W. Northway, R. C. Rosan, and D. Y. Porter, Pulmonary Disease Following Respirator Therapy of Hyaline-Membrane Disease, N Engl J Med, vol.276, issue.7, pp.357-68, 1967.

B. W. Buczynski, E. T. Maduekwe, O. Reilly, and M. A. , The Role of Hyperoxia in the Pathogenesis of Experimental BPD, Semin Perinatol, vol.37, issue.2, pp.69-78, 2013.

P. H. Jarreau and C. Delacourt, La dysplasie bronchopulmonaire : physiopathologie et évidences pour la prévention et la prise en charge, Néonatologie : bases scientifiques, pp.283-299, 2016.

A. H. Jobe, E. Bancalari, and . Bronchopulmonary-dysplasia, Am J Respir Crit Care Med, vol.163, issue.7, pp.1723-1732, 2001.

M. C. Walsh, D. Wilson-costello, A. Zadell, N. Newman, and A. Fanaroff, Safety, Reliability, and Validity of a Physiologic Definition of Bronchopulmonary Dysplasia, J Perinatol, vol.23, issue.6, pp.451-457, 2003.

P. Ancel, F. Goffinet, P. Kuhn, B. Langer, J. Matis et al., Survival and Morbidity of Preterm Children Born at 22 Through 34 Weeks' Gestation in France in 2011: Results of the EPIPAGE-2 Cohort Study, JAMA Pediatr, vol.169, issue.3, pp.230-238, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01258705

P. R. Chess, D. Angio, C. T. Pryhuber, G. S. Maniscalco, and W. M. , Pathogenesis of Bronchopulmonary Dysplasia, Semin Perinatol, vol.30, issue.4, pp.171-179, 2006.

A. Clement, Malformations du Système Respiratoire

, RespiRare Centre de référence des maladies respiratoires rares

. Disponible,

T. A. Parker and S. H. Abman, The pulmonary circulation in bronchopulmonary dysplasia, Semin Neonatol, vol.8, issue.1, pp.51-61, 2003.

J. P. Kinsella, A. Greenough, and S. H. Abman, Bronchopulmonary dysplasia. The Lancet, vol.367, pp.1421-1452, 2006.

A. J. Bhatt, G. S. Pryhuber, H. Huyck, R. H. Watkins, L. A. Metlay et al., Disrupted Pulmonary Vasculature and Decreased Vascular Endothelial Growth Factor, Flt-1, and TIE-2 in Human Infants Dying with Bronchopulmonary Dysplasia, Am J Respir Crit Care Med, vol.164, issue.10, pp.1971-80, 2001.

M. Jakkula, T. Cras, S. Gebb, K. P. Hirth, R. M. Tuder et al., Inhibition of angiogenesis decreases alveolarization in the developing rat lung, Am J Physiol-Lung Cell Mol Physiol, vol.279, issue.3, pp.600-607, 2000.

V. Bhandari and J. R. Gruen, The Genetics of Bronchopulmonary Dysplasia, Semin Perinatol, vol.30, issue.4, pp.185-91, 2006.

P. M. Lavoie, C. Pham, and K. L. Jang, Heritability of Bronchopulmonary Dysplasia, Defined According to the Consensus Statement of the National Institutes of Health, Pediatrics, vol.122, issue.3, pp.479-85, 2008.

J. Li, K. Yu, J. Oehlert, L. L. Jeliffe-pawlowski, J. B. Gould et al., Exome Sequencing of Neonatal Blood Spots and the Identification of Genes Implicated in Bronchopulmonary Dysplasia, Am J Respir Crit Care Med, vol.192, issue.5, pp.589-96, 2015.

A. Hadchouel, X. Durrmeyer, E. Bouzigon, R. Incitti, J. Huusko et al., Identification of SPOCK2 As a Susceptibility Gene for Bronchopulmonary Dysplasia, Am J Respir Crit Care Med, vol.184, issue.10, pp.1164-70, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00617002

A. Hilgendorff, C. Apitz, D. Bonnet, and G. Hansmann, Pulmonary hypertension associated with acute or chronic lung diseases in the preterm and term neonate and infant. The European Paediatric Pulmonary Vascular Disease Network, endorsed by ISHLT and DGPK, Heart, vol.102, issue.2, pp.49-56, 2016.

G. Altit, A. Dancea, C. Renaud, T. Perreault, L. C. Lands et al., screening and diagnosis of pulmonary hypertension in infants with bronchopulmonary dysplasia-A review of the literature, Paediatr Respir Rev, vol.23, pp.16-26, 2017.

P. M. Mourani, M. Mullen, and S. H. Abman, Pulmonary hypertension in bronchopulmonary dysplasia, Prog Pediatr Cardiol, vol.27, issue.1, pp.43-51, 2009.

E. Khemani, D. B. Mcelhinney, L. Rhein, O. Andrade, R. V. Lacro et al., Pulmonary Artery Hypertension in Formerly Premature Infants With Bronchopulmonary Dysplasia: Clinical Features and Outcomes in the Surfactant Era, Pediatrics, vol.120, issue.6, pp.1260-1269, 2007.

A. Dasgupta, L. Bowman, D. 'arsigny, C. Archer, and S. , Soluble Guanylate Cyclase: A New Therapeutic Target for Pulmonary Arterial Hypertension and Chronic Thromboembolic Pulmonary Hypertension, Clin Pharmacol Ther, vol.97, issue.1, pp.88-102, 2015.

S. K. Berkelhamer, K. K. Mestan, and R. H. Steinhorn, Pulmonary hypertension in bronchopulmonary dysplasia, Semin Perinatol, vol.37, issue.2, pp.124-155, 2013.

E. Dumas-de-la-roque, L. Storme, P. Mauriat, and S. Bonnet, Hypertension pulmonaire de l'enfant et du nouveau-né en réanimation. Partie II : diagnostic et traitement, Arch Pédiatrie, vol.18, issue.2, pp.195-203, 2011.

S. H. Abman, G. Hansmann, S. L. Archer, D. D. Ivy, I. Adatia et al., Pediatric Pulmonary Hypertension: Guidelines From the American Heart Association, Circulation, vol.132, issue.21, pp.2037-99, 2015.
DOI : 10.1161/cir.0000000000000329

P. M. Mourani, M. K. Sontag, A. Younoszai, D. D. Ivy, and S. H. Abman, Clinical Utility of Echocardiography for the Diagnosis and Management of Pulmonary Vascular Disease in Young Children With Chronic Lung Disease, Pediatrics, vol.121, issue.2, pp.317-342, 2008.

J. Stocks, A. Hislop, and S. Sonnappa, Early lung development: lifelong effect on respiratory health and disease, Lancet Respir Med, vol.1, issue.9, pp.728-770, 2013.
DOI : 10.1016/s2213-2600(13)70118-8

E. Baraldi and M. Filippone, Chronic Lung Disease after Premature Birth, N Engl J Med, vol.357, pp.1946-55, 2007.

E. Baraldi, S. Carraro, and M. Filippone, Bronchopulmonary dysplasia: Definitions and long-term respiratory outcome, Early Hum Dev, vol.85, issue.10, pp.1-3, 2009.
DOI : 10.1016/j.earlhumdev.2009.08.002

S. J. Kotecha, M. O. Edwards, W. J. Watkins, A. J. Henderson, S. Paranjothy et al., Effect of preterm birth on later FEV1: a systematic review and meta-analysis, Thorax, vol.68, issue.8, pp.760-766, 2013.

J. Fawke, S. Lum, J. Kirkby, E. Hennessy, N. Marlow et al., Lung Function and Respiratory Symptoms at 11 Years in Children Born Extremely Preterm, Am J Respir Crit Care Med, vol.182, issue.2, pp.237-282, 2010.

F. D. Martinez, Early-Life Origins of Chronic Obstructive Pulmonary Disease, N Engl J Med, vol.375, issue.9, pp.871-879, 2016.

. Sonnenschein-van-der, A. Voort, L. R. Arends, J. C. De-jongste, I. Annesi-maesano et al., Preterm birth, infant weight gain, and childhood asthma risk: A meta-analysis of 147,000 European children, J Allergy Clin Immunol, vol.133, issue.5, pp.1317-1346, 2014.

M. Renard and P. Truffert, Pronostic respiratoire clinique à 5 ans du grand prématuré, Cohorte EPIPAGE. Arch Pédiatrie, vol.15, issue.5, pp.592-596, 2008.
DOI : 10.1016/s0929-693x(08)71844-x

A. Lamarche-vadel, B. Blondel, P. Truffert, A. Burguet, G. Cambonie et al., Rehospitalization in infants younger than 29 weeks' gestation in the EPIPAGE cohort, Acta Paediatrica, vol.93, issue.10, pp.1340-1345, 2004.

D. Silva, C. Nardiello, A. Pozarska, and R. E. Morty, Recent advances in the mechanisms of lung alveolarization and the pathogenesis of bronchopulmonary dysplasia, Am J Physiol-Lung Cell Mol Physiol, vol.309, issue.11, pp.1239-72, 2015.

L. M. Davidson and S. K. Berkelhamer, Bronchopulmonary Dysplasia: Chronic Lung Disease of Infancy and Long-Term Pulmonary Outcomes, J Clin Med, vol.6, issue.1, p.4, 2017.

C. Karila, J. Saulnier, C. Elie, P. Taupin, P. Scheinmann et al., Hypoventilation alvéolaire à l'exercice chez des enfants avec dysplasie bronchopulmonaire, Rev Mal Respir, vol.25, issue.3, pp.303-315, 2008.
DOI : 10.1016/s0761-8425(08)71549-3

E. J. Short, N. K. Klein, B. A. Lewis, S. Fulton, S. Eisengart et al., Cognitive and Academic Consequences of Bronchopulmonary Dysplasia and Very Low Birth Weight: 8-YearOld Outcomes, Pediatrics, vol.112, issue.5, pp.359-359, 2003.

H. Nakanishi, A. Uchiyama, and S. Kusuda, Impact of pulmonary hypertension on neurodevelopmental outcome in preterm infants with bronchopulmonary dysplasia: a cohort study, J Perinatol, vol.36, issue.10, p.890, 2016.

J. K. Trittmann, L. D. Nelin, and M. A. Klebanoff, Bronchopulmonary dysplasia and neurodevelopmental outcome in extremely preterm neonates, Eur J Pediatr, vol.172, issue.9, pp.1173-80, 2013.

L. W. Doyle, J. L. Cheong, R. A. Ehrenkranz, and H. L. Halliday, Early (< 8 days) systemic postnatal corticosteroids for prevention of bronchopulmonary dysplasia in preterm infants, Cochrane Database of Systematic Reviews, vol.10, p.1146, 2017.

M. X. Rojas-reyes, C. J. Morley, and R. Soll, Prophylactic versus selective use of surfactant in preventing morbidity and mortality in preterm infants, Cochrane Database of Systematic Reviews, vol.3, p.510, 2012.

N. Seger and R. Soll, Animal derived surfactant extract for treatment of respiratory distress syndrome, Cochrane Database of Systematic Reviews, vol.2, p.7836, 2009.

T. Isayama, H. Iwami, S. Mcdonald, and J. Beyene, Association of Noninvasive Ventilation Strategies With Mortality and Bronchopulmonary Dysplasia Among Preterm Infants: A Systematic Review and Meta-analysis, JAMA, vol.316, issue.6, pp.611-635, 2016.

D. G. Sweet, V. Carnielli, G. Greisen, M. Hallman, E. Ozek et al., European Consensus Guidelines on the Management of Neonatal Respiratory Distress Syndrome in Preterm Infants2013 Update, Neonatology, vol.103, issue.4, pp.353-68, 2013.

, Committee on Fetus and Newborn. Respiratory Support in Preterm Infants at, Birth. Pediatrics, vol.133, issue.1, pp.171-175, 2014.

M. Keszler, Mechanical ventilation strategies, Semin Fetal Neonatal Med, vol.22, issue.4, pp.267-74, 2017.

, Early CPAP versus Surfactant in Extremely Preterm Infants, N Engl J Med, vol.362, issue.21, pp.1970-1979, 2010.

V. Bhandari, The potential of non-invasive ventilation to decrease BPD, Semin Perinatol, vol.37, issue.2, pp.108-122, 2013.

H. Kirpalani, D. Millar, B. Lemyre, B. A. Yoder, A. Chiu et al., A Trial Comparing Noninvasive Ventilation Strategies in Preterm Infants, N Engl J Med, vol.369, issue.7, pp.611-631, 2013.

S. H. Abman, J. M. Collaco, E. G. Shepherd, M. Keszler, M. Cuevas-guaman et al., Interdisciplinary Care of Children with Severe Bronchopulmonary Dysplasia, J Pediatr, vol.181, pp.12-28, 2017.
DOI : 10.1016/j.jpeds.2016.10.082

URL : http://europepmc.org/articles/pmc5562402?pdf=render

A. Greenough, Synchronized mechanical ventilation for respiratory support in newborn infants, Cochrane Database of Systematic Reviews, vol.9, p.456, 2016.

E. Lopez and P. H. Jarreau, Les différents modes de ventilation : quelles évidences ? In: Néonatologie : bases scientifiques, pp.137-147, 2016.
DOI : 10.1016/b978-2-294-73742-8.00013-3

C. Klingenberg, K. I. Wheeler, N. Mccallion, C. J. Morley, and P. G. Davis, Volume-targeted versus pressure-limited ventilation in neonates, Cochrane Database of Systematic Reviews, vol.10, p.3666, 2017.
DOI : 10.1002/14651858.cd003666.pub4

URL : https://www.cochranelibrary.com/cdsr/doi/10.1002/14651858.CD003666.pub4/media/CDSR/CD003666/CD003666.pdf

F. Cools, M. Offringa, and L. M. Askie, Elective high frequency oscillatory ventilation versus conventional ventilation for acute pulmonary dysfunction in preterm infants, Cochrane Database of Systematic Reviews, vol.3, p.104, 2015.

H. Stein, H. Alosh, P. Ethington, and D. B. White, Prospective crossover comparison between NAVA and pressure control ventilation in premature neonates less than 1500 grams, J Perinatol, vol.33, issue.6, p.452, 2013.
DOI : 10.1038/jp.2012.136

URL : https://www.nature.com/articles/jp2012136.pdf

J. Lee, H. Kim, J. A. Sohn, J. A. Lee, C. W. Choi et al., Randomized Crossover Study of Neurally Adjusted Ventilatory Assist in Preterm Infants, J Pediatr, vol.161, issue.5, pp.808-813, 2012.

T. E. Rossor, K. A. Hunt, S. Shetty, and A. Greenough, Neurally adjusted ventilatory assist compared to other forms of triggered ventilation for neonatal respiratory support, Cochrane Database of Systematic Reviews, vol.6, p.12251, 2017.

E. N. Kraybill, D. K. Runyan, C. L. Bose, and J. H. Khan, Risk factors for chronic lung disease in infants with birth weights of 751 to 1000 grams, J Pediatr, vol.115, issue.1, pp.115-135, 1989.

J. S. Garland, R. K. Buck, E. N. Allred, and A. Leviton, Hypocarbia Before Surfactant Therapy Appears to Increase Bronchopulmonary Dysplasia Risk in Infants With Respiratory Distress Syndrome, Arch Pediatr Adolesc Med, vol.149, issue.6, pp.617-639, 1995.
DOI : 10.1001/archpedi.1995.02170190027005

L. J. Graziani, A. R. Spitzer, D. G. Mitchell, D. A. Merton, C. Stanley et al., Mechanical Ventilation in Preterm Infants: Neurosonographic and Developmental Studies, Pediatrics, vol.90, issue.4, pp.515-537, 1992.

S. Fujimoto, H. Togari, N. Yamaguchi, F. Mizutani, S. Suzuki et al., Hypocarbia and cystic periventricular leukomalacia in premature infants, Arch Dis Child, vol.71, issue.2, pp.107-110, 1994.
DOI : 10.1136/fn.71.2.f107

URL : https://fn.bmj.com/content/fetalneonatal/71/2/F107.full.pdf

P. G. Woodgate and M. W. Davies, Permissive hypercapnia for the prevention of morbidity and mortality in mechanically ventilated newborn infants, Cochrane Database of Systematic Reviews, vol.2, p.2061, 2001.

U. H. Thome, O. Genzel-boroviczeny, B. Bohnhorst, M. Schmid, H. Fuchs et al., Permissive hypercapnia in extremely low birthweight infants (PHELBI): a randomised controlled multicentre trial, Lancet Respir Med, vol.3, issue.7, pp.534-577, 2015.

U. H. Thome, O. Genzel-boroviczeny, B. Bohnhorst, M. Schmid, H. Fuchs et al., Neurodevelopmental outcomes of extremely low birthweight infants randomised to different PCO2 targets: the PHELBI follow-up study, Arch Dis Child-Fetal Neonatal Ed, vol.102, issue.5, pp.376-82, 2017.

O. D. Saugstad and D. Aune, Optimal Oxygenation of Extremely Low Birth Weight Infants: A MetaAnalysis and Systematic Review of the Oxygen Saturation Target Studies, Neonatology, vol.105, issue.1, pp.55-63, 2014.

L. M. Askie, B. A. Darlow, P. G. Davis, N. Finer, B. Stenson et al., Effects of targeting lower versus higher arterial oxygen saturations on death or disability in preterm infants, Cochrane Database of Systematic Reviews, vol.4, p.11190, 2017.

P. Tarazona, S. , R. Esteban, S. , A. Diego et al., Guidelines for the follow up of patients with bronchopulmonary dysplasia. An Pediatría Engl Ed, vol.84, pp.61-62, 2016.

L. Ma, P. Zhou, J. Neu, and H. Lin, Potential Nutrients for Preventing or Treating Bronchopulmonary Dysplasia, Paediatr Respir Rev, vol.22, pp.83-91, 2017.

S. Wedgwood, C. Warford, S. C. Agvateesiri, P. Thai, S. K. Berkelhamer et al., Postnatal growth restriction augments oxygen-induced pulmonary hypertension in a neonatal rat model of bronchopulmonary dysplasia, Pediatr Res, vol.80, issue.6, pp.894-902, 2016.

M. Mataloun, C. M. Rebello, R. S. Mascaretti, M. Dohlnikoff, and C. R. Leone, Pulmonary responses to nutritional restriction and hyperoxia in premature rabbits, J Pediatr (Rio J), vol.82, issue.3, pp.179-85, 2006.

E. Zana-taïeb, L. Butruille, M. L. Franco-montoya, E. Lopez, F. Vernier et al., Effect of two models of intrauterine growth restriction on alveolarization in rat lungs : morphometric and gene expression analysis, PLoS ONE, vol.8, issue.11, 2013.

L. Ma, N. Li, X. Liu, L. Shaw, L. Calzi et al., Arginyl-glutamine dipeptide or docosahexaenoic acid attenuate hyperoxia-induced lung injury in neonatal mice, Nutrition, vol.28, issue.11, pp.1186-91, 2012.

L. K. Rogers, C. J. Valentine, M. Pennell, M. Velten, R. D. Britt et al., Maternal Docosahexaenoic Acid Supplementation Decreases Lung Inflammation in Hyperoxia-Exposed Newborn Mice, J Nutr, vol.141, issue.2, pp.214-236, 2011.

P. Zhang, P. M. Lavoie, T. Lacaze-masmonteil, M. Rhainds, and I. Marc, Omega-3 Long-Chain Polyunsaturated Fatty Acids for Extremely Preterm Infants: A Systematic Review, Pediatrics, vol.134, issue.1, pp.120-154, 2014.

C. T. Collins, M. Makrides, A. J. Mcphee, T. R. Sullivan, P. G. Davis et al., Docosahexaenoic Acid and Bronchopulmonary Dysplasia in Preterm Infants, N Engl J Med, vol.376, issue.13, pp.1245-55, 2017.

, Maternal Omega-3 Supplementation to Reduce Bronchopulmonary Dysplasia-Full Text ViewClinicalTrials.gov

. Disponible,

K. J. Barrington, E. Fortin-pellerin, and T. Pennaforte, Fluid restriction for treatment of preterm infants with chronic lung disease, Cochrane Database of Systematic Reviews, vol.2, p.5389, 2017.

R. I. Clyman, The role of patent ductus arteriosus and its treatments in the development of bronchopulmonary dysplasia, Semin Perinatol, vol.37, issue.2, pp.102-109, 2013.

D. Mccurnin, S. Seidner, L. Chang, N. Waleh, M. Ikegami et al., IbuprofenInduced Patent Ductus Arteriosus Closure: Physiologic, Histologic, and Biochemical Effects on the Premature Lung, Pediatrics, vol.121, issue.5, pp.945-56, 2008.

N. Waleh, D. C. Mccurnin, B. A. Yoder, P. W. Shaul, and R. I. Clyman, Patent Ductus Arteriosus Ligation Alters Pulmonary Gene Expression in Preterm Baboons, Pediatr Res, vol.69, issue.3, p.212, 2011.

D. Roberts, J. Brown, N. Medley, and S. R. Dalziel, Antenatal corticosteroids for accelerating fetal lung maturation for women at risk of preterm birth, Cochrane Database of Systematic Reviews, vol.3, p.4454, 2017.

A. Sotiriadis, A. Tsiami, S. Papatheodorou, A. A. Baschat, K. Sarafidis et al., Neurodevelopmental Outcome After a Single Course of Antenatal Steroids in Children Born Preterm: A Systematic Review and Meta-analysis, Obstet Gynecol, vol.125, issue.6, pp.1385-96, 2015.

O. Baud, C. Trousson, V. Biran, E. Leroy, D. Mohamed et al., Association Between Early Low-Dose Hydrocortisone Therapy in Extremely Preterm Neonates and Neurodevelopmental Outcomes at 2 Years of Age, JAMA, vol.317, issue.13, pp.1329-1366, 2017.

K. L. Watterberg, S. M. Scott, C. Backstrom, K. L. Gifford, and K. L. Cook, Links Between Early Adrenal Function and Respiratory Outcome in Preterm Infants: Airway Inflammation and Patent Ductus Arteriosus, Pediatrics, vol.105, issue.2, pp.320-324, 2000.
DOI : 10.1542/peds.105.2.320

V. S. Shah, A. Ohlsson, H. L. Halliday, and M. Dunn, Early administration of inhaled corticosteroids for preventing chronic lung disease in very low birth weight preterm neonates, Cochrane Database of Systematic Reviews, vol.1, p.1969, 2017.

S. S. Shah, A. Ohlsson, H. L. Halliday, and V. S. Shah, Inhaled versus systemic corticosteroids for preventing bronchopulmonary dysplasia in ventilated very low birth weight preterm neonates, Cochrane Database of Systematic Reviews, vol.10, p.2058, 2017.
DOI : 10.1002/14651858.cd002058.pub3

URL : https://www.cochranelibrary.com/cdsr/doi/10.1002/14651858.CD002058.pub3/media/CDSR/CD002058/CD002058.pdf

S. S. Shah, A. Ohlsson, H. L. Halliday, and V. S. Shah, Inhaled versus systemic corticosteroids for the treatment of bronchopulmonary dysplasia in ventilated very low birth weight preterm infants, Cochrane Database of Systematic Reviews, vol.10, p.2057, 2017.

O. Baud, L. Maury, F. Lebail, D. Ramful, E. Moussawi et al., Effect of early low-dose hydrocortisone on survival without bronchopulmonary dysplasia in extremely preterm infants (PREMILOC): a double-blind, placebo-controlled, multicentre, randomised trial, The Lancet, vol.387, pp.1827-1863, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01278609

L. W. Doyle, J. L. Cheong, R. A. Ehrenkranz, and H. L. Halliday, Late (> 7 days) systemic postnatal corticosteroids for prevention of bronchopulmonary dysplasia in preterm infants, Cochrane Database of Systematic Reviews, vol.10, p.1145, 2017.

W. Onland, M. Offringa, F. Cools, D. Jaegere, A. P. Rademaker et al., Systemic hydrocortisone to prevent bronchopulmonary dysplasia in preterm infants (the SToP-BPD study); a multicenter randomized placebo controlled trial, BMC Pediatr, vol.11, p.102, 2011.

N. Network, Hydrocortisone for BPD-Full Text ViewClinicalTrials.gov

. Disponible,

R. Venkataraman, M. Kamaluddeen, S. U. Hasan, H. L. Robertson, and A. Lodha, Intratracheal Administration of Budesonide-Surfactant in Prevention of Bronchopulmonary Dysplasia in Very Low Birth Weight Infants: A Systematic Review and Meta-Analysis, Pediatr Pulmonol, vol.52, issue.7, pp.968-75, 2017.

C. Fajardo, D. Levin, M. Garcia, D. Abrams, and I. Adamson, Surfactant versus Saline as a Vehicle for Corticosteroid Delivery to the Lungs of Ventilated Rabbits, Pediatr Res, vol.43, issue.4, p.542, 1998.

L. Li, C. Yang, X. Feng, Y. Du, Z. Zhang et al., Effects of intratracheal budesonide during early postnatal life on lung maturity of premature fetal rabbits, Pediatr Pulmonol, vol.53, issue.1, pp.28-35, 2018.

G. Ng, O. Da-silva, and A. Ohlsson, Bronchodilators for the prevention and treatment of chronic lung disease in preterm infants, Cochrane Database of Systematic Reviews, vol.12, p.3214, 2016.

A. N. Macritchie, K. H. Albertine, J. Sun, P. S. Lei, S. C. Jensen et al., Reduced endothelial nitric oxide synthase in lungs of chronically ventilated preterm lambs, Am J PhysiolLung Cell Mol Physiol, vol.281, issue.4, pp.1011-1031, 2001.

S. H. Abman, Impaired Vascular Endothelial Growth Factor Signaling in the Pathogenesis of Neonatal Pulmonary Vascular Disease, Membrane Receptors, Channels and Transporters in Pulmonary Circulation, pp.323-358, 2010.

R. Han, S. Babaei, M. Robb, T. Lee, R. Ridsdale et al., Defective Lung Vascular Development and Fatal Respiratory Distress in Endothelial NO Synthase-Deficient Mice: A Model of Alveolar Capillary Dysplasia?, Circ Res, vol.94, issue.8, pp.1115-1138, 2004.

J. Tang, N. E. Markham, Y. Lin, I. F. Mcmurtry, A. Maxey et al., Inhaled nitric oxide attenuates pulmonary hypertension and improves lung growth in infant rats after neonatal treatment with a VEGF receptor inhibitor, Am J Physiol-Lung Cell Mol Physiol, vol.287, issue.2, pp.344-51, 2004.

Y. Lin, N. E. Markham, V. Balasubramaniam, J. Tang, A. Maxey et al., Inhaled Nitric Oxide Enhances Distal Lung Growth after Exposure to Hyperoxia in Neonatal Rats, Pediatr Res, vol.58, issue.1, pp.22-31, 2005.

S. Afshar, L. L. Gibson, I. S. Yuhanna, T. S. Sherman, J. D. Kerecman et al., Pulmonary NO synthase expression is attenuated in a fetal baboon model of chronic lung disease, Am J Physiol-Lung Cell Mol Physiol, vol.284, issue.5, pp.749-58, 2003.

K. J. Barrington, N. Finer, and T. Pennaforte, Inhaled nitric oxide for respiratory failure in preterm infants, Cochrane Database of Systematic Reviews, vol.1, p.509, 2017.

D. J. Henderson-smart, D. Paoli, and A. G. , Prophylactic methylxanthine for prevention of apnoea in preterm infants, Cochrane Database of Systematic Reviews, vol.12, p.432, 2010.

B. Schmidt, R. S. Roberts, P. Davis, L. W. Doyle, K. J. Barrington et al., Caffeine Therapy for Apnea of Prematurity, N Engl J Med, vol.354, issue.20, pp.2112-2133, 2006.

B. Schmidt, R. S. Roberts, P. Davis, L. W. Doyle, K. J. Barrington et al., Long-Term Effects of Caffeine Therapy for Apnea of Prematurity, N Engl J Med, vol.357, pp.1893-902, 2007.

L. P. Brion, R. A. Primhak, A. , and I. , Diuretics acting on the distal renal tubule for preterm infants with (or developing) chronic lung disease, Cochrane Database of Systematic Reviews, vol.9, p.1817, 2002.

R. M. Viscardi and S. G. Kallapur, Role of Ureaplasma Respiratory Tract Colonization in Bronchopulmonary Dysplasia Pathogenesis: Current Concepts and Update, Clin Perinatol, vol.42, issue.4, pp.719-757, 2015.

J. Lowe, W. J. Watkins, M. O. Edwards, O. B. Spiller, E. Jacqz-aigrain et al., Association Between Pulmonary : Ureaplasma Colonization and Bronchopulmonary Dysplasia in Preterm Infants: Updated Systematic Review and Meta-analysis, Pediatr Infect Dis J, vol.33, issue.7, pp.697-702, 2014.

C. G. Mabanta, G. S. Pryhuber, G. A. Weinberg, and D. Phelps, Erythromycin for the prevention of chronic lung disease in intubated preterm infants at risk for, or colonized or infected with Ureaplasma urealyticum, Cochrane Database of Systematic Reviews, vol.4, p.3744, 2003.

Z. H. Aghai, A. Kode, J. G. Saslow, T. Nakhla, S. Farhath et al., Azithromycin Suppresses Activation of Nuclear Factor-kappa B and Synthesis of Pro-inflammatory Cytokines in Tracheal Aspirate Cells From Premature Infants, Pediatr Res, vol.62, issue.4, p.483, 2007.

P. Howard, Azithromycin-induced proarrhythmia and cardiovascular death, Ann Pharmacother, vol.47, issue.11, pp.1547-51, 2013.
DOI : 10.1177/1060028013504905

V. Nair, P. Loganathan, and A. S. Soraisham, Azithromycin and Other Macrolides for Prevention of Bronchopulmonary Dysplasia: A Systematic Review and Meta-Analysis, Neonatology, vol.106, issue.4, pp.337-384, 2014.

C. Smith, O. Egunsola, I. Choonara, S. Kotecha, E. Jacqz-aigrain et al., Use and safety of azithromycin in neonates: a systematic review, BMJ Open, vol.5, issue.12, p.8194, 2015.

H. O. Ballard, L. A. Shook, P. Bernard, M. I. Anstead, R. Kuhn et al., Use of azithromycin for the prevention of bronchopulmonary dysplasia in preterm infants: a randomized, double-blind, placebo controlled trial, Pediatr Pulmonol, vol.46, issue.2, pp.111-119, 2011.

H. O. Ballard, M. I. Anstead, and L. A. Shook, Azithromycin in the extremely low birth weight infant for the prevention of Bronchopulmonary Dysplasia: a pilot study, Respir Res, vol.8, p.41, 2007.

M. M. Gharehbaghi, A. Peirovifar, M. Ghojazadeh, and M. Mahallei, Efficacy of azithromycin for prevention of bronchopulmonary dysplasia (BDP), Turk J Med Sci, vol.42, issue.6, pp.1070-1075, 2012.

R. Ozdemir, O. Erdeve, E. A. Dizdar, S. S. Oguz, N. Uras et al., Clarithromycin in Preventing Bronchopulmonary Dysplasia in Ureaplasma urealyticum-Positive Preterm Infants, Pediatrics, vol.128, issue.6, pp.1496-501, 2011.

J. E. Tyson, L. L. Wright, W. Oh, K. A. Kennedy, L. Mele et al., Vitamin A Supplementation for Extremely-Low-Birth-Weight Infants, N Engl J Med, vol.340, issue.25, pp.1962-1970, 1999.

C. Agostoni, G. Buonocore, V. Carnielli, M. D. Curtis, D. Darmaun et al., Enteral Nutrient Supply for Preterm Infants: Commentary From the European Society of Paediatric Gastroenterology, Hepatology and Nutrition Committee on Nutrition, J Pediatr Gastroenterol Nutr, vol.50, issue.1, pp.85-91, 2010.

B. A. Darlow, P. J. Graham, and M. X. Rojas-reyes, Vitamin A supplementation to prevent mortality and short-and long-term morbidity in very low birth weight infants, Cochrane Database of Systematic Reviews, vol.8, p.501, 2016.

S. Meyer, K. Kronfeld, S. Gräber, R. Butzer, H. Wahl et al., Vitamin A to prevent bronchopulmonary dysplasia: The NeoVitaA Trial, J Matern Fetal Neonatal Med, vol.26, issue.5, pp.544-549, 2013.

D. L. Phelps, R. M. Ward, R. L. Williams, T. L. Nolen, K. L. Watterberg et al., Safety and pharmacokinetics of multiple dose myo-inositol in preterm infants, Pediatr Res, vol.80, issue.2, p.209, 2016.

M. Hallman, K. Bry, K. Hoppu, M. Lappi, and M. Pohjavuori, Inositol Supplementation in Premature Infants with Respiratory Distress Syndrome, N Engl J Med, vol.326, pp.1233-1242, 1992.

A. Howlett, A. Ohlsson, and N. Plakkal, Inositol in preterm infants at risk for or having respiratory distress syndrome, Cochrane Database of Systematic Reviews, vol.2, p.366, 2015.

, Inositol to Reduce Retinopathy of Prematurity-NCT01954082

. Clinicaltrials,

R. L. Morton, K. C. Das, X. Guo, D. N. Iklé, and C. W. White, Effect of oxygen on lung superoxide dismutase activities in premature baboons with bronchopulmonary dysplasia, Am J PhysiolLung Cell Mol Physiol, vol.276, issue.1, pp.64-74, 1999.

H. K. Poonyagariyagorn, S. Metzger, D. Dikeman, A. L. Mercado, A. Malinina et al., Superoxide Dismutase 3 Dysregulation in a Murine Model of Neonatal Lung Injury, Am J Respir Cell Mol Biol, vol.51, issue.3, pp.380-90, 2014.

W. N. Rosenfeld, J. M. Davis, L. Parton, S. E. Richter, A. Price et al., Safety and Pharmacokinetics of Recombinant Human Superoxide Dismutase Administered Intratracheally to Premature Neonates With Respiratory Distress Syndrome, Pediatrics, vol.97, issue.6, pp.811-818, 1996.

J. M. Davis, Superoxide Dismutase: A Role in the Prevention of Chronic Lung Disease, Biol Neonate, vol.74, pp.29-34, 1998.

G. Suresh, J. M. Davis, and R. Soll, Superoxide dismutase for preventing chronic lung disease in mechanically ventilated preterm infants, Cochrane Database of Systematic Reviews, vol.1, p.1968, 2001.

S. C. Langley and F. J. Kelly, N-acetylcysteine ameliorates hyperoxic lung injury in the preterm guinea pig, Biochem Pharmacol, vol.45, issue.4, pp.841-847, 1993.

L. M. Soghier and L. P. Brion, Cysteine, cystine or N-acetylcysteine supplementation in parenterally fed neonates, Cochrane Database of Systematic Reviews, vol.4, p.4869, 2006.

B. A. Darlow and N. Austin, Selenium supplementation to prevent short-term morbidity in preterm neonates, Cochrane Database of Systematic Reviews, vol.4, p.3312, 2003.

L. P. Brion, E. F. Bell, and T. S. Raghuveer, Vitamin E supplementation for prevention of morbidity and mortality in preterm infants, Cochrane Database of Systematic Reviews, vol.4, p.3665, 2003.

P. Manzoni, R. Guardione, P. Bonetti, C. Priolo, A. Maestri et al., Lutein and Zeaxanthin Supplementation in Preterm Very Low-Birth-Weight Neonates in Neonatal Intensive Care Units: A Multicenter Randomized Controlled Trial, Am J Perinatol, vol.30, issue.01, pp.25-32, 2013.

G. Distefano and P. Sciacca, Molecular physiopathogenetic mechanisms and development of new potential therapeutic strategies in persistent pulmonary hypertension of the newborn, Ital J Pediatr, vol.41, issue.1, p.6, 2015.

V. Balasubramaniam, C. F. Mervis, A. M. Maxey, N. E. Markham, and S. H. Abman, Hyperoxia reduces bone marrow, circulating, and lung endothelial progenitor cells in the developing lung: implications for the pathogenesis of bronchopulmonary dysplasia, Am J Physiol-Lung Cell Mol Physiol, vol.292, issue.5, pp.1073-84, 2007.

C. D. Baker, V. Balasubramaniam, P. M. Mourani, M. K. Sontag, C. P. Black et al., Cord blood angiogenic progenitor cells are decreased in bronchopulmonary dysplasia, Eur Respir J, vol.40, issue.6, pp.1516-1538, 2012.
DOI : 10.1183/09031936.00017312

URL : http://erj.ersjournals.com/content/40/6/1516.full.pdf

V. Balasubramaniam, S. L. Ryan, G. J. Seedorf, E. V. Roth, T. R. Heumann et al., Bone marrow-derived angiogenic cells restore lung alveolar and vascular structure after neonatal hyperoxia in infant mice, Am J Physiol-Lung Cell Mol Physiol, vol.298, issue.3, pp.315-338, 2009.

T. Van-haaften, R. Byrne, S. Bonnet, G. Y. Rochefort, J. Akabutu et al., Airway Delivery of Mesenchymal Stem Cells Prevents Arrested Alveolar Growth in Neonatal Lung Injury in Rats, Am J Respir Crit Care Med, vol.180, issue.11, pp.1131-1173, 2009.

M. Aslam, R. Baveja, O. D. Liang, A. Fernandez-gonzalez, C. Lee et al., Bone Marrow Stromal Cells Attenuate Lung Injury in a Murine Model of Neonatal Chronic Lung Disease, Am J Respir Crit Care Med, vol.180, issue.11, pp.1122-1152, 2009.

Y. S. Chang, S. Y. Ahn, H. S. Yoo, S. I. Sung, S. J. Choi et al., Mesenchymal Stem Cells for Bronchopulmonary Dysplasia: Phase 1 Dose-Escalation Clinical Trial, J Pediatr, vol.164, issue.5, pp.966-972, 2014.

S. Y. Ahn, Y. S. Chang, J. H. Kim, S. I. Sung, and W. S. Park, Two-Year Follow-Up Outcomes of Premature Infants Enrolled in the Phase I Trial of Mesenchymal Stem Cells Transplantation for Bronchopulmonary Dysplasia, J Pediatr, vol.185, pp.49-54, 2017.

S. Augustine, M. T. Avey, B. Harrison, T. Locke, M. Ghannad et al., Mesenchymal Stromal Cell Therapy in Bronchopulmonary Dysplasia: Systematic Review and Meta-Analysis of Preclinical Studies, STEM CELLS Transl Med, vol.6, issue.12, pp.2079-93, 2017.

M. E. Fung and B. Thébaud, Stem cell-based therapy for neonatal lung disease: it is in the juice, Pediatr Res, vol.75, issue.1-1, p.2, 2013.

M. Mueller and B. W. Kramer, Stem cells and Bronchopulmonary Dysplasia-The five questions: Which cells, when, in which dose, to which patients via which route?, Paediatr Respir Rev, vol.24, pp.54-63, 2017.

V. Meau-petit, G. Thouvenin, N. Guillemot-lambert, V. Champion, I. Tillous-borde et al., Hypertension artérielle pulmonaire des anciens grands prématurés bronchodysplasiques, Arch Pédiatrie, vol.20, issue.1, pp.44-53, 2013.

M. Kaestner, D. Schranz, G. Warnecke, C. Apitz, G. Hansmann et al., Expert consensus statement on the diagnosis and treatment of paediatric pulmonary hypertension. The European Paediatric Pulmonary Vascular Disease Network, endorsed by ISHLT and DGPK, Heart, vol.102, issue.2, pp.57-66, 2016.

G. Hansmann, C. Apitz, H. Abdul-khaliq, T. Alastalo, P. Beerbaum et al., Executive summary. Expert consensus statement on the diagnosis and treatment of paediatric pulmonary hypertension. The European Paediatric Pulmonary Vascular Disease Network, endorsed by ISHLT and DGPK, Heart, vol.102, issue.2, pp.86-100, 2016.

P. M. Mourani, M. K. Sontag, D. D. Ivy, and S. H. Abman, Effects of Long-Term Sildenafil Treatment for Pulmonary Hypertension in Infants with Chronic Lung Disease, J Pediatr, vol.154, issue.3, pp.379-384, 2009.

M. Nyp, T. Sandritter, N. Poppinga, C. Simon, and W. E. Truog, Sildenafil citrate, bronchopulmonary dysplasia and disordered pulmonary gas exchange: any benefits?, J Perinatol, vol.32, issue.1, p.64, 2012.

R. J. Barst, M. Beghetti, T. Pulido, G. Layton, I. Konourina et al., STARTS-2CLINICAL PERSPECTIVE: Long-Term Survival With Oral Sildenafil Monotherapy in Treatment-Naive Pediatric Pulmonary Arterial Hypertension, Circulation, vol.129, pp.1914-1937, 2014.

C. Research, Drug Safety and Availability-FDA Drug Safety Communication: FDA recommends against use of Revatio (sildenafil) in children with pulmonary hypertension

. Disponible,

. Revatio®, Communication sur l'augmentation du taux de mortalité lors son utilisation en pédiatrie à des doses supérieures aux doses recommandées dans le traitement de l'hypertension artérielle pulmonaire (HTAP)-Lettre aux professionnels de-ANSM : Agence nationale de sécurité du médicament et des produits de santé

. Disponible,

P. M. Mourani, D. D. Ivy, D. Gao, and S. H. Abman, Pulmonary Vascular Effects of Inhaled Nitric Oxide and Oxygen Tension in Bronchopulmonary Dysplasia, Am J Respir Crit Care Med, vol.170, issue.9, pp.1006-1019, 2004.

G. Hansmann, A. Fernandez-gonzalez, M. Aslam, S. H. Vitali, T. Martin et al., Mesenchymal Stem Cell-Mediated Reversal of Bronchopulmonary Dysplasia and Associated Pulmonary Hypertension, Pulm Circ, vol.2, issue.2, pp.170-81, 2012.

C. Nardiello, I. Mi?íková, and R. E. Morty, Looking ahead: where to next for animal models of bronchopulmonary dysplasia?, Cell Tissue Res, vol.367, issue.3, pp.457-68, 2017.

A. Porzionato, M. M. Sfriso, A. Mazzatenta, V. Macchi, D. Caro et al., Effects of hyperoxic exposure on signal transduction pathways in the lung, Respir Physiol Neurobiol, vol.209, pp.106-120, 2015.

T. M. Asikainen, L. Chang, J. J. Coalson, B. K. Schneider, N. S. Waleh et al., Improved lung growth and function through hypoxia-inducible factor in primate chronic lung disease of prematurity, FASEB J, vol.20, issue.10, pp.1698-700, 2006.

A. Vadivel, R. S. Alphonse, N. Etches, T. Van-haaften, J. Collins et al., HypoxiaInducible Factors Promote Alveolar Development and Regeneration, Am J Respir Cell Mol Biol, vol.50, issue.1, pp.96-105, 2013.

R. D. Rudic, E. G. Shesely, N. Maeda, O. Smithies, S. S. Segal et al., Direct evidence for the importance of endothelium-derived nitric oxide in vascular remodeling, J Clin Invest, vol.101, issue.4, pp.731-737, 1998.

A. Fernandez-gonzalez, S. A. Mitsialis, X. Liu, and S. Kourembanas, Vasculoprotective effects of heme oxygenase-1 in a murine model of hyperoxia-induced bronchopulmonary dysplasia, Am J Physiol-Lung Cell Mol Physiol, vol.302, issue.8, pp.775-84, 2012.

P. A. Dennery, Heme Oxygenase in Neonatal Lung Injury and Repair, Antioxid Redox Signal, vol.21, issue.13, pp.1881-92, 2014.
DOI : 10.1089/ars.2013.5791

URL : http://europepmc.org/articles/pmc4203111?pdf=render

G. Yang, C. Biswasa, Q. S. Lin, P. La, F. Namba et al., Heme oxygenase-1 regulates postnatal lung repair after hyperoxia: Role of ?-catenin/hnRNPK signaling, Redox Biol, vol.1, issue.1, pp.234-277, 2013.

T. R. Grover, T. M. Asikainen, J. P. Kinsella, S. H. Abman, and C. W. White, Hypoxia-inducible factors HIF1? and HIF-2? are decreased in an experimental model of severe respiratory distress syndrome in preterm lambs, Am J Physiol-Lung Cell Mol Physiol, vol.292, issue.6, pp.1345-51, 2007.

J. L. Shifren, N. Doldi, N. Ferrara, S. Mesiano, and R. B. Jaffe, In the human fetus, vascular endothelial growth factor is expressed in epithelial cells and myocytes, but not vascular endothelium: implications for mode of action, J Clin Endocrinol Metab, vol.79, issue.1, pp.316-338, 1994.

T. Cras, N. E. Markham, R. M. Tuder, N. F. Voelkel, and S. H. Abman, Treatment of newborn rats with a VEGF receptor inhibitor causes pulmonary hypertension and abnormal lung structure, Am J Physiol-Lung Cell Mol Physiol, vol.283, issue.3, pp.555-62, 2002.

B. Thébaud, F. Ladha, E. D. Michelakis, M. Sawicka, G. Thurston et al., Vascular Endothelial Growth Factor Gene Therapy Increases Survival, Promotes Lung Angiogenesis, and Prevents Alveolar Damage in Hyperoxia-Induced Lung Injury: Evidence That Angiogenesis Participates in Alveolarization, Circulation, vol.112, issue.16, pp.2477-86, 2005.

T. Cras, R. E. Spitzmiller, K. H. Albertine, J. M. Greenberg, J. A. Whitsett et al., VEGF causes pulmonary hemorrhage, hemosiderosis, and air space enlargement in neonatal mice, Am J Physiol-Lung Cell Mol Physiol, vol.287, issue.1, pp.134-176, 2004.

S. H. Abman, P. F. Shanley, and F. J. Accurso, Failure of postnatal adaptation of the pulmonary circulation after chronic intrauterine pulmonary hypertension in fetal lambs, J Clin Invest, vol.83, issue.6, pp.1849-58, 1989.

T. R. Grover, T. A. Parker, V. Balasubramaniam, N. E. Markham, and S. H. Abman, Pulmonary hypertension impairs alveolarization and reduces lung growth in the ovine fetus, Am J PhysiolLung Cell Mol Physiol, vol.288, issue.4, pp.648-54, 2005.
DOI : 10.1152/ajplung.00288.2004

. Roque-ed-de-la, G. Smeralda, J. Quignard, V. Freund-michel, A. Courtois et al., Altered vasoreactivity in neonatal rats with pulmonary hypertension associated with bronchopulmonary dysplasia: Implication of both eNOS phosphorylation and calcium signaling, PLOS ONE, vol.12, issue.2, p.173044, 2017.

F. K. Kuhr, K. A. Smith, M. Y. Song, I. Levitan, and J. Yuan, New mechanisms of pulmonary arterial hypertension: role of Ca2+ signaling, Am J Physiol-Heart Circ Physiol, vol.302, issue.8, pp.1546-62, 2012.

X. Yu, W. Tao, F. Jiang, C. Li, J. Lin et al., Celastrol Attenuates Hypertension-Induced Inflammation and Oxidative Stress in Vascular Smooth Muscle Cells via Induction of Heme Oxygenase-1, Am J Hypertens, vol.23, issue.8, pp.895-903, 2010.

X. Han, S. Sun, M. Zhao, X. Cheng, G. Chen et al., Celastrol Stimulates Hypoxia-Inducible Factor-1 Activity in Tumor Cells by Initiating the ROS/Akt/p70S6K Signaling Pathway and Enhancing Hypoxia-Inducible Factor-1? Protein Synthesis, PLOS ONE, vol.9, issue.11, p.112470, 2014.

G. F. Pinna, M. Fiorucci, J. Reimund, N. Taquet, Y. Arondel et al., Celastrol inhibits proinflammatory cytokine secretion in Crohn's disease biopsies, Biochem Biophys Res Commun, vol.322, issue.3, pp.778-86, 2004.
DOI : 10.1016/j.bbrc.2004.07.186

H. Li, Y. Zhang, H. Tan, Y. Jia, and D. Li, Therapeutic effect of tripterine on adjuvant arthritis in rats, J Ethnopharmacol, vol.118, issue.3, pp.479-84, 2008.

H. Li, Y. F. Jia, Y. Pan, D. J. Pan, D. Li et al., Effect of tripterine on collagen-induced arthritis in rats, Zhongguo Yao Li Xue Bao, vol.18, issue.3, pp.270-273, 1997.

R. Kannaiyan, M. K. Shanmugam, and G. Sethi, Molecular targets of celastrol derived from Thunder of God Vine: Potential role in the treatment of inflammatory disorders and cancer, Cancer Lett, vol.303, issue.1, pp.9-20, 2011.

W. Y. Seo, S. M. Ju, H. Y. Song, A. R. Goh, J. Jun et al., Celastrol suppresses IFNgamma-induced ICAM-1 expression and subsequent monocyte adhesiveness via the induction of heme oxygenase-1 in the HaCaT cells, Biochem Biophys Res Commun, vol.398, issue.1, pp.140-145, 2010.

J. Liu, J. Lee, S. Hernandez, M. A. Mazitschek, R. Ozcan et al., Treatment of Obesity with Celastrol. Cell, vol.161, issue.5, pp.999-1011, 2015.

. Sondes_calciques_synthetiques,

Y. Huang, Y. Zhou, Y. Fan, and D. Zhou, Celastrol inhibits the growth of human glioma xenografts in nude mice through suppressing VEGFR expression, Cancer Lett, vol.264, issue.1, pp.101-107, 2008.

X. Pang, Z. Yi, J. Zhang, B. Lu, B. Sung et al., Celastrol Suppresses Angiogenesis-Mediated Tumor Growth through Inhibition of AKT/Mammalian Target of Rapamycin Pathway, Cancer Res, vol.70, issue.5, pp.1951-1960, 2010.

J. Ma, J. Ma, L. Z. Han, L. Z. Han, H. Liang et al., Celastrol inhibits the HIF-1? pathway by inhibition of mTOR/p70S6K/eIF4E and ERK1/2 phosphorylation in human hepatoma cells, Oncol Rep, vol.32, issue.1, pp.235-277, 2014.

S. Huang, Y. Tang, X. Cai, X. Peng, X. Liu et al., Celastrol inhibits vasculogenesis by suppressing the VEGF-induced functional activity of bone marrow-derived endothelial progenitor cells, Biochem Biophys Res Commun, vol.423, issue.3, pp.467-72, 2012.

L. Huang, Z. Zhang, S. Zhang, J. Ren, R. Zhang et al., Inhibitory action of Celastrol on hypoxia-mediated angiogenesis and metastasis via the HIF-1? pathway, Int J Mol Med, vol.27, issue.3, pp.407-422, 2011.

S. W. Park, Y. Zhou, J. Lee, J. Lee, and U. Ozcan, Sarco(endo)plasmic reticulum Ca2+-ATPase 2b is a major regulator of endoplasmic reticulum stress and glucose homeostasis in obesity, Proc Natl Acad Sci, vol.107, issue.45, pp.19320-19325, 2010.

R. Goyal, A. Mittal, N. Chu, L. Zhang, and L. D. Longo, Alpha(1)-Adrenergic receptor subtype function in fetal and adult cerebral arteries, Am J Physiol Heart Circ Physiol, vol.298, issue.6, pp.1797-1806, 2010.