71 results on '"Kurakula K"'
Search Results
2. FHL2 is a novel target in pulmonary arterial hypertension
- Author
-
Nadeem, C, primary, Abbas, A L C, additional, Verhoeven, D T B, additional, Willems, L, additional, Goumans, M J, additional, and Kurakula, K, additional
- Published
- 2024
- Full Text
- View/download PDF
3. Synthesis and preclinical evaluation of [C-11]LR111 and [F-18]EW-7197 as PET tracers of the activin-receptor like kinase-5
- Author
-
Rotteveel, L., Kurakula, K., Kooijman, E.J.M., Schuit, R.C., Verlaan, M., Schreurs, M., Beaino, W., Dinther, M.A.H. van, Dijke, P. ten, Lammertsma, A.A., Poot, A.J., Bogaard, H.J., Windhorst, A.D., Pulmonary medicine, Physiology, ACS - Pulmonary hypertension & thrombosis, Radiology and nuclear medicine, AMS - Tissue Function & Regeneration, Amsterdam Neuroscience - Brain Imaging, Amsterdam Neuroscience - Mood, Anxiety, Psychosis, Stress & Sleep, Otolaryngology / Head & Neck Surgery, and CCA - Imaging and biomarkers
- Subjects
Cancer Research ,Positron emission tomography ,MDA-MB-231 tumour xenografts ,Molecular Medicine ,Radiology, Nuclear Medicine and imaging ,TGF beta type I receptor ,Cancer - Abstract
The transforming growth factor β (TGFβ) pathway plays a complex role in cancer biology, being involved in both tumour suppression as well as promotion. Overactive TGFβ signalling has been linked to multiple diseases, including cancer, pulmonary arterial hypertension, and fibrosis. One of the key meditators within this pathway is the TGFβ type I receptor, also termed activin receptor-like kinase 5 (ALK5). ALK5 expression level is a key determinant of TGFβ signalling intensity and duration, and perturbation has been linked to diseases. A validated ALK5 positron emission tomography (PET) tracer creates an opportunity, therefore, to study its role in human diseases. To develop ALK5 PET tracers, two small molecule ALK5 kinase inhibitors were selected as lead compounds, which were labelled with carbon-11 and fluorine-18, respectively. [11C]LR111 was synthesized with a yield of 17 ± 6%, a molar activity of 126 ± 79 GBq·μmol−1 and a purity of >95% (n = 44). [18F]EW-7197 was synthesized with a yield of 10 ± 5%, a molar activity of 183 ± 126 GBq·μmol−1 and a purity of >95% (n = 11). Metabolic stability was evaluated in vivo in mice, showing 39 ± 2% of intact [11C]LR111 and 21 ± 2% of intact [18F]EW-7197 in blood plasma at 45 min p.i. In vitro binding experiments were conducted in breast cancer MDA-MB-231 and lung cancer A431 cell lines. In addition, both tracers were used for PET imaging in MDA-MB-231 xenograft models. Selective uptake of [18F]EW-7197 and [11C]LR111 was observed in MDA-MB-231 cells, in the MDA-MB-231 tumour xenografts in vivo and in the autoradiograms. As [11C]LR111 and [18F]EW-7197 showed selectivity of binding to ALK5 in vivo and in vitro. Both tracers are thereby valuable tools for the detection of ALK5 activity.
- Published
- 2022
- Full Text
- View/download PDF
4. LIM-only protein FHL2 attenuates vascular tissue factor activity, inhibits thrombus formation in mice and FHL2 genetic variation associates with human venous thrombosis
- Author
-
Kroone, C., Vos, M., Rademakers, T., Kuijpers, M., Hoogenboezem, M., Buul, J. van, Heemskerk, J.W.M., Ruf, W., Vlieg, A.V., Versteeg, H.H., Goumans, M.J., Vries, C.J.M. de, Kurakula, K., INVENT Consortium, RS: Carim - B03 Cell biochemistry of thrombosis and haemostasis, Biochemie, MUMC+: HVC Pieken Trombose (9), Medical Biochemistry, ACS - Diabetes & metabolism, AGEM - Endocrinology, metabolism and nutrition, Amsterdam Gastroenterology Endocrinology Metabolism, Landsteiner Laboratory, ACS - Heart failure & arrhythmias, Physiology, and ACS - Pulmonary hypertension & thrombosis
- Subjects
SMOOTH-MUSCLE-CELLS ,LIM-Homeodomain Proteins ,Muscle Proteins ,COAGULATION ,Thromboplastin ,FACTOR EXPRESSION ,ACTIVATION ,03 medical and health sciences ,Tissue factor ,Mice ,0302 clinical medicine ,PROCOAGULANT ACTIVITY ,Medicine ,Animals ,Humans ,Thrombus ,Venous Thrombosis ,Gene knockdown ,Hemostasis ,business.industry ,INDUCTION ,THROMBOMODULIN ,Genetic Variation ,Hematology ,Articles ,medicine.disease ,Thrombosis ,ENDOTHELIAL-CELLS ,FHL2 ,RECEPTORS ,Venous thrombosis ,Coagulation ,MONOCYTES ,Cancer research ,Signal transduction ,business ,030215 immunology ,Transcription Factors - Abstract
Bleeding disorders and thrombotic complications are major causes of morbidity and mortality with many cases being unexplained. Thrombus formation involves aberrant expression and activation of tissue factor in vascular endothelial and smooth muscle cells. Here, we sought to identify factors that modulate tissue factor gene expression and activity in these vascular cells. The LIM-only protein FHL2 is a scaffolding protein modulating signal transduction pathways with crucial functions in endothelial and smooth muscle cells. However, the role of FHL2 in tissue factor regulation and thrombosis remains unexplored. Using a murine venous thrombosis model in mesenteric vessels, we demonstrated that FHL2 deficiency results in exacerbated thrombus formation. Gain and loss of function experiments revealed that FHL2 represses tissue factor expression in endothelial and smooth muscle cells through inhibition of the transcription factors NFκB and AP-1. Furthermore, we observed that FHL2 interacts with the cytoplasmic tail of tissue factor. In line with our in vivo observations, FHL2 decreases tissue factor activity in endothelial and smooth muscle cells whereas FHL2 knockdown or deficiency results in enhanced tissue factor activity. Finally, FHL2 single nucleotide polymorphism (SNP) rs4851770 was associated with venous thrombosis risk in a large population of venous thrombosis cases and control subjects from 12 studies (INVENT consortium). Altogether, our results highlight functional involvement of FHL2 in tissue factor-mediated coagulation and identify FHL2 as a novel gene associated with venous thrombosis in man.
- Published
- 2020
- Full Text
- View/download PDF
5. Deficiency of FHL2 attenuates airway inflammation in mice and genetic variation associates with human bronchial hyper-responsiveness
- Author
-
Kurakula, K., Vos, M., Logiantara, A., Roelofs, J. J. T. H., Nieuwenhuis, M. A., Koppelman, G. H., Postma, D. S., Brandsma, C. A., Sin, D. D., Bossé, Y., Nickle, D. C., van Rijt, L. S., and de Vries, C. J. M.
- Published
- 2015
- Full Text
- View/download PDF
6. Anti-Proliferative Therapy with 6-Mercaptopurine Improves Hemodynamics and BMPR2 Expression in Pulmonary Arterial Hypertension
- Author
-
Botros, L., primary, Szulcek, R., additional, Jansen, S., additional, Smits, J., additional, Phelp, P., additional, Kurakula, K., additional, Goumans, M.-J., additional, Nossent, E.J., additional, Boonstra, A., additional, Vonk Noordegraaf, A., additional, De Man, F., additional, Aman, J., additional, and Bogaard, H.J., additional
- Published
- 2020
- Full Text
- View/download PDF
7. LIM-only protein FHL2 attenuates inflammation in vascular smooth muscle cells through inhibition of the NFκB pathway
- Author
-
van de Pol, V., primary, Vos, M., additional, DeRuiter, M.C., additional, Goumans, M.J., additional, de Vries, C.J.M., additional, and Kurakula, K., additional
- Published
- 2020
- Full Text
- View/download PDF
8. Development of a patient-specific 3-Dimensional cell model to study right heart failure
- Author
-
Llucià-Valldeperas, A., primary, Smal, R., additional, Kurakula, K., additional, ten Dijke, P., additional, Bogaard, H.J., additional, Vonk-Noordegraaf, A., additional, Goumans, M.J., additional, and de Man, F.S., additional
- Published
- 2018
- Full Text
- View/download PDF
9. P548Four-and-a-half LIM-domain 2 secretion is increased in the dilated aorta of bicuspid aortic valve patients
- Author
-
Van De Pol, V, primary, Kurakula, K B, additional, Bons, L R, additional, Roos-Hesselink, J W, additional, Deruiter, M C, additional, and Goumans, M J, additional
- Published
- 2018
- Full Text
- View/download PDF
10. P570Pharmacological activation of nuclear receptor Nur77 decreases endothelial cell dysfunction and reduces experimental pulmonary hypertension
- Author
-
Kurakula, K B, primary, Sun, X, additional, Happe, C, additional, Goumans, M J, additional, and Bogaard, H J, additional
- Published
- 2018
- Full Text
- View/download PDF
11. Thoracic aortic aneurysm development in patients with bicuspid aortic valve: what is the role of endothelial cells?
- Author
-
Pol, V.V. de, Kurakula, K., DeRuiter, M.C., and Goumans, M.J.
- Subjects
parasitic diseases ,cardiovascular system ,complex mixtures ,digestive system diseases - Abstract
Bicuspid aortic valve (BAV) is the most common type of congenital cardiac malformation. Patients with a BAV have a predisposition for the development of thoracic aortic aneurysm (TAA). This pathological aortic dilation may result in aortic rupture, which is fatal in most cases. The abnormal aortic morphology of TAAs results from a complex series of events that alter the cellular structure and extracellular matrix (ECM) composition of the aortic wall. Because the major degeneration is located in the media of the aorta, most studies aim to unravel impaired smooth muscle cell (SMC) function in BAV TAA. However, recent studies suggest that endothelial cells play a key role in both the initiation and progression of TAAs by influencing the medial layer. Aortic endothelial cells are activated in BAV mediated TAAs and have a substantial influence on ECM composition and SMC phenotype, by secreting several key growth factors and matrix modulating enzymes. In recent years there have been significant advances in the genetic and molecular understanding of endothelial cells in BAV associated TAAs. In this review, the involvement of the endothelial cells in BAV TAA pathogenesis is discussed. Endothelial cell functioning in vessel homeostasis, flow response and signalling will be highlighted to give an overview of the importance and the under investigated potential of endothelial cells in BAV-associated TAA.
- Published
- 2017
12. Activation On Nuclear Receptor Nur77 Decreases Experimental Pulmonary Hypertension
- Author
-
Kurakula, K., Sun, X., Happe, C., Vries, C., Bogaard, H. J., Marie Jose Goumans, Pulmonary medicine, and ACS - Pulmonary hypertension & thrombosis
- Published
- 2017
13. REGULATORY RNAs CONTROLLING VASCULAR (DYS)FUNCTION BY AFFECTING TGF-beta FAMILY SIGNALLING
- Author
-
Kurakula, K., Goumans, M.J., and Dijke, P. ten
- Subjects
microRNA ,cardiovascular disease ,bone morphogenetic protein ,transforming growth factor-beta ,endothelial cells ,smooth muscle cells - Published
- 2015
14. Cardiac endothelial cells express Wilms' tumor-1 Wt1 expression in the developing, adult and infarcted heart
- Author
-
Duim, S.N., Kurakula, K., Goumans, M.J., and Kruithof, B.P.T.
- Subjects
Myocardial infarction ,Cardiac development ,Endothelial cells ,Wilm's tumor-1 ,Cardiac regeneration ,Epicardium - Published
- 2015
15. Bone marrow-specific deficiency of nuclear receptor nur77 enhances atherosclerosis
- Author
-
Hamers, A.A., Hamers, A.A., Vos, M., Rassam, F., Marinkovic, G., Kurakula, K., van Gorp, P.J., de Winther, M.P., Gijbels, M.J., de Waard, V., de Vries, C.J., Hamers, A.A., Hamers, A.A., Vos, M., Rassam, F., Marinkovic, G., Kurakula, K., van Gorp, P.J., de Winther, M.P., Gijbels, M.J., de Waard, V., and de Vries, C.J.
- Abstract
RATIONALE: Nuclear receptor Nur77, also known as NR4A1, TR3, or NGFI-B, is expressed in human atherosclerotic lesions in macrophages, endothelial cells, T cells and smooth muscle cells. Macrophages play a critical role in atherosclerosis and the function of Nur77 in lesion macrophages has not yet been investigated. OBJECTIVE: This study aims to delineate the function of Nur77 in macrophages and to assess the effect of bone marrow-specific deficiency of Nur77 on atherosclerosis. METHODS AND RESULTS: We investigated Nur77 in macrophage polarization using bone marrow-derived macrophages (BMM) from wild-type and Nur77-knockout (Nur77(-/-)) mice. Nur77(-/-) BMM exhibit changed expression of M2-specific markers and an inflammatory M1-phenotype with enhanced expression of interleukin-12, IFNgamma, and SDF-1alpha and increased NO synthesis in (non)-stimulated Nur77(-/-) BMM cells. SDF-1alpha expression in nonstimulated Nur77(-/-) BMM is repressed by Nur77 and the chemoattractive activity of Nur77(-/-) BMM is abolished by SDF-1alpha inhibiting antibodies. Furthermore, Nur77(-/-) mice show enhanced thioglycollate-elicited migration of macrophages and B cells. The effect of bone marrow-specific deficiency of Nur77 on atherosclerosis was studied in low density lipoprotein receptor-deficient (Ldlr(-/-)) mice. Ldlr(-/-) mice with a Nur77(-/-)-deficient bone marrow transplant developed 2.1-fold larger atherosclerotic lesions than wild-type bone marrow-transplanted mice. These lesions contain more macrophages, T cells, smooth muscle cells and larger necrotic cores. SDF-1alpha expression is higher in lesions of Nur77(-/-)-transplanted mice, which may explain the observed aggravation of lesion formation. CONCLUSIONS: In conclusion, in bone marrow-derived cells the nuclear receptor Nur77 has an anti-inflammatory function, represses SDF-1alpha expression and inhibits atherosclerosis.
- Published
- 2012
16. Lim-Only Protein Fhl2 Suppresses Pulmonary Artery Endothelial Cell Proliferation Through Activation Of Bmp Signalling
- Author
-
Kurakula, K., Rol, N., Smits, J., Happe, C. M., Bogaard, H. J., Ten Dijke, P., and Marie Jose Goumans
17. Selective inhibition of Histone deacetylases reverses vascular remodelling and improves right ventricle function in pulmonary hypertension
- Author
-
Kurakula, K. B., Sung, X. Q., Van der Feen, D. E., Hagdorn, Q. A. J., Bogaard, H. J., Berger, R. M., Goumans, M. J., and Cardiovascular Centre (CVC)
18. Pharmacological activation of nuclear receptor Nur77 decreases endothelial cell dysfunction and reduces experimental pulmonary hypertension
- Author
-
Kurakula, K. B., Sun, X., Happe, C., Marie Jose Goumans, Bogaard, H. J., Pulmonary medicine, and ACS - Pulmonary hypertension & thrombosis
19. P548Four-and-a-half LIM-domain 2 secretion is increased in the dilated aorta of bicuspid aortic valve patients.
- Author
-
Pol, V Van De, Kurakula, K B, Bons, L R, Roos-Hesselink, J W, Deruiter, M C, and Goumans, M J
- Subjects
- *
AORTIC valve , *CONGENITAL heart disease , *THORACIC aneurysms - Published
- 2018
- Full Text
- View/download PDF
20. Sex Dimorphism in Pulmonary Arterial Hypertension Associated With Autoimmune Diseases.
- Author
-
Krzyżewska A and Kurakula K
- Subjects
- Humans, Animals, Female, Male, Sex Factors, Sex Characteristics, Risk Factors, Genetic Predisposition to Disease, Chromosomes, Human, X genetics, Gonadal Steroid Hormones metabolism, Pulmonary Artery physiopathology, Pulmonary Artery metabolism, Chromosomes, Human, Y genetics, Health Status Disparities, Autoimmune Diseases genetics, Autoimmune Diseases epidemiology, Autoimmune Diseases immunology, Pulmonary Arterial Hypertension genetics, Pulmonary Arterial Hypertension epidemiology, Pulmonary Arterial Hypertension physiopathology
- Abstract
Pulmonary hypertension is a rare, incurable, and progressive disease. Although there is increasing evidence that immune disorders, particularly those associated with connective tissue diseases, are a strong predisposing factor in the development of pulmonary arterial hypertension (PAH), there is currently a lack of knowledge about the detailed molecular mechanisms responsible for this phenomenon. Exploring this topic is crucial because patients with an immune disorder combined with PAH have a worse prognosis and higher mortality compared with patients with other PAH subtypes. Moreover, data recorded worldwide show that the prevalence of PAH in women is 2× to even 4× higher than in men, and the ratio of PAH associated with autoimmune diseases is even higher (9:1). Sexual dimorphism in the pathogenesis of cardiovascular disease was explained for many years by the action of female sex hormones. However, there are increasing reports of interactions between sex hormones and sex chromosomes, and differences in the pathogenesis of cardiovascular disease may be controlled not only by sex hormones but also by sex chromosome pathways that are not dependent on the gonads. This review discusses the role of estrogen and genetic factors including the role of genes located on the X chromosome, as well as the potential protective role of the Y chromosome in sexual dimorphism, which is prominent in the occurrence of PAH associated with autoimmune diseases. Moreover, an overview of animal models that could potentially play a role in further investigating the aforementioned link was also reviewed., Competing Interests: None.
- Published
- 2024
- Full Text
- View/download PDF
21. Angiogenesis in Chronic Thromboembolic Pulmonary Hypertension: A Janus-Faced Player?
- Author
-
Willems L, Kurakula K, Verhaegen J, Klok FA, Delcroix M, Goumans MJ, and Quarck R
- Subjects
- Humans, Angiogenesis, Pulmonary Artery pathology, Chronic Disease, Endarterectomy adverse effects, Hypertension, Pulmonary etiology, Pulmonary Embolism therapy, Thrombosis pathology
- Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a rare form of pulmonary hypertension characterized by the presence of organized thrombi that obstruct pulmonary arteries, ultimately leading to right heart failure and death. Among others, impaired angiogenesis and inflammatory thrombosis have been shown to contribute to the progression of CTEPH. In this review, we summarize the 2-faced nature of angiogenesis in both thrombus formation and resolution in the context of CTEPH and highlight the dual role of angiogenesis and neovascularization in resolving venous thrombi. Furthermore, we discuss relevant in vitro and in vivo models that support the benefits or drawbacks of angiogenesis in CTEPH progression. We discuss the key pathways involved in modulating angiogenesis, particularly the underexplored role of TGFβ (transforming growth factor-beta) signaling in driving fibrosis as an integral element of CTEPH pathogenesis. We finally explore innovative treatment strategies that target angiogenic pathways. These strategies have the potential to pioneer preventive, inventive, or alternative therapeutic options for patients with CTEPH who may not qualify for surgical interventions. Moreover, they could be used synergistically with established treatments such as pulmonary endarterectomy or balloon pulmonary angioplasty. In summary, this review emphasizes the crucial role of angiogenesis in the development of in fibrothrombotic tissue, a major pathological characteristic of CTEPH., Competing Interests: Disclosures M. Delcroix received financial support from the Belgian Pulmonary Hypertension Patient Association. The other authors report no conflicts.
- Published
- 2024
- Full Text
- View/download PDF
22. Imaging the TGFβ type I receptor in pulmonary arterial hypertension.
- Author
-
Rotteveel L, Poot AJ, Kooijman EJM, Schuit RC, Schalij I, Sun X, Kurakula K, Happé C, Beaino W, Ten Dijke P, Lammertsma AA, Bogaard HJ, and Windhorst AD
- Abstract
Transforming growth factor β (TGFβ) activity is perturbed in remodelled pulmonary vasculature of patients with pulmonary arterial hypertension (PAH), cancer, vascular diseases and developmental disorders. Inhibition of TGFβ, which signals via activin receptor-like kinase 5 (ALK5), prevents progression and development of experimental PAH. The purpose of this study was to assess two ALK5 targeting positron emission tomography (PET) tracers ([
11 C]LR111 and [18 F]EW-7197) for imaging ALK5 in monocrotaline (MCT)- and Sugen/hypoxia (SuHx)-induced PAH. Both tracers were subjected to extensive in vitro and in vivo studies. [11 C]LR111 showed the highest metabolic stability, as 46 ± 2% of intact tracer was still present in rat blood plasma after 60 min. In autoradiography experiments, [11 C]LR111 showed high ALK5 binding in vitro compared with controls, 3.2 and 1.5 times higher in SuHx and MCT, respectively. In addition, its binding could be blocked by SB431542, an adenosine triphosphate competitive ALK5 kinase inhibitor. However, [18 F]EW-7197 showed the best in vivo results. 15 min after injection, uptake was 2.5 and 1.4 times higher in the SuHx and MCT lungs, compared with controls. Therefore, [18 F]EW-7197 is a promising PET tracer for ALK5 imaging in PAH., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
23. Inhibition of the prolyl isomerase Pin1 improves endothelial function and attenuates vascular remodelling in pulmonary hypertension by inhibiting TGF-β signalling.
- Author
-
Kurakula K, Hagdorn QAJ, van der Feen DE, Vonk Noordegraaf A, Ten Dijke P, de Boer RA, Bogaard HJ, Goumans MJ, and Berger RMF
- Subjects
- Animals, Disease Models, Animal, Endothelial Cells, Humans, NIMA-Interacting Peptidylprolyl Isomerase genetics, Peptidylprolyl Isomerase, Pulmonary Artery, Rats, Transforming Growth Factor beta, Vascular Remodeling, Hypertension, Pulmonary drug therapy
- Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease, characterized by obstructive pulmonary vascular remodelling ultimately leading to right ventricular (RV) failure and death. Disturbed transforming growth factor-β (TGF-β)/bone morphogenetic protein (BMP) signalling, endothelial cell dysfunction, increased proliferation of smooth muscle cells and fibroblasts, and inflammation contribute to this abnormal remodelling. Peptidyl-prolyl isomerase Pin1 has been identified as a critical driver of proliferation and inflammation in vascular cells, but its role in the disturbed TGF-β/BMP signalling, endothelial cell dysfunction, and vascular remodelling in PAH is unknown. Here, we report that Pin1 expression is increased in cultured pulmonary microvascular endothelial cells (MVECs) and lung tissue of PAH patients. Pin1 inhibitor, juglone significantly decreased TGF-β signalling, increased BMP signalling, normalized their hyper-proliferative, and inflammatory phenotype. Juglone treatment reversed vascular remodelling through reducing TGF-β signalling in monocrotaline + shunt-PAH rat model. Juglone treatment decreased Fulton index, but did not affect or harm cardiac function and remodelling in rats with RV pressure load induced by pulmonary artery banding. Our study demonstrates that inhibition of Pin1 reversed the PAH phenotype in PAH MVECs in vitro and in PAH rats in vivo, potentially through modulation of TGF-β/BMP signalling pathways. Selective inhibition of Pin1 could be a novel therapeutic option for the treatment of PAH., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
24. Derivation and characterisation of endothelial cells from patients with chronic thromboembolic pulmonary hypertension.
- Author
-
Tura-Ceide O, Smolders VFED, Aventin N, Morén C, Guitart-Mampel M, Blanco I, Piccari L, Osorio J, Rodríguez C, Rigol M, Solanes N, Malandrino A, Kurakula K, Goumans MJ, Quax PHA, Peinado VI, Castellà M, and Barberà JA
- Subjects
- Apoptosis, Case-Control Studies, Chronic Disease, Endothelium, Vascular pathology, Endothelium, Vascular physiopathology, Female, Humans, Hypertension, Pulmonary physiopathology, Male, Middle Aged, Mitochondria pathology, Oxidative Stress, Pulmonary Artery cytology, Pulmonary Artery pathology, Pulmonary Artery physiopathology, Pulmonary Embolism physiopathology, Endothelium, Vascular cytology, Hypertension, Pulmonary pathology, Pulmonary Embolism pathology
- Abstract
Pulmonary endarterectomy (PEA) resected material offers a unique opportunity to develop an in vitro endothelial cell model of chronic thromboembolic pulmonary hypertension (CTEPH). We aimed to comprehensively analyze the endothelial function, molecular signature, and mitochondrial profile of CTEPH-derived endothelial cells to better understand the pathophysiological mechanisms of endothelial dysfunction behind CTEPH, and to identify potential novel targets for the prevention and treatment of the disease. Isolated cells from specimens obtained at PEA (CTEPH-EC), were characterized based on morphology, phenotype, and functional analyses (in vitro and in vivo tubule formation, proliferation, apoptosis, and migration). Mitochondrial content, morphology, and dynamics, as well as high-resolution respirometry and oxidative stress, were also studied. CTEPH-EC displayed a hyperproliferative phenotype with an increase expression of adhesion molecules and a decreased apoptosis, eNOS activity, migration capacity and reduced angiogenic capacity in vitro and in vivo compared to healthy endothelial cells. CTEPH-EC presented altered mitochondrial dynamics, increased mitochondrial respiration and an unbalanced production of reactive oxygen species and antioxidants. Our study is the foremost comprehensive investigation of CTEPH-EC. Modulation of redox, mitochondrial homeostasis and adhesion molecule overexpression arise as novel targets and biomarkers in CTEPH., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
25. The Inflammatory Profile of CTEPH-Derived Endothelial Cells Is a Possible Driver of Disease Progression.
- Author
-
Smolders VFED, Lodder K, Rodríguez C, Tura-Ceide O, Barberà JA, Jukema JW, Quax PHA, Goumans MJ, and Kurakula K
- Subjects
- Endarterectomy, Female, Fluorescence, Gene Expression Regulation, Hemodynamics, Humans, Hypertension, Pulmonary genetics, Hypertension, Pulmonary physiopathology, Inflammation genetics, Inflammation physiopathology, Male, Middle Aged, Phosphorylation, Thromboembolism genetics, Thromboembolism physiopathology, Transcription Factor RelA antagonists & inhibitors, Transcription Factor RelA metabolism, Disease Progression, Endothelial Cells pathology, Hypertension, Pulmonary complications, Hypertension, Pulmonary pathology, Inflammation pathology, Thromboembolism complications, Thromboembolism pathology
- Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a form of pulmonary hypertension characterized by the presence of fibrotic intraluminal thrombi and causing obliteration of the pulmonary arteries. Although both endothelial cell (EC) dysfunction and inflammation are linked to CTEPH pathogenesis, regulation of the basal inflammatory response of ECs in CTEPH is not fully understood. Therefore, in the present study, we investigated the role of the nuclear factor (NF)-κB pro-inflammatory signaling pathway in ECs in CTEPH under basal conditions. Basal mRNA levels of interleukin (IL)-8, IL-1β, monocyte chemoattractant protein-1 (MCP-1), C-C motif chemokine ligand 5 (CCL5), and vascular cell adhesion molecule-1 (VCAM-1) were upregulated in CTEPH-ECs compared to the control cells. To assess the involvement of NF-κB signaling in basal inflammatory activation, CTEPH-ECs were incubated with the NF-κB inhibitor Bay 11-7085. The increase in pro-inflammatory cytokines was abolished when cells were incubated with the NF-κB inhibitor. To determine if NF-κB was indeed activated, we stained pulmonary endarterectomy (PEA) specimens from CTEPH patients and ECs isolated from PEA specimens for phospho-NF-κB-P65 and found that especially the vessels within the thrombus and CTEPH-ECs are positive for phospho-NF-κB-P65. In summary, we show that CTEPH-ECs have a pro-inflammatory status under basal conditions, and blocking NF-κB signaling reduces the production of inflammatory factors in CTEPH-ECs. Therefore, our results show that the increased basal pro-inflammatory status of CTEPH-ECs is, at least partially, regulated through activation of NF-κB signaling and potentially contributes to the pathophysiology and progression of CTEPH.
- Published
- 2021
- Full Text
- View/download PDF
26. Increased MAO-A Activity Promotes Progression of Pulmonary Arterial Hypertension.
- Author
-
Sun XQ, Peters EL, Schalij I, Axelsen JB, Andersen S, Kurakula K, Gomez-Puerto MC, Szulcek R, Pan X, da Silva Goncalves Bos D, Schiepers REJ, Andersen A, Goumans MJ, Vonk Noordegraaf A, van der Laarse WJ, de Man FS, and Bogaard HJ
- Subjects
- Animals, Clorgyline pharmacology, Clorgyline therapeutic use, Disease Models, Animal, Heart Ventricles drug effects, Heart Ventricles enzymology, Heart Ventricles pathology, Heart Ventricles physiopathology, Humans, Hypertrophy, Right Ventricular complications, Hypertrophy, Right Ventricular physiopathology, Indoles, Oxidative Stress drug effects, Pulmonary Arterial Hypertension chemically induced, Pulmonary Arterial Hypertension drug therapy, Pulmonary Arterial Hypertension physiopathology, Pulmonary Artery drug effects, Pulmonary Artery enzymology, Pulmonary Artery pathology, Pulmonary Artery physiopathology, Pyrroles, Rats, Vascular Remodeling drug effects, Vascular Stiffness drug effects, Vasodilation drug effects, Disease Progression, Monoamine Oxidase metabolism, Pulmonary Arterial Hypertension enzymology
- Abstract
Monoamine oxidases (MAOs), a class of enzymes bound to the outer mitochondrial membrane, are important sources of reactive oxygen species. Increased MAO-A activity in endothelial cells and cardiomyocytes contributes to vascular dysfunction and progression of left heart failure. We hypothesized that inhibition of MAO-A can be used to treat pulmonary arterial hypertension (PAH) and right ventricular (RV) failure. MAO-A levels in lung and RV samples from patients with PAH were compared with levels in samples from donors without PAH. Experimental PAH was induced in male Sprague-Dawley rats by using Sugen 5416 and hypoxia (SuHx), and RV failure was induced in male Wistar rats by using pulmonary trunk banding (PTB). Animals were randomized to receive either saline or the MAO-A inhibitor clorgyline at 10 mg/kg. Echocardiography and RV catheterization were performed, and heart and lung tissues were collected for further analysis. We found increased MAO-A expression in the pulmonary vasculature of patients with PAH and in experimental experimental PAH induced by SuHx. Cardiac MAO-A expression and activity was increased in SuHx- and PTB-induced RV failure. Clorgyline treatment reduced RV afterload and pulmonary vascular remodeling in SuHx rats through reduced pulmonary vascular proliferation and oxidative stress. Moreover, clorgyline improved RV stiffness and relaxation and reversed RV hypertrophy in SuHx rats. In PTB rats, clorgyline had no direct clorgyline had no direct effect on the right ventricle effect. Our study reveals the role of MAO-A in the progression of PAH. Collectively, these findings indicated that MAO-A may be involved in pulmonary vascular remodeling and consecutive RV failure.
- Published
- 2021
- Full Text
- View/download PDF
27. Volume Load-Induced Right Ventricular Failure in Rats Is Not Associated With Myocardial Fibrosis.
- Author
-
Hagdorn QAJ, Kurakula K, Koop AC, Bossers GPL, Mavrogiannis E, van Leusden T, van der Feen DE, de Boer RA, Goumans MTH, and Berger RMF
- Abstract
Background: Right ventricular (RV) function and failure are key determinants of morbidity and mortality in various cardiovascular diseases. Myocardial fibrosis is regarded as a contributing factor to heart failure, but its importance in RV failure has been challenged. This study aims to assess whether myocardial fibrosis drives the transition from compensated to decompensated volume load-induced RV dysfunction., Methods: Wistar rats were subjected to aorto-caval shunt (ACS, n = 23) or sham (control, n = 15) surgery, and sacrificed after 1 month, 3 months, or 6 months. Echocardiography, RV pressure-volume analysis, assessment of gene expression and cardiac histology were performed., Results: At 6 months, 6/8 ACS-rats (75%) showed clinical signs of RV failure (pleural effusion, ascites and/or liver edema), whereas at 1 month and 3 months, no signs of RV failure had developed yet. Cardiac output has increased two- to threefold and biventricular dilatation occurred, while LV ejection fraction gradually decreased. At 1 month and 3 months, RV end-systolic elastance (Ees) remained unaltered, but at 6 months, RV Ees had decreased substantially. In the RV, no oxidative stress, inflammation, pro-fibrotic signaling (TGFβ1 and pSMAD2/3), or fibrosis were present at any time point., Conclusions: In the ACS rat model, long-term volume load was initially well tolerated at 1 month and 3 months, but induced overt clinical signs of end-stage RV failure at 6 months. However, no myocardial fibrosis or increased pro-fibrotic signaling had developed. These findings indicate that myocardial fibrosis is not involved in the transition from compensated to decompensated RV dysfunction in this model., Competing Interests: The University Medical Center Groningen (UMCG) has received fees for consultancy activities of RMB for Actelion and Lilly outside the content of this manuscript. The UMCG has received research grants and/or fees from AstraZeneca, Abbott, Bristol-Myers Squibb, Novartis, Novo Nordisk, Roche, Trevena, and ThermoFisher GmbH for projects of RAB, outside the content of this manuscript. RAB is a minority shareholder of scPharmaceuticals, Inc. and received personal fees from MandalMed Inc., AstraZeneca, Novartis, Servier, and Vifor. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Hagdorn, Kurakula, Koop, Bossers, Mavrogiannis, van Leusden, van der Feen, de Boer, Goumans and Berger.)
- Published
- 2021
- Full Text
- View/download PDF
28. BMP Receptor Inhibition Enhances Tissue Repair in Endoglin Heterozygous Mice.
- Author
-
Bakker W, Dingenouts CKE, Lodder K, Wiesmeijer KC, de Jong A, Kurakula K, Mager HJ, Smits AM, de Vries MR, Quax PHA, and Goumans MJTH
- Subjects
- Animals, Bone Morphogenetic Protein Receptors genetics, Bone Morphogenetic Protein Receptors metabolism, Cells, Cultured, Endoglin genetics, Female, Heterozygote, Humans, Macrophages immunology, Macrophages metabolism, Male, Mice, Inbred C57BL, Mice, Knockout, Myocardial Infarction genetics, Myocardial Infarction metabolism, Telangiectasia, Hereditary Hemorrhagic genetics, Telangiectasia, Hereditary Hemorrhagic immunology, Telangiectasia, Hereditary Hemorrhagic metabolism, Wound Healing genetics, Mice, Bone Morphogenetic Protein Receptors antagonists & inhibitors, Disease Models, Animal, Endoglin metabolism, Myocardial Infarction prevention & control, Pyrazoles pharmacology, Pyrimidines pharmacology, Wound Healing drug effects
- Abstract
Hereditary hemorrhagic telangiectasia type 1 (HHT1) is a severe vascular disorder caused by mutations in the TGFβ/BMP co-receptor endoglin . Endoglin haploinsufficiency results in vascular malformations and impaired neoangiogenesis. Furthermore, HHT1 patients display an impaired immune response. To date it is not fully understood how endoglin haploinsufficient immune cells contribute to HHT1 pathology. Therefore, we investigated the immune response during tissue repair in Eng+/- mice, a model for HHT1. Eng +/- mice exhibited prolonged infiltration of macrophages after experimentally induced myocardial infarction. Moreover, there was an increased number of inflammatory M1-like macrophages (Ly6C
high /CD206- ) at the expense of reparative M2-like macrophages (Ly6Clow /CD206+ ). Interestingly, HHT1 patients also showed an increased number of inflammatory macrophages. In vitro analysis revealed that TGFβ-induced differentiation of Eng+/- monocytes into M2-like macrophages was blunted. Inhibiting BMP signaling by treating monocytes with LDN-193189 normalized their differentiation. Finally, LDN treatment improved heart function after MI and enhanced vascularization in both wild type and Eng+/- mice. The beneficial effect of LDN was also observed in the hind limb ischemia model. While blood flow recovery was hampered in vehicle-treated animals, LDN treatment improved tissue perfusion recovery in Eng+/- mice. In conclusion, BMPR kinase inhibition restored HHT1 macrophage imbalance in vitro and improved tissue repair after ischemic injury in Eng+/- mice.- Published
- 2021
- Full Text
- View/download PDF
29. Endothelial Dysfunction in Pulmonary Hypertension: Cause or Consequence?
- Author
-
Kurakula K, Smolders VFED, Tura-Ceide O, Jukema JW, Quax PHA, and Goumans MJ
- Abstract
Pulmonary arterial hypertension (PAH) is a rare, complex, and progressive disease that is characterized by the abnormal remodeling of the pulmonary arteries that leads to right ventricular failure and death. Although our understanding of the causes for abnormal vascular remodeling in PAH is limited, accumulating evidence indicates that endothelial cell (EC) dysfunction is one of the first triggers initiating this process. EC dysfunction leads to the activation of several cellular signalling pathways in the endothelium, resulting in the uncontrolled proliferation of ECs, pulmonary artery smooth muscle cells, and fibroblasts, and eventually leads to vascular remodelling and the occlusion of the pulmonary blood vessels. Other factors that are related to EC dysfunction in PAH are an increase in endothelial to mesenchymal transition, inflammation, apoptosis, and thrombus formation. In this review, we outline the latest advances on the role of EC dysfunction in PAH and other forms of pulmonary hypertension. We also elaborate on the molecular signals that orchestrate EC dysfunction in PAH. Understanding the role and mechanisms of EC dysfunction will unravel the therapeutic potential of targeting this process in PAH.
- Published
- 2021
- Full Text
- View/download PDF
30. Altered TGFβ/SMAD Signaling in Human and Rat Models of Pulmonary Hypertension: An Old Target Needs Attention.
- Author
-
Sanada TJ, Sun XQ, Happé C, Guignabert C, Tu L, Schalij I, Bogaard HJ, Goumans MJ, and Kurakula K
- Subjects
- Animals, Blood Pressure, Disease Models, Animal, Humans, Hypertension, Pulmonary genetics, Hypertension, Pulmonary physiopathology, Phosphorylation, Plasminogen Activator Inhibitor 1 metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Rats, Receptor, Transforming Growth Factor-beta Type I genetics, Receptor, Transforming Growth Factor-beta Type I metabolism, Receptor, Transforming Growth Factor-beta Type II genetics, Receptor, Transforming Growth Factor-beta Type II metabolism, Systole, Hypertension, Pulmonary metabolism, Signal Transduction, Smad Proteins metabolism, Transforming Growth Factor beta metabolism
- Abstract
Recent translational studies highlighted the inhibition of transforming growth factor (TGF)-β signaling as a promising target to treat pulmonary arterial hypertension (PAH). However, it remains unclear whether alterations in TGF-β signaling are consistent between PAH patients and animal models. Therefore, we compared TGF-β signaling in the lungs of PAH patients and rats with experimental PAH induced by monocrotaline (MCT) or SU5416+hypoxia (SuHx). In hereditary PAH (hPAH) patients, there was a moderate increase in both TGFβR2 and pSMAD2/3 protein levels, while these were unaltered in idiopathic PAH (iPAH) patients. Protein levels of TGFβR2 and pSMAD2/3 were locally increased in the pulmonary vasculature of PAH rats under both experimental conditions. Conversely, the protein levels of TGFβR2 and pSMAD2/3 were reduced in SuHx while slightly increased in MCT. mRNA levels of plasminogen activator inhibitor (PAI)-1 were increased only in MCT animals and such an increase was not observed in SuHx rats or in iPAH and hPAH patients. In conclusion, our data demonstrate considerable discrepancies in TGFβ-SMAD signaling between iPAH and hPAH patients, as well as between patients and rats with experimental PAH.
- Published
- 2021
- Full Text
- View/download PDF
31. Exacerbated inflammatory signaling underlies aberrant response to BMP9 in pulmonary arterial hypertension lung endothelial cells.
- Author
-
Szulcek R, Sanchez-Duffhues G, Rol N, Pan X, Tsonaka R, Dickhoff C, Yung LM, Manz XD, Kurakula K, Kiełbasa SM, Mei H, Timens W, Yu PB, Bogaard HJ, and Goumans MJ
- Subjects
- Adult, Aged, Endothelium, Vascular pathology, Female, Homeostasis, Humans, Interleukin-6 metabolism, Male, Microvessels pathology, Middle Aged, Neutralization Tests, Phenotype, Pulmonary Arterial Hypertension genetics, Transcription, Genetic, Endothelial Cells metabolism, Growth Differentiation Factor 2 metabolism, Inflammation metabolism, Inflammation pathology, Lung pathology, Pulmonary Arterial Hypertension metabolism, Pulmonary Arterial Hypertension pathology, Signal Transduction
- Abstract
Imbalanced transforming growth factor beta (TGFβ) and bone morphogenetic protein (BMP) signaling are postulated to favor a pathological pulmonary endothelial cell (EC) phenotype in pulmonary arterial hypertension (PAH). BMP9 is shown to reinstate BMP receptor type-II (BMPR2) levels and thereby mitigate hemodynamic and vascular abnormalities in several animal models of pulmonary hypertension (PH). Yet, responses of the pulmonary endothelium of PAH patients to BMP9 are unknown. Therefore, we treated primary PAH patient-derived and healthy pulmonary ECs with BMP9 and observed that stimulation induces transient transcriptional signaling associated with the process of endothelial-to-mesenchymal transition (EndMT). However, solely PAH pulmonary ECs showed signs of a mesenchymal trans-differentiation characterized by a loss of VE-cadherin, induction of transgelin (SM22α), and reorganization of the cytoskeleton. In the PAH cells, a prolonged EndMT signaling was found accompanied by sustained elevation of pro-inflammatory, pro-hypoxic, and pro-apoptotic signaling. Herein we identified interleukin-6 (IL6)-dependent signaling to be the central mediator required for the BMP9-induced phenotypic change in PAH pulmonary ECs. Furthermore, we were able to target the BMP9-induced EndMT process by an IL6 capturing antibody that normalized autocrine IL6 levels, prevented mesenchymal transformation, and maintained a functional EC phenotype in PAH pulmonary ECs. In conclusion, our results show that the BMP9-induced aberrant EndMT in PAH pulmonary ECs is dependent on exacerbated pro-inflammatory signaling mediated through IL6.
- Published
- 2020
- Full Text
- View/download PDF
32. Cellular senescence impairs the reversibility of pulmonary arterial hypertension.
- Author
-
van der Feen DE, Bossers GPL, Hagdorn QAJ, Moonen JR, Kurakula K, Szulcek R, Chappell J, Vallania F, Donato M, Kok K, Kohli JS, Petersen AH, van Leusden T, Demaria M, Goumans MTH, De Boer RA, Khatri P, Rabinovitch M, Berger RMF, and Bartelds B
- Subjects
- Animals, Cellular Senescence, Endothelial Cells, Familial Primary Pulmonary Hypertension, Humans, Rats, Heart Defects, Congenital, Pulmonary Arterial Hypertension
- Abstract
Pulmonary arterial hypertension (PAH) in congenital cardiac shunts can be reversed by hemodynamic unloading (HU) through shunt closure. However, this reversibility potential is lost beyond a certain point in time. The reason why PAH becomes irreversible is unknown. In this study, we used MCT+shunt-induced PAH in rats to identify a dichotomous reversibility response to HU, similar to the human situation. We compared vascular profiles of reversible and irreversible PAH using RNA sequencing. Cumulatively, we report that loss of reversibility is associated with a switch from a proliferative to a senescent vascular phenotype and confirmed markers of senescence in human PAH-CHD tissue. In vitro, we showed that human pulmonary endothelial cells of patients with PAH are more vulnerable to senescence than controls in response to shear stress and confirmed that the senolytic ABT263 induces apoptosis in senescent, but not in normal, endothelial cells. To support the concept that vascular cell senescence is causal to the irreversible nature of end-stage PAH, we targeted senescence using ABT263 and induced reversal of the hemodynamic and structural changes associated with severe PAH refractory to HU. The factors that drive the transition from a reversible to irreversible pulmonary vascular phenotype could also explain the irreversible nature of other PAH etiologies and provide new leads for pharmacological reversal of end-stage PAH., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
33. The Effects of Mercaptopurine on Pulmonary Vascular Resistance and BMPR2 Expression in Pulmonary Arterial Hypertension.
- Author
-
Botros L, Szulcek R, Jansen SMA, Kurakula K, Goumans MTH, van Kuilenburg ABP, Vonk Noordegraaf A, de Man FS, Aman J, and Bogaard HJ
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Antihypertensive Agents therapeutic use, Bone Morphogenetic Protein Receptors, Type II drug effects, Bone Morphogenetic Protein Receptors, Type II genetics, Mercaptopurine therapeutic use, Pulmonary Arterial Hypertension diagnosis, Pulmonary Arterial Hypertension drug therapy, Vascular Resistance drug effects
- Published
- 2020
- Full Text
- View/download PDF
34. The BMP Receptor 2 in Pulmonary Arterial Hypertension: When and Where the Animal Model Matches the Patient.
- Author
-
Happé C, Kurakula K, Sun XQ, da Silva Goncalves Bos D, Rol N, Guignabert C, Tu L, Schalij I, Wiesmeijer KC, Tura-Ceide O, Vonk Noordegraaf A, de Man FS, Bogaard HJ, and Goumans MJ
- Subjects
- Animals, Disease Models, Animal, Humans, Lung blood supply, Lung metabolism, Male, Models, Biological, Phosphorylation, Rats, Wistar, Smad Proteins metabolism, Bone Morphogenetic Protein Receptors, Type II metabolism, Pulmonary Arterial Hypertension metabolism
- Abstract
Background: Mutations in bone morphogenetic protein receptor type II (BMPR2) are leading to the development of hereditary pulmonary arterial hypertension (PAH). In non-hereditary forms of PAH, perturbations in the transforming growth factor-β (TGF-β)/BMP-axis are believed to cause deficient BMPR2 signaling by changes in receptor expression, the activity of the receptor and/or downstream signaling. To date, BMPR2 expression and its activity in the lungs of patients with non-hereditary PAH is poorly characterized. In recent decades, different animal models have been used to understand the role of BMPR2 signaling in PAH pathophysiology. Specifically, the monocrotaline (MCT) and Sugen-Hypoxia (SuHx) models are extensively used in interventional studies to examine if restoring BMPR2 signaling results in PAH disease reversal. While PAH is assumed to develop in patients over months or years, pulmonary hypertension in experimental animal models develops in days or weeks. It is therefore likely that modifications in BMP and TGF-β signaling in these models do not fully recapitulate those in patients. In order to determine the translational potential of the MCT and SuHx models, we analyzed the BMPR2 expression and activity in the lungs of rats with experimentally induced PAH and compared this to the BMPR2 expression and activity in the lungs of PAH patients. Methods: the BMPR2 expression was analyzed by Western blot analysis and immunofluorescence (IF) microscopy to determine the quantity and localization of the receptor in the lung tissue from normal control subjects and patients with hereditary or idiopathic PAH, as well as in the lungs of control rats and rats with MCT or SuHx-induced PAH. The activation of the BMP pathway was analyzed by determining the level and localization of phosphorylated Smad1/5/8 (pSmad 1/5/8), a downstream mediator of canonical BMPR2 signaling. Results: While BMPR2 and pSmad 1/5/8 expression levels were unaltered in whole lung lysates/homogenates from patients with hereditary and idiopathic PAH, IF analysis showed that BMPR2 and pSmad 1/5/8 levels were markedly decreased in the pulmonary vessels of both PAH patient groups. Whole lung BMPR2 expression was variable in the two PAH rat models, while in both experimental models the expression of BMPR2 in the lung vasculature was increased. However, in the human PAH lungs, the expression of pSmad 1/5/8 was downregulated in the lung vasculature of both experimental models. Conclusion: BMPR2 receptor expression and downstream signaling is reduced in the lung vasculature of patients with idiopathic and hereditary PAH, which cannot be appreciated when using human whole lung lysates. Despite increased BMPR2 expression in the lung vasculature, the MCT and SuHx rat models did develop PAH and impaired downstream BMPR2-Smad signaling similar to our findings in the human lung.
- Published
- 2020
- Full Text
- View/download PDF
35. Effects of 6-mercaptopurine in pressure overload induced right heart failure.
- Author
-
Axelsen JB, Andersen S, Sun XQ, Ringgaard S, Hyldebrandt JA, Kurakula K, Goumans MJ, de Man FS, Nielsen-Kudsk JE, Bogaard HJ, and Andersen A
- Subjects
- Animals, Apoptosis drug effects, Blood Pressure, Disease Models, Animal, Heart Failure drug therapy, Heart Failure pathology, Heart Ventricles, Hemodynamics, Male, Rats, Wistar, Ventricular Function, Right, Ventricular Remodeling, Heart Failure etiology, Heart Failure physiopathology, Hypertension, Pulmonary complications, Mercaptopurine pharmacology
- Abstract
Background: Several antineoplastic drugs have been proposed as new compounds for pulmonary arterial hypertension treatment but many have cardiotoxic side effects. The chemotherapeutic agent 6-mercaptopurine may have an effect in treatment of pulmonary arterial hypertension but at the same time, its effects on the afterload adaption of the right ventricle is unpredictable due to interaction with multiple downstream signalling pathways in the cardiomyocytes. We investigated the direct cardiac effects of 6-mercaptopurine in rats with isolated right heart failure caused by pulmonary trunk banding (PTB)., Methods: Male Wistar rat weanlings (112±2 g) were randomized to sham operation (sham, n = 10) or PTB. The PTB animals were randomized to placebo (PTB-control, n = 10) and 6-mercaptopurine (7.5 mg/kg/day) groups with treatment start before the PTB procedure (PTB-prevention, n = 10) or two weeks after (PTB-reversal, n = 10). Right ventricular effects were evaluated by echocardiography, cardiac MRI, invasive pressure-volume measurements, and histological and molecular analyses., Results: PTB increased right ventricular afterload and caused right ventricular hypertrophy and failure. 6-mercaptopurine did not improve right ventricular function nor reduce right ventricular remodelling in both prevention and reversal studies compared with placebo-treated rats., Conclusion: Treatment with 6-mercaptopurine did not have any beneficial or detrimental effects on right ventricular function or remodelling. Our data suggest that treatment of pulmonary arterial hypertension with 6-mercaptopurine is not harmful to the failing right ventricle., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
36. Reply to Piquereau and Perros and to Pullamsetti and de Jesus Perez.
- Author
-
Kurakula K, Boucherat O, Van der Feen DE, Bogaard HJ, Kulikowski E, Bartelds B, Provencher S, Berger RMF, Bonnet S, and Goumans MJ
- Subjects
- Familial Primary Pulmonary Hypertension, Humans, Pulmonary Arterial Hypertension
- Published
- 2019
- Full Text
- View/download PDF
37. Autophagy contributes to BMP type 2 receptor degradation and development of pulmonary arterial hypertension.
- Author
-
Gomez-Puerto MC, van Zuijen I, Huang CJ, Szulcek R, Pan X, van Dinther MA, Kurakula K, Wiesmeijer CC, Goumans MJ, Bogaard HJ, Morrell NW, Rana AA, and Ten Dijke P
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Arterial Pressure, Bone Morphogenetic Protein Receptors, Type II genetics, Cell Line, Cytokines metabolism, Disease Models, Animal, Endothelial Cells pathology, Female, Heterozygote, Humans, Inflammation Mediators metabolism, Lysosomes metabolism, Lysosomes pathology, Male, Microtubule-Associated Proteins metabolism, Middle Aged, Muscle, Smooth, Vascular pathology, Muscle, Smooth, Vascular physiopathology, Myocytes, Smooth Muscle pathology, Proteolysis, Pulmonary Arterial Hypertension pathology, Pulmonary Arterial Hypertension physiopathology, Pulmonary Artery metabolism, Pulmonary Artery pathology, Pulmonary Artery physiopathology, Rats, Signal Transduction, Young Adult, Autophagy, Bone Morphogenetic Protein Receptors, Type II metabolism, Endothelial Cells metabolism, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism, Pulmonary Arterial Hypertension metabolism
- Abstract
Pulmonary arterial hypertension (PAH) is characterised by an increase in mean pulmonary arterial pressure which almost invariably leads to right heart failure and premature death. More than 70% of familial PAH and 20% of idiopathic PAH patients carry heterozygous mutations in the bone morphogenetic protein (BMP) type 2 receptor (BMPR2). However, the incomplete penetrance of BMPR2 mutations suggests that other genetic and environmental factors contribute to the disease. In the current study, we investigate the contribution of autophagy in the degradation of BMPR2 in pulmonary vascular cells. We demonstrate that endogenous BMPR2 is degraded through the lysosome in primary human pulmonary artery endothelial (PAECs) and smooth muscle cells (PASMCs): two cell types that play a key role in the pathology of the disease. By means of an elegant HaloTag system, we show that a block in lysosomal degradation leads to increased levels of BMPR2 at the plasma membrane. In addition, pharmacological or genetic manipulations of autophagy allow us to conclude that autophagy activation contributes to BMPR2 degradation. It has to be further investigated whether the role of autophagy in the degradation of BMPR2 is direct or through the modulation of the endocytic pathway. Interestingly, using an iPSC-derived endothelial cell model, our findings indicate that BMPR2 heterozygosity alone is sufficient to cause an increased autophagic flux. Besides BMPR2 heterozygosity, pro-inflammatory cytokines also contribute to an augmented autophagy in lung vascular cells. Furthermore, we demonstrate an increase in microtubule-associated protein 1 light chain 3 beta (MAP1LC3B) levels in lung sections from PAH induced in rats. Accordingly, pulmonary microvascular endothelial cells (MVECs) from end-stage idiopathic PAH patients present an elevated autophagic flux. Our findings support a model in which an increased autophagic flux in PAH patients contributes to a greater decrease in BMPR2 levels. Altogether, this study sheds light on the basic mechanisms of BMPR2 degradation and highlights a crucial role for autophagy in PAH. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland., (© 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.)
- Published
- 2019
- Full Text
- View/download PDF
38. Multicenter Preclinical Validation of BET Inhibition for the Treatment of Pulmonary Arterial Hypertension.
- Author
-
Van der Feen DE, Kurakula K, Tremblay E, Boucherat O, Bossers GPL, Szulcek R, Bourgeois A, Lampron MC, Habbout K, Martineau S, Paulin R, Kulikowski E, Jahagirdar R, Schalij I, Bogaard HJ, Bartelds B, Provencher S, Berger RMF, Bonnet S, and Goumans MJ
- Subjects
- Animals, Apoptosis drug effects, Cell Cycle Proteins antagonists & inhibitors, Cell Cycle Proteins genetics, Cell Proliferation drug effects, DNA Repair, Disease Models, Animal, Endothelial Cells drug effects, Forkhead Box Protein M1 genetics, Gene Expression Regulation, Humans, Inflammation, Microvessels metabolism, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle drug effects, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins genetics, Pulmonary Arterial Hypertension metabolism, Pulmonary Artery cytology, Rats, Transcription Factors antagonists & inhibitors, Polo-Like Kinase 1, Cell Cycle Proteins metabolism, Endothelial Cells metabolism, Myocytes, Smooth Muscle metabolism, Pulmonary Arterial Hypertension genetics, Pulmonary Artery metabolism, Quinazolinones pharmacology, Transcription Factors metabolism, Vascular Remodeling drug effects
- Abstract
Rationale: Pulmonary arterial hypertension (PAH) is a degenerative arteriopathy that leads to right ventricular (RV) failure. BRD4 (bromodomain-containing protein 4), a member of the BET (bromodomain and extra-terminal motif) family, has been identified as a critical epigenetic driver for cardiovascular diseases. Objectives: To explore the therapeutic potential in PAH of RVX208, a clinically available BET inhibitor. Methods: Microvascular endothelial cells, smooth muscle cells isolated from distal pulmonary arteries of patients with PAH, rats with Sugen5416 + hypoxia- or monocrotaline + shunt-induced PAH, and rats with RV pressure overload induced by pulmonary artery banding were treated with RVX208 in three independent laboratories. Measurements and Main Results: BRD4 is upregulated in the remodeled pulmonary vasculature of patients with PAH, where it regulates FoxM1 and PLK1, proteins implicated in the DNA damage response. RVX208 normalized the hyperproliferative, apoptosis-resistant, and inflammatory phenotype of microvascular endothelial cells and smooth muscle cells isolated from patients with PAH. Oral treatment with RVX208 reversed vascular remodeling and improved pulmonary hemodynamics in two independent trials in Sugen5416 + hypoxia-PAH and in monocrotaline + shunt-PAH. RVX208 could be combined safely with contemporary PAH standard of care. RVX208 treatment also supported the pressure-loaded RV in pulmonary artery banding rats. Conclusions: RVX208, a clinically available BET inhibitor, modulates proproliferative, prosurvival, and proinflammatory pathways, potentially through interactions with FoxM1 and PLK1. This reversed the PAH phenotype in isolated PAH microvascular endothelial cells and smooth muscle cells in vitro , and in diverse PAH rat models. RVX208 also supported the pressure-loaded RV in vivo . Together, these data support the establishment of a clinical trial with RVX208 in patients with PAH.
- Published
- 2019
- Full Text
- View/download PDF
39. Prevention of progression of pulmonary hypertension by the Nur77 agonist 6-mercaptopurine: role of BMP signalling.
- Author
-
Kurakula K, Sun XQ, Happé C, da Silva Goncalves Bos D, Szulcek R, Schalij I, Wiesmeijer KC, Lodder K, Tu L, Guignabert C, de Vries CJM, de Man FS, Vonk Noordegraaf A, Ten Dijke P, Goumans MJ, and Bogaard HJ
- Subjects
- Animals, Cell Proliferation, Culture Media, Conditioned, Disease Models, Animal, Disease Progression, Endothelial Cells drug effects, HEK293 Cells, Humans, Inflammation, Lung drug effects, Male, Microcirculation, Rats, Rats, Sprague-Dawley, Signal Transduction, Vascular Remodeling, Bone Morphogenetic Proteins metabolism, Hypertension, Pulmonary drug therapy, Mercaptopurine pharmacology, Nuclear Receptor Subfamily 4, Group A, Member 1 agonists
- Abstract
Pulmonary arterial hypertension (PAH) is a progressive fatal disease characterised by abnormal remodelling of pulmonary vessels, leading to increased vascular resistance and right ventricle failure. This abnormal vascular remodelling is associated with endothelial cell dysfunction, increased proliferation of smooth muscle cells, inflammation and impaired bone morphogenetic protein (BMP) signalling. Orphan nuclear receptor Nur77 is a key regulator of proliferation and inflammation in vascular cells, but its role in impaired BMP signalling and vascular remodelling in PAH is unknown.We hypothesised that activation of Nur77 by 6-mercaptopurine (6-MP) would improve PAH by inhibiting endothelial cell dysfunction and vascular remodelling.Nur77 expression is decreased in cultured pulmonary microvascular endothelial cells (MVECs) and lungs of PAH patients. Nur77 significantly increased BMP signalling and strongly decreased proliferation and inflammation in MVECs. In addition, conditioned medium from PAH MVECs overexpressing Nur77 inhibited the growth of healthy smooth muscle cells. Pharmacological activation of Nur77 by 6-MP markedly restored MVEC function by normalising proliferation, inflammation and BMP signalling. Finally, 6-MP prevented and reversed abnormal vascular remodelling and right ventricle hypertrophy in the Sugen/hypoxia rat model of severe angioproliferative PAH.Our data demonstrate that Nur77 is a critical modulator in PAH by inhibiting vascular remodelling and increasing BMP signalling, and activation of Nur77 could be a promising option for the treatment of PAH., Competing Interests: Conflict of interest: X-Q. Sun has nothing to disclose. Conflict of interest: C. Happé has nothing to disclose. Conflict of interest: D. da Silva Goncalves Bos has nothing to disclose. Conflict of interest: R. Szulcek has nothing to disclose. Conflict of interest: I. Schalij has nothing to disclose. Conflict of interest: K.C. Wiesmeijer has nothing to disclose. Conflict of interest: K. Lodder has nothing to disclose. Conflict of interest: L. Tu has nothing to disclose. Conflict of interest: C. Guignabert has nothing to disclose. Conflict of interest: C.J.M. de Vries has nothing to disclose. Conflict of interest: F.S. de Man has nothing to disclose. Conflict of interest: A. Vonk Noordegraaf has nothing to disclose. Conflict of interest: P. ten Dijke has nothing to disclose. Conflict of interest: M-J. Goumans has nothing to disclose. Conflict of interest: H.J. Bogaard has nothing to disclose. Conflict of interest: K. Kurakula has nothing to disclose., (Copyright ©ERS 2019.)
- Published
- 2019
- Full Text
- View/download PDF
40. Nintedanib improves cardiac fibrosis but leaves pulmonary vascular remodelling unaltered in experimental pulmonary hypertension.
- Author
-
Rol N, de Raaf MA, Sun XQ, Kuiper VP, da Silva Gonçalves Bos D, Happé C, Kurakula K, Dickhoff C, Thuillet R, Tu L, Guignabert C, Schalij I, Lodder K, Pan X, Herrmann FE, van Nieuw Amerongen GP, Koolwijk P, Vonk-Noordegraaf A, de Man FS, Wollin L, Goumans MJ, Szulcek R, and Bogaard HJ
- Subjects
- Adult, Animals, Cell Proliferation drug effects, Cells, Cultured, Disease Models, Animal, Endothelial Cells drug effects, Endothelial Cells metabolism, Endothelial Cells pathology, Extracellular Matrix drug effects, Extracellular Matrix metabolism, Extracellular Matrix pathology, Female, Fibroblasts drug effects, Fibroblasts metabolism, Fibroblasts pathology, Fibrosis, Humans, Male, Myocardium metabolism, Pulmonary Arterial Hypertension metabolism, Pulmonary Arterial Hypertension pathology, Pulmonary Arterial Hypertension physiopathology, Pulmonary Artery metabolism, Pulmonary Artery pathology, Pulmonary Artery physiopathology, Pyrroles, Rats, Sprague-Dawley, Young Adult, Indoles pharmacology, Myocardium pathology, Protein Kinase Inhibitors pharmacology, Pulmonary Arterial Hypertension drug therapy, Pulmonary Artery drug effects, Vascular Remodeling drug effects, Ventricular Function, Right drug effects, Ventricular Remodeling drug effects
- Abstract
Aims: Pulmonary arterial hypertension (PAH) is associated with increased levels of circulating growth factors and corresponding receptors such as platelet derived growth factor, fibroblast growth factor and vascular endothelial growth factor. Nintedanib, a tyrosine kinase inhibitor targeting primarily these receptors, is approved for the treatment of patients with idiopathic pulmonary fibrosis. Our objective was to examine the effect of nintedanib on proliferation of human pulmonary microvascular endothelial cells (MVEC) and assess its effects in rats with advanced experimental pulmonary hypertension (PH)., Methods and Results: Proliferation was assessed in control and PAH MVEC exposed to nintedanib. PH was induced in rats by subcutaneous injection of Sugen (SU5416) and subsequent exposure to 10% hypoxia for 4 weeks (SuHx model). Four weeks after re-exposure to normoxia, nintedanib was administered once daily for 3 weeks. Effects of the treatment were assessed with echocardiography, right heart catheterization, and histological analysis of the heart and lungs. Changes in extracellular matrix production was assessed in human cardiac fibroblasts stimulated with nintedanib. Decreased proliferation with nintedanib was observed in control MVEC, but not in PAH patient derived MVEC. Nintedanib treatment did not affect right ventricular (RV) systolic pressure or total pulmonary resistance index in SuHx rats and had no effects on pulmonary vascular remodelling. However, despite unaltered pressure overload, the right ventricle showed less dilatation and decreased fibrosis, hypertrophy, and collagen type III with nintedanib treatment. This could be explained by less fibronectin production by cardiac fibroblasts exposed to nintedanib., Conclusion: Nintedanib inhibits proliferation of pulmonary MVECs from controls, but not from PAH patients. While in rats with experimental PH nintedanib has no effects on the pulmonary vascular pathology, it has favourable effects on RV remodelling.
- Published
- 2019
- Full Text
- View/download PDF
41. Structural and cellular mechanisms of peptidyl-prolyl isomerase Pin1-mediated enhancement of Tissue Factor gene expression, protein half-life, and pro-coagulant activity.
- Author
-
Kurakula K, Koenis DS, Herzik MA Jr, Liu Y, Craft JW Jr, van Loenen PB, Vos M, Tran MK, Versteeg HH, Goumans MTH, Ruf W, de Vries CJM, and Şen M
- Subjects
- Human Umbilical Vein Endothelial Cells, Humans, Multiprotein Complexes metabolism, NF-kappa B metabolism, Protein Binding, Protein Interaction Domains and Motifs, Proteolysis, Thromboplastin chemistry, Transcription Factor AP-1 metabolism, Coagulants metabolism, Gene Expression, NIMA-Interacting Peptidylprolyl Isomerase metabolism, Thromboplastin genetics, Thromboplastin metabolism
- Abstract
Tissue Factor is a cell-surface glycoprotein expressed in various cells of the vasculature and is the principal regulator of the blood coagulation cascade and hemostasis. Notably, aberrant expression of Tissue Factor is associated with cardiovascular pathologies such as atherosclerosis and thrombosis. Here, we sought to identify factors that regulate Tissue Factor gene expression and activity. Tissue Factor gene expression is regulated by various transcription factors, including activating protein-1 and nuclear factor-κ B. The peptidyl-prolyl isomerase Pin1 is known to modulate the activity of these two transcription factors, and we now show that Pin1 augments Tissue Factor gene expression in both vascular smooth muscle cells and activated endothelial cells via activating protein-1 and nuclear factor-κ B signaling. Furthermore, the cytoplasmic domain of Tissue Factor contains a well-conserved phospho-Ser258-Pro259 amino-acid motif recognized by Pin1. Using co-immunoprecipitation and solution nuclear magnetic resonance spectroscopy, we show that the WW-domain of Pin1 directly binds the cytoplasmic domain of Tissue Factor. This interaction occurs via the phospho-Ser258-Pro259 sequence in the Tissue Factor cytoplasmic domain and results in increased protein half-life and pro-coagulant activity. Taken together, our results establish Pin1 as an upstream regulator of Tissue Factor-mediated coagulation, thereby opening up new avenues for research into the use of specific Pin1 inhibitors for the treatment of diseases characterized by pathological coagulation, such as thrombosis and atherosclerosis., (Copyright © 2018 Ferrata Storti Foundation.)
- Published
- 2018
- Full Text
- View/download PDF
42. Endothelial dysfunction in pulmonary arterial hypertension: loss of cilia length regulation upon cytokine stimulation.
- Author
-
Dummer A, Rol N, Szulcek R, Kurakula K, Pan X, Visser BI, Bogaard HJ, DeRuiter MC, Goumans MJ, and Hierck BP
- Abstract
Pulmonary arterial hypertension (PAH) is a syndrome characterized by progressive lung vascular remodelling, endothelial cell (EC) dysfunction, and excessive inflammation. The primary cilium is a sensory antenna that integrates signalling and fine tunes EC responses to various stimuli. Yet, cilia function in the context of deregulated immunity in PAH remains obscure. We hypothesized that cilia function is impaired in ECs from patients with PAH due to their inflammatory status and tested whether cilia length changes in response to cytokines. Primary human pulmonary and mouse embryonic EC were exposed to pro- (TNFα, IL1β, and IFNγ) and/or anti-inflammatory (IL-10) cytokines and cilia length was quantified. Chronic treatment with all tested inflammatory cytokines led to a significant elongation of cilia in both control human and mouse EC (by ∼1 µm, P < 0.001). This structural response was PKA/PKC dependent. Intriguingly, withdrawal of the inflammatory stimulus did not reduce cilia length. IL-10, on the other hand, blocked and reversed the pro-inflammatory cytokine-induced cilia elongation in healthy ECs, but did not influence basal length. Conversely, primary cilia of ECs from PAH patients were significantly longer under basal conditions compared to controls (1.86 ± 0.02 vs. 2.43 ± 0.08 µm, P = 0.002). These cilia did not elongate further upon pro-inflammatory stimulation and anti-inflammatory treatment did not impact cilia length. The missing length modulation was specific to cytokine stimulation, as application of fluid shear stress led to increased cilia length in the PAH endothelium. We identified loss of cilia length regulation upon cytokine stimulation as part of the endothelial dysfunction in PAH.
- Published
- 2018
- Full Text
- View/download PDF
43. Contribution of Impaired Parasympathetic Activity to Right Ventricular Dysfunction and Pulmonary Vascular Remodeling in Pulmonary Arterial Hypertension.
- Author
-
da Silva Gonçalves Bós D, Van Der Bruggen CEE, Kurakula K, Sun XQ, Casali KR, Casali AG, Rol N, Szulcek R, Dos Remedios C, Guignabert C, Tu L, Dorfmüller P, Humbert M, Wijnker PJM, Kuster DWD, van der Velden J, Goumans MJ, Bogaard HJ, Vonk-Noordegraaf A, de Man FS, and Handoko ML
- Subjects
- Animals, Cells, Cultured, Disease Models, Animal, Humans, Hypertension, Pulmonary drug therapy, Male, Organ Culture Techniques, Rats, Rats, Sprague-Dawley, Vascular Remodeling, Ventricular Dysfunction, Right drug therapy, Ventricular Function, Right, Cholinesterase Inhibitors therapeutic use, Endothelium, Vascular pathology, Hypertension, Pulmonary metabolism, Parasympathetic Nervous System, Pulmonary Artery pathology, Pyridostigmine Bromide therapeutic use, Ventricular Dysfunction, Right metabolism
- Abstract
Background: The beneficial effects of parasympathetic stimulation have been reported in left heart failure, but whether it would be beneficial for pulmonary arterial hypertension (PAH) remains to be explored. Here, we investigated the relationship between parasympathetic activity and right ventricular (RV) function in patients with PAH, and the potential therapeutic effects of pyridostigmine (PYR), an oral drug stimulating the parasympathetic activity through acetylcholinesterase inhibition, in experimental pulmonary hypertension (PH)., Methods: Heart rate recovery after a maximal cardiopulmonary exercise test was used as a surrogate for parasympathetic activity. RV ejection fraction was assessed in 112 patients with PAH. Expression of nicotinic (α-7 nicotinic acetylcholine receptor) and muscarinic (muscarinic acetylcholine type 2 receptor) receptors, and acetylcholinesterase activity were evaluated in RV (n=11) and lungs (n=7) from patients with PAH undergoing heart/lung transplantation and compared with tissue obtained from controls. In addition, we investigated the effects of PYR (40 mg/kg per day) in experimental PH. PH was induced in male rats by SU5416 (25 mg/kg subcutaneously) injection followed by 4 weeks of hypoxia. In a subgroup, sympathetic/parasympathetic modulation was assessed by power spectral analysis. At week 6, PH status was confirmed by echocardiography, and rats were randomly assigned to vehicle or treatment (both n=12). At the end of the study, echocardiography was repeated, with additional RV pressure-volume measurements, along with lung, RV histological, and protein analyses., Results: Patients with PAH with lower RV ejection fraction (<41%) had a significantly reduced heart rate recovery in comparison with patients with higher RV ejection fraction. In PAH RV samples, α-7 nicotinic acetylcholine receptor was increased and acetylcholinesterase activity was reduced versus controls. No difference in muscarinic acetylcholine type 2 receptor expression was observed. Chronic PYR treatment in PH rats normalized the cardiovascular autonomic function, demonstrated by an increase in parasympathetic activity and baroreflex sensitivity. PYR improved survival, increased RV contractility, and reduced RV stiffness, RV hypertrophy, RV fibrosis, RV inflammation, and RV α-7 nicotinic acetylcholine receptor and muscarinic acetylcholine type 2 receptor expression, as well. Furthermore, PYR reduced pulmonary vascular resistance, RV afterload, and pulmonary vascular remodeling, which was associated with reduced local and systemic inflammation., Conclusions: RV dysfunction is associated with reduced systemic parasympathetic activity in patients with PAH, with an inadequate adaptive response of the cholinergic system in the RV. Enhancing parasympathetic activity by PYR improved survival, RV function, and pulmonary vascular remodeling in experimental PH., (© 2017 American Heart Association, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
44. Thoracic Aortic Aneurysm Development in Patients with Bicuspid Aortic Valve: What Is the Role of Endothelial Cells?
- Author
-
van de Pol V, Kurakula K, DeRuiter MC, and Goumans MJ
- Abstract
Bicuspid aortic valve (BAV) is the most common type of congenital cardiac malformation. Patients with a BAV have a predisposition for the development of thoracic aortic aneurysm (TAA). This pathological aortic dilation may result in aortic rupture, which is fatal in most cases. The abnormal aortic morphology of TAAs results from a complex series of events that alter the cellular structure and extracellular matrix (ECM) composition of the aortic wall. Because the major degeneration is located in the media of the aorta, most studies aim to unravel impaired smooth muscle cell (SMC) function in BAV TAA. However, recent studies suggest that endothelial cells play a key role in both the initiation and progression of TAAs by influencing the medial layer. Aortic endothelial cells are activated in BAV mediated TAAs and have a substantial influence on ECM composition and SMC phenotype, by secreting several key growth factors and matrix modulating enzymes. In recent years there have been significant advances in the genetic and molecular understanding of endothelial cells in BAV associated TAAs. In this review, the involvement of the endothelial cells in BAV TAA pathogenesis is discussed. Endothelial cell functioning in vessel homeostasis, flow response and signaling will be highlighted to give an overview of the importance and the under investigated potential of endothelial cells in BAV-associated TAA.
- Published
- 2017
- Full Text
- View/download PDF
45. Protein-protein interactions of the LIM-only protein FHL2 and functional implication of the interactions relevant in cardiovascular disease.
- Author
-
Tran MK, Kurakula K, Koenis DS, and de Vries CJ
- Subjects
- Cell Membrane metabolism, Cell Nucleus metabolism, Cytoplasm metabolism, Humans, LIM-Homeodomain Proteins chemistry, Models, Biological, Models, Molecular, Muscle Proteins chemistry, Protein Structure, Tertiary, Transcription Factors chemistry, Cardiovascular Diseases metabolism, LIM-Homeodomain Proteins metabolism, Muscle Proteins metabolism, Protein Interaction Mapping methods, Transcription Factors metabolism
- Abstract
FHL2 belongs to the LIM-domain only proteins and contains four and a half LIM domains, each of which are composed of two zinc finger structures. FHL2 exhibits specific interaction with proteins exhibiting diverse functions, including transmembrane receptors, transcription factors and transcription co-regulators, enzymes, and structural proteins. The function of these proteins is regulated by FHL2, which modulates intracellular signal transduction pathways involved in a plethora of cellular tasks. The present review summarizes the current knowledge on the protein interactome of FHL2 and provides an overview of the functional implication of these interactions in apoptosis, migration, and regulation of nuclear receptor function. FHL2 was originally identified in the heart and there is extensive literature available on the role of FHL2 in the cardiovascular system, which is also summarized in this review., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
46. LIM-only protein FHL2 regulates experimental pulmonary Schistosoma mansoni egg granuloma formation.
- Author
-
Kurakula K, Vos M, van Eijk M, Smits HH, and de Vries CJ
- Subjects
- Animals, Cell Movement immunology, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Granuloma, Respiratory Tract pathology, Immunohistochemistry, Inflammation immunology, Inflammation pathology, Macrophages immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Real-Time Polymerase Chain Reaction, Schistosomiasis mansoni pathology, Granuloma, Respiratory Tract immunology, LIM-Homeodomain Proteins immunology, Muscle Proteins immunology, Schistosomiasis mansoni immunology, Transcription Factors immunology
- Abstract
LIM-only protein FHL2 is associated with several immune and inflammatory diseases such as arthritis, influenza A virus infection, and lung inflammation. However, the role of FHL2 in macrophage differentiation and in the development of granuloma formation is unknown. Here, we show that expression of FHL2 is induced in mouse bone marrow derived macrophages (BMMs) following stimulation with M2 cytokines such as IL-4 and IL-10. FHL2-knockout (FHL2-KO) BMMs exhibit a proinflammatory M1 phenotype after LPS treatment and display a reduced anti-inflammatory M2 phenotype following IL-4 treatment. Furthermore, thioglycollate-induced migration of macrophages and B cells is enhanced in FHL2-KO mice. To evaluate the importance of FHL2 in the development of pulmonary granuloma formation, FHL2-KO mice were challenged with Schistosoma mansoni eggs. FHL2-KO mice show an enhanced number of granulomas and display decreased expression of Th2 markers and an exacerbated Th1 type of inflammation, characterized by enhanced expression of neutrophil markers and Th1 cytokines. Furthermore, the expression of barrier proteins is reduced in FHL2-KO lung compared to WT. Collectively, these data identify a previously unrecognized role for FHL2 in the pathogenesis of pulmonary granulomatous inflammation, partly through its effect on macrophage polarization, modulation of the Th1/Th2 balance and regulation of permeability in lung., (© 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2015
- Full Text
- View/download PDF
47. Nuclear Receptor Nur77 Attenuates Airway Inflammation in Mice by Suppressing NF-κB Activity in Lung Epithelial Cells.
- Author
-
Kurakula K, Vos M, Logiantara A, Roelofs JJ, Nieuwenhuis MA, Koppelman GH, Postma DS, van Rijt LS, and de Vries CJ
- Subjects
- Alleles, Animals, Asthma genetics, Asthma immunology, Asthma metabolism, Asthma pathology, Bronchoalveolar Lavage Fluid immunology, Cytokines metabolism, Disease Models, Animal, Enzyme Activation, Eosinophils pathology, Gene Expression, Immunoglobulin G blood, Immunoglobulin G immunology, Inflammation genetics, Inflammation immunology, Inflammation pathology, Leukocyte Count, Lymph Nodes immunology, Mice, Mice, Knockout, Mucin 5AC genetics, Mucin 5AC metabolism, NF-kappa B metabolism, Nuclear Receptor Subfamily 4, Group A, Member 1 deficiency, Nuclear Receptor Subfamily 4, Group A, Member 1 genetics, Odds Ratio, Polymorphism, Single Nucleotide, Respiratory Hypersensitivity genetics, Respiratory Hypersensitivity immunology, Respiratory Hypersensitivity pathology, Signal Transduction, Alveolar Epithelial Cells metabolism, Inflammation metabolism, Nuclear Receptor Subfamily 4, Group A, Member 1 metabolism, Respiratory Hypersensitivity metabolism
- Abstract
Allergic asthma is characterized by persistent chronic airway inflammation, which leads to mucus hypersecretion and airway hyperresponsiveness. Nuclear receptor Nur77 plays a pivotal role in distinct immune and inflammatory cells and is expressed in eosinophils and lung epithelium. However, the role of Nur77 in allergic airway inflammation has not been studied so far. In the present study, we determined the role of Nur77 in airway inflammation using a murine model of OVA-induced allergic airway inflammation. We found that OVA-challenged Nur77 knockout (KO) mice show significantly enhanced infiltration of inflammatory cells, including eosinophils and lymphocytes, and aggravated mucus production. The infiltration of macrophages is limited in this model and was similar in wild-type and Nur77 KO mice. Higher levels of Th2 cytokines were found in bronchoalveolar lavage fluid and draining lymph node cells of Nur77-KO mice, as well as increased serum IgG1 and IgG2a levels. Knockdown of Nur77 in human lung epithelial cells resulted in a marked increase in IκBα phosphorylation, corresponding with elevated NF-κB activity, whereas Nur77 overexpression decreased NF-κB activity. Consistently, Nur77 significantly decreased mRNA levels of inflammatory cytokines and Muc5ac expression and also attenuated mucus production in lung epithelial cells. To further corroborate these findings, we searched for association of single nucleotide polymorphisms in Nur77 gene with asthma and with the severity of bronchial hyperresponsiveness. We identified three Nur77 single nucleotide polymorphisms showing association with severity of bronchial hyperresponsiveness in asthma patients. Collectively, these findings support a protective role of Nur77 in OVA-induced airway inflammation and identify Nur77 as a novel therapeutic target for airway inflammation., (Copyright © 2015 by The American Association of Immunologists, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
48. Regulatory RNAs controlling vascular (dys)function by affecting TGF-ß family signalling.
- Author
-
Kurakula K, Goumans MJ, and Ten Dijke P
- Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide. Over the last few years, microRNAs (miRNAs) have emerged as master regulators of gene expression in cardiovascular biology and disease. miRNAs are small endogenous non-coding RNAs that usually bind to 3' untranslated region (UTR) of their target mRNAs and inhibit mRNA stability or translation of their target genes. miRNAs play a dynamic role in the pathophysiology of many CVDs through their effects on target mRNAs in vascular cells. Recently, numerous miRNAs have been implicated in the regulation of the transforming growth factor-β (TGF-β)/bone morphogenetic protein (BMP) signalling pathway which plays crucial roles in diverse biological processes, and is involved in pathogenesis of many diseases including CVD. This review gives an overview of current literature on the role of miRNAs targeting TGF-β/BMP signalling in vascular cells, including endothelial cells and smooth muscle cells. We also provide insight into how this miRNA-mediated regulation of TGF-β/BMP signalling might be used to harness CVD.
- Published
- 2015
- Full Text
- View/download PDF
49. 6-Mercaptopurine reduces cytokine and Muc5ac expression involving inhibition of NFκB activation in airway epithelial cells.
- Author
-
Kurakula K, Hamers AA, van Loenen P, and de Vries CJ
- Subjects
- Cell Line, Tumor, Cell Survival drug effects, Cell Survival physiology, Cytokines antagonists & inhibitors, Dose-Response Relationship, Drug, Epithelial Cells drug effects, Gene Expression Regulation, Humans, Mucin 5AC antagonists & inhibitors, NF-kappa B antagonists & inhibitors, Respiratory Mucosa drug effects, Cytokines biosynthesis, Epithelial Cells metabolism, Mercaptopurine pharmacology, Mucin 5AC biosynthesis, NF-kappa B metabolism, Respiratory Mucosa metabolism
- Abstract
Background: Mucus hypersecretion and excessive cytokine synthesis is associated with many of the pathologic features of chronic airway diseases such as asthma. 6-Mercaptopurine (6-MP) is an immunosuppressive drug that is widely used in several inflammatory disorders. Although 6-MP has been used to treat asthma, its function and mechanism of action in airway epithelial cells is unknown., Methods: Confluent NCI-H292 and MLE-12 epithelial cells were pretreated with 6-MP followed by stimulation with TNFα or PMA. mRNA levels of cytokines and mucins were measured by RT-PCR. Western blot analysis was performed to assess the phosphorylation of IκBα and luciferase assays were performed using an NFκB reporter plasmid to determine NFκB activity. Periodic Acid Schiff staining was used to assess the production of mucus., Results: 6-MP displayed no effect on cell viability up to a concentration of 15 μM. RT-PCR analysis showed that 6-MP significantly reduces TNFα- and PMA-induced expression of several proinflammatory cytokines in NCI-H292 and MLE-12 cells. Consistent with this, we demonstrated that 6-MP strongly inhibits TNFα-induced phosphorylation of IκBα and thus attenuates NFκB luciferase reporter activity. In addition, 6-MP decreases Rac1 activity in MLE-12 cells. 6-MP down-regulates gene expression of the mucin Muc5ac, but not Muc2, through inhibition of activation of the NFκB pathway. Furthermore, PMA- and TNFα-induced mucus production, as visualized by Periodic Acid Schiff (PAS) staining, is decreased by 6-MP., Conclusions: Our data demonstrate that 6-MP inhibits Muc5ac gene expression and mucus production in airway epithelial cells through inhibition of the NFκB pathway, and 6-MP may represent a novel therapeutic target for mucus hypersecretion in airway diseases.
- Published
- 2015
- Full Text
- View/download PDF
50. Cardiac endothelial cells express Wilms' tumor-1: Wt1 expression in the developing, adult and infarcted heart.
- Author
-
Duim SN, Kurakula K, Goumans MJ, and Kruithof BP
- Subjects
- Animals, Cell Hypoxia, Cell Movement, Cell Proliferation, Collagen chemistry, Coronary Vessels metabolism, Coronary Vessels pathology, Cyclin D1 genetics, Cyclin D1 metabolism, Drug Combinations, Embryo, Mammalian, Endothelial Cells pathology, Female, Gene Expression Regulation, Developmental, Laminin chemistry, Mice, Mice, Inbred C57BL, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardium pathology, Myocytes, Cardiac pathology, Pericardium pathology, Proteoglycans chemistry, Repressor Proteins metabolism, Signal Transduction, Stem Cells pathology, WT1 Proteins, Endothelial Cells metabolism, Myocardial Infarction genetics, Myocardium metabolism, Myocytes, Cardiac metabolism, Pericardium metabolism, Repressor Proteins genetics, Stem Cells metabolism
- Abstract
Myocardial infarction is the leading cause of death worldwide. Due to their limited regenerative capacity lost cardiomyocytes are replaced by a non-contractile fibrotic scar tissue. The epicardial layer of the heart provides cardiac progenitor cells during development. Because this layer regains embryonic characteristics in the adult heart after cardiac injury, it could serve as a promising source for resident cardiac progenitor cells. Wilms' tumor-1 (Wt1) is associated with the activation and reactivation of the epicardium and therefore potentially important for the differentiation and regenerative capacity of the epicardium. To gain more insight into the regulation of Wt1 we examined the spatiotemporal expression pattern of Wt1 during murine development and after cardiac injury. Interestingly, we found that Wt1 is expressed in the majority of the cardiac endothelial cells within the myocardial ventricular layer of the developing heart from E12.5 onwards. In the adult heart only a subset of coronary endothelial cells remains positive for Wt1. After myocardial infarction Wt1 is temporally upregulated in the endothelial cells of the infarcted area and the border zone of the heart. In vitro experiments show that endothelial Wt1 expression can be induced by hypoxia. We show that Wt1 is associated with endothelial cell proliferation: Wt1 expression is higher in proliferating endothelial cells, Wt1 knockdown inhibits the proliferation of endothelial cells, and Wt1 regulates CyclinD1 expression. Finally, endothelial cells lacking Wt1 are not capable to establish a proper vascular network in vitro. Together, these results suggest a possible role for Wt1 in cardiac vessel formation in development and disease., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.