Newby A. Matrix metalloproteinases regulate migration, proliferation, and death of vascular smooth muscle cells by degrading matrix and non-matrix substrates. Cardiovasc Res. 2006;69(3):614–624. doi:10.1016/j.cardiores.2005.08.002
  40. Bohara S, Bagheri A, Ertugral EG, et al. Integrative analysis of gene expression, protein abundance, and metabolomic profiling elucidates complex relationships in chronic hyperglycemia-induced changes in human aortic smooth muscle cells. J Biol Eng. 2024;18(1):61. doi:10.1186/s13036-024-00457-w
  41. Kirkgöz K. C-reactive protein in atherosclerosis: More than a biomarker, but not just a culprit. Rev Cardiovasc Med. 2023;24(10):297. doi:10.31083/j.rcm2410297
  42. Zhao Y, Peng J, Lu C, et al. Metabolomic heterogeneity of pulmonary arterial hypertension. PLoS One. 2014;9(2):e88727. doi:10.1371/journal.pone.0088727
  43. Kazimierczyk R, Szumowski P, Nekolla SG, et al. Prognostic role of PET/MRI hybrid imaging in patients with pulmonary arterial hypertension. Heart. 2021;107(1):54–60. doi:10.1136/heartjnl-2020-316741
  44. Hemnes AR, Luther JM, Rhodes CJ, et al. Human PAH is characterized by a pattern of lipid-related insulin resistance. JCI Insight. 2019;4(1):e123611. doi:10.1172/jci.insight.123611
  45. Hassoun PM, Mouthon L, Barberà JA, et al. Inflammation, growth factors, and pulmonary vascular remodeling. J Am Coll Cardiol. 2009;54(1 Suppl):S10–S19. doi:10.1016/j.jacc.2009.04.006
  46. Malhotra R, Paskin-Flerlage S, Zamanian RT, et al. Circulating angiogenic modulatory factors predict survival and functional class in pulmonary arterial hypertension. Pulm Circ. 2013;3(2):369–380. doi:10.4103/2045-8932.110445
  47. Blackburn JS, Brinckerhoff CE. Matrix metalloproteinase-1 and thrombin differentially activate gene expression in endothelial cells via PAR-1 and promote angiogenesis. Am J Pathol. 2008;173(6):1736–1746. doi:10.2353/ajpath.2008.080512
  48. Mazor R, Alsaigh T, Shaked H, et al. Matrix metalloproteinase-1-mediated up-regulation of vascular endothelial growth factor-2 in endothelial cells. J Biol Chem. 2013;288(1):598–607. doi:10.1074/jbc.M112.417451
  49. Merkus D, Van Beusekom HMM, Boomars KA. Protease-activated receptor 1 as potential therapeutic target in pulmonary arterial hypertension. Cardiovasc Res. 2019;115(8):1260–1261. doi:10.1093/cvr/cvz071
  50. Mickael C, Kheyfets VO, Langouët-Astrié C, et al. Peripheral blood inflammation profile of patients with pulmonary arterial hypertension using the High-Throughput Olink Proteomics Platform. Am J Respir Cell Mol Biol. 2022;66(5):580–581. doi:10.1165/rcmb.2021-0369LE
  51. Lepetit H, Eddahibi S, Fadel E, et al. Smooth muscle cell matrix metalloproteinases in idiopathic pulmonary arterial hypertension. Eur Respir J. 2005;25(5):834–842. doi:10.1183/09031936.05.00072504
  52. Hoffmann J, Marsh LM, Pieper M, et al. Compartment-specific expression of collagens and their processing enzymes in intrapulmonary arteries of IPAH patients. Am J Physiol Lung Cell Mol Physiol. 2015;308(10):L1002–L1013. doi:10.1152/ajplung.00383.2014
  53. Lenci E, Cosottini L, Trabocchi A. Novel matrix metalloproteinase inhibitors: An updated patent review (2014–2020). Exp Opin Ther Patents. 2021;31(6):509–523. doi:10.1080/13543776.2021.1881481
  54. Hardy E, Hardy-Sosa A, Fernandez-Patron C. MMP-2: Is too low as bad as too high in the cardiovascular system? Am J Physiol Heart Circ Physiol. 2018;315(5):H1332–H1340. doi:10.1152/ajpheart.00198.2018
  55. Chakraborti S, Sarkar J, Pramanik PK, Chakraborti T. Role of proteases in lung disease: A brief overview. In: Chakraborti S, Chakraborti T, Dhalla NS, eds. Proteases in Human Diseases. Singapore: Springer Singapore; 2017:333–374. doi:10.1007/978-981-10-3162-5_16
  56. Shamamian P, Schwartz JD, Pocock BJZ, et al. Activation of progelatinase A (MMP-2) by neutrophil elastase, cathepsin G, and proteinase-3: A role for inflammatory cells in tumor invasion and angiogenesis. J Cell Physiol. 2001;189(2):197–206. doi:10.1002/jcp.10014
  57. Monea S, Lehti K, Keski-Oja J, Mignatti P. Plasmin activates pro-matrix metalloproteinase-2 with a membrane-type 1 matrix metalloproteinase-dependent mechanism. J Cell Physiol. 2002;192(2):160–170. doi:10.1002/jcp.10126
  58. Miyamori H, Takino T, Kobayashi Y, et al. Claudin promotes activation of pro-matrix metalloproteinase-2 mediated by membrane-type matrix metalloproteinases. J Biol Chem. 2001;276(30):28204–28211. doi:10.1074/jbc.M103083200
  59. Grote K, Flach I, Luchtefeld M, et al. Mechanical stretch enhances mRNA expression and proenzyme release of matrix metalloproteinase-2 (MMP-2) via NAD(P)H oxidase-derived reactive oxygen species. Circ Res. 2003;92(11):e80–86. doi:10.1161/01.RES.0000077044.60138.7C
  60. Arvidsson M, Ahmed A, Bouzina H, Rådegran G. Matrix metalloproteinase 7 in diagnosis and differentiation of pulmonary arterial hypertension. Pulm Circ. 2019;9(4):2045894019895414. doi:10.1177/2045894019895414
  61. Schumann C, Lepper PM, Frank H, et al. Circulating biomarkers of tissue remodelling in pulmonary hypertension. Biomarkers. 2010;15(6):523–532. doi:10.3109/1354750X.2010.492431
  62. Tiede SL, Wassenberg M, Christ K, et al. Biomarkers of tissue remodeling predict survival in patients with pulmonary hypertension. Int J Cardiol. 2016;223:821–826. doi:10.1016/j.ijcard.2016.08.240
  63. Arvidsson M, Ahmed A, Säleby J, Hesselstrand R, Rådegran G. Plasma matrix metalloproteinase 2 is associated with severity and mortality in pulmonary arterial hypertension. Pulm Circ. 2022;12(1):e12041. doi:10.1002/pul2.12041
  64. Karamanian VA, Harhay M, Grant GR, et al. Erythropoietin upregulation in pulmonary arterial hypertension. Pulm Circ. 2014;4(2):269–279. doi:10.1086/675990
  65. Asosingh K, Aldred MA, Vasanji A, et al. Circulating angiogenic precursors in idiopathic pulmonary arterial hypertension. Am J Pathol. 2008;172(3):615–627. doi:10.2353/ajpath.2008.070705
  66. Cantini-Salignac C, Lartaud I, Schrijen F, Atkinson J, Chabot F. Metalloproteinase-9 in circulating monocytes in pulmonary hypertension. Fundam Clin Pharmacol. 2006;20(4):405–410. doi:10.1111/j.1472-8206.2006.00417.x
  67. Bauer Y, De Bernard S, Hickey P, et al. Identifying early pulmonary arterial hypertension biomarkers in systemic sclerosis: Machine learning on proteomics from the DETECT cohort. Eur Respir J. 2021;57(6):2002591. doi:10.1183/13993003.02591-2020
  68. Giannelli G, Iannone F, Marinosci F, Lapadula G, Antonaci S. The effect of bosentan on matrix metalloproteinase-9 levels in patients with systemic sclerosis-induced pulmonary hypertension. Curr Med Res Opin. 2005;21(3):327–332. doi:10.1185/030079905X30680
  69. Wan J, Zhang G, Li X, et al. Matrix metalloproteinase 3: A promoting and destabilizing factor in the pathogenesis of disease and cell differentiation. Front Physiol. 2021;12:663978. doi:10.3389/fphys.2021.663978
  70. Ito TK, Ishii G, Saito S, et al. Degradation of soluble VEGF receptor-1 by MMP-7 allows VEGF access to endothelial cells. Blood. 2009;5;113(10):2363-9. doi: 10.1182/blood-2008-08-172742
  71. Bonnema DD, Webb CS, Pennington WR, et al. Effects of age on plasma matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs). J Cardiac Fail. 2007;13(7):530–540. doi:10.1016/j.cardfail.2007.04.010
  72. Dieffenbach PB, Mallarino Haeger C, Rehman R, et al. A novel protective role for matrix metalloproteinase-8 in the pulmonary vasculature. Am J Respir Crit Care Med. 2021;204(12):1433–1451. doi:10.1164/rccm.202108-1863OC
  73. Ge Q, Zhang T, Yu J, et al. A new perspective on targeting pulmonary arterial hypertension: Programmed cell death pathways (autophagy, pyroptosis, ferroptosis) [Erratum in: Biomed Pharmacother. 2024;181:117741. doi:10.1016/j.biopha.2024.117741]. Biomed Pharmacother. 2024;181:117706. doi:10.1016/j.biopha.2024.117706
  74. Safdar Z, Tamez E, Chan W, et al. Circulating collagen biomarkers as indicators of disease severity in pulmonary arterial hypertension. JACC Heart Fail. 2014;2(4):412–421. doi:10.1016/j.jchf.2014.03.013
  75. Hiremath J, Thanikachalam S, Parikh K, et al. Exercise improvement and plasma biomarker changes with intravenous treprostinil therapy for pulmonary arterial hypertension: A placebo-controlled trial. J Heart Lung Transplant. 2010;29(2):137–149. doi:10.1016/j.healun.2009.09.005
  76. Vandooren J, Van Den Steen PE, Opdenakker G. Biochemistry and molecular biology of gelatinase B or matrix metalloproteinase-9 (MMP-9): The next decade. Crit Rev Biochem Mol Biol. 2013;48(3):222–272. doi:10.3109/10409238.2013.770819
  77. Grigoryev DN, Mathai SC, Fisher MR, et al. Identification of candidate genes in scleroderma-related pulmonary arterial hypertension. Transl Res. 2008;151(4):197–207. doi:10.1016/j.trsl.2007.12.010
  78. Jonigk D, Golpon H, Bockmeyer CL, et al. Plexiform lesions in pulmonary arterial hypertension. Am J Pathol. 2011;179(1):167–179. doi:10.1016/j.ajpath.2011.03.040
  79. Garbacki N, Di Valentin E, Piette J, Cataldo D, Crahay C, Colige A. Matrix metalloproteinase 12 silencing: A therapeutic approach to treat pathological lung tissue remodeling? Pulm Pharmacol Ther. 2009;22(4):267–278. doi:10.1016/j.pupt.2009.03.001
  80. Stracke JO, Hutton M, Stewart M, et al. Biochemical characterization of the catalytic domain of human matrix metalloproteinase 19. J Biol Chem. 2000;275(20):14809–14816. doi:10.1074/jbc.275.20.14809
  81. Ries C. Cytokine functions of TIMP-1. Cell Mol Life Sci. 2014;71(4):659–672. doi:10.1007/s00018-013-1457-3
  82. Almuntashiri S, Alhumaid A, Zhu Y, et al. TIMP-1 and its potential diagnostic and prognostic value in pulmonary diseases. Chin Med J Pulm Crit Care Med. 2023;1(2):67–76. doi:10.1016/j.pccm.2023.05.002
  83. Fassina G, Ferrari N, Brigati C, et al. Tissue inhibitors of metalloproteases: Regulation and biological activities. Clin Exp Metastasis. 2000;18(2):111–120. doi:10.1023/a:1006797522521
  84. Lindsay MM, Maxwell P, Dunn FG. TIMP-1: A marker of left ventricular diastolic dysfunction and fibrosis in hypertension. Hypertension. 2002;40(2):136–141. doi:10.1161/01.HYP.0000024573.17293.23
  85. Sundström J, Evans JC, Benjamin EJ, et al. Relations of plasma total TIMP-1 levels to cardiovascular risk factors and echocardiographic measures: The Framingham Heart Study. Eur Heart J. 2004;25(17):1509–1516. doi:10.1016/j.ehj.2004.05.029
  86. Caimi G, Hopps E, Montana M, et al. The function of matrix metalloproteinase-9 (MMP-9) and its tissue inhibitor (TIMP-1) in several clinical conditions: Results and analysis of our survey. Clin Hemorheol Microcirc. 2021;78(4):401–416. doi:10.3233/CH-201094
  87. Cavusoglu E, Ruwende C, Chopra V, et al. Tissue inhibitor of metalloproteinase-1 (TIMP-1) is an independent predictor of all-cause mortality, cardiac mortality, and myocardial infarction. Am Heart J. 2006;151(5):1101.e1–1101.e8. doi:10.1016/j.ahj.2006.02.029
  88. Fenster BE, Lasalvia L, Schroeder JD, et al. Galectin-3 levels are associated with right ventricular functional and morphologic changes in pulmonary arterial hypertension. Heart Vessels. 2016;31(6):939–946. doi:10.1007/s00380-015-0691-z
  89. Pendergrass SA, Hayes E, Farina G, et al. Limited systemic sclerosis patients with pulmonary arterial hypertension show biomarkers of inflammation and vascular injury. PLoS One. 2010;5(8):e12106. doi:10.1371/journal.pone.0012106
  90. Wetzl V, Tiede SL, Faerber L, et al. Plasma MMP2/TIMP4 ratio at follow-up assessment predicts disease progression of idiopathic pulmonary arterial hypertension. Lung. 2017;195(4):489–496. doi:10.1007/s00408-017-0014-5
  91. Jacobsen J, Visse R, Sørensen HP, et al. Catalytic properties of ADAM12 and its domain deletion mutants. Biochemistry (Moscow). 2008;47(2):537–547. doi:10.1021/bi701629c
  92. Boon L, Ugarte-Berzal E, Vandooren J, Opdenakker G. Protease propeptide structures, mechanisms of activation, and functions. Crit Rev Biochem Mol Biol. 2020;55(2):111–165. doi:10.1080/10409238.2020.1742090
  93. Moore L, Fan D, Basu R, Kandalam V, Kassiri Z. Tissue inhibitor of metalloproteinases (TIMPs) in heart failure. Heart Fail Rev. 2012;17(4–5):693–706. doi:10.1007/s10741-011-9266-y
  94. Kashiwagi M, Tortorella M, Nagase H, Brew K. TIMP-3 is a potent inhibitor of aggrecanase 1 (ADAM-TS4) and aggrecanase 2 (ADAM-TS5). J Biol Chem. 2001;276(16):12501–12504. doi:10.1074/jbc.C000848200
  95. Edgar AJ, Chacón MR, Bishop AE, Yacoub MH, Polak JM. Upregulated genes in sporadic, idiopathic pulmonary arterial hypertension. Respir Res. 2006;7(1):1. doi:10.1186/1465-9921-7-1
  96. Melendez-Zajgla J, Pozo L, Ceballos G, Maldonado V. Tissue inhibitor of metalloproteinases-4: The road less traveled. Mol Cancer. 2008;7(1):85. doi:10.1186/1476-4598-7-85
  97. Mochizuki S, Shimoda M, Shiomi T, Fujii Y, Okada Y. ADAM28 is activated by MMP-7 (matrilysin-1) and cleaves insulin-like growth factor binding protein-3. Biochem Biophys Res Commun. 2004;315(1):79–84. doi:10.1016/j.bbrc.2004.01.022
  98. Elias GJ, Ioannis M, Theodora P, et al. Circulating tissue inhibitor of matrix metalloproteinase-4 (TIMP-4) in systemic sclerosis patients with elevated pulmonary arterial pressure. Mediators Inflamm. 2008;164134. doi: 10.1155/2008/164134
  99. Paulissen G, Rocks N, Gueders MM, et al. Role of ADAM and ADAMTS metalloproteinases in airway diseases. Respir Res. 2009;10(1):127. doi:10.1186/1465-9921-10-127
  100. Black RA, White JM. ADAMs: focus on the protease domain. Curr Opin Cell Biol. 1998;10(5):654–659. doi:10.1016/S0955-0674(98)80042-2
  101. Zhong S, Khalil RA. A disintegrin and metalloproteinase (ADAM) and ADAM with thrombospondin motifs (ADAMTS) family in vascular biology and disease. Biochem Pharmacol. 2019;164:188–204. doi:10.1016/j.bcp.2019.03.033
  102. Omura J, Satoh K, Kikuchi N, et al. ADAMTS8 promotes the development of pulmonary arterial hypertension and right ventricular failure: A possible novel therapeutic target. Circ Res. 2019;125(10):884–906. doi:10.1161/CIRCRESAHA.119.315398
  103. Satoh K. Drug discovery focused on novel pathogenic proteins for pulmonary arterial hypertension. J Cardiol. 2021;78(1):1–11. doi:10.1016/j.jjcc.2021.01.009
  104. Newnham M, South K, Bleda M, et al. The ADAMTS13–VWF axis is dysregulated in chronic thromboembolic pulmonary hypertension. Eur Respir J. 2019;53(3):1801805. doi:10.1183/13993003.01805-2018
  105. Ahmed A, Ahmed S, Rådegran G. Plasma ADAMTS13 and von Willebrand factor in diagnosis and prediction of prognosis in pulmonary arterial hypertension. Pulm Circ. 2021;11(4):20458940211041500. doi:10.1177/20458940211041500
  106. Li Y, Chen Y, Yang L, et al. Increased plasma expression of a disintegrin and metalloproteinase with thrombospondin motifs like 4 in patients with idiopathic pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension. Pulm Circ. 2023;13(3):e12267. doi:10.1002/pul2.12267
  107. Liao S, Lin Y, Liu L, et al. ADAM10, a “multitasker” in sepsis: Focus on its posttranslational target. Inflamm Res. 2023;72(3):395–423. doi:10.1007/s00011-022-01673-0
  108. Tripathi P, Awasthi S, Gao P. ADAM metallopeptidase domain 33 (ADAM33): A promising target for asthma. Mediators Inflamm. 2014;2014:572025. doi:10.1155/2014/572025
  109. Rhodes CJ, Wharton J, Ghataorhe P, et al. Plasma proteome analysis in patients with pulmonary arterial hypertension: An observational cohort study. Lancet Respir Med. 2017;5(9):717–726. doi:10.1016/S2213-2600(17)30161-3
  110. Kopeć G, Skride A, Ereminiene E, et al. Emerging therapies and new directions in the treatment of pulmonary arterial hypertension. Pol Heart J. 2025;83(1):18–26. doi:10.33963/v.phj.104053
  111. Zolty R. Novel experimental therapies for treatment of pulmonary arterial hypertension. J Exp Pharmacol. 2021;13:817–857. doi:10.2147/JEP.S236743
  112. Shah AJ, Beckmann T, Vorla M, Kalra DK. New drugs and therapies in pulmonary arterial hypertension. Int J Mol Sci. 2023;24(6):5850. doi:10.3390/ijms24065850