1
|
Boscaro C, Schimdt G, Cignarella A, Dal Maso L, Bolego C, Trevisi L. The antiangiogenic effect of digitoxin is dependent on a ROS-elicited RhoA/ROCK pathway activation. Biochem Pharmacol 2024; 222:116049. [PMID: 38342347 DOI: 10.1016/j.bcp.2024.116049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/16/2024] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
We previously showed that digitoxin inhibits angiogenesis and cancer cell proliferation and migration and these effects were associated to protein tyrosine kinase 2 (FAK) inhibition. Considering the interactions between FAK and Rho GTPases regulating cell cytoskeleton and movement, we investigated the involvement of RhoA and Rac1 in the antiangiogenic effect of digitoxin. Phalloidin staining of human umbilical vein endothelial cells (HUVECs) showed the formation of stress fibers in cells treated with 10 nM digitoxin. By Rhotekin- and Pak1- pull down assays, detecting the GTP-bound form of GTPases, we observed that digitoxin (10-25 nM) induced sustained (0.5-6 h) RhoA activation with no effect on Rac1. Furthermore, inhibition of HUVEC migration and capillary-like tube formation by digitoxin was counteracted by hindering RhoA-ROCK axis with RhoA silencing or Y-27632 treatment. Digitoxin did not decrease p190RhoGAP phosphorylation at Tyr1105 (a site targeted by FAK), suggesting that RhoA activation was independent from FAK inhibition. Because increasing evidence points to a redox regulation of RhoA, we measured intracellular ROS and found that digitoxin treatment enhanced ROS levels in a concentration-dependent manner (1-25 nM). Notably, the flavoprotein inhibitor DPI or the pan-NADPH oxidase (NOX) inhibitor VAS-2870 antagonized both ROS increase and RhoA activation by digitoxin. Our results provide evidence that inhibition of HUVEC migration and tube formation by digitoxin is dependent on ROS production by endothelial NOX, which leads to the activation of RhoA/ROCK pathway. Digitoxin effects on proteins regulating cytoskeletal organization and cell motility could have a wider impact on cancer progression, beyond the antiangiogenic activity.
Collapse
Affiliation(s)
| | - Gudula Schimdt
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Lucia Dal Maso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Lucia Trevisi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.
| |
Collapse
|
2
|
Abdo AI, Tran HB, Hodge S, Beltrame JF, Zalewski PD. Zinc Homeostasis Alters Zinc Transporter Protein Expression in Vascular Endothelial and Smooth Muscle Cells. Biol Trace Elem Res 2021; 199:2158-2171. [PMID: 32776265 DOI: 10.1007/s12011-020-02328-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/03/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Zinc is an important essential micronutrient with anti-oxidative and anti-inflammatory properties in humans. The role of zinc in signalling has been characterized in the nervous, endocrine, gastrointestinal, renal and reproductive systems. Relatively little is known regarding its role in the vascular system, but the role of zinc homeostasis in augmenting vascular health and vasorelaxation is emerging. Zinc transport proteins are integral to the protective function of zinc, but knowledge of their expression in vascular endothelial and smooth muscle cells is lacking. METHODOLOGY Human coronary artery endothelial cells and pulmonary artery smooth muscle cells were assessed for gene expression (RT-PCR) of SLC39A (ZIP), SLC30A (ZnT) and metallothionein (MT) families of Zn transporters and storage proteins. Protein expression (fluorescence confocal microscopy) was then analysed for the proteins of interest that changed mRNA expression: ZIP2, ZIP12, ZnT1, ZnT2 and MT1/2. RESULTS Endothelial and smooth muscle cell mRNA expression of ZnT1, ZnT2 and MT1 was significantly downregulated by low and high Zn conditions, while ZIP2 and ZIP12 expression was induced by Zn depletion with the Zn chelator, TPEN. Changes in gene expression were consistent with protein expression levels for ZIP2, ZIP12 and MT1, where ZIP2 was localized to intracellular bodies and ZIP12 to lamellipodia. CONCLUSION Vascular endothelial and smooth muscle cells actively regulate specific Zn transport and metallothionein gene and protein expressions to achieve Zn homeostasis.
Collapse
Affiliation(s)
- Adrian I Abdo
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, 28 Woodville Rd, Woodville South, SA, 5011, Australia.
- Faculty of Health and Medical Sciences, University of Adelaide, 4 North Terrace, Adelaide, SA, 5000, Australia.
| | - Hai Bac Tran
- Faculty of Health and Medical Sciences, University of Adelaide, 4 North Terrace, Adelaide, SA, 5000, Australia
| | - Sandra Hodge
- Faculty of Health and Medical Sciences, University of Adelaide, 4 North Terrace, Adelaide, SA, 5000, Australia
| | - John F Beltrame
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, 28 Woodville Rd, Woodville South, SA, 5011, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, 4 North Terrace, Adelaide, SA, 5000, Australia
| | - Peter D Zalewski
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, 28 Woodville Rd, Woodville South, SA, 5011, Australia.
- Faculty of Health and Medical Sciences, University of Adelaide, 4 North Terrace, Adelaide, SA, 5000, Australia.
| |
Collapse
|
3
|
Kalkan BM, Akgol S, Ak D, Yucel D, Guney Esken G, Kocabas F. CASIN and AMD3100 enhance endothelial cell proliferation, tube formation and sprouting. Microvasc Res 2020; 130:104001. [PMID: 32198058 DOI: 10.1016/j.mvr.2020.104001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/27/2020] [Accepted: 03/14/2020] [Indexed: 01/12/2023]
Abstract
Endothelial dysfunction is prominent in atherosclerosis, hypertension, diabetes, peripheral and cardiovascular diseases, and stroke. Novel therapeutic approaches to these conditions often involve development of tissue-engineered veins with ex vivo expanded endothelial cells. However, high cell number requirements limit these approaches to become applicable to clinical applications and highlight the requirement of technologies that accelerate expansion of vascular-forming cells. We have previously shown that novel small molecules could induce hematopoietic stem cell expansion ex vivo. We hypothesized that various small molecules targeting hematopoietic stem cell quiescence and mobilization could be used to induce endothelial cell expansion and angiogenesis due to common origin and shared characteristics of endothelial and hematopoietic cells. Here, we have screened thirty-five small molecules and found that CASIN and AMD3100 increase endothelial cell expansion up to two-fold and induce tube formation and ex vivo sprouting. In addition, we have studied how CASIN and AMD3100 affect cell migration, apoptosis and cell cycle of endothelial cells. CASIN and AMD3100 upregulate key endothelial marker genes and downregulate a number of cyclin dependent kinase inhibitors. These findings suggest that CASIN and AMD3100 could be further tested in the development of artificial vascular systems and vascular gene editing technologies. Furthermore, these findings may have potential to contribute to the development of alternative treatment methods for diseases that cause endothelial damage.
Collapse
Affiliation(s)
- Batuhan Mert Kalkan
- Regenerative Biology Research Laboratory, Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey; Koc University, Istanbul, Turkey
| | - Sezer Akgol
- Regenerative Biology Research Laboratory, Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Deniz Ak
- Regenerative Biology Research Laboratory, Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey; Middle East Technical University, Ankara, Turkey
| | - Dogacan Yucel
- Faculty of Medicine, University of Minnesota, MN, USA
| | - Gulen Guney Esken
- Regenerative Biology Research Laboratory, Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Fatih Kocabas
- Regenerative Biology Research Laboratory, Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
4
|
Cannito S, Foglia B, Villano G, Turato C, C Delgado T, Morello E, Pin F, Novo E, Napione L, Quarta S, Ruvoletto M, Fasolato S, Zanus G, Colombatto S, Lopitz-Otsoa F, Fernández-Ramos D, Bussolino F, Sutti S, Albano E, Martínez-Chantar ML, Pontisso P, Parola M. SerpinB3 Differently Up-Regulates Hypoxia Inducible Factors -1α and -2α in Hepatocellular Carcinoma: Mechanisms Revealing Novel Potential Therapeutic Targets. Cancers (Basel) 2019; 11:1933. [PMID: 31817100 PMCID: PMC6966556 DOI: 10.3390/cancers11121933] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/15/2022] Open
Abstract
SerpinB3 (SB3) is a hypoxia and hypoxia-inducible factor (HIF)-2α-dependent cysteine-protease inhibitor up-regulated in hepatocellular carcinoma (HCC), released by cancer cells and able to stimulate proliferation and epithelial-to-mesenchymal-transition. Methods: In the study we employed transgenic and knock out SerpinB3 mice, liver cancer cell line, human HCC specimens, and mice receiving diethyl-nitrosamine (DEN) administration plus choline-deficient L-amino acid refined (CDAA) diet (DEN/CDAA protocol). Results: We provide detailed and mechanistic evidence that SB3 can act as a paracrine mediator able to affect the behavior of surrounding cells by differentially up-regulating, in normoxic conditions, HIF-1α and HIF-2α. SB3 acts by (i) up-regulating HIF-1α transcription, facilitating cell survival in a harsh microenvironment and promoting angiogenesis, (ii) increasing HIF-2α stabilization via direct/selective NEDDylation, promoting proliferation of liver cancer cells, and favoring HCC progression. Moreover (iii) the highest levels of NEDD8-E1 activating enzyme (NAE1) mRNA were detected in a subclass of HCC patients expressing the highest levels of HIF-2α transcripts; (iv) mice undergoing DEN/CDAA carcinogenic protocol showed a positive correlation between SB3 and HIF-2α transcripts with the highest levels of NAE1 mRNA detected in nodules expressing the highest levels of HIF-2α transcripts. Conclusions: These data outline either HIF-2α and NEDDylation as two novel putative therapeutic targets to interfere with the procarcinogenic role of SerpinB3 in the development of HCC.
Collapse
Affiliation(s)
- Stefania Cannito
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Beatrice Foglia
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Gianmarco Villano
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (G.V.); (M.R.); (S.F.)
| | - Cristian Turato
- Veneto Institute of Oncology IOV—IRCCS, 35128 Padova, Italy;
| | - Teresa C Delgado
- Liver Disease and Metabolism Laboratory, CIC bioGUNE, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain; (T.C.D.); (F.L.-O.); (D.F.-R.); (M.L.M.-C.)
| | - Elisabetta Morello
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Fabrizio Pin
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Erica Novo
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| | - Lucia Napione
- Department of Applied Science and Technology, Politecnico di Torino, 10129 Torino, Italy;
- Laboratory of Vascular Oncology Candiolo Cancer Institute—FPO IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), 10060 Candiolo, Italy;
| | - Santina Quarta
- Department of Medicine, University of Padova, 35128 Padova, Italy; (S.Q.); (P.P.)
| | - Mariagrazia Ruvoletto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (G.V.); (M.R.); (S.F.)
| | - Silvano Fasolato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (G.V.); (M.R.); (S.F.)
| | - Giacomo Zanus
- Hepatobiliary Surgery, University of Padova, 35128 Padova, Italy;
| | | | - Fernando Lopitz-Otsoa
- Liver Disease and Metabolism Laboratory, CIC bioGUNE, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain; (T.C.D.); (F.L.-O.); (D.F.-R.); (M.L.M.-C.)
| | - David Fernández-Ramos
- Liver Disease and Metabolism Laboratory, CIC bioGUNE, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain; (T.C.D.); (F.L.-O.); (D.F.-R.); (M.L.M.-C.)
| | - Federico Bussolino
- Laboratory of Vascular Oncology Candiolo Cancer Institute—FPO IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), 10060 Candiolo, Italy;
- Department of Oncology, University of Torino, 10125 Torino, Italy;
| | - Salvatore Sutti
- Department of Health Sciences and Interdisciplinary Research Center for Autoimmune Diseases, University Amedeo Avogadro of East Piedmont, 28100 Novara, Italy; (S.S.); (E.A.)
| | - Emanuele Albano
- Department of Health Sciences and Interdisciplinary Research Center for Autoimmune Diseases, University Amedeo Avogadro of East Piedmont, 28100 Novara, Italy; (S.S.); (E.A.)
| | - Maria Luz Martínez-Chantar
- Liver Disease and Metabolism Laboratory, CIC bioGUNE, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain; (T.C.D.); (F.L.-O.); (D.F.-R.); (M.L.M.-C.)
| | - Patrizia Pontisso
- Department of Medicine, University of Padova, 35128 Padova, Italy; (S.Q.); (P.P.)
| | - Maurizio Parola
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine & Clinical Pathology, University of Torino, 10125 Torino, Italy; (S.C.); (B.F.); (E.M.); (F.P.); (E.N.)
| |
Collapse
|
5
|
Keremu A, Yaoliwasi A, Tuerhong M, Kadeer N, Heyi, Yiming A, Yilike X. Research on the establishment of chronic stress-induced premature ovarian failure the rat model and effects of Chinese medicine Muniziqi treatment. Mol Reprod Dev 2018; 86:175-186. [PMID: 30512210 DOI: 10.1002/mrd.23092] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 10/30/2018] [Indexed: 12/20/2022]
Abstract
The purposes of this study were to establish and to explore the biological basis of the chronic stress-induced premature ovarian failure (POF) model and to explore the therapeutic effects of the traditional Chinese medicine Muniziqi. Sexually matured female Sprague-Dawley rats were fed with spinach and cilantro in cold and wet conditions for about 20 weeks until a chronic stress (CS) model was established. The CS rats were divided into a POF stress model group and a stress model group according to weekly biological characteristics and hormone level detection ( luteinizing hormone [LH], follicle stimulating hormone [FSH], and estrogen [E2]). To investigate the therapeutic effect of Muniziqi, the POF disease stress model group was divided into the high-, medium-, and low-drug intervention groups. The results showed that chronic stresses (special food, cold, damp) can lead to POF disease. The traditional Chinese medicine Muniziqi could not only improve the reproductive hormone level disorder, but also improve the function of the hypothalamus-pituitary-ovarian axis. The underlying mechanism may be a change in the E2, LH, and FSH hormone levels in serum and lower expression of ovarian premature aging-related protein PFN-1.
Collapse
Affiliation(s)
- Abulizi Keremu
- Department of Biology, School of Medicine, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Aziguli Yaoliwasi
- Department of Biology, School of Medicine, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Mayire Tuerhong
- Morphology Center, School of Medicine, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Nafeisha Kadeer
- Department of Biology, School of Medicine, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Heyi
- Houbo College of Xinjiang Medical University, Karamay, Xinjiang, China
| | - Adilijiang Yiming
- Department of Human Anatomy, School of Medicine, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xiamixinuer Yilike
- Department of Biology, School of Medicine, Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
6
|
Valla M, Mjønes PG, Engstrøm MJ, Ytterhus B, Bordin DL, van Loon B, Akslen LA, Vatten LJ, Opdahl S, Bofin AM. Characterization of FGD5 Expression in Primary Breast Cancers and Lymph Node Metastases. J Histochem Cytochem 2018; 66:787-799. [PMID: 30052477 DOI: 10.1369/0022155418792032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Faciogenital dysplasia 5 ( FGD5) amplification drives tumor cell proliferation, and is present in 9.5% of breast cancers. We describe FGD5 expression, assess associations between FGD5 amplification and FGD5 expression, and assess FGD5 expression in relation to proliferation and prognosis. FGD5 immunohistochemistry was done on primary tumors ( n=829) and lymph node metastases ( n=231) from a cohort of Norwegian patients. We explored associations between FGD5 amplification, FGD5 expression, and proliferation, and analyzed the prognostic value of FGD5 expression by estimating cumulative risks of death and hazard ratios (HRs). We identified nuclear and cytoplasmic expression in 64% and 73% of primary tumors, respectively, and found an association between gene amplification and nuclear expression ( p=0.02). The proportion of cases with FGD5 expression was higher in lymph node metastases, compared with primary tumors ( p=0.004 for nuclear and p=0.001 for cytoplasmic staining). Neither proliferation nor prognosis was associated with FGD5 expression (age-adjusted HR 1.12 [95% confidence interval = 0.89-1.41] for nuclear expression; and 0.88 [95% CI = 0.70-1.12] for cytoplasmic expression). FGD5 is expressed in a high proportion of breast cancers and lymph node metastases. There was a correlation between FGD5 amplification and nuclear expression, but no association between FGD5 expression and proliferation or prognosis.
Collapse
Affiliation(s)
- Marit Valla
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Patricia G Mjønes
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Pathology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Monica J Engstrøm
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Breast and Endocrine Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Borgny Ytterhus
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Diana L Bordin
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Barbara van Loon
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Pathology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Lars A Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Lars J Vatten
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Signe Opdahl
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anna M Bofin
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
7
|
Tagashira T, Fukuda T, Miyata M, Nakamura K, Fujita H, Takai Y, Hirata KI, Rikitake Y. Afadin Facilitates Vascular Endothelial Growth Factor–Induced Network Formation and Migration of Vascular Endothelial Cells by Inactivating Rho-Associated Kinase Through ArhGAP29. Arterioscler Thromb Vasc Biol 2018; 38:1159-1169. [DOI: 10.1161/atvbaha.118.310991] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 03/12/2018] [Indexed: 01/19/2023]
Abstract
Objective—
We previously reported that afadin, an actin filament-binding protein, regulated vascular endothelial growth factor–induced angiogenesis. However, the underlying molecular mechanisms are poorly understood. Here, we investigated the mechanisms of how Rho-associated kinase is activated in afadin-knockdown human umbilical vein endothelial cells (HUVECs) and how its activation is involved in defects of vascular endothelial growth factor–induced network formation and migration of the cells.
Approach and Results—
Knockdown of afadin or ArhGAP29, a GTPase-activating protein for RhoA, increased Rho-associated kinase activity and reduced the vascular endothelial growth factor–induced network formation and migration of cultured HUVECs, accompanied by the defective formation of membrane protrusions, such as lamellipodia and peripheral ruffles. Treatment of the afadin- or ArhGAP29-knockdown HUVECs with Rho-associated kinase inhibitors, Y-27632 or fasudil, partially restored the reduced network formation and migration as well as the defective formation of membrane protrusions. ArhGAP29 bound to afadin and was colocalized with afadin at the leading edge of migrating HUVECs. The defective formation of membrane protrusions in ArhGAP29-knockdown HUVECs was restored by expression of mutant ArhGAP29 that bound to afadin and contained a RhoGAP domain but not mutant ArhGAP29 that could bind to afadin and lacked the RhoGAP domain or mutant ArhGAP29 that could not bind to afadin and contained the RhoGAP domain. This suggested the requirement of both the interaction of afadin with ArhGAP29 and RhoGAP activity of ArhGAP29 for migration of HUVECs.
Conclusions—
Our results highlight a critical role of the afadin–ArhGAP29 axis for the regulation of Rho-associated kinase activity during vascular endothelial growth factor–induced network formation and migration of HUVECs.
Collapse
Affiliation(s)
- Toru Tagashira
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.T., T.F., K.-i.H.)
| | - Terunobu Fukuda
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.T., T.F., K.-i.H.)
| | - Muneaki Miyata
- Division of Signal Transduction, Department of Biochemistry and Molecular Biology (M.M., K.N., Y.R.)
| | - Kazuha Nakamura
- Division of Signal Transduction, Department of Biochemistry and Molecular Biology (M.M., K.N., Y.R.)
| | - Hidenobu Fujita
- Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, Japan (H.F., Y.R.)
| | - Yoshimi Takai
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology (Y.T.), Kobe University Graduate School of Medicine, Japan
| | - Ken-ichi Hirata
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.T., T.F., K.-i.H.)
| | - Yoshiyuki Rikitake
- Division of Signal Transduction, Department of Biochemistry and Molecular Biology (M.M., K.N., Y.R.)
- Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, Japan (H.F., Y.R.)
| |
Collapse
|
8
|
Lee MC, Shei W, Chan AS, Chua BT, Goh SR, Chong YF, Hilmy MH, Nongpiur ME, Baskaran M, Khor CC, Aung T, Hunziker W, Vithana EN. Primary angle closure glaucoma (PACG) susceptibility gene PLEKHA7 encodes a novel Rac1/Cdc42 GAP that modulates cell migration and blood-aqueous barrier function. Hum Mol Genet 2018; 26:4011-4027. [PMID: 29016860 DOI: 10.1093/hmg/ddx292] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/18/2017] [Indexed: 12/21/2022] Open
Abstract
PLEKHA7, a gene recently associated with primary angle closure glaucoma (PACG), encodes an apical junctional protein expressed in components of the blood aqueous barrier (BAB). We found that PLEKHA7 is down-regulated in lens epithelial cells and in iris tissue of PACG patients. PLEKHA7 expression also correlated with the C risk allele of the sentinel SNP rs11024102 with the risk allele carrier groups having significantly reduced PLEKHA7 levels compared to non-risk allele carriers. Silencing of PLEKHA7 in human immortalized non-pigmented ciliary epithelium (h-iNPCE) and primary trabecular meshwork cells, which are intimately linked to BAB and aqueous humor outflow respectively, affected actin cytoskeleton organization. PLEKHA7 specifically interacts with GTP-bound Rac1 and Cdc42, but not RhoA, and the activation status of the two small GTPases is linked to PLEKHA7 expression levels. PLEKHA7 stimulates Rac1 and Cdc42 GTP hydrolysis, without affecting nucleotide exchange, identifying PLEKHA7 as a novel Rac1/Cdc42 GAP. Consistent with the regulatory role of Rac1 and Cdc42 in maintaining the tight junction permeability, silencing of PLEKHA7 compromises the paracellular barrier between h-iNPCE cells. Thus, downregulation of PLEKHA7 in PACG may affect BAB integrity and aqueous humor outflow via its Rac1/Cdc42 GAP activity, thereby contributing to disease etiology.
Collapse
Affiliation(s)
- Mei-Chin Lee
- Ocular Genetics Research Group, Singapore Eye Research Institute, Singapore 169856, Singapore.,The Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - William Shei
- Ocular Genetics Research Group, Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Anita S Chan
- The Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore.,Department of Glaucoma, Singapore National Eye Centre, Singapore 168751, Singapore
| | - Boon-Tin Chua
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Singapore 138673, Singapore
| | - Shuang-Ru Goh
- The Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Yaan-Fun Chong
- The Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Maryam H Hilmy
- Department of Pathology, Singapore General Hospital, Singapore 169856, Singapore
| | - Monisha E Nongpiur
- Ocular Genetics Research Group, Singapore Eye Research Institute, Singapore 169856, Singapore.,The Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Mani Baskaran
- Ocular Genetics Research Group, Singapore Eye Research Institute, Singapore 169856, Singapore.,The Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore.,Department of Glaucoma, Singapore National Eye Centre, Singapore 168751, Singapore
| | - Chiea-Chuen Khor
- Ocular Genetics Research Group, Singapore Eye Research Institute, Singapore 169856, Singapore.,Department of Human Genetics, Genome Institute of Singapore, Agency for Science Technology and Research, Singapore 138672, Singapore.,Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore
| | - Tin Aung
- Ocular Genetics Research Group, Singapore Eye Research Institute, Singapore 169856, Singapore.,The Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore.,Department of Glaucoma, Singapore National Eye Centre, Singapore 168751, Singapore.,Department of Ophthalmology, National University of Singapore, Singapore 119228, Singapore
| | - Walter Hunziker
- Ocular Genetics Research Group, Singapore Eye Research Institute, Singapore 169856, Singapore.,Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Singapore 138673, Singapore.,Department of Physiology, National University of Singapore, Singapore 117593, Singapore
| | - Eranga N Vithana
- Ocular Genetics Research Group, Singapore Eye Research Institute, Singapore 169856, Singapore.,The Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore.,Department of Ophthalmology, National University of Singapore, Singapore 119228, Singapore
| |
Collapse
|
9
|
Chang SC, Yang WCV. Hyperglycemia, tumorigenesis, and chronic inflammation. Crit Rev Oncol Hematol 2016; 108:146-153. [PMID: 27931833 DOI: 10.1016/j.critrevonc.2016.11.003] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/08/2016] [Indexed: 12/21/2022] Open
Abstract
Hyperglycemia is the most prominent sign that characterizes diabetes. Hyperglycemia favors malignant cell growth by providing energy to cancer cells. Clinical studies also showed an increased risk of diabetes being associated with different types of cancers. In addition, poorly regulated glucose metabolism in diabetic patients is often found with increased levels of chronic inflammatory markers, e.g., interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α, and emerging evidence has highlighted activation of the immune response in the progression and development of cancer cells. Therefore, uncontrolled proinflammatory responses could conceivably create a chronic inflammatory state, promoting a tumor-favorable microenvironment and potentially triggering immune overactivation and cancer growth. To further understand how hyperglycemia contributes to immune overactivation, the tumor microenvironment and the development of chronic inflammation-associated tumors may provide insights into tumor biology and immunology. This paper provides a brief introduction to hyperglycemia-associated diseases, followed by a comprehensive overview of the current findings of regulatory molecular mechanisms of glycosylation on proteoglycans in the extracellular matrix under hyperglycemic conditions. Then, the authors discuss the role of hyperglycemia in tumorigenesis (particularly in prostate, liver, colorectal, and pancreatic cancers), as well as the contribution of hyperglycemia to chronic inflammation. The authors end with a brief discussion on the future perspectives of hyperglycemia/tumorigenesis and potential applications of alternative/effective therapeutic strategies for hyperglycemia-associated cancers.
Collapse
Affiliation(s)
- Shu-Chun Chang
- The Ph.D. Program for Translational Medicine, College for Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| | - Wei-Chung Vivian Yang
- The Ph.D. Program for Translational Medicine, College for Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
10
|
Hernández-García R, Iruela-Arispe ML, Reyes-Cruz G, Vázquez-Prado J. Endothelial RhoGEFs: A systematic analysis of their expression profiles in VEGF-stimulated and tumor endothelial cells. Vascul Pharmacol 2015; 74:60-72. [PMID: 26471833 DOI: 10.1016/j.vph.2015.10.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 12/18/2022]
Abstract
Rho guanine nucleotide exchange factors (RhoGEFs) integrate cell signaling inputs into morphological and functional responses. However, little is known about the endothelial repertoire of RhoGEFs and their regulation. Thus, we assessed the expression of 81 RhoGEFs (70 homologous to Dbl and 11 of the DOCK family) in endothelial cells. Further, in the case of DH-RhoGEFs, we also determined their responses to VEGF exposure in vitro and in the context of tumors. A phylogenetic analysis revealed the existence of four groups of DH-RhoGEFs and two of the DOCK family. Among them, we found that the most abundant endothelial RhoGEFs were: Tuba, FGD5, Farp1, ARHGEF17, TRIO, P-Rex1, ARHGEF15, ARHGEF11, ABR, Farp2, ARHGEF40, ALS, DOCK1, DOCK7 and DOCK6. Expression of RASGRF2 and PREX2 increased significantly in response to VEGF, but most other RhoGEFs were unaffected. Interestingly murine endothelial cells isolated from tumors showed that all four phylogenetic subgroups of DH-RhoGEFs were altered when compared to non-tumor endothelial cells. In summary, our results provide a detailed assessment of RhoGEFs expression profiles in the endothelium and set the basis to systematically address their regulation in vascular signaling.
Collapse
Affiliation(s)
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell, and Developmental Biology and Molecular Biology Institute,University of California,Los Angeles, CA,USA
| | | | | |
Collapse
|
11
|
Yamana S, Tokiyama A, Mizutani K, Hirata KI, Takai Y, Rikitake Y. The Cell Adhesion Molecule Necl-4/CADM4 Serves as a Novel Regulator for Contact Inhibition of Cell Movement and Proliferation. PLoS One 2015; 10:e0124259. [PMID: 25893857 PMCID: PMC4404054 DOI: 10.1371/journal.pone.0124259] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/11/2015] [Indexed: 01/16/2023] Open
Abstract
Contact inhibition of cell movement and proliferation is critical for proper organogenesis and tissue remodeling. We show here a novel regulatory mechanism for this contact inhibition using cultured vascular endothelial cells. When the cells were confluently cultured, Necl-4 was up-regulated and localized at cell–cell contact sites where it cis-interacted with the vascular endothelial growth factor (VEGF) receptor. This interaction inhibited the tyrosine-phosphorylation of the VEGF receptor through protein-tyrosine phosphatase, non-receptor type 13 (PTPN13), eventually reducing cell movement and proliferation. When the cells were sparsely cultured, Necl-4 was down-regulated but accumulated at leading edges where it inhibited the activation of Rho-associated protein kinase through PTPN13, eventually facilitating the VEGF-induced activation of Rac1 and enhancing cell movement. Necl-4 further facilitated the activation of extracellular signal-regulated kinase 1/2, eventually enhancing cell proliferation. Thus, Necl-4 serves as a novel regulator for contact inhibition of cell movement and proliferation cooperatively with the VEGF receptor and PTPN13.
Collapse
Affiliation(s)
- Shota Yamana
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Amina Tokiyama
- Division of Signal Transduction, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Kiyohito Mizutani
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Ken-ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yoshimi Takai
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yoshiyuki Rikitake
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Division of Signal Transduction, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- * E-mail:
| |
Collapse
|
12
|
RhoA/mDia-1/profilin-1 signaling targets microvascular endothelial dysfunction in diabetic retinopathy. Graefes Arch Clin Exp Ophthalmol 2015; 253:669-80. [PMID: 25791356 DOI: 10.1007/s00417-015-2985-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 02/21/2015] [Accepted: 03/02/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is a major cause of blindness in the working-age populations of developed countries, and effective treatments and prevention measures have long been the foci of study. Patients with DR invariably demonstrate impairments of the retinal microvascular endothelium. Many observational and preclinical studies have shown that angiogenesis and apoptosis play crucial roles in the pathogenesis of DR. Increasing evidence suggests that in DR, the small guanosine-5'-triphosphate-binding protein RhoA activates its downstream targets mammalian Diaphanous homolog 1 (mDia-1) and profilin-1, thus affecting important cellular functions, including cell morphology, motility, secretion, proliferation, and gene expression. However, the specific underlying mechanism of disease remains unclear. CONCLUSION This review focuses on the RhoA/mDia-1/profilin-1 signaling pathway that specifically triggers endothelial dysfunction in diabetic patients. Recently, RhoA and profilin-1 signaling has attracted a great deal of attention in the context of diabetes-related research. However, the precise molecular mechanism by which the RhoA/mDia-1/profilin-1 pathway is involved in progression of microvascular endothelial dysfunction (MVED) during DR has not been determined. This review briefly describes each feature of the cascade before exploring the most recent findings on how the pathway may trigger endothelial dysfunction in DR. When the underlying mechanisms are understood, novel therapies seeking to restore the endothelial homeostasis comprised in DR will become possible.
Collapse
|
13
|
The cholesterol biosynthesis enzyme oxidosqualene cyclase is a new target to impair tumour angiogenesis and metastasis dissemination. Sci Rep 2015; 5:9054. [PMID: 25761781 PMCID: PMC4357009 DOI: 10.1038/srep09054] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 02/16/2015] [Indexed: 12/13/2022] Open
Abstract
Aberrant cholesterol homeostasis and biosynthesis has been observed in different tumour types. This paper investigates the role of the post-squalenic enzyme of cholesterol biosynthesis, oxidosqualene cyclase (OSC), in regulating tumour angiogenesis and metastasis dissemination in mouse models of cancer. We showed that Ro 48-8071, a selective inhibitor of OSC, reduced vascular density and increased pericyte coverage, with a consequent inhibition of tumour growth in a spontaneous mouse model of pancreatic tumour (RIP-Tag2) and two metastatic mouse models of human colon carcinoma (HCT116) and pancreatic adenocarcinoma (HPAF-II). Remarkably, the inhibition of OSC hampered metastasis formation in HCT116 and HPAF-II models. Ro 48-8071 induced tumour vessel normalization and enhanced the anti-tumoral and anti-metastatic effects of 5-fluorouracil (5-FU) in HCT116 mice. Ro 48-8071 exerted a strong anti-angiogenic activity by impairing endothelial cell adhesion and migration, and by blocking vessel formation in angiogenesis assays. OSC inhibition specifically interfered with the PI3K pathway. According to in vitro results, Ro 48-8071 specifically inhibited Akt phosphorylation in both cancer cells and tumour vasculature in all treated models. Thus, our results unveil a crucial role of OSC in the regulation of cancer progression and tumour angiogenesis, and indicate Ro 48-8071 as a potential novel anti-angiogenic and anti-metastatic drug.
Collapse
|
14
|
Wilkins JR, Pike DB, Gibson CC, Li L, Shiu YT. The interplay of cyclic stretch and vascular endothelial growth factor in regulating the initial steps for angiogenesis. Biotechnol Prog 2014; 31:248-57. [PMID: 25376776 DOI: 10.1002/btpr.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 09/27/2014] [Indexed: 12/12/2022]
Abstract
Angiogenesis is regulated by chemical and mechanical factors in vivo. The regulatory role of mechanical factors and how chemical and mechanical angiogenic regulators work in concert remains to be explored. We investigated the effect of cyclic uniaxial stretch (20%, 1 Hz), with and without the stimulation of vascular endothelial growth factor (VEGF), on sprouting angiogenesis by employing a stretchable three-dimensional cell culture model. When compared to static controls, stretch alone significantly increased the density of endothelial sprouts, and these sprouts aligned perpendicular to the direction of stretch. The Rho-associated kinase (ROCK) inhibitor Y27632 suppressed stretch-induced sprouting angiogenesis and associated sprout alignment. While VEGF is a potent angiogenic stimulus through ROCK-dependent pathways, the combination of VEGF and stretch did not have an additive effect on angiogenesis. In the presence of VEGF stimulation, the ROCK inhibitor suppressed stretch-induced sprout alignment but did not affect stretch-induced sprout density; in contrast, the receptor tyrosine kinase (RTK) inhibitor sunitinib had no effect on stretch-induced alignment but trended toward suppressed stretch-induced sprout density. Our results suggest that the formation of sprouts and their directionality do not have completely identical regulatory pathways, and thus it is possible to separately manipulate the number and pattern of new sprouts.
Collapse
Affiliation(s)
- Justin R Wilkins
- Dept. of Medicine, Div. of Nephrology, University of Utah School of Medicine, Salt Lake City, UT, 84112; Dept. of Bioengineering, University of Utah, Salt Lake City, UT, 84112
| | | | | | | | | |
Collapse
|
15
|
Hoch E, Tovar GEM, Borchers K. Bioprinting of artificial blood vessels: current approaches towards a demanding goal. Eur J Cardiothorac Surg 2014; 46:767-78. [PMID: 24970571 DOI: 10.1093/ejcts/ezu242] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Free-form fabrication techniques, often referred to as '3D printing', are currently tested with regard to the processing of biological and biocompatible materials in general and for fabrication of vessel-like structures in particular. Such computer-controlled methods assemble 3D objects by layer-wise deposition or layer-wise cross-linking of materials. They use, for example, nozzle-based deposition of hydrogels and cells, drop-on-demand inkjet-printing of cell suspensions with subsequent cross-linking, layer-by-layer cross-linking of synthetic or biological polymers by selective irradiation with light and even laser-induced deposition of single cells. The need of vessel-like structures has become increasingly crucial for the supply of encapsulated cells for 3D tissue engineering, or even with regard to future application such as vascular grafts. The anticipated potential of providing tubes with tailored branching geometries made of biocompatible or biological materials pushes future visions of patient-specific vascularized tissue substitutions, tissue-engineered blood vessels and bio-based vascular grafts. We review here the early attempts of bringing together innovative free-form manufacturing processes with bio-based and biodegradable materials. The presented studies provide many important proofs of concepts such as the possibility to integrate viable cells into computer-controlled processes and the feasibility of supplying cells in a hydrogel matrix by generation of a network of perfused channels. Several impressive results in the generation of complex shapes and high-aspect-ratio tubular structures demonstrate the potential of additive assembly methods. Yet, it also becomes obvious that there remain major challenges to simultaneously match all material requirements in terms of biological functions (cell function supporting properties), physicochemical functions (mechanical properties of the printed material) and process-related (viscosity, cross-linkability) functions, towards the demanding goal of biofabricating artificial blood vessels.
Collapse
Affiliation(s)
- Eva Hoch
- Institute for Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany
| | - Günter E M Tovar
- Institute for Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Kirsten Borchers
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| |
Collapse
|
16
|
Ma J, Xue Y, Liu W, Yue C, Bi F, Xu J, Zhang J, Li Y, Zhong C, Chen Y. Role of activated Rac1/Cdc42 in mediating endothelial cell proliferation and tumor angiogenesis in breast cancer. PLoS One 2013; 8:e66275. [PMID: 23750283 PMCID: PMC3672132 DOI: 10.1371/journal.pone.0066275] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 05/03/2013] [Indexed: 01/09/2023] Open
Abstract
Angiogenesis is a well-established target in anti-cancer therapy. Although vascular endothelial growth factor (VEGF)-mediated angiogenesis apparently requires the Rho GTPases Rac1 and Cdc42, the relevant mechanisms are unclear. Here, we determined that activated Rac1/Cdc42 in MCF-7 breast cancer cells could decrease p53 protein levels and increase VEGF secretion to promote proliferation and tube formation of human umbilical vein endothelial cells (HUVECs). However, these effects are reversed after ubiquitin-proteasome breakage. In exploring potential mechanisms for this relationship, we confirmed that activated Rac1/Cdc42 could enhance p53 protein ubiquitination and weaken p53 protein stability to increase VEGF expression. Furthermore, in a xenograft model using nude mice that stably express active Rac1/Cdc42 protein, active Rac1/Cdc42 decreased p53 levels and increased VEGF expression. Additionally, tumor angiogenesis was inhibited, and p53 protein levels were augmented, by intratumoral injection of the ubiquitin-proteasome inhibitor MG132. Finally in 339 human breast cancer tissues, our analyses indicated that Rac1/Cdc42 expression was related to advanced TNM staging, high proliferation index, ER status, and positive invasive features. In particular, our data suggests that high Rac1/Cdc42 expression is correlated with low wt-p53 and high VEGF expression. We conclude that activated Rac1/Cdc42 is a vascular regulator of tumor angiogenesis and that it may reduce stability of the p53 protein to promote VEGF expression by enhancing p53 protein ubiquitin.
Collapse
Affiliation(s)
- Ji Ma
- Department of Oncology, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shannxi, China
- Department of Breast Surgery, Lanzhou General Hospital of People's Liberation Army, Lanzhou, Gansu, China
| | - Yan Xue
- Department of Oncology, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shannxi, China
- * E-mail:
| | - Wenchao Liu
- Department of Oncology, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shannxi, China
| | - Caixia Yue
- Laboratory of Signal Transduction and Molecular Targeted Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Bi
- Laboratory of Signal Transduction and Molecular Targeted Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junqing Xu
- Department of Radiology, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shannxi, China
| | - Jian Zhang
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, Shannxi, China
| | - Yan Li
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, Shannxi, China
| | - Cuiping Zhong
- Department of Ear Nose Throat Surgery, Lanzhou General Hospital of People's Liberation Army, Lanzhou, Gansu, China
| | - Yan Chen
- Department of Oncology, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shannxi, China
| |
Collapse
|
17
|
Nodularin exposure induces SOD1 phosphorylation and disrupts SOD1 co-localization with actin filaments. Toxins (Basel) 2012; 4:1482-99. [PMID: 23242317 PMCID: PMC3528258 DOI: 10.3390/toxins4121482] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 11/30/2012] [Accepted: 12/06/2012] [Indexed: 01/27/2023] Open
Abstract
Apoptotic cell death is induced in primary hepatocytes by the Ser/Thr protein phosphatase inhibiting cyanobacterial toxin nodularin after only minutes of exposure. Nodularin-induced apoptosis involves a rapid development of reactive oxygen species (ROS), which can be delayed by the Ca2+/calmodulin protein kinase II inhibitor KN93. This apoptosis model provides us with a unique population of highly synchronized dying cells, making it possible to identify low abundant phosphoproteins participating in apoptosis signaling. Here, we show that nodularin induces phosphorylation and possibly also cysteine oxidation of the antioxidant Cu,Zn superoxide dismutase (SOD1), without altering enzymatic SOD1 activity. The observed post-translational modifications of SOD1 could be regulated by Ca2+/calmodulin protein kinase II. In untreated hepatocytes, a high concentration of SOD1 was found in the sub-membranous area, co-localized with the cortical actin cytoskeleton. In the early phase of nodularin exposure, SOD1 was found in high concentration in evenly distributed apoptotic buds. Nodularin induced a rapid reorganization of the actin cytoskeleton and, at the time of polarized budding, SOD1 and actin filaments no longer co-localized.
Collapse
|
18
|
Wimmer R, Cseh B, Maier B, Scherrer K, Baccarini M. Angiogenic sprouting requires the fine tuning of endothelial cell cohesion by the Raf-1/Rok-α complex. Dev Cell 2011; 22:158-71. [PMID: 22209329 PMCID: PMC3268451 DOI: 10.1016/j.devcel.2011.11.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 11/09/2011] [Accepted: 11/28/2011] [Indexed: 11/30/2022]
Abstract
Sprouting angiogenesis, crucial for the development of new blood vessels, is a prime example of collective migration in which endothelial cells migrate as a group joined via cadherin-containing adherens junctions (AJ). The actomyosin apparatus is connected to AJ and generates contractile forces, which, depending on their strength and duration, increase or decrease cell cohesion. Thus, appropriate spatiotemporal control of junctional myosin is critical, but the mechanisms underlying it are incompletely understood. We show that Raf-1 is an essential component of this regulatory network and that its ablation impairs endothelial cell cohesion, sprouting, and tumor-induced angiogenesis. Mechanistically, Raf-1 is recruited to VE-cadherin complexes by a mechanism involving the small G protein Rap1 and is required to bring the Rho effector Rok-α to nascent AJs. This Raf-1-mediated fine tuning of Rok-α signaling allows the activation of junctional myosin and the timely maturation of AJ essential for maintaining cell cohesion during sprouting angiogenesis.
Collapse
Affiliation(s)
- Reiner Wimmer
- Department of Microbiology and Immunobiology, University of Vienna, Max F. Perutz Laboratories, Doktor-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Botond Cseh
- Department of Microbiology and Immunobiology, University of Vienna, Max F. Perutz Laboratories, Doktor-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Barbara Maier
- Department of Microbiology and Immunobiology, University of Vienna, Max F. Perutz Laboratories, Doktor-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Karina Scherrer
- Department of Microbiology and Immunobiology, University of Vienna, Max F. Perutz Laboratories, Doktor-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Manuela Baccarini
- Department of Microbiology and Immunobiology, University of Vienna, Max F. Perutz Laboratories, Doktor-Bohr-Gasse 9, 1030 Vienna, Austria
- Corresponding author
| |
Collapse
|
19
|
Fang X, Ueno M, Yamashita T, Ikuno Y. RhoA Activation and Effect of Rho-kinase Inhibitor in the Development of Retinal Neovascularization in a Mouse Model of Oxygen-induced Retinopathy. Curr Eye Res 2011; 36:1028-36. [DOI: 10.3109/02713683.2011.593110] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
20
|
Li H, Daculsi R, Grellier M, Bareille R, Bourget C, Remy M, Amedee J. The role of vascular actors in two dimensional dialogue of human bone marrow stromal cell and endothelial cell for inducing self-assembled network. PLoS One 2011; 6:e16767. [PMID: 21304816 PMCID: PMC3033416 DOI: 10.1371/journal.pone.0016767] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 01/14/2011] [Indexed: 11/25/2022] Open
Abstract
Angiogenesis is very important for vascularized tissue engineering. In this study, we found that a two-dimensional co-culture of human bone marrow stromal cell (HBMSC) and human umbical vein endothelial cell (HUVEC) is able to stimulate the migration of co-cultured HUVEC and induce self-assembled network formation. During this process, expression of vascular endothelial growth factor (VEGF165) was upregulated in co-cultured HBMSC. Meanwhile, VEGF165-receptor2 (KDR) and urokinase-type plasminogen activator (uPA) were upregulated in co-cultured HUVEC. Functional studies show that neutralization of VEGF165 blocked the migration and the rearrangement of the cells and downregulated the expression of uPA and its receptor. Blocking of vascular endothelial-cadherin (VE-cad) did not affect the migration of co-cultured HUVEC but suppressed the self-assembled network formation. In conclusion, co-cultures upregulated the expression of VEGF165 in co-cultured HBMSC; VEGF165 then activated uPA in co-cultured HUVEC, which might be responsible for initiating the migration and the self-assembled network formation with the participation of VE-cad. All of these results indicated that only the direct contact of HBMSC and HUVEC and their respective dialogue are sufficient to stimulate secretion of soluble factors and to activate molecules that are critical for self-assembled network formation which show a great application potential for vascularization in tissue engineering.
Collapse
Affiliation(s)
- Haiyan Li
- INSERM U577, Bordeaux and University Victor Segalen Bordeaux 2, Bordeaux, France.
| | | | | | | | | | | | | |
Collapse
|
21
|
Active Rac1 improves pathologic VEGF neovessel architecture and reduces vascular leak: mechanistic similarities with angiopoietin-1. Blood 2010; 117:1751-60. [PMID: 21030561 DOI: 10.1182/blood-2010-05-286831] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Architecturally defective, leaky blood vessels typify pathologic angiogenesis induced by vascular endothelial growth factor-A (VEGF-A). Such neovascular defects aggravate disease pathology and seriously compromise the therapeutic utility of VEGF. Endothelial cell (EC) transduction with active L61Rac1 strongly improved VEGF-driven angiogenesis in vivo as measured by increased neovascular density, enhanced lumen formation, and reduced vessel leakiness. Conversely, transduction with dominant-negative N17Rac1 strongly inhibited neovascularization. In vitro, active L61Rac1 promoted organization of cortical actin filaments and vascular cords and improved EC-EC junctions, indicating that improved cytoskeletal dynamics are important to the mechanism by which active L61Rac1 rectifies VEGF-driven angiogenesis. SEW2871, a sphingosine 1-phosphate receptor-1 agonist that activates Rac1 in ECs, improved cord formation and EC-EC junctions in vitro similarly to active L61Rac. Moreover, SEW2871 administration in vivo markedly improved VEGF neovessel architecture and reduced neovascular leak. Angiopoietin-1, a cytokine that "normalizes" VEGF neovessels in vivo, activated Rac1 and improved cord formation and EC-EC junctions in vitro comparably to active L61Rac1, and a specific Rac1 inhibitor blocked these effects. These studies distinguish augmentation of Rac1 activity as a means to rectify the pathologic angioarchitecture and dysfunctionality of VEGF neovessels, and they identify a rational pharmacologic strategy for improving VEGF angiogenesis.
Collapse
|
22
|
Qin L, Zhang M. Maspin regulates endothelial cell adhesion and migration through an integrin signaling pathway. J Biol Chem 2010; 285:32360-9. [PMID: 20713357 DOI: 10.1074/jbc.m110.131045] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Maspin has been identified as a potent angiogenesis inhibitor. However, the molecular mechanism responsible for its anti-angiogenic property is unclear. In this study, we examined the effect of maspin on endothelial cell (EC) adhesion and migration in a cell culture system. We found that maspin was expressed in blood vessels ECs and human umbilical vein endothelial cells (HUVECs). Maspin significantly enhanced HUVEC cell adhesion to various matrix proteins. This effect was dependent on the activation of integrin β(1), which subsequently led to distribution pattern changes of vinculin and F-actin. These results indicated that maspin affects cell adhesion and cytoskeleton reorganization through an integrin signal transduction pathway. Analysis of HUVECs following maspin treatment revealed increased integrin-linked kinase activities and phosphorylated FAK levels, consistent with increased cell adhesion. Interestingly, when HUVECs were induced to migrate by migration stimulatory factor bFGF, active Rac1 and cdc42 small GTPase levels were decreased dramatically at 30 min following maspin treatment. Using phosphorylated FAK at Tyr(397) as an indicator of focal adhesion disassembly, maspin-treated HUVECs had elevated FAK phosphorylation compared with the mock treated control. The results were a reduction in focal adhesion disassembly and the retardation in EC migration. This study uncovers a mechanism by which maspin exerts its effect on EC adhesion and migration through an integrin signal transduction pathway.
Collapse
Affiliation(s)
- Li Qin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | |
Collapse
|
23
|
Otranto M, Souza-Netto I, Aguila MB, Monte-Alto-Costa A. Male and female rats with severe protein restriction present delayed wound healing. Appl Physiol Nutr Metab 2010; 34:1023-31. [PMID: 20029510 DOI: 10.1139/h09-100] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Malnutrition remains a significant problem, not only in developing countries, but also in the developed world. The aim of this study was to investigate the effects of protein restriction on rat excisional cutaneous healing. Male and female rats (12 weeks old) were exposed to different degrees of protein restriction (23%, 12% (slight restriction), and 0% (severe restriction)) for 12 weeks. On week 9, a full-thickness excisional skin lesion was performed, and the lesion area was measured to evaluate wound contraction and re-epithelialization. Euthanasia was performed after 12 weeks, and the lesion and adjacent skin were removed, fixed in formalin, and embedded in paraffin. Sections were stained with hematoxylin-eosin, toluidine blue, picro-mallory, and sirius red, and were immunostained for alpha-smooth muscle actin. Animals (males and females) exposed to severe protein restriction (0% protein) presented impairment of wound contraction. Inflammatory cells were present in higher amounts in the protein-restricted groups than in the 23% group. Extracellular matrix was poorly deposited in the severely restricted group (0%), but only mildly disturbed in the slightly restricted group (12%). Neovascularization was disturbed in both restricted groups. Our study demonstrates that animals exposed to slight protein restriction present disturbed wound healing, but animals exposed to severe protein restriction present impaired wound healing.
Collapse
Affiliation(s)
- Marcela Otranto
- Histology and Embryology Departament, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | | | | | | |
Collapse
|
24
|
D'Amico G, Robinson SD, Germain M, Reynolds LE, Thomas GJ, Elia G, Saunders G, Fruttiger M, Tybulewicz V, Mavria G, Hodivala-Dilke KM. Endothelial-Rac1 is not required for tumor angiogenesis unless alphavbeta3-integrin is absent. PLoS One 2010; 5:e9766. [PMID: 20339539 PMCID: PMC2842301 DOI: 10.1371/journal.pone.0009766] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 03/01/2010] [Indexed: 12/25/2022] Open
Abstract
Endothelial cell migration is an essential aspect of tumor angiogenesis. Rac1 activity is needed for cell migration in vitro implying a requirement for this molecule in angiogenesis in vivo. However, a precise role for Rac1 in tumor angiogenesis has never been addressed. Here we show that depletion of endothelial Rac1 expression in adult mice, unexpectedly, has no effect on tumor growth or tumor angiogenesis. In addition, repression of Rac1 expression does not inhibit VEGF-mediated angiogenesis in vivo or ex vivo, nor does it affect chemotactic migratory responses to VEGF in 3-dimensions. In contrast, the requirement for Rac1 in tumor growth and angiogenesis becomes important when endothelial beta3-integrin levels are reduced or absent: the enhanced tumor growth, tumor angiogenesis and VEGF-mediated responses in beta3-null mice are all Rac1-dependent. These data indicate that in the presence of alphavbeta3-integrin Rac1 is not required for tumor angiogenesis.
Collapse
Affiliation(s)
- Gabriela D'Amico
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - Stephen D. Robinson
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - Mitchel Germain
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - Louise E. Reynolds
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - Gareth J. Thomas
- Centre for Tumour Biology, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - George Elia
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - Garry Saunders
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Victor Tybulewicz
- Division of Immune Cell Biology, National Institute for Medical Research, London, United Kingdom
| | - Georgia Mavria
- Cancer Research UK Centre for Cell and Molecular Biology, Institute of Cancer Research, London, United Kingdom
| | - Kairbaan M. Hodivala-Dilke
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| |
Collapse
|
25
|
Wu PK, Ringeisen BR. Development of human umbilical vein endothelial cell (HUVEC) and human umbilical vein smooth muscle cell (HUVSMC) branch/stem structures on hydrogel layers via biological laser printing (BioLP). Biofabrication 2010; 2:014111. [DOI: 10.1088/1758-5082/2/1/014111] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
26
|
D'Amico G, Jones DT, Nye E, Sapienza K, Ramjuan AR, Reynolds LE, Robinson SD, Kostourou V, Martinez D, Aubyn D, Grose R, Thomas GJ, Spencer-Dene B, Zicha D, Davies D, Tybulewicz V, Hodivala-Dilke KM. Regulation of lymphatic-blood vessel separation by endothelial Rac1. Development 2009; 136:4043-53. [PMID: 19906871 PMCID: PMC2778747 DOI: 10.1242/dev.035014] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2009] [Indexed: 12/29/2022]
Abstract
Sprouting angiogenesis and lymphatic-blood vessel segregation both involve the migration of endothelial cells, but the precise migratory molecules that govern the decision of blood vascular endothelial cells to segregate into lymphatic vasculature are unknown. Here, we deleted endothelial Rac1 in mice (Tie1-Cre(+);Rac1(fl/fl)) and revealed, unexpectedly, that whereas blood vessel morphology appeared normal, lymphatic-blood vessel separation was impaired, with corresponding edema, haemorrhage and embryonic lethality. Importantly, normal levels of Rac1 were essential for directed endothelial cell migratory responses to lymphatic-inductive signals. Our studies identify Rac1 as a crucial part of the migratory machinery required for endothelial cells to separate and form lymphatic vasculature.
Collapse
MESH Headings
- Animals
- Blood Vessels/metabolism
- Cell Separation/methods
- Cells, Cultured
- Embryo, Mammalian
- Endothelial Cells/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/embryology
- Endothelium, Vascular/metabolism
- Fluorescent Antibody Technique, Direct
- Fluorescent Dyes/metabolism
- Galactosides/metabolism
- Gene Deletion
- Gene Expression Regulation, Developmental
- Immunohistochemistry
- Indoles/metabolism
- Lymphatic Vessels/metabolism
- Mice
- Mice, Transgenic
- Neovascularization, Physiologic/genetics
- Neovascularization, Physiologic/physiology
- RNA, Small Interfering/metabolism
- Receptor, TIE-2/genetics
- Receptor, TIE-2/metabolism
- Transfection
- beta-Galactosidase/metabolism
- rac1 GTP-Binding Protein/analysis
- rac1 GTP-Binding Protein/genetics
- rac1 GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Gabriela D'Amico
- Adhesion and Angiogenesis Laboratory, Institute of Cancer and Cancer Research UK, Bart's & The London Queen Mary's School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Dylan T. Jones
- Adhesion and Angiogenesis Laboratory, Institute of Cancer and Cancer Research UK, Bart's & The London Queen Mary's School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Emma Nye
- Experimental Histopathology Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3PX, UK
| | - Karen Sapienza
- Centre for Tumour Biology, Institute of Cancer and Cancer Research UK, Bart's & The London Queen Mary's School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Antoine R. Ramjuan
- Adhesion and Angiogenesis Laboratory, Institute of Cancer and Cancer Research UK, Bart's & The London Queen Mary's School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Louise E. Reynolds
- Adhesion and Angiogenesis Laboratory, Institute of Cancer and Cancer Research UK, Bart's & The London Queen Mary's School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Stephen D. Robinson
- Adhesion and Angiogenesis Laboratory, Institute of Cancer and Cancer Research UK, Bart's & The London Queen Mary's School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Vassiliki Kostourou
- Adhesion and Angiogenesis Laboratory, Institute of Cancer and Cancer Research UK, Bart's & The London Queen Mary's School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
- BSRC Alexander Fleming, 34 Fleming street, 166 72 Vari, Athens, Greece
| | - Dolores Martinez
- Fluorescence Activated Cell Sorting Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3PX, UK
| | - Deborah Aubyn
- Light Microscopy Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3PX, UK
| | - Richard Grose
- Growth Factor Signalling Laboratory, Institute of Cancer and Cancer Research UK, Bart's & The London Queen Mary's School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Gareth J. Thomas
- Centre for Tumour Biology, Institute of Cancer and Cancer Research UK, Bart's & The London Queen Mary's School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Bradley Spencer-Dene
- Experimental Histopathology Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3PX, UK
- Histopathology, Imperial College London, London, W12 0NN, UK
| | - Daniel Zicha
- Light Microscopy Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3PX, UK
| | - Derek Davies
- Fluorescence Activated Cell Sorting Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3PX, UK
| | - Victor Tybulewicz
- Division of Immune Cell Biology, National Institute for Medical Research, Mill Hill, London, NW7 1AA, UK
| | - Kairbaan M. Hodivala-Dilke
- Adhesion and Angiogenesis Laboratory, Institute of Cancer and Cancer Research UK, Bart's & The London Queen Mary's School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| |
Collapse
|
27
|
Herbert JMJ, Buffa FM, Vorschmitt H, Egginton S, Bicknell R. A new procedure for determining the genetic basis of a physiological process in a non-model species, illustrated by cold induced angiogenesis in the carp. BMC Genomics 2009; 10:490. [PMID: 19852815 PMCID: PMC2771047 DOI: 10.1186/1471-2164-10-490] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Accepted: 10/23/2009] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Physiological processes occur in many species for which there is yet no sequenced genome and for which we would like to identify the genetic basis. For example, some species increase their vascular network to minimise the effects of reduced oxygen diffusion and increased blood viscosity associated with low temperatures. Since many angiogenic and endothelial genes have been discovered in man, functional homolog relationships between carp, zebrafish and human were used to predict the genetic basis of cold-induced angiogenesis in Cyprinus Carpio (carp). In this work, carp sequences were collected and built into contigs. Human-carp functional homolog relationships were derived via zebrafish using a new Conditional Stepped Reciprocal Best Hit (CSRBH) protocol. Data sources including publications, Gene Ontology and cDNA libraries were then used to predict the identity of known or potential angiogenic genes. Finally, re-analyses of cold carp microarray data identified carp genes up-regulated in response to low temperatures in heart and muscle. RESULTS The CSRBH approach outperformed all other methods and attained 8,726 carp to human functional homolog relationships for 16,650 contiguous sequences. This represented 3,762 non-redundant genes and 908 of them were predicted to have a role in angiogenesis. The total number of up-regulated differentially expressed genes was 698 and 171 of them were putatively angiogenic. Of these, 5 genes representing the functional homologs NCL, RHOA, MMP9, GRN and MAPK1 are angiogenesis-related genes expressed in response to low temperature. CONCLUSION We show that CSRBH functional homologs relationships and re-analyses of gene expression data can be combined in a non-model species to predict genes of biological interest before a genome sequence is fully available. Programs to run these analyses locally are available from http://www.cbrg.ox.ac.uk/~jherbert/.
Collapse
Affiliation(s)
- John M J Herbert
- Cancer Research UK Angiogenesis Group, Institute for Biomedical Research, Schools of Immunity and Infection and Cancer studies, College of Medicine and Dentistry, University of Birmingham, Birmingham, B15 2TT, UK.
| | | | | | | | | |
Collapse
|
28
|
Vallon M, Rohde F, Janssen KP, Essler M. Tumor endothelial marker 5 expression in endothelial cells during capillary morphogenesis is induced by the small GTPase Rac and mediates contact inhibition of cell proliferation. Exp Cell Res 2009; 316:412-21. [PMID: 19853600 DOI: 10.1016/j.yexcr.2009.10.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 10/09/2009] [Accepted: 10/16/2009] [Indexed: 12/21/2022]
Abstract
Tumor endothelial marker (TEM) 5 is an adhesion G-protein-coupled receptor upregulated in endothelial cells during tumor and physiologic angiogenesis. So far, the mechanisms leading to upregulation of TEM5 and its function during angiogenesis have not been identified. Here, we report that TEM5 expression in endothelial cells is induced during capillary-like network formation on Matrigel, during capillary morphogenesis in a three-dimensional collagen I matrix, and upon confluence on a two-dimensional matrix. TEM5 expression was not induced by a variety of soluble angiogenic factors, including VEGF and bFGF, in subconfluent endothelial cells. TEM5 upregulation was blocked by toxin B from Clostridium difficile, an inhibitor of the small GTPases Rho, Rac, and Cdc42. The Rho inhibitor C3 transferase from Clostridium botulinum did not affect TEM5 expression, whereas the Rac inhibitor NSC23766 suppressed TEM5 upregulation. An excess of the soluble TEM5 extracellular domain or an inhibitory monoclonal TEM5 antibody blocked contact inhibition of endothelial cell proliferation resulting in multilayered islands within the endothelial monolayer and increased vessel density during capillary formation. Based on our results we conclude that TEM5 expression during capillary morphogenesis is induced by the small GTPase Rac and mediates contact inhibition of proliferation in endothelial cells.
Collapse
Affiliation(s)
- Mario Vallon
- Nuklearmedizinische Klinik und Poliklinik, Technische Universität München, Ismaninger Strasse 22, 81675 Munich, Germany.
| | | | | | | |
Collapse
|
29
|
Yokomori H, Oda M, Yoshimura K, Nagai T, Fujimaki K, Watanabe S, Hibi T. Caveolin-1 and Rac regulate endothelial capillary-like tubular formation and fenestral contraction in sinusoidal endothelial cells. Liver Int 2009; 29:266-76. [PMID: 19067793 DOI: 10.1111/j.1478-3231.2008.01891.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
BACKGROUND/AIMS Rho guanidine triphosphatases (GTPases) are major regulators of cell migration. We investigated the cytoskeleton and Rho GTPases during cell migration and morphogenesis processes in isolated rat liver sinusoidal endothelial cells (LSECs) cultured on Matrigel while stimulated by the vascular endothelial growth factor (VEGF). METHODS To obtain primary monolayers, LSECs were cultured on Matrigel for 5-17 h with or without VEGF. Sinusoidal endothelial fenestrae (SEF) morphology was observed using scanning electron microscopy and transmission electron microscopy. RhoA, Rac1 and phosphorylated myosin light-chain kinase, Rho-binding domain of Rhotekin and the p21-binding domain of p21-activated protein kinase were analysed using Western blotting. RESULTS The LSECs showed cellular protrusions and or cords of aligned cells resembling primitive capillary-like structures, with SEF contraction. Time course analyses of Rac1 activation matched specific morphological changes. Rac1 activity increased progressively to 17 h in cells cultured without VEGF, but markedly increased at 7 h in the presence of VEGF. RhoA activity was slightly elevated at 5 h. The levels of endogenous caveolin-1 (CAV-1) expression increased in a time-dependent manner, reaching a peak at 7 h. CAV-1 expression occurred immediately before the formation of the capillary-like tube. Moreover, treatment with VEGF regulated CAV-1 expression in LSECs. CONCLUSIONS Spatial activation of Rac1 is involved in the formation of a capillary-like tubular network accompanying SEF contraction in LSECs, implying that endothelial migration and adhesion are necessary for LSECs tubular formation in the liver. CAV-1 might play an important positive role in the regulation of LSEC tubular formation.
Collapse
Affiliation(s)
- Hiroaki Yokomori
- Department of Internal Medicine, Kitasato Institute Medical Center Hospital, Kitasato University, Saitama, Japan.
| | | | | | | | | | | | | |
Collapse
|
30
|
Navarro A, Perez RE, Rezaiekhaligh M, Mabry SM, Ekekezie II. T1alpha/podoplanin is essential for capillary morphogenesis in lymphatic endothelial cells. Am J Physiol Lung Cell Mol Physiol 2008; 295:L543-51. [PMID: 18658274 DOI: 10.1152/ajplung.90262.2008] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The lymphatic vasculature functions to maintain tissue perfusion homeostasis. Defects in its formation or disruption of the vessels result in lymphedema, the effective treatment of which is hampered by limited understanding of factors regulating lymph vessel formation. Mice lacking T1alpha/podoplanin, a lymphatic endothelial cell transmembrane protein, have malformed lymphatic vasculature with lymphedema at birth, but the molecular mechanism for this phenotype is unknown. Here, we show, using primary human lung microvascular lymphatic endothelial cells (HMVEC-LLy), that small interfering RNA-mediated silence of podoplanin gene expression has the dramatic effect of blocking capillary tube formation in Matrigel. In addition, localization of phosphorylated ezrin/radixin/moesin proteins to plasma membrane extensions, an early event in the capillary morphogenic program in lymphatic endothelial cells, is impaired. We find that cells with decreased podoplanin expression fail to properly activate the small GTPase RhoA early (by 30 min) after plating on Matrigel, and Rac1 shows a delay in its activation. Further indication that podoplanin action is linked to RhoA activation is that use of a cell-permeable inhibitor of Rho inhibited lymphatic endothelial capillary tube formation in the same manner as did podoplanin gene silencing, which was not mimicked by treatment with a Rac1 inhibitor. These data clearly demonstrate that early activation of RhoA in the lymphangiogenic process, which is required for the successful establishment of the capillary network, is dependent on podoplanin expression. To our knowledge, this is the first time that a mechanism has been suggested to explain the role of podoplanin in lymphangiogenesis.
Collapse
Affiliation(s)
- Angels Navarro
- Section of Neonatology, Children's Mercy Hospitals & Clinics, Kansas City, MO 64108, USA
| | | | | | | | | |
Collapse
|
31
|
Flaiz C, Utermark T, Parkinson DB, Poetsch A, Hanemann CO. Impaired intercellular adhesion and immature adherens junctions in merlin-deficient human primary schwannoma cells. Glia 2008; 56:506-15. [PMID: 18240308 DOI: 10.1002/glia.20629] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Schwannomas that occur spontaneously or in patients with neurofibromatosis Type 2, lack both alleles for the tumor suppressor and plasma membrane-cytoskeleton linker merlin. We have shown that human primary schwannoma cells display activation of the RhoGTPases Rac1 and Cdc42 which results in highly dynamic and ongoing protrusive activity like ruffling. Ruffling is an initial and temporally limited step in the formation of intercellular contacts like adherens junctions that are based on the cadherin-catenin system. We tested if there is a connection between Rac1-induced ongoing ruffling and the maintenance, stabilization and functionality of adherens junctions and if this is of relevance in human, merlin-deficient schwannoma cells. We show intense ongoing ruffling is not limited to membranes of single human primary schwannoma cells, but occurs also in membranes of contacting cells, even when confluent. Live cell imaging shows that newly formed contacts are released after a short time, suggesting disturbed formation or stabilization of adherens junctions. Morphology, high phospho-tyrosine levels and cortactin staining indicate that adherens junctions are immature in human primary schwannoma cells, whereas they display characteristics of mature adherens junctions in human primary Schwann cells. When merlin is reintroduced, human primary schwannoma cells show only initial ruffling in contacting cells and adherens junctions appear more mature. We therefore propose that ongoing Rac-induced ruffling causes immature adherens junctions and leads to impaired, nonfunctional intercellular adhesion in aggregation assays in merlin-deficient schwannoma cells that could be an explanation for increased proliferation rates due to loss of contact inhibition or tumor development in general.
Collapse
Affiliation(s)
- C Flaiz
- Department of Clinical Neurobiology, Institute of Biomedical and Clinical Science, Peninsula College for Medicine and Dentistry, Plymouth, UK
| | | | | | | | | |
Collapse
|
32
|
Hata Y, Miura M, Nakao S, Kawahara S, Kita T, Ishibashi T. Antiangiogenic properties of fasudil, a potent Rho-Kinase inhibitor. Jpn J Ophthalmol 2008; 52:16-23. [PMID: 18369695 DOI: 10.1007/s10384-007-0487-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Accepted: 09/15/2007] [Indexed: 11/30/2022]
Abstract
PURPOSE Vascular endothelial growth factor (VEGF) plays a pivotal role in pathological angiogenesis. In this study, we addressed the therapeutic potential of fasudil, a potent Rho-kinase inhibitor, for VEGF-elicited angiogenesis and also for the intracellular signalings induced by VEGF. METHODS In vitro, the inhibitory effects of fasudil on the VEGF-dependent VEGF receptor 2 (VEFGR2 or KDR), extracellular signal-related kinase (ERK) 1/2, Akt and myosin light chain (MLC) phosphorylation, as well as on the migration and proliferation of bovine retinal microvascular endothelial cells (BRECs) were analyzed with Western blotting, [3H]-thymidine uptake, and modified Boyden chamber assay. VEGF-elicited in vivo angiogenesis was analyzed with a mouse corneal micropocket assay coembedded with or without fasudil. RESULTS VEGF caused enhanced MLC phosphorylation of BRECs, which was almost completely attenuated by 10microM fasudil. VEGF-dependent phosphorylation of ERK1/2 and Akt were also partially but significantly attenuated by treatment with fasudil without affecting VEGFR2 (KDR) phosphorylation. Moreover, both VEGF-induced [3H]-thymidine uptake and the migration of BRECs were significantly inhibited in the presence of fasudil. Finally, VEGF-elicited angiogenesis in the corneal micropocket assay was potently attenuated by coembedding with fasudil (P < 0.01). CONCLUSIONS These findings indicate that fasudil might have a therapeutic potential for ocular angiogenic diseases. The antiangiogenic effect of fasudil appears to be mediated through the blockade not only of Rho-kinase signaling but also of ERK and Akt signaling.
Collapse
Affiliation(s)
- Yasuaki Hata
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | | | | | | | | | | |
Collapse
|
33
|
Sayed KAE, Khanfar MA, Shallal HM, Muralidharan A, Awate B, Youssef DTA, Liu Y, Zhou YD, Nagle DG, Shah G. Latrunculin A and its C-17-O-carbamates inhibit prostate tumor cell invasion and HIF-1 activation in breast tumor cells. JOURNAL OF NATURAL PRODUCTS 2008; 71:396-402. [PMID: 18298079 PMCID: PMC2930178 DOI: 10.1021/np070587w] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The marine-derived macrolides latrunculins A ( 1) and B, from the Red Sea sponge Negombata magnifica, have been found to reversibly bind actin monomers, forming a 1:1 complex with G-actin and disrupting its polymerization. The microfilament protein actin is responsible for several essential functions within the cell such as cytokinesis and cell migration. One of the main binding pharmacophores of 1 to G-actin was identified as the C-17 lactol hydroxyl moiety that binds arginine 210 NH. Latrunculin A-17- O-carbamates 2- 6 were prepared by reaction with the corresponding isocyanates. Latrunculin A ( 1) and carbamates 4- 6 displayed potent anti-invasive activity against the human highly metastatic human prostate cancer PC-3M cells in a Matrigel assay at a concentration range of 50 nM to 1 microM. Latrunculin A ( 1, 500 nM) decreased the disaggregation and cell migration of PC-3M-CT+ spheroids by 3-fold. Carbamates 4 and 5 were 2.5- and 5-fold more active than 1, respectively, in this assay with less actin binding affinity. Latrunculin A ( 1, IC 50 6.7 microM) and its 17- O-[ N-(benzyl)carbamate ( 6, IC 50 29 microM) suppress hypoxia-induced HIF-1 activation in T47D breast tumor cells.
Collapse
Affiliation(s)
- Khalid A El Sayed
- Department of Basic Pharmaceutical Sciences, University of Louisana, Monroe, Louisana 71209, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kanthou C, Tozer GM. Tumour targeting by microtubule-depolymerising vascular disrupting agents. Expert Opin Ther Targets 2007; 11:1443-57. [DOI: 10.1517/14728222.11.11.1443] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
35
|
Chang F, Lemmon CA, Park D, Romer LH. FAK potentiates Rac1 activation and localization to matrix adhesion sites: a role for betaPIX. Mol Biol Cell 2006; 18:253-64. [PMID: 17093062 PMCID: PMC1751318 DOI: 10.1091/mbc.e06-03-0207] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
FAK, a cytoplasmic protein tyrosine kinase, is activated and localized to focal adhesions upon cell attachment to extracellular matrix. FAK null cells spread poorly and exhibit altered focal adhesion turnover. Rac1 is a member of the Rho-family GTPases that promotes membrane ruffling, leading edge extension, and cell spreading. We investigated the activation and subcellular location of Rac1 in FAK null and FAK reexpressing fibroblasts. FAK reexpressers had a more robust pattern of Rac1 activation after cell adhesion to fibronectin than the FAK null cells. Translocation of Rac1 to focal adhesions was observed in FAK reexpressers, but seldom in FAK null cells. Experiments with constitutively active L61Rac1 and dominant negative N17Rac1 indicated that the activation state of Rac1 regulated its localization to focal adhesions. We demonstrated that FAK tyrosine-phosphorylated betaPIX and thereby increased its binding to Rac1. In addition, betaPIX facilitated the targeting of activated Rac1 to focal adhesions and the efficiency of cell spreading. These data indicate that FAK has a role in the activation and focal adhesion translocation of Rac1 through the tyrosine phosphorylation of betaPIX.
Collapse
Affiliation(s)
- Fumin Chang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | |
Collapse
|
36
|
Varon C, Basoni C, Reuzeau E, Moreau V, Kramer IJ, Génot E. TGFbeta1-induced aortic endothelial morphogenesis requires signaling by small GTPases Rac1 and RhoA. Exp Cell Res 2006; 312:3604-19. [PMID: 16978608 DOI: 10.1016/j.yexcr.2006.08.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2006] [Revised: 08/03/2006] [Accepted: 08/05/2006] [Indexed: 12/14/2022]
Abstract
TGFbeta is a potent regulator of cell differentiation in many cell types. On aortic endothelial cells, TGFbeta1 displays angiogenic properties in inducing capillary-like tube formation in collagen I gels, in vitro. We investigated cytoskeletal changes that precede tube formation and related these alterations to the effects of TGFbeta1 on the activation state of members of the RhoGTPase family. TGFbeta1 promotes cell elongation and stress fiber formation in aortic endothelial cells. Using cell lines with inducible expression of Rac1 mutants, we show that these events are mimicked by expression of dominant-negative Rac1 whereas the constitutively active mutant prevents the TGFbeta1-mediated change of phenotype. Although TGFbeta1 induces an initial rise in the Rac1-GTP content, this phase is followed by a prolonged loss of the active form. In contrast, RhoA activity increases progressively and reaches a plateau when Rac1-GTP is no longer detectable. Prolonged inhibition of Rac1 appears necessary and sufficient for the increase in RhoA-GTP. In situ examination of Rho activity in TGFbeta1-treated cells provides evidence that active RhoA relocalizes to the tips of elongated cells. Inhibiting the Rho effector ROCK abrogates tube formation. Thus, Rac1 and RhoA are regulated by TGFbeta1 in the process of endothelial tube formation in collagen I gels.
Collapse
Affiliation(s)
- Christine Varon
- European Institute of Chemistry and Biology, University of Bordeaux I and Unité INSERM 441, Université Victor Segalen Bordeaux 2, Bordeaux, 2, rue Robert Escarpit, 33 600 Pessac, France
| | | | | | | | | | | |
Collapse
|
37
|
Singleton PA, Lingen MW, Fekete MJ, Garcia JGN, Moss J. Methylnaltrexone inhibits opiate and VEGF-induced angiogenesis: role of receptor transactivation. Microvasc Res 2006; 72:3-11. [PMID: 16820176 DOI: 10.1016/j.mvr.2006.04.004] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2006] [Revised: 04/18/2006] [Accepted: 04/19/2006] [Indexed: 11/22/2022]
Abstract
Angiogenesis or the formation of new blood vessels is important in the growth and metastatic potential of various cancers. Therefore, agents that inhibit angiogenesis have important therapeutic implications in numerous malignancies. We examined the effects of methylnaltrexone (MNTX), a peripheral mu opioid receptor antagonist, on agonist-induced human EC proliferation and migration, two key components in angiogenesis. Using human dermal microvascular EC, we observed that morphine sulfate (MS), the active metabolite, morphine-6-glucuronide (M6G), DAMGO ([d-Ala(2), N-Me-Phe(4), Gly(5)-ol]enkaphalin) and VEGF induced migration which were inhibited by pretreatment with MNTX at therapeutically relevant concentration (0.1 microM). The biologically inactive metabolite morphine-3-glucuronide (M3G) did not affect EC migration. We next examined the mechanism(s) by which MNTX inhibits opioid and VEGF-induced angiogenesis using human pulmonary microvascular EC. MS and DAMGO induced Src activation which was required for VEGF receptor transactivation and opioid-induced EC proliferation and migration. MNTX inhibited MS, DAMGO and VEGF induced tyrosine phosphorylation (transactivation) of VEGF receptors 1 and 2. Furthermore, MS, DAMGO and VEGF induced RhoA activation which was inhibited by MNTX or VEGF receptor tyrosine kinase inhibition. Finally, MNTX or silencing RhoA expression (siRNA) blocked MS, DAMGO and VEGF-induced EC proliferation and migration. Taken together, these results indicate that MNTX inhibits opioid-induced EC proliferation and migration via inhibition of VEGF receptor phosphorylation/transactivation with subsequent inhibition of RhoA activation. These results suggest that MNTX inhibition of angiogenesis can be a useful therapeutic intervention for cancer treatment.
Collapse
Affiliation(s)
- P A Singleton
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | |
Collapse
|
38
|
Loutrari H, Magkouta S, Pyriochou A, Koika V, Kolisis FN, Papapetropoulos A, Roussos C. Mastic Oil from Pistacia lentiscus var. chia Inhibits Growth and Survival of Human K562 Leukemia Cells and Attenuates Angiogenesis. Nutr Cancer 2006; 55:86-93. [PMID: 16965245 DOI: 10.1207/s15327914nc5501_11] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Mastic oil from Pistacia lentiscus var. chia, a natural plant extract traditionally used as a food additive, has been extensively studied for its antimicrobial activity attributed to the combination of its bioactive components. One of them, perillyl alcohol (POH), displays tumor chemopreventive, chemotherapeutic, and antiangiogenic properties. We investigated whether mastic oil would also suppress tumor cell growth and angiogenesis. We observed that mastic oil concentration and time dependently exerted an antiproliferative and proapoptotic effect on K562 human leukemia cells and inhibited the release of vascular endothelial growth factor (VEGF) from K562 and B16 mouse melanoma cells. Moreover, mastic oil caused a concentration-dependent inhibition of endothelial cell (EC) proliferation without affecting cell survival and a significant decrease of microvessel formation both in vitro and in vivo. Investigation of underlying mechanism(s) demonstrated that mastic oil reduced 1) in K562 cells the activation of extracellular signal-regulated kinases 1/2 (Erk1/2) known to control leukemia cell proliferation, survival, and VEGF secretion and 2) in EC the activation of RhoA, an essential regulator of neovessel organization. Overall, our results underscore that mastic oil, through its multiple effects on malignant cells and ECs, may be a useful natural dietary supplement for cancer prevention.
Collapse
Affiliation(s)
- Heleni Loutrari
- G.P. Livanos andM. Simou Laboratories, Evangelismos Hospital, Department of Critical Careand Pulmonary Services, Medical School, University of Athens, 10675 Athens, Greece.
| | | | | | | | | | | | | |
Collapse
|
39
|
Xue Y, Bi F, Zhang X, Zhang S, Pan Y, Liu N, Shi Y, Yao X, Zheng Y, Fan D. Role of Rac1 and Cdc42 in hypoxia induced p53 and von Hippel-Lindau suppression and HIF1alpha activation. Int J Cancer 2006; 118:2965-72. [PMID: 16395716 DOI: 10.1002/ijc.21763] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Low oxygen tension can influence tumor progression by enhancing angiogenesis, a process that may involve Rho GTPases whose activities have been implicated in tumorigenesis and metastasis. In the present study, we show that hypoxia can increase the mRNA levels and intracellular activities of Rac1 and Cdc42 in a time-dependent manner. The hypoxia-stimulated activities of Rac1 and Cdc42 could be blocked by the phosphatidylinositol 3'-kinase (PI3K) inhibitor LY294002 and the protein tyrosine kinase (PTK) inhibitor genistein but were not affected by the p38MAPK inhibitor SB203580 or the MEK-1 inhibitor PD98059, suggesting that the hypoxia-mediated signals were through PI3K and PTK. Correlating with the increased activities of Rac1 and Cdc42, the expression of the pro-angiogenesis factors HIF-1alpha and vascular endothelial growth factor (VEGF) was upregulated by hypoxia, whereas the expression of the tumor suppressors von Hippel-Lindau and p53 was down-regulated. Dominant negative N17Rac1 and N17Cdc42 could upregulate the expression of p53 and pVHL but downregulate that of HIF-1alpha and VEGF under hypoxia. Furthermore, the preconditioned medium from N17Rac1 or N17Cdc42-expressing gastric cancer cells was able to inhibit the proliferation of HUVECs. Our results indicate that PI3K and PTK-mediated activations of Rac1 and Cdc42 are involved in the hypoxia-induced production of angiogenesis-promoting factors and tumor suppressors, and suggest that the Rho family GTPases Rac1 and Cdc42 may contribute to the hypoxia-mediated angiogenesis.
Collapse
Affiliation(s)
- Yan Xue
- The State Key Laboratory of Cancer Biology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Singleton PA, Dudek SM, Chiang ET, Garcia JGN. Regulation of sphingosine 1-phosphate-induced endothelial cytoskeletal rearrangement and barrier enhancement by S1P1 receptor, PI3 kinase, Tiam1/Rac1, and alpha-actinin. FASEB J 2006; 19:1646-56. [PMID: 16195373 DOI: 10.1096/fj.05-3928com] [Citation(s) in RCA: 229] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelial cell (EC) barrier dysfunction results in increased vascular permeability observed in inflammation, tumor angiogenesis, and atherosclerosis. The platelet-derived phospholipid sphingosine-1-phosphate (S1P) decreases EC permeability in vitro and in vivo and thus has obvious therapeutic potential. We examined S1P-mediated human pulmonary artery EC signaling and barrier regulation in caveolin-enriched microdomains (CEM). Immunoblotting from S1P-treated EC revealed S1P-mediated rapid recruitment (1 microM, 5 min) to CEMs of the S1P receptors S1P1 and S1P3, p110 PI3 kinase alpha and beta catalytic subunits, the Rac1 GEF, Tiam1, and alpha-actinin isoforms 1 and 4. Immunoprecipitated p110 PI3 kinase catalytic subunits from S1P-treated EC exhibited PIP3 production in CEMs. Immunoprecipitation of S1P receptors from CEM fractions revealed complexes containing Tiam1 and S1P1. PI3 kinase inhibition (LY294002) attenuated S1P-induced Tiam1 association with S1P1, Tiam1/Rac1 activation, alpha-actinin-1/4 recruitment, and EC barrier enhancement. Silencing of either S1P1 or Tiam1 expression resulted in the loss of S1P-mediated Rac1 activation and alpha-actinin-1/4 recruitment to CEM. Finally, silencing S1P1, Tiam1, or both alpha-actinin isoforms 1/4 inhibits S1P-induced cortical F-actin rearrangement and S1P-mediated barrier enhancement. Taken together, these results suggest that S1P-induced recruitment of S1P1 to CEM fractions promotes PI3 kinase-mediated Tiam1/Rac1 activation required for alpha-actinin-1/4-regulated cortical actin rearrangement and EC barrier enhancement.
Collapse
MESH Headings
- Actinin/metabolism
- Actinin/physiology
- Catalytic Domain
- Caveolin 1/chemistry
- Cells, Cultured
- Cholesterol/chemistry
- Chromones/pharmacology
- Cytoskeleton/metabolism
- Electrophoresis, Polyacrylamide Gel
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Enzyme Inhibitors/pharmacology
- Gene Expression Regulation, Enzymologic
- Guanine Nucleotide Exchange Factors/metabolism
- Guanine Nucleotide Exchange Factors/physiology
- Humans
- Immunoblotting
- Immunoprecipitation
- Inflammation
- Microfilament Proteins/metabolism
- Microscopy, Fluorescence
- Models, Biological
- Morpholines/pharmacology
- Neoplasm Proteins/physiology
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphatidylinositol 3-Kinases/physiology
- Protein Isoforms
- Protein Structure, Tertiary
- Pulmonary Artery/pathology
- RNA, Small Interfering/metabolism
- Receptors, Lysosphingolipid/physiology
- Signal Transduction
- T-Lymphoma Invasion and Metastasis-inducing Protein 1
- Transfection
- rac1 GTP-Binding Protein/physiology
Collapse
Affiliation(s)
- Patrick A Singleton
- Division of Pulmonary and Critical Care Medicine, Center for Translational Respiratory Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|
41
|
Mavria G, Vercoulen Y, Yeo M, Paterson H, Karasarides M, Marais R, Bird D, Marshall CJ. ERK-MAPK signaling opposes Rho-kinase to promote endothelial cell survival and sprouting during angiogenesis. Cancer Cell 2006; 9:33-44. [PMID: 16413470 DOI: 10.1016/j.ccr.2005.12.021] [Citation(s) in RCA: 254] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2004] [Revised: 09/21/2005] [Accepted: 12/19/2005] [Indexed: 12/30/2022]
Abstract
Inhibition of ERK-MAPK signaling by expression of dominant-negative MEK1 in the tumor vasculature suppresses angiogenesis and tumor growth. In an organotypic tissue culture angiogenesis assay, ERK-MAPK inhibition during the migratory phase results in loss of bipolarity, detachment, and cell death of isolated endothelial cells and retraction of sprouting tubules. These effects are the consequence of upregulated Rho-kinase signaling. Transient inhibition of Rho-kinase rescues the effects of ERK-MAPK inhibition in vitro and in vivo, promotes sprouting, and increases vessel length in tumors. We propose a regulatory role of Rho-kinase by ERK-MAPK during angiogenesis that acts through the control of actomyosin contractility. Our data delineate a mechanism by which ERK-MAPK promotes endothelial cell survival and sprouting by downregulating Rho-kinase signaling.
Collapse
Affiliation(s)
- Georgia Mavria
- Institute of Cancer Research, Cancer Research UK Centre for Cell and Molecular Biology, 237 Fulham Road, London SW3 6JB, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Heo JC, Park JY, Woo SU, Rho JR, Lee HJ, Kim SU, Kho YH, Lee SH. Dykellic Acid Inhibits Cell Migration and Tube Formation by RhoA-GTP Expression. Biol Pharm Bull 2006; 29:2256-9. [PMID: 17077524 DOI: 10.1248/bpb.29.2256] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Dykellic acid, a novel factor initially identified from the culture broth of Westerdykella multispora F50733, has been shown to inhibit matrix metalloprotease 9 activity, caspase-3 activity, B cell proliferation and LPS-induced IgM production, suggesting that this factor may have anti-cancer effects. In an effort to further address the possible anti-tumoral effects of dykellic acid, we used wound healing, invasion and RhoA-GTP assays to examine the effects of dykellic acid on cell migration, invasion and angiogenesis. Our results revealed that dykellic acid dose-dependently inhibits B16 cell migration and motility, and inhibits HUVEC tube formation. Western blot analysis of the active form of RhoA (RhoA-GTP) showed that dykellic acid treatment decreased the levels of RhoA-GTP. These findings collectively suggest that dykellic acid may have both anti-metastatic and anti-angiogenic acitivites, and provides the first evidence for the involvement of RhoA in dykellic acid-induced effects.
Collapse
Affiliation(s)
- Jin-Chul Heo
- Department of Food Science & Technology, Kyungpook National University, Daegu, Korea
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Hudry-Clergeon H, Stengel D, Ninio E, Vilgrain I. Platelet-activating factor increases VE-cadherin tyrosine phosphorylation in mouse endothelial cells and its association with the PtdIns3'-kinase. FASEB J 2005; 19:512-20. [PMID: 15791001 PMCID: PMC4848345 DOI: 10.1096/fj.04-2202com] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Platelet-activating factor (PAF), a potent inflammatory mediator, is involved in endothelial permeability. This study was designed to characterize PAF receptor (PAF-R) expression and its specific contribution to the modifications of adherens junctions in mouse endothelial cells. We demonstrated that PAF-R was expressed in mouse endothelial cells and was functionally active in stimulating p42/p44 MAPK and phosphatidylinositol 3-kinase (PtdIns3'-kinase)/Akt activities. Treatment of cells with PAF induced a rapid time- and dose-dependent (10(-7) to 10(-10) M) increase in tyrosine phosphorylation of a subset of proteins ranging from 90 to 220 kDa, including the VE-cadherin, the latter effect being prevented by the tyrosine kinase inhibitors herbimycin A and bis-tyrphostin. We demonstrated that PAF promoted formation of multimeric aggregates of VE-cadherin with PtdIns3'-kinase, which was also inhibited by herbimycin and bis-tyrphostin. Finally, we show by immunostaining of endothelial cells VE-cadherin that PAF dissociated adherens junctions. The present data provide the first evidence that treatment of endothelial cells with PAF promoted activation of tyrosine kinases and the VE-cadherin tyrosine phosphorylation and PtdIns3'-kinase association, which ultimately lead to the dissociation of adherens junctions. Physical association between PtdIns3'-kinase, serving as a docking protein, and VE-cadherin may thus provide an efficient mechanism for amplification and perpetuation of PAF-induced cellular activation.
Collapse
Affiliation(s)
- Hélène Hudry-Clergeon
- Laboratoire de développement et vieillissement de l'endothélium
Université Joseph FourierINSERMCEA
| | - Dominique Stengel
- Génétique épidémiologique et moléculaire des pathologies cardiovasculaires
Université Pierre et Marie Curie - Paris 6INSERMIFR14Faculté de Médecine Pitié-Salpétrière 91, Boulevard de L'hôpital 75634 Paris cedex 13
| | - Ewa Ninio
- Génétique épidémiologique et moléculaire des pathologies cardiovasculaires
Université Pierre et Marie Curie - Paris 6INSERMIFR14Faculté de Médecine Pitié-Salpétrière 91, Boulevard de L'hôpital 75634 Paris cedex 13
| | - Isabelle Vilgrain
- Laboratoire de développement et vieillissement de l'endothélium
Université Joseph FourierINSERMCEA
- * Correspondence should be addressed to Isabelle Vilgrain
| |
Collapse
|
44
|
Milkiewicz M, Ispanovic E, Doyle JL, Haas TL. Regulators of angiogenesis and strategies for their therapeutic manipulation. Int J Biochem Cell Biol 2005; 38:333-57. [PMID: 16309946 DOI: 10.1016/j.biocel.2005.10.006] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2005] [Revised: 10/05/2005] [Accepted: 10/11/2005] [Indexed: 12/19/2022]
Abstract
Angiogenesis provides a mechanism by which delivery of oxygen and nutrients is adapted to compliment changes in tissue mass or metabolic activity. However, maladaptive angiogenesis is integral to the process of several diseases common in Western countries, including tumor growth, vascular insufficiency, diabetic retinopathy and rheumatoid arthritis. Understanding the process of capillary growth, including the identification and functional analyses of key pro- and anti-angiogenic factors, provides knowledge that can be applied to improve/reverse these pathological states. Initially, angiogenesis research focused predominantly on vascular endothelial growth factor (VEGF) as a main player in the angiogenesis cascade. It is apparent now that participation of multiple angiogenic factors and signal pathways is critical to enable effective growth and maturation of nascent capillaries. The purpose of this review is to focus on recent progress in identifying angiogenesis signaling pathways that show promise as targets for successful induction or inhibition of capillary growth. The strategies applied to achieve these contradictory tasks are discussed within the framework of our existing fundamental knowledge of angiogenesis signaling cascades, with an emphasis on comparing the employment of distinctive tactics in modulation of these pathways. Innovative developments that are presented include: (1) inducing a pleiotropic response via activation or inhibition of angiogenic transcription factors; (2) modulation of nitric oxide tissue concentration; (3) manipulating the kallikrein-kinin system; (4) use of endothelial progenitor cells as a means to either directly contribute to capillary growth or to be used as a vehicle to deliver "suicide genes" to tumor tissue.
Collapse
Affiliation(s)
- Malgorzata Milkiewicz
- School of Kinesiology and Health Sciences, York University, Toronto, Ont. M3J 1P3, Canada
| | | | | | | |
Collapse
|
45
|
Cascone I, Napione L, Maniero F, Serini G, Bussolino F. Stable interaction between alpha5beta1 integrin and Tie2 tyrosine kinase receptor regulates endothelial cell response to Ang-1. J Cell Biol 2005; 170:993-1004. [PMID: 16157706 PMCID: PMC2171441 DOI: 10.1083/jcb.200507082] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2005] [Accepted: 08/08/2005] [Indexed: 12/21/2022] Open
Abstract
During angiogenic remodeling, Ang-1, the ligand of Tie2 tyrosine kinase, is involved in vessel sprouting and stabilization through unclear effects on nascent capillaries and mural cells. In our study, we hypothesized that the Ang-1/Tie2 system could cross-talk with integrins, and be influenced by the dynamic interactions between extracellular matrix and endothelial cells (ECs). Here, we show that alpha5beta1 specifically sensitizes and modulates Tie2 receptor activation and signaling, allowing EC survival at low concentrations of Ang-1 and inducing persistent EC motility. Tie2 and alpha5beta1 interact constitutively; alpha5beta1 binding to fibronectin increases this association, whereas Ang-1 stimulation recruits p85 and FAK to this complex. Furthermore, we demonstrate that Ang-1 is able to mediate selectively alpha5beta1 outside-in FAK phosphorylation. Thus, Ang-1 triggers signaling pathways through Tie2 and alpha5beta1 receptors that could cross-talk when Tie2/alpha5beta1 interaction occurs in ECs plated on fibronectin. By using blocking antibodies, we consistently found that alpha5beta1, but not alphavbeta3 activation, is essential to Ang-1-dependent angiogenesis in vivo.
Collapse
Affiliation(s)
- Ilaria Cascone
- Department of Oncological Sciences and Institute for Cancer Research and Treatment, University of Turin, 10060 Candiolo, Italy.
| | | | | | | | | |
Collapse
|
46
|
Hyvelin JM, Howell K, Nichol A, Costello CM, Preston RJ, McLoughlin P. Inhibition of Rho-kinase attenuates hypoxia-induced angiogenesis in the pulmonary circulation. Circ Res 2005; 97:185-91. [PMID: 15961717 DOI: 10.1161/01.res.0000174287.17953.83] [Citation(s) in RCA: 164] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pulmonary hypertension (PH) is a common complication of chronic hypoxic lung diseases, which increase morbidity and mortality. Hypoxic PH has previously been attributed to structural changes in the pulmonary vasculature including narrowing of the vascular lumen and loss of vessels, which produce a fixed increase in resistance. Using quantitative stereology, we now show that chronic hypoxia caused PH and remodeling of the blood vessel walls in rats but that this remodeling did not lead to structural narrowing of the vascular lumen. Sustained inhibition of the RhoA/Rho-kinase pathway throughout the period of hypoxic exposure attenuated PH and prevented remodeling in intra-acinar vessels without enlarging the structurally determined lumen diameter. In chronically hypoxic lungs, acute Rho kinase inhibition markedly decreased PVR but did not alter the alveolar to arterial oxygen gap. In addition to increased vascular resistance, chronic hypoxia induced Rho kinase-dependent capillary angiogenesis. Thus, hypoxic PH was not caused by fixed structural changes in the vasculature but by sustained vasoconstriction, which was largely Rho kinase dependent. Importantly, this vasoconstriction had no role in ventilation-perfusion matching and optimization of gas exchange. Rho kinase also mediated hypoxia-induced capillary angiogenesis, a previously unrecognized but potentially important adaptive response.
Collapse
Affiliation(s)
- Jean-Marc Hyvelin
- Department of Physiology, University College, Earlsfort Terrace, Dublin 2, Ireland
| | | | | | | | | | | |
Collapse
|
47
|
Müller MM, Singer BB, Klaile E, Obrink B, Lucka L. Transmembrane CEACAM1 affects integrin-dependent signaling and regulates extracellular matrix protein-specific morphology and migration of endothelial cells. Blood 2005; 105:3925-34. [PMID: 15687237 DOI: 10.1182/blood-2004-09-3618] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1/CD66a), expressed on leukocytes, epithelia, and endothelia mediates homophilic cell adhesion. It plays an important role in cell morphogenesis and, recently, soluble CEACAM1 isoforms have been implicated in angiogenesis. In the present study, we investigated the function of long transmembrane isoform of CEACAM1 (CEACAM1-L) in cultured rat brain endothelial cells. We observed that expression of CEACAM1-L promotes network formation on basement membrane Matrigel and increased cell motility after monolayer injury. During cell-matrix adhesion, CEACAM1-L translocated into the Triton X-100-insoluble cytoskeletal fraction and affected cell spreading and cell morphology on Matrigel and laminin-1 but not on fibronectin. On laminin-1, CEACAM1-L-expressing cells developed protrusions with lamellipodia, showed less stress fiber formation, reduced focal adhesion kinase (FAK) tyrosine phosphorylation, and decreased focal adhesion formation leading to high motility. CEACAM1-L-mediated morphologic alterations were sensitive to RhoA activation via lysophosphatidic acid (LPA) treatment and dependent on Rac1 activation. Furthermore, we demonstrate a matrix protein-dependent association of CEACAM1-L with talin, an important regulator of integrin function. Taken together, our results suggest that transmembrane CEACAM1-L expressed on endothelial cells is implicated in the activation phase of angiogenesis by affecting the cytoskeleton architecture and integrin-mediated signaling.
Collapse
Affiliation(s)
- Mario M Müller
- Institut für Biochemie und Molekularbiologie, Charité-Universitätsmedizin Berlin, Berlin-Dahlem, Germany
| | | | | | | | | |
Collapse
|
48
|
Loutrari H, Hatziapostolou M, Skouridou V, Papadimitriou E, Roussos C, Kolisis FN, Papapetropoulos A. Perillyl alcohol is an angiogenesis inhibitor. J Pharmacol Exp Ther 2004; 311:568-75. [PMID: 15210838 DOI: 10.1124/jpet.104.070516] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Aberrant angiogenesis is essential for the progression of solid tumors and hematological malignancies. Thus, antiangiogenic therapy is one of the most promising approaches to control cancer. In the present work, we examined the ability of perillyl alcohol (POH), a dietary monoterpene with well-established tumor chemopreventive and chemotherapeutic activity, to interfere with the process of angiogenesis. POH remarkably prevented new blood vessel growth in the in vivo chicken embryo chorioallantoic membrane assay and proved to be effective in inhibiting the morphogenic differentiation of cultured endothelial cells into capillary-like networks both in collagen gel and Matrigel models. In addition, POH reduced the cell number in a proliferation assay and induced apoptosis of endothelial cells as indicated by the POH-mediated increase of caspase-3 activity and DNA fragmentation. Consistent with the observed antisurvival effect, POH treatment resulted in a significant inhibition of Akt phosphorylation in endothelial cells. Finally, POH was able to differentially modulate the release of two important angiogenic regulators: vascular endothelial growth factor (VEGF) and angiopoietin 2 (Ang2). POH decreased the release of VEGF from cancer cells but stimulated the expression of Ang2 by endothelial cells, indicating that it might suppress neovascularization and induce vessel regression. Overall, these data underscore the antiangiogenic potential of POH and suggest that POH, in addition to its anticancer activity, may be an effective agent in the treatment of angiogenesis-dependent diseases.
Collapse
Affiliation(s)
- Heleni Loutrari
- Laboratory for Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece 26504
| | | | | | | | | | | | | |
Collapse
|
49
|
Hong KH, Ryu J, Han KH. Monocyte chemoattractant protein-1-induced angiogenesis is mediated by vascular endothelial growth factor-A. Blood 2004; 105:1405-7. [PMID: 15498848 DOI: 10.1182/blood-2004-08-3178] [Citation(s) in RCA: 293] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Monocyte chemoattractant protein-1 (MCP-1) has been recognized as an angiogenic chemokine. In the present study, we investigated the detailed mechanism by which MCP-1 induces angiogenesis. We found that MCP-1 up-regulated hypoxia-inducible factor 1 alpha (HIF-1 alpha) gene expression in human aortic endothelial cells (HAECs), which induced vascular endothelial growth factor-A(165) (VEGF-A(165)) expression in the aortic wall and HAECs through activation of p42/44 mitogen-activated protein kinase (MAPK). In vivo angiogenesis assay using chick chorioallantoic membrane (CAM) showed that MCP-1-induced angiogenesis was as potent as that induced by VEGF-A(165) and completely inhibited by a VEGF inhibitor, Flt(2-11). The inhibition of RhoA small G protein did not affect MCP-1-induced VEGF-A(165) production and secretion but completely blocked both MCP-1- and VEGF-A-induced new vessel formation, as determined by CAM assay. These results suggest that MCP-1-induced angiogenesis is composed largely of 2 sequential steps: the induction of VEGF-A gene expression by MCP-1 and the subsequent VEGF-A-induced angiogenesis.
Collapse
MESH Headings
- Angiogenesis Inducing Agents/administration & dosage
- Animals
- Aorta, Abdominal/physiology
- Aorta, Thoracic/physiology
- Cells, Cultured
- Chemokine CCL2/administration & dosage
- Chemokine CCL2/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiology
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit
- Male
- Neovascularization, Physiologic/physiology
- RNA, Messenger/biosynthesis
- Rats
- Rats, Sprague-Dawley
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
- Vascular Endothelial Growth Factor A/biosynthesis
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/physiology
Collapse
Affiliation(s)
- Kyung Hee Hong
- Department of Cardiology, University of Ulsan College of Medicine, Asan Medical Center, 388-1 Pungnap-2 dong Songpa-gu 138-736, Seoul, South Korea
| | | | | |
Collapse
|
50
|
Xue Y, Bi F, Zhang X, Pan Y, Liu N, Zheng Y, Fan D. Inhibition of endothelial cell proliferation by targeting Rac1 GTPase with small interference RNA in tumor cells. Biochem Biophys Res Commun 2004; 320:1309-1315. [PMID: 15303276 DOI: 10.1016/j.bbrc.2004.06.088] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Hypoxia-induced angiogenesis plays an important role in the malignancy of solid tumors. A number of recent studies including our own have suggested that Rho family small GTPases are involved in this process, and Racl, a prominent member of the Rho family, may be critical in regulating hypoxia-induced gene activation of several angiogenesis factors and tumor suppressors. To fur-ther define Racl function in angiogenesis and to explore novel approaches to modulate angiogenesis, we employed the small interference RNA technique to knock down gene expression of Racl in gastric cancer cell line AGS that expresses a high level of Racl. Both the mRNA and protein levels of Racl in the AGS cells were decreased dramatically after transfection with a Racl-specific siRNA vector. When the conditioned medium derived from the Racl downregulated AGS cells was applied to the human endothelial cells. it could significantly inhibit the cell proliferation. Further study proved that, VEGF and HIF-la, two angiogenesis promoting factors, were found to be downregulated whereas p53 and VHL, which are tumor suppressors and angiogenesis inhibitors. were upregulated in the Racl siRNA transfected cells. Our results suggest that Racl may be involved in angiogenesis by controlling the expression of angiogenesis-related factors and provide a possible strategy for the treatment of tumor angiogenesis by targeting the Racl GTPase.
Collapse
Affiliation(s)
- Yan Xue
- Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, PR China
| | | | | | | | | | | | | |
Collapse
|