1
|
Lee YS, Choi JR, Kim JB. Gene Therapy for Cardiovascular Disease: Clinical Perspectives. Yonsei Med J 2024; 65:557-571. [PMID: 39313446 PMCID: PMC11427124 DOI: 10.3349/ymj.2024.0127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 09/25/2024] Open
Abstract
Cardiovascular disease (CVD) stands as one of the leading causes of death in the United States, with its prevalence steadily on the rise. Traditional therapeutic approaches, such as pharmacological treatment, cardiovascular intervention, and surgery, have inherent limitations. In response to these challenges, cardiac gene therapy has emerged as a promising alternative for treating CVD patients. However, several obstacles persist, including the low efficiency of gene transduction, immune reactions to vectors or transduced cells, and the occurrence of off-target effects. While preclinical research has demonstrated significant success in various CVD model in both small and large animals, the translation of these findings to clinical applications has, for the most part, yielded disappointing results, except for some early, albeit small, trials. This review aims to provide a comprehensive summary of recent preclinical and clinical studies on gene therapy for various CVDs. Additionally, we discuss the existing limitations and challenges that hinder the widespread clinical application of cardiac gene therapy.
Collapse
Affiliation(s)
- Young Shin Lee
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jung Ran Choi
- College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jin-Bae Kim
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, Korea
- College of Medicine, Kyung Hee University, Seoul, Korea.
| |
Collapse
|
2
|
Weng T, Yang M, Zhang W, Jin R, Xia S, Zhang M, Wu P, He X, Han C, Zhao X, Wang X. Dual gene-activated dermal scaffolds regulate angiogenesis and wound healing by mediating the coexpression of VEGF and angiopoietin-1. Bioeng Transl Med 2023; 8:e10562. [PMID: 37693053 PMCID: PMC10487340 DOI: 10.1002/btm2.10562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 05/22/2023] [Accepted: 06/01/2023] [Indexed: 09/12/2023] Open
Abstract
The vascularization of dermal substitutes is a key challenge in efforts to heal deep skin defects. In this study, dual gene-activated dermal scaffolds (DGADSs-1) were fabricated by loading nanocomposite particles of polyethylenimine (PEI)/multiple plasmid DNAs (pDNAs) encoding vascular endothelial growth factor and angiopoietin-1 at a ratio of 1:1. In a similar manner, DGADSs-2 were loaded with a chimeric plasmid encoding both VEGF and Ang-1. In vitro studies showed that both types of DGADSs released PEI/pDNA nanoparticles in a sustained manner; they demonstrated effective transfection ability, leading to upregulated expression of VEGF and Ang-1. Furthermore, both types of DGADSs promoted fibroblast proliferation and blood vessel formation, although DGADSs-1 showed a more obvious promotion effect. A rat full-thickness skin defect model showed that split-thickness skin transplanted using a one-step method could achieve full survival at the 12th day after surgery in both DGADSs-1 and DGADSs-2 groups, and the vascularization time of dermal substitutes was significantly shortened. Compared with the other three groups of scaffolds, the DGADSs-1 group had significantly greater cell infiltration, collagen deposition, neovascularization, and vascular maturation, all of which promoted wound healing. Thus, compared with single-gene-activated dermal scaffolds, DGADSs show greater potential for enhancing angiogenesis. DGADSs with different loading modes also exhibited differences in terms of angiogenesis; the effect of loading two genes (DGADSs-1) was better than the effect of loading a chimeric gene (DGADSs-2). In summary, DGADSs, which continuously upregulate VEGF and Ang-1 expression, offer a new functional tissue-engineered dermal substitute with the ability to activate vascularization.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
- Department of Burn and Plastic SurgeryChildren's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical CenterHangzhouChina
| | - Min Yang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Wei Zhang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Ronghua Jin
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Sizhan Xia
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Manjia Zhang
- The First Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Pan Wu
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Xiaojie He
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Chunmao Han
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Xiong Zhao
- Department of Burn and Plastic SurgeryChildren's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical CenterHangzhouChina
| | - Xingang Wang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
3
|
Li B, Li Y, Chen S, Wang Y, Zheng Y. VEGF mimetic peptide-conjugated nanoparticles for magnetic resonance imaging and therapy of myocardial infarction. J Control Release 2023; 360:44-56. [PMID: 37330014 DOI: 10.1016/j.jconrel.2023.06.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
To reduce the mortality of myocardial infarction (MI), accurate detection of the infarct and appropriate prevention against ischemia/reperfusion (I/R) induced cardiac dysfunction are highly desired. Considering that vascular endothelial growth factor (VEGF) receptors are overexpressed in the infarcted heart and VEGF mimetic peptide QK binds specifically to VEGF receptors and activates vascularization, the PEG-QK-modified, gadolinium-doped carbon dots (GCD-PEG-QK) were formulated. This research aims to investigate the magnetic resonance imaging (MRI) capability of GCD-PEG-QK on myocardial infarct and their therapeutic effect on I/R-induced myocardial injury. These multifunctional nanoparticles exhibited good colloidal stability, excellent fluorescent and magnetic property, and satisfactory biocompatibility. Intravenous injection of GCD-PEG-QK nanoparticles post myocardial I/R displayed accurate MRI of the infarct, enhanced efficacy of QK peptide on pro-angiogenesis, and amelioration of cardiac fibrosis, remodeling and dysfunction, probably via the improvement on QK's in vivo stability and MI-targeting. Collectively, the data suggested that this theranostic nanomedicine can realize precise MRI and effective therapy for acute MI in a non-invasive manner.
Collapse
Affiliation(s)
- Bing Li
- Department of Pharmacology, Capital Medical University, Beijing 100069, China
| | - Yingxu Li
- Department of Pharmacology, Capital Medical University, Beijing 100069, China
| | - Shuangling Chen
- Department of Chemical Biology, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Yuji Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Yuanyuan Zheng
- Department of Pharmacology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
4
|
Yao H, Xu H, Wu M, Lei W, Li L, Liu D, Wang Z, Ran H, Ma H, Zhou X. Targeted long-term noninvasive treatment of choroidal neovascularization by biodegradable nanoparticles. Acta Biomater 2023; 166:536-551. [PMID: 37196903 DOI: 10.1016/j.actbio.2023.05.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 04/24/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023]
Abstract
Choroidal neovascularization (CNV) is the main cause of vision loss in patients with wet age-related macular degeneration (AMD). Currently, treatment of these conditions requires repeated intravitreal injections, which may lead to complications such as infection and hemorrhage. So, we have developed a noninvasive method for treating CNV with nanoparticles, namely, Angiopoietin1-anti CD105-PLGA nanoparticles (AAP NPs), which targets the CNV to enhance drug accumulation at the site. These nanoparticles, with PLGA as a carrier, can slowly release encapsulated Angiopoietin 1 (Ang 1) and target the choroidal neovascularization marker CD105 to enhance drug accumulation, increases vascular endothelial cadherin (VE-cadherin) expression between vascular endothelial cells, effectively reduce neovascularization leakage and inhibit Angiopoietin 2(Ang 2) secretion by endothelial cells. In a rat model of laser-induced CNV, intravenous injection of AAP NPs exerted a good therapeutic effect in reducing CNV leakage and area. In short, these synthetic AAP NPs provide an effective alternative treatment for AMD and meet the urgent need for noninvasive treatment in neovascular ophthalmopathy. STATEMENT OF SIGNIFICANCE: This work describes the synthesis, injection-mediated delivery, in vitro and in vivo efficacy of targeted nanoparticles with encapsulated Ang1; via these nanoparticles, the drug can be targeted to choroidal neovascularization lesions for continuous treatment. The release of Ang1 can effectively reduce neovascularization leakage, maintain vascular stability, and inhibit Ang2 secretion and inflammation. This study provides a new approach for the treatment of wet age-related macular degeneration.
Collapse
Affiliation(s)
- Hao Yao
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400012, China; Chongqing Key Laboratory of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Huan Xu
- Chongqing Key Laboratory of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Mingxing Wu
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400012, China
| | - Wulong Lei
- Chongqing Key Laboratory of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Lanjiao Li
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400012, China; Chongqing Key Laboratory of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Danning Liu
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400012, China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Huafeng Ma
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400012, China.
| | - Xiyuan Zhou
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400012, China; Chongqing Key Laboratory of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China.
| |
Collapse
|
5
|
The role of PIWI-interacting RNA in naringin pro-angiogenesis by targeting HUVECs. Chem Biol Interact 2023; 371:110344. [PMID: 36623717 DOI: 10.1016/j.cbi.2023.110344] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 12/19/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
Angiogenesis is a biological process in which resting endothelial cells start proliferating, migrating and forming new blood vessels. Angiogenesis is particularly important in the repair of bone tissue defects. Naringin (NG) is the main active monomeric component of traditional Chinese medicine, which has various biological activities, such as anti-osteoporosis, anti-inflammatory, blood activation and microcirculation improvement. At present, the mechanism of naringin in the process of angiogenesis is not clear. PIWI protein-interacting RNA (piRNA) is a small noncoding RNA (sncRNA) that has the functions of regulating protein synthesis, regulating the structure of chromatin and the genome, stabilizing mRNA and others. Several studies have demonstrated that piRNAs can mediate the angiogenesis process. Whether naringin can interfere with the process of angiogenesis by regulating piRNAs and related target genes deserves further exploration. Thus, the purpose of this study was to validate the potential angiogenic and bone regeneration properties and related mechanisms of naringin both in vivo and in vitro.
Collapse
|
6
|
Huang P, Wan H, Shao C, Li C, Zhang L, He Y. Recent Advances in Chinese Herbal Medicine for Cerebral Ischemic Reperfusion Injury. Front Pharmacol 2022; 12:688596. [PMID: 35111041 PMCID: PMC8801784 DOI: 10.3389/fphar.2021.688596] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
Cerebral ischemic reperfusion injury (CI/RI) is a critical factor that leads to a poor prognosis in patients with ischemic stroke. It is an extremely complicated pathological process that is clinically characterized by high rates of disability and mortality. Current available treatments for CI/RI, including mechanical and drug therapies, are often accompanied by significant side effects. Therefore, it is necessary to discovery new strategies for treating CI/RI. Many studies confirm that Chinese herbal medicine (CHM) was used as a potential drug for treatment of CI/RI with the advantages of abundant resources, good efficacy, and few side effects. In this paper, we investigate the latest drug discoveries and advancements on CI/RI, make an overview of relevant CHM, and systematically summarize the pathophysiology of CI/RI. In addition, the protective effect and mechanism of related CHM, which includes extraction of single CHM and CHM formulation and preparation, are discussed. Moreover, an outline of the limitations of CHM and the challenges we faced are also presented. This review will be helpful for researchers further propelling the advancement of drugs and supplying more knowledge to support the application of previous discoveries in clinical drug applications against CI/RI.
Collapse
Affiliation(s)
- Ping Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haitong Wan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chongyu Shao
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chang Li
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ling Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
7
|
Cardiac-derived stem cell engineered with constitutively active HIF-1α gene enhances blood perfusion of hindlimb ischemia. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2021.09.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
8
|
Weng T, Wang J, Yang M, Zhang W, Wu P, You C, Han C, Wang X. Nanomaterials for the delivery of bioactive factors to enhance angiogenesis of dermal substitutes during wound healing. BURNS & TRAUMA 2022; 10:tkab049. [PMID: 36960274 PMCID: PMC8944711 DOI: 10.1093/burnst/tkab049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/14/2021] [Indexed: 11/14/2022]
Abstract
Dermal substitutes provide a template for dermal regeneration and reconstruction. They constitutes an ideal clinical treatment for deep skin defects. However, rapid vascularization remains as a major hurdle to the development and application of dermal substitutes. Several bioactive factors play an important regulatory role in the process of angiogenesis and an understanding of the mechanism of achieving their effective delivery and sustained function is vital. Nanomaterials have great potential for tissue engineering. Effective delivery of bioactive factors (including growth factors, peptides and nucleic acids) by nanomaterials is of increasing research interest. This paper discusses the process of dermal substitute angiogenesis and the roles of related bioactive factors in this process. The application of nanomaterials for the delivery of bioactive factors to enhance angiogenesis and accelerate wound healing is also reviewed. We focus on new systems and approaches for delivering bioactive factors for enhancing angiogenesis in dermal substitutes.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Jialiang Wang
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Min Yang
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Wei Zhang
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Pan Wu
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Chuangang You
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Chunmao Han
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | | |
Collapse
|
9
|
Yang X, Li X, Luo M, Guo Y, Li C, Lv D, Cheng Z, Huang L, Shang FF, Huang B, Shen J, Luo S, Yan J. Tubeimoside I promotes angiogenesis via activation of eNOS-VEGF signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113642. [PMID: 33264658 DOI: 10.1016/j.jep.2020.113642] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tubeimoside I (TBM) is a triterpenoid saponin purified from tubeimu (tuber of Bolbostemma paniculatum (Maxim.) Franquet). In traditional Chinese medicine, tubeimu had been used to treat acute mastitis, snake bites, detoxication, inflammatory diseases, and tumors for over 1000 years. AIM OF THE STUDY This study aimed to investigate whether TBM could promote angiogenesis and how to promote angiogenesis. MATERIALS AND METHODS In vivo, the pro-angiogenic effects of TBM were examined using the hindlimb ischemia model. After the ischemia operation, 1 mg/kg/day TBM was given via intraperitoneal injection for 28 days and the recovery of blood flow was monitored by Doppler scanner every 7 days. The capillary density in gastrocnemius muscle was detected by immunofluorescence. Expression of related proteins were determined by western blotting. In vitro, the pro-angiogenic effects of TBM on HUVECs were examined by Cell Counting Kit-8, scratch assay, endothelial cell tube formation assay and western blotting. RESULTS TBM improved recovery from hindlimb ischemia in C57BL/6 mice. TBM promoted endothelial cell viability, migration and tube formation in HUVECs. TBM could activate eNOS-VEGF signaling pathway by enhancing expression of eNOS. And TBM's pro-angiogenesis effects could be abolished by L-NAME (an inhibitor of eNOS). CONCLUSIONS TBM promoted angiogenesis via the activation of eNOS-VEGF signaling pathway and TBM could be a novel agent for therapeutic angiogenesis in ischemic diseases.
Collapse
Affiliation(s)
- Xiyang Yang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Xingbing Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Yongzheng Guo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Chang Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Dingyi Lv
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Zhe Cheng
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Longxiang Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Fei-Fei Shang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Bi Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Jian Shen
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Suxin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China.
| | - Jianghong Yan
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
10
|
Understanding angiogenesis and the role of angiogenic growth factors in the vascularisation of engineered tissues. Mol Biol Rep 2021; 48:941-950. [PMID: 33393005 DOI: 10.1007/s11033-020-06108-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022]
Abstract
Tissue engineering is a rapidly developing field with many potential clinical applications in tissue and organ regeneration. The development of a mature and stable vasculature within these engineered tissues (ET) remains a significant obstacle. Currently, several growth factors (GFs) have been identified to play key roles within in vivo angiogenesis, including vascular endothelial growth factor (VEGF), platelet derived growth factor (PDGF), FGF and angiopoietins. In this article we attempt to build on in vivo principles to review the single, dual and multiple GF release systems and their effects on promoting angiogenesis. We conclude that multiple GF release systems offer superior results compared to single and dual systems with more stable, mature and larger vessels produced. However, with more complex release systems this raises other problems such as increased cost and significant GF-GF interactions. Upstream regulators and pericyte-coated scaffolds could provide viable alternative to circumnavigate these issues.
Collapse
|
11
|
Hayashi SI, Rakugi H, Morishita R. Insight into the Role of Angiopoietins in Ageing-Associated Diseases. Cells 2020; 9:E2636. [PMID: 33302426 PMCID: PMC7762563 DOI: 10.3390/cells9122636] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 12/17/2022] Open
Abstract
Angiopoietin (Ang) and its receptor, TIE signaling, contribute to the development and maturation of embryonic vasculature as well as vascular remodeling and permeability in adult tissues. Targeting both this signaling pathway and the major pathway with vascular endothelial growth factor (VEGF) is expected to permit clinical applications, especially in antiangiogenic therapies against tumors. Several drugs targeting the Ang-TIE signaling pathway in cancer patients are under clinical development. Similar to how cancer increases with age, unsuitable angiogenesis or endothelial dysfunction is often seen in other ageing-associated diseases (AADs) such as atherosclerosis, Alzheimer's disease, type 2 diabetes, chronic kidney disease and cardiovascular diseases. Thus, the Ang-TIE pathway is a possible molecular target for AAD therapy. In this review, we focus on the potential role of the Ang-TIE signaling pathway in AADs, especially non-cancer-related AADs. We also suggest translational insights and future clinical applications of this pathway in those AADs.
Collapse
Affiliation(s)
- Shin-ichiro Hayashi
- Department of Clinical Gene Therapy, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan;
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
12
|
Yang Y, Tian W, Yang L, Zhang Q, Zhu M, Liu Y, Li J, Yang L, Liu J, Shen Y, Qi Z. Gemcitabine potentiates anti-tumor effect of resveratrol on pancreatic cancer via down-regulation of VEGF-B. J Cancer Res Clin Oncol 2020; 147:93-103. [PMID: 32897433 DOI: 10.1007/s00432-020-03384-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/02/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE In our previous study, we discovered that resveratrol (RSV) had potential tumor-promoting effect on pancreatic cancer (PaCa) via up-regulation of VEGF-B. Therefore, we assumed that a pharmacological inhibitor of VEGF-B should potentiate the anti-tumor effect of RSV on PaCa. METHODS Real-time PCR and western blotting were used to examine VEGF-B mRNA and protein levels. Cell viability and cell apoptosis were assessed by CCK-8 assay and flow cytometry analysis, respectively. PaCa cell-bearing nude mice were used to evaluate the anti-cancer effects of single treatment or co-administration of RSV and gemcitabine (GEM). RESULTS We found that treatment with GEM alone dramatically decreased VEGF-B expression in comparison with control group, indicating that GEM is a potential pharmacological inhibitor of VEGF-B in PaCa. The co-administration of RSV and GEM significantly lowered expression of VEGF-B and increased phosphorylation level of GSK3β at Ser9 when compared to RSV alone treatment either in vitro or in vivo. Combination of RSV and GEM significantly increased cell death and apoptosis of PaCa cells in vitro and inhibited tumor growth in vivo in comparison with RSV or GEM alone treatment. Furthermore, we found that the anti-tumor effect in combination group was dramatically weakened after VEGF-B overexpressed in PaCa cells. CONCLUSION These results suggest that VEGF-B signaling pathway plays an important role in the development of PaCa and combination of GEM and RSV would be a promising modality for clinical PaCa therapy.
Collapse
Affiliation(s)
- Yinan Yang
- Department of Histology and Embryology, School of Medicine, Nankai University, 94 Weijin Road, Nankai District, Tianjin, 300071, China.,Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
| | - Wencong Tian
- Department of Histology and Embryology, School of Medicine, Nankai University, 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Lei Yang
- Department of Histology and Embryology, School of Medicine, Nankai University, 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Qiong Zhang
- Department of Microbiology, School of Laboratory Medicine, Tianjin Medical University, 1 Guangdong Road, Hexi District, Tianjin, 300203, China
| | - Mengmeng Zhu
- Department of Histology and Embryology, School of Medicine, Nankai University, 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Yuansheng Liu
- Department of Histology and Embryology, School of Medicine, Nankai University, 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Jing Li
- Department of Histology and Embryology, School of Medicine, Nankai University, 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Liang Yang
- Department of Histology and Embryology, School of Medicine, Nankai University, 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Jie Liu
- Department of Histology and Embryology, School of Medicine, Nankai University, 94 Weijin Road, Nankai District, Tianjin, 300071, China
| | - Yanna Shen
- Department of Microbiology, School of Laboratory Medicine, Tianjin Medical University, 1 Guangdong Road, Hexi District, Tianjin, 300203, China.
| | - Zhi Qi
- Department of Histology and Embryology, School of Medicine, Nankai University, 94 Weijin Road, Nankai District, Tianjin, 300071, China. .,National Clinical Research Center of Kidney Diseases, Beijing, 100853, China.
| |
Collapse
|
13
|
VEGF-A in Cardiomyocytes and Heart Diseases. Int J Mol Sci 2020; 21:ijms21155294. [PMID: 32722551 PMCID: PMC7432634 DOI: 10.3390/ijms21155294] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
The vascular endothelial growth factor (VEGF), a homodimeric vasoactive glycoprotein, is the key mediator of angiogenesis. Angiogenesis, the formation of new blood vessels, is responsible for a wide variety of physio/pathological processes, including cardiovascular diseases (CVD). Cardiomyocytes (CM), the main cell type present in the heart, are the source and target of VEGF-A and express its receptors, VEGFR1 and VEGFR2, on their cell surface. The relationship between VEGF-A and the heart is double-sided. On the one hand, VEGF-A activates CM, inducing morphogenesis, contractility and wound healing. On the other hand, VEGF-A is produced by CM during inflammation, mechanical stress and cytokine stimulation. Moreover, high concentrations of VEGF-A have been found in patients affected by different CVD, and are often correlated with an unfavorable prognosis and disease severity. In this review, we summarized the current knowledge about the expression and effects of VEGF-A on CM and the role of VEGF-A in CVD, which are the most important cause of disability and premature death worldwide. Based on clinical studies on angiogenesis therapy conducted to date, it is possible to think that the control of angiogenesis and VEGF-A can lead to better quality and span of life of patients with heart disease.
Collapse
|
14
|
Pei X, Kim H, Lee M, Wang N, Shin J, Lee S, Yoon M, Yang VC, He H. Local delivery of cardiac stem cells overexpressing HIF-1α promotes angiogenesis and muscular tissue repair in a hind limb ischemia model. J Control Release 2020; 322:610-621. [DOI: 10.1016/j.jconrel.2020.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/23/2020] [Accepted: 03/13/2020] [Indexed: 12/14/2022]
|
15
|
Zhang Y, Kontos CD, Annex BH, Popel AS. Angiopoietin-Tie Signaling Pathway in Endothelial Cells: A Computational Model. iScience 2019; 20:497-511. [PMID: 31655061 PMCID: PMC6806670 DOI: 10.1016/j.isci.2019.10.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/21/2019] [Accepted: 09/30/2019] [Indexed: 12/13/2022] Open
Abstract
The angiopoietin-Tie signaling pathway is an important vascular signaling pathway involved in angiogenesis, vascular stability, and quiescence. Dysregulation in the pathway is linked to the impairments in vascular function associated with many diseases, including cancer, ocular diseases, systemic inflammation, and cardiovascular diseases. The present study uses a computational signaling pathway model validated against experimental data to quantitatively study various mechanistic aspects of the angiopoietin-Tie signaling pathway, including receptor activation, trafficking, turnover, and molecular mechanisms of its regulation. The model provides mechanistic insights into the controversial role of Ang2 and its regulators vascular endothelial protein tyrosine phosphatase (VE-PTP) and Tie1 and predicts synergistic effects of inhibition of VE-PTP, Tie1, and Tie2 cleavage on enhancing the vascular protective actions of Tie2.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Christopher D Kontos
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Brian H Annex
- Department of Medicine and the Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
16
|
Venneri MA, Barbagallo F, Fiore D, De Gaetano R, Giannetta E, Sbardella E, Pozza C, Campolo F, Naro F, Lenzi A, Isidori AM. PDE5 Inhibition Stimulates Tie2-Expressing Monocytes and Angiopoietin-1 Restoring Angiogenic Homeostasis in Diabetes. J Clin Endocrinol Metab 2019; 104:2623-2636. [PMID: 31102457 DOI: 10.1210/jc.2018-02525] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 03/08/2019] [Indexed: 02/13/2023]
Abstract
CONTEXT Vascular dysfunction is a common feature in end-organ complications of type 2 diabetes mellitus (T2DM). The endothelium-specific receptor tyrosine kinase Tie2 and its ligand, angiopoietin-1 (Ang1), participate in the processes of vessel repair, renewal, and maturation. However, their dysregulation in T2DM has seldom been investigated. OBJECTIVES To examine the relationship between angiogenic Tie2-expressing monocytes (TEMs) and Ang1, and their pharmacological modulation by the phosphodiesterase type 5 inhibitor (PDE5i) sildenafil, in T2DM and in db/db mouse model. DESIGN AND SETTING Randomized, double-blind, placebo-controlled study. PATIENTS AND INTERVENTION db/db male mice were randomly assigned to receive 8 weeks of sildenafil or vehicle. Diabetic men were randomly assigned to receive 4 weeks of sildenafil or placebo. MAIN OUTCOMES AND MEASURES Peripheral blood cells were investigated by flow cytometry to quantify inflammatory myeloid CD11b+ Gr1+ cells and proangiogenic TEMs in mice and classical CD14++CD16neg monocytes and proangiogenic TEMs in humans at baseline and after treatment. In vitro human tube formation assay was used to test serum angiogenic potential. RESULTS We show that TEMs and Ang1 are defective in mouse and human models of diabetes and are normalized by PDE5i treatment. Serum angiogenic properties are impaired in diabetes because they do not support the in vitro formation of capillary-like structures, but they are reestablished by in vivo PDE5i treatment. CONCLUSIONS Restoring a more physiological Tie2-Ang1 axis with sildenafil reestablishes serum angiogenic properties in diabetes, promoting angiogenic homeostasis.
Collapse
Affiliation(s)
- Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Federica Barbagallo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniela Fiore
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Rita De Gaetano
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Emilia Sbardella
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Carlotta Pozza
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Fabio Naro
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
17
|
Min S, Ko IK, Yoo JJ. State-of-the-Art Strategies for the Vascularization of Three-Dimensional Engineered Organs. Vasc Specialist Int 2019; 35:77-89. [PMID: 31297357 PMCID: PMC6609020 DOI: 10.5758/vsi.2019.35.2.77] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 02/07/2023] Open
Abstract
Engineering three-dimensional (3D) implantable tissue constructs is a promising strategy for replacing damaged or diseased tissues and organs with functional replacements. However, the efficient vascularization of new 3D organs is a major scientific and technical challenge since large tissue constructs or organs require a constant blood supply to survive in vivo. Current approaches to solving this problem generally fall into the following three major categories: (a) cell-based, (b) angiogenic factor-based, and (c) scaffold-based. In this review, we summarize state-of-the-art technologies that are used to develop complex, stable, and functional vasculature for engineered 3D tissue constructs and organs; additionally, we have suggested directions for future research.
Collapse
Affiliation(s)
- Sangil Min
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - In Kap Ko
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
18
|
Abstract
The ability to generate new microvessels in desired numbers and at desired locations has been a long-sought goal in vascular medicine, engineering, and biology. Historically, the need to revascularize ischemic tissues nonsurgically (so-called therapeutic vascularization) served as the main driving force for the development of new methods of vascular growth. More recently, vascularization of engineered tissues and the generation of vascularized microphysiological systems have provided additional targets for these methods, and have required adaptation of therapeutic vascularization to biomaterial scaffolds and to microscale devices. Three complementary strategies have been investigated to engineer microvasculature: angiogenesis (the sprouting of existing vessels), vasculogenesis (the coalescence of adult or progenitor cells into vessels), and microfluidics (the vascularization of scaffolds that possess the open geometry of microvascular networks). Over the past several decades, vascularization techniques have grown tremendously in sophistication, from the crude implantation of arteries into myocardial tunnels by Vineberg in the 1940s, to the current use of micropatterning techniques to control the exact shape and placement of vessels within a scaffold. This review provides a broad historical view of methods to engineer the microvasculature, and offers a common framework for organizing and analyzing the numerous studies in this area of tissue engineering and regenerative medicine. © 2019 American Physiological Society. Compr Physiol 9:1155-1212, 2019.
Collapse
Affiliation(s)
- Joe Tien
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Division of Materials Science and Engineering, Boston University, Brookline, Massachusetts, USA
| |
Collapse
|
19
|
Engineered delivery strategies for enhanced control of growth factor activities in wound healing. Adv Drug Deliv Rev 2019; 146:190-208. [PMID: 29879493 DOI: 10.1016/j.addr.2018.06.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/18/2018] [Accepted: 06/01/2018] [Indexed: 12/18/2022]
Abstract
Growth factors (GFs) are versatile signalling molecules that orchestrate the dynamic, multi-stage process of wound healing. Delivery of exogenous GFs to the wound milieu to mediate healing in an active, physiologically-relevant manner has shown great promise in laboratories; however, the inherent instability of GFs, accompanied with numerous safety, efficacy and cost concerns, has hindered the clinical success of GF delivery. In this article, we highlight that the key to overcoming these challenges is to enhance the control of the activities of GFs throughout the delivering process. We summarise the recent strategies based on biomaterials matrices and molecular engineering, which aim to improve the conditions of GFs for delivery (at the 'supply' end of the delivery), increase the stability and functions of GFs in extracellular matrix (in transportation to target cells), as well as enhance the GFs/receptor interaction on the cell membrane (at the 'destination' end of the delivery). Many of these investigations have led to encouraging outcomes in various in vitro and in vivo regenerative models with considerable translational potential.
Collapse
|
20
|
Esser TU, Roshanbinfar K, Engel FB. Promoting vascularization for tissue engineering constructs: current strategies focusing on HIF-regulating scaffolds. Expert Opin Biol Ther 2019; 19:105-118. [PMID: 30570406 DOI: 10.1080/14712598.2019.1561855] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Vascularization remains one of the greatest yet unmet challenges in tissue engineering. When engineered tissues are scaled up to therapeutically relevant dimensions, their demand of oxygen and nutrients can no longer be met by diffusion. Thus, there is a need for perfusable vascular structures. Hypoxia-inducible factors (HIF) act as transcriptional oxygen sensors and regulate a multitude of genes involved in adaptive processes to hypoxia, including angiogenesis. Thus, targeting HIFs is a promising strategy to induce vascularization of engineered tissues. AREAS COVERED Here we review current vascularization strategies and summarize the present knowledge regarding activation of HIF signaling by ions, iron chelating agents, α-Ketoglutarate (αKG) analogues, and the lipid-lowering drug simvastatin to induce angiogenesis. Specifically, we focus on the incorporation of HIF-activating agents into biomaterials and scaffolds for controlled release. EXPERT OPINION Vascularization of tissue constructs through activation of upstream regulators of angiogenesis offers advantages but also suffers from drawbacks. HIFs can induce a complete angiogenic program; however, this program appears to be too slow to vascularize larger constructs before cell death occurs. It is therefore crucial that HIF-activation is combined with cell protective strategies and prevascularization techniques to obtain fully vascularized, vital tissues of therapeutically relevant dimensions.
Collapse
Affiliation(s)
- Tilman U Esser
- a Experimental Renal and Cardiovascular Research, Department of Nephropathology , Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Kaveh Roshanbinfar
- a Experimental Renal and Cardiovascular Research, Department of Nephropathology , Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Felix B Engel
- a Experimental Renal and Cardiovascular Research, Department of Nephropathology , Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) , Erlangen , Germany
| |
Collapse
|
21
|
Wan J, Wan H, Yang R, Wan H, Yang J, He Y, Zhou H. Protective effect of Danhong Injection combined with Naoxintong Capsule on cerebral ischemia-reperfusion injury in rats. JOURNAL OF ETHNOPHARMACOLOGY 2018; 211:348-357. [PMID: 28986333 DOI: 10.1016/j.jep.2017.10.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/13/2017] [Accepted: 10/02/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Danhong Injection (DHI) and Naoxintong Capsule (NXT) are renowned traditional Chinese medicine in China. The drug combination of DHI and NXT is frequently applied for the treatment of cardiovascular and cerebrovascular diseases in clinic. However, there had been no pharmacological experiment studies of interaction between DHI and NXT. Due to the drug interactions, exploring their interaction profile is of great importance. MATERIAL AND METHODS In this study, focal cerebral I/R injury in adult male Sprague-Dawley rats were induced by transient middle cerebral artery occlusion (tMCAO) for 1h followed by reperfusion. Rats were divided into 5 groups: sham group, ischemia reperfusion untreated group (IRU), DHI group (DHI 10mL/kg/d), NXT group (NXT 0.5g/kg/d), DHI plus NXT group (DHI-NXT, DHI 10mL/kg/d plus NXT 0.5g/kg/d). All drug-treated groups were respectively successive administrated for 7 days after ischemia/ reperfusion (I/R) injury. The effects on rat neurological function were estimated by neurological defect scores. Brain infarct volumes were determined based on 2, 3, 5-triphenyltetrazolium chloride (TTC) staining. Pathological changes in brain tissues were observed using hematoxylin and eosin (H&E) staining and transmission electron microscope (TEM). Levels of nitric oxide (NO), granulocyte colony-stimulating factor (G-CSF) and granulocyte macrophage colony-stimulating factor (GM-CSF) in serum were determined with enzyme-linked immunosorbent assay (ELISA). Immunohisto-chemistry and Western blot were used to detect the expressions of basic fibroblast growth factor (bFGF), von Willebrand factor-microvessel vascular density (vWF-MVD), vascular endothelial cell growth factor (VEGF), transforming growth factor-β1 (TGF-β1), angiogenin-1 (Ang-1), angiogenin-2 (Ang-2) and platelet derived growth factor (PDGF) at day 7 after ischemia/reperfusion (I/R) injury. RESULTS Compared with IRU group and mono-therapy group (DHI group or NXT group), Danhong Injection combined with Naoxintong Capsule (DHI-NXT) group significantly ameliorated neurological deficits scores, infarct volume and pathological change, significantly decreased the overexpression of NO and the level of Ang-1, significantly increased the expressions of VEGF, Ang-2, G-CSF, GM-CSF, bFGF, PDGF, vWF, TGF-β1. CONCLUSION The protective benefits on rat brain against I/R injury were clearly produced when DHI and NXT were used in combination, which provided rational guidance for clinical combined application of DHI and NXT, and this protection maybe associated with the up-regulation expressions of the related chemokines and growth factors of angiogenesis.
Collapse
Affiliation(s)
- Jiayang Wan
- Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Haofang Wan
- Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Rongbin Yang
- Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Yu He
- Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
22
|
Salvianolic Acid Exerts Cardioprotection through Promoting Angiogenesis in Animal Models of Acute Myocardial Infarction: Preclinical Evidence. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8192383. [PMID: 28713492 PMCID: PMC5497657 DOI: 10.1155/2017/8192383] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/09/2017] [Accepted: 04/24/2017] [Indexed: 12/21/2022]
Abstract
Radix Salviae miltiorrhizae, danshen root (danshen), is one of the widely used Chinese herbal medicines in clinics, containing rich phenolic compounds. Salvianolic acid is the main active compound responsible for the pharmacologic effects of danshen. Here, we aimed to evaluate the effects of salvianolic acid on cardioprotection through promoting angiogenesis in experimental myocardial infarction. Studies of salvianolic acid in animal models of myocardial infarction were obtained from 6 databases until April 2016. The outcome measures were vascular endothelium growth factor (VEGF), blood vessel density (BVD), and myocardial infarct size. All the data were analyzed using Rev-Man 5.3 software. Ultimately, 14 studies were identified involving 226 animals. The quality score of studies ranged from 3 to 6. The meta-analysis of six studies showed significant effects of salvianolic acid on increasing VEGF expression compared with the control group (P < 0.01). The meta-analysis of the two salvianolic acid A studies and three salvianolic acid B studies showed significantly improving BVD compared with the control group (P < 0.01). The meta-analysis of five studies showed significant effects of salvianolic acid for decreasing myocardial infarct size compared with the control group (P < 0.01). In conclusion, these findings demonstrated that salvianolic acid can exert cardioprotection through promoting angiogenesis in animal models of myocardial infarction.
Collapse
|
23
|
Fan ZM, Wang DY, Yang JM, Lin ZX, Lin YX, Yang AL, Fan H, Cao M, Yuan SY, Liu ZJ, Zhou X, Wang YH. Dalbergia odorifera extract promotes angiogenesis through upregulation of VEGFRs and PI3K/MAPK signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2017; 204:132-141. [PMID: 28412217 DOI: 10.1016/j.jep.2017.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/21/2017] [Accepted: 04/07/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The heart wood of Dalbergia odorifera is a Chinese herbal medicine commonly used for the treatment of various ischemic diseases in Chinese medicine practice. AIM OF THE STUDY In this study, therapeutic angiogenesis effects of the Dalbergia odorifera extract (DOE) were investigated on transgenic zebrafish in vivo and human umbilical vein endothelial cells (HUVECs) in vitro. MATERIALS AND METHODS The pro-angiogenic effects of DOE on zebrafish were examined by subintestinal vessels (SIVs) sprouting assay and intersegmental vessels (ISVs) injury assay. And the pro-angiogenic effects of DOE on HUVECs were examined by MTT, scratch assay, protein chip and western blot. RESULTS In the in vivo studies, we found that DOE was able to dose-dependently promote angiogenesis in zebrafish SIVs area. In addition, DOE could also restore the injury in zebrafish ISVs area and upregulate the reduced mRNA expression of VEGFRs including kdr, kdrl and flt-1 induced by VEGF receptor kinase inhibitor II (VRI). In the in vitro studies, we observed that DOE promoted the proliferation, migration of HUVECs and also restored the injury induced by VRI. Moreover, protein chip and western blot experiments showed the PI3K/MAPK cell proliferation/migration pathway were activated by DOE. CONCLUSIONS DOE has a therapeutic effects on angiogenesis, and its mechanism may be related to adjusting the VEGFRs mRNA and activation of PI3K/MAPK signaling pathway. These results suggest a strong potential for Dalbergia odorifera to be developed as an angiogenesis-promoting therapeutic.
Collapse
Affiliation(s)
- Zhu-Ming Fan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Da-Ying Wang
- Central Hospital of Shanghai Xuhui District, Shanghai 200031, PR China
| | - Jian-Mei Yang
- Central Hospital of Shanghai Putuo District, Shanghai 200062, PR China
| | - Zhi-Xiu Lin
- Faculty of Science, School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Yun-Xiao Lin
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Ai-Lin Yang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Hua Fan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Min Cao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Su-Yun Yuan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Zong-Jun Liu
- Central Hospital of Shanghai Xuhui District, Shanghai 200031, PR China
| | - Xin Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China.
| | - You-Hua Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China.
| |
Collapse
|
24
|
Saharinen P, Eklund L, Alitalo K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov 2017; 16:635-661. [PMID: 28529319 DOI: 10.1038/nrd.2016.278] [Citation(s) in RCA: 412] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The endothelial angiopoietin (ANG)-TIE growth factor receptor pathway regulates vascular permeability and pathological vascular remodelling during inflammation, tumour angiogenesis and metastasis. Drugs that target the ANG-TIE pathway are in clinical development for oncological and ophthalmological applications. The aim is to complement current vascular endothelial growth factor (VEGF)-based anti-angiogenic therapies in cancer, wet age-related macular degeneration and macular oedema. The unique function of the ANG-TIE pathway in vascular stabilization also renders this pathway an attractive target in sepsis, organ transplantation, atherosclerosis and vascular complications of diabetes. This Review covers key aspects of the function of the ANG-TIE pathway in vascular disease and describes the recent development of novel therapeutics that target this pathway.
Collapse
Affiliation(s)
- Pipsa Saharinen
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Aapistie 5A, University of Oulu, 90220 Oulu, Finland
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| |
Collapse
|
25
|
Wang J, Zhang L, Pan X, Dai B, Sun Y, Li C, Zhang J. Discovery of multi-target receptor tyrosine kinase inhibitors as novel anti-angiogenesis agents. Sci Rep 2017; 7:45145. [PMID: 28332573 PMCID: PMC5362808 DOI: 10.1038/srep45145] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/17/2017] [Indexed: 01/18/2023] Open
Abstract
Recently, we have identified a biphenyl-aryl urea incorporated with salicylaldoxime (BPS-7) as an anti-angiogenesis agent. Herein, we disclosed a series of novel anti-angiogenesis agents with BPS-7 as lead compound through combining diarylureas with N-pyridin-2-ylcyclopropane carboxamide. Several title compounds exhibited simultaneous inhibition effects against three pro-angiogenic RTKs (VEGFR-2, TIE-2 and EphB4). Some of them displayed potent anti-proliferative activity against human vascular endothelial cell (EA.hy926). In particular, two potent compounds (CDAU-1 and CDAU-2) could be considered as promising anti-angiogenesis agents with triplet inhibition profile. The biological evaluation and molecular docking results indicate that N-pyridin-2-ylcyclopropane carboxamide could serve as a hinge-binding group (HBG) for the discovery of multi-target anti-angiogenesis agents. CDAU-2 also exhibited promising anti-angiogenic potency in a tissue model for angiogenesis.
Collapse
Affiliation(s)
- Jinfeng Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta West Road, Xi'an, 710061, P.R. China
| | - Lin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta West Road, Xi'an, 710061, P.R. China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta West Road, Xi'an, 710061, P.R. China
| | - Bingling Dai
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta West Road, Xi'an, 710061, P.R. China
| | - Ying Sun
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta West Road, Xi'an, 710061, P.R. China
| | - Chuansheng Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta West Road, Xi'an, 710061, P.R. China
| | - Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta West Road, Xi'an, 710061, P.R. China
| |
Collapse
|
26
|
Correlative Imaging of the Murine Hind Limb Vasculature and Muscle Tissue by MicroCT and Light Microscopy. Sci Rep 2017; 7:41842. [PMID: 28169309 PMCID: PMC5294414 DOI: 10.1038/srep41842] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 12/22/2016] [Indexed: 12/20/2022] Open
Abstract
A detailed vascular visualization and adequate quantification is essential for the proper assessment of novel angiomodulating strategies. Here, we introduce an ex vivo micro-computed tomography (microCT)-based imaging approach for the 3D visualization of the entire vasculature down to the capillary level and rapid estimation of the vascular volume and vessel size distribution. After perfusion with μAngiofil®, a novel polymerizing contrast agent, low- and high-resolution scans (voxel side length: 2.58–0.66 μm) of the entire vasculature were acquired. Based on the microCT data, sites of interest were defined and samples further processed for correlative morphology. The solidified, autofluorescent μAngiofil® remained in the vasculature and allowed co-registering of the histological sections with the corresponding microCT-stack. The perfusion efficiency of μAngiofil® was validated based on lectin-stained histological sections: 98 ± 0.5% of the blood vessels were μAngiofil®-positive, whereas 93 ± 2.6% were lectin-positive. By applying this approach we analyzed the angiogenesis induced by the cell-based delivery of a controlled VEGF dose. Vascular density increased by 426% mainly through the augmentation of medium-sized vessels (20–40 μm). The introduced correlative and quantitative imaging approach is highly reproducible and allows a detailed 3D characterization of the vasculature and muscle tissue. Combined with histology, a broad range of complementary structural information can be obtained.
Collapse
|
27
|
Rufaihah AJ, Johari NA, Vaibavi SR, Plotkin M, Di Thien DT, Kofidis T, Seliktar D. Dual delivery of VEGF and ANG-1 in ischemic hearts using an injectable hydrogel. Acta Biomater 2017; 48:58-67. [PMID: 27756647 DOI: 10.1016/j.actbio.2016.10.013] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 10/10/2016] [Accepted: 10/10/2016] [Indexed: 01/08/2023]
Abstract
Acute myocardial infarction (MI) caused by ischemia is the most common cause of cardiac dysfunction. While growth factor therapy is promising, the retention in the highly vascularized myocardium is limited and prevents sustained activation needed for adequate cellular responses. Here, we demonstrated the use of polyethylene glycol-fibrinogen (PF) hydrogels for sustained dual delivery of vascular endothelial growth factor (VEGF) and angiopoietin-1 (ANG-1) to enhance myocardial repair and function. VEGF and ANG-1 were incorporated in PF hydrogels and their in vitro characteristics were studied. Acute MI was generated in a rodent model with rats randomly assigned to 4 groups; sham, saline, PF and PF-VEGF-ANG1 (n=10 each group). Saline or hydrogel was injected in infarct and peri-infarct areas of the myocardium. After 4weeks, myocardial function was assessed using echocardiography. Tissue samples were harvested for Hematoxylin and Eosin, Masson Trichrome and capillary staining to assess the extent of fibrotic scar and arteriogenesis. Both VEGF and ANG-1 were released in a sustained and controlled manner over 30days. PF-VEGF-ANG1 treated animals showed the best improvement in cardiac function, highest degree of cardiac muscle preservation, and arteriogenesis. This study demonstrates that PF hydrogels can simultaneously provide mechanical support to attenuate adverse myocardial remodelling, and a pro-angiogenic benefit from the sustained VEGF and ANG1 delivery that culminates in a restorative effect following MI. The utility of this synergistic, biomaterial-based growth factor delivery may have clinical implications in the prevention of post-MI cardiac dysfunction. STATEMENT OF SIGNIFICANCE Acute myocardial infarction (MI) caused by ischemia is the most common cause of cardiac dysfunction. Here, we demonstrated the use of polyethylene glycol-fibrinogen (PF) hydrogels for sustained dual delivery of vascular endothelial growth factor (VEGF) and angiopoietin-1 (ANG-1) to enhance myocardial repair and function. Treated animals showed the best improvement in cardiac function, highest degree of cardiac muscle preservation, and arteriogenesis. This study demonstrates that PF hydrogels can simultaneously provide mechanical support to attenuate adverse myocardial remodelling, and a pro-angiogenic benefit from the sustained VEGF and ANG1 delivery that culminates in a restorative effect following MI.
Collapse
|
28
|
Hinkel R, Kupatt C. Selective Pressure-Regulated Retroinfusion for Gene Therapy Application in Ischemic Heart Disease. Methods Mol Biol 2017; 1521:249-260. [PMID: 27910055 DOI: 10.1007/978-1-4939-6588-5_18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Coronary heart disease is still the leading cause of death in industrialized nations. Even though revascularization strategies such as coronary artery bypass graft surgery, percutaneous coronary intervention and enhanced drug therapy significantly improved the outcome, about 30 % of patients develop chronic heart failure. Ischemic heart disease and heart failure are characterized by an adverse remodeling of the heart, featuring cardiomyocyte hypertrophy, increased fibrosis and capillary rarefaction. Therefore, gene therapeutic approaches for the treatment of heart failure, such as the modulating contractile function or therapeutic neovascularization, seem to be promising. To achieve an efficient transduction of the gene therapeutic agent, the time point and the application route seem to be important for the therapeutic success. In contrast to the classical systemic application regional intra-coronary application offers the possibility of higher transduction efficacy in the target area accompanied by a reduced off-target contamination. Antegrade delivery however, may be impaired by coronary heart disease, such as stenosis or occlusion of a coronary artery. Coronary veins appear not to be affected and might therefore be the preferable application route for gene therapy. For an effective and safe retrograde application in gene therapy, selective catheterization of the coronary vein draining the target area is necessary. In addition, to avoid coronary vein injury, a pressure regulated infusion enhances safety. Therefore, a selective pressure regulation of retroinfusion (SSR) seems to be a favorable approach for gene therapy transduction in combination with reduced systemic contamination.
Collapse
Affiliation(s)
- Rabea Hinkel
- Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, TU Munich, Ismaningerstr. 22, 81675, Munich, Germany. .,Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany. .,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Christian Kupatt
- Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, TU Munich, Ismaningerstr. 22, 81675, Munich, Germany.,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
29
|
Rychter M, Baranowska-Korczyc A, Lulek J. Progress and perspectives in bioactive agent delivery via electrospun vascular grafts. RSC Adv 2017. [DOI: 10.1039/c7ra04735e] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The review discusses the progress in the design and synthesis of bioactive agents incorporated into vascular grafts obtained by the electrospinning process.
Collapse
Affiliation(s)
- Marek Rychter
- Department of Pharmaceutical Technology
- Faculty of Pharmacy
- Poznan University of Medical Sciences
- 60-780 Poznan
- Poland
| | | | - Janina Lulek
- Department of Pharmaceutical Technology
- Faculty of Pharmacy
- Poznan University of Medical Sciences
- 60-780 Poznan
- Poland
| |
Collapse
|
30
|
Isidori AM, Venneri MA, Fiore D. Angiopoietin-1 and Angiopoietin-2 in metabolic disorders: therapeutic strategies to restore the highs and lows of angiogenesis in diabetes. J Endocrinol Invest 2016; 39:1235-1246. [PMID: 27344309 DOI: 10.1007/s40618-016-0502-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 06/08/2016] [Indexed: 12/14/2022]
Abstract
The morbidity and mortality of diabetes mellitus are mostly attributed to cardiovascular complications. Despite tremendous advancement in glycemic control, anti-diabetic medications have failed to revert vascular impairment once triggered by the metabolic disorder. The angiogenic growth factors, Angiopoietin-1 (Ang1) and Angiopoietin-2 (Ang2), are crucial regulators of vessel formation and maintenance starting with embryonic development and continuing through life. In mature vessels, angiopoietins control vascular permeability, inflammation and remodeling. A crucial role of angiopoietins is to drive vascular inflammation from the active to the quiescent state, enabling restoration of tissue homeostasis. The mechanism is of particular importance for healing and repair after damage, two conditions typically impaired in metabolic disorders. There is an emerging body of evidences suggesting that the imbalance of Ang1 and Ang2 regulation, leading to an increased Ang2/Ang1 ratio, represents a culprit of the vascular alterations of patients with type-2 diabetes mellitus. Pharmacological modulation of Ang1 or Ang2 actions may help prevent or delay the onset of diabetic vascular complications by restoring vessel function, favoring tissue repair and maintaining endothelial quiescence. In this review, we present a summary of the role of Ang1 and Ang2, their involvement in diabetic complications, and novel therapeutic strategies targeting angiopoietins to ameliorate vascular health in metabolic disorders.
Collapse
Affiliation(s)
- A M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy.
| | - M A Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - D Fiore
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| |
Collapse
|
31
|
A Designed Angiopoietin-1 Variant, Dimeric CMP-Ang1 Activates Tie2 and Stimulates Angiogenesis and Vascular Stabilization in N-glycan Dependent Manner. Sci Rep 2015; 5:15291. [PMID: 26478188 PMCID: PMC4609988 DOI: 10.1038/srep15291] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 09/22/2015] [Indexed: 01/09/2023] Open
Abstract
Angiopoietin-1 (Ang1), a potential growth factor for therapeutic angiogenesis and vascular stabilization, is known to specifically cluster and activate Tie2 in high oligomeric forms, which is a unique and essential process in this ligand-receptor interaction. However, highly oligomeric native Ang1 and Ang1 variants are difficult to produce, purify, and store in a stable and active form. To overcome these limitations, we developed a simple and active dimeric CMP-Ang1 by replacing the N-terminal of native Ang1 with the coiled-coil domain of cartilage matrix protein (CMP) bearing mutations in its cysteine residues. This dimeric CMP-Ang1 effectively increased the migration, survival, and tube formation of endothelial cells via Tie2 activation. Furthermore, dimeric CMP-Ang1 induced angiogenesis and suppressed vascular leakage in vivo. Despite its dimeric structure, the potencies of such Tie2-activation-induced effects were comparable to those of a previously engineered protein, COMP-Ang1. We also revealed that these effects of dimeric CMP-Ang1 were affected by specified N-glycosylation in its fibrinogen-like domain. Taken together, our results indicate that dimeric CMP-Ang1 is capable of activating Tie2 and stimulating angiogenesis in N-glycan dependent manner.
Collapse
|
32
|
Laakkonen JP, Ylä-Herttuala S. Recent Advancements in Cardiovascular Gene Therapy and Vascular Biology. Hum Gene Ther 2015; 26:518-24. [DOI: 10.1089/hum.2015.095] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Johanna P. Laakkonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, University of Eastern Finland, Kuopio, Finland
- Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
33
|
Chu F, Jin X, Ma H, Lu X, Zhu J. Effect of Musca domestic maggot polypeptide extract on HUVEC dysfunction induced by early-activated macrophages. PHARMACEUTICAL BIOLOGY 2015; 54:572-575. [PMID: 26931348 DOI: 10.3109/13880209.2015.1060506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
CONTEXT Musca domestica Linn. maggot is a traditional Chinese medicine. In our previous studies, Musca domestica maggot protein-enriched fraction and polypeptide extract (molecular weight <30 kD) were found to reverse endothelial cell dysfunction in atherosclerotic lesions. OBJECTIVE This study determines whether and how M. domestica maggot polypeptide extract affects the dysfunction of human umbilical vein endothelial cells (HUVEC) induced by macrophages (Mϕ). MATERIALS AND METHODS HUVEC and early-activated THP-1 Mϕ (incubated with LPS of 1 μg/ml for 2 h) were co-cultured in a Transwell system. The effects of Musca domestica maggot polypeptide extract (with increasing concentrations, i.e., 1.0, 2.5, 5.0, 10.0, 20.0, and 40.0 µg/ml) on the proliferation and migration HUVEC and their secretion of vascular endothelial growth factor (VEGF) were determined by flow cytometry, modified Boyden chamber assay, and enzyme-linked immunosorbent assay (ELISA) after incubation for 24 h. RESULTS Musca domestica maggot polypeptide extract decreased the proliferation of HUVEC in a concentration-dependent manner, with a 50% effective concentration (EC50) of 22.16 ± 1.48 µg/ml, and effectively inhibited HUVEC migration (EC50 = 35.15 ± 2.03 µg/ml) and VEGF secretion (EC50 = 28.64 ± 1.29 µg/ml). DISCUSSION AND CONCLUSION Musca domestica maggot polypeptide extract can inhibit the dysfunction of HUVEC induced by early-activated THP-1 Mϕ.
Collapse
Affiliation(s)
- Fujiang Chu
- a Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Basic Courses , Guangdong Pharmaceutical University , Guangzhou , China and
| | - Xiaobao Jin
- a Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Basic Courses , Guangdong Pharmaceutical University , Guangzhou , China and
| | - Hongyan Ma
- b School of Traditional Chinese Medicine, Guangdong Pharmaceutical University , Guangzhou , China
| | - Xuemei Lu
- a Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Basic Courses , Guangdong Pharmaceutical University , Guangzhou , China and
| | - Jiayong Zhu
- a Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Basic Courses , Guangdong Pharmaceutical University , Guangzhou , China and
| |
Collapse
|
34
|
Andersson L, Scharin Täng M, Lundqvist A, Lindbom M, Mardani I, Fogelstrand P, Shahrouki P, Redfors B, Omerovic E, Levin M, Borén J, Levin MC. Rip2 modifies VEGF-induced signalling and vascular permeability in myocardial ischaemia. Cardiovasc Res 2015; 107:478-86. [DOI: 10.1093/cvr/cvv186] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 06/12/2015] [Indexed: 01/04/2023] Open
|
35
|
Rivera LB, Meyronet D, Hervieu V, Frederick MJ, Bergsland E, Bergers G. Intratumoral myeloid cells regulate responsiveness and resistance to antiangiogenic therapy. Cell Rep 2015; 11:577-91. [PMID: 25892230 DOI: 10.1016/j.celrep.2015.03.055] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/02/2015] [Accepted: 03/25/2015] [Indexed: 12/21/2022] Open
Abstract
Antiangiogenic therapy is commonly used in the clinic, but its beneficial effects are short-lived, leading to tumor relapse within months. Here, we found that the efficacy of angiogenic inhibitors targeting the VEGF/VEGFR pathway was dependent on induction of the angiostatic and immune-stimulatory chemokine CXCL14 in mouse models of pancreatic neuroendocrine and mammary tumors. In response, tumors reinitiated angiogenesis and immune suppression by activating PI3K signaling in all CD11b+ cells, rendering tumors nonresponsive to VEGF/VEGFR inhibition. Adaptive resistance was also associated with an increase in Gr1+CD11b+ cells, but targeting Gr1+ cells was not sufficient to further sensitize angiogenic blockade because tumor-associated macrophages (TAMs) would compensate for the lack of such cells and vice versa, leading to an oscillating pattern of distinct immune-cell populations. However, PI3K inhibition in CD11b+ myeloid cells generated an enduring angiostatic and immune-stimulatory environment in which antiangiogenic therapy remained efficient.
Collapse
Affiliation(s)
- Lee B Rivera
- Department of Neurological Surgery, Brain Tumor Research Center, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - David Meyronet
- Université Lyon 1, Centre de Pathologie et Neuropathologie Est, Hospices Civils de Lyon, Bron Cedex 69677, France
| | - Valérie Hervieu
- Université Lyon 1, Service d'Anatomie Pathologique, Hôpital Édouard Herriot, Hospices Civils de Lyon, Lyon Cedex 69003, France
| | - Mitchell J Frederick
- Department of Head and Neck Surgery, Research Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Emily Bergsland
- Department of Medicine, UCSF Mount Zion Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gabriele Bergers
- Department of Neurological Surgery, Brain Tumor Research Center, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
36
|
Teichert M, Stumpf C, Booken N, Wobser M, Nashan D, Hallermann C, Mogler C, Müller CSL, Becker JC, Moritz RKC, Andrulis M, Nicolay JP, Goerdt S, Thomas M, Klemke CD, Augustin HG, Felcht M. Aggressive primary cutaneous B-cell lymphomas show increased Angiopoietin-2-induced angiogenesis. Exp Dermatol 2015; 24:424-9. [PMID: 25776770 DOI: 10.1111/exd.12688] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2015] [Indexed: 12/19/2022]
Abstract
Primary cutaneous large B-cell lymphomas, leg type (PCLBCL/LT) are primary cutaneous B-cell lymphoma (PCBCL) with an intermediate prognosis. Therefore, antracycline-based polychemotherapy combined with rituximab has been recommended as first-line treatment. Yet, despite this regimen, the 5-year survival rate remains 50-66% only. Angiogenesis, the formation of a vascular network, is essential for the pathogenesis of nodal lymphomas. So far, no study has analysed angiogenesis and its key factors in PCLBCL/LT. The present study was aimed at characterizing angiogenesis in PCLBCL/LT to identify the angiogenic molecules as potential therapeutic targets. The intra-tumoral microvessel density (MVD) was assessed by immunohistochemical studies of CD20 and CD31. The MVD was higher in PCLBCL/LT compared with indolent PCBCL. Analyses of open-source microarray data showed correlation between the angiogenic molecule angiopoietin-2 (Ang-2) and pan-endothelial cell markers. ELISA studies determined a shift between Ang-2 and Ang-1 towards Ang-2 in the peripheral blood of PCLBCL/LT patients. Immunofluorescence costainings against the Ang receptor Tie2/angiogenic integrins/CD34 revealed that the vasculature in both aggressive and indolent PCBCL tumors harbours an endothelial cell subpopulation with reduced expression of Tie2. In contrast, the alternative Ang-2 binding partners, angiogenic integrins, are strongly expressed in PCBCL. In line with these findings, downstream targets of Ang-2-integrin signalling, that is phosphorylation of focal adhesion kinase at Tyr397, and sprouting angiogenesis are enhanced in PCLBCL/LT. Our data present Ang-2 as a promising therapeutic target and anti-angiogenic therapy as a new line in treatment of PCLBCL/LT as a hitherto intractable disease.
Collapse
Affiliation(s)
- Martin Teichert
- Division of Vascular Oncology and Metastasis, German Cancer Research Centre Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Christine Stumpf
- Department of Dermatology, Venereology and Allergy, University Medical Centre Mannheim, Centre of Excellence of Dermatology of Baden-Württemberg, Ruprecht-Karls-University of Heidelberg, Mannheim, Germany
| | - Nina Booken
- Department of Dermatology, Venereology and Allergy, University Medical Centre Mannheim, Centre of Excellence of Dermatology of Baden-Württemberg, Ruprecht-Karls-University of Heidelberg, Mannheim, Germany
| | - Marion Wobser
- Department of Dermatology, Venereology and Allergy, Julius-Maximilians-University, Würzburg, Germany
| | - Dorothee Nashan
- Department of Dermatology, Hospital of Dortmund, Dortmund, Germany
| | | | - Carolin Mogler
- Division of Vascular Oncology and Metastasis, German Cancer Research Centre Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany.,Institute of Pathology, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
| | - Cornelia S L Müller
- Department of Dermatology, Venereology and Allergy, Saarland University Hospital, Homburg/Saar, Germany
| | - Jürgen C Becker
- Translational Skin Cancer Research, Deutsches Konsortium für Translationale Krebsforschung (DKTK), University Hospital Essen
| | - Rose K C Moritz
- Department of Dermatology, Venereology and Allergy, University of Bochum, Bochum, Germany
| | - Mindaugas Andrulis
- Institute of Pathology, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
| | - Jan P Nicolay
- Department of Dermatology, Venereology and Allergy, University Medical Centre Mannheim, Centre of Excellence of Dermatology of Baden-Württemberg, Ruprecht-Karls-University of Heidelberg, Mannheim, Germany
| | - Sergij Goerdt
- Department of Dermatology, Venereology and Allergy, University Medical Centre Mannheim, Centre of Excellence of Dermatology of Baden-Württemberg, Ruprecht-Karls-University of Heidelberg, Mannheim, Germany
| | - Markus Thomas
- Discovery Oncology, Pharmaceutical Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - Claus-Detlev Klemke
- Department of Dermatology, Venereology and Allergy, University Medical Centre Mannheim, Centre of Excellence of Dermatology of Baden-Württemberg, Ruprecht-Karls-University of Heidelberg, Mannheim, Germany
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Centre Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany.,Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moritz Felcht
- Division of Vascular Oncology and Metastasis, German Cancer Research Centre Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergy, University Medical Centre Mannheim, Centre of Excellence of Dermatology of Baden-Württemberg, Ruprecht-Karls-University of Heidelberg, Mannheim, Germany
| |
Collapse
|
37
|
Terzuoli E, Monti M, Vellecco V, Bucci M, Cirino G, Ziche M, Morbidelli L. Characterization of zofenoprilat as an inducer of functional angiogenesis through increased H2 S availability. Br J Pharmacol 2015; 172:2961-73. [PMID: 25631232 DOI: 10.1111/bph.13101] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 01/13/2015] [Accepted: 01/22/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Hydrogen sulfide (H2 S), an endogenous volatile mediator with pleiotropic functions, promotes vasorelaxation, exerts anti-inflammatory actions and regulates angiogenesis. Previously, the SH-containing angiotensin-converting enzyme inhibitor (ACEI), zofenopril, was identified as being effective in preserving endothelial function and inducing angiogenesis among ACEIs. Based on the H2 S donor property of its active metabolite zofenoprilat, the objective of this study was to evaluate whether zofenoprilat-induced angiogenesis was due to increased H2 S availability. EXPERIMENTAL APPROACH HUVECs were used for in vitro studies of angiogenesis, whereas the Matrigel plug assay was used for in vivo assessments. KEY RESULTS Zofenoprilat-treated HUVECs showed an increase in all functional features of the angiogenic process in vitro. As zofenoprilat induced the expression of CSE (cystathionine-γ-lyase) and the continuous production of H2 S, CSE inhibition or silencing blocked the ability of zofenoprilat to induce angiogenesis, both in vitro and in vivo. The molecular mechanisms underlying H2 S/zofenoprilat-induced angiogenesis were dependent on Akt, eNOS and ERK1/2 cascades. ATP-sensitive potassium (KATP ) channels, the molecular target that mediates part of the vascular functions of H2 S, were shown to be involved in the upstream activation of Akt and ERK1/2. Moreover, the up-regulation of fibroblast growth factor-2 was dependent on CSE-derived H2 S response to H2 S and KATP activation. CONCLUSIONS AND IMPLICATIONS Zofenoprilat induced a constant production of H2 S that stimulated the angiogenic process through a KATP channel/Akt/eNOS/ERK1/2 pathway. Thus, zofenopril can be considered as a pro-angiogenic drug acting through H2 S release and production, useful in cardiovascular pathologies where vascular functions need to be re-established and functional angiogenesis induced.
Collapse
Affiliation(s)
- E Terzuoli
- Department of Life Sciences, University of Siena, Siena, Italy
| | - M Monti
- Department of Life Sciences, University of Siena, Siena, Italy
| | - V Vellecco
- Department of Experimental Pharmacology, University of Naples Federico II, Naples, Italy
| | - M Bucci
- Department of Experimental Pharmacology, University of Naples Federico II, Naples, Italy
| | - G Cirino
- Department of Experimental Pharmacology, University of Naples Federico II, Naples, Italy
| | - M Ziche
- Department of Life Sciences, University of Siena, Siena, Italy
| | - L Morbidelli
- Department of Life Sciences, University of Siena, Siena, Italy
| |
Collapse
|
38
|
Abstract
The endothelial TIE1 and TIE2 receptor tyrosine kinases form a distinct subfamily characterized by their unique extracellular domains. Together with the angiopoietin growth factors (ANGPT1, ANGPT2, ANGPT4, also abbreviated as ANG), the TIE receptors form an endothelial specific signaling pathway with important functions in the regulation of lymphatic and cardiovascular development and vascular homeostasis. Angiopoietins exist in multimeric forms that activate the TIE receptors via unique mechanism. In endothelial cell–cell contacts, angiopoietins induce the formation of homomeric in trans TIE receptor complexes extending across the cell junctions, whereas matrix-bound angiopoietin-1 (ANG1) activates the TIE receptors in a cis configuration. In comparison to the vascular endothelial growth factor receptors, the TIE receptors undergo little ubiquitin-mediated degradation after activation, whereas TIE2 signaling is negatively regulated by the vascular endothelial protein tyrosine phosphatase, VE-PTP. ANG1 activation of TIE2 supports vascular stabilization, whereas angiopoietin-2 (ANG2), a context-dependent weak TIE2 agonist/antagonist, promotes pathological tumor angiogenesis, vascular permeability, and inflammation. Recently, ANG2 has been found to mediate some of its vascular destabilizing and angiogenic functions via integrin signalling. The circulating levels of ANG2 are increased in cancer, and in several human diseases associated with inflammation and vascular leak, for example, in sepsis. Blocking of ANG2 has emerged as a potential novel therapeutic strategy for these diseases. In addition, preclinical results demonstrate that genetic TIE1 deletion in mice inhibits the vascularization and growth of tumor isografts and protects from atherosclerosis, with little effect on normal vascular homeostasis in adult mice. The ability of the ANG-TIE pathway to control vessel stability and angiogenesis makes it an interesting vascular target for the treatment of the various diseases.
Collapse
|
39
|
Moon HE, Byun K, Park HW, Kim JH, Hur J, Park JS, Jun JK, Kim HS, Paek SL, Kim IK, Hwang JH, Kim JW, Kim DG, Sung YC, Koh GY, Song CW, Lee B, Paek SH. COMP-Ang1 Potentiates EPC Treatment of Ischemic Brain Injury by Enhancing Angiogenesis Through Activating AKT-mTOR Pathway and Promoting Vascular Migration Through Activating Tie2-FAK Pathway. Exp Neurobiol 2015; 24:55-70. [PMID: 25792870 PMCID: PMC4363334 DOI: 10.5607/en.2015.24.1.55] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 03/04/2015] [Accepted: 03/05/2015] [Indexed: 12/28/2022] Open
Abstract
Successful recovery from brain ischemia is limited due to poor vascularization surrounding the ischemic zone. Cell therapy with strong angiogenic factors could be an effective strategy to rescue the ischemic brain. We investigated whether cartilage oligomeric matrix protein (COMP)-Ang1, a soluble, stable and potent Ang1 variant, enhances the angiogenesis of human cord blood derived endothelial progenitor cells (hCB-EPCs) for rescuing brain from ischemic injury. COMP-Ang1 markedly improved the tube formation of capillaries by EPCs and incorporation of EPCs into tube formation with human umbilical vein endothelial cells (HUVECs) upon incubation on matrigel in vitro. COMP-Ang1 stimulated the migration of EPCs more than HUVECs in a scratch wound migration assay. The transplanted EPCs and COMP-Ang1 were incorporated into the blood vessels and decreased the infarct volume in the rat ischemic brain. Molecular studies revealed that COMP-Ang1 induced an interaction between Tie2 and FAK, but AKT was separated from the Tie2-FAK-AKT complex in the EPC plasma membrane. Tie2-FAK increased pp38, pSAPK/JNK, and pERK-mediated MAPK activation and interacted with integrins ανβ3, α4, β1, finally leading to migration of EPCs. AKT recruited mTOR, SDF-1, and HIF-1α to induce angiogenesis. Taken together, it is concluded that COMP-Ang1 potentiates the angiogenesis of EPCs and enhances the vascular morphogenesis indicating that combination of EPCs with COMP-Ang1 may be a potentially effective regimen for ischemic brain injury salvage therapy.
Collapse
Affiliation(s)
- Hyo Eun Moon
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Kyunghee Byun
- Center for Regenerative Medicine, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-840, Korea. ; Department of Anatomy and Cell Biology, Gachon University Medical School, Incheon 406-840, Korea
| | - Hyung Woo Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Jin Hyun Kim
- Clinical Research Institute, Gyeongsang National University Hospital, Jinju 660-702, Korea
| | - Jin Hur
- Innovative Research Institute for Cell Therapy (IRICT), Seoul National University Hospital, Seoul 110-744, Korea
| | - Joong Shin Park
- Department of Obstetrics & Gynecology, Seoul National University Hospital, Seoul 110-744, Korea
| | - Jong Kwan Jun
- Department of Obstetrics & Gynecology, Seoul National University Hospital, Seoul 110-744, Korea
| | - Hyo-Soo Kim
- Innovative Research Institute for Cell Therapy (IRICT), Seoul National University Hospital, Seoul 110-744, Korea
| | - Seung Leal Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Department of Neurosurgery, Mayo Clinic, USA
| | - In Keyoung Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Jae Ha Hwang
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Jin Wook Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Dong Gyu Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Young Chul Sung
- Division of Molecular and Life Science, Integrative Bioscience & Biotechnology, Pohang University of Science and Technology, Pohang 790-784, USA
| | - Gou-Young Koh
- Laboratory for Vascular Biology and Stem Cell, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-338, Korea
| | - Chang W Song
- Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Medical School, Minneapolis, USA
| | - Bonghee Lee
- Center for Regenerative Medicine, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-840, Korea. ; Department of Anatomy and Cell Biology, Gachon University Medical School, Incheon 406-840, Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-744, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 110-744, Korea
| |
Collapse
|
40
|
Scholz A, Plate KH, Reiss Y. Angiopoietin-2: a multifaceted cytokine that functions in both angiogenesis and inflammation. Ann N Y Acad Sci 2015; 1347:45-51. [PMID: 25773744 DOI: 10.1111/nyas.12726] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 01/19/2015] [Accepted: 01/23/2015] [Indexed: 12/12/2022]
Abstract
Angiogenesis and inflammation are two highly linked processes. In the last decade, several factors with dual function in both of these major pathways have been identified. This review focuses on angiopoietin-2 (Ang-2), an important proangiogenic factor that has more recently been implicated in mediating inflammatory processes as well. Ang-2 is upregulated in multiple inflammatory diseases and has been implicated in the direct control of inflammation-related signaling pathways. As a consequence of its multiple roles, designs for therapeutic targeting of Ang-2 should consider the dual function of this factor in regulating angiogenesis and inflammation.
Collapse
Affiliation(s)
- Alexander Scholz
- Department of Pathology, Laboratory of Immunology and Vascular Biology, Stanford University School of Medicine, Stanford, California
| | - Karl H Plate
- Edinger Institute/Institute of Neurology, Frankfurt University Medical School, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt, Germany.,German Cancer Consortium (DKTK), Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yvonne Reiss
- Edinger Institute/Institute of Neurology, Frankfurt University Medical School, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt, Germany.,German Cancer Consortium (DKTK), Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
41
|
Mammoto T, Jiang A, Jiang E, Mammoto A. Platelet-rich plasma extract prevents pulmonary edema through angiopoietin-Tie2 signaling. Am J Respir Cell Mol Biol 2015; 52:56-64. [PMID: 24960457 DOI: 10.1165/rcmb.2014-0076oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Increased vascular permeability contributes to life-threatening pathological conditions, such as acute respiratory distress syndrome. Current treatments for sepsis-induced pulmonary edema rely on low-tidal volume mechanical ventilation, fluid management, and pharmacological use of a single angiogenic or chemical factor with antipermeability activity. However, it is becoming clear that a combination of multiple angiogenic/chemical factors rather than a single factor is required for maintaining stable and functional blood vessels. We have demonstrated that mouse platelet-rich plasma (PRP) extract contains abundant angiopoietin (Ang) 1 and multiple other factors (e.g., platelet-derived growth factor), which potentially stabilize vascular integrity. Here, we show that PRP extract increases tyrosine phosphorylation levels of Tunica internal endothelial cell kinase (Tie2) and attenuates disruption of cell-cell junctional integrity induced by inflammatory cytokine in cultured human microvascular endothelial cells. Systemic injection of PRP extract also increases Tie2 phosphorylation in mouse lung and prevents endotoxin-induced pulmonary edema and the consequent decreases in lung compliance and exercise intolerance resulting from endotoxin challenge. Soluble Tie2 receptor, which inhibits Ang-Tie2 signaling, suppresses the ability of PRP extract to inhibit pulmonary edema in mouse lung. These results suggest that PRP extract prevents endotoxin-induced pulmonary edema mainly through Ang-Tie2 signaling, and PRP extract could be a potential therapeutic strategy for sepsis-induced pulmonary edema and various lung diseases caused by abnormal vascular permeability.
Collapse
Affiliation(s)
- Tadanori Mammoto
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | | | | |
Collapse
|
42
|
The angiopoietin/TIE receptor system: Focusing its role for ischemia-reperfusion injury. Cytokine Growth Factor Rev 2014; 26:281-91. [PMID: 25466648 DOI: 10.1016/j.cytogfr.2014.10.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 10/23/2014] [Accepted: 10/30/2014] [Indexed: 02/07/2023]
Abstract
Ischemia and reperfusion (I/R) are of fatal consequence for the affected organs, as they provoke a profound inflammatory reaction. This thoroughly destroys cells and tissues, inducing functional failure or even complete loss of organ function. Since I/R is primarily a vascular problem, the interaction between the endothelium and the surrounding environment is of great significance. The angiopoietins (ANG) and the TIE receptors are key players for the vascular homeostasis. This review summarizes biochemical and cellular mechanisms leading to I/R injury. After a brief introduction to the ANG/TIE system, a comprehensive overview of its role for the development of I/R syndrome is given. Finally, current therapeutic approaches to mitigate the consequences of I/R by modulating ANG/TIE signaling are reviewed in detail.
Collapse
|
43
|
Wang B, Yang Q, Bai WW, Xing YF, Lu XT, Sun YY, Zhao YX. Tongxinluo protects against pressure overload-induced heart failure in mice involving VEGF/Akt/eNOS pathway activation. PLoS One 2014; 9:e98047. [PMID: 24887083 PMCID: PMC4041651 DOI: 10.1371/journal.pone.0098047] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 04/28/2014] [Indexed: 02/01/2023] Open
Abstract
Background It has been demonstrated that Tongxinluo (TXL), a traditional Chinese medicine compound, improves ischemic heart disease in animal models via vascular endothelial growth factor (VEGF) and endothelial nitric oxide synthase (eNOS). The present study aimed to investigate whether TXL protects against pressure overload–induced heart failure in mice and explore the possible mechanism of action. Methods and Results Transverse aortic constriction (TAC) surgery was performed in mice to induce heart failure. Cardiac function was evaluated by echocardiography. Myocardial pathology was detected using hematoxylin and eosin or Masson trichrome staining. We investigated cardiomyocyte ultrastructure using transmission electron microscopy. Angiogenesis and oxidative stress levels were determined using CD31 and 8-hydroxydeoxyguanosine immunostaining and malondialdehyde assay, respectively. Fetal gene expression was measured using real-time PCR. Protein expression of VEGF, phosphorylated (p)-VEGF receptor 2 (VEGFR2), p–phosphatidylinositol 3-kinase (PI3K), p-Akt, p-eNOS, heme oxygenase-1 (HO-1), and NADPH oxidase 4 (Nox4) were measured with western blotting. Twelve-week low- and high-dose TXL treatment following TAC improved cardiac systolic and diastolic function and ameliorated left ventricular hypertrophy, fibrosis, and myocardial ultrastructure derangement. Importantly, TXL increased myocardial capillary density significantly and attenuated oxidative stress injury in failing hearts. Moreover, TXL upregulated cardiac nitrite content and the protein expression of VEGF, p-VEGFR2, p-PI3K, p-Akt, p-eNOS, and HO-1, but decreased Nox4 expression in mouse heart following TAC. Conclusion Our findings indicate that TXL protects against pressure overload–induced heart failure in mice. Activation of the VEGF/Akt/eNOS signaling pathway might be involved in TXL improvement of the failing heart.
Collapse
Affiliation(s)
- Bo Wang
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Qing Yang
- Institute of Pathogen Biology, Shandong University, Jinan, China
| | - Wen-wu Bai
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, China
| | - Yi-fan Xing
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao-ting Lu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, China
| | - Yuan-yuan Sun
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, China
| | - Yu-xia Zhao
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, China
- * E-mail:
| |
Collapse
|
44
|
Takeda K, Duan LJ, Takeda H, Fong GH. Improved vascular survival and growth in the mouse model of hindlimb ischemia by a remote signaling mechanism. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:686-96. [PMID: 24440788 DOI: 10.1016/j.ajpath.2013.11.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 11/15/2013] [Accepted: 11/19/2013] [Indexed: 02/07/2023]
Abstract
Deficiencies in prolyl hydroxylase domain proteins (PHDs) may lead to the accumulation of hypoxia-inducible factor-α proteins, the latter of which activate local angiogenic responses by paracrine mechanisms. Here, we investigate whether a keratinocyte-specific PHD deficiency may promote vascular survival and growth in a distantly located ischemic tissue by a remote signaling mechanism. We generated mice that carry a keratinocyte-specific Phd2 knockout (kPhd2KO) and performed femoral artery ligation. Relative to wild-type controls, kPhd2KO mice displayed improved vascular survival and arteriogenesis in ischemic hind limbs, leading to the accelerated recovery of hindlimb perfusion and superior muscle regeneration. Similar protective effects were also seen in type 1 and type 2 diabetic mice. Molecularly, both abundance of hypoxia-inducible factor-1α protein and expression of vascular endothelial growth factor-A were increased in epidermal tissues of kPhd2KO mice, accompanied by increased plasma concentration of vascular endothelial growth factor-A. Contrary to kPhd2KO mice, which are PHD2 deficient in all skin tissues, localized kPhd2KO in hindlimb skin tissues did not have similar effects, excluding paracrine signaling as a major mechanism. Confirming the existence of remote effects, hepatocyte-specific Phd2 knockout also protected hind limbs from ischemia injury. These data indicate that vascular survival and growth in ischemia-injured tissue may be stimulated by suppressing PHD2 in a remotely located tissue and may provide highly effective angiogenesis therapies without the need for directly accessing target tissues.
Collapse
Affiliation(s)
- Kotaro Takeda
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Li-Juan Duan
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Hiromi Takeda
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Guo-Hua Fong
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut.
| |
Collapse
|
45
|
Jeltsch M, Leppänen VM, Saharinen P, Alitalo K. Receptor tyrosine kinase-mediated angiogenesis. Cold Spring Harb Perspect Biol 2013; 5:5/9/a009183. [PMID: 24003209 DOI: 10.1101/cshperspect.a009183] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The endothelial cell is the essential cell type forming the inner layer of the vasculature. Two families of receptor tyrosine kinases (RTKs) are almost completely endothelial cell specific: the vascular endothelial growth factor (VEGF) receptors (VEGFR1-3) and the Tie receptors (Tie1 and Tie2). Both are key players governing the generation of blood and lymphatic vessels during embryonic development. Because the growth of new blood and lymphatic vessels (or the lack thereof) is a central element in many diseases, the VEGF and the Tie receptors provide attractive therapeutic targets in various diseases. Indeed, several drugs directed to these RTK signaling pathways are already on the market, whereas many are in clinical trials. Here we review the VEGFR and Tie families, their involvement in developmental and pathological angiogenesis, and the different possibilities for targeting them to either block or enhance angiogenesis and lymphangiogenesis.
Collapse
Affiliation(s)
- Michael Jeltsch
- Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, FIN-00014 Helsinki, Finland
| | | | | | | |
Collapse
|
46
|
Comparison of vascular growth factors in the murine brain reveals placenta growth factor as prime candidate for CNS revascularization. Blood 2013; 122:658-65. [PMID: 23803710 DOI: 10.1182/blood-2012-07-441527] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Vascular bypass procedures in the central nervous system (CNS) remain technically challenging, hindered by complications and often failing to prevent adverse outcome such as stroke. Thus, there is an unmet clinical need for a safe and effective CNS revascularization. Vascular endothelial growth factors (VEGFs) are promising candidates for revascularization; however, their effects appear to be tissue-specific and their potential in the CNS has not been fully explored. To test growth factors for angiogenesis in the CNS, we characterized the effects of endothelium-specific growth factors on the brain vasculature and parenchyma. Recombinant adeno-associated virus (AAV) vectors encoding the growth factors were injected transcranially to the frontoparietal cerebrum of mice. Angiogenesis, mural cell investment, leukocyte recruitment, vascular permeability, reactive gliosis and neuronal patterning were evaluated by 3-dimensional immunofluorescence, electron microscopy, optical projection tomography, and magnetic resonance imaging. Placenta growth factor (PlGF) stimulated robust angiogenesis and arteriogenesis without significant side effects, whereas VEGF and VEGF-C incited growth of aberrant vessels, severe edema, and inflammation. VEGF-B, angiopoietin-1, angiopoietin-2, and a VEGF/angiopoietin-1 chimera had minimal effects on the brain vessels or parenchyma. Of the growth factors tested, PlGF emerged as the most efficient and safe angiogenic factor, hence making it a candidate for therapeutic CNS revascularization.
Collapse
|
47
|
Hamm A, Veschini L, Takeda Y, Costa S, Delamarre E, Squadrito ML, Henze AT, Wenes M, Serneels J, Pucci F, Roncal C, Anisimov A, Alitalo K, De Palma M, Mazzone M. PHD2 regulates arteriogenic macrophages through TIE2 signalling. EMBO Mol Med 2013; 5:843-57. [PMID: 23616286 PMCID: PMC3779447 DOI: 10.1002/emmm.201302695] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 03/09/2013] [Accepted: 03/12/2013] [Indexed: 01/26/2023] Open
Abstract
Occlusion of the main arterial route redirects blood flow to the collateral circulation. We previously reported that macrophages genetically modified to express low levels of prolyl hydroxylase domain protein 2 (PHD2) display an arteriogenic phenotype, which promotes the formation of collateral vessels and protects the skeletal muscle from ischaemic necrosis. However, the molecular mechanisms underlying this process are unknown. Here, we demonstrate that femoral artery occlusion induces a switch in macrophage phenotype through angiopoietin-1 (ANG1)-mediated Phd2 repression. ANG blockade by a soluble trap prevented the downregulation of Phd2 expression in macrophages and their phenotypic switch, thus inhibiting collateral growth. ANG1-dependent Phd2 repression initiated a feed-forward loop mediated by the induction of the ANG receptor TIE2 in macrophages. Gene silencing and cell depletion strategies demonstrate that TIE2 induction in macrophages is required to promote their proarteriogenic functions, enabling collateral vessel formation following arterial obstruction. These results indicate an indispensable role for TIE2 in sustaining in situ programming of macrophages to a proarteriogenic, M2-like phenotype, suggesting possible new venues for the treatment of ischaemic disorders.
Collapse
Affiliation(s)
- Alexander Hamm
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|