ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

The epidemiology and pathogenesis of pulmonary arterial hypertension (Group 1)

The epidemiology and pathogenesis of pulmonary arterial hypertension (Group 1)
Literature review current through: Jan 2024.
This topic last updated: Nov 28, 2022.

INTRODUCTION — Pulmonary hypertension (PH) is a disease characterized by elevated pulmonary artery pressure (mean pulmonary artery pressure ≥20 mmHg at rest). The World Health Organization (WHO) classifies patients with PH into five groups (table 1) based upon etiology [1]:

Group 1 – Pulmonary arterial hypertension (PAH)

Group 2 – PH due to left heart disease

Group 3 – PH due to chronic lung disease and/or hypoxemia

Group 4 – PH due to pulmonary artery obstructions

Group 5 – PH due to unclear multifactorial mechanisms

The term PAH is used to describe those included in group 1, while the term PH is used when describing all five groups. The pathogenesis of PAH is described in this review. The classification of PH and the pathogenesis of groups 2, 3, and 4 PH are provided separately. (See "Pathophysiology of heart failure with preserved ejection fraction" and "Pulmonary hypertension due to lung disease and/or hypoxemia (group 3 pulmonary hypertension): Epidemiology, pathogenesis, and diagnostic evaluation in adults", section on 'Pathogenesis' and "Epidemiology, pathogenesis, clinical manifestations and diagnosis of chronic thromboembolic pulmonary hypertension", section on 'Pathogenesis' and "Pathophysiology of heart failure: Neurohumoral adaptations", section on 'Neurohumoral adaptations' and "Pathophysiology of heart failure with reduced ejection fraction: Hemodynamic alterations and remodeling" and "Clinical features and diagnosis of pulmonary hypertension of unclear etiology in adults", section on 'Postdiagnostic testing and classification'.)

EPIDEMIOLOGY — Pulmonary hypertension (PH) affects individuals of all age, race, and gender. However, because of the broad classification and multiple etiologies (table 1), obtaining accurate estimates of the prevalence of PH and its different forms, including PAH, has been challenging. Nonetheless, idiopathic PAH (IPAH) and heritable PAH (HPAH) are rare in the general population and estimated to be 5 to 15 cases per one million adults [2-4].

Although the prevalence of PAH is unknown in North America, several European registries have reported rates of 5 to 52 per million [5,6].

While schistosomiasis appears to be the most common cause of PAH worldwide, registry data in regions of the world without endemic schistosomiasis, report that over half of cases of PAH are idiopathic (IPAH) and up to 10 percent are heritable (HPAH) [2,5,6]. HPAH may be underdiagnosed. In one study, among five apparently unrelated families, 18 (out of 400) individuals had hereditary bone morphogenetic protein receptor 2 (BMPR2) mutations, 12 of whom were initially classified as IPAH [7]. Among the other associated etiologies of PAH, connective tissue disease and congenital heart disease appear to be the most common, the latter being the predominant cause of PAH in China [2,5,8].

Compared to other groups of PH, IPAH affects younger adults [9]. In contrast, in older populations, group 1 PAH is relatively uncommon. In one series, only 15 percent of 246 adults with PH older than 65 years had PAH, most frequently in association with connective tissue disease [10]. (See "Clinical features and diagnosis of pulmonary hypertension of unclear etiology in adults", section on 'Postdiagnostic testing and classification'.)

Although men and women can have PAH, women are more likely to be affected (female:male ratio ranges from 1.7 to 4.8:1.0) as well as symptomatic from PAH [4,9].

While in the past studies suggested that PAH affected young women in their thirties, PAH is now a disease that affects men and women most commonly presenting in midlife. (See "Clinical features and diagnosis of pulmonary hypertension of unclear etiology in adults", section on 'Clinical manifestations'.)

One United States registry reported a PAH predominance among White individuals (73 percent of registered cases), but PAH was also seen in African Americans (12 percent), Hispanic Americans (9 percent), and Asian Americans (3 percent) [9].

PAH-related hospitalizations and death may be decreasing. One national database reported that between 2007 and 2011, population-based death rates decreased from 4.6 to 1.7 per million and that the proportion of hospitalizations for PAH reduced from 79 to 38 per 100,000 hospitalizations [11]. Similarly, an analysis of the national inpatient sample database reported an over 50 percent reduction in the number of PAH-related hospitalizations between 2001 and 2012; however, there was no change in in-hospital mortality during that same time period (7.8 versus 6.3 percent) [12].

The epidemiology of other causes of PAH (table 1) are discussed in the followings sections:

Systemic sclerosis (see "Pulmonary arterial hypertension in systemic sclerosis (scleroderma): Definition, risk factors, and screening", section on 'Epidemiology')

Congenital heart disease (see "Pulmonary hypertension with congenital heart disease: Clinical manifestations and diagnosis", section on 'Introduction')

Human immunodeficiency virus (see "Pulmonary arterial hypertension associated with human immunodeficiency virus", section on 'Prevalence')

Portopulmonary hypertension (see "Portopulmonary hypertension", section on 'Epidemiology')

Schistosomiasis (see "Schistosomiasis: Epidemiology and clinical manifestations", section on 'Pulmonary complications')

Pulmonary veno-occlusive disease (see "Epidemiology, pathogenesis, clinical evaluation, and diagnosis of pulmonary veno-occlusive disease/pulmonary capillary hemangiomatosis in adults", section on 'Epidemiology')

Persistent pulmonary hypertension of the newborn (see "Persistent pulmonary hypertension of the newborn (PPHN): Clinical features and diagnosis", section on 'Epidemiology')

PATHOGENETIC MECHANISMS — Described in this section is the general physiologic principles behind the development of pulmonary hypertension (see 'General physiologic mechanisms' below) as well as the pathogenetic mechanisms specific to the individual forms of PAH. (See 'Idiopathic and heritable' below and 'Drugs and toxins' below and 'Other conditions' below and 'Pulmonary veno-occlusive disease/pulmonary capillary hemangiomatosis' below and 'Persistent pulmonary hypertension of the newborn' below.)

General physiologic mechanisms — Based upon a variation of Ohm’s Law (ie, change in pressure = flow x resistance), the mean pulmonary arterial pressure is determined by the following formula:

mean pulmonary artery pressure = (right ventricle cardiac output x pulmonary vascular resistance) + pulmonary alveolar occlusion pressure

The primary cause of significant pulmonary hypertension (PH) is almost always increased pulmonary vascular resistance (PVR). Increased flow alone (ie, right ventricle output) does not usually cause significant PH because the pulmonary vascular bed vasodilates and recruits vessels in response to increased flow. Similarly, increased pulmonary venous pressure (represented by the alveolar occlusion pressure) alone does not usually cause significant PH. However, a chronic increase of either flow and/or pulmonary venous pressure can increase pulmonary vascular resistance.

All variables can be altered by numerous medical conditions:

Increased PVR may be due to conditions associated with occlusive vasculopathy (ie, remodeling and altered vascular tone) of the small pulmonary arteries and arterioles (eg, conditions associated with PAH), conditions that decrease the cross sectional area of the pulmonary vascular bed (eg, pulmonary emboli, interstitial lung disease), or conditions that induce hypoxic vasoconstriction (eg, hypoventilation syndromes and parenchymal lung disease).

Increased flow through the pulmonary vasculature may be due to congenital heart defects with left-to-right shunt (eg, atrial septal defects, ventricular septal defects, patent ductus arteriosus), liver cirrhosis, anemia, arteriovenous malformations, or arteriovenous fistulas (dialysis). (See "Pulmonary hypertension in patients with end-stage kidney disease".)

Increased pulmonary venous pressure may be due to mitral valve disease, left ventricular systolic or diastolic dysfunction, constrictive pericarditis, restrictive cardiomyopathy or pulmonary venous obstruction (eg, pulmonary veno-occlusive disease).

Idiopathic and heritable — Patients with idiopathic PAH (IPAH) are clinically indistinguishable from patients with heritable PAH (HPAH). Although they share common pathologic and pathogenetic features, heritable PAH exists when heritable genetic defects known to cause PAH can be identified (most often bone morphogenetic protein receptor type II [BMPR2] mutations) while IPAH is sporadic.

Pathology — PAH is a proliferative vasculopathy, characterized by vasoconstriction, cell proliferation, fibrosis, and microthrombosis. Pathologic findings include hyperplasia and hypertrophy of all three layers of the vascular wall (intima, media, adventitia) in pulmonary arteries <50 microns (ie, localizes to the small pulmonary muscular arterioles). In addition, fibrosis and in situ thrombi of the small pulmonary arteries and arterioles (plexiform lesions) can be seen [13,14]. The pathologic appearance of the small pulmonary arteries and arterioles is qualitatively similar in all patients with group 1 PAH.

Pathologic classification of the pulmonary vascular abnormalities (Heath and Edwards classification) was first done using patients with congenital heart disease [15], but are now applied to all patients with PAH and represent increasing severity of PAH (picture 1):

Grade I and II changes are characterized by muscularization of the small pulmonary arterioles, followed by medial hypertrophy and intimal hyperplasia.

Grade III abnormalities are characterized by collagenous replacement of intimal cells, leading to an "onion-skin" appearance.

Grade IV through VI abnormalities overlap, include plexiform lesions, and can be considered one stage [16-18].

Historically, it has been felt that Grade I, II, and III lesions are reversible while Grade IV through VI lesions are not. However, reversibility is poorly understood since no primary data exists as to the reversibility of these lesions in patients on PAH targeted therapy. However, many patients with a positive response to therapy do exhibit a drop in their pulmonary vascular resistance suggesting some degree of reversibility of the occlusive vasculopathy. Grade IV through VI lesions are associated with a poor outcome in patients that undergo surgery for congenital shunts [19,20].

Genetic mutations — Patients with IPAH may have an underlying genetic predisposition to pulmonary vascular disease, while patients with HPAH have PAH due to an inheritable genetic mutation. The most common heritable genetic mutation is BMPR2, which is transmitted as an autosomal dominant trait with incomplete penetrance and variable expressivity [21].

Mutations in the following genes have been variably associated with familial, idiopathic, or hereditary hemorrhagic telangiectasia (HHT)-associated PAH (table 2):

Bone morphogenetic protein receptor type II – BMPR2 is a member of the transforming growth factor (TGF)-beta family. Several lines of evidence support a role for abnormal BMPR2 in PAH:

Human – Up to 25 percent of patients with IPAH have abnormal BMPR2 structure or function [22-27], while up to 80 percent of hereditary PAH is due to mutations in BMPR2 [28].

Animal – Transgenic mice with smooth muscle specific deletion of BMPR2 have pulmonary hypertension [29].

Cellular – The BMPR2 pathway induces apoptosis, which when mutated, may permit excess endothelial cell proliferation in response to a variety of injuries (figure 1) [30].

Activin-like kinase type 1 receptor (ALK1; ACVRL1; also known as serine/threonine-protein kinase receptor R3) – The ALK1 receptor is also a member of the TGF-beta family. Mutations have been identified in some patients with HHT and PAH [31-33].

5-hydroxytryptamine (serotonin) transporter (5HTT) – Increased 5HTT activity may induce pulmonary artery smooth muscle hypertrophy. The L-allelic variant of the 5HTT gene promoter is associated with increased activity of 5HTT and is found in a greater percentage of patients with idiopathic PAH compared to controls [34,35].

Endoglin (ENG) – ENG is a protein involved in vasculogenesis. Mutations of the ENG gene have been associated with HHT and idiopathic PAH [36,37].

Mothers against decapentaplegic homologue 9 (SMAD9) – SMAD9 is an important intracellular signaling molecule downstream of the TGF-beta receptor. Rare mutations of SMAD9 have been found in patients with idiopathic PAH [38].

Caveolin 1 (CAV1) – CAV1 is a scaffolding plasma membrane-associated protein involved in cell cycle progression, mutations of which have been described in familial and idiopathic PAH [39].

Potassium channel subfamily K member 3 (KCNK3) – KCNK3 encodes a potassium channel, which can be remedied by pharmacologic manipulation. Mutations in KCNK3 were identified in familial and idiopathic PAH [40,41].

Mutations in the eukaryotic translation initiation factor 2-alpha kinase (EIF2AK4) gene are predominantly found in PAH associated with pulmonary veno-occlusive disease (PVOD). They have also been rarely found in patients with IPAH/HPAH [42,43]. However, one study reported that although EIF2AK4 mutations were found in 1 percent of patients clinically classified as having IPAH/HPAH, their clinical features and pathology suggested they were likely misclassified and probably had PVOD [42]; for example these patients tended to be younger (<50 years), had a low diffusing capacity (<50 percent predicted), and had a worse prognosis (ie, clinical features consistent with PVOD). (See "Epidemiology, pathogenesis, clinical evaluation, and diagnosis of pulmonary veno-occlusive disease/pulmonary capillary hemangiomatosis in adults", section on 'Genetic factors'.)

Vascular mediators — In addition, to a genetic cause or predisposition, superimposed modifying factors activate or perpetuate the disease, thereby explaining the progressive nature of PAH.

Modifying factors involved in the pathogenesis of PAH include one or more of the following [13,44-56]:

Increased endothelin levels (endothelin is a vasoconstrictor and mitogen)

Decreased nitric oxide levels (nitric oxide is a vasodilator and is antiproliferative)

Decreased prostacyclin levels (prostacyclin is a vasodilator, is antiproliferative, and inhibits platelet function)

Studies implicating these molecules in the pathogenesis of PH have resulted in the development of drugs that augment prostanoid-mediated pathways, block endothelin receptors, or augment NO signaling by inhibiting breakdown of cGMP (eg, phosphodiesterase inhibitors), by directly activating guanylate cyclase(eg, riociguat), or by NO replacement (eg, inhaled NO). (See "Treatment of pulmonary arterial hypertension (group 1) in adults: Pulmonary hypertension-specific therapy".)

Potassium channel dysfunction — Some data suggest that IPAH may result from functional impairment of the voltage-gated potassium channel (Kv) in pulmonary artery smooth muscle cells [57]. Such impairment could lead sequentially to a change in resting membrane potential, elevation of the intracytoplasmic free calcium concentration, and an increase in pulmonary vascular tone [58]. Kv dysfunction may also play an important role in the development of PAH due to anorectic agents (eg, fenfluramine, dexfenfluramine, and aminorex) or PH due to hypoxia [59].

Estrogen — Estrogen is considered a risk factor for the development of PAH. As an example, one preclinical study reported that estrogen inhibition prevented and treated PAH in BMPR2 mutant mice [60]. Human studies are lacking.

Drugs and toxins — The mechanism by which drugs cause pulmonary hypertension is unknown but thought to be due to altered growth factor biology (eg, serotonin, platelet derived growth factor). The following drugs are considered definite risk factors for PAH: appetite suppressants, toxic rapeseed oil, and benfluorex [1]. Drugs that are considered possible risk factors for PAH include the following: amphetamines, L-tryptophan, methamphetamines, cocaine, phenylpropanolamine, St. John's Wort, dasatinib, and interferon.

Definite risk factors include the following:

Appetite suppressants (eg, aminorex, fenfluramine, dexfenfluramine, and methamphetamines) increase the risk of developing PAH [61-66]. While the reason for this is largely unknown, altered serotonin biology may play a role. Serotonin has been shown to induce growth of pulmonary artery smooth muscle cells from patients with PAH; fenfluramine derivatives can interact directly with the serotonin transporter [67-70]. In a case control study that compared 95 patients with PAH to 335 control patients, appetite suppressants increased the risk of PAH (odds ratio [OR] 6.3, 95% CI 3-13) [62]. The risk was particularly high when the appetite suppressants were used in the preceding year or for more than three months (OR 10, 95% CI 3-30). Appetite suppressant use reported by patients with all types of PH is greater than that reported by the general population, suggesting that appetite suppressants may initiate PH in patients with underlying conditions that are associated with PH, or that obesity is associated with PH [71].

Rapeseed oil, when consumed in excess amounts, has been reported in the past to be associated with PAH, although amounts in commercially available vegetable oil are safe [72].

Benfluorex, a drug that is used in Europe for the treatment of diabetes and metabolic syndrome, shares an active metabolite with fenfluramine and has also been associated with the development of PAH with and without coexistent valvular heart disease [73-75].

Dasatinib, a tyrosine kinase inhibitor (TKI) used for the treatment of chronic myelogenous leukemia, has been reported to cause PAH [76]. TKIs are known inhibitors of platelet derived growth factor (PDGF) receptor and its downstream signaling molecules (eg, Src kinase). PDGF has a well-defined role in the pathogenesis of PAH so the association between TKIs and PAH is somewhat paradoxical and unexplained. Prior treatment with imatinib (another TKI that is less potent than dasatinib) and female gender appear to be risk factors for the development of dasatinib-induced PAH. Dasatinib-associated PAH may be partially reversible upon discontinuation of the drug. (See "Pulmonary toxicity associated with antineoplastic therapy: Molecularly targeted agents".)

Interferon (IFN) therapy has been associated with PAH. PAH has been reported in a small number of patients receiving interferon (IFN) therapy for hepatitis (IFN-alpha) and multiple sclerosis (IFN-beta) through an unknown mechanism [77-81]. IFN-associated PAH may be partially reversible upon discontinuation of the drug.

Several drugs have also been associated with the development of pulmonary veno-occlusive disease (mostly chemotherapeutic agents), which are discussed separately. (See "Epidemiology, pathogenesis, clinical evaluation, and diagnosis of pulmonary veno-occlusive disease/pulmonary capillary hemangiomatosis in adults", section on 'Drugs and toxins'.)

Possible risk factors include the following:

Chronic use of cocaine or amphetamines or use of diethylpropion either inhaled or intravenous, has also been associated with PAH [62,82-84]. The mechanism is thought to relate to the shared pharmacologic properties with fenfluramine. In one study, cocaine or amphetamine use tripled the risk of developing PAH [62].

PAH has also been associated with amphetamine use and with recreational use of the designer amphetamine analog, 4-methyl-aminorex (ie, ice, euphoria, U-4-E-uh), as well as with leflunomide, phentermine, and mazindol (used for the treatment of narcolepsy and obesity) [71,85-87]. Several drugs that have mechanisms of action that are similar to amphetamines (eg, methylphenidate and ropinirole) have no clear association with PAH. However, clinicians should be aware in case patients develop symptoms of PAH.

Other drugs that have been listed as possible agents associated with PAH include phenylpropanolamine, L-tryptophan, St. John’s wort, alkylating agents, bosutinib, direct-acting antiviral agents against hepatitis C virus, and indirubin (chinese herb Qing-Dai) [1,88-90].

While no clear association with adult PAH has been demonstrated with selective serotonin reuptake inhibitors (SSRIs), they are associated with a poor prognosis in those with established PAH [91]. They have also been associated with the development of persistent pulmonary hypertension of the newborn when taken by pregnant mothers [92-96]. (See "Persistent pulmonary hypertension of the newborn (PPHN): Clinical features and diagnosis".)

Other conditions — There are several other conditions that can be complicated by PAH, the pathogenesis of which is less clear.

Connective tissue diseases — While several connective tissue diseases (systemic sclerosis, Raynaud’s disease, systemic lupus erythematosus, mixed connective tissue disease, rheumatoid arthritis) can be complicated by PAH alone they can also be complicated by other forms of pulmonary hypertension (eg, PH from interstitial lung disease and from heart failure, or both), the pathogenesis of which is poorly understood. (See "Overview of pulmonary complications of systemic sclerosis (scleroderma)", section on 'Pulmonary vascular disease'.)

Congenital heart disease — The pathogenesis of pulmonary hypertension in patients with congenital heart disease (eg, septal defects, Eisenmenger syndrome) is discussed separately. (See "Pulmonary hypertension with congenital heart disease: Clinical manifestations and diagnosis".)

Eisenmenger syndrome is the most severe and end-stage form of shunt-related PAH. This group also includes patients that have PAH with coincidental or small defects and those with persistent or worsening PAH despite closure of the defect. (See "Pathophysiology of left-to-right shunts", section on 'Pulmonary hypertension' and "Pulmonary hypertension with congenital heart disease: Clinical manifestations and diagnosis", section on 'Pathogenesis'.)

Human immunodeficiency virus — A small proportion of patients with human immunodeficiency virus (HIV) develop PAH, the pathogenesis of which is discussed separately. (See "Pulmonary arterial hypertension associated with human immunodeficiency virus", section on 'Pathogenesis'.)

Portopulmonary hypertension — Patients with portal hypertension, typically from chronic liver disease, may develop PAH, the pathogenesis of which is discussed separately. (See "Portopulmonary hypertension", section on 'Pathogenesis'.)

Schistosomiasis — PAH can develop in patients infected with schistosomiasis species, particularly those with hepatosplenic involvement, the details of which are provided separately. (See "Schistosomiasis: Epidemiology and clinical manifestations", section on 'Pathogenesis' and "Schistosomiasis: Epidemiology and clinical manifestations", section on 'Pulmonary complications'.)

Miscellaneous — One case of PAH has been reported to be induced by profound vitamin C-deficiency [97].

Vasoreactivity to calcium channel blockers — A small subset (5 to 10 percent) of patients with significant PAH exhibit an acute response to pulmonary vasodilators (usually calcium channel blockers; "responders") [98]. This population may be physiologically different to the remaining majority who do not respond to vasodilators ("non-responders"). Limited data suggest that recruitment of pulmonary flow from the precapillary microvessels (ie, functional capillary surface area [FCSA]) to distal capillary microvessels is different between responders and non-responders. One observational study of 14 drug-naïve patients with PAH measured FCSA before and after vasodilator testing in responders (12 patients) and non-responders (two patients) at the time of diagnosis [99]. Responders had a higher resting FCSA that readily increased during vasodilator testing when compared with non-responders. In contrast, non-responders were unable to recruit FCSA flow during vasodilator testing. These observations suggested that the primary pathology in vasodilator-responsive patients is vasoconstriction at the precapillary level, compared with non-responders who may be unable to recruit flow due to obstructed vessels at the precapillary level. Further exploration of this theory is warranted with the inclusion of larger numbers of responders. Measurement of the vasodilator response is discussed separately. (See "Treatment of pulmonary arterial hypertension (group 1) in adults: Pulmonary hypertension-specific therapy", section on 'Vasoreactive patients'.)

Pulmonary veno-occlusive disease/pulmonary capillary hemangiomatosis — Pulmonary veno-occlusive disease-associated PH (classified as group 1’) (table 1) has a pathophysiology that appears to be distinct from other forms of PAH, the details of which are provided separately. (See "Epidemiology, pathogenesis, clinical evaluation, and diagnosis of pulmonary veno-occlusive disease/pulmonary capillary hemangiomatosis in adults", section on 'Pathogenesis and risk factors'.)

Persistent pulmonary hypertension of the newborn — Persistent pulmonary hypertension of the newborn (PPHN) is classified as group 1. Details regarding the pathogenesis of PPHN are provided separately. (See "Persistent pulmonary hypertension of the newborn (PPHN): Clinical features and diagnosis", section on 'Pathogenesis'.)

SUMMARY AND RECOMMENDATIONS

Definition – Pulmonary hypertension (PH) is classified into five groups based on the World Health Organization (WHO) classification system. The term PAH is used to describe those included in WHO group 1, while the term PH is used when collectively describing all five groups. (See 'Introduction' above.)

Epidemiology – Idiopathic PAH (IPAH) and heritable PAH (HPAH) are rare in the general population and estimated to be 5 to 15 cases per one million adults, although schistosomiasis is the most common cause worldwide. PAH affects younger adults and although it occurs in both genders, women are affected more often and are generally more symptomatic than men. (See 'Epidemiology' above.)

Pathogenetic mechanisms – PAH is a proliferative vasculopathy of the small pulmonary muscular arterioles (<50 microns). It is characterized by vasoconstriction, hyperplasia, hypertrophy, fibrosis, and thrombosis that involves all three layers of the vascular wall (intima, media, adventitia).

Idiopathic and heritable PAH – Patients with IPAH and heritable variants may have a genetic predisposition to PAH (eg, bone morphogenetic protein receptor 2 mutations) with additional contributing mechanisms including vasoactive mediators, potassium channel dysfunction and abnormal response to estrogen. (See 'Pathogenetic mechanisms' above and 'Idiopathic and heritable' above.)

Other causes – The pathogenesis of other forms of PAH (eg, drugs and toxins, connective tissue disease, congenital heart disease, human immunodeficiency virus, portopulmonary hypertension, schistosomiasis, pulmonary venoocclusive disease, persistent pulmonary hypertension of the newborn) is poorly understood. (See 'Pathogenetic mechanisms' above and 'Drugs and toxins' above and 'Other conditions' above and 'Pulmonary veno-occlusive disease/pulmonary capillary hemangiomatosis' above and 'Persistent pulmonary hypertension of the newborn' above.)

  1. Simonneau G, Montani D, Celermajer DS, et al. Haemodynamic definitions and updated clinical classification of pulmonary hypertension. Eur Respir J 2019; 53.
  2. Humbert M, Sitbon O, Chaouat A, et al. Pulmonary arterial hypertension in France: results from a national registry. Am J Respir Crit Care Med 2006; 173:1023.
  3. Ling Y, Johnson MK, Kiely DG, et al. Changing demographics, epidemiology, and survival of incident pulmonary arterial hypertension: results from the pulmonary hypertension registry of the United Kingdom and Ireland. Am J Respir Crit Care Med 2012; 186:790.
  4. McGoon MD, Benza RL, Escribano-Subias P, et al. Pulmonary arterial hypertension: epidemiology and registries. J Am Coll Cardiol 2013; 62:D51.
  5. Badesch DB, Raskob GE, Elliott CG, et al. Pulmonary arterial hypertension: baseline characteristics from the REVEAL Registry. Chest 2010; 137:376.
  6. Peacock AJ, Murphy NF, McMurray JJ, et al. An epidemiological study of pulmonary arterial hypertension. Eur Respir J 2007; 30:104.
  7. Newman JH, Wheeler L, Lane KB, et al. Mutation in the gene for bone morphogenetic protein receptor II as a cause of primary pulmonary hypertension in a large kindred. N Engl J Med 2001; 345:319.
  8. Zhang R, Dai LZ, Xie WP, et al. Survival of Chinese patients with pulmonary arterial hypertension in the modern treatment era. Chest 2011; 140:301.
  9. Frost AE, Badesch DB, Barst RJ, et al. The changing picture of patients with pulmonary arterial hypertension in the United States: how REVEAL differs from historic and non-US Contemporary Registries. Chest 2011; 139:128.
  10. Pugh ME, Sivarajan L, Wang L, et al. Causes of pulmonary hypertension in the elderly. Chest 2014; 146:159.
  11. Stein PD, Matta F, Hughes PG. Scope of problem of pulmonary arterial hypertension. Am J Med 2015; 128:844.
  12. Anand V, Roy SS, Archer SL, et al. Trends and Outcomes of Pulmonary Arterial Hypertension-Related Hospitalizations in the United States: Analysis of the Nationwide Inpatient Sample Database From 2001 Through 2012. JAMA Cardiol 2016; 1:1021.
  13. Humbert M, Morrell NW, Archer SL, et al. Cellular and molecular pathobiology of pulmonary arterial hypertension. J Am Coll Cardiol 2004; 43:13S.
  14. Pietra GG, Capron F, Stewart S, et al. Pathologic assessment of vasculopathies in pulmonary hypertension. J Am Coll Cardiol 2004; 43:25S.
  15. HEATH D, EDWARDS JE. The pathology of hypertensive pulmonary vascular disease; a description of six grades of structural changes in the pulmonary arteries with special reference to congenital cardiac septal defects. Circulation 1958; 18:533.
  16. Yamaki S, Kumate M, Yonesaka S, et al. Lung biopsy diagnosis of operative indication in secundum atrial septal defect with severe pulmonary vascular disease. Chest 2004; 126:1042.
  17. Remetz MS, Cleman MW, Cabin HS. Pulmonary and pleural complications of cardiac disease. Clin Chest Med 1989; 10:545.
  18. Egito ES, Aiello VD, Bosisio IB, et al. Vascular remodeling process in reversibility of pulmonary arterial hypertension secondary to congenital heart disease. Pathol Res Pract 2003; 199:521.
  19. Hoffman JI, Rudolph AM, Heymann MA. Pulmonary vascular disease with congenital heart lesions: pathologic features and causes. Circulation 1981; 64:873.
  20. Haworth SG, Sauer U, Bũhlmeyer K, Reid L. Development of the pulmonary circulation in ventricular septal defect: a quantitative structural study. Am J Cardiol 1977; 40:781.
  21. Garcia-Rivas G, Jerjes-Sánchez C, Rodriguez D, et al. A systematic review of genetic mutations in pulmonary arterial hypertension. BMC Med Genet 2017; 18:82.
  22. Deng Z, Morse JH, Slager SL, et al. Familial primary pulmonary hypertension (gene PPH1) is caused by mutations in the bone morphogenetic protein receptor-II gene. Am J Hum Genet 2000; 67:737.
  23. International PPH Consortium, Lane KB, Machado RD, et al. Heterozygous germline mutations in BMPR2, encoding a TGF-beta receptor, cause familial primary pulmonary hypertension. Nat Genet 2000; 26:81.
  24. Newman JH, Trembath RC, Morse JA, et al. Genetic basis of pulmonary arterial hypertension: current understanding and future directions. J Am Coll Cardiol 2004; 43:33S.
  25. Liu D, Wu WH, Mao YM, et al. BMPR2 mutations influence phenotype more obviously in male patients with pulmonary arterial hypertension. Circ Cardiovasc Genet 2012; 5:511.
  26. Deng Z, Haghighi F, Helleby L, et al. Fine mapping of PPH1, a gene for familial primary pulmonary hypertension, to a 3-cM region on chromosome 2q33. Am J Respir Crit Care Med 2000; 161:1055.
  27. Girerd B, Coulet F, Jaïs X, et al. Characteristics of pulmonary arterial hypertension in affected carriers of a mutation located in the cytoplasmic tail of bone morphogenetic protein receptor type 2. Chest 2015; 147:1385.
  28. Fessel JP, Loyd JE, Austin ED. The genetics of pulmonary arterial hypertension in the post-BMPR2 era. Pulm Circ 2011; 1:305.
  29. West J, Fagan K, Steudel W, et al. Pulmonary hypertension in transgenic mice expressing a dominant-negative BMPRII gene in smooth muscle. Circ Res 2004; 94:1109.
  30. Kimura N, Matsuo R, Shibuya H, et al. BMP2-induced apoptosis is mediated by activation of the TAK1-p38 kinase pathway that is negatively regulated by Smad6. J Biol Chem 2000; 275:17647.
  31. Trembath RC, Thomson JR, Machado RD, et al. Clinical and molecular genetic features of pulmonary hypertension in patients with hereditary hemorrhagic telangiectasia. N Engl J Med 2001; 345:325.
  32. Johnson DW, Berg JN, Baldwin MA, et al. Mutations in the activin receptor-like kinase 1 gene in hereditary haemorrhagic telangiectasia type 2. Nat Genet 1996; 13:189.
  33. Harrison RE, Flanagan JA, Sankelo M, et al. Molecular and functional analysis identifies ALK-1 as the predominant cause of pulmonary hypertension related to hereditary haemorrhagic telangiectasia. J Med Genet 2003; 40:865.
  34. Marcos E, Fadel E, Sanchez O, et al. Serotonin-induced smooth muscle hyperplasia in various forms of human pulmonary hypertension. Circ Res 2004; 94:1263.
  35. Eddahibi S, Humbert M, Fadel E, et al. Serotonin transporter overexpression is responsible for pulmonary artery smooth muscle hyperplasia in primary pulmonary hypertension. J Clin Invest 2001; 108:1141.
  36. McAllister KA, Grogg KM, Johnson DW, et al. Endoglin, a TGF-beta binding protein of endothelial cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat Genet 1994; 8:345.
  37. Abdalla SA, Pece-Barbara N, Vera S, et al. Analysis of ALK-1 and endoglin in newborns from families with hereditary hemorrhagic telangiectasia type 2. Hum Mol Genet 2000; 9:1227.
  38. Nasim MT, Ogo T, Ahmed M, et al. Molecular genetic characterization of SMAD signaling molecules in pulmonary arterial hypertension. Hum Mutat 2011; 32:1385.
  39. Austin ED, Ma L, LeDuc C, et al. Whole exome sequencing to identify a novel gene (caveolin-1) associated with human pulmonary arterial hypertension. Circ Cardiovasc Genet 2012; 5:336.
  40. Ma L, Roman-Campos D, Austin ED, et al. A novel channelopathy in pulmonary arterial hypertension. N Engl J Med 2013; 369:351.
  41. Antigny F, Hautefort A, Meloche J, et al. Potassium Channel Subfamily K Member 3 (KCNK3) Contributes to the Development of Pulmonary Arterial Hypertension. Circulation 2016; 133:1371.
  42. Hadinnapola C, Bleda M, Haimel M, et al. Phenotypic Characterization of EIF2AK4 Mutation Carriers in a Large Cohort of Patients Diagnosed Clinically With Pulmonary Arterial Hypertension. Circulation 2017; 136:2022.
  43. Mason CR, Baker JF. Effects of optokinetic velocity and medial vestibulo-cerebellum lesions on vestibulo-ocular reflex direction adaptation in the cat. Brain Res 1989; 490:373.
  44. Giaid A, Saleh D. Reduced expression of endothelial nitric oxide synthase in the lungs of patients with pulmonary hypertension. N Engl J Med 1995; 333:214.
  45. Giaid A, Yanagisawa M, Langleben D, et al. Expression of endothelin-1 in the lungs of patients with pulmonary hypertension. N Engl J Med 1993; 328:1732.
  46. Christman BW, McPherson CD, Newman JH, et al. An imbalance between the excretion of thromboxane and prostacyclin metabolites in pulmonary hypertension. N Engl J Med 1992; 327:70.
  47. Yoshibayashi M, Nishioka K, Nakao K, et al. Plasma endothelin concentrations in patients with pulmonary hypertension associated with congenital heart defects. Evidence for increased production of endothelin in pulmonary circulation. Circulation 1991; 84:2280.
  48. Ishikawa S, Miyauchi T, Sakai S, et al. Elevated levels of plasma endothelin-1 in young patients with pulmonary hypertension caused by congenital heart disease are decreased after successful surgical repair. J Thorac Cardiovasc Surg 1995; 110:271.
  49. Fuse S, Kamiya T. Plasma thromboxane B2 concentration in pulmonary hypertension associated with congenital heart disease. Circulation 1994; 90:2952.
  50. Ventetuolo CE, Baird GL, Barr RG, et al. Higher Estradiol and Lower Dehydroepiandrosterone-Sulfate Levels Are Associated with Pulmonary Arterial Hypertension in Men. Am J Respir Crit Care Med 2016; 193:1168.
  51. Nong Z, Hoylaerts M, Van Pelt N, et al. Nitric oxide inhalation inhibits platelet aggregation and platelet-mediated pulmonary thrombosis in rats. Circ Res 1997; 81:865.
  52. Fagan KA, Fouty BW, Tyler RC, et al. The pulmonary circulation of homozygous or heterozygous eNOS-null mice is hyperresponsive to mild hypoxia. J Clin Invest 1999; 103:291.
  53. Zhao YD, Courtman DW, Deng Y, et al. Rescue of monocrotaline-induced pulmonary arterial hypertension using bone marrow-derived endothelial-like progenitor cells: efficacy of combined cell and eNOS gene therapy in established disease. Circ Res 2005; 96:442.
  54. Kaneko FT, Arroliga AC, Dweik RA, et al. Biochemical reaction products of nitric oxide as quantitative markers of primary pulmonary hypertension. Am J Respir Crit Care Med 1998; 158:917.
  55. Demoncheaux EA, Higenbottam TW, Kiely DG, et al. Decreased whole body endogenous nitric oxide production in patients with primary pulmonary hypertension. J Vasc Res 2005; 42:133.
  56. Granton J, Langleben D, Kutryk MB, et al. Endothelial NO-Synthase Gene-Enhanced Progenitor Cell Therapy for Pulmonary Arterial Hypertension: The PHACeT Trial. Circ Res 2015; 117:645.
  57. Yuan XJ, Wang J, Juhaszova M, et al. Attenuated K+ channel gene transcription in primary pulmonary hypertension. Lancet 1998; 351:726.
  58. Yu Y, Fantozzi I, Remillard CV, et al. Enhanced expression of transient receptor potential channels in idiopathic pulmonary arterial hypertension. Proc Natl Acad Sci U S A 2004; 101:13861.
  59. Weir EK, Reeve HL, Huang JM, et al. Anorexic agents aminorex, fenfluramine, and dexfenfluramine inhibit potassium current in rat pulmonary vascular smooth muscle and cause pulmonary vasoconstriction. Circulation 1996; 94:2216.
  60. Chen X, Austin ED, Talati M, et al. Oestrogen inhibition reverses pulmonary arterial hypertension and associated metabolic defects. Eur Respir J 2017; 50.
  61. Abramowicz MJ, Van Haecke P, Demedts M, Delcroix M. Primary pulmonary hypertension after amfepramone (diethylpropion) with BMPR2 mutation. Eur Respir J 2003; 22:560.
  62. Abenhaim L, Moride Y, Brenot F, et al. Appetite-suppressant drugs and the risk of primary pulmonary hypertension. International Primary Pulmonary Hypertension Study Group. N Engl J Med 1996; 335:609.
  63. Voelkel NF, Clarke WR, Higenbottam T. Obesity, dexfenfluramine, and pulmonary hypertension. A lesson not learned? Am J Respir Crit Care Med 1997; 155:786.
  64. Brenot F, Herve P, Petitpretz P, et al. Primary pulmonary hypertension and fenfluramine use. Br Heart J 1993; 70:537.
  65. Delcroix M, Kurz X, Walckiers D, et al. High incidence of primary pulmonary hypertension associated with appetite suppressants in Belgium. Eur Respir J 1998; 12:271.
  66. Zamanian RT, Hedlin H, Greuenwald P, et al. Features and Outcomes of Methamphetamine-associated Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2018; 197:788.
  67. Eddahibi S, Humbert M, Fadel E, et al. Hyperplasia of pulmonary artery smooth muscle cells is causally related to overexpression of the serotonin transporter in primary pulmonary hypertension. Chest 2002; 121:97S.
  68. Eddahibi S, Raffestin B, Hamon M, Adnot S. Is the serotonin transporter involved in the pathogenesis of pulmonary hypertension? J Lab Clin Med 2002; 139:194.
  69. Eddahibi S, Adnot S. Anorexigen-induced pulmonary hypertension and the serotonin (5-HT) hypothesis: lessons for the future in pathogenesis. Respir Res 2002; 3:9.
  70. Hervé P, Launay JM, Scrobohaci ML, et al. Increased plasma serotonin in primary pulmonary hypertension. Am J Med 1995; 99:249.
  71. Rich S, Rubin L, Walker AM, et al. Anorexigens and pulmonary hypertension in the United States: results from the surveillance of North American pulmonary hypertension. Chest 2000; 117:870.
  72. Garcia-Dorado D, Miller DD, Garcia EJ, et al. An epidemic of pulmonary hypertension after toxic rapeseed oil ingestion in Spain. J Am Coll Cardiol 1983; 1:1216.
  73. Savale L, Chaumais MC, Cottin V, et al. Pulmonary hypertension associated with benfluorex exposure. Eur Respir J 2012; 40:1164.
  74. Boutet K, Frachon I, Jobic Y, et al. Fenfluramine-like cardiovascular side-effects of benfluorex. Eur Respir J 2009; 33:684.
  75. Frachon I, Etienne Y, Jobic Y, et al. Benfluorex and unexplained valvular heart disease: a case-control study. PLoS One 2010; 5:e10128.
  76. Montani D, Bergot E, Günther S, et al. Pulmonary arterial hypertension in patients treated by dasatinib. Circulation 2012; 125:2128.
  77. Dhillon S, Kaker A, Dosanjh A, et al. Irreversible pulmonary hypertension associated with the use of interferon alpha for chronic hepatitis C. Dig Dis Sci 2010; 55:1785.
  78. Caravita S, Secchi MB, Wu SC, et al. Sildenafil therapy for interferon-β-1a-induced pulmonary arterial hypertension: a case report. Cardiology 2011; 120:187.
  79. Ledinek AH, Jazbec SS, Drinovec I, Rot U. Pulmonary arterial hypertension associated with interferon beta treatment for multiple sclerosis: a case report. Mult Scler 2009; 15:885.
  80. Savale L, Sattler C, Günther S, et al. Pulmonary arterial hypertension in patients treated with interferon. Eur Respir J 2014; 44:1627.
  81. Medication and use guides for PLEGRIDY peginterferon beta-1a. US Food and Drug Administration, 2023. https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/125499s028lbl.pdf (Accessed on September 06, 2023).
  82. Schaiberger PH, Kennedy TC, Miller FC, et al. Pulmonary hypertension associated with long-term inhalation of "crank" methamphetamine. Chest 1993; 104:614.
  83. Albertson TE, Walby WF, Derlet RW. Stimulant-induced pulmonary toxicity. Chest 1995; 108:1140.
  84. Chin KM, Channick RN, Rubin LJ. Is methamphetamine use associated with idiopathic pulmonary arterial hypertension? Chest 2006; 130:1657.
  85. Gaine SP, Rubin LJ, Kmetzo JJ, et al. Recreational use of aminorex and pulmonary hypertension. Chest 2000; 118:1496.
  86. Alvarez PA, Saad AK, Flagel S, et al. Leflunomide-induced pulmonary hypertension in a young woman with rheumatoid arthritis: a case report. Cardiovasc Toxicol 2012; 12:180.
  87. Hagiwara M, Tsuchida A, Hyakkoku M, et al. Delayed onset of pulmonary hypertension associated with an appetite suppressant, mazindol: a case report. Jpn Circ J 2000; 64:218.
  88. Riou M, Seferian A, Savale L, et al. Deterioration of pulmonary hypertension and pleural effusion with bosutinib following dasatinib lung toxicity. Eur Respir J 2016; 48:1517.
  89. Renard S, Borentain P, Salaun E, et al. Severe Pulmonary Arterial Hypertension in Patients Treated for Hepatitis C With Sofosbuvir. Chest 2016; 149:e69.
  90. Nishio M, Hirooka K, Doi Y. Chinese herbal drug natural indigo may cause pulmonary artery hypertension. Eur Heart J 2016; 37:1992.
  91. Sadoughi A, Roberts KE, Preston IR, et al. Use of selective serotonin reuptake inhibitors and outcomes in pulmonary arterial hypertension. Chest 2013; 144:531.
  92. Chambers CD, Johnson KA, Dick LM, et al. Birth outcomes in pregnant women taking fluoxetine. N Engl J Med 1996; 335:1010.
  93. Andrade SE, McPhillips H, Loren D, et al. Antidepressant medication use and risk of persistent pulmonary hypertension of the newborn. Pharmacoepidemiol Drug Saf 2009; 18:246.
  94. Wichman CL, Moore KM, Lang TR, et al. Congenital heart disease associated with selective serotonin reuptake inhibitor use during pregnancy. Mayo Clin Proc 2009; 84:23.
  95. Wilson KL, Zelig CM, Harvey JP, et al. Persistent pulmonary hypertension of the newborn is associated with mode of delivery and not with maternal use of selective serotonin reuptake inhibitors. Am J Perinatol 2011; 28:19.
  96. Kieler H, Artama M, Engeland A, et al. Selective serotonin reuptake inhibitors during pregnancy and risk of persistent pulmonary hypertension in the newborn: population based cohort study from the five Nordic countries. BMJ 2012; 344:d8012.
  97. Gayen SK, Abdelrahman AA, Preston IR, et al. Vitamin C Deficiency-Induced Pulmonary Arterial Hypertension. Chest 2020; 157:e21.
  98. Sitbon O, Humbert M, Jaïs X, et al. Long-term response to calcium channel blockers in idiopathic pulmonary arterial hypertension. Circulation 2005; 111:3105.
  99. Langleben D, Orfanos SE, Giovinazzo M, et al. Acute vasodilator responsiveness and microvascular recruitment in idiopathic pulmonary arterial hypertension. Ann Intern Med 2015; 162:154.
Topic 8247 Version 37.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟