1
|
Mongiat M, Pascal G, Poletto E, Williams DM, Iozzo RV. Proteoglycans of basement membranes: Crucial controllers of angiogenesis, neurogenesis, and autophagy. PROTEOGLYCAN RESEARCH 2024; 2:e22. [PMID: 39184370 PMCID: PMC11340296 DOI: 10.1002/pgr2.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/02/2024] [Indexed: 08/27/2024]
Abstract
Anti-angiogenic therapy is an established method for the treatment of several cancers and vascular-related diseases. Most of the agents employed target the vascular endothelial growth factor A, the major cytokine stimulating angiogenesis. However, the efficacy of these treatments is limited by the onset of drug resistance. Therefore, it is of fundamental importance to better understand the mechanisms that regulate angiogenesis and the microenvironmental cues that play significant role and influence patient treatment and outcome. In this context, here we review the importance of the three basement membrane heparan sulfate proteoglycans (HSPGs), namely perlecan, agrin and collagen XVIII. These HSPGs are abundantly expressed in the vasculature and, due to their complex molecular architecture, they interact with multiple endothelial cell receptors, deeply affecting their function. Under normal conditions, these proteoglycans exert pro-angiogenic functions. However, in pathological conditions such as cancer and inflammation, extracellular matrix remodeling leads to the degradation of these large precursor molecules and the liberation of bioactive processed fragments displaying potent angiostatic activity. These unexpected functions have been demonstrated for the C-terminal fragments of perlecan and collagen XVIII, endorepellin and endostatin. These bioactive fragments can also induce autophagy in vascular endothelial cells which contributes to angiostasis. Overall, basement membrane proteoglycans deeply affect angiogenesis counterbalancing pro-angiogenic signals during tumor progression, and represent possible means to develop new prognostic biomarkers and novel therapeutic approaches for the treatment of solid tumors.
Collapse
Affiliation(s)
- Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Gabriel Pascal
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Evelina Poletto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Davion M. Williams
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Renato V. Iozzo
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
2
|
Petrenko O, Badziukh S, Korsa V, Kolosovych I, Tykhomyrov A. Topical Application of Autologous Plasma-Derived Plasminogen Accelerates Healing of Chronic Foot Ulcers in Type 2 Diabetes Patients. INT J LOW EXTR WOUND 2024:15347346241256025. [PMID: 38758187 DOI: 10.1177/15347346241256025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Plasminogen (Pg) is currently considered a master regulator of wound healing, but the molecular mechanisms of its efficacy in improving impaired closure of chronic skin ulcers in type 2 diabetes patients remain unclear. Here, we investigated wound healing effects of autologous plasma-derived Pg in diabetes patients with chronic foot ulcers and evaluated Pg-induced changes in levels of key protein markers related to wound repair. Type 2 diabetes patients with chronic wounds of lower extremities were included in the study and received topical applications of Pg in a dose of 1.0 mg/mL every 2 days during 20 days, in addition to the standard wound management treatment. Patients treated only according to conventional protocol served as a control. Wound closure rates were monitored by digital planimetry of wound areas. Plasminogen supplementary treatment significantly accelerated relative wound closure as compared with diabetes patients from the control group (24 ± 4 days vs 120 ± 17 days, respectively, P < .01). As shown by Western blot, Pg application reduced expression of protein regulators of hypoxia events, angiogenesis, and autophagy such as hypoxia-inducible factor-1α (by 6.3-folds, P < .01), angiostatins (by 2.5-folds, P < .05), and autophagy marker LC3-II/LC3-I (by 8.6-folds, P < .05), while increasing vascular endothelial growth factor level by 1.9-folds (P < .05). Gelatin zymography showed that Pg-supplemented therapy decreased activity of matrix metalloproteinase-9 (MMP-9) by 3.5-folds at the end of treatment period (P < .01). We report here for the first time that topically applied plasma-derived Pg has a pronounced beneficial effect in promoting foot ulcer healing in patients with type 2 diabetes through preventing hypoxia-induced signaling, reducing autophagy flux, diminishing excessive MMP activity, and enhancing angiogenesis.
Collapse
Affiliation(s)
- Oleg Petrenko
- Department of Surgery, Bogomolets National Medical University, Kyiv, Ukraine
| | - Sergiy Badziukh
- Department of Surgery, Bogomolets National Medical University, Kyiv, Ukraine
| | - Victoria Korsa
- Department of Enzyme Chemistry & Biochemistry, Palladin Institute of Biochemistry of NAS of Ukraine, Kyiv, Ukraine
| | - Ihor Kolosovych
- Department of Surgery, Bogomolets National Medical University, Kyiv, Ukraine
| | - Artem Tykhomyrov
- Department of Enzyme Chemistry & Biochemistry, Palladin Institute of Biochemistry of NAS of Ukraine, Kyiv, Ukraine
| |
Collapse
|
3
|
Lund N, Wieboldt H, Fischer L, Muschol N, Braun F, Huber T, Sorriento D, Iaccarino G, Müllerleile K, Tahir E, Adam G, Kirchhof P, Fabritz L, Patten M. Overexpression of VEGFα as a biomarker of endothelial dysfunction in aortic tissue of α-GAL-Tg/KO mice and its upregulation in the serum of patients with Fabry's disease. Front Cardiovasc Med 2024; 11:1355033. [PMID: 38374995 PMCID: PMC10875336 DOI: 10.3389/fcvm.2024.1355033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/17/2024] [Indexed: 02/21/2024] Open
Abstract
Introduction Fabry's disease is an X-linked lysosomal storage disorder caused by reduced activity of α-galactosidase A (GAL), leading to premature death on account of renal, cardiac, and vascular organ failure. Accumulation of the GAL substrate globotriaosylceramide (Gb3) in endothelial and smooth muscle cells is associated with early vascular cell damage, suggesting endothelial dysfunction as a driver of cardiorenal organ failure. Here, we studied the vascular expression of the key angiogenic factors, VEGFα and its antagonist angiostatin, in Fabry α-GAL-Tg/KO mice and determined circulating VEGFα and angiostatin serum levels in patients with Fabry's disease and healthy controls. Methods Cryopreserved aortic vessels from six α-GAL-Tg/KO and six wild-type (WT) mice were obtained and VEGFα and angiostatin levels were determined by performing Western blot analysis. VEGFα expression was visualized by an immunohistochemical staining of paraffin aortic rings. In addition, VEGFα and angiostatin serum levels were measured by using an enzyme-linked immunosorbent assay in 48 patients with genetically verified Fabry's disease (50% male) and 22 healthy controls and correlated with disease severity markers such as lyso-Gb3, albuminuria, NTproBNP, high-sensitive troponin T (hsTNT), and myocardial wall thickness. Results It was found that there was a significant increase in VEGFα protein expression (1.66 ± 0.35 vs. 0.62 ± 0.16, p = 0.0009) and a decrease in angiostatin expression (0.024 ± 0.007 vs. 0.053 ± 0.02, p = 0.038) in aortic lysates from α-GAL-Tg/KO compared with that from WT mice. Immunohistochemical staining revealed an adventitial VEGFα signal in α-GAL-Tg/KO mice, whereas no VEGFα signal could be detected in WT mice aortas. No differences in aortic angiostatin expression between α-GAL-Tg/KO- and WT mice could be visualized. The serum levels of VEGFα were significantly upregulated in patients with Fabry's disease compared with that in healthy controls (708.5 ± 426.3 vs. 458.5 ± 181.5 pg/ml, p = 0.048) and positively associated with albuminuria (r = 0.82, p < 0.0001) and elevated NTproBNP (r = 0.87, p < 0.0001) and hsTNT values (r = 0.41, p = 0.048) in male patients with Fabry's disease. For angiostatin, no significant difference was found between patients with Fabry's disease and healthy controls (747.6 ± 390.3 vs. 858.8 ± 599.3 pg/ml). Discussion In conclusion, an overexpression of VEGFα and downregulation of its counter player angiostatin in aortic tissue of α-GAL-Tg/KO mice support the hypothesis of an underlying vasculopathy in Fabry's disease. Elevated VEGFα serum levels were also observed in patients with Fabry's disease and were positively associated with elevated markers of organ manifestation in males. These findings suggest that angiogenetic markers, such as VEGFα, may be potentially useful biomarkers for the detection of endothelial dysfunction in classical Fabry's disease.
Collapse
Affiliation(s)
- N. Lund
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Intensive Care Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - H. Wieboldt
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - L. Fischer
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - N. Muschol
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - F. Braun
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - T. Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - D. Sorriento
- Department of Advanced Biomedical Sciences, Interdepartmental Center of Research on Hypertension and Related Conditions of the Federico II University, Naples, Italy
| | - G. Iaccarino
- Department of Clinical Medicine and Surgery, Interdepartmental Center of Research on Hypertension and Related Conditions of the Federico II University, Naples, Italy
| | - K. Müllerleile
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - E. Tahir
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - G. Adam
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - P. Kirchhof
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - L. Fabritz
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - M. Patten
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
4
|
Chen X, Wang Y, Liu H, Zhang J, Wang J, Jin X, Ma Y. CSP I-plus modified rEndostatin inhibits hepatocellular carcinoma metastasis via down-regulation of VEGFA and integrinβ1. BMC Cancer 2022; 22:1200. [PMID: 36419008 PMCID: PMC9682839 DOI: 10.1186/s12885-022-10318-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND In our previous study, N end of the Circumsporozoite protein (CSP I-plus) modified recombinant human Endostatin (rEndostatin, endostar) (rES-CSP) was constructed, which had antiangiogenic capability and bound to hepatocellular carcinoma in vivo and in vitro. In this study, the inhibition of rES-CSP on hepatocellular carcinoma metastasis was verified in vivo and in vitro, and its possible mechanism was explored. METHODS Firstly, the impact of rES-CSP on the migration, adhesion of hepatoma cell HCCLM3 was identified by wound healing, transwell, and on metastasis of orthotopic xenograft model was identified in nude mouse. Then the expression of metastasis-associated molecules (MMP2, E-cadherin, integrinβ1) and angiogenesis-related factors (VEGFA) in vitro and in vivo were detected by real-time PCR, western blotting, immunohistochemistry. RESULTS Finally, we found that rES-CSP could inhibit the migration and invasion of HCCLM3, and decrease tumor metastasis and growth in nude mouse orthotopic xenograft models. The tumor inhibiting rates of rES-CSP and Endostar were 42.46 ± 5.39% and 11.1 ± 1.88%. The lung metastasis rates of the control, Endostar and rES-CSP were 71, 50, and 42.8%, respectively. Compared with Endostar, rES-CSP significantly down-regulated the expression of VEGFA and integrinβ1. Heparin, a competitive inhibitor of CSP I-plus, which can be bind to the highly-sulfated heparan sulfate proteoglycans (HSPGs) over-expressed in liver and hepatocellular carcinoma, alleviated the down-regulation of VEGFA and integrinβ1. CONCLUSIONS These indicate that rES-CSP may play a role in inhibiting tumor growth and metastasis by down-regulating the angiogenic factor VEGF and the metastasis-related molecules or by interfering with HSPGs-mediated tumor metastasis.
Collapse
Affiliation(s)
- Xueqin Chen
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Higher Education Mega Center, Guangzhou, 510006 China
| | - Yan Wang
- grid.411847.f0000 0004 1804 4300Zhongshan Campus Laboratory Center, Guangdong Pharmaceutical University, Guangzhou, 510006 China
| | - Hancong Liu
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Higher Education Mega Center, Guangzhou, 510006 China
| | - Jingjing Zhang
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Higher Education Mega Center, Guangzhou, 510006 China
| | - Jie Wang
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Higher Education Mega Center, Guangzhou, 510006 China
| | - Xiaobao Jin
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Higher Education Mega Center, Guangzhou, 510006 China
| | - Yan Ma
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Higher Education Mega Center, Guangzhou, 510006 China
| |
Collapse
|
5
|
Araújo NM, Rubio IGS, Toneto NPA, Morale MG, Tamura RE. The use of adenoviral vectors in gene therapy and vaccine approaches. Genet Mol Biol 2022; 45:e20220079. [PMID: 36206378 PMCID: PMC9543183 DOI: 10.1590/1678-4685-gmb-2022-0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 07/12/2022] [Indexed: 11/04/2022] Open
Abstract
Adenovirus was first identified in the 1950s and since then this pathogenic group
of viruses has been explored and transformed into a genetic transfer vehicle.
Modification or deletion of few genes are necessary to transform it into a
conditionally or non-replicative vector, creating a versatile tool capable of
transducing different tissues and inducing high levels of transgene expression.
In the early years of vector development, the application in monogenic diseases
faced several hurdles, including short-term gene expression and even a fatality.
On the other hand, an adenoviral delivery strategy for treatment of cancer was
the first approved gene therapy product. There is an increasing interest in
expressing transgenes with therapeutic potential targeting the cancer hallmarks,
inhibiting metastasis, inducing cancer cell death or modulating the immune
system to attack the tumor cells. Replicative adenovirus as vaccines may be even
older and date to a few years of its discovery, application of non-replicative
adenovirus for vaccination against different microorganisms has been
investigated, but only recently, it demonstrated its full potential being one of
the leading vaccination tools for COVID-19. This is not a new vector nor a new
technology, but the result of decades of careful and intense work in this
field.
Collapse
Affiliation(s)
- Natália Meneses Araújo
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil.
| | - Ileana Gabriela Sanchez Rubio
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil. ,Universidade Federal de São Paulo, Laboratório de Ciências
Moleculares da Tireóide, Diadema, SP, Brazil.
| | | | - Mirian Galliote Morale
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil. ,Universidade Federal de São Paulo, Laboratório de Ciências
Moleculares da Tireóide, Diadema, SP, Brazil.
| | - Rodrigo Esaki Tamura
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil.
| |
Collapse
|
6
|
Chen CG, Iozzo RV. Extracellular matrix guidance of autophagy: a mechanism regulating cancer growth. Open Biol 2022; 12:210304. [PMID: 34982945 PMCID: PMC8727153 DOI: 10.1098/rsob.210304] [Citation(s) in RCA: 14] [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: 10/11/2021] [Accepted: 12/01/2021] [Indexed: 01/09/2023] Open
Abstract
The extracellular matrix (ECM) exists as a dynamic network of biophysical and biochemical factors that maintain tissue homeostasis. Given its sensitivity to changes in the intra- and extracellular space, the plasticity of the ECM can be pathological in driving disease through aberrant matrix remodelling. In particular, cancer uses the matrix for its proliferation, angiogenesis, cellular reprogramming and metastatic spread. An emerging field of matrix biology focuses on proteoglycans that regulate autophagy, an intracellular process that plays both critical and contextual roles in cancer. Here, we review the most prominent autophagic modulators from the matrix and the current understanding of the cellular pathways and signalling cascades that mechanistically drive their autophagic function. We then critically assess how their autophagic functions influence tumorigenesis, emphasizing the complexities and stage-dependent nature of this relationship in cancer. We highlight novel emerging data on immunoglobulin-containing and proline-rich receptor-1, heparanase and thrombospondin 1 in autophagy and cancer. Finally, we further discuss the pro- and anti-autophagic modulators originating from the ECM, as well as how these proteoglycans and other matrix constituents specifically influence cancer progression.
Collapse
Affiliation(s)
- Carolyn G. Chen
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
7
|
Li M, Popovic Z, Chu C, Krämer BK, Hocher B. Endostatin in Renal and Cardiovascular Diseases. KIDNEY DISEASES (BASEL, SWITZERLAND) 2021; 7:468-481. [PMID: 34901193 PMCID: PMC8613550 DOI: 10.1159/000518221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/29/2021] [Indexed: 04/21/2023]
Abstract
UNLABELLED Endostatin, a protein derived from the cleavage of collagen XVIII by the action of proteases, is an endogenous inhibitor known for its ability to inhibit proliferation and migration of endothelial cells, angiogenesis, and tumor growth. Angiogenesis is defined as the formation of new blood vessels from pre-existing vasculature, which is crucial in many physiological processes, such as embryogenesis, tissue regeneration, and neoplasia. SUMMARY Increasing evidence shows that dysregulation of angiogenesis is crucial for the pathogenesis of renal and cardiovascular diseases. Endostatin plays a pivotal role in the regulation of angiogenesis. Recent studies have provided evidence that circulating endostatin increases significantly in patients with kidney and heart failure and may also contribute to disease progression. KEY MESSAGE In the current review, we summarize the latest findings on preclinical and clinical studies analyzing the impact of endostatin on renal and cardiovascular diseases.
Collapse
Affiliation(s)
- Mei Li
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- *Berthold Hocher,
| | - Zoran Popovic
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Chang Chu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Nephrology, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Bernhard K. Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- European Center for Angioscience, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
- Center for Innate Immunoscience, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
- Institute of Medical Diagnostics, IMD Berlin, Berlin, Germany
| |
Collapse
|
8
|
Zhang Z, Liu X, Shen Z, Quan J, Lin C, Li X, Hu G. Endostatin in fibrosis and as a potential candidate of anti-fibrotic therapy. Drug Deliv 2021; 28:2051-2061. [PMID: 34595978 PMCID: PMC8491667 DOI: 10.1080/10717544.2021.1983071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Fibrotic diseases pose significant clinical challenges due to their broadness and complexity. Thus, a better understanding of fibrogenesis and the development of more effective treatments is imperative. Recent evidence suggests a significant antifibrotic potential of an endogenous glycoprotein, endostatin. While endostatin has been widely studied for its role as an anticancer adjuvant by inhibiting tumor angiogenesis, its possible implication in fibrosis remains largely unclear. Here, we review the role of endostatin in various cellular processes and highlight its antifibrotic activity. We hypothesize that endostatin conveys a homeostatic function in the process of fibrosis by regulating (a) TGF-β1 and its downstream signaling; (b) RhoA/ROCK pathway; (c) NF-κB signaling pathway; (d) expression of EGR-1; (e) PDGF/PDGFR pathway; (f) autophagy-related pathways; (g) pathways associated with cell proliferation and apoptosis. Finally, we propose a schematic model of the antifibrotic roles and mechanisms of endostatin; also, we outline future research directions of endostatin and aim to present a potential therapeutic approach for fibrosis.
Collapse
Affiliation(s)
- Zequn Zhang
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xi Liu
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaolong Shen
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun Quan
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Changwei Lin
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaorong Li
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Gui Hu
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
9
|
Chen CG, Iozzo RV. Angiostatic cues from the matrix: Endothelial cell autophagy meets hyaluronan biology. J Biol Chem 2020; 295:16797-16812. [PMID: 33020183 PMCID: PMC7864073 DOI: 10.1074/jbc.rev120.014391] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/02/2020] [Indexed: 01/21/2023] Open
Abstract
The extracellular matrix encompasses a reservoir of bioactive macromolecules that modulates a cornucopia of biological functions. A prominent body of work posits matrix constituents as master regulators of autophagy and angiogenesis and provides molecular insight into how these two processes are coordinated. Here, we review current understanding of the molecular mechanisms underlying hyaluronan and HAS2 regulation and the role of soluble proteoglycan in affecting autophagy and angiogenesis. Specifically, we assess the role of proteoglycan-evoked autophagy in regulating angiogenesis via the HAS2-hyaluronan axis and ATG9A, a novel HAS2 binding partner. We discuss extracellular hyaluronan biology and the post-transcriptional and post-translational modifications that regulate its main synthesizer, HAS2. We highlight the emerging group of proteoglycans that utilize outside-in signaling to modulate autophagy and angiogenesis in cancer microenvironments and thoroughly review the most up-to-date understanding of endorepellin signaling in vascular endothelia, providing insight into the temporal complexities involved.
Collapse
Affiliation(s)
- Carolyn G Chen
- Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Renato V Iozzo
- Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
10
|
Wu Y, Qin J, He J, Shen Y, Wang H, Li Y, Zeng Q, Dong J, An Y, Xiong S, Feng M, Wen F. Serum Endostatin Is a Novel Marker for COPD Associated with Lower Lung Function, Exacerbation and Systemic Inflammation. Int J Chron Obstruct Pulmon Dis 2020; 15:397-407. [PMID: 32158205 PMCID: PMC7049271 DOI: 10.2147/copd.s234760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/30/2019] [Indexed: 02/05/2023] Open
Abstract
Backgrounds and Aims It is well known that angiogenesis contributes to the progression of chronic obstructive pulmonary disease (COPD) by initiating the remodeling of bronchial vasculature. However, the specific molecular mechanisms are incompletely understood. This research aimed to explore whether endostatin, a member of endogenous antiangiogenic proteins, is a biomarker in COPD and plays a role in the angiogenesis of COPD. Methods 100 stable COPD patients, 130 patients with acute exacerbation (AECOPD) and 68 healthy volunteers were recruited in this research. Lung function test was conducted in the healthy people and stable COPD patients. Serum endostatin, C-reactive protein (CRP) and vascular endothelial growth factor (VEGF) of all the subjects were measured by Human Magnetic Luminex Screening Assay. Results Serum endostatin level was significantly higher in stable COPD compared with healthy control and even more in AECOPD patients (P<0.001). Besides, stable COPD patients with frequent exacerbation (≥2 exacerbations per year) in the last 1 year had a higher concentration of endostatin in the circulation compared to the patients with less exacerbation (P=0.037). Furthermore, circulatory endostatin was negatively associated with forced expiratory volume in 1 s % predicted (FEV1%pre), an index of lung function in the stable COPD group (P=0.009). Finally, endostatin was positively correlated to serum CRP in COPD group (including stable and AECOPD) (P=0.005) and all the subjects (P<0.001), but only associated with VEGF in the total participants (P=0.002), not in the COPD group. Conclusion These results suggested that endostatin is a biomarker for COPD and associated with lower lung function, exacerbation, and systemic inflammation. Endostatin potentially contributes to the pathogenesis of COPD.
Collapse
Affiliation(s)
- Yanqiu Wu
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jiangyue Qin
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Junyun He
- Department of Respiratory and Critical Care Medicine, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan, People's Republic of China
| | - Yongchun Shen
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hao Wang
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yanping Li
- Department of Respiratory and Critical Care Medicine, Third People's Hospital of Chengdu, Sichuan, People's Republic of China
| | - Qianglin Zeng
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, People's Republic of China
| | - Jiajia Dong
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yunfei An
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shuguang Xiong
- Department of Respiratory and Critical Care Medicine, 416 Hospital of Nuclear Industry, Chengdu, Sichuan, People's Republic of China
| | - Mei Feng
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Fuqiang Wen
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
11
|
Gubbiotti MA, Buraschi S, Kapoor A, Iozzo RV. Proteoglycan signaling in tumor angiogenesis and endothelial cell autophagy. Semin Cancer Biol 2019; 62:1-8. [PMID: 31078640 PMCID: PMC7864242 DOI: 10.1016/j.semcancer.2019.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/24/2022]
Abstract
The need for more effective cancer therapies is omnipresent as the ever-complex, and highly adaptive, mechanisms of tumor biology allow this disease to elude even the most stringent treatment options. The expanding field of proteoglycan signaling is enticing as a reservoir of potential drug targets and prospects for novel therapeutic strategies. The newest trend in proteoglycan biology is the interplay between extracellular signaling and autophagy fueled by the close link between autophagy and angiogenesis. Here we summarize the most current evidence surrounding proteoglycan signaling in both of these biological processes featuring the well-known suspects, decorin and perlecan, as well as other up-and-coming neophytes in this evolving signaling web.
Collapse
Affiliation(s)
- Maria A Gubbiotti
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Sidney Kimmel Medical College and Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Simone Buraschi
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Sidney Kimmel Medical College and Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Aastha Kapoor
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Sidney Kimmel Medical College and Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Sidney Kimmel Medical College and Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
12
|
Nyakas M, Aamdal E, Jacobsen KD, Guren TK, Aamdal S, Hagene KT, Brunsvig P, Yndestad A, Halvorsen B, Tasken KA, Aukrust P, Maelandsmo GM, Ueland T. Prognostic biomarkers for immunotherapy with ipilimumab in metastatic melanoma. Clin Exp Immunol 2019; 197:74-82. [PMID: 30821848 PMCID: PMC6591141 DOI: 10.1111/cei.13283] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2019] [Indexed: 12/23/2022] Open
Abstract
New therapies, including the anti‐cytotoxic T lymphocyte antigen (CTLA)‐4 antibody, ipilimumab, is approved for metastatic melanoma. Prognostic biomarkers need to be identified, because the treatment has serious side effects. Serum samples were obtained before and during treatment from 56 patients with metastatic or unresectable malignant melanoma, receiving treatment with ipilimumab in a national Phase IV study (NCT0268196). Expression of a panel of 17 inflammatory‐related markers reflecting different pathways including extracellular matrix remodeling and fibrosis, vascular inflammation and monocyte/macrophage activation were measured at baseline and the second and/or third course of treatment with ipilimumab. Six candidate proteins [endostatin, osteoprotegerin (OPG), C‐reactive protein (CRP), pulmonary and activation‐regulated chemokine (PARC), growth differentiation factor 15 (GDF15) and galectin‐3 binding‐protein (Gal3BP)] were persistently higher in non‐survivors. In particular, high Gal3BP and endostatin levels were also independently associated with poor 2‐year survival after adjusting for lactate dehydrogenase, M‐stage and number of organs affected. A 1 standard deviation increase in endostatin gave 1·74 times [95% confidence interval (CI) = 1·10–2·78, P = 0·019] and for Gal3BP 1·52 times (95% CI = 1·01–2·29, P = 0·047) higher risk of death in the adjusted model. Endostatin and Gal3BP may represent prognostic biomarkers for patients on ipilimumab treatment in metastatic melanoma and should be further evaluated. Owing to the non‐placebo design, we could only relate our findings to prognosis during ipilimumab treatment.
Collapse
Affiliation(s)
- M Nyakas
- Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - E Aamdal
- Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - K D Jacobsen
- Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - T K Guren
- Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - S Aamdal
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - K T Hagene
- Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - P Brunsvig
- Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - A Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,K.G. Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway
| | - B Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,K.G. Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway
| | - K A Tasken
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - P Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,K.G. Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway.,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| | - G M Maelandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - T Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| |
Collapse
|
13
|
Kanno Y. The Role of Fibrinolytic Regulators in Vascular Dysfunction of Systemic Sclerosis. Int J Mol Sci 2019; 20:ijms20030619. [PMID: 30709025 PMCID: PMC6387418 DOI: 10.3390/ijms20030619] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 02/08/2023] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease of autoimmune origin characterized by vascular dysfunction and extensive fibrosis of the skin and visceral organs. Vascular dysfunction is caused by endothelial cell (EC) apoptosis, defective angiogenesis, defective vasculogenesis, endothelial-to-mesenchymal transition (EndoMT), and coagulation abnormalities, and exacerbates the disease. Fibrinolytic regulators, such as plasminogen (Plg), plasmin, α2-antiplasmin (α2AP), tissue-type plasminogen activator (tPA), urokinase-type plasminogen activator (uPA) and its receptor (uPAR), plasminogen activator inhibitor 1 (PAI-1), and angiostatin, are considered to play an important role in the maintenance of endothelial homeostasis, and are associated with the endothelial dysfunction of SSc. This review considers the roles of fibrinolytic factors in vascular dysfunction of SSc.
Collapse
Affiliation(s)
- Yosuke Kanno
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's College of Liberal Arts, 97-1 Kodo Kyo-tanabe, Kyoto 610-0395, Japan.
| |
Collapse
|
14
|
Wu L, Wu Y, Xiong Z, Yao C, Zeng M, Zhang R, Hua Y. Effects and possible mechanism of a picosecond pulsed electric field on angiogenesis in cervical cancer in vitro. Oncol Lett 2019; 17:1517-1522. [PMID: 30675207 PMCID: PMC6341837 DOI: 10.3892/ol.2018.9782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 10/12/2018] [Indexed: 11/15/2022] Open
Abstract
Picosecond pulsed electric field (psPEF) is an athermal, minimally invasive and local ablative biomedical engineering technique used in cancer therapy. However, to the best of our knowledge, the effect of psPEF on angiogenesis in cervical cancer is unknown. Therefore, the aim of the current study was to investigate the effects and possible mechanism of psPEF on angiogenesis in cervical cancer in vitro. HeLa cell and human umbilical vein endothelial cell (HUVEC) suspensions were exposed to psPEF with an increasing gradient of electric field intensity (0, 200, 400 and 600 kV/cm). A Cell Counting kit-8 assay and flow cytometry were used to investigate the effect of psPEF on the proliferation and apoptosis of HUVECs. The invasion, migration and tube formation capabilities of HUVECs following psPEF treatment were investigated by Transwell invasion assay, scratch test and lumen formation assay, respectively. Changes in the protein and mRNA levels of angiogenesis-associated factors in HeLa cells were detected by western blot analysis and reverse transcription-quantitative polymerase chain reaction. psPEF was identified to inhibit proliferation and tube formation, and induce apoptosis and necrosis of HUVECs in a dose-dependent manner. psPEF was revealed to decrease the protein and mRNA expression levels of vascular endothelial growth factor and hypoxia-inducible factor 1α in HeLa cells. In summary, psPEF exhibited anti-angiogenic effects in cervical cancer in vitro by exerting direct effects on HUVECs and indirect effects on angiogenesis-associated factors in HeLa cells.
Collapse
Affiliation(s)
- Limei Wu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yutong Wu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Zhengai Xiong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Chenguo Yao
- State Key Laboratory of Power Transmission Equipment and System Security and New Technology, Chongqing University, Chongqing 400044, P.R. China
| | - Manman Zeng
- Department of Gynecology, Women and Children's Hospital of Guangdong, Guangzhou, Guangdong 511442, P.R. China
| | - Ruizhe Zhang
- Equipment Status Evaluation Center, Beijing Electric Power Research Institute, Beijing 100075, P.R. China
| | - Yuanyuan Hua
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
15
|
Chen Y, Mathy NW, Lu H. The role of VEGF in the diagnosis and treatment of malignant pleural effusion in patients with non‑small cell lung cancer (Review). Mol Med Rep 2018; 17:8019-8030. [PMID: 29693703 PMCID: PMC5983970 DOI: 10.3892/mmr.2018.8922] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/15/2018] [Indexed: 12/21/2022] Open
Abstract
Malignant pleural effusion (MPE) is a severe medical condition, which can result in breathlessness, pain, cachexia and reduced physical activity. It can occur in almost all types of malignant tumors; however, lung cancer is the most common cause of MPE, accounting for ~1/3 of clinical cases. Although there are numerous therapeutic approaches currently available for the treatment of MPE, none are fully effective and the majority can only alleviate the symptoms of the patients. Vascular endothelial growth factor (VEGF) has now been recognized as one of the most important regulatory factors in tumor angiogenesis, which participates in the entire process of tumor growth through its function to stimulate tumor angiogenesis, activate host vascular endothelial cells and promote malignant proliferation. Novel drugs targeting VEGF, including endostar and bevacizumab, have been developed and approved for the treatment of various tumors. Data from recent clinical studies have demonstrated that drugs targeting VEGF are effective and safe for the clinical management of MPE. Therefore, VEGF‑targeting represents a promising novel strategy for the diagnosis and treatment of MPE. The present review summarized recent advances in the role of VEGF in the pathogenesis, diagnosis and clinical management of MPE in patients with non‑small cell lung cancer.
Collapse
Affiliation(s)
- Yao Chen
- Department of Oncology, Jianghan University School of Medicine, Wuhan, Hubei 430056, P.R. China
| | | | - Hongda Lu
- Department of Oncology, Wuhan Central Hospital, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
16
|
Antitumor activities of Liver-targeting peptide modified Recombinant human Endostatin in BALB/c-nu mice with Hepatocellular carcinoma. Sci Rep 2017; 7:14074. [PMID: 29075040 PMCID: PMC5658401 DOI: 10.1038/s41598-017-14320-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/09/2017] [Indexed: 12/13/2022] Open
Abstract
In our previous study, a liver-targeting peptide CSP I-plus modified recombinant human Endostatin (rEndostatin, endostar) (rES-CSP) was constructed and showed potent antiangiogenic capability and could specifically bind to human hepatocellular carcinoma cells to make a direct inhibition in vitro. In this study, the biological activities of rES-CSP in vivo were evaluated by subcutaneous and orthotopic xenograft nude mice model of human hepatocellular carcinoma cells HepG2. We found that rES-CSP significantly decreased tumor volume to 54.9% in the nude mice with subcutaneous xenograft compared with the control. In orthotopic xenograft model, rES-CSP not only decreased tumor volume (to 39.6% compared with the control) and tumor weight, it also increased its biodistribution in the liver tissue and hepatoma tissue. Moreover, lower microvessel density (MVD) and higher apoptotic index (AI) were also observed in the tumor tissues. It had no significant side-effects on the heart, liver, spleen, lung and kidney of mice. Results indicated CSP I-plus modified Endostar may be a potential candidate for a targeting therapy on hepatocellular carcinoma.
Collapse
|
17
|
From Inflammation to Current and Alternative Therapies Involved in Wound Healing. Int J Inflam 2017; 2017:3406215. [PMID: 28811953 PMCID: PMC5547704 DOI: 10.1155/2017/3406215] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/01/2017] [Accepted: 06/06/2017] [Indexed: 02/08/2023] Open
Abstract
Wound healing is a complex event that develops in three overlapping phases: inflammatory, proliferative, and remodeling. These phases are distinct in function and histological characteristics. However, they depend on the interaction of cytokines, growth factors, chemokines, and chemical mediators from cells to perform regulatory events. In this article, we will review the pathway in the skin healing cascade, relating the major chemical inflammatory mediators, cellular and molecular, as well as demonstrating the local and systemic factors that interfere in healing and disorders associated with tissue repair deficiency. Finally, we will discuss the current therapeutic interventions in the wounds treatment, and the alternative therapies used as promising results in the development of new products with healing potential.
Collapse
|
18
|
Negative regulators of angiogenesis: important targets for treatment of exudative AMD. Clin Sci (Lond) 2017; 131:1763-1780. [PMID: 28679845 DOI: 10.1042/cs20170066] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/17/2017] [Accepted: 04/03/2017] [Indexed: 12/12/2022]
Abstract
Angiogenesis contributes to the pathogenesis of many diseases including exudative age-related macular degeneration (AMD). It is normally kept in check by a tightly balanced production of pro- and anti-angiogenic factors. The up-regulation of the pro-angiogenic factor, vascular endothelial growth factor (VEGF), is intimately linked to the pathogenesis of exudative AMD, and its antagonism has been effectively targeted for treatment. However, very little is known about potential changes in expression of anti-angiogenic factors and the role they play in choroidal vascular homeostasis and neovascularization associated with AMD. Here, we will discuss the important role of thrombospondins and pigment epithelium-derived factor, two major endogenous inhibitors of angiogenesis, in retinal and choroidal vascular homeostasis and their potential alterations during AMD and choroidal neovascularization (CNV). We will review the cell autonomous function of these proteins in retinal and choroidal vascular cells. We will also discuss the potential targeting of these molecules and use of their mimetic peptides for therapeutic development for exudative AMD.
Collapse
|
19
|
|
20
|
Li Q, Yuan DM, Ma LH, Ma CH, Liu YF, Lv TF, Song Y. Chloroquine inhibits tumor growth and angiogenesis in malignant pleural effusion. Tumour Biol 2016; 37:10.1007/s13277-016-5441-z. [PMID: 27771855 DOI: 10.1007/s13277-016-5441-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 09/23/2016] [Indexed: 12/28/2022] Open
Abstract
Malignant pleural effusion (MPE) is associated with a poor prognosis in lung cancer. Currently, no effective cure exists for MPE. Chloroquine (CQ) has been demonstrated to induce vascular normalization and inhibit tumor growth. The aim of this study was to assess whether CQ affects MPE. The xenografts mice were divided into normal saline (NS), CQ, or bevacizumab (BE) group. Tumor growth and microvascular density (MVD) were monitored. We explored the effect of CQ on the proliferation, survival, and proangiogenic signaling of tumor cells in vitro. We further evaluated the effects of CQ on the viability, migration, and tube formation of human umbilical vein endothelial cells (HUVECs). A chicken chorioallantoic membrane (CAM) model was used to elucidate the effects of CQ on angiogenesis. Finally, an MPE mouse model were treated by CQ, BE, or NS. The volume of pleural effusion, tumor foci, and MVD was evaluated. CQ therapy group exhibited decreased tumor volume, tumor weight, and MVD in the mouse xenografts. CQ inhibited the proliferation of the tumor cells. However, the expression of vascular endothelial growth factor was not affected. Additionally, CQ inhibited the proliferation, migration, and tube formation of HUVECs and also restrained angiogenesis in the CAM. Western blot showed that CQ might suppress angiogenesis by downregulating p-Akt, Jagged1, and Ang2 in HUVECs. In MPE mice, the volume of the pleural effusion, the number of pleural tumor foci, and the MVD were significantly reduced in the CQ group. Our work demonstrated that CQ played the role of an efficient treatment for MPE.
Collapse
Affiliation(s)
- Qian Li
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Dong-Mei Yuan
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Li-Hong Ma
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Chen-Hui Ma
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Ya-Fang Liu
- Department of Respiratory Medicine, Jinling Hospital, Southern Medical University (Guangzhou), Nanjing, China
| | - Tang-Feng Lv
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China.
- Department of Respiratory Medicine, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, Jiangsu Province, 210002, China.
| |
Collapse
|
21
|
Aydemir EA, Şimşek E, Korcum AF, Fişkin K. Endostatin and irradiation modifies the activity of ADAM10 and neprilysin in breast cancer cells. Mol Med Rep 2016; 14:2343-51. [PMID: 27430992 DOI: 10.3892/mmr.2016.5463] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 06/06/2016] [Indexed: 11/05/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels, is regarded as a key cancer cell property. Endostatin (ES) is a potential antiangiogenic agent and it may be useful when implemented in combination with other cancer therapeutic strategies. The present study investigated the in vitro effects of ES, radiotherapy (RT) or combination therapy (ES + RT) on two important proteases, a disintegrin and metalloproteinase domain‑containing protein 10 (ADAM10) and neprilysin (NEP) in 4T1 mouse breast cancer cells and the more metastatic phenotype of 4THMpc breast cancer cells. 4T1 and 4THMpc cells were treated with recombinant murine ES (4 µg/ml) alone, RT (45 Gy) alone or with ES + RT. ADAM10 enzyme activity was determined using a tumor necrosis factor‑α converting enzyme (α‑secretase) activity assay kit, and NEP enzyme activity was measured with a fluorometric assay based on the generation of free dansyl‑D‑Ala‑Gly from N-dansyl-Ala-Gly-D-nitro-Phe-Gly, the substrate of NEP. Western blotting analysis was performed to determine whether the altered enzyme activity levels of the two cell lines occurred due to changes in expression level. These data indicate that ES independently potentiates the activity of ADAM10 and NEP enzymes in 4T1 and 4THMpc breast cancer cells.
Collapse
Affiliation(s)
- Esra Arslan Aydemir
- Department of Biology, Science Faculty, Akdeniz University, Antalya 07058, Turkey
| | - Ece Şimşek
- Department of Nutrition and Dietetics, Antalya School of Health, Akdeniz University, Antalya 07058, Turkey
| | - Aylin Fidan Korcum
- Department of Radiation Oncology, School of Medicine, Akdeniz University, Antalya 07058, Turkey
| | - Kayahan Fişkin
- Department of Nutrition and Dietetics, Antalya School of Health, Akdeniz University, Antalya 07058, Turkey
| |
Collapse
|
22
|
The GA genotype of the −1154 G/A (rs1570360) vascular endothelial growth factor (VEGF) is protective against hypertension-related chronic kidney disease incidence. Mol Cell Biochem 2016; 418:159-65. [DOI: 10.1007/s11010-016-2741-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/15/2016] [Indexed: 12/15/2022]
|
23
|
Luo X, Andres ML, Timiryasova TM, Fodor I, Slater JM, Gridley DS. Radiation-enhanced Endostatin Gene Expression and Effects of Combination Treatment. Technol Cancer Res Treat 2016; 4:193-202. [PMID: 15773788 DOI: 10.1177/153303460500400208] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Targeting cells that support tumor growth by administering potent angiogenesis inhibitors is currently an area of intense interest. In the present study, a unique plasmid vector for the mouse endostatin gene, pXLG-mEndo, was constructed and evaluated with and without radiation using the Lewis lung carcinoma (LLC) cell line. The physical properties of the expressed endostatin protein were validated by PCR, gel electrophoresis, and Western blot. Enzyme-linked immunosorbent and immunocytochemical analyses for the therapeutic gene demonstrated that transfected LLC cells secreted the protein into the medium. Exposure of the cells to 2 gray (Gy) γ-rays reduced the time to reach the maximum expression level of the endostatin gene and also increased the amount of secreted endostatin protein (P<0.001). Biological activity of the endostatin was demonstrated by the inhibition of tube formation by human umbilical vein endothelial cells (HUVEC). Based on 3 H-thymidine incorporation, endostatin expression significantly depressed DNA synthesis in HUVEC and LLC cells compared to controls transfected with parental vector or no vector (P>0.005). In addition, radiation increased the efficiency of endostatin-mediated inhibition of both cell types over a 3-day period post-exposure (P<0.05 or less). Intratumoral injection of 100 μg pXLG-mEndo combined with 10 Gy radiation significantly delayed LLC tumor growth, especially when each modality was delivered twice (P<0.05 or less compared to all other groups). No toxicity was observed. These findings are very promising and suggest that endostatin therapy with a plasmid vector, such as pXLG-mEndo, may enhance the efficacy of radiotherapy for lung cancer.
Collapse
Affiliation(s)
- Xian Luo
- Department of Radiation Medicine, Chan Shun Pavilion, Room A-1010, 11175 Campus Street, Loma Linda University Medical Center, Loma Linda, CA 92354, USA
| | | | | | | | | | | |
Collapse
|
24
|
Poluzzi C, Iozzo RV, Schaefer L. Endostatin and endorepellin: A common route of action for similar angiostatic cancer avengers. Adv Drug Deliv Rev 2016; 97:156-73. [PMID: 26518982 DOI: 10.1016/j.addr.2015.10.012] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/15/2015] [Accepted: 10/16/2015] [Indexed: 12/17/2022]
Abstract
Traditional cancer therapy typically targets the tumor proper. However, newly-formed vasculature exerts a major role in cancer development and progression. Autophagy, as a biological mechanism for clearing damaged proteins and oxidative stress products released in the tumor milieu, could help in tumor resolution by rescuing cells undergoing modifications or inducing autophagic-cell death of tumor blood vessels. Cleaved fragments of extracellular matrix proteoglycans are emerging as key players in the modulation of angiogenesis and endothelial cell autophagy. An essential characteristic of cancer progression is the remodeling of the basement membrane and the release of processed forms of its constituents. Endostatin, generated from collagen XVIII, and endorepellin, the C-terminal segment of the large proteoglycan perlecan, possess a dual activity as modifiers of both angiogenesis and endothelial cell autophagy. Manipulation of these endogenously-processed forms, located in the basement membrane within tumors, could represent new therapeutic approaches for cancer eradication.
Collapse
Affiliation(s)
- Chiara Poluzzi
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany.
| |
Collapse
|
25
|
Toro Bejarano M, Merchan JR. Targeting tumor vasculature through oncolytic virotherapy: recent advances. Oncolytic Virother 2015; 4:169-81. [PMID: 27512680 PMCID: PMC4918394 DOI: 10.2147/ov.s66045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The oncolytic virotherapy field has made significant advances in the last decade, with a rapidly increasing number of early- and late-stage clinical trials, some of them showing safety and promising therapeutic efficacy. Targeting tumor vasculature by oncolytic viruses (OVs) is an attractive strategy that offers several advantages over nontargeted viruses, including improved tumor viral entry, direct antivascular effects, and enhanced antitumor efficacy. Current understanding of the biological mechanisms of tumor neovascularization, novel vascular targets, and mechanisms of resistance has allowed the development of oncolytic viral vectors designed to target tumor neovessels. While some OVs (such as vaccinia and vesicular stomatitis virus) can intrinsically target tumor vasculature and induce vascular disruption, the majority of reported vascular-targeted viruses are the result of genetic manipulation of their viral genomes. Such strategies include transcriptional or transductional endothelial targeting, "armed" viruses able to downregulate angiogenic factors, or to express antiangiogenic molecules. The above strategies have shown preclinical safety and improved antitumor efficacy, either alone, or in combination with standard or targeted agents. This review focuses on the recent efforts toward the development of vascular-targeted OVs for cancer treatment and provides a translational/clinical perspective into the future development of new generation biological agents for human cancers.
Collapse
Affiliation(s)
- Marcela Toro Bejarano
- Division of Hematology-Oncology, Department of Medicine, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Jaime R Merchan
- Division of Hematology-Oncology, Department of Medicine, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| |
Collapse
|
26
|
Behl T, Kotwani A. Possible role of endostatin in the antiangiogenic therapy of diabetic retinopathy. Life Sci 2015; 135:131-7. [DOI: 10.1016/j.lfs.2015.06.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 05/15/2015] [Accepted: 06/03/2015] [Indexed: 02/03/2023]
|
27
|
Zhao P, Luo R, Wu J, Xie F, Li H, Xiao X, Fu L, Zhu X, Liu R, Zhu Y, Liang Z, Huang W. E10A, an adenovirus carrying human endostatin gene, in combination with docetaxel treatment inhibits prostate cancer growth and metastases. J Cell Mol Med 2015; 14:381-91. [PMID: 26065034 PMCID: PMC3837610 DOI: 10.1111/j.1582-4934.2008.00548.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
E10A, a replication-defective adenovirus carrying human endostatin gene, has finished Phase I clinical trials for solid cancers. We assessed whether the combination of E10A with docetaxel would enhance antiangiogenic activities and inhibit prostate cancer growth and metastases. Combination use of conditioned medium from prostate cancer cells infected by E10A and docetaxel exerted synergistic inhibition of HUVECs proliferation, migration and tube formation, compared with either agent alone. In prostate cancer s.c. xenograft models, combined therapy resulted in significant tumor growth inhibition and survival improvement. The antitumoral effect was tightly correlated with a remarkable decrease in tumor cell proliferation, microvessel, especially immature vasculature and significant increase in apoptosis induction. Systemic administration of E10A and docetaxel also effectively inhibited orthotopic growth and metastases of prostate cancer and achieved better in vivo antiangiogenic effects than either agent alone. Our data indicate that E10A in combination with docetaxel exert enhanced antiangiogenic activities and inhibit prostate cancer growth and metastases. Therefore, this approach may be an effective treatment for advanced prostate cancer and deserves more extensive investigation.
Collapse
Affiliation(s)
- Peng Zhao
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Yadav L, Puri N, Rastogi V, Satpute P, Sharma V. Tumour Angiogenesis and Angiogenic Inhibitors: A Review. J Clin Diagn Res 2015; 9:XE01-XE05. [PMID: 26266204 PMCID: PMC4525594 DOI: 10.7860/jcdr/2015/12016.6135] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 04/24/2015] [Indexed: 02/06/2023]
Abstract
Angiogenesis is a complex process depending on the coordination of many regulators and there by activating angiogenic switch. Recent advances in understanding of angiogenic mechanism have lead to the development of several anti-angiogenic and anti-metastatic agents that use the strategy of regulation of angiogenic switch. Antiangiogenic therapy is a form of treatment not cure for cancer and represents a highly effective strategy for destroying tumour because vascular supply is the fundamental requirement for growth of tumour. Because of the quiescent nature of normal adult vasculature, angiogenic inhibitors are expected to confer a degree of specificity when compared to nonspecific modalities of chemo and radiotherapy, so it has the advantage of less toxicities, does not induce drug resistance and deliver a relatively non toxic, long term treatment of tumour.
Collapse
Affiliation(s)
- Lalita Yadav
- Senior Lecturer, Department of Oral & Maxillofacial Pathology, Kalka Dental College, Meerut Uttar Pradesh, India
| | - Naveen Puri
- Professor and HOD, Department of Oral Pathology, Kalka Dental College & Hospital, Meerut Uttar Pradesh, India
| | - Varun Rastogi
- Reader, Department of Oral Pathology, Kalka Dental College & Hospital, Meerut Uttar Pradesh, India
| | - Pranali Satpute
- Senior Lecturer, Department of Oral Pathology, Guardian College of Dental Sciences & Research Centre, Ambarnath (W), Thane Maharashtra, India
| | - Vandana Sharma
- Senior Lecturer, Department of Oral & Maxillofacial Pathology, Kalka Dental College, Meerut Uttar Pradesh, India
| |
Collapse
|
29
|
VEGF overexpression is a valuable prognostic factor for non-Hodgkin's lymphoma evidence from a systemic meta-analysis. DISEASE MARKERS 2015; 2015:786790. [PMID: 25810565 PMCID: PMC4355555 DOI: 10.1155/2015/786790] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 01/25/2015] [Accepted: 01/26/2015] [Indexed: 12/13/2022]
Abstract
Vascular endothelial growth factor (VEGF) plays a vital role in the progression of Non-Hodgkin's lymphoma (NHL). Although multiple studies have investigated the relationship between VEGF expression and prognosis of NHL, these studies have yielded conflicting results. Therefore, we performed a meta-analysis to evaluate the role of VEGF in the prognosis of NHL patients. We systematically searched eligible studies from databases and determined that there was a significant correlation between VEGF overexpression and overall survival (HR (hazard ratio) = 1.66, 95% CI: 1.25–2.22, P = 0.001). Based on subgroup analysis by study location, number of patients, the source of VEGF expression, and study design, we found that VEGF overexpression in surgically resected tissue (HR = 1.95, 95% CI: 1.41–2.69, P = 0.000), but not in serum (HR = 1.37, 95% CI: 0.96–1.95, P = 0.087), was associated with poorer prognosis. Additionally, VEGF overexpression did not correlate with performance status, LDH level, IPI score, tumor staging, B symptoms, or NHL relapse. In summary, overexpression of VEGF in lymphoma tissue represents a promising potential prognostic factor in NHL.
Collapse
|
30
|
Tsuji T, Kelly NJ, Takahashi S, Leme AS, Houghton AM, Shapiro SD. Macrophage elastase suppresses white adipose tissue expansion with cigarette smoking. Am J Respir Cell Mol Biol 2015; 51:822-9. [PMID: 24914890 DOI: 10.1165/rcmb.2014-0083oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Macrophage elastase (MMP12) is a key mediator of cigarette smoke (CS)-induced emphysema, yet its role in other smoking related pathologies remains unclear. The weight suppressing effects of smoking are a major hindrance to cessation efforts, and MMP12 is known to suppress the vascularization on which adipose tissue growth depends by catalyzing the formation of antiangiogenic peptides endostatin and angiostatin. The goal of this study was to determine the role of MMP12 in adipose tissue growth and smoking-related suppression of weight gain. Whole body weights and white adipose depots from wild-type and Mmp12-deficient mice were collected during early postnatal development and after chronic CS exposure. Adipose tissue specimens were analyzed for angiogenic and adipocytic markers and for content of the antiangiogenic peptides endostatin and angiostatin. Cultured 3T3-L1 adipocytes were treated with adipose tissue homogenate to examine its effects on vascular endothelial growth factor (VEGF) expression and secretion. MMP12 content and activity were increased in the adipose tissue of wild-type mice at 2 weeks of age, leading to elevated endostatin production, inhibition of VEGF secretion, and decreased adipose tissue vascularity. By 8 weeks of age, adipose MMP12 levels subsided, and the protein was no longer detectable. However, chronic CS exposure led to macrophage accumulation and restored adipose MMP12 activity, thereby suppressing adipose tissue mass and vascularity. Our results reveal a novel systemic role for MMP12 in postnatal adipose tissue expansion and smoking-associated weight loss by suppressing vascularity within the white adipose tissue depots.
Collapse
Affiliation(s)
- Takao Tsuji
- 1 Department of Medicine, UPMC and University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | | | | | | |
Collapse
|
31
|
Tsuji T, Yamaguchi K, Kikuchi R, Itoh M, Nakamura H, Nagai A, Aoshiba K. Promotion of adipogenesis by an EP2 receptor agonist via stimulation of angiogenesis in pulmonary emphysema. Prostaglandins Other Lipid Mediat 2014; 112:9-15. [PMID: 24911647 DOI: 10.1016/j.prostaglandins.2014.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 05/15/2014] [Accepted: 05/15/2014] [Indexed: 02/07/2023]
Abstract
Body weight loss is a common manifestation in patients with chronic obstructive pulmonary disease (COPD), particularly those with severe emphysema. Adipose angiogenesis is a key mediator of adipogenesis and use of pro-angiogenic agents may serve as a therapeutic option for lean COPD patients. Since angiogenesis is stimulated by PGE2, we examined whether ONO-AE1-259, a selective E-prostanoid (EP) 2 receptor agonist, might promote adipose angiogenesis and adipogenesis in a murine model of elastase-induced pulmonary emphysema (EIE mice). Mice were intratracheally instilled with elastase or saline, followed after 4 weeks by intraperitoneal administration of ONO-AE1-259 for 4 weeks. The subcutaneous adipose tissue (SAT) weight decreased in the EIE mice, whereas in the EIE mice treated with ONO-AE1-259, the SAT weight was largely restored, which was associated with significant increases in SAT adipogenesis, angiogenesis, and VEGF protein production. In contrast, ONO-AE1-259 administration induced no alteration in the weight of the visceral adipose tissue. These results suggest that in EIE mice, ONO-AE1-259 stimulated adipose angiogenesis possibly via VEGF production, and thence, adipogenesis. Our data pave the way for the development of therapeutic interventions for weight loss in emphysema patients, e.g., use of pro-angiogenic agents targeting the adipose tissue vascular component.
Collapse
Affiliation(s)
- Takao Tsuji
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami, Inashiki, Ibaraki 300-0395, Japan.
| | - Kazuhiro Yamaguchi
- Comprehensive and Internal Medicine, Tokyo Women's Medical University Medical Center East, 2-1-10 Nishi-ogu, Arakawa-ku 116-8567, Japan.
| | - Ryota Kikuchi
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami, Inashiki, Ibaraki 300-0395, Japan.
| | - Masayuki Itoh
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami, Inashiki, Ibaraki 300-0395, Japan.
| | - Hiroyuki Nakamura
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami, Inashiki, Ibaraki 300-0395, Japan.
| | - Atsushi Nagai
- The First Department of Medicine, Tokyo Women's Medical University, 8-1 Kawata-cho, Shinjyuku-ku 162-8666, Japan.
| | - Kazutetsu Aoshiba
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, 3-20-1 Chuou, Ami, Inashiki, Ibaraki 300-0395, Japan.
| |
Collapse
|
32
|
Plasmid transfer of plasminogen K1-5 reduces subcutaneous hepatoma growth by affecting inflammatory factors. BIOMED RESEARCH INTERNATIONAL 2014; 2014:656527. [PMID: 24895598 PMCID: PMC4034484 DOI: 10.1155/2014/656527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 04/09/2014] [Accepted: 04/10/2014] [Indexed: 11/18/2022]
Abstract
There is evidence that plasminogen K1-5 (PlgK1-5) directly affects tumour cells and inflammation. Therefore, we analysed if PlgK1-5 has immediate effects on hepatoma cells and inflammatory factors in vitro and in vivo. In vitro, effects of plasmid encoding PlgK1-5 (pK1-5) on Hepa129, Hepa1-6, and HuH7 cell viability, apoptosis, and proliferation as well as VEGF and TNF-alpha expression and STAT3-phosphorylation were investigated. In vivo, tumour growth, proliferation, vessel density, and effects on vascular endothelial growth factor (VEGF) and tumour necrosis factor alpha (TNF-alpha) expression were examined following treatment with pK1-5. In vivo, pK1-5 halved cell viability; cell death was increased by up to 15% compared to the corresponding controls. Proliferation was not affected. VEGF, TNF-alpha, and STAT3-phosphorylation were affected following treatment with pK1-5. In vivo, ten days after treatment initiation, pK1-5 reduced subcutaneous tumour growth by 32% and mitosis by up to 77% compared to the controls. Vessel density was reduced by 50%. TNF-alpha levels in tumour and liver tissue were increased, whereas VEGF levels in tumours and livers were reduced after pK1-5 treatment. Taken together, plasmid gene transfer of PlgK1-5 inhibits hepatoma (cell) growth not only by reducing vessel density but also by inducing apoptosis, inhibiting proliferation, and triggering inflammation.
Collapse
|
33
|
Qi X, Liu Y, Wei W, Huang X, Zuo Y. Effects of the C-terminal of endostatin on the tumorigenic potential of H22 cells. Biomed Rep 2014; 1:761-765. [PMID: 24649025 DOI: 10.3892/br.2013.147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 07/04/2013] [Indexed: 11/05/2022] Open
Abstract
Endostatin is an endogenous angiogenesis inhibitor whose specific functional site has not yet been determined. In the present experiment, 13 amino acids (LCIENSFMTSFSK) were selectively deleted from the C-terminal of endostatin and the resulting mutant endostatin was named EM13. To determine the effect of the C-terminal deletion on the biological activity of endostatin, EM13, wild-type endostatin and empty plasmid were transfected into H22 cells. After 48 h, the three types of transfected cells were harvested and injected into nude mice. The results demonstrated that there was no significant difference in tumor size, as determined by hematoxylin and eosin staining, between the EM13-transfected group and the endostatin and empty plasmid groups, although the nude mice that were injected with EM13-transfected H22 cells exhibited smaller tumors and lower density of blood vessels compared to those injected with endostatin- and empty plasmid-transfected H22 cells. The results suggested that the 13 amino acids of the C-terminal of endostatin do not play an important role in the tumorigenic potential of H22 cells. This experiment was unsuccessful in reproducing the results of several investigators. Therefore, the mechanism underlying the tumorigenesis of H22 cells remains to be elucidated.
Collapse
Affiliation(s)
- Xia Qi
- Department of Clinical Biochemistry, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yuejian Liu
- The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Wei Wei
- Department of Clinical Biochemistry, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Xiaohua Huang
- Department of Clinical Biochemistry, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yunfei Zuo
- Department of Clinical Biochemistry, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
34
|
Pushpakumar SB, Kundu S, Metreveli N, Sen U. Folic acid mitigates angiotensin-II-induced blood pressure and renal remodeling. PLoS One 2013; 8:e83813. [PMID: 24386282 PMCID: PMC3873386 DOI: 10.1371/journal.pone.0083813] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 11/08/2013] [Indexed: 12/11/2022] Open
Abstract
Clinical data suggests an association between systolic hypertension, renal function and hyperhomocysteinemia (HHcy). HHcy is a state of elevated plasma homocysteine (Hcy) levels and is known to cause vascular complications. In this study, we tested the hypothesis whether Ang II-induced hypertension increases plasma Hcy levels and contributes to renovascular remodeling. We also tested whether folic acid (FA) treatment reduces plasma Hcy levels by enhancing Hcy remethylation and thus mitigating renal remodeling. Hypertension was induced in WT mice by infusing Ang II using Alzet mini osmotic pumps. Blood pressure, Hcy level, renal vascular density, oxidative stress, inflammation and fibrosis markers, and angiogenic- and anti-angiogenic factors were measured. Ang II hypertension increased plasma Hcy levels and reduced renal cortical blood flow and microvascular density. Elevated Hcy in Ang II hypertension was associated with decreased 4, 5-Diaminofluorescein (DAF-2DA) staining suggesting impaired endothelial function. Increased expression of Nox-2, -4 and dihydroethidium stain revealed oxidative stress. Excess collagen IV deposition in the peri-glomerular area and increased MMP-2, and -9 expression and activity indicated renal remodeling. The mRNA and protein expression of asymmetric dimethylarginine (ADMA) was increased and eNOS protein was decreased suggesting the involvement of this pathway in Hcy mediated hypertension. Decreased expressions of VEGF and increased anti-angiogenic factors, angiostatin and endostatin indicated impaired vasculogenesis. FA treatment partially reduced hypertension by mitigating HHcy in Ang II-treated animals and alleviated pro-inflammatory, pro-fibrotic and anti-angiogenic factors. These results suggest that renovascular remodeling in Ang II-induced hypertension is, in part, due to HHcy.
Collapse
Affiliation(s)
- Sathnur B. Pushpakumar
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Sourav Kundu
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Naira Metreveli
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Utpal Sen
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
35
|
Jiang WG, Lu XA, Shang BY, Fu Y, Zhang SH, Zhou D, Li L, Li Y, Luo Y, Zhen YS. Genetically engineered endostatin-lidamycin fusion proteins effectively inhibit tumor growth and metastasis. BMC Cancer 2013; 13:479. [PMID: 24128285 PMCID: PMC4016579 DOI: 10.1186/1471-2407-13-479] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Accepted: 09/20/2013] [Indexed: 01/07/2023] Open
Abstract
Background Endostatin (ES) inhibits endothelial cell proliferation, migration, invasion, and tube formation. It also shows antiangiogenesis and antitumor activities in several animal models. Endostatin specifically targets tumor vasculature to block tumor growth. Lidamycin (LDM), which consists of an active enediyne chromophore (AE) and a non-covalently bound apo-protein (LDP), is a member of chromoprotein family of antitumor antibiotics with extremely potent cytotoxicity to cancer cells. Therefore, we reasoned that endostatin-lidamycin (ES-LDM) fusion proteins upon energizing with enediyne chromophore may obtain the combined capability targeting tumor vasculature and tumor cell by respective ES and LDM moiety. Methods In this study, we designed and obtained two new endostatin-based fusion proteins, endostatin-LDP (ES-LDP) and LDP-endostatin (LDP-ES). In vitro, the antiangiogenic effect of fusion proteins was determined by the wound healing assay and tube formation assay and the cytotoxicity of their enediyne-energized analogs was evaluated by CCK-8 assay. Tissue microarray was used to analyze the binding affinity of LDP, ES or ES-LDP with specimens of human lung tissue and lung tumor. The in vivo efficacy of the fusion proteins was evaluated with human lung carcinoma PG-BE1 xenograft and the experimental metastasis model of 4T1-luc breast cancer. Results ES-LDP and LDP-ES disrupted the formation of endothelial tube structures and inhibited endothelial cell migration. Evidently, ES-LDP accumulated in the tumor and suppressed tumor growth and metastasis. ES-LDP and ES show higher binding capability than LDP to lung carcinoma; in addition, ES-LDP and ES share similar binding capability. Furthermore, the enediyne-energized fusion protein ES-LDP-AE demonstrated significant efficacy against lung carcinoma xenograft in athymic mice. Conclusions The ES-based fusion protein therapy provides some fundamental information for further drug development. Targeting both tumor vasculature and tumor cells by endostatin-based fusion proteins and their enediyne-energized analogs probably provides a promising modality in cancer therapy.
Collapse
Affiliation(s)
- Wen-guo Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P, R, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Holstein JH, Karabin-Kehl B, Scheuer C, Garcia P, Histing T, Meier C, Benninger E, Menger MD, Pohlemann T. Endostatin inhibits Callus remodeling during fracture healing in mice. J Orthop Res 2013; 31:1579-84. [PMID: 23720153 DOI: 10.1002/jor.22401] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 05/06/2013] [Indexed: 02/04/2023]
Abstract
Information on the impact of endogenous anti-angiogenic factors on bone repair is limited. The hypothesis of the present study was endostatin, an endogenous inhibitor of angiogenesis, disturbs fracture healing. We evaluated this hypothesis in a closed femoral fracture model studying two groups of mice, one that was treated by a daily injection of 10 µg recombinant endostatin subcutaneously (n = 38) and a second one that received the vehicle for control (n = 37). Histomorphometric analysis showed a significantly increased callus formation in endostatin-treated animals at 2 and 5 weeks post-fracture. This was associated with a significantly higher callus tissue fraction of cartilage and fibrous tissue at 2 weeks and a significantly higher fraction of bone at 5 weeks post-fracture. Biomechanical testing revealed a significantly higher torsional stiffness in the endostatin group at 2 weeks. For both groups, we could demonstrate the expression of the endostatin receptor unit integrin alpha5 in endothelial cells, osteoblasts, osteoclasts, and chondrocytes at 2 weeks. Immunohistochemical fluorescence staining of CD31 showed a lower number of blood vessels in endostatin-treated animals compared to controls. The results of the present study indicate endostatin promotes soft callus formation but inhibits callus remodeling during fracture healing most probably by an inhibition of angiogenesis.
Collapse
Affiliation(s)
- Joerg H Holstein
- Department of Trauma, Hand and Reconstructive Surgery, University of Saarland, Kirrberger Strasse, 66421, Homburg/Saar, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Wan YY, Tian GY, Guo HS, Kang YM, Yao ZH, Li XL, Liu QH, Lin DJ. Endostatin, an angiogenesis inhibitor, ameliorates bleomycin-induced pulmonary fibrosis in rats. Respir Res 2013; 14:56. [PMID: 23688086 PMCID: PMC3668162 DOI: 10.1186/1465-9921-14-56] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 05/15/2013] [Indexed: 12/26/2022] Open
Abstract
Background Recent evidence has demonstrated the role of angiogenesis in the pathogenesis of pulmonary fibrosis. Endostatin, a proteolytic fragment of collagen XVIII, is a potent inhibitor of angiogenesis. The aim of our study was to assess whether endostatin has beneficial effects on bleomycin (BLM)-induced pulmonary fibrosis in rats. Methods The rats were randomly divided into five experimental groups: (A) saline only, (B) BLM only, (C) BLM plus early endostatin treatment, (D) BLM plus late endostatin treatment, and (F) BLM plus whole-course endostatin treatment. We investigated the microvascular density (MVD), inflammatory response and alveolar epithelial cell apoptosis in rat lungs in each group at different phases of disease development. Results Early endostatin administration attenuated fibrotic changes in BLM-induced pulmonary fibrosis in rats. Endostatin treatment decreased MVD by inhibiting the expression of VEGF/VEGFR-2 (Flk-1) and the activation of extracellular signal-regulated protein kinase 1/2 (ERK1/2). Endostatin treatment also decreased the number of inflammatory cells infiltrating the bronchoalveolar lavage fluid during the early inflammatory phase of BLM-induced pulmonary fibrosis. In addition, the levels of tumour necrosis factor-α (TNF-α) and transforming growth factor β1 (TGF-β1) were reduced by endostatin treatment. Furthermore, endostatin decreased alveolar type II cell apoptosis and had an epithelium-protective effect. These might be the mechanism underlying the preventive effect of endostatin on pulmonary fibrosis. Conclusions Our findings suggest that endostatin treatment inhibits the increased MVD, inflammation and alveolar epithelial cell apoptosis, consequently ameliorating BLM-induced pulmonary fibrosis in rats.
Collapse
Affiliation(s)
- Yun-Yan Wan
- Department of Respiratory Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province 250021, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Braga MS, Turaça TL, Foguer K, Chaves KCB, Pesquero JB, Chammas R, Schor N, Bellini MH. Vascular endothelial growth factor as a biomarker for endostatin gene therapy. Biomed Pharmacother 2013; 67:511-5. [PMID: 23726969 DOI: 10.1016/j.biopha.2013.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 04/19/2013] [Indexed: 12/26/2022] Open
Abstract
Renal cell carcinoma (RCC) is characterized by high vascular endothelial growth factor (VEGF) production and, consequently, excessive angiogenesis. Several strategies have been developed to target angiogenesis as a method for treating metastatic RCC (mRCC). Endostatin (ES) is a C-terminal fragment of collagen XVIII that has antiangiogenic activity. The aim of this study was to investigate the predictive value of circulating VEGF-A in a murine model of mRCC after ES gene therapy. ES therapy did not affect the levels of collagen XVIII/ES or ES in the tissue. The circulating level of ES was increased in the control and ES-treated groups (normal vs. control, P<0.05 and ES-treated vs. control, P<0.001), and the intratumoral vessels were significantly decreased (ES-treated vs. control, P<0.05). ES therapy decreased the VEGF mRNA levels. The tissue and circulating levels of VEGF in the control group were significantly higher than normal (P<0.01 and P<0.05, respectively). Treatment with ES significantly reduced the VEGF concentrations in both compartments (P<0.001 for tissue and P<0.05 for plasma). Our findings indicate that in addition to the directly targeted tumor vessels, ES exerts its antitumor effect by down-regulating VEGF gene expression in renal tumor cells. Additionally, our findings point to the predictive value of VEGF for ES therapy.
Collapse
|
39
|
Chen X, Liu Y, Yin Y, Jin S, Ping G, Røe OD, Yan K, Shu Y, Guo R. Recombinant human endostatin (endostar) decreased recurrent ascites, pleural fluid and ascitic VEGF in a case of advanced mesothelioma. J Chemother 2013; 24:231-6. [PMID: 23040690 DOI: 10.1179/1973947812y.0000000023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
We report a case of 43-year-old male with advanced malignant mesothelioma (MM) with a large amount of fluid in the pleural and peritoneal cavity. The addition of endostar to the gemcitabine-cisplatin regimen gave a prompt and significant improvement of clinical symptoms and disappearance of ascites. The patient is still progression free after 27 months. Endostar, in combination with chemotherapy should be explored in the treatment of MM, especially its effect on pleural and ascitic fluid.
Collapse
Affiliation(s)
- Xiaofeng Chen
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ma X, Yao Y, Yuan D, Liu H, Wang S, Zhou C, Song Y. Recombinant human endostatin endostar suppresses angiogenesis and lymphangiogenesis of malignant pleural effusion in mice. PLoS One 2012; 7:e53449. [PMID: 23285296 PMCID: PMC3532165 DOI: 10.1371/journal.pone.0053449] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 11/28/2012] [Indexed: 01/02/2023] Open
Abstract
Background Malignant pleural effusion (MPE) is a common complication of lung cancer. One widely used treatment for MPE is Endostar, a recombined humanized endostatin based treatment. However, the mechanism of this treatment is still unclear. The aim of this study was to investigate the effects of Endostar in mice with MPE. Methods and Materials Lewis lung carcinoma (LLC) cell line expressing enhanced green fluorescent protein (EGFP) was injected into pleural cavity to establish MPE mice model. Mice were randomly divided into four groups. High dose of Endostar (30 mg/kg), low dose of Endostar (8 mg/kg), normal saline, or Bevacizumab (5 mg/kg) was respectively injected into pleural cavity three times with 3-day interval in each group. Transverse computed tomography (CT) was performed to observe pleural fluid formation 14 days after LLC cells injection. Mice were anesthetized and sacrificed 3 days after final administration. The volume of pleural effusion n was measured using 1 ml syringe. Micro blood vessel density (MVD), Lymphatic micro vessel density (LMVD), the expression level of vascular endothelial growth factor A (VEGF-A) and VEGF-C were observed by immunohistochemistry (IHC) staining. Results The volume of pleural effusion as well as the number of pleural tumor foci, MVD and the expression of VEGF-A were significantly reduced in high dose of Endostar treat group. More importantly, LMVD and the expression of VEGF-C were markedly lower in treat group than those in the other three control groups. Conclusion Our work demonstrated that Endostar played an efficient anti-cancer role in MPE through its suppressive effect on angiogenesis and lymphangiogenesis, which provided a certain theoretical basis for the effectiveness of Endostar on the MPE treatment.
Collapse
MESH Headings
- Animals
- Carcinoma, Lewis Lung/complications
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/pathology
- Cell Line, Tumor
- Down-Regulation/drug effects
- Drug Evaluation, Preclinical
- Endostatins/pharmacology
- Endostatins/therapeutic use
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Lymphangiogenesis/drug effects
- Lymphangiogenesis/physiology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Transplantation
- Neovascularization, Pathologic/etiology
- Neovascularization, Pathologic/prevention & control
- Pleural Effusion, Malignant/complications
- Pleural Effusion, Malignant/drug therapy
- Pleural Effusion, Malignant/pathology
- Recombinant Proteins/pharmacology
- Recombinant Proteins/therapeutic use
Collapse
Affiliation(s)
- Xingqun Ma
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yanwen Yao
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Dongmei Yuan
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Hongbing Liu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Shouju Wang
- Department of Medical Imaging, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Changsheng Zhou
- Department of Medical Imaging, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
- * E-mail:
| |
Collapse
|
41
|
ZhiYong W, Fei S, LianJu X, Yingkai L, Chun Q, Shuliang L, XiQiao W. Endostar Injection Inhibits Rabbit Ear Hypertrophic Scar Formation. INT J LOW EXTR WOUND 2012; 11:271-6. [DOI: 10.1177/1534734612463698] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This study aims to investigate the effect of Endostar injection on the rabbit ear hypertrophic scar formation and expand the use of Endostar. The rabbit ear hypertrophic scar models were established 4 weeks postoperation and were treated with Endostar injection; the control group was injected with saline, once a week, 3 times totally. At the seventh week, the scar tissue was harvested and processed with hematoxylin and eosin (HE) staining and CD34 immunohistochemistry and cell apoptosis assay. In addition, the endothelial cell was cultured and seeded on Martrigel with different concentrations of Endostar to observe the vessel tube formation. The results showed that the volume of the hypertrophic scar with Endostar injection was greatly reduced compared with what was seen in the control group; meanwhile, HE staining showed that the cell number decreased and collagen density became looser. In addition, the CD34 staining indicated that microvessel formation in the study group also decreased and cell apoptosis increased. In vitro, the addition of Endostar could reduce vessel tube formation in a dose-dependent manner. In conclusion, the Endostar is effective for hypertrophic scar inhibition and could be a potential tool to treat scars.
Collapse
Affiliation(s)
| | - Song Fei
- Shanghai Jiaotong University, Shanghai, China
| | - Xu LianJu
- Shanghai Jiaotong University, Shanghai, China
| | - Liu Yingkai
- Shanghai Jiaotong University, Shanghai, China
| | - Qing Chun
- Shanghai Jiaotong University, Shanghai, China
| | - Lu Shuliang
- Shanghai Jiaotong University, Shanghai, China
| | - Wang XiQiao
- Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
42
|
Sen U, Sathnur PB, Kundu S, Givvimani S, Coley DM, Mishra PK, Qipshidze N, Tyagi N, Metreveli N, Tyagi SC. Increased endogenous H2S generation by CBS, CSE, and 3MST gene therapy improves ex vivo renovascular relaxation in hyperhomocysteinemia. Am J Physiol Cell Physiol 2012; 303:C41-51. [PMID: 22517358 DOI: 10.1152/ajpcell.00398.2011] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Hydrogen sulfide (H(2)S) has recently been identified as a regulator of various physiological events, including vasodilation, angiogenesis, antiapoptotic, and cellular signaling. Endogenously, H(2)S is produced as a metabolite of homocysteine (Hcy) by cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE), and 3-mercaptopyruvate sulfurtransferase (3MST). Although Hcy is recognized as vascular risk factor at an elevated level [hyperhomocysteinemia (HHcy)] and contributes to vascular injury leading to renovascular dysfunction, the exact mechanism is unclear. The goal of the current study was to investigate whether conversion of Hcy to H(2)S improves renovascular function. Ex vivo renal artery culture with CBS, CSE, and 3MST triple gene therapy generated more H(2)S in the presence of Hcy, and these arteries were more responsive to endothelial-dependent vasodilation compared with nontransfected arteries treated with high Hcy. Cross section of triple gene-delivered renal arteries immunostaining suggested increased expression of CD31 and VEGF and diminished expression of the antiangiogenic factor endostatin. In vitro endothelial cell culture demonstrated increased mitophagy during high levels of Hcy and was mitigated by triple gene delivery. Also, dephosphorylated Akt and phosphorylated FoxO3 in HHcy were reversed by H(2)S or triple gene delivery. Upregulated matrix metalloproteinases-13 and downregulated tissue inhibitor of metalloproteinase-1 in HHcy were normalized by overexpression of triple genes. Together, these results suggest that H(2)S plays a key role in renovasculopathy during HHcy and is mediated through Akt/FoxO3 pathways. We conclude that conversion of Hcy to H(2)S by CBS, CSE, or 3MST triple gene therapy improves renovascular function in HHcy.
Collapse
Affiliation(s)
- Utpal Sen
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Kentucky, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Chen J, Hamm LL, Kleinpeter MA, Husserl F, Khan IE, Chen CS, Liu Y, Mills KT, He C, Rifai N, Simon EE, He J. Elevated plasma levels of endostatin are associated with chronic kidney disease. Am J Nephrol 2012; 35:335-40. [PMID: 22456114 DOI: 10.1159/000336109] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 12/23/2011] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Angiogenesis may play an important role in the renal repair process after injury. We investigated the association between plasma endostatin, an endothelial-specific antiangiogenic factor, and chronic kidney disease (CKD). METHODS We compared plasma endostatin levels in 201 CKD patients and 201 controls. CKD was defined as estimated glomerular filtration rate (eGFR) <60 ml/min/1.73 m(2) or presence of albuminuria (≥30 mg/24 h). RESULTS After adjustment for established CKD risk factors, the median (interquartile range) of plasma endostatin was 276.7 ng/dl (199.3-357.5) in patients with CKD and 119.4 ng/dl (103.7-134.6) in controls without CKD (p < 0.0001 for group difference). log-transformed plasma endostatin was significantly and inversely correlated with eGFR (r = -0.83, p < 0.0001) and positively correlated with log-transformed urine albumin (r = 0.66, p < 0.0001) in the study participants. In addition, one standard deviation increase in log-transformed plasma endostatin (0.55 ng/dl) was associated with a decline in eGFR of -26.2 ml/min and an increase in urine albumin of 3.26 mg/ 24 h after adjusting for multiple covariables. Furthermore, the multivariable-adjusted odds ratio for CKD comparing the highest tertile (≥131.4 ng/dl) to the two lower tertiles of plasma endostatin was 21.6 (95% CI: 10.2-45.5; p < 0.0001). CONCLUSION These data indicate that elevated plasma endostatin is strongly and independently associated with CKD. Prospective cohort studies and clinical trials are warranted to further examine the causal relationship between endostatin and risk of CKD and to develop novel interventions targeting circulating endostatin aimed at reducing CKD risk.
Collapse
Affiliation(s)
- Jing Chen
- Department of Medicine, Tulane University School of Medicine, New Orleans, La. 70112, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Qipshidze N, Metreveli N, Mishra PK, Lominadze D, Tyagi SC. Hydrogen sulfide mitigates cardiac remodeling during myocardial infarction via improvement of angiogenesis. Int J Biol Sci 2012; 8:430-41. [PMID: 22419888 PMCID: PMC3303169 DOI: 10.7150/ijbs.3632] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 01/12/2012] [Indexed: 11/17/2022] Open
Abstract
Exogenous hydrogen sulfide (H2S) leads to down-regulation of inflammatory responses and provides myocardial protection during acute ischemia/reperfusion injury; however its role during chronic heart failure (CHF) due to myocardial infarction (MI) is yet to be unveiled. We previously reported that H2S inhibits antiangiogenic factors such, as endostatin and angiostatin, but a little is known about its effect on parstatin (a fragment of proteinase-activated receptor-1, PAR-1). We hypothesize that H2S inhibits parstatin formation and promotes VEGF activation, thus promoting angiogenesis and significantly limiting the extent of MI injury. To verify this hypothesis MI was created in 12 week-old male mice by ligation of left anterior descending artery (LAD). Sham surgery was performed except LAD ligation. After the surgery mice were treated with sodium hydrogen sulfide (30 μmol/l NaHS, a donor for H2S, in drinking water) for 4 weeks. The LV tissue was analyzed for VEGF, flk-1 and flt-1, endostatin, angiostatin and parstatin. The expression of VEGF, flk-1 and flt-1 were significantly increased in treated mice while the level of endostatin, angiostatin and parstatin were decreased compared to in untreated mice. The echocardiography in mice treated with H2S showed the improvement of heart function compared to in untreated mice. The X-ray and Doppler blood flow measurements showed enhancement of cardiac-angiogenesis in mice treated with H2S. This observed cytoprotection was associated with an inhibition of anti-angiogenic proteins and stimulation of angiogenic factors. We established that administration of H2S at the time of MI ameliorated infarct size and preserved LV function during development of MI in mice. These results suggest that H2S is cytoprotective and angioprotective during evolution of MI.
Collapse
Affiliation(s)
- Natia Qipshidze
- Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| | | | | | | | | |
Collapse
|
45
|
Chan SF, Wang HT, Huang KW, Torng PL, Lee HI, Hwang LH. Anti-angiogenic therapy renders large tumors vulnerable to immunotherapy via reducing immunosuppression in the tumor microenvironment. Cancer Lett 2012; 320:23-30. [PMID: 22266191 DOI: 10.1016/j.canlet.2012.01.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 01/12/2012] [Accepted: 01/13/2012] [Indexed: 10/14/2022]
Abstract
We have recently demonstrated that a 4-in-1 gene therapy strategy that contains two anti-angiogenic genes [endostatin and pigment epithelium-derived factor] and two cytokine genes [granulocyte macrophage colony-stimulating factor and interleukin 12] has a considerable antitumor effect on large tumors in a woodchuck hepatoma model. The current study further investigates the underlying mechanisms for the antitumor effect observed by using small rodent models. We found that immunotherapy alone increased immunosuppressive cells in large tumors over time, whereas the anti-angiogenic therapy contained in the 4-in-1 strategy alleviated immunosuppression and made tumors vulnerable to immunotherapy, thus resulting in a synergistic antitumor effect.
Collapse
Affiliation(s)
- Suit-Fong Chan
- Graduate Institute of Microbiology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | | | | | | | | | | |
Collapse
|
46
|
Jang JA, Kang JH, Sa KH, Han SW, Seo JS, Kim KH, Nam EJ, Kim IS, Kang YM. Therapeutic Effect of a Recombinant βig-h3 Fragment-RGD Peptide for Chronic Inflammatory Arthritis. JOURNAL OF RHEUMATIC DISEASES 2012. [DOI: 10.4078/jrd.2012.19.2.73] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Ji Ae Jang
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
- Department of Biochemistry and Cellular Biology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Jin Hee Kang
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
- Department of Biochemistry and Cellular Biology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Keum Hee Sa
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
- Department of Biochemistry and Cellular Biology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Seung Woo Han
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Jae Seok Seo
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Kyung Hoon Kim
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Eon Jeong Nam
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - In San Kim
- Department of Biochemistry and Cellular Biology, Kyungpook National University School of Medicine, Daegu, Korea
- Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, Korea
| | - Young Mo Kang
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
- Department of Biochemistry and Cellular Biology, Kyungpook National University School of Medicine, Daegu, Korea
- Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, Korea
| |
Collapse
|
47
|
Baker M, Robinson SD, Lechertier T, Barber PR, Tavora B, D'Amico G, Jones DT, Vojnovic B, Hodivala-Dilke K. Use of the mouse aortic ring assay to study angiogenesis. Nat Protoc 2011; 7:89-104. [PMID: 22193302 DOI: 10.1038/nprot.2011.435] [Citation(s) in RCA: 377] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Here we provide a protocol for quantitative three-dimensional ex vivo mouse aortic ring angiogenesis assays, in which developing microvessels undergo many key features of angiogenesis over a timescale similar to that observed in vivo. The aortic ring assay allows analysis of cellular proliferation, migration, tube formation, microvessel branching, perivascular recruitment and remodeling-all without the need for cellular dissociation-thus providing a more complete picture of angiogenic processes compared with traditional cell-based assays. Our protocol can be applied to aortic rings from embryonic stage E18 through to adulthood and can incorporate genetic manipulation, treatment with growth factors, drugs or siRNA. This robust assay allows assessment of the salient steps in angiogenesis and quantification of the developing microvessels, and it can be used to identify new modulators of angiogenesis. The assay takes 6-14 d to complete, depending on the age of the mice, treatments applied and whether immunostaining is performed.
Collapse
Affiliation(s)
- Marianne Baker
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Boosani CS, Sudhakar YA. Proteolytically Derived Endogenous Angioinhibitors Originating from the Extracellular Matrix. Pharmaceuticals (Basel) 2011; 4:1551-1577. [PMID: 22267953 PMCID: PMC3260939 DOI: 10.3390/ph4121551] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis, a neovascularization process induced from the existing parent blood vessels, is a prerequisite for many physiological and pathological conditions. Under physiological conditions it is regulated by a balance between endogenous angioinhibitors and angioactivators, and an imbalance between them would lead to pathological conditions such as cancer, age-related macular degeneration (AMD), diabetic retinopathy, cardiovascular diseases, etc. Several proteolytically generated endogenous molecules have been identified which exhibit angioinhibition and/or antitumor activities. These angioinhibitors interact with endothelial and tumor cells by binding to distinct integrins and initiate many of their intracellular signaling mechanisms regulating the cell survival and or apoptotic pathways. The present review will focus on the extracellular matrix derived angioinhibitors, and their mechanisms of actions that point to the clinical significance and therapeutic implications.
Collapse
Affiliation(s)
- Chandra Shekhar Boosani
- Cell Signaling, Retinal and Tumor Angiogenesis Laboratory, Department of Genetics, Boys Town National Research Hospital, Omaha, NE 68131, USA; E-Mail:
| | - Yakkanti A. Sudhakar
- Cell Signaling, Retinal and Tumor Angiogenesis Laboratory, Department of Genetics, Boys Town National Research Hospital, Omaha, NE 68131, USA; E-Mail:
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-402-498-6681; Fax: +1-402-498-6331
| |
Collapse
|
49
|
Nilsson Åberg UW, Saarinen N, Abrahamsson A, Nurmi T, Engblom S, Dabrosin C. Tamoxifen and flaxseed alter angiogenesis regulators in normal human breast tissue in vivo. PLoS One 2011; 6:e25720. [PMID: 21984941 PMCID: PMC3184168 DOI: 10.1371/journal.pone.0025720] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 09/09/2011] [Indexed: 01/01/2023] Open
Abstract
The incidence of breast cancer is increasing in the Western world and there is an urgent need for studies of the mechanisms of sex steroids in order to develop novel preventive strategies. Diet modifications may be among the means for breast cancer prevention. Angiogenesis, key in tumor progression, is regulated by the balance between pro- and anti-angiogenic factors, which are controlled in the extracellular space. Sampling of these molecules at their bioactive compartment is therefore needed. The aims of this study were to explore if tamoxifen, one of the most used anti-estrogen treatments for breast cancer affected some of the most important endogenous angiogenesis regulators, vascular endothelial growth factor (VEGF), angiogenin, and endostatin in normal breast tissue in vivo and if a diet supplementation with flaxseed had similar effects as tamoxifen in the breast. Microdialysis was used for in situ sampling of extracellular proteins in normal breast tissue of women before and after six weeks of tamoxifen treatment or before and after addition of 25 g/day of ground flaxseed to the diet or in control women. We show significant correlations between estradiol and levels of VEGF, angiogenin, and endostatin in vivo, which was verified in ex vivo breast tissue culture. Moreover, tamoxifen decreased the levels of VEGF and angiogenin in the breast whereas endostatin increased significantly. Flaxseed did not alter VEGF or angiogenin levels but similar to tamoxifen the levels of endostatin increased significantly. We conclude that one of the mechanisms of tamoxifen in normal breast tissue include tipping of the angiogenic balance into an anti-angiogenic state and that flaxseed has limited effects on the pro-angiogenic factors whereas the anti-angiogenic endostatin may be modified by diet. Further studies of diet modifications for breast cancer prevention are warranted.
Collapse
Affiliation(s)
- Ulrika W. Nilsson Åberg
- Division of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Niina Saarinen
- Division of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
- Functional Foods Forum, University of Turku, Turku, Finland
| | - Annelie Abrahamsson
- Division of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
- Department of Oncology, County Council of Östergötland, Linköping, Sweden
| | - Tarja Nurmi
- University of Eastern Finland, Kuopio Campus, Kuopio, Finland
| | - Sofia Engblom
- Division of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
- Department of Oncology, County Council of Östergötland, Linköping, Sweden
| | - Charlotta Dabrosin
- Division of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
- Department of Oncology, County Council of Östergötland, Linköping, Sweden
- * E-mail:
| |
Collapse
|
50
|
OLFERT IMARK, BIROT OLIVIER. Importance of Anti-angiogenic Factors in the Regulation of Skeletal Muscle Angiogenesis. Microcirculation 2011; 18:316-30. [DOI: 10.1111/j.1549-8719.2011.00092.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|