1
|
Du J, Liu P, Zhou Y, Misener S, Sharma I, Leeaw P, Thomson BR, Jin J, Quaggin SE. The mechanosensory channel PIEZO1 functions upstream of angiopoietin/TIE/FOXO1 signaling in lymphatic development. J Clin Invest 2024; 134:e176577. [PMID: 38747287 PMCID: PMC11093609 DOI: 10.1172/jci176577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/12/2024] [Indexed: 05/19/2024] Open
Abstract
Lymphedema is a debilitating disease with no effective cure and affects an estimated 250 million individuals worldwide. Prior studies have identified mutations in piezo-type mechanosensitive ion channel component 1 (PIEZO1), angiopoietin 2 (ANGPT2), and tyrosine kinase with Ig-like and EGF-like domains 1 (TIE1) in patients with primary lymphedema. Here, we identified crosstalk between these molecules and showed that activation of the mechanosensory channel PIEZO1 in lymphatic endothelial cells (LECs) caused rapid exocytosis of the TIE ligand ANGPT2, ectodomain shedding of TIE1 by disintegrin and metalloproteinase domain-containing protein 17 (ADAM17), and increased TIE/PI3K/AKT signaling, followed by nuclear export of the transcription factor FOXO1. These data establish a functional network between lymphedema-associated genes and provide what we believe to be the first molecular mechanism bridging channel function with vascular signaling and intracellular events culminating in transcriptional regulation of genes expressed in LECs. Our study provides insights into the regulation of lymphatic function and molecular pathways involved in human disease.
Collapse
Affiliation(s)
- Jing Du
- Feinberg Cardiovascular and Renal Research Institute
| | - Pan Liu
- Feinberg Cardiovascular and Renal Research Institute
| | - Yalu Zhou
- Feinberg Cardiovascular and Renal Research Institute
| | - Sol Misener
- Feinberg Cardiovascular and Renal Research Institute
| | - Isha Sharma
- Feinberg Cardiovascular and Renal Research Institute
| | - Phoebe Leeaw
- Feinberg Cardiovascular and Renal Research Institute
| | - Benjamin R. Thomson
- Feinberg Cardiovascular and Renal Research Institute
- Department of Ophthalmology, and
| | - Jing Jin
- Feinberg Cardiovascular and Renal Research Institute
- Division of Nephrology, Department of Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Susan E. Quaggin
- Feinberg Cardiovascular and Renal Research Institute
- Division of Nephrology, Department of Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
2
|
Meltzer M, Eliash N, Azoulay Z, Hadad U, Papo N. In vitro inhibition of cancer angiogenesis and migration by a nanobody that targets the orphan receptor Tie1. Cell Mol Life Sci 2022; 79:312. [PMID: 35604495 PMCID: PMC11072481 DOI: 10.1007/s00018-022-04336-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/28/2022]
Abstract
The human signaling molecules Tie1 and Tie2 receptor tyrosine kinases (RTKs) play important pathophysiological roles in many diseases, including different cancers. The activity of Tie1 is mediated mainly through the downstream angiopoietin-1 (Ang1)-dependent activation of Tie2, rendering both Tie 1 and the Tie1/Tie2/Ang1 axis attractive putative targets for therapeutic intervention. However, the development of inhibitors that target Tie1 and an understanding of their effect on Tie2 and on the Tie1/Tie2/Ang1 axis remain unfulfilled tasks, due, largely, to the facts that Tie1 is an orphan receptor and is difficult to produce and use in the quantities required for immune antibody library screens. In a search for a selective inhibitor of this orphan receptor, we sought to exploit the advantages (e.g., small size that allows binding to hidden epitopes) of non-immune nanobodies and to simultaneously overcome their limitations (i.e., low expression and stability). We thus performed expression, stability, and affinity screens of yeast-surface-displayed naïve and predesigned synthetic (non-immune) nanobody libraries against the Tie1 extracellular domain. The screens yielded a nanobody with high expression and good affinity and specificity for Tie1, thereby yielding preferential binding for Tie1 over Tie2. The stability, selectivity, potency, and therapeutic potential of this synthetic nanobody were profiled using in vitro and cell-based assays. The nanobody triggered Tie1-dependent inhibition of RTK (Tie2, Akt, and Fak) phosphorylation and angiogenesis in endothelial cells, as well as suppression of human glioblastoma cell viability and migration. This study opens the way to developing nanobodies as therapeutics for different cancers associated with Tie1 activation.
Collapse
Affiliation(s)
- May Meltzer
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, 1 Ben-Gurion Avenue, 8410501, Beer-Sheva, Israel
| | - Noam Eliash
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, 1 Ben-Gurion Avenue, 8410501, Beer-Sheva, Israel
| | - Ziv Azoulay
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, 1 Ben-Gurion Avenue, 8410501, Beer-Sheva, Israel
| | - Uzi Hadad
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Niv Papo
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, 1 Ben-Gurion Avenue, 8410501, Beer-Sheva, Israel.
| |
Collapse
|
3
|
Angiopoietin-2-induced lymphatic endothelial cell migration drives lymphangiogenesis via the β1 integrin-RhoA-formin axis. Angiogenesis 2022; 25:373-396. [PMID: 35103877 DOI: 10.1007/s10456-022-09831-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 11/08/2021] [Indexed: 11/01/2022]
Abstract
Lymphangiogenesis is an essential physiological process but also a determining factor in vascular-related pathological conditions. Angiopoietin-2 (Ang2) plays an important role in lymphatic vascular development and function and its upregulation has been reported in several vascular-related diseases, including cancer. Given the established role of the small GTPase RhoA on cytoskeleton-dependent endothelial functions, we investigated the relationship between RhoA and Ang2-induced cellular activities. This study shows that Ang2-driven human dermal lymphatic endothelial cell migration depends on RhoA. We demonstrate that Ang2-induced migration is independent of the Tie receptors, but dependent on β1 integrin-mediated RhoA activation with knockdown, pharmacological approaches, and protein sequencing experiments. Although the key proteins downstream of RhoA, Rho kinase (ROCK) and myosin light chain, were activated, blockade of ROCK did not abrogate the Ang2-driven migratory effect. However, formins, an alternative target of RhoA, were identified as key players, and especially FHOD1. The Ang2-RhoA relationship was explored in vivo, where lymphatic endothelial RhoA deficiency blocked Ang2-induced lymphangiogenesis, highlighting RhoA as an important target for anti-lymphangiogenic treatments.
Collapse
|
4
|
Crook T, Patil D, Nagarkar R, Gaya A, Plowman N, Limaye S, Srivastava N, Akolkar D, Ranade A, Bhatt A, Datta V, Bose C, Apurwa S, Patil S, Kumar P, Srinivasan A, Datar R. Angiogenesis Inhibitors in Personalized Combination Regimens for the Treatment of Advanced Refractory Cancers. FRONTIERS IN MOLECULAR MEDICINE 2021; 1:749283. [PMID: 39087078 PMCID: PMC11285706 DOI: 10.3389/fmmed.2021.749283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/02/2021] [Indexed: 08/02/2024]
Abstract
Background: Angiogenic factors are commonly activated in solid tumors and present a viable therapeutic target. However, anticancer treatment with angiogenesis inhibitors (AGI) is limited to a few cancers, mostly as monotherapy and not selected based on molecular indications. We aimed to determine whether patient-specific combination regimens with AGI and other anticancer agents when selected based on multi-analyte tumor interrogation (ETA: Encyclopedic Tumor Analysis) can expand the scope of AGIs in advanced refractory solid organ cancers with improved treatment responses. Methods: We evaluated treatment outcomes in 60 patients with advanced, refractory solid organ cancers who received ETA-guided combination regimens of AGI with other targeted, endocrine or cytotoxic agents. Radiological evaluation of treatment response was followed by determination of Objective Response Rate (ORR), Disease Control Rate (DCR), Progression Free Survival (PFS) and Overall Survival (OS). Results: Among the 60 patients, Partial Response (PR) was observed in 28 cases (46.7%), Stable Disease (SD) was observed in 29 cases (48.3%) and Disease Progression (PD, within 60 days) was observed in 3 cases (5.0%). The ORR was 46.7% and DCR was 95.0%. At the most recent follow-up the median PFS (mPFS) was 5.0 months and median OS (mOS) was 8.9 months. There were no Grade 4 therapy related adverse events or treatment related deaths. Conclusion: ETA-guided patient-specific combination regimens with AGI and other anti-neoplastic agents, can yield improved outcomes over AGI monotherapy. Trial Registration: Details of all trials are available at WHO-ICTRP: https://apps.who.int/trialsearch/. RESILIENT ID CTRI/2018/02/011,808. LIQUID IMPACT ID CTRI/2019/02/017,548.
Collapse
Affiliation(s)
| | | | | | | | | | - Sewanti Limaye
- Kokilaben Dhirubhai Ambani Hospital and Medical Research Institute, Mumbai, India
| | | | | | | | | | | | | | | | | | - Prashant Kumar
- Institute of Bioinformatics, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
- Somaiya Vidyavihar University, Mumbai, India
| | | | | |
Collapse
|
5
|
Huang D, Gao W, Zhong X, Ge J. NLRP3 activation in endothelia promotes development of diabetes-associated atherosclerosis. Aging (Albany NY) 2020; 12:18181-18191. [PMID: 32966239 PMCID: PMC7585081 DOI: 10.18632/aging.103666] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/22/2020] [Indexed: 01/24/2023]
Abstract
Inflammatory damage to endothelial cells plays a pivotal role in the diabetes-provoked atherosclerosis (AS). PYD domains-containing protein 3 (NLRP3) induces formation of inflammasome activates caspase-1, which subsequently cleaves the precursor form of IL-1β (pro-IL-1β) into the processed, secreted form IL-1β to promote the immune responses in AS. However, it is not known whether NLRP3 activation specifically in endothelial cells causes AS. Here, in an in vitro model for AS, we showed that NLRP3-depleted human aortic endothelial cells (HAECs) became resistant to apoptotic cell death, maintained proliferative potential and reduced reactive oxygen species (ROS) production upon treatment with oxidized low-density lipoprotein (ox-LDL). Next, the role of NLRP3 in endothelial cells in the development of diabetes-associated AS was assessed in endothelial cell-specific NLRP3 mutant, ApoE (-/-) mice (APOEKO/Tie2p-Cre/NLRP3MKO), compared to control ApoE (-/-) mice (APOEKO), supplied with either high-fat diet (HFD), or normal diet (ND). We found that endothelia-specific NLRP3-depletion significantly attenuated AS severity in mice treated with HFD, likely through reduced apoptotic death of endothelial cells and production of ROS. Together, our data suggest that NLRP3 activation in endothelial cells promotes development of diabetes-associated AS.
Collapse
Affiliation(s)
- Dong Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Wei Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Xin Zhong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| |
Collapse
|
6
|
Abstract
Vascularization is a major hurdle in complex tissue and organ engineering. Tissues greater than 200 μm in diameter cannot rely on simple diffusion to obtain nutrients and remove waste. Therefore, an integrated vascular network is required for clinical translation of engineered tissues. Microvessels have been described as <150 μm in diameter, but clinically they are defined as <1 mm. With new advances in super microsurgery, vessels less than 1 mm can be anastomosed to the recipient circulation. However, this technical advancement still relies on the creation of a stable engineered microcirculation that is amenable to surgical manipulation and is readily perfusable. Microvascular engineering lays on the crossroads of microfabrication, microfluidics, and tissue engineering strategies that utilize various cellular constituents. Early research focused on vascularization by co-culture and cellular interactions, with the addition of angiogenic growth factors to promote vascular growth. Since then, multiple strategies have been utilized taking advantage of innovations in additive manufacturing, biomaterials, and cell biology. However, the anatomy and dynamics of native blood vessels has not been consistently replicated. Inconsistent results can be partially attributed to cell sourcing which remains an enigma for microvascular engineering. Variations of endothelial cells, endothelial progenitor cells, and stem cells have all been used for microvascular network fabrication along with various mural cells. As each source offers advantages and disadvantages, there continues to be a lack of consensus. Furthermore, discord may be attributed to incomplete understanding about cell isolation and characterization without considering the microvascular architecture of the desired tissue/organ.
Collapse
|
7
|
Loganathan K, Salem Said E, Winterrowd E, Orebrand M, He L, Vanlandewijck M, Betsholtz C, Quaggin SE, Jeansson M. Angiopoietin-1 deficiency increases renal capillary rarefaction and tubulointerstitial fibrosis in mice. PLoS One 2018; 13:e0189433. [PMID: 29293543 PMCID: PMC5749705 DOI: 10.1371/journal.pone.0189433] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 11/24/2017] [Indexed: 01/06/2023] Open
Abstract
Presence of tubulointerstitial fibrosis is predictive of progressive decline in kidney function, independent of its underlying cause. Injury to the renal microvasculature is a major factor in the progression of fibrosis and identification of factors that regulate endothelium in fibrosis is desirable as they might be candidate targets for treatment of kidney diseases. The current study investigates how loss of Angipoietin-1 (Angpt1), a ligand for endothelial tyrosine-kinase receptor Tek (also called Tie2), affects tubulointerstitial fibrosis and renal microvasculature. Inducible Angpt1 knockout mice were subjected to unilateral ureteral obstruction (UUO) to induce fibrosis, and kidneys were collected at different time points up to 10 days after obstruction. Staining for aSMA showed that Angpt1 deficient kidneys had significantly more fibrosis compared to wildtype mice 3, 6, and 10 days after UUO. Further investigation 3 days after UUO showed a significant increase of Col1a1 and vimentin in Angpt1 deficient mice, as well as increased gene expression of Tgfb1, Col1a1, Fn1, and CD44. Kidney injury molecule 1 (Kim1/Havcr1) was significantly more increased in Angpt1 deficient mice 1 and 3 days after UUO, suggesting a more severe injury early in the fibrotic process in Angpt1 deficient mice. Staining for endomucin showed that capillary rarefaction was evident 3 days after UUO and Angpt1 deficient mice had significantly less capillaries 6 and 10 days after UUO compared to UUO kidneys in wildtype mice. RNA sequencing revealed downregulation of several markers for endothelial cells 3 days after UUO, and that Angpt1 deficient mice had a further downregulation of Emcn, Plvap, Pecam1, Erg, and Tek. Our results suggest that loss of Angpt1 is central in capillary rarefaction and fibrogenesis and propose that manipulations to maintain Angpt1 levels may slow down fibrosis progression.
Collapse
Affiliation(s)
| | - Ebtisam Salem Said
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Emily Winterrowd
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Martina Orebrand
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Liqun He
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Michael Vanlandewijck
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Integrated Cardio Metabolic Centre, Karolinska Institutet, Huddinge, Sweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Integrated Cardio Metabolic Centre, Karolinska Institutet, Huddinge, Sweden
| | - Susan E. Quaggin
- Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL, United States of America
- Division of Nephrology and Hypertension, Northwestern University, Chicago, IL, United States of America
| | - Marie Jeansson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
8
|
Matkar PN, Ariyagunarajah R, Leong-Poi H, Singh KK. Friends Turned Foes: Angiogenic Growth Factors beyond Angiogenesis. Biomolecules 2017; 7:biom7040074. [PMID: 28974056 PMCID: PMC5745456 DOI: 10.3390/biom7040074] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/15/2017] [Accepted: 09/22/2017] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels from pre-existing ones is a biological process that ensures an adequate blood flow is maintained to provide the cells with a sufficient supply of nutrients and oxygen within the body. Numerous soluble growth factors and inhibitors, cytokines, proteases as well as extracellular matrix proteins and adhesion molecules stringently regulate the multi-factorial process of angiogenesis. The properties and interactions of key angiogenic molecules such as vascular endothelial growth factors (VEGFs), fibroblast growth factors (FGFs) and angiopoietins have been investigated in great detail with respect to their molecular impact on angiogenesis. Since the discovery of angiogenic growth factors, much research has been focused on their biological actions and their potential use as therapeutic targets for angiogenic or anti-angiogenic strategies in a context-dependent manner depending on the pathologies. It is generally accepted that these factors play an indispensable role in angiogenesis. However, it is becoming increasingly evident that this is not their only role and it is likely that the angiogenic factors have important functions in a wider range of biological and pathological processes. The additional roles played by these molecules in numerous pathologies and biological processes beyond angiogenesis are discussed in this review.
Collapse
Affiliation(s)
- Pratiek N Matkar
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | | | - Howard Leong-Poi
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Krishna K Singh
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada.
- Division of Vascular Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada.
- Department of Surgery, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
9
|
Singh S, Manson SR, Lee H, Kim Y, Liu T, Guo Q, Geminiani JJ, Austin PF, Chen YM. Tubular Overexpression of Angiopoietin-1 Attenuates Renal Fibrosis. PLoS One 2016; 11:e0158908. [PMID: 27454431 PMCID: PMC4959721 DOI: 10.1371/journal.pone.0158908] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/23/2016] [Indexed: 01/22/2023] Open
Abstract
Emerging evidence has highlighted the pivotal role of microvasculature injury in the development and progression of renal fibrosis. Angiopoietin-1 (Ang-1) is a secreted vascular growth factor that binds to the endothelial-specific Tie2 receptor. Ang-1/Tie2 signaling is critical for regulating blood vessel development and modulating vascular response after injury, but is dispensable in mature, quiescent vessels. Although dysregulation of vascular endothelial growth factor (VEGF) signaling has been well studied in renal pathologies, much less is known about the role of the Ang-1/Tie2 pathway in renal interstitial fibrosis. Previous studies have shown contradicting effects of overexpressing Ang-1 systemically on renal tubulointerstitial fibrosis when different engineered forms of Ang-1 are used. Here, we investigated the impact of site-directed expression of native Ang-1 on the renal fibrogenic process and peritubular capillary network by exploiting a conditional transgenic mouse system [Pax8-rtTA/(TetO)7 Ang-1] that allows increased tubular Ang-1 production in adult mice. Using a murine unilateral ureteral obstruction (UUO) fibrosis model, we demonstrate that targeted Ang-1 overexpression attenuates myofibroblast activation and interstitial collagen I accumulation, inhibits the upregulation of transforming growth factor β1 and subsequent phosphorylation of Smad 2/3, dampens renal inflammation, and stimulates the growth of peritubular capillaries in the obstructed kidney. Our results suggest that Ang-1 is a potential therapeutic agent for targeting microvasculature injury in renal fibrosis without compromising the physiologically normal vasculature in humans.
Collapse
Affiliation(s)
- Sudhir Singh
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Scott R. Manson
- Division of Urology, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Heedoo Lee
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Yeawon Kim
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Tuoen Liu
- Oncology Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Qiusha Guo
- Division of Urology, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Julio J. Geminiani
- Division of Urology, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Paul F. Austin
- Division of Urology, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Ying Maggie Chen
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
- * E-mail:
| |
Collapse
|
10
|
Lack of TEK Gene Mutation in Patients with Cutaneomucosal Venous Malformations from the North-Western Region of Algeria. GENETICS RESEARCH INTERNATIONAL 2014; 2013:784789. [PMID: 24386570 PMCID: PMC3872160 DOI: 10.1155/2013/784789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/21/2013] [Indexed: 11/18/2022]
Abstract
Background. Venous malformations (VM) result from an error in vascular morphogenesis. The first gene suspected in their development is the TEK gene (tyrosine kinase, endothelial). Mutations of this gene have been identified in several Belgian families with a dominant form of the disease. Therefore, we investigated whether mutations in this TEK gene could explain the MV development in patients of families from Tlemcen region (north-western Algeria). Methods. Genomic DNA was extracted from leucocytes of ten patients. The search for mutations in all the 23 exons and in the 5' and 3' intronic sequences flanking the TEK gene was performed using PCR amplification and direct sequencing of amplified genomic DNA. Additionally, a search for somatic mutations of the gene TEK was performed on a biopsy of the venous malformation from one of the ten eligible patients. Results. The sequencing of the 23 exons of the TEK gene revealed neither germinal mutation in our ten patients nor somatic mutation in the tissue of the biopsy. Conclusion. The absence of mutation in the TEK gene in the population studied suggests that the TEK gene is not necessarily involved in the onset of VM; its association with these malformations may differ from one population to another.
Collapse
|
11
|
Pearce TR, Shroff K, Kokkoli E. Peptide targeted lipid nanoparticles for anticancer drug delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2012; 24:3803-22, 3710. [PMID: 22674563 DOI: 10.1002/adma.201200832] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Indexed: 05/21/2023]
Abstract
Encapsulating anticancer drugs in nanoparticles has proven to be an effective mechanism to alter the pharmacokinetic and pharmacodynamic profiles of the drugs, leading to clinically useful cancer therapeutics like Doxil and DaunoXome. Underdeveloped tumor vasculature and lymphatics allow these first-generation nanoparticles to passively accumulate within the tumor, but work to create the next-generation nanoparticles that actively participate in the tumor targeting process is underway. Lipid nanoparticles functionalized with targeting peptides are among the most often studied. The goal of this article is to review the recently published literature of targeted nanoparticles to highlight successful designs that improved in vivo tumor therapy, and to discuss the current challenges of designing these nanoparticles for effective in vivo performance.
Collapse
Affiliation(s)
- Timothy R Pearce
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
12
|
Urich E, Lazic SE, Molnos J, Wells I, Freskgård PO. Transcriptional profiling of human brain endothelial cells reveals key properties crucial for predictive in vitro blood-brain barrier models. PLoS One 2012; 7:e38149. [PMID: 22675443 PMCID: PMC3364980 DOI: 10.1371/journal.pone.0038149] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 04/30/2012] [Indexed: 12/24/2022] Open
Abstract
Brain microvascular endothelial cells (BEC) constitute the blood-brain barrier (BBB) which forms a dynamic interface between the blood and the central nervous system (CNS). This highly specialized interface restricts paracellular diffusion of fluids and solutes including chemicals, toxins and drugs from entering the brain. In this study we compared the transcriptome profiles of the human immortalized brain endothelial cell line hCMEC/D3 and human primary BEC. We identified transcriptional differences in immune response genes which are directly related to the immortalization procedure of the hCMEC/D3 cells. Interestingly, astrocytic co-culturing reduced cell adhesion and migration molecules in both BECs, which possibly could be related to regulation of immune surveillance of the CNS controlled by astrocytic cells within the neurovascular unit. By matching the transcriptome data from these two cell lines with published transcriptional data from freshly isolated mouse BECs, we discovered striking differences that could explain some of the limitations of using cultured BECs to study BBB properties. Key protein classes such as tight junction proteins, transporters and cell surface receptors show differing expression profiles. For example, the claudin-5, occludin and JAM2 expression is dramatically reduced in the two human BEC lines, which likely explains their low transcellular electric resistance and paracellular leakiness. In addition, the human BEC lines express low levels of unique brain endothelial transporters such as Glut1 and Pgp. Cell surface receptors such as LRP1, RAGE and the insulin receptor that are involved in receptor-mediated transport are also expressed at very low levels. Taken together, these data illustrate that BECs lose their unique protein expression pattern outside of their native environment and display a more generic endothelial cell phenotype. A collection of key genes that seems to be highly regulated by the local surroundings of BEC within the neurovascular unit are presented and discussed.
Collapse
Affiliation(s)
- Eduard Urich
- CNS Research, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Stanley E. Lazic
- Bioinformatics and Exploratory Data Analysis, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Juliette Molnos
- Translational Research Science, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Isabelle Wells
- Bioinformatics and Exploratory Data Analysis, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | |
Collapse
|
13
|
Jeansson M, Gawlik A, Anderson G, Li C, Kerjaschki D, Henkelman M, Quaggin SE. Angiopoietin-1 is essential in mouse vasculature during development and in response to injury. J Clin Invest 2011; 121:2278-89. [PMID: 21606590 PMCID: PMC3104773 DOI: 10.1172/jci46322] [Citation(s) in RCA: 348] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 04/20/2011] [Indexed: 12/19/2022] Open
Abstract
Angiopoietin-1/Tek signaling is a critical regulator of blood vessel development, with conventional knockout of angiopoietin-1 or Tek in mice being embryonically lethal due to vascular defects. In addition, angiopoietin-1 is thought to be required for the stability of mature vessels. Using a Cre-Lox conditional gene targeting approach, we have studied the role of angiopoietin-1 in embryonic and adult vasculature. We report here that angiopoietin-1 is critical for regulating both the number and diameter of developing vessels but is not required for pericyte recruitment. Cardiac-specific knockout of angiopoietin-1 reproduced the phenotype of the conventional knockout, demonstrating that the early vascular abnormalities arise from flow-dependent defects. Strikingly, deletion in the entire embryo after day E13.5 produced no immediate vascular phenotype. However, when combined with injury or microvascular stress, angiopoietin-1 deficiency resulted in profound organ damage, accelerated angiogenesis, and fibrosis. These findings redefine our understanding of the biological roles of angiopoietin-1: it is dispensable in quiescent vessels but has a powerful ability to modulate the vascular response after injury.
Collapse
MESH Headings
- Angiopoietin-1/deficiency
- Angiopoietin-1/genetics
- Angiopoietin-1/physiology
- Animals
- Blood Vessels/cytology
- Blood Vessels/embryology
- Blood Vessels/injuries
- Diabetes Mellitus, Experimental/physiopathology
- Diabetic Nephropathies/physiopathology
- Fetal Heart/growth & development
- Fetal Heart/pathology
- Gene Expression Regulation, Developmental
- Humans
- Kidney Glomerulus/blood supply
- Kidney Glomerulus/pathology
- Liver/blood supply
- Mice
- Mice, Knockout
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/physiology
- Neovascularization, Pathologic/embryology
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/physiopathology
- Neovascularization, Physiologic/physiology
- Pericytes/metabolism
- Receptor Protein-Tyrosine Kinases/physiology
- Receptor, TIE-1/physiology
- Receptor, TIE-2
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/physiology
- Wound Healing/physiology
Collapse
Affiliation(s)
- Marie Jeansson
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada.
Rheinisch-Westfälische Technische Hochschule (RWTH) University of Aachen, Aachen, Germany.
Mouse Imaging Centre, Toronto Centre for Phenogenomics, Toronto, Canada.
Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria.
Division of Nephrology, St. Michael’s Hospital, and Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| | - Alexander Gawlik
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada.
Rheinisch-Westfälische Technische Hochschule (RWTH) University of Aachen, Aachen, Germany.
Mouse Imaging Centre, Toronto Centre for Phenogenomics, Toronto, Canada.
Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria.
Division of Nephrology, St. Michael’s Hospital, and Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| | - Gregory Anderson
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada.
Rheinisch-Westfälische Technische Hochschule (RWTH) University of Aachen, Aachen, Germany.
Mouse Imaging Centre, Toronto Centre for Phenogenomics, Toronto, Canada.
Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria.
Division of Nephrology, St. Michael’s Hospital, and Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| | - Chengjin Li
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada.
Rheinisch-Westfälische Technische Hochschule (RWTH) University of Aachen, Aachen, Germany.
Mouse Imaging Centre, Toronto Centre for Phenogenomics, Toronto, Canada.
Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria.
Division of Nephrology, St. Michael’s Hospital, and Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| | - Dontscho Kerjaschki
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada.
Rheinisch-Westfälische Technische Hochschule (RWTH) University of Aachen, Aachen, Germany.
Mouse Imaging Centre, Toronto Centre for Phenogenomics, Toronto, Canada.
Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria.
Division of Nephrology, St. Michael’s Hospital, and Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| | - Mark Henkelman
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada.
Rheinisch-Westfälische Technische Hochschule (RWTH) University of Aachen, Aachen, Germany.
Mouse Imaging Centre, Toronto Centre for Phenogenomics, Toronto, Canada.
Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria.
Division of Nephrology, St. Michael’s Hospital, and Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| | - Susan E. Quaggin
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada.
Rheinisch-Westfälische Technische Hochschule (RWTH) University of Aachen, Aachen, Germany.
Mouse Imaging Centre, Toronto Centre for Phenogenomics, Toronto, Canada.
Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria.
Division of Nephrology, St. Michael’s Hospital, and Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
14
|
Limaye N, Wouters V, Uebelhoer M, Tuominen M, Wirkkala R, Mulliken JB, Eklund L, Boon LM, Vikkula M. Somatic mutations in angiopoietin receptor gene TEK cause solitary and multiple sporadic venous malformations. Nat Genet 2009; 41:118-24. [PMID: 19079259 PMCID: PMC2670982 DOI: 10.1038/ng.272] [Citation(s) in RCA: 339] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Accepted: 10/07/2008] [Indexed: 12/01/2022]
Abstract
Germline substitutions in the endothelial cell tyrosine kinase receptor TIE2 (encoded by TEK) cause a rare, inherited form of venous anomaly known as a mucocutaneous venous malformation (VMCM; refs. 1, 2, 3 and V.W., N.L., M.U., A. Irrthum, L.M.B. et al., unpublished data). We identified a somatic 'second hit' causing loss of function of TIE2 in a resected VMCM and assessed whether such localized, tissue-specific events have a role in the etiology of sporadic venous malformations, which are far more common. We identified eight somatic TEK mutations in lesions from 28 of 57 individuals (49.1%) with sporadic venous malformations; the mutations were absent from the individuals' blood and control tissues. The somatic mutations included one causing a frequent L914F substitution and several double mutations in cis, all of which resulted in ligand-independent TIE2 hyperphosphorylation in vitro. When overexpressed in human umbilical vein endothelial cells, the L914F mutant was abnormally localized and responded to ligand, in contrast to wild-type TIE2 and the common, inherited R849W mutant, suggesting that the mutations have distinct effects. The presence of the same mutations in multifocal sporadic venous malformations in two individuals suggests a common origin for the abnormal endothelial cells at the distant sites. These data show that a sporadic disease may be explained by somatic changes in a gene causing rare, inherited forms and pinpoint TIE2 pathways as potential therapeutic targets for venous malformations.
Collapse
Affiliation(s)
- Nisha Limaye
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Vinciane Wouters
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Melanie Uebelhoer
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Marjut Tuominen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Department of Medical Biochemistry and Molecular Biology, University of Oulu, Finland
| | - Riikka Wirkkala
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Department of Medical Biochemistry and Molecular Biology, University of Oulu, Finland
| | - John B. Mulliken
- Department of Plastic Surgery, Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Department of Medical Biochemistry and Molecular Biology, University of Oulu, Finland
| | - Laurence M. Boon
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Center for Vascular Anomalies, Division of Plastic Surgery, Cliniques Universitaires St-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Miikka Vikkula
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
15
|
Aikawa T, Whipple CA, Lopez ME, Gunn J, Young A, Lander AD, Korc M. Glypican-1 modulates the angiogenic and metastatic potential of human and mouse cancer cells. J Clin Invest 2008; 118:89-99. [PMID: 18064304 DOI: 10.1172/jci32412] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Accepted: 10/17/2007] [Indexed: 12/23/2022] Open
Abstract
Cells isolated from many types of human cancers express heparin-binding growth factors (HBGFs) that drive tumor growth, metastasis, and angiogenesis. The heparan sulfate proteoglycan glypican-1 (GPC1) is a coreceptor for HBGFs. Here we show that both cancer cell-derived and host-derived GPC1 are crucial for efficient growth, metastasis, and angiogenesis of human and mouse cancer cells. Thus downregulation of GPC1 in the human pancreatic cancer cell line PANC-1, using antisense approaches, resulted in prolonged doubling times and decreased anchorage-independent growth in vitro as well as attenuated tumor growth, angiogenesis, and metastasis when these cells were transplanted into athymic mice. Moreover, athymic mice that lacked GPC1 exhibited decreased tumor angiogenesis and metastasis following intrapancreatic implantation with either PANC-1 or T3M4 human pancreatic cancer cells and fewer pulmonary metastases following intravenous injection of murine B16-F10 melanoma cells. In addition, hepatic endothelial cells isolated from these mice exhibited an attenuated mitogenic response to VEGF-A. These data indicate that cancer cell- and host-derived GPC1 are crucial for full mitogenic, angiogenic, and metastatic potential of cancer cells. Thus targeting GPC1 might provide new avenues for cancer therapy and for the prevention of cancer metastasis.
Collapse
Affiliation(s)
- Takuma Aikawa
- Department of Medicine, Dartmouth Hitchcock Medical Center and Dartmouth Medical School, Hanover, New Hampshire 03756, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
McCarter SD, Mei SHJ, Lai PFH, Zhang QW, Parker CH, Suen RS, Hood RD, Zhao YD, Deng Y, Han RNN, Dumont DJ, Stewart DJ. Cell-based Angiopoietin-1 Gene Therapy for Acute Lung Injury. Am J Respir Crit Care Med 2007; 175:1014-26. [PMID: 17322110 DOI: 10.1164/rccm.200609-1370oc] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The acute respiratory distress syndrome is a significant cause of morbidity and mortality in critically ill patients. Angiopoietin-1 (Ang-1), a ligand for the endothelial Tie2 receptor, is an endothelial survival and vascular stabilization factor that reduces endothelial permeability and inhibits leukocyte-endothelium interactions. OBJECTIVES We hypothesized that Ang-1 counteracts vascular inflammation and pulmonary vascular leak in experimental acute lung injury. METHODS We used cell-based gene therapy in a rat model of ALI. Transgenic mice overexpressing Ang-1 or deficient in the Tie2 receptor were also studied to better elucidate the mechanisms of protection. MEASUREMENTS AND MAIN RESULTS The present report provides data that support a strong protective role for the Ang-1/Tie2 system in two experimental models of LPS-induced acute lung injury. In a rat model, cell-based Ang-1 gene transfer improved morphological, biochemical, and molecular indices of lung injury and inflammation. These findings were confirmed in a gain-of-function conditional, targeted transgenic mouse model, in which Ang-1 reduced endothelial cell activation and the expression of adhesion molecules, associated with a marked improvement in airspace inflammation and intraalveolar septal thickening. Moreover, heterozygous Tie2-deficient mice demonstrated enhanced evidence of lung injury and increased early mortality. CONCLUSIONS These results support a critical role for the Ang-1/Tie2 axis in modulating the pulmonary vascular response to lung injury and suggest that Ang-1 therapy may represent a potential new strategy for the treatment and/or prevention of acute respiratory distress syndrome in critically ill patients.
Collapse
|
17
|
Jin YL, Enzan H, Kuroda N, Hayashi Y, Toi M, Miyazaki E, Hamauzu T, Hiroi M, Guo LM, Shen ZS, Saibara T. Vascularization in tissue remodeling after rat hepatic necrosis induced by dimethylnitrosamine. Med Mol Morphol 2006; 39:33-43. [PMID: 16575513 DOI: 10.1007/s00795-005-0306-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2005] [Accepted: 11/14/2005] [Indexed: 01/31/2023]
Abstract
We observed postnecrotic tissue remodeling to examine vascularization in adult rat livers. Livers, bone marrow, and peripheral blood from rats at 24 h to 14 days after an injection of dimethylnitrosamine (DMN) were examined by light microscopic, immunohistochemical, and ultrastructural methods. Numerous ED-1 (a marker for rat monocytes/macrophages)-positive round mononuclear cells infiltrated in the necrotic areas at 36 h after DMN treatment. On day 5, when necrotic tissues were removed, some of the cells were transformed from round to spindle in shape. On day 7, these cells were contacted with residual reticulin fibers and became positive for SE-1, a marker of hepatic sinusoidal endothelial cells and Tie-1, an endothelial cell-specific surface receptor, associated with frequent occurrence of ED-1/SE-1 and ED-1/Tie-1 double-positive spindle cells. Ultrastructurally, the spindle cells simultaneously showed phagocytosis and endothelial cell-like morphology. With time necrotic areas diminished, and on day 14, the necrotic tissues were almost replaced by regenerated liver tissues and thin bundles of central-to-central bridging fibrosis. Bone marrow from 12 h to day 2 showed an increase of BrdU-positive mononuclear cells. Some of them were positive for ED-1. The BrdU-labeled and ED-1-positive cells appeared as early as 12 h after DMN injection and reached a peak in number at 36 h. They were similar in structure to ED-1-positive cells in necrotic liver tissues. These findings suggest that round mononuclear ED-1-positive cells proliferate first in bone marrow after DMN treatment, reach necrotic areas of the liver through the circulation, and differentiate to sinusoidal endothelial cells. Namely, hepatic sinusoids in DMN-induced necrotic areas may partly be reorganized possibly by vasculogenesis.
Collapse
Affiliation(s)
- Yu-Lan Jin
- Department of Pathology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Saharinen P, Kerkelä K, Ekman N, Marron M, Brindle N, Lee GM, Augustin H, Koh GY, Alitalo K. Multiple angiopoietin recombinant proteins activate the Tie1 receptor tyrosine kinase and promote its interaction with Tie2. ACTA ACUST UNITED AC 2005; 169:239-43. [PMID: 15851516 PMCID: PMC2171878 DOI: 10.1083/jcb.200411105] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The Tie1 receptor tyrosine kinase was isolated over a decade ago, but so far no ligand has been found to activate this receptor. Here, we have examined the potential of angiopoietins, ligands for the related Tie2 receptor, to mediate Tie1 activation. We show that a soluble Ang1 chimeric protein, COMP-Ang1, stimulates Tie1 phosphorylation in endothelial cells with similar kinetics and angiopoietin dose dependence when compared with Tie2. The phosphorylation of overexpressed Tie1 was weakly induced by COMP-Ang1 also in transfected cells that do not express Tie2. When cotransfected, Tie2 formed heteromeric complexes with Tie1, enhanced Tie1 activation, and induced phosphorylation of a kinase-inactive Tie1 in a ligand-dependent manner. Tie1 phosphorylation was also induced by native Ang1 and Ang4, although less efficiently than with COMP-Ang1. In conclusion, we show that Tie1 phosphorylation is induced by multiple angiopoietin proteins and that the activation is amplified via Tie2. These results should be important in dissecting the signal transduction pathways and biological functions of Tie1.
Collapse
Affiliation(s)
- Pipsa Saharinen
- Molecular/Cancer Biology Laboratory and Ludwig Institute for Cancer Research, University of Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Popkov M, Jendreyko N, McGavern DB, Rader C, Barbas CF. Targeting Tumor Angiogenesis with Adenovirus-Delivered Anti-Tie-2 Intrabody. Cancer Res 2005. [DOI: 10.1158/0008-5472.972.65.3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Inhibition of tumor angiogenesis is a promising approach for cancer therapy. As an endothelial cell–specific receptor kinase expressed almost exclusively on the surface of vascular endothelium, Tie-2 has an important role in tumor angiogenesis. To explore the therapeutic potential of blocking Tie-2 receptor-interaction pathway, an adenoviral vector was used to deliver a recombinant single-chain antibody fragment rabbit intrabody (pAd-2S03) capable of inhibition of both mouse and human Tie-2 surface expression. pAd-2S03 was given to mice with well-established primary tumors, either a human Kaposi's sarcoma (SLK) or a human colon carcinoma (SW1222). The intrabody significantly inhibited growth of both tumors (75% and 63%, respectively) when compared with pAd-GFP control-treated tumors (P < 0.01). Histopathologic analysis of cryosections taken from mice treated with pAd-2S03 revealed a marked decrease in vessel density, which was reduced by >87% in both tumor models when compared with control-treated tumors (P < 0.01). In contrast, human Tie-2-monospecific pAd-1S05 intrabody did not affect the growth of tumors, indicating that the antitumor effect of pAd-2S03 was due to the inhibition of tumor angiogenesis in these murine models. Our results show that the Tie-2 receptor pathway is essential for both SLK sarcoma and SW1222 colon carcinoma xenograft growth. The present study shows the potential utility of antiangiogenic agents that target the endothelium-specific receptor Tie-2 for down-regulation or genetic deletion.
Collapse
Affiliation(s)
- Mikhail Popkov
- 1Department of Molecular Biology and Skaggs Institute for Chemical Biology and
| | - Nina Jendreyko
- 1Department of Molecular Biology and Skaggs Institute for Chemical Biology and
| | - Dorian B. McGavern
- 2Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California and
| | - Christoph Rader
- 3Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Carlos F. Barbas
- 1Department of Molecular Biology and Skaggs Institute for Chemical Biology and
| |
Collapse
|
20
|
Iraha F, Saito Y, Yoshida K, Kawakami M, Izutsu Y, Daar IO, Maéno M. Common and distinct signals specify the distribution of blood and vascular cell lineages in Xenopus laevis embryos. Dev Growth Differ 2002; 44:395-407. [PMID: 12392573 DOI: 10.1046/j.1440-169x.2002.00653.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In an effort to elucidate the regulatory mechanisms that determine the fate of blood cells and vascular cells in the ventral blood island mesoderm, the embryonic expression of Xtie-2, a Xenopus homolog of the tie-2 receptor tyrosine kinase, was examined. Whole-mount in situ hybridization analysis revealed that Xtie-2 mRNA is expressed at the late tailbud stage within the regions where endothelial precursor cells exist. On the ventral side of embryos, Xtie-2-positive cells are predominantly present just outside the boundary of alpha-globin-positive cells, thus the expression pattern of these two markers seems mutually exclusive. Further experiments revealed that there is a consistent and strong correlation between the induction of Xtie-2 and alpha-globin expression in embryos and explant tissues. First, these two markers displayed overlapping expression in embryos ventralized by the removal of a "dorsal determinant" from the vegetal cytoplasm at the 1-cell stage. Second, expression of both Xtie-2 and alpha-globin were markedly induced in ectodermal explants (animal caps) from embryos co-injected with activin and bone morphogenetic protein (BMP)-4 RNA. Furthermore, both Xtie-2 and alpha-globin messages were strongly positive in dorsal marginal zone explants that had been injected with BMP-4 RNA. In contrast, however, there was a clear distinction in the localization of these two transcripts in embryos dorsalized by LiCl treatment. Distinct localization was also found in the ventral marginal zone (VMZ) explants. Using the VMZ explant system, we demonstrate a role of fibroblast growth factor (FGF) signaling in enhancing the vascular cell marker and reducing the blood cell marker. The present study suggests that the early steps of blood and vascular cell differentiation are regulated by a common BMP-4-dependent signaling; however, distinct factor(s) such as FGF are involved in different distribution of these two cell lineages.
Collapse
Affiliation(s)
- Fumie Iraha
- Graduate School of Science and Technology, Niigata University, 8050 Ikarashi-2, Niigata 950-2181, Japan
| | | | | | | | | | | | | |
Collapse
|
21
|
Shih SC, Robinson GS, Perruzzi CA, Calvo A, Desai K, Green JE, Ali IU, Smith LEH, Senger DR. Molecular profiling of angiogenesis markers. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 161:35-41. [PMID: 12107087 PMCID: PMC1850687 DOI: 10.1016/s0002-9440(10)64154-5] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The goal of this study was to develop a sensitive, simple, and widely applicable assay to measure copy numbers of specific mRNAs using real-time quantitative reverse transcriptase-polymerase chain reaction (RT-PCR), and identify a profile of gene expression closely associated with angiogenesis. We measured a panel of nine potential angiogenesis markers from a mouse transgenic model of prostate adenocarcinoma (TRAMP) and a mouse skin model of vascular endothelial growth factor (VEGF)-driven angiogenesis. In both models, expression of VEGF correlated with expression of mRNAs encoding other angiogenic cytokines (angiopoietin-1 and angiopoietin-2), endothelial cell receptor tyrosine kinases (Flt-1, KDR, Tie-1), and endothelial cell adhesion molecules (VE-cadherin, PECAM-1). Relative to control, in dermis highly stimulated by VEGF, the Ang-2 mRNA transcript numbers increased 35-fold, PECAM-1 and VE-cadherin increased 10-fold, Tie-1 increased 8-fold, KDR and Flt-1 each increased 4-fold, and Ang-1 increased 2-fold. All transcript numbers were correspondingly reduced in skin with less VEGF expression, indicating a relationship of each of these seven markers with VEGF. Thus, this study identifies a highly efficient method for precise quantification of a panel of seven specific mRNAs that correlate with VEGF expression and VEGF-induced neovascularization, and it provides evidence that real-time quantitative RT-PCR offers a highly sensitive strategy for monitoring angiogenesis.
Collapse
Affiliation(s)
- Shu-Ching Shih
- Department of Ophthalmology, Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Tsiamis AC, Morris PN, Marron MB, Brindle NPJ. Vascular endothelial growth factor modulates the Tie-2:Tie-1 receptor complex. Microvasc Res 2002; 63:149-58. [PMID: 11866538 DOI: 10.1006/mvre.2001.2377] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The receptor tyrosine kinase Tie-1 is expressed predominantly in endothelial cells where it physically associates with the related receptor Tie-2. Positive signalling through Tie-2 is associated with microvessel stability and suppression of this signal is thought to be required for vascular endothelial growth factor (VEGF)-induced microvessel remodelling or growth. Here we examine the effects of VEGF on Tie-1 and the Tie-2:Tie-1 complex. We show that VEGF induces generation of the Tie-1 endodomain and loss of the full-length receptor. The effects of VEGF on endodomain formation are not suppressed by inhibitors of protein kinase C and do not involve the nitric oxide signalling pathway. Tyrosine kinase inhibitors, in contrast, do abolish endodomain generation in response to the endothelial growth factor. VEGF stimulation of cells does not cause dissociation of the Tie-2:Tie-1 complex; rather the complex is converted to a form comprising the full-length-Tie-2 and Tie-1 endodomain. VEGF can therefore switch the Tie-2:Tie-1 complex between two different forms in endothelial cells. The ability of VEGF to modulate Tie-1 and the Tie-2:Tie-1 complex provides a mechanism whereby this initiator of vessel growth and remodelling can directly modulate receptors involved in vessel stabilization. Such cross-talk is likely to be important in the coordinate control of blood vessel formation during development and in postnatal angiogenesis.
Collapse
Affiliation(s)
- Achilleas C Tsiamis
- Department of Surgery, University of Leicester, RKCSB, Leicester, LE2 7LX, United Kingdom
| | | | | | | |
Collapse
|
23
|
Kontos CD, Cha EH, York JD, Peters KG. The endothelial receptor tyrosine kinase Tie1 activates phosphatidylinositol 3-kinase and Akt to inhibit apoptosis. Mol Cell Biol 2002; 22:1704-13. [PMID: 11865050 PMCID: PMC135589 DOI: 10.1128/mcb.22.6.1704-1713.2002] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tie1 is an orphan receptor tyrosine kinase that is expressed almost exclusively in endothelial cells and that is required for normal embryonic vascular development. Genetic studies suggest that Tie1 promotes endothelial cell survival, but other studies have suggested that the Tie1 kinase has little to no activity, and Tie1-mediated signaling pathways are unknown. To begin to study Tie1 signaling, a recombinant glutathione S-transferase (GST)-Tie1 kinase fusion protein was produced in insect cells and found to be autophosphorylated in vitro. GST-Tie1 but not a kinase-inactive mutant associated with a recombinant p85 SH2 domain protein in vitro, suggesting that Tie1 might signal through phosphatidylinositol (PI) 3-kinase. To study Tie1 signaling in a cellular context, a c-fms-Tie1 chimeric receptor (fTie1) was expressed in NIH 3T3 cells. Ligand stimulation of fTie1 resulted in Tie1 autophosphorylation and downstream activation of PI 3-kinase and Akt. Stimulation of fTie1-expressing cells potently inhibited UV irradiation-induced apoptosis in a PI 3-kinase-dependent manner. Moreover, both Akt phosphorylation and inhibition of apoptosis were abrogated by mutation of tyrosine 1113 to phenylalanine, suggesting that this residue is an important PI 3-kinase binding site. These findings are the first biochemical demonstration of a signal transduction pathway and corresponding cellular function for Tie1, and the antiapoptotic effect of Tie1 is consistent with the results of previous genetic studies.
Collapse
Affiliation(s)
- Christopher D Kontos
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | |
Collapse
|
24
|
Reyes M, Dudek A, Jahagirdar B, Koodie L, Marker PH, Verfaillie CM. Origin of endothelial progenitors in human postnatal bone marrow. J Clin Invest 2002; 109:337-46. [PMID: 11827993 PMCID: PMC150857 DOI: 10.1172/jci14327] [Citation(s) in RCA: 244] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
This study demonstrates that a CD34(-), vascular endothelial cadherin(-) (VE-cadherin(-)), AC133(+), and fetal liver kinase(+) (Flk1(+)) multipotent adult progenitor cell (MAPC) that copurifies with mesenchymal stem cells from postnatal human bone marrow (BM) is a progenitor for angioblasts. In vitro, MAPCs cultured with VEGF differentiate into CD34(+), VE-cadherin(+), Flk1(+) cells - a phenotype that would be expected for angioblasts. They subsequently differentiate into cells that express endothelial markers, function in vitro as mature endothelial cells, and contribute to neoangiogenesis in vivo during tumor angiogenesis and wound healing. This in vitro model of preangioblast-to-endothelium differentiation should prove very useful in studying commitment to the angioblast and beyond. In vivo, MAPCs can differentiate in response to local cues into endothelial cells that contribute to neoangiogenesis in tumors. Because MAPCs can be expanded in culture without obvious senescence for more than 80 population doublings, they may be an important source of endothelial cells for cellular pro- or anti-angiogenic therapies.
Collapse
MESH Headings
- AC133 Antigen
- Adolescent
- Adult
- Antigens, CD
- Antigens, CD34/metabolism
- Bone Marrow Cells/cytology
- Bone Marrow Cells/immunology
- Bone Marrow Cells/metabolism
- Cadherins/metabolism
- Cell Differentiation
- Child
- Child, Preschool
- Endothelium, Vascular/cytology
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Glycoproteins/metabolism
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/metabolism
- Humans
- Middle Aged
- Neoplasms/blood supply
- Neovascularization, Pathologic
- Neovascularization, Physiologic
- Peptides/metabolism
- Phenotype
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptors, Growth Factor/metabolism
- Receptors, Vascular Endothelial Growth Factor
Collapse
Affiliation(s)
- Morayma Reyes
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | |
Collapse
|
25
|
Reyes M, Dudek A, Jahagirdar B, Koodie L, Marker PH, Verfaillie CM. Origin of endothelial progenitors in human postnatal bone marrow. J Clin Invest 2002. [DOI: 10.1172/jci0214327] [Citation(s) in RCA: 809] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
26
|
Chen BP, Li YS, Zhao Y, Chen KD, Li S, Lao J, Yuan S, Shyy JY, Chien S. DNA microarray analysis of gene expression in endothelial cells in response to 24-h shear stress. Physiol Genomics 2001; 7:55-63. [PMID: 11595792 DOI: 10.1152/physiolgenomics.2001.7.1.55] [Citation(s) in RCA: 160] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The recently developed DNA microarray technology provides a powerful and efficient tool to rapidly compare the differential expression of a large number of genes. Using the DNA microarray approach, we investigated gene expression profiles in cultured human aortic endothelial cells (HAECs) in response to 24 h of laminar shear stress at 12 dyn/cm(2). This relatively long-term shearing of cultured HAECs led to the modulation of the expression of a number of genes. Several genes related to inflammation and EC proliferation were downregulated, suggesting that 24-h shearing may keep ECs in a relatively noninflammatory and nonproliferative state compared with static cells. Some genes were significantly upregulated by the 24-h shear stress; these includes genes involved in EC survival and angiogenesis (Tie2 and Flk-1) and vascular remodeling (matrix metalloproteinase 1). These results provide information on the profile of gene expression in shear-adapted ECs, which is the case for the native ECs in the straight part of the aorta in vivo.
Collapse
Affiliation(s)
- B P Chen
- Department of Bioengineering and the Whitaker Institute of Biomedical Engineering, University of California, San Diego, La Jolla 92093-0427, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Rajantie I, Ekman N, Iljin K, Arighi E, Gunji Y, Kaukonen J, Palotie A, Dewerchin M, Carmeliet P, Alitalo K. Bmx tyrosine kinase has a redundant function downstream of angiopoietin and vascular endothelial growth factor receptors in arterial endothelium. Mol Cell Biol 2001; 21:4647-55. [PMID: 11416142 PMCID: PMC87133 DOI: 10.1128/mcb.21.14.4647-4655.2001] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Bmx gene, a member of the Tec tyrosine kinase gene family, is known to be expressed in subsets of hematopoietic and endothelial cells. In this study, mice were generated in which the first coding exon of the Bmx gene was replaced with the lacZ reporter gene by a knock-in strategy. The homozygous mice lacking Bmx activity were fertile and had a normal life span without an obvious phenotype. Staining of their tissues using beta-galactosidase substrate to assess the sites of Bmx expression revealed strong signals in the endothelial cells of large arteries and in the endocardium starting between days 10.5 and 12.5 of embryogenesis and continuing in adult mice, while the venular endothelium showed a weak signal only in the superior and inferior venae cavae. Of the five known endothelial receptor tyrosine kinases tested, activated Tie-2 induced tyrosyl phosphorylation of the Bmx protein and both Tie-2 and vascular endothelial growth factor receptor 1 (VEGFR-1) stimulated Bmx tyrosine kinase activity. Thus, the Bmx tyrosine kinase has a redundant role in arterial endothelial signal transduction downstream of the Tie-2 and VEGFR-1 growth factor receptors.
Collapse
Affiliation(s)
- I Rajantie
- Molecular/Cancer Biology Laboratory, Haartman Institute and Biomedicum Helsinki, 00014 University of Helsinki, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
As the sequencing of the human genome is completed by the Human Genome Project, the analysis of this rich source of information will illuminate many areas in medicine and biology. The protein tyrosine kinases are a large multigene family with particular relevance to many human diseases, including cancer. A search of the human genome for tyrosine kinase coding elements identified several novel genes and enabled the creation of a nonredundant catalog of tyrosine kinase genes. Ninety unique kinase genes can be identified in the human genome, along with five pseudogenes. Of the 90 tyrosine kinases, 58 are receptor type, distributed into 20 subfamilies. The 32 nonreceptor tyrosine kinases can be placed in 10 subfamilies. Additionally, mouse orthologs can be identified for nearly all the human tyrosine kinases. The completion of the human tyrosine kinase family tree provides a framework for further advances in biomedical science.
Collapse
Affiliation(s)
- D R Robinson
- Department of Biological Chemistry, UC Davis School of Medicine, UC Davis Cancer Center, Sacramento, California, CA 95817, USA
| | | | | |
Collapse
|