Introduction
Pulmonary Hypertension (PH): Overview and Pathogenesis
Pulmonary hypertension (PH) is caused by various diseases and mechanisms.1 It involves structural and/or functional changes in pulmonary blood vessels that increase pulmonary vascular resistance and pulmonary arterial pressure, leading to clinical and pathophysiological syndromes that progress to heart failure or even death.1 The key features of PH include excessive proliferation of pulmonary artery smooth muscle cells (PASMCs), increased extracellular matrix deposition, and inflammatory factor accumulation in the pulmonary vascular wall, all contributing to elevated pulmonary vascular resistance.2 The diagnostic criteria for PH included mean pulmonary arterial pressure (mPAP) ≥ 25 mmHg at rest as measured by right heart catheterization.3 Although the 6th World Symposium on PH lowered this threshold to 20 mmHg, this value is higher than that in healthy adults (14.0 ± 3.3 mmHg).4 Epidemiological studies reported that various PH subtypes affect approximately 1% of the global population, and the prevalence of PH is as high as 10% in individuals older than 65.5 The increase in smoking and global population aging have increased the prevalence of chronic obstructive pulmonary disease (COPD), which is closely related to smoking. Approximately 90% of patients with COPD have mPAP > 20 mmHg, with the prevalence of PH associated with COPD estimated at 39.2%.6 On average, there is a 2-year gap between PH symptom onset and diagnosis. Early symptoms are often subtle, and progression can be rapid, leading to an average survival of only 2.8 years for untreated patients.7 The pathogenesis of PH involves pulmonary arterial remodeling caused by excessive PASMC proliferation and pulmonary arterial endothelial cell (PAEC) injury, leading to immune responses by various cell types (B cells, T cells, neutrophils, dendritic cells).8 Current treatments include non-pharmacological approaches such as lung transplantation and medications such as prostacyclins (prostaglandin I2, iloprost, or treprostinil), endothelin receptor antagonists (bosentan, ambrisentan, or macitentan), phosphodiesterase-5 (PDE5) inhibitors (sildenafil, tadalafil, or vardenafil), and L-type calcium channel blockers.1 Despite improvements in pulmonary function and reduced hospitalizations, the 5-year mortality rate remains approximately 50%.9
Hypoxia-induced pulmonary hypertension (HPH) is a subtype of PH characterized by chronic lung disease or prolonged hypoxia, with exertional dyspnea as the primary symptom.10 Current guidelines recommend long-term oxygen therapy as the primary treatment for HPH. However, this therapy only alleviates symptoms, and it does not address oxidative stress, enzyme imbalance, or related issues.1 With the continuous exploration of reverse pharmacology and pre-clinical and clinical studies, new drugs are gradually emerging. Traditional Chinese medicine (TCM) has received extensive attention because of its advantages including minimal adverse reactions, multiple targets, and effectiveness through multiple pathways. Moreover, TCM is extracted from pure natural plants, thus meeting the requirements of safety and low costs. Currently, many Chinese herbal medicines are being studied clinically for HPH treatment. This paper reviews the use of TCM for HPH treatment over the past two decades, focusing on TCM monomers, compounds, and extracts. It discusses the inhibition of abnormal proliferation of PASMCs via antioxidative pathways and summarizes the mechanisms of action of TCM and its use in HPH treatment strategies.
PH: Pathogenic Factors and Subtypes
The pathogenesis of PH is multifactorial, resulting from the complex interaction of various factors and pathways, including external and internal influences, interactive factors, various vasoactive molecules, multiple ion channels, and multiple signaling pathways (Figure 1).1 Pulmonary vasoconstriction, pulmonary vascular remodeling (PVR), and in situ thrombosis formation are the main pathological changes in PH, and they are primarily associated with impaired pulmonary vascular endothelial function caused by an imbalance between vasodilators and vasoconstrictors (nitric oxide [NO]/endothelin-1 [ET-1]),11 abnormal PASMC proliferation, local inflammation accumulation, immune dysregulation, mitochondrial dysfunction, and metabolic reprogramming, among other factors.12 Excessive PASMC proliferation and resistance to apoptosis leading to vascular remodeling are the primary pathological features of PH.13,14 PH can be classified into five subtypes based on differences in pathophysiology and treatment strategies:15 pulmonary arterial hypertension attributable to arterial causes, PH caused by left heart disease, PH attributable to lung diseases and/or hypoxia, chronic thromboembolic PH and other pulmonary artery obstructions, and pulmonary arterial hypertension of unclear multifactorial mechanisms. Each subtype can be divided into numerous subclasses (Figure 1).
Figure 1 Pathogenic factors and subtypes of pulmonary hypertension.
|
HPH: Mechanisms and Current Treatments
HPH is a progressive syndrome triggered by chronic hypoxia or lung disease, and exertional dyspnea is a key symptom.16,17 The pathogenesis of HPH involves a shift from compensatory functional changes to decompensatory structural damage, progressing from hypoxic pulmonary vasoconstriction (HPV)18 to PVR.19
Under physiological conditions, the pulmonary circulation features high flow, low pressure, and low resistance. During hypoxia, small pulmonary arterioles constrict, thereby redirecting blood flow to well-ventilated lung segments, optimizing gas exchange.20 If hypoxia is localized, then HPV can improve the partial pressure of oxygen in arterial blood (PaO2), most effectively when the hypoxic lung volume is 30%–70%. The impact on PaO2 can be negligible if the area of vascular constriction is small. Thus, transient HPV can divert blood flow from hypoxic lung segments, serving as a physiological compensatory mechanism to adapt to adverse reactions and protect PaO2.21 However, prolonged hypoxia causes sustained pulmonary vasoconstriction, leading to increased vascular stiffness, narrowed arterial lumina, and elevated blood viscosity. Consequently, chronic HPV is detrimental, promoting HPH and increasing the workload of the right ventricle.
Continuous functional changes in the pulmonary vasculature inevitably result in structural damage. When hypoxia persists beyond the organism’s compensatory capacity, the pulmonary vessels undergo structural remodeling. This process involves several mechanisms. Specifically, pulmonary arteries, composed of the intima, media (PASMCs), and adventitia, generate excess reactive oxygen species (ROS) under prolonged hypoxia. Chemotactic factors such as CCR5 recruit inflammatory factors including interleukin (IL)-1β, IL-6, macrophage migration inhibitory factor, and high mobility group box 1 protein (HMGB1), which accumulate around the vessels.22 As pulmonary vessels contract, ROS and inflammatory factors enter the pulmonary arterial vasculature, initially causing endothelial cell injury in response to chronic inflammation. Because of impaired endothelial barrier function, there is an imbalance in released vasodilatory substances (such as NO, angiotensin II, thromboxane A2), further exacerbating vascular constriction. Simultaneously, because of changes in vascular morphology, the cellular interstitial spaces widen. Growth factors produced by the intima (such as platelet-derived growth factor [PDGF], transforming growth factor-β1 [TGF-β1], fibroblast growth factor-2, and vascular endothelial growth factor [VEGF]23) interact with PASMCs through these spaces, leading to the abnormal proliferation and migration of these cells.24 ROS can also promote the activation and expression of multiple growth factors, including phosphatidylinositol 3-kinase (PI3K)/Akt, p38 mitogen-activated protein kinase (MAPK), c-Src, TGF-β1, and VEGF. These growth factors can in turn stimulate ROS production, forming a local positive feedback loop. In addition, the mitochondrial apoptosis pathway mediated by ROS plays a crucial role in pulmonary arterial remodeling. Using rat models of HPH, multiple studies demonstrated that the Bax/Bcl-2 ratio is decreased, mitochondrial membrane potential is increased, permeability is reduced, apoptosome formation is decreased, and cleaved caspase-3 expression is decreased, thereby inhibiting PASMC apoptosis and ultimately causing pulmonary arterial remodeling.25,26 The development of HPH represents an adaptive response of pulmonary vascular cells to hypoxia, including gene expression changes and structural readjustments in response to long-term hypoxic conditions. The process of PVR involves various factors such as local immune inflammatory responses, oxidative imbalance, the dysfunction of endothelial cells and fibroblasts, and the abnormal proliferation and apoptosis inhibition of PASMCs.27,28
HPH represents an adaptive response to chronic hypoxia involving gene expression changes in the pulmonary vasculature that lead to metabolic, functional, and structural readjustments. Endothelial-to-mesenchymal transition (EndMT) also plays a significant role in PVR.29,30 In chronic lung diseases such as obstructive pulmonary disease or pulmonary fibrosis, hypoxia-sensitive genes are highly expressed, contributing to the progression of HPH.31,32 This uncontrolled pressure eventually increases the load on the right heart, potentially leading to structural changes or failure, earning HPH the nickname “cancer of cardiovascular diseases”17 (Figure 2).
![]() |
Figure 2 Pathogenesis of HPH.
|
Unlike other forms of PH, HPH lacks specialized targeted drugs. Treatment primarily relies on consensus-driven combination therapies, as recommended by domestic and international experts. The Diagnosis and Treatment Guidelines for Pulmonary Arterial Hypertension in China (2021 Edition) recommend long-term oxygen therapy for HPH caused by lung diseases and/or hypoxia.1 Nevertheless, these combinatorial regimens elevate risks of adverse effects and impose substantial economic burdens, underscoring the unmet need for superior therapeutic options.4
TCM offers promising potential for HPH treatment. TCM is known for its wide range of pharmacological activities, including anti-cancer, antioxidant, neuroprotective, anti-inflammatory, and anti–COVID-19 effects.33 Research has demonstrated that herbal TCMs can protect against HPH by reversing the abnormal proliferation of PASMCs caused by hypoxia,34,35 thereby reducing pulmonary arterial pressure.
Chinese Herbs: Compounds, Monomers, and Derivatives
TCM originated in early human history, rooted in the long-term interactions of the Chinese people with nature. The legend of Shennong, known as the “Divine Farmer” or “Emperor of Medicine” symbolizes the beginnings of TCM during the Neolithic Age. During this period, primitive agriculture emerged, and people began to understand the medicinal properties of various crops and natural substances. Over generations, ancient scholars documented and clinically applied approximately 12,000 medicinal plants, leading to TCM’s widespread recognition and global influence.36
Today, more than 80% of the population in developing countries relies on traditional herbal medicine for basic healthcare.37 TCM is typified by its unique medicinal substances and its multi-level, multi-target therapeutic effects, which have garnered international attention. However, because of the complexity of TCM components and limited analytical methods, only a few TCM products, such as sodium tanshinone IIA sulfonate (STS), beetroot juice, epicatechin, tetramethylpyrazine phosphate, and rosuvastatin combined with garlic extract, have been approved by the FDA and CFDA for therapeutic use.38 Although large-scale randomized controlled trials have not provided convincing evidence of its therapeutic effects, TCM is widely used in routine medical care and preclinical research. TCM is applied in forms such as decoctions (herbal soups made by boiling medicinal herbs in water and then straining out the residue), powders (ground into powder form and encapsulated), pills, ointments, tinctures, tablets, infusions, and injections.
Chinese Herbal Compounds (CHCs)
CHCs, as multi-component natural medicines, offer multi-target efficacy and synergistic effects across multiple pathways, differing fundamentally from Western medicine in HPH diagnosis and treatment.39 CHC prescriptions often focus on promoting qi and blood circulation, improving fluid metabolism, and resolving phlegm.40 Various CHC forms (decoctions, capsules, injections) are widely used in pre-clinical HPH models, displaying effectiveness in delaying or inhibiting disease progression.
For example, Tongxinluo is a CHC that reduces hypoxia-induced endothelial damage by downregulating intercellular adhesion molecules (ICAM-1, ICP-1), hypoxia-inducible factor 1α (HIF-1α), VEGF, and ET-1 while regulating eNOS and iNOS expression, thus increasing NO activity in HPH rats.41,42 Another study demonstrated that San-huang-xie-xin-tang, composed of Huanglian (Coptis chinensis Franch.), Huangqin (Scutellaria baicalensis Georgi), and Dahuang (Rheum palmatum L.), significantly reduces U46619-induced arterial hypertension by downregulating PDE5, Rho kinase (ROCK) II, and cyclooxygenase-2 (COX-2) and upregulating soluble guanylate cyclase 1 (sGC1).43 Lu et al reported that Qiliqiangxin capsules composed of Huangqi (Astragalus membranaceus (Fisch.) Bunge), Renshen (Panax ginseng C.A.Mey.), Wutou (Aconitum carmichaelii Debeaux), Danshen (Salvia miltiorrhiza Bunge), Rougui (Cinnamomum cassia (L.) J.Presl), Honghua (Carthamus tinctorius L.), Gangliao (Polygonum aviculare L.), and Chenpi (Citrus reticulata Blanco) directly restore mitochondrial function and structure, modulate apoptosis pathways, and reverse right ventricular remodeling attributable to HPH.44 Wu et al conducted network pharmacology analysis, revealing that the Qishen Yiqi formula targets therapeutic pathways in pulmonary arterial hypertension and significantly modulates PI3K-Akt, MAPK, and Hif-1 signaling pathways.45
Highly cited studies from China also highlighted CHCs such as Xuefu Zhuyu decoction and Qibai Pingfei lung capsules, which improve PVR and reduce inflammation by modulating pathways such as PI3K/Akt and JAK1/STAT3.46,47 Details of the composition and mechanisms of CHCs are presented in Table 1.
![]() |
Table 1 The Mechanism of Action of Chinese Herbal Compounds and the Diseases They Treat
|
Chinese Herbal Monomers (HMs) and Their Derivatives: Mechanisms
The therapeutic mechanism of HMs involves reducing inflammatory and vascular growth factor levels, regulating Ca2+ and K+ channels, restoring mitochondrial function, and controlling signaling pathways to achieve pulmonary artery vasodilation, protect endothelial function, and reverse PVR.55
Anti-Inflammatory
Inflammation plays a crucial role HPH development, with lesions in patients with HPH containing chemotactic factors and inflammatory mediators such as TNF, IL-6, IL-1β, and TGF-β1.12 In HPH rat models, inflammatory cells, including T cells, B cells, macrophages, dendritic cells, and mast cells, infiltrate around pulmonary vessels and contribute to PVR.56 These immune cells are correlated with disease severity and survival, making them potential therapeutic targets for HPH.57
Monocyte chemoattractant protein-1 (MCP-1), also known as CC-motif chemokine ligand 2 (CCL2), belongs to the CC chemokine family. MCP-1 promotes the progression of inflammation by attracting inflammatory factors to migrate and infiltrate the local microenvironment. Activation of the NF-κB pathway increases MCP-1 expression and stimulates the proliferation of PASMCs.58 Resveratrol can downregulate MCP-1 by inhibiting the NF-κB signaling pathway, thereby reducing damage to PAECs and suppressing abnormal PASMC proliferation.59,60 The NF-κB signaling pathway is a classical anti-inflammatory pathway widely targeted in PH treatment. Puerarin, betaine, and baicalin can also alleviate the accumulation of inflammatory factors around pulmonary arteries by inhibiting the NF-κB pathway, thereby improving pulmonary artery obstruction caused by pulmonary vascular resistance and mitigating PH.61–64 Elevated expression of inflammatory factors such as TNF-α, IL-6, and IL-1β is commonly observed around the pulmonary arteries in PH. These inflammatory factors accelerate the remodeling process and disrupt local immune balance.65 Allicin (Allium sativum L.) and grape seed proanthocyanidins can reduce the expression of related inflammatory factors in the lungs of mice, alleviating perivascular inflammation and inhibiting HPV and PVR.66,67
Oxidative Stress
Oxidative stress occurs when the balance between oxidation and antioxidants is disrupted in a low oxygen environment, leading to an oxidative state. This imbalance, driven by free radicals, plays a key role in disease development. When ROS, central to oxidative stress, accumulate beyond the capacity of antioxidant systems, they induce neutrophil infiltration, protease secretion, and the production of oxidative intermediates.68 In HPH, ROS accumulation is linked to pulmonary microvascular endothelial dysfunction, growth factor feedback, and mitochondrial apoptosis pathways, all contributing to HPH progression and right heart failure.69–71 NADPH oxidase, a major ROS source, can induce PVR directly or through ROS generation.72,73 Given the role of oxidative stress in HPH, natural herbal antioxidant therapies have emerged as promising prevention and treatment options.
Nuclear factor E2-related factor 2 (Nrf2), the main antioxidant response regulator, reduces oxidative stress by controlling antioxidant enzymes such as heme oxygenase-1 (HO-1).74 Salvianolic acid A reduces intracellular ROS production, inhibits TGF-β1 and EndMT, and counters PVR by activating the Nrf2/HO-1 pathway.75 The synergistic action of NADPH oxidase (NOX) and VPO1 exacerbates oxidative damage in HPH.76 MLB and trimethoxy stilbene, a new resveratrol analog, prevent PVR and right heart failure in HPH rats by inhibiting oxidative stress and inflammation via the NOX/VPO1 pathway and extracellular signal-regulated kinase (ERK) signaling.77,78 Resveratrol further regulates the MAPK/ERK1 and PI3K/Akt pathways, enhances the Nrf2/thioredoxin-1 (Trx-1) axis, inhibits HIF-1α expression, and reduces hypoxia-induced ROS production. Ligustrazine exerts anti-inflammatory, antioxidant, and anti-PASMC proliferation effects in HPH by inhibiting the ROS/HIF/VEGF pathway and modulating the ROS/iNOS/PKG axis.79,80
Ion Channels
Ion channel dysfunction, especially Ca2+ and K+ channel dysfunction, plays a critical role in HPH progression.81 Elevation of the intracellular calcium concentration ([Ca2+]i) is a major factor in PVR development.82 Prolonged hypoxia and inflammation impair K+ channel activity in PASMCs, leading to membrane depolarization and voltage-gated Ca2+ channel activation, thereby increasing [Ca2+]i and causing vasoconstriction.83 Thus, strategies to reduce [Ca2+]i and restore K+ channel function are key to treating HPH, with natural herbal medicines and its derivatives often targeting these ion channels.
Danshensu IIA induces pulmonary vasodilation by inhibiting calcium influx, suppressing intracellular calcium release, and activating K+ channels.84 TRPC channels regulate [Ca2+]i by facilitating calcium influx or modulating membrane potential.81 Under hypoxic conditions, TRPC1 and TRPC6 expression is increased in PASMCs, thereby enhancing store-operated calcium entry (SOCE). STS reduces SOCE and basal calcium levels by inhibiting elevated TRPC1 and TRPC6 expression in rats with HPH, alleviating pulmonary vascular resistance, and improving pulmonary vascular and right ventricular remodeling.85 Calcium-sensing receptors (CaSRs) are crucial for regulating [Ca2+]i, vasoconstriction, and PASMC proliferation. Ligustilide inhibits CaSR activation in PASMCs and reduces [Ca2+]i.86 Hypoxia reduces Kv1.5 and Kv2.1 expression in small pulmonary arteries, lowering IKV currents in PASMCs.87 STS significantly inhibits the hypoxia-induced proliferation of PASMCs by affecting the expression of Kv2.1.88 Eulophia maculata (Eulophia maculata (Lindl.) Rchb.f.) extract reduces extracellular calcium levels, inhibits adrenaline-induced intracellular calcium release, and suppresses PDE5, thereby enhancing pulmonary vasodilation.89 Pretreatment with Rhodiola tangutica (Maxim.) S.H.Fu upregulates K+ channel protein expression, inhibits Ca2+ channel protein expression, and suppresses hypoxia-induced PASMC proliferation.90
Apoptosis Regulation
Dysregulated apoptosis represents a core pathological mechanism in pulmonary vascular remodeling (PVR) associated with hypoxic pulmonary hypertension (HPH). Imbalanced apoptosis and proliferation across major pulmonary vascular cell types, particularly pulmonary arterial endothelial cells (PAECs) and smooth muscle cells (PASMCs), are central to disease progression, exhibiting paradoxical effects that contribute to its complexity.91
Apoptosis of PAECs serves as a trigger of HPH. This process is a key early event in HPH pathogenesis. Chronic hypoxia, inflammatory cytokines (eg, TNF-α, IL-1β), and oxidative stress directly induce PAEC apoptosis.22 This compromises endothelial barrier function and vasoregulation, reducing vasodilatory factors (eg, NO, prostacyclin) while increasing vasoconstrictors (eg, ET-1, TXA2), exacerbating vasoconstriction.23 Critically, apoptotic debris and inflammatory mediators from dying PAECs activate local inflammation, recruiting immune cells and promoting the release of growth factors (eg, PDGF, TGF-β) and cytokines. These signals drive the phenotypic shift of adjacent PASMCs from a contractile to a proliferative/secretory state, characterized by excessive proliferation, migration, and apoptosis resistance, ultimately leading to fixed PVR and elevated pulmonary artery pressure.24 Thus, PAEC apoptosis initiates and sustains a pathological vascular cycle, making its inhibition a key therapeutic target. Active compounds from traditional herbs, such as Astragaloside IV (AST IV),92 Salvianolic acid A (SalA),38 and Resveratrol,93 demonstrate protective effects against hypoxia/inflammation-induced PAEC apoptosis in HPH. They enhance endothelial survival and NO bioavailability via pathways like PI3K/Akt and Nrf2/HO-1 activation.
PASMC apoptosis resistance constitutes the structural basis for PVR.94 Characterized by downregulated pro-apoptotic proteins (eg, Bax) and upregulated anti-apoptotic proteins (eg, Bcl-2) under hypoxia, this phenomenon reduces the Bax/Bcl-2 ratio, stabilizes mitochondrial membrane potential, and suppresses mPTP opening, apoptosome formation, and Caspase-3 activation.95 Concurrent activation of pro-survival pathways (eg, PI3K/Akt, ERK) further enhances PASMC viability, enabling sustained proliferation and accumulation that directly drives medial hypertrophy, vascular obliteration, and increased resistance. Herbal agents including AST IV,96 Puerarin,97 Baicalin,98 Apigenin99 and Salidroside100 counter this resistance by modulating apoptotic protein expression (eg, reducing Bcl-2/Bax ratio, activating Caspase-3) through pathways such as the mitochondrial pathway (Salidroside via A2aR), AMPKα1/p53/Bax/Bcl-2/caspase cascade, HIF-1α/Kv1.5 signaling, or PI3K/Akt inhibition, thereby inducing PASMC apoptosis and attenuating medial thickening.
Therapeutic Implication of Dual Regulation
The paradoxical effects of apoptosis in PAECs (detrimental) versus PASMCs (protective resistance) necessitate treatment strategies capable of bidirectional regulation, ie protecting PAECs from apoptosis while inducing apoptosis in hyperproliferative PASMCs. Thus, effective HPH management requires: Preserving endothelial integrity and function by reducing PAEC apoptosis; Selectively inducing apoptosis in apoptosis-resistant, proliferating PASMCs. Many herbal compounds exhibit this dual potential through pleiotropic actions – antioxidant, anti-inflammatory, and pathway modulation – simultaneously protecting endothelium and promoting PASMC apoptosis. Future research should delineate the specific molecular mechanisms of different compounds in cell-specific apoptosis regulation, assess their relative contributions to restoring pulmonary vascular homeostasis, and explore potential synergies. Harnessing this cell-specific apoptosis modulation is central to developing more effective and safer herbal therapies for HPH (Figure 3).
![]() |
Figure 3 Mechanisms underlying HPH treatment by TCMs.
|
Chinese HMs and Their Derivatives: Classification
In preclinical HPH treatment, different CHCs often exhibit similar therapeutic effects, mainly because of similarities in key HMs or their classification. For instance, although the compositions of Qili Qiangxin capsules44 and Qibai Pingfei capsules47,48 differ, both CHCs share Astragalus as a key HM,101 a flavonoid known for its immunomodulatory, cardiotonic, hypotensive, and anti-viral properties. Consequently, both CHCs reduce inflammation and modulate the NO pathway, providing anti-inflammatory and endothelial-protecting effects. Key HMs in CHC such as plantain, rhodiola, and peony (Paeonia lactiflora Pall.) contain potent antioxidants such as catalpol,102 salidroside,103 and paeoniflorin,104 which belong to the glycoside class and share the mechanisms of clearing ROS and activating antioxidant pathways. The mechanisms of various Chinese herbal monomers are shown in Table 2.
![]() |
Table 2 The Pharmacological Mechanisms of Various Components in Chinese Herbal Medicines and the Diseases They Treat
|
Flavonoids in Chinese HMs
Flavonoids, a group of organic compounds found abundantly in plants, are characterized by a flavan nucleus, and they exhibit anti-inflammatory, antioxidant, anti-angiogenic, anti-microbial, and anti-viral effects.150 They also reduce vascular fragility, improve vascular permeability, and regulate lipid and cholesterol levels in vascular diseases.151
An experimental study demonstrated that breviscapine can stimulate NO production by increasing the expression of protein kinase G1 (PKG-1) and activate the NO signaling pathway to improve pulmonary vascular dilation. Additionally, breviscapine significantly improved right ventricular hypertrophy in HPH rats by inhibiting calcium influx and activating calmodulin.105 The natural flavonoid compound puerarin inhibits abnormal PASMC proliferation by arresting the cell cycle at the G1 phase and improve PVR. Additionally, the reduced expression of the cell cycle-related proteins cyclin A, cyclin E, and cyclin D1 is also attributed to puerarin-induced autophagy.106 Another in vitro study revealed that puerarin improves hypoxia-induced pulmonary vascular contraction and oxidative stress by activating the BMPR2/Smad and PPARγ/PI3K/Akt signaling pathways, which reduces ROS and ET-1 levels in hypoxia-induced HPAECs.107 In antioxidant therapy research for HPH, genistein restored the imbalance of ROS and H2O2 in hypoxia-induced PASMCs from the source and improved oxidative balance in the body through the EPO/EPOR signaling pathway.108 Baicalein (baicalin) intervention in rats with HPH can reduce the levels of inflammatory factors such as TNF-α, IL-1β, and IL-6, improving the local inflammatory storm caused by hypoxia. Subsequent studies suggested that baicalein can exert its anti-inflammatory properties by modulating the NF-κB and BMP/Smad pathways.62,109 Furthermore, the ability of baicalein to improve PVR might involve the synergistic action of multiple pathways, including the TNF-α/BMPR2, MAPK/matrix metalloproteinase-9 (MMP-9) signaling pathway, Akt/eNOS, ERK, and NF-κB signaling pathways.110 Quercetin, another common flavonoid compound, was found in in vitro experiments to inhibit abnormal PASMC proliferation and induce PASMC apoptosis by blocking the tyrosine receptor kinase A/AKT signaling pathway.111 Additionally, its homologous compound isoquercetin also exerted protective effects on HPH rats, with potential mechanisms related to the PDGF receptor β signaling pathway.112 In summary, flavonoid compounds improve HPH by modulating vascular constriction factors, exerting anti-inflammatory and antioxidative effects, and inhibiting abnormal PASMC proliferation.
Alkaloids in Chinese HMs
Alkaloids, nitrogen-containing organic compounds found in plants in the Polygonaceae, Fabaceae, Apocynaceae, and Solanaceae families, are key active ingredients in many TCMs. With more than 3000 types identified, these compounds exhibit diverse biological activities, including anti-cancer, anti-angiogenesis, anti-inflammatory, and anti-proliferative effects.152,153 Recently, their therapeutic potential in HPH has gained attention. Mechanistically, alkaloids can prevent and treat HPH through anti-inflammatory, antioxidant, and autophagic processes, promoting PASMC apoptosis, disrupting the cell cycle, and inhibiting cell proliferation and migration.118,119
For example, ligustrazine delays PVR in HPH by modulating PASMC calcium homeostasis and regulating vasoactive factors (ET-1, NO)114 while also inhibiting PASMC proliferation by blocking the PI3K/Akt signaling pathway, thereby preventing cell cycle progression from G0/G1 to S phase.115 Tetrandrine, another alkaloid, significantly reduces pulmonary arterial pressure in rats with HPH by stimulating endothelial NO production by NOS via PKG-1 and blocking myocardial Ca2+ channels, thereby delaying right ventricular hypertrophy.116,117 Berberine, a natural alkaloid organic compound extracted from the herb Coptis chinensis, was found in recent studies to primarily treat HPH by modulating the BMPR2 and TGF-β pathways and inhibiting the Trx1/β-catenin pathway. Another study demonstrated that berberine inhibits PASMC proliferation induced by adrenaline primarily via the PP2A pathway, and these findings were validated in both patients with HPH and animal models.118,119 Betaine, a biogenic amine from goji berries (Lycium barbarum L.), was originally used as a dietary supplement, and it was revealed in preclinical studies of HPH to possess notable anti-inflammatory effects. It regulates the levels of inflammatory factors such as NF-κB, TNF-α, and IL-1β, thereby improving mPAP.120 Aloperine, extracted from the Chinese herbal medicine Sophora alopecuroides L., exerts significant protective effects against vascular diseases. Both in vitro and in vivo HPH studies indicated that aloperine suppresses PASMC proliferation and reduces mean mPAP through modulating the RhoA/ROCK and NF-κB pathways.121,122
Glycosides in Chinese HMs
The study of glycosides began in the early 19th century with the discovery of amygdalin in 1830. Glycosides are organic compounds characterized by a glycosidic bond between a monosaccharide or oligosaccharide and another molecule such as alcohol, sugar, purine, or pyrimidine. This unique structure gives them notable stability, water solubility, and bioavailability.154 Glycosides play a crucial role in drug development, with many TCMs, such as Plantago,102 Rhodiola,103 and Peony,104 containing effective glycosides. Research indicates that glycosides exert therapeutic effects through pathways involving inflammation, oxidative stress, cell proliferation, apoptosis, cell cycle progression, and ion channels. Their anti-inflammatory and antioxidant effects are particularly valuable in HPH treatment.
AST IV, a glycoside isolated from Astragalus membranaceus, has displayed anti-inflammatory effects in rats with HPH, reducing vasoconstrictive and inflammatory factors (ET-1, Ang II, TNF-α, and IL-6) and mitigating PVR by inhibiting the NLRP-3/calpain-1 pathway.97,123 In vitro, AST IV suppressed T follicular helper cell responses, expanded T follicular regulatory cell responses, and regulated the Notch signaling pathway, thereby reversing remodeling progression.92,124 Furthermore, AST IV has been demonstrated to exert cardioprotective effects by regulating calcium ion homeostasis, suppressing cardiomyocyte apoptosis in rats with HPH, and delaying right ventricular remodeling.155 The efficacy of the traditional Chinese herb Rhodiola rosea in treating HPH has been confirmed.125 However, its specific mechanisms of action are unclear, prompting in-depth research into its key active component salidroside. Salidroside promoted PASMC apoptosis via an A2aR-related mitochondrial-dependent pathway, thereby alleviating HPH.100 Additionally, salidroside inhibited chronic hypoxia-induced PASMC proliferation via the AMPKα1/p53/p27/p21 pathway and reversed resistance to apoptosis through the AMPKα1/p53/Bax/Bcl-2/caspase 9/caspase 3 pathway.126 Therefore, apoptosis modulation is considered a key mechanism of salidroside in treating HPH. Asiaticoside, a saponin monomer isolated from the traditional Chinese herb Centella asiatica (L.) Urb., primarily treats HPH by blocking TGF-β1/SMAD family member 2/3 signaling to induce PASMC apoptosis,127 and it can also prevent endothelial cell apoptosis by regulating the NO pathway.128 Recent studies revealed that notoginsenoside R1 extracted from Panax notoginseng (Burkill) F.H.Chen inhibits hypoxic pulmonary vasoconstriction induced by low oxygen and high carbon dioxide levels by attenuating ERK pathway activation.129 Notoginsenoside R1 also prevents HPH by modulating the p38 MAPK pathway.130 Other glycosides such as icariin, epimedium glycoside, and paeoniflorin have also displayed therapeutic effects against HPH. For instance, icariin effectively inhibits EndMT by blocking HIF-2α/Arg1 signaling, thereby improving pulmonary endothelial dysfunction and PVR.131 Epimedium glycoside improves HPH by inhibiting the TGF-β1/Smad2/3 pathway,132 and paeoniflorin ameliorates HPH by improving BMPR2 downregulation-mediated EndMT.30
In summary, glycosides compounds influence HPH by modulating inflammation, oxidative stress, cell apoptosis, and vascular constriction, with their apoptosis-modulating and antioxidant effects being particularly significant. Aucubin, a β-
Polyphenols in Chinese HMs
Phenolic acids, secondary metabolites widely found in plants, possesses a range of biological activities, including anti-inflammatory, anti-microbial, anti-cancer, anti-allergic, anti-viral, and anti-thrombotic effects.159–161 As natural antioxidants, they hold promise in TCM and as resources for developing new drugs and products. However, further research is needed to determine their safety, efficacy, and mechanisms of action for specific applications. Recently, phenolic acids have attracted interest as potential therapeutic agents for preventing and treating HPH.
Resveratrol, a non-flavonoid polyphenolic compound, is synthesized by plants in response to stress, injury, infection, or UV radiation. It is found in various plants, especially grapes, Polygonum cuspidatum, and peanuts. Resveratrol exhibits anti-inflammatory, antioxidant, anti-proliferative, and anti-fibrotic properties.162 Its therapeutic effects on the cardiovascular system have led to its widespread use in preclinical HPH studies. Sphingosine-1-phosphate (SphK1/S1P) signaling induces PVR by activating NF-κB and upregulating cyclin D1. Resveratrol reverses this process by inhibiting the SphK1/S1P/NF-κB/Cyclin D1 signaling pathway.133 Resveratrol activates the PI3K/AKT signaling pathway to inhibit the expression of arginase II induced by hypoxia, thereby preventing PASMC proliferation.93,134 In terms of antioxidation, resveratrol regulates the MAPK/ERK1 and PI3K/Akt pathways, enhances the NRF2/Trx-1 axis, suppresses HIF-1α expression, and reduces hypoxia-induced ROS production, concurrently decreasing the levels of TNF-α, IL-1β, and IL-6 in rat lungs.135 Furthermore, resveratrol can achieve similar therapeutic effects by inhibiting the RhoA/ROCK signaling pathway, activating SIRT1, and suppressing Th17 cell differentiation.136 Furthermore, resveratrol can prevent right ventricular hypertrophy and cardiac fibrosis in patients with HPH while also reducing acetylation levels in the right ventricle.163 Phenolic acids extracted from HMs such as (salvianolic acid A) and magnesium lithospermate B (MLB) have displayed significant efficacy in treating HPH. Danshensu inhibits PASMC proliferation, with its protective effects involving regulation of the TGF-β/Smad3 pathway.137 Salvianolic acid A delays PVR by activating the BMPR2/Smad pathway while inhibiting apoptosis and EndMT.38 MLB exerts its therapeutic effects on HPH by inhibiting the NOX/ROS/ERK and NOX/VPO1 pathways.138,139 MLB also inhibits hypoxia-induced EndMT and downregulates HIF-1α, MCP-1, NF-κB, PCNA, CDK4, and other factors, thereby halting disease progression. These findings suggest that polyphenolic natural antioxidants could effectively improve HPH.
Terpenes in Chinese HMs
Terpenoids, a class of naturally occurring organic compounds, are known for their distinctive aromas and are found in many herbal medicines with expectorant, anti-tussive, anti-spasmodic, diaphoretic, insecticidal, and analgesic effects. Terpenoids can be isolated from natural sources through distillation, direct steam distillation, freezing, and extraction. Terpenoids exhibit a range of biological activities including antioxidant, anti-inflammatory, and anti-microbial properties.164
Paclitaxel, is a terpenoid known for its anti-proliferative effects against vascular diseases, and it is FDA-approved for preventing restenosis caused by drug-eluting stents.165 In HPH studies, paclitaxel exerted beneficial effects in model mice, which were attributed to the upregulation of p27Kip1 and shortening of the cell cycle.140 Another study indicates that paclitaxel exerts protective effects against HPH by inhibiting FoxO1-mediated autophagy.141 Dihydroartemisinin, derived from Artemisia annua L., is a potent antimalarial drug, and it also improves endothelial cell proliferation and migration while reducing oxidative stress by lowering ROS levels in hypoxia-induced pulmonary arterial hypertension.142 Basigin (Bsg) is a transmembrane glycoprotein that promotes myofibroblast differentiation, cell proliferation, and MMP activation. Triptolide improves right ventricular dysfunction in HPH model animals by inhibiting Bsg and its ligand CyPA, suppressing PASMC proliferation, and reducing ROS and inflammatory cytokine levels.143 HMGB1 is a pro-inflammatory cytokine governing tissue remodeling and angiogenesis in pulmonary arterial hypertension. Glycyrrhizin, a terpenoid compound, improves HPH-induced right ventricular systolic pressure elevation and right ventricular hypertrophy, reduces pulmonary inflammation, and delays PVR by inhibiting HMGB1.144
Quinones in Chinese HMs
Quinone compounds are bioactive molecules found in plants, including naphthoquinones, benzoquinones, and anthraquinones. Their unsaturated cyclohexadienedione structure makes them essential in the electron transfer chain of biological oxidation. This redox property underpins their roles in regulating apoptosis, autophagy, proliferation, and angiogenesis.166 STS is a notable quinone that alleviates HPH by reducing [Ca2+]i and restoring K+ channel activity.84,85 STS also inhibits hypoxia-induced abnormal PASMC proliferation by modulating pathways such as mTOR/eIF 2α,145 PI3K/AKT/mTOR,146 and PKG/PPARγ,147 thus regulating pulmonary inflammation and edema. In another in vitro experiment, thymoquinone induced apoptosis in PDGF-BB–induced PASMC proliferation by enhancing the mitochondrial-dependent apoptotic pathway associated with p38 MAPK and regulating the expression of apoptosis genes (Bax, Bcl-2). Hydroxysafflor yellow A (HSYA), a key component of traditional Honghua medicine, has anti-inflammatory, antioxidant, and anti-thrombotic effects. A network pharmacology study revealed that HSYA can inhibit PASMC proliferation and PVR and reduce pulmonary arterial wall thickness and muscularization by modulating genes such as ANXA5, SRC, PGR, EGFR, PPARG, and ESR1, thereby improving HPH.
Chinese Herbs Improve HPH Through Antioxidant Signaling Pathways
Chronic hypoxia-induced oxidative stress is a key factor in HPH development. Although the exact mechanisms of HPH are not fully understood, international guidelines recognize chronic hypoxia as a primary trigger. Compared with other modeling methods, cellular or mouse models generated via long-term hypoxia are more consistent with oxidation- and inflammation-related indicators in clinical patients with HPH.167
ROS, as markers of oxidative stress, influence cellular growth, proliferation, transcription factor activity, and apoptosis. Additionally, ROS act as second messengers, activating various signal transduction pathways that can lead to cellular and tissue damage, organ dysfunction, and even carcinogenesis.168 Therefore, targeting oxidative stress at its source is a vital strategy for HPH therapy.
Previous sections discussed various HPH treatment strategies, including strategies targeting anti-inflammatory and antioxidant pathways, ion channel modulation, and signaling pathways, as well as the inhibition of EndMT. Herbal medicines play a significant role in regulating oxidative stress through signaling pathways and restoring the ROS balance, potentially reversing the disease at its root. Research has established that Ros have strong links with excessive PASMC proliferation, PVR progression, and right heart failure.169 Thus, exploring the influence of herbal medicines on signaling pathways, PVR, and antioxidant activity could offer new therapeutic approaches for HPH. The following sections of this article summarize the mechanisms by which herbal medicines regulate signaling pathways, exert antioxidant effects, inhibit abnormal PASMC proliferation, and reverse PVR.
Oxidation
Oxygen is essential for cellular signaling, gene expression, growth, development, and apoptosis, primarily through its role in glucose metabolism, which provides energy for bodily functions.170 Hypoxia-induced oxidative stress occurs when harmful stimuli lead to the excessive production of ROS by oxidases in mitochondria and the endoplasmic reticulum. This stress surpasses the antioxidant capacity of the body, resulting in neutrophil infiltration, increased protease secretion, and damage to DNA, proteins, lipids, and other macromolecules, ultimately causing cell necrosis and apoptosis.171 To counteract ROS damage, organisms have evolved a self-antioxidant defense system comprising enzymatic antioxidants such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px), as well as non-enzymatic antioxidants including vitamin C, vitamin E, coenzyme Q, lipoic acid, and glutathione.172 SOD, CAT, and GSH-Px are indicators of ROS levels in HPH cell and animal models, as they reflect the degree of oxidative stress. Normally, ROS production and elimination are balanced. However, in HPH, prolonged hypoxia in pulmonary arteries causes ROS levels to exceed the body’s clearance capacity. To compensate, the body constricts pulmonary blood vessels to enhance oxygenation. When this constriction surpasses the compensatory limit, irreversible PVR occurs.173 Recent studies found that various natural herbs can effectively reduce ROS levels in HPH models through multiple signaling pathways. This reduction delays or reverses the abnormal proliferation of PASMCs and decreases PVR, leading to lower mPAP and protecting right heart function (Figure 4).
![]() |
Figure 4 Antioxidant effects of Chinese herbal medicines. ① There are many types of ROS. Those primarily involved in oxidative stress include singlet oxygen (1O2), hydroxyl radical (•HO), superoxide anion (O2−), hydrogen peroxide (H2O2), nitric oxide (NO), and peroxynitrite (ONOO−), whereas those mainly participating in cellular signaling pathways are O2 and H2O2. ② Intracellular ROS are generated through various pathways:174 primarily by O2− generated by the mitochondrial respiratory chain; ROS generated through the uncoupling process of nitric oxide synthase (NOS); oxidative enzymes such as NOX generate ROS by donating electrons to oxygen molecules, and NOX4 is primarily located in mitochondria, serving as a major source of ROS; xanthine oxidase oxidation produces uric acid accompanied by the generation of ROS; and reactions catalyzed by myeloperoxidase also result in ROS production. ③ Monomers of TCM antioxidants: MLB, asiaticoside, dioscin, ginsenoside Rg1, paclitaxel, AST IV, magnolol, andrographolide, wogonin, cannabidiol, genistein, salidroside, caffeic acid phenethyl, chrysin, isoliquiritigenin, resveratrol, pucrarin, paeoniflorin, rutin, baicalin, STS, artemisinin, dihydroartemisinin, tanshinone IIA, polydatin, halofuginone, quercetin, allicin, sulforaphane, perillyl alcohol, umbelliferone, ligustrazine.
|
Antioxidant Signaling Pathways
Signaling pathways are key regulators of cellular homeostasis, physiological processes, development, the determination of cellular fate, and disease progression, and they are potential therapeutic targets. Understanding these pathways is fundamental to studying biological functions and disease mechanisms and developing new treatments.175 Antioxidant signaling pathways, representing significant branches of these pathways, are crucial for regulating cellular responses to oxidative stress, protecting cells from oxidative damage. These effects are conferred through regulating the activity and expression of antioxidant enzymes, scavenging free radicals, maintaining redox balance, and activating antioxidant response genes such as Nrf2. This activation promotes the expression of antioxidant stress genes, including antioxidant enzymes and heat shock proteins, which enhance cells’ antioxidant capacity.176 Additionally, oxidative stress can trigger cellular apoptosis and inflammatory responses. Antioxidant signaling helps to regulate these processes by modulating the expression and activity of relevant signaling molecules, thereby maintaining cellular stability.177
The PI3K/Akt signaling pathway, a classic antioxidant pathway, plays a crucial role in HPH treatment by mediating antioxidation, anti-inflammation, inhibition of abnormal PASMC proliferation, and vasodilation while also protecting endothelial cells. PI3K activation leads to the phosphorylation and activation of Akt, which in turn regulates downstream targets involved in antioxidant defense. Phosphorylated Akt activates transcription factors such as NF-κB and Nrf2, which bind to antioxidant response elements (AREs) in the promoter regions of antioxidant enzyme genes. This process increases the transcription of antioxidant enzymes such as SOD, CAT, and GSH-Px, helping to eliminate ROS that accumulate around pulmonary artery vessels because of chronic hypoxia.178
Additionally, the Akt pathway can affect the mitochondrial apoptosis pathway by regulating the activity of apoptosis-related protein kinases such as Bax, Bcl-2, and Bad through phosphorylation, thereby controlling cellular apoptosis. These mechanisms collectively help cells manage oxidative stress, maintain redox balance, and protect against oxidative damage. In patients with COPD, macrolide antibiotics reduce chronic inflammation caused by oxidative stress by activating Nrf2 via the PI3K/Akt pathway. This activation decreases secretion by airway epithelial cells, inhibits smooth muscle cell proliferation, improves airway remodeling, and enhances lung function.179
During HPH progression, impairment of the NO pathway increases pulmonary vascular constriction, worsening the disease. NO is a key vasodilator synthesized in pulmonary vascular endothelial cells from
ERK, a member of the MAPK family, plays a key role in cell growth, differentiation, and proliferation. The ERK pathway is typically activated by receptor-triggered molecular cascades, starting with the activation of Ras, followed by Raf kinase, and cumulating in ERK activation. Once active, ERK enters the nucleus to regulate gene transcription by phosphorylating transcription factors and nuclear proteins, thus influencing cellular functions and fate.182 In HPH, excessive ERK activation affects PASMC survival by altering the expression of cell survival factors and apoptotic regulators, leading to abnormal PASMC proliferation. Cysteine-rich 61 (CCN1), a protein in the CCN family, is crucial for regulating proliferation, differentiation, apoptosis, angiogenesis, and fibrosis. AST IV counters abnormal PASMC proliferation and PVR by inhibiting CCN1 and reducing ERK pathway activation.183
The Nrf2 signaling pathway is a critical defense mechanism against oxidative stress. Under normal conditions, Nrf2 is bound to Kelch-like ECH-associated protein 1 (Keap1) in the cytoplasm and is degraded via the ubiquitin–proteasome degradation pathway, thereby maintaining low Nrf2 levels. During oxidative stress, inhibition of Keap1 or structural changes of Nrf2 allow Nrf2 to escape, translocate to the nucleus, and bind to AREs with cofactors such as Maf proteins. This binding activates the transcription of genes encoding antioxidant enzymes, detoxification enzymes, heat shock proteins, and apoptosis regulators, such as NAD(P)H:quinone oxidoreductase 1 (NQO1), HO-1, and glutamate–cysteine ligase.184 Some natural herbal medicines can activate the Nrf2 pathway by stabilizing the Nrf2–Keap1 complex or enhancing Nrf2 translocation, with Akt phosphorylation also contributing to this activation. Sulforaphane, a potent Nrf2 activator, has been demonstrated to prevent right ventricular dysfunction and remodeling in mice with HPH and right ventricular inflammation and fibrosis. Furthermore, an in vivo study demonstrated that SFN alleviates SuHx-induced PVR, inflammation, and fibrosis by activating the Nrf2/NQO1 pathway.185 Overall, the Nrf2 pathway serves as a vital defense system for cells, playing a key role in maintaining cellular homeostasis and protecting against oxidative and toxic stresses.
The NADPH pathway is vital for cellular antioxidant defense, as NADPH provides reducing power for various redox reactions. It is primarily synthesized by enzymes such as glucose-6-phosphate dehydrogenase and isopentenol-2,4-diol-1,4-dehydrogenase, which help to maintain the reduced state of antioxidant molecules such as GSH and Trx. These molecules are essential for scavenging free radicals and mitigating oxidative damage.186 Additionally, NADPH supports the synthesis of reduced thiol and sulfur compounds, further bolstering cellular antioxidant capacity. Many HMs enhance resistance to oxidative stress and treat HPH by modulating the NADPH pathway. Wogonin, a key component of Scutellaria baicalensis, is known for its anti-inflammatory, anti-angiogenic, and anti-fibrotic properties. In a network pharmacology study, researchers identified 40 potential targets of wogonin in the treatment of HPH. Their analyses, supported by in vitro experiments, suggest that wogonin’s anti-proliferative effects are mediated through the regulation of the HIF-1/NOX4 pathway.187
NF-κB is a nuclear transcription factor involved in regulating inflammation, immune responses, and apoptosis.188 In its inactive state, NF-κB is bound to inhibitor of κB (IκB) in the cytoplasm. External signals, such as inflammatory mediators, cytokines, or oxidative stress, trigger the phosphorylation and degradation of IκB by specific kinases (eg, IKK). This process releases NF-κB, allowing it to translocate to the nucleus, bind DNA, and activate gene transcription. The PI3K/Akt pathway can also activate NF-κB. Activated NF-κB induces the expression of inflammation-related genes, including inflammatory mediators (IL-1β, TNF-α), cell adhesion molecules (ICAM-1, VCAM-1), and pro-inflammatory chemokines (IL-8).189 Additionally, NF-κB influences the proliferation, differentiation, and survival of immune cells, which are crucial for normal immune function. Many TCMs inhibit NF-κB activity, thereby reducing inflammatory factor levels around pulmonary vessels in HPH and improving the local microenvironment. Andrographolide, a TCM with anti-inflammatory, antioxidant, and anti-proliferative properties, regulates oxidative stress through the NOX/Nrf2 pathway and inflammation via NF-κB. It inhibits increases in [Ca2+]i, blocks ROS production, and prevents the upregulation of IL-6, IL-8, ET-1, and VEGF in PASMCs, thereby reversing PVR in patients with HPH.190
The RhoA/ROCK pathway is essential for cellular signal transduction, as it regulates various processes such as cytoskeletal remodeling, cell motility, cell proliferation, and gene transcription. The pathway centers on RhoA, a small GTPase, and ROCK, a protein kinase. RhoA, activated by external stimuli, regulates ROCK, which then phosphorylates substrates such as myosin and actin. This affects cellular contraction, morphology, and signaling. Aberrant activation of the RhoA/ROCK signaling pathway is linked to various diseases, including vascular conditions and tumor metastasis.191 In HPH, RhoA/ROCK pathway activation promotes PASMC proliferation, migration, and contraction, leading to vascular constriction and increased pulmonary arterial resistance, thereby worsening the disease. RhoA/ROCK pathway inhibition is a potential treatment strategy for HPH. Studies reported that ROCK inhibitors, such as fasudil, can reduce pulmonary vascular resistance, improve vascular remodeling, and decrease the right ventricular load, thus enhancing patient outcomes. Sage (Salvia officinalis L.), a traditional TCM with anti-bacterial, anti-viral, antioxidant, and anti-thrombotic properties, improved antioxidant enzyme activities and repair tissue damage in hypoxia-preconditioned mice with HPH. Sage achieved this by downregulating factors such as HIF-1α, PCNA), Bcl-2, CDK4, cyclin D1, and P27Kip1, as well as inhibiting pro-inflammatory cytokines and the RhoA/ROCK pathway.192 Thus, the RhoA/ROCK signaling pathway is crucial for HPH treatment, and further research might lead to new therapeutic advancements.
The mitochondrial apoptosis pathway is a major mechanism of programmed cell death, guiding cells toward apoptosis by regulating mitochondrial structure and function. Apoptotic signals increase mitochondrial outer membrane permeability, causing the release of specific apoptotic inducers such as Cytc into the cytosol. Cytc then binds to other apoptotic proteins, such as apoptosis-inducing factor, to form complexes that activate caspases, including caspase-9, initiating a caspase cascade.193 This cascade leads to apoptosis, which is characterized by DNA degradation, cell membrane rupture, cytoplasmic condensation, and apoptotic body formation. Hypoxia decreases mitochondrial membrane potential, disrupts the electron transport chain, and increases free radical production, thereby damaging mitochondrial function. This disruption can alter intracellular Ca2+ homeostasis, leading to high Ca2+ concentrations, which activate mitochondrial permeability transition pores and further increase outer membrane permeability. Regulatory factors such as Bcl-2 family proteins within mitochondria are involved in this pathway. TCMs often regulate Bcl-2 family proteins, such as Bax and Bak, through antioxidant signaling pathways, influencing mitochondrial outer membrane permeability and mediating PASMC apoptosis194 (Figure 5).
![]() |
Figure 5 Chinese herbal medicine trigger mitochondrial apoptosis through signaling pathway to relieve abnormal proliferation of pulmonary artery smooth muscle in hypoxic pulmonary hypertension.
|
Clinical Translation Challenges and Current Status of Clinical Trials
While robust preclinical studies (including in vitro and in vivo models) compellingly demonstrate the potential of numerous Traditional Chinese Medicine (TCM) monomers, extracts, and formulas in treating hypoxic pulmonary hypertension (HPH) – with mechanisms involving anti-inflammatory, antioxidant, and anti-proliferative effects on pulmonary artery smooth muscle cells (PASMCs), promotion of PASMC apoptosis, protection of endothelial function, and regulation of ion channels and signaling pathways – significant challenges remain in translating these promising preclinical findings into clinical practice. Currently, the efficacy and safety of TCM for HPH still require rigorous validation through well-designed clinical studies.
Limitations of Preclinical Studies and Translation Barriers
The translational potential of current preclinical evidence for human pulmonary hypertension (HPH) faces significant limitations and hurdles. Widely used rodent models, employing chronic hypoxia or monocrotaline (MCT), successfully capture key pathological hallmarks like pulmonary vascular remodeling and right ventricular hypertrophy. However, they fail to recapitulate the full complexity, heterogeneity, and protracted course of human HPH.195 Moreover, inherent species differences in drug metabolism, receptor sensitivity, and immune responses between rodents and humans can lead to divergent pharmacological outcomes, while the predominant use of young, healthy animals inadequately models the common clinical scenario of HPH patients with significant comorbidities (eg, COPD, OSAHS, interstitial lung disease) and age-related factors.195 Compounding this, preclinical studies often utilize high drug dosages and non-clinically relevant routes of administration (eg, intraperitoneal injection), creating a disconnect with feasible human therapeutic regimens; establishing a clinically applicable safe and effective therapeutic window necessitates systematic pharmacokinetic and toxicological bridging studies. Furthermore, research predominantly focuses on single phytochemicals or derivatives (eg, salidroside,100,126 sodium tanshinone IIA sulfonate,84,85 resveratrol,93,133,134 astragaloside IV92,123,124), whose mechanisms are relatively tractable, yet this contrasts sharply with clinical practice utilizing complex traditional Chinese medicine (TCM) formulas (eg, Tongxinluo41,42 or Qili Qiangxin Capsule44). The intricate synergistic, additive, or antagonistic interactions within these multi-component mixtures pose substantial challenges for standardization, quality control, mechanistic elucidation, and complicate clinical validation due to their inherent multi-target nature. Critically, the relatively short duration of preclinical studies hinders comprehensive evaluation of potential long-term adverse effects, such as organ toxicity and drug interactions, associated with TCM constituents, especially given their broad biological activities; long-term safety profiles in HPH patients remain largely undefined. Finally, a fundamental misalignment exists in endpoints: preclinical investigations rely heavily on surrogate metrics like hemodynamics (eg, right ventricular systolic pressure, RVSP), histology (eg, vascular remodeling severity, RV hypertrophy index), and molecular biomarkers (eg, protein expression, cytokine levels), whereas clinical trials prioritize patient-centric outcomes including exercise capacity (eg, 6-minute walk distance, 6MWD), functional class (WHO FC), quality of life, time to clinical worsening, survival, and hemodynamics measured via clinically applicable methods (eg, echocardiography, right heart catheterization).196 This discordance constitutes a major translational barrier.
Current Evidence from Clinical Trials
The current clinical evidence supporting Traditional Chinese Medicine (TCM) for human pulmonary hypertension (HPH) treatment is markedly limited by a critical shortage of high-quality, large-sample randomized controlled trials (RCTs). Available evidence predominantly originates from exploratory investigations, small-scale trials, observational data, or studies targeting broader pulmonary hypertension (PH) cohorts that may incidentally include subsets of HPH patients. Regarding Chinese Herbal Compounds (CHCs), Qiliqiangxin Capsules44 (approved by China’s NMPA for chronic heart failure) have shown promise in exploratory and small RCTs for improving cardiac function, exercise capacity, and quality of life in PH patients, including those with left heart or lung disease etiologies, with proposed mechanisms involving ventricular remodeling amelioration, diuresis, and anti-inflammatory/neuroendocrine modulation; however, robust RCTs dedicated specifically to HPH populations are absent. Similarly, while preclinical studies of Tongxinluo Capsules41,42 (primarily indicated for coronary heart disease and ischemic stroke) in animal HPH models suggest benefits via endothelial protection, anti-inflammation, anti-oxidation, and attenuated pulmonary vascular remodeling, clinical research specifically evaluating its efficacy in human HPH remains exceedingly scarce. For other CHCs such as Xuefuzhuyu Decoction46 or Qibaipingfei Capsules,47 systematic clinical trial data supporting their use in HPH are largely unavailable, as research has been predominantly confined to animal models. Concerning Chinese Herbal Monomers/Extracts, Breviscapine Injection54 (Scutellarin, approved for ischemic cardio-cerebrovascular diseases) has limited low-quality clinical reports suggesting potential symptomatic or hemodynamic benefits in connective tissue disease-associated PAH or chronic cor pulmonale, yet no dedicated studies adequately evaluate its role in HPH. Sodium Tanshinone IIA Sulfonate (STS),145–147 another angina-approved agent, possesses microcirculation-improving and potential anti-remodeling properties; preliminary small studies exploring it as adjuvant therapy for PH/PAH hint at possible benefits in exercise tolerance and hemodynamics, but high-quality RCTs specifically powered for HPH assessment are similarly absent. Potential benefits observed for other monomers like Aucubin102 in preclinical settings await validation through formal clinical investigations in HPH patients. In summary, although preliminary data and mechanistic plausibility suggest potential utility for certain TCM compounds and monomers in HPH management, the clinical evidence landscape remains fragmented and critically constrained by the profound absence of adequately designed, sufficiently powered RCTs specifically enrolling HPH cohorts. This fundamental evidence gap precludes definitive conclusions regarding their efficacy and safety profiles within this distinct patient population.
Critical Knowledge Gaps and Future Directions
Given the profound methodological limitations and the current state of clinical evidence outlined previously, the clinical translation of Traditional Chinese Medicine (TCM) for human pulmonary hypertension (HPH) faces critical challenges and mandates focused future research. The most fundamental gap remains the acute need for rigorously designed, large-sample, multicenter, placebo-controlled randomized controlled trials (RCTs) dedicated to evaluating the efficacy (using primary endpoints such as 6-minute walk distance [6MWD] and time to clinical worsening) and long-term safety profiles of specific TCM interventions (whether monocomponent agents or standardized complex formulas) within well-defined HPH populations, characterized by precise inclusion criteria reflecting underlying lung disease etiology and PH severity alongside standardized treatment protocols. Addressing the inherent heterogeneity among HPH patients, arising from diverse etiologies, varying disease severities, and distinct comorbidities, requires future research to actively pursue patient stratification strategies leveraging biomarkers of inflammation, oxidative stress, vasoactive factors, and advanced imaging phenotypes to identify specific subgroups most likely to derive benefit from particular TCM therapies. Concurrently, the limited efficacy of current standard therapies (eg, long-term oxygen therapy, pulmonary disease management) underscores the critical importance of exploring TCM as an adjuvant to conventional or emerging Western pharmacotherapies within integrated combination strategies; research must rigorously evaluate potential synergistic effects, safety profiles, and long-term outcomes of such combinations. The inherent complexity of TCM formulations necessitates an unwavering commitment to ensuring batch-to-batch consistency, stability of active constituents, and safety through the development and stringent enforcement of comprehensive quality standards and Good Manufacturing Practices (GMP). Furthermore, bridging the translational divide requires embedding mechanistic investigations (eg, biomarker analyses, advanced imaging to assess vascular remodeling) within clinical trials to validate whether mechanisms identified preclinically hold true in humans and provide a biological rationale for observed clinical effects. Given the chronic, progressive nature of HPH, the persistent efficacy of any TCM intervention must be evaluated through long-term follow-up studies spanning several years, incorporating assessments of sustained clinical benefits, impacts on survival, and systematic surveillance for potential late-emerging adverse effects including hepatic, renal, and immunomodulatory consequences. Finally, complementing objective clinical measures, a dedicated emphasis on patient-reported outcomes (PROs) focusing on improvements in dyspnea, fatigue levels, functional capacity in daily activities, and overall health-related quality of life is essential to fully capture the therapeutic value of TCM from the patient perspective.
Deficiencies and Prospects
Currently, diagnostic examinations for PH include right heart catheterization, blood gas analysis, X-ray, computed tomography (CT), and echocardiography. Of these, right heart catheterization is the only invasive procedure that provides direct hemodynamic assessment of the pulmonary artery and right ventricle through peripheral venous access, making it the gold standard for diagnosing PH. However, invasive procedures carry risks such as infection and mortality, and human error can alter normal pulmonary vascular and cardiac functions in animal models,196 potentially leading to unnecessary resource waste.
Among non-invasive methods, blood gas analysis can only assess hypoxemia and the acid-base status without quantifying disease progression. Chest X-ray and CT can observe the size and diameter of pulmonary arteries to assess disease progression, but such changes are negligible in mouse models. Echocardiography uses tricuspid regurgitation velocity to estimate pulmonary artery pressure via the Bernoulli equation; however, even the smallest animal ultrasound probes are significantly larger than the radiation area of a mouse’s cardiovascular system. Furthermore, all of these tests detect established pulmonary arterial hypertension, but they cannot identify high-risk individuals or monitor disease progression in animal models.
Focusing on the disease itself, chronic hypoxia often leads to ROS accumulation in the lungs, and ROS levels around the pulmonary vasculature in HPH can reflect disease progression to some extent. Therefore, targeting ROS and synthesizing probes to monitor the progression of HPH represent novel examination and monitoring approaches.
In recent years, significant research has explored the use of probes in medical biology, particularly for acute kidney injury (AKI), a condition with high incidence and mortality rates. Early diagnosis is critical for AKI treatment and prognosis. Although traditional urinalysis methods enable non-invasive molecular-level diagnoses of AKI, their sensitivity remains insufficient. Vanin-1 is considered as an early and sensitive biomarker of AKI. Recent studies developed CL-Pa as a novel chemiluminescent probe targeting vanin-1. This probe significantly aids in the early diagnosis of AKI and assessment of the efficacy of anti-AKI drugs.197 Another study on atherosclerosis reported a novel small-molecule fluorescent probe capable of sequentially imaging and detecting gamma-glutamyl transferase and hypobromous acid. This probe both accurately identifies the locations of mature plaques and successfully predicts the occurrence of atherosclerotic plaques before they become detectable by conventional immunofluorescence or visual inspection. This technology enables imaging of the locations of mature atherosclerotic plaques and provides early indications of plaque formation.198 In conclusion, chemical fluorescent probes possess high sensitivity, real-time capability, and intuitiveness. When combined with appropriate targets, they hold promise for quantifying the progression of HPH in disease models and providing early diagnosis.
In recent years, network pharmacology, bulk RNA-seq, single-cell sequencing, and spatial transcriptomics have flourished in biomedical research, but they have yet to be widely applied in the treatment of HPH with TCM. Network pharmacology is an emerging branch of pharmacology based on the principles of systems biology. It reveals the efficacy, toxicity, and metabolic characteristics of drugs through the construction and analysis of biological networks. The research methods of network pharmacology include constructing a multi-layer biological network of “drug–component–target–disease” and utilizing gene functional annotation and pathway enrichment analysis to investigate the mechanisms of drug action in depth. For instance, in a study of the TCM compound Shenkang injection (SKI) in the treatment of diabetes, researchers screened 280 drug targets from the TCMSP database and identified 1197 diabetic kidney disease (DKD) targets from five disease databases: GenGards, OMIM, Drugbank, TTD, and Disgenet. They performed a protein-protein interaction (PPI) network analysis of the intersection, ultimately identifying 118 overlapping targets. The PPI analysis identified core targets such as NOS3, PTGS2, CASP3, CCL2, CXCL8, HIF1A, and AKT1. In the “drug-component-target-disease” network, quercetin, kaempferol, and luteolin exhibited the highest degrees of interaction, suggesting they are the key active components of SKI in the treatment of DKD.199 This method both predicts the material basis and mechanisms of TCM efficacy and elucidates the rules of drug combinations and formula compatibility, providing new insights for the complex system research of TCM. Furthermore, the applications of network pharmacology in the field of TCM include identifying potential active ingredients in single herbs or formulas, thus elucidating the mechanisms of action of TCM or formulas and interpreting the scientific connotations of compatibility contraindications in TCM. The advent of single-cell sequencing has enabled researchers to gain deeper insights into cellular heterogeneity, which is challenging to achieve with traditional sequencing technologies. Additionally, single-cell sequencing technology can measure mRNA levels in individual cells, providing insights into the gene expression status of those cells. In cancer research, single-cell sequencing can assist in identifying the roles of different cell types within the tumor microenvironment and their influence on treatment responses in tumor development. In a study on liver cancer,200 single cell RNA-seq revealed unique immune characteristics of early recurrent hepatocellular carcinoma, including a reduction in Treg counts and an increase in dendritic cell counts. In addition, CD8+ T cells exhibited an innate-like functional impairment state characterized by low cytotoxicity and a low clonal expansion phenotype, and early-relapse tumors exhibited low proliferative capacity and high immune evasion ability. Recurrent malignant cells can disrupt antigen presentation by dendritic cells via the PD-L1/CD80 and CTLA4/CD80 axes and recruit innate CD161+ CD8+ T cells through the CCL20/CCR6 axis, thereby creating a dysfunctional anti-tumor immune response in early recurrent hepatocellular carcinoma. These findings provide a theoretical and experimental basis for improving the efficacy of immunotherapy in liver cancer and identifying strategies to prevent recurrence and metastasis.
Conclusions
This comprehensive analysis establishes that Traditional Chinese Medicine (TCM) exerts multi-target therapeutic effects against hypoxic pulmonary hypertension (HPH) through synergistic modulation of critical pathological pathways. Bioactive components—including Qiliqiangxin compound formulations, salidroside, resveratrol, and sodium tanshinone IIA sulfonate—concurrently ameliorate oxidative stress by activating Nrf2/HO-1 and PI3K/Akt signaling to reduce ROS accumulation; suppress inflammatory cascades via NF-κB inhibition to lower TNF-α, IL-6, and MCP-1 levels; reverse pulmonary vascular remodeling through regulation of K+/Ca²+ channels, induction of PASMC apoptosis, and inhibition of abnormal proliferation via AMPK/p53 and RhoA/ROCK pathways; and protect endothelial function by activating BMPR2/Smad signaling and the NO-sGC-cGMP axis. Critically, TCM’s holistic approach addresses HPH’s multifactorial complexity more effectively than monotherapies by concurrently targeting complementary disease mechanisms.
Nevertheless, clinical translation faces significant challenges: inconsistent quality control in compound preparations risks batch-dependent efficacy variations, pharmacokinetic uncertainties of multi-component systems impede dosage optimization, and rigorous validation of long-term safety and efficacy in human HPH remains limited. Future research should prioritize three critical domains: standardizing bioactive constituents using chromatographic fingerprinting techniques; validating novel targets such as Aucubin-mediated redox balance through advanced methodologies like single-cell sequencing and spatial transcriptomics; and accelerating clinical innovation via ROS-responsive diagnostic probes, network pharmacology-guided TCM combinations, and randomized trials benchmarking TCM against existing therapies.
In summary, TCM represents a promising paradigm for HPH treatment by integrating multi-target efficacy with holistic intervention. Bridging its empirical foundation with modern scientific rigor—through standardized compound development, mechanism-driven target discovery, and tailored clinical validation—will catalyze the development of next-generation HPH therapies that synergize therapeutic potency with regulatory robustness.
Abbreviations
AREs, antioxidant response elements; AKI, acute kidney injury; AST IV, Astragaloside IV; AMPK, AMP-activated protein kinase; Bsg, Basigin; COPD, chronic obstructive pulmonary disease; CHCs, Chinese herbal compounds; COX-2, cyclooxygenase-2; HMs, Chinese herbal monomers; CCL2, CC-motif chemokine ligand 2; CaSRs, Calcium-sensing receptors; Cytc, Cytochrome C; CT, computed tomography; CAT, catalase; DKD, diabetic kidney disease; ERK, extracellular signal-regulated kinase; ET-1, endothelin-1; EndMT, Endothelial-to-mesenchymal transition; GSH-Px, glutathione peroxidase; HPH, Hypoxic pulmonary hypertension; HPV, hypoxic pulmonary vasoconstriction; HSYA, Hydroxysafflor yellow A; HIF-1α, hypoxia-inducible factor 1α; HMGB1, high mobility group box 1 protein; HO-1, heme oxygenase-1; [Ca2+]I, intracellular calcium concentration; IκB, inhibitor of κB; Keap1, Kelch-like ECH-associated protein 1; MCP-1, Monocyte chemoattractant protein-1; MMP-9, matrix metalloproteinase-9; MLB, magnesium lithospermate B; mPAP, mean pulmonary arterial pressure; Nrf2, Nuclear factor E2-related factor 2; NFATc3, nuclear factor of activated T cells; NQO1, NAD(P)H:quinone oxidoreductase 1; NO, nitric oxide; PH, Pulmonary hypertension; PASMCs, pulmonary artery smooth muscle cells; PAEC, pulmonary arterial endothelial cell; PDE5, phosphodiesterase-5; PVR, pulmonary vascular remodeling; PaO2, pressure of oxygen in arterial blood; PPARγ, proliferator-activated receptor γ; PCNA, proliferating cell nuclear antigen; PKG-1, protein kinase G1; ROS, reactive oxygen species; ROCK, Rho kinase; STS, sodium tanshinone IIA sulfonate; sGC1, soluble guanylate cyclase 1; SphK1/S1P, Sphingosine-1-phosphate; SKI, Shenkang injection; SOD, superoxide dismutase; SOCE, store-operated calcium entry; TNF, tumor necrosis factor; TCM, Traditional Chinese medicine; Trx-1, thioredoxin-1; TGF-β1, transforming growth factor-β1; VEGF, vascular endothelial growth factor; VCAM-1, vascular cell adhesion molecule-1.
Acknowledgment
We thank Medjaden Inc. for scientific editing of this paper.
Funding
This work was supported by the National Natural Science Foundation of China (NSFC.8246036282470511); Hainan Key Research and Development Social Development Project (ZDYF2022SHFZ293 ZDYF2024SHFZ120 ZDYF2025SHFZ050); Natural Science Foundation of Hainan Province (823MS146); Hainan Provincial Health Commission Scientific Research Project (22A200032) and Hainan Province Clinical Medical Center (2021).
Disclosure
The authors declare that they have no conflict of interest.
References
1. Humbert M, Kovacs G, Hoeper MM, et al. 2022 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Respir J. 2023;61(1):1.
2. Ulloa NA, Cook J. Altitude-induced pulmonary hypertension. In: StatPearls. Treasure Island (FL): StatPearls Publishing LLC.; 2024.
3. Pullamsetti SS, Mamazhakypov A, Weissmann N, Seeger W, Savai R. Hypoxia-inducible factor signaling in pulmonary hypertension. J Clin Invest. 2020;130(11):5638–5651. doi:10.1172/JCI137558
4. Galiè N, Humbert M, Vachiery JL, et al. 2015 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension: the joint task force for the diagnosis and treatment of pulmonary hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS): endorsed by: association for European Paediatric and Congenital Cardiology (AEPC), International Society for Heart and Lung Transplantation (ISHLT). Eur Heart J. 2016;37(1):67–119. doi:10.1093/eurheartj/ehv317
5. Hoeper MM, Humbert M, Souza R, et al. A global view of pulmonary hypertension. Lancet Respir Med. 2016;4(4):306–322. doi:10.1016/S2213-2600(15)00543-3
6. 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
7. de Jesus Perez VA. Molecular pathogenesis and current pathology of pulmonary hypertension. Heart Fail Rev. 2016;21(3):239–257. doi:10.1007/s10741-015-9519-2
8. Humbert M, Guignabert C, Bonnet S, et al. Pathology and pathobiology of pulmonary hypertension: state of the art and research perspectives. Eur Respir J. 2019;53(1):1801887. doi:10.1183/13993003.01887-2018
9. Thenappan T, Ormiston ML, Ryan JJ, Archer SL. Pulmonary arterial hypertension: pathogenesis and clinical management. BMJ. 2018;360:j5492. doi:10.1136/bmj.j5492
10. Vazquez ZGS, Klinger JR. Guidelines for the Treatment of Pulmonary Arterial Hypertension. Lung. 2020;198(4):581–596. doi:10.1007/s00408-020-00375-w
11. Goldberg AB, Mazur W, Kalra DK. Pulmonary hypertension: diagnosis, imaging techniques, and novel therapies. Cardiovasc Diagn Ther. 2017;7(4):405–417. doi:10.21037/cdt.2017.04.11
12. Hu Y, Chi L, Kuebler WM, Goldenberg NM. Perivascular inflammation in pulmonary arterial hypertension. Cells. 2020;9(11):2338. doi:10.3390/cells9112338
13. Tuder RM, Abman SH, Braun T, et al. Development and pathology of pulmonary hypertension. J Am Coll Cardiol. 2009;54(1 Suppl):S3–s9. doi:10.1016/j.jacc.2009.04.009
14. Walsh-Sukys MC, Tyson JE, Wright LL, et al. Persistent pulmonary hypertension of the newborn in the era before nitric oxide: practice variation and outcomes. Pediatrics. 2000;105(1 Pt 1):14–20. doi:10.1542/peds.105.1.14
15. Lau EMT, Giannoulatou E, Celermajer DS, Humbert M. Epidemiology and treatment of pulmonary arterial hypertension. Nat Rev Cardiol. 2017;14(10):603–614. doi:10.1038/nrcardio.2017.84
16. Vogelmeier CF, Criner GJ, Martinez FJ, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease 2017 report. GOLD executive summary. Am J Respir Crit Care Med. 2017;195(5):557–582. doi:10.1164/rccm.201701-0218PP
17. Nathan SD, Barbera JA, Gaine SP, et al. Pulmonary hypertension in chronic lung disease and hypoxia. Eur Respir J. 2019;53(1):1801914. doi:10.1183/13993003.01914-2018
18. Licker M, Hagerman A, Jeleff A, Schorer R, Ellenberger C. The hypoxic pulmonary vasoconstriction: from physiology to clinical application in thoracic surgery. Saudi J Anaesth. 2021;15(3):250–263. doi:10.4103/sja.sja_1216_20
19. 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
20. Arriaza K, Cuevas C, Pena E, Siques P, Brito J. Impact of zinc on oxidative signaling pathways in the development of pulmonary vasoconstriction induced by hypobaric hypoxia. Int J Mol Sci. 2022;23(13):6974. doi:10.3390/ijms23136974
21. Gassmann M, Cowburn A, Gu H, et al. Hypoxia-induced pulmonary hypertension-Utilizing experiments of nature. Br J Pharmacol. 2021;178(1):121–131. doi:10.1111/bph.15144
22. Wedgwood S, Devol JM, Grobe A, et al. Fibroblast growth factor-2 expression is altered in lambs with increased pulmonary blood flow and pulmonary hypertension. Pediatr Res. 2007;61(1):32–36. doi:10.1203/01.pdr.0000250013.77008.28
23. Mata-Greenwood E, Meyrick B, Steinhorn RH, Fineman JR, Black SM. Alterations in TGF-beta1 expression in lambs with increased pulmonary blood flow and pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2003;285(1):L209–221. doi:10.1152/ajplung.00171.2002
24. Aimaier S, Tao Y, Lei F, et al. Protective effects of the Terminalia bellirica tannin-induced Nrf2/HO-1 signaling pathway in rats with high-altitude pulmonary hypertension. BMC Complement Med Ther. 2023;23(1):150. doi:10.1186/s12906-023-03981-2
25. Chen J, Jiang W, Zhu F, Wang Q, Yang H, Wu J. Curcumin improves pulmonary hypertension rats by regulating mitochondrial function. Biomed Res Int. 2021;2021:1078019. doi:10.1155/2021/1078019
26. Cao X, He Y, Li X, Xu Y, Liu X. The IRE1α-XBP1 pathway function in hypoxia-induced pulmonary vascular remodeling, is upregulated by quercetin, inhibits apoptosis and partially reverses the effect of quercetin in PASMCs. Am J Transl Res. 2019;11(2):641–654.
27. Liu J, Wang W, Wang L, et al. IL-33 initiates vascular remodelling in hypoxic pulmonary hypertension by up-regulating HIF-1α and VEGF expression in vascular endothelial cells. EBioMedicine. 2018;33:196–210. doi:10.1016/j.ebiom.2018.06.003
28. Pena E, Brito J, El Alam S, Siques P. Oxidative stress, kinase activity and inflammatory implications in right ventricular hypertrophy and heart failure under hypobaric hypoxia. Int J Mol Sci. 2020;21(17):6421. doi:10.3390/ijms21176421
29. Zhang B, Niu W, Dong HY, Liu ML, Luo Y, Li ZC. Hypoxia induces endothelial‑mesenchymal transition in pulmonary vascular remodeling. Int J Mol Med. 2018;42(1):270–278. doi:10.3892/ijmm.2018.3584
30. Yu M, Peng L, Liu P, et al. Paeoniflorin ameliorates chronic Hypoxia/SU5416-induced pulmonary arterial hypertension by inhibiting endothelial-to-mesenchymal transition. Drug Des Devel Ther. 2020;14:1191–1202. doi:10.2147/DDDT.S235207
31. Gierhardt M, Pak O, Walmrath D, et al. Impairment of hypoxic pulmonary vasoconstriction in acute respiratory distress syndrome. Eur Respir Rev. 2021;30(161):210059. doi:10.1183/16000617.0059-2021
32. Sydykov A, Mamazhakypov A, Maripov A, et al. Pulmonary hypertension in acute and chronic high altitude maladaptation disorders. Int J Environ Res Public Health. 2021;18(4):1692.
33. Zhang L, Yu J, Zhou Y, Shen M, Sun L. Becoming a faithful defender: traditional Chinese medicine against coronavirus disease 2019 (COVID-19). Am J Chin Med. 2020;48(4):763–777. doi:10.1142/S0192415X2050038X
34. Fan Y, Ma Z, Zhao L, et al. Anti-tumor activities and mechanisms of traditional Chinese medicines formulas: a review. Biomed Pharmacother. 2020;132:110820. doi:10.1016/j.biopha.2020.110820
35. Rao T, Tan Z, Peng J, et al. The pharmacogenetics of natural products: a pharmacokinetic and pharmacodynamic perspective. Pharmacol Res. 2019;146:104283. doi:10.1016/j.phrs.2019.104283
36. Shi J, Yang Y, Zhou X, et al. The current status of old traditional medicine introduced from Persia to China. Front Pharmacol. 2022;13:953352. doi:10.3389/fphar.2022.953352
37. Tang JL, Liu BY, Ma KW. Traditional Chinese medicine. Lancet. 2008;372(9654):1938–1940. doi:10.1016/S0140-6736(08)61354-9
38. Xue Z, Li Y, Zhou M, et al. Traditional herbal medicine discovery for the treatment and prevention of pulmonary arterial hypertension. Front Pharmacol. 2021;12:720873. doi:10.3389/fphar.2021.720873
39. Chan E, Tan M, Xin J, Sudarsanam S, Johnson DE. Interactions between traditional Chinese medicines and Western therapeutics. Curr Opin Drug Discovery Dev. 2010;13(1):50–65.
40. Jiang WY. Therapeutic wisdom in traditional Chinese medicine: a perspective from modern science. Discov Med. 2005;5(29):455–461.
41. Wang Y, Kuang X, Yin Y, et al. Tongxinluo prevents chronic obstructive pulmonary disease complicated with atherosclerosis by inhibiting ferroptosis and protecting against pulmonary microvascular barrier dysfunction. Biomed Pharmacother. 2022;145:112367. doi:10.1016/j.biopha.2021.112367
42. Wang Y, Ma TT, Gao NN, et al. Effect of Tongxinluo on pulmonary hypertension and pulmonary vascular remodeling in rats exposed to a low pressure hypoxic environment. J Ethnopharmacol. 2016;194:668–673. doi:10.1016/j.jep.2016.10.004
43. Tsai HH, Chen IJ, Lo YC. Effects of San-Huang-Xie-Xin-Tang on U46619-induced increase in pulmonary arterial blood pressure. J Ethnopharmacol. 2008;117(3):457–462. doi:10.1016/j.jep.2008.02.024
44. Lu Y, Wu J, Sun Y, et al. Qiliqiangxin prevents right ventricular remodeling by inhibiting apoptosis and improving metabolism reprogramming with pulmonary arterial hypertension. Am J Transl Res. 2020;12(9):5655–5669.
45. Wu P, Xie X, Chen M, et al. Elucidation of the mechanisms and molecular targets of Qishen Yiqi formula for the treatment of pulmonary arterial hypertension using a bioinformatics/network topology-based strategy. Comb Chem High Throughput Screen. 2021;24(5):701–715. doi:10.2174/1386207323666201019145354
46. Li MJ, Guo L, Yan JP, Chen Y, Dong L, Cai WR. Study on effect of Xuefu Zhuyu decoction on pulmonary vascular remodeling in hypoxic pulmonary hypertension rats via PI3K/AKT/mTOR signaling pathway. Chin J Mod Appl Pharm. 2020;37(21):2576–2581.
47. Zhu ZQ, Zhu J, Tong JB, Li ZG, Fang L. Effects of Qibai Pingfei capsule on rats with COPD complicated with PAH based on JAK1/STAT3 signal pathway. Chin J Tradit Chin Med Pharm. 2023;38(1):324–330.
48. Zhang L, Tong XL, Wang XL, Zhu J, Li ZG. Qibai pingfei capsule promotes the apoptosis of pulmonary artery smooth muscle cells in rats with hypoxia by inhibiting the phosphoinositide 3-kinase/protein kinase b pathway. J Anhui Univ Chin Med. 2022;41(02):55–60.
49. Yang ZT. Effect and mechanism of Tibetan MedicineTsantan Sumtang on right ventricular function in hypoxia-induced pulmonary hypertension rats. Qinghai University; 2021.
50. Wu ZY. Base on microrna-130a targeting gax to explore the effect of compound xiebai capsule on vascular remodeling in pulmonary arterial hypertension. Nanjing University of Traditional Chinese Medicine; 2020.
51. Tan JL, Yi J, Cao XY, Wang FY, Ding RZ, Dai AQ. Exploration of the effects and mechanisms of feixin decoction on monocrotaline-lnduced pulmonary arterial hypertension in rats based on ppar-γ/nf-κb signaling pathway. Trad Chin Drug Res Clin Pharm. 2024;35(03):307–316.
52. Li YM, Cao DY, Chen B, Li ZY, Yu XL. Study on the mechanism of Qilongtian Capsules in the treatment of hypoxic pulmonary hypertension based on UPLC-Q-Exactive-Orbitrap-MS/MS and network pharmacology. Chin J Hosp Pharm. 2023;44(20):1–8.
53. Ma YT, Si MD, Zhang XY, Jia YX, Yan CH, Ma DL. Therapy Mechanism of Fuling Gancao Decoction on Pulmonary Hypertension based on Network Pharmacology and Molecular Docking. Clin J Trad Med. 2022;34(04):675–682.
54. Li YY, Guo RJ, Xie YM, et al. Expert consensus on injection of breviscapine in clinical practice. Zhongguo Zhong Yao Za Zhi. 2020;45(10):2296–2299. doi:10.19540/j.cnki.cjcmm.20200217.501
55. Zeng Z, Wang X, Cui L, Wang H, Guo J, Chen Y. Natural products for the treatment of pulmonary hypertension: mechanism, progress, and future opportunities. Curr Issues Mol Biol. 2023;45(3):2351–2371. doi:10.3390/cimb45030152
56. Liang S, Desai AA, Black SM, Tang H. Cytokines, Chemokines, and Inflammation in Pulmonary Arterial Hypertension. Adv Exp Med Biol. 2021;1303:275–303.
57. Mercurio V, Cuomo A, Naranjo M, Hassoun PM. Inflammatory mechanisms in the pathogenesis of pulmonary arterial hypertension: recent advances. Compr Physiol. 2021;11(2):1805–1829. doi:10.1002/j.2040-4603.2021.tb00165.x
58. Langer F, Schramm R, Bauer M, Tscholl D, Kunihara T, Schäfers HJ. Cytokine response to pulmonary thromboendarterectomy. Chest. 2004;126(1):135–141. doi:10.1378/chest.126.1.135
59. Chun C, Yang W, Xueding C, et al. Resveratrol downregulates acute pulmonary thromboembolism-induced pulmonary artery hypertension via p38 mitogen-activated protein kinase and monocyte chemoattractant protein-1 signaling in rats. Life Sci. 2012;90(19–20):721–727. doi:10.1016/j.lfs.2012.03.008
60. Lin JW, Yang LH, Ren ZC, et al. Resveratrol downregulates TNF-α-induced monocyte chemoattractant protein-1 in primary rat pulmonary artery endothelial cells by P38 mitogen-activated protein kinase signaling. Drug Des Devel Ther. 2019;13:1843–1853. doi:10.2147/DDDT.S184785
61. Pang Y, Liang MT, Gong Y, et al. HGF reduces disease severity and inflammation by attenuating the NF-κB signaling in a rat model of pulmonary artery hypertension. Inflammation. 2018;41(3):924–931. doi:10.1007/s10753-018-0747-1
62. Shi R, Wei Z, Zhu D, et al. Baicalein attenuates monocrotaline-induced pulmonary arterial hypertension by inhibiting vascular remodeling in rats. Pulm Pharmacol Ther. 2018;48:124–135. doi:10.1016/j.pupt.2017.11.003
63. Zhang F, Wang Y, Liu P, Di P, Li M, Wang C. Puerarin exhibits antiinflammatory properties in gunpowder smog-induced acute lung injury in rats via regulation of the renin-angiotensin system and the NFκB signaling pathway. Exp Ther Med. 2021;22(2):809. doi:10.3892/etm.2021.10241
64. Hu G, Song Y, Ke S, et al. Tanshinone IIA protects against pulmonary arterial hypertension in broilers. Poult Sci. 2017;96(5):1132–1138. doi:10.3382/ps/pew322
65. Groth A, Vrugt B, Brock M, Speich R, Ulrich S, Huber LC. Inflammatory cytokines in pulmonary hypertension. Respir Res. 2014;15(1):47. doi:10.1186/1465-9921-15-47
66. Chen F, Wang H, Zhao J, et al. Grape seed proanthocyanidin inhibits monocrotaline-induced pulmonary arterial hypertension via attenuating inflammation: in vivo and in vitro studies. J Nutr Biochem. 2019;67:72–77. doi:10.1016/j.jnutbio.2019.01.013
67. Sánchez-Gloria JL, Martínez-Olivares CE, Rojas-Morales P, et al. Anti-inflammatory effect of allicin associated with fibrosis in pulmonary arterial hypertension. Int J Mol Sci. 2021;22(16):8600. doi:10.3390/ijms22168600
68. Ornatowski W, Lu Q, Yegambaram M, et al. Complex interplay between autophagy and oxidative stress in the development of pulmonary disease. Redox Biol. 2020;36:101679. doi:10.1016/j.redox.2020.101679
69. Zheng W, Wang Z, Jiang X, Zhao Q, Shen J. Targeted drugs for treatment of pulmonary arterial hypertension: past, present, and future perspectives. J Med Chem. 2020;63(24):15153–15186. doi:10.1021/acs.jmedchem.0c01093
70. Yu Z, Xiao J, Chen X, et al. Bioactivities and mechanisms of natural medicines in the management of pulmonary arterial hypertension. Chin Med. 2022;17(1):13. doi:10.1186/s13020-022-00568-w
71. Bonnet S, Boucherat O. The ROS controversy in hypoxic pulmonary hypertension revisited. Eur Respir J. 2018;51(3):1800276. doi:10.1183/13993003.00276-2018
72. Li Y, Pagano PJ. Microvascular NADPH oxidase in health and disease. Free Radic Biol Med. 2017;109:33–47. doi:10.1016/j.freeradbiomed.2017.02.049
73. Nisbet RE, Graves AS, Kleinhenz DJ, et al. The role of NADPH oxidase in chronic intermittent hypoxia-induced pulmonary hypertension in mice. Am J Respir Cell Mol Biol. 2009;40(5):601–609. doi:10.1165/2008-0145OC
74. Harris C, Hansen JM. Nrf2-mediated resistance to oxidant-induced redox disruption in embryos. Birth Defects Res B Dev Reprod Toxicol. 2012;95(3):213–218. doi:10.1002/bdrb.21005
75. Chen Y, Yuan T, Zhang H, et al. Activation of Nrf2 attenuates pulmonary vascular remodeling via inhibiting endothelial-to-mesenchymal transition: an insight from a plant polyphenol. Int J Biol Sci. 2017;13(8):1067–1081. doi:10.7150/ijbs.20316
76. Zhang YS, He L, Liu B, et al. A novel pathway of NADPH oxidase/vascular peroxidase 1 in mediating oxidative injury following ischemia-reperfusion. Basic Res Cardiol. 2012;107(3):266. doi:10.1007/s00395-012-0266-4
77. Li T, Peng JJ, Wang EL, et al. Magnesium Lithospermate B derived from Salvia miltiorrhiza ameliorates right ventricle remodeling in pulmonary hypertensive rats via inhibition of NOX/VPO1 pathway. Planta Med. 2019;85(9–10):708–718. doi:10.1055/a-0863-4741
78. Liu B, Luo XJ, Yang ZB, et al. Inhibition of NOX/VPO1 pathway and inflammatory reaction by trimethoxystilbene in prevention of cardiovascular remodeling in hypoxia-induced pulmonary hypertensive rats. J Cardiovasc Pharmacol. 2014;63(6):567–576. doi:10.1097/FJC.0000000000000082
79. Zhang L, Deng M, Zhou S. Tetramethylpyrazine inhibits hypoxia-induced pulmonary vascular leakage in rats via the ROS-HIF-VEGF pathway. Pharmacology. 2011;87(5–6):265–273. doi:10.1159/000326082
80. Yang DP, Dong WP, Yang YC, et al. Tetramethylpyrazine improves monocrotaline-induced pulmonary hypertension through the ROS/iNOS/PKG-1 axis. J Healthc Eng. 2022;2022:1890892. doi:10.1155/2022/1890892
81. Lambert M, Capuano V, Olschewski A, et al. Ion channels in pulmonary hypertension: a therapeutic interest? Int J Mol Sci. 2018;19(10):3162. doi:10.3390/ijms19103162
82. Lai N, Lu W, Wang J. Ca(2+) and ion channels in hypoxia-mediated pulmonary hypertension. Int J Clin Exp Pathol. 2015;8(2):1081–1092.
83. Mondéjar-Parreño G, Cogolludo A, Perez-Vizcaino F. Potassium (K(+)) channels in the pulmonary vasculature: implications in pulmonary hypertension Physiological, pathophysiological and pharmacological regulation. Pharmacol Ther. 2021;225:107835. doi:10.1016/j.pharmthera.2021.107835
84. Wang J, Dong MQ, Liu ML, et al. Tanshinone IIA modulates pulmonary vascular response to agonist and hypoxia primarily via inhibiting Ca2+ influx and release in normal and hypoxic pulmonary hypertension rats. Eur J Pharmacol. 2010;640(1–3):129–138. doi:10.1016/j.ejphar.2010.04.047
85. Wang J, Jiang Q, Wan L, et al. Sodium tanshinone IIA sulfonate inhibits canonical transient receptor potential expression in pulmonary arterial smooth muscle from pulmonary hypertensive rats. Am J Respir Cell Mol Biol. 2013;48(1):125–134. doi:10.1165/rcmb.2012-0071OC
86. Zhang H, Chang Z, Mehmood K, et al. Tetramethylpyrazine inhibited hypoxia-induced expression of calcium-sensing receptors in pulmonary artery smooth muscle cells in chickens. J Biol Regul Homeost Agents. 2018;32(3):489–495.
87. Hong Z, Weir EK, Nelson DP, Olschewski A. Subacute hypoxia decreases voltage-activated potassium channel expression and function in pulmonary artery myocytes. Am J Respir Cell Mol Biol. 2004;31(3):337–343. doi:10.1165/rcmb.2003-0386OC
88. Huang YF, Liu ML, Dong MQ, et al. Effects of sodium tanshinone II A sulphonate on hypoxic pulmonary hypertension in rats in vivo and on Kv2.1 expression in pulmonary artery smooth muscle cells in vitro. J Ethnopharmacol. 2009;125(3):436–443. doi:10.1016/j.jep.2009.07.020
89. Wisutthathum S, Demougeot C, Totoson P, et al. Eulophia macrobulbon extract relaxes rat isolated pulmonary artery and protects against monocrotaline-induced pulmonary arterial hypertension. Phytomedicine. 2018;50:157–165. doi:10.1016/j.phymed.2018.05.014
90. Zhang R, Li Z, Liu C, et al. Pretreatment with the active fraction of Rhodiola tangutica (Maxim.) S.H. Fu rescues hypoxia-induced potassium channel inhibition in rat pulmonary artery smooth muscle cells. J Ethnopharmacol. 2022;283:114734. doi:10.1016/j.jep.2021.114734
91. Guignabert C, Dorfmuller P. Pathology and pathobiology of pulmonary hypertension. Semin Respir Crit Care Med. 2013;34(5):551–559. doi:10.1055/s-0033-1356496
92. Li C, Zhu H, Zhang S, et al. Astragaloside IV ameliorates pulmonary vascular remodeling in hypoxia-induced pulmonary hypertension by restraining the T follicular helper cell response and expanding T follicular regulatory cell response. Phytomedicine. 2022;102:154171. doi:10.1016/j.phymed.2022.154171
93. Guan Z, Shen L, Liang H, et al. Resveratrol inhibits hypoxia-induced proliferation and migration of pulmonary artery vascular smooth muscle cells by inhibiting the phosphoinositide 3-kinase/protein kinase B signaling pathway. Mol Med Rep. 2017;16(2):1653–1660. doi:10.3892/mmr.2017.6814
94. Li MX, Jiang DQ, Wang Y, et al. Signal mechanisms of vascular remodeling in the development of pulmonary arterial hypertension. J Cardiovasc Pharmacol. 2016;67(2):182–190. doi:10.1097/FJC.0000000000000328
95. Piantadosi CA, Suliman HB. Mitochondrial Dysfunction in Lung Pathogenesis. Annu Rev Physiol. 2017;79:495–515. doi:10.1146/annurev-physiol-022516-034322
96. Teng C, Li B, Lin C, et al. Targeted delivery of baicalein-p53 complex to smooth muscle cells reverses pulmonary hypertension. J Control Release. 2022;341:591–604. doi:10.1016/j.jconrel.2021.12.006
97. 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
98. Zhu Z, Xu Y, Zou H, Zhang Z, Ni W, Chen S. Effects of puerarin on pulmonary vascular remodeling and protein kinase C-alpha in chronic cigarette smoke exposure smoke-exposed rats. J Huazhong Univ Sci Technolog Med Sci. 2008;28(1):27–32. doi:10.1007/s11596-008-0107-8
99. He Y, Fang X, Shi J, Li X, Xie M, Liu X. Apigenin attenuates pulmonary hypertension by inducing mitochondria-dependent apoptosis of PASMCs via inhibiting the hypoxia inducible factor 1α-KV1.5 channel pathway. Chem Biol Interact. 2020;317:108942. doi:10.1016/j.cbi.2020.108942
100. Huang X, Zou L, Yu X, et al. Salidroside attenuates chronic hypoxia-induced pulmonary hypertension via adenosine A2a receptor related mitochondria-dependent apoptosis pathway. J Mol Cell Cardiol. 2015;82:153–166. doi:10.1016/j.yjmcc.2015.03.005
101. Zheng Y, Ren W, Zhang L, Zhang Y, Liu D, Liu Y. A review of the pharmacological action of astragalus polysaccharide. Front Pharmacol. 2020;11:349. doi:10.3389/fphar.2020.00349
102. Wang H, Zhou XM, Wu LY, et al. Aucubin alleviates oxidative stress and inflammation via Nrf2-mediated signaling activity in experimental traumatic brain injury. J Neuroinflammation. 2020;17(1):188. doi:10.1186/s12974-020-01863-9
103. Lei W, Chen MH, Huang ZF, et al. Salidroside protects pulmonary artery endothelial cells against hypoxia-induced apoptosis via the AhR/NF-κB and Nrf2/HO-1 pathways. Phytomedicine. 2024;128:155376. doi:10.1016/j.phymed.2024.155376
104. Ren S, Wang Y, Zhang Y, et al. Paeoniflorin alleviates AngII-induced cardiac hypertrophy in H9c2 cells by regulating oxidative stress and Nrf2 signaling pathway. Biomed Pharmacother. 2023;165:115253. doi:10.1016/j.biopha.2023.115253
105. Yang JJ, Zheng XJ, Zhao YJ, Ruan HL, Ren ZF, Luo H. Clinical observation of breviscapine injection in treatment of connective tissue diease pulmonary aterial hypertention. Chin Arch Tradit Chin Med. 2019;37:2762–2764.
106. Zhang X, Liu Q, Zhang C, et al. Puerarin prevents progression of experimental hypoxia-induced pulmonary hypertension via inhibition of autophagy. J Pharmacol Sci. 2019;141(2):97–105. doi:10.1016/j.jphs.2019.09.010
107. Yuan T, Zhang H, Chen D, et al. Puerarin protects pulmonary arteries from hypoxic injury through the BMPRII and PPARγ signaling pathways in endothelial cells. Pharmacol Rep. 2019;71(5):855–861. doi:10.1016/j.pharep.2019.05.002
108. Zhang M, Wu Y, Wang M, Wang Y, Tausif R, Yang Y. Genistein rescues hypoxia-induced pulmonary arterial hypertension through estrogen receptor and β-adrenoceptor signaling. J Nutr Biochem. 2018;58:110–118. doi:10.1016/j.jnutbio.2018.04.016
109. Zhang Z, Zhang L, Sun C, et al. Baicalin attenuates monocrotaline-induced pulmonary hypertension through bone morphogenetic protein signaling pathway. Oncotarget. 2017;8(38):63430–63441. doi:10.18632/oncotarget.18825
110. Yan G, Wang J, Yi T, et al. Baicalin prevents pulmonary arterial remodeling in vivo via the AKT/ERK/NF-κB signaling pathways. Pulm Circ. 2019;9(4):2045894019878599. doi:10.1177/2045894019878599
111. He Y, Cao X, Liu X, et al. Quercetin reverses experimental pulmonary arterial hypertension by modulating the TrkA pathway. Exp Cell Res. 2015;339(1):122–134. doi:10.1016/j.yexcr.2015.10.013
112. Zhang Y, Cui Y, Deng W, et al. Isoquercitrin protects against pulmonary hypertension via inhibiting PASMCs proliferation. Clin Exp Pharmacol Physiol. 2017;44(3):362–370. doi:10.1111/1440-1681.12705
113. Li Q, Wang J, Zhu X, et al. Dihydromyricetin prevents monocrotaline-induced pulmonary arterial hypertension in rats. Biomed Pharmacother. 2017;96:825–833. doi:10.1016/j.biopha.2017.10.007
114. Chen Y, Lu W, Yang K, et al. Tetramethylpyrazine: a promising drug for the treatment of pulmonary hypertension. Br J Pharmacol. 2020;177(12):2743–2764. doi:10.1111/bph.15000
115. Huang H, Kong L, Luan S, Qi C, Wu F. Ligustrazine suppresses platelet-derived growth factor-BB-induced pulmonary artery smooth muscle cell proliferation and inflammation by regulating the PI3K/AKT signaling pathway. Am J Chin Med. 2021;49(2):437–459. doi:10.1142/S0192415X21500208
116. Wang X, Yang Y, Yang D, et al. Tetrandrine prevents monocrotaline-induced pulmonary arterial hypertension in rats through regulation of the protein expression of inducible nitric oxide synthase and cyclic guanosine monophosphate-dependent protein kinase type 1. J Vasc Surg. 2016;64(5):1468–1477. doi:10.1016/j.jvs.2015.09.016
117. King VF, Garcia ML, Himmel D, et al. Interaction of tetrandrine with slowly inactivating calcium channels. Characterization of calcium channel modulation by an alkaloid of Chinese medicinal herb origin. J Biol Chem. 1988;263(5):2238–2244. doi:10.1016/S0021-9258(18)69196-3
118. Luo J, Gu Y, Liu P, et al. Berberine attenuates pulmonary arterial hypertension via protein phosphatase 2A signaling pathway both in vivo and in vitro. J Cell Physiol. 2018;233(12):9750–9762. doi:10.1002/jcp.26940
119. Chen M, Shen H, Zhu L, et al. Berberine attenuates hypoxia-induced pulmonary arterial hypertension via bone morphogenetic protein and transforming growth factor-β signaling. J Cell Physiol. 2019;234(10):17482–17493. doi:10.1002/jcp.28370
120. Yang JM, Zhou R, Zhang M, Tan HR, Yu JQ. Betaine attenuates monocrotaline-induced pulmonary arterial hypertension in rats via inhibiting inflammatory response. Molecules. 2018;23(6):1274.
121. Chang Z, Zhang P, Zhang M, et al. Aloperine suppresses human pulmonary vascular smooth muscle cell proliferation via inhibiting inflammatory response. Chin J Physiol. 2019;62(4):157–165. doi:10.4103/CJP.CJP_27_19
122. Wu F, Yao W, Yang J, et al. Protective effects of aloperin on monocroline-induced pulmonary hypertension via regulation of Rho A/Rho kinsase pathway in rats. Biomed Pharmacother. 2017;95:1161–1168. doi:10.1016/j.biopha.2017.08.126
123. Sun Y, Lu M, Sun T, Wang H. Astragaloside IV attenuates inflammatory response mediated by NLRP-3/calpain-1 is involved in the development of pulmonary hypertension. J Cell Mol Med. 2021;25(1):586–590. doi:10.1111/jcmm.15671
124. Yao J, Fang X, Zhang C, et al. Astragaloside IV attenuates hypoxia‑induced pulmonary vascular remodeling via the Notch signaling pathway. Mol Med Rep. 2021;23(1):1.
125. Kosanovic D, Tian X, Pak O, et al. Rhodiola: an ordinary plant or a promising future therapy for pulmonary hypertension? a brief review. Pulm Circ. 2013;3(3):499–506. doi:10.1086/674303
126. Chen M, Cai H, Yu C, et al. Salidroside exerts protective effects against chronic hypoxia-induced pulmonary arterial hypertension via AMPKα1-dependent pathways. Am J Transl Res. 2016;8(1):12–27.
127. Wang XB, Wang W, Zhu XC, et al. The potential of asiaticoside for TGF-β1/Smad signaling inhibition in prevention and progression of hypoxia-induced pulmonary hypertension. Life Sci. 2015;137:56–64. doi:10.1016/j.lfs.2015.07.016
128. Wang J, Wong YK, Liao F. What has traditional Chinese medicine delivered for modern medicine? Expert Rev Mol Med. 2018;20:e4. doi:10.1017/erm.2018.3
129. Xu Y, Lin L, Tang L, et al. Notoginsenoside R1 attenuates hypoxia and hypercapnia-induced vasoconstriction in isolated rat pulmonary arterial rings by reducing the expression of ERK. Am J Chin Med. 2014;42(4):799–816. doi:10.1142/S0192415X14500517
130. Zhao S, Zheng MX, Chen HE, Wu CY, Wang WT. Effect of panax notoginseng saponins injection on the p38MAPK pathway in lung tissue in a rat model of hypoxic pulmonary hypertension. Chin J Integr Med. 2015;21(2):147–151. doi:10.1007/s11655-014-1790-2
131. Chen X, He Y, Yu Z, et al. Polydatin glycosides improve monocrotaline-induced pulmonary hypertension injury by inhibiting endothelial-to-mesenchymal transition. Front Pharmacol. 2022;13:862017. doi:10.3389/fphar.2022.862017
132. Xiang Y, Cai C, Wu Y, et al. Icariin attenuates monocrotaline-induced pulmonary arterial hypertension via the inhibition of TGF-β1/Smads pathway in rats. Evid Based Complement Alternat Med. 2020;2020:9238428. doi:10.1155/2020/9238428
133. Shi W, Zhai C, Feng W, et al. Resveratrol inhibits monocrotaline-induced pulmonary arterial remodeling by suppression of SphK1-mediated NF-κB activation. Life Sci. 2018;210:140–149. doi:10.1016/j.lfs.2018.08.071
134. Chen B, Xue J, Meng X, Slutzky JL, Calvert AE, Chicoine LG. Resveratrol prevents hypoxia-induced arginase II expression and proliferation of human pulmonary artery smooth muscle cells via Akt-dependent signaling. Am J Physiol Lung Cell Mol Physiol. 2014;307(4):L317–325. doi:10.1152/ajplung.00285.2013
135. Xu D, Li Y, Zhang B, et al. Resveratrol alleviate hypoxic pulmonary hypertension via anti-inflammation and anti-oxidant pathways in rats. Int J Med Sci. 2016;13(12):942–954. doi:10.7150/ijms.16810
136. Li C, Peng G, Long J, Xiao P, Zeng X, Yang H. Protective effects of resveratrol and SR1001 on hypoxia-induced pulmonary hypertension in rats. Clin Exp Hypertens. 2020;42(6):519–526. doi:10.1080/10641963.2020.1714643
137. Zhang N, Dong M, Luo Y, Zhao F, Li Y. Danshensu prevents hypoxic pulmonary hypertension in rats by inhibiting the proliferation of pulmonary artery smooth muscle cells via TGF-β-smad3-associated pathway. Eur J Pharmacol. 2018;820:1–7. doi:10.1016/j.ejphar.2017.12.010
138. Li T, Luo XJ, Wang EL, et al. Magnesium lithospermate B prevents phenotypic transformation of pulmonary arteries in rats with hypoxic pulmonary hypertension through suppression of NADPH oxidase. Eur J Pharmacol. 2019;847:32–41. doi:10.1016/j.ejphar.2019.01.020
139. Wang Y, Duo D, Yan Y, He R, Wu X. Magnesium lithospermate B ameliorates hypobaric hypoxia-induced pulmonary arterial hypertension by inhibiting endothelial-to-mesenchymal transition and its potential targets. Biomed Pharmacother. 2020;130:110560. doi:10.1016/j.biopha.2020.110560
140. Zhao J, Yang M, Wu X, et al. Effects of paclitaxel intervention on pulmonary vascular remodeling in rats with pulmonary hypertension. Exp Ther Med. 2019;17(2):1163–1170. doi:10.3892/etm.2018.7045
141. Feng W, Wang J, Yan X, et al. Paclitaxel alleviates monocrotaline-induced pulmonary arterial hypertension via inhibition of FoxO1-mediated autophagy. Naunyn Schmiedebergs Arch Pharmacol. 2019;392(5):605–613. doi:10.1007/s00210-019-01615-4
142. Yu H, Liu J, Dong Y, et al. Anti-hypoxic effect of dihydroartemisinin on pulmonary artery endothelial cells. Biochem Biophys Res Commun. 2018;506(4):840–846. doi:10.1016/j.bbrc.2018.10.176
143. Kurosawa R, Satoh K, Nakata T, et al. Identification of celastrol as a novel therapeutic agent for pulmonary arterial hypertension and right ventricular failure through suppression of Bsg (Basigin)/CyPA (Cyclophilin A). Arterioscler Thromb Vasc Biol. 2021;41(3):1205–1217. doi:10.1161/ATVBAHA.120.315731
144. Yang PS, Kim DH, Lee YJ, et al. Glycyrrhizin, inhibitor of high mobility group box-1, attenuates monocrotaline-induced pulmonary hypertension and vascular remodeling in rats. Respir Res. 2014;15:148. doi:10.1186/s12931-014-0148-4
145. Yang L, Zheng JX, Shi XD, Qian H, Sun JL. Sodium tanshinone IIA sulfonate inhibits the proliferation of hypoxia-induced PASMC of rat and its potential mechanism. J Jiangsu Univ. 2018;28(3):190–194.
146. Bao YR, Chen JW, Jiang Y, et al. Sodium Tanshinone II Sulfonate A Ameliorates Hypoxia-Induced Pulmonary Hypertension. Front Pharmacol. 2020;11:687. doi:10.3389/fphar.2020.00687
147. Jiang Q, Lu W, Yang K, et al. Sodium tanshinone IIA sulfonate inhibits hypoxia-induced enhancement of SOCE in pulmonary arterial smooth muscle cells via the PKG-PPAR-γ signaling axis. Am J Physiol Cell Physiol. 2016;311(1):C136–149. doi:10.1152/ajpcell.00252.2015
148. Zhu N, Xiang Y, Zhao X, et al. Thymoquinone suppresses platelet-derived growth factor-BB-induced vascular smooth muscle cell proliferation, migration and neointimal formation. J Cell Mol Med. 2019;23(12):8482–8492. doi:10.1111/jcmm.14738
149. Ji XY, Lei CJ, Kong S, et al. Hydroxy-Safflower Yellow A Mitigates Vascular Remodeling in Rat Pulmonary Arterial Hypertension. Drug Des Devel Ther. 2024;18:475–491. doi:10.2147/DDDT.S439686
150. Slika H, Mansour H, Wehbe N, et al. Therapeutic potential of flavonoids in cancer: ROS-mediated mechanisms. Biomed Pharmacother. 2022;146:112442. doi:10.1016/j.biopha.2021.112442
151. Wei Q, Zhang YH. Flavonoids with anti-angiogenesis function in cancer. Molecules. 2024;29(7):1570.
152. Alasvand M, Assadollahi V, Ambra R, Hedayati E, Kooti W, Peluso I. Antiangiogenic effect of alkaloids. Oxid Med Cell Longev. 2019;2019:9475908. doi:10.1155/2019/9475908
153. Mondal A, Gandhi A, Fimognari C, Atanasov AG, Bishayee A. Alkaloids for cancer prevention and therapy: current progress and future perspectives. Eur J Pharmacol. 2019;858:172472. doi:10.1016/j.ejphar.2019.172472
154. Niu Y, Zhang J, Shi D, Zang W, Niu J. Glycosides as potential medicinal components for ulcerative colitis: a review. Molecules. 2023;28(13):5210. doi:10.3390/molecules28135210
155. Jiang S, Jiao G, Chen Y, Han M, Wang X, Liu W. Astragaloside IV attenuates chronic intermittent hypoxia-induced myocardial injury by modulating Ca(2+) homeostasis. Cell Biochem Funct. 2020;38(6):710–720. doi:10.1002/cbf.3538
156. Potočnjak I, Marinić J, Batičić L, Šimić L, Broznić D, Domitrović R. Aucubin administered by either oral or parenteral route protects against cisplatin-induced acute kidney injury in mice. Food Chem Toxicol. 2020;142:111472. doi:10.1016/j.fct.2020.111472
157. Huang TL, Yang SH, Chen YR, Liao JY, Tang Y, Yang KC. The therapeutic effect of aucubin-supplemented hyaluronic acid on interleukin-1beta-stimulated human articular chondrocytes. Phytomedicine. 2019;53:1–8. doi:10.1016/j.phymed.2018.09.233
158. Xue HY, Gao GZ, Lin QY, Jin LJ, Xu YP. Protective effects of aucubin on H2O2-induced apoptosis in PC12 cells. Phytother Res. 2012;26(3):369–374. doi:10.1002/ptr.3562
159. Pacheco-Ordaz R, Wall-Medrano A, Goñi MG, Ramos-Clamont-Montfort G, Ayala-Zavala JF, González-Aguilar GA. Effect of phenolic compounds on the growth of selected probiotic and pathogenic bacteria. Lett Appl Microbiol. 2018;66(1):25–31. doi:10.1111/lam.12814
160. Abotaleb M, Liskova A, Kubatka P, Büsselberg D. Therapeutic potential of plant phenolic acids in the treatment of cancer. Biomolecules. 2020;10(2):221. doi:10.3390/biom10020221
161. Cardoso RR, Neto RO, Dos Santos D’Almeida CT, et al. Kombuchas from green and black teas have different phenolic profile, which impacts their antioxidant capacities, antibacterial and antiproliferative activities. Food Res Int. 2020;128:108782. doi:10.1016/j.foodres.2019.108782
162. Chudzińska M, Rogowicz D, Wołowiec Ł, et al. Resveratrol and cardiovascular system-the unfulfilled hopes. Ir J Med Sci. 2021;190(3):981–986. doi:10.1007/s11845-020-02441-x
163. Bernal-Ramírez J, Silva-Platas C, Jerjes-Sánchez C, et al. Resveratrol prevents right ventricle dysfunction, calcium mishandling, and energetic failure via SIRT3 stimulation in pulmonary arterial hypertension. Oxid Med Cell Longev. 2021;2021:9912434. doi:10.1155/2021/9912434
164. Wang Q, Zhao X, Jiang Y, Jin B, Wang L. Functions of representative terpenoids and their biosynthesis mechanisms in medicinal plants. Biomolecules. 2023;13(12):1725. doi:10.3390/biom13121725
165. Leopardi M, Houbballah R, Becquemin JP. Effectiveness of Zilver PTX eluting stent in TASC C/D lesions and restenosis. J Cardiovasc Surg. 2014;55(2):229–234.
166. Ying HZ, Yu CH, Chen HK, et al. Quinonoids: therapeutic potential for lung cancer treatment. Biomed Res Int. 2020;2020:2460565. doi:10.1155/2020/2460565
167. Jiang Y, Song S, Liu J, et al. Epigenetic regulation of programmed cell death in hypoxia-induced pulmonary arterial hypertension. Front Immunol. 2023;14:1206452. doi:10.3389/fimmu.2023.1206452
168. Geng Y, Hu Y, Zhang F, Tuo Y, Ge R, Bai Z. Mitochondria in hypoxic pulmonary hypertension, roles and the potential targets. Front Physiol. 2023;14:1239643. doi:10.3389/fphys.2023.1239643
169. Wan JJ, Yi J, Wang FY, Zhang C, Dai AG. Expression and regulation of HIF-1a in hypoxic pulmonary hypertension: focus on pathological mechanism and pharmacological treatment. Int J Med Sci. 2024;21(1):45–60. doi:10.7150/ijms.88216
170. Singh A, Kukreti R, Saso L, Kukreti S. Oxidative Stress: a key modulator in neurodegenerative diseases. Molecules. 2019;24(8):1583. doi:10.3390/molecules24081583
171. Okoye CN, Koren SA, Wojtovich AP. Mitochondrial complex I ROS production and redox signaling in hypoxia. Redox Biol. 2023;67:102926. doi:10.1016/j.redox.2023.102926
172. Mishra N, Jiang C, Chen L, Paul A, Chatterjee A, Shen G. Achieving abiotic stress tolerance in plants through antioxidative defense mechanisms. Front Plant Sci. 2023;14:1110622. doi:10.3389/fpls.2023.1110622
173. Grüning NM, Ralser M. Monogenic disorders of ROS production and the primary anti-oxidative defense. Biomolecules. 2024;14(2):206. doi:10.3390/biom14020206
174. Richalet JP, Hermand E, Lhuissier FJ. Cardiovascular physiology and pathophysiology at high altitude. Nat Rev Cardiol. 2024;21(2):75–88. doi:10.1038/s41569-023-00924-9
175. Newton K, Strasser A, Kayagaki N, Dixit VM. Cell death. Cell. 2024;187(2):235–256. doi:10.1016/j.cell.2023.11.044
176. Liu S, Pi J, Zhang Q. Signal amplification in the KEAP1-NRF2-ARE antioxidant response pathway. Redox Biol. 2022;54:102389. doi:10.1016/j.redox.2022.102389
177. Jomova K, Raptova R, Alomar SY, et al. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: chronic diseases and aging. Arch Toxicol. 2023;97(10):2499–2574. doi:10.1007/s00204-023-03562-9
178. Liu Y, Kong H, Cai H, Chen G, Chen H, Ruan W. Progression of the PI3K/Akt signaling pathway in chronic obstructive pulmonary disease. Front Pharmacol. 2023;14:1238782. doi:10.3389/fphar.2023.1238782
179. Sun X, Chen L, He Z. PI3K/Akt-Nrf2 and Anti-inflammation effect of macrolides in chronic obstructive pulmonary disease. Curr Drug Metab. 2019;20(4):301–304. doi:10.2174/1389200220666190227224748
180. Andrabi SM, Sharma NS, Karan A, et al. Nitric Oxide: physiological Functions, Delivery, and Biomedical Applications. Adv Sci. 2023;10(30):e2303259. doi:10.1002/advs.202303259
181. Kuriyama S, Morio Y, Toba M, et al. Genistein attenuates hypoxic pulmonary hypertension via enhanced nitric oxide signaling and the erythropoietin system. Am J Physiol Lung Cell Mol Physiol. 2014;306(11):L996–l1005. doi:10.1152/ajplung.00276.2013
182. Park JI. MAPK-ERK Pathway. Int J Mol Sci. 2023;24(11):9666.
183. Liu Y, Tang BL, Lu ML, Wang HX. Astragaloside IV improves pulmonary arterial hypertension by increasing the expression of CCN1 and activating the ERK1/2 pathway. J Cell Mol Med. 2023;27(5):622–633. doi:10.1111/jcmm.17681
184. Adinolfi S, Patinen T, Jawahar Deen A, et al. The KEAP1-NRF2 pathway: targets for therapy and role in cancer. Redox Biol. 2023;63:102726. doi:10.1016/j.redox.2023.102726
185. Kang Y, Zhang G, Huang EC, et al. Sulforaphane prevents right ventricular injury and reduces pulmonary vascular remodeling in pulmonary arterial hypertension. Am J Physiol Heart Circ Physiol. 2020;318(4):H853–h866. doi:10.1152/ajpheart.00321.2019
186. Cipriano A, Viviano M, Feoli A, et al. NADPH oxidases: from molecular mechanisms to current inhibitors. J Med Chem. 2023;66(17):11632–11655. doi:10.1021/acs.jmedchem.3c00770
187. Cui L, Zeng Z, Wang X, et al. Deciphering the mechanism of Wogonin, a natural flavonoid, on the proliferation of pulmonary arterial smooth muscle cells by integrating network pharmacology and in vitro validation. Curr Issues Mol Biol. 2023;45(1):555–570. doi:10.3390/cimb45010037
188. Yu H, Lin L, Zhang Z, Zhang H, Hu H. Targeting NF-κB pathway for the therapy of diseases: mechanism and clinical study. Signal Transduct Target Ther. 2020;5(1):209. doi:10.1038/s41392-020-00312-6
189. Guo Q, Jin Y, Chen X, et al. NF-κB in biology and targeted therapy: new insights and translational implications. Signal Transduct Target Ther. 2024;9(1):53. doi:10.1038/s41392-024-01757-9
190. Nie X, Shen C, Tan J, et al. Andrographolide attenuates established pulmonary hypertension via rescue of vascular remodeling. Biomolecules. 2021;11(12):1801. doi:10.3390/biom11121801
191. Lv C, Huang Y, Yan R, Gao Y. Vascular endothelial growth factor induces the migration of human airway smooth muscle cells by activating the RhoA/ROCK pathway. BMC Pulm Med. 2023;23(1):505. doi:10.1186/s12890-023-02803-y
192. Wang Y, Duo D, Yan Y, et al. Extract of Salvia przewalskii repair tissue damage in chronic hypoxia maybe through the RhoA-ROCK signalling pathway. Biol Pharm Bull. 2020;43(3):432–439. doi:10.1248/bpb.b19-00775
193. Flores-Romero H, Dadsena S, García-Sáez AJ. Mitochondrial pores at the crossroad between cell death and inflammatory signaling. Mol Cell. 2023;83(6):843–856. doi:10.1016/j.molcel.2023.02.021
194. Chen C, Chen C, Wang Z, et al. Puerarin induces mitochondria-dependent apoptosis in hypoxic human pulmonary arterial smooth muscle cells. PLoS One. 2012;7(3):e34181. doi:10.1371/journal.pone.0034181
195. Dignam JP, Scott TE, Kemp-Harper BK, Hobbs AJ. Animal models of pulmonary hypertension: getting to the heart of the problem. Br J Pharmacol. 2022;179(5):811–837. doi:10.1111/bph.15444
196. Virgincar RS, Nouls JC, Wang Z, et al. Quantitative (129)Xe MRI detects early impairment of gas-exchange in a rat model of pulmonary hypertension. Sci Rep. 2020;10(1):7385. doi:10.1038/s41598-020-64361-1
197. Feng Y, Xu S, Guo H, et al. Vanin-1-activated Chemiluminescent Probe: help to early diagnosis of acute kidney injury with high signal-to-noise ratio through urinalysis. Anal Chem. 2023;95(39):14754–14761. doi:10.1021/acs.analchem.3c02875
198. Wang H, Zhang X, Li P, et al. Prediction of early atherosclerotic plaques using a sequence-activated fluorescence probe for the simultaneous detection of γ-Glutamyl transpeptidase and hypobromous acid. Angew Chem Int Ed Engl. 2024;63(1):e202315861. doi:10.1002/anie.202315861
199. Liu Y, Wang S, Jin G, et al. Network pharmacology-based study on the mechanism of ShenKang injection in diabetic kidney disease through Keap1/Nrf2/Ho-1 signaling pathway. Phytomedicine. 2023;118:154915. doi:10.1016/j.phymed.2023.154915
200. Sun Y, Wu L, Zhong Y, et al. Single-cell landscape of the ecosystem in early-relapse hepatocellular carcinoma. Cell. 2021;184(2):404–421.e416. doi:10.1016/j.cell.2020.11.041