1
|
Thapa K, Khan H, Kaur G, Kumar P, Singh TG. Therapeutic targeting of angiopoietins in tumor angiogenesis and cancer development. Biochem Biophys Res Commun 2023; 687:149130. [PMID: 37944468 DOI: 10.1016/j.bbrc.2023.149130] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023]
Abstract
The formation and progression of tumors in humans are linked to the abnormal development of new blood vessels known as neo-angiogenesis. Angiogenesis is a broad word that encompasses endothelial cell migration, proliferation, tube formation, and intussusception, as well as peri-EC recruitment and extracellular matrix formation. Tumor angiogenesis is regulated by angiogenic factors, out of which some of the most potent angiogenic factors such as vascular endothelial growth factor and Angiopoietins (ANGs) in the body are produced by macrophages and other immune cells within the tumor microenvironment. ANGs have a distinct function in tumor angiogenesis and behavior. ANG1, ANG 2, ANG 3, and ANG 4 are the family members of ANG out of which ANG2 has been extensively investigated owing to its unique role in modifying angiogenesis and its tight association with tumor progression, growth, and invasion/metastasis, which makes it an excellent candidate for therapeutic intervention in human malignancies. ANG modulators have demonstrated encouraging outcomes in the treatment of tumor development, either alone or in conjunction with VEGF inhibitors. Future development of more ANG modulators targeting other ANGs is needed. The implication of ANG1, ANG3, and ANG4 as probable therapeutic targets for anti-angiogenesis treatment in tumor development should be also evaluated. The article has described the role of ANG in tumor angiogenesis as well as tumor growth and the treatment strategies modulating ANGs in tumor angiogenesis as demonstrated in clinical studies. The pharmacological modulation of ANGs and ANG-regulated pathways that are responsible for tumor angiogenesis and cancer development should be evaluated for the development of future molecular therapies.
Collapse
Affiliation(s)
- Komal Thapa
- Chitkara School of Pharmacy, Chitkara University, 174103, Himachal Pradesh, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Gagandeep Kaur
- Chitkara School of Pharmacy, Chitkara University, 174103, Himachal Pradesh, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, 151401, Bathinda, India
| | | |
Collapse
|
2
|
Somanath PR, Chernoff J, Cummings BS, Prasad SM, Homan HD. Targeting P21-Activated Kinase-1 for Metastatic Prostate Cancer. Cancers (Basel) 2023; 15:2236. [PMID: 37190165 PMCID: PMC10137274 DOI: 10.3390/cancers15082236] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 05/17/2023] Open
Abstract
Metastatic prostate cancer (mPCa) has limited therapeutic options and a high mortality rate. The p21-activated kinase (PAK) family of proteins is important in cell survival, proliferation, and motility in physiology, and pathologies such as infectious, inflammatory, vascular, and neurological diseases as well as cancers. Group-I PAKs (PAK1, PAK2, and PAK3) are involved in the regulation of actin dynamics and thus are integral for cell morphology, adhesion to the extracellular matrix, and cell motility. They also play prominent roles in cell survival and proliferation. These properties make group-I PAKs a potentially important target for cancer therapy. In contrast to normal prostate and prostatic epithelial cells, group-I PAKs are highly expressed in mPCA and PCa tissue. Importantly, the expression of group-I PAKs is proportional to the Gleason score of the patients. While several compounds have been identified that target group-I PAKs and these are active in cells and mice, and while some inhibitors have entered human trials, as of yet, none have been FDA-approved. Probable reasons for this lack of translation include issues related to selectivity, specificity, stability, and efficacy resulting in side effects and/or lack of efficacy. In the current review, we describe the pathophysiology and current treatment guidelines of PCa, present group-I PAKs as a potential druggable target to treat mPCa patients, and discuss the various ATP-competitive and allosteric inhibitors of PAKs. We also discuss the development and testing of a nanotechnology-based therapeutic formulation of group-I PAK inhibitors and its significant potential advantages as a novel, selective, stable, and efficacious mPCa therapeutic over other PCa therapeutics in the pipeline.
Collapse
Affiliation(s)
- Payaningal R. Somanath
- Department of Clinical & Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
- MetasTx LLC, Basking Ridge, NJ 07920, USA
| | - Jonathan Chernoff
- MetasTx LLC, Basking Ridge, NJ 07920, USA
- Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Brian S. Cummings
- MetasTx LLC, Basking Ridge, NJ 07920, USA
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Sandip M. Prasad
- Morristown Medical Center, Atlantic Health System, Morristown, NJ 07960, USA
| | | |
Collapse
|
3
|
Abstract
The angiogenesis process was described in its basic concepts in the works of the Scottish surgeon John Hunter and terminologically assessed in the early twentieth century. An aberrant angiogenesis is a prerequisite for cancer cells in solid tumors to grow and metastasize. The sprouting of new blood vessels is one of the major characteristics of cancer and represents a gateway for tumor cells to enter both the blood and lymphatic circulation systems. In vivo, ex vivo, and in vitro models of angiogenesis have provided essential tools for cancer research and antiangiogenic drug screening. Several in vivo studies have been performed to investigate the various steps of tumor angiogenesis and in vitro experiments contributed to dissecting the molecular bases of this phenomenon. Moreover, coculture of cancer and endothelial cells in 2D and 3D matrices have contributed to improve the recapitulation of the complex process of tumor angiogenesis, including the peculiar conditions of tumor microenvironment.
Collapse
Affiliation(s)
- Gianfranco Natale
- Department of Translational Research and New Technologies in Medicine and Surgery, School of Medicine, University of Pisa, Pisa, Italy
- Museum of Human Anatomy "Filippo Civinini", School of Medicine, University of Pisa, Pisa, Italy
| | - Guido Bocci
- Department of Clinical and Experimental Medicine, School of Medicine, University of Pisa, Pisa, Italy.
| |
Collapse
|
4
|
Laschke MW, Gu Y, Menger MD. Replacement in angiogenesis research: Studying mechanisms of blood vessel development by animal-free in vitro, in vivo and in silico approaches. Front Physiol 2022; 13:981161. [PMID: 36060683 PMCID: PMC9428454 DOI: 10.3389/fphys.2022.981161] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/21/2022] [Indexed: 01/10/2023] Open
Abstract
Angiogenesis, the development of new blood vessels from pre-existing ones, is an essential process determining numerous physiological and pathological conditions. Accordingly, there is a high demand for research approaches allowing the investigation of angiogenic mechanisms and the assessment of pro- and anti-angiogenic therapeutics. The present review provides a selective overview and critical discussion of such approaches, which, in line with the 3R principle, all share the common feature that they are not based on animal experiments. They include in vitro assays to study the viability, proliferation, migration, tube formation and sprouting activity of endothelial cells in two- and three-dimensional environments, the degradation of extracellular matrix compounds as well as the impact of hemodynamic forces on blood vessel formation. These assays can be complemented by in vivo analyses of microvascular network formation in the chorioallantoic membrane assay and early stages of zebrafish larvae. In addition, the combination of experimental data and physical laws enables the mathematical modeling of tissue-specific vascularization, blood flow patterns, interstitial fluid flow as well as oxygen, nutrient and drug distribution. All these animal-free approaches markedly contribute to an improved understanding of fundamental biological mechanisms underlying angiogenesis. Hence, they do not only represent essential tools in basic science but also in early stages of drug development. Moreover, their advancement bears the great potential to analyze angiogenesis in all its complexity and, thus, to make animal experiments superfluous in the future.
Collapse
|
5
|
Colón-Bolea P, García-Gómez R, Casar B. RAC1 Activation as a Potential Therapeutic Option in Metastatic Cutaneous Melanoma. Biomolecules 2021; 11:1554. [PMID: 34827551 PMCID: PMC8615836 DOI: 10.3390/biom11111554] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/21/2022] Open
Abstract
Metastasis is a complex process by which cancer cells escape from the primary tumor to colonize distant organs. RAC1 is a member of the RHO family of small guanosine triphosphatases that plays an important role in cancer migration, invasion, angiogenesis and metastasis. RAC1 activation has been related to most cancers, such as cutaneous melanoma, breast, lung, and pancreatic cancer. RAC1P29S driver mutation appears in a significant number of cutaneous melanoma cases. Likewise, RAC1 is overexpressed or hyperactivated via signaling through oncogenic cell surface receptors. Thus, targeting RAC1 represents a promising strategy for cutaneous melanoma therapy, as well as for inhibition of other signaling activation that promotes resistance to targeted therapies. In this review, we focus on the role of RAC1 in metastatic cutaneous melanoma emphasizing the anti-metastatic potential of RAC1- targeting drugs.
Collapse
Affiliation(s)
- Paula Colón-Bolea
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas—Universidad de Cantabria, 39011 Santander, Spain; (P.C.-B.); (R.G.-G.)
| | - Rocío García-Gómez
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas—Universidad de Cantabria, 39011 Santander, Spain; (P.C.-B.); (R.G.-G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Berta Casar
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas—Universidad de Cantabria, 39011 Santander, Spain; (P.C.-B.); (R.G.-G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
6
|
Lin PK, Salvador J, Xie J, Aguera KN, Koller GM, Kemp SS, Griffin CT, Davis GE. Selective and Marked Blockade of Endothelial Sprouting Behavior Using Paclitaxel and Related Pharmacologic Agents. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:2245-2264. [PMID: 34563512 DOI: 10.1016/j.ajpath.2021.08.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/10/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022]
Abstract
Whether alterations in the microtubule cytoskeleton affect the ability of endothelial cells (ECs) to sprout and form branching networks of tubes was investigated in this study. Bioassays of human EC tubulogenesis, where both sprouting behavior and lumen formation can be rigorously evaluated, were used to demonstrate that addition of the microtubule-stabilizing drugs, paclitaxel, docetaxel, ixabepilone, and epothilone B, completely interferes with EC tip cells and sprouting behavior, while allowing for EC lumen formation. In bioassays mimicking vasculogenesis using single or aggregated ECs, these drugs induce ring-like lumens from single cells or cyst-like spherical lumens from multicellular aggregates with no evidence of EC sprouting behavior. Remarkably, treatment of these cultures with a low dose of the microtubule-destabilizing drug, vinblastine, led to an identical result, with complete blockade of EC sprouting, but allowing for EC lumen formation. Administration of paclitaxel in vivo markedly interfered with angiogenic sprouting behavior in developing mouse retina, providing corroboration. These findings reveal novel biological activities for pharmacologic agents that are widely utilized in multidrug chemotherapeutic regimens for the treatment of human malignant cancers. Overall, this work demonstrates that manipulation of microtubule stability selectively interferes with the ability of ECs to sprout, a necessary step to initiate and form branched capillary tube networks.
Collapse
Affiliation(s)
- Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Jocelynda Salvador
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Jun Xie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kalia N Aguera
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Gretchen M Koller
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Scott S Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida.
| |
Collapse
|
7
|
Zheng Y, Yuan W, Liu H, Huang S, Bian L, Guo R. Injectable supramolecular gelatin hydrogel loading of resveratrol and histatin-1 for burn wound therapy. Biomater Sci 2020; 8:4810-4820. [PMID: 32744545 DOI: 10.1039/d0bm00391c] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Prolonged inflammatory response and insufficient vascularization cause delayed and poor wound healing. In this study, we fabricated a supramolecular host-guest gelatin (HGM) hydrogel loaded with resveratrol (Res) and histatin-1 (His-1) to suppress inflammation and promote vascularization at skin burn wound sites. The HGM hydrogel showed good properties of shear-thinning and injectability, thereby allowing easy in situ injection and fast adaption to irregular wounds. Res and His-1 were demonstrated to enhance angiogenesis in vitro using cell migration and tube formation assays based on human umbilical vein endothelial cells (HUVECs). In an established rat burn wound model, HGM/Res/His-1 hydrogel treatment promoted wound healing by inhibiting expression of the pro-inflammatory factors of interleukin 6 (IL-6), interleukin 1β (IL-1β) and tumor necrosis factor α (TNF-α) and increasing the expression of transforming growth factor β1 (TGF-β1) and platelet endothelial cell adhesion molecule-1 (CD31). HGM/Res/His-1 hydrogel treatment showed comparable efficacy with that of the commercial dressing, Tegaderm™, and therefore shows promising potential for clinical translation.
Collapse
Affiliation(s)
- Yuanyuan Zheng
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | | | | | | | | | | |
Collapse
|
8
|
Goyal D, Goyal R. Angiogenic Transformation in Human Brain Micro Endothelial Cells: Whole Genome DNA Methylation and Transcriptomic Analysis. Front Physiol 2019; 10:1502. [PMID: 31920707 PMCID: PMC6917667 DOI: 10.3389/fphys.2019.01502] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/26/2019] [Indexed: 01/22/2023] Open
Abstract
We tested the hypothesis that endothelial capillary tube formation in 3D cultures in basement membrane extract (BME) is secondary to the altered DNA promoter methylation and mRNA expression in human brain micro endothelial cells (HBMECs). We conducted a whole-genome transcriptomic and methylation microarray and CRISPR/Cas9-mediated gene knockdown to test our hypothesis. The data demonstrated that with angiogenic transformation 1318 and 1490 genes were significantly (p < 0.05) upregulated and downregulated, respectively. We compared our gene expression data with the published databases on GEO and found several genes in common. PTGS2, SELE, ID2, HSPA6, DLX2, HEY2, FOSB, SMAD6, SMAD7, and SMAD9 showed a very high level of expression during capillary tube formation. Among downregulated gene were ITGB4, TNNT1, PRSS35, TXNIP, IGFBP5. The most affected canonical pathways were ATM signaling and cell cycle G2/M DNA damage checkpoint regulation. The top upstream regulators of angiogenic transformation were identified to be VEGF, TP53, HGF, ESR1, and CDKN1A. We compared the changes in gene expression with the change in gene methylation and found hypomethylation of the CpG sites was associated with upregulation of 515 genes and hypermethylation was associated with the downregulation of 31 genes. Furthermore, the silencing of FOSB, FZD7, HEY2, HSPA6, NR4A3, SELE, PTGS2, SMAD6, SMAD7, and SMAD9 significantly inhibited angiogenic transformation as well as cell migration of HBMECs. We conclude that the angiogenic transformation is associated with altered DNA methylation and gene expression changes.
Collapse
Affiliation(s)
- Dipali Goyal
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
| | - Ravi Goyal
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
9
|
Pulmonary Vascular Platform Models the Effects of Flow and Pressure on Endothelial Dysfunction in BMPR2 Associated Pulmonary Arterial Hypertension. Int J Mol Sci 2018; 19:ijms19092561. [PMID: 30158434 PMCID: PMC6164056 DOI: 10.3390/ijms19092561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/22/2018] [Accepted: 08/22/2018] [Indexed: 12/15/2022] Open
Abstract
Endothelial dysfunction is a known consequence of bone morphogenetic protein type II receptor (BMPR2) mutations seen in pulmonary arterial hypertension (PAH). However, standard 2D cell culture models fail to mimic the mechanical environment seen in the pulmonary vasculature. Hydrogels have emerged as promising platforms for 3D disease modeling due to their tunable physical and biochemical properties. In order to recreate the mechanical stimuli seen in the pulmonary vasculature, we have created a novel 3D hydrogel-based pulmonary vasculature model (“artificial arteriole”) that reproduces the pulsatile flow rates and pressures seen in the human lung. Using this platform, we studied both Bmpr2R899X and WT endothelial cells to better understand how the addition of oscillatory flow and physiological pressure influenced gene expression, cell morphology, and cell permeability. The addition of oscillatory flow and pressure resulted in several gene expression changes in both WT and Bmpr2R899X cells. However, for many pathways with relevance to PAH etiology, Bmpr2R899X cells responded differently when compared to the WT cells. Bmpr2R899X cells were also found not to elongate in the direction of flow, and instead remained stagnant in morphology despite mechanical stimuli. The increased permeability of the Bmpr2R899X layer was successfully reproduced in our artificial arteriole, with the addition of flow and pressure not leading to significant changes in permeability. Our artificial arteriole is the first to model many mechanical properties seen in the lung. Its tunability enables several new opportunities to study the endothelium in pulmonary vascular disease with increased control over environmental parameters.
Collapse
|
10
|
Liu X, Gu X, Ma W, Oxendine M, Gil HJ, Davis GE, Cleaver O, Oliver G. Rasip1 controls lymphatic vessel lumen maintenance by regulating endothelial cell junctions. Development 2018; 145:dev165092. [PMID: 30042182 PMCID: PMC6141773 DOI: 10.1242/dev.165092] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/11/2018] [Indexed: 01/04/2023]
Abstract
Although major progress in our understanding of the genes and mechanisms that regulate lymphatic vasculature development has been made, we still do not know how lumen formation and maintenance occurs. Here, we identify the Ras-interacting protein Rasip1 as a key player in this process. We show that lymphatic endothelial cell-specific Rasip1-deficient mouse embryos exhibit enlarged and blood-filled lymphatics at embryonic day 14.5. These vessels have patent lumens with disorganized junctions. Later on, as those vessels become fragmented and lumens collapse, cell junctions become irregular. In addition, Rasip1 deletion at later stages impairs lymphatic valve formation. We determined that Rasip1 is essential for lymphatic lumen maintenance during embryonic development by regulating junction integrity, as Rasip1 loss results in reduced levels of junction molecules and defective cytoskeleton organization in vitro and in vivo We determined that Rasip1 regulates Cdc42 activity, as deletion of Cdc42 results in similar phenotypes to those seen following the loss of Rasip1 Furthermore, ectopic Cdc42 expression rescues the phenotypes in Rasip1-deficient lymphatic endothelial cells, supporting the suggestion that Rasip1 regulates Cdc42 activity to regulate cell junctions and cytoskeleton organization, which are both activities required for lymphatic lumen maintenance.
Collapse
Affiliation(s)
- Xiaolei Liu
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Xiaowu Gu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wanshu Ma
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Michael Oxendine
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hyea Jin Gil
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - George E Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Guillermo Oliver
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
11
|
Thewke DP, Kou J, Fulmer ML, Xie Q. The HGF/MET Signaling and Therapeutics in Cancer. CURRENT HUMAN CELL RESEARCH AND APPLICATIONS 2018. [DOI: 10.1007/978-981-10-7296-3_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
12
|
Liu Y, Xu G, Wei J, Wu Q, Li X. Cardiomyocyte coculture on layered fibrous scaffolds assembled from micropatterned electrospun mats. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 81:500-510. [DOI: 10.1016/j.msec.2017.08.042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/12/2017] [Accepted: 08/10/2017] [Indexed: 12/14/2022]
|
13
|
Torres P, Díaz J, Arce M, Silva P, Mendoza P, Lois P, Molina-Berríos A, Owen GI, Palma V, Torres VA. The salivary peptide histatin-1 promotes endothelial cell adhesion, migration, and angiogenesis. FASEB J 2017; 31:4946-4958. [PMID: 28751526 DOI: 10.1096/fj.201700085r] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 07/10/2017] [Indexed: 01/19/2023]
Abstract
Saliva is a key factor that contributes to the high efficiency of wound healing in the oral mucosa. This is not only attributed to physical cues but also to the presence of specific peptides in the saliva, such as histatins. Histatin-1 is a 38 aa antimicrobial peptide, highly enriched in human saliva, which has been previously reported to promote the migration of oral keratinocytes and fibroblasts in vitro However, the participation of histatin-1 in other crucial events required for wound healing, such as angiogenesis, is unknown. Here we demonstrate that histatin-1 promotes angiogenesis, as shown in vivo, using the chick chorioallantoic membrane model, and by an in vitro tube formation assay, using both human primary cultured endothelial cells (HUVECs) and the EA.hy926 cell line. Specifically, histatin-1 promoted endothelial cell adhesion and spreading onto fibronectin, as well as endothelial cell migration in the wound closure and Boyden chamber assays. These actions required the activation of the Ras and Rab interactor 2 (RIN2)/Rab5/Rac1 signaling axis, as histatin-1 increased the recruitment of RIN2, a Rab5-guanine nucleotide exchange factor (GEF) to early endosomes, leading to sequential Rab5/Rac1 activation. Accordingly, interfering with either Rab5 or Rac1 activities prevented histatin-1-dependent endothelial cell migration. Finally, by immunodepletion assays, we showed that salivary histatin-1 is required for the promigratory effects of saliva on endothelial cells. In conclusion, we report that salivary histatin-1 is a novel proangiogenic factor that may contribute to oral wound healing.-Torres, P., Díaz, J., Arce, M., Silva, P., Mendoza, P., Lois, P., Molina-Berríos, A., Owen, G. I., Palma, V., Torres, V. A. The salivary peptide histatin-1 promotes endothelial cell adhesion, migration, and angiogenesis.
Collapse
Affiliation(s)
- Pedro Torres
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Jorge Díaz
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - Maximiliano Arce
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile.,Faculty of Biological Sciences Pontificia Universidad Católica de Chile, Santiago, Chile.,Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricio Silva
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Faculty of Health Sciences, Universidad Central de Chile, Santiago, Chile
| | - Pablo Mendoza
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Pablo Lois
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Alfredo Molina-Berríos
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Gareth I Owen
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile.,Faculty of Biological Sciences Pontificia Universidad Católica de Chile, Santiago, Chile.,Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Verónica Palma
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Vicente A Torres
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile; .,Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| |
Collapse
|
14
|
D'Angelo L, Myer NM, Myers KA. MCAK-mediated regulation of endothelial cell microtubule dynamics is mechanosensitive to myosin-II contractility. Mol Biol Cell 2017; 28:1223-1237. [PMID: 28298485 PMCID: PMC5415018 DOI: 10.1091/mbc.e16-05-0306] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 02/21/2017] [Accepted: 02/24/2017] [Indexed: 01/16/2023] Open
Abstract
This study indicates that MCAK contributes to the mechanosensing-mediated regulation of MT dynamics through a myosin-II–dependent mechanism that becomes uncoupled in response to 3D ECM engagement specifically within EC branches. Compliance and dimensionality mechanosensing, the processes by which cells sense the physical attributes of the extracellular matrix (ECM), are known to drive cell branching and shape change largely through a myosin-II–mediated reorganization of the actin and microtubule (MT) cytoskeletons. Subcellular regulation of MT dynamics is spatially controlled through a Rac1–Aurora-A kinase pathway that locally inhibits the MT depolymerizing activity of mitotic centromere–associated kinesin (MCAK), thereby promoting leading-edge MT growth and cell polarization. These results suggest that the regulation of MT growth dynamics is intimately linked to physical engagement of the cell with the ECM. Here, we tested the hypothesis that MCAK contributes to compliance and dimensionality mechanosensing-mediated regulation of MT growth dynamics through a myosin-II–dependent signaling pathway. We cultured endothelial cells (ECs) on collagen-coupled stiff or compliant polyacrylamide ECMs to examine the effects of MCAK expression on MT growth dynamics and EC branching morphology. Our results identify that MCAK promotes fast MT growth speeds in ECs cultured on compliant 2D ECMs but promotes slow MT growth speeds in ECs cultured on compliant 3D ECMs, and these effects are myosin-II dependent. Furthermore, we find that 3D ECM engagement uncouples MCAK-mediated regulation of MT growth persistence from myosin-II–mediated regulation of growth persistence specifically within EC branched protrusions.
Collapse
Affiliation(s)
- Lauren D'Angelo
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | - Nicole M Myer
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | - Kenneth A Myers
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| |
Collapse
|
15
|
Prieto CP, Ortiz MC, Villanueva A, Villarroel C, Edwards SS, Elliott M, Lattus J, Aedo S, Meza D, Lois P, Palma V. Netrin-1 acts as a non-canonical angiogenic factor produced by human Wharton's jelly mesenchymal stem cells (WJ-MSC). Stem Cell Res Ther 2017; 8:43. [PMID: 28241866 PMCID: PMC5330133 DOI: 10.1186/s13287-017-0494-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 01/16/2017] [Accepted: 02/08/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Angiogenesis, the process in which new blood vessels are formed from preexisting ones, is highly dependent on the presence of classical angiogenic factors. Recent evidence suggests that axonal guidance proteins and their receptors can also act as angiogenic regulators. Netrin, a family of laminin-like proteins, specifically Netrin-1 and 4, act via DCC/Neogenin-1 and UNC5 class of receptors to promote or inhibit angiogenesis, depending on the physiological context. METHODS Mesenchymal stem cells secrete a broad set of classical angiogenic factors. However, little is known about the expression of non-canonical angiogenic factors such as Netrin-1. The aim was to characterize the possible secretion of Netrin ligands by Wharton's jelly-derived mesenchymal stem cells (WJ-MSC). We evaluated if Netrin-1 presence in the conditioned media from these cells was capable of inducing angiogenesis both in vitro and in vivo, using human umbilical vein endothelial cells (HUVEC) and chicken chorioallantoic membrane (CAM), respectively. In addition, we investigated if the RhoA/ROCK pathway is responsible for the integration of Netrin signaling to control vessel formation. RESULTS The paracrine angiogenic effect of the WJ-MSC-conditioned media is mediated at least in part by Netrin-1 given that pharmacological blockage of Netrin-1 in WJ-MSC resulted in diminished angiogenesis on HUVEC. When HUVEC were stimulated with exogenous Netrin-1 assayed at physiological concentrations (10-200 ng/mL), endothelial vascular migration occurred in a concentration-dependent manner. In line with our determination of Netrin-1 present in WJ-MSC-conditioned media we were able to obtain endothelial tubule formation even in the pg/mL range. Through CAM assays we validated that WJ-MSC-secreted Netrin-1 promotes an increased angiogenesis in vivo. Netrin-1, secreted by WJ-MSC, might mediate its angiogenic effect through specific cell surface receptors on the endothelium, such as UNC5b and/or integrin α6β1, expressed in HUVEC. However, the angiogenic response of Netrin-1 seems not to be mediated through the RhoA/ROCK pathway. CONCLUSIONS Thus, here we show that stromal production of Netrin-1 is a critical component of the vascular regulatory machinery. This signaling event may have deep implications in the modulation of several processes related to a number of diseases where angiogenesis plays a key role in vascular homeostasis.
Collapse
Affiliation(s)
- Catalina P. Prieto
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - María Carolina Ortiz
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - Andrea Villanueva
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - Cynthia Villarroel
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - Sandra S. Edwards
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - Matías Elliott
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - José Lattus
- Campus Oriente, Department of Obstetrics and Gynecology, Faculty of Medicine, University of Chile, Santiago de Chile, Chile
| | - Sócrates Aedo
- Campus Oriente, Department of Obstetrics and Gynecology, Faculty of Medicine, University of Chile, Santiago de Chile, Chile
| | - Daniel Meza
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - Pablo Lois
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - Verónica Palma
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| |
Collapse
|
16
|
Hargreaves A, Bigley A, Price S, Kendrew J, Barry ST. Automated image analysis of intra-tumoral and peripheral endocrine organ vascular bed regression using 'Fibrelength' as a novel structural biomarker. J Appl Toxicol 2017; 37:902-912. [PMID: 28186326 DOI: 10.1002/jat.3438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 12/15/2016] [Accepted: 12/15/2016] [Indexed: 12/16/2022]
Abstract
The study of vascular modulation has received a great deal of attention in recent years as knowledge has increased around the role of angiogenesis within disease contexts such as cancer. Despite rapidly expanding insights into the molecular processes involved and the concomitant generation of a number of anticancer vascular modulating chemotherapeutics, techniques used in the measurement of structural vascular change have advanced more modestly, particularly with regard to the preclinical quantification of off-target vascular regression within systemic, notably endocrine, blood vessels. Such changes translate into a number of major clinical side effects and there remains a need for improved preclinical screening and analysis. Here we present the generation of a novel structural biomarker, which can be incorporated into a number of contemporary image analysis platforms and used to compare tumour versus systemic host tissue vascularity. By contrasting the measurements obtained, the preclinical efficacy of vascular modulating chemotherapies can be evaluated in light of the predicted therapeutic window. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Adam Hargreaves
- PathCelerate Ltd, The BioHub, Alderley Park, Mereside, Alderley Edge, Cheshire, UK
| | - Alison Bigley
- Oraclebio Ltd, BioCity Scotland, North Lanarkshire, Scotland, UK
| | - Shirley Price
- University of Surrey, 3660 Office of the Vice-Provost, Guildford, Surrey, UK
| | - Jane Kendrew
- AstraZeneca PLC, Oncology iMED, Alderley Edge, Cheshire, UK
| | - Simon T Barry
- AstraZeneca PLC, Oncology iMED, Alderley Edge, Cheshire, UK
| |
Collapse
|
17
|
Gambino TJ, Williams SP, Caesar C, Resnick D, Nowell CJ, Farnsworth RH, Achen MG, Stacker SA, Karnezis T. A Three-Dimensional Lymphatic Endothelial Cell Tube Formation Assay to Identify Novel Kinases Involved in Lymphatic Vessel Remodeling. Assay Drug Dev Technol 2017; 15:30-43. [DOI: 10.1089/adt.2016.764] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- T. Jessica Gambino
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Steven P. Williams
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Carol Caesar
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Daniel Resnick
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Cameron J. Nowell
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Rae H. Farnsworth
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Marc G. Achen
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Steven A. Stacker
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Tara Karnezis
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- O'Brien Institute, a Department of St. Vincent's Institute, Fitzroy, Victoria, Australia
| |
Collapse
|
18
|
CdGAP/ARHGAP31, a Cdc42/Rac1 GTPase regulator, is critical for vascular development and VEGF-mediated angiogenesis. Sci Rep 2016; 6:27485. [PMID: 27270835 PMCID: PMC4895392 DOI: 10.1038/srep27485] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/17/2016] [Indexed: 02/06/2023] Open
Abstract
Mutations in the CdGAP/ARHGAP31 gene, which encodes a GTPase-activating protein for Rac1 and Cdc42, have been reported causative in the Adams-Oliver developmental syndrome often associated with vascular defects. However, despite its abundant expression in endothelial cells, CdGAP function in the vasculature remains unknown. Here, we show that vascular development is impaired in CdGAP-deficient mouse embryos at E15.5. This is associated with superficial vessel defects and subcutaneous edema, resulting in 44% embryonic/perinatal lethality. VEGF-driven angiogenesis is defective in CdGAP(-/-) mice, showing reduced capillary sprouting from aortic ring explants. Similarly, VEGF-dependent endothelial cell migration and capillary formation are inhibited upon CdGAP knockdown. Mechanistically, CdGAP associates with VEGF receptor-2 and controls VEGF-dependent signaling. Consequently, CdGAP depletion results in impaired VEGF-mediated Rac1 activation and reduced phosphorylation of critical intracellular mediators including Gab1, Akt, PLCγ and SHP2. These findings are the first to demonstrate the importance of CdGAP in embryonic vascular development and VEGF-induced signaling, and highlight CdGAP as a potential therapeutic target to treat pathological angiogenesis and vascular dysfunction.
Collapse
|
19
|
Abstract
Angiogenesis involves the generation of new blood vessels from the existing vasculature and is dependent on many growth factors and signaling events. In vivo angiogenesis is dynamic and complex, meaning assays are commonly utilized to explore specific targets for research into this area. Tube-forming assays offer an excellent overview of the molecular processes in angiogenesis. The Matrigel tube forming assay is a simple-to-implement but powerful tool for identifying biomolecules involved in angiogenesis. A detailed experimental protocol on the implementation of the assay is described in conjunction with an in-depth review of methods that can be applied to the analysis of the tube formation. In addition, an ImageJ plug-in is presented which allows automatic quantification of tube images reducing analysis times while removing user bias and subjectivity.
Collapse
Affiliation(s)
- Ryan M Brown
- Centre for Doctoral Training in Physical Sciences of Imaging in the Biomedical Sciences and School of Chemistry, University of Birmingham, Birmingham, UK
| | - Christopher J Meah
- School of Immunity and Infection and Cancer Studies, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Victoria L Heath
- School of Immunity and Infection and Cancer Studies, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Iain B Styles
- School of Computer Science, University of Birmingham, Birmingham, UK
| | - Roy Bicknell
- School of Immunity and Infection and Cancer Studies, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
20
|
LPS-Stimulated Human Skin-Derived Stem Cells Enhance Neo-Vascularization during Dermal Regeneration. PLoS One 2015; 10:e0142907. [PMID: 26565617 PMCID: PMC4643997 DOI: 10.1371/journal.pone.0142907] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 10/28/2015] [Indexed: 11/26/2022] Open
Abstract
High numbers of adult stem cells are still required to improve the formation of new vessels in scaffolds to accelerate dermal regeneration. Recent data indicate a benefit for vascularization capacity by stimulating stem cells with lipopolysaccharide (LPS). In this study, stem cells derived from human skin (SDSC) were activated with LPS and seeded in a commercially available dermal substitute to examine vascularization in vivo. Besides, in vitro assays were performed to evaluate angiogenic factor release and tube formation ability. Results showed that LPS-activated SDSC significantly enhanced vascularization of the scaffolds, compared to unstimulated stem cells in vivo. Further, in vitro assays confirmed higher secretion rates of proangiogenic as well as proinflammatoric factors in the presence of LPS-activated SDSC. Our results suggest that combining activated stem cells and a dermal substitute is a promising option to enhance vascularization in scaffold-mediated dermal regeneration.
Collapse
|
21
|
Activation of Apoptotic Signal in Endothelial Cells through Intracellular Signaling Molecules Blockade in Tumor-Induced Angiogenesis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:908757. [PMID: 26346668 PMCID: PMC4539440 DOI: 10.1155/2015/908757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 06/07/2015] [Accepted: 06/11/2015] [Indexed: 11/25/2022]
Abstract
Tumor-induced angiogenesis is the bridge between avascular and vascular tumor growth phases. In tumor-induced angiogenesis, endothelial cells start to migrate and proliferate toward the tumor and build new capillaries toward the tumor. There are two stages for sprout extension during angiogenesis. The first stage is prior to anastomosis, when single sprouts extend. The second stage is after anastomosis when closed flow pathways or loops are formed and blood flows in the closed loops. Prior to anastomosis, biochemical and biomechanical signals from extracellular matrix regulate endothelial cell phenotype; however, after anastomosis, blood flow is the main regulator of endothelial cell phenotype. In this study, the critical signaling pathways of each stage are introduced. A Boolean network model is used to map environmental and flow induced signals to endothelial cell phenotype (proliferation, migration, apoptosis, and lumen formation). Using the Boolean network model, blockade of intracellular signaling molecules of endothelial cell is investigated prior to and after anastomosis and the cell fate is obtained in each case. Activation of apoptotic signal in endothelial cell can prevent the extension of new vessels and may inhibit angiogenesis. It is shown that blockade of a few signaling molecules in endothelial cell activates apoptotic signal that are proposed as antiangiogenic strategies.
Collapse
|
22
|
Bazmara H, Soltani M, Sefidgar M, Bazargan M, Mousavi Naeenian M, Rahmim A. Blood flow and endothelial cell phenotype regulation during sprouting angiogenesis. Med Biol Eng Comput 2015; 54:547-58. [DOI: 10.1007/s11517-015-1341-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 07/01/2015] [Indexed: 11/24/2022]
|
23
|
Xia Y, Cai XY, Fan JQ, Zhang LL, Ren JH, Chen J, Li ZY, Zhang RG, Zhu F, Wu G. Rho Kinase Inhibitor Fasudil Suppresses the Vasculogenic Mimicry of B16 Mouse Melanoma Cells Both In Vitro and In Vivo. Mol Cancer Ther 2015; 14:1582-90. [PMID: 25934709 DOI: 10.1158/1535-7163.mct-14-0523] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 04/24/2015] [Indexed: 11/16/2022]
Abstract
The aim of this study was to investigate the biologic role of the Rho kinase inhibitor fasudil in the vasculogenic mimicry (VM) of B16 mouse melanoma cells. It was previously reported that RhoA plays a critical role in angiogenesis by coordinating endothelial cell cytoskeleton remodeling and promoting endothelial cell motility. Although RhoA has been implicated in the regulation of angiogenesis, little has been described regarding its control of these tumor cell-lined channels. In this study, we established an in vitro model of VM using 3-dimensional cell culturing of mouse B16 melanoma cells and studied VM in vivo by transplanting B16 cells into C57/BL mice. Next, we explored the effect of RhoA and Rho-associated, coiled-coil containing protein kinase (ROCK) on VM formation using the Rho kinase inhibitor fasudil. We provide direct evidence that fasudil leads to reduced vascular-like channels in Matrigel. Additional experiments suggested that fasudil prevents both initial cellular architecture changes and cell migration in vitro. Finally, we provide in-depth evidence for the underlying mechanisms of fasudil-induced VM destruction using the Rho-GTPase agonist lysophosphatidic acid. In vivo studies revealed that fasudil reduced B16 melanoma cell xenograft tumor growth without causing significant toxicity in mice. Fasudil-treated tumors also displayed fewer VM channels. These results suggest that fasudil may be an emerging therapeutic option for targeting cancer VM.
Collapse
Affiliation(s)
- Yun Xia
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian-Yi Cai
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji-Quan Fan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Ling Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing-Hua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen-Yu Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui-Guang Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Zhu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
24
|
Jacobo SMP, Kazlauskas A. Insulin-like growth factor 1 (IGF-1) stabilizes nascent blood vessels. J Biol Chem 2015; 290:6349-60. [PMID: 25564613 DOI: 10.1074/jbc.m114.634154] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Here we report that VEGF-A and IGF-1 differ in their ability to stabilize newly formed blood vessels and endothelial cell tubes. Although VEGF-A failed to support an enduring vascular response, IGF-1 stabilized neovessels generated from primary endothelial cells derived from various vascular beds and mouse retinal explants. In these experimental systems, destabilization/regression was driven by lysophosphatidic acid (LPA). Because previous studies have established that Erk antagonizes LPA-mediated regression, we considered whether Erk was an essential component of IGF-dependent stabilization. Indeed, IGF-1 lost its ability to stabilize neovessels when the Erk pathway was inhibited pharmacologically. Furthermore, stabilization was associated with prolonged Erk activity. In the presence of IGF-1, Erk activity persisted longer than in the presence of VEGF or LPA alone. These studies reveal that VEGF and IGF-1 can have distinct inputs in the angiogenic process. In contrast to VEGF, IGF-1 stabilizes neovessels, which is dependent on Erk activity and associated with prolonged activation.
Collapse
Affiliation(s)
- Sarah Melissa P Jacobo
- From the Department of Ophthalmology, Harvard Medical School, The Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02115
| | - Andrius Kazlauskas
- From the Department of Ophthalmology, Harvard Medical School, The Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02115
| |
Collapse
|
25
|
Elsherif L, Ozler M, Zayed MA, Shen JH, Chernoff J, Faber JE, Parise LV. Potential compensation among group I PAK members in hindlimb ischemia and wound healing. PLoS One 2014; 9:e112239. [PMID: 25379771 PMCID: PMC4224450 DOI: 10.1371/journal.pone.0112239] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 10/14/2014] [Indexed: 12/04/2022] Open
Abstract
PAKs are serine/threonine kinases that regulate cytoskeletal dynamics and cell migration. PAK1 is activated by binding to the small EF hand protein, CIB1, or to the Rho GTPases Rac1 or Cdc42. The role of PAK1 in angiogenesis was established based only on in vitro studies and its role in angiogenesis in vivo has never been examined. Here we tested the hypothesis that PAK1 is an essential regulator of ischemic neovascularization (arteriogenesis and angiogenesis) and wound healing using a global PAK1 knockout mouse. Neovascularization was assessed using unilateral hindlimb ischemia. We found that plantar perfusion, limb use and appearance were not significantly different between 6-8 week old PAK1-/- and PAK1+/+ mice throughout the 21-day period following hindlimb ischemia; however a slightly delayed healing was observed in 16 week old PAK1-/- mice. In addition, the wound healing rate, as assessed with an ear punch assay, was unchanged in PAK1-/- mice. Surprisingly, however, we observed a notable increase in PAK2 expression and phosphorylation in ischemic gastrocnemius tissue from PAK1-/- but not PAK1+/+ mice. Furthermore, we observed higher levels of activated ERK2, but not AKT, in ischemic and non-ischemic muscle of PAK1-/- mice upon hindlimb ischemic injury. A group I PAK inhibitor, IPA3, significantly inhibited endothelial cell sprouting from aortic rings in both PAK1-/- and PAK1+/+ mice, implying that PAK2 is a potential contributor to this process. Taken together, our data indicate that while PAK1 has the potential to contribute to neovascularization and wound healing, PAK2 may functionally compensate when PAK1 is deficient.
Collapse
Affiliation(s)
- Laila Elsherif
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Mehmet Ozler
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Mohamed A. Zayed
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Jessica H. Shen
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Jonathan Chernoff
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, United States of America
| | - James E. Faber
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Leslie V. Parise
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
26
|
|
27
|
Morjen M, Honoré S, Bazaa A, Abdelkafi-Koubaa Z, Ellafi A, Mabrouk K, Kovacic H, El Ayeb M, Marrakchi N, Luis J. PIVL, a snake venom Kunitz-type serine protease inhibitor, inhibits in vitro and in vivo angiogenesis. Microvasc Res 2014; 95:149-56. [PMID: 25173589 DOI: 10.1016/j.mvr.2014.08.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 08/17/2014] [Accepted: 08/21/2014] [Indexed: 12/22/2022]
Abstract
Development and homeostasis of the vascular system requires integrin-promoting endothelial cell adhesion, migration and survival. Nowadays, integrins represent potential targets for pharmacological agents and open new avenues for the control of metastatic spread in the treatment of tumor malignancies. We have already reported that PIVL, a serine protease inhibitor isolated from Macrovipera lebetina venom, displays an anti-tumor effect through interference with integrin receptor function. Here, we report that PIVL inhibits human vascular endothelial cell adhesion and migration onto fibrinogen and fibronectin in a dose-dependent manner without any cytotoxicity. Furthermore, we show that PIVL increases microtubule dynamic instability in HMEC-1 transfected with EGFP-tagged α-tubulin. Using Matrigel™ and chick chorioallantoic membrane assays, we demonstrate that PIVL exhibits a strong anti-angiogenic effect both in vitro and in vivo. Interestingly, results herein reveal that the potent anti-angiogenic properties of PIVL are mediated by its RGD-like motif ((41)RGN(43)).
Collapse
Affiliation(s)
- Maram Morjen
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Tunisia.
| | - Stéphane Honoré
- Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, UMR_S 911, Marseille, France; APHM, Hôpital Timone, Service Pharmacie, Marseille, France
| | - Amine Bazaa
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Tunisia
| | | | - Ameneallah Ellafi
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Tunisia
| | - Kamel Mabrouk
- Equipe CROPS, Institut de Chimie Radicalaire - UMR 7273, Université d'Aix-Marseille, Site de Saint Jérôme, Av. Escadrille Normandie Niemen, 13397 Marseille, France
| | - Hervé Kovacic
- APHM, Hôpital Timone, Service Pharmacie, Marseille, France
| | - Mohamed El Ayeb
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Tunisia
| | - Naziha Marrakchi
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Tunisia; Faculté de Médecine de Tunis, Tunisia
| | - José Luis
- APHM, Hôpital Timone, Service Pharmacie, Marseille, France
| |
Collapse
|
28
|
Interleukin-1β-induced Wnt5a enhances human corneal endothelial cell migration through regulation of Cdc42 and RhoA. Mol Cell Biol 2014; 34:3535-45. [PMID: 25022753 DOI: 10.1128/mcb.01572-13] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Wnt5a can activate β-catenin-independent pathways for regulation of various cellular functions, such as migration, that play critical roles in wound repair. Investigation of Wnt5a signaling may help identify therapeutic targets for enhancing corneal endothelial wound healing that could provide an alternative to corneal transplantation in patients with blindness from endothelial dysfunction. However, Wnt5a signaling in corneal endothelial cells (CECs) has not been well characterized. In this study, we show transient induction of Wnt5a by interleukin-1β (IL-1β) stimulation proceeds through NF-κB in human CECs. This leads to binding of Fzd5 to Ror2, resulting in activation of disheveled protein (Dvl) and subsequently disheveled-associated activator of morphogenesis 1 (DAAM1). This leads to activation of Cdc42 and subsequent inhibition of RhoA. Inhibition of RhoA leads to parallel dephosphorylation and inactivation of LIM domain kinase 2 along with dephosphorylation and activation of slingshot 1, resulting in dephosphorylation and activation of cofilin and leading to enhanced cell migration. These findings suggest that Wnt5a enhances cell migration through activation of Cdc42 and inactivation of RhoA in human CECs.
Collapse
|
29
|
Zhou W, Fong MY, Min Y, Somlo G, Liu L, Palomares MR, Yu Y, Chow A, O'Connor STF, Chin AR, Yen Y, Wang Y, Marcusson EG, Chu P, Wu J, Wu X, Li AX, Li Z, Gao H, Ren X, Boldin MP, Lin PC, Wang SE. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 2014; 25:501-15. [PMID: 24735924 PMCID: PMC4016197 DOI: 10.1016/j.ccr.2014.03.007] [Citation(s) in RCA: 1100] [Impact Index Per Article: 110.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 08/23/2013] [Accepted: 03/07/2014] [Indexed: 12/11/2022]
Abstract
Cancer-secreted microRNAs (miRNAs) are emerging mediators of cancer-host crosstalk. Here we show that miR-105, which is characteristically expressed and secreted by metastatic breast cancer cells, is a potent regulator of migration through targeting the tight junction protein ZO-1. In endothelial monolayers, exosome-mediated transfer of cancer-secreted miR-105 efficiently destroys tight junctions and the integrity of these natural barriers against metastasis. Overexpression of miR-105 in nonmetastatic cancer cells induces metastasis and vascular permeability in distant organs, whereas inhibition of miR-105 in highly metastatic tumors alleviates these effects. miR-105 can be detected in the circulation at the premetastatic stage, and its levels in the blood and tumor are associated with ZO-1 expression and metastatic progression in early-stage breast cancer.
Collapse
Affiliation(s)
- Weiying Zhou
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA; Department of Pharmacology, College of Pharmacy, The Third Military Medical University, Chongqing, 400038, China
| | - Miranda Y Fong
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Yongfen Min
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - George Somlo
- Department of Medical Oncology, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Liang Liu
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA; Department of Biotherapy and Key Laboratory of Cancer Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Melanie R Palomares
- Department of Medical Oncology, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA; Department of Population Sciences, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Yang Yu
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA; Department of Biotherapy and Key Laboratory of Cancer Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Amy Chow
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | | | - Andrew R Chin
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA; City of Hope Irell & Manella Graduate School of Biological Sciences, Duarte, CA 91010, USA
| | - Yun Yen
- Department of Molecular Pharmacology, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA; Core of Translational Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Yafan Wang
- Core of Translational Research Laboratory, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Eric G Marcusson
- Oncology and Basic Mechanisms, Regulus Therapeutics, San Diego, CA 92121, USA
| | - Peiguo Chu
- Department of Pathology, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Jun Wu
- Department of Comparative Medicine, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Xiwei Wu
- Core of Integrative Genomics, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Arthur Xuejun Li
- Department of Information Science, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Zhuo Li
- Core of Electron Microscopy, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Hanlin Gao
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA; Core of Integrative Genomics, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | - Xiubao Ren
- Department of Biotherapy and Key Laboratory of Cancer Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Mark P Boldin
- Department of Molecular and Cellular Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA
| | | | - Shizhen Emily Wang
- Department of Cancer Biology, City of Hope Beckman Research Institute and Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
30
|
BNIP3 supports melanoma cell migration and vasculogenic mimicry by orchestrating the actin cytoskeleton. Cell Death Dis 2014; 5:e1127. [PMID: 24625986 PMCID: PMC3973222 DOI: 10.1038/cddis.2014.94] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 02/10/2014] [Accepted: 02/11/2014] [Indexed: 01/23/2023]
Abstract
BNIP3 is an atypical BH3-only member of the BCL-2 family of proteins with reported pro-death as well as pro-autophagic and cytoprotective functions, depending on the type of stress and cellular context. In line with this, the role of BNIP3 in cancer is highly controversial and increased BNIP3 levels in cancer patients have been linked with both good as well as poor prognosis. In this study, using small hairpin RNA (shRNA) lentiviral transduction to stably knockdown BNIP3 (BNIP3-shRNA) expression levels in melanoma cells, we show that BNIP3 supports cancer cell survival and long-term clonogenic growth. Although BNIP3-shRNA increased mitochondrial mass and baseline levels of reactive oxygen species production, which are features associated with aggressive cancer cell behavior, it also prevented cell migration and completely abolished the ability to form a tubular-like network on matrigel, a hallmark of vasculogenic mimicry (VM). We found that this attenuated aggressive behavior of these melanoma cells was underscored by severe changes in cell morphology and remodeling of the actin cytoskeleton associated with loss of BNIP3. Indeed, BNIP3-silenced melanoma cells displayed enhanced formation of actin stress fibers and membrane ruffles, while lamellopodial protrusions and filopodia, tight junctions and adherens junctions were reduced. Moreover, loss of BNIP3 resulted in re-organization of focal adhesion sites associated with increased levels of phosphorylated focal adhesion kinase. Remarkably, BNIP3 silencing led to a drop of the protein levels of the integrin-associated protein CD47 and its downstream signaling effectors Rac1 and Cdc42. These observations underscore that BNIP3 is required to maintain steady-state levels of intracellular complexes orchestrating the plasticity of the actin cytoskeleton, which is integral to cell migration and other vital processes stimulating cancer progression. All together these results unveil an unprecedented pro-tumorigenic role of BNIP3 driving melanoma cell's aggressive features, like migration and VM.
Collapse
|
31
|
Liu Y, Li H, Yan S, Wei J, Li X. Hepatocyte cocultures with endothelial cells and fibroblasts on micropatterned fibrous mats to promote liver-specific functions and capillary formation capabilities. Biomacromolecules 2014; 15:1044-54. [PMID: 24547870 DOI: 10.1021/bm401926k] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The maintenance of hepatocyte phenotype and functions remains as a great challenge in the generation of functional liver tissue and in vitro model for drug metabolism studies. The use of hepatocyte coculture systems plays essential roles in the establishment of cell-cell and cell-extracellular matrix communications similar to native liver tissues. In the current study, micropatterned electrospun fibrous mats were created to load hepatocytes, fibroblasts, and endothelial cells (ECs), which were precisely assembled to establish their spatially controlled coculture for mimicking the in vivo structure of hepatic lobules. Hepatocytes formed compact polyhedral spheroids with an average diameter of 80-100 μm, reorganized actin filaments in the cell-cell contact regions, and well-developed bile canaliculi. Compared with hepatocytes cultured alone, the coculture of hepatocytes with either fibroblasts or ECs led to significantly higher albumin secretion, urea synthesis and cytochrome P-450 expression, which were dramatically improved by the coculture of hepatocytes with both fibroblasts and ECs. The cocultured ECs well spread on patterned regions with little organized filamentous actin, and significantly higher densities and deeper penetration into patterned scaffolds were determined for ECs after coculture with fibroblasts and hepatocytes compared with those after cultured alone or coculture with either fibroblasts or hepatocytes. A Matrigel overlay assay showed that the capabilities of ECs to form capillary tubes were significantly enhanced by micropatterned coculture with fibroblasts and hepatocytes. Thus, the coculture of hepatocytes, fibroblasts, and ECs on micropatterned fibrous mats helps both hepatocytes in the maintenance of hepatic functions and ECs in the formation of capillary-like structures. It is suggested that the micropatterned coculture model described here not only provides functional hepatic tissues for predictions of drug metabolism profiles, but also will enable investigations on more complex and physiological cell-cell communications.
Collapse
Affiliation(s)
- Yaowen Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University , Chengdu 610031, People's Republic of China
| | | | | | | | | |
Collapse
|
32
|
Kim SJ, Wan Q, Cho E, Han B, Yoder MC, Voytik-Harbin SL, Na S. Matrix rigidity regulates spatiotemporal dynamics of Cdc42 activity and vacuole formation kinetics of endothelial colony forming cells. Biochem Biophys Res Commun 2014; 443:1280-5. [PMID: 24393843 DOI: 10.1016/j.bbrc.2013.12.135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 12/26/2013] [Indexed: 12/15/2022]
Abstract
Recent evidence has shown that endothelial colony forming cells (ECFCs) may serve as a cell therapy for improving blood vessel formation in subjects with vascular injury, largely due to their robust vasculogenic potential. The Rho family GTPase Cdc42 is known to play a primary role in this vasculogenesis process, but little is known about how extracellular matrix (ECM) rigidity affects Cdc42 activity during the process. In this study, we addressed two questions: Does matrix rigidity affect Cdc42 activity in ECFC undergoing early vacuole formation? How is the spatiotemporal activation of Cdc42 related to ECFC vacuole formation? A fluorescence resonance energy transfer (FRET)-based Cdc42 biosensor was used to examine the effects of the rigidity of three-dimensional (3D) collagen matrices on spatiotemporal activity of Cdc42 in ECFCs. Collagen matrix stiffness was modulated by varying the collagen concentration and therefore fibril density. The results showed that soft (150 Pa) matrices induced an increased level of Cdc42 activity compared to stiff (1 kPa) matrices. Time-course imaging and colocalization analysis of Cdc42 activity and vacuole formation revealed that Cdc42 activity was colocalized to the periphery of cytoplasmic vacuoles. Moreover, soft matrices generated faster and larger vacuoles than stiff matrices. The matrix-driven vacuole formation was enhanced by a constitutively active Cdc42 mutant, but significantly inhibited by a dominant-negative Cdc42 mutant. Collectively, the results suggest that matrix rigidity is a strong regulator of Cdc42 activity and vacuole formation kinetics, and that enhanced activity of Cdc42 is an important step in early vacuole formation in ECFCs.
Collapse
Affiliation(s)
- Seung Joon Kim
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Qiaoqiao Wan
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Eunhye Cho
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sherry L Voytik-Harbin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Sungsoo Na
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| |
Collapse
|
33
|
Salvati E, Zizza P, Rizzo A, Iachettini S, Cingolani C, D’Angelo C, Porru M, Randazzo A, Pagano B, Novellino E, Pisanu ME, Stoppacciaro A, Spinella F, Bagnato A, Gilson E, Leonetti C, Biroccio A. Evidence for G-quadruplex in the promoter of vegfr-2 and its targeting to inhibit tumor angiogenesis. Nucleic Acids Res 2013; 42:2945-57. [PMID: 24335081 PMCID: PMC3950687 DOI: 10.1093/nar/gkt1289] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Tumor angiogenesis is mainly mediated by vascular endothelial growth factor (VEGF), a pro-angiogenic factor produced by cancer cells and active on the endothelium through the VEGF receptor 2 (VEGFR-2). Here we identify a G-rich sequence within the proximal promoter region of vegfr-2, able to form an antiparallel G-quadruplex (G4) structure. This G4 structure can be efficiently stabilized by small molecules with the consequent inhibition of vegfr-2 expression. Functionally, the G4-mediated reduction of VEGFR-2 protein causes a switching off of signaling components that, converging on actin cytoskeleton, regulate the cellular events leading to endothelial cell proliferation, migration and differentiation. As a result of endothelial cell function impairment, angiogenic process is strongly inhibited by G4 ligands both in vitro and in vivo. Interestingly, the G4-mediated antiangiogenic effect seems to recapitulate that observed by using a specific interference RNA against vegfr-2, and it is strongly antagonized by overexpressing the vegfr-2 gene. In conclusion, we describe the evidence for the existence of G4 in the promoter of vegfr-2, whose expression and function can be markedly inhibited by G4 ligands, thereby revealing a new, and so far undescribed, way to block VEGFR-2 as target for anticancer therapy.
Collapse
Affiliation(s)
- Erica Salvati
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Pasquale Zizza
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Angela Rizzo
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Sara Iachettini
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Chiara Cingolani
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Carmen D’Angelo
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Manuela Porru
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Antonio Randazzo
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Bruno Pagano
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Ettore Novellino
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Maria Elena Pisanu
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Antonella Stoppacciaro
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Francesca Spinella
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Anna Bagnato
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Eric Gilson
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Carlo Leonetti
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
| | - Annamaria Biroccio
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy, Laboratory of Molecular Pathology, Department of Pharmacy, University of Naples “Federico II”, Naples, Italy, Department of Clinical and Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy, Laboratory of Molecular Pathology, Regina Elena National Cancer Institute, Rome, Italy, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France and Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, France
- *To whom correspondence should be addressed. Tel: +39 06 52662569; Fax: +39 06 52662592;
| |
Collapse
|
34
|
Rath S, Liebl J, Fürst R, Vollmar AM, Zahler S. Regulation of endothelial signaling and migration by v-ATPase. Angiogenesis 2013; 17:587-601. [PMID: 24254321 DOI: 10.1007/s10456-013-9408-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 11/12/2013] [Indexed: 01/04/2023]
Abstract
The vacuolar ATPase (v-ATPase) is a proton pump, able to acidify intracellular compartments and the pericellular space. v-ATPase has extensively been studied in various functional contexts, e.g., migration of tumor cells, and inhibition of v-ATPase has been proven as intriguing novel therapeutic concept. Since the role of v-ATPase in endothelial cell migration and angiogenesis has scarcely been investigated, we examined the consequences of pharmacological inhibition of v-ATPase (by concanamycin) on proliferation, migration, VEGF-receptor 2 (VEGFR2) trafficking and signaling, as well as Notch-mediated transcription in endothelial cells [human microvascular endothelial cells (HMEC-1) and human umbilical vein endothelial cells (HUVEC)] Treatment of the cells with 3 or 10 nM of the v-ATPase inhibitor concanamycin for 48 h or longer inhibited proliferation and arrested cell cycle in the G2/M phase in HMEC-1, while a G1 phase arrest occurred in HUVEC. Already after 24 h these concentrations reduced migration (scratch assay, chemotactic gradient). Activation of the small GTPase Rac1 in freshly adherent cells was reduced by concanamycin. Downstream signaling of the VEGFR2 (phosphorylation of ERK1/2 and AKT), as well as autophosphorylation of VEGFR2 were inhibited. VEGFR2 on the cell surface was reduced, and sequestered in a lysosomal compartment. In addition, concanamycin blocked transcription of the Notch target genes Hey1 and Hey2 after stimulation with DLL4. Since the impaired signaling pathways (Rac-1, VEGFR2, Notch) all depend on vesicular recycling circuits, we conclude that the disturbance of these is the main mode of action of v-ATPase inhibition in endothelial cells, offering an attractive multi-factorial anti-angiogenic approach.
Collapse
Affiliation(s)
- Sebastian Rath
- Department of Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377, Munich, Germany
| | | | | | | | | |
Collapse
|
35
|
Zhou Q, Anderson C, Zhang H, Li X, Inglis F, Jayagopal A, Wang S. Repression of choroidal neovascularization through actin cytoskeleton pathways by microRNA-24. Mol Ther 2013; 22:378-389. [PMID: 24297048 DOI: 10.1038/mt.2013.243] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 10/10/2013] [Indexed: 01/15/2023] Open
Abstract
Actin cytoskeleton is critical for cell motility and division, both of which are important for angiogenesis. MicroRNAs (miRNA/miR) are emerging as pivotal modulators of vascular development and disease. How miRNAs regulate actin cytoskeleton dynamics in endothelial cells (EC) and neovascularization is still unclear. Here, we report that miR-24 regulates actin dynamics in ECs through targeting multiple members downstream of Rho signaling, including Pak4, Limk2, and Diaph1 proteins. Overexpression of miR-24 in ECs blocks stress fiber and lamellipodia formation, represses EC migration, proliferation, and tube formation in vitro, as well as angiogenesis in an ex vivo aortic ring assay. Moreover, subretinal delivery of miR-24 mimics represses laser-induced choroidal neovascularization (CNV) in vivo. Mechanistically, knockdown of miR-24 target protein LIMK2 or PAK4 inhibits stress fiber formation and tube formation in vitro, mimicking miR-24 overexpression phenotype in angiogenesis, while overexpression of LIMK2 and PAK4 by adenoviruses partially rescued the tube formation defects in miR-24 overexpressing ECs. Taken together, these findings suggest that miR-24 represses angiogenesis by simultaneously regulating multiple components in the actin cytoskeleton pathways. Manipulation of actin cytoskeleton pathways by miR-24 may represent an attractive therapeutic solution for the treatment of wet age-related macular degeneration (AMD) and other vascular diseases.
Collapse
Affiliation(s)
- Qinbo Zhou
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, USA
| | - Chastain Anderson
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, USA
| | - Hongmei Zhang
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xinyu Li
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Fiona Inglis
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, USA
| | - Ashwath Jayagopal
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Shusheng Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, USA; Department of Ophthalmology, Tulane University, New Orleans, Louisiana, USA.
| |
Collapse
|
36
|
Bid HK, Roberts RD, Manchanda PK, Houghton PJ. RAC1: an emerging therapeutic option for targeting cancer angiogenesis and metastasis. Mol Cancer Ther 2013; 12:1925-34. [PMID: 24072884 DOI: 10.1158/1535-7163.mct-13-0164] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Angiogenesis and metastasis are well recognized as processes fundamental to the development of malignancy. Both processes involve the coordination of multiple cellular and chemical activities through myriad signaling networks, providing a mass of potential targets for therapeutic intervention. This review will focus on one master regulator of cell motility, RAC1, and the existing data with regard to its role in cell motility, including particular roles for tumor angiogenesis and invasion/metastasis. We also emphasize the preclinical investigations carried out with RAC1 inhibitors to evaluate the therapeutic potential of this target. Herein, we explore potential future directions as well as the challenges of targeting RAC1 in the treatment of cancer. Recent insights at the molecular and cellular levels are paving the way for a more directed and detailed approach to target mechanisms of RAC1 regulating angiogenesis and metastasis. Understanding these mechanisms may provide insight into RAC1 signaling components as alternative therapeutic targets for tumor angiogenesis and metastasis.
Collapse
Affiliation(s)
- Hemant K Bid
- Corresponding Author: Peter J. Houghton, Center for Childhood Cancer, The Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205.
| | | | | | | |
Collapse
|
37
|
Tung JJ, Tattersall IW, Kitajewski J. Tips, stalks, tubes: notch-mediated cell fate determination and mechanisms of tubulogenesis during angiogenesis. Cold Spring Harb Perspect Med 2013; 2:a006601. [PMID: 22355796 DOI: 10.1101/cshperspect.a006601] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Angiogenesis is the process of developing vascular sprouts from existing blood vessels. Luminal endothelial cells convert into "tip" cells that contribute to the development of a multicellular stalk, which then undergoes lumen formation. In this review, we consider a variety of cellular and molecular pathways that mediate these transitions. We focus first on Notch signaling in cell fate determination as a mechanism to define tip and stalk cells. We next discuss the current models of lumen formation and describe new players in this process, such as chloride intracellular channel proteins. Finally, we consider the possible medical therapeutic benefits of understanding these processes and acknowledge potential obstacles in drug development.
Collapse
Affiliation(s)
- Jennifer J Tung
- Department of Obstetrics/Gynecology and Pathology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA
| | | | | |
Collapse
|
38
|
Bryant PW, Zheng Q, Pumiglia KM. Focal adhesion kinase is a phospho-regulated repressor of Rac and proliferation in human endothelial cells. Biol Open 2012; 1:723-30. [PMID: 23213465 PMCID: PMC3507225 DOI: 10.1242/bio.20121008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 05/15/2012] [Indexed: 11/06/2022] Open
Abstract
Focal adhesion kinase (FAK) is critically positioned to integrate signals from the extracellular matrix and cellular adhesion. It is essential for normal vascular development and has been implicated in a wide range of cellular functions including the regulation of cell proliferation, migration, differentiation, and survival. It is currently being actively targeted therapeutically using different approaches. We have used human endothelial cells as a model system to compare the effects of inhibiting FAK through several different approaches including dominant negatives, kinase inhibitors and shRNA. We find that manipulations of FAK signaling that result in inhibition of FAK 397 phosphorylation inhibit proliferation and migration. However, abolition of FAK expression using stable (shRNA) or transient (siRNA) approaches does not interfere with these cellular functions. The ability to regulate cell proliferation by FAK manipulation is correlated with the activation status of Rac, an essential signal for the regulation of cyclin-dependent kinase inhibitors. The knockdown of FAK, while not affecting cellular proliferation or migration, dramatically interferes with vascular morphogenesis and survival, mirroring in vivo findings. We propose a novel model of FAK signaling whereby one of the multifunctional roles of FAK as a signaling protein includes FAK as a phospho-regulated repressor of Rac activation, with important implications on interpretation of research experiments and therapeutic development.
Collapse
Affiliation(s)
- Patrick W Bryant
- Cell Biology and Cancer Research, Albany Medical College , Albany, NY 12208 , USA
| | | | | |
Collapse
|
39
|
Zhu W, Nelson CM. PI3K signaling in the regulation of branching morphogenesis. Biosystems 2012; 109:403-11. [PMID: 22525052 DOI: 10.1016/j.biosystems.2012.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2012] [Revised: 04/03/2012] [Accepted: 04/11/2012] [Indexed: 11/25/2022]
Abstract
Branching morphogenesis drives the formation of epithelial organs including the mammary gland, lung, kidney, salivary gland and prostate. Branching at the cellular level also drives development of the nervous and vascular systems. A variety of signaling pathways are orchestrated together to establish the pattern of these branched organs. The phosphoinositide 3-kinase (PI3K) signaling network is of particular interest because of the diverse outcomes it generates, including proliferation, motility, growth, survival and cell death. Here, we focus on the role of the PI3K pathway in the development of branched tissues. Cultured cells, explants and transgenic mice have revealed that the PI3K pathway is critical for the regulation of cell proliferation, apoptosis and motility during branching of tissues.
Collapse
Affiliation(s)
- Wenting Zhu
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | | |
Collapse
|
40
|
Zhu J, He P, Lin L, Jones DR, Marchant RE. Biomimetic poly(ethylene glycol)-based hydrogels as scaffolds for inducing endothelial adhesion and capillary-like network formation. Biomacromolecules 2012; 13:706-13. [PMID: 22296572 PMCID: PMC3310151 DOI: 10.1021/bm201596w] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The extracellular matrix (ECM) is an attractive model for designing synthetic scaffolds with a desirable environment for tissue engineering. Here, we report on the synthesis of ECM-mimetic poly(ethylene glycol) (PEG) hydrogels for inducing endothelial cell (EC) adhesion and capillary-like network formation. A collagen type I-derived peptide GPQGIAGQ (GIA)-containing PEGDA (GIA-PEGDA) was synthesized with the collagenase-sensitive GIA sequence attached in the middle of the PEGDA chain, which was then copolymerized with RGD capped-PEG monoacrylate (RGD-PEGMA) to form biomimetic hydrogels. The hydrogels degraded in vitro with the rate dependent on the concentration of collagenase and also supported the adhesion of human umbilical vein ECs (HUVECs). Biomimetic RGD/GIA-PEGDA hydrogels with incorporation of 1% RGD-PEGDA into GIA-PEGDA hydrogels induced capillary-like organization when HUVECs were seeded on the hydrogel surface, while RGD/PEGDA and GIA-PEGDA hydrogels did not. These results indicate that both cell adhesion and biodegradability of scaffolds play important roles in the formation of capillary-like networks.
Collapse
Affiliation(s)
- Junmin Zhu
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| | - Ping He
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| | - Lin Lin
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| | - Derek R. Jones
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| | - Roger E. Marchant
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| |
Collapse
|
41
|
Kasuya J, Sudo R, Mitaka T, Ikeda M, Tanishita K. Spatio-temporal control of hepatic stellate cell-endothelial cell interactions for reconstruction of liver sinusoids in vitro. Tissue Eng Part A 2012; 18:1045-56. [PMID: 22220631 DOI: 10.1089/ten.tea.2011.0351] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Vascularization of engineered tissues in vitro remains a major challenge in liver tissue engineering. Liver microvessels, termed liver sinusoids, have highly specialized structures, and recapturing these sinusoidal structures is essential for reconstruction of functional liver tissue in vitro. Liver sinusoids are composed of hepatocytes, hepatic stellate cells (HSCs), and endothelial cells (ECs). Direct HSC-EC contacts are increasingly recognized for their roles in EC capillary morphogenesis. However, the hypothetical role of HSC-EC contacts in morphogenesis remains unclear in hepatocyte-HSC-EC triculture. In the present study, we first determined the effects of direct HSC-EC contacts on EC capillary morphogenesis using a hepatocyte-HSC-EC triculture model where HSC behavior was spatially controlled to achieve HSC-mediated proximal layers of hepatocytes and ECs. EC capillary morphogenesis was induced by overlaying Matrigel on an EC layer. Direct HSC-EC contacts inhibited EC capillary morphogenesis, suggesting that the HSC-EC contacts may be an important factor in capillary formation. We next tested the hypothesis that, in addition to spatial control, temporal control of HSC behavior is also important in achieving capillary morphogenesis in the triculture. ECs responded to the induction of capillary morphogenesis before the formation of direct HSC-EC contacts, while the ECs remained to form monolayers when capillary morphogenesis was induced after the HSC-EC contacts were established. When capillary morphogenesis was successfully achieved in the triculture, HSCs tended to preferably localize near the preformed capillary-like structures, resulting in the reconstruction of liver sinusoidal structures. In these structures, hepatocyte maturation was induced. Our findings indicate that control, both spatial and temporal, of HSC behavior is a key engineering strategy for the vascularization of engineered liver tissue in vitro.
Collapse
Affiliation(s)
- Junichi Kasuya
- Center for System Integration Engineering, School of Integrated Design Engineering, Keio University, Kohoku-ku, Yokohama, Japan.
| | | | | | | | | |
Collapse
|
42
|
Liu J, Zeng L, Kennedy RM, Gruenig NM, Childs SJ. βPix plays a dual role in cerebral vascular stability and angiogenesis, and interacts with integrin αvβ8. Dev Biol 2011; 363:95-105. [PMID: 22206757 DOI: 10.1016/j.ydbio.2011.12.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 12/09/2011] [Accepted: 12/14/2011] [Indexed: 11/19/2022]
Abstract
The growth of new blood vessels by angiogenesis and their stabilization by the recruitment of perivascular mural cells are thought to be two sequential, yet independent events. Here we identify molecular links between both processes through the βPix and integrin α(v)β(8) proteins. Bubblehead (bbh) mutants with a genetic mutation in βPix show defective vascular stabilization. βPix is a guanine nucleotide exchange factor and scaffold protein that binds many proteins including Git1, which bridges βPix to integrins at focal adhesions. Here we show that the ability of βPix to stabilize vessels requires Git1 binding residues. Knockdown of Git1 leads to a hemorrhage phenotype similar to loss of integrin α(v), integrin β(8) or βPix, suggesting that vascular stabilization through βPix involves interactions with integrins. Furthermore, double loss of function of βPix and integrin α(v) shows enhanced hemorrhage rates. Not only is vascular stability impaired in these embryos, but we also uncover a novel role of both βPix and integrin α(v)β(8) in cerebral angiogenesis. Downregulation of either βPix or integrin α(v)β(8) results in fewer and morphologically abnormal cerebral arteries penetrating the hindbrain. We show that this is coupled with a significant reduction in endothelial cell proliferation in bbh mutants or integrin α(v)β(8) morphants. These data suggest that a complex involving βPix, GIT1 and integrin α(v)β(8) may regulate vascular stability, cerebral angiogenesis and endothelial cell proliferation in the developing embryo.
Collapse
Affiliation(s)
- Jing Liu
- Biochemistry and Molecular Biology, and Smooth Muscle Research Group, University of Calgary, AB, Canada
| | | | | | | | | |
Collapse
|
43
|
Johnson JA, Hemnes AR, Perrien DS, Schuster M, Robinson LJ, Gladson S, Loibner H, Bai S, Blackwell TR, Tada Y, Harral JW, Talati M, Lane KB, Fagan KA, West J. Cytoskeletal defects in Bmpr2-associated pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2011; 302:L474-84. [PMID: 22180660 DOI: 10.1152/ajplung.00202.2011] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The heritable form of pulmonary arterial hypertension (PAH) is typically caused by a mutation in bone morphogenic protein receptor type 2 (BMPR2), and mice expressing Bmpr2 mutations develop PAH with features similar to human disease. BMPR2 is known to interact with the cytoskeleton, and human array studies in PAH patients confirm alterations in cytoskeletal pathways. The goal of this study was to evaluate cytoskeletal defects in BMPR2-associated PAH. Expression arrays on our Bmpr2 mutant mouse lungs revealed cytoskeletal defects as a prominent molecular consequence of universal expression of a Bmpr2 mutation (Rosa26-Bmpr2(R899X)). Pulmonary microvascular endothelial cells cultured from these mice have histological and functional cytoskeletal defects. Stable transfection of different BMPR2 mutations into pulmonary microvascular endothelial cells revealed that cytoskeletal defects are common to multiple BMPR2 mutations and are associated with activation of the Rho GTPase, Rac1. Rac1 defects are corrected in cell culture and in vivo through administration of exogenous recombinant human angiotensin-converting enzyme 2 (rhACE2). rhACE2 reverses 77% of gene expression changes in Rosa26-Bmpr2(R899X) transgenic mice, in particular, correcting defects in cytoskeletal function. Administration of rhACE2 to Rosa26-Bmpr2(R899X) mice with established PAH normalizes pulmonary pressures. Together, these findings suggest that cytoskeletal function is central to the development of BMPR2-associated PAH and that intervention against cytoskeletal defects may reverse established disease.
Collapse
Affiliation(s)
- Jennifer A Johnson
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2650, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Examining the role of Rac1 in tumor angiogenesis and growth: a clinically relevant RNAi-mediated approach. Angiogenesis 2011; 14:457-66. [PMID: 21789714 DOI: 10.1007/s10456-011-9229-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 07/15/2011] [Indexed: 12/30/2022]
Abstract
Angiogenesis, the sprouting of new blood vessels from the pre-existing vasculature, is a well established target in anti-cancer therapy. It is thought that the Rho GTPase Rac1 is required during vascular endothelial growth factor (VEGF)-mediated angiogenesis. In the present study, we have used a clinically relevant RNA interference approach to silence Rac1 expression. Human umbilical vein endothelial cells were transiently transfected with non-specific control siRNA (siNS) or Rac1 siRNA (siRac1) using electroporation or Lipofectamine 2000. Functional assays with transfected endothelial cells were performed to determine the effect of Rac1 knockdown on angiogenesis in vitro. Silencing of Rac1 inhibited VEGF-mediated tube formation, cell migration, invasion and proliferation. In addition, treatment with Rac1 siRNA inhibited angiogenesis in an in vivo Matrigel plug assay. Intratumoral injections of siRac1 almost completely inhibited the growth of grafted Neuro2a tumors and reduced tumor angiogenesis. Together, these data indicate that Rac1 is an important regulator of VEGF-mediated angiogenesis. Knockdown of Rac1 may represent an attractive approach to inhibit tumor angiogenesis and growth.
Collapse
|
45
|
Xu K, Sacharidou A, Fu S, Chong DC, Skaug B, Chen ZJ, Davis GE, Cleaver O. Blood vessel tubulogenesis requires Rasip1 regulation of GTPase signaling. Dev Cell 2011; 20:526-39. [PMID: 21396893 DOI: 10.1016/j.devcel.2011.02.010] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Revised: 01/06/2011] [Accepted: 02/23/2011] [Indexed: 01/30/2023]
Abstract
Cardiovascular function depends on patent blood vessel formation by endothelial cells (ECs). However, the mechanisms underlying vascular "tubulogenesis" are only beginning to be unraveled. We show that endothelial tubulogenesis requires the Ras interacting protein 1, Rasip1, and its binding partner, the RhoGAP Arhgap29. Mice lacking Rasip1 fail to form patent lumens in all blood vessels, including the early endocardial tube. Rasipl null angioblasts fail to properly localize the polarity determinant Par3 and display defective cell polarity, resulting in mislocalized junctional complexes and loss of adhesion to extracellular matrix (ECM). Similarly, depletion of either Rasip1 or Arhgap29 in cultured ECs blocks in vitro lumen formation, fundamentally alters the cytoskeleton, and reduces integrin-dependent adhesion to ECM. These defects result from increased RhoA/ROCK/myosin II activity and blockade of Cdc42 and Rac1 signaling. This study identifies Rasip1 as a unique, endothelial-specific regulator of Rho GTPase signaling, which is essential for blood vessel morphogenesis.
Collapse
Affiliation(s)
- Ke Xu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Uncovering the behaviors of individual cells within a multicellular microvascular community. Proc Natl Acad Sci U S A 2011; 108:5133-8. [PMID: 21383144 DOI: 10.1073/pnas.1007508108] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although individual cells vary in behavior during the formation of tissues, the nature of such variations are largely uncharacterized. Here, we tracked the morphologies and motilities of ~300 human endothelial cells from an initial dispersed state to the formation of capillary-like structures, distilling the dynamics of tissue morphogenesis into an array of ~36,000 numerical phenotypes. Quantitative analysis of population averages revealed two previously unidentified phases in which the cells spread before forming connections with neighboring cells and where the microvascular plexus stabilized before spatially reorganizing. Analysis at the single-cell level showed that in contrast to the population-averaged behavior, most cells followed distinct temporal patterns that were not reflected in the bulk average. Interestingly, some of these behavioral patterns correlated to the cells' final structural role within the plexus. Knowledge of how individual cells or groups of cells behave enhances our understanding of how native tissues self-organize and could ultimately enable more precise approaches for engineering tissues and synthesizing multicellular communities.
Collapse
|
47
|
MacKenzie AE, Lappin JE, Taylor DL, Nicklin SA, Milligan G. GPR35 as a Novel Therapeutic Target. Front Endocrinol (Lausanne) 2011; 2:68. [PMID: 22654822 PMCID: PMC3356001 DOI: 10.3389/fendo.2011.00068] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 10/16/2011] [Indexed: 12/21/2022] Open
Abstract
G protein-coupled receptors (GPCRs) remain the best studied class of cell surface receptors and the most tractable family of proteins for novel small molecule drug discovery. Despite this, a considerable number of GPCRs remain poorly characterized and in a significant number of cases, endogenous ligand(s) that activate them remain undefined or are of questionable physiological relevance. GPR35 was initially discovered over a decade ago but has remained an "orphan" receptor. Recent publications have highlighted novel ligands, both endogenously produced and synthetic, which demonstrate significant potency at this receptor. Furthermore, evidence is accumulating which highlights potential roles for GPR35 in disease and therefore, efforts to characterize GPR35 more fully and develop it as a novel therapeutic target in conditions that range from diabetes and hypertension to asthma are increasing. Recently identified ligands have shown marked species selective properties, indicating major challenges for future drug development. As we begin to understand these issues, the continuing efforts to identify novel agonist and antagonist ligands for GPR35 will help to decipher its true physiological relevance; translating multiple assay systems in vitro, to animal disease systems in vivo and finally to man.
Collapse
Affiliation(s)
- A. E. MacKenzie
- Molecular Pharmacology Group, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgow, UK
| | - J. E. Lappin
- Molecular Pharmacology Group, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgow, UK
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgow, UK
| | - D. L. Taylor
- Centre for Therapeutics Discovery, MRC TechnologyLondon, UK
| | - S. A. Nicklin
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgow, UK
| | - G. Milligan
- Molecular Pharmacology Group, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgow, UK
- *Correspondence: G. Milligan, Molecular Pharmacology Group, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland G12 8QQ, UK. e-mail:
| |
Collapse
|
48
|
Cseh B, Fernandez-Sauze S, Grall D, Schaub S, Doma E, Van Obberghen-Schilling E. Autocrine fibronectin directs matrix assembly and crosstalk between cell–matrix and cell–cell adhesion in vascular endothelial cells. J Cell Sci 2010; 123:3989-99. [DOI: 10.1242/jcs.073346] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Cellular fibronectin (cFN) variants harboring extra FN type 3 repeats, namely extra domains B and A, are major constituents of the extracellular matrix around newly forming blood vessels during development and angiogenesis. Their expression is induced by angiogenic stimuli and their assembly into fibrillar arrays is driven by cell-generated tension at α5β1 integrin-based adhesions. Here, we examined the role and functional redundancy of cFN variants in cultured endothelial cells by isoform-selective RNA interference. We show that FN fibrillogenesis is a cell-autonomous process whereby basally directed secretion and assembly of cellular FN are tightly coupled events that play an important role not only in signaling at cell–matrix adhesions but also at cell–cell contacts. Silencing of cFN variants differentially affects integrin usage, cell spreading, motility and capillary morphogenesis in vitro. cFN-deficient cells undergo a switch from α5β1- to αvβ3-based adhesion, accompanied by a Src-regulated disruption of adherens junctions. These studies identify a crucial role for autocrine FN in subendothelial matrix assembly and junctional integrity that provides spatially and temporally restricted control of endothelial plasticity during angiogenic blood vessel remodeling.
Collapse
Affiliation(s)
- Botond Cseh
- University of Nice-Sophia Antipolis, CNRS UMR 6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| | - Samantha Fernandez-Sauze
- University of Nice-Sophia Antipolis, CNRS UMR 6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| | - Dominique Grall
- University of Nice-Sophia Antipolis, CNRS UMR 6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| | - Sébastien Schaub
- University of Nice-Sophia Antipolis, CNRS UMR 6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| | - Eszter Doma
- University of Nice-Sophia Antipolis, CNRS UMR 6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| | - Ellen Van Obberghen-Schilling
- University of Nice-Sophia Antipolis, CNRS UMR 6543, Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France
| |
Collapse
|
49
|
Mori T, Ono K, Kariya Y, Ogawa T, Higashi S, Miyazaki K. Laminin-3B11, a novel vascular-type laminin capable of inducing prominent lamellipodial protrusions in microvascular endothelial cells. J Biol Chem 2010; 285:35068-78. [PMID: 20805229 PMCID: PMC2966121 DOI: 10.1074/jbc.m110.146126] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 08/09/2010] [Indexed: 11/06/2022] Open
Abstract
The basement membrane (BM) proteins laminins, which consist of α, β, and γ chains, support tissue structures and cellular functions. To date only α4 and α5 types of laminins have been identified in the BMs of blood vessels. Our recent study suggested the presence of novel α3B-containing laminins in vascular BMs. Here we identified and characterized the third member of vascular laminins, laminin-3B11 (Lm3B11). RT-PCR analysis showed that microvascular endothelial (MVE) cells and umbilical vein endothelial cells expressed the messages for the α3B, β1, β2, and γ1 chains. In the culture of MVE cells, α3B was associated with β1 and γ1, producing Lm3B11. Recombinant Lm3B11 was overexpressed by introducing the cDNAs of the three chains into HEK-293 cells and purified to homogeneity. Purified Lm3B11 exhibited relatively weak cell adhesion activity through both α3β1 and α6β1 integrins. Most characteristically, Lm3B11 strongly stimulated MVE cells to extend many lamellipodial protrusions. This pseudopodial branching was blocked by an inhibitor for Src or phosphatidylinositol 3-kinase. Consistently, Lm3B11 stimulated the phosphorylation of Src and Akt more strongly than other laminins, suggesting that the integrin-derived signaling is mediated by these factors. The unique activity of Lm3B11 appears to be favorable to the branching of capillaries and venules.
Collapse
Affiliation(s)
- Taizo Mori
- From the Graduate School of Integrated Sciences and
| | - Kota Ono
- From the Graduate School of Integrated Sciences and
| | - Yoshinobu Kariya
- the Division of Cell Biology, Kihara Institute for Biological Research, Yokohama City University, 641-12 Maioka-cho, Totsuka-ku, Yokohama 244-0813, Japan
| | - Takashi Ogawa
- the Division of Cell Biology, Kihara Institute for Biological Research, Yokohama City University, 641-12 Maioka-cho, Totsuka-ku, Yokohama 244-0813, Japan
| | - Shouichi Higashi
- From the Graduate School of Integrated Sciences and
- the Division of Cell Biology, Kihara Institute for Biological Research, Yokohama City University, 641-12 Maioka-cho, Totsuka-ku, Yokohama 244-0813, Japan
| | - Kaoru Miyazaki
- From the Graduate School of Integrated Sciences and
- the Division of Cell Biology, Kihara Institute for Biological Research, Yokohama City University, 641-12 Maioka-cho, Totsuka-ku, Yokohama 244-0813, Japan
| |
Collapse
|
50
|
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: 2.7] [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
|