1
|
Silva JAF, Qi X, Grant MB, Boulton ME. Spatial and temporal VEGF receptor intracellular trafficking in microvascular and macrovascular endothelial cells. Sci Rep 2021; 11:17400. [PMID: 34462507 PMCID: PMC8405636 DOI: 10.1038/s41598-021-96964-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 08/12/2021] [Indexed: 01/18/2023] Open
Abstract
The vascular endothelial growth factor receptors (VEGFRs) can shape the neovascular phenotype of vascular endothelial cells when translocated to the nucleus, however the spatial and temporal changes in the intracellular distribution and translocation of VEGFRs to the nucleus and the organelles involved in this process is unclear. This study reports the effect of exogenous VEGF on translocation of VEGFRs and organelles in micro- and macrovascular endothelial cells. We showed that VEGF is responsible for: a rapid and substantial nuclear translocation of VEGFRs; VEGFR1 and VEGFR2 exhibit distinct spatial, temporal and structural translocation characteristics both in vitro and in vivo and this determines the nuclear VEGFR1:VEGFR2 ratio which differs between microvascular and macrovascular cells; VEGFR2 nuclear translocation is associated with the endosomal pathway transporting the receptor from Golgi in microvascular endothelial cells; and an increase in the volume of intracellular organelles. In conclusion, the nuclear translocation of VEGFRs is both receptor and vessel (macro versus micro) dependent and the endosomal pathway plays a key role in the translocation of VEGFRs to the nucleus and the subsequent export to the lysosomal system. Modulating VEGF-mediated VEGFR1 and VEGFR2 intracellular transmigration pathways may offer an alternative for the development of new anti-angiogenic therapies.
Collapse
Affiliation(s)
- Juliete A F Silva
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, 1670 University Blvd, Volker Hall, Room 472, Birmingham, AL, 35233, USA.
| | - Xiaoping Qi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, 1670 University Blvd, Volker Hall, Room 472, Birmingham, AL, 35233, USA
| | - Maria B Grant
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, 1670 University Blvd, Volker Hall, Room 472, Birmingham, AL, 35233, USA
| | - Michael E Boulton
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, 1670 University Blvd, Volker Hall, Room 472, Birmingham, AL, 35233, USA.
| |
Collapse
|
2
|
Wewers TM, Schulz A, Nolte I, Pavenstädt H, Brand M, Di Marco GS. Circulating Soluble Fms-like Tyrosine Kinase in Renal Diseases Other than Preeclampsia. J Am Soc Nephrol 2021; 32:1853-1863. [PMID: 34155060 PMCID: PMC8455271 DOI: 10.1681/asn.2020111579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/20/2021] [Indexed: 02/04/2023] Open
Abstract
Soluble Fms-like tyrosine kinase (sFlt-1/sVEGFR1) is a naturally occurring antagonist of vascular endothelial growth factor (VEGF). Despite being a secreted, soluble protein lacking cytoplasmic and transmembrane domains, sFlt-1 can act locally and be protective against excessive microenvironmental VEGF concentration or exert autocrine functions independently of VEGF. Circulating sFlt-1 may indiscriminately affect endothelial function and the microvasculature of distant target organs. The clinical significance of excess sFlt-1 in kidney disease was first shown in preeclampsia, a major renal complication of pregnancy. However, circulating sFlt-1 levels appear to be increased in various diseases with varying degrees of renal impairment. Relevant clinical associations between circulating sFlt-1 and severe outcomes (e.g., endothelial dysfunction, renal impairment, cardiovascular disease, and all-cause mortality) have been observed in patients with CKD and after kidney transplantation. However, sFlt-1 appears to be protective against renal dysfunction-associated aggravation of atherosclerosis and diabetic nephropathy. Therefore, in this study, we provide an update on sFlt-1 in several kidney diseases other than preeclampsia, discuss clinical findings and experimental studies, and briefly consider its use in clinical practice.
Collapse
Affiliation(s)
- Theresa M. Wewers
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany,Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Annika Schulz
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Ingo Nolte
- Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Hermann Pavenstädt
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Marcus Brand
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Giovana S. Di Marco
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany,Correspondence: Giovana S. Di Marco, Albert-Schweitzer-Campus 1, Building A14, 48149 Münster, Germany.
| |
Collapse
|
3
|
Yoon Y, Voloudakis G, Doran N, Zhang E, Dimovasili C, Chen L, Shao Z, Darmanis S, Tang C, Tang J, Wang VX, Hof PR, Robakis NK, Georgakopoulos A. PS1 FAD mutants decrease ephrinB2-regulated angiogenic functions, ischemia-induced brain neovascularization and neuronal survival. Mol Psychiatry 2021; 26:1996-2012. [PMID: 32541930 PMCID: PMC7736163 DOI: 10.1038/s41380-020-0812-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
Abstract
Microvascular pathology and ischemic lesions contribute substantially to neuronal dysfunction and loss that lead to Alzheimer disease (AD). To facilitate recovery, the brain stimulates neovascularization of damaged tissue via sprouting angiogenesis, a process regulated by endothelial cell (EC) sprouting and the EphB4/ephrinB2 system. Here, we show that in cultures of brain ECs, EphB4 stimulates the VE-cadherin/Rok-α angiogenic complexes known to mediate sprouting angiogenesis. Importantly, brain EC cultures expressing PS1 FAD mutants decrease the EphB4-stimulated γ-secretase cleavage of ephrinB2 and reduce production of the angiogenic peptide ephrinB2/CTF2, the VE-cadherin angiogenic complexes and EC sprouting and tube formation. These data suggest that FAD mutants may attenuate ischemia-induced brain angiogenesis. Supporting this hypothesis, ischemia-induced VE-cadherin angiogenic complexes, levels of neoangiogenesis marker Endoglin, vascular density, and cerebral blood flow recovery, are all decreased in brains of mouse models expressing PS1 FAD mutants. Ischemia-induced brain neuronal death and cognitive deficits also increase in these mice. Furthermore, a small peptide comprising the C-terminal sequence of peptide ephrinB2/CTF2 rescues angiogenic functions of brain ECs expressing PS1 FAD mutants. Together, our data show that PS1 FAD mutations impede the EphB4/ephrinB2-mediated angiogenic functions of ECs and impair brain neovascularization, neuronal survival and cognitive recovery following ischemia. Furthermore, our data reveal a novel brain angiogenic mechanism targeted by PS1 FAD mutants and a potential therapeutic target for ischemia-induced neurodegeneration. Importantly, FAD mutant effects occur in absence of neuropathological hallmarks of AD, supporting that such hallmarks may form downstream of mutant effects on neoangiogenesis and neuronal survival.
Collapse
Affiliation(s)
- YoneJung Yoon
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Georgios Voloudakis
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nathan Doran
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily Zhang
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christina Dimovasili
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lei Chen
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Zhiping Shao
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Spyros Darmanis
- Departments of Bioengineering and Applied Physics, Stanford University and Chan Zuckerberg Biohub, Stanford, CA, 94305, USA
| | - Cheuk Tang
- Department of Radiology, Neuroscience and Psychiatry Translational and Molecular Imaging Institute at Mount Sinai, New York, NY, USA
| | - Jun Tang
- Department of Radiology, Neuroscience and Psychiatry Translational and Molecular Imaging Institute at Mount Sinai, New York, NY, USA
| | - Victoria X Wang
- Department of Radiology, Translational and Molecular Imaging Institute at Mount Sinai, New York, NY, USA
| | - Patrick R Hof
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nikolaos K Robakis
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Anastasios Georgakopoulos
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Niepolski L, Drzewiecka H, Warchoł W. Circulating vascular endothelial growth factor receptor 2 levels and their association with lipid abnormalities in patients on hemodialysis. Biomed Rep 2021; 14:37. [PMID: 33692900 PMCID: PMC7938296 DOI: 10.3892/br.2021.1413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 02/08/2021] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to examine the association between the levels of circulating vascular endothelial growth factor receptor (VEGFR)2 levels, serum lipid composition and plasma receptor for advanced glycation end-products (RAGE) expression in patients undergoing hemodialysis (HD). A total of 50 patients on HD (27 men and 23 women; median age, 66 years; age range 28-88 years; HD mean time, 29.0, 3.9-157.0 months) were enrolled. Age-matched healthy subjects (n=26) were used as the control group. Plasma VEGFR2 and RAGE levels were determined using ELISA. Dyslipidemia (D) in patients on HD was diagnosed according to the Kidney Disease Outcomes Quality Initiative Clinical Practice Guidelines for Managing Dyslipidemias in Chronic Kidney Disease. Circulating VEGFR2, RAGE and serum lipids were compared between dyslipidemic and non-dyslipidemic patients on HD and controls. In patients on HD, the plasma VEGFR2 levels were lower compared with those in the healthy population. D was associated with high plasma VEGFR2 levels. The triglyceride/HDL-cholesterol ratio was strongly associated with plasma VEGFR2 levels. The plasma VEGFR2 concentration was associated with circulating RAGE levels. Therefore, circulating VEGFR2 levels may be partly associated with lipid abnormalities and plasma RAGE levels in patients receiving HD.
Collapse
Affiliation(s)
- Leszek Niepolski
- Department of Physiology, Poznan University of Medical Sciences, Poznań 60-781, Poland
| | - Hanna Drzewiecka
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, Poznań 60-781, Poland
| | - Wojciech Warchoł
- Department of Ophthalmology and Optometry, Poznan University of Medical Sciences, Poznań 60-781, Poland
| |
Collapse
|
5
|
Brain expression of the vascular endothelial growth factor gene family in cognitive aging and alzheimer's disease. Mol Psychiatry 2021; 26:888-896. [PMID: 31332262 PMCID: PMC6980445 DOI: 10.1038/s41380-019-0458-5] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/19/2019] [Accepted: 05/17/2019] [Indexed: 01/18/2023]
Abstract
Vascular endothelial growth factor (VEGF) is associated with the clinical manifestation of Alzheimer's disease (AD). However, the role of the VEGF gene family in neuroprotection is complex due to the number of biological pathways they regulate. This study explored associations between brain expression of VEGF genes with cognitive performance and AD pathology. Genetic, cognitive, and neuropathology data were acquired from the Religious Orders Study and Rush Memory and Aging Project. Expression of ten VEGF ligand and receptor genes was quantified using RNA sequencing of prefrontal cortex tissue. Global cognitive composite scores were calculated from 17 neuropsychological tests. β-amyloid and tau burden were measured at autopsy. Participants (n = 531) included individuals with normal cognition (n = 180), mild cognitive impairment (n = 148), or AD dementia (n = 203). Mean age at death was 89 years and 37% were male. Higher prefrontal cortex expression of VEGFB, FLT4, FLT1, and PGF was associated with worse cognitive trajectories (p ≤ 0.01). Increased expression of VEGFB and FLT4 was also associated with lower cognition scores at the last visit before death (p ≤ 0.01). VEGFB, FLT4, and FLT1 were upregulated among AD dementia compared with normal cognition participants (p ≤ 0.03). All four genes associated with cognition related to elevated β-amyloid (p ≤ 0.01) and/or tau burden (p ≤ 0.03). VEGF ligand and receptor genes, specifically genes relevant to FLT4 and FLT1 receptor signaling, are associated with cognition, longitudinal cognitive decline, and AD neuropathology. Future work should confirm these observations at the protein level to better understand how changes in VEGF transcription and translation relate to neurodegenerative disease.
Collapse
|
6
|
Affiliation(s)
- Vladimir Jurisic
- Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
7
|
Tomatidine Represses Invasion and Migration of Human Osteosarcoma U2OS and HOS Cells by Suppression of Presenilin 1 and c-Raf-MEK-ERK Pathway. Molecules 2020; 25:molecules25020326. [PMID: 31941156 PMCID: PMC7024336 DOI: 10.3390/molecules25020326] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma, which is the most prevalent malignant bone tumor, is responsible for the great majority of bone cancer-associated deaths because of its highly metastatic potential. Although tomatidine is suggested to serve as a chemosensitizer in multidrug-resistant tumors, the anti-metastatic effect of tomatidine in osteosarcoma is still unknown. Here, we tested the hypothesis that tomatidine suppresses migration and invasion, features that are associated with metastatic process in human osteosarcoma cells and also investigate its underlying pathway. Tomatidine, up to 100 μM, without cytotoxicity, inhibited the invasion and migration capabilities of human osteosarcoma U2OS and HOS cells and repressed presenilin 1 (PS-1) expression of U2OS cells. After the knockdown of PS-1, U2OS and HOS cells’ biological behaviors of cellular invasion and migratory potential were significantly reduced. While tomatidine significantly decreased the phosphorylation of c-Raf, mitogen/extracellular signal-regulated kinase (MEK), and extracellular signal-regulated protein kinase (ERK)1/2 in U2OS cells, no obvious influences on p-Jun N-terminal kinase, p38, and Akt, including their phosphorylation, were observed. In ERK 1 silencing U2 OS cells, tomatidine further enhanced the decrease of their migratory potential and invasive activities. We conclude that both PS-1 derived from U2OS and HOS cells and the c-Raf–MEK–ERK pathway contribute to cellular invasion and migration and tomatidine could inhibit the phenomenons. These findings indicate that tomatidine might be a potential candidate for anti-metastasis treatment of human osteosarcoma.
Collapse
|
8
|
Han KY, Chang JH, Azar DT. MMP14-Containing Exosomes Cleave VEGFR1 and Promote VEGFA-Induced Migration and Proliferation of Vascular Endothelial Cells. Invest Ophthalmol Vis Sci 2019; 60:2321-2329. [PMID: 31117124 PMCID: PMC6532701 DOI: 10.1167/iovs.18-26277] [Citation(s) in RCA: 22] [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: 11/27/2018] [Accepted: 04/17/2019] [Indexed: 12/17/2022] Open
Abstract
Purpose Investigate the impact matrix metalloproteinase 14 (MMP14) delivered via exosomes produced by corneal fibroblasts on vascular endothelial growth factor receptor 1 (VEGFR1) cleavage on endothelial cells, and other key processes of angiogenesis. Methods Proteolysis of VEGFR1 and R2 by the catalytic domain of MMP14 was investigated via immunocytochemistry with anti-VEGFR1, anti-VEGFR2, and anti-MMP14 antibodies. Exosomes were isolated via precipitation and serial ultracentrifugation from wild-type (WT) and MMP14 exon4-deficient corneal fibroblasts. Transmission electron microscopy and nanotracking analysis were used to characterize the isolated exosomes. The presence of MMP14 in exosomes from WT fibroblasts was confirmed by Western blotting. VEGFR1 cleavage upon treatment with WT-derived exosomes, Δexon4-derived exosomes, or the pan-MMP inhibitor GM60001 was examined via in vitro proteolysis analysis using recombinant mouse (rm) VEGFR1/R2. Endothelial cell migration and proliferation were investigated using a Boyden chamber assay and BrdU incorporation, respectively. Results WT-derived exosomes specifically cleaved rmVEGFR1 in vitro, whereas Δexon4-derived exosomes did not. Treatment with the pan-MMP inhibitor GM6001 effectively inhibited VEGFR1 cleavage by WT-derived exosomes, confirming the role of MMP14 in this cleavage. WT-derived exosomes induced greater endothelial cell migration (P < 0.01) and proliferation (P < 0.5) compared to Δexon4-derived exosomes. Conclusions MMP14-containing exosomes may be involved in the regulation of corneal neovascularization through degradation of VEGFR1 and VEGFA-induced endothelial cell proliferation and migration.
Collapse
Affiliation(s)
- Kyu-Yeon Han
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States
| | - Dimitri T. Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States
| |
Collapse
|
9
|
Merilahti JAM, Elenius K. Gamma-secretase-dependent signaling of receptor tyrosine kinases. Oncogene 2018; 38:151-163. [PMID: 30166589 PMCID: PMC6756091 DOI: 10.1038/s41388-018-0465-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/28/2022]
Abstract
Human genome harbors 55 receptor tyrosine kinases (RTK). At least half of the RTKs have been reported to be cleaved by gamma-secretase-mediated regulated intramembrane proteolysis. The two-step process involves releasing the RTK ectodomain to the extracellular space by proteolytic cleavage called shedding, followed by cleavage in the RTK transmembrane domain by the gamma-secretase complex resulting in release of a soluble RTK intracellular domain. This intracellular domain, including the tyrosine kinase domain, can in turn translocate to various cellular compartments, such as the nucleus or proteasome. The soluble intracellular domain may interact with transcriptional regulators and other proteins to induce specific effects on cell survival, proliferation, and differentiation, establishing an additional signaling mode for the cleavable RTKs. On the other hand, the same process can facilitate RTK turnover and proteasomal degradation. In this review we focus on the regulation of RTK shedding and gamma-secretase cleavage, as well as signaling promoted by the soluble RTK ICDs. In addition, therapeutic implications of increased knowledge on RTK cleavage on cancer drug development are discussed.
Collapse
Affiliation(s)
- Johannes A M Merilahti
- Institute of Biomedicine, University of Turku, 20520, Turku, Finland.,Medicity Research Laboratory, University of Turku, 20520, Turku, Finland.,Turku Doctoral Programme of Molecular Medicine, University of Turku, 20520, Turku, Finland
| | - Klaus Elenius
- Institute of Biomedicine, University of Turku, 20520, Turku, Finland. .,Medicity Research Laboratory, University of Turku, 20520, Turku, Finland. .,Department of Oncology, Turku University Hospital, 20520, Turku, Finland.
| |
Collapse
|
10
|
Lacal PM, Graziani G. Therapeutic implication of vascular endothelial growth factor receptor-1 (VEGFR-1) targeting in cancer cells and tumor microenvironment by competitive and non-competitive inhibitors. Pharmacol Res 2018; 136:97-107. [PMID: 30170190 DOI: 10.1016/j.phrs.2018.08.023] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 08/24/2018] [Accepted: 08/26/2018] [Indexed: 12/12/2022]
Abstract
The vascular endothelial growth factor receptor-1 (VEGFR-1) is a tyrosine kinase receptor for VEGF-A, VEGF-B, and placental growth factor (PlGF) ligands that is expressed in endothelial, myelomonocytic and tumor cells. VEGF-B and PlGF exclusively bind to VEGFR-1, whereas VEGF-A also binds to VEGFR-2. At variance with VEGFR-2, VEGFR-1 does not play a relevant role in physiological angiogenesis in the adult, while it is important in tumor-associated angiogenesis. VEGFR-1 and PlGF are expressed in a variety of tumors, promote invasiveness and contribute to resistance to anti-VEGF-A therapy. The currently approved antiangiogenic therapies for the treatment of a variety of solid tumors hamper VEGF-A signaling mediated by both VEGFR-2 and VEGFR-1 [i.e., the monoclonal antibody (mAb) anti-VEGF-A bevacizumab, the chimeric molecule aflibercept and several small molecule tyrosine kinase inhibitors] or exclusively by VEGFR-2 (i.e., the mAb anti-VEGFR-2 ramucirumab). However, molecules that interfere with VEGF-A/VEGFR-2 signaling determine severe adverse effects due to inhibition of physiological angiogenesis and their efficacy is hampered by tumor infiltration of protumoral myeloid cells. Blockade of VEGFR-1 may exert anti-tumor activity by multiple mechanisms: a) inhibition of tumor-associated angiogenesis; b) reduction of myeloid progenitor mobilization and tumor infiltration by VEGFR-1 expressing M2 macrophages, which contribute to tumor progression and spreading; c) inhibition of invasiveness, vasculogenic mimicry and survival of VEGFR-1 positive tumor cells. As a consequence of these properties, molecules targeting VEGFR-1 are expected to produce less adverse effects and to counteract resistance towards anti-VEGF-A therapies. More interestingly, selective VEGFR-1 inhibition might enhance the efficacy of immunotherapy with immune checkpoint inhibitors. In this review, we will examine the experimental evidence available so far that supports targeting VEGFR-1 signal transduction pathway for cancer treatment by competitive inhibitors that prevent growth factor interaction with the receptor and non-competitive inhibitors that hamper receptor activation without affecting ligand binding.
Collapse
Affiliation(s)
- Pedro Miguel Lacal
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via Monti di Creta 104, 00167 Rome, Italy.
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy.
| |
Collapse
|
11
|
Raikwar NS, Shibuya M, Thomas CP. VEGF-A selectively inhibits FLT1 ectodomain shedding independent of receptor activation and receptor endocytosis. Am J Physiol Cell Physiol 2018; 315:C214-C224. [PMID: 29719170 PMCID: PMC6139503 DOI: 10.1152/ajpcell.00247.2017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 03/28/2018] [Accepted: 04/11/2018] [Indexed: 11/22/2022]
Abstract
Ectodomain shedding and regulated intracellular proteolysis can determine the fate or function of cell surface proteins. Fms-related tyrosine kinase (FLT) or VEGF receptor 1 is a high-affinity cell surface VEGF-A receptor tyrosine kinase that is constitutively cleaved to release an NH2-terminal VEGF-A binding ectodomain that, once shed, can antagonize the effects of VEGF-A in the extracellular milieu. We evaluated the effect of VEGF-A on FLT1 cleavage in native cells and in transient and stable expression systems. We demonstrate that VEGF-A inhibits FLT1 ectodomain cleavage in a time- and dose-dependent manner, whereas VEGF-A knockdown in HEK293 cells increases ectodomain shedding. Although kinase insert domain receptor (KDR) or VEGF receptor 2, analogous to FLT1, is also subject to extracellular and intracellular cleavage, VEGF-A does not inhibit KDR cleavage. VEGF-A inhibition of FLT1 cleavage is not dependent on FLT1 tyrosine kinase activity or the intracellular FLT1 residues. N-acetylleucylleucylnorleucinal (ALLN), a proteasomal inhibitor; bafilomycin A, an inhibitor of endosomal acidification; and dynasore, a dynamin inhibitor, all increase the abundance of FLT1 and the shed ectodomain, indicating that FLT1 is subject to dynamin-mediated endocytosis and susceptible to proteasomal and lysosomal degradation. VEGF-A inhibition of cleavage is not reversed by ALLN, bafilomycin A, or dynasore. However, a 30 AA deletion in the extracellular immunoglobulin 7 domain leads to enhanced cleavage of Flt1 with a significant reduction of the VEGF inhibitory effect. Our results indicate that the inhibition of FLT1 ectodomain cleavage by VEGF-A is dependent neither on receptor activation nor on internalization nor a consequence of receptor degradation and likely represents a direct inhibitory effect on receptor cleavage.
Collapse
Affiliation(s)
- Nandita S Raikwar
- Department of Internal Medicine, University of Iowa Carver College of Medicine , Iowa City, Iowa
| | - Masabumi Shibuya
- Institute of Physiology and Medicine, Jobu University, Takasaki, Gunma, Japan
| | - Christie P Thomas
- Department of Internal Medicine, University of Iowa Carver College of Medicine , Iowa City, Iowa
- Department of Pediatrics, University of Iowa Carver College of Medicine , Iowa City, Iowa
- Department of Obstetrics, University of Iowa Carver College of Medicine , Iowa City, Iowa
- Graduate Program in Molecular Medicine, University of Iowa Carver College of Medicine , Iowa City, Iowa
- Veterans Affairs Medical Center , Iowa City, Iowa
| |
Collapse
|
12
|
Capossela S, Bertolo A, Gunasekera K, Pötzel T, Baur M, Stoyanov JV. VEGF vascularization pathway in human intervertebral disc does not change during the disc degeneration process. BMC Res Notes 2018; 11:333. [PMID: 29784013 PMCID: PMC5963106 DOI: 10.1186/s13104-018-3441-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 05/11/2018] [Indexed: 12/30/2022] Open
Abstract
Objective During degeneration of the intervertebral disc ingrowth of blood vessels and nerves into the disc are associated with back pain. Vascular endothelial growth factors promote vasculogenesis by binding to the membrane vascular endothelial growth factor receptor 1, while shorter soluble forms of this receptor can inhibit vascularization. We hypothesized that membrane and soluble receptor forms might change between stages of intervertebral disc degeneration. Results Expression of soluble and membrane forms of vascular endothelial growth factor receptor 1 in human degenerated intervertebral discs and healthy bovine caudal discs was assessed by qRT-PCR and immunoblot. Comparative microarray meta-analysis across disc degeneration grades showed that membrane and soluble forms of this receptor, together with other components of classic vascularization pathways, are constitutively expressed across human disc degeneration stages. Contrary to our hypothesis, we observed that expression of the classic vascularization pathway is stable across degeneration stages and we assume that soluble vascular endothelial growth factor receptor 1 does not contribute to prevent disc degeneration. However, we observed increased expression levels of genes involved in alternative vascularization signalling pathways in severely degenerated discs, suggesting that abnormal vascularization is part of the pathological progression of disc degeneration. Electronic supplementary material The online version of this article (10.1186/s13104-018-3441-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Simona Capossela
- Biomedical Laboratories, Swiss Paraplegic Research, 6207, Nottwil, Switzerland
| | - Alessandro Bertolo
- Biomedical Laboratories, Swiss Paraplegic Research, 6207, Nottwil, Switzerland
| | - Kapila Gunasekera
- Biomedical Laboratories, Swiss Paraplegic Research, 6207, Nottwil, Switzerland
| | | | - Martin Baur
- Swiss Paraplegic Centre, Nottwil, Switzerland.,Cantonal Hospital of Lucerne, Lucerne, Switzerland
| | - Jivko V Stoyanov
- Biomedical Laboratories, Swiss Paraplegic Research, 6207, Nottwil, Switzerland.
| |
Collapse
|
13
|
Palmer KR, Tong S, Kaitu'u-Lino TJ. Placental-specific sFLT-1: role in pre-eclamptic pathophysiology and its translational possibilities for clinical prediction and diagnosis. Mol Hum Reprod 2018; 23:69-78. [PMID: 27986932 DOI: 10.1093/molehr/gaw077] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/09/2016] [Indexed: 11/12/2022] Open
Abstract
Pre-eclampsia is a common obstetric complication globally responsible for a significant burden of maternal and perinatal morbidity and mortality. Central to its pathophysiology is the anti-angiogenic protein, soluble fms-like tyrosine kinase-1 (sFLT-1). sFLT-1 is released from a range of tissues into the circulation, where it antagonizes the activity of vascular endothelial growth factor and placental growth factor leading to endothelial dysfunction. It is this widespread endothelial dysfunction that produces the clinical features of pre-eclampsia including hypertension and proteinuria. There are multiple splice variants of sFLT-1. One, known as sFLT-1 e15a, evolved quite recently and is only present in humans and higher order primates. This sFLT-1 variant is also the main sFLT-1 secreted from the placenta. Recent work has shown that sFLT-1 e15a is significantly elevated in the placenta and circulation of women with pre-eclampsia. It is also biologically active, capable of causing endothelial dysfunction and the end-organ dysfunction seen in pre-eclampsia. Indeed, the over-expression of sFLT-1 e15a in mice recapitulates the pre-eclamptic phenotype in pregnancy. Therefore, here we propose that sFLT-1 e15a may be the sFLT-1 variant primarily responsible for pre-eclampsia, a uniquely human disease. Furthermore, this placental-specific sFLT-1 variant provides promise for use as an accurate biomarker in the prediction or diagnosis of pre-eclampsia.
Collapse
Affiliation(s)
- K R Palmer
- Department of Obstetrics and Gynaecology, Monash University, Monash Medical Centre, 246 Clayton Rd, Clayton, 3168 Victoria, Australia.,Translational Obstetric Group, University of Melbourne, Mercy Hospital for Women, 163 Studley Rd, Heidelberg, 3084 Victoria, Australia
| | - S Tong
- Translational Obstetric Group, University of Melbourne, Mercy Hospital for Women, 163 Studley Rd, Heidelberg, 3084 Victoria, Australia
| | - T J Kaitu'u-Lino
- Translational Obstetric Group, University of Melbourne, Mercy Hospital for Women, 163 Studley Rd, Heidelberg, 3084 Victoria, Australia
| |
Collapse
|
14
|
Palmer K. Assessing the Circulating Placental-Specific Anti-angiogenic Protein sFLT-1 e15a in Preeclampsia. Methods Mol Biol 2018; 1710:27-37. [PMID: 29196992 DOI: 10.1007/978-1-4939-7498-6_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Preeclampsia is a common obstetric complication globally responsible for a significant burden of maternal and perinatal morbidity and mortality. The anti-angiogenic protein, sFLT-1, plays a central role in its pathophysiology. sFLT-1 is released from a range of tissues into the circulation, where it antagonizes the activity of vascular endothelial growth factor and placental growth factor leading to endothelial dysfunction. The resulting widespread endothelial dysfunction produces the clinical features of preeclampsia including hypertension and proteinuria. Multiple splice variants of sFLT-1 have been identified, with one, known as sFLT-1 e15a, present only in humans and higher-order primates. This sFLT-1 variant is also the main form of sFLT-1 produced by the placenta. Recent work has shown that sFLT-1 e15a is significantly elevated in the placenta and circulation of women with preeclampsia. It is also biologically active, capable of causing endothelial dysfunction and end-organ dysfunction seen in preeclampsia. Indeed, overexpression of sFLT-1 e15a in mice recapitulates the preeclamptic phenotype in pregnancy. No commercial assay currently exists to analyze sFLT-1 e15a protein levels. Here, a new ELISA method to determine circulating sFLT-1 variant levels is described.
Collapse
Affiliation(s)
- Kirsten Palmer
- Department of Obstetrics and Gynecology, School of Clinical Sciences, Monash University, Level 5, 246 Clayton Rd, Clayton, 3168, VIC, Australia.
| |
Collapse
|
15
|
Merilahti JAM, Ojala VK, Knittle AM, Pulliainen AT, Elenius K. Genome-wide screen of gamma-secretase-mediated intramembrane cleavage of receptor tyrosine kinases. Mol Biol Cell 2017; 28:3123-3131. [PMID: 28904208 PMCID: PMC5662267 DOI: 10.1091/mbc.e17-04-0261] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/11/2017] [Accepted: 09/06/2017] [Indexed: 12/20/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) have been demonstrated to signal via regulated intramembrane proteolysis, in which ectodomain shedding and subsequent intramembrane cleavage by gamma-secretase leads to release of a soluble intracellular receptor fragment with functional activity. For most RTKs, however, it is unknown whether they can exploit this new signaling mechanism. Here we used a system-wide screen to address the frequency of susceptibility to gamma-secretase cleavage among human RTKs. The screen covering 45 of the 55 human RTKs identified 12 new as well as all nine previously published gamma-secretase substrates. We biochemically validated the screen by demonstrating that the release of a soluble intracellular fragment from endogenous AXL was dependent on the sheddase disintegrin and metalloprotease 10 (ADAM10) and the gamma-secretase component presenilin-1. Functional analysis of the cleavable RTKs indicated that proliferation promoted by overexpression of the TAM family members AXL or TYRO3 depends on gamma-secretase cleavage. Taken together, these data indicate that gamma-secretase-mediated cleavage provides an additional signaling mechanism for numerous human RTKs.
Collapse
Affiliation(s)
- Johannes A M Merilahti
- Department of Medical Biochemistry and Genetics, University of Turku, 20520 Turku, Finland.,Medicity Research Laboratory, University of Turku, 20520 Turku, Finland.,Turku Doctoral Programme of Molecular Medicine, University of Turku, 20520 Turku, Finland
| | - Veera K Ojala
- Department of Medical Biochemistry and Genetics, University of Turku, 20520 Turku, Finland
| | - Anna M Knittle
- Department of Medical Biochemistry and Genetics, University of Turku, 20520 Turku, Finland
| | - Arto T Pulliainen
- Department of Medical Biochemistry and Genetics, University of Turku, 20520 Turku, Finland
| | - Klaus Elenius
- Department of Medical Biochemistry and Genetics, University of Turku, 20520 Turku, Finland .,Medicity Research Laboratory, University of Turku, 20520 Turku, Finland.,Department of Oncology, Turku University Hospital, 20520 Turku, Finland
| |
Collapse
|
16
|
Tien WS, Chen JH, Wu KP. SheddomeDB: the ectodomain shedding database for membrane-bound shed markers. BMC Bioinformatics 2017; 18:42. [PMID: 28361715 PMCID: PMC5374707 DOI: 10.1186/s12859-017-1465-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A number of membrane-anchored proteins are known to be released from cell surface via ectodomain shedding. The cleavage and release of membrane proteins has been shown to modulate various cellular processes and disease pathologies. Numerous studies revealed that cell membrane molecules of diverse functional groups are subjected to proteolytic cleavage, and the released soluble form of proteins may modulate various signaling processes. Therefore, in addition to the secreted protein markers that undergo secretion through the secretory pathway, the shed membrane proteins may comprise an additional resource of noninvasive and accessible biomarkers. In this context, identifying the membrane-bound proteins that will be shed has become important in the discovery of clinically noninvasive biomarkers. Nevertheless, a data repository for biological and clinical researchers to review the shedding information, which is experimentally validated, for membrane-bound protein shed markers is still lacking. RESULTS In this study, the database SheddomeDB was developed to integrate publicly available data of the shed membrane proteins. A comprehensive literature survey was performed to collect the membrane proteins that were verified to be cleaved or released in the supernatant by immunological-based validation experiments. From 436 studies on shedding, 401 validated shed membrane proteins were included, among which 199 shed membrane proteins have not been annotated or validated yet by existing cleavage databases. SheddomeDB attempted to provide a comprehensive shedding report, including the regulation of shedding machinery and the related function or diseases involved in the shedding events. In addition, our published tool ShedP was embedded into SheddomeDB to support researchers for predicting the shedding event on unknown or unrecorded membrane proteins. CONCLUSIONS To the best of our knowledge, SheddomeDB is the first database for the identification of experimentally validated shed membrane proteins and currently may provide the most number of membrane proteins for reviewing the shedding information. The database included membrane-bound shed markers associated with numerous cellular processes and diseases, and some of these markers are potential novel markers because they are not annotated or validated yet in other databases. SheddomeDB may provide a useful resource for discovering membrane-bound shed markers. The interactive web of SheddomeDB is publicly available at http://bal.ym.edu.tw/SheddomeDB/ .
Collapse
Affiliation(s)
- Wei-Sheng Tien
- Institute of Biomedical Informatics, National Yang Ming University, Taipei, 112, Taiwan.,Bioinformatics Program, Taiwan International Graduate Program, Academia Sinica, Taipei, 115, Taiwan
| | - Jun-Hong Chen
- Department of Computer Science, National Taipei University of Education, Taipei, 106, Taiwan
| | - Kun-Pin Wu
- Institute of Biomedical Informatics, National Yang Ming University, Taipei, 112, Taiwan.
| |
Collapse
|
17
|
Miller MA, Sullivan RJ, Lauffenburger DA. Molecular Pathways: Receptor Ectodomain Shedding in Treatment, Resistance, and Monitoring of Cancer. Clin Cancer Res 2016; 23:623-629. [PMID: 27895032 DOI: 10.1158/1078-0432.ccr-16-0869] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/01/2016] [Accepted: 11/01/2016] [Indexed: 12/21/2022]
Abstract
Proteases known as sheddases cleave the extracellular domains of their substrates from the cell surface. The A Disintegrin and Metalloproteinases ADAM10 and ADAM17 are among the most prominent sheddases, being widely expressed in many tissues, frequently overexpressed in cancer, and promiscuously cleaving diverse substrates. It is increasingly clear that the proteolytic shedding of transmembrane receptors impacts pathophysiology and drug response. Receptor substrates of sheddases include the cytokine receptors TNFR1 and IL6R; the Notch receptors; type-I and -III TGFβ receptors; receptor tyrosine kinases (RTK) such as HER2, HER4, and VEGFR2; and, in particular, MET and TAM-family RTKs AXL and Mer (MerTK). Activation of receptor shedding by mechanical cues, hypoxia, radiation, and phosphosignaling offers insight into mechanisms of drug resistance. This particularly holds for kinase inhibitors targeting BRAF (such as vemurafenib and dabrafenib) and MEK (such as trametinib and cobimetinib), along with direct sheddase inhibitors. Receptor proteolysis can be detected in patient fluids and is especially relevant in melanoma, glioblastoma, lung cancer, and triple-negative breast cancer where RTK substrates, MAPK signaling, and ADAMs are frequently dysregulated. Translatable strategies to exploit receptor shedding include combination kinase inhibitor regimens, recombinant decoy receptors based on endogenous counterparts, and, potentially, immunotherapy. Clin Cancer Res; 23(3); 623-9. ©2016 AACR.
Collapse
Affiliation(s)
- Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ryan J Sullivan
- Division of Medical Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| |
Collapse
|
18
|
Ectodomain cleavage of FLT1 regulates receptor activation and function and is not required for its downstream intracellular cleavage. Exp Cell Res 2016; 344:103-111. [PMID: 27017929 DOI: 10.1016/j.yexcr.2016.03.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/18/2016] [Accepted: 03/22/2016] [Indexed: 01/18/2023]
Abstract
FLT1 is a cell surface VEGF receptor which is cleaved to release an N-terminal ectodomain which binds VEGF and PlGF and can antagonize the effects of VEGF in the extracellular milieu. To further evaluate FLT1 processing we expressed tagged FLT1 constructs in HEK293 and COS7 cells where we demonstrate, by deletion mapping, that the cleavage site is immediately adjacent to the transmembrane domain (TMD) between residues 759 and 763. Cleavage reciprocally regulates free VEGF in conditioned media and we show that the cleavage site is also transferable to another transmembrane receptor. A second cleavage event downstream of the ectodomain cleavage releases a cytosolic C-terminal FLT1 fragment and this intracellular cleavage of FLT1 is not catalyzed or regulated by the upstream ectodomain cleavage since abolition of the ectodomain cleavage has no impact on the downstream cleavage event. The downstream cleavage event is not susceptible to γ-secretase inhibitors and overexpression of presenilin 1, the catalytic subunit of γ-secretase did not change the downstream intracellular cleavage event. Furthermore, this cleavage did not occur via a previously published valine residue (767V) in the TMD of FLT1, indicating the existence of another cleavage pathway. We tested the impact of the ectodomain cleavage on p44/42 MAP kinase activation and demonstrate that compared to wild type FLT1, cleavage resistant FLT1 constructs failed to stimulate p44/42 MAP kinase activation. Our results indicate that FLT1 ectodomain cleavage not only regulates the availability of free VEGF in the extracellular milieu but also regulates cellular signaling via the ERK kinase pathway.
Collapse
|
19
|
Li P, Lin X, Zhang JR, Li Y, Lu J, Huang FC, Zheng CH, Xie JW, Wang JB, Huang CM. The expression of presenilin 1 enhances carcinogenesis and metastasis in gastric cancer. Oncotarget 2016; 7:10650-62. [PMID: 26872378 PMCID: PMC4891148 DOI: 10.18632/oncotarget.7298] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 01/19/2016] [Indexed: 12/12/2022] Open
Abstract
Presenilin 1 (PS-1, encoded by PSEN1) is a part of the gamma- (γ-) secretase complex. Mutations in PSEN1 cause the majority of cases of familial Alzheimer's disease (FAD). Although in recent years PS-1 has been implicated as a tumor enhancer in various cancers, nothing is known regarding its role in gastric cancer (GC). In the present study, we investigate the role and clinical significance of PS-1 in GC. We observed that PS-1 was significantly upregulated and amplified in GC tissues and cell lines, and its aberrant expression was positively correlated with lymph node metastasis and with poor overall survival. Furthermore, PS-1 promoted tumor invasion and metastasis of GC both in vitro and vivo without affecting the proliferation of GC cells (MGC-803 and MKN-45). The results of treatment with the γ-secretase inhibitor DAPT were consistent with the outcomes of PS-1 silencing. PS-1/γ-secretase cleaves E-cadherin and releases its bound protein partner, β-catenin, from the actin cytoskeleton, thereby allowing it to translocate into the nucleus and to activate the TCF/LEF-1 transcriptional activator, which may promote GC invasion and metastasis.In conclusion, PS-1 promotes invasion and metastasis in GC and may represent a novel prognostic biomarker and potential therapeutic target for GC treatment.
Collapse
Affiliation(s)
- Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Xi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Jun-Rong Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Yun Li
- Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Fei-Chao Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| |
Collapse
|
20
|
Morine KJ, Paruchuri V, Qiao X, Mohammad N, Mcgraw A, Yunis A, Jaffe I, Kapur NK. Circulating multimarker profile of patients with symptomatic heart failure supports enhanced fibrotic degradation and decreased angiogenesis. Biomarkers 2015; 21:91-7. [PMID: 26667393 DOI: 10.3109/1354750x.2015.1118539] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Heart failure (HF) involves myocardial fibrosis and dysregulated angiogenesis. OBJECTIVE We explored whether biomarkers of fibrosis and angiogenesis correlate with HF severity. METHODS Biomarkers of fibrosis [procollagen types I and III (PIP and P3NP), carboxyterminal-telopeptide of type I collagen (ICTP), matrix metalloproteases (MMP2 and MMP9), tissue inhibitor of MMP1 (TIMP1)]; and angiogenesis [placental growth factor (PGF), vascular endothelial growth factor (VEGF), soluble Fms-like tyrosine kinase-1 (sFlt1)] were measured in 52 HF patients and 19 controls. RESULTS P3NP, ICTP, MMP2, TIMP1, PGF, and sFlt1 levels were elevated in HF, while PIP/ICTP, PGF/sFlt1, and VEGF/sFlt1 ratios were reduced. PIP/ICTP, MMP-9/TIMP1, and VEGF/sFlt1 ratios were lowest among patients with severe HF. CONCLUSIONS Severe HF is associated with collagen breakdown and reduced angiogenesis. A multimarker approach may guide therapeutic targeting of fibrosis and angiogenesis in HF.
Collapse
Affiliation(s)
- Kevin J Morine
- a Division of Cardiology , Department of Medicine, Tufts Medical Center, Molecular Cardiology Research Institute , Boston , MA , USA
| | - Vikram Paruchuri
- a Division of Cardiology , Department of Medicine, Tufts Medical Center, Molecular Cardiology Research Institute , Boston , MA , USA
| | - Xiaoying Qiao
- a Division of Cardiology , Department of Medicine, Tufts Medical Center, Molecular Cardiology Research Institute , Boston , MA , USA
| | - Najwa Mohammad
- a Division of Cardiology , Department of Medicine, Tufts Medical Center, Molecular Cardiology Research Institute , Boston , MA , USA
| | - Adam Mcgraw
- a Division of Cardiology , Department of Medicine, Tufts Medical Center, Molecular Cardiology Research Institute , Boston , MA , USA
| | - Adil Yunis
- a Division of Cardiology , Department of Medicine, Tufts Medical Center, Molecular Cardiology Research Institute , Boston , MA , USA
| | - Iris Jaffe
- a Division of Cardiology , Department of Medicine, Tufts Medical Center, Molecular Cardiology Research Institute , Boston , MA , USA
| | - Navin K Kapur
- a Division of Cardiology , Department of Medicine, Tufts Medical Center, Molecular Cardiology Research Institute , Boston , MA , USA
| |
Collapse
|
21
|
Hod T, Cerdeira AS, Karumanchi SA. Molecular Mechanisms of Preeclampsia. Cold Spring Harb Perspect Med 2015; 5:cshperspect.a023473. [PMID: 26292986 DOI: 10.1101/cshperspect.a023473] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Preeclampsia is a pregnancy-specific disease characterized by new onset hypertension and proteinuria after 20 wk of gestation. It is a leading cause of maternal and fetal morbidity and mortality worldwide. Exciting discoveries in the last decade have contributed to a better understanding of the molecular basis of this disease. Epidemiological, experimental, and therapeutic studies from several laboratories have provided compelling evidence that an antiangiogenic state owing to alterations in circulating angiogenic factors leads to preeclampsia. In this review, we highlight the role of key circulating antiangiogenic factors as pathogenic biomarkers and in the development of novel therapies for preeclampsia.
Collapse
Affiliation(s)
- Tammy Hod
- Department of Medicine, Obstetrics & Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02214
| | - Ana Sofia Cerdeira
- Department of Medicine, Obstetrics & Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02214 Gulbenkian Program for Advanced Medical Education, 1067-001 Lisbon, Portugal
| | - S Ananth Karumanchi
- Department of Medicine, Obstetrics & Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02214 Howard Hughes Medical Institute, Chevy Chase, Maryland 20815
| |
Collapse
|
22
|
The cellular response to vascular endothelial growth factors requires co-ordinated signal transduction, trafficking and proteolysis. Biosci Rep 2015; 35:BSR20150171. [PMID: 26285805 PMCID: PMC4613718 DOI: 10.1042/bsr20150171] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/18/2015] [Indexed: 01/18/2023] Open
Abstract
VEGFs (vascular endothelial growth factors) are a family of conserved disulfide-linked soluble secretory glycoproteins found in higher eukaryotes. VEGFs mediate a wide range of responses in different tissues including metabolic homoeostasis, cell proliferation, migration and tubulogenesis. Such responses are initiated by VEGF binding to soluble and membrane-bound VEGFRs (VEGF receptor tyrosine kinases) and co-receptors. VEGF and receptor splice isoform diversity further enhances complexity of membrane protein assembly and function in signal transduction pathways that control multiple cellular responses. Different signal transduction pathways are simultaneously activated by VEGFR-VEGF complexes with membrane trafficking along the endosome-lysosome network further modulating signal output from multiple enzymatic events associated with such pathways. Balancing VEGFR-VEGF signal transduction with trafficking and proteolysis is essential in controlling the intensity and duration of different intracellular signalling events. Dysfunction in VEGF-regulated signal transduction is important in chronic disease states including cancer, atherosclerosis and blindness. This family of growth factors and receptors is an important model system for understanding human disease pathology and developing new therapeutics for treating such ailments.
Collapse
|
23
|
Chen MK, Hung MC. Proteolytic cleavage, trafficking, and functions of nuclear receptor tyrosine kinases. FEBS J 2015; 282:3693-721. [PMID: 26096795 DOI: 10.1111/febs.13342] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/18/2015] [Accepted: 06/09/2015] [Indexed: 01/18/2023]
Abstract
Intracellular localization has been reported for over three-quarters of receptor tyrosine kinase (RTK) families in response to environmental stimuli. Internalized RTK may bind to non-canonical substrates and affect various cellular processes. Many of the intracellular RTKs exist as fragmented forms that are generated by γ-secretase cleavage of the full-length receptor, shedding, alternative splicing, or alternative translation initiation. Soluble RTK fragments are stabilized and intracellularly transported into subcellular compartments, such as the nucleus, by binding to chaperone or transcription factors, while membrane-bound RTKs (full-length or truncated) are transported from the plasma membrane to the ER through the well-established Rab- or clathrin adaptor protein-coated vesicle retrograde trafficking pathways. Subsequent nuclear transport of membrane-bound RTK may occur via two pathways, INFS or INTERNET, with the former characterized by release of receptors from the ER into the cytosol and the latter characterized by release of membrane-bound receptor from the ER into the nucleoplasm through the inner nuclear membrane. Although most non-canonical intracellular RTK signaling is related to transcriptional regulation, there may be other functions that have yet to be discovered. In this review, we summarize the proteolytic processing, intracellular trafficking and nuclear functions of RTKs, and discuss how they promote cancer progression, and their clinical implications.
Collapse
Affiliation(s)
- Mei-Kuang Chen
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mien-Chie Hung
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Center of Molecular Medicine and Graduate Institute of Cancer Biology, China Medical University, Taichung, Taiwan.,Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
24
|
Souders CA, Maynard SE, Yan J, Wang Y, Boatright NK, Sedan J, Balyozian D, Cheslock PS, Molrine DC, Simas TAM. Circulating Levels of sFlt1 Splice Variants as Predictive Markers for the Development of Preeclampsia. Int J Mol Sci 2015; 16:12436-53. [PMID: 26042465 PMCID: PMC4490453 DOI: 10.3390/ijms160612436] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/06/2015] [Accepted: 05/25/2015] [Indexed: 11/17/2022] Open
Abstract
Angiogenic biomarkers, including soluble fms-like tyrosine kinase 1 (sFlt1), are thought to be predictors of preeclampsia onset; however, improvement is needed before a widespread diagnostic test can be utilized. Here we describe the development and use of diagnostic monoclonal antibodies specific to the two main splice variants of sFlt1, sFlt1-1 and sFlt1-14. These antibodies were selected for their sensitivity and specificity to their respective sFlt1 isoform in a capture ELISA format. Data from this pilot study suggest that sFlt1-1 may be more predictive of preeclampsia than total sFlt1. It may be possible to improve current diagnostic platforms if more specific antibodies are utilized.
Collapse
Affiliation(s)
- Colby A Souders
- MassBiologics of the University of Massachusetts Medical School, Boston, MA 02126, USA.
| | - Sharon E Maynard
- Department of Medicine, Division of Nephrology, Lehigh Valley Health Network, University of South Florida Morsani College of Medicine, Allentown, PA 18105, USA.
| | - Jing Yan
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Yang Wang
- MassBiologics of the University of Massachusetts Medical School, Boston, MA 02126, USA.
| | - Naomi K Boatright
- MassBiologics of the University of Massachusetts Medical School, Boston, MA 02126, USA.
| | - Jessica Sedan
- MassBiologics of the University of Massachusetts Medical School, Boston, MA 02126, USA.
| | - David Balyozian
- MassBiologics of the University of Massachusetts Medical School, Boston, MA 02126, USA.
| | - Peter S Cheslock
- MassBiologics of the University of Massachusetts Medical School, Boston, MA 02126, USA.
| | - Deborah C Molrine
- MassBiologics of the University of Massachusetts Medical School, Boston, MA 02126, USA.
| | - Tiffany A Moore Simas
- Department of Obstetrics and Gynecology, University of Massachusetts Medical School/ UMass Memorial Health Care, Worcester, MA 01605, USA.
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
25
|
Bertram A, Lovric S, Engel A, Beese M, Wyss K, Hertel B, Park JK, Becker JU, Kegel J, Haller H, Haubitz M, Kirsch T. Circulating ADAM17 Level Reflects Disease Activity in Proteinase-3 ANCA-Associated Vasculitis. J Am Soc Nephrol 2015; 26:2860-70. [PMID: 25788529 DOI: 10.1681/asn.2014050477] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 01/04/2015] [Indexed: 12/14/2022] Open
Abstract
ANCA-associated vasculitides are characterized by inflammatory destruction of small vessels accompanied by enhanced cleavage of membrane-bound proteins. One of the main proteases responsible for ectodomain shedding is disintegrin and metalloproteinase domain-containing protein 17 (ADAM17). Given its potential role in aggravating vascular dysfunction, we examined the role of ADAM17 in active proteinase-3 (PR3)-positive ANCA-associated vasculitis (AAV). ADAM17 concentration was significantly increased in plasma samples from patients with active PR3-AAV compared with samples from patients in remission or from other controls with renal nonvascular diseases. Comparably, plasma levels of the ADAM17 substrate syndecan-1 were significantly enhanced in active AAV. We also observed that plasma-derived ADAM17 retained its specific proteolytic activity and was partly located on extracellular microparticles. Transcript levels of ADAM17 were increased in blood samples of patients with active AAV, but those of ADAM10 or tissue inhibitor of metalloproteinases 3, which inhibits ADAMs, were not. We also performed a microRNA (miR) screen and identified miR-634 as significantly upregulated in blood samples from patients with active AAV. In vitro, miR-634 mimics induced a proinflammatory phenotype in monocyte-derived macrophages, with enhanced expression and release of ADAM17 and IL-6. These data suggest that ADAM17 has a prominent role in AAV and might account for the vascular complications associated with this disease.
Collapse
Affiliation(s)
- Anna Bertram
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Svjetlana Lovric
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Alissa Engel
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Michaela Beese
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Kristin Wyss
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Barbara Hertel
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Joon-Keun Park
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Jan U Becker
- Institute for Forensic Medicine, Hannover Medical School, Hannover, Germany; and
| | - Johanna Kegel
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Hermann Haller
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| | - Marion Haubitz
- Department of Nephrology and Hypertension, Center for Internal Medicine and Medical Clinic III, Klinikum Fulda, Fulda, Germany
| | - Torsten Kirsch
- Department of Nephrology and Hypertension, Center for Internal Medicine and
| |
Collapse
|
26
|
Li C, Raikwar NS, Santillan MK, Santillan DA, Thomas CP. Aspirin inhibits expression of sFLT1 from human cytotrophoblasts induced by hypoxia, via cyclo-oxygenase 1. Placenta 2015; 36:446-53. [PMID: 25638730 DOI: 10.1016/j.placenta.2015.01.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 12/29/2014] [Accepted: 01/08/2015] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Elevated circulating soluble FLT1 (sFLT1) levels seen in preeclampsia may play a role in its development. Aspirin is recommended for prevention of preeclampsia. We hypothesized that aspirin may inhibit the production of sFlt1. METHODS Placentas from women with and without preeclampsia were collected. Primary cytotrophoblasts (CTBs) were cultured from normal placentas and treated with aspirin, sc-560, a COX1 inhibitor or celecoxib, a COX2 inhibitor. The expression of sFLT1, FLT1, COX1 and COX2 was studied. The effect of aspirin on sFlt1 expression was also studied in HEK293 cells and in HTR-8/SVNeo cells. RESULTS The expression of sFLT1 was increased in preeclamptic placentas compared to control placentas and the expression and release of sFLT1 increased in CTBs exposed to 2% O2 compared to controls. Aspirin at 3 and 12 mM concentration reduced the expression and release of sFLT1 in CTBs. Aspirin also inhibited sFlt1 expression from HTR-8/SVNeo and HEK293 cells. Sc-560, but not celecoxib, reduced sFLT1 expression and release from CTBs. Aspirin and sc-560 also reduced hypoxia-induced FLT1 mRNA expression and inhibited COX1 mRNA in CTBs. DISCUSSION This study confirms that sFLT1 expression is increased in preeclamptic placentas and in CTBs exposed to hypoxia. Aspirin inhibits the production sFLT1 in CTBs and in HTR-8/SVNeo. Sc-560 recapitulated the effects of aspirin on sFLT1 expression and release in CTBs suggesting that the aspirin effect may be mediated via inhibition of COX1. The study increases our understanding of the mechanisms regulating sFlt1 expression and provides a plausible explanation for the effect of aspirin to prevent preeclampsia.
Collapse
Affiliation(s)
- C Li
- Department of Obstetrics and Gynecology, Iowa City, IA, USA
| | - N S Raikwar
- Division of Nephrology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - M K Santillan
- Department of Obstetrics and Gynecology, Iowa City, IA, USA
| | - D A Santillan
- Department of Obstetrics and Gynecology, Iowa City, IA, USA
| | - C P Thomas
- Department of Obstetrics and Gynecology, Iowa City, IA, USA; Division of Nephrology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Veterans Affairs Medical Center, Iowa City, IA, USA.
| |
Collapse
|
27
|
Raikwar NS, Liu KZ, Thomas CP. N-terminal cleavage and release of the ectodomain of Flt1 is mediated via ADAM10 and ADAM 17 and regulated by VEGFR2 and the Flt1 intracellular domain. PLoS One 2014; 9:e112794. [PMID: 25387128 PMCID: PMC4227870 DOI: 10.1371/journal.pone.0112794] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 10/20/2014] [Indexed: 11/25/2022] Open
Abstract
Flt is one of the cell surface VEGF receptors which can be cleaved to release an N-terminal extracellular fragment which, like alternately transcribed soluble Flt1 (sFlt1), can antagonize the effects of VEGF. In HUVEC and in HEK293 cells where Flt1 was expressed, metalloprotease inhibitors reduced Flt1 N-terminal cleavage. Overexpression of ADAM10 and ADAM17 increased cleavage while knockdown of ADAM10 and ADAM17 reduced N-terminal cleavage suggesting that these metalloproteases were responsible for Flt1 cleavage. Protein kinase C (PKC) activation increased the abundance and the cleavage of Flt1 but this did not require any residues within the intracellular portion of Flt1. ALLN, a proteasomal inhibitor, increased the abundance of Flt1 which was additive to the effect of PKC. Removal of the entire cytosolic region of Flt1 appeared to stimulate cleavage of Flt1 and Flt1 was no longer sensitive to ALLN suggesting that the cytosolic region contained a degradation domain. Knock down of c-CBL, a ring finger ubiquitin ligase, in HEK293 cells increased the expression of Flt1 although it did not appear to require a previously published tyrosine residue (1333Y) in the C-terminus of Flt1. Increasing VEGFR2 expression increased VEGF-stimulated sFlt1 expression and progressively reduced the cleavage of Flt1 with Flt1 staying bound to VEGFR2 as a heterodimer. Our results imply that secreted sFlt1 and cleaved Flt1 will tend to have local effects as a VEGF antagonist when released from cells expressing VEGFR2 and more distant effects when released from cells lacking VEGFR2.
Collapse
Affiliation(s)
- Nandita S. Raikwar
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, IA, United States of America
| | - Kang Z. Liu
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, IA, United States of America
| | - Christie P. Thomas
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, IA, United States of America
- Department of Pediatrics, University of Iowa College of Medicine, Iowa City, IA, United States of America
- Department of Obstetrics, University of Iowa College of Medicine, Iowa City, IA, United States of America
- Department of Molecular and Cellular Biology, University of Iowa College of Medicine, Iowa City, IA, United States of America
- Veterans Affairs Medical Center, Iowa City, IA, United States of America
- * E-mail:
| |
Collapse
|
28
|
Han KY, Chang JH, Dugas-Ford J, Alexander JS, Azar DT. Involvement of lysosomal degradation in VEGF-C-induced down-regulation of VEGFR-3. FEBS Lett 2014; 588:4357-63. [PMID: 25281926 DOI: 10.1016/j.febslet.2014.09.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 09/22/2014] [Indexed: 11/17/2022]
Abstract
The vascular endothelial growth factor (VEGF)-C-induced down-regulation of VEGF receptor (VEGFR)-3 is important in lymphangiogenesis. Here, we demonstrate that VEGF-C, -D, and -C156S, but not VEGF-A, down-regulate VEGFR-3. VEGF-C stimulates VEGFR-3 tyrosyl phosphorylation and transient phosphorylation of extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinases in lymphatic endothelial cells. VEGF-C-induced down-regulation of VEGFR-3 was blocked by a VEGF-C trap, tyrosine kinase inhibitor, and leupeptin, pepstatin, and E64 (LPE), but was unaffected by Notch 1 activator and γ-secretase inhibitors. Our findings indicate that VEGF-C down-regulates VEGFR-3 in lymphatic endothelial cells through VEGFR-3 kinase activation and, in part, via lysosomal degradation.
Collapse
Affiliation(s)
- Kyu-Yeon Han
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, United States
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, United States.
| | - Jennifer Dugas-Ford
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, United States
| | - Jonathan S Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States. http://www.nature.com/nm/journal/v15/n9/abs/nm.2018.html-a8
| | - Dimitri T Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
29
|
Loss of presenilin 2 is associated with increased iPLA2 activity and lung tumor development. Oncogene 2014; 33:5193-200. [PMID: 24858037 PMCID: PMC4287650 DOI: 10.1038/onc.2014.128] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/11/2014] [Accepted: 04/14/2014] [Indexed: 12/17/2022]
Abstract
Presenilins are the enzymatic components of γ-secretase complex that cleaves amyloid precursor protein, Notch and β-catenin, which has critical roles in the development of Alzheimer's disease and cancer cell growth. Therefore, in the present study, we studied the effects and mechanisms of PS2 knockout on lung cancer development and possible mechanisms as a key regulator of lung tumor development. We compared carcinogen-induced tumor growth between PS2 knockout mice and wild-type mice. PS2 knockout mice showed increased urethane (1 mg/g)-induced lung tumor incidence when compared with that of wild-type mice with decreased activity of γ-secretase in the lung tumor tissues. Consequently, iPLA2 activities in lung tumor tissues of PS2 knockout mice were much higher than in tumor tissues of wild-type mice. Furthermore, knockdown of PS2 using PS2 siRNA decreased γ-secretase activity with increased iPLA2 activity in the lung cancer cells (A549 and NCI-H460), leading to increased lung cancer cell growth. PS2 knockout mice and PS2 knockdown lung cancer cells showed increased DNA-binding activities of nuclear factor kappa-beta, signal transducer and activator of transcription 3 (STAT3) and AP-1 which are critical transcriptional factors of iPLA2 than those of PS2 wild-type mice and control lung cancer cells. Taken together, these results suggest that the loss of PS2 could have a critical role in lung tumor development through the upregulation of iPLA2 activity by reducing γ-secretase.
Collapse
|
30
|
Di Marco GS, Pavenstädt H, Brand M. Soluble Flt-1 release response to heparin use: implications for dialysis patients? Nephrol Dial Transplant 2014; 29:1112-5. [PMID: 24578469 DOI: 10.1093/ndt/gfu043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
| | - Hermann Pavenstädt
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Marcus Brand
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| |
Collapse
|
31
|
Unlocking Doors without Keys: Activation of Src by Truncated C-terminal Intracellular Receptor Tyrosine Kinases Lacking Tyrosine Kinase Activity. Cells 2014; 3:92-111. [PMID: 24709904 PMCID: PMC3980740 DOI: 10.3390/cells3010092] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 02/07/2014] [Accepted: 02/07/2014] [Indexed: 01/07/2023] Open
Abstract
One of the best examples of the renaissance of Src as an open door to cancer has been the demonstration that just five min of Src activation is sufficient for transformation and also for induction and maintenance of cancer stem cells [1]. Many tyrosine kinase receptors, through the binding of their ligands, become the keys that unlock the structure of Src and activate its oncogenic transduction pathways. Furthermore, intracellular isoforms of these receptors, devoid of any tyrosine kinase activity, still retain the ability to unlock Src. This has been shown with a truncated isoform of KIT (tr-KIT) and a truncated isoform of VEGFR-1 (i21-VEGFR-1), which are intracellular and require no ligand binding, but are nonetheless able to activate Src and induce cell migration and invasion of cancer cells. Expression of the i21-VEGFR-1 is upregulated by the Notch signaling pathway and repressed by miR-200c and retinoic acid in breast cancer cells. Both Notch inhibitors and retinoic acid have been proposed as potential therapies for invasive breast cancer.
Collapse
|
32
|
Lavainne F, Meffray E, Pepper RJ, Néel M, Delcroix C, Salama AD, Fakhouri F. Heparin use during dialysis sessions induces an increase in the antiangiogenic factor soluble Flt1. Nephrol Dial Transplant 2014; 29:1225-31. [DOI: 10.1093/ndt/gft517] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
33
|
Murdoch CE, Shuler M, Haeussler DJF, Kikuchi R, Bearelly P, Han J, Watanabe Y, Fuster JJ, Walsh K, Ho YS, Bachschmid MM, Cohen RA, Matsui R. Glutaredoxin-1 up-regulation induces soluble vascular endothelial growth factor receptor 1, attenuating post-ischemia limb revascularization. J Biol Chem 2014; 289:8633-44. [PMID: 24482236 DOI: 10.1074/jbc.m113.517219] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Glutaredoxin-1 (Glrx) is a cytosolic enzyme that regulates diverse cellular function by removal of GSH adducts from S-glutathionylated proteins including signaling molecules and transcription factors. Glrx is up-regulated during inflammation and diabetes, and Glrx overexpression inhibits VEGF-induced EC migration. The aim was to investigate the role of up-regulated Glrx in EC angiogenic capacities and in vivo revascularization in the setting of hind limb ischemia. Glrx-overexpressing EC from Glrx transgenic (TG) mice showed impaired migration and network formation and secreted higher levels of soluble VEGF receptor 1 (sFlt), an antagonizing factor to VEGF. After hind limb ischemia surgery Glrx TG mice demonstrated impaired blood flow recovery, associated with lower capillary density and poorer limb motor function compared with wild type littermates. There were also higher levels of anti-angiogenic sFlt expression in the muscle and plasma of Glrx TG mice after surgery. Noncanonical Wnt5a is known to induce sFlt. Wnt5a was highly expressed in ischemic muscles and EC from Glrx TG mice, and exogenous Wnt5a induced sFlt expression and inhibited network formation in human microvascular EC. Adenoviral Glrx-induced sFlt in EC was inhibited by a competitive Wnt5a inhibitor. Furthermore, Glrx overexpression removed GSH adducts on p65 in ischemic muscle and EC and enhanced NF-κB activity, which was responsible for Wnt5a-sFlt induction. Taken together, up-regulated Glrx induces sFlt in EC via NF-κB-dependent Wnt5a, resulting in attenuated revascularization in hind limb ischemia. The Glrx-induced sFlt explains part of the mechanism of redox-regulated VEGF signaling.
Collapse
|
34
|
Golde TE, Koo EH, Felsenstein KM, Osborne BA, Miele L. γ-Secretase inhibitors and modulators. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1828:2898-907. [PMID: 23791707 PMCID: PMC3857966 DOI: 10.1016/j.bbamem.2013.06.005] [Citation(s) in RCA: 215] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 06/04/2013] [Indexed: 12/11/2022]
Abstract
γ-Secretase is a fascinating, multi-subunit, intramembrane cleaving protease that is now being considered as a therapeutic target for a number of diseases. Potent, orally bioavailable γ-secretase inhibitors (GSIs) have been developed and tested in humans with Alzheimer's disease (AD) and cancer. Preclinical studies also suggest the therapeutic potential for GSIs in other disease conditions. However, due to inherent mechanism based-toxicity of non-selective inhibition of γ-secretase, clinical development of GSIs will require empirical testing with careful evaluation of benefit versus risk. In addition to GSIs, compounds referred to as γ-secretase modulators (GSMs) remain in development as AD therapeutics. GSMs do not inhibit γ-secretase, but modulate γ-secretase processivity and thereby shift the profile of the secreted amyloid β peptides (Aβ) peptides produced. Although GSMs are thought to have an inherently safe mechanism of action, their effects on substrates other than the amyloid β protein precursor (APP) have not been extensively investigated. Herein, we will review the current state of development of GSIs and GSMs and explore pertinent biological and pharmacological questions pertaining to the use of these agents for select indications. This article is part of a Special Issue entitled: Intramembrane Proteases.
Collapse
Affiliation(s)
- Todd E Golde
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | | | | | | | | |
Collapse
|
35
|
Increased Soluble Flt-1 Correlates With Delayed Graft Function and Early Loss of Peritubular Capillaries in the Kidney Graft. Transplantation 2013; 96:739-44. [DOI: 10.1097/tp.0b013e31829f4772] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
36
|
Abstract
To date, 18 distinct receptor tyrosine kinases (RTKs) are reported to be trafficked from the cell surface to the nucleus in response to ligand binding or heterologous agonist exposure. In most cases, an intracellular domain (ICD) fragment of the receptor is generated at the cell surface and translocated to the nucleus, whereas for a few others the intact receptor is translocated to the nucleus. ICD fragments are generated by several mechanisms, including proteolysis, internal translation initiation, and messenger RNA (mRNA) splicing. The most prevalent mechanism is intramembrane cleavage by γ-secretase. In some cases, more than one mechanism has been reported for the nuclear localization of a specific RTK. The generation and use of RTK ICD fragments to directly communicate with the nucleus and influence gene expression parallels the production of ICD fragments by a number of non-RTK cell-surface molecules that also influence cell proliferation. This review will be focused on the individual RTKs and to a lesser extent on other growth-related cell-surface transmembrane proteins.
Collapse
Affiliation(s)
- Graham Carpenter
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146
| | | |
Collapse
|
37
|
Raikwar NS, Liu KZ, Thomas CP. Protein kinase C regulates FLT1 abundance and stimulates its cleavage in vascular endothelial cells with the release of a soluble PlGF/VEGF antagonist. Exp Cell Res 2013; 319:2578-87. [PMID: 23911939 DOI: 10.1016/j.yexcr.2013.07.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 06/25/2013] [Accepted: 07/09/2013] [Indexed: 01/18/2023]
Abstract
FLT1 and its soluble form (sFLT1) arise as alternate transcripts from the same gene and sFLT1 can antagonize the effect of vascular endothelial growth factor (VEGF) on its cognate receptors. We investigated the effect of VEGF and protein kinase C (PKC) activation on sFLT1 abundance. We demonstrated that VEGF stimulates sFLT1 and FLT1 mRNA and protein levels in vascular endothelial cells via VEGFR2 and PKC. Using an FLT1 expression vector with N and C-terminal epitope tags, we show that PKC activation increases the cleavage of FLT1 into an N-terminal extracellular fragment and a C-terminal intracellular fragment with the cleavage occurring adjacent to the transmembrane domain. The trafficking and glycosylation inhibitors brefeldin, monensin and tunicamycin substantially reduced cleavage and release of the N-terminal ectodomain of FLT1 and inhibited secretion of the isoforms of sFLT1. The shed FLT1 ectodomain can bind VEGF and PlGF and inhibit VEGF-induced vascular tube formation thus confirming that it is functionally equivalent to the alternately spliced and secreted sFLT1 isoforms.
Collapse
Affiliation(s)
- Nandita S Raikwar
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, IA, USA
| | | | | |
Collapse
|
38
|
Martin A, Tisch RM, Getts DR. Manipulating T cell-mediated pathology: Targets and functions of monoclonal antibody immunotherapy. Clin Immunol 2013; 148:136-47. [DOI: 10.1016/j.clim.2013.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 04/17/2013] [Accepted: 04/18/2013] [Indexed: 12/16/2022]
|
39
|
Tang YT, Jiang F, Guo L, Si MY, Jiao XY. The soluble VEGF receptor 1 and 2 expression in cerebral spinal fluid as an indicator for leukemia central nervous system metastasis. J Neurooncol 2013; 112:329-38. [PMID: 23400753 DOI: 10.1007/s11060-013-1066-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 02/04/2013] [Indexed: 02/05/2023]
Abstract
Over-expression of vascular endothelial growth factor A (VEGF-A) is correlated with leukemia metastasis. VEGF-A acts by binding to its membrane receptors R1 and R2 present in soluble forms (sVEGFR1, sVEGFR2) with different functions. sVEGFR could inhibit VEGF-A bioactivities, associated with favorable prognosis in solid tumors. However, its role is obscure in central nervous system leukemia (CNSL). The aim of this study was to investigate sVEGFR1, R2 as biomarkers in CNSL. Paired cerebrospinal fluid (CSF) and serum samples were collected from 35 leukemia cases with or without CNS metastasis. Levels of sVEGFR1 and sVEGFR2 in both CSF (sVEGFR1CSF, sVEGFR2CSF) and serum (sVEGFR1Serum, sVEGFR2Serum) were detected by ELISA. Other risk factors related to CNSL prognosis were also analyzed. sVEGFRSerum levels were 2.54-fold (sVEGFR1) and 25.6-fold (sVEGFR2) higher than sVEGFRCSF in both leukemic groups. sVEGFR1CSF in CNSL were 33 % higher than in the non-CNSL, and the levels of sVEGFR2CSF and sVEGFR2Serum had the same trend. Elevated sVEGFR1CSF and sVEGFR2CSF is closely correlated with blood-brain barrier (BBB) values and WBCCSF that is an indicator of CNSL disease burden. Cox regression analysis showed that the sVEGFR2CSF had a positive effect on event-free survival. Our data suggest that sVEGFR2CSF may be more potent than sVEGFR1CSF in predicting the outcome of leukemia patients, the balance between sVEGFR2CSF and VEGF-ACSF levels might be crucial for the progression of CNSL.
Collapse
Affiliation(s)
- Yue-Ting Tang
- Department of Hematology Laboratory, First Affiliated Hospital of Shantou University Medical College, 57 Changping Road, Shantou 515041, Guangdong, China
| | | | | | | | | |
Collapse
|
40
|
Cho YK, Zhang X, Uehara H, Young JR, Archer B, Ambati B. Vascular Endothelial Growth Factor Receptor 1 morpholino increases graft survival in a murine penetrating keratoplasty model. Invest Ophthalmol Vis Sci 2012; 53:8458-71. [PMID: 23150613 DOI: 10.1167/iovs.12-10408] [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
PURPOSE This study sought to determine whether a Vascular Endothelial Growth Factor Receptor 1 (VEGFR1)-specific morpholino (MO) could decrease neovascularization, thereby enhancing murine cornea transplant survival, and if this effect is synergistic with steroid therapy. METHODS Graft survival, corneal neovascularization, and corneal lymphangiogenesis were compared among the VEGFR1_MO, STD MO and PBS groups following subconjunctival injection in mice that underwent normal risk penetrating keratoplasty (NR PK) and high-risk penetrating keratoplasty (HR PK). Graft survival, corneal neovascularization, and corneal lymphangiogenesis in groups treated with both VEGFR1_MO and steroid therapy were also analyzed in HR PK. RESULTS In NR PK, the VEGFR1_MO decreased angiogenesis, lymphangiogenesis, and increased graft survival compared with the PBS group (P = 0.055, P = 0.003, P = 0.043, respectively). In HR PK, VEGFR1_MO decreased angiogenesis, lymphangiogenesis, and increased graft survival compared with the STD MO (P = 0.000, P = 0.000, P = 0.029, respectively) and PBS groups (P = 0.004, P = 0.002, P = 0.024). In HR PK, when the VEGFR1_MO was combined with steroid therapy, a significant increase in graft survival was seen compared with steroid treatment alone (P = 0.045). The 2-month graft survival rate for HR PK was 27% in the combination group compared with 0% in the triamcinolone only group. CONCLUSIONS VEGFR1_MO decreased angiogenesis and lymphangiogenesis, resulting in increased graft survival in both NR PK and HR PK. This beneficial effect is synergistically enhanced with steroid treatment in HR PK.
Collapse
Affiliation(s)
- Yang Kyung Cho
- Department of Ophthalmology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | | | | | | | | | | |
Collapse
|
41
|
Cerdeira AS, Karumanchi SA. Angiogenic factors in preeclampsia and related disorders. Cold Spring Harb Perspect Med 2012; 2:cshperspect.a006585. [PMID: 23125198 DOI: 10.1101/cshperspect.a006585] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During fetal development, the human placenta undergoes high levels of both angiogenesis and vasculogenesis. Additionally, the developing placenta undergoes a process of vascular mimicry (referred to as pseudovasculogenesis) as cytotrophoblasts convert from an epithelial to an endothelial phenotype. The initiation, maturation, and maintenance of the placental vasculature are of critical importance. Failure to do so can lead to adverse obstetric outcomes such as preeclampsia and/or intrauterine growth restriction (IUGR). Furthermore, the foundation of many aspects of adult health is laid in utero. In this context, normal placental function is not only critical for normal fetal development but can also permanently influence long-term health and disease. Understanding the mechanisms that regulate placental vasculogenesis and angiogenesis is therefore of critical importance. This chapter will focus on placental vascular development with a particular emphasis on the role of angiogenic factors in the pathogenesis of the maternal syndrome of preeclampsia and related disorders.
Collapse
Affiliation(s)
- Ana Sofia Cerdeira
- Department of Medicine, Obstetrics & Gynecology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02114, USA
| | | |
Collapse
|
42
|
Carpenter G, Pozzi A. Cell responses to growth factors: the role of receptor tyrosine kinase intracellular domain fragments. Sci Signal 2012; 5:pe42. [PMID: 23012654 DOI: 10.1126/scisignal.2003526] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Growth factor activation of receptor tyrosine kinases (RTKs) provokes well-described canonical second messenger pathways that transmit biochemical signals in the cytoplasm and to the nucleus to initiate cellular responses. The proteolytic liberation of intracellular domain fragments (ICDs) from activated RTKs and the nuclear translocation of these ICDs represent a more recently identified and noncanonical mechanism by which RTKs communicate with the nucleus. Several reports have added previously unknown facets to the ICD mechanism and have enlarged the scope of ICDs as second messengers.
Collapse
Affiliation(s)
- Graham Carpenter
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA.
| | | |
Collapse
|
43
|
Cai J, Qi X, Kociok N, Skosyrski S, Emilio A, Ruan Q, Han S, Liu L, Chen Z, Bowes Rickman C, Golde T, Grant MB, Saftig P, Serneels L, de Strooper B, Joussen AM, Boulton ME. β-Secretase (BACE1) inhibition causes retinal pathology by vascular dysregulation and accumulation of age pigment. EMBO Mol Med 2012; 4:980-91. [PMID: 22903875 PMCID: PMC3491829 DOI: 10.1002/emmm.201101084] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 06/23/2012] [Accepted: 06/28/2012] [Indexed: 01/18/2023] Open
Abstract
β-Secretase (BACE1) is a major drug target for combating Alzheimer's disease (AD). Here we show that BACE1(-/-) mice develop significant retinal pathology including retinal thinning, apoptosis, reduced retinal vascular density and an increase in the age pigment, lipofuscin. BACE1 expression is highest in the neural retina while BACE2 was greatest in the retinal pigment epithelium (RPE)/choroid. Pigment epithelial-derived factor, a known regulator of γ-secretase, inhibits vascular endothelial growth factor (VEGF)-induced in vitro and in vivo angiogenesis and this is abolished by BACE1 inhibition. Moreover, intravitreal administration of BACE1 inhibitor or BACE1 small interfering RNA (siRNA) increases choroidal neovascularization in mice. BACE1 induces ectodomain shedding of vascular endothelial growth factor receptor 1 (VEGFR1) which is a prerequisite for γ-secretase release of a 100 kDa intracellular domain. The increase in lipofuscin following BACE1 inhibition and RNAI knockdown is associated with lysosomal perturbations. Taken together, our data show that BACE1 plays a critical role in retinal homeostasis and that the use of BACE inhibitors for AD should be viewed with extreme caution as they could lead to retinal pathology and exacerbate conditions such as age-related macular degeneration.
Collapse
Affiliation(s)
- Jun Cai
- Department of Anatomy & Cell Biology, University of Florida, Gainesville, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Soluble fms-like tyrosine kinase 1 and soluble endoglin are elevated circulating anti-angiogenic factors in pre-eclampsia. Pregnancy Hypertens 2012; 2:358-67. [PMID: 26105603 DOI: 10.1016/j.preghy.2012.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 06/24/2012] [Indexed: 01/05/2023]
Abstract
Pre-eclampsia, characterized by hypertension and proteinuria, affects approximately 3-5% of all pregnancies worldwide and is a major cause of maternal and fetal morbidity and mortality. Maternal endothelial dysfunction is associated with disease pathogenesis. Recently, reports have shown that elevated levels of circulating soluble fms-like tyrosine kinase 1 [sFlt1] and soluble endoglin [sEng] are associated with pre-eclampsia. Flt1 is a receptor for vascular endothelial growth factor receptor [VEGF], whereas endoglin [Eng] is an auxiliary receptor for transforming growth factor-β [TGF-β] super-family members. Both signaling pathways modulate angiogenesis and are involved in vascular homeostasis. Increased levels of sFlt1 and sEng dysregulate VEGF and TGF-β signaling respectively, resulting in endothelial dysfunction of maternal blood vessels. This review summarizes our current knowledge of Flt1 and endoglin and soluble forms in pre-eclampsia. Furthermore, it highlights the predictive and early-screening value of circulating levels of sFlt1 and sEng for the risk of developing pre-eclampsia.
Collapse
|
45
|
Na HW, Shin WS, Ludwig A, Lee ST. The cytosolic domain of protein-tyrosine kinase 7 (PTK7), generated from sequential cleavage by a disintegrin and metalloprotease 17 (ADAM17) and γ-secretase, enhances cell proliferation and migration in colon cancer cells. J Biol Chem 2012; 287:25001-9. [PMID: 22665490 DOI: 10.1074/jbc.m112.348904] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein-tyrosine kinase 7 (PTK7) is a member of the defective receptor protein-tyrosine kinases and is known to function as a regulator of planar cell polarity during development. Its expression is up-regulated in some cancers including colon carcinomas. A 100-kDa fragment of PTK7 was detected in the culture media from colon cancer cells and HEK293 cells. The shed fragment was named sPTK7-Ig1-7 because its molecular mass was very similar to that of the entire extracellular domain of PTK7 that contains immunoglobulin-like loops 1 to 7 (Ig1-7). The shedding of sPTK7-Ig1-7 was enhanced by treatment with phorbol 12-myristate 13-acetate. In addition to the sPTK7-Ig1-7 found in the culture medium, two C-terminal fragments of PTK7 were detected in the cell lysates: PTK7-CTF1, which includes a transmembrane segment and a cytoplasmic domain, and PTK7-CTF2, which lacks most of the transmembrane segment from PTK7-CTF1. Analysis of PTK7 processing in the presence of various protease inhibitors or after knockdown of potential proteases suggests that shedding of PTK7 into sPTK7-Ig1-7 and PTK7-CTF1 is catalyzed by ADAM17, and further cleavage of PTK7-CTF1 into PTK7-CTF2 is mediated by the γ-secretase complex. PTK7-CTF2 localizes to the nucleus and enhances proliferation, migration, and anchorage-independent colony formation. Our findings demonstrate a novel role for PTK7 in the tumorigenesis via generation of PTK7-CTF2 by sequential cleavage of ADAM17 and γ-secretase.
Collapse
Affiliation(s)
- Hye-Won Na
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | | | | | | |
Collapse
|
46
|
Cho YK, Uehara H, Young JR, Archer B, Zhang X, Ambati BK. Vascular endothelial growth factor receptor 1 morpholino decreases angiogenesis in a murine corneal suture model. Invest Ophthalmol Vis Sci 2012; 53:685-92. [PMID: 22199251 DOI: 10.1167/iovs.11-8391] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE This study sought to determine whether a vascular endothelial growth factor receptor 1 (VEGFR1)-specific morpholino could induce the alternative splicing of Flt-1 pre-mRNA to downregulate membrane-bound Flt-1 (mFlt-1) and increase the production of soluble Flt-1 (sFlt-1), thereby limiting angiogenesis and inflammation in a mouse corneal suture injury model. METHODS A murine corneal suture model was used to investigate the effects of a VEGFR1-specific morpholino in vivo. Western blot analysis and RT-PCR were used to compare the impact of the Flt morpholino on mFlt-1 and sFlt-1 levels. For vascular regression modeling, two corneal sutures were placed and injected with Flt morpholino, standard morpholino, and PBS on days 8 and 10. Corneas were harvested on day 14. The grade of neovascularization (graded 0-5; 0, no neovascularization; 5, thick tortuous new vessel growth over the suture and toward the center of the cornea) was compared on days 8, 10, and 14. Immunohistochemistry, fluorescent microscopy, and confocal microscopy were used to digitally quantify the area and volume of neovascularization and inflammatory infiltration. RESULTS Western blot analysis revealed that the Flt morpholino decreased mFlt-1 levels while increasing sFlt-1 levels. An increased sFlt-1/mFlt-1 ratio in the Flt morpholino group was seen with RT-PCR. Based on the neovascularization grading, there was a decrease in neovascularization area in the Flt morpholino group (3.29 ± 0.19 to 2.92 ± 0.13) from day 8 to 14 (P < 0.05) compared with that in both the standard morpholino (2.68 ± 0.19 to 3.14 ± 0.22) and in the PBS (2.96 ± 0.14 to 3.42 ± 0.19) groups, both of which showed an increase in neovascularization (P < 0.05). The Flt morpholino group also showed reduced neovascularization volume compared with that of the PBS (P = 0.001) and STD morpholino groups (P = 0.000). CONCLUSIONS Flt morpholinos decrease mFlt-1 and increase sFlt-1 levels, resulting in decreased neovascularization in a murine corneal suture model.
Collapse
Affiliation(s)
- Yang Kyung Cho
- Department of Ophthalmology, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
| | | | | | | | | | | |
Collapse
|
47
|
Raghu H, Nalla AK, Gondi CS, Gujrati M, Dinh DH, Rao JS. uPA and uPAR shRNA inhibit angiogenesis via enhanced secretion of SVEGFR1 independent of GM-CSF but dependent on TIMP-1 in endothelial and glioblastoma cells. Mol Oncol 2011; 6:33-47. [PMID: 22177802 DOI: 10.1016/j.molonc.2011.11.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 11/22/2011] [Accepted: 11/23/2011] [Indexed: 12/31/2022] Open
Abstract
The uPA/uPAR system is known to play a critical role in angiogenesis of glioblastoma. Previously, we have shown that shRNA against uPA and uPAR attenuates angiogenesis by blocking nuclear translocation of angiogenin, inhibition of angiopoietin/Tie2 signaling, and regulating several other pro-angiogenic, angiostatic and anti-angiogenic molecules. Further analysis revealed that GM-CSF, a pleiotropic cytokine, was significantly inhibited in U87MG and 4910 co-cultures with endothelial cells transfected with shRNA against uPA and uPAR. The role of the uPA/uPAR system in this process is not completely understood. Analysis of tumor conditioned medium of U87MG, 4910 and HMECs transfected with shRNA against uPA or uPAR alone or in combination (pU2) revealed inhibition of GM-CSF-enhanced secretion of SVEGFR1 as shown by Western blotting and ELISA. Moreover, phosphorylation of JAK2 and STAT5, the downstream effectors of GM-CSF signaling, was also inhibited in all three cell lines. Phosphorylation at Tyr 166 position of the GM-CSFRβ subunit, the signal activating subunit of the GM-CSF receptor, was inhibited in HMEC, U87MG and 4910 cells. Further analysis revealed that shRNA against uPA and/or uPAR increased secretion of TIMP-1, which is known to enhance SVEGFR1 secretion in endothelial cells. Moreover, addition of purified uPA (with and without GM-CSF) activated JAK2/STAT5 signaling in HMEC. Exogenous addition of SVEGFR1 to pU2 tumor conditioned medium enhanced inhibition of VEGF-induced endothelial capillary tube formation as assessed by an in vitro angiogenesis assay. To determine the significance of these events in vivo, nude mice with pre-established tumors treated with shRNA against uPA and/or uPAR showed decreased levels of GM-CSF and increased levels of SVEGFR1 and TIMP-1 when compared with controls. Enhanced secretion of SVEGFR1 by puPA, puPAR and pU2 in endothelial and GBM cells was mediated indirectly by MMP-7 and augmented by ectodomain shedding of VEGFr1 by tyrosine phosphorylation at the 1213 position. Taken together, these results suggest that the uPA/uPAR system could prove beneficial as an indirect target for inhibition of angiogenesis in glioblastoma.
Collapse
Affiliation(s)
- Hari Raghu
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL 61605, USA
| | | | | | | | | | | |
Collapse
|
48
|
Rosenberg VA, Buhimschi IA, Lockwood CJ, Paidas MJ, Dulay AT, Ramma W, Abdel-Razeq SS, Zhao G, Ahmad S, Ahmed A, Buhimschi CS. Heparin elevates circulating soluble fms-like tyrosine kinase-1 immunoreactivity in pregnant women receiving anticoagulation therapy. Circulation 2011; 124:2543-53. [PMID: 22082677 DOI: 10.1161/circulationaha.111.046821] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Alterations in circulating levels of pro- and antiangiogenic factors have been associated with adverse pregnancy outcomes. Heparin is routinely administered to pregnant women, but without clear knowledge of its impact on these factors. METHODS AND RESULTS We conducted a longitudinal study of 42 pregnant women. Twenty-one women received prophylactic heparin anticoagulation, and 21 healthy pregnant women served as controls. Compared with gestational age-matched controls, heparin treatment was associated with increased circulating levels of soluble fms-like tyrosine kinase-1 (sFlt-1) in the third trimester (P<0.05), in the absence of preeclampsia, placental abruption, or fetal growth restriction. Heparin had no effect on circulating levels of vascular endothelial growth factor, placenta growth factor, or soluble endoglin as assessed by ELISA. In vitro, low-molecular weight and unfractionated heparins stimulated sFlt-1 release from placental villous explants, in a dose- and time-dependent manner. This effect was not due to placental apoptosis, necrosis, alteration in protein secretion, or increased transcription. Western blot analysis demonstrated that heparin induced shedding of the N-terminus of Flt-1 both in vivo and in vitro as indicated by a predominant band of 100-112 kDa. By using an in vitro angiogenesis assay, we demonstrated that serum of heparin-treated cases inhibited both basal and vascular endothelial growth factor-induced capillary-like tube formation. CONCLUSIONS Heparin likely increases the maternal sFlt-1 through shedding of the extracellular domain of Flt-1 receptor. Our results imply that upregulation of circulating sFlt-1 immunoreactivity in pregnancy is not always associated with adverse outcomes, and that heparin's protective effects, if any, cannot be explained by promotion of angiogenesis.
Collapse
Affiliation(s)
- Victor A Rosenberg
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University, 333 Cedar Street, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Le Roux S, Pepper RJ, Dufay A, Néel M, Meffray E, Lamandé N, Rimbert M, Josien R, Hamidou M, Hourmant M, Cook HT, Charreau B, Larger E, Salama AD, Fakhouri F. Elevated soluble Flt1 inhibits endothelial repair in PR3-ANCA-associated vasculitis. J Am Soc Nephrol 2011; 23:155-64. [PMID: 22034638 DOI: 10.1681/asn.2010080858] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis exhibits endothelial damage, but the capacity for vessel repair in this disorder is not well understood. Here, we observed a marked increase in serum levels of soluble Flt1 (sFlt1), a potent inhibitor of vascular endothelial growth factor, in patients with active ANCA-associated vasculitis compared with patients during remission and other controls. Serum levels of sFlt1 correlated with C5a, an anaphylatoxin released after complement activation. Serum from patients with acute ANCA-associated vasculitis disrupted blood flow in the chicken chorioallantoic membrane assay, suggesting an antiangiogenic effect. Preincubation with excess human vascular endothelial growth factor prevented this effect. Anti-proteinase-3 (PR3) mAb and serum containing PR3-ANCA from patients with active vasculitis both induced a significant and sustained release of sFlt1 from monocytes, whereas anti-myeloperoxidase (MPO) mAb or polyclonal antibodies did not. However, the serum containing polyclonal PR3-ANCA did not induce release of sFlt1 from cultured human umbilical vein endothelial cells. In summary, these data suggest that anti-PR3 antibodies, and to a much lesser extent anti-MPO antibodies, increase sFlt1 during acute ANCA-associated vasculitis, leading to an antiangiogenic state that hinders endothelial repair.
Collapse
Affiliation(s)
- Sandrine Le Roux
- ITUN, U643, Department of Nephrology and Immunology, CHU de Nantes, 44000 Nantes, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bruce D, Tan PH. Vascular endothelial growth factor receptors and the therapeutic targeting of angiogenesis in cancer: where do we go from here? ACTA ACUST UNITED AC 2011; 18:85-103. [PMID: 22017472 DOI: 10.3109/15419061.2011.619673] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Abstract Vascular Endothelial Growth Factor receptors (VEGFRs), the interactions with their ligands and the subsequent signalling pathways are known to play a vital role in tumour angiogenesis. Initial clinical trials of VEGFR inhibitors were disappointing but over the past decade some therapies have been successfully brought to market. At present, VEGFR inhibitors appear to be most promising as adjuvants to conventional chemotherapy. However, several interacting signalling molecules and downstream pathways have recently been shown to interact with VEGFR signalling and provide promising novel targets, such as the platelet-derived growth factor (PDGF), epithelial growth factor (EGF), human epithelial receptor-2, (HER-2) Tie-2 and oestrogen receptors. Elucidation of this web of signalling pathways may identify new therapeutic strategies which may be used in combination with VEGFR inhibitors to augment the efficacy of anti-angiogenic cancer treatments. This review assesses the role of modulating VEGFR activity in cancer and systematically examines current evidence and trials in this area.
Collapse
Affiliation(s)
- David Bruce
- Nuffield Department of Surgical Science, Oxford University, The John Radcliffe, Headley Way, Oxford, UK
| | | |
Collapse
|