1
|
Kargl CK, Jia Z, Shera DA, Sullivan BP, Burton LC, Kim KH, Nie Y, Hubal MJ, Shannahan JH, Kuang S, Gavin TP. Angiogenic potential of skeletal muscle derived extracellular vesicles differs between oxidative and glycolytic muscle tissue in mice. Sci Rep 2023; 13:18943. [PMID: 37919323 PMCID: PMC10622454 DOI: 10.1038/s41598-023-45787-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 10/24/2023] [Indexed: 11/04/2023] Open
Abstract
Skeletal muscle fibers regulate surrounding endothelial cells (EC) via secretion of numerous angiogenic factors, including extracellular vesicles (SkM-EV). Muscle fibers are broadly classified as oxidative (OXI) or glycolytic (GLY) depending on their metabolic characteristics. OXI fibers secrete more pro-angiogenic factors and have greater capillary densities than GLY fibers. OXI muscle secretes more EV than GLY, however it is unknown whether muscle metabolic characteristics regulate EV contents and signaling potential. EVs were isolated from primarily oxidative or glycolytic muscle tissue from mice. MicroRNA (miR) contents were determined and endothelial cells were treated with OXI- and GLY-EV to investigate angiogenic signaling potential. There were considerable differences in miR contents between OXI- and GLY-EV and pathway analysis identified that OXI-EV miR were predicted to positively regulate multiple endothelial-specific pathways, compared to GLY-EV. OXI-EV improved in vitro angiogenesis, which may have been mediated through nitric oxide synthase (NOS) related pathways, as treatment of endothelial cells with a non-selective NOS inhibitor abolished the angiogenic benefits of OXI-EV. This is the first report to show widespread differences in miR contents between SkM-EV isolated from metabolically different muscle tissue and the first to demonstrate that oxidative muscle tissue secretes EV with greater angiogenic signaling potential than glycolytic muscle tissue.
Collapse
Affiliation(s)
- Christopher K Kargl
- Department of Health and Kinesiology, Max E. Wastl Human Performance Laboratory, Purdue University, West Lafayette, IN, USA
| | - Zhihao Jia
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Deborah A Shera
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Brian P Sullivan
- Department of Health and Kinesiology, Max E. Wastl Human Performance Laboratory, Purdue University, West Lafayette, IN, USA
| | - Lundon C Burton
- Department of Health and Kinesiology, Max E. Wastl Human Performance Laboratory, Purdue University, West Lafayette, IN, USA
| | - Kun Ho Kim
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Yaohui Nie
- Department of Health and Kinesiology, Max E. Wastl Human Performance Laboratory, Purdue University, West Lafayette, IN, USA
| | - Monica J Hubal
- Department of Kinesiology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | | | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Timothy P Gavin
- Department of Health and Kinesiology, Max E. Wastl Human Performance Laboratory, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
2
|
Zhang Y, Liu W, Wang X, Liu Y, Wei H. Nanozyme-Enabled Treatment of Cardio- and Cerebrovascular Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204809. [PMID: 36192166 DOI: 10.1002/smll.202204809] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/18/2022] [Indexed: 06/16/2023]
Abstract
Cardio- and cerebrovascular diseases are two major vascular-related diseases that lead to death worldwide. Reactive oxygen species (ROS) play a vital role in the occurrence and exacerbation of diseases. Excessive ROS induce cellular context damage and lead to tissue dysfunction. Nanozymes, as emerging enzyme mimics, offer a unique perspective for therapy through multifunctional activities, achieving essential results in the treatment of ROS-related cardio- and cerebrovascular diseases by directly scavenging excess ROS or regulating pathologically related molecules. This review first introduces nanozyme-enabled therapeutic mechanisms at the cellular level. Then, the therapies for several typical cardio- and cerebrovascular diseases with nanozymes are discussed, mainly including cardiovascular diseases, ischemia reperfusion injury, and neurological disorders. Finally, the challenges and outlooks for the application of nanozymes are also presented. This review will provide some instructive perspectives on nanozymes and promote the development of enzyme-mimicking strategies in cardio- and cerebrovascular disease therapy.
Collapse
Affiliation(s)
- Yihong Zhang
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Wanling Liu
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Xiaoyu Wang
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
- Department of Chemistry and Material Science, College of Science, Nanjing Forestry University, Nanjing, Jiangsu, 210037, China
| | - Yufeng Liu
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Hui Wei
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, Jiangsu, 210023, China
| |
Collapse
|
3
|
Li S, Pritchard DM, Yu LG. Regulation and Function of Matrix Metalloproteinase-13 in Cancer Progression and Metastasis. Cancers (Basel) 2022; 14:3263. [PMID: 35805035 PMCID: PMC9265061 DOI: 10.3390/cancers14133263] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022] Open
Abstract
Matrix metalloproteinase-13 (MMP-13) is a member of the Matrix metalloproteinases (MMPs) family of endopeptidases. MMP-13 is produced in low amounts and is well-regulated during normal physiological conditions. Its expression and secretion are, however, increased in various cancers, where it plays multiple roles in tumour progression and metastasis. As an interstitial collagenase, MMP-13 can proteolytically cleave not only collagens I, II and III, but also a range of extracellular matrix proteins (ECMs). Its action causes ECM remodelling and often leads to the release of various sequestered growth and angiogenetic factors that promote tumour cell growth, invasion and angiogenesis. This review summarizes our current understanding of the regulation of MMP-13 expression and secretion and discusses the actions of MMP-13 in cancer progression and metastasis.
Collapse
Affiliation(s)
- Shun Li
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK;
| | - David Mark Pritchard
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK;
| | - Lu-Gang Yu
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK;
| |
Collapse
|
4
|
Fisher RA, Miners JS, Love S. Pathological changes within the cerebral vasculature in Alzheimer's disease: New perspectives. Brain Pathol 2022; 32:e13061. [PMID: 35289012 PMCID: PMC9616094 DOI: 10.1111/bpa.13061] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/11/2022] [Accepted: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
Cerebrovascular disease underpins vascular dementia (VaD), but structural and functional changes to the cerebral vasculature contribute to disease pathology and cognitive decline in Alzheimer's disease (AD). In this review, we discuss the contribution of cerebral amyloid angiopathy and non‐amyloid small vessel disease in AD, and the accompanying changes to the density, maintenance and remodelling of vessels (including alterations to the composition and function of the cerebrovascular basement membrane). We consider how abnormalities of the constituent cells of the neurovascular unit – particularly of endothelial cells and pericytes – and impairment of the blood‐brain barrier (BBB) impact on the pathogenesis of AD. We also discuss how changes to the cerebral vasculature are likely to impair Aβ clearance – both intra‐periarteriolar drainage (IPAD) and transport of Aβ peptides across the BBB, and how impaired neurovascular coupling and reduced blood flow in relation to metabolic demand increase amyloidogenic processing of APP and the production of Aβ. We review the vasoactive properties of Aβ peptides themselves, and the probable bi‐directional relationship between vascular dysfunction and Aβ accumulation in AD. Lastly, we discuss recent methodological advances in transcriptomics and imaging that have provided novel insights into vascular changes in AD, and recent advances in assessment of the retina that allow in vivo detection of vascular changes in the early stages of AD.
Collapse
Affiliation(s)
- Robert A Fisher
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| | - J Scott Miners
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| | - Seth Love
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| |
Collapse
|
5
|
Influence of Genotype on Endometrial Angiogenesis during Early Pregnancy in Piau and Commercial Line Gilts. Animals (Basel) 2022; 12:ani12050553. [PMID: 35268121 PMCID: PMC8908842 DOI: 10.3390/ani12050553] [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: 10/11/2021] [Revised: 02/04/2022] [Accepted: 02/16/2022] [Indexed: 11/17/2022] Open
Abstract
This study aimed to evaluate the endometrial angiogenesis of pregnant commercial line and Piau gilts during early pregnancy. We used 27 gilts, divided into three groups according to the type of mating: Commercial (n = 9), commercial line females mated with commercial line males; Cross-mated (n = 9), Piau females mated with commercial line males; and Piau (n = 9), Piau females mated with Piau males. Each group was divided into three subgroups based on gestational age at the time of slaughter (7, 15, and 30 days of pregnancy). Immediately after slaughter, endometrial samples were obtained for histological evaluation and for analysis of the relative transcript abundance (RTA) of angiogenesis-related genes (HIF1α, FGF9, ANG1, TEK, VEGFA, ANGPT1, and ANGPT2). The number of endometrial glands was similar among groups but decreased with gestational age (p < 0.05). Piau females showed a higher number of blood vessels (p < 0.05) at 7 and 15 days of pregnancy, but no differences were observed among groups at 30 days, suggesting an influence of the male genotype on the pattern of uterine vascularization. There were no differences among groups for RTA of the FGF9, HIF1α, TEK, VEGFA, ANGPT1, and ANGPT2 genes. The HIF1α-gene RTA was higher at 7 and 15 days of pregnancy; for TEK and ANGPT1, the RTA was higher at 15 days of pregnancy; and the RTA of VEGFA and ANGPT2 genes were higher at 30 days of pregnancy. The ANG1 RTA was similar for pregnancies in the commercial and Piau groups but was higher (p < 0.05) at 15 days in the Cross-mated group, suggesting an interaction between genotypes. Overall, the pattern found for the RTA of angiogenesis-related genes was similar among the groups in this study, although some phenotypic differences could be noted, such as the highest number of blood vessels being found during early pregnancy of Piau gilts. The results of the gene RTA when crossed with phenotypic data led to conclusions that are conflicting with those reported in the literature. However, noteworthy is that angiogenesis is a complex process in which the balance between stimulatory and inhibitory factors may be related to time.
Collapse
|
6
|
Associations between increased circulating endothelial progenitor cell levels and anxiety/depressive severity, cognitive deficit and function disability among patients with major depressive disorder. Sci Rep 2021; 11:18221. [PMID: 34521977 PMCID: PMC8440504 DOI: 10.1038/s41598-021-97853-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 05/31/2021] [Indexed: 12/11/2022] Open
Abstract
The association of major depressive disorder (MDD) with cardiovascular diseases (CVDs) through endothelial dysfunction is bidirectional. Circulating endothelial progenitor cells (cEPCs), essential for endothelial repair and function, are associated with risks of various CVDs. Here, the relationship of cEPC counts with MDD and the related clinical presentations were investigated in 50 patients with MDD and 46 healthy controls. In patients with MDD, a battery of clinical domains was analysed: depressed mood with Hamilton Depression Rating Scale (HAMD) and Montgomery–Åsberg Depression Rating Scale (MADRS), anxiety with Hamilton Anxiety Rating Scale (HAMA), cognitive dysfunction and deficit with Digit Symbol Substitution Test (DSST) and Perceived Deficits Questionnaire-Depression (PDQ-D), somatic symptoms with Depressive and Somatic Symptom Scale (DSSS), quality of life with 12-Item Short Form Health Survey (SF-12) and functional disability with Sheehan Disability Scale (SDS). Immature and mature cEPC counts were measured through flow cytometry. Increased mature and immature cEPC counts were significantly associated with higher anxiety after controlling the confounding effect of systolic blood pressure, and potentially associated with more severe depressive symptoms, worse cognitive performance and increased cognitive deficit, higher social disability, and worse mental health outcomes. Thus, cEPCs might have pleiotropic effects on MDD-associated symptoms and psychosocial outcomes.
Collapse
|
7
|
Šalandová M, van Hengel IAJ, Apachitei I, Zadpoor AA, van der Eerden BCJ, Fratila‐Apachitei LE. Inorganic Agents for Enhanced Angiogenesis of Orthopedic Biomaterials. Adv Healthc Mater 2021; 10:e2002254. [PMID: 34036754 PMCID: PMC11469191 DOI: 10.1002/adhm.202002254] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/30/2021] [Indexed: 01/02/2023]
Abstract
Aseptic loosening of a permanent prosthesis remains one of the most common reasons for bone implant failure. To improve the fixation between implant and bone tissue as well as enhance blood vessel formation, bioactive agents are incorporated into the surface of the biomaterial. This study reviews and compares five bioactive elements (copper, magnesium, silicon, strontium, and zinc) with respect to their effect on the angiogenic behavior of endothelial cells (ECs) when incorporated on the surface of biomaterials. Moreover, it provides an overview of the state-of-the-art methodologies used for the in vitro assessment of the angiogenic properties of these elements. Two databases are searched using keywords containing ECs and copper, magnesium, silicon, strontium, and zinc. After applying the defined inclusion and exclusion criteria, 59 articles are retained for the final assessment. An overview of the angiogenic properties of five bioactive elements and the methods used for assessment of their in vitro angiogenic potential is presented. The findings show that silicon and strontium can effectively enhance osseointegration through the simultaneous promotion of both angiogenesis and osteogenesis. Therefore, their integration onto the surface of biomaterials can ultimately decrease the incidence of implant failure due to aseptic loosening.
Collapse
Affiliation(s)
- Monika Šalandová
- Additive Manufacturing LaboratoryDepartment of Biomechanical EngineeringFaculty of Mechanical, Maritime, and Materials EngineeringDelft University of TechnologyMekelweg 2Delft2628 CDThe Netherlands
| | - Ingmar A. J. van Hengel
- Additive Manufacturing LaboratoryDepartment of Biomechanical EngineeringFaculty of Mechanical, Maritime, and Materials EngineeringDelft University of TechnologyMekelweg 2Delft2628 CDThe Netherlands
| | - Iulian Apachitei
- Additive Manufacturing LaboratoryDepartment of Biomechanical EngineeringFaculty of Mechanical, Maritime, and Materials EngineeringDelft University of TechnologyMekelweg 2Delft2628 CDThe Netherlands
| | - Amir A. Zadpoor
- Additive Manufacturing LaboratoryDepartment of Biomechanical EngineeringFaculty of Mechanical, Maritime, and Materials EngineeringDelft University of TechnologyMekelweg 2Delft2628 CDThe Netherlands
| | - Bram C. J. van der Eerden
- Department of Internal MedicineErasmus Medical CenterDoctor Molewaterplein 40Rotterdam3015 GDThe Netherlands
| | - Lidy E. Fratila‐Apachitei
- Additive Manufacturing LaboratoryDepartment of Biomechanical EngineeringFaculty of Mechanical, Maritime, and Materials EngineeringDelft University of TechnologyMekelweg 2Delft2628 CDThe Netherlands
| |
Collapse
|
8
|
Rosanto YB, Hasan CY, Rahardjo R, Pangestiningsih TW. Effect of snail mucus on angiogenesis during wound healing. F1000Res 2021; 10:181. [PMID: 38912381 PMCID: PMC11190653 DOI: 10.12688/f1000research.51297.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/07/2021] [Indexed: 06/25/2024] Open
Abstract
Background: Angiogenesis is the process through which new blood vessels are formed from existing ones. This process plays an important role in supplying the oxygen and nutrients needed for cellular metabolism and eliminating cell debris during wound healing. Snail mucus can bind to several factors that stimulate angiogenesis, including vascular endothelial growth factor, platelet-derived growth factor, and fibroblast growth factor. The aim of this study is to observe changes in angiogenesis during the healing of wounds topically applied with snail mucus. Methods: Punch biopsy was performed on the back of male Wistar rats to obtain four wounds, and different concentrations of snail mucus were applied to each of these wounds. The animals were sacrificed on days 2, 4, and 7 to observe the extent of angiogenesis during wound healing by microscopy. Results: Two-way ANOVA showed differences in number of blood vessels formed (p = 0.00) and day of observation (p = 0.00) between groups. Post hoc Tukey's HSD test showed that 24% snail mucus treatment does not significantly affect wound healing (p = 0.488); by contrast, treatment with 48% and 96% snail mucus demonstrated significant effects on angiogenesis (p = 0.01). Spearman's test showed interactive effects between snail mucus concentration and day of observation on the extent of angiogenesis (p = 0.001, R = 0.946). Conclusion: Topical application of snail mucus gel can increase angiogenesis during wound healing in Wistar rat skin.
Collapse
Affiliation(s)
- Yosaphat Bayu Rosanto
- Oral and Maxillofacial Surgery, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia, 55281, Indonesia
| | - Cahya Yustisia Hasan
- Oral and Maxillofacial Surgery, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia, 55281, Indonesia
| | - Rahardjo Rahardjo
- Oral and Maxillofacial Surgery, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia, 55281, Indonesia
| | - Tri Wahyu Pangestiningsih
- Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia, 55281, Indonesia
| |
Collapse
|
9
|
Beheshtizadeh N, Asgari Y, Nasiri N, Farzin A, Ghorbani M, Lotfibakhshaiesh N, Azami M. A network analysis of angiogenesis/osteogenesis-related growth factors in bone tissue engineering based on in-vitro and in-vivo data: A systems biology approach. Tissue Cell 2021; 72:101553. [PMID: 33975231 DOI: 10.1016/j.tice.2021.101553] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/01/2021] [Accepted: 04/27/2021] [Indexed: 12/18/2022]
Abstract
The principal purpose of tissue engineering is to stimulate the injured or unhealthy tissues to revive their primary function through the simultaneous use of chemical agents, cells, and biocompatible materials. Still, choosing the appropriate protein as a growth factor (GF) for tissue engineering is vital to fabricate artificial tissues and accelerate the regeneration procedure. In this study, the angiogenesis and osteogenesis-related proteins' interactions are studied using their related network. Three major biological processes, including osteogenesis, angiogenesis, and angiogenesis regulation, were investigated by creating a protein-protein interaction (PPI) network (45 nodes and 237 edges) of bone regeneration efficient proteins. Furthermore, a gene ontology and a centrality analysis were performed to identify essential proteins within a network. The higher degree in this network leads to higher interactions between proteins and causes a considerable effect. The most highly connected proteins in the PPI network are the most remarkable for their employment. The results of this study showed that three significant proteins including prostaglandin endoperoxide synthase 2 (PTGS2), TEK receptor tyrosine kinase (TEK), and fibroblast growth factor 18 (FGF18) were involved simultaneously in osteogenesis, angiogenesis, and their positive regulatory. Regarding the available literature, the results of this study confirmed that PTGS2 and FGF18 could be used as a GF in bone tissue engineering (BTE) applications to promote angiogenesis and osteogenesis. Nevertheless, TEK was not used in BTE applications until now and should be considered in future works to be examined in-vitro and in-vivo.
Collapse
Affiliation(s)
- Nima Beheshtizadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran; School of Engineering, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, 2109, Australia
| | - Yazdan Asgari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran
| | - Noushin Nasiri
- School of Engineering, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, 2109, Australia
| | - Ali Farzin
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Ghorbani
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran
| | - Nasrin Lotfibakhshaiesh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
10
|
Zakeri N, Mirdamadi ES, Kalhori D, Solati-Hashjin M. Signaling molecules orchestrating liver regenerative medicine. J Tissue Eng Regen Med 2020; 14:1715-1737. [PMID: 33043611 DOI: 10.1002/term.3135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/06/2020] [Accepted: 09/09/2020] [Indexed: 12/19/2022]
Abstract
The liver is in charge of more than 500 functions in the human body, which any damage and failure to the liver can significantly compromise human life. Numerous studies are being carried out in regenerative medicine, as a potential driving force, toward alleviating the need for liver donors and fabrication of a 3D-engineered transplantable hepatic tissue. Liver tissue engineering brings three main factors of cells, extracellular matrix (ECM), and signaling molecules together, while each of these three factors tries to mimic the physiological state of the tissue to direct tissue regeneration. Signaling molecules play a crucial role in directing tissue fabrication in liver tissue engineering. When mimicking the natural in vivo process of regeneration, it is tightly associated with three main phases of differentiation, proliferation (progression), and tissue maturation through vascularization while directing each of these phases is highly regulated by the specific signaling molecules. The understanding of how these signaling molecules guide the dynamic behavior of regeneration would be a tool for further tailoring of bioengineered systems to help the liver regeneration with many cellular, molecular, and tissue-level functions. Hence, the signaling molecules come to aid all these phases for further improvements toward the clinical use of liver tissue engineering as the goal.
Collapse
Affiliation(s)
- Nima Zakeri
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Elnaz Sadat Mirdamadi
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Dianoosh Kalhori
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Mehran Solati-Hashjin
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
11
|
Reichardt W, von Elverfeldt D. Preclinical Applications of Magnetic Resonance Imaging in Oncology. Recent Results Cancer Res 2020; 216:405-437. [PMID: 32594394 DOI: 10.1007/978-3-030-42618-7_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The evolving possibilities of molecular imaging (MI) are fundamentally changing the way we look at cancer, with imaging paradigms now shifting away from basic morphological measures toward the longitudinal assessment of functional, metabolic, cellular, and molecular information in vivo. Recent developments of imaging methodology and probe molecules utilizing the vast number of novel animal models of human cancers have enhanced our ability to non-invasively characterize neoplastic tissue and follow anticancer treatments. While preclinical molecular imaging offers a whole palette of excellent methodology to choose from, we will focus on magnetic resonance imaging (MRI) techniques, since they provide excellent molecular imaging capabilities and bear high potential for clinical translation. Prerequisites and consequences of using animal models as surrogates of human cancers in preclinical molecular imaging are outlined. We present physical principles, values, and limitations of MRI as molecular imaging modality and comment on its high potential to non-invasively assess information on metabolism, hypoxia, angiogenesis, and cell trafficking in preclinical cancer research.
Collapse
Affiliation(s)
- Wilfried Reichardt
- Medical Physics, Department of Radiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany. .,German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Dominik von Elverfeldt
- Medical Physics, Department of Radiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
12
|
Díaz E, Valle MB, Ribeiro S, Lanceros‑Mendez S, Barandiarán JM. A New Approach for the Fabrication of Cytocompatible PLLA-Magnetite Nanoparticle Composite Scaffolds. Int J Mol Sci 2019; 20:E4664. [PMID: 31547060 PMCID: PMC6801398 DOI: 10.3390/ijms20194664] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/10/2019] [Accepted: 09/15/2019] [Indexed: 11/24/2022] Open
Abstract
Magnetic biomimetic scaffolds of poly(L-lactide) (PLLA) and nanoparticles of magnetite (nFe3O4) are prepared in a wide ratio of compositions by lyophilization for bone regeneration. The magnetic properties, cytotoxicity, and the in vitro degradation of these porous materials are closely studied. The addition of magnetite at 50 °C was found to produce an interaction reaction between the ester groups of the PLLA and the metallic cations of the magnetite, causing the formation of complexes. This fact was confirmed by the analysis of the infrared spectroscopy and the gel permeation chromatography test results. They, respectively, showed a displacement of the absorption bands of the carbonyl group (C=O) of the PLLA and a scission of the polymer chains. The iron from the magnetite acted as a catalyser of the macromolecular scission reaction, which determines the final biomedical applications of the scaffolds-it does so because the reaction shortens the degradation process without appearing to influence its toxicity. None of the samples studied in the tests presented cytotoxicity, even at 70% magnetite concentrations.
Collapse
Affiliation(s)
- Esperanza Díaz
- Escuela de Ingeniería de Bilbao, Departamento de Ingeniería Minera, Metalúrgica y Ciencia de Materiales, Universidad del País Vasco (UPV/EHU), 48920 Portugalete, Spain
- BCMaterials, Basque Centre for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain (J.M.B.)
| | - María Blanca Valle
- Facultad de Ciencia y Tecnología, Departamento de Electricidad y Electrónica, University of the Basque Country (UPV/EHU), Sarriena s/n, 48940 Leioa, Spain;
| | - Sylvie Ribeiro
- Centro de Física, Universidade do Minho, 4710-057 Braga, Portugal;
- Centre of Molecular and Environmental Biology (CBMA), Universidade do Minho, 4710-057 Braga, Portugal
| | - Senentxu Lanceros‑Mendez
- BCMaterials, Basque Centre for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain (J.M.B.)
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| | - José Manuel Barandiarán
- BCMaterials, Basque Centre for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain (J.M.B.)
- Facultad de Ciencia y Tecnología, Departamento de Electricidad y Electrónica, University of the Basque Country (UPV/EHU), Sarriena s/n, 48940 Leioa, Spain;
| |
Collapse
|
13
|
Paun IA, Calin BS, Mustaciosu CC, Mihailescu M, Moldovan A, Crisan O, Leca A, Luculescu CR. 3D Superparamagnetic Scaffolds for Bone Mineralization under Static Magnetic Field Stimulation. MATERIALS 2019; 12:ma12172834. [PMID: 31484381 PMCID: PMC6747966 DOI: 10.3390/ma12172834] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/20/2019] [Accepted: 08/26/2019] [Indexed: 01/22/2023]
Abstract
We reported on three-dimensional (3D) superparamagnetic scaffolds that enhanced the mineralization of magnetic nanoparticle-free osteoblast cells. The scaffolds were fabricated with submicronic resolution by laser direct writing via two photons polymerization of Ormocore/magnetic nanoparticles (MNPs) composites and possessed complex and reproducible architectures. MNPs with a diameter of 4.9 ± 1.5 nm and saturation magnetization of 30 emu/g were added to Ormocore, in concentrations of 0, 2 and 4 mg/mL. The homogenous distribution and the concentration of the MNPs from the unpolymerized Ormocore/MNPs composite were preserved after the photopolymerization process. The MNPs in the scaffolds retained their superparamagnetic behavior. The specific magnetizations of the scaffolds with 2 and 4 mg/mL MNPs concentrations were of 14 emu/g and 17 emu/g, respectively. The MNPs reduced the shrinkage of the structures from 80.2 ± 5.3% for scaffolds without MNPs to 20.7 ± 4.7% for scaffolds with 4 mg/mL MNPs. Osteoblast cells seeded on scaffolds exposed to static magnetic field of 1.3 T deformed the regular architecture of the scaffolds and evoked faster mineralization in comparison to unstimulated samples. Scaffolds deformation and extracellular matrix mineralization under static magnetic field (SMF) exposure increased with increasing MNPs concentration. The results are discussed in the frame of gradient magnetic fields of ~3 × 10−4 T/m generated by MNPs over the cells bodies.
Collapse
Affiliation(s)
- Irina Alexandra Paun
- Center for Advanced Laser Technologies (CETAL), National Institute for Laser, Plasma and Radiation Physics, RO-077125 Magurele-Ilfov, Romania.
- Physics Department, Faculty of Applied Sciences, University Politehnica of Bucharest, RO-060042 Bucharest, Romania.
| | - Bogdan Stefanita Calin
- Center for Advanced Laser Technologies (CETAL), National Institute for Laser, Plasma and Radiation Physics, RO-077125 Magurele-Ilfov, Romania
- Physics Department, Faculty of Applied Sciences, University Politehnica of Bucharest, RO-060042 Bucharest, Romania
| | - Cosmin Catalin Mustaciosu
- Horia Hulubei National Institute for Physics and Nuclear Engineering IFIN-HH, RO-077125 Magurele-Ilfov, Romania
| | - Mona Mihailescu
- Physics Department, Faculty of Applied Sciences, University Politehnica of Bucharest, RO-060042 Bucharest, Romania
| | - Antoniu Moldovan
- National Institute for Laser, Plasma and Radiation Physics, RO-077125 Magurele-Ilfov, Romania
| | - Ovidiu Crisan
- National Institute of Materials Physics, RO-077125 Magurele-Ilfov, Romania
| | - Aurel Leca
- National Institute of Materials Physics, RO-077125 Magurele-Ilfov, Romania
| | - Catalin Romeo Luculescu
- Center for Advanced Laser Technologies (CETAL), National Institute for Laser, Plasma and Radiation Physics, RO-077125 Magurele-Ilfov, Romania
| |
Collapse
|
14
|
Faria VRD, Pinho RO, Camilo BS, Guimarães JD, Fonseca E Silva F, Lopes PS, Silva PV, Teixeira SA, Veroneze R, Penitente-Filho JM, Guimarães SEF. Genes expression and phenotypic differences in corpus luteum and cumulus cells of commercial line and piau breed gilts. Theriogenology 2019; 136:111-117. [PMID: 31254724 DOI: 10.1016/j.theriogenology.2019.06.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 11/27/2022]
Abstract
We aimed to characterize the expression of angiogenesis-related genes in corpus luteum (CL) and cumulus cells (CC) during estrous cycle in gilts of different genetic groups, as well as to study the relation between gene expression and phenotypic data. Forty five gilts were used as follows: L1, Commercial Line 1 (Large White x Landrace x Duroc) (n = 15); L2, Commercial Line 2 (Large White x Landrace x Pietrain) (n = 15); and Piau, Piau breed gilts (n = 15). Estrus observation started from 120 days of age. After the second observed estrus females were slaughtered (n = 3) on days 3, 5, 10, 14 and 18 of estrous cycle (first day of estrous cycle as Day 0). CL sampling was performed on days 3, 5, 10 and 14 and collection of CC and follicular fluid on days 14 and 18. Follicular fluid was used for analysis of estradiol levels and CC and CL samples for analysis of angiogenesis-related genes expression, ANGPT-1/-2 and TEK in CC and MMP-2, VEGFA, VEGFR-1/-2, ANGPT-1/-2 and TEK in CL. Piau gilts showed lower ovulation rate than both L1 and L2 gilts (P < 0.05), lower number of large antral follicles (>6 mm) at 18 days than L2 gilts (P < 0.05), and smaller diameter of the largest follicles at 14 days than L1 gilts (P < 0.05). Piau and L2 gilts showed higher estradiol levels in follicular fluid on day 18. Expression of ANGPT-1 and -2 genes in CC did not differ among genetic groups neither among days of the estrous cycle, but TEK gene expression was higher in L1 than L2 gilts on day 18. Expression of VEGFA, VEGFR-2 and MMP-2 genes in CL did not differ among genetic groups and days of cycle, but VEGFR-1 expression was higher in Piau than L2 gilts on days 10 and 14, and it was higher in L1 than L2 gilts on day 14. The ANGPT-1/-2 and TEK genes expression in CL were significantly higher in Piau than L1 gilts on day 10. The ANGPT-2/ANGPT-1 gene expression ratio in CL was higher in L1 than Piau and L2 gilts at 14 days, suggesting a shorter luteal phase for L1 gilts. Results indicated differences among genetic groups for the pattern of the angiogenesis-related genes expression in CL along estrous cycles, which may be reflected in phenotypic traits such as ovulation rate, estradiol levels in follicular fluid and number and diameter of antral follicles.
Collapse
Affiliation(s)
| | | | - Breno Soares Camilo
- Department of Veterinary, Universidade Federal de Viçosa, Viçosa, MG, Brazil.
| | | | | | - Paulo Sávio Lopes
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, MG, Brazil.
| | | | | | - Renata Veroneze
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, MG, Brazil.
| | | | | |
Collapse
|
15
|
Zheng LC, Wang XQ, Lu K, Deng XL, Zhang CW, Luo H, Xu XD, Chen XM, Yan L, Wang YQ, Shi SL. Ephrin-B2/Fc promotes proliferation and migration, and suppresses apoptosis in human umbilical vein endothelial cells. Oncotarget 2018; 8:41348-41363. [PMID: 28489586 PMCID: PMC5522204 DOI: 10.18632/oncotarget.17298] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 04/03/2017] [Indexed: 01/12/2023] Open
Abstract
Tumor growth and metastasis are angiogenesis dependent. Angiogenic growth involves endothelial cell proliferation, migration, and invasion. Ephrin-B2 is a ligand for Eph receptor tyrosine kinases and is an important mediator in vascular endothelial growth factor-mediated angiogenesis. However, research offer controversial information regarding effects of ephrin-B2 on vascular endothelial cells. In this paper, proteome analyses showed that ephrin-B2/Fc significantly activates multiple signaling pathways related to cell proliferation, survival, and migration and suppresses apoptosis and cell death. Cytological experiments further confirm that ephrin-B2/Fc stimulates endothelial cell proliferation, triggers dose-dependent migration, and suppresses cell apoptosis. Results demonstrate that soluble dose-dependent ephrinB2 can promote proliferation and migration and inhibit apoptosis of human umbilical vein endothelial cells. These results also suggest that ephrinB2 prevents ischemic disease and can potentially be a new therapeutic target for treating angiogenesis-related diseases and tumors.
Collapse
Affiliation(s)
- Li-Chun Zheng
- Medical College of Xiamen University, Jinshan Community Health Service Center, Xiamen Traditional Chinese Medical Hospital, Xiamen 361000, P.R. China.,Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Xiao-Qing Wang
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Kun Lu
- Department of Basic Medicine, Medical College of Xiamen University, Cancer Research Center of Xiamen University, Xiamen 361102, P.R. China
| | - Xiao-Ling Deng
- Department of Basic Medicine, Medical College of Xiamen University, Cancer Research Center of Xiamen University, Xiamen 361102, P.R. China
| | - Cheng-Wei Zhang
- Department of Cardiology, Affiliated Dongnan Hospital of Xiamen University, Zhangzhou 363000, P.R. China
| | - Hong Luo
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Xu-Dong Xu
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Xiao-Man Chen
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Lu Yan
- Department of Basic Medicine, Medical College of Xiamen University, Xiamen 361102, P.R. China
| | - Yi-Qing Wang
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Song-Lin Shi
- Department of Basic Medicine, Medical College of Xiamen University, Cancer Research Center of Xiamen University, Xiamen 361102, P.R. China
| |
Collapse
|
16
|
Lee SR, Nilius B, Han J. Gaseous Signaling Molecules in Cardiovascular Function: From Mechanisms to Clinical Translation. Rev Physiol Biochem Pharmacol 2018; 174:81-156. [PMID: 29372329 DOI: 10.1007/112_2017_7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Carbon monoxide (CO), hydrogen sulfide (H2S), and nitric oxide (NO) constitute endogenous gaseous molecules produced by specific enzymes. These gases are chemically simple, but exert multiple effects and act through shared molecular targets to control both physiology and pathophysiology in the cardiovascular system (CVS). The gases act via direct and/or indirect interactions with each other in proteins such as heme-containing enzymes, the mitochondrial respiratory complex, and ion channels, among others. Studies of the major impacts of CO, H2S, and NO on the CVS have revealed their involvement in controlling blood pressure and in reducing cardiac reperfusion injuries, although their functional roles are not limited to these conditions. In this review, the basic aspects of CO, H2S, and NO, including their production and effects on enzymes, mitochondrial respiration and biogenesis, and ion channels are briefly addressed to provide insight into their biology with respect to the CVS. Finally, potential therapeutic applications of CO, H2S, and NO with the CVS are addressed, based on the use of exogenous donors and different types of delivery systems.
Collapse
Affiliation(s)
- Sung Ryul Lee
- Department of Convergence Biomedical Science, Cardiovascular and Metabolic Disease Center, College of Medicine, Inje University, Busan, Republic of Korea
| | - Bernd Nilius
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea.
| |
Collapse
|
17
|
Migrino RQ, Davies HA, Truran S, Karamanova N, Franco DA, Beach TG, Serrano GE, Truong D, Nikkhah M, Madine J. Amyloidogenic medin induces endothelial dysfunction and vascular inflammation through the receptor for advanced glycation endproducts. Cardiovasc Res 2017; 113:1389-1402. [PMID: 28859297 PMCID: PMC6676393 DOI: 10.1093/cvr/cvx135] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/17/2017] [Accepted: 07/07/2017] [Indexed: 12/25/2022] Open
Abstract
AIMS Medin is a common amyloidogenic protein in humans that accumulates in arteries with advanced age and has been implicated in vascular degeneration. Medin's effect on endothelial function remains unknown. The aims are to assess medin's effects on human arteriole endothelial function and identify potential mechanisms underlying medin-induced vascular injury. METHODS AND RESULTS Ex vivo human adipose and leptomeningeal arterioles were exposed (1 h) to medin (0.1, 1, or 5 µM) without or with FPS-ZM1 [100 µM, receptor for advanced glycation endproducts (RAGE)-specific inhibitor] and endothelium-dependent function (acetylcholine dilator response) and endothelium-independent function (dilator response to nitric oxide donor diethylenetriamine NONOate) were compared with baseline control. Human umbilical vein endothelial cells were exposed to medin without or with FPS-ZM1 and oxidative and nitrative stress, cell viability, and pro-inflammatory signaling measures were obtained. Medin caused impaired endothelial function (vs. baseline response: -45.2 ± 5.1 and -35.8 ± 7.9% in adipose and leptomeningeal arterioles, respectively, each P < 0.05). Dilator response to NONOate was not significantly changed. Medin decreased arteriole and endothelial cell nitric oxide production, increased superoxide production, reduced endothelial cell viability, proliferation, and migration. Medin increased gene and protein expression of interleukin-6 and interleukin-8 via activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB). Medin-induced endothelial dysfunction and oxidative stress were reversed by antioxidant polyethylene glycol superoxide dismutase and by RAGE inhibitor FPS-ZM1. CONCLUSIONS Medin causes human microvascular endothelial dysfunction through oxidative and nitrative stress and promotes pro-inflammatory signaling in endothelial cells. These effects appear to be mediated via RAGE. The findings represent a potential novel mechanism of vascular injury.
Collapse
Affiliation(s)
- Raymond Q. Migrino
- Office of Research, Phoenix Veterans Affairs Health Care System, 650 E. Indian School Road, Phoenix, AZ 85022, USA
- Department of Medicine, University of Arizona College of Medicine-Phoenix, AZ, USA
| | - Hannah A. Davies
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Seth Truran
- Office of Research, Phoenix Veterans Affairs Health Care System, 650 E. Indian School Road, Phoenix, AZ 85022, USA
| | - Nina Karamanova
- Office of Research, Phoenix Veterans Affairs Health Care System, 650 E. Indian School Road, Phoenix, AZ 85022, USA
| | - Daniel A. Franco
- Office of Research, Phoenix Veterans Affairs Health Care System, 650 E. Indian School Road, Phoenix, AZ 85022, USA
| | - Thomas G. Beach
- Department of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Geidy E. Serrano
- Department of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Danh Truong
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Jillian Madine
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|
18
|
Mao H, Xie L, Pi X. Low-Density Lipoprotein Receptor-Related Protein-1 Signaling in Angiogenesis. Front Cardiovasc Med 2017; 4:34. [PMID: 28589128 PMCID: PMC5438976 DOI: 10.3389/fcvm.2017.00034] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/01/2017] [Indexed: 11/13/2022] Open
Abstract
Low-density lipoprotein receptor-related protein-1 (LRP1) plays multifunctional roles in lipid homeostasis, signaling transduction, and endocytosis. It has been recognized as an endocytic receptor for many ligands and is involved in the signaling pathways of many growth factors or cytokines. Dysregulation of LRP1-dependent signaling events contributes to the development of pathophysiologic processes such as Alzheimer’s disease, atherosclerosis, inflammation, and coagulation. Interestingly, recent studies have linked LRP1 with endothelial function and angiogenesis, which has been underappreciated for a long time. During zebrafish embryonic development, LRP1 is required for the formation of vascular network, especially for the venous development. LRP1 depletion in the mouse embryo proper leads to angiogenic defects and disruption of endothelial integrity. Moreover, in a mouse oxygen-induced retinopathy model, specific depletion of LRP1 in endothelial cells results in abnormal development of neovessels. These loss-of-function studies suggest that LRP1 plays a pivotal role in angiogenesis. The review addresses the recent advances in the roles of LRP1-dependent signaling during angiogenesis.
Collapse
Affiliation(s)
- Hua Mao
- Department of Medicine, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Liang Xie
- Department of Medicine, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Xinchun Pi
- Department of Medicine, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
19
|
Chao J, Li P, Chao L. Kallistatin suppresses cancer development by multi-factorial actions. Crit Rev Oncol Hematol 2017; 113:71-78. [PMID: 28427524 PMCID: PMC5441310 DOI: 10.1016/j.critrevonc.2017.03.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 02/17/2017] [Accepted: 03/11/2017] [Indexed: 01/07/2023] Open
Abstract
Kallistatin was first identified in human plasma as a tissue kallikrein-binding protein and a serine proteinase inhibitor. Kallistatin via its two structural elements regulates differential signaling cascades, and thus a wide spectrum of biological functions. Kallistatin's active site is essential for: inhibiting tissue kallikrein's activity; stimulating endothelial nitric oxide synthase and sirtuin 1 expression and activation; and modulating the synthesis of the microRNAs, miR-34a, miR-21 and miR-203. Kallistatin's heparin-binding site is crucial for antagonizing the signaling pathways of vascular endothelial growth factor, tumor necrosis factor-α, Wnt, transforming growth factor-β and epidermal growth factor. Circulating kallistatin levels are markedly reduced in patients with prostate and colon cancer. Kallistatin administration attenuates angiogenesis, inflammation, tumor growth and invasion in animal models and cultured cells. Therefore, tumor progression may be substantially suppressed by kallistatin's pleiotropic activities. In this review, we will discuss the role and mechanisms of kallistatin in the regulation of cancer development.
Collapse
Affiliation(s)
- Julie Chao
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA.
| | - Pengfei Li
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Lee Chao
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
20
|
Zonta YR, Martinez M, Camargo ICC, Domeniconi RF, Lupi Júnior LA, Pinheiro PFF, Reiter RJ, Martinez FE, Chuffa LGA. Melatonin Reduces Angiogenesis in Serous Papillary Ovarian Carcinoma of Ethanol-Preferring Rats. Int J Mol Sci 2017; 18:ijms18040763. [PMID: 28398226 PMCID: PMC5412347 DOI: 10.3390/ijms18040763] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 03/24/2017] [Accepted: 03/30/2017] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis is a hallmark of ovarian cancer (OC); the ingrowth of blood vessels promotes rapid cell growth and the associated metastasis. Melatonin is a well-characterized indoleamine that possesses important anti-angiogenic properties in a set of aggressive solid tumors. Herein, we evaluated the role of melatonin therapy on the angiogenic signaling pathway in OC of an ethanol-preferring rat model that mimics the same pathophysiological conditions occurring in women. OC was chemically induced with a single injection of 7,12-dimethylbenz(a)anthracene (DMBA) under the ovarian bursa. After the rats developed serous papillary OC, half of the animals received intraperitoneal injections of melatonin (200 µg/100 g body weight/day) for 60 days. Melatonin-treated animals showed a significant reduction in OC size and microvessel density. Serum levels of melatonin were higher following therapy, and the expression of its receptor MT1 was significantly increased in OC-bearing rats, regardless of ethanol intake. TGFβ1, a transforming growth factor-beta1, was reduced only after melatonin treatment. Importantly, vascular endothelial growth factor (VEGF) was severely reduced after melatonin therapy in animals given or not given ethanol. Conversely, the levels of VEGF receptor 1 (VEGFR1) was diminished after ethanol consumption, regardless of melatonin therapy, and VEGFR2 was only reduced following melatonin. Hypoxia-inducible factor (HIF)-1α was augmented with ethanol consumption, and, notably, melatonin significantly reduced their levels. Collectively, our results suggest that melatonin attenuates angiogenesis in OC in an animal model of ethanol consumption; this provides a possible complementary therapeutic opportunity for concurrent OC chemotherapy.
Collapse
MESH Headings
- Alcohol Drinking/physiopathology
- Animals
- Antioxidants/administration & dosage
- Antioxidants/pharmacology
- Blotting, Western
- Cystadenocarcinoma, Papillary/blood supply
- Cystadenocarcinoma, Papillary/drug therapy
- Cystadenocarcinoma, Papillary/metabolism
- Cystadenocarcinoma, Serous/blood supply
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/metabolism
- Ethanol/administration & dosage
- Female
- Food Preferences
- Immunohistochemistry
- Injections, Intraperitoneal
- Melatonin/administration & dosage
- Melatonin/pharmacology
- Microscopy, Fluorescence
- Neovascularization, Pathologic/prevention & control
- Ovarian Neoplasms/blood supply
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/metabolism
- Rats
- Receptor, Melatonin, MT1/metabolism
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor Receptor-1/metabolism
Collapse
Affiliation(s)
- Yohan Ricci Zonta
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Marcelo Martinez
- Department of Morphology and Pathology, Universidade Federal de São Carlos (UFSCar), São Carlos-SP 13565-905, Brazil.
| | - Isabel Cristina C Camargo
- Department of Biotechnology, School of Sciences, Humanities and Languages, São Paulo State University (UNESP), Assis-SP 19806-900, Brazil.
| | - Raquel F Domeniconi
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Luiz Antonio Lupi Júnior
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Patricia Fernanda F Pinheiro
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX 78229, USA.
| | - Francisco Eduardo Martinez
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Luiz Gustavo A Chuffa
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| |
Collapse
|
21
|
Nusrat O, Belotte J, Fletcher NM, Memaj I, Saed MG, Diamond MP, Saed GM. The Role of Angiogenesis in the Persistence of Chemoresistance in Epithelial Ovarian Cancer. Reprod Sci 2016; 23:1484-1492. [PMID: 27122375 DOI: 10.1177/1933719116645191] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Chemoresistance remains a major challenge in the treatment of ovarian cancer. As part of a survival mechanism, tumor cells have been shown to release proangiogenic factors, such as vascular endothelial growth factor (VEGF), through a mechanism that involves the upregulation of hypoxia-induced factor (HIF)-1α. The objective of this study was to compare the expression of VEGF and its receptors (R1 and R2) as well as HIF-1α in chemoresistant epithelial ovarian cancer (EOC) cells to their chemosensitive counterparts and determine their impact on angiogenesis. METHODS Two human EOC cell lines, MDAH-2774 and SKOV-3, and their cisplatin- or taxotere-resistant counterparts were used. Total RNA and protein were subjected to real-time reverse transcriptase-polymerase chain reaction, immunoprecipitation/Western blot and enzyme-linked immunosorbent assay to evaluate the expression of VEGF, VEGF receptors (R1 and R2), and HIF-1α. Angiogenesis was assessed with an in vitro angiogenesis assay. Data were analyzed using independent Student t tests and chi-square. RESULTS Both taxotere- and cisplatin-resistant MDAH-2774 and SKOV-3 EOC cell lines manifested a significant decrease in VEGF, VEGF receptors, HIF-1α messenger RNA, and protein levels as compared to their chemosensitive counterparts. There was a significant decrease in the number and thickness of polygon blood vessel formation in chemoresistant EOC cells compared to chemosensitive counterparts. CONCLUSION Cisplatin- and taxotere-resistant EOC cells are characterized by lower VEGF, VEGF receptors, and HIF-1α, and decreased angiogenesis. These findings may indicate a decrease in drug delivery at the tumor site, hence allowing the persistence of chemoresistant EOC cells.
Collapse
Affiliation(s)
- Osama Nusrat
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jimmy Belotte
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nicole M Fletcher
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ira Memaj
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mohammed G Saed
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA
| | - Michael P Diamond
- Department of Obstetrics and Gynecology, Georgia Regents University, Augusta, GA, USA
| | - Ghassan M Saed
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
22
|
Talaat RM, Abo-El-Atta AS, Farou SM, El-Dosoky KI. Therapeutic effect of dimethyl dimethoxy biphenyl dicarboxylate on collagen-induced arthritis in rats. Chin J Integr Med 2015; 21:846-854. [PMID: 24584757 DOI: 10.1007/s11655-014-1746-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To study the effect of oral administration of dimethyl dimethoxy biphenyl dicarboxylate (DDB) on adjusting angiogeneic/inflammatory mediators and ameliorating the pathology of bones in rats with collagen-induced arthritis (CIA). METHODS Wistar rat model of CIA was set up using bovine collagen type II. Fifty rats were divided into five groups randomly: normal, CIA model, DDB treatment, methotrexate (MTX) treatment, and combined DDB+MTX treatment. Ankle joints of rats were imaged with digital X-ray machine to show the destruction of joints. Fore and hind paw and knee joints were removed above the ankle joint then processed for haematoxylin and eosin staining. Plasma levels of vascular endothelial growth factor (VEGF), platelet derived growth factor, interleukin-8 (IL-8), IL-4, tumor necrosis factor α (TNF-α), and cyclooxygenase-2 (COX-2) were quantified by enzyme-linked immunosorbent assay. Nitric oxide levels were detected by Griess reagent. RESULTS Compared with the CIA model group, a remarkable reduction in various angiogenic (VEGF and IL-8) and inflammatory mediators (TNF-α, IL-4 and COX-2) after treatment with DDB either alone or combined with MTX P<0.05 or P<0.01). Histopathological and X-ray findings were confirmatory to the observed DDB anti-arthritic effect. The DDB-treated group showed amelioration in signs of arthritis which appeared essentially similar to normal. CONCLUSION Our data shed light on the therapeutic efficacy of DDB in experimental rheumatoid arthritis (RA) compared with a choice drug (MTX) and it may be offered as a second-line drug in the treatment of RA.
Collapse
Affiliation(s)
- Roba M Talaat
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute, Menofia University, Sadat Branch, Sadat, 32897, Egypt.
| | - Amira S Abo-El-Atta
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute, Menofia University, Sadat Branch, Sadat, 32897, Egypt
| | - Sabah M Farou
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute, Menofia University, Sadat Branch, Sadat, 32897, Egypt
| | - Karima I El-Dosoky
- Pathology Department, Faculty of Medicine, Tanta University, Tanta, 31111, Egypt
| |
Collapse
|
23
|
Romero M, Zanuy M, Rosell E, Cascante M, Piulats J, Font-Bardia M, Balzarini J, De Clerq E, Pujol MD. Optimization of xanthatin extraction from Xanthium spinosum L. and its cytotoxic, anti-angiogenesis and antiviral properties. Eur J Med Chem 2015; 90:491-6. [PMID: 25481815 PMCID: PMC7115430 DOI: 10.1016/j.ejmech.2014.11.060] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 11/10/2014] [Accepted: 11/29/2014] [Indexed: 11/26/2022]
Abstract
The aqueous extraction of the sesquiterpene lactone xanthatin from Xanthium spinosum L. favours the conversion of xanthinin (1) to xanthatin (2) via the loss of acetic acid. The cytotoxic (Hep-G2 and L1210 human cell lines) and antiviral activities of isolated xanthatin are established. This natural compound shows significant cytotoxicity against the Hep-G2 cell line and our experimental results reveal its strong anti-angiogenesis capacity in vitro. The structure of xanthatin is determined by spectroscopic methods and for the first time confirmed by X-ray diffraction.
Collapse
Affiliation(s)
- M Romero
- Laboratory of Pharmaceutical Chemistry (associated to CSIC), Faculty of Pharmacy, University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain
| | - M Zanuy
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
| | - E Rosell
- Oryzon, Parc Cientific de Barcelona, Baldiri Reixac, 12, 08028 Barcelona, Spain
| | - M Cascante
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
| | - J Piulats
- Oryzon, Parc Cientific de Barcelona, Baldiri Reixac, 12, 08028 Barcelona, Spain
| | - M Font-Bardia
- Cristal.lografia, Mineralogia i Dipòsits Minerals, Universitat de Barcelona, Martí i Franquès s/n, 08028 Barcelona, Spain
| | - J Balzarini
- Rega Institute for Medical Research, Katholieke Universiteit Leuve, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| | - E De Clerq
- Rega Institute for Medical Research, Katholieke Universiteit Leuve, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| | - M D Pujol
- Laboratory of Pharmaceutical Chemistry (associated to CSIC), Faculty of Pharmacy, University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain.
| |
Collapse
|
24
|
Moon JH, Lee JW, Kang JK. Angiogenic effects of recombinant thymosin β4 in a mouse hindlimb ischemia model. J Biomed Res 2014. [DOI: 10.12729/jbr.2014.15.4.156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
25
|
The beneficial effect of melatonin in brain endothelial cells against oxygen-glucose deprivation followed by reperfusion-induced injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:639531. [PMID: 25126203 PMCID: PMC4122057 DOI: 10.1155/2014/639531] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/07/2014] [Accepted: 06/20/2014] [Indexed: 12/31/2022]
Abstract
Melatonin has a cellular protective effect in cerebrovascular and neurodegenerative diseases. Protection of brain endothelial cells against hypoxia and oxidative stress is important for treatment of central nervous system (CNS) diseases, since brain endothelial cells constitute the blood brain barrier (BBB). In the present study, we investigated the protective effect of melatonin against oxygen-glucose deprivation, followed by reperfusion- (OGD/R-) induced injury, in bEnd.3 cells. The effect of melatonin was examined by western blot analysis, cell viability assays, measurement of intracellular reactive oxygen species (ROS), and immunocytochemistry (ICC). Our results showed that treatment with melatonin prevents cell death and degradation of tight junction protein in the setting of OGD/R-induced injury. In response to OGD/R injury of bEnd.3 cells, melatonin activates Akt, which promotes cell survival, and attenuates phosphorylation of JNK, which triggers apoptosis. Thus, melatonin protects bEnd.3 cells against OGD/R-induced injury.
Collapse
|
26
|
Dworacka M, Krzyżagórska E, Wesołowska A, Zharmakhanova G, Iskakova S, Dworacki G. Circulating monocyte chemotactic protein 1 (MCP-1), vascular cell adhesion molecule 1 (VCAM-1) and angiogenin in type 2 diabetic patients treated with statins in low doses. Eur J Pharmacol 2014; 740:474-9. [PMID: 24991787 DOI: 10.1016/j.ejphar.2014.06.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 06/19/2014] [Accepted: 06/20/2014] [Indexed: 01/29/2023]
Abstract
Statins are known as agents promoting a biphasic dose-dependent effect on angiogenesis under experimental conditions. Dysregulation of angiogenesis plays an important role in the development of atherosclerosis and it may be affected by metabolic factors. The aim of this research was to explain how low doses of statins modify serum concentrations of pro-angiogenic factors MCP-1 and angiogenin in type 2 diabetic patients. Measurements of metabolic control parameters were performed in 30 patients with type 2 diabetes treated with low doses of statin, and in 34 statin-free patients with type 2 diabetes. The serum levels of MCP-1 and VCAM-1 in statin-treated patients were lower than those of the statin-free group. ANCOVA results revealed that these effects were dependent only on the use of statins. In type 2 diabetic subjects, overall positive correlation was found between total cholesterol or LDL serum concentration and MCP-1 serum level. The angiogenin concentration in the serum did not show differences and was comparable in both groups. The angiogenin serum level correlated negatively with HDL, LDL and with HbA1c. Multivariate regression analysis indicated that angiogenin serum levels in type 2 diabetic patients were determined mainly by HbA1c, HDL-cholesterol and diabetes duration. It has been shown that statins used in low doses in type 2 diabetic subjects decrease MCP-1 and VCAM-1serum levels, most likely due to the statins-related effect on the lipid profile, while angiogenin serum levels in this group are determined rather by the current metabolic control.
Collapse
Affiliation(s)
- Marzena Dworacka
- Department of Pharmacology Poznan University of Medical Sciences, Rokietnicka 5a, 60-805 Poznań, Poland.
| | - Ewa Krzyżagórska
- Poznan Specialist Center of Medical Care, Diabetology Out-patient Clinic, Poznan, Poland.
| | - Anna Wesołowska
- Department of Pharmacology Poznan University of Medical Sciences, Rokietnicka 5a, 60-805 Poznań, Poland.
| | - Gulmira Zharmakhanova
- Department of Pharmacology, West Kazakhstan State Medical University, Maresev str. 68, Aktobe, Kazakhstan.
| | - Saule Iskakova
- Department of Pharmacology, West Kazakhstan State Medical University, Maresev str. 68, Aktobe, Kazakhstan.
| | - Grzegorz Dworacki
- Department of Clinical Immunology Poznan University of Medical Sciences, Rokietnicka 5d, 60-805 Poznan, Poland.
| |
Collapse
|
27
|
Huang K, Huang X, Xiao G, Yang H, Lin J, Diao Y. Kallistatin, a novel anti-angiogenesis agent, inhibits angiogenesis via inhibition of the NF-κB signaling pathway. Biomed Pharmacother 2014; 68:455-61. [DOI: 10.1016/j.biopha.2014.03.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 03/04/2014] [Indexed: 10/25/2022] Open
|
28
|
Bir SC, Pattillo CB, Pardue S, Kolluru GK, Shen X, Giordano T, Kevil CG. Nitrite anion therapy protects against chronic ischemic tissue injury in db/db diabetic mice in a NO/VEGF-dependent manner. Diabetes 2014; 63:270-81. [PMID: 24009258 PMCID: PMC4179307 DOI: 10.2337/db13-0890] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Nitrite anion has been demonstrated to be a prodrug of nitric oxide (NO) with positive effects on tissue ischemia/reperfusion injury, cytoprotection, and vasodilation. However, effects of nitrite anion therapy for ischemic tissue vascular remodeling during diabetes remain unknown. We examined whether sodium nitrite therapy altered ischemic revascularization in BKS-Lepr(db/db) mice subjected to permanent unilateral femoral artery ligation. Sodium nitrite therapy completely restored ischemic hind limb blood flow compared with nitrate or PBS therapy. Importantly, delayed nitrite therapy 5 days after ischemia restored ischemic limb blood flow in aged diabetic mice. Restoration of blood flow was associated with increases in ischemic tissue angiogenesis activity and cell proliferation. Moreover, nitrite but not nitrate therapy significantly prevented ischemia-mediated tissue necrosis in aged mice. Nitrite therapy significantly increased ischemic tissue vascular endothelial growth factor (VEGF) protein expression that was essential for nitrite-mediated reperfusion of ischemic hind limbs. Nitrite significantly increased ischemic tissue NO bioavailability along with concomitant reduction of superoxide formation. Lastly, nitrite treatment also significantly stimulated hypoxic endothelial cell proliferation and migration in the presence of high glucose in an NO/VEGF-dependent manner. These results demonstrate that nitrite therapy effectively stimulates ischemic tissue vascular remodeling in the setting of metabolic dysfunction that may be clinically useful.
Collapse
Affiliation(s)
- Shyamal C. Bir
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | - Christopher B. Pattillo
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | - Sibile Pardue
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | - Gopi K. Kolluru
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | - Xinggui Shen
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | | | - Christopher G. Kevil
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
- TheraVasc Inc., Cleveland, OH
- Corresponding author: Christopher G. Kevil,
| |
Collapse
|
29
|
de Toledo-Piza AR, Maria DA. Angiogenesis enhanced byPhyllocaulis boraceiensismucus in human cells. FEBS J 2013; 280:5118-27. [DOI: 10.1111/febs.12487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 08/02/2013] [Accepted: 08/12/2013] [Indexed: 01/04/2023]
Affiliation(s)
| | - Durvanei A. Maria
- Laboratory of Biochemistry and Biophysics; Butantan Institute; São Paulo; Brazil
| |
Collapse
|
30
|
Abstract
Molecular imaging fundamentally changes the way we look at cancer. Imaging paradigms are now shifting away from classical morphological measures towards the assessment of functional, metabolic, cellular, and molecular information in vivo. Interdisciplinary driven developments of imaging methodology and probe molecules utilizing animal models of human cancers have enhanced our ability to non-invasively characterize neoplastic tissue and follow anti-cancer treatments. Preclinical molecular imaging offers a whole palette of excellent methodology to choose from. We will focus on positron emission tomography (PET) and magnetic resonance imaging (MRI) techniques, since they provide excellent and complementary molecular imaging capabilities and bear high potential for clinical translation. Prerequisites and consequences of using animal models as surrogates of human cancers in preclinical molecular imaging are outlined. We present physical principles, values and limitations of PET and MRI as molecular imaging modalities and comment on their high potential to non-invasively assess information on hypoxia, angiogenesis, apoptosis, gene expression, metabolism, and cell trafficking in preclinical cancer research.
Collapse
Affiliation(s)
- Gunter Wolf
- University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.
| | | |
Collapse
|
31
|
Endogenous and induced angiogenic characteristics of human chorion-derived stem cells. Cell Biol Int 2012; 36:1145-53. [DOI: 10.1042/cbi20120044] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
32
|
Elevated angiopoietin-1 serum levels in patients with Alzheimer's disease. Int J Alzheimers Dis 2012; 2012:324016. [PMID: 23094194 PMCID: PMC3474986 DOI: 10.1155/2012/324016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 09/13/2012] [Indexed: 12/17/2022] Open
Abstract
Background. Alzheimer's disease (AD) is the most common cause of dementia in the elderly. AD is characterized by the accumulation of amyloid plaques and neurofibrillary tangles and by massive neuronal loss in the brain. There is epidemiologic and pathologic evidence that AD is associated with vascular risk factors and vascular diseases, contributing to cerebral hypoperfusion with consecutive stimulation of angiogenesis and upregulation of proangiogenic factors such as Angiopoietin-1 (Ang-1). Methods. In the present study, we measured Ang-1 serum levels in 42 patients with AD, 20 patients with mild cognitive impairment (MCI), and in 40 healthy elderly controls by ELISA. Results. We found significantly increased Ang-1 serum levels in patients with AD compared to control subjects (P = 0.003). There was no significant difference between MCI patients and healthy controls (P = 0.553) or between AD and MCI patients (P = 0.054). The degree of cognitive impairment as measured by the mini-mental status examination (MMSE) score was significantly correlated with the Ang-1 serum levels in all patients and healthy controls. Conclusions. We found significantly increased Ang-1 serum levels in AD patients. We could also show an association between Ang-1 serum levels and the cognitive status in all patients and healthy controls. Thus, serum Ang-1 could be a potential candidate for a biomarker panel for AD diagnosis.
Collapse
|
33
|
Novak S, Paradis F, Patterson JL, Pasternak JA, Oxtoby K, Moore HS, Hahn M, Dyck MK, Dixon WT, Foxcroft GR. Temporal candidate gene expression in the sow placenta and embryo during early gestation and effect of maternal Progenos supplementation on embryonic and placental development. Reprod Fertil Dev 2012; 24:550-8. [PMID: 22541543 DOI: 10.1071/rd10312] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 09/20/2011] [Indexed: 11/23/2022] Open
Abstract
The present study characterised gene expression associated with embryonic muscle development and placental vascularisation during early gestation in the pig and examined effects of Progenos supplementation in early pregnancy. Tissues were collected from commercial multiparous sows (n = 48) from Days 16 to 49 of gestation. In the placenta, qPCR revealed that vascular endothelial growth factor (VEGFA) expression did not change from Day 17 to 49 of gestation; however, KDR receptor and angiopoietin-1 and -2 expression were differentially regulated, with periods of high expression corresponding to two critical phases of angiogenesis in the pig. In the embryo, the pattern of myogenesis-related gene expression was consistent with available literature. A commercially available nutritional supplement Progenos (20 g day⁻¹ L-arginine) added to the diet of sows from either Day 15 to 29 (P15-29; n = 33), Day 30 to 44 (n = 29) or from Day 15 to 44 (n = 76) of gestation tended to increase (P = 0.058) embryonic growth rate compared with non-supplemented controls (n = 79) and angiogenin expression was higher (P = 0.028) at Day 30 of gestation in placentae from sows on the P15-29 Progenos treatment. These results are consistent with proposed beneficial effects of l-arginine on early embryonic development and placental vascularisation.
Collapse
Affiliation(s)
- S Novak
- Swine Reproduction-Development Program, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kobus K, Kopycinska J, Kozlowska-Wiechowska A, Urasinska E, Kempinska-Podhorodecka A, Haas TL, Milkiewicz P, Milkiewicz M. Angiogenesis within the duodenum of patients with cirrhosis is modulated by mechanosensitive Kruppel-like factor 2 and microRNA-126. Liver Int 2012; 32:1222-32. [PMID: 22574900 DOI: 10.1111/j.1478-3231.2012.02791.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 02/22/2012] [Indexed: 12/22/2022]
Abstract
BACKGROUND The mechanism involved in neovascularization in splanchnic circulation and the main trigger that induces angiogenesis in patients with cirrhosis are not fully recognized. AIMS To explore the involvement of flow sensitive lung Kruppel-like factor (KLF2), microRNA-126 (miR-126), angiopoietin-2 (Ang-2) and heme oxygenase-1 (HO-1) in modulation of vascular endothelial growth factor (VEGF) signalling that have a critical effect on growth of new blood vessels. METHODS Duodenal biopsies from 22 patients with cirrhosis and 10 controls were obtained during routine endoscopy. The process of angiogenesis was evaluated by a measurement of CD31 concentration, immunodetection of CD34 protein and estimation of capillary densities. Messenger RNA (mRNA) and protein expressions were analysed by real-time PCR, Western blot or ELISA respectively. RESULTS Markers of angiogenesis (both, CD31 and CD34) were significantly enhanced in cirrhotic patients. In comparison to healthy controls, levels of Ang-2 and KLF-2 mRNAs as well as Ang-2, KLF-2, HO-1, VEGF protein expressions were considerably increased. Levels of sCD163, a surrogate marker of portal hypertension, correlated with levels of Ang-2, (P = 0.021) and VEGF (P = 0.009). The expression of miR-126, a KLF2-mediated regulator of the VEGF signalling was enhanced in cirrhotic patients. CONCLUSIONS Our results demonstrate, for the first time in humans, that neovascularization is induced in duodenal tissue of patients with cirrhosis and proangiogenic factors such as KLF-2, Ang-2, miR-126 and VEGF can contribute to the angiogenesis induced by hemodynamic forces. Thus, cirrhosis-induced blood flow and pressure within splanchnic vessels may be important hemodynamic triggers that initiate the angiogenic signalling cascade.
Collapse
Affiliation(s)
- Karolina Kobus
- Medical Biology Laboratory, Pomeranian Medical University, Szczecin, Poland
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Lu Q, Lu S, Gao X, Luo Y, Tong B, Wei Z, Lu T, Xia Y, Chou G, Wang Z, Dai Y. Norisoboldine, an alkaloid compound isolated from Radix Linderae, inhibits synovial angiogenesis in adjuvant-induced arthritis rats by moderating Notch1 pathway-related endothelial tip cell phenotype. Exp Biol Med (Maywood) 2012; 237:919-32. [PMID: 22875342 DOI: 10.1258/ebm.2012.011416] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Synovial angiogenesis is well recognized as participating in the pathogenesis of rheumatoid arthritis (RA) and has been regarded as a potential target for RA therapy. Previously, we have shown that norisoboldine (NOR) can protect joints from destruction in mice with collagen II-induced arthritis (CIA). Here, we investigate the effect of NOR on synovial angiogenesis in adjuvant-induced arthritis (AA) rats, and clarify the mechanisms in vitro. NOR, administered orally, significantly reduced the number of blood vessels and expression of growth factors in the synovium of AA rats. In vitro, it markedly prevented the migration and sprouting of endothelial cells. Notably, the endothelial tip cell phenotype, which is essential for the migration of endothelial cells and subsequent angiogenesis, was significantly inhibited by NOR. This inhibitory effect was attenuated by pretreatment with N-{N-[2-(3,5-difluorophenyl) acetyl]-(S)-alanyl}-(S)-phenylglycine tert-butyl ester, a Notch1 inhibitor, suggesting that the action of NOR was related to the Notch1 pathway. A molecular docking study further confirmed that NOR was able to promote Notch1 activation by binding the Notch1 transcription complex. In conclusion, NOR was able to prevent synovial angiogenesis in AA rats, which is a putatively new mechanism responsible for its anti-rheumatoid effect. The anti-angiogenesis action of NOR was likely achieved by moderating the Notch1 pathway-related endothelial tip cell phenotype with a potential action target of the Notch1 transcription complex.
Collapse
Affiliation(s)
- Qian Lu
- Department of Pharmacology of Chinese Materia Medica, State Key Laboratory of Natural Medicines
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Rosuvastatin promotes angiogenesis and reverses isoproterenol-induced acute myocardial infarction in rats: role of iNOS and VEGF. Eur J Pharmacol 2012; 691:134-42. [PMID: 22732653 DOI: 10.1016/j.ejphar.2012.06.022] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 06/08/2012] [Accepted: 06/13/2012] [Indexed: 12/24/2022]
Abstract
Several reports highlighted the cardioprotective effect of statins after different types of ischemic injury. We studied the effect of rosuvastatin on acute myocardial infarction induced experimentally in rats focusing on angiogenesis as a potential mechanism underlying the drug effect. Acute myocardial infarction was induced by injecting the rats with two doses of isoproterenol (85 mg/kg/24 h, s.c.). Rats were examined for their electrocardiographic pattern and myocardial fibrosis one week after injection of isoproterenol (time for initiating therapy) and eight weeks thereafter (the end of therapeutic period) to examine the progression of the injury. Examination of the heart tissues at the end of week 9 showed a non significant decrease in the degree of myocardial fibrosis compared to those observed at week 1, indicating a slow rate of recovery from isoproterenol-induced injury. Treatment with rosuvastatin (5 or 10 mg/kg) for 8 weeks in myocardial-infarct rats enhanced the electrocardiographic pattern, reduced serum cardiac biomarkers, reduced tissue tumor necrosis factor-α (TNF-α) and upregulated vascular endothelial growth factor (VEGF) level. In addition, immunohistochemical staining revealed higher expression of inducible nitric oxide synthase (iNOS), VEGF and CD(34) (a marker for microvessel density) in the cardiac tissues after treatment with rosuvastatin compared to control group. The immunostaining for VEGF was positively correlated with microvessel density and iNOS. Overall, the current results provide evidence that the effect of rosuvastatin on myocardial-infarct rats involves induction of angiogenesis.
Collapse
|
37
|
Alef MJ, Tzeng E, Zuckerbraun BS. Nitric oxide and nitrite-based therapeutic opportunities in intimal hyperplasia. Nitric Oxide 2012; 26:285-94. [PMID: 22504069 DOI: 10.1016/j.niox.2012.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 03/27/2012] [Accepted: 03/30/2012] [Indexed: 12/22/2022]
Abstract
Vascular intimal hyperplasia (IH) limits the long term efficacy of current surgical and percutaneous therapies for atherosclerotic disease. There are extensive changes in gene expression and cell signaling in response to vascular therapies, including changes in nitric oxide (NO) signaling. NO is well recognized for its vasoregulatory properties and has been investigated as a therapeutic treatment for its vasoprotective abilities. The circulating molecules nitrite (NO(2)(-)) and nitrate (NO(3)(-)), once thought to be stable products of NO metabolism, are now recognized as important circulating reservoirs of NO and represent a complementary source of NO in contrast to the classic L-arginine-NO-synthase pathway. Here we review the background of IH, its relationship with the NO and nitrite/nitrate pathways, and current and future therapeutic opportunities for these molecules.
Collapse
Affiliation(s)
- Matthew J Alef
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | | |
Collapse
|
38
|
Daleprane JB, Schmid T, Dehne N, Rudnicki M, Menrad H, Geis T, Ikegaki M, Ong TP, Brüne B, Abdalla DSP. Suppression of hypoxia-inducible factor-1α contributes to the antiangiogenic activity of red propolis polyphenols in human endothelial cells. J Nutr 2012; 142:441-7. [PMID: 22279137 DOI: 10.3945/jn.111.150706] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Polyphenol-enriched fractions from natural sources have been proposed to interfere with angiogenesis in pathological conditions. We recently reported that red propolis polyphenols (RPP) exert antiangiogenic activity. However, molecular mechanisms of this activity remain unclear. Here, we aimed at characterizing molecular mechanisms to explain the impact of RPP on endothelial cells' (EC) physiology. We used in vitro and ex and in vivo models to test the hypothesis that RPP inhibit angiogenesis by affecting hypoxia-inducible factor-1α (HIF1α) stabilization in EC. RPP (10 mg/L) affected angiogenesis by reducing migration and sprouting of EC, attenuated the formation of new blood vessels, and decreased the differentiation of embryonic stem cells into CD31-positive cells. Moreover, RPP (10 mg/L) inhibited hypoxia- or dimethyloxallylglycine-induced mRNA and protein expression of the crucial angiogenesis promoter vascular endothelial growth factor (VEGF) in a time-dependent manner. Under hypoxic conditions, RPP at 10 mg/L, supplied for 1-4 h, decreased HIF1α protein accumulation, which in turn attenuated VEGF gene expression. In addition, RPP reduced the HIF1α protein half-life from ~58 min to 38 min under hypoxic conditions. The reduced HIF1α protein half-life was associated with an increase in the von Hippel-Lindau (pVHL)-dependent proteasomal degradation of HIF1α. RPP (10 mg/L, 4 h) downregulated Cdc42 protein expression. This caused a corresponding increase in pVHL protein levels and a subsequent degradation of HIF1α. In summary, we have elucidated the underlying mechanism for the antiangiogenic action of RPP, which attenuates HIF1α protein accumulation and signaling.
Collapse
Affiliation(s)
- Julio B Daleprane
- Department of Clinical and Toxicology Analysis, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Oliver G, Novak S, Patterson JL, Pasternak JA, Paradis F, Norrby M, Oxtoby K, Dyck MK, Dixon WT, Foxcroft GR. Restricted feed intake in lactating primiparous sows. II. Effects on subsequent litter sex ratio and embryonic gene expression. Reprod Fertil Dev 2012; 23:899-911. [PMID: 21871209 DOI: 10.1071/rd11013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2011] [Accepted: 04/08/2011] [Indexed: 11/23/2022] Open
Abstract
Expression of panels of candidate genes controlling myogenesis, angiogenesis and gender-specific imprinting of development were analysed in embryonic, placental and endometrial tissues recovered at Day 30 of gestation from a subset of primiparous sows that were either feed restricted (Restrict; n=17) or fed to appetite (Control; n=15) during the last week of the previous lactation. Embryos were also sex typed to investigate gender bias in response to treatments. Average embryonic weight was lower in the subset of Restrict compared with Control litters (1.38±0.07vs 1.59±0.08g, respectively) and the male:female sex ratio was higher (P<0.05) in embryos (litters) recovered from Restrict sows. Treatment affected (P≤0.05) the expression of embryonic and placental genes involved in insulin-like growth factor (IGF) 2 signalling, including IGF2, INSR and IGF2R. Embryonic expression of ESR1 was also affected by treatment (P<0.03) and sex×treatment interactions were observed for the expression of embryonic ESR1 (P<0.05) and placental ANGPT2 (P<0.03). At the molecular level, these results support the suggestion that changes in placental function are not the primary mechanism mediating detrimental effects of previous sow catabolism on early embryonic development in the feed-restricted lactational sow model. However, perturbations in the IGF2 system are implicated as mediators of these effects.
Collapse
Affiliation(s)
- G Oliver
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Soleti R, Martinez MC. Sonic Hedgehog on microparticles and neovascularization. VITAMINS AND HORMONES 2012; 88:395-438. [PMID: 22391314 DOI: 10.1016/b978-0-12-394622-5.00018-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neovascularization represents a pivotal process consisting in the development of vascular network during embryogenesis and adult life. Postnatally, it arises mainly through angiogenesis, which has physiological and pathological roles in health and disease. Blood vessel formation results as tightly regulated multistep process which needs coordination and precise regulation of the balance of proangiogenic and antiangiogenic factors. Sonic Hedgehog (SHH), a morphogen belonging to Hedgehog (HH) family proteins, is implicated in a remarkably wide variety of process, including vessel development. Recent evidence demonstrate that, in addition to the classic factors, microvesicles (MVs), both microparticles (MPs) and exosomes, small vesicles released distinct cellular compartments, are involved in modulation of neovascularization. MPs generated from T lymphocytes undergoing both activation and apoptosis harbor at their surface SHH and play a crucial role in modulation of neovascularization. They are able to modulate the different steps implicated in angiogenesis process in vitro and to enhance postischemic neovascularization in vivo. As the consequence, we suggest that the MPs carrying SHH contribute to generation of a vascular network and may represent a new therapeutic approach to treat pathologies associated with failed angiogenesis.
Collapse
|
41
|
Inhibitory effects of Rap1GAP overexpression on proliferation and migration of endothelial cells via ERK and Akt pathways. ACTA ACUST UNITED AC 2011; 31:721-727. [DOI: 10.1007/s11596-011-0667-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Indexed: 01/28/2023]
|
42
|
Rana M, von See C, Rücker M, Schumann P, Essig H, Kokemüller H, Lindhorst D, Gellrich NC. Increase in periosteal angiogenesis through heat shock conditioning. Head Face Med 2011; 7:22. [PMID: 22098710 PMCID: PMC3253043 DOI: 10.1186/1746-160x-7-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 11/18/2011] [Indexed: 11/10/2022] Open
Abstract
Objective It is widely known that stress conditioning can protect microcirculation and induce the release of vasoactive factors for a period of several hours. Little, however, is known about the long-term effects of stress conditioning on microcirculation, especially on the microcirculation of the periosteum of the calvaria. For this reason, we used intravital fluorescence microscopy to investigate the effects of heat shock priming on the microcirculation of the periosteum over a period of several days. Methods Fifty-two Lewis rats were randomized into eight groups. Six groups underwent heat shock priming of the periosteum of the calvaria at 42.5°C, two of them (n = 8) for 15 minutes, two (n = 8) for 25 minutes and two (n = 8) for 35 minutes. After 24 hours, a periosteal chamber was implanted into the heads of the animals of one of each of the two groups mentioned above. Microcirculation and inflammatory responses were studied repeatedly over a period of 14 days using intravital fluorescence microscopy. The expression of heat shock protein (HSP) 70 was examined by immunohistochemistry in three further groups 24 hours after a 15-minute (n = 5), a 25-minute (n = 5) or a 35-minute (n = 5) heat shock treatment. Two groups that did not undergo priming were used as controls. One control group (n = 8) was investigated by intravital microscopy and the other (n = 5) by immunohistochemistry. Results During the entire observation period of 14 days, the periosteal chambers revealed physiological microcirculation of the periosteum of the calvaria without perfusion failures. A significant (p < 0.05) and continuous increase in functional capillary density was noted from day 5 to day 14 after 25-minute heat shock priming. Whereas a 15-minute exposure did not lead to an increase in functional capillary density, 35-minute priming caused a significant but reversible perfusion failure in capillaries. Non-perfused capillaries in the 35-minute treatment group were reperfused by day 10. Immunohistochemistry demonstrated an increase in cytoprotective HSP70 expression in the periosteum after a 15-minute and a 35-minute heat shock pretreatment when compared with the control group. The level of HSP70 expression that was measured in the periosteum after 25 minutes of treatment was significantly higher than the levels observed after 15 or 35 minutes of heat shock exposure. Conclusion A few days after heat shock priming over an appropriate period of time, a continuous increase in functional capillary density is seen in the periosteum of the calvaria. This increase in perfusion appears to be the result of the induction of angiogenesis.
Collapse
Affiliation(s)
- Majeed Rana
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Boreddy SR, Sahu RP, Srivastava SK. Benzyl isothiocyanate suppresses pancreatic tumor angiogenesis and invasion by inhibiting HIF-α/VEGF/Rho-GTPases: pivotal role of STAT-3. PLoS One 2011; 6:e25799. [PMID: 22016776 PMCID: PMC3189946 DOI: 10.1371/journal.pone.0025799] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 09/11/2011] [Indexed: 12/22/2022] Open
Abstract
Our previous studies have shown that benzyl isothiocyanate (BITC) suppresses pancreatic tumor growth by inhibiting STAT-3; however, the exact mechanism of tumor growth suppression was not clear. Here we evaluated the effects and mechanism of BITC on pancreatic tumor angiogenesis. Our results reveal that BITC significantly inhibits neovasularization on rat aorta and Chicken-Chorioallantoic membrane. Furthermore, BITC blocks the migration and invasion of BxPC-3 and PanC-1 pancreatic cancer cells in a dose dependant manner. Moreover, secretion of VEGF and MMP-2 in normoxic and hypoxic BxPC-3 and PanC-1 cells was significantly suppressed by BITC. Both VEGF and MMP-2 play a critical role in angiogenesis and metastasis. Our results reveal that BITC significantly suppresses the phosphorylation of VEGFR-2 (Tyr-1175), and expression of HIF-α. Rho-GTPases, which are regulated by VEGF play a crucial role in pancreatic cancer progression. BITC treatment reduced the expression of RhoC whereas up-regulated the expression of tumor suppressor RhoB. STAT-3 over-expression or IL-6 treatment significantly induced HIF-1α and VEGF expression; however, BITC substantially suppressed STAT-3 as well as STAT-3-induced HIF-1α and VEGF expression. Finally, in vivo tumor growth and matrigel-plug assay show reduced tumor growth and substantial reduction of hemoglobin content in the matrigel plugs and tumors of mice treated orally with 12 µmol BITC, indicating reduced tumor angiogenesis. Immunoblotting of BITC treated tumors show reduced expression of STAT-3 phosphorylation (Tyr-705), HIF-α, VEGFR-2, VEGF, MMP-2, CD31 and RhoC. Taken together, our results suggest that BITC suppresses pancreatic tumor growth by inhibiting tumor angiogenesis through STAT-3-dependant pathway.
Collapse
Affiliation(s)
- Srinivas Reddy Boreddy
- Department of Biomedical Sciences and Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| | - Ravi P. Sahu
- Department of Biomedical Sciences and Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| | - Sanjay K. Srivastava
- Department of Biomedical Sciences and Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| |
Collapse
|
44
|
Abstract
(1) Angiogenesis (growth of new capillaries from an existing capillary bed) may result from a mismatch in microvascular supply and metabolic demand (metabolic error signal). Krogh examined the distribution and number of capillaries to explore the correlation between O(2) delivery and O(2) consumption. Subsequently, the heterogeneity in angiogenic response within a muscle has been shown to reflect either differences in fibre type composition or mechanical load. However, local control leads to targetted angiogenesis in the vicinity of glycolytic fibre types following muscle stimulation, or oxidative fibres following endurance training, while heterogeneity of capillary spacing is maintained during ontogenetic growth. (2) Despite limited microscopy resolution and lack of specific markers, Krogh's interest in the structure of the capillary wall paved the way for understanding the mechanisms of capillary growth. Angiogenesis may be influenced by the response of perivascular or stromal cells (fibroblasts, macrophages and pericytes) to altered activity, likely acting as a source for chemical signals modulating capillary growth such as vascular endothelial growth factor. In addition, haemodynamic factors such as shear stress and muscle stretch play a significant role in adaptive remodelling of the microcirculation. (3) Most indices of capillarity are highly dependent on fibre size, resulting in possible bias because of scaling. To examine the consequences of capillary distribution, it is therefore helpful to quantify the area of tissue supplied by individual capillaries. This allows the spatial limitations inherent in most models of tissue oxygenation to be overcome generating an alternative approach to Krogh's tissue cylinder, the capillary domain, to improve descriptions of intracellular oxygen diffusion.
Collapse
Affiliation(s)
- S Egginton
- Department of Physiology, University of Birmingham, Birmingham, UK.
| |
Collapse
|
45
|
Amsden BG. Delivery approaches for angiogenic growth factors in the treatment of ischemic conditions. Expert Opin Drug Deliv 2011; 8:873-90. [DOI: 10.1517/17425247.2011.577412] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
46
|
Suppression of vascular endothelial growth factor (VEGF) induced angiogenic responses by fucodiphloroethol G. Process Biochem 2011. [DOI: 10.1016/j.procbio.2011.01.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
47
|
Sathish S, Shanthi P, Sachdanandam P. Mitigation of DMBA-induced mammary carcinoma in experimental rats by antiangiogenic property of Kalpaamruthaa. J Diet Suppl 2011; 8:144-57. [PMID: 22432686 DOI: 10.3109/19390211.2011.561824] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Extra cellular matrix (ECM) and basement membrane (BM) are important layers that regulate cell structure, cell migration, and cellular proliferation. Degradation of both ECM and BM mediated by proteases favors the tumor invasion and promotes angiogenesis. Female Sprague-Dawley rats weighing 180 ± 10 g were categorized into 6 groups. Group-1 animals served as vehicle control. Group-2 to Group-4 animals were administered with 7,12-dimethylbenz(a)anthracene (25 mg/rat dissolved in olive oil, orally) on day 1 of experimental period to induce mammary carcinoma. (After 90 days, mammary carcinoma was confirmed by histopathological examination). Group-3 and Group-4 rats were subsequently treated with Semecarpus anacardium nut milk extract (SA) and Kalpaamruthaa (KA), respectively. Group-5 and Group-6 animals served as drug control for SA and KA, respectively. Pro-angiogenic factors like proteases, cyclooxygenase-2, and vascular endothelial growth factor were elevated in tumor-bearing animals and decreased in SA- and KA-supplemented rats. Increased levels of these angiogenic factors in tumor-bearing rats indicate the progression of mammary tumor. The decreased levels of these angiogenic in SA- and KA-treated rats may be due to the ameliorative effect of phenolic compounds such as flavonoids, tannins, and other compounds present in the drug.
Collapse
Affiliation(s)
- Sivaprakasam Sathish
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, India
| | | | | |
Collapse
|
48
|
Valapala M, Thamake SI, Vishwanatha JK. A competitive hexapeptide inhibitor of annexin A2 prevents hypoxia-induced angiogenic events. J Cell Sci 2011; 124:1453-64. [PMID: 21486955 DOI: 10.1242/jcs.079236] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Extracellular proteolysis is an indispensable requirement for the formation of new blood vessels during neovascularization and is implicated in the generation of several angiogenic regulatory molecules. Anti-proteolytic agents have become attractive therapeutic strategies in diseases associated with excessive neovascularization. Annexin A2 (AnxA2) is an endothelial cell-surface receptor for the generation of active proteolytic factors, such as plasmin. Here, we show that AnxA2 is abundantly expressed in the neovascular tufts in a murine model of neovascularization. Exposure to hypoxic conditions results in elevation of AnxA2 and tissue plasminogen activator (tPA) in human retinal microvascular endothelial cells (RMVECs). We show that the hexapeptide competitive inhibitor LCKLSL, which targets the N-terminal tPA-binding site of AnxA2, binds efficiently to cell-surface AnxA2 compared with binding of the control peptide LGKLSL. Treatment with the competitive peptide inhibits the generation of plasmin and suppresses the VEGF-induced activity of tPA under hypoxic conditions. Application of the competitive peptide in two in vivo models of angiogenesis demonstrated suppression of the angiogenic responses, which was also associated with significant changes in the vascular sprouting. These results suggest that AnxA2-mediated plasmin generation is an important event in angiogenesis and is inhibited by a specific competitive peptide that inhibits the binding of tPA to AnxA2.
Collapse
Affiliation(s)
- Mallika Valapala
- Department of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | | | | |
Collapse
|
49
|
Grammas P. Neurovascular dysfunction, inflammation and endothelial activation: implications for the pathogenesis of Alzheimer's disease. J Neuroinflammation 2011; 8:26. [PMID: 21439035 PMCID: PMC3072921 DOI: 10.1186/1742-2094-8-26] [Citation(s) in RCA: 311] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 03/25/2011] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related disorder characterized by progressive cognitive decline and dementia. Alzheimer's disease is an increasingly prevalent disease with 5.3 million people in the United States currently affected. This number is a 10 percent increase from previous estimates and is projected to sharply increase to 8 million by 2030; it is the sixth-leading cause of death. In the United States the direct and indirect costs of Alzheimer's and other dementias to Medicare, Medicaid and businesses amount to more than $172 billion each year. Despite intense research efforts, effective disease-modifying therapies for this devastating disease remain elusive. At present, the few agents that are FDA-approved for the treatment of AD have demonstrated only modest effects in modifying clinical symptoms for relatively short periods and none has shown a clear effect on disease progression. New therapeutic approaches are desperately needed. Although the idea that vascular defects are present in AD and may be important in disease pathogenesis was suggested over 25 years ago, little work has focused on an active role for cerebrovascular mechanisms in the pathogenesis of AD. Nevertheless, increasing literature supports a vascular-neuronal axis in AD as shared risk factors for both AD and atherosclerotic cardiovascular disease implicate vascular mechanisms in the development and/or progression of AD. Also, chronic inflammation is closely associated with cardiovascular disease, as well as a broad spectrum of neurodegenerative diseases of aging including AD. In this review we summarize data regarding, cardiovascular risk factors and vascular abnormalities, neuro- and vascular-inflammation, and brain endothelial dysfunction in AD. We conclude that the endothelial interface, a highly synthetic bioreactor that produces a large number of soluble factors, is functionally altered in AD and contributes to a noxious CNS milieu by releasing inflammatory and neurotoxic species.
Collapse
Affiliation(s)
- Paula Grammas
- Garrison Institute on Aging, and Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, Texas, USA.
| |
Collapse
|
50
|
Vicari D, Foy KC, Liotta EM, Kaumaya PTP. Engineered conformation-dependent VEGF peptide mimics are effective in inhibiting VEGF signaling pathways. J Biol Chem 2011; 286:13612-25. [PMID: 21321115 DOI: 10.1074/jbc.m110.216812] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Angiogenesis, or formation of new blood vessels, is crucial to cancer tumor growth. Tumor growth, progression, and metastasis are critically influenced by the production of the pro-angiogenic vascular endothelial growth factor (VEGF). Promising anti-angiogenic drugs are currently available; however, their susceptibilities to drug resistance and long term toxicity are serious impediments to their use, thus requiring the development of new therapeutic approaches for safe and effective angiogenic inhibitors. In this work, peptides were designed to mimic the VEGF-binding site to its receptor VEGFR-2. The VEGF conformational peptide mimic, VEGF-P3(CYC), included two artificial cysteine residues, which upon cyclization constrained the peptide in a loop native-like conformation to better mimic the anti-parallel structure of VEGF. The engineered cyclic VEGF mimic peptide demonstrated the highest affinity to VEGFR-2 by surface plasmon resonance assay. The VEGF peptide mimics were evaluated as inhibitors in several in vitro assays in which VEGF-dependent signaling pathways were observed. All VEGF mimics inhibited VEGFR-2 phosphorylation with VEGF-P3(CYC) showing the highest inhibitory effects when compared with unstructured peptides. Additionally, we show in several angiogenic in vitro assays that all the VEGF mimics inhibited endothelial cell proliferation, migration, and network formation with the conformational VEGF-P3 (CYC) being the best. The VEGF-P3(CYC) also caused a significant delay in tumor development in a transgenic model of VEGF(+/-)Neu2-5(+/-). These results indicate that the structure-based design is important for the development of this peptidomimetic and for its anti-angiogenic effects.
Collapse
Affiliation(s)
- Daniele Vicari
- Department of Microbiology, Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | |
Collapse
|