1
|
Pruitt L, Abbott RK. Hypoxia-adenosinergic regulation of B cell responses. Front Immunol 2024; 15:1478506. [PMID: 39559353 PMCID: PMC11570280 DOI: 10.3389/fimmu.2024.1478506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/15/2024] [Indexed: 11/20/2024] Open
Abstract
Hypoxic microenvironments induce widespread metabolic changes that have been shown to be critical in regulating innate and adaptive immune responses. Hypoxia-induced changes include the generation of extracellular adenosine followed by subsequent signaling through adenosine receptors on immune cells. This evolutionarily conserved "hypoxia-adenosinergic" pathway of hypoxia → extracellular adenosine → adenosine receptor signaling has been shown to be critical in limiting and redirecting T cell responses including in tumor microenvironments and the gut mucosa. However, the question of whether hypoxic microenvironments are involved in the development of B cell responses has remained unexplored until recently. The discovery that germinal centers (GC), the anatomic site in which B cells undergo secondary diversification and affinity maturation, develop a hypoxic microenvironment has sparked new interest in how this evolutionarily conserved pathway affects antibody responses. In this review we will summarize what is known about hypoxia-adenosinergic microenvironments in lymphocyte development and ongoing immune responses. Specific focus will be placed on new developments regarding the role of the hypoxia-adenosinergic pathway in regulating GC development and humoral immunity.
Collapse
Affiliation(s)
| | - Robert K. Abbott
- Department of Pathology, University of Texas Medical Branch,
Galveston, TX, United States
| |
Collapse
|
2
|
Liu J, Wu F, Li Z, Zheng S, Huang Y, Chen H. Salvianic acid A sodium facilitates cardiac microvascular endothelial cell proliferation by enhancing the hypoxia-inducible factor-1 alpha/vascular endothelial growth factor signalling pathway post-myocardial infarction. Clin Exp Pharmacol Physiol 2024; 51:e13855. [PMID: 38636942 DOI: 10.1111/1440-1681.13855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 02/24/2024] [Accepted: 03/05/2024] [Indexed: 04/20/2024]
Abstract
Cardiac microvascular endothelial cells (CMECs) are important cells surrounding the cardiomyocytes in the heart that maintain microenvironment homeostasis. Salvianic acid A sodium (SAAS) has been reported to prevent myocardial infarction (MI) injury. However, the role of SAAS on CMEC proliferation remains unclear. CEMCs exposed to oxygen glucose deprivation (OGD) were used to explore the angiogenic abilities of SAAS. In vivo, C57BL/6 mice were divided into three groups: sham, MI and SAAS + MI groups. Compared to OGD group, SAAS led to a reduction in the apoptotic rate and an increase of the proliferation in vitro. Additionally, SAAS increased the protein levels of Bcl2, HIF-1α and vascular endothelial growth factor (VEGF) with the reduction of Bax. In terms of the specific mechanisms, SAAS might inhibit HIF-1α ubiquitination and enhance the HIF-1α/VEGF signalling pathway to increase CMEC proliferation. Furthermore, SAAS increased the density of vessels, inhibited myocardial fibrosis and improved cardiac dysfunction in vivo. The present study has revealed that SAAS could potentially be used as an active substance to facilitate CMEC proliferation post-MI.
Collapse
Affiliation(s)
- Jichun Liu
- Department of Cardiology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Fei Wu
- School of Clinical Medicine, Wannan Medical College, Wuhu, China
- Department of Oncology, Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Zhenhan Li
- School of Clinical Medicine, Wannan Medical College, Wuhu, China
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shengwei Zheng
- School of Clinical Medicine, Wannan Medical College, Wuhu, China
| | - Yanqiang Huang
- Research Center for the Prevention and Treatment of Drug Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Baise, China
| | - Hao Chen
- Department of Pathology, Wannan Medical College, Wuhu, China
- Postdoctoral Research Station of Clinical Medicine, Jinan University, Guangzhou, China
- Graduate School, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
3
|
Choy JS, Hubbard T, Golts EM, Bhatt DL, Navia JA, Kassab GS. Pre-arterialization of coronary veins prior to retroperfusion of ischemic myocardium: percutaneous closure device. Front Cardiovasc Med 2023; 10:1208903. [PMID: 37790598 PMCID: PMC10543752 DOI: 10.3389/fcvm.2023.1208903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/31/2023] [Indexed: 10/05/2023] Open
Abstract
Background Chronic coronary retroperfusion to treat myocardial ischemia has previously failed due to edema and hemorrhage of coronary veins suddenly exposed to arterial pressures. The objective of this study was to selectively adapt the coronary veins to become arterialized prior to coronary venous retroperfusion to avoid vascular edema and hemorrhage. Methods and results In 32 animals (Group I = 19 and Group II = 13), the left anterior descending (LAD) artery was occluded using an ameroid occlusion model. In Group I, the great cardiac vein was blocked with suture ligation (Group IA = 11) or with occlusion device (Group IB = 8) to arterialize the venous system within 2 weeks at intermediate pressure (between arterial and venous levels) before a coronary venous bypass graft (CVBG) was implemented through a left internal mammary artery (LIMA) anastomosis. Group II only received the LAD artery occlusion and served as control. Serial echocardiograms showed recovery of left ventricular (LV) function with this adaptation-arterialization approach, with an increase in ejection fraction (EF) in Group I from 38% ± 5% after coronary occlusion to 53% ± 7% eight weeks after CVBG, whereas in Group II the EF never recovered (41% ± 2%-33% ± 7%). The remodeling of the venous system not only allowed restoration of myocardial function when CVBG was implemented but possibly promoted a novel form of "collateralization" between the native arterioles and the newly arterialized venules, which revascularized the ischemic myocardium. Conclusions These findings form a potential rationale for a venous arterialization-revascularization treatment for the refractory angina and the "no-option" patients using a hybrid percutaneous (closure device for arterialization)/surgical approach (CVBG) to revascularize the myocardium.
Collapse
Affiliation(s)
- Jenny S. Choy
- Department of Biomedical Engineering, California Medical Innovations Institute, San Diego, CA, United States
| | | | - Eugene M. Golts
- Division of Cardiovascular and Thoracic Surgery, University of California, San Diego, CA, United States
| | - Deepak L. Bhatt
- Icahn School of Medicine at Mount Sinai Health System, New York, NY, United States
| | - José A. Navia
- Department of Cardiac Surgery, Austral University, Pilar, Buenos Aires, Argentina
| | - Ghassan S. Kassab
- Department of Biomedical Engineering, California Medical Innovations Institute, San Diego, CA, United States
- 3DT Holdings, LLC, San Diego, CA, United States
| |
Collapse
|
4
|
Kar A, Gupta S, Matilal A, Kumar D, Sarkar S. Nanotherapeutics for the Myocardium: A Potential Alternative for Treating Cardiac Diseases. J Cardiovasc Pharmacol 2023; 82:180-188. [PMID: 37341530 DOI: 10.1097/fjc.0000000000001444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 06/03/2023] [Indexed: 06/22/2023]
Abstract
ABSTRACT Cardiovascular diseases (CVDs) are the foremost cause of morbidity and mortality worldwide. Current clinical interventions include invasive approaches for progressed conditions and pharmacological assistance for initial stages, which has systemic side effects. Preventive, curative, diagnostic, and theranostic (therapeutic + diagnostic) approaches till date are not very useful in combating the ongoing CVD epidemic, which demands a promising efficient alternative approach. To combat the growing CVD outbreak globally, the ideal strategy is to make the therapeutic intervention least invasive and direct to the heart to reduce the bystander effects on other organs and increase the bioavailability of the therapeutics to the myocardium. The application of nanoscience and nanoparticle-mediated approaches have gained a lot of momentum because of their efficient passive and active myocardium targeting capability owing to their improved specificity and controlled release. This review provides extensive insight into the various types of nanoparticles available for CVDs, their mechanisms of targeting (eg, direct or indirect), and the utmost need for further development of bench-to-bedside cardiac tissue-based nanomedicines. Furthermore, the review aims to summarize the different ideas and methods of nanoparticle-mediated therapeutic approaches to the myocardium till date with present clinical trials and future perspectives. This review also reflects the potential of such nanoparticle-mediated tissue-targeted therapies to contribute to the sustainable development goals of good health and well-being.
Collapse
Affiliation(s)
- Abhik Kar
- Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | | | | | | | | |
Collapse
|
5
|
Mohan M, Mannan A, Singh TG. Therapeutic implication of Sonic Hedgehog as a potential modulator in ischemic injury. Pharmacol Rep 2023:10.1007/s43440-023-00505-0. [PMID: 37347388 DOI: 10.1007/s43440-023-00505-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/23/2023]
Abstract
Sonic Hedgehog (SHh) is a homology protein that is involved in the modeling and development of embryonic tissues. As SHh plays both protective and harmful roles in ischemia, any disruption in the transduction and regulation of the SHh signaling pathway causes ischemia to worsen. The SHh signal activation occurs when SHh binds to the receptor complex of Ptc-mediated Smoothened (Smo) (Ptc-smo), which initiates the downstream signaling cascade. This article will shed light on how pharmacological modifications to the SHh signaling pathway transduction mechanism alter ischemic conditions via canonical and non-canonical pathways by activating certain downstream signaling cascades with respect to protein kinase pathways, angiogenic cytokines, inflammatory mediators, oxidative parameters, and apoptotic pathways. The canonical pathway includes direct activation of interleukins (ILs), angiogenic cytokines like hepatocyte growth factor (HGF), platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), and hypoxia-inducible factor alpha (HIF-), which modulate ischemia. The non-canonical pathway includes indirect activation of certain pathways like mTOR, PI3K/Akt, MAPK, RhoA/ROCK, Wnt/-catenin, NOTCH, Forkhead box protein (FOXF), Toll-like receptors (TLR), oxidative parameters such as GSH, SOD, and CAT, and some apoptotic parameters such as Bcl2. This review provides comprehensive insights that contribute to our knowledge of how SHh impacts the progression and outcomes of ischemic injuries.
Collapse
Affiliation(s)
- Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
6
|
Jones IC, Dass CR. Roles of pigment epithelium-derived factor in cardiomyocytes: implications for use as a cardioprotective therapeutic. J Pharm Pharmacol 2023:7146108. [PMID: 37104852 DOI: 10.1093/jpp/rgad037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 04/12/2023] [Indexed: 04/29/2023]
Abstract
OBJECTIVES Cardiovascular diseases are the leading cause of death worldwide, with patients having limited options for treatment. Pigment epithelium-derived factor (PEDF) is an endogenous multifunctional protein with several mechanisms of action. Recently, PEDF has emerged as a potential cardioprotective agent in response to myocardial infarction. However, PEDF is also associated with pro-apoptotic effects, complicating its role in cardioprotection. This review summarises and compares knowledge of PEDF's activity in cardiomyocytes with other cell types and draws links between them. Following this, the review offers a novel perspective of PEDF's therapeutic potential and recommends future directions to understand the clinical potential of PEDF better. KEY FINDINGS PEDF's mechanisms as a pro-apoptotic and pro-survival protein are not well understood, despite PEDF's implication in several physiological and pathological activities. However, recent evidence suggests that PEDF may have significant cardioprotective properties mediated by key regulators dependent on cell type and context. CONCLUSIONS While PEDF's cardioprotective activity shares some key regulators with its apoptotic activity, cellular context and molecular features likely allow manipulation of PEDF's cellular activity, highlighting the importance of further investigation into its activities and its potential to be applied as a therapeutic to mitigate damage from a range of cardiac pathologies.
Collapse
Affiliation(s)
- Isobel C Jones
- Curtin Medical School, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| | - Crispin R Dass
- Curtin Medical School, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| |
Collapse
|
7
|
Fang T, Ma C, Zhang Z, Sun L, Zheng N. Roxadustat, a HIF-PHD inhibitor with exploitable potential on diabetes-related complications. Front Pharmacol 2023; 14:1088288. [PMID: 36843948 PMCID: PMC9950780 DOI: 10.3389/fphar.2023.1088288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/26/2023] [Indexed: 02/12/2023] Open
Abstract
Diabetes mellitus (DM) is a group of metabolic diseases caused by absolute or relative deficiency of insulin secretion and characterized by chronic hyperglycemia. Its complications affect almost every tissue of the body, usually leading to blindness, renal failure, amputation, etc. and in the final stage, it mostly develops into cardiac failure, which is the main reason why diabetes mellitus manifests itself as a high clinical lethality. The pathogenesis of diabetes mellitus and its complications involves various pathological processes including excessive production of mitochondrial reactive oxygen species (ROS) and metabolic imbalance. Hypoxia-inducible Factor (HIF) signaling pathway plays an important role in both of the above processes. Roxadustat is an activator of Hypoxia-inducible Factor-1α, which increases the transcriptional activity of Hypoxia-inducible Factor-1α by inhibiting hypoxia-inducible factor prolyl hydroxylase (HIF-PHD). Roxadustat showed regulatory effects on maintaining metabolic stability in the hypoxic state of the body by activating many downstream signaling pathways such as vascular endothelial growth factor (VEGF), glucose transporter protein-1 (GLUT1), lactate dehydrogenase (LDHA), etc. This review summarizes the current research findings of roxadustat on the diseases of cardiomyopathy, nephropathy, retinal damage and impaired wound healing, which also occur at different stages of diabetes and greatly contribute to the damage caused by diabetes to the organism. We attempts to uncover a more comprehensive picture of the therapeutic effects of roxadustat, and inform its expanding research about diabetic complications treatment.
Collapse
Affiliation(s)
- Tingting Fang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China
| | - Congcong Ma
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China
| | - Zhanming Zhang
- Pharmaceutical Sciences, China Medical University-The Queen’s University of Belfast Joint College, Shenyang, Liaoning, China
| | - Luning Sun
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China
| | - Ningning Zheng
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China,*Correspondence: Ningning Zheng,
| |
Collapse
|
8
|
Qiu ZK, Zhang MZ, Zhang WC, Li ZJ, Si LB, Long X, Yu NZ, Wang XJ. Role of HIF-1α in pathogenic mechanisms of keloids. J Cosmet Dermatol 2023; 22:1436-1448. [PMID: 36718786 DOI: 10.1111/jocd.15601] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/08/2022] [Accepted: 12/12/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUDS AND OBJECTIVE Keloids are defined as overrepairing products that develop after skin lesions. Keloids are characterized by the proliferation of fibroblasts and the overaccumulation of extracellular matrix components (mainly collagen), leading to a locally hypoxic microenvironment. Hence, this article was aimed to review hypoxia in pathogenesis of keloids. METHODS We reviewed and summarized the relevant published studies. RESULTS Hypoxia results in the accumulation of hypoxia-inducible factor 1α (HIF-1α) in keloids, contributing to overactivation of the fibrotic signaling pathway, epithelial-mesenchymal transition, and changes in metabolism, eventually leading to aggravated fibrosis, infiltrative growth, and radiotherapy resistance. CONCLUSION It is, therefore, essential to understand the role of HIF-1α in the pathogenic mechanisms of keloids in order to develop new therapeutic approaches.
Collapse
Affiliation(s)
- Zi-Kai Qiu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Zi Zhang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen-Chao Zhang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Jin Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lou-Bin Si
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Long
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan-Ze Yu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Jun Wang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
de Castro AL, Fernandes RO, Ortiz VD, Campos C, Bonetto JHP, Fernandes TRG, Conzatti A, Siqueira R, Tavares AV, Belló-Klein A, Araujo ASDR. Cardioprotective doses of thyroid hormones improve NO bioavailability in erythrocytes and increase HIF-1α expression in the heart of infarcted rats. Arch Physiol Biochem 2022; 128:1516-1523. [PMID: 32551929 DOI: 10.1080/13813455.2020.1779752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
CONTEXT Infarction leads to a decrease in NO bioavailability in the erythrocytes. Thyroid hormones (TH) present positive effects after infarction. However, there are no studies evaluating the effects of cardioprotective doses of TH in the erythrocytes after infarction. OBJECTIVE This study aimed to evaluate the effects of TH in NO bioavailability and oxidative stress parameters in the erythrocytes of infarcted rats. MATERIAL AND METHODS Wistar rats were allocated into the three groups: Sham-operated (SHAM), infarcted (AMI) and infarcted + TH (AMIT). AMIT rats received T4 and T3 for 12 days by gavage. Subsequently, the animals were evaluated by echocardiography and the LV and erythrocytes were collected. RESULTS TH improved NO bioavailability and increased catalase activity in the erythrocytes. Besides that, TH increased HIF-1α in the heart. CONCLUSION TH seems to be positive for erythrocytes preventing a decrease in NO bioavailability and increasing antioxidant enzymatic defense after infarction.
Collapse
Affiliation(s)
- Alexandre Luz de Castro
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rafael Oliveira Fernandes
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Vanessa D Ortiz
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Cristina Campos
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jéssica H P Bonetto
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tânia Regina G Fernandes
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Adriana Conzatti
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rafaela Siqueira
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Angela Vicente Tavares
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Adriane Belló-Klein
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Alex Sander da Rosa Araujo
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
10
|
Sharma V, Manhas A, Gupta S, Dikshit M, Jagavelu K, Verma RS. Fabrication, characterization and in vivo assessment of cardiogel loaded chitosan patch for myocardial regeneration. Int J Biol Macromol 2022; 222:3045-3056. [DOI: 10.1016/j.ijbiomac.2022.10.079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
11
|
O’Connor C, Brady E, Zheng Y, Moore E, Stevens KR. Engineering the multiscale complexity of vascular networks. NATURE REVIEWS. MATERIALS 2022; 7:702-716. [PMID: 35669037 PMCID: PMC9154041 DOI: 10.1038/s41578-022-00447-8] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 05/14/2023]
Abstract
The survival of vertebrate organisms depends on highly regulated delivery of oxygen and nutrients through vascular networks that pervade nearly all tissues in the body. Dysregulation of these vascular networks is implicated in many common human diseases such as hypertension, coronary artery disease, diabetes and cancer. Therefore, engineers have sought to create vascular networks within engineered tissues for applications such as regenerative therapies, human disease modelling and pharmacological testing. Yet engineering vascular networks has historically remained difficult, owing to both incomplete understanding of vascular structure and technical limitations for vascular fabrication. This Review highlights the materials advances that have enabled transformative progress in vascular engineering by ushering in new tools for both visualizing and building vasculature. New methods such as bioprinting, organoids and microfluidic systems are discussed, which have enabled the fabrication of 3D vascular topologies at a cellular scale with lumen perfusion. These approaches to vascular engineering are categorized into technology-driven and nature-driven approaches. Finally, the remaining knowledge gaps, emerging frontiers and opportunities for this field are highlighted, including the steps required to replicate the multiscale complexity of vascular networks found in nature.
Collapse
Affiliation(s)
- Colleen O’Connor
- Department of Bioengineering, University of Washington, Seattle, WA USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA USA
| | - Eileen Brady
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA USA
- Department of Molecular and Cellular Biology, University of Washington, Seattle, WA USA
| | - Ying Zheng
- Department of Bioengineering, University of Washington, Seattle, WA USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA USA
| | - Erika Moore
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL USA
| | - Kelly R. Stevens
- Department of Bioengineering, University of Washington, Seattle, WA USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA USA
- Brotman Baty Institute, Seattle, WA USA
| |
Collapse
|
12
|
Cyclosporine A and Tacrolimus Induce Functional Impairment and Inflammatory Reactions in Endothelial Progenitor Cells. Int J Mol Sci 2021; 22:ijms22189696. [PMID: 34575860 PMCID: PMC8472421 DOI: 10.3390/ijms22189696] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/29/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
Immunosuppressants are a mandatory therapy for transplant patients to avoid rejection of the transplanted organ by the immune system. However, there are several known side effects, including alterations of the vasculature, which involve a higher occurrence of cardiovascular events. While the effects of the commonly applied immunosuppressive drugs cyclosporine A (CsA) and tacrolimus (Tac) on mature endothelial cells have been addressed in several studies, we focused our research on the unexplored effects of CsA and Tac on endothelial colony-forming cells (ECFCs), a subgroup of endothelial progenitor cells, which play an important role in vascular repair and angiogenesis. We hypothesized that CsA and Tac induce functional defects and activate an inflammatory cascade via NF-κB signaling in ECFCs. ECFCs were incubated with different doses (0.01 µM–10 µM) of CsA or Tac. ECFC function was determined using in vitro models. The expression of inflammatory cytokines and adhesion molecules was explored by quantitative real-time PCR and flow cytometry. NF-κB subunit modification was assessed by immunoblot and immunofluorescence. CsA and Tac significantly impaired ECFC function, including proliferation, migration, and tube formation. TNF-α, IL-6, VCAM, and ICAM mRNA expression, as well as PECAM and VCAM surface expression, were enhanced. Furthermore, CsA and Tac led to NF-κB p65 subunit phosphorylation and nuclear translocation. Pharmacological inhibition of NF-κB by parthenolide diminished CsA- and Tac-mediated proinflammatory effects. The data of functional impairment and activation of inflammatory signals provide new insight into mechanisms associated with CsA and Tac and cardiovascular risk in transplant patients.
Collapse
|
13
|
Adini A, Adini I, Grad E, Tal Y, Danenberg HD, Kang PM, Matthews BD, D’Amato RJ. The Prominin-1-Derived Peptide Improves Cardiac Function Following Ischemia. Int J Mol Sci 2021; 22:5169. [PMID: 34068392 PMCID: PMC8153573 DOI: 10.3390/ijms22105169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 12/11/2022] Open
Abstract
Myocardial infarction (MI) remains the leading cause of death in the western world. Despite advancements in interventional revascularization technologies, many patients are not candidates for them due to comorbidities or lack of local resources. Non-invasive approaches to accelerate revascularization within ischemic tissues through angiogenesis by providing Vascular Endothelial Growth Factor (VEGF) in protein or gene form has been effective in animal models but not in humans likely due to its short half-life and systemic toxicity. Here, we tested the hypothesis that PR1P, a small VEGF binding peptide that we developed, which stabilizes and upregulates endogenous VEGF, could be used to improve outcome from MI in rodents. To test this hypothesis, we induced MI in mice and rats via left coronary artery ligation and then treated animals with every other day intraperitoneal PR1P or scrambled peptide for 14 days. Hemodynamic monitoring and echocardiography in mice and echocardiography in rats at 14 days showed PR1P significantly improved multiple functional markers of heart function, including stroke volume and cardiac output. Furthermore, molecular biology and histological analyses of tissue samples showed that systemic PR1P targeted, stabilized and upregulated endogenous VEGF within ischemic myocardium. We conclude that PR1P is a potential non-invasive candidate therapeutic for MI.
Collapse
Affiliation(s)
- Avner Adini
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.D.M.); (R.J.D.)
- Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Irit Adini
- Department of Surgery, Harvard Medical School, The Center for Engineering in Medicine, Mass General Hospital, Shriners Hospitals for Children Boston, Boston, MA 02114, USA;
| | - Etty Grad
- Interventional Cardiology, Heart Institute, Hadassah Hebrew University Medical Center, Jerusalem 91200, Israel; (E.G.); (H.D.D.)
| | - Yuval Tal
- Allergy and Clinical Immunology Unit and Department of Medicine, Hadassah University Medical Center, Jerusalem 91200, Israel;
| | - Haim D. Danenberg
- Interventional Cardiology, Heart Institute, Hadassah Hebrew University Medical Center, Jerusalem 91200, Israel; (E.G.); (H.D.D.)
| | - Peter M. Kang
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA;
| | - Benjamin D. Matthews
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.D.M.); (R.J.D.)
- Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Robert J. D’Amato
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.D.M.); (R.J.D.)
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
14
|
Puri S, Panza G, Mateika JH. A comprehensive review of respiratory, autonomic and cardiovascular responses to intermittent hypoxia in humans. Exp Neurol 2021; 341:113709. [PMID: 33781731 PMCID: PMC8527806 DOI: 10.1016/j.expneurol.2021.113709] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/17/2021] [Accepted: 03/24/2021] [Indexed: 01/08/2023]
Abstract
This review explores forms of respiratory and autonomic plasticity, and associated outcome measures, that are initiated by exposure to intermittent hypoxia. The review focuses primarily on studies that have been completed in humans and primarily explores the impact of mild intermittent hypoxia on outcome measures. Studies that have explored two forms of respiratory plasticity, progressive augmentation of the hypoxic ventilatory response and long-term facilitation of ventilation and upper airway muscle activity, are initially reviewed. The role these forms of plasticity might have in sleep disordered breathing are also explored. Thereafter, the role of intermittent hypoxia in the initiation of autonomic plasticity is reviewed and the role this form of plasticity has in cardiovascular and hemodynamic responses during and following intermittent hypoxia is addressed. The role of these responses in individuals with sleep disordered breathing and spinal cord injury are subsequently addressed. Ultimately an integrated picture of the respiratory, autonomic and cardiovascular responses to intermittent hypoxia is presented. The goal of the integrated picture is to address the types of responses that one might expect in humans exposed to one-time and repeated daily exposure to mild intermittent hypoxia. This form of intermittent hypoxia is highlighted because of its potential therapeutic impact in promoting functional improvement and recovery in several physiological systems.
Collapse
Affiliation(s)
- Shipra Puri
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI 48201, United States of America; Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, United States of America
| | - Gino Panza
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI 48201, United States of America; Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, United States of America
| | - Jason H Mateika
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI 48201, United States of America; Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, United States of America; Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI 48201, United States of America.
| |
Collapse
|
15
|
Implicación de la isoforma antiangiogénica VEGF-A165b en la angiogénesis y la función sistólica tras un infarto de miocardio reperfundido. Rev Esp Cardiol 2021. [DOI: 10.1016/j.recesp.2020.03.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
16
|
Dobbin SJ, Petrie MC, Myles RC, Touyz RM, Lang NN. Cardiotoxic effects of angiogenesis inhibitors. Clin Sci (Lond) 2021; 135:71-100. [PMID: 33404052 PMCID: PMC7812690 DOI: 10.1042/cs20200305] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
The development of new therapies for cancer has led to dramatic improvements in survivorship. Angiogenesis inhibitors represent one such advancement, revolutionising treatment for a wide range of malignancies. However, these drugs are associated with cardiovascular toxicities which can impact optimal cancer treatment in the short-term and may lead to increased morbidity and mortality in the longer term. Vascular endothelial growth factor inhibitors (VEGFIs) are associated with hypertension, left ventricular systolic dysfunction (LVSD) and heart failure as well as arterial and venous thromboembolism, QTc interval prolongation and arrhythmia. The mechanisms behind the development of VEGFI-associated LVSD and heart failure likely involve the combination of a number of myocardial insults. These include direct myocardial effects, as well as secondary toxicity via coronary or peripheral vascular damage. Cardiac toxicity may result from the 'on-target' effects of VEGF inhibition or 'off-target' effects resulting from inhibition of other tyrosine kinases. Similar mechanisms may be involved in the development of VEGFI-associated right ventricular (RV) dysfunction. Some VEGFIs can be associated with QTc interval prolongation and an increased risk of ventricular and atrial arrhythmia. Further pre-clinical and clinical studies and trials are needed to better understand the impact of VEGFI on the cardiovascular system. Once mechanisms are elucidated, therapies can be investigated in clinical trials and surveillance strategies for identifying VEGFI-associated cardiovascular complications can be developed.
Collapse
Affiliation(s)
- Stephen J.H. Dobbin
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Mark C. Petrie
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Rachel C. Myles
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Rhian M. Touyz
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Ninian N. Lang
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| |
Collapse
|
17
|
Inhibition of MLKL Attenuates Necroptotic Cell Death in a Murine Cell Model of Ischaemia Injury. J Clin Med 2021; 10:jcm10020212. [PMID: 33435617 PMCID: PMC7826539 DOI: 10.3390/jcm10020212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/01/2021] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Steatosis in donor livers poses a major risk of organ dysfunction due to their susceptibility to ischaemia-reperfusion (I/R) injury during transplant. Necroptosis, a novel form of programmed cell death, is orchestrated by receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3) and mixed-lineage kinase domain-like pseudokinase (MLKL), has been implicated in I/R injury. Here we investigated the mechanisms of cell death pathways in an in vitro model of hepato-steatotic ischaemia. METHODS Free fatty acid (FFA) treated alpha mouse liver 12 (AML-12) cells were incubated in oxygen-glucose-deprivation (OGD) conditions as seen during ischaemia. RESULTS We found that OGD triggered upregulation of insoluble fraction of RIPK3 and MLKL in FFA + OGD cells compared to FFA control cells. We report that intervention with small interfering (si) MLKL and siRIPK3 significantly attenuated cell death in FFA + OGD cells. Absence of activated CASPASE8 and cleaved-CASPASE3, no change in the expression of CASPASE1 and prostaglandin-endoperoxide synthase 2 (Ptgs2) in FFA + OGD treated cells compared to FFA control cells indicated that apoptosis, pyroptosis and ferroptosis, respectively, are unlikely to be active in this model. CONCLUSION Our findings indicate that RIPK3-MLKL dependent necroptosis contributed to cell death in our in vitro model. Both MLKL and RIPK3 are promising therapeutic targets to inhibit necroptosis during ischaemic injury in fatty liver.
Collapse
|
18
|
Role of antiangiogenic VEGF-A 165b in angiogenesis and systolic function after reperfused myocardial infarction. ACTA ACUST UNITED AC 2020; 74:131-139. [PMID: 32474003 DOI: 10.1016/j.rec.2020.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/30/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION AND OBJECTIVES Angiogenesis helps to reestablish microcirculation after myocardial infarction (MI). In this study, we aimed to further understand the role of the antiangiogenic isoform vascular endothelial growth factor (VEGF)-A165b after MI and to explore its potential as a coadjuvant therapy to coronary reperfusion. METHODS Two mice MI models were formed: a) permanent coronary ligation (nonreperfused MI); b) transient 45-minute coronary occlusion followed by reperfusion (reperfused MI); in both models, animals underwent echocardiography before euthanasia at day 21 after MI induction. We determined serum and myocardial VEGF-A165b levels. In both experimental MI models, we assessed the functional and structural role of VEGF-A165b blockade. In a cohort of 104 ST-segment elevation MI patients, circulating VEGF-A165b levels were correlated with cardiovascular magnetic resonance-derived left ventricular ejection fraction at 6 months and with the occurrence of adverse events (death, heart failure, and/or reinfarction). RESULTS In both models, circulating and myocardial VEGF-A165b levels were increased 21 days after MI induction. Serum VEGF-A165b levels inversely correlated with systolic function evaluated by echocardiography. VEGF-A165b blockade increased capillary density, reduced infarct size, and enhanced left ventricular function in reperfused, but not in nonreperfused, MI experiments. In patients, higher VEGF-A165b levels correlated with depressed ejection fraction and worse outcomes. CONCLUSIONS In experimental and clinical studies, higher serum VEGF-A165b levels are associated with worse systolic function. Their blockade enhances neoangiogenesis, reduces infarct size, and increases ejection fraction in reperfused, but not in nonreperfused, MI experiments. Therefore, VEGF-A165b neutralization represents a potential coadjuvant therapy to coronary reperfusion.
Collapse
|
19
|
Zhou YH, Han QF, Gao L, Sun Y, Tang ZW, Wang M, Wang W, Yao HC. HMGB1 Protects the Heart Against Ischemia-Reperfusion Injury via PI3K/AkT Pathway-Mediated Upregulation of VEGF Expression. Front Physiol 2020; 10:1595. [PMID: 32063860 PMCID: PMC7000523 DOI: 10.3389/fphys.2019.01595] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 12/19/2019] [Indexed: 01/02/2023] Open
Abstract
Delivery of exogenous high mobility group box 1 (HMGB1) may exert a beneficial effect on myocardial ischemia-reperfusion (I/R) injury. Since the expression of vascular endothelial growth factor (VEGF) and phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) in the myocardium mediates the cardioprotective function of basic fibroblast growth factor, we hypothesized that VEGF and the PI3K/Akt signaling pathway also mediate the protective effects of intravenously delivered HMGB1. Thus, the objective of the present study was to analyze the impact of intravenous administration of HMGB1 on the myocardial expression of VEGF, myocardial fibrosis, and cardiac function in rats subjected to acute myocardial I/R. The ischemia was induced by ligation of the left anterior descending coronary artery for 30 min and was followed by 3 h of reperfusion. Myocardial malondialdehyde content, infarct size, and collagen volume fraction decreased, while the activity of superoxide dismutase was increased, the expression of VEGF and p-Akt was upregulated, and cardiac function was improved in the HMGB1-treated group when compared with rats subjected to I/R only (all P < 0.05). However, these effects of HMGB1 were abolished by LY294002. The obtained results demonstrate that the cardioprotective effects of intravenous administration of HMGB1 prior to I/R may be mediated by upregulation of myocardial expression of VEGF, which may activate the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Yan-Hong Zhou
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Shandong First Medical University, Liaocheng, China
| | - Qian-Feng Han
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Shandong First Medical University, Liaocheng, China
| | - Lei Gao
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Shandong University, Liaocheng, China
| | - Ying Sun
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Shandong First Medical University, Liaocheng, China
| | - Zhan-Wei Tang
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Shandong First Medical University, Liaocheng, China
| | - Meng Wang
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Shandong First Medical University, Liaocheng, China.,Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Wei Wang
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Shandong First Medical University, Liaocheng, China
| | - Heng-Chen Yao
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong University and Clinical School of Shandong First Medical University, Liaocheng, China
| |
Collapse
|
20
|
Danastas K, Miller EJ, Hey-Cunningham AJ, Murphy CR, Lindsay LA. Expression of vascular endothelial growth factor A isoforms is dysregulated in women with endometriosis. Reprod Fertil Dev 2019; 30:651-657. [PMID: 29017687 DOI: 10.1071/rd17184] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 09/12/2017] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis is a critical step in the development of ectopic lesions during endometriosis. Although total vascular endothelial growth factor (VEGF) A is elevated in the peritoneal fluid of women with endometriosis, there are contradictory reports on how levels of total endometrial VEGFA are altered in this disease. Furthermore, limited research is available on different VEGFA isoforms in women with endometriosis. Thus, the aim of the present study was to analyse levels of various VEGFA isoforms in women with and without endometriosis at different stages of the menstrual cycle. Quantitative polymerase chain reaction analysis showed that total VEGFA was highest during menstruation in endometriosis compared with controls (P=0.0373). VEGF121 and VEGF189 were similarly highest during menstruation in endometriosis compared with controls (P=0.0165 and 0.0154 respectively). The present study is also the first to identify the natural expression of VEGF111 in human tissue, which is also highest during menstruation in endometriosis (P=0.0464). This discovery of the natural production of VEGF111 in human endometrium, as well as the upregulation of VEGFA isoforms during menstruation in endometriosis, may shed further light on the development and progression of the disease, and improve our understanding of the regulation of endometrial angiogenesis.
Collapse
Affiliation(s)
- Kevin Danastas
- Discipline of Anatomy and Histology, School of Medical Sciences and Bosch Institute, F13 Anderson Stuart Building, The University of Sydney, Sydney, NSW 2006, Australia
| | - Emily J Miller
- Department of Obstetrics, Gynaecology and Neonatology, K25 Medical Foundation Building, The University of Sydney, Sydney, NSW 2006, Australia
| | - Alison J Hey-Cunningham
- Department of Obstetrics, Gynaecology and Neonatology, K25 Medical Foundation Building, The University of Sydney, Sydney, NSW 2006, Australia
| | - Christopher R Murphy
- Discipline of Anatomy and Histology, School of Medical Sciences and Bosch Institute, F13 Anderson Stuart Building, The University of Sydney, Sydney, NSW 2006, Australia
| | - Laura A Lindsay
- Discipline of Anatomy and Histology, School of Medical Sciences and Bosch Institute, F13 Anderson Stuart Building, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
21
|
Prajnamitra RP, Chen HC, Lin CJ, Chen LL, Hsieh PCH. Nanotechnology Approaches in Tackling Cardiovascular Diseases. Molecules 2019; 24:molecules24102017. [PMID: 31137787 PMCID: PMC6572019 DOI: 10.3390/molecules24102017] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/24/2019] [Accepted: 05/25/2019] [Indexed: 01/14/2023] Open
Abstract
Cardiovascular diseases have continued to remain a leading cause of mortality and morbidity worldwide. Poor proliferation capability of adult cardiomyocytes disables the heart from regenerating new myocardium after a myocardial ischaemia event and therefore weakens the heart in the long term, which may result in heart failure and death. Delivery of cardioprotective therapeutics soon after the event can help to protect the heart from further cell death and improve cardiac function, but delivery methods and potential side effects of these therapeutics may be an issue. Advances in nanotechnology, particularly nanoparticles for drug delivery, have enabled researchers to obtain better drug targeting capability, thus increasing the therapeutic outcome. Detailed study of nanoparticles in vivo is useful as it can provide insight for future treatments. Nanogel can help to create a more favourable environment, not only for a sustained delivery of therapeutics, but also for a better navigation of the therapeutics to the targeted sites. Finally, if the damage to the myocardium is too severe for drug treatment, nanopatch can help to improve cardiac function and healing by becoming a platform for pluripotent stem cell-derived cardiomyocytes to grow for the purpose of cell-based regenerative therapy.
Collapse
Affiliation(s)
- Ray Putra Prajnamitra
- Institute of Biomedical Sciences, Academia Sinica, 128 Section 2 Academia Road, Nangang District, Taipei 115, Taiwan.
| | - Hung-Chih Chen
- Institute of Biomedical Sciences, Academia Sinica, 128 Section 2 Academia Road, Nangang District, Taipei 115, Taiwan.
| | - Chen-Ju Lin
- Institute of Biomedical Sciences, Academia Sinica, 128 Section 2 Academia Road, Nangang District, Taipei 115, Taiwan.
| | - Li-Lun Chen
- Institute of Biomedical Sciences, Academia Sinica, 128 Section 2 Academia Road, Nangang District, Taipei 115, Taiwan.
| | - Patrick Ching-Ho Hsieh
- Institute of Biomedical Sciences, Academia Sinica, 128 Section 2 Academia Road, Nangang District, Taipei 115, Taiwan.
| |
Collapse
|
22
|
Abstract
Cardioprotective engineering is an emerging bioengineering discipline aiming to develop engineering strategies to optimize cardioprotective actions against cardiac injuries and disorders. Although there exist innate cardioprotective mechanisms capable of supporting cardiomyocyte survival in response to an insult, not all these mechanisms are optimized in promptness and effectiveness, suggesting the necessity of cardioprotective engineering. Various cardioprotective strategies have been developed and used in experimental and clinical investigations; however, few of these strategies have exerted a significant clinical impact. There are two major challenges in cardioprotective engineering - understanding the innate cardioprotective mechanisms and developing engineering strategies for precise control of the types, levels, timing, and coordination of cardioprotective actions to facilitate recovery from injuries and disorders. Understanding the innate mechanisms is the foundation for developing cardioprotective engineering strategies. Here, ischemic myocardial injury is used as an example to demonstrate the concept of cardioprotective engineering.
Collapse
Affiliation(s)
- Shu Q Liu
- Biomedical Engineering Department, Northwestern University, 2145 Sheridan Road, Evanston IL, 60208-3107
| |
Collapse
|
23
|
Sousa Fialho MDL, Abd Jamil AH, Stannard GA, Heather LC. Hypoxia-inducible factor 1 signalling, metabolism and its therapeutic potential in cardiovascular disease. Biochim Biophys Acta Mol Basis Dis 2019; 1865:831-843. [DOI: 10.1016/j.bbadis.2018.09.024] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/24/2018] [Accepted: 09/18/2018] [Indexed: 12/20/2022]
|
24
|
Park MK, Ko EJ, Jeon KY, Kim H, Jo JO, Baek KW, Kang YJ, Choi YH, Hong Y, Ock MS, Cha HJ. Induction of Angiogenesis by Malarial Infection through Hypoxia Dependent Manner. THE KOREAN JOURNAL OF PARASITOLOGY 2019; 57:117-125. [PMID: 31104403 PMCID: PMC6526210 DOI: 10.3347/kjp.2019.57.2.117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 03/12/2019] [Accepted: 03/14/2019] [Indexed: 11/23/2022]
Abstract
Malarial infection induces tissue hypoxia in the host through destruction of red blood cells. Tissue hypoxia in malarial infection may increase the activity of HIF1α through an intracellular oxygen-sensing pathway. Activation of HIF1α may also induce vascular endothelial growth factor (VEGF) to trigger angiogenesis. To investigate whether malarial infection actually generates hypoxia-induced angiogenesis, we analyzed severity of hypoxia, the expression of hypoxia-related angiogenic factors, and numbers of blood vessels in various tissues infected with Plasmodium berghei. Infection in mice was performed by intraperitoneal injection of 2×106 parasitized red blood cells. After infection, we studied parasitemia and survival. We analyzed hypoxia, numbers of blood vessels, and expression of hypoxia-related angiogenic factors including VEGF and HIF1α. We used Western blot, immunofluorescence, and immunohistochemistry to analyze various tissues from Plasmodium berghei-infected mice. In malaria-infected mice, parasitemia was increased over the duration of infection and directly associated with mortality rate. Expression of VEGF and HIF1α increased with the parasitemia in various tissues. Additionally, numbers of blood vessels significantly increased in each tissue type of the malaria-infected group compared to the uninfected control group. These results suggest that malarial infection in mice activates hypoxia-induced angiogenesis by stimulation of HIF1α and VEGF in various tissues.
Collapse
Affiliation(s)
- Mi-Kyung Park
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea
| | - Eun-Ji Ko
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea
- Department of Biological Science, Pusan National University, Busan 46241, Korea
| | - Kyung-Yoon Jeon
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea
| | - Hyunsu Kim
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea
- Department of Biological Science, Pusan National University, Busan 46241, Korea
| | - Jin-Ok Jo
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea
| | - Kyung-Wan Baek
- Department of Parasitology, College of Medicine, Pusan National University, Busan 50612, Korea
| | - Yun-Jeong Kang
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center and Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 47227, Korea
| | - Yeonchul Hong
- Department of Parasitology and Tropical Medicine, Kyungpook National University School of Medicine, Daegu 41944, Korea
| | - Mee Sun Ock
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea
| |
Collapse
|
25
|
Slater T, Haywood NJ, Matthews C, Cheema H, Wheatcroft SB. Insulin-like growth factor binding proteins and angiogenesis: from cancer to cardiovascular disease. Cytokine Growth Factor Rev 2019; 46:28-35. [PMID: 30954375 DOI: 10.1016/j.cytogfr.2019.03.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/15/2019] [Indexed: 12/22/2022]
Abstract
Angiogenesis is a tightly regulated activity that is vital during embryonic development and for normal physiological repair processes and reproduction in healthy adults. Pathological angiogenesis is a driving force behind a variety of diseases including cancer and retinopathies, and inhibition of angiogenesis is a therapeutic option that has been the subject of much research, with several inhibitory agents now available for medical therapy. Conversely, therapeutic angiogenesis has been mooted as having significant potential in the treatment of ischemic conditions such as angina pectoris and peripheral arterial disease, but so far there has been less translation from lab to bedside. The insulin-like growth factor binding proteins (IGFBP) are a family of seven proteins essential for the binding and transport of the insulin-like growth factors (IGF). It is being increasingly recognised that IGFBPs have a significant role beyond simply modulating IGF activity, with evidence of both IGF dependent and independent actions through a variety of mechanisms. Moreover, the action of the IGFBPs can be stimulatory or inhibitory depending on the cell type and environment. Specifically the IGFBPs have been heavily implicated in angiogenesis, both pathological and physiological, and they have significant promise as targeted cell therapy agents for both pathological angiogenesis inhibition and therapeutic angiogenesis following ischemic injury. In this short review we will explore the current understanding of the individual impact of each IGFBP on angiogenesis, and the pathways through which these effects occur.
Collapse
Affiliation(s)
- Thomas Slater
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Natalie J Haywood
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Connor Matthews
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Harneet Cheema
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom.
| |
Collapse
|
26
|
Kikuchi R, Stevens M, Harada K, Oltean S, Murohara T. Anti-angiogenic isoform of vascular endothelial growth factor-A in cardiovascular and renal disease. Adv Clin Chem 2019; 88:1-33. [PMID: 30612603 DOI: 10.1016/bs.acc.2018.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Accumulating evidence suggests that pathologic interactions between the heart and the kidney can contribute to the progressive dysfunction of both organs. Recently, there has been an increase in the prevalence of cardiovascular disease (CVD) and chronic kidney disease (CKD) due to increasing obesity rates. It has been reported that obesity causes various heart and renal disorders and appears to accelerate their progression. Vascular endothelial growth factor-A (VEGF-A) is a major regulator of angiogenesis and vessel permeability, and is associated with CVD and CKD. It is now recognized that alternative VEGF-A gene splicing generates VEGF-A isoforms that differ in their biological actions. Proximal splicing that includes an exon 8a sequence results in pro-angiogenic VEGF-A165a, whereas distal splicing inclusive of exon 8b yields the anti-angiogenic isoform of VEGF-A (VEGF-A165b). This review highlights several recent preclinical and clinical studies on the role of VEGF-A165b in CVD and CKD as a novel function of VEGF-A. This review also discusses potential therapeutic approaches of the use of VEGF-A in clinical settings as a potential circulating biomarker for CVD and CKD.
Collapse
Affiliation(s)
- Ryosuke Kikuchi
- Department of Medical Technique, Nagoya University Hospital, Nagoya, Japan.
| | - Megan Stevens
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Kazuhiro Harada
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sebastian Oltean
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
27
|
Effects of cardiosphere-derived cell transplantation on cardiac mitochondrial oxygen consumption after myocardial infarction in rats. Biomed Pharmacother 2018; 108:883-892. [DOI: 10.1016/j.biopha.2018.09.117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 12/31/2022] Open
|
28
|
Emerson SR, Sciarrillo CM, Kurti SP, Emerson EM, Rosenkranz SK. High-Fat Meal–Induced Changes in Markers of Inflammation and Angiogenesis in Healthy Adults Who Differ by Age and Physical Activity Level. Curr Dev Nutr 2018. [PMCID: PMC6367518 DOI: 10.1093/cdn/nzy098] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Inflammation and angiogenesis are key facets of cardiovascular disease pathophysiology. Age and physical activity level can influence fasting systemic inflammation, but the impact of these factors on postprandial inflammation is unknown. In addition, markers of angiogenesis have never been tested in the context of a single high-fat meal (HFM). Objective The purpose of this study was to investigate the effects of an HFM on markers of inflammation and angiogenesis in individuals of different ages and physical activity levels. Methods Twenty-two healthy adults—8 younger active (YA) adults (4 men, 4 women; mean ± SD age: 25 ± 5 y), 8 older active (OA) adults (4 men, 4 women; 67 ± 5 y), and 6 older inactive (OI) adults (3 men, 3 women; 68 ± 7 y)—consumed an HFM [63% fat (39% saturated fat, 14% monounsaturated fat, 10% polyunsaturated fat), 34% carbohydrate; 12 kcal/kg body mass; 927 ± 154 kcal]. Fourteen inflammatory and 9 angiogenic markers were measured at baseline and 3 and 6 h postmeal. Results Significant group effects were observed in interleukin (IL)-10 (YA > OA; P = 0.02), IL-23 (YA > OA; P = 0.02), tumor necrosis factor (TNF)-α (OA < OI; P = 0.04), and vascular endothelial growth factor (VEGF)-C (YA < OA; P = 0.001). IL-8, VEGF-A, VEGF-C, and heparin-binding epidermal growth factor–like growth factor significantly increased, whereas granulocyte-macrophage colony-stimulating factor, interferon-γ, IL-1β, IL-5, IL-10, IL-12, IL-13, IL-17A, IL-23, TNF-α, leptin, angiopoietin-2, and follistatin significantly decreased after HFM consumption (P’s < 0.05). Notably, VEGF-A and VEGF-C were significantly higher at 3 h [mean difference: 22.5 pg/mL (VEGF-A); 73.5 pg/mL (VEGF-C)] and 6 h postmeal [mean difference: 26.9 pg/mL (VEGF-A); 81.2 pg/mL (VEGF-C)]. Conclusions A novel finding of this study was the robust increase in VEGF after an HFM. There were also group differences in several inflammatory markers (IL-10 and IL-23 greater in YA than OA, and TNF-α lower in OA than OI) that suggest a potential influence of age and physical activity level.
Collapse
Affiliation(s)
- Sam R Emerson
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK
- Physical Activity and Nutrition Clinical Research Consortium (PAN-CRC), College of Human Ecology
- Department of Food, Nutrition, Dietetics, and Health
| | | | - Stephanie P Kurti
- Physical Activity and Nutrition Clinical Research Consortium (PAN-CRC), College of Human Ecology
- Department of Kinesiology, Kansas State University, Manhattan, KS
- Department of Kinesiology, James Madison University, Harrisonburg, VA
| | - Emily M Emerson
- Physical Activity and Nutrition Clinical Research Consortium (PAN-CRC), College of Human Ecology
- Department of Food, Nutrition, Dietetics, and Health
| | - Sara K Rosenkranz
- Physical Activity and Nutrition Clinical Research Consortium (PAN-CRC), College of Human Ecology
- Department of Food, Nutrition, Dietetics, and Health
| |
Collapse
|
29
|
Marks ECA, Wilkinson TM, Frampton CM, Skelton L, Pilbrow AP, Yandle TG, Pemberton CJ, Doughty RN, Whalley GA, Ellis CJ, Troughton RW, Owen MC, Pattinson NR, Cameron VA, Richards AM, Gieseg SP, Palmer BR. Plasma levels of soluble VEGF receptor isoforms, circulating pterins and VEGF system SNPs as prognostic biomarkers in patients with acute coronary syndromes. BMC Cardiovasc Disord 2018; 18:169. [PMID: 30111293 PMCID: PMC6094571 DOI: 10.1186/s12872-018-0894-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 07/23/2018] [Indexed: 12/17/2022] Open
Abstract
Background Development of collateral circulation in coronary artery disease is cardio-protective. A key process in forming new blood vessels is attraction to occluded arteries of monocytes with their subsequent activation as macrophages. In patients from a prospectively recruited post-acute coronary syndromes cohort we investigated the prognostic performance of three products of activated macrophages, soluble vascular endothelial growth factor (VEGF) receptors (sFlt-1 and sKDR) and pterins, alongside genetic variants in VEGF receptor genes, VEGFR-1 and VEGFR-2. Methods Baseline levels of sFlt-1 (VEGFR1), sKDR (VEGFR2) and pterins were measured in plasma samples from subgroups (n = 513; 211; 144, respectively) of the Coronary Disease Cohort Study (CDCS, n = 2067). DNA samples from the cohort were genotyped for polymorphisms from the VEGFR-1 gene SNPs (rs748252 n = 2027, rs9513070 n = 2048) and VEGFR-2 gene SNPs (rs2071559 n = 2050, rs2305948 n = 2066, rs1870377 n = 2042). Results At baseline, levels of sFlt-1 were significantly correlated with age, alcohol consumption, NTproBNP, BNP and other covariates relevant to cardiovascular pathophysiology. Total neopterin levels were associated with alcohol consumption at baseline. 7,8 dihydroneopterin was associated with BMI. The A allele of VEGFR-2 variant rs1870377 was associated with higher plasma sFlt-1 and lower levels of sKDR at baseline. Baseline plasma sFlt-1 was univariately associated with all cause mortality with (p < 0.001) and in a Cox’s proportional hazards regression model sFlt-1 and pterins were both associated with mortality independent of established predictors (p < 0.027). Conclusions sFlt-1 and pterins may have potential as prognostic biomarkers in acute coronary syndromes patients. Genetic markers from VEGF system genes warrant further investigation as markers of levels of VEGF system components in these patients. Trial registration Australian New Zealand Clinical Trials Registry. ACTRN12605000431628. 16 September 2005, Retrospectively registered. Electronic supplementary material The online version of this article (10.1186/s12872-018-0894-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Edward C A Marks
- Christchurch Heart institute, Department of Medicine, University of Otago, PO Box 4345, Christchurch, New Zealand.,School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Tom M Wilkinson
- Christchurch Heart institute, Department of Medicine, University of Otago, PO Box 4345, Christchurch, New Zealand
| | - Chris M Frampton
- Christchurch Heart institute, Department of Medicine, University of Otago, PO Box 4345, Christchurch, New Zealand
| | - Lorraine Skelton
- Christchurch Heart institute, Department of Medicine, University of Otago, PO Box 4345, Christchurch, New Zealand
| | - Anna P Pilbrow
- Christchurch Heart institute, Department of Medicine, University of Otago, PO Box 4345, Christchurch, New Zealand
| | - Tim G Yandle
- Christchurch Heart institute, Department of Medicine, University of Otago, PO Box 4345, Christchurch, New Zealand
| | - Chris J Pemberton
- Christchurch Heart institute, Department of Medicine, University of Otago, PO Box 4345, Christchurch, New Zealand
| | - Robert N Doughty
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Gillian A Whalley
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand.,Department of Medicine, Dunedin School of Medicine, University of Otago, Auckland, New Zealand
| | - Chris J Ellis
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard W Troughton
- Christchurch Heart institute, Department of Medicine, University of Otago, PO Box 4345, Christchurch, New Zealand
| | - Maurice C Owen
- Canterbury Scientific Ltd, 71 Whiteleigh Ave, Christchurch, New Zealand
| | - Neil R Pattinson
- Canterbury Scientific Ltd, 71 Whiteleigh Ave, Christchurch, New Zealand
| | - Vicky A Cameron
- Christchurch Heart institute, Department of Medicine, University of Otago, PO Box 4345, Christchurch, New Zealand
| | - A Mark Richards
- Christchurch Heart institute, Department of Medicine, University of Otago, PO Box 4345, Christchurch, New Zealand.,Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - Steven P Gieseg
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Barry R Palmer
- Christchurch Heart institute, Department of Medicine, University of Otago, PO Box 4345, Christchurch, New Zealand. .,School of Health Sciences, College of Health, Massey University Wellington, Wellington, New Zealand.
| |
Collapse
|
30
|
Shi C, Zhao Y, Yang Y, Chen C, Hou X, Shao J, Yao H, Li Q, Xia Y, Dai J. Collagen-binding VEGF targeting the cardiac extracellular matrix promotes recovery in porcine chronic myocardial infarction. Biomater Sci 2018; 6:356-363. [PMID: 29266144 DOI: 10.1039/c7bm00891k] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
An effective therapy for chronic myocardial infarction (MI) has yet to be developed. Vascular endothelial growth factor (VEGF) promotes angiogenesis and improves cardiac function after MI. However, non-targeted delivery of VEGF decreases its therapeutic efficacy. In this study, for targeting the cardiac extracellular matrix, a collagen-binding domain (CBD) VEGF was used to bind specifically to the collagen-rich cardiac extracellular matrix. When intramyocardially injected into the peri-infarct region of a chronically infarcted porcine heart, CBD-VEGF attenuated the remodeling of the left ventricle with a decreased infarct size and promoted cardiomyocyte survival and angiogenesis 3 months after injection. In the 12-month trial, mature vessel networks and myocardium-like tissues were observed in the infarct region after CBD-VEGF injection. Also these beneficial effects might derive from CBD-VEGF significantly protecting cardiomyocytes from apoptosis and recruiting cardiac progenitor cells to the infarcted region. These results demonstrated that CBD-VEGF could be a promising therapeutic strategy for chronic MI.
Collapse
Affiliation(s)
- Chunying Shi
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao, 266021, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Puddighinu G, D'Amario D, Foglio E, Manchi M, Siracusano A, Pontemezzo E, Cordella M, Facchiano F, Pellegrini L, Mangoni A, Tafani M, Crea F, Germani A, Russo MA, Limana F. Molecular mechanisms of cardioprotective effects mediated by transplanted cardiac ckit + cells through the activation of an inflammatory hypoxia-dependent reparative response. Oncotarget 2017; 9:937-957. [PMID: 29416668 PMCID: PMC5787525 DOI: 10.18632/oncotarget.22946] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 11/12/2017] [Indexed: 12/16/2022] Open
Abstract
The regenerative effects of cardiac ckit+ stem cells (ckit+CSCs) in acute myocardial infarction (MI) have been studied extensively, but how these cells exert a protective effect on cardiomyocytes is not well known. Growing evidences suggest that in adult stem cells injury triggers inflammatory signaling pathways which control tissue repair and regeneration. Aim of the present study was to determine the mechanisms underlying the cardioprotective effects of ckit+CSCs following transplantation in a murine model of MI. Following isolation and in vitro expansion, cardiac ckit+CSCs were subjected to normoxic and hypoxic conditions and assessed at different time points. These cells adapted to hypoxia as showed by the activation of HIF-1α and the expression of a number of genes, such as VEGF, GLUT1, EPO, HKII and, importantly, of alarmin receptors, such as RAGE, P2X7R, TLR2 and TLR4. Activation of these receptors determined an NFkB-dependent inflammatory and reparative gene response (IRR). Importantly, hypoxic ckit+CSCs increased the secretion of the survival growth factors IGF-1 and HGF. To verify whether activation of the IRR in a hypoxic microenvironment could exert a beneficial effect in vivo, autologous ckit+CSCs were transplanted into mouse heart following MI. Interestingly, transplantation of ckit+CSCs lowered apoptotic rates and induced autophagy in the peri-infarct area; further, it reduced hypertrophy and fibrosis and, most importantly, improved cardiac function. ckit+CSCs are able to adapt to a hypoxic environment and activate an inflammatory and reparative response that could account, at least in part, for a protective effect on stressed cardiomyocytes following transplantation in the infarcted heart.
Collapse
Affiliation(s)
- Giovanni Puddighinu
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Domenico D'Amario
- Department of Cardiovascular Sciences, Catholic University of The Sacred Heart, Rome, Italy
| | - Eleonora Foglio
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Melissa Manchi
- Department of Cardiovascular Sciences, Catholic University of The Sacred Heart, Rome, Italy
| | - Andrea Siracusano
- Department of Cardiovascular Sciences, Catholic University of The Sacred Heart, Rome, Italy
| | - Elena Pontemezzo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Martina Cordella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Pellegrini
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico (CCP), Milan, Italy
| | - Antonella Mangoni
- Department of Pathological Anatomy, Catholic University of The Sacred Heart, Rome, Italy
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Filippo Crea
- Department of Cardiovascular Sciences, Catholic University of The Sacred Heart, Rome, Italy
| | - Antonia Germani
- Laboratory of Vascular Pathology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Fondazione Luigi Maria Monti, Rome, Italy
| | - Matteo Antonio Russo
- IRCCS San Raffaele Pisana, Rome, Italy.,MEBIC Consortium, San Raffaele Roma Open University, Rome, Italy
| | - Federica Limana
- IRCCS San Raffaele Pisana, Rome, Italy.,San Raffaele Roma Open University, Rome, Italy
| |
Collapse
|
32
|
Schaefer JA, Guzman PA, Riemenschneider SB, Kamp TJ, Tranquillo RT. A cardiac patch from aligned microvessel and cardiomyocyte patches. J Tissue Eng Regen Med 2017; 12:546-556. [PMID: 28875579 DOI: 10.1002/term.2568] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 08/28/2017] [Accepted: 09/01/2017] [Indexed: 12/18/2022]
Abstract
Cardiac tissue engineering aims to produce replacement tissue patches in the lab to replace or treat infarcted myocardium. However, current patches lack preformed microvascularization and are therefore limited in thickness and force production. In this study, we sought to assess whether a bilayer patch composed of a layer made from human induced pluripotent stem cell-derived cardiomyocytes and a microvessel layer composed of self-assembled human blood outgrowth endothelial cells and pericytes was capable of engrafting on the epicardial surface of a nude rat infarct model and becoming perfused by the host 4 weeks after acute implantation. The bilayer configuration was found to increase the twitch force production, improve human induced pluripotent stem cell-derived cardiomyocyte survival and maturation, and increase patent microvessel lumens compared with time-matched single layer controls after 2 weeks of in vitro culture. Upon implantation, the patch microvessels sprouted into the cardiomyocyte layer of the patch and inosculated with the host vasculature as evidenced by species-specific perfusion labels and erythrocyte staining. Our results demonstrate that the added microvessel layer of a bilayer patch substantially improves in vitro functionality and that the bilayer patch is capable of engraftment with rapid microvessel inosculation on injured myocardium. The bilayer format will allow for scaling up in size through the addition of layers to obtain thicker tissues generating greater force in the future.
Collapse
Affiliation(s)
- Jeremy A Schaefer
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Pilar A Guzman
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Sonja B Riemenschneider
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA
| | - Timothy J Kamp
- Department of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Robert T Tranquillo
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA.,Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
33
|
Sruthi TV, Edatt L, Raji GR, Kunhiraman H, Shankar SS, Shankar V, Ramachandran V, Poyyakkara A, Kumar SVB. Horizontal transfer of miR-23a from hypoxic tumor cell colonies can induce angiogenesis. J Cell Physiol 2017; 233:3498-3514. [PMID: 28929578 DOI: 10.1002/jcp.26202] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 09/14/2017] [Indexed: 12/16/2022]
Abstract
Neo vessel formation by angiogenesis is an important event during many pathological conditions including cancer, where it is indispensable for tumor growth and survival. Although, various pro-angiogenic cytokines and soluble factors, secreted by tumor cells, have been reported to promote angiogenesis, recent studies have shown regulatory role of exosomes, secreted by tumor cells in the process of angiogenesis. These exosomes are capable of carrying nucleic acids, proteins, etc., as their cargo. Under the light of these facts and considering the presence of miRNAs, the non-coding RNAs capable of regulating target gene expression, as one of the major cargos in the exosomes, we investigated, whether exosomes derived from normoxic and hypoxic tumor cell colonies exhibit difference in levels of miR-23∼27∼24 cluster members and if so, to check the significance of their horizontal transfer on the process of angiogenesis. Results of our study showed that exosomes secreted by hypoxic tumor cell colonies possess significantly higher levels of miR23a and can induce angiogenesis. Further, we have shown that exosomes secreted by cells that ectopically over express miR23a is capable of inducing angiogenesis in different angiogenic model systems such as CAM, in ovo Xenograft and HUVEC models systems. Further, mechanistic analysis revealed that miR23a driven regulation of angiogenesis is brought about by down regulation of SIRT1 in the recipient cells. Collectively, the results presented here suggest that exosomal transfer of miR23a from tumor cell colonies can induce the process of angiogenesis by targeting SIRT1 in the recipient endothelial cells.
Collapse
Affiliation(s)
- T V Sruthi
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Nileshwar, Kasargod, Kerala, India
| | - Lincy Edatt
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Nileshwar, Kasargod, Kerala, India
| | - Grace R Raji
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Nileshwar, Kasargod, Kerala, India
| | - Haritha Kunhiraman
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Nileshwar, Kasargod, Kerala, India
| | - Sharath S Shankar
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Nileshwar, Kasargod, Kerala, India.,Chemical Sciences and Technology Division, National Institute for Interdisciplinary Science and Technology, CSIR, Trivandrum, Kerala, India
| | - Vandana Shankar
- Agroprocessing and Technology Division, National Institute for Interdisciplinary Science and Technology, CSIR, Trivandrum, Kerala, India
| | - Vishnu Ramachandran
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Nileshwar, Kasargod, Kerala, India
| | - Aswini Poyyakkara
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Nileshwar, Kasargod, Kerala, India
| | - Sameer V B Kumar
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Nileshwar, Kasargod, Kerala, India
| |
Collapse
|
34
|
Casieri V, Matteucci M, Cavallini C, Torti M, Torelli M, Lionetti V. Long-term Intake of Pasta Containing Barley (1-3)Beta-D-Glucan Increases Neovascularization-mediated Cardioprotection through Endothelial Upregulation of Vascular Endothelial Growth Factor and Parkin. Sci Rep 2017; 7:13424. [PMID: 29044182 PMCID: PMC5647408 DOI: 10.1038/s41598-017-13949-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/03/2017] [Indexed: 01/17/2023] Open
Abstract
Barley (1-3)β-D-Glucan (BBG) enhances angiogenesis. Since pasta is very effective in providing a BBG-enriched diet, we hypothesized that the intake of pasta containing 3% BBG (P-BBG) induces neovascularization-mediated cardioprotection. Healthy adult male C57BL/6 mice fed P-BBG (n = 15) or wheat pasta (Control, n = 15) for five-weeks showed normal glucose tolerance and cardiac function. With a food intake similar to the Control, P-BBG mice showed a 109% survival rate (P < 0.01 vs. Control) after cardiac ischemia (30 min)/reperfusion (60 min) injury. Left ventricular (LV) anion superoxide production and infarct size in P-BBG mice were reduced by 62 and 35% (P < 0.0001 vs. Control), respectively. The capillary and arteriolar density of P-BBG hearts were respectively increased by 12 and 18% (P < 0.05 vs. Control). Compared to the Control group, the VEGF expression in P-BBG hearts was increased by 87.7% (P < 0.05); while, the p53 and Parkin expression was significantly increased by 125% and cleaved caspase-3 levels were reduced by 33% in P-BBG mice. In vitro, BBG was required to induce VEGF, p53 and Parkin expression in human umbelical vascular endothelial cells. Moreover, the BBG-induced Parkin expression was not affected by pifithrin-α (10 uM/7days), a p53 inhibitor. In conclusion, long-term dietary supplementation with P-BBG confers post-ischemic cardioprotection through endothelial upregulation of VEGF and Parkin.
Collapse
Affiliation(s)
| | - Marco Matteucci
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Claudia Cavallini
- ATTRE (Advanced Therapies and Tissue Regeneration) Laboratory, Innovation Accelerator CNR, Bologna, Italy
| | - Milena Torti
- Research and Development Unit, Pastificio Attilio Matromauro Granoro s.r.l, Corato, Italy
| | - Michele Torelli
- Research and Development Unit, Pastificio Attilio Matromauro Granoro s.r.l, Corato, Italy
| | - Vincenzo Lionetti
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy. .,UOS Anesthesia and Intensive Care, Fondazione Toscana "G. Monasterio", Pisa, Italy.
| |
Collapse
|
35
|
Tao B, Wang R, Sun C, Zhu Y. 3-Mercaptopyruvate Sulfurtransferase, Not Cystathionine β-Synthase Nor Cystathionine γ-Lyase, Mediates Hypoxia-Induced Migration of Vascular Endothelial Cells. Front Pharmacol 2017; 8:657. [PMID: 28979207 PMCID: PMC5611563 DOI: 10.3389/fphar.2017.00657] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 09/04/2017] [Indexed: 01/06/2023] Open
Abstract
Hypoxia-induced angiogenesis is a common phenomenon in many physiological and patho-physiological processes. However, the potential differential roles of three hydrogen sulfide producing systems cystathionine γ-lyase (CSE)/H2S, cystathionine β-synthase (CBS)/H2S, and 3-mercaptopyruvate sulfurtransferase (MPST)/H2S in hypoxia-induced angiogenesis are still unknown. We found that minor hypoxia (10% oxygen) significantly increased the migration of vascular endothelial cells while hypoxia (8% oxygen) significantly inhibited cell migration. The present study was performed using cells cultured in 10% oxygen. RNA interference was used to block the endogenous generation of hydrogen sulfide by CSE, CBS, or MPST in a vascular endothelial cell migration model in both normoxia and hypoxia. The results showed that CBS had a promoting effect on the migration of vascular endothelial cells cultured in both normoxic and hypoxic conditions. In contrast, CSE had an inhibitory effect on cell migration. MPST had a promoting effect on the migration of vascular endothelial cells cultured in hypoxia; however, it had no effect on the cells cultured in normoxia. Importantly, it was found that the hypoxia-induced increase in vascular endothelial cell migration was mediated by MPST, but not CSE or CBS. The western blot analyses showed that hypoxia significantly increased MPST protein levels, decreased CSE protein levels and did not change CBS levels, suggesting that these three hydrogen sulfide-producing systems respond differently to hypoxic conditions. Interestingly, MPST protein levels were elevated by hypoxia in a bi-phasic manner and MPST mRNA levels increased later than the first stage elevation of the protein levels, implying that the expression of MPST induced by hypoxia was also regulated at a post-transcriptional level. RNA pull-down assay showed that some candidate RNA binding proteins, such as nucleolin and Annexin A2, were dissociated from the 3'-UTR of MPST mRNA in hypoxia which implied their involvement in MPST mRNA regulation.
Collapse
Affiliation(s)
- Beibei Tao
- Shanghai Key Laboratory of Bioactive Small Molecules and Research Center on Aging and Medicine, Department of Physiology and Pathophysiology, Fudan University Shanghai Medical CollegeShanghai, China
| | - Rui Wang
- Shanghai Key Laboratory of Bioactive Small Molecules and Research Center on Aging and Medicine, Department of Physiology and Pathophysiology, Fudan University Shanghai Medical CollegeShanghai, China
| | - Chen Sun
- Shanghai Key Laboratory of Bioactive Small Molecules and Research Center on Aging and Medicine, Department of Physiology and Pathophysiology, Fudan University Shanghai Medical CollegeShanghai, China
| | - Yichun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Research Center on Aging and Medicine, Department of Physiology and Pathophysiology, Fudan University Shanghai Medical CollegeShanghai, China
| |
Collapse
|
36
|
Kim J, Mirando AC, Popel AS, Green JJ. Gene delivery nanoparticles to modulate angiogenesis. Adv Drug Deliv Rev 2017; 119:20-43. [PMID: 27913120 PMCID: PMC5449271 DOI: 10.1016/j.addr.2016.11.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 10/01/2016] [Accepted: 11/24/2016] [Indexed: 01/19/2023]
Abstract
Angiogenesis is naturally balanced by many pro- and anti-angiogenic factors while an imbalance of these factors leads to aberrant angiogenesis, which is closely associated with many diseases. Gene therapy has become a promising strategy for the treatment of such a disordered state through the introduction of exogenous nucleic acids that express or silence the target agents, thereby engineering neovascularization in both directions. Numerous non-viral gene delivery nanoparticles have been investigated towards this goal, but their clinical translation has been hampered by issues associated with safety, delivery efficiency, and therapeutic effect. This review summarizes key factors targeted for therapeutic angiogenesis and anti-angiogenesis gene therapy, non-viral nanoparticle-mediated approaches to gene delivery, and recent gene therapy applications in pre-clinical and clinical trials for ischemia, tissue regeneration, cancer, and wet age-related macular degeneration. Enhanced nanoparticle design strategies are also proposed to further improve the efficacy of gene delivery nanoparticles to modulate angiogenesis.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Adam C Mirando
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Departments of Ophthalmology, Neurosurgery, and Materials Science & Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
37
|
Dynamics and implications of circulating anti-angiogenic VEGF-A 165b isoform in patients with ST-elevation myocardial infarction. Sci Rep 2017; 7:9962. [PMID: 28855597 PMCID: PMC5577291 DOI: 10.1038/s41598-017-10505-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/09/2017] [Indexed: 12/22/2022] Open
Abstract
Angiogenesis is crucial to restore microvascular perfusion in the jeopardized myocardium in the weeks following reperfused ST-segment elevation myocardial infarction (STEMI). (VEGF)-A165b, an anti-angiogenic factor, has been identified as a regulator of vascularization; however, it has not been previously implicated in acute myocardial infarction. We sought to investigate the dynamics of circulating VEGF-A165b and its association with cardiac magnetic resonance-derived infarct size and left ventricular ejection fraction (LVEF). 50 STEMI patients and 23 controls were included. Compared with control individuals, serum VEGF-A165b was elevated in STEMI patients prior to primary percutaneous coronary intervention (PCI). Following PCI, serum VEGF-A165b increased further, reaching a maximum level at 24 h and decreased one month after reperfusion. VEGF-A165b levels at 24 h were associated with a large infarct size and inversely related to LVEF. VEGF-A165b expression was increased in myocardial infarct areas from patients with previous history of AMI. An ex vivo assay using serum from STEMI patients showed that neutralization of VEGF-A165b increased tubulogenesis. Overall, the study suggests that VEGF-A165b might play a deleterious role after AMI as an inhibitor of angiogenesis in the myocardium. Accordingly, neutralization of VEGF-A165b could represent a novel pro-angiogenic therapy for reperfusion of myocardium in STEMI.
Collapse
|
38
|
Herrfurth L, Theis V, Matschke V, May C, Marcus K, Theiss C. Morphological Plasticity of Emerging Purkinje Cells in Response to Exogenous VEGF. Front Mol Neurosci 2017; 10:2. [PMID: 28194096 PMCID: PMC5276996 DOI: 10.3389/fnmol.2017.00002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 01/04/2017] [Indexed: 12/12/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is well known as the growth factor with wide-ranging functions even in the central nervous system (CNS). Presently, most attention is given to the investigation of its role in neuronal protection, growth and maturation processes, whereby most effects are mediated through VEGF receptor 2 (VEGFR-2). The purpose of our current study is to provide new insights into the impact of VEGF on immature and mature Purkinje cells (PCs) in accordance with maturity and related receptor expression. Therefore, to expand our knowledge of VEGF effects in PCs development and associated VEGFR-2 expression, we used cultivated organotypic cerebellar slice cultures in immunohistochemical or microinjection studies, followed by confocal laser scanning microscopy (CLSM) and morphometric analysis. Additionally, we incorporated in our study the method of laser microdissection, followed by quantitative polymerase chain reaction (qPCR). For the first time we could show the age-dependent VEGF sensitivity of PCs with the largest promoting effects being on dendritic length and cell soma size in neonatal and juvenile stages. Once mature, PCs were no longer susceptible to VEGF stimulation. Analysis of VEGFR-2 expression revealed its presence in PCs throughout development, which underlined its mediating functions in neuronal cells.
Collapse
Affiliation(s)
- Leonard Herrfurth
- Medizinische Fakultät, Institut für Anatomie, Abteilung für Cytologie, Ruhr-Universität Bochum Bochum, Germany
| | - Verena Theis
- Medizinische Fakultät, Institut für Anatomie, Abteilung für Cytologie, Ruhr-Universität Bochum Bochum, Germany
| | - Veronika Matschke
- Medizinische Fakultät, Institut für Anatomie, Abteilung für Cytologie, Ruhr-Universität Bochum Bochum, Germany
| | - Caroline May
- Abteilung für Medizinische Proteomik/Bioanalytik, Medizinisches Proteom-Center, Ruhr-University Bochum Bochum, Germany
| | - Katrin Marcus
- Abteilung für Medizinische Proteomik/Bioanalytik, Medizinisches Proteom-Center, Ruhr-University Bochum Bochum, Germany
| | - Carsten Theiss
- Medizinische Fakultät, Institut für Anatomie, Abteilung für Cytologie, Ruhr-Universität Bochum Bochum, Germany
| |
Collapse
|
39
|
Tao YK, Zeng H, Zhang GQ, Chen ST, Xie XJ, He X, Wang S, Wen H, Chen JX. Notch3 deficiency impairs coronary microvascular maturation and reduces cardiac recovery after myocardial ischemia. Int J Cardiol 2017; 236:413-422. [PMID: 28131704 DOI: 10.1016/j.ijcard.2017.01.096] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/13/2017] [Indexed: 12/29/2022]
Abstract
RATIONALE Vascular maturation plays an important role in wound repair post-myocardial infarction (MI). The Notch3 is critical for pericyte recruitment and vascular maturation during embryonic development. OBJECTIVE This study is to test whether Notch3 deficiency impairs vascular maturation and blunts cardiac functional recovery post-MI. APPROACH AND RESULTS Wild type (WT) and Notch3 knockout (Notch3KO) mice were subjected to MI by the ligation of left anterior descending coronary artery (LAD). Cardiac function and coronary blood flow reserve (CFR) were measured by echocardiography. The expression of angiogenic growth factor, pericyte/capillary coverage and arteriolar formation were analyzed. Loss of Notch3 in mice resulted in a significant reduction of pericytes and small arterioles. Notch3 KO mice had impaired pericyte/capillary coverage and CFR compared to WT mice. Notch3 KO mice were more prone to ischemic injury with larger infarcted size and higher rates of mortality. The expression of CXCR-4 and VEGF/Ang-1 was significantly decreased in Notch3 KO mice. Notch3 KO mice also had few NG2+/Sca1+ and NG2+/c-kit+ progenitor cells in the ischemic area and exhibited worse cardiac function recovery at 2weeks after MI. These were accompanied by a significant reduction of pericyte/capillary coverage and arteriolar maturation. Furthermore, Notch3 KO mice subjected to MI had increased intracellular adhesion molecule-2 (ICAM-2) expression and CD11b+ macrophage infiltration into ischemic areas compared to that of WT mice. CONCLUSION Notch3 mutation impairs recovery of cardiac function post-MI by the mechanisms involving the pre-existing coronary microvascular dysfunction conditions, and impairment of pericyte/progenitor cell recruitment and microvascular maturation.
Collapse
Affiliation(s)
- Yong-Kang Tao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; Emergency Department of China-Japan Friendship Hospital, Beijing 100029, China
| | - Heng Zeng
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Guo-Qiang Zhang
- Emergency Department of China-Japan Friendship Hospital, Beijing 100029, China.
| | - Sean T Chen
- Duke University School of Medicine, Durham, USA
| | - Xue-Jiao Xie
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Xiaochen He
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Shuo Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Hongyan Wen
- Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| |
Collapse
|
40
|
Bonaventura A, Montecucco F, Dallegri F. Update on the effects of treatment with recombinant tissue-type plasminogen activator (rt-PA) in acute ischemic stroke. Expert Opin Biol Ther 2016; 16:1323-1340. [PMID: 27548625 DOI: 10.1080/14712598.2016.1227779] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Acute ischemic stroke (AIS) represents a major cause of death and disability all over the world. The recommended therapy aims at dissolving the clot to re-establish quickly the blood flow to the brain and reduce neuronal injury. Intravenous administration of recombinant tissue-type plasminogen activator (rt-PA) is clinically used with this goal. AREAS COVERED A description of beneficial and detrimental effects of rt-PA treatment is addressed. An overview of new therapies against AIS, such as new thrombolytics, sonolysis and sonothrombolysis, endovascular procedures, and association therapies is provided. Updates on the pathophysiological process leading to intracranial hemorrhage (ICH) is also discussed. EXPERT OPINION rt-PA treatment in AIS patients is beneficial to recovery outcomes. To weaken risks and improve benefits, it might be relevant to consider: i) a definitive identification of risk factors for symptomatic ICH; ii). a better organization of the health care system to reduce time-to-treatment and enhance discharge management. The pharmacological improvement of new thrombolytic drugs (such as tenecteplase and desmoteplase) targeting harmful and maximally exploiting beneficial effects might further reduce mortality and disability in AIS.
Collapse
Affiliation(s)
- Aldo Bonaventura
- a First Clinic of Internal Medicine, Department of Internal Medicine , University of Genoa School of Medicine , Genoa , Italy
- b IRCCS AOU San Martino - IST, Genoa , Genoa , Italy
| | - Fabrizio Montecucco
- a First Clinic of Internal Medicine, Department of Internal Medicine , University of Genoa School of Medicine , Genoa , Italy
- b IRCCS AOU San Martino - IST, Genoa , Genoa , Italy
- c Centre of Excellence for Biomedical Research (CEBR) , University of Genoa , Genoa , Italy
| | - Franco Dallegri
- a First Clinic of Internal Medicine, Department of Internal Medicine , University of Genoa School of Medicine , Genoa , Italy
- b IRCCS AOU San Martino - IST, Genoa , Genoa , Italy
| |
Collapse
|
41
|
Klenke S, Renckhoff K, Engler A, Peters J, Frey UH. Easy-to-use strategy for reference gene selection in quantitative real-time PCR experiments. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:1353-1366. [PMID: 27650728 DOI: 10.1007/s00210-016-1305-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 09/09/2016] [Indexed: 12/19/2022]
Abstract
Real-time PCR is an indispensable technique for mRNA expression analysis but conclusions depend on appropriate reference gene selection. However, while reference gene selection has been a topic of publications, this issue is often disregarded when measuring target mRNA expression. Therefore, we (1) evaluated the frequency of appropriate reference gene selection, (2) suggest an easy-to-use tool for least variability reference gene selection, (3) demonstrate application of this tool, and (4) show effects on target gene expression profiles. All 2015 published articles in Naunyn-Schmiedeberg's Archives of Pharmacology were screened for the use of quantitative real-time PCR analysis and selection of reference genes. Target gene expression (Vegfa, Grk2, Sirt4, and Timp3) in H9c2 cells was analyzed following various interventions (hypoxia, hyperglycemia, and/or isoflurane exposure with and without subsequent hypoxia) in relation to putative reference genes (Actb, Gapdh, B2m, Sdha, and Rplp1) using the least variability method vs. an arbitrarily selected but established reference gene. In the vast majority (18 of 21) of papers, no information was provided regarding selection of an appropriate reference gene. In only 1 of 21 papers, a method of appropriate reference gene selection was described and in 2 papers reference gene selection remains unclear. The method of reference gene selection had major impact on interpretation of target gene expression. With hypoxia, for instance, the least variability gene was Rplp1 and target gene expression (Vefga) heavily showed a 2-fold up-regulation (p = 0.022) but no change (p = 0.3) when arbitrarily using Gapdh. Frequency of appropriate reference gene selection in this journal is low, and we propose our strategy for reference gene selection as an easy tool for proper target gene expression.
Collapse
Affiliation(s)
- Stefanie Klenke
- Klinik für Anästhesiologie & Intensivmedizin, Universität Duisburg-Essen and Universitätsklinikum Essen, Hufelandstr. 55, D-45122, Essen, Germany.
| | - Kristina Renckhoff
- Klinik für Anästhesiologie & Intensivmedizin, Universität Duisburg-Essen and Universitätsklinikum Essen, Hufelandstr. 55, D-45122, Essen, Germany
| | - Andrea Engler
- Klinik für Anästhesiologie & Intensivmedizin, Universität Duisburg-Essen and Universitätsklinikum Essen, Hufelandstr. 55, D-45122, Essen, Germany
| | - Jürgen Peters
- Klinik für Anästhesiologie & Intensivmedizin, Universität Duisburg-Essen and Universitätsklinikum Essen, Hufelandstr. 55, D-45122, Essen, Germany
| | - Ulrich H Frey
- Klinik für Anästhesiologie & Intensivmedizin, Universität Duisburg-Essen and Universitätsklinikum Essen, Hufelandstr. 55, D-45122, Essen, Germany
| |
Collapse
|
42
|
Topsakal R, Eryol NK, Abaci A, Oymak S, Ozdoğru I, Yilmaz Y, Seyfeli E, Oğuzhan A, Ergin A. The Relation Between Chronic Obstructive Pulmonary Disease and Coronary Collateral Vessels. Angiology 2016; 56:651-6. [PMID: 16327940 DOI: 10.1177/000331970505600601] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Coronary collateral vessels can provide a perfusion reserve in case of increased myocardial oxygen demand. Development of coronary collateral vessels (CCV) is triggered by the pressure gradient between the coronary bed of arteries caused by an obstruction and myocardial ischemia. Myocardial hypoxia can facilitate development of CCVs. There is a chronic hypoxemia in patients with chronic obstructive pulmonary disease (COPD). The aim of this study was to evaluate the effect of COPD on CCVs. The study included 98 patients with COPD who underwent coronary angiography. Those patients in whom coronary angiography is normal or severity of coronary artery stenosis in thought not to be sufficient for the development of CCVs (<80%) were excluded from the study. A total of 98 patients (mean age, 62 ±9 years) met the criteria for the COPD group. For case-control matching, 98 consecutive without COPD patients (mean age 62 ±10) who had one or more diseased vessels with 80% or greater stenosis were included in the control group. The CCVs were graded according to the Rentrop scoring system, and the collateral score was calculated by summing the Rentrop numbers of every patient. The mean number of diseased vessels in patients with COPD and without COPD were 1.61 ±0.69 and 1.77 ±0.89 (p=0.155), respectively. The mean collateral score was 2.15 ±2.03 in the COPD group and 1.32 ±1.54 in the control group. After confounding variables were controlled for, the collateral score in patients with COPD group was significantly different from that in patients without COPD group (p=0.002). These findings suggest that CCV development is better in patients with COPD than in those patients without COPD. Thus, COPD may be an important factor affecting CCV development, which may be related to the presence of chronic hypoxemia in patients with COPD.
Collapse
Affiliation(s)
- Ramazan Topsakal
- Department of Cardiology, Erciyes University, Medical Faculty, Kayseri, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Chhokar V, Tucker AL. Angiogenesis: Basic Mechanisms and Clinical Applications. Semin Cardiothorac Vasc Anesth 2016. [DOI: 10.1177/108925320300700304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The development and maintenance of an adequate vascular supply is critical for the viability of normal and neoplastic tissues. Angiogenesis, the development of new blood vessels from preexisting capillary networks, plays an important role in a number of physiologic and pathologic processes, including reproduction, wound repair, inflammatory diseases, and tumor growth. Angiogenesis involves sequential steps that are triggered in response to angiogenic growth factors released by inflammatory, mesenchymal, or tumor cells that act as ligands for endothelial cell receptor tyrosine kinases. Stimulated endothelial cells detach from neighboring cells and migrate, proliferate, and form tubes. The immature tubes are subsequently invested and stabilized by pericytes or smooth muscle cells. Angiogenesis depends upon complex interactions among various classes of molecules, including adhesion molecules, proteases, structural proteins, cell surface receptors, and growth factors. The therapeutic manipulation of angiogenesis targeted against ischemic and neoplastic diseases has been investigated in preclinical animal models and in clinical trials. Proangiogenic trials that have stimulated vessel growth in ischemic coronary or peripheral tissues through expression, delivery, or stimulated release of growth factors have shown efficacy in animal models and mixed results in human clinical trials. Antiangiogenic trials have used strategies to block the function of molecules critical for new vessel growth or maturation in the treatment of a variety of malignancies, mostly with results less encouraging than those seen in preclinical models. Pro-and antiangiogenic clinical trials demonstrate that strategies for optimal drug delivery, dosing schedules, patient selection, and endpoint measurements need further investigation and refinement before the therapeutic manipulation of angiogenesis will realize its full clinical potential.
Collapse
Affiliation(s)
- Vikram Chhokar
- Department of Internal Medicine, Salem VA Health System, Roanoke, Virginia
| | - Amy L. Tucker
- Department of Internal Medicine, Cardiovascular Division; Cardiovascular Research Center; Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
45
|
Schwarz ER, Meven DA, Sulemanjee NZ, Kersting PH, Tussing T, Skobel EC, Hanrath P, Uretsky BF. Monocyte Chemoattractant Protein 1-Induced Monocyte Infiltration Produces Angiogenesis but Not Arteriogenesis in Chronically Infarcted Myocardium. J Cardiovasc Pharmacol Ther 2016; 9:279-89. [PMID: 15678247 DOI: 10.1177/107424840400900408] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Monocyte chemoattractant protein 1 (MCP-1) stimulates the invasion of monocytes into ischemic tissue with concomitant adhesion to endothelial cells. Monocyte stimulation has been shown to be involved in the induction of arteriogenesis, which is the development of functional arterioles resulting in improvement of perfusion. However, angiogenesis (newly developed capillaries contribute to improved tissue perfusion) in several models has not resulted in any improvement in blood flow. Objective: The effects of MCP-1 on potential angiogenesis and arteriogenesis as well as changes in left ventricular function were tested in a chronic infarct model in rat hearts. Methods: Anesthetized rats were subjected to open-chest ligation of the left coronary artery with subsequent myocardial infarction. After 6 weeks, animals were randomized to receive either MCP-1 (3 µL in 0.15 mL NaCl, group 1, n = 9) or saline (0.15 mL, group 2, n = 9), which was injected into the myocardium at the border zones of the infarcts. For assessment of left ventricular dimensions and global cardiac function, transthoracic two-dimensional echocardiography was performed at baseline, 6 weeks after myocardial infarction, and 4 weeks after MCP-1 or saline injection, by use of a 12-MHz pediatric transducer. For light microscopic analysis, myocardial tissue was stained with Elastica-van-Giesson and von Willebrand factor for blood vessels and endothelial cells, respectively. In a subset of animals, hearts were excised 24 hours after MCP-1 administration (n = 4) or saline administration (n = 4) for assessment of monocyte infiltration by immunohistologic staining of the CD31 antigen. Results: Left ventricular dimensions and ejection fraction changed after coronary occlusion (from 60.4% ± 2.85% to 24.8% ± 5.01% ejection fraction in group 1, and from 58.4% ±2.06% to 26.3% ± 4.3% ejection fraction in group 2 at 6 weeks, P < .005) without any further change 4 weeks after treatment (ejection fraction in group 1, 26.3% ± 2.7%, ejection fraction in group 2, 25.0% ± 5.18%). The MCP-1 group resulted in 390.6 ± 10.36 endothelial cells compared with 285.2 ± 13.56 in group 2 ( P < .005) at the injection site. Monocyte infiltration was observed at the MCP-1 injection site with an increase in capillary growth (angiogenesis). However, there was no difference in the number of arteriolar structures between animals treated with MCP-1 and saline animals (group 1, 19.0 ± 1.52 vs group 2,16.4 ± 0.68, P > .05). Conclusion: A single intramyocardial injection of MCP-1 into the infarct border zone resulted in neo-angiogenesis and monocyte infiltration but not arteriogenesis in the rat heart. There was no functional change of chronically infarcted myocardium in the present model.
Collapse
Affiliation(s)
- Ernst R Schwarz
- Department of Cardiology, Rheinisch-Westfaelisch Technische Hochschule University Hospital Aachen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Mateika JH, Komnenov D. Intermittent hypoxia initiated plasticity in humans: A multipronged therapeutic approach to treat sleep apnea and overlapping co-morbidities. Exp Neurol 2016; 287:113-129. [PMID: 27170208 DOI: 10.1016/j.expneurol.2016.05.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/18/2016] [Accepted: 05/03/2016] [Indexed: 10/21/2022]
Abstract
Over the past three decades exposure to intermittent hypoxia (IH) has generally been considered a stimulus associated with a number of detrimental outcomes. However, there is sufficient evidence to link IH to many beneficial outcomes but they have largely been ignored, particularly in the field of sleep medicine in the United States. Recent reviews have postulated that this apparent contradiction is related to the severity and duration of exposure to IH; mild forms of IH initiate beneficial outcomes while severe forms of IH are coupled to detrimental consequences. In the present review we explore the role that IH has in initiating respiratory plasticity and the potential this form of plasticity has to mitigate obstructive sleep apnea (OSA) in humans. In taking this approach, we address the possibility that IH could serve as an adjunct therapy coupled with continuous positive airway pressure (CPAP) to treat OSA. Our working hypothesis is that exposure to mild IH leads to respiratory plasticity that manifests in increased stability of the upper airway, which could ultimately reduce the CPAP required to treat OSA. In turn, this reduction could increase CPAP compliance and extend the length of treatment each night, which might improve the magnitude of outcome measures. Improved treatment compliance coupled with the direct effect that IH has on numerous overlapping conditions (i.e. asthma, chronic obstructive pulmonary disease, spinal cord injury) may well lead to substantial improvements that exceed outcomes following treatment with CPAP alone. Overall, this review will consider evidence from the published literature which suggests that IH could serve as an effective multipronged therapeutic approach to treat sleep apnea and its overlapping co-morbidities.
Collapse
Affiliation(s)
- Jason H Mateika
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI 48201, United States; Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, United States; Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI 48201, United States.
| | - Dragana Komnenov
- John D. Dingell Veterans Affairs Medical Center, Detroit, MI 48201, United States; Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, United States
| |
Collapse
|
47
|
Suzuki Y, Nagai N, Umemura K. A Review of the Mechanisms of Blood-Brain Barrier Permeability by Tissue-Type Plasminogen Activator Treatment for Cerebral Ischemia. Front Cell Neurosci 2016; 10:2. [PMID: 26834557 PMCID: PMC4724711 DOI: 10.3389/fncel.2016.00002] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/04/2016] [Indexed: 11/13/2022] Open
Abstract
Cerebrovascular homeostasis is maintained by the blood-brain barrier (BBB), which forms a mechanical and functional barrier between systemic circulation and the central nervous system (CNS). In patients with ischemic stroke, the recombinant tissue-type plasminogen activator (rt-PA) is used to accelerate recanalization of the occluded vessels. However, rt-PA is associated with a risk of increasing intracranial bleeding (ICB). This effect is thought to be caused by the increase in cerebrovascular permeability though various factors such as ischemic reperfusion injury and the activation of matrix metalloproteinases (MMPs), but the detailed mechanisms are unknown. It was recently found that rt-PA treatment enhances BBB permeability not by disrupting the BBB, but by activating the vascular endothelial growth factor (VEGF) system. The VEGF regulates both the dissociation of endothelial cell (EC) junctions and endothelial endocytosis, and causes a subsequent increase in vessel permeability through the VEGF receptor-2 (VEGFR-2) activation in ECs. Here, we review the possibility that rt-PA increases the penetration of toxic molecules derived from the bloodstream including rt-PA itself, without disrupting the BBB, and contributes to these detrimental processes in the cerebral parenchyma.
Collapse
Affiliation(s)
- Yasuhiro Suzuki
- Department of Pharmacology, Hamamatsu University School of MedicineHamamatsu, Japan; School of Pharmaceutical Sciences, Ohu UniversityKoriyama, Japan
| | - Nobuo Nagai
- Faculty of Bioscience, Department of Animal Bioscience, Nagahama Institute of Bio-Science and Technology Nagahama, Japan
| | - Kazuo Umemura
- Department of Pharmacology, Hamamatsu University School of Medicine Hamamatsu, Japan
| |
Collapse
|
48
|
Paz Y, Shinfeld A. Re: “Transcatheter treatment for refractory angina with the coronary sinus Reducer” by Maayan Konigstein et al. EUROINTERVENTION 2015; 11:727. [DOI: 10.4244/eijy14m07_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
49
|
Zhu H, Jiang X, Li X, Hu M, Wan W, Wen Y, He Y, Zheng X. Intramyocardial delivery of VEGF165 via a novel biodegradable hydrogel induces angiogenesis and improves cardiac function after rat myocardial infarction. Heart Vessels 2015; 31:963-75. [PMID: 26142379 DOI: 10.1007/s00380-015-0710-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 06/24/2015] [Indexed: 12/20/2022]
Abstract
Vascular endothelial growth factor (VEGF), an independent mitogen, has been reported to induce angiogenesis and thus attenuates the damage induced by myocardial infarction (MI). VEGF165 is the most abundant and predominant isoform of VEGF. This study investigates whether this effect could be strengthened by local intramyocardial injection of VEGF165 along with a novel biodegradable Dex-PCL-HEMA/PNIPAAm hydrogel and ascertains its possible mechanism of action. Rat models of myocardial infarction were induced by coronary artery ligation. Phosphate-buffered saline (PBS group), Dex-PCL-HEMA/PNIPAAm hydrogel (Gel group), phosphate-buffered saline containing VEGF165 (VP group), and hydrogel containing VEGF165 (VPG group) were injected into a peri-infarcted area of cardiac tissue immediately after myocardial infarction, respectively. The sham group was thoracic but without myocardial infarction. The injection of VEGF165 along with a hydrogel induced angiogenesis, reduced collagen content and MI area, inhibited cell apoptosis, increased the level of VEGF165 protein and the expression of flk-1 and flt-1, and improved cardiac function compared with the injection of either alone after MI in rats. The results suggest that injection of VEGF165 along with a hydrogel acquires more cardioprotective effects than either alone in rat with MI by sustained release of VEGF165, then may enhance the feedback between VEGF and its receptors flk-1 and flt-1.
Collapse
Affiliation(s)
- Hongling Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.
| | - Xiaoyan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Miaoyang Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Weiguo Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Ying Wen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Yiyu He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Xiaoxin Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| |
Collapse
|
50
|
Yang Y, Shi C, Hou X, Zhao Y, Chen B, Tan B, Deng Z, Li Q, Liu J, Xiao Z, Miao Q, Dai J. Modified VEGF targets the ischemic myocardium and promotes functional recovery after myocardial infarction. J Control Release 2015; 213:27-35. [PMID: 26144351 DOI: 10.1016/j.jconrel.2015.06.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 06/25/2015] [Accepted: 06/29/2015] [Indexed: 02/07/2023]
Abstract
Vascular endothelial growth factor (VEGF) promotes angiogenesis and improves cardiac function after myocardial infarction (MI). However, the non-targeted delivery of VEGF decreases its therapeutic efficacy due to an insufficient local concentration in the ischemic myocardium. In this study, we used a specific peptide to modify VEGF and determined that this modified VEGF (IMT-VEGF) localized to the ischemic myocardium through intravenous injection by interacting with cardiac troponin I (cTnI). When IMT-VEGF was used to mediate cardiac repair in a rat model of ischemia-reperfusion (I-R) injury, we observed a decreased scar size, enhanced angiogenesis and improved cardiac function. Moreover, an alternative treatment using the repeated administration of a low-dose IMT-VEGF also promoted angiogenesis and functional recovery. The therapeutic effects of IMT-VEGF were further confirmed in a pig model of MI as the result of the conserved properties of its interacting protein, cTnI. These results suggest a promising therapeutic strategy for MI based on the targeted delivery of IMT-VEGF.
Collapse
Affiliation(s)
- Yun Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Graduate School, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China
| | - Chunying Shi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Institute for Translational Medicine, College of Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266021, China
| | - Xianglin Hou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Road, Chongqing, 400038, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Road, Chongqing, 400038, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Road, Chongqing, 400038, China
| | - Bo Tan
- Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, 398 Ruoshui Road, Suzhou, 215123, China
| | - Zongwu Deng
- Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, 398 Ruoshui Road, Suzhou, 215123, China
| | - Qingguo Li
- Department of Cardiothoracic Surgery, the affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Jianzhou Liu
- Department of Cardiac Surgery, Peking Union Medical College Hospital, Peking Union Medical College, 1 Shuaifuyuan, Beijing, 100730, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Road, Chongqing, 400038, China
| | - Qi Miao
- Department of Cardiac Surgery, Peking Union Medical College Hospital, Peking Union Medical College, 1 Shuaifuyuan, Beijing, 100730, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Road, Chongqing, 400038, China.
| |
Collapse
|