Biomarkers of chronic obstructive pulmonary disease with pulmonary hyp

Introduction

Chronic obstructive pulmonary disease (COPD) is a progressive respiratory condition characterized by persistent symptoms and airflow limitation that is not fully reversible with bronchodilator therapy. It has become a significant global health issue.1 According to the World Health Organization, COPD was the third leading cause of death worldwide in 2019, accounting for 3.23 million deaths. Additionally, it ranks as the seventh leading cause of poor health globally, as measured by disability-adjusted life years. The prevalence of COPD is notably higher among current and former smokers, and it increases with age.2

It has been shown that the pathogenesis of COPD is closely related to abnormal inflammatory response, protease- antiprotease imbalance, and oxidative stress damage. Inflammatory cells release pro-inflammatory factors such as interleukin (IL-6, IL-8) and tumor necrosis factor alpha (TNF-α), leading to thickening of the airway wall, excessive mucus secretion, and ciliary dysfunction. Chronic inflammation further damages the alveolar structure, leading to emphysema and small airway fibrosis. In addition, protease-antiprotease imbalance and the imbalance between oxidative stress and antioxidation, exacerbate lung tissue damage and inflammation amplification. As the pathophysiology of COPD progresses, it can lead to an increase in mean pulmonary arterial pressure (mPAP). This increase is primarily driven by three key mechanisms: first, hypoxic pulmonary vasoconstriction contributes to increased pulmonary circulation resistance; second, the loss of small pulmonary vessels and pulmonary vascular remodeling—characterized by intimal proliferation of poorly differentiated smooth muscle cells and deposition of elastic and collagen fibers—also plays a significant role; third, hypoxia induces an increase in red blood cell count and blood viscosity. These three factors can contribute to pulmonary hypertension (PH) either individually or simultaneously.3 The prevalence of PH is very high in patients with advanced COPD. A meta-analysis shows that the combined prevalence of COPD-related PH is 39.2%, and the prevalence of PH increases with the severity of COPD.4 A study in patients with severe airway obstruction indicates that up to 90% of such patients have an mPAP greater than 20 mmHg.5 In COPD, PH is typically of moderate severity and progresses slowly, often without affecting right ventricular function in most patients. However, a small percentage (1–3%) may experience disproportionate PH, where pulmonary arterial pressure significantly exceeds the degree of airway impairment.6 Persistent PH leads to increased right ventricular afterload, ultimately causing right ventricular remodeling and functional failure, known as chronic cor pulmonale (CCP). Furthermore, since COPD and cardiovascular disease share common risk factors—such as advanced age, smoking, and systemic inflammation—COPD can lead to chronic systemic inflammation in addition to pulmonary inflammation. This systemic inflammation may result in myocardial inflammation and subsequent fibrosis, which affect the mechanical, electrical, and vasomotor functions of the myocardium. Consequently, this also increases the risk of cardiovascular disease in COPD patients, particularly right heart failure due to PH.7 Importantly, a key characteristic of COPD is that an individual’s prognosis is significantly influenced by comorbidities, with PH and cardiovascular disease increasingly recognized as exacerbating factors associated with higher mortality rates in COPD patients.8 A research displays that the 5-year all-cause mortality of COPD patients is significantly higher in combined with PH group than without PH group (32.0% vs 13.0%).9 The mPAP and pulmonary vascular resistance values are negatively correlated with survival rates. Reports indicate that the five-year survival rate for COPD patients with mPAP values greater than 25 mmHg is only 36%. Additionally, pulmonary hemodynamics have been shown to be a stronger predictor of survival than forced expiratory volume in one second (FEV1) or gas exchange variables.10

Recognizing PH can be challenging, as its symptoms often overlap with those of COPD. A high index of suspicion for PH is warranted if clinical deterioration does not correlate with a decline in pulmonary function, especially in cases of profound hypoxemia or significant reductions in carbon monoxide diffusion capacity.6 Patients with suspected PH should undergo evaluation by Doppler echocardiography, a widely accepted non-invasive diagnostic tool that uses ultrasound to generate cardiac images. PH can be diagnosed when pulmonary artery systolic pressure exceeds 50 mmHg, in conjunction with a clinical diagnosis. The gold standard for diagnosing PH, however, is catheterization, which involves inserting a catheter into the pulmonary artery to directly measure pressure, with PH defined as an mPAP of 25 mmHg or higher. Despite its accuracy, this invasive technique presents challenges in clinical practice due to its complexity and associated risks.11 In view of the high incidence of PH and CCP in COPD patients, along with the associated adverse outcomes, there is an urgent need for convenient and non-invasive diagnostic methods that offer good specificity and sensitivity. Circulating biomarkers and intracellular molecular markers in COPD patients are expected to play a key role in diagnosing the presence of PH and CCP, complementing existing diagnostic methods.

Brain Natriuretic Peptide

Brain natriuretic peptide (BNP) is a biologically active molecule composed of 32 amino acids, primarily synthesized in the cardiac ventricles and released into circulation in response to pressure overload, volume expansion, and increased myocardial wall stress. Its precursor, pro-BNP, is cleaved by blood proteases to produce BNP and N-terminal pro-BNP (NT-proBNP). BNP has a half-life of approximately 22 minutes, while NT-proBNP has a half-life of about 120 minutes, making NT-proBNP relatively stable and potentially more accurate for disease diagnosis.12 COPD accompanied by PH results in increased pulmonary vascular pressure and right heart afterload, which heightens the strain on the ventricular wall and triggers the release of NT-proBNP. This mechanism may be a significant contributor to the upregulation of NT-proBNP levels in these patients.13 Secondly, hypoxia has been identified as a potential contributor to elevated NT-proBNP levels. A study by Hopkins et al on adult patients with cyanotic congenital heart disease demonstrated a significant increase in BNP levels, highlighting that hypoxia serves as a direct stimulus for BNP secretion in human cardiac myocytes.14 Casals et al conducted in vitro experiments using cultured human ventricular myocytes and demonstrated that hypoxia may stimulate the synthesis and secretion of BNP in these cells through the enhanced transcriptional activity of hypoxia-inducible factor 1 (HIF-1).15 Oxygen therapy can significantly reduce pulmonary artery pressure and NT-proBNP levels in COPD patients. Non-invasive positive pressure ventilation can decrease the risk of acute exacerbation and improve prognosis in patients with severe-to-very-severe COPD complicated by PH.16 Finally, the elevation of NT-proBNP levels can also be linked to the activity of various pro-inflammatory cytokines. Previous studies have indicated that cytokines such as IL-1β, TNF-α, and IL-6 may act as stimuli for the release of NT-proBNP from cardiac myocytes.17

Historically, BNP has been closely associated with heart failure (HF). In cases of left ventricular heart failure (LVHF), elevated BNP levels have been linked to reduced exercise tolerance and a poorer prognosis.18 Additionally, it has been reported that even in the absence of LVEF, PH secondary to end-stage lung disease can further exacerbate right ventricular afterload, potentially leading to right heart failure in COPD patients. This condition can also result in increased BNP concentrations.19 A meta-analysis showed that compared to COPD patients without PH and HF, patients with combined PH and HF had significantly elevated NT-proBNP levels. Moreover, compared to the stable COPD group, the NT-proBNP levels in acute exacerbation of chronic obstructive pulmonary disease (AECOPD) patients were significantly elevated. Elevated NT-proBNP levels are associated with a higher mortality risk in hospitalized AECOPD patients and serve as an important prognostic indicator for poor outcomes in these individuals.20 The study by Agoston-Coldea et al demonstrated that NT-proBNP levels have significant predictive value for right ventricular dysfunction, with an area under the receiver operating characteristic (ROC) curve (AUC) of 0.945 (p < 0.0001), indicating extremely strong diagnostic accuracy. When using 311 pg/mL as the optimal cutoff value, NT-proBNP exhibited 100% sensitivity and 84.0% specificity. These findings suggest that at this threshold, NT-proBNP can serve as a highly effective screening tool for right ventricular dysfunction.21 It is worth pondering whether NT-proBNP can differentiate between right heart failure caused by isolated HF and right heart failure induced by COPD combined with CCP. In the study by Dewan et al, 12.3% of HF patients with reduced ejection fraction were found to have COPD. Compared to those without COPD, participants with COPD exhibited higher levels of NT-proBNP, experienced more severe symptoms and functional limitations, and faced a higher risk of composite outcomes, including HF deterioration and cardiovascular death.22 Another study found no significant difference in NT-proBNP levels between patients with HF combined with COPD and those with HF alone.20 Therefore, although NT-proBNP cannot differentiate between right heart failure caused by isolated HF and that caused by COPD combined with CCP, it can still serve as a useful exclusion criterion for COPD-induced PH and CCP. A normal NT-proBNP level effectively rules out these complications.

Inflammatory Mediators

The histopathological characteristics of PH include thickening of the vascular intima and media, muscularization of distal pulmonary arteries, vascular occlusion, and the presence of complex plexiform lesions. These features are closely associated with pulmonary artery smooth muscle cells (PASMCs), endothelial cells (ECs), and immune cells.23,24 ECs and immune cells secrete growth factors and inflammatory cytokines, triggering a phenotypic shift in PASMCs from a contractile or differentiated state to a proliferative or dedifferentiated state. This shift promotes and sustains PASMC proliferation and contributes to vascular remodeling.

Macrophages are key effectors of pulmonary inflammation in patients with PH. Among them, alveolar macrophages contribute to local immune homeostasis and possess surfactant properties, while pulmonary interstitial macrophages, such as perivascular macrophages, play a predominant role in driving pulmonary inflammation.25 Florentin et al’s study further demonstrated that hypoxia results in a gradual depletion of alveolar macrophages and a substantial increase in interstitial macrophages. The reduction in alveolar macrophages under hypoxic conditions may be attributed to increased expression of cleaved caspase-3, which promotes apoptosis, along with decreased levels of granulocyte colony-stimulating factor (G-CSF), which is insufficient to stimulate macrophage proliferation. The increase in interstitial macrophages under hypoxic conditions may be attributed to elevated levels of chemokines and their receptors. Both animal experiments and clinical studies of PH patients have shown higher chemokine levels: hypoxic mice exhibited elevated pulmonary levels of chemokines such as chemokine (C-X3-C motif) ligand 1 (CX3CL1) and C-C chemokine ligand (CCL2), while PH patients showed increased levels of pulmonary chemokines including CCL1, CCL2, CCL3, CCL4, CCL18, and CX3CL1, which promote monocyte migration. Additionally, expression of corresponding chemokine receptors like C-C chemokine receptor 1 (CCR1), CCR2, CCR5, and CX3CR1 was found to be elevated in the circulating monocytes of PH patients. The increase in lung chemokines and the upregulation of chemokine receptor expression on blood monocytes trigger the mobilization of inflammatory monocytes to the lungs in PH patients, where they subsequently differentiate into interstitial perivascular macrophages.25 Inflammation in the right ventricle (RV), driven by the activation of the nucleotide-binding domain, leucine-rich repeat-containing family, pyrin domain-containing-3 (NLRP3) inflammasome in macrophages, contributes to increased RV fibrosis and worsening RV function. The specific pathway of action includes the following steps: 1. Priming and Activation: Initiated by various signals, such as damage-associated molecular patterns, potassium efflux, calcium influx, and mitochondrial dysfunction; 2. Aggregation: Formation of the apoptosis-associated speck-like protein containing a caspase activation and recruitment domain, which is a key adaptor molecule in inflammasome signaling; 3. Gasdermin D Pore Formation and IL-1β Release: Activation leads to the formation of gasdermin D pores and subsequent release of IL-1β; 4. Induction of Pro-inflammatory Response: IL-1β triggers a pro-inflammatory response in the RV, including the release of additional inflammatory cytokines such as IL-6; 5. Downstream IL-6 Signaling: IL-6 signals downstream through signal transducer and activator of transcription 3 (STAT3) in monocytes, aiding in the recruitment of new monocytes/macrophages to the RV. Animal models have confirmed that the inflammatory and fibrotic changes associated with PH are chamber-specific. When comparing the RV and left ventricle, only the RV shows elevated levels of collagen-III, atrial natriuretic peptide, and macrophage counts.26 Xingchen et al demonstrated that treatment with the glutaminase 1 inhibitor BPTES significantly ameliorated pulmonary artery pressure, right ventricular function, and pulmonary vascular remodeling in a rat model of PH. Concurrently, this intervention suppressed M1 macrophage polarization, NLRP3 inflammasome activation, and the release of pro-inflammatory cytokines. These findings further validate that macrophage inhibition may represent a promising therapeutic approach for PH.27

The T-helper 17 (Th17) and T-helper 2 (Th2) subsets of CD4+ T cells are pivotal in the development of chronic hypoxia-induced PH. IL-6, in combination with transforming growth factor-β (TGF-β), promotes the differentiation of naive T cells into Th17 cells and sustains STAT3 activation.28 Th17 cells then produce the pro-inflammatory cytokine IL-17, which has multiple roles in promoting airway inflammation. IL-17 facilitates the recruitment of neutrophils by inducing CXC chemokines such as CXCL2, which is also known as macrophage inflammatory protein-2 (MIP-2), and upregulating G-CSF expression. Additionally, IL-17 binds to TNF-α, stabilizing mRNA and enhancing the production of other pro-inflammatory cytokines like IL-6 and IL-8, thereby intensifying airway inflammation.29–32 Moreover, IL-17 stimulates the release of matrix metalloproteinase-12 (MMP-12), contributing to tissue degradation.33,34 It also activates the Wnt3a/β-catenin/CyclinD1 pathway in pulmonary artery endothelial cells (PAECs), leading to PAECs dysfunction.35 This dysfunction further stimulates PASMCs proliferation, intensifying hypoxia-induced vascular remodeling. The cytokine IL-4, produced by Th2 cells, is a multifunctional cytokine that not only stimulates antibody production by B lymphocytes but also plays a pivotal role in Th2-mediated inflammatory responses.36 IL-4 is crucial for the collagen accumulation and proliferative effects of hypoxia-induced mitogenic factor (HIMF) in pulmonary arteries (PAs). Additionally, HIMF significantly enhances the expression of vascular endothelial growth factor (VEGF) and themonocyte chemoattractant protein-1 (MCP-1, also known as CCL2) in pulmonary microvascular endothelial cells through an IL-4-dependent pathway, facilitating the recruitment of circulating monocytes. This suggests that HIMF, in the presence of IL-4, fosters a pro-inflammatory and chemotactic microenvironment. Therefore, IL-4 signaling likely plays a critical role in HIMF-induced pulmonary inflammation and vascular remodeling.37–39The pathways by which the above inflammatory factors affect vascular remodeling are summarized in Figure 1.

Figure 1 The pathways of inflammatory factors involved in vascular remodeling.

Abbreviations: ASC, Apoptosis-associated speck-like protein; B cell, B lymphocyte; CCL2, Chemokine (C-C motif) ligand 2; CXCL2, Chemokine (C-X-C motif) ligand 2; IL, Interleukin; G-CSF, Granulocyte Colony-Stimulating Factor; MMP-12, matrix metalloproteinase −12; NLRP3, Nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3; STAT3, Signal and transducer of transcription 3; TGF-β, Transforming growth factor-β; Th cell, T helper cell; TNF-α, Tumor necrosis factor-α; VEGF, Vascular endothelial growth factor.

It is worth noting that the above inflammatory cytokines produced by immune cell-related pathways are closely associated with the diagnosis of comorbidities and disease severity in COPD patients, demonstrating significant clinical significance. Xu et al’s research has demonstrated that CCL18 and CX3CL1 are significantly elevated in patients with COPD and CCP (COPD&CCP). The combination of CCL18 and CX3CL1 shows high precision for discriminating COPD&CCP with high AUC values (0.828), sensitivity (66.1%), and specificity (92.5%). The combined detection of the two can achieve high-specificity differentiation of COPD&CCP and reduce misdiagnosis (such as differentiation from simple COPD or LVEF). Moreover, both are independent predictors of poor clinical outcomes, associated with reduced therapeutic benefits and an unfavorable prognosis in COPD&CCP patients.40 Eissa et al’s study shows that IL-1 can be used for early warning and initial screening of COPD-PH. ROC analysis of IL-1 in predicting the probability of PH shows a significant AUC value of 0.722, indicating its moderate predictive ability for PH risk. The best cutoff point is >86, with a sensitivity of 78.6% at this point, meaning it can identify approximately 79% of COPD-PH patients. However, the specificity is 58.1%, indicating some false positives, so clinical exclusion of interference such as infection is required.41 Yang et al’s study confirmed that IL-6 demonstrated significant value in the diagnosis and treatment of COPD&PH. ROC analysis showed that the AUC of IL-6 for diagnosing COPD&PH reached 0.929, approaching perfect predictive efficacy. When the cutoff value was set at 98.99 pg/mL, its sensitivity was 87.27%, meaning it can accurately identify over 80% of COPD&PH patients, and the specificity was 89.47%, effectively reducing misdiagnosis. These results indicated that IL-6 can serve as a non-invasive and highly efficient screening marker to assist in the early clinical detection of high-risk populations for COPD&PH. The study further confirmed that elevated IL-6 levels were an independent risk factor for the onset of COPD&PH (OR value: 2.564, 95% confidence interval: 1.114–4.789), suggesting that IL-6 is not only useful for diagnosis but also a potential target for monitoring disease progression. Clinically, dynamic monitoring of IL-6 levels helps assess disease severity and guide treatment decisions.42 Interestingly, a study by Zou et al demonstrated that serum IL-1β and IL-17 levels in the AECOPD group were significantly higher than those in the stable COPD group or control group. These findings suggest that serum IL-1β and IL-17 may serve as critical biomarkers for distinguishing COPD patients from healthy subjects and could be instrumental in evaluating the severity of COPD and predicting clinical outcomes.43

Inflammatory mediators contribute to the pathogenesis of PH in COPD patients by affecting PASMCs, leading to an imbalance between vasoconstriction and vasodilation and promoting vascular remodeling.44 Nitric oxide (NO), produced by endothelial nitric oxide synthase (eNOS), possesses vasodilatory and anti-proliferative properties. Bao et al demonstrated that NO expression is significantly downregulated in AECOPD patients combined with PH. Moreover, decreased NO levels were identified as an independent risk factor for concurrent PH in this population and can help clinically predict the occurrence of PH.45 Similarly, prostacyclin I2 (PGI2) is a vasodilator that also inhibits vascular remodeling and is synthesized via prostacyclin synthase activity. In patients with COPD and PH, both the synthesis and release of NO in the lungs are diminished, and the expression of prostacyclin synthase mRNA is reduced.46,47 Inflammatory mediators that contribute to vasoconstriction and vascular remodeling include angiotensin II (AngII), thromboxane A2 (TxA2), prostaglandin E2 (PGE2), 5-hydroxytryptamine (5-HT), and endothelin-1 (ET-1).

Ang II is generated from angiotensin I through the catalytic action of angiotensin-converting enzyme (ACE). Ang II functions as a vasoconstrictor and also promotes the proliferation of PASMCs.44 An animal experiment demonstrated that during the progression of hypoxic PH, there is an increase in local ACE expression and Ang II production within the pulmonary artery walls. This increase promotes vascular contraction and induces distal muscularization of typically non-muscular vessels, ultimately contributing to the development of PH.48

Like PGI2, TXA2 and PGE2 are prostaglandins—lipid mediators produced through the cyclooxygenase-initiated metabolism of arachidonic acid.49 TXA2 induces contraction of PAs by binding to the thromboxane receptor. PGE2 regulates various functions of lung fibroblasts, including proliferation, migration, and collagen synthesis.50 Multiple factors stimulate the endogenous production of PGE2 in lung fibroblasts, including IL-1, TNF-α, and TGF-β. Notably, TGF-β promotes fibroblast-mediated contraction of the collagen matrix, which serves as an indicator of repair function. In contrast, IL-1 and TNF-α inhibit this contraction. This suggests that PGE2 may play a regulatory role in fibroblast-mediated tissue repair functions in the lung under different condition.51,52 PGE2 mediates its effects through four specific receptors, with the vasoconstrictive receptor EP3 being particularly notable for its response to hypoxic conditions. In both human and mouse PASMCs, the expression of the EP3 receptor is upregulated in response to hypoxia. This upregulation may contribute to pulmonary vasoconstriction under hypoxic conditions, as the EP3 receptor is associated with reduced cAMP levels, often leading to contraction and other vasoconstrictive effects.53 The specific pathways involved in this process include: 1. Rho/ROCK-dependent actin remodeling: This mechanism facilitates the trafficking of membrane type 1–matrix MMP (MT1-MMP) to the cell surface; 2. MT1-MMP activity: Once on the cell surface, MT1-MMP cleaves extracellular pro-MMP-2, enhancing the activity of extracellular MMP-2. This process degrades the extracellular matrix and vascular basement membrane, promoting angiogenesis; 3. Promotion of TGF-β signaling: Hypoxia-activated pathways increase TGF-β signaling and the expression of fibrotic proteins, leading to vascular wall remodeling; 4. Feedback loop with TGF-β1 and PGE2: TGF-β1 further stimulates the release of endogenous PGE2, potentially amplifying the response. In PASMCs lacking the EP3 receptor, MT1-MMP is less present on the membrane and more retained within the cytoplasm, resulting in decreased extracellular MMP-2 activity. Conversely, reintroducing EP3a and EP3b isoforms in these cells restores membrane-localized MT1-MMP, enabling them to respond more effectively to hypoxic conditions.54

5-HT, commonly known as serotonin, plays a role in promoting cell mitosis. Research indicates that hypoxia serves as a potent inducer of serotonin transporter (5-HTT) expression.55 Both in vitro and in vivo exposure to hypoxia has been shown to increase 5-HTT expression twofold in PASMCs via a transcriptional mechanism, specifically by upregulating 5-HTT mRNA. This increase makes PASMCs more sensitive to the growth-promoting effects of 5-HT. The induction of 5-HTT expression by hypoxia may ultimately influence the degree of pulmonary vascular remodeling and the severity of PH in patients with advanced hypoxic COPD.

Growth Factors

Growth factors, including basic fibroblast growth factor (FGF), VEGF, platelet-derived growth factor (PDGF), and epidermal growth factor (EGF), play crucial roles in vascular remodeling in PH. Specifically, FGF-1 enhances the expression of the endothelin-1 subtype A receptor (ET-AR) in PASMCs and activates ET-AR through endothelin-1 (ET-1)—a primary mediator of hypoxia-induced PH—leading to PASMCs contraction. In addition, FGF-2 and VEGF are considered the strongest activators of angiogenesis, which can stimulate the migration and proliferation of endothelial cells in existing blood vessels to generate and stabilize new blood vessels.56 A recent study has demonstrated that elevated serum levels of FGF-2 and VEGF in COPD patients are strongly correlated with an increased likelihood of developing PH. Clinically, serum FGF-2 and VEGF concentrations may serve as valuable biomarkers for assessing PH risk. Furthermore, serum FGF-2 and VEGF levels were significantly higher during acute exacerbation phase than in the remission and stable phases in COPD, indicating a direct association with disease severity in COPD patients.57 Another study investigated VEGF levels in sputum samples of COPD patients, demonstrating a significant positive correlation between exercise-induced PH severity and sputum VEGF concentrations. As a non-invasive diagnostic modality, sputum analysis offers direct insights into airway inflammation, epithelial injury, and vascular remodeling due to its direct origin from the respiratory tract. However, sputum specimen integrity is susceptible to multiple confounders, including salivary contamination, insufficient sputum volume, or tenacious consistency. Future research may optimize diagnostic accuracy by standardizing sputum collection protocols, integrating serum biomarker profiling, and incorporating echocardiographic pulmonary artery pressure assessments.58

The generation of growth factors requires transcriptional activation factors HIF-1a and STAT3.59 Research has shown that, compared to normoxia (20% O2), exposure to moderate hypoxia (5% O2) increases the proliferation response of PASMCs to mitogens such as FGF-2 and PDGF, a process likely associated with hypoxia-inducible factors (HIFs).60 HIFs refer to a group of heterodimeric transcription factors composed of α and β subunits. Targeted deletion of either the α or β subunit of HIF-1 is lethal to early embryos and is associated with vascular developmental defects, which play a crucial role in the proliferation and/or survival of PASMCs during early vascular development.61 We searched for literature on the effects of HIF-1α and growth factors on PH over the past 10 years and summarized the findings in Table 1. Hypoxia increases the intracellular level of HIF-1α. Then the α subunit binds to the β subunit into a heterodimer, which binds to numerous promoters containing hypoxia response elements (HREs), and finally promotes the expression of growth factors and the development of PH.62 Knockdown of HIF-1α with specific small interfering RNAs inhibited FGF-2-stimulated and PDGF-stimulated proliferation of PASMCs,60 reduced the muscularization of small pulmonary arterioles.63 Hypoxia serves as the primary stimulus for the upregulation of HIF-1α. Under hypoxic conditions, the expression of OTU deubiquitinase 6B (Otud6b) increases, reducing the rate of ubiquitin-mediated degradation.64 Simultaneously, the expression of small ubiquitin-like modifier 1 (SUMO-1) also increases, which, in contrast to ubiquitin, is thought to primarily prevent proteasome-mediated protein degradation.65 Consequently, the expression of HIF-1α increases. Additionally, during hypoxia, the expression of the matrix protein periostin rises, which is believed to interact with various integrins, including αVβ3, αVβ5, and α6β4. This interaction initiates processes such as cell proliferation, cell migration, and epithelial-to-mesenchymal transition. Nie et al found that the increased periostin is mainly localized at the nuclear section of cells, where it may interact with HIF-1α.66,67 In addition to hypoxia, RAS mutations, phosphatase and tensin homolog (PTEN) deficiency, increased expression of EGFR, and the interaction between MMP-2 and integrin-αVβ3 can also upregulate the level of HIF-1α by activating the PI3K/AKT/mTOR pathway.68 Activation of the PI3K/mTOR pathway increases HIF-1α protein levels without altering HIF-1α mRNA levels, possibly by increasing HIF-1α translation.61,63

Table 1 Hypoxia Facilitates Vascular Remodeling by Enhancing the Expression of Growth Factors

In COPD patients, ECs and macrophages produce various regulatory mediators, including IL-6, IL-8, and TGF-β. These pro-inflammatory markers can activate the STAT3 pathway, leading to increased STAT3 phosphorylation.81,82 STAT3 forms homodimers and binds with HIF-1α simultaneously to the VEGF promoter, where they form a molecular complex with the transcription coactivators CBP/p300 and Ref-1/APE. Moreover, the negative expression of HIF-1α or STAT3 significantly impairs promoter activity, leading to a reduction in VEGF expression. This finding suggests that the cooperative binding of both STAT3 and HIF-1α to the VEGF promoter is essential for optimal transcription of VEGF mRNA in response to hypoxic conditions. The resultant upregulation of growth factors enhances the proliferation, migration, and proteoglycan synthesis in lung fibroblasts, thereby facilitating vascular remodeling.83,84

The Forkhead Box M1 Transcription Factor

The forkhead box M1 (FOXM1) transcription factor belongs to the FOX family of transcription factors, with the FOXM1 gene located on human chromosome 12p13.3, encoding a protein of 747 amino acids.85 The FOXM1 transcription factor is recognized as a key regulator of cell cycle progression, promoting the G1/S and G2/M transitions, which in turn advances mitotic progression through its downstream targets. It is widely acknowledged for its role in promoting the proliferation of cancer cell lines.86 However, FOXM1 is also essential for normal pulmonary vascular development and plays an important role in the proliferation of PASMCs stimulated by hypoxia.87

Two additional members of the FOX family, FOXO1 and FOXO3, have been demonstrated to regulate FOXM1 transcription. In COPD patients, on the one hand, hypoxia and exposure to cigarette smoke promote oxidative stress, leading to an increase in the expression of miR-214 in PASMCs while simultaneously reducing the expression of PTEN.88 Conversely, dysfunctional ECs secrete multiple factors, such as PDGF-B, CXCL12, ET-1, and MIF, which activate the PI3K/Akt pathway. This activation leads to the phosphorylation and subsequent nuclear exclusion of FOXOs, thereby diminishing their transcriptional inhibition of FOXM1 and resulting in the upregulation of FOXM1 expression.89

In addition to HIF-2α and FOXOs, the expression of FOXM1 in PASMCs is also regulated by miR-204 and the epigenetic reader bromodomain-containing protein 4 (BRD4). Hypoxia has been shown to reduce poly ADP ribose polymerase 1–dependent miR-204 expression in PASMCs, resulting in overexpression of BRD, ultimately leading to the pro-proliferative and anti-apoptotic phenotype in these cells. The specific molecular mechanisms underlying this process include: 1. Inhibition of the cell cycle regulator p21, thereby promoting cell proliferation; 2. Activation of the nuclear factor of activated T cells; 3. Upregulation of pulmonary hypertension-related oncogenes, such as B-cell lymphoma 2 (Bcl-2) and survivin; 4. Mitochondrial membrane hyperpolarization in PASMCs, leading to increased resistance to apoptosis and enhanced proliferation.90 Furthermore, BRD4 can bind to the promoters of pro-inflammatory cytokines, such as IL-6 and TNF-α, thereby enhancing their expression in activated macrophages in chronic inflammatory diseases. This increased expression can lead to DNA damage in PASMCs, resulting in elevated expression and activation of PARP1. Consequently, this cascade further downregulates miR-204.91,92 Thus, BRD4 may not only play a role in the onset of PH, but also in the sustainability of PH by maintaining this inflammatory state.

Additionally, the promotion of PASMCs proliferation through increased FOXM1 expression may be associated with several factors. First, FOXM1 has been demonstrated to directly enhance the expression of multiple genes involved in metabolic reprogramming, particularly in glycolysis and cell cycle progression. Key targets include GLUT1, HK2, Cyclin D1, and STAT3, all of which are linked to the progression of PH.93–95 Secondly, studies indicate that FOXM1 could enhance the activity of Nuclear Factor Kappa-B (NF-κB) and β-catenin, two transcription factors that significantly influence various mechanisms of PH progression. These mechanisms encompass stress responses, cell proliferation, survival, and immune responses.96,97 Additionally, it is associated with decreased TGF-β/ Smad3-dependent signaling, resulting in down-regulated expression of contractile proteins in PASMCs. This downregulation represents a de-differentiated phenotype in these cells.98 Finally, FOXM1 enhances DNA repair capacity by stimulating the expression of DNA damage sensor protein Nijmegen breakage syndrome 1(NBS1), thereby promoting hyperproliferation of PASMCs and contributing to disease progression.99 We summarized that hypoxia promotes vascular remodeling through FOXM1 pathway as shown in Figure 2. In patients with COPD, FOXM1 contributes to the development of PH through the aforementioned pathways. As an intracellular molecular marker, FOXM1 requires detection via tissue biopsy or enriched cell analysis, positioning it as a promising tool for disease screening.

Figure 2 Hypoxia promotes vascular remodeling through the FOXM1 pathway.

Abbreviations: Akt, Protein Kinase B, PKB; Bcl-2, B-cell lymphoma-2; BRD4, Bromodomain-containing protein 4; CXCL12, Chemokine (C-X-C motif) ligand 12; ET-1, Endothelin-1; FOXM1, Forkhead box M1; GLUT1, Glucose Transporter 1; HIF-2α, Hypoxia-inducible factor 2α; HK2, Hexokinase 2; IL-6, Interleukin-6; MIF, Macrophage migration inhibitory factor; NBS1, Nijmegen breakage syndrome protein 1; NFATs, Nuclear factor of activated T cells; NF-kB, Nuclear factor-kappa B; PARP1, Poly (ADP-ribose) polymerase 1; PDGF-BB, Platelet-derived growth factor; PI3K, Phosphatidylinositol 3-kinase; PIP3, Phosphatidylinositol phosphate 3; PTEN, phosphatase and tensin homolog deleted on chromosome ten; P21, Cyclin-dependent kinase inhibitor 1A; STAT3, Signal and transducer of transcription 3; TGF-β, Transforming growth factor-β; TNF-α, Tumor necrosis factor-α.

Conclusion

Comorbid PH and CCP in patients with COPD are associated with dismal prognoses and heightened mortality, underscoring the urgent need for early detection, precise diagnosis, and timely intervention. This review synthesizes emerging evidence on promising biomarkers for COPD-PH/CCP, highlighting their roles in improving diagnostic accuracy and guiding therapeutic strategies. Hypoxia-driven crosstalk between endothelial cells and immune cells orchestrates the release of proinflammatory cytokines (such as IL-1, IL-6, IL-17), chemokines (such as CCL18, CX3CL1), and angiogenic growth factors (such as VEGF, FGF), which synergize with the neurohumoral marker NT-proBNP to form a quantifiable diagnostic network. These biomarkers exhibit significant upregulation in COPD-PH/CCP patients, with robust diagnostic performance characterized by high sensitivity and specificity. Their utility in non-invasive screening and risk stratification offers clinical advantages over traditional imaging, particularly in resource-limited settings. The transcription factor FOXM1, a key driver of vascular remodeling, represents a novel intracellular molecular marker. While current detection requires tissue biopsy or enriched cell analysis, future advancements in liquid biopsy technologies may enable peripheral blood-based FOXM1 quantification, overcoming current limitations in accessibility. Beyond conventional oxygen therapy and supportive care, biomarker-guided precision therapies are emerging as transformative approaches. Targeting interleukin pathways or chemokine-receptor interactions may disrupt the vicious cycle of inflammation and vascular remodeling. FOXM1 related inhibitors are being explored in preclinical models to block smooth muscle cell proliferation and pulmonary artery thickening. Integrating circulating biomarkers (eg, NT-proBNP, VEGF) with tissue-based FOXM1 analysis could enable dynamic risk assessment and personalized treatment adjustment. Prospective studies are needed to validate these biomarkers in large, diverse cohorts and to define their roles in predicting treatment response. Additionally, translating targeted therapies from bench to bedside requires addressing drug specificity and systemic safety. By bridging biomarker discovery with mechanistic insights, this framework holds promise for achieving early detection, early intervention, and improved outcomes in COPD-PH/CCP, ultimately reducing morbidity and mortality in this vulnerable population.

Acknowledgments

This work was supported by Grants from the Natural Science Foundation of China (No. 81970084) and the Bethune Fund for Scientific Research Project (BJ-RW2020014J).

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Disclosure

There are no conflicts of interest to declare.

References

1. Yigit E, Manav A, Ture M, Karabag T. The impact of septoplasty on cardiopulmonary functions in patients with nasal septal deviation: a prospective comprehensive analysis of echocardiographic outcome and serum N-Terminal pro BNP levels. J Craniofac Surg. 2022;33(1):35–40. doi:10.1097/scs.0000000000007801

2. World Health Organization. Tobacco and chronic obstructive pulmonary disease (COPD). WHO tobacco knowledge summaries. World Health Organization. 2023.

3. Gredic M, Blanco I, Kovacs G, et al. Pulmonary hypertension in chronic obstructive pulmonary disease. Br J Pharmacol. 2021;178(1):132–151. doi:10.1111/bph.14979

4. Zhang L, Liu Y, Zhao S, et al. The incidence and prevalence of pulmonary hypertension in the COPD population: a systematic review and meta-analysis. Int J Chron Obstruct Pulmon Dis. 2022;17:1365–1379. doi:10.2147/COPD.S359873

5. Scharf SM, Iqbal M, Keller C, Criner G, Lee S, Fessler HE; National Emphysema Treatment Trial (NETT) Group. Hemodynamic characterization of patients with severe emphysema. Am J Respir Crit Care Med. 2002;166(3):314–322. doi:10.1164/rccm.2107027

6. Barberà JA, Blanco I. Pulmonary hypertension in patients with chronic obstructive pulmonary disease: advances in pathophysiology and management. Drugs. 2009;69(9):1153–1171. doi:10.2165/00003495-200969090-00002

7. Ehteshami-Afshar S, Mooney L, Dewan P, et al. Clinical characteristics and outcomes of patients with heart failure with reduced ejection fraction and chronic obstructive pulmonary disease: insights from PARADIGM-HF. J Am Heart Assoc. 2021;10(4):e019238. doi:10.1161/jaha.120.019238

8. Vestbo J, Anderson JA, Brook RD, et al. Fluticasone furoate and vilanterol and survival in chronic obstructive pulmonary disease with heightened cardiovascular risk (SUMMIT): a double-blind randomised controlled trial. Lancet. 2016;387(10030):1817–1826. doi:10.1016/s0140-6736(16)30069-1

9. Li Y, Zhang R, Shan H, et al. FVC/DLCO identifies pulmonary hypertension and predicts 5-year all-cause mortality in patients with COPD. Eur J Med Res. 2023;28(1):174. doi:10.1186/s40001-023-01130-6

10. Oswald-Mammosser M, Weitzenblum E, Quoix E, et al. Prognostic factors in COPD patients receiving long-term oxygen therapy. Importance of pulmonary artery pressure. Chest. 1995;107(5):1193–1198. doi:10.1378/chest.107.5.1193

11. Hoeper MM, Bogaard HJ, Condliffe R, et al. Definitions and diagnosis of pulmonary hypertension. J Am Coll Cardiol. 2013;62(25 Suppl):D42–50. doi:10.1016/j.jacc.2013.10.032

12. Vanderheyden M, Bartunek J, Goethals M. Brain and other natriuretic peptides: molecular aspects. Eur J Heart Fail. 2004;6(3):261–268. doi:10.1016/j.ejheart.2004.01.004

13. Ozdemirel TS, Ulaşli SS, Yetiş B, Karaçağlar E, Bayraktar N, Ulubay G. Effects of right ventricular dysfunction on exercise capacity and quality of life and associations with serum NT-proBNP levels in COPD: an observational study. Anadolu Kardiyol Derg. 2014;14(4):370–377. doi:10.5152/akd.2014.4687

14. Hopkins WE, Chen Z, Fukagawa NK, Hall C, Knot HJ, LeWinter MM. Increased atrial and brain natriuretic peptides in adults with cyanotic congenital heart disease: enhanced understanding of the relationship between hypoxia and natriuretic peptide secretion. Circulation. 2004;109(23):2872–2877. doi:10.1161/01.Cir.0000129305.25115.80

15. Casals G, Ros J, Sionis A, Davidson MM, Morales-Ruiz M, Jiménez W. Hypoxia induces B-type natriuretic peptide release in cell lines derived from human cardiomyocytes. Am J Physiol Heart Circ Physiol. 2009;297(2):H550–5. doi:10.1152/ajpheart.00250.2009

16. Ge N, Cai X, XiaoC, Wu, M. 夜间无创正压通气对重度-极重度慢性阻塞性肺疾病相关肺动脉高压的影响 [Effects of Nocturnal Non-invasive Positive Pressure Ventilation on Severe to Extremely Severe Chronic Obstructive Pulmonary Disease-related Pulmonary Hypertension]. 中国现代药物应用 [Chinese Journal of Modern Drug Application]. 2017;11(1):49–51. Chinese. doi:10.14164/j.cnki.cn11-5581/r.2017.01.019

17. Ma KK, Ogawa T, de Bold AJ. Selective upregulation of cardiac brain natriuretic peptide at the transcriptional and translational levels by pro-inflammatory cytokines and by conditioned medium derived from mixed lymphocyte reactions via p38 MAP kinase. J Mol Cell Cardiol. 2004;36(4):505–513. doi:10.1016/j.yjmcc.2004.01.001

18. Krüger S, Graf J, Kunz D, Stickel T, Hanrath P, Janssens U. brain natriuretic peptide levels predict functional capacity in patients with chronic heart failure. J Am Coll Cardiol. 2002;40(4):718–722. doi:10.1016/s0735-1097(02)02032-6

19. Ishii J, Nomura M, Ito M, et al. Plasma concentration of brain natriuretic peptide as a biochemical marker for the evaluation of right ventricular overload and mortality in chronic respiratory disease. Clin Chim Acta. 2000;301(1–2):19–30. doi:10.1016/s0009-8981(00)00312-0

20. Su X, Lei T, Yu H, et al. NT-proBNP in different patient groups of COPD: a systematic review and meta-analysis. Int J Chron Obstruct Pulmon Dis. 2023;18:811–825. doi:10.2147/copd.S396663

21. Agoston-Coldea L, Lupu S, Hicea S, Paradis A, Mocan T. Serum levels of the soluble IL-1 receptor family member ST2 and right ventricular dysfunction. Biomarker Med. 2014;8(1):95–106. doi:10.2217/bmm.13.116

22. Dewan P, Docherty KF, Bengtsson O, et al. Effects of dapagliflozin in heart failure with reduced ejection fraction and chronic obstructive pulmonary disease: an analysis of DAPA-HF. Eur J Heart Fail. 2021;23(4):632–643. doi:10.1002/ejhf.2083

23. 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

24. Stacher E, Graham BB, Hunt JM, et al. Modern age pathology of pulmonary arterial hypertension. Am J Respir Crit Care Med. 2012;186(3):261–272. doi:10.1164/rccm.201201-0164OC

25. Florentin J, Coppin E, Vasamsetti SB, et al. Inflammatory macrophage expansion in pulmonary hypertension depends upon mobilization of blood-borne monocytes. J Immunol. 2018;200(10):3612–3625. doi:10.4049/jimmunol.1701287

26. Al-Qazazi R, Lima PDA, Prisco SZ, et al. Macrophage-NLRP3 activation promotes right ventricle failure in pulmonary arterial hypertension. Am J Respir Crit Care Med. 2022;206(5):608–624. doi:10.1164/rccm.202110-2274OC

27. Chen X, Li L, Deng Y, et al. Inhibition of glutaminase 1 reduces M1 macrophage polarization to protect against monocrotaline-induced pulmonary arterial hypertension. Immunol Lett. 2025;272:106974. doi:10.1016/j.imlet.2025.106974

28. Kimura A, Naka T, Kishimoto T. IL-6-dependent and -independent pathways in the development of interleukin 17-producing T helper cells. Proc Natl Acad Sci U S A. 2007;104(29):12099–12104. doi:10.1073/pnas.0705268104

29. Miyamoto M, Prause O, Sjöstrand M, Laan M, Lötvall J, Lindén A. Endogenous IL-17 as a mediator of neutrophil recruitment caused by endotoxin exposure in mouse airways. J Immunol. 2003;170(9):4665–4672. doi:10.4049/jimmunol.170.9.4665

30. Thatcher TH, McHugh NA, Egan RW, et al. Role of CXCR2 in cigarette smoke-induced lung inflammation. Am J Physiol Lung Cell Mol Physiol. 2005;289(2):L322–8. doi:10.1152/ajplung.00039.2005

31. van den Berg A, Kuiper M, Snoek M, et al. Interleukin-17 induces hyperresponsive interleukin-8 and interleukin-6 production to tumor necrosis factor-alpha in structural lung cells. Am J Respir Cell Mol Biol. 2005;33(1):97–104. doi:10.1165/rcmb.2005-0022OC

32. Ye P, Rodriguez FH, Kanaly S, et al. Requirement of interleukin 17 receptor signaling for lung CXC chemokine and granulocyte colony-stimulating factor expression, neutrophil recruitment, and host defense. J Exp Med. 2001;194(4):519–527. doi:10.1084/jem.194.4.519

33. Elkington PT, Friedland JS. Matrix metalloproteinases in destructive pulmonary pathology. Thorax. 2006;61(3):259–266. doi:10.1136/thx.2005.051979

34. Shan M, Cheng HF, Song LZ, et al. Lung myeloid dendritic cells coordinately induce TH1 and TH17 responses in human emphysema. Sci Transl Med. 2009;1(4):4ra10. doi:10.1126/scitranlsmed.3000154

35. Wang L, Liu J, Wang W, et al. Targeting IL-17 attenuates hypoxia-induced pulmonary hypertension through downregulation of β-catenin. Thorax. 2019;74(6):564–578. doi:10.1136/thoraxjnl-2018-211846

36. Snapper CM, Paul WE. Interferon-gamma and B cell stimulatory factor-1 reciprocally regulate Ig isotype production. Science. 1987;236(4804):944–947. doi:10.1126/science.3107127

37. Rollins BJ, Pober JS. Interleukin-4 induces the synthesis and secretion of MCP-1/JE by human endothelial cells. Am J Pathol. 1991;138(6):1315–1319.

38. Wen FQ, Liu X, Manda W, et al. TH2 Cytokine-enhanced and TGF-beta-enhanced vascular endothelial growth factor production by cultured human airway smooth muscle cells is attenuated by IFN-gamma and corticosteroids. J Allergy Clin Immunol. 2003;111(6):1307–1318. doi:10.1067/mai.2003.1455

39. Yamaji-Kegan K, Su Q, Angelini DJ, Myers AC, Cheadle C, Johns RA. Hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMalpha) increases lung inflammation and activates pulmonary microvascular endothelial cells via an IL-4-dependent mechanism. J Immunol. 2010;185(9):5539–5548. doi:10.4049/jimmunol.0904021

40. Xu QH, Huang SP, Li WL, et al. Expression of CCL-18 and CX3CL1 in serum, and their potential roles as two diagnostic and prognostic markers in chronic obstructive pulmonary disease and chronic cor pulmonale (COPD&CCP): a pilot study. Clin Lab. 2020;66(10). doi:10.7754/Clin.Lab.2020.200244

41. Eissa SA, Soliman YMA, Essawy TS, et al. Incidence of pulmonary hypertension in COPD and its relation to inflammatory marker interleukin-1. Egypt J Bronchol. 2024;18(27). doi:10.1186/s43168-024-00278-x

42. Yang D, Wang L, Jiang P, Kang R, Xie Y. Correlation between hs-CRP, IL-6, IL-10, ET-1, and chronic obstructive pulmonary disease combined with pulmonary hypertension. J Healthc Eng. 2022;2022:3247807. doi:10.1155/2022/3247807

43. Zou Y, Chen X, Liu J, et al. Serum IL-1β and IL-17 levels in patients with COPD: associations with clinical parameters. Int J Chron Obstruct Pulmon Dis. 2017;12:1247–1254. doi:10.2147/COPD.S131877

44. Shujaat A, Minkin R, Eden E. Pulmonary hypertension and chronic cor pulmonale in COPD. Int J Chron Obstruct Pulmon Dis. 2007;2(3):273–282.

45. Bao C, Yang W, Luo W. 外周血Apelin, NO, NOS水平对慢阻肺急性加重期并发肺动脉高压的诊断价值 [Diagnostic Value of Peripheral Blood Apelin, NO, and NOS Levels in Chronic Obstructive Pulmonary Disease Complicated with Pulmonary Hypertension during Acute Exacerbation]. 临床肺科杂志 [Journal of Clinical Pulmonary Medicine]. 2023;28(9):1352–1356. Chinese. doi:10.3969/j.issn.1009-6663.2023.09.011

46. Clini E, Cremona G, Campana M, et al. Production of endogenous nitric oxide in chronic obstructive pulmonary disease and patients with cor pulmonale. Correlates with echo-Doppler assessment. Am J Respir Crit Care Med. 2000;162(2 Pt 1):446–450. doi:10.1164/ajrccm.162.2.9909105

47. Lee JD, Taraseviciene-Stewart L, Keith R, Geraci MW, Voelkel NF. The expression of prostacyclin synthase is decreased in the small pulmonary arteries from patients with emphysema. Chest. 2005;128(6 Suppl):575s. doi:10.1378/chest.128.6_suppl.575S

48. Morrell NW, Atochina EN, Morris KG, Danilov SM, Stenmark KR. Angiotensin converting enzyme expression is increased in small pulmonary arteries of rats with hypoxia-induced pulmonary hypertension. J Clin Invest. 1995;96(4):1823–1833. doi:10.1172/jci118228

49. Breyer RM, Bagdassarian CK, Myers SA, Breyer MD. Prostanoid receptors: subtypes and signaling. Annu Rev Pharmacol Toxicol. 2001;41(1):661–690. doi:10.1146/annurev.pharmtox.41.1.661

50. Huang S, Wettlaufer SH, Hogaboam C, Aronoff DM, Peters-Golden M. Prostaglandin E(2) inhibits collagen expression and proliferation in patient-derived normal lung fibroblasts via E prostanoid 2 receptor and cAMP signaling. Am J Physiol Lung Cell Mol Physiol. 2007;292(2):L405–13. doi:10.1152/ajplung.00232.2006

51. Tingström A, Heldin CH, Rubin K. Regulation of fibroblast-mediated collagen gel contraction by platelet-derived growth factor, interleukin-1 alpha and transforming growth factor-beta 1. J Cell Sci. 1992;102(Pt 2):315–322. doi:10.1242/jcs.102.2.315

52. Zhu YK, Liu XD, Sköld MC, et al. Cytokine inhibition of fibroblast-induced gel contraction is mediated by PGE(2) and NO acting through separate parallel pathways. Am J Respir Cell Mol Biol. 2001;25(2):245–253. doi:10.1165/ajrcmb.25.2.4383

53. Sugimoto Y, Narumiya S. Prostaglandin E receptors. J Biol Chem. 2007;282(16):11613–11617. doi:10.1074/jbc.R600038200

54. Lu A, Zuo C, He Y, et al. EP3 receptor deficiency attenuates pulmonary hypertension through suppression of Rho/TGF-β1 signaling. J Clin Invest. 2015;125(3):1228–1242. doi:10.1172/jci77656

55. Eddahibi S, Fabre V, Boni C, et al. Induction of serotonin transporter by hypoxia in pulmonary vascular smooth muscle cells. Relationship with the mitogenic action of serotonin. Circ Res. 1999;84(3):329–336. doi:10.1161/01.res.84.3.329

56. Laddha AP, Kulkarni YA. VEGF and FGF-2: promising targets for the treatment of respiratory disorders. Respir Med. 2019;156:33–46. doi:10.1016/j.rmed.2019.08.003

57. Wang Y. 慢性阻塞性肺疾病患者血清VEGF, bFGF水平与肺动脉高压的关系研究 [Research on the Relationship between Serum VEGF and bFGF Levels and Pulmonary Hypertension in Patients with Chronic Obstructive Pulmonary Disease]. 临床肺科杂志 [Journal of Clinical Pulmonary Medicine]. 2019;24(5):849–852. Chinese. doi:10.3969/j.issn.1009-6663.2019.05.018

58. Kanazawa H, Asai K, Nomura S. Vascular endothelial growth factor as a non-invasive marker of pulmonary vascular remodeling in patients with bronchitis-type of COPD. Respir Res. 2007;8(1):22. doi:10.1186/1465-9921-8-22

59. Gray MJ, Zhang J, Ellis LM, et al. HIF-1alpha, STAT3, CBP/p300 and Ref-1/APE are components of a transcriptional complex that regulates Src-dependent hypoxia-induced expression of VEGF in pancreatic and prostate carcinomas. Oncogene. 2005;24(19):3110–3120. doi:10.1038/sj.onc.1208513

60. Schultz K, Fanburg BL, Beasley D. Hypoxia and hypoxia-inducible factor-1alpha promote growth factor-induced proliferation of human vascular smooth muscle cells. Am J Physiol Heart Circ Physiol. 2006;290(6):H2528–34. doi:10.1152/ajpheart.01077.2005

61. Iyer NV, Kotch LE, Agani F, et al. Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor 1 alpha. Genes Dev. 1998;12(2):149–162. doi:10.1101/gad.12.2.149

62. Karar J, Maity A. PI3K/AKT/mTOR Pathway in Angiogenesis. Front Mol Neurosci. 2011;4:51. doi:10.3389/fnmol.2011.00051

63. Semenza GL. HIF-1 and mechanisms of hypoxia sensing. Curr Opin Cell Biol. 2001;13(2):167–171. doi:10.1016/s0955-0674(00)00194-0

64. Yu AY, Shimoda LA, Iyer NV, et al. Impaired physiological responses to chronic hypoxia in mice partially deficient for hypoxia-inducible factor 1alpha. J Clin Invest. 1999;103(5):691–696. doi:10.1172/jci5912

65. Liu Y, Tang B, Wang H, Lu M. Otud6b induces pulmonary arterial hypertension by mediating the Calpain-1/HIF-1α signaling pathway. Cell Mol Life Sci. 2024;81(1):258. doi:10.1007/s00018-024-05291-3

66. Jiang Y, Wang J, Tian H, et al. Increased SUMO-1 expression in response to hypoxia: interaction with HIF-1α in hypoxic pulmonary hypertension. Int J Mol Med. 2015;36(1):271–281. doi:10.3892/ijmm.2015.2209

67. Li G, Jin R, Norris RA, et al. Periostin mediates vascular smooth muscle cell migration through the integrins alphavbeta3 and alphavbeta5 and focal adhesion kinase (FAK) pathway. Atherosclerosis. 2010;208(2):358–365. doi:10.1016/j.atherosclerosis.2009.07.046

68. Nie X, Shen C, Tan J, et al. Periostin: a potential therapeutic target for pulmonary hypertension? Circ Res. 2020;127(9):1138–1152. doi:10.1161/circresaha.120.316943

69. Ntokou A, Dave JM, Kauffman AC, et al. Macrophage-derived PDGF-B induces muscularization in murine and human pulmonary hypertension. JCI Insight. 2021;6(6). doi:10.1172/jci.insight.139067

70. Cheng CC, Chi PL, Shen MC, et al. Caffeic acid phenethyl ester rescues pulmonary arterial hypertension through the inhibition of AKT/ERK-dependent PDGF/HIF-1α in vitro and in vivo. Int J Mol Sci. 2019;20(6):1468. doi:10.3390/ijms20061468

71. Chen J, Song M, Qian D, et al. Atorvastatin rescues pulmonary artery hypertension by inhibiting the AKT/ERK-dependent PDGF-BB/HIF-1α axis. Panminerva Med. 2024;66(1):4–9. doi:10.23736/s0031-0808.20.03910-5

72. Wang RR, Yuan TY, Chen D, et al. Dan-Shen-Yin granules prevent hypoxia-induced pulmonary hypertension via STAT3/HIF-1α/VEGF and FAK/AKT signaling pathways. Front Pharmacol. 2022;13:844400. doi:10.3389/fphar.2022.844400

73. Wang L, Huang J, Zhang R, et al. Cullin 5 aggravates hypoxic pulmonary hypertension by activating TRAF6/NF-κB/HIF-1α/VEGF. iScience. 2023;26(11):108199. doi:10.1016/j.isci.2023.108199

74. Jin H, Jiao Y, Guo L, et al. Astragaloside IV blocks monocrotaline‑induced pulmonary arterial hypertension by improving inflammation and pulmonary artery remodeling. Int J Mol Med. 2021;47(2):595–606. doi:10.3892/ijmm.2020.4813

75. Wang G, Tao X, Peng L. miR-155-5p regulates hypoxia-induced pulmonary artery smooth muscle cell function by targeting PYGL. Bioengineered. 2022;13(5):12985–12997. doi:10.1080/21655979.2022.2079304

76. Yang C, Rong R, Li Y, Cheng M, Luo Y. Decrease in LINC00963 attenuates the progression of pulmonary arterial hypertension via microRNA-328-3p/profilin 1 axis. J Clin Lab Anal. 2022;36(5):e24383. doi:10.1002/jcla.24383

77. Wang J, Ma YR, Chang YE, et al. Preventive effect of LCZ696 on hypoxic pulmonary hypertension in rats via regulating the PI3K/AKT signaling pathway. Pulm Pharmacol Ther. 2023;82:102229. doi:10.1016/j.pupt.2023.102229

78. Favre S, Gambini E, Nigro P, et al. Sildenafil attenuates hypoxic pulmonary remodelling by inhibiting bone marrow progenitor cells. J Cell Mol Med. 2017;21(5):871–880. doi:10.1111/jcmm.13026

79. Zhou F, Dai A, Jiang Y, Tan X, Zhang X. SENP‑1 enhances hypoxia‑induced proliferation of rat pulmonary artery smooth muscle cells by regulating hypoxia‑inducible factor‑1α. Mol Med Rep. 2016;13(4):3482–3490. doi:10.3892/mmr.2016.4969

80. Fu D, Dai A, Hu R, Tian H, Chen Y, Zhu L. Regulatory mechanism of Siah1 in the pathogenesis of rat hypoxic pulmonary hypertension. Zhonghua Yi Xue Za Zhi. 2015;95(26):2093–2098.

81. Boreddy SR, Sahu RP, Srivastava SK. Benzyl isothiocyanate suppresses pancreatic tumor angiogenesis and invasion by inhibiting HIF-α/VEGF/Rho-GTPases: pivotal role of STAT-3. PLoS One. 2011;6(10):e25799. doi:10.1371/journal.pone.0025799

82. Yoshida S, Ono M, Shono T, et al. Involvement of interleukin-8, vascular endothelial growth factor, and basic fibroblast growth factor in tumor necrosis factor alpha-dependent angiogenesis. Mol Cell Biol. 1997;17(7):4015–4023. doi:10.1128/mcb.17.7.4015

83. Chetty C, Lakka SS, Bhoopathi P, Rao JS. MMP-2 alters VEGF expression via alphaVbeta3 integrin-mediated PI3K/AKT signaling in A549 lung cancer cells. Int, J, Cancer. 2010;127(5):1081–1095. doi:10.1002/ijc.25134

84. Korver W, Roose J, Heinen K, et al. The human TRIDENT/HFH-11/FKHL16 gene: structure, localization, and promoter characterization. Genomics. 1997;46(3):435–442. doi:10.1006/geno.1997.5065

85. Kim IM, Ackerson T, Ramakrishna S, et al. The forkhead box m1 transcription factor stimulates the proliferation of tumor cells during development of lung cancer. Cancer Res. 2006;66(4):2153–2161. doi:10.1158/0008-5472.Can-05-3003

86. Kalin TV, Wang IC, Meliton L, et al. Forkhead Box m1 transcription factor is required for perinatal lung function. Proc Natl Acad Sci U S A. 2008;105(49):19330–19335. doi:10.1073/pnas.0806748105

87. Lam EW, Brosens JJ, Gomes AR, Koo CY. Forkhead box proteins: tuning forks for transcriptional harmony. Nat Rev Cancer. 2013;13(7):482–495. doi:10.1038/nrc3539

88. Liu H, Yin T, Yan W, et al. Dysregulation of microRNA-214 and PTEN contributes to the pathogenesis of hypoxic pulmonary hypertension. Int J Chron Obstruct Pulmon Dis. 2017;12:1781–1791. doi:10.2147/copd.S104627

89. Savai R, Al-Tamari HM, Sedding D, et al. Pro-proliferative and inflammatory signaling converge on FoxO1 transcription factor in pulmonary hypertension. Nat Med. 2014;20(11):1289–1300. doi:10.1038/nm.3695

90. Meloche J, Potus F, Vaillancourt M, et al. Bromodomain-containing protein 4: the epigenetic origin of pulmonary arterial hypertension. Circ Res. 2015;117(6):525–535. doi:10.1161/circresaha.115.307004

91. Belkina AC, Nikolajczyk BS, Denis GV. BET protein function is required for inflammation: brd2 genetic disruption and BET inhibitor JQ1 impair mouse macrophage inflammatory responses. J Immunol. 2013;190(7):3670–3678. doi:10.4049/jimmunol.1202838

92. Meloche J, Pflieger A, Vaillancourt M, et al. Role for DNA damage signaling in pulmonary arterial hypertension. Circulation. 2014;129(7):786–797. doi:10.1161/circulationaha.113.006167

93. Gong AH, Wei P, Zhang S, et al. FoxM1 drives a feed-forward STAT3-activation signaling loop that promotes the self-renewal and tumorigenicity of glioblastoma stem-like cells. Cancer Res. 2015;75(11):2337–2348. doi:10.1158/0008-5472.Can-14-2800

94. Hu C, Liu D, Zhang Y, et al. LXRα-mediated downregulation of FOXM1 suppresses the proliferation of hepatocellular carcinoma cells. Oncogene. 2014;33(22):2888–2897. doi:10.1038/onc.2013.250

95. Wang Y, Yun Y, Wu B, et al. FOXM1 promotes reprogramming of glucose metabolism in epithelial ovarian cancer cells via activation of GLUT1 and HK2 transcription. Oncotarget. 2016;7(30):47985–47997. doi:10.18632/oncotarget.10103

96. Zhang N, Wei P, Gong A, et al. FoxM1 promotes β-catenin nuclear localization and controls wnt target-gene expression and glioma tumorigenesis. Cancer Cell. 2011;20(4):427–442. doi:10.1016/j.ccr.2011.08.016

97. Zhao B, Barrera LA, Ersing I, et al. The NF-κB genomic landscape in lymphoblastoid B cells. Cell Rep. 2014;8(5):1595–1606. doi:10.1016/j.celrep.2014.07.037

98. Dai J, Zhou Q, Tang H, et al. Smooth muscle cell-specific FoxM1 controls hypoxia-induced pulmonary hypertension. Cell Signal. 2018;51:119–129. doi:10.1016/j.cellsig.2018.08.003

99. Bourgeois A, Lambert C, Habbout K, et al. FOXM1 promotes pulmonary artery smooth muscle cell expansion in pulmonary arterial hypertension. J Mol Med. 2018;96(2):223–235. doi:10.1007/s00109-017-1619-0

Continue Reading