1
|
Peng Q, Shan D, Cui K, Li K, Zhu B, Wu H, Wang B, Wong S, Norton V, Dong Y, Lu YW, Zhou C, Chen H. The Role of Endothelial-to-Mesenchymal Transition in Cardiovascular Disease. Cells 2022; 11:1834. [PMID: 35681530 PMCID: PMC9180466 DOI: 10.3390/cells11111834] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 02/07/2023] Open
Abstract
Endothelial-to-mesenchymal transition (EndoMT) is the process of endothelial cells progressively losing endothelial-specific markers and gaining mesenchymal phenotypes. In the normal physiological condition, EndoMT plays a fundamental role in forming the cardiac valves of the developing heart. However, EndoMT contributes to the development of various cardiovascular diseases (CVD), such as atherosclerosis, valve diseases, fibrosis, and pulmonary arterial hypertension (PAH). Therefore, a deeper understanding of the cellular and molecular mechanisms underlying EndoMT in CVD should provide urgently needed insights into reversing this condition. This review summarizes a 30-year span of relevant literature, delineating the EndoMT process in particular, key signaling pathways, and the underlying regulatory networks involved in CVD.
Collapse
Affiliation(s)
- Qianman Peng
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Q.P.); (D.S.); (K.C.); (K.L.); (B.Z.); (H.W.); (B.W.); (S.W.); (V.N.); (Y.D.); (Y.W.L.)
| | - Dan Shan
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Q.P.); (D.S.); (K.C.); (K.L.); (B.Z.); (H.W.); (B.W.); (S.W.); (V.N.); (Y.D.); (Y.W.L.)
| | - Kui Cui
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Q.P.); (D.S.); (K.C.); (K.L.); (B.Z.); (H.W.); (B.W.); (S.W.); (V.N.); (Y.D.); (Y.W.L.)
| | - Kathryn Li
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Q.P.); (D.S.); (K.C.); (K.L.); (B.Z.); (H.W.); (B.W.); (S.W.); (V.N.); (Y.D.); (Y.W.L.)
| | - Bo Zhu
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Q.P.); (D.S.); (K.C.); (K.L.); (B.Z.); (H.W.); (B.W.); (S.W.); (V.N.); (Y.D.); (Y.W.L.)
| | - Hao Wu
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Q.P.); (D.S.); (K.C.); (K.L.); (B.Z.); (H.W.); (B.W.); (S.W.); (V.N.); (Y.D.); (Y.W.L.)
| | - Beibei Wang
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Q.P.); (D.S.); (K.C.); (K.L.); (B.Z.); (H.W.); (B.W.); (S.W.); (V.N.); (Y.D.); (Y.W.L.)
| | - Scott Wong
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Q.P.); (D.S.); (K.C.); (K.L.); (B.Z.); (H.W.); (B.W.); (S.W.); (V.N.); (Y.D.); (Y.W.L.)
| | - Vikram Norton
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Q.P.); (D.S.); (K.C.); (K.L.); (B.Z.); (H.W.); (B.W.); (S.W.); (V.N.); (Y.D.); (Y.W.L.)
| | - Yunzhou Dong
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Q.P.); (D.S.); (K.C.); (K.L.); (B.Z.); (H.W.); (B.W.); (S.W.); (V.N.); (Y.D.); (Y.W.L.)
| | - Yao Wei Lu
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Q.P.); (D.S.); (K.C.); (K.L.); (B.Z.); (H.W.); (B.W.); (S.W.); (V.N.); (Y.D.); (Y.W.L.)
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA;
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Q.P.); (D.S.); (K.C.); (K.L.); (B.Z.); (H.W.); (B.W.); (S.W.); (V.N.); (Y.D.); (Y.W.L.)
| |
Collapse
|
2
|
Armant DR, Aberdeen GW, Kilburn BA, Pepe GJ, Albrecht ED. Baboon placental heparin-binding epidermal growth factor-like growth factor. Reproduction 2021; 160:31-37. [PMID: 32272450 DOI: 10.1530/rep-19-0487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 04/09/2020] [Indexed: 11/08/2022]
Abstract
Placental extravillous trophoblast remodeling of the uterine spiral arteries is important for promoting blood flow to the placenta and fetal development. Heparin-binding EGF-like growth factor (HB-EGF), an EGF family member, stimulates differentiation and invasive capacity of extravillous trophoblasts in vitro. Trophoblast expression and maternal levels of HB-EGF are reduced at term in women with preeclampsia, but it is uncertain whether HB-EGF is downregulated earlier when it may contribute to placental insufficiency. A nonhuman primate model has been established in which trophoblast remodeling of the uterine spiral arteries is suppressed by shifting the rise in estrogen from the second to the first trimester of baboon pregnancy. In the present study, we used this model to determine if placental HB-EGF is altered by prematurely elevating estrogen early in baboon gestation. Uterine spiral artery remodeling and placental expression of HB-EGF and other EGF family members were assessed on day 60 of gestation in baboons treated with estradiol (E2) daily between days 25 and 59 of gestation (term = 184 days). The percentages of spiral artery remodeling were 90, 84 and 70% lower (P < 0.01), respectively, for vessels of 26-50, 51-100 and >100 µm diameter in E2-treated compared with untreated baboons. HB-EGF protein quantified by immunocytochemical staining/image analysis was decreased three-fold (P < 0.01) in the placenta of E2-treated versus untreated baboons, while amphiregulin (AREG) and EGF expression was unaltered. Therefore, we propose that HB-EGF modulates the estrogen-sensitive remodeling of the uterine spiral arteries by the extravillous trophoblast in early baboon pregnancy.
Collapse
Affiliation(s)
- D Randall Armant
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Graham W Aberdeen
- Departments of Obstetrics, Gynecology and Reproductive Sciences and Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Brian A Kilburn
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Gerald J Pepe
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Eugene D Albrecht
- Departments of Obstetrics, Gynecology and Reproductive Sciences and Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Inoue Y, Shimazawa M, Nakamura S, Takata S, Hashimoto Y, Izawa H, Masuda T, Tsuruma K, Sakaue T, Nakayama H, Higashiyama S, Hara H. Both Autocrine Signaling and Paracrine Signaling of HB-EGF Enhance Ocular Neovascularization. Arterioscler Thromb Vasc Biol 2017; 38:174-185. [PMID: 29191924 DOI: 10.1161/atvbaha.117.310337] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 11/17/2017] [Indexed: 12/27/2022]
Abstract
OBJECTIVE The incidence of blindness is increasing because of the increase in abnormal ocular neovascularization. Anti-VEGF (vascular endothelial growth factor) therapies have led to good results, although they are not a cure for the blindness. The purpose of this study was to determine what role HB-EGF (heparin-binding epidermal growth factor-like growth factor) plays in ocular angiogenesis. APPROACH AND RESULTS We examined the role played by HB-EGF in ocular neovascularization in 2 animal models of neovascularization: laser-induced choroidal neovascularization (CNV) and oxygen-induced retinopathy. We also studied human retinal microvascular endothelial cells in culture. Our results showed that the neovascularization was decreased in both the CNV and oxygen-induced retinopathy models in HB-EGF conditional knockout mice compared with that in wild-type mice. Moreover, the expressions of HB-EGF and VEGF were increased after laser-induced CNV and oxygen-induced retinopathy, and their expression sites were located around the neovascular areas. Exposure of human retinal microvascular endothelial cells to HB-EGF and VEGF increased their proliferation and migration, and CRM-197 (cross-reactive material-197), an HB-EGF inhibitor, decreased the HB-EGF-induced and VEGF-induced cell proliferation and migration. VEGF increased the expression of HB-EGF mRNA. VEGF-dependent activation of EGFR (epidermal growth factor receptor)/ERK1/2 (extracellular signal-regulated kinase 1/2) signaling and cell proliferation of endothelial cells required stimulation of the ADAM17 (a disintegrin and metalloprotease) and ADAM12. CRM-197 decreased the grades of the fluorescein angiograms and size of the CNV areas in marmoset monkeys. CONCLUSIONS These findings suggest that HB-EGF plays an important role in the development of CNV. Therefore, further investigations of HB-EGF are needed as a potential therapeutic target in the treatment of exudative age-related macular degeneration.
Collapse
Affiliation(s)
- Yuki Inoue
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Masamitsu Shimazawa
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Shinsuke Nakamura
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Shinsuke Takata
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Yuhei Hashimoto
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Hiroshi Izawa
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Tomomi Masuda
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Kazuhiro Tsuruma
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Tomohisa Sakaue
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Hironao Nakayama
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Shigeki Higashiyama
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Hideaki Hara
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.).
| |
Collapse
|
4
|
Faiotto VB, Franci D, Enz Hubert RM, de Souza GR, Fiusa MML, Hounkpe BW, Santos TM, Carvalho-Filho MA, De Paula EV. Circulating levels of the angiogenesis mediators endoglin, HB-EGF, BMP-9 and FGF-2 in patients with severe sepsis and septic shock. J Crit Care 2017; 42:162-167. [PMID: 28746898 DOI: 10.1016/j.jcrc.2017.07.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 06/18/2017] [Accepted: 07/16/2017] [Indexed: 02/06/2023]
Abstract
PURPOSE Endothelial barrier dysfunction is a hallmark of sepsis, and is at least partially mediated by pathways that regulate endothelial barrier assembly during angiogenesis. Not surprisingly, increased levels of key angiogenic proteins such as VEGF-A and Angiopoietin-2 have been described in sepsis. The purpose of this study was to investigate if additional pathways that regulate endothelial barrier integrity during angiogenesis could also be involved in the host response of sepsis. MATERIAL AND METHODS We evaluated circulating levels of four proteins involved in angiogenesis, not previously studied in sepsis, in a cohort of 50 patients with severe sepsis and septic shock. RESULTS Circulating levels of BMP-9 and FGF-2 were similar in patients and healthy volunteers. In contrast, patients with septic shock presented 1.5-fold higher levels of endoglin (P=0.004), and 2-fold lower levels of Heparin-Binding EGF-like growth factor (HB-EGF) (P=0.002) when compared to healthy individuals. Of note, HB-EGF deficiency has been recently demonstrated to be detrimental to survival in a murine model of sepsis. CONCLUSIONS Endoglin and HB-EGF could be involved in the host response of sepsis. Additional studies are warrant to investigate their role as biomarker or therapeutic targets in sepsis.
Collapse
Affiliation(s)
| | - Daniel Franci
- School of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | | | | | | | | | | | | | - Erich Vinicius De Paula
- School of Medical Sciences, University of Campinas, Campinas, SP, Brazil; Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil.
| |
Collapse
|
5
|
Shim JW, Sandlund J, Hameed MQ, Blazer-Yost B, Zhou FC, Klagsbrun M, Madsen JR. Excess HB-EGF, which promotes VEGF signaling, leads to hydrocephalus. Sci Rep 2016; 6:26794. [PMID: 27243144 PMCID: PMC4886677 DOI: 10.1038/srep26794] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/10/2016] [Indexed: 01/19/2023] Open
Abstract
Heparin binding epidermal growth factor-like growth factor (HB-EGF) is an angiogenic factor mediating radial migration of the developing forebrain, while vascular endothelial growth factor (VEGF) is known to influence rostral migratory stream in rodents. Cell migratory defects have been identified in animal models of hydrocephalus; however, the relationship between HB-EGF and hydrocephalus is unclear. We show that mice overexpressing human HB-EGF with β-galactosidase reporter exhibit an elevated VEGF, localization of β-galactosidase outside the subventricular zone (SVZ), subarachnoid hemorrhage, and ventriculomegaly. In Wistar polycystic kidney rats with hydrocephalus, alteration of migratory trajectory is detected. Furthermore, VEGF infusions into the rats result in ventriculomegaly with an increase of SVZ neuroblast in rostral migratory stream, whereas VEGF ligand inhibition prevents it. Our results support the idea that excess HB-EGF leads to a significant elevation of VEGF and ventricular dilatation. These data suggest a potential pathophysiological mechanism that elevated HB-EGF can elicit VEGF induction and hydrocephalus.
Collapse
Affiliation(s)
- Joon W Shim
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.,Department of Biology, Indiana University Purdue University, Indianapolis, IN 46202, USA.,Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Johanna Sandlund
- Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.,Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive L235, Stanford, CA 94305, USA.,Clinical Microbiology Laboratory, Stanford University Medical Center, 3375 Hillview Avenue Palo, Alto, CA 94304, USA
| | - Mustafa Q Hameed
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.,Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Bonnie Blazer-Yost
- Department of Biology, Indiana University Purdue University, Indianapolis, IN 46202, USA
| | - Feng C Zhou
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Michael Klagsbrun
- Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.,Department of Surgery and Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Joseph R Madsen
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Zeng F, Kloepfer LA, Finney C, Diedrich A, Harris RC. Specific endothelial heparin-binding EGF-like growth factor deletion ameliorates renal injury induced by chronic angiotensin II infusion. Am J Physiol Renal Physiol 2016; 311:F695-F707. [PMID: 27226110 DOI: 10.1152/ajprenal.00377.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 05/18/2016] [Indexed: 12/28/2022] Open
Abstract
Transactivation of EGF receptor (EGFR) by angiotensin II (Ang II) plays important roles in the initiation and progression of chronic kidney diseases. Studies suggest that heparin-binding EGF-like factor (HB-EGF) may be a critical mediator in this process, but its role in vivo has not been investigated. In the current study, we found that in response to Ang II infusion, kidneys from endothelial HB-EGF deletion mice had significantly reduced EGFR activation compared with controls. Meanwhile, deletion of endothelial HB-EGF expression decreased Ang II infusion related renal injury, as demonstrated by 1) less albuminuria; 2) less glomerulosclerosis; 3) preserved endothelial integrity and decreased podocyte injury, as shown by greater glomerular tuft area and WT1-positive cells, and fewer apoptotic cells measured by cleaved caspase 3 staining; 4) reduced inflammation in the perivascular area and interstitium measured by F4/80 and CD3 immunostaining; and 5) reduced renal fibrosis. In conclusion, our results suggest that shedding of HB-EGF from endothelium plays an important role in Ang II-induced renal injury by linking Ang II-AT1R with EGFR transactivation. Inhibition of HB-EGF shedding could be a potential therapeutic strategy for chronic kidney disease.
Collapse
Affiliation(s)
- Fenghua Zeng
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Lance A Kloepfer
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Charlene Finney
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - André Diedrich
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Veterans Affairs, Nashville, Tennessee
| |
Collapse
|
7
|
Long DL, Ulici V, Chubinskaya S, Loeser RF. Heparin-binding epidermal growth factor-like growth factor (HB-EGF) is increased in osteoarthritis and regulates chondrocyte catabolic and anabolic activities. Osteoarthritis Cartilage 2015; 23:1523-31. [PMID: 25937027 PMCID: PMC4558365 DOI: 10.1016/j.joca.2015.04.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 04/14/2015] [Accepted: 04/22/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE We determined if the epidermal growth factor receptor ligand HB-EGF is produced in cartilage and if it regulates chondrocyte anabolic or catabolic activity. METHODS HB-EGF expression was measured by quantitative PCR using RNA isolated from mouse knee joint tissues and from normal and osteoarthritis (OA) human chondrocytes. Immunohistochemistry was performed on normal and OA human cartilage and meniscus sections. Cultured chondrocytes were treated with fibronectin fragments (FN-f) as a catabolic stimulus and osteogenic protein 1 (OP-1) as an anabolic stimulus. Effects of HB-EGF on cell signaling were analyzed by immunoblotting of selected signaling proteins. MMP-13 was measured in conditioned media, proteoglycan synthesis was measured by sulfate incorporation, and matrix gene expression by quantitative PCR. RESULTS HB-EGF expression was increased in 12-month old mice at 8 weeks after surgery to induce OA and increased amounts of HB-EGF were noted in human articular cartilage from OA knees. FN-f stimulated chondrocyte HB-EGF expression and HB-EGF stimulated chondrocyte MMP-13 production. However, HB-EGF was not required for FN-f stimulation of MMP-13 production. HB-EGF activated the ERK and p38 MAP kinases and stimulated phosphorylation of Smad1 at an inhibitory serine site which was associated with inhibition of OP-1 mediated proteoglycan synthesis and reduced aggrecan (ACAN) but not COL2A1 expression. CONCLUSION HB-EGF is a new factor identified in OA cartilage that promotes chondrocyte catabolic activity while inhibiting anabolic activity suggesting it could contribute to the catabolic-anabolic imbalance seen in OA cartilage.
Collapse
Affiliation(s)
- D L Long
- Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | - V Ulici
- Thurston Arthritis Research Center, Division of Rheumatology, Allergy and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| | - S Chubinskaya
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA.
| | - R F Loeser
- Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Thurston Arthritis Research Center, Division of Rheumatology, Allergy and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
8
|
Bhattacharya D, Chaudhuri S, Singh MK, Chaudhuri S. T11TS inhibits Angiopoietin-1/Tie-2 signaling, EGFR activation and Raf/MEK/ERK pathway in brain endothelial cells restraining angiogenesis in glioma model. Exp Mol Pathol 2015; 98:455-66. [DOI: 10.1016/j.yexmp.2015.03.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 03/17/2015] [Indexed: 12/31/2022]
|
9
|
Shoni M, Lui KO, Vavvas DG, Muto MG, Berkowitz RS, Vlahos N, Ng SW. Protein kinases and associated pathways in pluripotent state and lineage differentiation. Curr Stem Cell Res Ther 2015; 9:366-87. [PMID: 24998240 DOI: 10.2174/1574888x09666140616130217] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 06/07/2014] [Accepted: 06/12/2014] [Indexed: 02/06/2023]
Abstract
Protein kinases (PKs) mediate the reversible conversion of substrate proteins to phosphorylated forms, a key process in controlling intracellular signaling transduction cascades. Pluripotency is, among others, characterized by specifically expressed PKs forming a highly interconnected regulatory network that culminates in a finely-balanced molecular switch. Current high-throughput phosphoproteomic approaches have shed light on the specific regulatory PKs and their function in controlling pluripotent states. Pluripotent cell-derived endothelial and hematopoietic developments represent an example of the importance of pluripotency in cancer therapeutics and organ regeneration. This review attempts to provide the hitherto known kinome profile and the individual characterization of PK-related pathways that regulate pluripotency. Elucidating the underlying intrinsic and extrinsic signals may improve our understanding of the different pluripotent states, the maintenance or induction of pluripotency, and the ability to tailor lineage differentiation, with a particular focus on endothelial cell differentiation for anti-cancer treatment, cell-based tissue engineering, and regenerative medicine strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shu-Wing Ng
- 221 Longwood Avenue, BLI- 449A, Boston MA 02115, USA.
| |
Collapse
|
10
|
Rattik S, Wigren M, Björkbacka H, Fredrikson GN, Hedblad B, Siegbahn A, Bengtsson E, Schiopu A, Edsfeldt A, Dunér P, Grufman H, Gonçalves I, Nilsson J. High plasma levels of heparin-binding epidermal growth factor are associated with a more stable plaque phenotype and reduced incidence of coronary events. Arterioscler Thromb Vasc Biol 2014; 35:222-8. [PMID: 25359857 DOI: 10.1161/atvbaha.114.304369] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Rupture of atherosclerotic plaques is the major cause of acute coronary events (CEs). Plaque destabilization is the consequence of an imbalance between inflammatory-driven degradation of fibrous tissue and smooth muscle cell-dependent tissue repair. Proinflammatory factors have been documented extensively as biomarkers of cardiovascular risk but factors that contribute to stabilization of atherosclerotic plaques have received less attention. The present study aimed to investigate whether plasma levels of the smooth muscle cell growth factor epidermal growth factor (EGF), heparin-binding-EGF (HB-EGF), and platelet-derived growth factor correlate with plaque phenotype and incidence of CEs. APPROACH AND RESULTS HB-EGF, EGF and platelet-derived growth factor were measured in plasma from 202 patients undergoing carotid endarterectomy and in 384 incident CE cases and 409 matched controls recruited from the Malmö Diet and Cancer cohort. Significant positive associations were found between the plasma levels of all 3 growth factors and the collagen and elastin contents of the removed plaques. CE cases in the Malmö Diet and Cancer cohort had lower levels of HB-EGF in plasma, whereas no significant differences were found for EGF and platelet-derived growth factor. After adjusting for cardiovascular risk factors in a Cox proportional hazard model, the hazard ratio for the highest HB-EGF tertile was 0.61 (95% confidence interval, 0.47-0.82; P<0.001). CONCLUSIONS The associations between high levels of smooth muscle cell growth factors in plasma and a more fibrous plaque phenotype as well as the association between low levels of HB-EGF and incident CEs point to a potential clinically important role for factors that contribute to plaque stabilization by stimulating smooth muscle cells.
Collapse
Affiliation(s)
- Sara Rattik
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (S.R., M.W., H.B., G.N.F., B.H., E.B., A.S., A.E., P.D., H.G., I.G., J.N.); Department of Cardiology-Coronary diseases, Skåne University Hospital, Malmö, Sweden (A.S., A.E., I.G.); and Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden (A.S.).
| | - Maria Wigren
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (S.R., M.W., H.B., G.N.F., B.H., E.B., A.S., A.E., P.D., H.G., I.G., J.N.); Department of Cardiology-Coronary diseases, Skåne University Hospital, Malmö, Sweden (A.S., A.E., I.G.); and Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden (A.S.)
| | - Harry Björkbacka
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (S.R., M.W., H.B., G.N.F., B.H., E.B., A.S., A.E., P.D., H.G., I.G., J.N.); Department of Cardiology-Coronary diseases, Skåne University Hospital, Malmö, Sweden (A.S., A.E., I.G.); and Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden (A.S.)
| | - Gunilla Nordin Fredrikson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (S.R., M.W., H.B., G.N.F., B.H., E.B., A.S., A.E., P.D., H.G., I.G., J.N.); Department of Cardiology-Coronary diseases, Skåne University Hospital, Malmö, Sweden (A.S., A.E., I.G.); and Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden (A.S.)
| | - Bo Hedblad
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (S.R., M.W., H.B., G.N.F., B.H., E.B., A.S., A.E., P.D., H.G., I.G., J.N.); Department of Cardiology-Coronary diseases, Skåne University Hospital, Malmö, Sweden (A.S., A.E., I.G.); and Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden (A.S.)
| | - Agneta Siegbahn
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (S.R., M.W., H.B., G.N.F., B.H., E.B., A.S., A.E., P.D., H.G., I.G., J.N.); Department of Cardiology-Coronary diseases, Skåne University Hospital, Malmö, Sweden (A.S., A.E., I.G.); and Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden (A.S.)
| | - Eva Bengtsson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (S.R., M.W., H.B., G.N.F., B.H., E.B., A.S., A.E., P.D., H.G., I.G., J.N.); Department of Cardiology-Coronary diseases, Skåne University Hospital, Malmö, Sweden (A.S., A.E., I.G.); and Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden (A.S.)
| | - Alexandru Schiopu
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (S.R., M.W., H.B., G.N.F., B.H., E.B., A.S., A.E., P.D., H.G., I.G., J.N.); Department of Cardiology-Coronary diseases, Skåne University Hospital, Malmö, Sweden (A.S., A.E., I.G.); and Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden (A.S.)
| | - Andreas Edsfeldt
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (S.R., M.W., H.B., G.N.F., B.H., E.B., A.S., A.E., P.D., H.G., I.G., J.N.); Department of Cardiology-Coronary diseases, Skåne University Hospital, Malmö, Sweden (A.S., A.E., I.G.); and Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden (A.S.)
| | - Pontus Dunér
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (S.R., M.W., H.B., G.N.F., B.H., E.B., A.S., A.E., P.D., H.G., I.G., J.N.); Department of Cardiology-Coronary diseases, Skåne University Hospital, Malmö, Sweden (A.S., A.E., I.G.); and Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden (A.S.)
| | - Helena Grufman
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (S.R., M.W., H.B., G.N.F., B.H., E.B., A.S., A.E., P.D., H.G., I.G., J.N.); Department of Cardiology-Coronary diseases, Skåne University Hospital, Malmö, Sweden (A.S., A.E., I.G.); and Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden (A.S.)
| | - Isabel Gonçalves
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (S.R., M.W., H.B., G.N.F., B.H., E.B., A.S., A.E., P.D., H.G., I.G., J.N.); Department of Cardiology-Coronary diseases, Skåne University Hospital, Malmö, Sweden (A.S., A.E., I.G.); and Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden (A.S.)
| | - Jan Nilsson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (S.R., M.W., H.B., G.N.F., B.H., E.B., A.S., A.E., P.D., H.G., I.G., J.N.); Department of Cardiology-Coronary diseases, Skåne University Hospital, Malmö, Sweden (A.S., A.E., I.G.); and Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden (A.S.)
| |
Collapse
|
11
|
Abstract
Recruitment of mural cells (MCs), namely pericytes and smooth muscle cells (SMCs), is essential to improve the maturation of newly formed vessels. Sonic hedgehog (Shh) has been suggested to promote the formation of larger and more muscularized vessels, but the underlying mechanisms of this process have not yet been elucidated. We first identified Shh as a target of platelet-derived growth factor BB (PDGF-BB) and found that SMCs respond to Shh by upregulating extracellular signal-regulated kinase 1/2 and Akt phosphorylation. We next showed that PDGF-BB-induced SMC migration was reduced after inhibition of Shh or its signaling pathway. Moreover, we found that PDGF-BB-induced SMC migration involves Shh-mediated motility. In vivo, in the mouse model of corneal angiogenesis, Shh is expressed by MCs of newly formed blood vessels. PDGF-BB inhibition reduced Shh expression, demonstrating that Shh is a target of PDGF-BB, confirming in vitro experiments. Finally, we found that in vivo inhibition of either PDGF-BB or Shh signaling reduces NG2(+) MC recruitment into neovessels and subsequently reduces neovessel life span. Our findings demonstrate, for the first time, that Shh is involved in PDGF-BB-induced SMC migration and recruitment of MCs into neovessels and elucidate the molecular signaling pathway involved in this process.
Collapse
|
12
|
Low nitric oxide bioavailability upregulates renal heparin binding EGF-like growth factor expression. Kidney Int 2013; 84:1176-88. [PMID: 23760291 PMCID: PMC3796048 DOI: 10.1038/ki.2013.214] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 03/29/2013] [Accepted: 04/04/2013] [Indexed: 12/20/2022]
Abstract
Decreased nitric oxide bioavailability plays an important role in the initiation and progression of diabetic nephropathy, but the underlying mechanisms remain unclear. Here, we found that heparin binding epidermal growth factor-like growth factor (HB-EGF) expression levels increased in the kidneys of both endothelial nitric oxide synthase (eNOS) knockout and eNOS knockout diabetic (Lepr db/db) mice as early as 8 weeks of age. Further increases in expression were only seen in eNOS knockout diabetic mice and paralleled the progression of glomerulopathy. HB-EGF expression increased in endothelium, podocytes, and tubular epithelial cells. In cultured glomerular endothelial cells, the nitric oxide synthase inhibitors NG-nitro-L-arginine methyl ester (L-NAME) or L-N5-(1-Iminoethyl) ornithine increased HB-EGF protein expression. Administration of L-NAME dramatically increased renal HB-EGF expression and urinary HB-EGF excretion in diabetic mice. On the other hand, replenishing nitric oxide with sodium nitrate in eNOS knockout diabetic mice reduced urinary HB-EGF excretion and inhibited the progression of diabetic nephropathy. Furthermore, specific deletion of HB-EGF expression in endothelium attenuated renal injury in diabetic eNOS knockout mice. Thus, our results suggest that decreased nitric oxide bioavailability leads to increased HB-EGF expression, which may be an important mediator of the resulting progressive diabetic nephropathy in eNOS knockout diabetic mice.
Collapse
|
13
|
Two-stage hepatectomy with effective perioperative chemotherapy does not induce tumor growth or growth factor expression in liver metastases from colorectal cancer. Surgery 2013; 153:179-88. [DOI: 10.1016/j.surg.2012.06.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 06/08/2012] [Indexed: 12/29/2022]
|
14
|
Vourtsis SA, Spyriounis PK, Agrogiannis GD, Ionac M, Papalois AE. VEGF Application on Rat Skin Flap Survival. J INVEST SURG 2012; 25:14-9. [DOI: 10.3109/08941939.2011.593693] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
15
|
Abstract
The human endometrium is exposed to repeated inflammation every month, culminating in tissue breakdown and menstruation. Subsequently, the endometrium has a remarkable capacity for efficient repair and remodeling to enable implantation if fertilization takes place. Endometrial function is known to be governed by the ovarian hormones estradiol and progesterone. This review paper focuses on hormonal control of the cyclical tissue injury and repair that takes place in the local endometrial environment at the time of menstruation. Progesterone levels decline premenstrually as the corpus luteum regresses in the absence of pregnancy, and estradiol levels increase during the postmenstrual phase. The functional impact of these significant changes is discussed, including their immediate and downstream effects. Finally, we examine the contribution of aberrant endometrial function to the presentation of heavy menstrual bleeding and identify potential therapeutic targets for the treatment of this common gynecological problem.
Collapse
Affiliation(s)
- Jacqueline A Maybin
- The University of Edinburgh's Centre for Reproductive Biology, The Queen's Medical Research Institute, Edinburgh, UK
| | | |
Collapse
|
16
|
Maybin JA, Critchley HOD, Jabbour HN. Inflammatory pathways in endometrial disorders. Mol Cell Endocrinol 2011; 335:42-51. [PMID: 20723578 DOI: 10.1016/j.mce.2010.08.006] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 08/11/2010] [Accepted: 08/11/2010] [Indexed: 01/19/2023]
Abstract
Complex interactions between the endocrine and immune systems govern the key endometrial events of implantation and menstruation. In contrast to other tissue sites, cyclical endometrial inflammation is physiological. However, dysregulation of this inflammatory response can lead to endometrial disorders. This review examines the inflammatory processes occurring in the normal endometrium during menstruation and implantation, highlighting recent advances in our understanding and gaps in current knowledge. Subsequently, the role of inflammatory pathways in the pathology of various common endometrial conditions is discussed, including heavy menstrual bleeding, dysmenorrhoea (painful periods), uterine fibroids, endometriosis and recurrent miscarriage.
Collapse
Affiliation(s)
- Jacqueline A Maybin
- University of Edinburgh Centre for Reproductive Biology, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | | | | |
Collapse
|
17
|
VEGF non-angiogenic functions in adult organ homeostasis: therapeutic implications. J Mol Med (Berl) 2011; 89:635-45. [DOI: 10.1007/s00109-011-0739-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 01/19/2011] [Accepted: 02/14/2011] [Indexed: 12/21/2022]
|
18
|
Hernández Vera R, Genové E, Alvarez L, Borrós S, Kamm R, Lauffenburger D, Semino CE. Interstitial fluid flow intensity modulates endothelial sprouting in restricted Src-activated cell clusters during capillary morphogenesis. Tissue Eng Part A 2009; 15:175-85. [PMID: 18636940 DOI: 10.1089/ten.tea.2007.0314] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Development of tissues in vitro with dimensions larger than 150 to 200 microm requires the presence of a functional vascular network. Therefore, we have studied capillary morphogenesis under controlled biological and biophysical conditions with the aim of promoting vascular structures in tissue constructs. We and others have previously demonstrated that physiological values of interstitial fluid flow normal to an endothelial monolayer in combination with vascular endothelial growth factor play a critical role during capillary morphogenesis by promoting cell sprouting. In the present work, we studied the effect that a range of interstitial flow velocities (0-50 microm/min) has in promoting the amount, length, and branching of developing sprouts during capillary morphogenesis. The number of capillary-like structures developed from human umbilical vein endothelial cell monolayers across the interstitial flow values tested was not significantly affected. Instead, the length and branching degree of the sprouts presented a significant maximum at flow velocities of 10 to 20 microm/min. More-over, at these same flow values, the phosphorylation level of Src also showed its peak. We discovered that capillary morphogenesis is restricted to patches of Src-activated cells (phosphorylated Src (pSrc)) at the monolayer, suggesting that the transduction pathway in charge of sensing the mechanical stimulus induced by flow is promoting predetermined mechanically sensitive areas (pSrc) to undergo capillary morphogenesis
Collapse
Affiliation(s)
- Rodrigo Hernández Vera
- Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Belmadani S, Matrougui K, Kolz C, Pung YF, Palen D, Prockop DJ, Chilian WM. Amplification of coronary arteriogenic capacity of multipotent stromal cells by epidermal growth factor. Arterioscler Thromb Vasc Biol 2009; 29:802-8. [PMID: 19342596 DOI: 10.1161/atvbaha.109.186189] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We determined whether increasing adherence of multipotent stromal cells (MSCs) would amplify their effects on coronary collateral growth (CCG). METHODS AND RESULTS Adhesion was established in cultured coronary endothelials cells (CECs) or MSCs treated with epidermal growth factor (EGF). EGF increased MSCs adhesion to CECs, and increased intercellular adhesion molecule (ICAM-1) or vascular cell adhesion molecule (VCAM-1) expression. Increased adherence was blocked by EGF receptor antagonism or antibodies to the adhesion molecules. To determine whether adherent MSCs, treated with EGF, would augment CCG, repetitive episodes of myocardial ischemia (RI) were introduced and CCG was measured from the ratio of collateral-dependent (CZ) and normal zone (NZ) flows. CZ/NZ was increased by MSCs without treatment versus RI-control and was further increased by EGF-treated MSCs. EGF-treated MSCs significantly improved myocardial function versus RI or RI+MSCs demonstrating that the increase in collateral flow was functionally significant. Engraftment of MSCs into myocardium was also increased by EGF treatment. CONCLUSIONS These results reveal the importance of EGF in MSCs adhesion to endothelium and suggest that MSCs may be effective therapies for the stimulation of coronary collateral growth when interventions are used to increase their adhesion and homing (in vitro EGF treatment) to the jeopardized myocardium.
Collapse
|
20
|
Liver regeneration and tumor stimulation--a review of cytokine and angiogenic factors. J Gastrointest Surg 2008; 12:966-80. [PMID: 18181006 DOI: 10.1007/s11605-007-0459-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Accepted: 12/06/2007] [Indexed: 01/31/2023]
Abstract
Liver resection for metastatic (colorectal carcinoma) tumors is often followed by a significant incidence of tumor recurrence. Cellular and molecular changes resulting from hepatectomy and the subsequent liver regeneration process may influence the kinetics of tumor growth and contribute to recurrence. Clinical and experimental evidence suggests that factors involved in liver regeneration may also stimulate the growth of occult tumors and the reactivation of dormant micrometastases. An understanding of the underlying changes may enable alternative strategies to minimize tumor recurrence and improve patient survival after hepatectomy.
Collapse
|
21
|
Aquaporin 1 is required for hypoxia-inducible angiogenesis in human retinal vascular endothelial cells. Microvasc Res 2008; 75:297-301. [DOI: 10.1016/j.mvr.2007.12.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Revised: 12/01/2007] [Accepted: 12/04/2007] [Indexed: 12/23/2022]
|
22
|
Zania P, Papaconstantinou M, Flordellis CS, Maragoudakis ME, Tsopanoglou NE. Thrombin mediates mitogenesis and survival of human endothelial cells through distinct mechanisms. Am J Physiol Cell Physiol 2008; 294:C1215-26. [PMID: 18367587 DOI: 10.1152/ajpcell.00452.2007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Thrombin has been reported to play a pivotal role in the initiation of angiogenesis by indirectly regulating and organizing a network of angiogenic molecules. In addition, it has been proposed that thrombin can directly activate endothelial cell proliferation. However, in this report it was shown that thrombin is a poor growth factor for human endothelial cells, and its modest mitogenic activity is mediated indirectly by the release of heparin-binding epidermal growth factor, subsequent to proteinase-activated receptor 1 (PAR1) activation. On the other hand, it was demonstrated that thrombin is a potent anti-apoptotic factor for endothelial cells, pointing to a novel role of thrombin in vascular protection. Analysis by annexin V-propidium iodide double staining revealed that thrombin, specifically, promoted survival of serum-starved endothelial cells in a concentration-dependent manner. In contrast to its mitogenic effect, the anti-apoptotic effect of thrombin was largely independent of its catalytic activity and was mediated through interaction with alphanubeta3 and alpha5beta1 integrins, whereas the involvement of PAR1 was limited. These results provide new insights in understanding the role of thrombin in endothelial cell signaling and vascular biology.
Collapse
Affiliation(s)
- Panagiota Zania
- Department of Pharmacology, Medical School, University of Patras, 26500 Patras, Greece
| | | | | | | | | |
Collapse
|
23
|
De Luca A, Carotenuto A, Rachiglio A, Gallo M, Maiello MR, Aldinucci D, Pinto A, Normanno N. The role of the EGFR signaling in tumor microenvironment. J Cell Physiol 2008; 214:559-67. [PMID: 17894407 DOI: 10.1002/jcp.21260] [Citation(s) in RCA: 270] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The epidermal growth factor receptor (EGFR) family comprehends four different tyrosine kinases (EGFR, ErbB-2, ErbB-3, and ErbB-4) that are activated following binding to epidermal growth factor (EGF)-like growth factors. It has been long established that the EGFR system is involved in tumorigenesis. These proteins are frequently expressed in human carcinomas and support proliferation and survival of cancer cells. However, activation of the EGFR in non-malignant cell populations of the neoplastic microenvironment might also play an important role in cancer progression. EGFR signaling regulates in tumor cells the synthesis and secretion of several different angiogenic growth factors, including vascular endothelial growth factor (VEGF), interleukin-8 (IL-8), and basic fibroblast growth factor (bFGF). Overexpression of ErbB-2 also leads to increased expression of angiogenic growth factors, whereas treatment with anti-EGFR or anti-ErbB-2 agents produces a significant reduction of the synthesis of these proteins by cancer cells. EGFR expression and function in tumor-associated endothelial cells has also been described. Therefore, EGFR signaling might regulate angiogenesis both directly and indirectly. In addition, activation of EGFR is involved in the pathogenesis of bone metastases. Within the bone marrow microenvironment, cancer cells stimulate the synthesis of osteoclastogenic factors by residing stromal cells, a phenomenon that leads to bone destruction. It has been shown that EGFR signaling regulates the ability of bone marrow stromal cells to produce osteoclastogenic factors and to sustain osteoclast activation. Taken together, these findings suggest that the EGFR system is an important mediator, within the tumor microenvironment, of autocrine and paracrine circuits that result in enhanced tumor growth.
Collapse
Affiliation(s)
- Antonella De Luca
- Cell Biology and Preclinical Models Unit, INT-Fondazione Pascale, Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
OBJECTIVE Heparin-binding EGF-like growth factor (HB-EGF) belongs to the epidermal growth factor (EGF) superfamily of ligands. It has been implicated as a regulator of angiogenesis. However, the mechanisms by which HB-EGF promotes angiogenesis are unknown. The goal of the present study was to define the pathways by which HB-EGF stimulates angiogenesis in endothelial cells. METHODS To characterize the angiogenic activity of HB-EGF, we treated human umbilical vein endothelial cells (HUVEC) with HB-EGF and analyzed the effects on cell proliferation, migration and tube formation. Side-by-side assays with EGF were used for comparison. RESULTS Both HB-EGF and EGF stimulated HUVEC migration in scratch assays and promoted vascular tube formation in 2D-angiogenesis assays, without inducing cell proliferation. HB-EGF- and EGF-induced HUVEC migration and capillary tube formation were dependent upon activation of PI3K, MAPK and eNOS. Importantly, HB-EGF-and EGF-induced tube formation was comparable to, but were independent of tube formation induced by VEGF. CONCLUSIONS We have demonstrated that HB-EGF and EGF induce angiogenesis via activation of PI3K, MAPK and eNOS in a VEGF-independent fashion. Thus, the role played by HB-EGF in stimulating physiologic processes such as wound healing in vivo may be dependent, in part, on its ability to promote angiogenesis.
Collapse
Affiliation(s)
- Veela B Mehta
- Department of Pediatric Surgery, The Research Institute at Nationwide Children's Hospital, Center for Perinatal Research, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA
| | | |
Collapse
|
25
|
Abstract
Cardiovascular diseases are the major cause of morbidity and mortality in both men and women in industrially developed countries. These disorders may result from impaired angiogenesis, particularly in response to hypoxia. Despite many limitations, gene therapy is still emerging as a potential alternative for patients who are not candidates for traditional revascularization procedures, like angioplasty or vein grafts. This review focuses on recent approaches in the development of new gene delivery vectors, with great respect to newly discovered AAV serotypes and their modified forms. Moreover, some new cardiovascular gene therapy strategies have been highlighted, such as combination of different angiogenic growth factors or simultaneous application of genes and progenitor cells in order to obtain stable and functional blood vessels in ischemic tissue.
Collapse
Affiliation(s)
| | | | - J. Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland; Tel: +48-12-664-63-75; Fax: +48-12-664-69-18; E-mail:
| |
Collapse
|
26
|
Malhas A, Lee CF, Sanders R, Saunders NJ, Vaux DJ. Defects in lamin B1 expression or processing affect interphase chromosome position and gene expression. J Cell Biol 2007; 176:593-603. [PMID: 17312019 PMCID: PMC2064018 DOI: 10.1083/jcb.200607054] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2006] [Accepted: 01/24/2007] [Indexed: 11/22/2022] Open
Abstract
Radial organization of nuclei with peripheral gene-poor chromosomes and central gene-rich chromosomes is common and could depend on the nuclear boundary as a scaffold or position marker. To test this, we studied the role of the ubiquitous nuclear envelope (NE) component lamin B1 in NE stability, chromosome territory position, and gene expression. The stability of the lamin B1 lamina is dependent on lamin endoproteolysis (by Rce1) but not carboxymethylation (by Icmt), whereas lamin C lamina stability is not affected by the loss of full-length lamin B1 or its processing. Comparison of wild-type murine fibroblasts with fibroblasts lacking full-length lamin B1, or defective in CAAX processing, identified genes that depend on a stable processed lamin B1 lamina for normal expression. We also demonstrate that the position of mouse chromosome 18 but not 19 is dependent on such a stable nuclear lamina. The results implicate processed lamin B1 in the control of gene expression as well as chromosome position.
Collapse
Affiliation(s)
- Ashraf Malhas
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, England, UK
| | | | | | | | | |
Collapse
|
27
|
Ohnishi S, Yasuda T, Kitamura S, Nagaya N. Effect of hypoxia on gene expression of bone marrow-derived mesenchymal stem cells and mononuclear cells. Stem Cells 2007; 25:1166-77. [PMID: 17289933 DOI: 10.1634/stemcells.2006-0347] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
MSC have self-renewal and multilineage differentiation potential, including differentiation into endothelial cells and vascular smooth muscle cells. Although bone marrow-derived mononuclear cells (MNC) have been applied for therapeutic angiogenesis in ischemic tissue, little information is available regarding comparison of the molecular foundation between MNC and their MSC subpopulation, as well as their response to ischemic conditions. Thus, we investigated the gene expression profiles between MSC and MNC of rat bone marrow under normoxia and hypoxia using a microarray containing 31,099 genes. In normoxia, 2,232 (7.2%) and 2,193 genes (7.1%) were preferentially expressed more than threefold in MSC and MNC, respectively, and MSC expressed a number of genes involved in development, morphogenesis, cell adhesion, and proliferation, whereas various genes highly expressed in MNC were involved in inflammatory response and chemotaxis. Under hypoxia, 135 (0.44%) and 49 (0.16%) genes were upregulated (>threefold) in MSC and MNC, respectively, and a large number of those upregulated genes were involved in glycolysis and metabolism. Focusing on genes encoding secretory proteins, the upregulated genes in MSC under hypoxia included several molecules involved in cell proliferation and survival, such as vascular endothelial growth factor-D, placenta growth factor, pre-B-cell colony-enhancing factor 1, heparin-binding epidermal growth factor-like growth factor, and matrix metalloproteinase-9, whereas the upregulated genes in MNC under hypoxia included proinflammatory cytokines such as chemokine (C-X-C motif) ligand 2 and interleukin-1alpha. Our results may provide information on the differential molecular mechanisms regulating the properties of MSC and MNC under ischemic conditions. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Shunsuke Ohnishi
- Department of Regenerative Medicine and Tissue Engineering, National Cardiovascular Center, 5-7-1 Fujishirodai, Osaka 565-8565, Japan.
| | | | | | | |
Collapse
|
28
|
Malik S, Day K, Perrault I, Charnock-Jones DS, Smith SK. Reduced levels of VEGF-A and MMP-2 and MMP-9 activity and increased TNF-alpha in menstrual endometrium and effluent in women with menorrhagia. Hum Reprod 2006; 21:2158-66. [PMID: 16585124 DOI: 10.1093/humrep/del089] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Heavy regular menstrual periods (menorrhagia) are an important cause of ill health in women and remain the leading indication for hysterectomy. Abnormalities of the endometrial blood vessels are among the possible causes of this condition. Many different factors affect endothelial cell growth, function and vessel remodelling. We sought to determine whether the levels of vascular endothelial growth factor-A (VEGF-A), tumour necrosis factor-alpha (TNF-alpha), matrix metalloproteinase (MMP)-2 and MMP-9 and soluble VEGF receptor-1 (VEGF-R1) were altered in the menstrual effluent of women with objective menorrhagia. We have also quantitated the VEGF-A mRNA in the menstruated endometrium. METHODS AND RESULTS We recruited 37 women and determined their menstrual blood loss (MBL) over two cycles and collected menstrual effluent during the 2nd day of bleeding for 4 h. There was no difference in the total level of VEGF-A, and neither latent MMP. However, the concentration of VEGF-A was significantly reduced in the women with menorrhagia, as was the VEGF-A mRNA level. In addition, the active forms of both MMPs were markedly reduced and the total sVEGF-R1 as well as the TNF-alpha content were increased. CONCLUSIONS This is the first study to show abnormalities of factors important for endothelial cell behaviour in the endometrium of women with menorrhagia. This may underlie the disordered vessel structure and/or function in this condition.
Collapse
Affiliation(s)
- Shazia Malik
- Department of Obstetrics and Gynaecology, The Rosie Hospital, Robinson Way, Cambridge, UK.
| | | | | | | | | |
Collapse
|
29
|
Wheatley C. A scarlet pimpernel for the resolution of inflammation? The role of supra-therapeutic doses of cobalamin, in the treatment of systemic inflammatory response syndrome (SIRS), sepsis, severe sepsis, and septic or traumatic shock. Med Hypotheses 2006; 67:124-42. [PMID: 16545917 DOI: 10.1016/j.mehy.2006.01.036] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Accepted: 01/19/2006] [Indexed: 02/06/2023]
Abstract
Cobalamin carrier proteins,the Transcobalamins (TCS), are elevated during trauma, infections and chronic inflammatory conditions. This remains un-explained. It is proposed that such TC elevations signal a need for cobalamin central to the resolution of inflammation. Thus Cobalamin may regulate the transcription factor, NFkappaB, activation or suppression of which determines the inflammatory response and its resolution. Such regulation may involve at least 5 separate mechanisms: (i) hormone-like regulation of TNFalpha, through reduction of excess NO by cobalamin, as well as through the selective inhibition, in tandem with glutathione, of inducible nitric oxide synthase; (ii) quenching of nitric oxide radicals and reactive oxygen species, enhanced by cobalamin's glutathione sparing effect; (iii) the promotion of acetylcholine synthesis, central to the neuro-immune cholinergic anti-inflammatory pathway; (iv) the promotion of oxidative phosphorylation; (v) and a bacteriostatic role of the TCS released by neutrophil secondary granules during phagocytosis, which also appears to modulate the inflammatory response. TC elevations are dependent on NFkappaB activation, through crosstalk between NFkappaB and Sp1, another member of the helix-loop-helix protein family, which directly mediates transcription of the TCII gene. Sp1 also has binding sites on the TNFalpha and EGF gene promoters. NFkappaB may thus ensure sufficient cobalamin to determine its own eventual suppression. Cobalamin's established regulation of EGF may additionally preserve normal function of macrophages and the coagulation cascade in wound healing. By regulating NFkappaB, Cobalamin may also be the as yet unidentified mediator needed to potentiate the anti-inflammatory action of eicosanoids derived from omega-3 essential fatty acids. Moreover, animal and human clinical data suggests that high dose cobalamin may prove a promising approach to SIRS/sepsis/septic and traumatic shock.
Collapse
Affiliation(s)
- Carmen Wheatley
- Orthomolecular Oncology, 4, Richmond Road, Oxford OX1 2JJ, United Kingdom.
| |
Collapse
|
30
|
Paquin A, Barnabé-Heider F, Kageyama R, Miller FD. CCAAT/enhancer-binding protein phosphorylation biases cortical precursors to generate neurons rather than astrocytes in vivo. J Neurosci 2006; 25:10747-58. [PMID: 16291948 PMCID: PMC6725854 DOI: 10.1523/jneurosci.2662-05.2005] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The intracellular mechanisms that bias mammalian neural precursors to generate neurons versus glial cells are not well understood. We demonstrated previously that the growth factor-regulated mitogen-activated protein kinase kinase (MEK) and its downstream target, the CCAAT/enhancer-binding protein (C/EBP) family of transcription factors, are essential for neurogenesis in cultured cortical precursor cells (Ménard et al., 2002). Here, we examined a role for this pathway during cortical cell fate determination in vivo using in utero electroporation of the embryonic cortex. These studies demonstrate that inhibition of the activity of either MEK or the C/EBPs inhibits the genesis of neurons in vivo. Moreover, the MEK pathway mediates phosphorylation of C/EBPbeta in cortical precursors, and expression of a C/EBPbeta construct in which the MEK pathway phosphorylation sites are mutated inhibits neurogenesis. Conversely, expression of a C/EBPbeta construct, in which the same sites are mutated to glutamate and therefore are "constitutively" phosphorylated, enhances neurogenesis in the early embryonic cortex. A subpopulation of precursors in which C/EBP activity is inhibited are maintained as cycling precursors in the ventricular/subventricular zone of the cortex until early in postnatal life, when they have an enhanced propensity to generate astrocytes, presumably in response to gliogenic signals in the neonatal environment. Thus, activation of an MEK-C/EBP pathway in cortical precursors in vivo biases them to become neurons and against becoming astrocytes, thereby acting as a growth factor-regulated switch.
Collapse
Affiliation(s)
- Annie Paquin
- Developmental Biology, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
31
|
van Cruijsen H, Giaccone G, Hoekman K. Epidermal growth factor receptor and angiogenesis: Opportunities for combined anticancer strategies. Int J Cancer 2005; 117:883-8. [PMID: 16152621 DOI: 10.1002/ijc.21479] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tumor-induced angiogenesis is essential for malignant growth. This mini review focuses on the role of vascular endothelial growth factor (VEGF) and epidermal growth factor (EGF) and their receptors in this process, and the rationale to combine inhibitors of these growth factors as anticancer therapy. Concomitantly, targeting the VEGF(R) and the EGF(R) signaling pathway may circumvent the problem of acquired resistance to EGFR inhibitors. By targeting both pathways, the antiangiogenic effect may be more pronounced, which may lead to greater antitumor activity. Preliminary efficacy data from clinical trials encourage further exploration of this combined anticancer strategy.
Collapse
Affiliation(s)
- Hester van Cruijsen
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
32
|
Eng E, Holgren C, Hubchak S, Naaz P, Schnaper HW. Hypoxia regulates PDGF-B interactions between glomerular capillary endothelial and mesangial cells. Kidney Int 2005; 68:695-703. [PMID: 16014047 DOI: 10.1111/j.1523-1755.2005.00448.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Platelet-derived growth factor (PDGF)-B regulates mesangial cell and vessel development during embryogenesis, and contributes to the pathogenesis of adult renal and vascular diseases. Endothelial cell PDGF-B exerts paracrine effects on mesangial cells, but its regulation is not well defined. We examined the impact of hypoxia on PDGF-B-mediated interactions between glomerular endothelial and mesangial cells, a condition of potential relevance in developing, and diseased adult, kidneys. METHODS Glomerular endothelial or mesangial cells were subjected to hypoxia and responses compared to normoxic cells. Endothelial PDGF-B was studied by Northern and Western analysis. Mesangial proliferative responses to PDGF-B were assessed by (3)H-thymidine incorporation, and migration by a modified Boyden chamber assay. Hypoxia-induced changes in receptor specific binding capacity were studied by saturation binding assays. RESULTS Hypoxia stimulated increases in endothelial PDGF-B mRNA and protein. In normoxic mesangial cells, PDGF-B stimulated dose-dependent proliferation, but the proliferative response of hypoxic cells was two to three times greater. Exogenous PDGF-B also caused prompter migration in hypoxic mesangial cells. Mesangial cells were treated with endothelial cell-conditioned medium. More cells migrated when hypoxic cells were stimulated with hypoxic conditioned medium, than when normoxic cells were stimulated with normoxic conditioned medium. Preincubating conditioned medium with PDGF-B neutralizing antibody greatly decreased the chemoattractant activity. Binding studies demonstrated increased specific binding capacity in hypoxic cells. CONCLUSION Hypoxia enhances PDGF-B paracrine interactions between glomerular endothelial and mesangial cells. These hypoxia-regulated interactions may be important during glomerulogenesis in fetal life and during the pathogenesis of adult glomerular disease.
Collapse
Affiliation(s)
- Eudora Eng
- Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA.
| | | | | | | | | |
Collapse
|
33
|
Dreux AC, Lamb DJ, Modjtahedi H, Ferns GAA. The epidermal growth factor receptors and their family of ligands: their putative role in atherogenesis. Atherosclerosis 2005; 186:38-53. [PMID: 16076471 DOI: 10.1016/j.atherosclerosis.2005.06.038] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2005] [Revised: 06/21/2005] [Accepted: 06/23/2005] [Indexed: 12/12/2022]
Abstract
The epidermal growth factor receptor is a member of type-I growth factor receptor family with tyrosine kinase activity that is activated following the binding of multiple cognate ligands. Several members of the EGF family of ligands are expressed by cells involved in atherogenesis. EGF receptor mediated processes have been well characterised within epithelial, smooth muscle and tumour cell lines in vitro, and the EGF receptor has been identified immunocytochemically on intimal smooth muscle cells within atherosclerotic plaques. There is also limited evidence for the expression of the EGF receptor family on leukocytes, although their function has yet to be clarified. In this review, we will discuss the biological functions of this receptor and its ligands and their potential to modulate the function of cells involved in the atherosclerotic process.
Collapse
Affiliation(s)
- Alys C Dreux
- Centre for Clinical Science & Measurement, School of Biomedical & Molecular Sciences, University of Surrey, Guildford, Surrey GU2 7XH, UK.
| | | | | | | |
Collapse
|
34
|
Chalothorn D, Moore SM, Zhang H, Sunnarborg SW, Lee DC, Faber JE. Heparin-binding epidermal growth factor-like growth factor, collateral vessel development, and angiogenesis in skeletal muscle ischemia. Arterioscler Thromb Vasc Biol 2005; 25:1884-90. [PMID: 15994441 DOI: 10.1161/01.atv.0000175761.59602.16] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Heparin-binding epidermal growth factor-like growth factor (HB-EGF) is a potent mitogen for smooth muscle cells and has been implicated in atherosclerosis, tissue regeneration after ischemia, vascular development, and tumor angiogenesis. We examined the hypothesis that HB-EGF participates in angiogenesis and collateral growth in ischemia. METHODS AND RESULTS During 3 weeks after femoral artery ligation, no attenuation occurred in recovery of hindlimb perfusion or distal saphenous artery flow in HB-EGF-null (HB-EGF(-/-)) versus wild-type mice. Lumen diameters of remodeled collaterals in gracilis muscle were similar by morphometry (87+/-8 versus 94+/-6 microm) and angiography, although medial thickening was reduced. Gastrocnemius muscle underwent comparable angiogenesis (41% and 33% increase in capillary-to-muscle fiber ratio). Renal renin mRNA, arterial pressure, and heart rate during anesthesia or conscious unrestrained conditions were similar between groups. These latter findings validate comparisons of perfusion data and also suggest that differences in arterial pressure and/or renin-angiotensin activity are not masking an otherwise inhibitory effect of HB-EGF absence. Four days after ligation, EGF receptor phosphorylation increased in muscle by 104% in wild-type but by only 30% in HB-EGF(-/-) mice. This argues against compensation by other EGF receptor ligands. CONCLUSIONS Our results suggest that HB-EGF is not required for arteriogenesis or angiogenesis in hindlimb ischemia.
Collapse
Affiliation(s)
- Dan Chalothorn
- Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, NC 27599-7545, USA
| | | | | | | | | | | |
Collapse
|
35
|
Affiliation(s)
- Ian Zachary
- Department of Medicine, University College London, 5 University Street, London WC1E 6JJ, UK.
| |
Collapse
|
36
|
Ongusaha PP, Kwak JC, Zwible AJ, Macip S, Higashiyama S, Taniguchi N, Fang L, Lee SW. HB-EGF is a potent inducer of tumor growth and angiogenesis. Cancer Res 2004; 64:5283-90. [PMID: 15289334 DOI: 10.1158/0008-5472.can-04-0925] [Citation(s) in RCA: 165] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Heparin-binding epidermal growth factor-like growth factor (HB-EGF) has been shown to stimulate the growth of a variety of cells in an autocrine or paracrine manner. Although HB-EGF is widely expressed in tumors compared with normal tissue, its contribution to tumorigenicity is unknown. HB-EGF can be produced as a membrane-anchored form (pro-HB-EGF) and later processed to a soluble form (s-HB-EGF), although a significant amount of pro-HB-EGF remains uncleaved on the cell surface. To understand the roles of two forms of HB-EGF in promoting tumor growth, we have studied the effects of HB-EGF expression in the process of tumorigenesis using in vitro and in vivo systems. We demonstrate here that in EJ human bladder cancer cells containing a tetracycline-regulatable s-HB-EGF or pro-HB-EGF expression system, s-HB-EGF expression increased their transformed phenotypes, including growth rate, colony-forming ability, and activation of cyclin D1 promoter, as well as induction of vascular endothelial growth factor in vitro. Moreover, s-HB-EGF or wild-type HB-EGF induced the expression and activities of the metalloproteases, MMP-9 and MMP-3, leading to enhanced cell migration. In vivo studies also demonstrated that tumor cells expressing s-HB-EGF or wild-type HB-EGF significantly enhanced tumorigenic potential in athymic nude mice and exerted an angiogenic effect, increasing the density and size of tumor blood vessels. However, cells expressing solely pro-HB-EGF did not exhibit any significant tumorigenic potential. These findings establish s-HB-EGF as a potent inducer of tumor growth and angiogenesis and suggest that therapeutic intervention aimed at the inhibition of s-HB-EGF functions may be useful in cancer treatment.
Collapse
Affiliation(s)
- Pat P Ongusaha
- Cancer Biology Program, Hematology and Oncology Division, Beth Israel Deaconess Medical Center and Harvard Medical School, 4 Blackfan Circle, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Li W, Hamada Y, Nakashima E, Naruse K, Kamiya H, Akiyama N, Hirooka H, Takahashi N, Horiuchi S, Hotta N, Oiso Y, Nakamura J. Suppression of 3-deoxyglucosone and heparin-binding epidermal growth factor-like growth factor mRNA expression by an aldose reductase inhibitor in rat vascular smooth muscle cells. Biochem Biophys Res Commun 2004; 314:370-6. [PMID: 14733914 DOI: 10.1016/j.bbrc.2003.12.095] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Reactive carbonyl compounds and oxidative stress have been recently shown to up-regulate the expression of heparin-binding epidermal growth factor-like growth factor (HB-EGF), a potent mitogen for vascular smooth muscle cells (SMCs) produced by SMC themselves. Because the polyol pathway has been reported to influence the formation of carbonyl compounds and the oxidative stress in various cells, we conducted this study to investigate whether the polyol pathway affects HB-EGF expression along with the generation of carbonyl compounds and the oxidative stress in SMCs. We found that, compared with those cultured with 5.5mM glucose, SMCs cultured with 40 mM glucose showed the accelerated thymidine incorporation, elevated levels of intracellular sorbitol, 3-deoxyglucosone (3-DG), advanced glycation end products (AGEs), and thiobarbituric acid-reactive substances (TBARS) along with the enhanced expression of HB-EGF mRNA. An aldose reductase inhibitor (ARI), SNK-860, significantly inhibited all of these abnormalities, while aminoguanidine suppressed 3-DG levels and HB-EGF mRNA expression independent of sorbitol levels. The results suggest that the polyol pathway may play a substantial role in SMC hyperplasia under hyperglycemic condition in part by affecting HB-EGF mRNA expression via the production of carbonyl compounds and oxidative stress.
Collapse
Affiliation(s)
- Weiguo Li
- Division of Metabolic Diseases, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Liu D, Jia H, Holmes DIR, Stannard A, Zachary I. Vascular endothelial growth factor-regulated gene expression in endothelial cells: KDR-mediated induction of Egr3 and the related nuclear receptors Nur77, Nurr1, and Nor1. Arterioscler Thromb Vasc Biol 2003; 23:2002-7. [PMID: 14525795 DOI: 10.1161/01.atv.0000098644.03153.6f] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The program of gene expression regulated by vascular endothelial growth factor (VEGF) remains poorly understood. The aim of this study was to identify VEGF-regulated genes in human umbilical vein endothelial cells. METHODS AND RESULTS VEGF-regulated gene expression was analyzed by screening Affymetrix oligonucleotide arrays and quantitative, real-time, reverse transcription-polymerase chain reaction. The most strongly induced genes were the NR4A nuclear receptor family members Nur77, Nurr1, and Nor1 and the zinc-finger transcription factor Egr3. VEGF also induced rapid expression of Down syndrome candidate region 1, cyclooxygenase-2, tissue factor, stanniocalcin-1, the serine/threonine kinase Cot, and eps15 homology domain-containing protein. VEGF-induced NR4A family and Egr3 expression was blocked by a KDR inhibitor, and placental growth factor and basic fibroblast growth factor weakly increased expression of these genes. Induction of NR4A genes was mediated via intracellular Ca2+, protein kinase C- and calcineurin-dependent pathways. VEGF increased protein expression of Nurr1 and Nur77 and decreased Nur77 phosphorylation at the negative regulatory site serine 351. CONCLUSIONS VEGF induces expression of NR4A nuclear receptors and Egr3 via KDR and KDR-mediated signaling mechanisms. The genes identified here are novel candidates as key early mediators of VEGF-induced endothelial functions.
Collapse
Affiliation(s)
- Dan Liu
- BHF Laboratories, Department of Medicine, University College London, London, UK
| | | | | | | | | |
Collapse
|
39
|
Iivanainen E, Nelimarkka L, Elenius V, Heikkinen SM, Junttila TT, Sihombing L, Sundvall M, Maatta JA, Laine VJO, Yla-Herttuala S, Higashiyama S, Alitalo K, Elenius K. Angiopoietin-regulated recruitment of vascular smooth muscle cells by endothelial-derived heparin binding EGF-like growth factor. FASEB J 2003; 17:1609-21. [PMID: 12958167 DOI: 10.1096/fj.02-0939com] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Recruitment of vascular smooth muscle cells (SMC) by endothelial cells (EC) is essential for angiogenesis. Endothelial-derived heparin binding EGF-like growth factor (HB-EGF) was shown to mediate this process by signaling via ErbB1 and ErbB2 receptors in SMCs. 1) Analysis of ErbB-ligands demonstrated that primary ECs expressed only HB-EGF and neuregulin-1. 2) Primary SMCs expressed ErbB1 and ErbB2, but not ErbB3 or ErbB4. 3) Consistent with their known receptor specificities, recombinant HB-EGF, but not neuregulin-1, stimulated tyrosine phosphorylation of ErbB1 and ErbB2 and migration in SMCs. 4) Neutralization of HB-EGF or inhibition of ErbB1 or ErbB2 blocked 70-90% of the potential of ECs to stimulate SMC migration. Moreover, 5) angiopoietin-1, an EC effector with a role in recruitment of SMC-like cells to vascular structures in vivo, enhanced EC-stimulated SMC migration by a mechanism involving up-regulation of endothelial HB-EGF. Finally, 6) immunohistochemical analysis of developing human tissues demonstrated that HB-EGF was expressed in vivo in ECs associated with SMCs or pericytes but not in ECs of the hyaloid vessels not associated with SMCs. These results suggest an important role for HB-EGF and ErbB receptors in the recruitment of SMCs by ECs and elaborate on the mechanism by which angiopoietins exert their vascular effects.
Collapse
MESH Headings
- Angiogenesis Inducing Agents/pharmacology
- Angiopoietin-1
- Cell Movement
- Cells, Cultured
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiology
- Epidermal Growth Factor/genetics
- Epidermal Growth Factor/pharmacology
- Epidermal Growth Factor/physiology
- ErbB Receptors/metabolism
- ErbB Receptors/physiology
- Heparin-binding EGF-like Growth Factor
- Humans
- Intercellular Signaling Peptides and Proteins
- Membrane Glycoproteins/pharmacology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Paracrine Communication
- Phosphorylation
- RNA, Messenger/biosynthesis
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/physiology
Collapse
Affiliation(s)
- Erika Iivanainen
- Medicity Research Laboratories, Department of Medical Biochemistry and Molecular Biology, University of Turku, FIN-20520 Turku, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Pang Y, Lineaweaver WC, Lei MP, Oswald T, Shamburger S, Cai Z, Zhang F. Evaluation of the mechanism of vascular endothelial growth factor improvement of ischemic flap survival in rats. Plast Reconstr Surg 2003; 112:556-64. [PMID: 12900614 DOI: 10.1097/01.prs.0000070965.28811.2c] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This study evaluated the effects of exogenous vascular endothelial growth factor (VEGF) on the regulation of cytokines in a rat dorsal ischemic skin flap model. Exogenous VEGF (1 microg/ml) was injected subdermally into the flaps of 12 rats before the flaps were sutured back in place. Another 12 rats with flaps received saline injections, as a control group. Biopsy specimens were obtained from the flaps treated with VEGF or saline solution, at positions 2.5, 5.5, and 8.5 cm from the distal edge of the flaps, at 12 hours (n = 6 for each group) and 24 hours (n = 6 for each group) after suturing of the flaps. Expression of cytokine, growth factor, and inducible nitric oxide synthase was measured. The results demonstrated that expression of tumor necrosis factor-alpha and nitric oxide synthase in the distal part of the VEGF-treated flaps was significantly decreased, compared with the control values, at 12 and 24 hours postoperatively. It was concluded that administration of exogenous VEGF could protect flaps from ischemia-reperfusion injury through the regulation of proinflammatory cytokines and the inhibition of cytotoxic nitric oxide production.
Collapse
Affiliation(s)
- Yi Pang
- Department of Pediatrics, Division of Plasic Surgery, University of Mississippoi Medical Center, Jackson, 39216, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Jackson LF, Qiu TH, Sunnarborg SW, Chang A, Zhang C, Patterson C, Lee DC. Defective valvulogenesis in HB-EGF and TACE-null mice is associated with aberrant BMP signaling. EMBO J 2003; 22:2704-16. [PMID: 12773386 PMCID: PMC156761 DOI: 10.1093/emboj/cdg264] [Citation(s) in RCA: 314] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Heparin-binding epidermal growth factor (HB-EGF) and betacellulin (BTC) are activating ligands for EGF receptor (EGFR/ErbB1) and ErbB4. To identify their physiological functions, we disrupted mouse HB-EGF and BTC alleles by homologous recombination. Most HB-EGF(-/-) mice died before weaning, and survivors had enlarged, dysfunctional hearts and reduced lifespans. Although BTC(-/-) mice were viable and fertile and displayed no overt defects, the lifespan of double null HB-EGF(-/-)/BTC(-/-) mice was further reduced, apparently due to accelerated heart failure. HB-EGF(-/-) newborns had enlarged and malformed semilunar and atrioventricular heart valves, and hypoplastic, poorly differentiated lungs. Defective cardiac valvulogenesis was the result of abnormal mesenchymal cell proliferation during remodeling, and was associated with dramatic increases in activated Smad1/5/8. Consistent with the phenotype, HB-EGF transcripts were localized to endocardial cells lining the margins of wild-type valves. Similarly defective valvulogenesis was observed in newborn mice lacking EGFR and tumor necrosis factor-alpha converting enzyme (TACE). These results suggest that cardiac valvulogenesis is dependent on EGFR activation by TACE-derived soluble HB-EGF, and that EGFR signaling is required to regulate bone morphogenetic protein signaling in this context.
Collapse
Affiliation(s)
- Leslie F Jackson
- Department of Biochemistry & Biophysics, UNC Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
CHOBOTOVA KATYA, MUCHMORE MARYELIZABETH, CARVER JANET, YOO HYUNGJ, MANEK SANJIV, GULLICK WILLIAMJ, BARLOW DAVIDH, MARDON HELENJ. The mitogenic potential of heparin-binding epidermal growth factor in the human endometrium is mediated by the epidermal growth factor receptor and is modulated by tumor necrosis factor-alpha. J Clin Endocrinol Metab 2002; 87:5769-77. [PMID: 12466384 PMCID: PMC1635788 DOI: 10.1210/jc.2002-020069] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Heparin-binding epidermal growth factor (HB-EGF), a member of the epidermal growth factor (EGF) family, is implicated in a variety of biological processes, including reproduction. Previous studies describe increased levels of HB-EGF in the human endometrium during the midsecretory stage of the menstrual cycle, suggesting a function for HB-EGF in implantation of the human blastocyst. Here we have investigated the expression and function of the soluble and transmembrane forms of HB-EGF in the human endometrium. We show that the expression of the transmembrane form of HB-EGF in the human endometrium is modulated according to the stage of the menstrual cycle. We present data demonstrating that both the soluble and transmembrane forms of HB-EGF induce DNA synthesis in human endometrial stromal cells. Furthermore, TNFalpha has a cooperative effect on HB-EGF, EGF, TGFalpha, and betacellulin-induced DNA synthesis in stromal cells, suggesting roles for the EGF family and TNFalpha in regeneration and maturation of human endometrium. Induction of DNA synthesis by HB-EGF and its modulation by TNFalpha in endometrial stromal cells are mediated by the EGF receptor and not the HB-EGF receptor ErbB4. Our data suggest key functions for HB-EGF, TNFalpha, and the EGF receptor in endometrial maturation, via autocrine/paracrine and juxtacrine pathways, in preparation for embryo implantation.
Collapse
Key Words
- btc, betacellulin
- cho, chinese hamster ovary
- egf, epidermal growth factor
- egfr, epidermal growth factor receptor
- fcs, fetal calf serum
- fitc, fluorescein isothiocyanate
- hb-egf, heparin-binding epidermal growth factor
- hrp, horseradish peroxidase
- pmsf, phenylmethylsulfonylfluoride
- sol-, soluble form
- tm-, transmembrane form
Collapse
Affiliation(s)
- KATYA CHOBOTOVA
- Nuffield Department of Obstetrics and Gynecology, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom OX3 9DU; and Research School of Biosciences, University of Kent (W.J.G.), Canterbury, Kent, United Kingdom CT2 7NJ
| | - MARY-ELIZABETH MUCHMORE
- Nuffield Department of Obstetrics and Gynecology, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom OX3 9DU; and Research School of Biosciences, University of Kent (W.J.G.), Canterbury, Kent, United Kingdom CT2 7NJ
| | - JANET CARVER
- Nuffield Department of Obstetrics and Gynecology, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom OX3 9DU; and Research School of Biosciences, University of Kent (W.J.G.), Canterbury, Kent, United Kingdom CT2 7NJ
| | - HYUNG-J YOO
- Nuffield Department of Obstetrics and Gynecology, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom OX3 9DU; and Research School of Biosciences, University of Kent (W.J.G.), Canterbury, Kent, United Kingdom CT2 7NJ
| | - SANJIV MANEK
- Nuffield Department of Obstetrics and Gynecology, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom OX3 9DU; and Research School of Biosciences, University of Kent (W.J.G.), Canterbury, Kent, United Kingdom CT2 7NJ
| | - WILLIAM J. GULLICK
- Nuffield Department of Obstetrics and Gynecology, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom OX3 9DU; and Research School of Biosciences, University of Kent (W.J.G.), Canterbury, Kent, United Kingdom CT2 7NJ
| | - DAVID H. BARLOW
- Nuffield Department of Obstetrics and Gynecology, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom OX3 9DU; and Research School of Biosciences, University of Kent (W.J.G.), Canterbury, Kent, United Kingdom CT2 7NJ
| | - HELEN J. MARDON
- Nuffield Department of Obstetrics and Gynecology, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom OX3 9DU; and Research School of Biosciences, University of Kent (W.J.G.), Canterbury, Kent, United Kingdom CT2 7NJ
| |
Collapse
|
44
|
O-Charoenrat P, Rhys-Evans P, Eccles S. A synthetic matrix metalloproteinase inhibitor prevents squamous carcinoma cell proliferation by interfering with epidermal growth factor receptor autocrine loops. Int J Cancer 2002; 100:527-33. [PMID: 12124801 DOI: 10.1002/ijc.10531] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is characterized by its capacity to invade adjacent tissues and to metastasize locoregionally. Evidence suggests that matrix metalloproteinases (MMPs) may play a causal role in HNSCC progression. While evaluating the role of MMPs in the invasion process, we made the surprising observation that a broad-spectrum MMP inhibitor, (marimastat, BB2516), inhibited the growth in vitro of some HNSCC cell lines. This inhibitory effect was only found in HNSCC cell lines overexpressing epidermal growth factor receptors. The effects of the MMP inhibitor could be reversed by adding exogenous c-erbB ligands, suggesting that the phenomenon may be related to autocrine ligand processing. This hypothesis was supported by the finding that the growth-inhibitory effect of marimastat was directly related to its ability to prevent the release of major c-erbB ligands including transforming growth factor-alpha, betacellulin and heregulin beta1 from HNSCC. Marimastat was also found to potentiate the cytotoxic effects of cisplatin both in vitro and in vivo. Our results indicate that the cleavage of several c-erbB ligands from membrane-anchored precursors requires MMP activity. We conclude that MMP inhibitors could prevent tumor progression not only by inhibiting invasion and angiogenesis, as previously shown, but also by their ability to inhibit autocrine signaling through the c-erbB receptors. Clinical trials to test this hypothesis in HNSCC should be considered.
Collapse
Affiliation(s)
- Pornchai O-Charoenrat
- Division of Head and Neck Surgery, Department of Surgery, Siriraj Hospital Medical School, Bangkok, Thailand.
| | | | | |
Collapse
|
45
|
Frerich B, Lindemann N, Kurtz-Hoffmann J, Oertel K. In vitro model of a vascular stroma for the engineering of vascularized tissues. Int J Oral Maxillofac Surg 2001; 30:414-20. [PMID: 11720044 DOI: 10.1054/ijom.2001.0130] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A major problem for the in vitro engineering of larger tissue equivalents like those required in reconstructive surgery is the lack of solutions for sufficient nutrition and oxygenation. The starting point of our investigation was the question of whether the principles of in vitro angiogenesis can be applied and utilized for tissue engineering. A soft tissue model was developed, consisting of human adipose tissue stromal cells and umbilical vein endothelial cells in a fibrin-microcarrier scaffold. Capillary-like structures were visualized using UEA-I-lectin labelling and confocal laser scanning microscopy. Length of capillary-like structures was measured in an image analysis system. Under serum-free culture conditions, maintenance of capillary-like structures was significantly increased in comparison to serum-containing cultures. The application of vascular endothelial growth factor (VEGF) resulted in a high initial angiogenic response; long-term stabilization of capillary-like structures could not be achieved, however supplementation with IGF-1 resulted in the highest values and the slightest decrease in length of capillary-like structures, so that the results could be interpreted as an improved stabilization.
Collapse
Affiliation(s)
- B Frerich
- Department of Oral and Maxillofacial Surgery and Plastic Facial Surgery, University of Leipzig, Germany.
| | | | | | | |
Collapse
|
46
|
O-charoenrat P, Rhys-Evans P, Eccles SA. Expression of vascular endothelial growth factor family members in head and neck squamous cell carcinoma correlates with lymph node metastasis. Cancer 2001; 92:556-68. [PMID: 11505400 DOI: 10.1002/1097-0142(20010801)92:3<556::aid-cncr1355>3.0.co;2-q] [Citation(s) in RCA: 150] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The expression of vascular endothelial growth factor (VEGF)-A isoforms (121, 165, 189, 206), VEGF-B, VEGF-C and VEGF-D in both experimental and clinical models of head and neck squamous cell carcinoma (HNSCC) was determined and correlated with conventional clinicopathologic parameters, with particular reference to cervical nodal metastasis. METHODS The mRNA expression of VEGFs in 14 HNSCC cell lines was compared with 4 normal keratinocyte cultures and 10 fibroblast cultures using a semiquantitative reverse transcription polymerase chain reaction (RT-PCR) assay. Protein levels were determined by Western blotting and enzyme-linked immunosorbent assay (ELISA). The authors then examined the expression of VEGFs in tissues from 54 patients including histologically normal epithelium (n = 32), early invasive squamous cell carcinomas (SCCs) (n = 23), advanced primary SCCs (n = 31), and lymph node metastases (n = 27). RESULTS Increased levels of VEGF-A (all four isoforms) and VEGF-C were found in tumor cell lines compared with normal cells, whereas no differences in VEGF-B levels were found. VEGF-D expression, however, was lower in HNSCC cells. Studies in clinical samples showed highly significant increases in mRNA expression of all four isoforms of VEGF-A and VEGF-C in tumors versus normal epithelium. In contrast, the levels of VEGF-D were significantly decreased in tumors, and VEGF-B expression appeared similar in both normal and malignant tissues. Multivariate analysis demonstrated that an infiltrative mode of invasion and enhanced expression of VEGF-A (isoforms 121 and 165) and VEGF-C had predictive value for the presence of cervical nodal metastases. CONCLUSIONS Up-regulation of VEGF-A (two isoforms) and VEGF-C and down-regulation of VEGF-D have been common features in HNSCC. Thus VEGF-A and VEGF-C appeared to play a vital role in the metastatic process of HNSCC.
Collapse
Affiliation(s)
- P O-charoenrat
- Department of Head and Neck Surgery, Royal Marsden Hospital, London, SW3 6JJ, United Kingdom.
| | | | | |
Collapse
|
47
|
Koh YH, Suzuki K, Che W, Park YS, Miyamoto Y, Higashiyama S, Taniguchi N. Inactivation of glutathione peroxidase by NO leads to the accumulation of H2O2 and the induction of HB-EGF via c-Jun NH2-terminal kinase in rat aortic smooth muscle cells. FASEB J 2001; 15:1472-4. [PMID: 11387261 DOI: 10.1096/fj.00-0572fje] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Y H Koh
- Department of Biochemistry, Osaka University Medical School, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
48
|
Nguyen LL, D'Amore PA. Cellular interactions in vascular growth and differentiation. INTERNATIONAL REVIEW OF CYTOLOGY 2001; 204:1-48. [PMID: 11243594 DOI: 10.1016/s0074-7696(01)04002-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In nature, mammalian cells do not exist in isolation, but rather are involved in interactions with other cells and matrix. In this review, several aspects of cellular interactions that are important in vascular growth and development will be highlighted. The cardiovascular system is the earliest to develop in the embryo. A number of growth factors and their receptors mediate the complex stages of migration, assembly, organization, and stabilization of developing vessels. In the adult organism, normal angiogenesis is restricted primarily to tissue growth (such as muscle and fat), the wound healing process and the female reproductive system. However, pathological angiogenesis, such as with tumor growth, diabetic retinopathy, and arthritis, is of great concern. The identification and/or development of exogenous and endogenous angiogenesis inhibitors has added to the understanding of these pathological processes. In addition to cellular interactions via ligands and receptors, cells also interact directly through physical contacts. These interactions facilitate anchorage, communication, and permeability. Since vessels serve as non-leaky conduits for blood flow as well as interfaces for molecular diffusion, the physical interactions between the cells that make up vessels must be specific for the function at hand. Permeability is a specialized function of vessels and is mediated by intracellular mechanisms and intercellular interactions. Cells also interact with the surrounding extracellular matrix. Integrin-matrix interaction is a two-way exchange critical for angiogenesis. Matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases play major roles in embryonic remodeling, adult injury, and pathological conditions. Several experimental model systems have been useful in our understanding of cellular interactions. These in vitro models incorporate heterotypic cell-cell interactions and/or allow cell-matrix interactions to occur.
Collapse
Affiliation(s)
- L L Nguyen
- Schepens Eye Research Institute and Department of Surgery, Children's Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | |
Collapse
|
49
|
Sundberg C, Nagy JA, Brown LF, Feng D, Eckelhoefer IA, Manseau EJ, Dvorak AM, Dvorak HF. Glomeruloid microvascular proliferation follows adenoviral vascular permeability factor/vascular endothelial growth factor-164 gene delivery. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 158:1145-60. [PMID: 11238063 PMCID: PMC1850349 DOI: 10.1016/s0002-9440(10)64062-x] [Citation(s) in RCA: 161] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Glomeruloid bodies are a defining histological feature of glioblastoma multiforme and some other tumors and vascular malformations. Little is known about their pathogenesis. We injected a nonreplicating adenoviral vector engineered to express vascular permeability factor/vascular endothelial growth factor-164 (VPF/VEGF(164)) into the ears of athymic mice. This vector infected local cells that strongly expressed VPF/VEGF(164) mRNA for 10 to 14 days, after which expression gradually declined. Locally expressed VPF/VEGF(164) induced an early increase in microvascular permeability, leading within 24 hours to edema and deposition of extravascular fibrin; in addition, many pre-existing microvessels enlarged to form thin-walled, pericyte-poor, "mother" vessels. Glomeruloid body precursors were first detected at 3 days as focal accumulations of rapidly proliferating cells in the endothelial lining of mother vessels, immediately adjacent to cells expressing VPF/VEGF(164). Initially, glomeruloid bodies were comprised of endothelial cells but subsequently pericytes and macrophages also participated. As they enlarged by endothelial cell and pericyte proliferation, glomeruloid bodies severely compromised mother vessel lumens and blood flow. Subsequently, as VPF/VEGF(164) expression declined, glomeruloid bodies devolved throughout a period of weeks by apoptosis and reorganization into normal-appearing microvessels. These results provide the first animal model for inducing glomeruloid bodies and indicate that VPF/VEGF(164) is sufficient for their induction and necessary for their maintenance.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Apoptosis
- Capillary Permeability
- Cell Division
- Cytokines/biosynthesis
- Cytokines/genetics
- Endothelial Growth Factors/biosynthesis
- Endothelial Growth Factors/genetics
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Endothelium, Vascular/ultrastructure
- Female
- In Situ Hybridization
- Lymphokines/biosynthesis
- Lymphokines/genetics
- Mice
- Mice, Nude
- Models, Animal
- Neovascularization, Pathologic
- Protein Isoforms/biosynthesis
- Protein Isoforms/genetics
- RNA, Messenger/biosynthesis
- Receptor Protein-Tyrosine Kinases/biosynthesis
- Receptor Protein-Tyrosine Kinases/genetics
- Receptors, Cytokine/biosynthesis
- Receptors, Cytokine/genetics
- Receptors, Growth Factor/biosynthesis
- Receptors, Growth Factor/genetics
- Receptors, Vascular Endothelial Growth Factor
- Time Factors
- Transcription, Genetic
- Transgenes
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- C Sundberg
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Xue L, Tassiopoulos AK, Woloson SK, Stanton DL, Ms CS, Hampton B, Burgess WH, Greisler HP. Construction and biological characterization of an HB-GAM/FGF-1 chimera for vascular tissue engineering. J Vasc Surg 2001; 33:554-60. [PMID: 11241127 DOI: 10.1067/mva.2001.112229] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Cardiovascular tissue engineering approaches to vessel wall restoration have focused on the potent but relatively nonspecific and heparin-dependent mesenchymal cell mitogen fibroblast growth factor 1 (FGF-1). We hypothesized that linking FGF-1 to a sequence likely to bind to cell surface receptors relatively more abundant on endothelial cells (ECs) might induce a relative greater EC bioavailability of the FGF-1. We constructed a heparin-binding growth-associated molecule (HB-GAM)/FGF-1 chimera by linking full-length human HB-GAM to the amino-terminus of human FGF-1beta (21-154) and tested its activities on smooth muscle cells (SMCs) and ECs. METHODS Primary canine carotid SMCs and jugular vein ECs were plated in 96-well plates in media containing 10% fetal bovine serum and grown to approximately 80% confluence. After being growth arrested in serum-free media for 24 hours, the cells were exposed to concentration ranges of cytokines and heparin, and proliferation was measured with tritiated-thymidine incorporation. Twenty percent fetal bovine serum was used as positive control, and phosphate-buffered saline was used as negative control. RESULTS In the presence of heparin the HB-GAM/FGF-1 chimera stimulated less SMC proliferation than did the wild-type FGF-1 with a median effective dose of approximately 0.3 nmol versus approximately 0.1 nmol (P <.001). By contrast, the chimera retained full stimulating activity on EC proliferation with a median effective dose of 0.06 nmol for both cytokines. Unlike the wild-type protein, the chimera possessed heparin-independent activity. In the absence of heparin, the chimera induced dose-dependent EC and SMC proliferation at 0.06 nmol or more compared with the wild-type FGF-1, which stimulated minimal DNA synthesis at 6.0-nmol concentrations. CONCLUSIONS The HB-GAM/FGF-1 chimera displays significantly greater and uniquely heparin-independent mitogenic activity for both cell types, and in the presence of heparin it displays a significantly greater EC specificity.
Collapse
Affiliation(s)
- L Xue
- Department of Surgery, Loyola University Medical Center, Maywood, IL 60153, USA
| | | | | | | | | | | | | | | |
Collapse
|