Pulmonary Hypertension: Molecular Mechanisms and Clinical Studies

Joseph Adu-Amankwaah , Qiang You , Xiaoer Liu , Jiayi Jiang , Dongqi Yang , Kuntao Liu , Jinxiang Yuan , Yanfang Wang , Qinghua Hu , Rubin Tan

MedComm ›› 2025, Vol. 6 ›› Issue (3) : e70134

PDF
MedComm ›› 2025, Vol. 6 ›› Issue (3) : e70134 DOI: 10.1002/mco2.70134
REVIEW

Pulmonary Hypertension: Molecular Mechanisms and Clinical Studies

Author information +
History +
PDF

Abstract

Pulmonary hypertension (PH) stands as a tumor paradigm cardiovascular disease marked by hyperproliferation of cells and vascular remodeling, culminating in heart failure. Complex genetic and epigenetic mechanisms collectively contribute to the disruption of pulmonary vascular homeostasis. In recent years, advancements in research technology have identified numerous gene deletions and mutations, in addition to bone morphogenetic protein receptor type 2, that are closely associated with the vascular remodeling process in PH. Additionally, epigenetic modifications such as RNA methylation, DNA methylation, histone modification, and noncoding RNAs have been shown to precisely regulate PH molecular networks in a cell-type-specific manner, emerging as potential biomarkers and therapeutic targets. This review summarizes and analyzes the roles and molecular mechanisms of currently identified genes and epigenetic factors in PH, emphasizing the pivotal role of long ncRNAs in its regulation. Additionally, it examines current clinical and preclinical therapies for PH targeting these genes and epigenetic factors and explores potential new treatment strategies.

Keywords

clinical therapeutics / epigenetic / genetic / noncoding RNAs / pulmonary hypertension

Cite this article

Download citation ▾
Joseph Adu-Amankwaah, Qiang You, Xiaoer Liu, Jiayi Jiang, Dongqi Yang, Kuntao Liu, Jinxiang Yuan, Yanfang Wang, Qinghua Hu, Rubin Tan. Pulmonary Hypertension: Molecular Mechanisms and Clinical Studies. MedComm, 2025, 6(3): e70134 DOI:10.1002/mco2.70134

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

J. Zhang, X. Hu, T. Wang, et al., “Extracellular Vesicles in Venous Thromboembolism and Pulmonary Hypertension,” Journal of Nanobiotechnology 21, no. 1 (2023): 461.

[2]

R. Naeije, M. J. Richter, and L. J. Rubin, “The Physiological Basis of Pulmonary Arterial Hypertension,” European Respiratory Journal 59, no. 6 (2022): 2102334.

[3]

R. Tan, Q. You, D. Yu, et al., “Novel Hub Genes Associated with Pulmonary Artery Remodeling in Pulmonary Hypertension,” Frontiers in Cardiovascular Medicine 9 (2022): 945854.

[4]

S. S. Virani, A. Alonso, H. J. Aparicio, et al., “Heart Disease and Stroke Statistics-2021 Update: a Report from the American Heart Association,” Circulation 143, no. 8 (2021): e254–e743.

[5]

G. A. Heresi, D. M. Platt, W. Wang, et al., “Healthcare Burden of Pulmonary Hypertension Owing to Lung Disease and/or Hypoxia,” BMC Pulmonary Medicine 17, no. 1 (2017): 58.

[6]

M. Humbert, G. Kovacs, M. M. Hoeper, et al., “2022 ESC/ERS Guidelines for the Diagnosis and Treatment of Pulmonary Hypertension,” European Heart Journal 43, no. 38 (2022): 3618–3731.

[7]

G. Simonneau, D. Montani, D. S. Celermajer, et al., “Haemodynamic Definitions and Updated Clinical Classification of Pulmonary Hypertension,” European Respiratory Journal 53, no. 1 (2019): 1801913.

[8]

M. W. Dodson, L. M. Brown, and C. G. Elliott, “Pulmonary Arterial Hypertension,” Heart Failure Clinics 14, no. 3 (2018): 255–269.

[9]

A. R. Hemnes and M. Humbert, “Pathobiology of Pulmonary Arterial Hypertension: Understanding the Roads Less Travelled,” European Respiratory Review 26, no. 146 (2017): 170093.

[10]

R. M. Tuder, S. L. Archer, P. Dorfmüller, et al., “Relevant Issues in the Pathology and Pathobiology of Pulmonary Hypertension,” Journal of the American College of Cardiology 62, no. 25 Suppl (2013): D4–D12.

[11]

R. Tan, J. Li, F. Liu, et al., “Phenylalanine Induces Pulmonary Hypertension through Calcium-sensing Receptor Activation,” American Journal of Physiology. Lung Cellular and Molecular Physiology 319, no. 6 (2020): L1010–L1020.

[12]

B. Liu, Y. P. Wei, X. Fan, et al., “Calcium Sensing Receptor Variants Increase Pulmonary Hypertension Susceptibility,” Hypertension 79, no. 7 (2022): 1348–1360.

[13]

R. Xiao, Y. Su, T. Feng, et al., “Monocrotaline Induces Endothelial Injury and Pulmonary Hypertension by Targeting the Extracellular Calcium-sensing Receptor,” Journal of the American Heart Association 6, no. 4 (2017): e004865.

[14]

E. L. Wang, J. J. Zhang, F. M. Luo, et al., “Cerebellin-2 Promotes Endothelial-mesenchymal Transition in Hypoxic Pulmonary Hypertension Rats by Activating NF-κB/HIF-1α/Twist1 Pathway,” Life Sciences 328 (2023): 121879.

[15]

S. J. Cassady and G. V. Ramani, “Right Heart Failure in Pulmonary Hypertension,” Cardiology Clinics 38, no. 2 (2020): 243–255.

[16]

M. A. Aldred, N. W. Morrell, and C. Guignabert, “New Mutations and Pathogenesis of Pulmonary Hypertension: Progress and Puzzles in Disease Pathogenesis,” Circulation Research 130, no. 9 (2022): 1365–1381.

[17]

L. Ho, N. Hossen, T. Nguyen, A. Vo, and F. Ahsan, “Epigenetic Mechanisms as Emerging Therapeutic Targets and Microfluidic Chips Application in Pulmonary Arterial Hypertension,” Biomedicines 10, no. 1 (2022): 170.

[18]

Ł. Wołowiec, M. Mędlewska, J. Osiak, et al., “MicroRNA and lncRNA as the Future of Pulmonary Arterial Hypertension Treatment,” International Journal of Molecular Sciences 24, no. 11 (2023): 9735.

[19]

F. Kopp and J. T. Mendell, “Functional Classification and Experimental Dissection of Long Noncoding RNAs,” Cell 172, no. 3 (2018): 393–407.

[20]

C. A Santos-Ferreira, M. T. Abreu, C. I. Marques, L. M. Gonçalves, R. Baptista, and H. M. Girão, “Micro-RNA Analysis in Pulmonary Arterial Hypertension: Current Knowledge and Challenges,” JACC: Basic to Translational Science 5, no. 11 (2020): 1149–1162.

[21]

B. Tatius, W. Wasityastuti, F. D. Astarini, and D. A. A. Nugrahaningsih, “Significance of BMPR2 Mutations in Pulmonary Arterial Hypertension,” Respiratory Investigation 59, no. 4 (2021): 397–407.

[22]

A. Hautefort, P. Mendes-Ferreira, J. Sabourin, et al., “Bmpr2 mutant Rats Develop Pulmonary and Cardiac Characteristics of Pulmonary Arterial Hypertension,” Circulation 139, no. 7 (2019): 932–948.

[23]

A. L. Theilmann, L. G. Hawke, L. R. Hilton, et al., “Endothelial BMPR2 Loss Drives a Proliferative Response to BMP (Bone Morphogenetic Protein) 9 via Prolonged Canonical Signaling,” Arteriosclerosis, Thrombosis, and Vascular Biology 40, no. 11 (2020): 2605–2618.

[24]

L. A. Hurst, B. J. Dunmore, L. Long, et al., “TNFα Drives Pulmonary Arterial Hypertension by Suppressing the BMP Type-II Receptor and Altering NOTCH Signalling,” Nature Communications 8 (2017): 14079.

[25]

L. Wang, J. R. Moonen, A. Cao, et al., “Dysregulated Smooth Muscle Cell BMPR2-ARRB2 Axis Causes Pulmonary Hypertension,” Circulation Research 132, no. 5 (2023): 545–564.

[26]

L. R. Hilton, M. T. Rätsep, M. M. VandenBroek, et al., “Impaired Interleukin-15 Signaling via BMPR2 Loss Drives Natural Killer Cell Deficiency and Pulmonary Hypertension,” Hypertension 79, no. 11 (2022): 2493–2504.

[27]

D. Li, N. Y. Shao, J. R. Moonen, et al., “ALDH1A3 coordinates Metabolism with Gene Regulation in Pulmonary Arterial Hypertension,” Circulation 143, no. 21 (2021): 2074–2090.

[28]

Z. C. Nwosu, M. P. Ebert, S. Dooley, and C. Meyer, “Caveolin-1 in the Regulation of Cell Metabolism: a Cancer Perspective,” Molecular Cancer 15, no. 1 (2016): 71.

[29]

J. Fan, S. Zheng, M. Wang, and X. Yuan, “The Critical Roles of Caveolin-1 in Lung Diseases,” Frontiers in Pharmacology 15 (2024): 1417834.

[30]

Z. Daneva, C. Marziano, M. Ottolini, et al., “Caveolar Peroxynitrite Formation Impairs Endothelial TRPV4 Channels and Elevates Pulmonary Arterial Pressure in Pulmonary Hypertension,” PNAS 118, no. 17 (2021): e2023130118.

[31]

K. Yang, M. Zhao, J. Huang, et al., “Pharmacological Activation of PPARγ Inhibits Hypoxia-induced Proliferation through a Caveolin-1-targeted and -dependent Mechanism in PASMCs,” American Journal of Physiology. Cell Physiology 314, no. 4 (2018): C428–C438.

[32]

S. D. S Oliveira, J. Chen, M. Castellon, et al., “Injury-induced Shedding of Extracellular Vesicles Depletes Endothelial Cells of Cav-1 (Caveolin-1) and Enables TGF-β (Transforming Growth Factor-β)-dependent Pulmonary Arterial Hypertension,” Arteriosclerosis, Thrombosis, and Vascular Biology 39, no. 6 (2019): 1191–1202.

[33]

S. Tomita, N. Nakanishi, T. Ogata, et al., “The Cavin-1/Caveolin-1 Interaction Attenuates BMP/Smad Signaling in Pulmonary Hypertension by Interfering with BMPR2/Caveolin-1 Binding,” Communications Biology 7, no. 1 (2024): 40.

[34]

Y. Marinho, E. S. Villarreal, S. Y. Aboagye, et al., “Schistosomiasis-associated Pulmonary Hypertension Unveils Disrupted Murine Gut-lung Microbiome and Reduced Endoprotective Caveolin-1/BMPR2 Expression,” Frontiers in Immunology 14 (2023): 1254762.

[35]

E. D. Willers, J. H. Newman, J. E. Loyd, et al., “Serotonin Transporter Polymorphisms in Familial and Idiopathic Pulmonary Arterial Hypertension,” American Journal of Respiratory and Critical Care Medicine 173, no. 7 (2006): 798–802.

[36]

K. E. Roberts, M. B. Fallon, M. J. Krowka, et al., “Serotonin Transporter Polymorphisms in Patients with Portopulmonary Hypertension,” Chest 135, no. 6 (2009): 1470–1475.

[37]

W. Ren, S. W. Watts, and B. L. Fanburg, “Serotonin Transporter Interacts with the PDGFβ Receptor in PDGF-BB-induced Signaling and Mitogenesis in Pulmonary Artery Smooth Muscle Cells,” American Journal of Physiology. Lung Cellular and Molecular Physiology 300, no. 3 (2011): L486–L497.

[38]

K. White, L. Loughlin, Z. Maqbool, et al., “Serotonin Transporter, Sex, and Hypoxia: Microarray Analysis in the Pulmonary Arteries of Mice Identifies Genes with Relevance to human PAH,” Physiological Genomics 43, no. 8 (2011): 417–437.

[39]

K. White, Y. Dempsie, M. Nilsen, A. F. Wright, L. Loughlin, and M. R. MacLean, “The Serotonin Transporter, Gender, and 17β Oestradiol in the Development of Pulmonary Arterial Hypertension,” Cardiovascular Research 90, no. 2 (2011): 373–382.

[40]

A. Baloira, M. Núñez, J. Cifrian, C. Vilariño, M. Ojeda, and D. Valverde, “Polymorphisms in the Serotonin Transporter Protein (SERT) Gene in Patients with Pulmonary Arterial Hypertension,” Archivos De Bronconeumologia 48, no. 3 (2012): 77–80.

[41]

H. Zhang, M. Xu, J. Xia, and R. Y. Qin, “Association between Serotonin Transporter (SERT) Gene Polymorphism and Idiopathic Pulmonary Arterial Hypertension: a Meta-analysis and Review of the Literature,” Metabolism 62, no. 12 (2013): 1867–1875.

[42]

N. Gallego-Zazo, A. Cruz-Utrilla, M. J. Del Cerro, et al., “Description of Two New Cases of AQP1 Related Pulmonary Arterial Hypertension and Review of the Literature,” Genes 13, no. 5 (2022): 927.

[43]

K. W. Liang, S. K. Chang, Y. W. Chen, W. J. Tsai, and K. Y. Wang, “A Cluster of Heritable Pulmonary Arterial Hypertension Cases in a family with all Three Siblings Carrying the Same Novel AQP1 c.273C>G Variant-a Case Report,” Pulmonary Circulation 13, no. 2 (2023): e12211.

[44]

S. Gräf, M. Haimel, M. Bleda, et al., “Identification of Rare Sequence Variation Underlying Heritable Pulmonary Arterial Hypertension,” Nature Communications 9, no. 1 (2018): 1416.

[45]

M. Liu, Q. Liu, Y. Pei, et al., “Aqp-1 Gene Knockout Attenuates Hypoxic Pulmonary Hypertension of Mice,” Arteriosclerosis, Thrombosis, and Vascular Biology 39, no. 1 (2019): 48–62.

[46]

K. Leggett, J. Maylor, C. Undem, et al., “Hypoxia-induced Migration in Pulmonary Arterial Smooth Muscle Cells Requires Calcium-dependent Upregulation of Aquaporin 1,” American Journal of Physiology. Lung Cellular and Molecular Physiology 303, no. 4 (2012): L343–L353.

[47]

N. Lai, J. Lade, K. Leggett, et al., “The Aquaporin 1 C-terminal Tail Is Required for Migration and Growth of Pulmonary Arterial Myocytes,” American Journal of Respiratory Cell and Molecular Biology 50, no. 6 (2014): 1010–1020.

[48]

X. Yun, H. Jiang, N. Lai, J. Wang, and L. A. Shimoda, “Aquaporin 1-mediated Changes in Pulmonary Arterial Smooth Muscle Cell Migration and Proliferation Involve β-catenin,” American Journal of Physiology. Lung Cellular and Molecular Physiology 313, no. 5 (2017): L889–L898.

[49]

X. Yun, S. Niedermeyer, M. R. Andrade, et al., “Aquaporin 1 Confers Apoptosis Resistance in Pulmonary Arterial Smooth Muscle Cells from the SU5416 Hypoxia Rat Model,” Physiological Reports 12, no. 16 (2024): e16156.

[50]

K. W. Liang, S. K. Chang, Y. W. Chen, W. W. Lin, W. J. Tsai, and K. Y. Wang, “Whole Exome Sequencing of Patients with Heritable and Idiopathic Pulmonary Arterial Hypertension in central Taiwan,” Frontiers in Cardiovascular Medicine 9 (2022): 911649.

[51]

A. G. Vassiliou, C. Keskinidou, A. Kotanidou, et al., “Knockdown of Bone Morphogenetic Protein Type II Receptor Leads to Decreased Aquaporin 1 Expression and Function in human Pulmonary Microvascular Endothelial Cells,” Canadian Journal of Physiology and Pharmacology 98, no. 11 (2020): 834–839.

[52]

N. S. Lotsios, C. Keskinidou, I. Dimopoulou, et al., “Effects of Modulating BMP9, BMPR2, and AQP1 on BMP Signaling in human Pulmonary Microvascular Endothelial Cells,” International Journal of Molecular Sciences 25, no. 15 (2024): 8043.

[53]

M. T. Wang, K. P. Weng, S. K. Chang, W. C. Huang, and L. W. Chen, “Hemodynamic and Clinical Profiles of Pulmonary Arterial Hypertension Patients with GDF2 and BMPR2 Variants,” International Journal of Molecular Sciences 25, no. 5 (2024): 2734.

[54]

C. L. Welch and W. K. Chung, “Channelopathy Genes in Pulmonary Arterial Hypertension,” Biomolecules 12, no. 2 (2022): 265.

[55]

C. Barozzi, M. Galletti, L. Tomasi, et al., “A Combined Targeted and Whole Exome Sequencing Approach Identified Novel Candidate Genes Involved in Heritable Pulmonary Arterial Hypertension,” Scientific Reports 9, no. 1 (2019): 753.

[56]

C. A. Eichstaedt, Z. Saßmannshausen, M. Shaukat, et al., “Gene Panel Diagnostics Reveals New Pathogenic Variants in Pulmonary Arterial Hypertension,” Respiratory Research 23, no. 1 (2022): 74.

[57]

S. M. Gelinas, C. E. Benson, M. A. Khan, et al., “Whole Exome Sequence Analysis Provides Novel Insights into the Genetic Framework of Childhood-onset Pulmonary Arterial Hypertension,” Genes 11, no. 11 (2020): 1328.

[58]

L. Stevens, E. Colglazier, C. Parker, et al., “Genetics Dictating Therapeutic Decisions in Pediatric Pulmonary Hypertension? A Case Report Suggesting We Are Getting Closer,” Pulmonary Circulation 12, no. 1 (2022): e12033.

[59]

R. D. Machado, C. L. Welch, M. Haimel, et al., “Biallelic Variants of ATP13A3 Cause Dose-dependent Childhood-onset Pulmonary Arterial Hypertension Characterised by Extreme Morbidity and Mortality,” Journal of Medical Genetics 59, no. 9 (2022): 906–911.

[60]

M. Lerche, C. A. Eichstaedt, K. Hinderhofer, et al., “Mutually Reinforcing Effects of Genetic Variants and Interferon-β 1a Therapy for Pulmonary Arterial Hypertension Development in Multiple Sclerosis Patients,” Pulmonary Circulation 9, no. 3 (2019): 2045894019872192.

[61]

B. Liu, M. Azfar, E. Legchenko, et al., “ATP13A3 variants Promote Pulmonary Arterial Hypertension by Disrupting Polyamine Transport,” Cardiovascular Research 120, no. 7 (2024): 756–768.

[62]

L. Southgate, R. D. Machado, S. Gräf, and N. W. Morrell, “Molecular Genetic Framework Underlying Pulmonary Arterial Hypertension,” Nature Reviews Cardiology 17, no. 2 (2020): 85–95.

[63]

Y. Wu, J. Wharton, R. Walters, et al., “The Pathophysiological Role of Novel Pulmonary Arterial Hypertension Gene SOX17,” European Respiratory 58, no. 3 (2021): 2004172.

[64]

H. Ishida, J. Maeda, K. Uchida, and H. Yamagishi, “Unique Pulmonary Hypertensive Vascular Diseases Associated with Heart and Lung Developmental Defects,” Journal of Cardiovascular Development and Disease 10, no. 8 (2023): 333.

[65]

H. Ostler, C. Fall, H. El-Said, et al., “Early Identification of SOX17 Deficiency in Infants to Guide Management of Heritable Pulmonary Arterial Hypertension Using PDA Stent to Create Reverse Potts Shunt Physiology,” Pulmonary Circulation 14, no. 2 (2024): e12366.

[66]

T. M. Wang, S. S. Wang, Y. J. Xu, et al., “SOX17 loss-of-function Mutation Underlying Familial Pulmonary Arterial Hypertension,” International Heart Journal 62, no. 3 (2021): 566–574.

[67]

T. Hiraide, M. Kataoka, H. Suzuki, et al., “SOX17 mutations in Japanese Patients with Pulmonary Arterial Hypertension,” American Journal of Respiratory and Critical Care Medicine 198, no. 9 (2018): 1231–1233.

[68]

N. Zhu, C. L. Welch, J. Wang, et al., “Rare Variants in SOX17 Are Associated with Pulmonary Arterial Hypertension with Congenital Heart Disease,” Genome Medicine 10, no. 1 (2018): 56.

[69]

D. Montani, B. Lechartier, B. Girerd, et al., “An Emerging Phenotype of Pulmonary Arterial Hypertension Patients Carrying SOX17 Variants,” European Respiratory Journal 60, no. 6 (2022): 2200656.

[70]

J. Aman, N. W. Morrell, C. J. Rhodes, M. R. Wilkins, and H. J. Bogaard, “The SOX17 Phenotype in Pulmonary Arterial Hypertension: Lessons for Pathobiology and Clinical Management,” European Respiratory Journal 60, no. 6 (2022): 2201438.

[71]

S. Gu, R. Kumar, M. H. Lee, C. Mickael, and B. B. Graham, “Common Genetic Variants in Pulmonary Arterial Hypertension,” The Lancet Respiratory Medicine 7, no. 3 (2019): 190–191.

[72]

C. J. Rhodes, K. Batai, M. Bleda, et al., “Genetic Determinants of Risk in Pulmonary Arterial Hypertension: International Genome-wide Association Studies and Meta-analysis,” The Lancet Respiratory Medicine 7, no. 3 (2019): 227–238.

[73]

X. Sun, B. L. Sun, A. Babicheva, et al., “Direct Extracellular NAMPT Involvement in Pulmonary Hypertension and Vascular Remodeling. Transcriptional Regulation by SOX and HIF-2α” American Journal of Respiratory Cell and Molecular Biology 63, no. 1 (2020): 92–103.

[74]

D. Yi, B. Liu, H. Ding, et al., “E2F1 Mediates SOX17 Deficiency-induced Pulmonary Hypertension,” Hypertension 80, no. 11 (2023): 2357–2371.

[75]

S. Sangam, X. Sun, T. H Schwantes-An, et al., “SOX17 deficiency Mediates Pulmonary Hypertension: at the Crossroads of Sex, Metabolism, and Genetics,” American Journal of Respiratory and Critical Care Medicine 207, no. 8 (2023): 1055–1069.

[76]

S. M. Lin and D. B. Frank, “SOX17 at the Intersection of Sex, Transcription, and Metabolism in Pulmonary Hypertension,” American Journal of Respiratory and Critical Care Medicine 207, no. 8 (2023): 971–972.

[77]

C. S. Park, S. H. Kim, H. Y. Yang, et al., “Sox17 deficiency Promotes Pulmonary Arterial Hypertension via HGF/c-Met Signaling,” Circulation Research 131, no. 10 (2022): 792–806.

[78]

A. J. Ainscough, T. J. Smith, M. Haensel, et al., “An Organ-on-chip Model of Pulmonary Arterial Hypertension Identifies a BMPR2-SOX17-prostacyclin Signalling Axis,” Communications Biology 5, no. 1 (2022): 1192.

[79]

S. L. Mirza, P. D. Upton, J. Hodgson, S. Gräf, N. W. Morrell, and B. J. Dunmore, “SEMA3G Regulates BMP9 Inhibition of VEGF-mediated Migration and Network Formation in Pulmonary Endothelial Cells,” Vascular Pharmacology 155 (2024): 107381.

[80]

X. Zou, T. Liu, Z. Huang, et al., “SOX17 is a Critical Factor in Maintaining Endothelial Function in Pulmonary Hypertension by an Exosome-mediated Autocrine Manner,” Advanced Science(Weinh) 10, no. 14 (2023): e2206139.

[81]

R. Walters, E. Vasilaki, J. Aman, et al., “SOX17 enhancer Variants Disrupt Transcription Factor Binding and enhancer Inactivity Drives Pulmonary Hypertension,” Circulation 147, no. 21 (2023): 1606–1621.

[82]

C. L. Welch and W. K. Chung, “Genetics and Other Omics in Pediatric Pulmonary Arterial Hypertension,” Chest 157, no. 5 (2020): 1287–1295.

[83]

C. L. Welch, E. D. Austin, and W. K. Chung, “Genes That Drive the Pathobiology of Pediatric Pulmonary Arterial Hypertension,” Pediatric Pulmonology 56, no. 3 (2021): 614–620.

[84]

N. Zhu, E. M. Swietlik, C. L. Welch, et al., “Rare Variant Analysis of 4241 Pulmonary Arterial Hypertension Cases from an International Consortium Implicates FBLN2, PDGFD, and Rare De Novo Variants in PAH,” Genome Medicine 13, no. 1 (2021): 80.

[85]

F. Taha and L. Southgate, “Molecular Genetics of Pulmonary Hypertension in Children,” Current Opinion in Genetics & Development 75 (2022): 101936.

[86]

N. Zhu, M. W. Pauciulo, C. L. Welch, et al., “Novel Risk Genes and Mechanisms Implicated by Exome Sequencing of 2572 Individuals with Pulmonary Arterial Hypertension,” Genome Medicine 11, no. 1 (2019): 69.

[87]

C. L. Welch, M. A. Aldred, S. Balachandar, et al., “Defining the Clinical Validity of Genes Reported to Cause Pulmonary Arterial Hypertension,” Genetics in Medicine 25, no. 11 (2023): 100925.

[88]

S. Hirose, Y. Hosoda, S. Furuya, T. Otsuki, and E. Ikeda, “Expression of Vascular Endothelial Growth Factor and Its Receptors Correlates Closely with Formation of the Plexiform Lesion in human Pulmonary Hypertension,” Pathology International 50, no. 6 (2000): 472–479.

[89]

T. D. Le Cras, D. H. Kim, N. E. Markham, and A. S. Abman, “Early Abnormalities of Pulmonary Vascular Development in the Fawn-Hooded Rat Raised at Denver’s Altitude,” American Journal of Physiology. Lung Cellular and Molecular Physiology 279, no. 2 (2000): L283–L291.

[90]

M. Riou, M. Canuet, M. R. Ghigna, et al., “First Histological Description of Pulmonary and Vascular Abnormalities of Pulmonary Hypertension Associated with KDR Pathogenic Variant,” European Respiratory Journal 60, no. 5 (2022): 2201197.

[91]

E. M. Swietlik, D. Greene, N. Zhu, et al., “Bayesian Inference Associates Rare KDR Variants with Specific Phenotypes in Pulmonary Arterial Hypertension,” Circulation: Genomic and Medicine 14, no. 1 (2020): e003155.

[92]

C. Delaney, R. H. Wright, J. R. Tang, et al., “Lack of EC-SOD Worsens Alveolar and Vascular Development in a Neonatal Mouse Model of Bleomycin-induced Bronchopulmonary Dysplasia and Pulmonary Hypertension,” Pediatric Research 78, no. 6 (2015): 634–640.

[93]

M. Eyries, D. Montani, B. Girerd, et al., “Familial Pulmonary Arterial Hypertension by KDR Heterozygous Loss of Function,” European Respiratory Journal 55, no. 4 (2020): 1902165.

[94]

M. P. Winter, S. Sharma, J. Altmann, et al., “Interruption of Vascular Endothelial Growth Factor Receptor 2 Signaling Induces a Proliferative Pulmonary Vasculopathy and Pulmonary Hypertension,” Basic Research in Cardiology 115, no. 6 (2020): 58.

[95]

D. Liu and N. W. Morrell, “Genetics and the Molecular Pathogenesis of Pulmonary Arterial Hypertension,” Current Hypertension Reports 15, no. 6 (2013): 632–637.

[96]

P. Navas Tejedor, Tenorio Castaño, J. Palomino Doza, et al., “An Homozygous Mutation in KCNK3 Is Associated with an Aggressive Form of Hereditary Pulmonary Arterial Hypertension,” Clinical Genetics 91, no. 3 (2017): 453–457.

[97]

B. Girerd, J. Weatherald, D. Montani, and M. Humbert, “Heritable Pulmonary Hypertension: from Bench to Bedside,” European Respiratory Review 26, no. 145 (2017): 170037.

[98]

A. Maurac, É. Lardenois, M. Eyries, et al., “T-box Protein 4 Mutation Causing Pulmonary Arterial Hypertension and Lung Disease,” European Respiratory Journal 54, no. 2 (2019): 1900388.

[99]

I. Hernandez-Gonzalez, J. Tenorio, J. Palomino-Doza, et al., “Clinical Heterogeneity of Pulmonary Arterial Hypertension Associated with Variants in TBX4,” PLoS ONE 15, no. 4 (2020): e0232216.

[100]

M. G. Haarman, W. S Kerstjens-Frederikse, and R. M. F. Berger, “TBX4 variants and Pulmonary Diseases: Getting out of the ‘Box’” Current Opinion in Pulmonary Medicine 26, no. 3 (2020): 277–284.

[101]

P. Thoré, B. Girerd, X. Jaïs, et al., “Phenotype and Outcome of Pulmonary Arterial Hypertension Patients Carrying a TBX4 Mutation,” European Respiratory Journal 55, no. 5 (2020): 1902340.

[102]

E. D. Austin and C. G. Elliott, “TBX4 syndrome: a Systemic Disease Highlighted by Pulmonary Arterial Hypertension in Its Most Severe Form,” European Respiratory Journal 55, no. 5 (2020): 2000585.

[103]

L. M. van den Heuvel, S. M. A. Jansen, S. I. M. Alsters, et al., “Genetic Evaluation in a Cohort of 126 Dutch Pulmonary Arterial Hypertension Patients,” Genes 11, no. 10 (2020): 1191.

[104]

M. Prapa, M. Lago-Docampo, E. M. Swietlik, et al., “First Genotype-phenotype Study in TBX4 Syndrome: Gain-of-function Mutations Causative for Lung Disease,” American Journal of Respiratory and Critical Care Medicine 206, no. 12 (2022): 1522–1533.

[105]

P. Szafranski, S. Patrizi, T. Gambin, et al., “Diminished TMEM100 Expression in a Newborn with Acinar Dysplasia and a Novel TBX4 Variant: a Case Report,” Pediatric and Developmental Pathology 27, no. 3 (2024): 255–259.

[106]

P. Navas, J. Tenorio, C. A. Quezada, et al., “Molecular Analysis of BMPR2, TBX4, and KCNK3 and Genotype-phenotype Correlations in spanish Patients and Families with Idiopathic and Hereditary Pulmonary Arterial Hypertension,” Revista Española de Cardiología (English Edition) 69, no. 11 (2016): 1011–1019.

[107]

M. Nimmakayalu, H. Major, V. Sheffield, et al., “Microdeletion of 17q22q23.2 Encompassing TBX2 and TBX4 in a Patient with Congenital Microcephaly, Thyroid Duct Cyst, Sensorineural Hearing Loss, and Pulmonary Hypertension,” American Journal of Medical Genetics. Part A 155a, no. 2 (2011): 418–423.

[108]

M. Eyries and F. Soubrier, “Molecular Genetic Diagnosis of Pulmonary Arterial Hypertension: an Increased Complexity,” Revista Española de Cardiología (English Edition) 69, no. 11 (2016): 1003–1004.

[109]

P. Szafranski, Z. H Coban-Akdemir, R. Rupps, et al., “Phenotypic Expansion of TBX4 Mutations to Include Acinar Dysplasia of the Lungs,” American Journal of Medical Genetics. Part A 170, no. 9 (2016): 2440–2444.

[110]

F. O. Flanagan, A. M. Holtz, S. O. Vargas, et al., “An Intronic Variant in TBX4 in a Single family with Variable and Severe Pulmonary Manifestations,” NPJ Genomic Medicine 8, no. 1 (2023): 7.

[111]

W. S Kerstjens-Frederikse, E. M. Bongers, M. T. Roofthooft, et al., “TBX4 mutations (small patella syndrome) Are Associated with Childhood-onset Pulmonary Arterial Hypertension,” Journal of Medical Genetics 50, no. 8 (2013): 500–5006.

[112]

M. Eyries, D. Montani, S. Nadaud, et al., “Widening the Landscape of Heritable Pulmonary Hypertension Mutations in Paediatric and Adult Cases,” European Respiratory Journal 53, no. 3 (2019): 1801371.

[113]

S. M. Tsoi, K. Jones, E. Colglazier, et al., “Persistence of Persistent Pulmonary Hypertension of the Newborn: a Case of De Novo TBX4 Variant,” Pulmonary Circulation 12, no. 3 (2022): e12108.

[114]

A. Cruz-Utrilla, N. Gallego-Zazo, J. A. Tenorio-Castaño, et al., “Clinical Implications of the Genetic Background in Pediatric Pulmonary Arterial Hypertension: Data from the Spanish REHIPED Registry,” International Journal of Molecular Sciences 23, no. 18 (2022): 10433.

[115]

H. V. Neves da Silva, J. P. Weinman, E. K. Englund, R. R. Deterding, D. D. Ivy, and L. P. Browne, “Computed Tomographic Findings in TBX4 Mutation: a Common Cause of Severe Pulmonary Artery Hypertension in Children,” Pediatric Radiology 54, no. 2 (2024): 199–207.

[116]

C. Maddaloni, S. Ronci, D. U. De Rose, et al., “Neonatal Persistent Pulmonary Hypertension Related to a Novel TBX4 Mutation: Case Report and Review of the Literature,” Italian Journal of Pediatrics 50, no. 1 (2024): 41.

[117]

M. Levy, M. Eyries, I. Szezepanski, et al., “Genetic Analyses in a Cohort of Children with Pulmonary Hypertension,” European Respiratory Journal 48, no. 4 (2016): 1118–1126.

[118]

C. Galambos, M. P. Mullen, J. T. Shieh, et al., “Phenotype Characterisation of TBX4 Mutation and Deletion Carriers with Neonatal and Paediatric Pulmonary Hypertension,” European Respiratory Journal 54, no. 2 (2019): 1801965.

[119]

J. A. Karolak, P. Szafranski, D. Kilner, et al., “Heterozygous CTNNB1 and TBX4 Variants in a Patient with Abnormal Lung Growth, Pulmonary Hypertension, Microcephaly, and Spasticity,” Clinical Genetics 96, no. 4 (2019): 366–370.

[120]

J. A. Karolak, T. Gambin, E. M. Honey, T. Slavik, E. Popek, and P. Stankiewicz, “A De Novo 2.2 Mb Recurrent 17q23.1q23.2 Deletion Unmasks Novel Putative Regulatory Non-coding SNVs Associated with Lethal Lung Hypoplasia and Pulmonary Hypertension: A Case Report,” BMC Medical Genomics 13, no. 1 (2020): 34.

[121]

C. Yin, K. Li, Y. Yu, et al., “Genome-wide Association Study Identifies Loci and Candidate Genes for Non-idiopathic Pulmonary Hypertension in Eastern Chinese Han Population,” BMC Pulmonary Medicine 18, no. 1 (2018): 158.

[122]

O. K. Abou Hassan, W. Haidar, G. Nemer, H. Skouri, F. Haddad, and I. BouAkl, “Clinical and Genetic Characteristics of Pulmonary Arterial Hypertension in Lebanon,” BMC Medical Genetics 19, no. 1 (2018): 89.

[123]

Y. Yoshida, K. Uchida, K. Kodo, et al., “Genetic and Functional Analyses of TBX4 Reveal Novel Mechanisms Underlying Pulmonary Arterial Hypertension,” Journal of Molecular and Cellular Cardiology 171 (2022): 105–116.

[124]

P. Chelladurai, C. Kuenne, A. Bourgeois, et al., “Epigenetic Reactivation of Transcriptional Programs Orchestrating Fetal Lung Development in human Pulmonary Hypertension,” Science Translational Medicine 14, no. 648 (2022): eabe5407.

[125]

E. D. Austin and J. E. Loyd, “The Genetics of Pulmonary Arterial Hypertension,” Circulation Research 115, no. 1 (2014): 189–202.

[126]

B. Girerd, D. Montani, X. Jaïs, et al., “Genetic Counselling in a National Referral Centre for Pulmonary Hypertension,” European Respiratory Journal 47, no. 2 (2016): 541–552.

[127]

L. Liang, G. Ma, K. Chen, et al., “EIF2AK4 mutation in Pulmonary Veno-occlusive Disease: a Case Report and Review of the Literature,” Medicine(Baltimore) 95, no. 39 (2016): e5030.

[128]

J. Song, C. A. Eichstaedt, R. R. Viales, et al., “Identification of Genetic Defects in Pulmonary Arterial Hypertension by a New Gene Panel Diagnostic Tool,” Clinical Science 130, no. 22 (2016): 2043–2052.

[129]

D. H. Best, K. L. Sumner, B. P. Smith, et al., “EIF2AK4 mutations in Patients Diagnosed with Pulmonary Arterial Hypertension,” Chest 151, no. 4 (2017): 821–828.

[130]

H. Yang, Q. Zeng, Y. Ma, et al., “Genetic Analyses in a Cohort of 191 Pulmonary Arterial Hypertension Patients,” Respiratory Research 19, no. 1 (2018): 87.

[131]

H. S. Zhang, Q. Liu, C. M. Piao, et al., “Genotypes and Phenotypes of chinese Pediatric Patients with Idiopathic and Heritable Pulmonary Arterial Hypertension-a Single-center Study,” Canadian Journal of Cardiology 35, no. 12 (2019): 1851–1856.

[132]

Q. Zeng, H. Yang, B. Liu, et al., “Clinical Characteristics and Survival of Chinese Patients Diagnosed with Pulmonary Arterial Hypertension Who Carry BMPR2 or EIF2KAK4 Variants,” BMC Pulmonary Medicine 20, no. 1 (2020): 150.

[133]

M. Eyries, D. Montani, B. Girerd, et al., “EIF2AK4 mutations Cause Pulmonary Veno-occlusive Disease, a Recessive Form of Pulmonary Hypertension,” Nature Genetics 46, no. 1 (2014): 65–69.

[134]

P. Laveneziana, D. Montani, P. Dorfmüller, et al., “Mechanisms of Exertional Dyspnoea in Pulmonary Veno-occlusive Disease with EIF2AK4 Mutations,” European Respiratory Journal 44, no. 4 (2014): 1069–1072.

[135]

M. L. den Toom, G. Grinwis, R. J. van Suylen, et al., “Pulmonary Veno-occlusive Disease as a Cause of Severe Pulmonary Hypertension in a Dog,” Acta Veterinaria Scandinavica 60, no. 1 (2018): 78.

[136]

L. Liang, H. Su, X. Ma, and R. Zhang, “Good Response to PAH-targeted Drugs in a PVOD Patient Carrying Biallelic EIF2AK4 Mutation,” Respiratory Research 19, no. 1 (2018): 192.

[137]

J. Weatherald, P. Dorfmüller, F. Perros, et al., “Pulmonary Capillary Haemangiomatosis: a Distinct Entity?,” European Respiratory Review 29, no. 156 (2020): 190168.

[138]

J. E. Park, S. A. Chang, S. Y. Jang, K. S. Lee, D. K. Kim, and C. S. Ki, “Differential Diagnosis of Pulmonary Veno-occlusive Disease and/or Pulmonary Capillary Hemangiomatosis after Identification of Two Novel EIF2AK4 Variants by Whole-exome Sequencing,” Molecular Syndromology 14, no. 3 (2023): 254–257.

[139]

I. Oriaku, M. N. LeSieur, W. C. Nichols, R. Barrios, C. G. Elliott, and A. Frost, “A Novel BMPR2 Mutation with Widely Disparate Heritable Pulmonary Arterial Hypertension Clinical Phenotype,” Pulmonary Circulation 10, no. 3 (2020): 2045894020931315.

[140]

X. Zeng, F. Chen, A. Rathinasabapathy, T. Li, A. Adnan Ali Mohammed Mohammed, and Z. Yu, “Rapid Disease Progress in a PVOD Patient Carrying a Novel EIF(2)AK(4) Mutation: a Case Report,” BMC Pulmonary Medicine 20, no. 1 (2020): 186.

[141]

Y. Li, Y. Gu, C. Hong, Y. Deng, Z. Chen, and J. Jiang, “Pulmonary Capillary Hemangiomatosis in Chinese Patients without EIF2AK4 Mutations,” Pathology, Research and Practice 216, no. 9 (2020): 153100.

[142]

G. Treffel, A. Guillaumot, E. Gomez, et al., “Familial Pulmonary Veno-occlusive Disease with a Composite Biallelic Heterozygous EIF2AK4 Mutation,” Revue Des Maladies Respiratoires 37, no. 10 (2020): 823–828.

[143]

L. Zhang, Y. Wang, and R. Zhang, “Good Response to Pulmonary Arterial Hypertension-targeted Therapy in 2 Pulmonary Veno-occlusive Disease Patients: a Case Report,” Medicine(Baltimore) 100, no. 41 (2021): e27334.

[144]

L. M. Kimmig, M. R. Stutz, A. N. Husain, and R. Bag, “Identification of a Novel EIF2AK Variant and Genetics-assisted Approach to Diagnosis of Pulmonary Capillary Hemangiomatosis,” Lung 200, no. 2 (2022): 217–219.

[145]

M. Katrien, S. Dieter, D. Anke, D. Amelie, M. De Pauw, and D. Eric, “Case Report: Pulmonary Veno-occlusive Disease: a Rare but Fatal Cause of Pulmonary Hypertension in a Patient Following Allogeneic Stem Cell Transplantation,” Acta Clinica Belgica 77, no. 3 (2022): 697–702.

[146]

B. Lechartier, B. Girerd, M. Eyries, A. Beurnier, M. Humbert, and D. Montani, “Screening for Pulmonary Veno-occlusive Disease in Heterozygous EIF2AK4 Variant Carriers,” European Respiratory Journal 60, no. 2 (2022): 2200760.

[147]

C. Zhang, Q. Du, S. Wang, and R. Zhang, “A Rare Compound Heterozygous EIF2AK4 Mutation in Pulmonary Veno-occlusive Disease,” BMC Pulmonary Medicine 22, no. 1 (2022): 455.

[148]

M. Mahdavi, H. Shahzadi, M. Ghasemnezhad, and S. Hoseinzadeh Moghadam, “Pulmonary Capillary Hemangiomatosis as a Rare Underlying Cause of Primary Pulmonary Hypertension: a Case Report in an Adolescent,” Iranian Journal of Medical Sciences 49, no. 6 (2024): 394–398.

[149]

C. Hadinnapola, M. Bleda, M. Haimel, et al., “Phenotypic Characterization of EIF2AK4 Mutation Carriers in a Large Cohort of Patients Diagnosed Clinically with Pulmonary Arterial Hypertension,” Circulation 136, no. 21 (2017): 2022–2033.

[150]

O. K. Abou Hassan, W. Haidar, M. Arabi, et al., “Novel EIF2AK4 Mutations in Histologically Proven Pulmonary Capillary Hemangiomatosis and Hereditary Pulmonary Arterial Hypertension,” BMC Medical Genetics 20, no. 1 (2019): 176.

[151]

J. Gómez, J. R. Reguero, C. Alvarez, et al., “A Semiconductor Chip-based next Generation Sequencing Procedure for the Main Pulmonary Hypertension Genes,” Lung 193, no. 4 (2015): 571–574.

[152]

J. Tenorio, P. Navas, E. Barrios, et al., “A Founder EIF2AK4 Mutation Causes an Aggressive Form of Pulmonary Arterial Hypertension in Iberian Gypsies,” Clinical Genetics 88, no. 6 (2015): 579–583.

[153]

C. A. Eichstaedt, J. Song, N. Benjamin, et al., “EIF2AK4 mutation as “Second Hit” in Hereditary Pulmonary Arterial Hypertension,” Respiratory Research 17, no. 1 (2016): 141.

[154]

M. M. Zhu, J. Dai, Z. Dai, Y. Peng, and Y. Y. Zhao, “GCN2 kinase Activation Mediates Pulmonary Vascular Remodeling and Pulmonary Arterial Hypertension,” JCI Insight 9, no. 20 (2024): e177926.

[155]

M. S. Bohnen, L. Ma, N. Zhu, et al., “Loss-of-function ABCC8 Mutations in Pulmonary Arterial Hypertension,” Circ Genom Precis Med 11, no. 10 (2018): e002087.

[156]

M. Lago-Docampo, J. Tenorio, I. Hernandez-Gonzalez, et al., “Characterization of Rare ABCC8 Variants Identified in Spanish Pulmonary Arterial Hypertension Patients,” Scientific Reports 10, no. 1 (2020): 15135.

[157]

H. Le Ribeuz, B. Masson, V. Capuano, et al., “SUR1 as a New Therapeutic Target for Pulmonary Arterial Hypertension,” American Journal of Respiratory Cell and Molecular Biology 66, no. 5 (2022): 539–554.

[158]

C. McClenaghan, K. V. Woo, and C. G. Nichols, “Pulmonary Hypertension and ATP-sensitive Potassium Channels,” Hypertension 74, no. 1 (2019): 14–22.

[159]

J. K. Tie and D. W. Stafford, “Structural and Functional Insights into Enzymes of the Vitamin K Cycle,” Journal of Thrombosis and Haemostasis 14, no. 2 (2016): 236–247.

[160]

J. Stenflo, P. Fernlund, W. Egan, and P. Roepstorff, “Vitamin K Dependent Modifications of Glutamic Acid Residues in Prothrombin,” PNAS 71, no. 7 (1974): 2730–2733.

[161]

L. Suleiman, C. Negrier, and H. Boukerche, “Protein S: a Multifunctional Anticoagulant Vitamin K-dependent Protein at the Crossroads of Coagulation, Inflammation, Angiogenesis, and Cancer,” Critical Reviews in Oncology/Hematology 88, no. 3 (2013): 637–654.

[162]

S. Cavaco, C. S. Viegas, M. S. Rafael, et al., “Gla-rich Protein Is Involved in the Cross-talk between Calcification and Inflammation in Osteoarthritis,” Cellular and Molecular Life Sciences 73, no. 5 (2016): 1051–1065.

[163]

S. Feng, S. E. Jacobsen, and W. Reik, “Epigenetic Reprogramming in Plant and Animal Development,” Science 330, no. 6004 (2010): 622–627.

[164]

G. H. Kim, J. J. Ryan, G. Marsboom, and S. L. Archer, “Epigenetic Mechanisms of Pulmonary Hypertension,” Pulmonary Circulation 1, no. 3 (2011): 347–356.

[165]

J. B. Huang, J. Liang, X. F. Zhao, W. S. Wu, and F. Zhang, “Epigenetics: Novel Mechanism of Pulmonary Hypertension,” Lung 191, no. 6 (2013): 601–610.

[166]

N. M. Al Aboud, C. Tupper, and I. Jialal, Genetics, epigenetic mechanism (Treasure Island, FL: StatPearls Publishing, 2025), Accessed January 28, 2025. https://www.ncbi.nlm.nih.gov/books/NBK532999/.

[167]

L. Liu, Y. Li, and T. O. Tollefsbol, “Gene-environment Interactions and Epigenetic Basis of human Diseases,” Current Issues in Molecular Biology 10, no. 1-2 (2008): 25–36.

[168]

J. P. Hamilton, “Epigenetics: Principles and Practice,” Digestive Diseases 29, no. 2 (2011): 130–135.

[169]

D. E. Handy, R. Castro, and J. Loscalzo, “Epigenetic Modifications: Basic Mechanisms and Role in Cardiovascular Disease,” Circulation 123, no. 19 (2011): 2145–2156.

[170]

S. L. Berger, T. Kouzarides, R. Shiekhattar, and A. Shilatifard, “An Operational Definition of Epigenetics,” Genes & Development 23, no. 7 (2009): 781–783.

[171]

Y. Jiang, S. Song, J. Liu, et al., “Epigenetic Regulation of Programmed Cell Death in Hypoxia-induced Pulmonary Arterial Hypertension,” Frontiers in Immunology 14 (2023): 1206452.

[172]

L. Zhang, Q. Lu, and C. Chang, “Epigenetics in Health and Disease,” Advances in Experimental Medicine and Biology 1253 (2020): 3–55.

[173]

A. Baccarelli, M. Rienstra, and E. J. Benjamin, “Cardiovascular Epigenetics: Basic Concepts and Results from Animal and human Studies,” Circulation: Cardiovascular Genetics 3, no. 6 (2010): 567–573.

[174]

L. Sun, H. Zhang, and P. Gao, “Metabolic Reprogramming and Epigenetic Modifications on the Path to Cancer,” Protein Cell 13, no. 12 (2022): 877–919.

[175]

G. Gao, A. Chen, J. Gong, et al., “Comprehensive Analyses of m6A RNA Methylation Patterns and Related Immune Microenvironment in Idiopathic Pulmonary Arterial Hypertension,” Frontiers in Genetics 14 (2023): 1222368.

[176]

Y. Xia, Y. Zhang, J. Huang, et al., “N6-methyladenosine Modifications in Pulmonary Hypertension,” Pharmacology 108, no. 6 (2023): 497–503.

[177]

S. S. Zhao, J. Liu, Q. C. Wu, and X. L. Zhou, “Role of Histone Lactylation Interference RNA M(6)A Modification and Immune Microenvironment Homeostasis in Pulmonary Arterial Hypertension,” Frontiers in Cell and Developmental Biology 11 (2023): 1268646.

[178]

T. Huang, Y. Zeng, Y. Yang, et al., “Comprehensive Analysis of M(6)A Methylomes in Idiopathic Pulmonary Arterial Hypertension,” Epigenetics 18, no. 1 (2023): 2242225.

[179]

Z. Wang, Y. X. Zhang, J. Z. Shi, et al., “RNA m6A Methylation and Regulatory Proteins in Pulmonary Arterial Hypertension,” Hypertension Research 47, no. 5 (2024): 1273–1287.

[180]

Y. Qin, Y. Qiao, L. Li, et al., “The M(6)A Methyltransferase METTL3 Promotes Hypoxic Pulmonary Arterial Hypertension,” Life Sciences 274 (2021): 119366.

[181]

K. Kang, C. Sun, H. Li, et al., “N6-methyladenosine-driven miR-143/145-KLF4 Circuit Orchestrates the Phenotypic Switch of Pulmonary Artery Smooth Muscle Cells,” Cellular and Molecular Life Sciences 81, no. 1 (2024): 256.

[182]

S. Xu, X. Xu, Z. Zhang, L. Yan, L. Zhang, and L. Du, “The Role of RNA M(6)A Methylation in the Regulation of Postnatal Hypoxia-induced Pulmonary Hypertension,” Respiratory Research 22, no. 1 (2021): 121.

[183]

K. Kang, J. Xiang, X. Zhang, et al., “N6-methyladenosine Modification of KLF2 May Contribute to Endothelial-to-mesenchymal Transition in Pulmonary Hypertension,” Cellular & Molecular Biology Letters 29, no. 1 (2024): 69.

[184]

Y. Jiang, H. Liu, R. Shi, et al., “Methyltransferase-Like 3-mediated N6-methyladenosine RNA Methylation Regulates Hypoxia-induced Pulmonary Arterial Smooth Muscle Cell Pyroptosis by Targeting PTEN,” Journal of the American Heart Association 13, no. 19 (2024): e034470.

[185]

L. Hu, J. Wang, H. Huang, et al., “YTHDF1 regulates Pulmonary Hypertension through Translational Control of MAGED1,” American Journal of Respiratory and Critical Care Medicine 203, no. 9 (2021): 1158–1172.

[186]

T. Kang, L. Liu, F. Tan, et al., “Inhibition of YTHDF1 Prevents Hypoxia-induced Pulmonary Artery Smooth Muscle Cell Proliferation by Regulating Foxm1 Translation in an m6A-dependent Manner,” Experimental Cell Research 424, no. 2 (2023): 113505.

[187]

X. Wang, Q. Li, S. He, et al., “LncRNA FENDRR with m6A RNA Methylation Regulates Hypoxia-induced Pulmonary Artery Endothelial Cell Pyroptosis by Mediating DRP1 DNA Methylation,” Molecular Medicine 28, no. 1 (2022): 126.

[188]

J. Wang, Y. Shen, Y. Zhang, et al., “Smooth Muscle Ythdf2 Abrogation Ameliorates Pulmonary Vascular Remodeling by Regulating Myadm Transcript Stability,” Hypertension 81, no. 8 (2024): 1785–1798.

[189]

N. Gu, Y. Shen, Y. He, et al., “Loss of m6A Demethylase ALKBH5 Alleviates Hypoxia-induced Pulmonary Arterial Hypertension via Inhibiting Cyp1a1 mRNA Decay,” Journal of Molecular and Cellular Cardiology 194 (2024): 16–31.

[190]

J. Zhang, W. Q. Huang, Y. R. Zhang, et al., “Upregulation of eIF2α by M(6)A Modification Accelerates the Proliferation of Pulmonary Artery Smooth Muscle Cells in MCT-induced Pulmonary Arterial Hypertension Rats,” Journal of Cardiovascular Translational Research 17, no. 3 (2024): 598–608.

[191]

B. Jin, Y. Li, and K. D. Robertson, “DNA Methylation: Superior or Subordinate in the Epigenetic Hierarchy?,” Genes & Cancer 2, no. 6 (2011): 607–617.

[192]

M. Paulsen and A. C. Ferguson-Smith, “DNA Methylation in Genomic Imprinting, Development, and Disease,” Journal of Pathology 195, no. 1 (2001): 97–110.

[193]

K. D. Robertson, “DNA Methylation and human Disease,” Nature Reviews Genetics 6, no. 8 (2005): 597–610.

[194]

I. M. Emon, R. Al-Qazazi, M. J. Rauh, and S. L. Archer, “The Role of Clonal Hematopoiesis of Indeterminant Potential and DNA (Cytosine-5)-methyltransferase Dysregulation in Pulmonary Arterial Hypertension and Other Cardiovascular Diseases,” Cells 12, no. 21 (2023): 2528.

[195]

R. M. Kohli and Y. Zhang, “TET Enzymes, TDG and the Dynamics of DNA Demethylation,” Nature 502, no. 7472 (2013): 472–479.

[196]

M. Bochtler, A. Kolano, and G. L. Xu, “DNA Demethylation Pathways: Additional Players and Regulators,” BioEssays 39, no. 1 (2017): 1–13.

[197]

A. Chakraborty and P. Viswanathan, “Methylation-demethylation Dynamics: Implications of Changes in Acute Kidney Injury,” Analytical Cellular Pathology (Amsterdam) 2018 (2018): 8764384.

[198]

G. Benincasa, B. A. Maron, O. Affinito, et al., “Association between Circulating CD4(+) T Cell Methylation Signatures of Network-oriented SOCS3 Gene and Hemodynamics in Patients Suffering Pulmonary Arterial Hypertension,” Journal of Cardiovascular Translational Research 16, no. 1 (2023): 17–30.

[199]

F. Perros, S. Cohen-Kaminsky, N. Gambaryan, et al., “Cytotoxic Cells and Granulysin in Pulmonary Arterial Hypertension and Pulmonary Veno-occlusive Disease,” American Journal of Respiratory and Critical Care Medicine 187, no. 2 (2013): 189–196.

[200]

S. Bonnet, E. D. Michelakis, C. J. Porter, et al., “An Abnormal Mitochondrial-hypoxia Inducible Factor-1alpha-Kv Channel Pathway Disrupts Oxygen Sensing and Triggers Pulmonary Arterial Hypertension in Fawn Hooded Rats: Similarities to human Pulmonary Arterial Hypertension,” Circulation 113, no. 22 (2006): 2630–2641.

[201]

S. L. Archer, G. Marsboom, G. H. Kim, et al., “Epigenetic Attenuation of Mitochondrial Superoxide Dismutase 2 in Pulmonary Arterial Hypertension: a Basis for Excessive Cell Proliferation and a New Therapeutic Target,” Circulation 121, no. 24 (2010): 2661–2671.

[202]

N. W. Morrell, M. A. Aldred, W. K. Chung, et al., “Genetics and Genomics of Pulmonary Arterial Hypertension,” European Respiratory Journal 53, no. 1 (2019): 1801899.

[203]

R. Quarck and F. Perros, “Rescuing BMPR2-driven Endothelial Dysfunction in PAH: a Novel Treatment Strategy for the Future?,” Stem Cell Investigation 4 (2017): 56.

[204]

D. Liu, Y. Yan, J. W. Chen, et al., “Hypermethylation of BMPR2 Promoter Occurs in Patients with Heritable Pulmonary Arterial Hypertension and Inhibits BMPR2 Expression,” American Journal of Respiratory and Critical Care Medicine 196, no. 7 (2017): 925–928.

[205]

G. Pousada, A. Baloira, and D. Valverde, “Methylation Analysis of the BMPR2 Gene Promoter Region in Patients with Pulmonary Arterial Hypertension,” Archivos De Bronconeumologia 52, no. 6 (2016): 293–298.

[206]

A. Ulrich, Y. Wu, H. Draisma, et al., “Blood DNA Methylation Profiling Identifies Cathepsin Z Dysregulation in Pulmonary Arterial Hypertension,” Nature Communications 15, no. 1 (2024): 330.

[207]

Q. Liu, Y. Yang, M. Wu, et al., “Hub Gene ELK3-mediated Reprogramming Lipid Metabolism Regulates Phenotypic Switching of Pulmonary Artery Smooth Muscle Cells to Develop Pulmonary Arterial Hypertension Induced by PM(2.5),” Journal of Hazardous Materials 465 (2024): 133190.

[208]

Q. Yang, Z. Lu, R. Ramchandran, L. D. Longo, and J. U. Raj, “Pulmonary Artery Smooth Muscle Cell Proliferation and Migration in Fetal Lambs Acclimatized to High-altitude Long-term Hypoxia: Role of Histone Acetylation,” American Journal of Physiology. Lung Cellular and Molecular Physiology 303, no. 11 (2012): L1001–L1010.

[209]

Z. Zhang, X. Luo, Y. Lv, et al., “Intrauterine Growth Restriction Programs Intergenerational Transmission of Pulmonary Arterial Hypertension and Endothelial Dysfunction via Sperm Epigenetic Modifications,” Hypertension 74, no. 5 (2019): 1160–1171.

[210]

P. Crosswhite and Z. Sun, “TNFα Induces DNA and Histone Hypomethylation and Pulmonary Artery Smooth Muscle Cell Proliferation Partly via Excessive Superoxide Formation,” Antioxidants (Basel) 13, no. 6 (2024): 677.

[211]

S. T. da Rocha and A. V. Gendrel, “The Influence of DNA Methylation on Monoallelic Expression,” Essays in Biochemistry 63, no. 6 (2019): 663–676.

[212]

M. A. Aldred, S. A. Comhair, M. Varella-Garcia, et al., “Somatic Chromosome Abnormalities in the Lungs of Patients with Pulmonary Arterial Hypertension,” American Journal of Respiratory and Critical Care Medicine 182, no. 9 (2010): 1153–1160.

[213]

B. L. Carman, S. Qin, D. N. Predescu, et al., “Dysregulation of the Long Noncoding RNA X-inactive-specific Transcript Expression in Male Patients with Pulmonary Arterial Hypertension,” American Journal of Pathology 194, no. 8 (2024): 1592–1606.

[214]

A. R. Cutter and J. J. Hayes, “A Brief Review of Nucleosome Structure,” Febs Letters 589, no. 20 Pt A (2015): 2914–2922.

[215]

L. Mariño-Ramírez, M. G. Kann, B. A. Shoemaker, and D. Landsman, “Histone Structure and Nucleosome Stability,” Expert Review of Proteomics 2, no. 5 (2005): 719–729.

[216]

S. G. Swygert and C. L. Peterson, “Chromatin Dynamics: Interplay between Remodeling Enzymes and Histone Modifications,” Biochimica Et Biophysica Acta 1839, no. 8 (2014): 728–736.

[217]

F. Andreoli and A. Del Rio, “Physicochemical Modifications of Histones and Their Impact on Epigenomics,” Drug Discovery Today 19, no. 9 (2014): 1372–1379.

[218]

V. Cavalieri, “The Expanding Constellation of Histone Post-translational Modifications in the Epigenetic Landscape,” Genes (Basel) 12, no. 10 (2021): 1596.

[219]

C. E. Barnes, D. M. English, and S. M. Cowley, “Acetylation & Co: an Expanding Repertoire of Histone Acylations Regulates Chromatin and Transcription,” Essays in Biochemistry 63, no. 1 (2019): 97–107.

[220]

E. L. Greer and Y. Shi, “Histone Methylation: a Dynamic Mark in Health, Disease and Inheritance,” Nature Reviews Genetics 13, no. 5 (2012): 343–357.

[221]

E. Gamen, W. Seeger, and S. S. Pullamsetti, “The Emerging Role of Epigenetics in Pulmonary Hypertension,” European Respiratory Journal 48, no. 3 (2016): 903–917.

[222]

R. C. P Luna, Y. de Oliveira, J. V. C. Lisboa, et al., “Insights on the Epigenetic Mechanisms Underlying Pulmonary Arterial Hypertension,” Brazilian Journal of Medical and Biological Research 51, no. 12 (2018): e7437.

[223]

P. Chelladurai, O. Boucherat, K. Stenmark, et al., “Targeting Histone Acetylation in Pulmonary Hypertension and Right Ventricular Hypertrophy,” British Journal of Pharmacology 178, no. 1 (2021): 54–71.

[224]

Y. Meng, H. Li, C. Liu, L. Zheng, and B. Shen, “Jumonji Domain-containing Protein family: the Functions beyond Lysine Demethylation,” Journal of Molecular Cell Biology 10, no. 4 (2018): 371–373.

[225]

Y. Li, X. Wei, R. Xiao, et al., “SMYD2-methylated PPARγ Facilitates Hypoxia-induced Pulmonary Hypertension by Activating Mitophagy,” Circulation Research 135, no. 1 (2024): 93–109.

[226]

D. Chen, Y. Yang, X. Cheng, et al., “Megakaryocytic Leukemia 1 Directs a Histone H3 Lysine 4 Methyltransferase Complex to Regulate Hypoxic Pulmonary Hypertension,” Hypertension 65, no. 4 (2015): 821–833.

[227]

Q. Yang, Z. Lu, D. Singh, and J. U. Raj, “BIX-01294 Treatment Blocks Cell Proliferation, Migration and Contractility in Ovine Foetal Pulmonary Arterial Smooth Muscle Cells,” Cell Proliferation 45, no. 4 (2012): 335–344.

[228]

S. A. Aljubran, R. Cox Jr, P. Tamarapu Parthasarathy, et al., “Enhancer of Zeste Homolog 2 Induces Pulmonary Artery Smooth Muscle Cell Proliferation,” PLoS ONE 7, no. 5 (2012): e37712.

[229]

Z. L. Shi, K. Fang, Z. H. Li, D. H. Ren, J. Y. Zhang, and J. Sun, “EZH2 inhibition Ameliorates Transverse Aortic Constriction-induced Pulmonary Arterial Hypertension in Mice,” Canadian Respiratory Journal 2018 (2018): 9174926.

[230]

N. Gambaryan, C. Meng, M. Humbert, I. Adcock, and S. Wort, “H3K27 histone Lysine Methylation as Potential Therapeutic Target in Pulmonary Arterial Hypertension,” European Respiratory Journal 42 (2013): P5157.

[231]

L. H. Pojoga, J. S. Williams, T. M. Yao, et al., “Histone Demethylase LSD1 Deficiency during High-salt Diet Is Associated with Enhanced Vascular Contraction, Altered NO-cGMP Relaxation Pathway, and Hypertension,” American Journal of Physiology. Heart and Circulatory Physiology 301, no. 5 (2011): H1862–H1871.

[232]

R. Marmorstein and M. M. Zhou, “Writers and Readers of Histone Acetylation: Structure, Mechanism, and Inhibition,” Cold Spring Harbor Perspectives in Biology 6, no. 7 (2014): a018762.

[233]

I. Cohen, E. Poręba, K. Kamieniarz, and R. Schneider, “Histone Modifiers in Cancer: Friends or Foes?,” Genes & Cancer 2, no. 6 (2011): 631–647.

[234]

B. M. Dancy and P. A. Cole, “Protein Lysine Acetylation by p300/CBP,” Chem. Rev. 115, no. 6 (2015): 2419–2452.

[235]

E. Seto and M. Yoshida, “Erasers of Histone Acetylation: the Histone Deacetylase Enzymes,” Cold Spring Harbor Perspectives in Biology 6, no. 4 (2014): a018713.

[236]

Y. Su, R. Tan, M. Sun, et al., “MiR-1249 on Endothelial Extracellular Vesicles Mediates Cigarette Smoke-induced Pulmonary Hypertension by Inhibiting HDAC10 (Histone Deacetylase 10)-NFκB (Nuclear Factor κB)-CaSR (Calcium-Sensing Receptor) Cascade,” Hypertension 79, no. 12 (2022): 2721–2732.

[237]

X. F. Xu, X. L. Ma, Z. Shen, X. L. Wu, F. Cheng, and L. Z. Du, “Epigenetic Regulation of the Endothelial Nitric Oxide Synthase Gene in Persistent Pulmonary Hypertension of the Newborn Rat,” Journal of Hypertension 28, no. 11 (2010): 2227–2235.

[238]

Y. K. Cho, G. H. Eom, H. J. Kee, et al., “Sodium Valproate, a Histone Deacetylase Inhibitor, but Not Captopril, Prevents Right Ventricular Hypertrophy in Rats,” Circulation Journal 74, no. 4 (2010): 760–770.

[239]

B. Lan, E. Hayama, N. Kawaguchi, Y. Furutani, and T. Nakanishi, “Therapeutic Efficacy of Valproic Acid in a Combined Monocrotaline and Chronic Hypoxia Rat Model of Severe Pulmonary Hypertension,” PLoS ONE 10, no. 1 (2015): e0117211.

[240]

G. Zurlo, J. Piquereau, M. Moulin, et al., “Sirtuin 1 Regulates Pulmonary Artery Smooth Muscle Cell Proliferation: Role in Pulmonary Arterial Hypertension,” Journal of Hypertension 36, no. 5 (2018): 1164–1177.

[241]

E. Nozik-Grayck, C. Woods, R. S. Stearman, et al., “Histone Deacetylation Contributes to Low Extracellular Superoxide Dismutase Expression in human Idiopathic Pulmonary Arterial Hypertension,” American Journal of Physiology. Lung Cellular and Molecular Physiology 311, no. 1 (2016): L124–L134.

[242]

P. Chelladurai, S. Dabral, S. R. Basineni, et al., “Isoform-specific Characterization of Class I Histone Deacetylases and Their Therapeutic Modulation in Pulmonary Hypertension,” Scientific Reports 10, no. 1 (2020): 12864.

[243]

O. Boucherat, S. Chabot, R. Paulin, et al., “HDAC6: A Novel Histone Deacetylase Implicated in Pulmonary Arterial Hypertension,” Scientific Reports 7, no. 1 (2017): 4546.

[244]

L. Zhao, C. N. Chen, N. Hajji, et al., “Histone Deacetylation Inhibition in Pulmonary Hypertension: Therapeutic Potential of Valproic Acid and Suberoylanilide Hydroxamic Acid,” Circulation 126, no. 4 (2012): 455–467.

[245]

H. J. Bogaard, S. Mizuno, A. A. Hussaini, et al., “Suppression of Histone Deacetylases Worsens Right Ventricular Dysfunction after Pulmonary Artery Banding in Rats,” American Journal of Respiratory and Critical Care Medicine 183, no. 10 (2011): 1402–1410.

[246]

E. S. Lander, “Initial Impact of the Sequencing of the human Genome,” Nature 470, no. 7333 (2011): 187–197.

[247]

Q. Jin, Z. Zhao, Q. Zhao, et al., “Long Noncoding RNAs: Emerging Roles in Pulmonary Hypertension,” Heart Failure Reviews 25, no. 5 (2020): 795–815.

[248]

R. W. Carthew and E. J. Sontheimer, “Origins and Mechanisms of miRNAs and siRNAs,” Cell 136, no. 4 (2009): 642–655.

[249]

D. P. Bartel, “MicroRNAs: Genomics, Biogenesis, Mechanism, and Function,” Cell 116, no. 2 (2004): 281–297.

[250]

V. Ambros, B. Bartel, D. P. Bartel, et al., “A Uniform System for microRNA Annotation,” Rna 9, no. 3 (2003): 277–279.

[251]

M. Lu, Q. Zhang, M. Deng, et al., “An Analysis of human microRNA and Disease Associations,” PLoS ONE 3, no. 10 (2008): e3420.

[252]

T. Bertero, Y. Lu, S. Annis, et al., “Systems-level Regulation of microRNA Networks by miR-130/301 Promotes Pulmonary Hypertension,” Journal of Clinical Investigation 124, no. 8 (2014): 3514–3528.

[253]

L. Deng, F. J. Blanco, H. Stevens, et al., “MicroRNA-143 Activation Regulates Smooth Muscle and Endothelial Cell Crosstalk in Pulmonary Arterial Hypertension,” Circulation Research 117, no. 10 (2015): 870–883.

[254]

L. C. Huber, S. Ulrich, C. Leuenberger, et al., “Featured Article: MicroRNA-125a in Pulmonary Hypertension: Regulator of a Proliferative Phenotype of Endothelial Cells,” Experimental Biology and Medicine (Maywood, N.J.) 240, no. 12 (2015): 1580–1589.

[255]

P. Caruso, M. R. MacLean, R. Khanin, et al., “Dynamic Changes in Lung microRNA Profiles during the Development of Pulmonary Hypertension due to Chronic Hypoxia and Monocrotaline,” Arteriosclerosis, Thrombosis, and Vascular Biology 30, no. 4 (2010): 716–723.

[256]

A. S. I Ahmed, A. B. Blood, and L. Zhang, “MicroRNA-210 Mediates Hypoxia-induced Pulmonary Hypertension by Targeting Mitochondrial Bioenergetics and mtROS Flux,” Acta Physiol (Oxf) 240, no. 9 (2024): e14212.

[257]

B. Wang, Y. Xu, Y. Huang, et al., “miR-210-5p Promotes Pulmonary Hypertension by Blocking ATP2A2,” Cardiovascular Drugs and Therapy (2024). Online ahead of print.

[258]

T. Bertero, K. Cottrill, A. Krauszman, et al., “The microRNA-130/301 family Controls Vasoconstriction in Pulmonary Hypertension,” Journal of Biological Chemistry 290, no. 4 (2015): 2069–2085.

[259]

M. Brock, M. Trenkmann, R. E. Gay, et al., “Interleukin-6 Modulates the Expression of the Bone Morphogenic Protein Receptor Type II through a Novel STAT3–microRNA Cluster 17/92 Pathway,” Circulation Research 104, no. 10 (2009): 1184–1191.

[260]

W. Zhang, Y. Qi, and B. Wu, “MicroRNA-146-5p Promotes Pulmonary Artery Endothelial Cell Proliferation under Hypoxic Conditions through Regulating USP3,” Disease Markers 2021, no. 1 (2021): 3668422.

[261]

A. Courboulin, R. Paulin, N. J. Giguère, et al., “Role for miR-204 in human Pulmonary Arterial Hypertension,” Journal of Experimental Medicine 208, no. 3 (2011): 535–548.

[262]

Z. Cai, J. Li, Q. Zhuang, et al., “MiR-125a-5p Ameliorates Monocrotaline-induced Pulmonary Arterial Hypertension by Targeting the TGF-β1 and IL-6/STAT3 Signaling Pathways,” Experimental & Molecular Medicine 50, no. 4 (2018): 1–11.

[263]

L. Luo, L. Xiao, G. Lian, H. Wang, and L. J. A. Xie, “miR-125a-5p Inhibits Glycolysis by Targeting Hexokinase-II to Improve Pulmonary Arterial Hypertension,” Aging (Albany NY) 12, no. 10 (2020): 9014.

[264]

G. Zhu, W. Zhang, Y. Liu, and S. Wang, “miR-371b-5p Inhibits Endothelial Cell Apoptosis in Monocrotaline-Induced Pulmonary Arterial Hypertension via PTEN/PI3K/Akt Signaling Pathways,” Molecular Medicine Reports 18, no. 6 (2018): 5489–5501.

[265]

J. Kim, Y. Kang, Y. Kojima, et al., “An Endothelial Apelin-FGF Link Mediated by miR-424 and miR-503 Is Disrupted in Pulmonary Arterial Hypertension,” Nature Medicine 19, no. 1 (2013): 74–82.

[266]

C. Ma, Q. Xu, S. Huang, et al., “The HIF-1α/miR-26a-5p/PFKFB3/ULK1/2 Axis Regulates Vascular Remodeling in Hypoxia-induced Pulmonary Hypertension by Modulation of Autophagy,” Faseb Journal 37, no. 5 (2023): e22906.

[267]

Y. Qin, B. Zhu, L. Li, et al., “Overexpressed lncRNA AC068039.4 Contributes to Proliferation and Cell Cycle Progression of Pulmonary Artery Smooth Muscle Cells via Sponging miR-26a-5p/TRPC6 in Hypoxic Pulmonary Arterial Hypertension,” Shock (Augusta, Ga.) 55, no. 2 (2021): 244–255.

[268]

S. Cantoni, M. Galletti, F. Zambelli, et al., “Sodium Butyrate Inhibits Platelet-derived Growth Factor-induced Proliferation and Migration in Pulmonary Artery Smooth Muscle Cells through A Kt Inhibition,” Febs Journal 280, no. 9 (2013): 2042–2055.

[269]

C. P. Ponting, P. L. Oliver, and W. J. C. Reik, “Evolution and Functions of Long Noncoding RNAs,” Cell 136, no. 4 (2009): 629–641.

[270]

P. Ji, S. Diederichs, W. Wang, et al., “MALAT-1, a Novel Noncoding RNA, and Thymosin beta4 Predict Metastasis and Survival in Early-stage Non-small Cell Lung Cancer,” Oncogene 22, no. 39 (2003): 8031–8041.

[271]

Y. Zhuo, Q. Zeng, P. Zhang, G. Li, Q. Xie, and Y. Cheng, “Functional Polymorphism of lncRNA MALAT1 Contributes to Pulmonary Arterial Hypertension Susceptibility in Chinese People,” Clinical Chemistry and Laboratory Medicine 55, no. 1 (2017): 38–46.

[272]

A. Gorelova, M. Berman, and I. Al Ghouleh, “Endothelial-to-mesenchymal Transition in Pulmonary Arterial Hypertension,” Antioxidants & Redox Signaling 34, no. 12 (2021): 891–914.

[273]

B. Ranchoux, F. Antigny, C. Rucker-Martin, et al., “Endothelial-to-mesenchymal Transition in Pulmonary Hypertension,” Circulation 131, no. 11 (2015): 1006–1018.

[274]

T. Suzuki, E. J. Carrier, M. H. Talati, et al., “Isolation and Characterization of Endothelial-to-mesenchymal Transition Cells in Pulmonary Arterial Hypertension,” American Journal of Physiology Lung Cellular and Molecular Physiology 314, no. 1 (2018): L118–L126.

[275]

K. R. Zahid, U. Raza, J. Chen, U. J. Raj, and D. Gou, “Pathobiology of Pulmonary Artery Hypertension: Role of Long Non-coding RNAs,” Cardiovascular Research 116, no. 12 (2020): 1937–1947.

[276]

Y. Xiang, Y. Zhang, Y. Tang, and Q. Li, “MALAT1 Modulates TGF-β1-Induced Endothelial-to-Mesenchymal Transition through Downregulation of miR-145,” Cellular Physiology and Biochemistry 42, no. 1 (2017): 357–372.

[277]

H. Li, Q. Zhao, L. Chang, et al., “LncRNA MALAT1 Modulates Ox-LDL Induced EndMT through the Wnt/β-catenin Signaling Pathway,” Lipids Health Disease 18, no. 1 (2019): 62.

[278]

D. Wang, H. Xu, B. Wu, et al., “Long Non-Coding RNA MALAT1 Sponges miR-124-3p.1/KLF5 to Promote Pulmonary Vascular Remodeling and Cell Cycle Progression of Pulmonary Artery Hypertension,” International Journal of Molecular Medicine 44, no. 3 (2019): 871–884.

[279]

X. Li, Y. He, Y. Xu, et al., “KLF5 mediates Vascular Remodeling via HIF-1α in Hypoxic Pulmonary Hypertension,” American Journal of Physiology Lung Cellular and Molecular Physiology 310, no. 4 (2016): L299–L310.

[280]

A. Courboulin, V. L. Tremblay, M. Barrier, et al., “Krüppel-Like Factor 5 Contributes to Pulmonary Artery Smooth Muscle Proliferation and Resistance to Apoptosis in human Pulmonary Arterial Hypertension,” Respiratory Research 12, no. 1 (2011): 128.

[281]

S. Ghafouri-Fard, M. Esmaeili, and M. Taheri, “H19 lncRNA: Roles in Tumorigenesis,” Biomedicine & Pharmacotherapy 123 (2020): 109774.

[282]

J. Omura, K. Habbout, T. Shimauchi, et al., “Identification of Long Noncoding RNA H19 as a New Biomarker and Therapeutic Target in Right Ventricular Failure in Pulmonary Arterial Hypertension,” Circulation 142, no. 15 (2020): 1464–1484.

[283]

H. Su, X. Xu, C. Yan, et al., “LncRNA H19 Promotes the Proliferation of Pulmonary Artery Smooth Muscle Cells through AT(1)R via Sponging Let-7b in Monocrotaline-induced Pulmonary Arterial Hypertension,” Respiratory Research 19, no. 1 (2018): 254.

[284]

A. C. Montezano, A. Nguyen Dinh Cat, F. J. Rios, and R. M. Touyz, “Angiotensin II and Vascular Injury,” Current Hypertension Reports 16, no. 6 (2014): 431.

[285]

X. Yu, J. Huang, X. Liu, et al., “LncRNAH19 acts as a ceRNA of Let-7 G to Facilitate Endothelial-to-mesenchymal Transition in Hypoxic Pulmonary Hypertension via Regulating TGF-β Signalling Pathway,” Respiratory Research 25, no. 1 (2024): 270.

[286]

M.-H. Li, X. Liu, Y.-L. Xie, et al., “Sodium Butyrate Alleviates Right Ventricular Hypertrophy in Pulmonary Arterial Hypertension by Inhibiting H19 and Affecting the Activation of Let-7G-5p/IGF1 Receptor/ERK,” European Journal of Pharmacology 965 (2024): 176315.

[287]

R. Wang, S. Zhou, P. Wu, et al., “Identifying Involvement of H19-miR-675-3p-IGF1R and H19-miR-200a-PDCD4 in Treating Pulmonary Hypertension with Melatonin,” Mol Ther Nucleic Acids 13 (2018): 44–54.

[288]

C. Guo, Y. Qi, J. Qu, L. Gai, Y. Shi, and C. Yuan, “Pathophysiological Functions of the lncRNA TUG1,” Current Pharmaceutical Design 26, no. 6 (2020): 688–700.

[289]

T. L. Young, T. Matsuda, and C. L. Cepko, “The Noncoding RNA Taurine Upregulated Gene 1 Is Required for Differentiation of the Murine Retina,” Current Biology 15, no. 6 (2005): 501–512.

[290]

S. Wang, W. Cao, S. Gao, et al., “TUG1 regulates Pulmonary Arterial Smooth Muscle Cell Proliferation in Pulmonary Arterial Hypertension,” Canadian Journal of Cardiology 35, no. 11 (2019): 1534–1545.

[291]

S. Bonnet, O. Boucherat, S. Provencher, and R. Paulin, “Early Evidence for the Role of lncRNA TUG1 in Vascular Remodelling in Pulmonary Hypertension,” Canadian Journal of Cardiology 35, no. 11 (2019): 1433–1434.

[292]

L. Yang, H. Liang, L. Shen, Z. Guan, and X. Meng, “LncRNA Tug1 Involves in the Pulmonary Vascular Remodeling in Mice with Hypoxic Pulmonary Hypertension via the microRNA-374c-mediated Foxc1,” Life Sciences 237 (2019): 116769.

[293]

C. Yu, L. Li, F. Xie, et al., “LncRNA TUG1 Sponges miR-204-5p to Promote Osteoblast Differentiation through Upregulating Runx2 in Aortic Valve Calcification,” Cardiovascular Research 114, no. 1 (2018): 168–179.

[294]

J. Liu, Y. Liu, F. Wang, and M. Liang, “MiR-204: Molecular Regulation and Role in Cardiovascular and Renal Diseases,” Hypertension 78, no. 2 (2021): 270–281.

[295]

Z. Lv, R. Jiang, X. Hu, et al., “Dysregulated lncRNA TUG1 in Different Pulmonary Artery Cells under Hypoxia,” Annals of translational medicine 9, no. 10 (2021): 879.

[296]

R. L. Strausberg, E. A. Feingold, L. H. Grouse, et al., “Generation and Initial Analysis of More than 15, 000 Full-length human and Mouse cDNA Sequences,” PNAS 99, no. 26 (2002): 16899–16903.

[297]

Y. Jiang, B. Hei, W. Hao, et al., “Clinical Value of lncRNA SOX2-OT in Pulmonary Arterial Hypertension and Its Role in Pulmonary Artery Smooth Muscle Cell Proliferation, Migration, Apoptosis, and Inflammatory,” Heart & Lung 55 (2022): 16–23.

[298]

Y. Yao, H. Li, X. Da, et al., “SUMOylation of Vps34 by SUMO1 Promotes Phenotypic Switching of Vascular Smooth Muscle Cells by Activating Autophagy in Pulmonary Arterial Hypertension,” Pulmonary Pharmacology & Therapeutics 55 (2019): 38–49.

[299]

Y. Jiang, J. Wang, H. Tian, et al., “Increased SUMO-1 Expression in Response to Hypoxia: Interaction with HIF-1α in Hypoxic Pulmonary Hypertension,” International Journal of Molecular Medicine 36, no. 1 (2015): 271–281.

[300]

A. Leung, C. Trac, W. Jin, et al., “Novel Long Noncoding RNAs Are Regulated by Angiotensin II in Vascular Smooth Muscle Cells,” Circulation Research 113, no. 3 (2013): 266–278.

[301]

G. Chen, S. Huang, F. Song, Y. Zhou, and X. He, “Lnc-Ang362 Is a Pro-fibrotic Long Non-coding RNA Promoting Cardiac Fibrosis after Myocardial Infarction by Suppressing Smad7,” Archives of Biochemistry and Biophysics 685 (2020): 108354.

[302]

H. Wang, R. Qin, and Y. Cheng, “LncRNA-Ang362 Promotes Pulmonary Arterial Hypertension by Regulating miR-221 and miR-222,” Shock (Augusta, Ga.) 53, no. 6 (2020): 723–729.

[303]

X. X. Zhu, Y. W. Yan, D. Chen, et al., “Long Non-coding RNA HoxA-AS3 Interacts with EZH2 to Regulate Lineage Commitment of Mesenchymal Stem Cells,” Oncotarget 7, no. 39 (2016): 63561–63570.

[304]

D. Duboule, “The Rise and Fall of Hox Gene Clusters,” Development (Cambridge, England) 134, no. 14 (2007): 2549–2560.

[305]

H. Zhang, Y. Liu, L. Yan, et al., “Long Noncoding RNA Hoxaas3 Contributes to Hypoxia-induced Pulmonary Artery Smooth Muscle Cell Proliferation,” Cardiovascular Research 115, no. 3 (2019): 647–657.

[306]

Z. K. Li, L. F. Gao, X. A. Zhu, and D. K. Xiang, “LncRNA HOXA-AS3 Promotes the Progression of Pulmonary Arterial Hypertension through Mediation of miR-675-3p/PDE5A Axis,” Biochemical Genetics 59, no. 5 (2021): 1158–1172.

[307]

B. A. Cockrill and A. B. Waxman, “Phosphodiesterase-5 Inhibitors,” Handbook of Experimental Pharmacology. 218 (2013): 229–255.

[308]

H. Barnes, Z. Brown, A. Burns, and T. Williams, “Phosphodiesterase 5 Inhibitors for Pulmonary Hypertension,” Cochrane Database of Systematic Reviews (Online) 1, no. 1 (2019): Cd012621.

[309]

T. Weirick, G. Militello, Y. Ponomareva, et al., “Logic Programming to Infer Complex RNA Expression Patterns from RNA-seq Data,” Brief Bioinformation 19, no. 2 (2018): 199–209.

[310]

K. Jandl, H. Thekkekara Puthenparampil, L. M. Marsh, et al., “Long Non-coding RNAs Influence the Transcriptome in Pulmonary Arterial Hypertension: the Role of PAXIP1-AS1,” Journal of Pathology 247, no. 3 (2019): 357–370.

[311]

C. Veith, L. M. Marsh, M. Wygrecka, et al., “Paxillin Regulates Pulmonary Arterial Smooth Muscle Cell Function in Pulmonary Hypertension,” American Journal of Pathology 181, no. 5 (2012): 1621–1633.

[312]

R. Song, S. Lei, S. Yang, and S. J. Wu, “LncRNA PAXIP1-AS1 Fosters the Pathogenesis of Pulmonary Arterial Hypertension via ETS1/WIPF1/RhoA Axis,” Journal of Cellular and Molecular Medicine 25, no. 15 (2021): 7321–7334.

[313]

H. Yamamura, Y. Suzuki, and A. Yamamura, “[Pathophysiological roles of TRPC6 channels in pulmonary arterial hypertension],” Nihon Yakurigaku Zasshi 155, no. 4 (2020): 230–235.

[314]

C. M. Zehendner, C. Valasarajan, A. Werner, et al., “Long Noncoding RNA TYKRIL Plays a Role in Pulmonary Hypertension via the p53-mediated Regulation of PDGFRβ” American Journal of Respiratory and Critical Care Medicine 202, no. 10 (2020): 1445–1457.

[315]

N. F. Hosseini, H. Manoochehri, S. G. Khoei, and M. Sheykhhasan, “The Functional Role of Long Non-coding RNA UCA1 in human Multiple Cancers: a Review Study,” Current Molecular Medicine 21, no. 2 (2021): 96–110.

[316]

S. Ghafouri-Fard and M. Taheri, “UCA1 long Non-coding RNA: an Update on Its Roles in Malignant Behavior of Cancers,” Biomedicine & Pharmacotherapy 120 (2019): 109459.

[317]

X. S. Wang, Z. Zhang, H. C. Wang, et al., “Rapid Identification of UCA1 as a Very Sensitive and Specific Unique Marker for human Bladder Carcinoma,” Clinical Cancer Research 12, no. 16 (2006): 4851–4858.

[318]

T. T. Zhu, R. L. Sun, Y. L. Yin, et al., “Long Noncoding RNA UCA1 Promotes the Proliferation of Hypoxic human Pulmonary Artery Smooth Muscle Cells,” Pflugers Archiv: European journal of physiology 471, no. 2 (2019): 347–355.

[319]

J. F. Geng, X. Liu, H. B. Zhao, W. F. Fan, J. J. Geng, and X. Z. Liu, “LncRNA UCA1 Inhibits Epilepsy and Seizure-induced Brain Injury by Regulating miR-495/Nrf2-ARE Signal Pathway,” International Journal of Biochemistry & Cell Biology 99 (2018): 133–139.

[320]

J. Fu, P. Bai, Y. Chen, T. Yu, and F. Li, “Inhibition of miR-495 Improves both Vascular Remodeling and Angiogenesis in Pulmonary Hypertension,” Journal of Vascular Research 56, no. 2 (2019): 97–106.

[321]

K. Kimura, A. Wakamatsu, Y. Suzuki, et al., “Diversification of Transcriptional Modulation: Large-scale Identification and Characterization of Putative Alternative Promoters of human Genes,” Genome Research 16, no. 1 (2006): 55–65.

[322]

A. D. Mahmoud, M. D. Ballantyne, V. Miscianinov, et al., “The human-specific and Smooth Muscle Cell-enriched lncRNA SMILR Promotes Proliferation by Regulating Mitotic CENPF mRNA and Drives Cell-cycle Progression Which Can be Targeted to Limit Vascular Remodeling,” Circulation Research 125, no. 5 (2019): 535–551.

[323]

S. Lei, F. Peng, M. L. Li, W. B. Duan, C. Q. Peng, and S. J. Wu, “LncRNA-SMILR Modulates RhoA/ROCK Signaling by Targeting miR-141 to Regulate Vascular Remodeling in Pulmonary Arterial Hypertension,” American Journal of Physiology. Heart and Circulatory Physiology 319, no. 2 (2020): H377–H391.

[324]

G. Loirand, P. Guérin, and P. Pacaud, “Rho Kinases in Cardiovascular Physiology and Pathophysiology,” Circulation Research 98, no. 3 (2006): 322–334.

[325]

N. Ishii, K. Ozaki, H. Sato, et al., “Identification of a Novel Non-coding RNA, MIAT, That Confers Risk of Myocardial Infarction,” Journal of Human Genetics 51, no. 12 (2006): 1087–1099.

[326]

W. W. Li, A. H. Cao, and F. Y. Sun, “LncRNA MIAT Stimulates Oxidative Stress in the Hypoxic Pulmonary Hypertension Model by Sponging miR-29a-5p and Inhibiting Nrf2 Pathway,” European Review for Medical and Pharmacological Sciences 24, no. 17 (2020): 9022–9029.

[327]

Z. Sun, Y. Liu, F. Yu, Y. Xu, L. Yanli, and N. Liu, “Long Non-coding RNA and mRNA Profile Analysis of Metformin to Reverse the Pulmonary Hypertension Vascular Remodeling Induced by Monocrotaline,” Biomedicine & Pharmacotherapy 115 (2019): 108933.

[328]

Z. Sun, Y. Liu, R. Hu, T. Wang, Y. Li, and N. Liu, “Metformin Inhibits Pulmonary Artery Smooth Muscle Cell Proliferation by Upregulating p21 via NONRATT015587.2,” International Journal of Molecular Medicine 49, no. 4 (2022): 49.

[329]

E. Wang, B. Zhang, L. Huang, et al., “LncRNA MIR210HG Promotes Phenotype Switching of Pulmonary Arterial Smooth Muscle Cells through Autophagy-dependent Ferroptosis Pathway,” Apoptosis 29, no. 9-10 (2024): 1648–1662.

[330]

C. Liu, J. Chen, X. Huang, et al., “lncRNA VELRP Modulates Pulmonary Arterial Smooth Muscle Cell Proliferation and Promotes Vascular Remodeling in Pulmonary Hypertension,” Arteriosclerosis, Thrombosis, and Vascular Biology 44, no. 12 (2024): 2560–2576.

[331]

J. Chen, J. Guo, J. Chen, and D. Gou, “The Vascular LNCRNA VELRP Defines an Epigenetic Checkpoint in Hyperproliferation of Pulmonary Arterial Smooth Muscle Cell,” American Journal of Respiratory and Critical Care Medicine 197 (2018): A4621.

[332]

A. Mahajan, A. Kumar, L. Chen, and N. K. Dhillon. LncRNA-536 and RNA Binding Protein RBM25 Interactions in Pulmonary Arterial Hypertension. bioRxiv [Preprint]. 2024; Aug 28:2024.08.27.610011.

[333]

M. Chinnappan, S. Gunewardena, P. Chalise, and N. Dhillon, “Analysis of lncRNA-miRNA-mRNA Interactions in Hyper-proliferative human Pulmonary Arterial Smooth Muscle Cells,” Scientific Reports 9, no. 1 (2019): 10533.

[334]

Y.-Y. Tai, Q. Yu, Y. Tang, et al., “Allele-specific Control of Rodent and human lncRNA KMT2E-AS1 Promotes Hypoxic Endothelial Pathology in Pulmonary Hypertension,” Science Translational Medicine 16, no. 729 (2024): eadd2029.

[335]

S.-E. Lemay, S. Provencher, S. Bonnet, F. Potus, and O. Boucherat, “A HIF-2α-dependent KMT2E-AS1/KMT2E Axis Orchestrates Endothelial Epigenetic and Metabolic Dysfunction in Pulmonary Hypertension,” Annals of translational medicine 12, no. 3 (2024): 55.

[336]

C. Schneider, R. M. King, and L. Philipson, “Genes Specifically Expressed at Growth Arrest of Mammalian Cells,” Cell 54, no. 6 (1988): 787–793.

[337]

K. Liu, C. Liu, and Z. Zhang, “lncRNA GAS5 Acts as a ceRNA for miR-21 in Suppressing PDGF-bb-induced Proliferation and Migration in Vascular Smooth Muscle Cells,” Journal of Cellular Biochemistry 120, no. 9 (2019): 15233–15240.

[338]

X. Hao, H. Li, P. Zhang, X. Yu, J. Jiang, and S. Chen, “Down-regulation of lncRNA Gas5 Promotes Hypoxia-induced Pulmonary Arterial Smooth Muscle Cell Proliferation by Regulating KCNK3 Expression,” European Journal of Pharmacology 889 (2020): 173618.

[339]

X. Feng, K. Wang, T. Yang, Y. Liu, and X. Wang, “LncRNA-GAS5/miR-382-3p Axis Inhibits Pulmonary Artery Remodeling and Promotes Autophagy in Chronic Thromboembolic Pulmonary Hypertension,” Genes Genomics 44, no. 4 (2022): 395–404.

[340]

G. Huang, X. Wu, S. Li, et al., “The Long Noncoding RNA CASC2 Functions as a Competing Endogenous RNA by Sponging miR-18a in Colorectal Cancer,” Scientific Reports 6 (2016): 26524.

[341]

J. Gong, Z. Chen, Y. Chen, et al., “Long Non-coding RNA CASC2 Suppresses Pulmonary Artery Smooth Muscle Cell Proliferation and Phenotypic Switch in Hypoxia-induced Pulmonary Hypertension,” Respiratory Research 20, no. 1 (2019): 53.

[342]

Y. Han, Y. Liu, C. Yang, C. Gao, X. Guo, and J. Cheng, “LncRNA CASC2 Inhibits Hypoxia-induced Pulmonary Artery Smooth Muscle Cell Proliferation and Migration by Regulating the miR-222/ING5 Axis,” Cellular & Molecular Biology Letters 25 (2020): 21.

[343]

Y. Liu, H. Zhang, Y. Li, et al., “Long Noncoding RNA Rps4l Mediates the Proliferation of Hypoxic Pulmonary Artery Smooth Muscle Cells,” Hypertension 76, no. 4 (2020): 1124–1133.

[344]

Y. Li, J. Zhang, H. Sun, et al., “lnc-Rps4l-encoded Peptide RPS4XL Regulates RPS6 Phosphorylation and Inhibits the Proliferation of PASMCs Caused by Hypoxia,” Molecular Therapy 29, no. 4 (2021): 1411–1424.

[345]

Y. Li, J. Zhang, H. Sun, et al., “RPS4XL encoded by lnc-Rps4l Inhibits Hypoxia-induced Pyroptosis by Binding HSC70 Glycosylation Site,” Molecular Therapy Nucleic Acids 28 (2022): 920–934.

[346]

J. C. Venter, M. D. Adams, E. W. Myers, et al., “The Sequence of the human Genome,” Science 291, no. 5507 (2001): 1304–1351.

[347]

C. P. Barnett, J. J. Mencel, J. Gecz, et al., “Choreoathetosis, Congenital Hypothyroidism and Neonatal respiratory Distress Syndrome with Intact NKX2-1,” American Journal of Medical Genetics. Part A 158a, no. 12 (2012): 3168–3173.

[348]

Y. Liu, R. Hu, J. Zhu, et al., “The lncRNA PAHRF Functions as a Competing Endogenous RNA to Regulate MST1 Expression by Sponging miR-23a-3p in Pulmonary Arterial Hypertension,” Vascular Pharmacology 139 (2021): 106886.

[349]

B. J. Thompson and E. Sahai, “MST Kinases in Development and Disease,” Journal of Cell Biology 210, no. 6 (2015): 871–882.

[350]

H. Ono, T. Ichiki, H. Ohtsubo, et al., “Critical Role of Mst1 in Vascular Remodeling after Injury,” Arteriosclerosis, Thrombosis, and Vascular Biology 25, no. 9 (2005): 1871–1876.

[351]

T. V. Kudryashova, S. Dabral, S. Nayakanti, et al., “Noncanonical HIPPO/MST Signaling via BUB3 and FOXO Drives Pulmonary Vascular Cell Growth and Survival,” Circulation Research 130, no. 5 (2022): 760–778.

[352]

E. Pasmant, I. Laurendeau, D. Héron, M. Vidaud, D. Vidaud, and I. Bièche, “Characterization of a Germ-line Deletion, Including the Entire INK4/ARF Locus, in a Melanoma-neural System Tumor family: Identification of ANRIL, an Antisense Noncoding RNA whose Expression Coclusters with ARF,” Cancer Research 67, no. 8 (2007): 3963–3969.

[353]

Y. Kong, C. H. Hsieh, and L. C. Alonso, “ANRIL: a lncRNA at the CDKN2A/B Locus with Roles in Cancer and Metabolic Disease,” Frontiers in Endocrinology (Lausanne) 9 (2018): 405.

[354]

N. Lou, G. Liu, and Y. Pan, “Long Noncoding RNA ANRIL as a Novel Biomarker in human Cancer,” Future oncology 16, no. 35 (2020): 2981–2995.

[355]

Z. W. Wang, J. J. Pan, J. F. Hu, et al., “SRSF3-mediated Regulation of N6-methyladenosine Modification-related lncRNA ANRIL Splicing Promotes Resistance of Pancreatic Cancer to Gemcitabine,” Cell reports 39, no. 6 (2022): 110813.

[356]

W. Deng, Y. Zhang, J. Cai, et al., “LncRNA-ANRIL Promotes Gastric Cancer Progression by Enhancing NF-kB Signaling,” Experimental Biology and Medicine (Maywood, N.J.) 244, no. 12 (2019): 953–959.

[357]

C. Sun, C. Shen, Y. Zhang, and C. Hu, “LncRNA ANRIL Negatively Regulated Chitooligosaccharide-induced Radiosensitivity in Colon Cancer Cells by Sponging miR-181a-5p,” Advances in clinical and experimental medicine: official organ Wroclaw Medical University 30, no. 1 (2021): 55–65.

[358]

A. Sanchez, J. Lhuillier, G. Grosjean, L. Ayadi, and S. Maenner, “The Long Non-coding RNA ANRIL in Cancers,” Cancers (Basel) 15, no. 16 (2023): 4160.

[359]

Z. Li, X. Yu, and J. Shen, “ANRIL: a Pivotal Tumor Suppressor Long Non-coding RNA in human Cancers,” Tumour Biology: The Journal of the International Society for Oncodevelopmental Biology and Medicine 37, no. 5 (2016): 5657–5661.

[360]

Z. Du, F. Zhang, L. Liu, et al., “LncRNA ANRIL Promotes HR Repair through Regulating PARP1 Expression by Sponging miR-7-5p in Lung Cancer,” BMC cancer 23, no. 1 (2023): 130.

[361]

I. Razeghian-Jahromi, A. Karimi Akhormeh, and M. J. Zibaeenezhad, “The Role of ANRIL in Atherosclerosis,” Disease Markers 2022 (2022): 8859677.

[362]

L. Chen, H. Qu, M. Guo, et al., “ANRIL and Atherosclerosis,” Journal of Clinical Pharmacy and Therapeutics 45, no. 2 (2020): 240–248.

[363]

J. S. Chi, J. Z. Li, J. J. Jia, T. Zhang, X. M. Liu, and L. Yi, “Long Non-coding RNA ANRIL in Gene Regulation and Its Duality in Atherosclerosis,” Journal of Huazhong University of Science and Technology. Medical Sciences = Hua Zhong Ke Ji Da Xue Xue Bao. Yi Xue Ying De Wen Ban = Huazhong Keji Daxue Xuebao. Yixue Yingdewen Ban 37, no. 6 (2017): 816–822.

[364]

I. Gareev, V. Kudriashov, A. Sufianov, et al., “The Role of Long Non-coding RNA ANRIL in the Development of Atherosclerosis,” Non-coding RNA Research 7, no. 4 (2022): 212–216.

[365]

T. Huang, H. Y. Zhao, X. B. Zhang, et al., “LncRNA ANRIL Regulates Cell Proliferation and Migration via Sponging miR-339-5p and Regulating FRS2 Expression in Atherosclerosis,” European Review for Medical and Pharmacological Sciences 24, no. 4 (2020): 1956–1969.

[366]

A. Congrains, K. Kamide, R. Oguro, et al., “Genetic Variants at the 9p21 Locus Contribute to Atherosclerosis through Modulation of ANRIL and CDKN2A/B,” Atherosclerosis 220, no. 2 (2012): 449–455.

[367]

C. Tan, J. Liu, J. Wei, and S. Yang, “Effects of ANRIL Variants on the Risk of Ischemic Stroke: a Meta-analysis,” Bioscience Reports 39, no. 5 (2019): BSR20182127.

[368]

S. Hao, K. Zhou, P. Yu, et al., “ANRIL Polymorphisms in Psoriasis vulgaris Patients in northern China,” European Journal of Dermatology 32, no. 2 (2022): 259–268.

[369]

S. Mirzajani, S. Ghafouri-Fard, J. M. Habibabadi, M. C. Glassy, A. Sayad, and M. Taheri, “Altered ANRIL Methylation in Epileptic Patients,” Journal of Molecular Neuroscience 71, no. 1 (2021): 193–199.

[370]

W. C. Ng, J. Chan, T. N. Tarkas, N. Meharban, W. Munir, and M. Bashir, “The Association of ANRIL with Coronary Artery Disease and Aortic Aneurysms, How Far Does The Gene Desert Go?,” Annals of Vascular Surgery 80 (2022): 345–357.

[371]

A. Öztürk and A. O. Ada, “The Roles of ANRIL Polymorphisms in Periodontitis: a Systematic Review and Meta-analysis,” Clinical Oral Investigations 26, no. 2 (2022): 1121–1135.

[372]

P. Sooshtari, B. Feng, S. Biswas, et al., “ANRIL Regulates Multiple Molecules of Pathogenetic Significance in Diabetic Nephropathy,” PLoS ONE 17, no. 8 (2022): e0270287.

[373]

P. Wan, S. Huang, Y. Luo, et al., “Reciprocal Regulation between lncRNA ANRIL and p15 in Steroid-induced Glaucoma,” Cells 11, no. 9 (2022): 1468.

[374]

H. Su, B. Liu, H. Chen, et al., “LncRNA ANRIL Mediates Endothelial Dysfunction through BDNF Downregulation in Chronic Kidney Disease,” Cell death & disease 13, no. 7 (2022): 661.

[375]

A. A. Thomas, B. Feng, and S. Chakrabarti, “ANRIL: a Regulator of VEGF in Diabetic Retinopathy,” Investigative Ophthalmology & Visual Science 58, no. 1 (2017): 470–480.

[376]

M. Akbari, B. M. Hussen, S. Eslami, S. M. Neishabouri, and S. Ghafouri-Fard, “Association between ANRIL Polymorphisms and Risk of Obsessive-compulsive Disorder,” Heliyon 9, no. 3 (2023): e14081.

[377]

G. Li, L. Gao, J. Zhao, D. Liu, H. Li, and M. Hu, “LncRNA ANRIL/miR-7-5p/TCF4 Axis Contributes to the Progression of T Cell Acute Lymphoblastic Leukemia,” Cancer cell international 20 (2020): 335.

[378]

S. Wang, C. Zhang, and X. Zhang, “Downregulation of Long Non-Coding RNA ANRIL Promotes Proliferation and Migration in Hypoxic human Pulmonary Artery Smooth Muscle Cells,” Molecular Medicine Reports 21, no. 2 (2020): 589–596.

[379]

M. S. Leisegang, C. Fork, I. Josipovic, et al., “Long Noncoding RNA MANTIS Facilitates Endothelial Angiogenic Function,” Circulation 136, no. 1 (2017): 65–79.

[380]

Q. Yang, W. Fan, B. Lai, B. Liao, and M. Deng, “lncRNA-TCONS_00008552 Expression in Patients with Pulmonary Arterial Hypertension due to Congenital Heart Disease,” PLoS ONE 18, no. 3 (2023): e0281061.

[381]

Y. Liu, Z. Sun, J. Zhu, B. Xiao, J. Dong, and X. Li, “LncRNA-TCONS_00034812 in Cell Proliferation and Apoptosis of Pulmonary Artery Smooth Muscle Cells and Its Mechanism,” Journal of Cellular Physiology 233, no. 6 (2018): 4801–4814.

[382]

L. Benetatos, G. Vartholomatos, and E. Hatzimichael, “MEG3 imprinted Gene Contribution in Tumorigenesis,” International Journal of Cancer 129, no. 4 (2011): 773–779.

[383]

B. Zhu, Y. Gong, G. Yan, et al., “Down-regulation of lncRNA MEG3 Promotes Hypoxia-induced human Pulmonary Artery Smooth Muscle Cell Proliferation and Migration via Repressing PTEN by Sponging miR-21,” Biochemical and Biophysical Research Communications 495, no. 3 (2018): 2125–2132.

[384]

A. Kitagawa, C. Jacob, and S. A. Gupte, “Glucose-6-phosphate Dehydrogenase and MEG3 Controls Hypoxia-induced Expression of Serum Response Factor (SRF) and SRF-dependent Genes in Pulmonary Smooth Muscle Cell,” Journal of Smooth Muscle Research 58, no. 0 (2022): 34–49.

[385]

Z. Sun, X. Nie, S. Sun, et al., “Long Non-coding RNA MEG3 Downregulation Triggers human Pulmonary Artery Smooth Muscle Cell Proliferation and Migration via the p53 Signaling Pathway,” Cellular Physiology and Biochemistry 42, no. 6 (2017): 2569–2581.

[386]

Y. Xing, X. Zheng, Y. Fu, et al., “Long Noncoding RNA-maternally Expressed Gene 3 Contributes to Hypoxic Pulmonary Hypertension,” Molecular Therapy 27, no. 12 (2019): 2166–2181.

[387]

J. Chen, J. Guo, X. Cui, et al., “The Long Noncoding RNA LnRPT Is Regulated by PDGF-BB and Modulates the Proliferation of Pulmonary Artery Smooth Muscle Cells,” American Journal of Respiratory Cell and Molecular Biology 58, no. 2 (2018): 181–193.

[388]

X. Zhao, C. Wang, M. Liu, F. Meng, and K. Liu, “LncRNA FENDRR Servers as a Possible Marker of Essential Hypertension and Regulates human Umbilical Vein Endothelial Cells Dysfunction via miR-423-5p/Nox4 Axis,” International Journal of General Medicine 15 (2022): 2529–2540.

[389]

X. Li, Y. Zhang, L. Su, et al., “FGF21 alleviates Pulmonary Hypertension by Inhibiting mTORC1/EIF4EBP1 Pathway via H19,” Journal of Cellular and Molecular Medicine 26, no. 10 (2022): 3005–3021.

[390]

C. Yang, R. Rong, Y. Li, M. Cheng, and Y. Luo, “Decrease in LINC00963 Attenuates the Progression of Pulmonary Arterial Hypertension via microRNA-328-3p/Profilin 1 Axis,” Journal of Clinical Laboratory Analysis 36, no. 5 (2022): e24383.

[391]

S. T. Xue, B. Zheng, S. Q. Cao, et al., “Long Non-coding RNA LINC00680 Functions as a ceRNA to Promote Esophageal Squamous Cell Carcinoma Progression through the miR-423-5p/PAK6 Axis,” Molecular cancer 21, no. 1 (2022): 69.

[392]

R. Montiel-Manriquez, C. Castro-Hernández, C. Arriaga-Canon, and L. A. Herrera, “Antisense Oligonucleotides as a Tool for Prolonged Knockdown of Nuclear lncRNAs in human Cell Lines,” Journal of visualized experiments: JoVE, no. 199 (2023): e65124.

[393]

P. Jafar-Nejad, B. Powers, A. Soriano, et al., “The Atlas of RNase H Antisense Oligonucleotide Distribution and Activity in the CNS of Rodents and Non-human Primates Following central Administration,” Nucleic Acids Research 49, no. 2 (2021): 657–673.

[394]

H. Wang, R. B. Chen, S. N. Zhang, and R. F. Zhang, “N7-methylguanosine Modification of lncRNAs in a Rat Model of Hypoxic Pulmonary Hypertension: a Comprehensive Analysis,” BMC Genomics 23, no. 1 (2022): 33.

[395]

P. Singh, A. Singh, S. Shah, J. Vataliya, A. Mittal, and D. Chitkara, “RNA Interference Nanotherapeutics for Treatment of Glioblastoma Multiforme,” Molecular Pharmaceutics 17, no. 11 (2020): 4040–4066.

[396]

L. Stojic, “Tuning the Expression of Long Noncoding RNA Loci with CRISPR Interference,” Methods in Molecular Biology 2161 (2020): 1–16.

[397]

H. Geng, H. F. Bu, F. Liu, et al., “In Inflamed Intestinal Tissues and Epithelial Cells, Interleukin 22 Signaling Increases Expression of H19 Long Noncoding RNA, Which Promotes Mucosal Regeneration,” Gastroenterology 155, no. 1 (2018): 144–155.

[398]

O. Khorkova, J. Stahl, A. Joji, C. H. Volmar, Z. Zeier, and C. Wahlestedt, “Natural Antisense Transcripts as Drug Targets,” Frontiers in Molecular Biosciences 9 (2022): 978375.

[399]

C. Wahlestedt, “Natural Antisense and Noncoding RNA Transcripts as Potential Drug Targets,” Drug Discovery Today 11, no. 11-12 (2006): 503–508.

[400]

C. Yang, L. Zhang, X. Hao, M. Tang, B. Zhou, and J. Hou, “Identification of a Novel N7-methylguanosine-related lncRNA Signature Predicts the Prognosis of Hepatocellular Carcinoma and Experiment Verification,” Current Oncology 30, no. 1 (2022): 430–448.

[401]

S. Mocellin and M. Provenzano, “RNA Interference: Learning Gene Knock-Down from Cell Physiology,” Journal of translational medicine 2, no. 1 (2004): 39.

[402]

L. Deng, J. Chen, B. Chen, et al., “LncPTSR Triggers Vascular Remodeling in Pulmonary Hypertension by Regulating [Ca(2+)](i) in Pulmonary Arterial Smooth Muscle Cells,” American Journal of Respiratory Cell and Molecular Biology 66, no. 5 (2022): 524–538.

[403]

M. Czarnek, J. Kochan, M. Wawro, R. Myrczek, and J. Bereta, “Construction of a Set of Novel Transposon Vectors for Efficient Silencing of Protein and lncRNA Genes via CRISPR Interference,” Molecular Biotechnology 65, no. 10 (2023): 1598–1607.

[404]

S. Hu, J. Du, N. Chen, et al., “In Vivo CRISPR/Cas9-mediated Genome Editing Mitigates Photoreceptor Degeneration in a Mouse Model of X-linked Retinitis pigmentosa,” Investigative Ophthalmology & Visual Science 61, no. 4 (2020): 31.

[405]

J. Zhao, W. Ma, Y. Zhong, et al., “Transcriptional Inhibition of lncRNA gadd7 by CRISPR/dCas9-KRAB Protects Spermatocyte Viability,” Frontiers in Molecular Biosciences 8 (2021): 652392.

[406]

J. A. R Gordon, C. E. Tye, B. Banerjee, et al., “LINC01638 sustains human Mesenchymal Stem Cell Self-renewal and Competency for Osteogenic Cell Fate,” Scientific Reports 13, no. 1 (2023): 20314.

[407]

S. J. Liu, M. A. Horlbeck, J. S. Weissman, and D. A. Lim, “Genome-Scale Perturbation of Long Noncoding RNA Expression Using CRISPR Interference,” Methods in Molecular Biology 2254 (2021): 323–338.

[408]

M. J. Ramaiah, A. D. Tangutur, and R. R. Manyam, “Epigenetic Modulation and Understanding of HDAC Inhibitors in Cancer Therapy,” Life Sciences 277 (2021): 119504.

[409]

D. Sarkar, E. Y. Leung, B. C. Baguley, G. J. Finlay, and M. E. Askarian-Amiri, “Epigenetic Regulation in human Melanoma: Past and Future,” Epigenetics 10, no. 2 (2015): 103–121.

[410]

H. Wang, M. Guo, H. Wei, and Y. Chen, “Targeting p53 Pathways: Mechanisms, Structures, and Advances in Therapy,” Signal Transduction and Targeted Therapy 8, no. 1 (2023): 92.

[411]

H. Wang, W. Yu, Y. Wang, et al., “P53 contributes to Cardiovascular Diseases via Mitochondria Dysfunction: a New Paradigm,” Free Radical Biology and Medicine 208 (2023): 846–858.

[412]

T. Wakasugi, I. Shimizu, Y. Yoshida, et al., “Role of Smooth Muscle Cell p53 in Pulmonary Arterial Hypertension,” PLoS ONE 14, no. 2 (2019): e0212889.

[413]

D. P. Lane, “Cancer. P53, Guardian of the Genome,” Nature 358, no. 6381 (1992): 15–16.

[414]

L. J. Hernández Borrero and W. S. El-Deiry, “Tumor Suppressor p53: Biology, Signaling Pathways, and Therapeutic Targeting,” Biochimica et Biophysica Acta Reviews on Cancer 1876, no. 1 (2021): 188556.

[415]

B. J. Aubrey, A. Strasser, and G. L. Kelly, “Tumor-suppressor Functions of the TP53 Pathway,” Cold Spring Harbor perspectives in medicine 6, no. 5 (2016): a026062.

[416]

H. H. Hasbullah and M. Musa, “Gene Therapy Targeting p53 and KRAS for Colorectal Cancer Treatment: a Myth or the Way Forward?,” International Journal of Molecular Sciences 22, no. 21 (2021): 11941.

[417]

J. K. Hennigs, A. Cao, C. G. Li, et al., “PPARγ-p53-mediated Vasculoregenerative Program to Reverse Pulmonary Hypertension,” Circulation Research 128, no. 3 (2021): 401–418.

[418]

A. R. Bhagwani, M. Ali, B. Piper, et al., “A p53-TLR3 Axis Ameliorates Pulmonary Hypertension by Inducing BMPR2 via IRF3,” Iscience 26, no. 2 (2023): 105935.

[419]

C. Teng, B. Li, C. Lin, et al., “Targeted Delivery of Baicalein-p53 Complex to Smooth Muscle Cells Reverses Pulmonary Hypertension,” Journal of Control Release 341 (2022): 591–604.

[420]

S. Mizuno, H. J. Bogaard, D. Kraskauskas, et al., “p53 Gene Deficiency Promotes Hypoxia-induced Pulmonary Hypertension and Vascular Remodeling in Mice,” American Journal of Physiology. Lung Cellular and Molecular Physiology 300, no. 5 (2011): L753–761.

[421]

Z. Wang, K. Yang, Q. Zheng, et al., “Divergent Changes of p53 in Pulmonary Arterial Endothelial and Smooth Muscle Cells Involved in the Development of Pulmonary Hypertension,” American Journal of Physiology. Lung Cellular and Molecular Physiology 316, no. 1 (2019): L216–L228.

[422]

S. Jacquin, V. Rincheval, B. Mignotte, et al., “Inactivation of p53 Is Sufficient to Induce Development of Pulmonary Hypertension in Rats,” PLoS ONE 10, no. 6 (2015): e0131940.

[423]

W. Zhuang, G. Lian, B. Huang, et al., “CPT1 regulates the Proliferation of Pulmonary Artery Smooth Muscle Cells through the AMPK-p53-p21 Pathway in Pulmonary Arterial Hypertension,” Molecular and Cellular Biochemistry 455, no. 1-2 (2019): 169–183.

[424]

X. Wang, Y. F. Ibrahim, D. Das, M. Zungu-Edmondson, N. V. Shults, and Y. J. Suzuki, “Carfilzomib Reverses Pulmonary Arterial Hypertension,” Cardiovascular Research 110, no. 2 (2016): 188–199.

[425]

R. Yamanaka, A. Hoshino, K. Fukai, et al., “TIGAR Reduces Smooth Muscle Cell Autophagy to Prevent Pulmonary Hypertension,” American Journal of Physiology. Heart and Circulatory Physiology 319, no. 5 (2020): H1087–H1096.

[426]

Y. Y. Kim, J. H. Um, J. H. Yoon, et al., “p53 regulates Mitochondrial Dynamics by Inhibiting Drp1 Translocation into Mitochondria during Cellular Senescence,” Faseb Journal 34, no. 2 (2020): 2451–2464.

[427]

E. Bonda, G. Rahav, A. Kaya, and M. Bakhanashvili, “p53 in the Mitochondria, as a Trans-acting Protein, Provides Error-correction Activities during the Incorporation of Non-canonical dUTP into DNA,” Oncotarget 7, no. 45 (2016): 73323–73336.

[428]

S. De, J. Kumari, R. Mudgal, et al., “RECQL4 is Essential for the Transport of p53 to Mitochondria in Normal human Cells in the Absence of Exogenous Stress,” Journal of Cell Science 125, no. Pt 10 (2012): 2509–2522.

[429]

F. Li, X. Chen, B. Xu, and H. Zhou, “Curcumin Induces p53-independent Necrosis in H1299 Cells via a Mitochondria-associated Pathway,” Molecular Medicine Reports 12, no. 5 (2015): 7806–7814.

[430]

Y. Han, Y. Liu, J. Zhen, et al., “P53 regulates Mitochondrial Biogenesis via Transcriptionally Induction of Mitochondrial Ribosomal Protein L12,” Experimental Cell Research 418, no. 1 (2022): 113249.

[431]

S. Soberanes, V. Panduri, G. M. Mutlu, A. Ghio, G. R. Bundinger, and D. W. Kamp, “P53 mediates Particulate Matter-induced Alveolar Epithelial Cell Mitochondria-regulated Apoptosis,” American Journal of Respiratory and Critical Care Medicine 174, no. 11 (2006): 1229–1238.

[432]

W. Lv, L. Liang, D. Liu, C. Li, L. Jia, and Y. Jin, “Aquaporin 1 Facilitates Ferroptosis, M1 Polarization, Mitochondrial Dysfunction, and Autophagy Damage on Lipopolysaccharide-induced Macrophage through Down-regulation of p53 Signaling Pathway,” Dna and Cell Biology 42, no. 8 (2023): 456–480.

[433]

K. Zhang, Y. Zheng, G. Bao, et al., “Flt3 activation Mitigates Mitochondrial Fragmentation and Heart Dysfunction through Rebalanced L-OPA1 Processing by Hindering the Interaction between Acetylated p53 and PHB2 in Cardiac Remodeling,” Antioxidants 12, no. 9 (2023): 1657.

[434]

T. L. Ma, K. F. Chang, X. F. Huang, H. C. Lai, C. Y. Hsiao, and N. M. Tsai, “Angelica sinensis Extract Induces Telomere Dysfunction, Cell Cycle Arrest, and Mitochondria-mediated Apoptosis in human Glioblastoma Cells,” Chinese Journal of Physiology 66, no. 3 (2023): 119–128.

[435]

Y. F. Wang, Y. D. Wang, S. Gao, and W. Sun, “Implications of p53 in Mitochondrial Dysfunction and Parkinson’s Disease,” International Journal of Neuroscience 134, no. 8 (2024): 906–917.

[436]

S. Rius-Pérez, S. Pérez, M. B. Toledano, and J. Sastre, “p53 drives Necroptosis via Downregulation of Sulfiredoxin and Peroxiredoxin 3,” Redox Biology 56 (2022): 102423.

[437]

R. A. Eid, M. M Bin-Meferij, A. F. El-Kott, et al., “Exendin-4 Protects against Myocardial Ischemia-reperfusion Injury by Upregulation of SIRT1 and SIRT3 and Activation of AMPK,” Journal of Cardiovascular Translational Research 14, no. 4 (2021): 619–635.

[438]

Y. H. Jiang, L. Y. Jiang, Y. C. Wang, D. F. Ma, and X. Li, “Quercetin Attenuates Atherosclerosis via Modulating Oxidized LDL-induced Endothelial Cellular Senescence,” Frontiers in pharmacology 11 (2020): 512.

[439]

C. Zhang, P. Liao, R. Liang, X. Zheng, and J. Jian, “Epigallocatechin Gallate Prevents Mitochondrial Impairment and Cell Apoptosis by Regulating miR-30a/p53 Axis,” Phytomedicine 61 (2019): 152845.

[440]

J. H. Lee, D. H. Kim, M. Kim, K. H. Jung, and K. H. Lee, “Mitochondrial ROS-mediated Metabolic and Cytotoxic Effects of Isoproterenol on Cardiomyocytes Are p53-dependent and Reversed by Curcumin,” Molecules (Basel, Switzerland) 27, no. 4 (2022): 1346.

[441]

F. Forini, C. Kusmic, G. Nicolini, et al., “Triiodothyronine Prevents Cardiac Ischemia/Reperfusion Mitochondrial Impairment and Cell Loss by Regulating miR30a/p53 Axis,” Endocrinology 155, no. 11 (2014): 4581–4590.

[442]

Y. Wang, B. Zhong, Q. Wu, J. Tong, T. Zhu, and M. Zhang, “Effect of Aldosterone on Senescence and Proliferation Inhibition of Endothelial Progenitor Cells Induced by Sirtuin 1 (SIRT1) in Pulmonary Arterial Hypertension,” Medical Science Monitor 26 (2020): e920678.

[443]

W. S. Hu, W. Y. Liao, C. H. Chang, and T. S. Chen, “Paracrine IGF-1 Activates SOD2 Expression and Regulates ROS/p53 Axis in the Treatment of Cardiac Damage in D-Galactose-induced Aging Rats after Receiving Mesenchymal Stem Cells,” Journal of Clinical Medicine 11, no. 15 (2022): 4419.

[444]

M. Maurer, M. Bader, M. Bas, et al., “New Topics in Bradykinin Research,” Allergy 66, no. 11 (2011): 1397–1406.

[445]

W. Feng, X. Xu, G. Zhao, et al., “Increased Age-related Cardiac Dysfunction in Bradykinin B2 Receptor-deficient Mice,” Journals of Gerontology. Series A, Biological Sciences and Medical Sciences 71, no. 2 (2016): 178–187.

[446]

S. Rana, R. Datta, R. D. Chaudhuri, E. Chatterjee, M. Chawla-Sarkar, and S. Sarkar, “Nanotized PPARα Overexpression Targeted to Hypertrophied Myocardium Improves Cardiac Function by Attenuating the p53-GSK3β-mediated Mitochondrial Death Pathway,” Antioxidants & Redox Signaling 30, no. 5 (2019): 713–732.

[447]

Y. Zhu, X. Q. Han, X. J. Sun, R. Yang, W. Q. Ma, and N. F. Liu, “Lactate Accelerates Vascular Calcification through NR4A1-regulated Mitochondrial Fission and BNIP3-related Mitophagy,” Apoptosis 25, no. 5-6 (2020): 321–340.

[448]

A. Yin, M. Feng, L. Zhang, Z. Cheng, Y. Li, and L. Qian, “Identification of a Novel Native Peptide Derived from 60S Ribosomal Protein L23a That Translationally Regulates p53 to Reduce Myocardial Ischemia-reperfusion,” Pharmacological Research 175 (2022): 105988.

[449]

X. Liu, C. C. Chua, J. Gao, et al., “Pifithrin-alpha Protects against Doxorubicin-induced Apoptosis and Acute Cardiotoxicity in Mice,” American Journal of Physiology. Heart and Circulatory Physiology 286, no. 3 (2004): H933–H939.

[450]

J. Liu, W. Mao, B. Ding, and C. S. Liang, “ERKs/p53 Signal Transduction Pathway Is Involved in Doxorubicin-induced Apoptosis in H9c2 Cells and Cardiomyocytes,” American Journal of Physiology. Heart and Circulatory Physiology 295, no. 5 (2008): H1956–H1965.

[451]

V. A. Sardão, P. J. Oliveira, J. Holy, C. R. Oliveira, and K. B. Wallace, “Doxorubicin-induced Mitochondrial Dysfunction Is Secondary to Nuclear p53 Activation in H9c2 Cardiomyoblasts,” Cancer Chemotheraphy and Pharmacology 64, no. 4 (2009): 811–827.

[452]

J. Gu, S. Wang, H. Guo, et al., “Inhibition of p53 Prevents Diabetic Cardiomyopathy by Preventing Early-stage Apoptosis and Cell Senescence, Reduced Glycolysis, and Impaired Angiogenesis,” Cell death & disease 9, no. 2 (2018): 82.

[453]

H. J. Wang, E. Y. Lee, S. J. Han, et al., “Dual Pathways of p53 Mediated Glucolipotoxicity-induced Apoptosis of Rat Cardiomyoblast Cell: Activation of p53 Proapoptosis and Inhibition of Nrf2-NQO1 Antiapoptosis,” Metabolism 61, no. 4 (2012): 496–503.

[454]

H. Men, H. Cai, Q. Cheng, et al., “The Regulatory Roles of p53 in Cardiovascular Health and Disease,” Cellular and Molecular Life Sciences 78, no. 5 (2021): 2001–2018.

[455]

X. Jiang, K. Zhang, C. Gao, et al., “Activation of FMS-Like Tyrosine Kinase 3 Protects against Isoprenaline-induced Cardiac Hypertrophy by Improving Autophagy and Mitochondrial Dynamics,” Faseb Journal 36, no. 12 (2022): e22672.

[456]

N. Mouraret, E. Marcos, S. Abid, et al., “Activation of Lung p53 by Nutlin-3a Prevents and Reverses Experimental Pulmonary Hypertension,” Circulation 127, no. 16 (2013): 1664–1676.

[457]

X. Han, C. Li, P. Yang, and T. Jiang, “Potential Mechanisms of Qili Qiangxin capsule to Prevent Pulmonary Arterial Hypertension Based on Network Pharmacology Analysis in a Rat Model,” Annals of translational medicine 10, no. 8 (2022): 453.

[458]

J. Liu, J. Li, C. Xie, L. Xuan, and B. Tang, “MSCs Attenuate Hypoxia Induced Pulmonary Hypertension by Activating P53 and NF-kB Signaling Pathway through TNFα Secretion,” Biochemical and Biophysical Research Communications 532, no. 3 (2020): 400–405.

[459]

Y. Zhang, K. Köhler, J. Xu, et al., “Inhibition of p53 after Acute Myocardial Infarction: Reduction of Apoptosis Is Counteracted by Disturbed Scar Formation and Cardiac Rupture,” Journal of Molecular and Cellular Cardiology 50, no. 3 (2011): 471–478.

[460]

A. M. Popović, A. Huđek Turković, K. Žuna, V. Bačun-Družina, I. Rubelj, and M. Matovinović, “FTO Gene Polymorphisms at the Crossroads of Metabolic Pathways of Obesity and Epigenetic Influences,” Food Technology and Biotechnology 61, no. 1 (2023): 14–26.

[461]

B. I. Fedeles, V. Singh, J. C. Delaney, D. Li, and J. M. Essigmann, “The AlkB family of Fe(II)/α-Ketoglutarate-dependent Dioxygenases: Repairing Nucleic Acid Alkylation Damage and Beyond,” Journal of Biological Chemistry 290, no. 34 (2015): 20734–20742.

[462]

J. Wei, F. Liu, Z. Lu, et al., “Differential M(6)A, M(6)A(m), and M(1)A Demethylation Mediated by FTO in the Cell Nucleus and Cytoplasm,” Molecular Cell 71, no. 6 (2018): 973–985.e5.

[463]

Y. Cui, P. Wang, M. Li, et al., “Cinnamic Acid Mitigates Left Ventricular Hypertrophy and Heart Failure in Part through Modulating FTO-dependent N(6)-methyladenosine RNA Modification in Cardiomyocytes,” Biomedicine & Pharmacotherapy 165 (2023): 115168.

[464]

H. Kang, Z. Zhang, L. Yu, Y. Li, M. Liang, and L. Zhou, “FTO Reduces Mitochondria and Promotes Hepatic Fat Accumulation through RNA Demethylation,” Journal of Cellular Biochemistry 119, no. 7 (2018): 5676–5685.

[465]

Z. Cockova, O. Honc, P. Telensky, M. J. Olsen, and J. Novotny, “Streptozotocin-induced Astrocyte Mitochondrial Dysfunction Is Ameliorated by FTO Inhibitor MO-I-500,” Acs Chemical Neuroscience 12, no. 20 (2021): 3818–3828.

[466]

A. Bravard, E. Lefai, E. Meugnier, et al., “FTO Is Increased in Muscle during Type 2 Diabetes, and Its Overexpression in Myotubes Alters Insulin Signaling, Enhances Lipogenesis and ROS Production, and Induces Mitochondrial Dysfunction,” Diabetes 60, no. 1 (2011): 258–268.

[467]

A. Lim, J. Zhou, R. A. Sinha, et al., “Hepatic FTO Expression Is Increased in NASH and Its Silencing Attenuates Palmitic Acid-induced Lipotoxicity,” Biochemical and Biophysical Research Communications 479, no. 3 (2016): 476–481.

[468]

J. Xu, D. Yin, W. Zhang, and Y. Xu, “The Role and Mechanism of FTO in Pulmonary Vessels,” Biotechnology & Genetic Engineering Reviews 40, no. 4 (2023): 4284–4299.

[469]

M. K. Andersen, L. Ängquist, J. Bork-Jensen, et al., “Physical Activity and Insulin Sensitivity Independently Attenuate the Effect of FTO rs9939609 on Obesity,” Diabetes Care 46, no. 5 (2023): 985–992.

[470]

C. Celis-Morales, C. F. Marsaux, K. M. Livingstone, et al., “Physical Activity Attenuates the Effect of the FTO Genotype on Obesity Traits in European Adults: the Food4Me Study,” obesity 24, no. 4 (2016): 962–969.

[471]

T. O. Kilpeläinen, L. Qi, S. Brage, et al., “Physical Activity Attenuates the Influence of FTO Variants on Obesity Risk: a Meta-analysis of 218, 166 Adults and 19, 268 Children,” Plos Medicine 8, no. 11 (2011): e1001116.

[472]

J. Petkeviciene, A. Smalinskiene, J. Klumbiene, V. Petkevicius, V. Kriaucioniene, and V. Lesauskaite, “Physical Activity, but Not Dietary Intake, Attenuates the Effect of the FTO rs9939609 Polymorphism on Obesity and Metabolic Syndrome in Lithuanian Adult Population,” Public Health 135 (2016): 23–29.

[473]

K. S. Vimaleswaran, S. Li, J. H. Zhao, et al., “Physical Activity Attenuates the Body Mass Index-increasing Influence of Genetic Variation in the FTO Gene,” American Journal of Clinical Nutrition 90, no. 2 (2009): 425–428.

[474]

P. F. Todendi, C. Brand, J. F. C. Silveira, et al., “Physical Fitness Attenuates the Genetic Predisposition to Obesity in Children and Adolescents,” Scandinavian Journal of Medicine & Science in Sports 31, no. 4 (2021): 894–902.

[475]

J. R. Ruiz, I. Labayen, F. B. Ortega, et al., “Attenuation of the Effect of the FTO rs9939609 Polymorphism on Total and central Body Fat by Physical Activity in Adolescents: the HELENA Study,” Archives of Pediatrics & Adolescent Medicine 164, no. 4 (2010): 328–333.

[476]

P. T. Williams, “Attenuating Effect of Vigorous Physical Activity on the Risk for Inherited Obesity: a Study of 47, 691 Runners,” PLoS ONE 7, no. 2 (2012): e31436.

[477]

Y. Qing, L. Dong, L. Gao, et al., “R-2-hydroxyglutarate Attenuates Aerobic Glycolysis in Leukemia by Targeting the FTO/M(6)A/PFKP/LDHB Axis,” Molecular Cell 81, no. 5 (2021): 922–939.

[478]

C. Huang, Y. Luo, B. Zeng, et al., “Branched-chain Amino Acids Prevent Obesity by Inhibiting the Cell Cycle in an NADPH-FTO-m(6)A Coordinated Manner,” Journal of Nutritional Biochemistry 122 (2023): 109437.

[479]

C. Gu, H. Zhang, Q. Li, S. Zhao, and Y. Gao, “MiR-192 Attenuates High Glucose-induced Pyroptosis in Retinal Pigment Epithelial Cells via Inflammasome Modulation,” Bioengineered 13, no. 4 (2022): 10362–10372.

[480]

R. Wang, L. Wang, L. Wang, et al., “FGF2 is Protective towards Cisplatin-induced KGN Cell Toxicity by Promoting FTO Expression and Autophagy,” Front Endocrinol (Lausanne) 13 (2022): 890623.

[481]

B. Liu, L. Xia, Y. Li, et al., “Prenatal Nicotine Exposure Raises Male Blood Pressure via FTO-mediated NOX2/ROS Signaling,” Hypertension 81, no. 2 (2023): 240–251.

[482]

X. Li and Y. X. Tao, “Intrathecal Administration of the Fat-mass and Obesity-associated Protein Inhibitor Mitigates Neuropathic Pain in Female Rats,” Translational Perioperative and Pain Medicine 9, no. 4 (2022): 478–487.

[483]

Z. Xingxing, L. Tao, W. Liangbing, S. Junmei, and G. Jiarong, “Qiteng Xiaozhuo Granules Medicated Serum Inhibits Excessive Proliferation and Promotes Apoptosis of human Glomerular Mesangial Cells by Targeting Fat Mass and Obesity Associated Proteins,” Journal of Traditional Chinese Medicine 43, no. 6 (2023): 1072–1080.

[484]

N. Lu, X. Li, J. Yu, et al., “Curcumin Attenuates Lipopolysaccharide-induced Hepatic Lipid Metabolism Disorder by Modification of M(6) A RNA Methylation in Piglets,” Lipids 53, no. 1 (2018): 53–63.

[485]

Y. Zhang, H. Zhou, and C. Ding, “The Ameliorative Effect of CangFu Daotan Decoction on Polycystic Ovary Syndrome of Rodent Model Is Associated with m6A Methylation and Wnt/β-catenin Pathway,” Gynecological Endocrinology 39, no. 1 (2023): 2181637.

[486]

M. V. Sukhanova, A. S. Singatulina, D. Pastré, and O. I. Lavrik, “Fused in Sarcoma (FUS) in DNA Repair: Tango with Poly(ADP-ribose) Polymerase 1 and Compartmentalisation of Damaged DNA,” International Journal of Molecular Sciences 21, no. 19 (2020): 7020.

[487]

M. Chiusa, W. Hu, J. Zienkiewicz, et al., “EGF Receptor-mediated FUS Phosphorylation Promotes Its Nuclear Translocation and Fibrotic Signaling,” Journal of Cell Biology 219, no. 9 (2020): e202001120.

[488]

Q. Wang, Y. Sun, Q. Zhao, et al., “Circular RNAs in Pulmonary Hypertension: Emerging Biological Concepts and Potential Mechanism,” Animal Models and Experimental Medicine 5, no. 1 (2022): 38–47.

[489]

W. Jia, S. H. Kim, M. A. Scalf, et al., “Fused in Sarcoma Regulates DNA Replication Timing and Kinetics,” Journal of Biological Chemistry 297, no. 3 (2021): 101049.

[490]

W. P. Shi, W. J. Lin, W. Y. Ge, et al., “Curcumin Inhibits Liquid-liquid Phase Separation of Fused in Sarcoma and Attenuates the Sequestration of Pyruvate Kinase to Restore Cellular Metabolism,” Food & Function 14, no. 10 (2023): 4621–4631.

[491]

S. J. Cha, S. Lee, H. J. Choi, et al., “Therapeutic Modulation of GSTO Activity Rescues FUS-associated Neurotoxicity via Deglutathionylation in ALS Disease Models,” Developmental Cell 57, no. 6 (2022): 783–798.e8.

[492]

L. Wang, “MiR-141-3p Overexpression Suppresses the Malignancy of Osteosarcoma by Targeting FUS to Degrade LDHB,” Bioscience Reports 40, no. 6 (2020): BSR20193404.

[493]

S. Bian, “miR-4319 Inhibited the Development of Thyroid Cancer by Modulating FUS-stabilized SMURF1,” Journal of Cellular Biochemistry 121, no. 1 (2020): 174–182.

[494]

L. Wang, Y. Liu, H. Li, et al., “miR-4478 Sensitizes Ovarian Cancer Cells to Irradiation by Inhibiting Fus and Attenuating Autophagy,” Molecular Therapy Nucleic Acids 23 (2021): 1110–1119.

[495]

X. Li, X. Wang, Z. Cheng, and Q. Zhu, “AGO2 and Its Partners: a Silencing Complex, a Chromatin Modulator, and New Features,” Critical Reviews in Biochemistry and Molecular Biology 55, no. 1 (2020): 33–53.

[496]

M. Connolly, B. E. Garfield, A. Crosby, N. W. Morrell, S. J. Wort, and P. R. Kemp, “miR-322-5p Targets IGF-1 and Is Suppressed in the Heart of Rats with Pulmonary Hypertension,” FEBS Open Bio 8, no. 3 (2018): 339–348.

[497]

J. Zhan, K. Jin, R. Xie, et al., “Ago2 Protects against Diabetic Cardiomyopathy by Activating Mitochondrial Gene Translation,” Circulation 149, no. 14 (2023): 1102–1120.

[498]

M. Glaß and S. Hüttelmaier, “IGF2BP1-an Oncofetal RNA-binding Protein Fuels Tumor Virus Propagation,” Viruses. 15, no. 7 (2023): 1431.

[499]

K. Belanger, C. A. Nutter, J. Li, et al., “CELF1 contributes to Aberrant Alternative Splicing Patterns in the Type 1 Diabetic Heart,” Biochemical and Biophysical Research Communications 503, no. 4 (2018): 3205–3211.

[500]

M. D. Díaz-Muñoz, V. Y. Kiselev, N. Le Novère, T. Curk, J. Ule, and M. Turner, “Tia1 dependent Regulation of mRNA Subcellular Location and Translation Controls p53 Expression in B Cells,” Nature Communications 8, no. 1 (2017): 530.

[501]

B. M. Hussen, M. Taheri, R. K. Yashooa, et al., “Revolutionizing Medicine: Recent Developments and Future Prospects in Stem-cell Therapy,” International Journal of Surgery 110, no. 12 (2024): 8002–8024.

[502]

K. P. Maremanda, I. K. Sundar, and I. Rahman, “Protective Role of Mesenchymal Stem Cells and Mesenchymal Stem Cell-derived Exosomes in Cigarette Smoke-induced Mitochondrial Dysfunction in Mice,” Toxicology and Applied Pharmacology 385 (2019): 114788.

[503]

M. Zhao, S. Liu, C. Wang, et al., “Mesenchymal Stem Cell-derived Extracellular Vesicles Attenuate Mitochondrial Damage and Inflammation by Stabilizing Mitochondrial DNA,” ACS Nano 15, no. 1 (2021): 1519–1538.

[504]

S. Yao, M. Pang, Y. Wang, et al., “Mesenchymal Stem Cell Attenuates Spinal Cord Injury by Inhibiting Mitochondrial Quality Control-associated Neuronal Ferroptosis,” Redox Biology 67 (2023): 102871.

[505]

R. Zhu, T. Yan, Y. Feng, et al., “Mesenchymal Stem Cell Treatment Improves Outcome of COVID-19 Patients via Multiple Immunomodulatory Mechanisms,” Cell Research 31, no. 12 (2021): 1244–1262.

[506]

M. Zhang, Q. Liu, H. Meng, et al., “Ischemia-reperfusion Injury: Molecular Mechanisms and Therapeutic Targets,” Signal Transduction and Targeted Therapy 9, no. 1 (2024): 12.

[507]

J. H. Xu, J. P. Liang, C. J. Zhu, and Y. J. Lian, “Mesenchymal Stem Cell-derived Extracellular Vesicles Therapy for Pulmonary Hypertension: a Comprehensive Review of Preclinical Studies,” Journal of Interventional Cardiology 2022 (2022): 5451947.

[508]

L. Hu, J. Wang, D. Lin, et al., “Mesenchymal Stem Cell-derived Nanovesicles as a Credible Agent for Therapy of Pulmonary Hypertension,” American Journal of Respiratory Cell and Molecular Biology 67, no. 1 (2022): 61–75.

[509]

S. Oh, A. Y. Jang, S. Chae, et al., “Comparative Analysis on the Anti-inflammatory/Immune Effect of Mesenchymal Stem Cell Therapy for the Treatment of Pulmonary Arterial Hypertension,” Scientific Reports 11, no. 1 (2021): 2012.

[510]

Y. Huang and B. Zhou, “Mitochondrial Dysfunction in Cardiac Diseases and Therapeutic Strategies,” Biomedicines 11, no. 5 (2023): 1500.

[511]

S. M. Emani, B. L. Piekarski, D. Harrild, P. J. Del Nido, and J. D. McCully, “Autologous Mitochondrial Transplantation for Dysfunction after Ischemia-reperfusion Injury,” Journal of Thoracic and Cardiovascular Surgery 154, no. 1 (2017): 286–289.

[512]

A. Guariento, B. L. Piekarski, I. P. Doulamis, et al., “Autologous Mitochondrial Transplantation for Cardiogenic Shock in Pediatric Patients Following Ischemia-reperfusion Injury,” Journal of Thoracic and Cardiovascular Surgery 162, no. 3 (2021): 992–1001.

[513]

T. Wei, W. Chen, L. Wen, et al., “G Protein-coupled Estrogen Receptor Deficiency Accelerates Liver Tumorigenesis by Enhancing Inflammation and Fibrosis,” Cancer Letters 382, no. 2 (2016): 195–202.

[514]

L. Zhu, J. Zhang, J. Zhou, et al., “Mitochondrial Transplantation Attenuates Hypoxic Pulmonary Hypertension,” Oncotarget 7, no. 31 (2016): 48925–48940.

[515]

R. Xiao, J. Liu, S. Luo, et al., “Orally-administrated Mitochondria Attenuate Pulmonary Hypertension with the Aid of Erythrocytes as Carriers,” Clinical and translational medicine 12, no. 9 (2022): e1033.

[516]

V. J. Clemente-Suárez, A. Martín-Rodríguez, R. Yáñez-Sepúlveda, and J. F. Tornero-Aguilera, “Mitochondrial Transfer as a Novel Therapeutic Approach in Disease Diagnosis and Treatment,” International Journal of Molecular Sciences 24, no. 10 (2023): 8848.

[517]

I. Ahmad, A. Valverde, F. Ahmad, and A. R. Naqvi, “Long Noncoding RNA in Myeloid and Lymphoid Cell Differentiation, Polarization and Function,” Cells 9, no. 2 (2020): 269.

[518]

R. B. Perry and I. Ulitsky, “The Functions of Long Noncoding RNAs in Development and Stem Cells,” Development (Cambridge, England) 143, no. 21 (2016): 3882–3894.

[519]

D. Busscher, R. A. Boon, and R. P. Juni, “The Multifaceted Actions of the lncRNA H19 in Cardiovascular Biology and Diseases,” Clinical Science (London, England: 1979) 136, no. 15 (2022): 1157–1178.

[520]

D. Li, C. Zhang, J. Li, et al., “Long Non-coding RNA MALAT1 Promotes Cardiac Remodeling in Hypertensive Rats by Inhibiting the Transcription of MyoD,” Aging 11, no. 20 (2019): 8792–8809.

[521]

B. Ranchoux, L. D. Harvey, R. J. Ayon, et al., “Endothelial Dysfunction in Pulmonary Arterial Hypertension: an Evolving Landscape (2017 Grover Conference Series),” Pulmonary Circulation 8, no. 1 (2018): 2045893217752912.

[522]

K. M. Michalik, X. You, Y. Manavski, et al., “Long Noncoding RNA MALAT1 Regulates Endothelial Cell Function and Vessel Growth,” Circulation Research 114, no. 9 (2014): 1389–1397.

[523]

C. Ejikeme and Z. Safdar, “Exploring the Pathogenesis of Pulmonary Vascular Disease,” Frontiers in Medicine (Lausanne) 11 (2024): 1402639.

[524]

Y. Qin, G. Yan, Y. Qiao, et al., “Emerging Role of Long Non-coding RNAs in Pulmonary Hypertension and Their Molecular Mechanisms (Review),” Experimental and Therapeutic Medicine 20, no. 6 (2020): 164.

[525]

A. Jusic and Y. Devaux, “Noncoding RNAs in Hypertension,” Hypertension 74, no. 3 (2019): 477–492.

[526]

J. Chery, “RNA Therapeutics: RNAi and Antisense Mechanisms and Clinical Applications,” Postdoc J 4, no. 7 (2016): 35–50.

[527]

J. Wang, Z. Lu, M. G. Wientjes, and J. L. Au, “Delivery of siRNA Therapeutics: Barriers and Carriers,” The Aaps Journal [Electronic Resource] 12, no. 4 (2010): 492–503.

[528]

R. Juliano, M. R. Alam, V. Dixit, and H. Kang, “Mechanisms and Strategies for Effective Delivery of Antisense and siRNA Oligonucleotides,” Nucleic Acids Res. 36, no. 12 (2008): 4158–4171.

[529]

A. van Rijt, E. Stefanek, and K. Valente, “Preclinical Testing Techniques: Paving the Way for New Oncology Screening Approaches,” Cancers (Basel) 15, no. 18 (2023): 4466.

[530]

L. J. Marshall, J. Bailey, M. Cassotta, K. Herrmann, and F. Pistollato, “Poor Translatability of Biomedical Research Using Animals—A Narrative Review,” Alternatives to Laboratory Animals 51, no. 2 (2023): 102–135.

[531]

A. E. Frost, R. J. Barst, M. M. Hoeper, et al., “Long-term Safety and Efficacy of imatinib in Pulmonary Arterial Hypertension,” Journal of Heart and Lung Transplantation 34, no. 11 (2015): 1366–1375.

[532]

D. F. Condon, S. Agarwal, A. Chakraborty, et al., “Novel Mechanisms Targeted by Drug Trials in Pulmonary Arterial Hypertension,” Chest 161, no. 4 (2022): 1060–1072.

[533]

Positioning imatinib for pulmonary arterial hypertension (PIPAH). ClinicalTrials.gov identifier: NCT04416750. Updated October 10, 2023. Accessed January 28, 2025, https://clinicaltrials.gov/ct2/show/NCT04416750.

[534]

R. P. Frantz, R. L. Benza, R. N. Channick, et al., “TORREY, a Phase 2 Study to Evaluate the Efficacy and Safety of Inhaled Seralutinib for the Treatment of Pulmonary Arterial Hypertension,” Pulmonary Circulation 11, no. 4 (2021): 20458940211057071.

[535]

R. P. Frantz, V. V. McLaughlin, S. Sahay, et al., “Seralutinib in Adults with Pulmonary Arterial Hypertension (TORREY): a Randomised, Double-blind, Placebo-controlled Phase 2 Trial,” The Lancet Respiratory Medicine 12, no. 7 (2024): 523–534.

[536]

A. Andruska and E. Spiekerkoetter, “Consequences of BMPR2 Deficiency in the Pulmonary Vasculature and beyond: Contributions to Pulmonary Arterial Hypertension,” International Journal of Molecular Sciences 19, no. 9 (2018): 2499.

[537]

Z. C. Lyu, L. Wang, J. H. Lin, et al., “The Features of Rare Pathogenic BMPR2 Variants in Pulmonary Arterial Hypertension: Comparison between Patients and Reference Population,” International Journal of Cardiology 318 (2020): 138–143.

[538]

X. J. Wang, T. Y. Lian, X. Jiang, et al., “Germline BMP9 Mutation Causes Idiopathic Pulmonary Arterial Hypertension,” European Respiratory Journal 53, no. 3 (2019): 1801609.

[539]

L. M. Yung, P. Yang, S. Joshi, et al., “ACTRIIA-Fc Rebalances Activin/GDF versus BMP Signaling in Pulmonary Hypertension,” Science Translational Medicine 12, no. 543 (2020): eaaz5660.

[540]

M. Humbert, V. McLaughlin, J. S. R. Gibbs, et al., “Sotatercept for the Treatment of Pulmonary Arterial Hypertension,” New England Journal of Medicine 384, no. 13 (2021): 1204–1215.

[541]

R. A. Lewis, C. Durrington, R. Condliffe, and D. G. Kiely, “BNP/NT-proBNP in Pulmonary Arterial Hypertension: Time for Point-of-care Testing?,” European Respiratory Review 29, no. 156 (2020): 200009.

[542]

A. B. Waxman, D. M. Systrom, S. Manimaran, J. de Oliveira Pena, J. Lu, and F. P. Rischard, “SPECTRA Phase 2b Study: Impact of sotatercept on Exercise Tolerance and Right Ventricular Function in Pulmonary Arterial Hypertension,” Circulation: Heart Failure 17, no. 5 (2024): e011227.

[543]

M. M. Hoeper, D. B. Badesch, H. A. Ghofrani, et al., “Phase 3 Trial of sotatercept for Treatment of Pulmonary Arterial Hypertension,” New England Journal of Medicine 388, no. 16 (2023): 1478–1490.

[544]

K. Liao, H. Mackenzie, S. Ait-Oudhia, et al., “The Impact of Immunogenicity on the Pharmacokinetics, Efficacy, and Safety of sotatercept in a Phase III Study of Pulmonary Arterial Hypertension,” Clinical Pharmacology & Therapeutics 115, no. 3 (2024): 478–487.

[545]

S. M. Kawut, C. L Archer-Chicko, A. DeMichele, et al., “Anastrozole in Pulmonary Arterial Hypertension. A Randomized, Double-blind, Placebo-controlled Trial,” American Journal of Respiratory and Critical Care Medicine 195, no. 3 (2017): 360–368.

[546]

S. M. Kawut, R. Feng, S. S. Ellenberg, et al., “Pulmonary Hypertension and Anastrozole (PHANTOM): a Randomized, Double-Blind, Placebo-Controlled Trial,” American Journal of Respiratory and Critical Care Medicine 210, no. 9 (2024): 1143–1151.

[547]

Tamoxifen therapy to treat pulmonary arterial hypertension (T2PAH). ClinicalTrials.gov identifier: NCT03528902. Updated August 07, 2024. Accessed January 28, 2025, https://clinicaltrials.gov/ct2/show/NCT03528902.

[548]

Subcutaneous elafin in healthy subjects. ClinicalTrials.gov ClinicalTrials.gov. identifier: NCT03522935. Updated August 15, 2024. Accessed January 28, 2025, https://clinicaltrials.gov/ct2/show/NCT03522935.

[549]

Pulmonary hypertension: assessment of cell therapy (PHACeT). ClinicalTrials.gov identifier: NCT00469027. Updated October 17, 2016. Accessed January 28, 2025, https://clinicaltrials.gov/ct2/show/NCT00469027.

[550]

J. Granton, D. Langleben, M. B. Kutryk, et al., “Endothelial NO-synthase Gene-enhanced Progenitor Cell Therapy for Pulmonary Arterial Hypertension: the PHACeT Trial,” Circulation Research 117, no. 7 (2015): 645–654.

[551]

Study of angiogenic cell therapy for progressive pulmonary hypertension: intervention with repeat dosing of eNOS-enhanced EPCs (SAPPHIRE). ClinicalTrials.gov identifier: NCT03001414. Updated February 01, 2021. Accessed January 28, 2025, https://clinicaltrials.gov/ct2/show/NCT03001414

[552]

A. Olschewski, E. M. Berghausen, C. A. Eichstaedt, et al., “Pathobiology, Pathology and Genetics of Pulmonary Hypertension: Update from the Cologne Consensus Conference 2018,” International Journal of Cardiology 272s (2018): 4–10.

[553]

Apabetalone for pulmonary arterial hypertension: a pilot study (APPRoAcH-p). ClinicalTrials.gov identifier: NCT03655704. Updated April 25, 2022. Accessed January 28, 2025, https://clinicaltrials.gov/ct2/show/NCT03655704.

[554]

K. M. Holmström, L. Baird, Y. Zhang, et al., “Nrf2 impacts Cellular Bioenergetics by Controlling Substrate Availability for Mitochondrial Respiration,” Biol Open 2, no. 8 (2013): 761–770.

[555]

R. J. Oudiz, C. Meyer, M. Chin, et al., “Results of Interim Analysis of the Efficacy and Safety of Bardoxolone Methyl in Patients with Pulmonary Arterial Hypertension Associated with Connective Tissue Disease (CTD) (the lariat study),” American Journal of Respiratory and Critical Care Medicine 195 (2017): A6869.

[556]

Bardoxolone methyl evaluation in patients with pulmonary hypertension (PH)— LARIAT. ClinicalTrials.gov identifier: NCT02036970. Updated February 05, 2024. Accessed January 28, 2025, https://clinicaltrials.gov/ct2/show/NCT02036970.

[557]

Bardoxolone methyl in patients with connective tissue disease-associated pulmonary arterial hypertension—CATALYST. ClinicalTrials.gov identifier: NCT02657356. Updated February 06, 2024. Accessed January 28, 2025, https://clinicaltrials.gov/ct2/show/NCT02657356.

[558]

Extended access program to assess long-term safety of bardoxolone methyl in patients with pulmonary hypertension RANGER (RANGER). ClinicalTrials.gov identifier: NCT03068130. Updated February 06, 2021. Accessed January 28, 2025, https://clinicaltrials.gov/ct2/show/NCT03068130.

[559]

M. Bader, “Inhibition of Serotonin Synthesis: a Novel Therapeutic Paradigm,” Pharmacology & Therapeutics 205 (2020): 107423.

[560]

A study of rodatristat ethyl in patients with pulmonary arterial hypertension (ELEVATE2). ClinicalTrials.gov identifier: NCT04712669. Updated November 05, 2023. Accessed January 28, 2025, https://clinicaltrials.gov/ct2/show/NCT04712669.

[561]

O. Sitbon, A. Skride, J. Feldman, et al., “Safety and Efficacy of Rodatristat ethyl for the Treatment of Pulmonary Arterial Hypertension (ELEVATE-2): a Dose-ranging, Randomised, Multicentre, Phase 2b Trial,” The Lancet Respiratory Medicine 12, no. 11 (2024): 865–876.

[562]

E. K. Bajwa, D. Cislak, A. Kumar, et al., “Phase 1 Study of MK-5475, an Inhaled Soluble Guanylate Cyclase Stimulator, in Participants with Pulmonary Hypertension Associated with Chronic Obstructive Pulmonary Disease,” International Journal of Chronic Obstructive Pulmonary Disease 19 (2024): 1105–1121.

[563]

K. Hosokawa, H. Watanabe, Y. Taniguchi, et al., “A Multicenter, Single-blind, Randomized, warfarin-controlled Trial of edoxaban in Patients with Chronic Thromboembolic Pulmonary Hypertension: KABUKI Trial,” Circulation 149, no. 5 (2024): 406–409.

[564]

H. A. Ghofrani, G. Simonneau, A. M. D’Armini, et al., “Macitentan for the Treatment of Inoperable Chronic Thromboembolic Pulmonary Hypertension (MERIT-1): Results from the Multicentre, Phase 2, Randomised, Double-blind, Placebo-controlled Study,” The Lancet Respiratory Medicine 12, no. 4 (2024): e21–e30.

[565]

M. M. Hoeper, B. Oerke, M. Wissmüller, et al., “Tadalafil for Treatment of Combined Postcapillary and Precapillary Pulmonary Hypertension in Patients with Heart Failure and Preserved Ejection Fraction: a Randomized Controlled Phase 3 Study,” Circulation 150, no. 8 (2024): 600–610.

[566]

E. Grünig, P. Jansa, F. Fan, et al., “Randomized Trial of macitentan/tadalafil Single-tablet Combination Therapy for Pulmonary Arterial Hypertension,” Journal of the American College of Cardiology 83, no. 4 (2024): 473–484.

[567]

K. Schlosser, J. Hanson, P. J. Villeneuve, et al., “Assessment of Circulating lncRNAs under Physiologic and Pathologic Conditions in Humans Reveals Potential Limitations as Biomarkers,” Scientific Reports 6 (2016): 36596.

RIGHTS & PERMISSIONS

2025 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

306

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/