101
|
Lai H, Chen A, Cai H, Fu J, Salem F, Li Y, He JC, Schlondorff D, Lee K. Podocyte and endothelial-specific elimination of BAMBI identifies differential transforming growth factor-β pathways contributing to diabetic glomerulopathy. Kidney Int 2020; 98:601-614. [PMID: 32739209 DOI: 10.1016/j.kint.2020.03.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 12/28/2022]
Abstract
Transforming growth factor-β (TGF-β) is a central mediator of diabetic nephropathy. The effect of TGF-β, mediated by the type I TGF-β receptor, ALK5, and subsequent Smad2/3 activation results in podocyte apoptosis and loss. Previously, we demonstrated that the genetic deletion of the BMP and Activin Membrane-Bound Inhibitor (BAMBI), a negative modulator TGF-β signaling, accelerates diabetic nephropathy in mice. This was associated with heightened ALK1-mediated activation of Smad1/5 in the glomerular endothelial cells (ECs). Therefore, to evaluate the glomerular cell-specific effects of TGF-β in diabetic nephropathy we examined the effects of the podocyte- or EC-specific loss of Bambi (Pod-Bambi-/- or EC-Bambi-/-) in streptozotocin-induced diabetic mice with endothelial nitric oxide synthase deficiency. Interestingly, although hyperglycemia and body weight loss were similar in all groups of diabetic mice, significant hypertension was present only in the diabetic EC-Bambi-/- mice. While the podocyte or EC-specific loss of BAMBI both accelerated the progression of diabetic nephropathy, the worsened podocyte injury and loss observed in the diabetic Pod-Bambi-/- mice were associated with enhanced Smad3 activation. Increased Smad1/5 activation and EC proliferation were apparent only in the glomeruli of diabetic EC-Bambi-/- mice. The enhanced Smad1/5 activation in diabetic EC-Bambi-/- mice was associated with increased glomerular expression of plasmalemma vesicle-associated protein, pointing to the involvement of immature or dedifferentiated glomerular ECs in diabetic nephropathy. Notably, diabetic EC-Bambi-/- mice displayed podocyte injury and loss that were comparable to diabetic Pod-Bambi-/- mice. Thus, our results highlight the glomerular cell-specific contribution of TGF-β signaling and the intricate cross-talk between injured glomerular cells in the progression of diabetic nephropathy.
Collapse
Affiliation(s)
- Han Lai
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Anqun Chen
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Division of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Hong Cai
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jia Fu
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fadi Salem
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yu Li
- Division of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - John C He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Kidney Center at James J Peters Veterans Affairs Medical Center, Bronx, New York, USA
| | - Detlef Schlondorff
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kyung Lee
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
102
|
Dessein H, Duflot N, Romano A, Opio C, Pereira V, Mola C, Kabaterene N, Coutinho A, Dessein A. Genetic algorithms identify individuals with high risk of severe liver disease caused by schistosomes. Hum Genet 2020; 139:821-831. [PMID: 32277285 DOI: 10.1007/s00439-020-02160-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/28/2020] [Indexed: 02/06/2023]
Abstract
Schistosomes induce severe hepatic disease, which is fatal in 2-10% of cases, mortality being higher in cases of co-infection with HBV or HCV. Hepatic disease occurs as a consequence of the chronic inflammation caused by schistosome eggs trapped in liver sinusoids. In certain individuals, the repair process leads to a massive accumulation of fibrosis in the periportal spaces. We and others have shown that genetic variants play a crucial role in disease progression from mild to severe fibrosis and explain why hepatic fibrosis progresses rapidly in certain subjects only. We will review here published findings concerning the strategies that have been used in the analysis of hepatic fibrosis in schistosome-infected individuals, the genetic variants that have associated with fibrosis, and variants in new pathways crucial for fibrosis progression. Together, these studies show that the development of fibrosis is under the tight genetic control of various common variants with moderate effects. This polygenic control has made it possible to develop models that identify schistosome-infected individual at risk of severe hepatic disease. We discuss the performances and limitations of these models.
Collapse
Affiliation(s)
- Hélia Dessein
- BILHI Genetics, 60 Avenue André Roussin, 13016, Marseille, France
- UMR_S906-Génétique Et Immunologie Des Maladies Parasitaires, Aix Marseille Université-INSERM, Marseille, France
| | - Nicolas Duflot
- BILHI Genetics, 60 Avenue André Roussin, 13016, Marseille, France
- UMR_S906-Génétique Et Immunologie Des Maladies Parasitaires, Aix Marseille Université-INSERM, Marseille, France
| | - Audrey Romano
- BILHI Genetics, 60 Avenue André Roussin, 13016, Marseille, France
- UMR_S906-Génétique Et Immunologie Des Maladies Parasitaires, Aix Marseille Université-INSERM, Marseille, France
| | - Christopher Opio
- Department of Medicine, Mulago Hospital, Makerere University College of Health Sciences, Kampala, Uganda
| | - Valeria Pereira
- Instituto Aggeu Magalhães, Fiocruz, Fundaçao Oswaldo Cruz, Av. Professor Moraes Rego, S/N Cidade Universitária, Recife, PE, 50740-465, Brazil
| | - Carla Mola
- Instituto Aggeu Magalhães, Fiocruz, Fundaçao Oswaldo Cruz, Av. Professor Moraes Rego, S/N Cidade Universitária, Recife, PE, 50740-465, Brazil
| | - Narcis Kabaterene
- Vector Control Division Uganda, Ministry of Health, Queen's Ln, Kampala, Uganda
| | - Ana Coutinho
- Fundação Oswaldo Cruz Rio de Janeiro, Av. Brasil, 4365, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Alain Dessein
- BILHI Genetics, 60 Avenue André Roussin, 13016, Marseille, France.
- UMR_S906-Génétique Et Immunologie Des Maladies Parasitaires, Aix Marseille Université-INSERM, Marseille, France.
| |
Collapse
|
103
|
Peacock HM, Tabibian A, Criem N, Caolo V, Hamard L, Deryckere A, Haefliger JA, Kwak BR, Zwijsen A, Jones EAV. Impaired SMAD1/5 Mechanotransduction and Cx37 (Connexin37) Expression Enable Pathological Vessel Enlargement and Shunting. Arterioscler Thromb Vasc Biol 2020; 40:e87-e104. [PMID: 32078368 DOI: 10.1161/atvbaha.119.313122] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Impaired ALK1 (activin receptor-like kinase-1)/Endoglin/BMP9 (bone morphogenetic protein 9) signaling predisposes to arteriovenous malformations (AVMs). Activation of SMAD1/5 signaling can be enhanced by shear stress. In the genetic disease hereditary hemorrhagic telangiectasia, which is characterized by arteriovenous malformations, the affected receptors are those involved in the activation of mechanosensitive SMAD1/5 signaling. To elucidate how genetic and mechanical signals interact in AVM development, we sought to identify targets differentially regulated by BMP9 and shear stress. Approach and Results: We identify Cx37 (Connexin37) as a differentially regulated target of ligand-induced and mechanotransduced SMAD1/5 signaling. We show that stimulation of endothelial cells with BMP9 upregulated Cx37, whereas shear stress inhibited this expression. This signaling was SMAD1/5-dependent, and in the absence of SMAD1/5, there was an inversion of the expression pattern. Ablated SMAD1/5 signaling alone caused AVM-like vascular malformations directly connecting the dorsal aorta to the inlet of the heart. In yolk sacs of mouse embryos with an endothelial-specific compound heterozygosity for SMAD1/5, addition of TNFα (tumor necrosis factor-α), which downregulates Cx37, induced development of these direct connections bypassing the yolk sac capillary bed. In wild-type embryos undergoing vascular remodeling, Cx37 was globally expressed by endothelial cells but was absent in regions of enlarging vessels. TNFα and endothelial-specific compound heterozygosity for SMAD1/5 caused ectopic regions lacking Cx37 expression, which correlated to areas of vascular malformations. Mechanistically, loss of Cx37 impairs correct directional migration under flow conditions. CONCLUSIONS Our data demonstrate that Cx37 expression is differentially regulated by shear stress and SMAD1/5 signaling, and that reduced Cx37 expression is permissive for capillary enlargement into shunts.
Collapse
Affiliation(s)
- Hanna M Peacock
- From the Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (H.M.P., A.T., N.C., A.Z., E.A.V.J.), KU Leuven, Belgium
| | - Ashkan Tabibian
- From the Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (H.M.P., A.T., N.C., A.Z., E.A.V.J.), KU Leuven, Belgium
| | - Nathan Criem
- From the Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (H.M.P., A.T., N.C., A.Z., E.A.V.J.), KU Leuven, Belgium
| | - Vincenza Caolo
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (V.C.)
| | - Lauriane Hamard
- Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Switzerland (L.H., J.-A.H.)
| | | | - Jacques-Antoine Haefliger
- Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Switzerland (L.H., J.-A.H.)
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, Switzerland (B.R.K.)
| | - An Zwijsen
- From the Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (H.M.P., A.T., N.C., A.Z., E.A.V.J.), KU Leuven, Belgium
| | - Elizabeth A V Jones
- From the Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology (H.M.P., A.T., N.C., A.Z., E.A.V.J.), KU Leuven, Belgium
| |
Collapse
|
104
|
TMEM100 is a key factor for specification of lymphatic endothelial progenitors. Angiogenesis 2020; 23:339-355. [PMID: 32112176 DOI: 10.1007/s10456-020-09713-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 02/15/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND TMEM100 is identified as a downstream gene of bone morphogenetic protein 9 (BMP9) signaling via activin receptor-like kinase 1 (ALK1), which is known to participate in lymphangiogenesis as well as angiogenesis. TMEM100 has been shown to be important for blood vessel formation and maintenance, but its role in the development of lymphatic vasculature remains unknown. The objective is to investigate the role of TMEM100 in development of the lymphatic system. METHODS AND RESULTS Global Tmem100 gene deletion was induced by tamoxifen on 10.5 days post-coitus. Tmem100-inducible knockout (iKO) embryos in embryonic days (E)14.5-16.5 exhibited edema and blood-filled enlarged lymphatics with misconnections between veins and lymphatic vessels. For a reciprocal approach, we have generated a novel mouse line in which TMEM100 overexpression (OE) can be induced in endothelial cells by intercrossing with Tie2-Cre driver. TMEM100-OE embryos at E12.5-14.5 exhibited edema with small size and number of lymphatic vessels, the exact opposite phenotypes of Tmem100-iKOs. In Tmem100-iKO embryos, the number of progenitors of lymphatic endothelial cells (LECs) in the cardinal vein was increased, while it was decreased in TMEM100-OE embryos. The activity of NOTCH signaling, which limits the number of progenitors of LECs in the cardinal vein, was decreased in Tmem100-iKO embryos, whereas it was increased in TMEM100-OE embryos. CONCLUSION TMEM100 plays an important role in the specification of LECs in the cardinal veins, at least in part, by regulating the NOTCH signaling.
Collapse
|
105
|
Jiang H, Zhang L, Liu X, Sun W, Kato K, Chen C, Li X, Li T, Sun Z, Han W, Zhang F, Xiao Q, Yang Z, Hu J, Qin Z, Adams RH, Gao X, He Y. Angiocrine FSTL1 (Follistatin-Like Protein 1) Insufficiency Leads to Atrial and Venous Wall Fibrosis via SMAD3 Activation. Arterioscler Thromb Vasc Biol 2020; 40:958-972. [PMID: 32078339 DOI: 10.1161/atvbaha.119.313901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Angiocrine factors, mediating the endothelial-mural cell interaction in vascular wall construction as well as maintenance, are incompletely characterized. This study aims to investigate the role of endothelial cell-derived FSTL1 (follistatin-like protein 1) in vascular homeostasis. Approach and Results: Using conditional knockout mouse models, we show that loss of FSTL1 in endothelial cells (Fstl1ECKO) led to an increase of pulmonary vascular resistance, resulting in the heart regurgitation especially with tricuspid valves. However, this abnormality was not detected in mutant mice with Fstl1 knockout in smooth muscle cells or hematopoietic cells. We further showed that there was excessive αSMA (α-smooth muscle actin) associated with atrial endocardia, heart valves, veins, and microvessels after the endothelial FSTL1 deletion. There was also an increase in collagen deposition, as demonstrated in livers of Fstl1ECKO mutants. The SMAD3 (mothers against decapentaplegic homolog 3) phosphorylation (pSMAD3) was significantly enhanced, and pSMAD3 staining was colocalized with αSMA in vein walls, suggesting the activation of TGFβ (transforming growth factor β) signaling in vascular mural cells of Fstl1ECKO mice. Consistently, treatment with a TGFβ pathway inhibitor reduced the abnormal association of αSMA with the atria and blood vessels in Fstl1ECKO mutant mice. CONCLUSIONS The findings imply that endothelial FSTL1 is critical for the homeostasis of vascular walls, and its insufficiency may favor cardiovascular fibrosis leading to heart failure.
Collapse
Affiliation(s)
- Haijuan Jiang
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| | - Luqing Zhang
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.).,MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.)
| | - Xuelian Liu
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| | - Wei Sun
- MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.).,Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, China (W.S.)
| | - Katsuhiro Kato
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Germany (K.K., R.H.A.).,Department of Cardiology, Nagoya University Hospital, Japan (K.K.)
| | - Chuankai Chen
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| | - Xiao Li
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| | - Taotao Li
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| | - Zhiliang Sun
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| | - Wencan Han
- MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.)
| | - Fujing Zhang
- MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.)
| | - Qi Xiao
- MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.)
| | - Zhongzhou Yang
- MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.)
| | - Junhao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, China (J.H.)
| | - Zhihai Qin
- The First Affiliated Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, China (Z.Q.)
| | - Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Germany (K.K., R.H.A.)
| | - Xiang Gao
- MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, China (L.Z., W.S., W.H., F.Z., Q.X., Z.Y., X.G.)
| | - Yulong He
- From the Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Cam-Su Genomic Resources Center, Soochow University, Suzhou, China (H.J., L.Z., X. Liu, C.C., X. Li, T.L., Z.S., Y.H.)
| |
Collapse
|
106
|
Schoonderwoerd MJA, Goumans MJTH, Hawinkels LJAC. Endoglin: Beyond the Endothelium. Biomolecules 2020; 10:biom10020289. [PMID: 32059544 PMCID: PMC7072477 DOI: 10.3390/biom10020289] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 02/06/2023] Open
Abstract
Keywords: endoglin; CD105 TGF-β; BMP9; ALK-1; TRC105; tumor microenvironment.
Collapse
Affiliation(s)
- Mark J. A. Schoonderwoerd
- Department of Gastrenterology-Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | | | - Lukas J. A. C. Hawinkels
- Department of Gastrenterology-Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
- Correspondence: ; Tel.: +31-71-526-6736
| |
Collapse
|
107
|
Do HS, Park SW, Im I, Seo D, Yoo HW, Go H, Kim YH, Koh GY, Lee BH, Han YM. Enhanced thrombospondin-1 causes dysfunction of vascular endothelial cells derived from Fabry disease-induced pluripotent stem cells. EBioMedicine 2020; 52:102633. [PMID: 31981984 PMCID: PMC6992938 DOI: 10.1016/j.ebiom.2020.102633] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/04/2020] [Accepted: 01/07/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Fabry disease (FD) is a recessive X-linked lysosomal storage disorder caused by α-galactosidase A (GLA) deficiency. Although the mechanism is unclear, GLA deficiency causes an accumulation of globotriaosylceramide (Gb3), leading to vasculopathy. METHODS To explore the relationship between the accumulation of Gb3 and vasculopathy, induced pluripotent stem cells generated from four Fabry patients (FD-iPSCs) were differentiated into vascular endothelial cells (VECs). Genome editing using CRISPR-Cas9 system was carried out to correct the GLA mutation or to delete Thrombospondin-1 (TSP-1). Global transcriptomes were compared between wild-type (WT)- and FD-VECs by RNA-sequencing analysis. FINDINGS Here, we report that overexpression of TSP-1 contributes to the dysfunction of VECs in FD. VECs originating from FD-iPSCs (FD-VECs) showed aberrant angiogenic functionality even upon treatment with recombinant α-galactosidase. Intriguingly, FD-VECs produced more p-SMAD2 and TSP-1 than WT-VECs. We also found elevated TSP-1 in the peritubular capillaries of renal tissues biopsied from FD patients. Inhibition of SMAD2 signaling or knock out of TSP-1 (TSP-1-/-) rescues normal vascular functionality in FD-VECs, like in gene-corrected FD-VECs. In addition, the enhanced oxygen consumption rate is reduced in TSP-1-/- FD-VECs. INTERPRETATION The overexpression of TSP-1 secondary to Gb3 accumulation is primarily responsible for the observed FD-VEC dysfunction. Our findings implicate dysfunctional VEC angiogenesis in the peritubular capillaries in some of the complications of Fabry disease. FUNDING This study was supported by grant 2018M3A9H1078330 from the National Research Foundation of the Republic of Korea.
Collapse
Affiliation(s)
- Hyo-Sang Do
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Sang-Wook Park
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea; New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Ilkyun Im
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea
| | - Donghyuk Seo
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Han-Wook Yoo
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Heounjeong Go
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Yoo Hyung Kim
- College of Natural Sciences, KAIST, Daejeon 34141, Republic of Korea; Center for Vascular Research, Institute for Basic Sciences, Daejeon 34141, Republic of Korea
| | - Gou Young Koh
- Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea; Center for Vascular Research, Institute for Basic Sciences, Daejeon 34141, Republic of Korea
| | - Beom-Hee Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| | - Yong-Mahn Han
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea.
| |
Collapse
|
108
|
Lin X, Qiu W, Xiao Y, Ma J, Xu F, Zhang K, Gao Y, Chen Q, Li Y, Li H, Qian A. MiR-199b-5p Suppresses Tumor Angiogenesis Mediated by Vascular Endothelial Cells in Breast Cancer by Targeting ALK1. Front Genet 2020; 10:1397. [PMID: 32082362 PMCID: PMC7002562 DOI: 10.3389/fgene.2019.01397] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/20/2019] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis is a crucial event during cancer progression that regulates tumor growth and metastasis. Activin receptor-like kinase 1 (ALK1), predominantly expressed in endothelial cells, plays a key role in the organization of neo-angiogenic vessels. Therapeutic targeting of ALK1 has been proposed as a promising strategy for cancer treatment, and microRNAs (miRNAs) are increasingly being explored as modulators of angiogenesis. However, the regulation of ALK1 by miRNAs is unclear. In this study, we identified that ALK1 is directly targeted by miR-199b-5p, which was able to inhibit angiogenesis in vitro and in vivo. Moreover, it was found that miR-199b-5p was repressed in breast cancer cells and its expression was decreased during the VEGF-induced angiogenesis process of human umbilical vein endothelial cells (HUVECs). Overexpression of miR-199b-5p inhibited the formation of capillary-like tubular structures and migration of HUVECs. Furthermore, overexpression of miR-199b-5p inhibited the mRNA and protein expression of ALK1 in HUVECs by directly binding to its 3’UTR. Additionally, overexpression of miR-199b-5p attenuated the induction of ALK1/Smad/Id1 pathway by BMP9 in HUVECs. Finally, overexpression of miR-199b-5p reduced tumor growth and angiogenesis in in vivo. Taken together, these findings demonstrate the anti-angiogenic role of miR-199b-5p, which directly targets ALK1, suggesting that miR-199b-5p might be a potential anti-angiogenic target for cancer therapy.
Collapse
Affiliation(s)
- Xiao Lin
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Wuxia Qiu
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yunyun Xiao
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Jianhua Ma
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Fang Xu
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Kewen Zhang
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yongguang Gao
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Qiang Chen
- State Key Laboratory of Solidification Processing, Northwestern Polytechnical University, Xi'an, China
| | - Yu Li
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Hui Li
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Airong Qian
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
109
|
Grandon B, Rincheval-Arnold A, Jah N, Corsi JM, Araujo LM, Glatigny S, Prevost E, Roche D, Chiocchia G, Guénal I, Gaumer S, Breban M. HLA-B27 alters BMP/TGFβ signalling in Drosophila, revealing putative pathogenic mechanism for spondyloarthritis. Ann Rheum Dis 2019; 78:1653-1662. [PMID: 31563893 DOI: 10.1136/annrheumdis-2019-215832] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/08/2019] [Accepted: 09/09/2019] [Indexed: 01/16/2023]
Abstract
OBJECTIVES The human leucocyte antigen (HLA)-B27 confers an increased risk of spondyloarthritis (SpA) by unknown mechanism. The objective of this work was to uncover HLA-B27 non-canonical properties that could explain its pathogenicity, using a new Drosophila model. METHODS We produced transgenic Drosophila expressing the SpA-associated HLA-B*27:04 or HLA-B*27:05 subtypes, or the non-associated HLA-B*07:02 allele, alone or in combination with human β2-microglobulin (hβ2m), under tissue-specific drivers. Consequences of transgenes expression in Drosophila were examined and affected pathways were investigated by the genetic interaction experiments. Predictions of the model were further tested in immune cells from patients with SpA. RESULTS Loss of crossveins in the wings and a reduced eye phenotype were observed after expression of HLA-B*27:04 or HLA-B*27:05 in Drosophila but not in fruit flies expressing the non-associated HLA-B*07:02 allele. These HLA-B27-induced phenotypes required the presence of hβ2m that allowed expression of well-folded HLA-B conformers at the cell surface. Loss of crossveins resulted from a dominant negative effect of HLA-B27 on the type I bone morphogenetic protein (BMP) receptor saxophone (Sax) with which it interacted, resulting in elevated mothers against decapentaplegic (Mad, a Drosophila receptor-mediated Smad) phosphorylation. Likewise, in immune cells from patients with SpA, HLA-B27 specifically interacted with activin receptor-like kinase-2 (ALK2), the mammalian Sax ortholog, at the cell surface and elevated Smad phosphorylation was observed in response to activin A and transforming growth factor β (TGFβ). CONCLUSIONS Antagonistic interaction of HLA-B27 with ALK2, which exerts inhibitory functions on the TGFβ/BMP signalling pathway at the cross-road between inflammation and ossification, could adequately explain SpA development.
Collapse
Affiliation(s)
- Benjamin Grandon
- LGBC, EA4589, UVSQ/Université Paris-Saclay, EPHE/PSL Research University, Montigny-le-Bretonneux, France
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/ Université Paris Saclay, Montigny-le-Bretonneux, France
| | - Aurore Rincheval-Arnold
- LGBC, EA4589, UVSQ/Université Paris-Saclay, EPHE/PSL Research University, Montigny-le-Bretonneux, France
| | - Nadège Jah
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/ Université Paris Saclay, Montigny-le-Bretonneux, France
| | - Jean-Marc Corsi
- LGBC, EA4589, UVSQ/Université Paris-Saclay, EPHE/PSL Research University, Montigny-le-Bretonneux, France
| | - Luiza M Araujo
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/ Université Paris Saclay, Montigny-le-Bretonneux, France
| | - Simon Glatigny
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/ Université Paris Saclay, Montigny-le-Bretonneux, France
| | - Erwann Prevost
- LGBC, EA4589, UVSQ/Université Paris-Saclay, EPHE/PSL Research University, Montigny-le-Bretonneux, France
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/ Université Paris Saclay, Montigny-le-Bretonneux, France
| | - Delphine Roche
- LGBC, EA4589, UVSQ/Université Paris-Saclay, EPHE/PSL Research University, Montigny-le-Bretonneux, France
| | - Gilles Chiocchia
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/ Université Paris Saclay, Montigny-le-Bretonneux, France
| | - Isabelle Guénal
- LGBC, EA4589, UVSQ/Université Paris-Saclay, EPHE/PSL Research University, Montigny-le-Bretonneux, France
| | - Sébastien Gaumer
- LGBC, EA4589, UVSQ/Université Paris-Saclay, EPHE/PSL Research University, Montigny-le-Bretonneux, France
| | - Maxime Breban
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/ Université Paris Saclay, Montigny-le-Bretonneux, France
- Rheumatology, Ambroise Paré Hospital, Boulogne Billancourt, France
| |
Collapse
|
110
|
Epigallocatechin-3-gallate inhibits tumor angiogenesis: involvement of endoglin/Smad1 signaling in human umbilical vein endothelium cells. Biomed Pharmacother 2019; 120:109491. [DOI: 10.1016/j.biopha.2019.109491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/16/2019] [Accepted: 09/22/2019] [Indexed: 02/06/2023] Open
|
111
|
Barbosa Do Prado L, Han C, Oh SP, Su H. Recent Advances in Basic Research for Brain Arteriovenous Malformation. Int J Mol Sci 2019; 20:ijms20215324. [PMID: 31731545 PMCID: PMC6862668 DOI: 10.3390/ijms20215324] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 10/11/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023] Open
Abstract
Arteriovenous malformations (AVMs) are abnormal connections of vessels that shunt blood directly from arteries into veins. Rupture of brain AVMs (bAVMs) can cause life-threatening intracranial bleeding. Even though the majority of bAVM cases are sporadic without a family history, some cases are familial. Most of the familial cases of bAVMs are associated with a genetic disorder called hereditary hemorrhagic telangiectasia (HHT). The mechanism of bAVM formation is not fully understood. The most important advances in bAVM basic science research is the identification of somatic mutations of genes in RAS-MAPK pathways. However, the mechanisms by which mutations of these genes lead to AVM formation are largely unknown. In this review, we summarized the latest advance in bAVM studies and discussed some pathways that play important roles in bAVM pathogenesis. We also discussed the therapeutic implications of these pathways.
Collapse
Affiliation(s)
- Leandro Barbosa Do Prado
- Center for Cerebrovascular Research, Department of Anesthesia, University of California, San Francisco, CA 94143, USA;
| | - Chul Han
- Barrow Aneurysm & AVM Research Center, Barrow Neurological Institute/Dignity Health, Phoenix, AZ 85013, USA; (C.H.); (S.P.O.)
| | - S. Paul Oh
- Barrow Aneurysm & AVM Research Center, Barrow Neurological Institute/Dignity Health, Phoenix, AZ 85013, USA; (C.H.); (S.P.O.)
| | - Hua Su
- Center for Cerebrovascular Research, Department of Anesthesia, University of California, San Francisco, CA 94143, USA;
- Correspondence: ; Tel.: +01-415-206-3162
| |
Collapse
|
112
|
Abstract
The systemic circulation depends upon a highly organized, hierarchal blood vascular network that requires the successful specification of arterial and venous endothelial cells during development. This process is driven by a cascade of signaling events (including Hedgehog, vascular endothelial growth factor (VEGF), Notch, connexin (Cx), transforming growth factor-beta (TGF- β), and COUP transcription factor 2 (COUP-TFII)) to influence endothelial cell cycle status and expression of arterial or venous genes and is further regulated by hemodynamic flow. Failure of endothelial cells to properly undergo arteriovenous specification may contribute to vascular malformation and dysfunction, such as in hereditary hemorrhagic telangiectasia (HHT) and capillary malformation-arteriovenous malformation (CM-AVM) where abnormal vessel structures, such as large shunts lacking clear arteriovenous identity and function, form and compromise peripheral blood flow. This review provides an overview of recent findings in the field of arteriovenous specification and highlights key regulators of this process.
Collapse
Affiliation(s)
- Jennifer Fang
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, Irvine, CA, 92697, USA
| | - Karen Hirschi
- 2Departments of Medicine, Genetics, and Biomedical Engineering, Yale Cardiovascular Research Center, Yale Stem Cell Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| |
Collapse
|
113
|
Li KN, Jain P, He CH, Eun FC, Kang S, Tumbar T. Skin vasculature and hair follicle cross-talking associated with stem cell activation and tissue homeostasis. eLife 2019; 8:e45977. [PMID: 31343406 PMCID: PMC6684267 DOI: 10.7554/elife.45977] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 07/25/2019] [Indexed: 12/21/2022] Open
Abstract
Skin vasculature cross-talking with hair follicle stem cells (HFSCs) is poorly understood. Skin vasculature undergoes dramatic remodeling during adult mouse hair cycle. Specifically, a horizontal plexus under the secondary hair germ (HPuHG) transiently neighbors the HFSC activation zone during the quiescence phase (telogen). Increased density of HPuHG can be induced by reciprocal mutations in the epithelium (Runx1) and endothelium (Alk1) in adult mice, and is accompanied by prolonged HFSC quiescence and by delayed entry and progression into the hair growth phase (anagen). Suggestively, skin vasculature produces BMP4, a well-established HFSC quiescence-inducing factor, thus contributing to a proliferation-inhibitory environment near the HFSC. Conversely, the HFSC activator Runx1 regulates secreted proteins with previously demonstrated roles in vasculature remodeling. We suggest a working model in which coordinated remodeling and molecular cross-talking of the adult epithelial and endothelial skin compartments modulate timing of HFSC activation from quiescence for proper tissue homeostasis of adult skin.
Collapse
Affiliation(s)
- Kefei Nina Li
- Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| | - Prachi Jain
- Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| | - Catherine Hua He
- Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| | - Flora Chae Eun
- Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| | - Sangjo Kang
- Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| | - Tudorita Tumbar
- Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| |
Collapse
|
114
|
Akla N, Viallard C, Popovic N, Lora Gil C, Sapieha P, Larrivée B. BMP9 (Bone Morphogenetic Protein-9)/Alk1 (Activin-Like Kinase Receptor Type I) Signaling Prevents Hyperglycemia-Induced Vascular Permeability. Arterioscler Thromb Vasc Biol 2019; 38:1821-1836. [PMID: 29880487 DOI: 10.1161/atvbaha.118.310733] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Objective- Diabetic macular edema is a major cause of visual impairment. It is caused by blood-retinal barrier breakdown that leads to vascular hyperpermeability. Current therapeutic approaches consist of retinal photocoagulation or targeting VEGF (vascular endothelial growth factor) to limit vascular leakage. However, long-term intravitreal use of anti-VEGFs is associated with potential safety issues, and the identification of alternative regulators of vascular permeability may provide safer therapeutic options. The vascular specific BMP (bone morphogenetic protein) receptor ALK1 (activin-like kinase receptor type I) and its circulating ligand BMP9 have been shown to be potent vascular quiescence factors, but their role in the context of microvascular permeability associated with hyperglycemia has not been evaluated. Approach and Results- We investigated Alk1 signaling in hyperglycemic endothelial cells and assessed whether BMP9/Alk1 signaling could modulate vascular permeability. We show that high glucose concentrations impair Alk1 signaling, both in cultured endothelial cells and in a streptozotocin model of mouse diabetes mellitus. We observed that Alk1 signaling participates in the maintenance of vascular barrier function, as Alk1 haploinsufficiency worsens the vascular leakage observed in diabetic mice. Conversely, sustained delivery of BMP9 by adenoviral vectors significantly decreased the loss of retinal barrier function in diabetic mice. Mechanistically, we demonstrate that Alk1 signaling prevents VEGF-induced phosphorylation of VE-cadherin and induces the expression of occludin, thus strengthening vascular barrier functions. Conclusions- From these data, we suggest that by preventing retinal vascular permeability, BMP9 could serve as a novel therapeutic agent for diabetic macular edema.
Collapse
Affiliation(s)
- Naoufal Akla
- From the Department of Biochemistry (N.A., P.S.).,University of Montreal, Quebec, Canada; and Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada (N.A., C.V., N.P., C.L.G., P.S., B.L.)
| | - Claire Viallard
- Department of Molecular Biology (C.V., B.L.).,University of Montreal, Quebec, Canada; and Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada (N.A., C.V., N.P., C.L.G., P.S., B.L.)
| | - Natalija Popovic
- Department of Biomedical Sciences (N.P., C.L.G., B.L.).,University of Montreal, Quebec, Canada; and Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada (N.A., C.V., N.P., C.L.G., P.S., B.L.)
| | - Cindy Lora Gil
- Department of Biomedical Sciences (N.P., C.L.G., B.L.).,University of Montreal, Quebec, Canada; and Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada (N.A., C.V., N.P., C.L.G., P.S., B.L.)
| | - Przemyslaw Sapieha
- From the Department of Biochemistry (N.A., P.S.).,Department of Ophthalmology (P.S., B.L.).,University of Montreal, Quebec, Canada; and Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada (N.A., C.V., N.P., C.L.G., P.S., B.L.)
| | - Bruno Larrivée
- Department of Molecular Biology (C.V., B.L.).,Department of Biomedical Sciences (N.P., C.L.G., B.L.).,Department of Ophthalmology (P.S., B.L.).,University of Montreal, Quebec, Canada; and Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada (N.A., C.V., N.P., C.L.G., P.S., B.L.)
| |
Collapse
|
115
|
Li Q, Zhou X, Zhou X. Downregulation of miR‑98 contributes to hypoxic pulmonary hypertension by targeting ALK1. Mol Med Rep 2019; 20:2167-2176. [PMID: 31322216 PMCID: PMC6691262 DOI: 10.3892/mmr.2019.10482] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 05/31/2019] [Indexed: 12/21/2022] Open
Abstract
Chronic hypoxia is one of the most common causes of secondary pulmonary hypertension, the mechanisms of which remain unclear. MicroRNAs (miRNAs) are small, noncoding RNAs that inhibit the translation or accelerate the degradation of mRNA. Previous studies have demonstrated that deregulated miRNA expression contributes to various cellular processes including cell apoptosis and proliferation, which are mediated by hypoxia. In the present study, the expression of miR‑98 was identified to be decreased in the lung tissue of a hypoxic pulmonary hypertension (HPH) rat model and pulmonary artery (PA) smooth muscle cells (PASMCs), which was induced by hypoxia. By transfecting miR‑98 mimics into PASMCs, the high expression of miR‑98 inhibited cell proliferation, but upregulated hypoxia‑induced PASMCs apoptosis. However, these effects of miR‑98 mimics on PASMCs were reversed by ALK1 (activin receptor‑like kinase‑1) overexpression. ALK1 was identified as a candidate target of miR‑98. In addition, overexpressing miR‑98 markedly decreased the pulmonary artery wall thickness and the right ventricular systolic pressure in rats induced by hypoxia. These results provided clear evidence that miR‑98 was a direct regulator of ALK1, and that the downregulation of miR‑98 contributed to the pathogenesis of HPH. These results provide a novel potential therapeutic strategy for the treatment of HPH.
Collapse
Affiliation(s)
- Qingling Li
- Department of Respiratory Medicine, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xincan Zhou
- Department of Respiratory Medicine, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xianghui Zhou
- Department of Respiratory Medicine, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
116
|
Ye F, Xu H, Yin H, Zhao X, Li D, Zhu Q, Wang Y. The role of BMP6 in the proliferation and differentiation of chicken cartilage cells. PLoS One 2019; 14:e0204384. [PMID: 31260450 PMCID: PMC6602178 DOI: 10.1371/journal.pone.0204384] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 05/28/2019] [Indexed: 11/18/2022] Open
Abstract
Previous studies have indicated that bone morphogenetic protein (BMP) 6 may play an important role in skeletal system development and progression. However, the mechanism underlying the effects of BMP6 in cartilage cell proliferation and differentiation remains unknown. In this study, cartilage cells were isolated from shanks of chicken embryos and treated with different concentrations of Growth Hormone. Cell proliferation potential was assessed using real-time polymerase chain reaction (RT-PCR), western blotting and CCK-8 assays in vitro. The results showed that at 48 h, the Collagen II and BMP6 expression levels in 50 ng/μl GH-treated cartilage cells were significantly higher than in groups treated with 100 ng/μl or 200 ng/μl GH. We further observed that knockdown of BMP6 in cartilage cells led to significantly decreased expression mRNAs and proteins of Collagen II and Collagen X. Moreover, the suppression of BMP6 expression by a specific siRNA led to significantly decreased expression mRNA levels of IGF1R, JAK2, PKC, PTH, IHH and PTHrP and decreased protein levels of PKC, IHH and PTHrP. Taken together, our data suggest that BMP6 may play a critical role in chicken cartilage cell proliferation and differentiation through the regulation of IGF1, JAK2, PKC, PTH, and IHH-PTHrP signaling pathways.
Collapse
Affiliation(s)
- Fei Ye
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| | - Hengyong Xu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| | - Huadong Yin
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| | - Xiaoling Zhao
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| | - Diyan Li
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| | - Qing Zhu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| | - Yan Wang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
117
|
Oliveira SDS, Chen J, Castellon M, Mao M, Raj JU, Comhair S, Erzurum S, Silva CLM, Machado RF, Bonini MG, Minshall RD. Injury-Induced Shedding of Extracellular Vesicles Depletes Endothelial Cells of Cav-1 (Caveolin-1) and Enables TGF-β (Transforming Growth Factor-β)-Dependent Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol 2019; 39:1191-1202. [PMID: 30943774 PMCID: PMC7297129 DOI: 10.1161/atvbaha.118.312038] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Objective- To determine whether pulmonary arterial hypertension is associated with endothelial cell (EC)-Cav-1 (caveolin-1) depletion, EC-derived extracellular vesicle cross talk with macrophages, and proliferation of Cav-1 depleted ECs via TGF-β (transforming growth factor-β) signaling. Approach and Results- Pulmonary vascular disease was induced in Sprague-Dawley rats by exposure to a single injection of VEGFRII (vascular endothelial growth factor receptor II) antagonist SU5416 (Su) followed by hypoxia (Hx) plus normoxia (4 weeks each-HxSu model) and in WT (wild type; Tie2.Cre-; Cav1 lox/lox) and EC- Cav1-/- (Tie2.Cre+; Cav1 fl/fl) mice (Hx: 4 weeks). We observed reduced lung Cav-1 expression in the HxSu rat model in association with increased Cav-1+ extracellular vesicle shedding into the circulation. Whereas WT mice exposed to hypoxia exhibited increased right ventricular systolic pressure and pulmonary microvascular thickening compared with the group maintained in normoxia, the remodeling was further increased in EC- Cav1-/- mice indicating EC Cav-1 expression protects against hypoxia-induced pulmonary hypertension. Depletion of EC Cav-1 was associated with reduced BMPRII (bone morphogenetic protein receptor II) expression, increased macrophage-dependent TGF-β production, and activation of pSMAD2/3 signaling in the lung. In vitro, in the absence of Cav-1, eNOS (endothelial NO synthase) dysfunction was implicated in the mechanism of EC phenotype switching. Finally, reduced expression of EC Cav-1 in lung histological sections from human pulmonary arterial hypertension donors was associated with increased plasma concentration of Cav-1, extracellular vesicles, and TGF-β, indicating Cav-1 may be a plasma biomarker of vascular injury and key determinant of TGF-β-induced pulmonary vascular remodeling. Conclusions- EC Cav-1 depletion occurs, in part, via Cav-1+ extracellular vesicle shedding into the circulation, which contributes to increased TGF-β signaling, EC proliferation, vascular remodeling, and pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Suellen D S Oliveira
- From the Department of Anesthesiology (S.D.S.O., M.C., R.D.M.), University of Illinois at Chicago
| | - Jiwang Chen
- Department of Medicine (J.C., M.M., R.F.M., M.G.B.), University of Illinois at Chicago
- Research Resources Center Cardiovascular Research Core (J.C., M.C.), University of Illinois at Chicago
| | - Maricela Castellon
- From the Department of Anesthesiology (S.D.S.O., M.C., R.D.M.), University of Illinois at Chicago
- Research Resources Center Cardiovascular Research Core (J.C., M.C.), University of Illinois at Chicago
| | - Mao Mao
- Department of Medicine (J.C., M.M., R.F.M., M.G.B.), University of Illinois at Chicago
| | - J Usha Raj
- Department of Pediatrics (J.U.R.), University of Illinois at Chicago
| | - Suzy Comhair
- Lerner Research Institute (S.C., S.E.), Cleveland Clinic Foundation, OH
| | - Serpil Erzurum
- Lerner Research Institute (S.C., S.E.), Cleveland Clinic Foundation, OH
| | - Claudia L M Silva
- Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil (C.L.M.S.)
| | - Roberto F Machado
- Department of Medicine (J.C., M.M., R.F.M., M.G.B.), University of Illinois at Chicago
| | - Marcelo G Bonini
- Department of Medicine (J.C., M.M., R.F.M., M.G.B.), University of Illinois at Chicago
| | - Richard D Minshall
- From the Department of Anesthesiology (S.D.S.O., M.C., R.D.M.), University of Illinois at Chicago
- Department of Pharmacology (R.D.M.), University of Illinois at Chicago
| |
Collapse
|
118
|
Voss MH, Bhatt RS, Vogelzang NJ, Fishman M, Alter RS, Rini BI, Beck JT, Joshi M, Hauke R, Atkins MB, Burgess E, Logan TF, Shaffer D, Parikh R, Moazzam N, Zhang X, Glasser C, Sherman ML, Plimack ER. A phase 2, randomized trial evaluating the combination of dalantercept plus axitinib in patients with advanced clear cell renal cell carcinoma. Cancer 2019; 125:2400-2408. [PMID: 30951193 DOI: 10.1002/cncr.32061] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/28/2019] [Accepted: 02/05/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND In a prior open-label study, the combination of dalantercept, a novel antiangiogenic targeting activin receptor-like kinase 1 (ALK1), plus axitinib was deemed safe and tolerable with a promising efficacy signal in patients with advanced renal cell carcinoma (RCC). METHODS In the current phase 2, randomized, double-blind, placebo-controlled study, patients with clear cell RCC previously treated with 1 prior angiogenesis inhibitor were randomized 1:1 to receive axitinib plus dalantercept versus axitinib plus placebo. Randomization was stratified by the type of prior therapy. The primary endpoint was progression-free survival (PFS). Secondary endpoints were PFS in patients with ≥2 prior lines of anticancer therapy, overall survival, and the objective response rate. RESULTS Between June 10, 2014, and February 23, 2017, a total of 124 patients were randomly assigned to receive axitinib plus dalantercept (59 patients) or placebo (65 patients). The median PFS was not found to be significantly different between the treatment groups (median, 6.8 months vs 5.6 months; hazard ratio, 1.11 [95% CI, 0.71-1.73; P = .670]). Neither group reached the median overall survival (hazard ratio, 1.39 [95% CI, 0.70-2.77; P = .349]). The objective response rate was 19.0% (11 of 58 patients; 95% CI, 9.9%-31.4%) in the dalantercept plus axitinib group and 24.6% (15 of 61 patients; 95% CI, 14.5%-37.3%) in the placebo plus axitinib group. At least 1 treatment-emergent adverse event of ≥grade 3 was observed in 59% of patients (34 of 58 patients) in the dalantercept group and 64% of patients (39 of 61 patients) in the placebo group. One treatment-related death occurred in the placebo plus axitinib group. CONCLUSIONS Although well tolerated, the addition of dalantercept to axitinib did not appear to improve treatment-related outcomes in previously treated patients with advanced RCC.
Collapse
Affiliation(s)
- Martin H Voss
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rupal S Bhatt
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | - Robert S Alter
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, New Jersey
| | - Brian I Rini
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | | | - Monika Joshi
- Penn State Cancer Institute, Hershey, Pennsylvania
| | - Ralph Hauke
- Nebraska Methodist Hospital, Omaha, Nebraska
| | | | - Earle Burgess
- Levine Cancer Institute, Carolinas Medical Center, Charlotte, North Carolina
| | - Theodore F Logan
- Indiana University Health Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | | | - Rahul Parikh
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | | | | | | | | | | |
Collapse
|
119
|
Hong Q, Zhang L, Fu J, Verghese DA, Chauhan K, Nadkarni GN, Li Z, Ju W, Kretzler M, Cai GY, Chen XM, D'Agati VD, Coca SG, Schlondorff D, He JC, Lee K. LRG1 Promotes Diabetic Kidney Disease Progression by Enhancing TGF- β-Induced Angiogenesis. J Am Soc Nephrol 2019; 30:546-562. [PMID: 30858225 DOI: 10.1681/asn.2018060599] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 01/28/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Glomerular endothelial dysfunction and neoangiogenesis have long been implicated in the pathogenesis of diabetic kidney disease (DKD). However, the specific molecular pathways contributing to these processes in the early stages of DKD are not well understood. Our recent transcriptomic profiling of glomerular endothelial cells identified a number of proangiogenic genes that were upregulated in diabetic mice, including leucine-rich α-2-glycoprotein 1 (LRG1). LRG1 was previously shown to promote neovascularization in mouse models of ocular disease by potentiating endothelial TGF-β/activin receptor-like kinase 1 (ALK1) signaling. However, LRG1's role in the kidney, particularly in the setting of DKD, has been unclear. METHODS We analyzed expression of LRG1 mRNA in glomeruli of diabetic kidneys and assessed its localization by RNA in situ hybridization. We examined the effects of genetic ablation of Lrg1 on DKD progression in unilaterally nephrectomized, streptozotocin-induced diabetic mice at 12 and 20 weeks after diabetes induction. We also assessed whether plasma LRG1 was associated with renal outcome in patients with type 2 diabetes. RESULTS LRG1 localized predominantly to glomerular endothelial cells, and its expression was elevated in the diabetic kidneys. LRG1 ablation markedly attenuated diabetes-induced glomerular angiogenesis, podocyte loss, and the development of diabetic glomerulopathy. These improvements were associated with reduced ALK1-Smad1/5/8 activation in glomeruli of diabetic mice. Moreover, increased plasma LRG1 was associated with worse renal outcome in patients with type 2 diabetes. CONCLUSIONS These findings identify LRG1 as a potential novel pathogenic mediator of diabetic glomerular neoangiogenesis and a risk factor in DKD progression.
Collapse
Affiliation(s)
- Quan Hong
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing, China
| | - Lu Zhang
- Department of Nephrology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jia Fu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Divya A Verghese
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kinsuk Chauhan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Girish N Nadkarni
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zhengzhe Li
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Wenjun Ju
- Division of Nephrology, University of Michigan, Ann Arbor, Michigan
| | | | - Guang-Yan Cai
- Department of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing, China
| | - Xiang-Mei Chen
- Department of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing, China
| | - Vivette D D'Agati
- Department of Pathology, Columbia University Medical Center, New York, New York; and
| | - Steven G Coca
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Detlef Schlondorff
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - John C He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; .,Renal Section, James J. Peters Veterans Affair Medical Center, Bronx, New York
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York;
| |
Collapse
|
120
|
Abstract
Venous endothelial cells are molecularly and functionally distinct from their arterial counterparts. Although veins are often considered the default endothelial state, genetic manipulations can modulate both acquisition and loss of venous fate, suggesting that venous identity is the result of active transcriptional regulation. However, little is known about this process. Here we show that BMP signalling controls venous identity via the ALK3/BMPR1A receptor and SMAD1/SMAD5. Perturbations to TGF-β and BMP signalling in mice and zebrafish result in aberrant vein formation and loss of expression of the venous-specific gene Ephb4, with no effect on arterial identity. Analysis of a venous endothelium-specific enhancer for Ephb4 shows enriched binding of SMAD1/5 and a requirement for SMAD binding motifs. Further, our results demonstrate that BMP/SMAD-mediated Ephb4 expression requires the venous-enriched BMP type I receptor ALK3/BMPR1A. Together, our analysis demonstrates a requirement for BMP signalling in the establishment of Ephb4 expression and the venous vasculature. The establishment of functional vasculatures requires the specification of newly formed vessels into veins and arteries. Here, Neal et al. use a combination of genetic approaches in mice and zebrafish to show that BMP signalling, via ALK3 and SMAD1/5, is required for venous specification during blood vessel development.
Collapse
|
121
|
Budi EH, Mamai O, Hoffman S, Akhurst RJ, Derynck R. Enhanced TGF-β Signaling Contributes to the Insulin-Induced Angiogenic Responses of Endothelial Cells. iScience 2019; 11:474-491. [PMID: 30684493 PMCID: PMC6348203 DOI: 10.1016/j.isci.2018.12.038] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 11/12/2018] [Accepted: 12/28/2018] [Indexed: 12/17/2022] Open
Abstract
Angiogenesis, the development of new blood vessels, is a key process in disease. We reported that insulin promotes translocation of transforming growth factor β (TGF-β) receptors to the plasma membrane of epithelial and fibroblast cells, thus enhancing TGF-β responsiveness. Since insulin promotes angiogenesis, we addressed whether increased autocrine TGF-β signaling participates in endothelial cell responses to insulin. We show that insulin enhances TGF-β responsiveness and autocrine TGF-β signaling in primary human endothelial cells, by inducing a rapid increase in cell surface TGF-β receptor levels. Autocrine TGF-β/Smad signaling contributed substantially to insulin-induced gene expression associated with angiogenesis, including TGF-β target genes encoding angiogenic mediators; was essential for endothelial cell migration; and participated in endothelial cell invasion and network formation. Blocking TGF-β signaling impaired insulin-induced microvessel outgrowth from neonatal aortic rings and modified insulin-stimulated blood vessel formation in zebrafish. We conclude that enhanced autocrine TGF-β signaling is integral to endothelial cell and angiogenic responses to insulin.
Collapse
Affiliation(s)
- Erine H Budi
- Department of Cell and Tissue Biology, University of California at San Francisco Broad Center, Room RMB-1027, 35 Medical Center Way, San Francisco, CA 94143-0669, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Ons Mamai
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Steven Hoffman
- Department of Cell and Tissue Biology, University of California at San Francisco Broad Center, Room RMB-1027, 35 Medical Center Way, San Francisco, CA 94143-0669, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Rosemary J Akhurst
- Department of Anatomy, University of California at San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Rik Derynck
- Department of Cell and Tissue Biology, University of California at San Francisco Broad Center, Room RMB-1027, 35 Medical Center Way, San Francisco, CA 94143-0669, USA; Department of Anatomy, University of California at San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
122
|
Abstract
PURPOSE OF REVIEW Mutations in the Endoglin (Eng) gene, an auxiliary receptor in the transforming growth factor beta (TGFβ)-superfamily signaling pathway, are responsible for the human vascular disorder hereditary hemorrhagic telangiectasia (HHT) type 1, characterized in part by blood vessel enlargement. A growing body of work has uncovered an autonomous role for Eng in endothelial cells. We will highlight the influence of Eng on distinct cellular behaviors, such as migration and shape control, which are ultimately important for the assignment of proper blood vessel diameters. RECENT FINDINGS How endothelial cells establish hierarchically ordered blood vessel trees is one of the outstanding questions in vascular biology. Mutations in components of the TGFβ-superfamily of signaling molecules disrupt this patterning and cause arteriovenous malformations (AVMs). Eng is a TGFβ coreceptor enhancing signaling through the type I receptor Alk1. Recent studies identified bone morphogenetic proteins (BMPs) 9 and 10 as the primary ligands for Alk1/Eng. Importantly, Eng potentiated Alk1 pathway activation downstream of hemodynamic forces. New results furthermore revealed how Eng affects endothelial cell migration and cell shape control in response to these forces, thereby providing new avenues for our understanding of AVM cause. SUMMARY We will discuss the interplay of Eng and hemodynamic forces, such as shear stress, in relation to Alk1 receptor activation. We will furthermore detail how this signaling pathway influences endothelial cell behaviors important for the establishment of hierarchically ordered blood vessel trees. Finally, we will provide an outlook how these insights might help in developing new therapies for the treatment of HHT.
Collapse
|
123
|
Galaris G, Thalgott JH, Lebrin FPG. Pericytes in Hereditary Hemorrhagic Telangiectasia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1147:215-246. [PMID: 31147880 DOI: 10.1007/978-3-030-16908-4_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a genetic disorder characterized by multi-systemic vascular dysplasia affecting 1 in 5000 people worldwide. Individuals with HHT suffer from many complications including nose and gastrointestinal bleeding, anemia, iron deficiency, stroke, abscess, and high-output heart failure. Identification of the causative gene mutations and the generation of animal models have revealed that decreased transforming growth factor-β (TGF-β)/bone morphogenetic protein (BMP) signaling and increased vascular endothelial growth factor (VEGF) signaling activity in endothelial cells are responsible for the development of the vascular malformations in HHT. Perturbations in these key pathways are thought to lead to endothelial cell activation resulting in mural cell disengagement from the endothelium. This initial instability state causes the blood vessels to response inadequately when they are exposed to angiogenic triggers resulting in excessive blood vessel growth and the formation of vascular abnormalities that are prone to bleeding. Drugs promoting blood vessel stability have been reported as effective in preclinical models and in clinical trials indicating possible interventional targets based on a normalization approach for treating HHT. Here, we will review how disturbed TGF-β and VEGF signaling relates to blood vessel destabilization and HHT development and will discuss therapeutic opportunities based on the concept of vessel normalization to treat HHT.
Collapse
Affiliation(s)
- Georgios Galaris
- Department of Internal Medicine (Nephrology), Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jérémy H Thalgott
- Department of Internal Medicine (Nephrology), Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Franck P G Lebrin
- Department of Internal Medicine (Nephrology), Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.
- Physics for Medicine, ESPCI, INSERM U1273, CNRS, Paris, France.
- MEMOLIFE Laboratory of Excellence and PSL Research University, Paris, France.
| |
Collapse
|
124
|
Abdullahi W, Brzica H, Hirsch NA, Reilly BG, Ronaldson PT. Functional Expression of Organic Anion Transporting Polypeptide 1a4 Is Regulated by Transforming Growth Factor- β/Activin Receptor-like Kinase 1 Signaling at the Blood-Brain Barrier. Mol Pharmacol 2018; 94:1321-1333. [PMID: 30262595 PMCID: PMC6207918 DOI: 10.1124/mol.118.112912] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/26/2018] [Indexed: 12/12/2022] Open
Abstract
Central nervous system (CNS) drug delivery can be achieved by targeting drug uptake transporters such as Oatp1a4. In fact, many drugs that can improve neurologic outcomes in CNS diseases [3-hydroxy-3-methylglutaryl-CoA reductase inhibitors (i.e., statins)] are organic anion transporting polypeptide (OATP) transport substrates. To date, transport properties and regulatory mechanisms of Oatp1a4 at the blood-brain barrier (BBB) have not been rigorously studied. Such knowledge is critical to develop Oatp1a4 for optimization of CNS drug delivery and for improved treatment of neurological diseases. Our laboratory has demonstrated that the transforming growth factor-β (TGF-β)/activin receptor-like kinase 1 (ALK1) signaling agonist bone morphogenetic protein 9 (BMP-9) increases functional expression of Oatp1a4 in rat brain microvessels. Here, we expand on this work and show that BMP-9 treatment increases blood-to-brain transport and brain exposure of established OATP transport substrates (i.e., taurocholate, atorvastatin, and pravastatin). We also demonstrate that BMP-9 activates the TGF-β/ALK1 pathway in brain microvessels as indicated by increased nuclear translocation of specific Smad proteins associated with signaling mediated by the ALK1 receptor (i.e., pSmad1/5/8). Furthermore, we report that an activated Smad protein complex comprised of phosphorylated Smad1/5/8 and Smad4 is formed following BMP-9 treatment and binds to the promoter of the Slco1a4 gene (i.e., the gene that encodes Oatp1a4). This signaling mechanism causes increased expression of Slco1a4 mRNA. Overall, this study provides evidence that Oatp1a4 transport activity at the BBB is directly regulated by TGF-β/ALK1 signaling and indicates that this pathway can be targeted for control of CNS delivery of OATP substrate drugs.
Collapse
Affiliation(s)
- Wazir Abdullahi
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Hrvoje Brzica
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Nicholas A Hirsch
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Bianca G Reilly
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
125
|
Astrocytes and the TGF-β1 Pathway in the Healthy and Diseased Brain: a Double-Edged Sword. Mol Neurobiol 2018; 56:4653-4679. [DOI: 10.1007/s12035-018-1396-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/14/2018] [Indexed: 12/14/2022]
|
126
|
Sweeney M, Foldes G. It Takes Two: Endothelial-Perivascular Cell Cross-Talk in Vascular Development and Disease. Front Cardiovasc Med 2018; 5:154. [PMID: 30425990 PMCID: PMC6218412 DOI: 10.3389/fcvm.2018.00154] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/10/2018] [Indexed: 12/26/2022] Open
Abstract
The formation of new blood vessels is a crucial step in the development of any new tissue both during embryogenesis and in vitro models as without sufficient perfusion the tissue will be unable to grow beyond the size where nutrition and oxygenation can be managed by diffusion alone. Endothelial cells are the primary building block of blood vessels and are capable of forming tube like structures independently however they are unable to independently form functional vasculature which is capable of conducting blood flow. This requires support from other structures including supporting perivascular cells and the extracellular matrix. The crosstalk between endothelial cells and perivascular cells is vital in regulating vasculogenesis and angiogenesis and the consequences when this is disrupted can be seen in a variety of congenital and acquired disease states. This review details the mechanisms of vasculogenesis in vivo during embryogenesis and compares this to currently employed in vitro techniques. It also highlights clinical consequences of defects in the endothelial cell-pericyte cross-talk and highlights therapies which are being developed to target this pathway. Improving the understanding of the intricacies of endothelial-pericyte signaling will inform pathophysiology of multiple vascular diseases and allow the development of effective in vitro models to guide drug development and assist with approaches in tissue engineering to develop functional vasculature for regenerative medicine applications.
Collapse
Affiliation(s)
- Mark Sweeney
- Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Gabor Foldes
- Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| |
Collapse
|
127
|
The Controversial Role of TGF-β in Neovascular Age-Related Macular Degeneration Pathogenesis. Int J Mol Sci 2018; 19:ijms19113363. [PMID: 30373226 PMCID: PMC6275040 DOI: 10.3390/ijms19113363] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 12/18/2022] Open
Abstract
The multifunctional transforming growth factors-beta (TGF-βs) have been extensively studied regarding their role in the pathogenesis of neovascular age-related macular degeneration (nAMD), a major cause of severe visual loss in the elderly in developed countries. Despite this, their effect remains somewhat controversial. Indeed, both pro- and antiangiogenic activities have been suggested for TGF-β signaling in the development and progression of nAMD, and opposite therapies have been proposed targeting the inhibition or activation of the TGF-β pathway. The present article summarizes the current literature linking TGF-β and nAMD, and reviews experimental data supporting both pro- and antiangiogenic hypotheses, taking into account the limitations of the experimental approaches.
Collapse
|
128
|
Abou-Alfa GK, Miksad RA, Tejani MA, Williamson S, Gutierrez ME, Olowokure OO, Sharma MR, El Dika I, Sherman ML, Pandya SS. A Phase Ib, Open-Label Study of Dalantercept, an Activin Receptor-Like Kinase 1 Ligand Trap, plus Sorafenib in Advanced Hepatocellular Carcinoma. Oncologist 2018; 24:161-e70. [PMID: 30352941 PMCID: PMC6369956 DOI: 10.1634/theoncologist.2018-0654] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 09/27/2018] [Indexed: 11/25/2022] Open
Abstract
Lessons Learned. Patients with hepatocellular carcinoma (HCC) often have limited therapeutic responses to the vascular endothelial growth factor (VEGF) tyrosine kinase inhibitor sorafenib, which is standard of care in advanced HCC. Targeting the activin receptor‐like kinase 1 (ALK1) and VEGF pathways simultaneously by combining the ALK1 ligand trap dalantercept with sorafenib may result in more effective angiogenic blockade and delay tumor progression in patients with advanced HCC. Although the combination was generally well tolerated, there was no additive antitumor activity with the combination of dalantercept plus sorafenib in patients with advanced HCC. No complete or partial responses were observed, and overall survival ranged from 1.9 to 23.3 months. These results suggest that, in this patient population, further development of the possible limited benefits of combination therapy with dalantercept plus sorafenib is not warranted.
Background. Targeting the activin receptor‐like kinase 1 (ALK1) and vascular endothelial growth factor (VEGF) pathways may result in more effective angiogenic blockade in patients with hepatocellular carcinoma (HCC). Methods. In this phase Ib study, patients with advanced HCC were enrolled to dose‐escalation cohorts, starting at 0.6 mg/kg dalantercept subcutaneously every 3 weeks plus 400 mg sorafenib orally once daily, or to a dose expansion cohort. The primary objective was to determine the safety and tolerability and the dalantercept maximum tolerated dose (MTD) level. Secondary objectives were to assess the preliminary activity and the association of pharmacodynamic biomarkers with tumor response. Results. A total of 21 patients were enrolled in the study. Five patients received 0.6 mg/kg dalantercept in the first dose escalation cohort. Based on the initial safety results, the dose level was de‐escalated to 0.4 mg/kg in the second cohort (n = 6). The MTD was identified as 0.4 mg/kg and used for the dose expansion cohort (n = 10). At this dose level, the combination was generally well tolerated. Overall survival ranged from 1.9 to 23.3 months, and the best overall response was stable disease. Conclusion. The addition of dalantercept to sorafenib did not improve antitumor activity in patients with HCC. The dalantercept program in this population was discontinued.
Collapse
Affiliation(s)
- Ghassan K Abou-Alfa
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| | - Rebecca A Miksad
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | | | | | | | | | - Imane El Dika
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | |
Collapse
|
129
|
Rol N, Kurakula KB, Happé C, Bogaard HJ, Goumans MJ. TGF-β and BMPR2 Signaling in PAH: Two Black Sheep in One Family. Int J Mol Sci 2018; 19:ijms19092585. [PMID: 30200294 PMCID: PMC6164161 DOI: 10.3390/ijms19092585] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 08/21/2018] [Accepted: 08/23/2018] [Indexed: 12/14/2022] Open
Abstract
Knowledge pertaining to the involvement of transforming growth factor β (TGF-β) and bone morphogenetic protein (BMP) signaling in pulmonary arterial hypertension (PAH) is continuously increasing. There is a growing understanding of the function of individual components involved in the pathway, but a clear synthesis of how these interact in PAH is currently lacking. Most of the focus has been on signaling downstream of BMPR2, but it is imperative to include the role of TGF-β signaling in PAH. This review gives a state of the art overview of disturbed signaling through the receptors of the TGF-β family with respect to vascular remodeling and cardiac effects as observed in PAH. Recent (pre)-clinical studies in which these two pathways were targeted will be discussed with an extended view on cardiovascular research fields outside of PAH, indicating novel future perspectives.
Collapse
Affiliation(s)
- Nina Rol
- Department of Pulmonology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081HV Amsterdam, The Netherlands.
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081HV Amsterdam, The Netherlands.
| | - Konda Babu Kurakula
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333ZA Leiden, The Netherlands.
| | - Chris Happé
- Department of Pulmonology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081HV Amsterdam, The Netherlands.
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081HV Amsterdam, The Netherlands.
| | - Harm Jan Bogaard
- Department of Pulmonology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081HV Amsterdam, The Netherlands.
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333ZA Leiden, The Netherlands.
| |
Collapse
|
130
|
Ouarné M, Bouvard C, Boneva G, Mallet C, Ribeiro J, Desroches-Castan A, Soleilhac E, Tillet E, Peyruchaud O, Bailly S. BMP9, but not BMP10, acts as a quiescence factor on tumor growth, vessel normalization and metastasis in a mouse model of breast cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:209. [PMID: 30165893 PMCID: PMC6118004 DOI: 10.1186/s13046-018-0885-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Angiogenesis has become an attractive target for cancer therapy. However, despite the initial success of anti-VEGF (Vascular endothelial growth factor) therapies, the overall survival appears only modestly improved and resistance to therapy often develops. Other anti-angiogenic targets are thus urgently needed. The predominant expression of the type I BMP (bone morphogenetic protein) receptor ALK1 (activin receptor-like kinase 1) in endothelial cells makes it an attractive target, and phase I/II trials are currently being conducted. ALK1 binds with strong affinity to two ligands that belong to the TGF-ß family, BMP9 and BMP10. In the present work, we addressed their specific roles in tumor angiogenesis, cancer development and metastasis in a mammary cancer model. METHODS For this, we used knockout (KO) mice for BMP9 (constitutive Gdf2-deficient), for BMP10 (inducible Bmp10-deficient) and double KO mice (Gdf2 and Bmp10) in a syngeneic immunocompetent orthotopic mouse model of spontaneous metastatic breast cancer (E0771). RESULTS Our studies demonstrate a specific role for BMP9 in the E0771 mammary carcinoma model. Gdf2 deletion increased tumor growth while inhibiting vessel maturation and tumor perfusion. Gdf2 deletion also increased the number and the mean size of lung metastases. On the other hand, Bmp10 deletion did not significantly affect the E0771 mammary model and the double deletion (Gdf2 and Bmp10) did not lead to a stronger phenotype than the single Gdf2 deletion. CONCLUSIONS Altogether, our data show that in a tumor environment BMP9 and BMP10 play different roles and thus blocking their shared receptor ALK1 is maybe not appropriate. Indeed, BMP9, but not BMP10, acts as a quiescence factor on tumor growth, lung metastasis and vessel normalization. Our results also support that activating rather than blocking the BMP9 pathway could be a new strategy for tumor vessel normalization in order to treat breast cancer.
Collapse
Affiliation(s)
- Marie Ouarné
- Univ. Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000, Grenoble, France
| | - Claire Bouvard
- Univ. Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000, Grenoble, France
| | - Gabriela Boneva
- Univ. Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000, Grenoble, France
| | - Christine Mallet
- Univ. Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000, Grenoble, France
| | - Johnny Ribeiro
- Inserm, U1033, Lyon, France.,Université Claude Bernard Lyon 1, Villeurbanne, France.,, Faculté de Médecine de Lyon Est, Lyon, France
| | - Agnès Desroches-Castan
- Univ. Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000, Grenoble, France
| | - Emmanuelle Soleilhac
- Univ. Grenoble Alpes, Inserm, CEA, BIG-Biologie à Grande Echelle, 38000, Grenoble, France
| | - Emmanuelle Tillet
- Univ. Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000, Grenoble, France
| | - Olivier Peyruchaud
- Inserm, U1033, Lyon, France.,Université Claude Bernard Lyon 1, Villeurbanne, France.,, Faculté de Médecine de Lyon Est, Lyon, France
| | - Sabine Bailly
- Univ. Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000, Grenoble, France.
| |
Collapse
|
131
|
Bocci M, Sjölund J, Kurzejamska E, Lindgren D, Marzouka NAD, Bartoschek M, Höglund M, Pietras K. Activin receptor-like kinase 1 is associated with immune cell infiltration and regulates CLEC14A transcription in cancer. Angiogenesis 2018; 22:117-131. [PMID: 30132150 PMCID: PMC6510886 DOI: 10.1007/s10456-018-9642-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 08/13/2018] [Indexed: 12/11/2022]
Abstract
Cancer cells sustain their metabolic needs through nutrients and oxygen supplied by the bloodstream. The requirement for tumor angiogenesis has been therapeutically exploited in the clinical setting mainly by means of inhibition of the vascular endothelial growth factor family of ligands and receptors. Despite promising results in preclinical models, the benefits for patients proved to be limited. Inadequate efficacy similarly halted the development of agents impinging on the activity of the activin receptor-like kinase (ALK)1, a member of the transforming growth factor-β superfamily. Notwithstanding its characterization as an endothelial cell marker, the full spectrum of biological processes associated with ALK1 is essentially unexplored. Here, we present data revealing the genetic network associated with ACVRL1 (the gene encoding for ALK1) expression in human cancer tissues. Computational analysis unveiled a hitherto unknown role for ACVRL1 in relation to genes modulating the functionality of the immune cell compartment. Moreover, we generated a signature of 8 genes co-expressed with ACVRL1 across different tumor types and characterized the c-type lectin domain containing protein (CLEC)14A as a potential downstream target of ACVRL1. Considering the lack of reagents for ALK1 detection that has hampered the field to date, our work provides the opportunity to validate the 8-gene signature and CLEC14A as biomarkers for ALK1 activity. Ultimately, this may help revisit the clinical development of already existing ALK1-blocking compounds as precision medicines for cancer.
Collapse
Affiliation(s)
- Matteo Bocci
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Building 404:A3, 223 81, Lund, Sweden
| | - Jonas Sjölund
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Building 404:A3, 223 81, Lund, Sweden
| | - Ewa Kurzejamska
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Building 404:A3, 223 81, Lund, Sweden
| | - David Lindgren
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Building 404:A3, 223 81, Lund, Sweden
| | - Nour-Al-Dain Marzouka
- Unit of Urothelial Cancer Genomics, Department of Oncology and Pathology, Lund University, Scheelevägen 8, 22363, Lund, Sweden
| | - Michael Bartoschek
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Building 404:A3, 223 81, Lund, Sweden
| | - Mattias Höglund
- Unit of Urothelial Cancer Genomics, Department of Oncology and Pathology, Lund University, Scheelevägen 8, 22363, Lund, Sweden
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Building 404:A3, 223 81, Lund, Sweden.
| |
Collapse
|
132
|
Abstract
The cytokine transforming growth factor (TGF)-β1 is highly induced after encephalopathic brain injury, with data showing that it can both contribute to the pathophysiology and aid in disease resolution. In the immature brain, sustained TGFβ-signaling after injury may prolong inflammation to both exacerbate acute stage damage and perturb the normal course of development. Yet in adult encephalopathy, elevated TGFβ1 may promote a reparative state. In this review, we highlight the context-dependent actions of TGFβ-signaling in the brain during resolution of encephalopathy and focus on neuronal survival mechanisms that are affected by TGFβ1. We discuss the mechanisms that contribute to the disparate actions of TGFβ1 toward elucidating the long-term neurological and neuropsychiatric consequences that follow encephalopathic injury.
Collapse
Affiliation(s)
- Brian H Kim
- 1 Department of Pharmacology, Physiology and Neuroscience, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Steven W Levison
- 1 Department of Pharmacology, Physiology and Neuroscience, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
133
|
Chen GZ, Ke Y, Qin K, Dong MQ, Zeng SJ, Lin XF, Zhan SQ, Tang K, Peng C, Ding XW, Zhou D. Analysis of the Expression of Angioarchitecture-related Factors in Patients with Cerebral Arteriovenous Malformation. Chin Med J (Engl) 2018; 130:2465-2472. [PMID: 29052569 PMCID: PMC5684641 DOI: 10.4103/0366-6999.216413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Cerebral arteriovenous malformation (cAVM) is a type of vascular malformation associated with vascular remodeling, hemodynamic imbalance, and inflammation. We detected four angioarchitecture-related cytokines to make a better understanding of the potential aberrant signaling in the pathogenesis of cAVM and found useful proteins in predicting the risk of cerebral hemorrhage. Methods: Immunohistochemical analysis was conducted on specimens from twenty patients with cAVM diagnosed via magnetic resonance imaging and digital subtraction angiography and twenty primary epilepsy controls using antibodies against vascular endothelial growth factor receptor-2 (VEGFR-2), matrix metalloproteinase-9 (MMP-9), vascular cell adhesion molecule (VCAM-1), and endothelial nitric oxide synthase (eNOS). Western blotting and real-time fluorescent quantitative polymerase chain reaction (PCR) were performed to determine protein and mRNA expression levels. Student's t-test was used for statistical analysis. Results: VEGFR-2, MMP-9, VCAM-1, and eNOS expression levels increased in patients with cAVM compared with those in normal cerebral vascular tissue, as determined by immunohistochemical analysis. In addition, Western blotting and real-time PCR showed that the protein and mRNA expression levels of VEGFR-2, MMP-9, VCAM-1, and eNOS were higher in the cAVM group than in the control group, all the differences mentioned were statistically significant (P < 0.05). Conclusions: VEGFR-2, MMP-9, VCAM-1, and eNOS are upregulated in patients with cAVM and might play important roles in angiogenesis, vascular remodeling, and migration in patients with cAVM. MMP-9, VEGFR-2, VCAM-1, and eNOS might be potential excellent group proteins in predicting the risk of cerebral hemorrhage at arteriovenous malformation.
Collapse
Affiliation(s)
- Guang-Zhong Chen
- Department of Neurosurgery, Guangdong General Hospital, Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Yu Ke
- Department of Biomedical Engineering, Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong 510632, China
| | - Kun Qin
- Department of Neurosurgery, Guangdong General Hospital, Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Meng-Qi Dong
- Department of Neurosurgery, Guangdong General Hospital, Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Shao-Jian Zeng
- Department of Neurosurgery, Guangdong General Hospital, Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Xiao-Feng Lin
- Department of Neurosurgery, Guangdong General Hospital, Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Sheng-Quan Zhan
- Department of Neurosurgery, Guangdong General Hospital, Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Kai Tang
- Department of Neurosurgery, Guangdong General Hospital, Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Chao Peng
- Department of Neurosurgery, Guangdong General Hospital, Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Xiao-Wen Ding
- Department of Neurosurgery, Guangdong General Hospital, Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Dong Zhou
- Department of Neurosurgery, Guangdong General Hospital, Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| |
Collapse
|
134
|
Schütz E, Bochenek ML, Riehl DR, Bosmann M, Münzel T, Konstantinides S, Schäfer K. Absence of transforming growth factor beta 1 in murine platelets reduces neointima formation without affecting arterial thrombosis. Thromb Haemost 2018; 117:1782-1797. [PMID: 28726976 DOI: 10.1160/th17-02-0112] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 06/11/2017] [Indexed: 12/15/2022]
Abstract
Platelet degranulation at the site of vascular injury prevents bleeding and may affect the chronic vascular wound healing response. Transforming Growth Factor (TGF)-β1 is a major component of platelet α-granules known to accumulating in thrombi. It was our aim to determine the role of TGFβ1 released from activated platelets for neointima formation following arterial injury and thrombosis. Mice with platelet-specific deletion of TGFβ1 (Plt.TGFβ-KO) underwent carotid artery injury. Immunoassays confirmed the absence of active TGFβ1 in platelet releasates and plasma of Plt.TGFβ-KO mice. Whole blood analyses revealed similar haematological parameters, and tail cut assays excluded major bleeding defects. Platelet aggregation and the acute thrombotic response to injury in vivo did not differ between Plt.TGFβ-KO and Plt.TGFβ-WT mice. Morphometric analysis revealed that absence of TGFβ1 in platelets resulted in a significant reduction of neointima formation with lower neointima area, intima-to-media ratio, and lumen stenosis. On the other hand, the media area was enlarged in mice lacking TGFβ1 in platelets and contained increased amounts of proteases involved in latent TGFβ activation, including MMP2, MMP9 and thrombin. Significantly increased numbers of proliferating cells and cells expressing the mesenchymal markers platelet-derived growth factor receptor-β or fibroblast-specific protein-1, and the macrophage antigen F4/80, were observed in the media of Plt.TGFβ-KO mice, whereas the medial smooth muscle-actin-immunopositive area and collagen content did not differ between genotypes. Our findings support an essential role for platelet-derived TGFβ1 for the vascular remodelling response to arterial injury, apparently independent from the role of platelets in thrombosis or haemostasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Katrin Schäfer
- Katrin Schäfer, MD, FESC, FAHA, Center for Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany, Tel.: +49 6131 17 4221, Fax: +49 6131 17 8047, E-mail:
| |
Collapse
|
135
|
Kritharis A, Al-Samkari H, Kuter DJ. Hereditary hemorrhagic telangiectasia: diagnosis and management from the hematologist's perspective. Haematologica 2018; 103:1433-1443. [PMID: 29794143 PMCID: PMC6119150 DOI: 10.3324/haematol.2018.193003] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 05/14/2018] [Indexed: 12/11/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT), also known as Osler-Weber-Rendu syndrome, is an autosomal dominant disorder that causes abnormal blood vessel formation. The diagnosis of hereditary hemorrhagic telangiectasia is clinical, based on the Curaçao criteria. Genetic mutations that have been identified include ENG, ACVRL1/ALK1, and MADH4/SMAD4, among others. Patients with HHT may have telangiectasias and arteriovenous malformations in various organs and suffer from many complications including bleeding, anemia, iron deficiency, and high-output heart failure. Families with the same mutation exhibit considerable phenotypic variation. Optimal treatment is best delivered via a multidisciplinary approach with appropriate diagnosis, screening and local and/or systemic management of lesions. Anti-angiogenic agents such as bevacizumab have emerged as a promising systemic therapy in reducing bleeding complications but are not curative. Other pharmacological agents include iron supplementation, antifibrinolytics and hormonal treatment. This review discusses the biology of HHT, management issues that face the practising hematologist, and considerations of future directions in HHT treatment.
Collapse
Affiliation(s)
- Athena Kritharis
- Division of Blood Disorders, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Hanny Al-Samkari
- Hematology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David J Kuter
- Hematology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
136
|
Tillet E, Ouarné M, Desroches-Castan A, Mallet C, Subileau M, Didier R, Lioutsko A, Belthier G, Feige JJ, Bailly S. A heterodimer formed by bone morphogenetic protein 9 (BMP9) and BMP10 provides most BMP biological activity in plasma. J Biol Chem 2018; 293:10963-10974. [PMID: 29789425 DOI: 10.1074/jbc.ra118.002968] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 05/04/2018] [Indexed: 11/06/2022] Open
Abstract
Bone morphogenetic protein 9 (BMP9) and BMP10 are the two high-affinity ligands for the endothelial receptor activin receptor-like kinase 1 (ALK1) and are key regulators of vascular remodeling. They are both present in the blood, but their respective biological activities are still a matter of debate. The aim of the present work was to characterize their circulating forms to better understand how their activities are regulated in vivo First, by cotransfecting BMP9 and BMP10, we found that both can form a disulfide-bonded heterodimer in vitro and that this heterodimer is functional on endothelial cells via ALK1. Next, we developed an ELISA that could specifically recognize the BMP9-BMP10 heterodimer and which indicated its presence in both human and mouse plasma. In addition to using available Bmp9-KO mice, we generated a conditional Bmp10-KO mouse strain. The plasma from Bmp10-KO mice, similarly to that of Bmp9-KO mice, completely lacked the ability to activate ALK1-transfected 3T3 cells or phospho-Smad1-5 on endothelial cells, indicating that the circulating BMP activity is mostly due to the BMP9-BMP10 heterodimeric form. This result was confirmed in human plasma that had undergone affinity chromatography to remove BMP9 homodimer. Finally, we provide evidence that hepatic stellate cells in the liver could be the source of the BMP9-BMP10 heterodimer. Together, our findings demonstrate that BMP9 and BMP10 can heterodimerize and that this heterodimer is responsible for most of the biological BMP activity found in plasma.
Collapse
Affiliation(s)
- Emmanuelle Tillet
- From the University of Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000 Grenoble, France
| | - Marie Ouarné
- From the University of Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000 Grenoble, France
| | - Agnès Desroches-Castan
- From the University of Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000 Grenoble, France
| | - Christine Mallet
- From the University of Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000 Grenoble, France
| | - Mariela Subileau
- From the University of Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000 Grenoble, France
| | - Robin Didier
- From the University of Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000 Grenoble, France
| | - Anna Lioutsko
- From the University of Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000 Grenoble, France
| | - Guillaume Belthier
- From the University of Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000 Grenoble, France
| | - Jean-Jacques Feige
- From the University of Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000 Grenoble, France
| | - Sabine Bailly
- From the University of Grenoble Alpes, Inserm, CEA, BIG-Biologie du Cancer et de l'Infection, 38000 Grenoble, France
| |
Collapse
|
137
|
Smyth LCD, Rustenhoven J, Park TIH, Schweder P, Jansson D, Heppner PA, O'Carroll SJ, Mee EW, Faull RLM, Curtis M, Dragunow M. Unique and shared inflammatory profiles of human brain endothelia and pericytes. J Neuroinflammation 2018; 15:138. [PMID: 29751771 PMCID: PMC5948925 DOI: 10.1186/s12974-018-1167-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/18/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Pericytes and endothelial cells are critical cellular components of the blood-brain barrier (BBB) and play an important role in neuroinflammation. To date, the majority of inflammation-related studies in endothelia and pericytes have been carried out using immortalised cell lines or non-human-derived cells. Whether these are representative of primary human cells is unclear and systematic comparisons of the inflammatory responses of primary human brain-derived pericytes and endothelia has yet to be performed. METHODS To study the effects of neuroinflammation at the BBB, primary brain endothelial cells and pericytes were isolated from human biopsy tissue. Culture purity was examined using qPCR and immunocytochemistry. Electrical cell-substrate impedance sensing (ECIS) was used to determine the barrier properties of endothelial and pericyte cultures. Using immunocytochemistry, cytometric bead array, and ECIS, we compared the responses of endothelia and pericytes to a panel of inflammatory stimuli (IL-1β, TNFα, LPS, IFN-γ, TGF-β1, IL-6, and IL-4). Secretome analysis was performed to identify unique secretions of endothelia and pericytes in response to IL-1β. RESULTS Endothelial cells were pure, moderately proliferative, retained the expression of BBB-related junctional proteins and transporters, and generated robust TEER. Both endothelia and pericytes have the same pattern of transcription factor activation in response to inflammatory stimuli but respond differently at the secretion level. Secretome analysis confirmed that endothelia and pericytes have overlapping but distinct secretome profiles in response to IL-1β. We identified several cell-type specific responses, including G-CSF and GM-CSF (endothelial-specific), and IGFBP2 and IGFBP3 (pericyte-specific). Finally, we demonstrated that direct addition of IL-1β, TNFα, LPS, and IL-4 contributed to the loss of endothelial barrier integrity in vitro. CONCLUSIONS Here, we identify important cell-type differences in the inflammatory response of brain pericytes and endothelia and provide, for the first time, a comprehensive profile of the secretions of primary human brain endothelia and pericytes which has implications for understanding how inflammation affects the cerebrovasculature.
Collapse
Affiliation(s)
- Leon C D Smyth
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Justin Rustenhoven
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Thomas I-H Park
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Patrick Schweder
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Auckland City Hospital, Auckland, 1023, New Zealand
| | - Deidre Jansson
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Peter A Heppner
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Auckland City Hospital, Auckland, 1023, New Zealand
| | - Simon J O'Carroll
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Edward W Mee
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Auckland City Hospital, Auckland, 1023, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Maurice Curtis
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand. .,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|
138
|
Huang L, Deng J, Xu W, Wang H, Shi L, Wu F, Wu D, Nei W, Zhao M, Mao P, Zhou X. CD8+ T cells with high TGF‑β1 expression cause lymph node fibrosis following HIV infection. Mol Med Rep 2018; 18:77-86. [PMID: 29749506 PMCID: PMC6059705 DOI: 10.3892/mmr.2018.8964] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 03/20/2018] [Indexed: 12/11/2022] Open
Abstract
Lymph node (LN) fibrosis resulting in cluster of differentiation (CD) 4+ T cell reduction following human immunodeficiency virus (HIV) infection is an important step in the pathogenesis of acquired immunodeficiency syndrome. The mechanisms mediating LN fibrosis following HIV infection have not been completely elucidated. In order to investigate the mechanism of LN fibrosis, the expression of transforming growth factor (TGF)‑β1 was determined in the LNs of HIV‑infected individuals by immunohistochemistry and fluorescence‑based flow cytometry. The effect of stimulated CD8+ T cells on collagen secretion by fibroblasts was detected using immunofluorescence staining and western blot analysis. The results demonstrated that the LNs of HIV‑infected individuals exhibited a significantly increased proportion of CD8+ T cells with high TGF‑β1 expression. These CD8+ T cells demonstrated increased CD38 and programmed cell death protein 1 expression and decreased CD127 expression compared with the controls. CD8+ T cells from the LNs of non‑HIV infected individuals expressed a high TGF‑β1 level following stimulation with phorbol‑12‑myristate 13‑acetate. These CD8+T cells subsequently induced the secretion of a large amount of type I collagen in human lymphatic fibroblasts. The results of the present study indicated that CD8+ T cells with high TGF‑β1 expression served an important role in LN fibrosis following HIV infection.
Collapse
Affiliation(s)
- Lei Huang
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing 100039, P.R. China
| | - Jianning Deng
- Guangxi AIDS Clinical Treatment Center, The Fourth People's Hospital of Nanning, Nanning, Guangxi 530023, P.R. China
| | - Wen Xu
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing 100039, P.R. China
| | - Hongbo Wang
- The Second Center of Hepatobiliary Surgery Department, 302 Military Hospital of China, Beijing 100039, P.R. China
| | - Lei Shi
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing 100039, P.R. China
| | - Fengyao Wu
- Guangxi AIDS Clinical Treatment Center, The Fourth People's Hospital of Nanning, Nanning, Guangxi 530023, P.R. China
| | - Dan Wu
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing 100039, P.R. China
| | - Weimin Nei
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing 100039, P.R. China
| | - Min Zhao
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing 100039, P.R. China
| | - Panyong Mao
- Research Clinical Center for Translational Medicine, 302 Military Hospital of China, Beijing 100039, P.R. China
| | - Xianzhi Zhou
- The Fourth Military Medical University, Xian, Shaanxi 710032, P.R. China
| |
Collapse
|
139
|
Verrecchia F, Rédini F. Transforming Growth Factor-β Signaling Plays a Pivotal Role in the Interplay Between Osteosarcoma Cells and Their Microenvironment. Front Oncol 2018; 8:133. [PMID: 29761075 PMCID: PMC5937053 DOI: 10.3389/fonc.2018.00133] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/13/2018] [Indexed: 12/12/2022] Open
Abstract
Osteosarcomas are the most frequent form of primary bone tumors and mainly affect children, adolescents, and young adults. Despite encouraging progress in therapeutic management, including the advent of multidrug chemotherapy, the survival rates have remained unchanged for more than four decades: 75% at 5 years for localized disease, but two groups of patients are still at high risk: metastatic at diagnosis (overall survival around 40% at 5 years) and/or poor responders to chemotherapy (20% at 5 years). Because these tumors are classified as “complex genomic,” it is extremely difficult to determine the signaling pathways that might be targeted by specific therapies. A hypothesis has thus emerged, stating that the particular microenvironment of these tumors may interfere with the tumor cells that promote chemoresistance and the dissemination of metastases. The stroma is composed of a large number of cell types (immune cells, endothelial cells, mesenchymal stromal cells, etc.) which secrete growth factors, such as transforming growth factor-β (TGF-β), which favors the development of primary tumors and dissemination of metastases by constituting a permissive niche at primary and distant sites. Rather than targeting the tumor cells themselves, which are very heterogeneous in osteosarcoma, the hypothesis is instead to target the key actors secreted in the microenvironment, such as TGF-βs, which play a part in tumor progression. In the last decade, numerous studies have shown that overexpression of TGF-β is a hallmark of many cancers, including primary bone tumors. In this context, TGF-β signaling has emerged as a crucial factor in the cross talk between tumor cells and stroma cells in poor-prognosis cancers. Secretion of TGF-β by tumor cells or stroma cells can effectively act in a paracrine manner to regulate the phenotype and functions of the microenvironment to stimulate protumorigenic microenvironmental changes. TGF-β can thus exert its protumorigenic function in primary bone tumors by promoting angiogenesis, bone remodeling and cell migration, and by inhibiting immunosurveillance. This review focuses on the involvement of TGF-β signaling in primary bone tumor development, and the related therapeutic options that may be possible for these tumors.
Collapse
Affiliation(s)
- Franck Verrecchia
- UMR1238 INSERM, Université de Nantes, PHY-OS, "Bone Sarcomas and Remodeling of Calcified Tissues", Medical School, Nantes, France
| | - Françoise Rédini
- UMR1238 INSERM, Université de Nantes, PHY-OS, "Bone Sarcomas and Remodeling of Calcified Tissues", Medical School, Nantes, France
| |
Collapse
|
140
|
Yuan SM. Pulmonary artery hypertension in childhood: The transforming growth factor-β superfamily-related genes. Pediatr Neonatol 2018; 59:112-119. [PMID: 28967497 DOI: 10.1016/j.pedneo.2016.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 10/20/2016] [Accepted: 12/05/2016] [Indexed: 01/08/2023] Open
Abstract
Pulmonary artery hypertension (PAH) is very rare in childhood, and it can be divided into heritable, idiopathic drug- and toxin-induced and other disease (connective tissue disease, human immunodeficiency virus infection, portal hypertension, congenital heart disease, or schistosomiasis)-associated types. PAH could not be interpreted solely by pathophysiological theories. The impact of the transforming growth factor-β superfamily-related genes on the development of PAH in children remains to be clarified. Pertinent literature on the transforming growth factor-β superfamily-related genes in relation to PAH in children published after the year 2000 was reviewed and analyzed. Bone morphogenetic protein receptor type II gene mutation promotes cell division or prevents cell death, resulting in an overgrowth of cells in small arteries throughout the lungs. About 20% of individuals with a bone morphogenetic protein receptor type II gene mutation develop symptomatic PAH. In heritable PAH, bone morphogenetic protein receptor type II mutations may be absent; while mutations of other genes, such as type I receptor activin receptor-like kinase 1 and the type III receptor endoglin (both associated with hereditary hemorrhagic telangiectasia), caveolin-1 and KCNK3, the gene encoding potassium channel subfamily K, member 3, can be detected, instead. Gene mutations, environmental changes and acquired adjustment, etc. may explain the development of PAH. The researches on PAH rat model and familial PAH members may facilitate the elucidations of the mechanisms and further provide theories for prophylaxis and treatment of PAH.
Collapse
Affiliation(s)
- Shi-Min Yuan
- Department of Cardiothoracic Surgery, The First Hospital of Putian, Teaching Hospital, Fujian Medical University, Putian, Fujian Province, People's Republic of China.
| |
Collapse
|
141
|
Ying Y, Ueta T, Jiang S, Lin H, Wang Y, Vavvas D, Wen R, Chen YG, Luo Z. Metformin inhibits ALK1-mediated angiogenesis via activation of AMPK. Oncotarget 2018; 8:32794-32806. [PMID: 28427181 PMCID: PMC5464828 DOI: 10.18632/oncotarget.15825] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/12/2017] [Indexed: 11/25/2022] Open
Abstract
Anti-VEGF therapy has been proven to be effective in the treatment of pathological angiogenesis. However, therapy resistance often occurs, leading to development of alternative approaches. The present study examines if AMPK negatively regulates ALK1-mediated signaling events and associated angiogenesis. Thus, we treated human umbilical vein endothelial cells with metformin as well as other pharmacological AMPK activators and showed that activation of AMPK inhibited Smad1/5 phosphorylation and tube formation induced by BMP9. This event was mimicked by expression of the active mutant of AMPKα1 and prevented by the dominant negative AMPKα1. Metformin inhibition of BMP9 signaling is possibly mediated by upregulation of Smurf1, leading to degradation of ALK1. Furthermore, metformin suppressed BMP9-induced angiogenesis in mouse matrigel plug. In addition, laser photocoagulation was employed to evaluate the effect of metformin. The data revealed that metformin significantly reduced choroidal neovascularization to a level comparable to LDN212854, an ALK1 specific inhibitor. In conjunction, metformin diminished expression of ALK1 in endothelium of the lesion area. Collectively, our study for the first time demonstrates that AMPK inhibits ALK1 and associated angiogenesis/neovascularization. This may offer us a new avenue for the treatment of related diseases using clinically used pharmacological AMPK activators like metformin in combination with other strategies to enhance the treatment efficacy or in the case of anti-VEGF resistance.
Collapse
Affiliation(s)
- Ying Ying
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Takashi Ueta
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Shanshan Jiang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Hui Lin
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Yuanyuan Wang
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Demetrios Vavvas
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Rong Wen
- Bascom Palmer Eye Institute, University of Miami Miller Medical School, Miami, FL, USA
| | - Ye-Guang Chen
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing, China
| | - Zhijun Luo
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.,Windsor University School of Medicine, Brighton's Estate, Cayon, St. Kitts
| |
Collapse
|
142
|
Harrell CR, Simovic Markovic B, Fellabaum C, Arsenijevic A, Djonov V, Volarevic V. Molecular mechanisms underlying therapeutic potential of pericytes. J Biomed Sci 2018; 25:21. [PMID: 29519245 PMCID: PMC5844098 DOI: 10.1186/s12929-018-0423-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/21/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Pericytes are multipotent cells present in every vascularized tissue in the body. Despite the fact that they are well-known for more than a century, pericytes are still representing cells with intriguing properties. This is mainly because of their heterogeneity in terms of definition, tissue distribution, origin, phenotype and multi-functional properties. The body of knowledge illustrates importance of pericytes in the regulation of homeostatic and healing processes in the body. MAIN BODY In this review, we summarized current knowledge regarding identification, isolation, ontogeny and functional characteristics of pericytes and described molecular mechanisms involved in the crosstalk between pericytes and endothelial or immune cells. We highlighted the role of pericytes in the pathogenesis of fibrosis, diabetes-related complications (retinopathy, nephropathy, neuropathy and erectile dysfunction), ischemic organ failure, pulmonary hypertension, Alzheimer disease, tumor growth and metastasis with the focus on their therapeutic potential in the regenerative medicine. The functions and capabilities of pericytes are impressive and, as yet, incompletely understood. Molecular mechanisms responsible for pericyte-mediated regulation of vascular stability, angiogenesis and blood flow are well described while their regenerative and immunomodulatory characteristics are still not completely revealed. Strong evidence for pericytes' participation in physiological, as well as in pathological conditions reveals a broad potential for their therapeutic use. Recently published results obtained in animal studies showed that transplantation of pericytes could positively influence the healing of bone, muscle and skin and could support revascularization. However, the differences in their phenotype and function as well as the lack of standardized procedure for their isolation and characterization limit their use in clinical trials. CONCLUSION Critical to further progress in clinical application of pericytes will be identification of tissue specific pericyte phenotype and function, validation and standardization of the procedure for their isolation that will enable establishment of precise clinical settings in which pericyte-based therapy will be efficiently applied.
Collapse
Affiliation(s)
- C. Randall Harrell
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N Palm Harbor, Palm Harbor, Florida USA
| | - Bojana Simovic Markovic
- Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Serbia, Faculty of Medical Sciences, 69 Svetozar Markovic Street, Kragujevac, 34000 Serbia
| | - Crissy Fellabaum
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N Palm Harbor, Palm Harbor, Florida USA
| | - Aleksandar Arsenijevic
- Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Serbia, Faculty of Medical Sciences, 69 Svetozar Markovic Street, Kragujevac, 34000 Serbia
| | - Valentin Djonov
- University of Bern, Institute of Anatomy, Baltzerstrasse 2, Bern, Switzerland
| | - Vladislav Volarevic
- Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Serbia, Faculty of Medical Sciences, 69 Svetozar Markovic Street, Kragujevac, 34000 Serbia
| |
Collapse
|
143
|
Eleftheriou NM, Sjölund J, Bocci M, Cortez E, Lee SJ, Cunha SI, Pietras K. Compound genetically engineered mouse models of cancer reveal dual targeting of ALK1 and endoglin as a synergistic opportunity to impinge on angiogenic TGF-β signaling. Oncotarget 2018; 7:84314-84325. [PMID: 27741515 PMCID: PMC5341292 DOI: 10.18632/oncotarget.12604] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 10/03/2016] [Indexed: 01/21/2023] Open
Abstract
Angiogenesis occurs early in tumor development, sustains primary tumor growth and provides a route for metastatic escape. The TGF-β family receptors modulate angiogenesis via endothelial-cell specific pathways. Here we investigate the interaction of two such receptors, ALK1 and endoglin, in pancreatic neuroendocrine tumors (PanNET). Independently, ALK1 and endoglin deficiencies exhibited genetically divergent phenotypes, while both highly correlate to an endothelial metagene in human and mouse PanNETs. A concurrent deficiency of both receptors synergistically decreased tumor burden to a greater extent than either individual knockdown. Furthermore, the knockout of Gdf2 (BMP9), the primary ligand for ALK1 and endoglin, exhibited a mixed phenotype from each of ALK1 and endoglin deficiencies; overall primary tumor burden decreased, but hepatic metastases increased. Tumors lacking BMP9 display a hyperbranching vasculature, and an increase in vascular mesenchymal-marker expression, which may be implicit in the increase in metastases. Taken together, our work cautions against singular blockade of BMP9 and instead demonstrates the utility of dual blockade of ALK1 and endoglin as a strategy for anti-angiogenic therapy in PanNET.
Collapse
Affiliation(s)
- Nikolas M Eleftheriou
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Jonas Sjölund
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Matteo Bocci
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Eliane Cortez
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Se-Jin Lee
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sara I Cunha
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| |
Collapse
|
144
|
Serum/glucocorticoid-regulated kinase 1 as a novel transcriptional target of bone morphogenetic protein-ALK1 receptor signaling in vascular endothelial cells. Angiogenesis 2018; 21:415-423. [DOI: 10.1007/s10456-018-9605-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 02/16/2018] [Indexed: 12/11/2022]
|
145
|
Wesseling M, Sakkers TR, de Jager SCA, Pasterkamp G, Goumans MJ. The morphological and molecular mechanisms of epithelial/endothelial-to-mesenchymal transition and its involvement in atherosclerosis. Vascul Pharmacol 2018; 106:1-8. [PMID: 29471141 DOI: 10.1016/j.vph.2018.02.006] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/05/2018] [Accepted: 02/17/2018] [Indexed: 12/26/2022]
Abstract
Cell transdifferentiation occurs during cardiovascular development or remodeling either as a pathologic feature in the progression of disease or as a response to injury. Endothelial-to-Mesenchymal Transition (EndMT) is a process that is classified as a specialized form of Epithelial-to-Mesenchymal Transition (EMT), in which epithelial cells lose their epithelial characteristics and gain a mesenchymal phenotype. During transdifferentiation, cells lose both cell-cell contacts and their attachment to the basement membrane. Subsequently, the shape of the cells changes from a cuboidal to an elongated shape. A rearrangement of actin filaments facilitates the cells to become motile and prime their migration into the underlying tissue. EMT is a key process during embryonic development, wound healing and tissue regeneration, but has also been implicated in pathophysiological processes, such organ fibrosis and tumor metastases. EndMT has been associated with additional pathophysiological processes in cardiovascular related diseases, including atherosclerosis. Recent studies prove a significant role for EndMT in the progression and destabilization of atherosclerotic plaques, as a consequence of EndMT-derived fibroblast infiltration and the increased secretion of matrix metalloproteinase respectively. In this review we will discuss the essential molecular and morphological mechanisms of EMT and EndMT, along with their common denominators and key differences. Finally, we will discuss the role of EMT/EndMT in developmental and pathophysiological processes, focusing on the potential role of EndMT in atherosclerosis in more depth.
Collapse
Affiliation(s)
- M Wesseling
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands; Laboratory of Clinical Chemistry and Histology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - T R Sakkers
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S C A de Jager
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands; Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - G Pasterkamp
- Laboratory of Clinical Chemistry and Histology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M J Goumans
- Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
146
|
Vascular deficiency of Smad4 causes arteriovenous malformations: a mouse model of Hereditary Hemorrhagic Telangiectasia. Angiogenesis 2018; 21:363-380. [PMID: 29460088 PMCID: PMC5878194 DOI: 10.1007/s10456-018-9602-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/28/2018] [Indexed: 12/18/2022]
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant vascular disorder that leads to abnormal connections between arteries and veins termed arteriovenous malformations (AVM). Mutations in TGFβ pathway members ALK1, ENG and SMAD4 lead to HHT. However, a Smad4 mouse model of HHT does not currently exist. We aimed to create and characterize a Smad4 endothelial cell (EC)-specific, inducible knockout mouse (Smad4f/f;Cdh5-CreERT2) that could be used to study AVM development in HHT. We found that postnatal ablation of Smad4 caused various vascular defects, including the formation of distinct AVMs in the neonate retina. Our analyses demonstrated that increased EC proliferation and size, altered mural cell coverage and distorted artery-vein gene expression are associated with Smad4 deficiency in the vasculature. Furthermore, we show that depletion of Smad4 leads to decreased Vegfr2 expression, and concurrent loss of endothelial Smad4 and Vegfr2 in vivo leads to AVM enlargement. Our work provides a new model in which to study HHT-associated phenotypes and links the TGFβ and VEGF signaling pathways in AVM pathogenesis.
Collapse
|
147
|
Alsina-Sanchís E, García-Ibáñez Y, Figueiredo AM, Riera-Domingo C, Figueras A, Matias-Guiu X, Casanovas O, Botella LM, Pujana MA, Riera-Mestre A, Graupera M, Viñals F. ALK1 Loss Results in Vascular Hyperplasia in Mice and Humans Through PI3K Activation. Arterioscler Thromb Vasc Biol 2018; 38:1216-1229. [PMID: 29449337 DOI: 10.1161/atvbaha.118.310760] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/31/2018] [Indexed: 12/21/2022]
Abstract
OBJECTIVE ALK1 (activin-receptor like kinase 1) is an endothelial cell-restricted receptor with high affinity for BMP (bone morphogenetic protein) 9 TGF-β (transforming growth factor-β) family member. Loss-of-function mutations in ALK1 cause a subtype of hereditary hemorrhagic telangiectasia-a rare disease characterized by vasculature malformations. Therapeutic strategies are aimed at reducing potential complications because of vascular malformations, but currently, there is no curative treatment for hereditary hemorrhagic telangiectasia. APPROACH AND RESULTS In this work, we report that a reduction in ALK1 gene dosage (heterozygous ALK1+/- mice) results in enhanced retinal endothelial cell proliferation and vascular hyperplasia at the sprouting front. We found that BMP9/ALK1 represses VEGF (vascular endothelial growth factor)-mediated PI3K (phosphatidylinositol 3-kinase) by promoting the activity of the PTEN (phosphatase and tensin homolog). Consequently, loss of ALK1 function in endothelial cells results in increased activity of the PI3K pathway. These results were confirmed in cutaneous telangiectasia biopsies of patients with hereditary hemorrhagic telangiectasia 2, in which we also detected an increase in endothelial cell proliferation linked to an increase on the PI3K pathway. In mice, genetic and pharmacological inhibition of PI3K is sufficient to abolish the vascular hyperplasia of ALK1+/- retinas and in turn normalize the vasculature. CONCLUSIONS Overall, our results indicate that the BMP9/ALK1 hub critically mediates vascular quiescence by limiting PI3K signaling and suggest that PI3K inhibitors could be used as novel therapeutic agents to treat hereditary hemorrhagic telangiectasia.
Collapse
Affiliation(s)
- Elisenda Alsina-Sanchís
- From the Program Against Cancer Therapeutic Resistance, Institut Català d'Oncologia, Hospital Duran i Reynals (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., O.C., M.A.P., M.G., F.V.).,Institut d'Investigació Biomèdica de Bellvitge, Spain (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., X.M.-G., O.C., M.A.P., F.V.)
| | - Yaiza García-Ibáñez
- From the Program Against Cancer Therapeutic Resistance, Institut Català d'Oncologia, Hospital Duran i Reynals (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., O.C., M.A.P., M.G., F.V.).,Institut d'Investigació Biomèdica de Bellvitge, Spain (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., X.M.-G., O.C., M.A.P., F.V.)
| | - Ana M Figueiredo
- From the Program Against Cancer Therapeutic Resistance, Institut Català d'Oncologia, Hospital Duran i Reynals (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., O.C., M.A.P., M.G., F.V.).,Vascular Signaling Laboratory, Institut d´Investigació Biomèdica de Bellvitge (A.M.F., M.G.), L'Hospitalet de Llobregat, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge, Spain (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., X.M.-G., O.C., M.A.P., F.V.)
| | - Carla Riera-Domingo
- From the Program Against Cancer Therapeutic Resistance, Institut Català d'Oncologia, Hospital Duran i Reynals (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., O.C., M.A.P., M.G., F.V.).,Institut d'Investigació Biomèdica de Bellvitge, Spain (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., X.M.-G., O.C., M.A.P., F.V.)
| | - Agnès Figueras
- From the Program Against Cancer Therapeutic Resistance, Institut Català d'Oncologia, Hospital Duran i Reynals (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., O.C., M.A.P., M.G., F.V.).,Institut d'Investigació Biomèdica de Bellvitge, Spain (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., X.M.-G., O.C., M.A.P., F.V.)
| | - Xavier Matias-Guiu
- Institut d'Investigació Biomèdica de Bellvitge, Spain (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., X.M.-G., O.C., M.A.P., F.V.).,Servei d'Anatomia Patològica (X.M.-G.).,Institut d'Investigació Biomèdica de Bellvitge, Hospital Universitari de Bellvitge, Spain; Hospital Universitari Arnau de Vilanova, Lleida, Spain (X.M.-G.).,Universitat de Lleida, Spain (X.M.-G.)
| | - Oriol Casanovas
- From the Program Against Cancer Therapeutic Resistance, Institut Català d'Oncologia, Hospital Duran i Reynals (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., O.C., M.A.P., M.G., F.V.).,Institut d'Investigació Biomèdica de Bellvitge, Spain (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., X.M.-G., O.C., M.A.P., F.V.)
| | - Luisa M Botella
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain (L.M.B.)
| | - Miquel A Pujana
- From the Program Against Cancer Therapeutic Resistance, Institut Català d'Oncologia, Hospital Duran i Reynals (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., O.C., M.A.P., M.G., F.V.).,Institut d'Investigació Biomèdica de Bellvitge, Spain (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., X.M.-G., O.C., M.A.P., F.V.)
| | - Antoni Riera-Mestre
- HHT Unit, Internal Medicine Department (A.R.-M.).,Departament de Ciències Clíniques, Universitat de Barcelona, Spain (A.R.-M.)
| | - Mariona Graupera
- From the Program Against Cancer Therapeutic Resistance, Institut Català d'Oncologia, Hospital Duran i Reynals (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., O.C., M.A.P., M.G., F.V.) .,Vascular Signaling Laboratory, Institut d´Investigació Biomèdica de Bellvitge (A.M.F., M.G.), L'Hospitalet de Llobregat, Barcelona, Spain.,CIBERONC, Madrid, Spain (M.G.)
| | - Francesc Viñals
- From the Program Against Cancer Therapeutic Resistance, Institut Català d'Oncologia, Hospital Duran i Reynals (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., O.C., M.A.P., M.G., F.V.) .,Institut d'Investigació Biomèdica de Bellvitge, Spain (E.A.-S., Y.G.-I., A.M.F., C.R.-D., A.F., X.M.-G., O.C., M.A.P., F.V.).,Departament de Ciències Fisiològiques, Campus de Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain (F.V.)
| |
Collapse
|
148
|
Goumans MJ, Ten Dijke P. TGF-β Signaling in Control of Cardiovascular Function. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a022210. [PMID: 28348036 DOI: 10.1101/cshperspect.a022210] [Citation(s) in RCA: 251] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Genetic studies in animals and humans indicate that gene mutations that functionally perturb transforming growth factor β (TGF-β) signaling are linked to specific hereditary vascular syndromes, including Osler-Rendu-Weber disease or hereditary hemorrhagic telangiectasia and Marfan syndrome. Disturbed TGF-β signaling can also cause nonhereditary disorders like atherosclerosis and cardiac fibrosis. Accordingly, cell culture studies using endothelial cells or smooth muscle cells (SMCs), cultured alone or together in two- or three-dimensional cell culture assays, on plastic or embedded in matrix, have shown that TGF-β has a pivotal effect on endothelial and SMC proliferation, differentiation, migration, tube formation, and sprouting. Moreover, TGF-β can stimulate endothelial-to-mesenchymal transition, a process shown to be of key importance in heart valve cushion formation and in various pathological vascular processes. Here, we discuss the roles of TGF-β in vasculogenesis, angiogenesis, and lymphangiogenesis and the deregulation of TGF-β signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Marie-José Goumans
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
149
|
Goumans MJ, Zwijsen A, Ten Dijke P, Bailly S. Bone Morphogenetic Proteins in Vascular Homeostasis and Disease. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a031989. [PMID: 28348038 DOI: 10.1101/cshperspect.a031989] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is well established that control of vascular morphogenesis and homeostasis is regulated by vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), Delta-like 4 (Dll4), angiopoietin, and ephrin signaling. It has become clear that signaling by bone morphogenetic proteins (BMPs), which have a long history of studies in bone and early heart development, are also essential for regulating vascular function. Indeed, mutations that cause deregulated BMP signaling are linked to two human vascular diseases, hereditary hemorrhagic telangiectasia and pulmonary arterial hypertension. These observations are corroborated by data obtained with vascular cells in cell culture and in mouse models. BMPs are required for normal endothelial cell differentiation and for venous/arterial and lymphatic specification. In adult life, BMP signaling orchestrates neo-angiogenesis as well as vascular inflammation, remodeling, and calcification responses to shear and oxidative stress. This review emphasizes the pivotal role of BMPs in the vascular system, based on studies of mouse models and human vascular disorders.
Collapse
Affiliation(s)
- Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - An Zwijsen
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium.,KU Leuven Department of Human Genetics, 3000 Leuven, Belgium
| | - Peter Ten Dijke
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Mécale (INSERM), U1036, 38000 Grenoble, France.,Laboratoire Biologie du Cancer et de l'Infection, Commissariat à l'Énergie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France.,University of Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
150
|
Activation of Activin receptor-like kinases curbs mucosal inflammation and proliferation in chronic rhinosinusitis with nasal polyps. Sci Rep 2018; 8:1561. [PMID: 29367682 PMCID: PMC5784055 DOI: 10.1038/s41598-018-19955-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 01/09/2018] [Indexed: 01/17/2023] Open
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) is a widespread disease causing obstruction of the nasal cavity. Its cause remains unclear. The transforming growth-factor beta (TGF-β) superfamily and their receptors, termed Activin receptor-like kinases (ALKs), have recently been suggested to play a role in local airway inflammation, but have so far not been evaluated in human nasal epithelial cells (HNECs) from CRSwNP patients. We demonstrated that ALK1–7 were expressed in the nasal polyp epithelium, and the expression of ALK1-6 was markedly elevated in polyps compared to nasal mucosa from healthy controls. Stimulation with the ALK ligand TGF-β1 decreased Ki67 expression in HNECs from CRSwNP patients, not evident in controls. Likewise, TGF-β1, Activin A and Activin B, all ALK ligands, decreased IL-8 release and Activin A and Activin B reduced ICAM1 expression on HNECs from CRSwNP patients, not seen in controls. Pre-stimulation with TGF-β1, Activin A, BMP4 and Activin B attenuated a TNF-α-induced ICAM1 upregulation on HNECs of CRSwNP. No effect was evident in controls. In conclusion, an increased expression of ALK1-6 was found on polyp epithelial cells and ligand stimulation appeared to reduce proliferation and local inflammation in polyps.
Collapse
|