1
|
Alshehri MS, Alqahtani FS. Unusual presentation of tractional retinal detachment in beta thalassemia minor. Am J Ophthalmol Case Rep 2023; 32:101946. [PMID: 37942050 PMCID: PMC10630591 DOI: 10.1016/j.ajoc.2023.101946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 10/01/2023] [Accepted: 10/18/2023] [Indexed: 11/10/2023] Open
Abstract
Purpose To report a case of proliferative retinopathy with tractional retinal detachment associated with beta thalassemia minor in a 27-year-old female. Observations A young lady having beta thalassemia minor presented with decreased vision in both eyes, the effect being more severe in her right eye. The patient's other systemic history, including ophthalmic history, was unremarkable. The fundus examination revealed peripheral retinal ischemia in both eyes and tractional retinal detachment in the right eye. Conclusion Beta thalassemia minor is not associated with striking retinal pathology, nevertheless proliferative retinopathy. However, in this case the patient developed tractional retinal detachment that required surgery. This indicates that proliferative changes may develop in patients with beta thalassemia, and routine fundus examinations could be recommended for these patients.
Collapse
|
2
|
Fu J, Zong X, Jin M, Min J, Wang F, Wang Y. Mechanisms and regulation of defensins in host defense. Signal Transduct Target Ther 2023; 8:300. [PMID: 37574471 PMCID: PMC10423725 DOI: 10.1038/s41392-023-01553-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/11/2023] [Accepted: 06/26/2023] [Indexed: 08/15/2023] Open
Abstract
As a family of cationic host defense peptides, defensins are mainly synthesized by Paneth cells, neutrophils, and epithelial cells, contributing to host defense. Their biological functions in innate immunity, as well as their structure and activity relationships, along with their mechanisms of action and therapeutic potential, have been of great interest in recent years. To highlight the key research into the role of defensins in human and animal health, we first describe their research history, structural features, evolution, and antimicrobial mechanisms. Next, we cover the role of defensins in immune homeostasis, chemotaxis, mucosal barrier function, gut microbiota regulation, intestinal development and regulation of cell death. Further, we discuss their clinical relevance and therapeutic potential in various diseases, including infectious disease, inflammatory bowel disease, diabetes and obesity, chronic inflammatory lung disease, periodontitis and cancer. Finally, we summarize the current knowledge regarding the nutrient-dependent regulation of defensins, including fatty acids, amino acids, microelements, plant extracts, and probiotics, while considering the clinical application of such regulation. Together, the review summarizes the various biological functions, mechanism of actions and potential clinical significance of defensins, along with the challenges in developing defensins-based therapy, thus providing crucial insights into their biology and potential clinical utility.
Collapse
Affiliation(s)
- Jie Fu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Hangzhou, Zhejiang Province, China
| | - Xin Zong
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Hangzhou, Zhejiang Province, China
| | - Mingliang Jin
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Hangzhou, Zhejiang Province, China
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China.
| | - Yizhen Wang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China.
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
3
|
Role of Defensins in Tumor Biology. Int J Mol Sci 2023; 24:ijms24065268. [PMID: 36982340 PMCID: PMC10049535 DOI: 10.3390/ijms24065268] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/02/2023] [Accepted: 03/04/2023] [Indexed: 03/12/2023] Open
Abstract
Defensins have long been considered as merely antimicrobial peptides. Throughout the years, more immune-related functions have been discovered for both the α-defensin and β-defensin subfamily. This review provides insights into the role of defensins in tumor immunity. Since defensins are present and differentially expressed in certain cancer types, researchers started to unravel their role in the tumor microenvironment. The human neutrophil peptides have been demonstrated to be directly oncolytic by permealizing the cell membrane. Further, defensins can inflict DNA damage and induce apoptosis of tumor cells. In the tumor microenvironment, defensins can act as chemoattractants for subsets of immune cells, such as T cells, immature dendritic cells, monocytes and mast cells. Additionally, by activating the targeted leukocytes, defensins generate pro-inflammatory signals. Moreover, immuno-adjuvant effects have been reported in a variety of models. Therefore, the action of defensins reaches beyond their direct antimicrobial effect, i.e., the lysis of microbes invading the mucosal surfaces. By causing an increase in pro-inflammatory signaling events, cell lysis (generating antigens) and attraction and activation of antigen presenting cells, defensins could have a relevant role in activating the adaptive immune system and generating anti-tumor immunity, and could thus contribute to the success of immune therapy.
Collapse
|
4
|
Decreased leukocyte exhaustion is associated with decreased IFN-β and increased α-defensin-1 levels in type-2 diabetes. Cytokine 2022; 156:155918. [DOI: 10.1016/j.cyto.2022.155918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/15/2022] [Accepted: 05/16/2022] [Indexed: 11/21/2022]
|
5
|
Moeinabadi-Bidgoli K, Rezaee M, Rismanchi H, Mohammadi MM, Babajani A. Mesenchymal Stem Cell-Derived Antimicrobial Peptides as Potential Anti-Neoplastic Agents: New Insight into Anticancer Mechanisms of Stem Cells and Exosomes. Front Cell Dev Biol 2022; 10:900418. [PMID: 35874827 PMCID: PMC9298847 DOI: 10.3389/fcell.2022.900418] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 06/20/2022] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs), as adult multipotent cells, possess considerable regenerative and anti-neoplastic effects, from inducing apoptosis in the cancer cells to reducing multidrug resistance that bring them up as an appropriate alternative for cancer treatment. These cells can alter the behavior of cancer cells, the condition of the tumor microenvironment, and the activity of immune cells that result in tumor regression. It has been observed that during inflammatory conditions, a well-known feature of the tumor microenvironment, the MSCs produce and release some molecules called “antimicrobial peptides (AMPs)” with demonstrated anti-neoplastic effects. These peptides have remarkable targeted anticancer effects by attaching to the negatively charged membrane of neoplastic cells, disrupting the membrane, and interfering with intracellular pathways. Therefore, AMPs could be considered as a part of the wide-ranging anti-neoplastic effects of MSCs. This review focuses on the possible anti-neoplastic effects of MSCs-derived AMPs and their mechanisms. It also discusses preconditioning approaches and using exosomes to enhance AMP production and delivery from MSCs to cancer cells. Besides, the clinical administration of MSCs-derived AMPs, along with their challenges in clinical practice, were debated.
Collapse
Affiliation(s)
- Kasra Moeinabadi-Bidgoli
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Basic and Molecular Epidemiology of Gastroenterology Disorders Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Malihe Rezaee
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Rismanchi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Amirhesam Babajani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Basingab F, Alsaiary A, Almontashri S, Alrofaidi A, Alharbi M, Azhari S, Algothmi K, Alhazmi S. Alterations in Immune-Related Defensin Alpha 4 ( DEFA4) Gene Expression in Health and Disease. Int J Inflam 2022; 2022:9099136. [PMID: 35668817 PMCID: PMC9167129 DOI: 10.1155/2022/9099136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 03/31/2022] [Accepted: 05/05/2022] [Indexed: 12/15/2022] Open
Abstract
Defensin Alpha 4 (DEFA4) is the fourth member of the Alpha Defensins family known as a part of antimicrobial peptides in the innate immune system. DEFA4 has a strong preference to kill Gram-negative bacteria more than Gram-positive bacteria. In addition, DEFA4 exhibits antiviral activity against human immunodeficiency virus type 1 (HIV-1) in vitro. Moreover, DEFA4 can act as an inhibitor of corticosterone production (Corticostatin). On the other hand, alternations in DEFA4 gene expression have been reported in different disorders such as diseases related to inflammation and immunity dysfunction, brain-related disorders, and various cancers. The up-regulation of DEFA4 appears to be involved in the malignant transformation or aggressive form of cancer. Interestingly, the modified version of DEFA4 fragment (1-11) was potent and efficient against antibiotic-resistant bacteria. This review provides a general background abSaudi Arabia out DEFA4 and sheds light on changes in DEFA4 gene expression in different diseases. The paper also discusses other aspects related to DEFA4 as an antimicrobial and antiviral agent. The research was conducted based on available articles obtained from databases starting from 1988 to the present.
Collapse
Affiliation(s)
- Fatemah Basingab
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Unit, King Fahad for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abeer Alsaiary
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Biology Department, College of Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Shahad Almontashri
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Aisha Alrofaidi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mona Alharbi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sheren Azhari
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khloud Algothmi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Safiah Alhazmi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
7
|
Overview of Host Defense Peptides with Promising Anti-Breast Cancer Activity. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2022. [DOI: 10.2478/sjecr-2021-0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Breast cancer is the leading cause of death among women worldwide. The main limitations of conventional anti-cancer therapy, including breast cancer treatment, are side effects and the development of resistance to chemotherapeutics. Host defense peptides (HDPs) are bioactive compounds of innate immunity isolated from almost all living organisms, which exhibit wide range of biological activities. This review focuses on the anti-cancer effects of HDPs and their therapeutic potential against breast cancer. Numerous HDPs from different sources, including mammalian and amphibian origin, and their chemically modified analogues, exert the spectrum of anti-cancer activities. These effects include direct disruption of cancer cell membrane, induction of apoptosis, inhibition of angiogenesis and cancer cell proliferation, but also the modulation of anti-cancer immune response. Selected examples of HDPs of different origin and their anti-breast cancer capacities have been reviewed. Conclusively, due to their anti-cancer effects accompanied by substantial selectivity for cancer cells and low toxicity for normal cells, HDPs have been widely recognized as possible therapeutic agents.
Collapse
|
8
|
Zhang MY, Zhu L, Zheng X, Xie TH, Wang W, Zou J, Li Y, Li HY, Cai J, Gu S, Yao Y, Wei TT. TGR5 Activation Ameliorates Mitochondrial Homeostasis via Regulating the PKCδ/Drp1-HK2 Signaling in Diabetic Retinopathy. Front Cell Dev Biol 2022; 9:759421. [PMID: 35096809 PMCID: PMC8795816 DOI: 10.3389/fcell.2021.759421] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/07/2021] [Indexed: 01/07/2023] Open
Abstract
Background: Diabetic retinopathy (DR) is one of the most important microvascular diseases of diabetes. Our previous research demonstrated that bile acid G-protein-coupled membrane receptor (TGR5), a novel cell membrane receptor of bile acid, ameliorates the vascular endothelial cell dysfunction in DR. However, the precise mechanism leading to this alteration remains unknown. Thus, the mechanism of TGR5 in the progress of DR should be urgently explored. Methods: In this study, we established high glucose (HG)-induced human retinal vascular endothelial cells (RMECs) and streptozotocin-induced DR rat in vitro and in vivo. The expression of TGR5 was interfered through the specific agonist or siRNA to study the effect of TGR5 on the function of endothelial cell in vitro. Western blot, immunofluorescence and fluorescent probes were used to explore how TGR5 regulated mitochondrial homeostasis and related molecular mechanism. The adeno-associated virus serotype 8-shTGR5 (AAV8-shTGR5) was performed to evaluate retinal dysfunction in vivo and further confirm the role of TGR5 in DR by HE staining, TUNEL staining, PAS staining and Evans Blue dye. Results: We found that TGR5 activation alleviated HG-induced endothelial cell apoptosis by improving mitochondrial homeostasis. Additionally, TGR5 signaling reduced mitochondrial fission by suppressing the Ca2+-PKCδ/Drp1 signaling and enhanced mitophagy through the upregulation of the PINK1/Parkin signaling pathway. Furthermore, our result indicated that Drp1 inhibited mitophagy by facilitating the hexokinase (HK) 2 separation from the mitochondria and HK2-PINK1/Parkin signaling. In vivo, intraretinal microvascular abnormalities, including retinal vascular leakage, acellular capillaries and apoptosis, were poor in AAV8-shTGR5-treated group under DR, but this effect was reversed by pretreatment with the mitochondrial fission inhibitor Mdivi-1 or autophagy agonist Rapamycin. Conclusion: Overall, our findings indicated that TGR5 inhibited mitochondrial fission and enhanced mitophagy in RMECs by regulating the PKCδ/Drp1-HK2 signaling pathway. These results revealed the molecular mechanisms underlying the protective effects of TGR5 and suggested that activation of TGR5 might be a potential therapeutic strategy for DR.
Collapse
Affiliation(s)
- Meng-Yuan Zhang
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Lingpeng Zhu
- Center of Clinical Research, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Xinhua Zheng
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Tian-Hua Xie
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Wenjuan Wang
- Center of Clinical Research, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Jian Zou
- Center of Clinical Research, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Yan Li
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Hong-Ying Li
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Jiping Cai
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Shun Gu
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Yong Yao
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China.,Department of Ophthalmology, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, China
| | - Ting-Ting Wei
- Center of Clinical Research, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
9
|
Li D, Guo F, Chen K, Yang X, Wang Y. Preclinical Safety Evaluation of a Recombinant Plasmid Vector Encoding Mature Human Neutrophil Peptide-1 by Repeated Local Administrations in Nonhuman Primates. Hum Gene Ther 2021; 32:1382-1389. [PMID: 33858232 DOI: 10.1089/hum.2020.289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
In our previous studies, a novel gene therapy approach was developed based on a plasmid vector pSecTag2B in which recombinant HNP1 gene was regulated under a cytomegalovirus promoter to encode a mature human neutrophil peptide-1 (HNP1) form. We showed for the first time in various tumor models, including human cancer xenografts, that overexpression of HNP1 in the tumor milieu by intratumoral pSecTag-HNP1 (pHNP1) administration efficiently attenuated in vivo tumor progression, mediated host immune responses to tumors, and produced a synergistic effect when combined with chemotherapeutics. In this study, a preclinical safety investigation of HNP1 gene therapy was conducted in nonhuman primates. Eleven cynomolgus monkeys were divided into three groups of three to four animals each and received either repeated s.c. injections of pHNP1/cationic liposome complexes at a low (0.625 mg/kg) or a high (2.5 mg/kg) dose or glucose as control. Significant HNP1 in vivo accumulation was detected after consecutive administrations. All primates reached the end of the study with good body conditions. Injection site inflammation was the only obvious toxic reaction during observation period. In addition, elevation of monocyte/macrophage and neutrophil as well as decline of lymphocyte were detected in the peripheral blood of pHNP1-treated primates. These alterations were partially alleviated at the end of observation period. Besides, dose-related histopathological changes of the immune organs were observed at necropsy, including a minimal thymic lymphocyte decrease and a minimal-to-mild lymph node erythrocyte increase, but which cannot be excluded from HNP1-induced immune reactions. Together, these data support future clinical studies of pHNP1-based local gene delivery in tumor patients.
Collapse
Affiliation(s)
- Dan Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, and Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Fuchun Guo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ke Chen
- WestChina-Frontier Pharma Tech Co., Ltd. (WCFP), National Chengdu Center for Safety Evaluation of Drugs (NCCSED), Chengdu, China
| | - Xunning Yang
- WestChina-Frontier Pharma Tech Co., Ltd. (WCFP), National Chengdu Center for Safety Evaluation of Drugs (NCCSED), Chengdu, China
| | - Yongsheng Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,Institute of Drug Clinical Trial, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Abstract
Defensins are a major family of host defense peptides expressed predominantly in neutrophils and epithelial cells. Their broad antimicrobial activities and multifaceted immunomodulatory functions have been extensively studied, cementing their role in innate immunity as a core host-protective component against bacterial, viral and fungal infections. More recent studies, however, paint defensins in a bad light such that they are "alleged" to promote viral and bacterial infections in certain biological settings. This mini review summarizes the latest findings on the potential pathogenic properties of defensins against the backdrop of their protective roles in antiviral and antibacterial immunity. Further, a succinct description of both tumor-proliferative and -suppressive activities of defensins is also given to highlight their functional and mechanistic complexity in antitumor immunity. We posit that given an enabling environment defensins, widely heralded as the "Swiss army knife," can function as a "double-edged sword" in host immunity.
Collapse
Affiliation(s)
- Dan Xu
- Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Sciences and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Wuyuan Lu
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
11
|
Hajishengallis G. New developments in neutrophil biology and periodontitis. Periodontol 2000 2019; 82:78-92. [DOI: 10.1111/prd.12313] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- George Hajishengallis
- Department of Microbiology Penn Dental Medicine University of Pennsylvania Philadelphia Pennsylvania, USA
| |
Collapse
|
12
|
Sprott D, Poitz DM, Korovina I, Ziogas A, Phieler J, Chatzigeorgiou A, Mitroulis I, Deussen A, Chavakis T, Klotzsche-von Ameln A. Endothelial-Specific Deficiency of ATG5 (Autophagy Protein 5) Attenuates Ischemia-Related Angiogenesis. Arterioscler Thromb Vasc Biol 2019; 39:1137-1148. [DOI: 10.1161/atvbaha.119.309973] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Pathological angiogenesis, such as exuberant retinal neovascularization during proliferative retinopathies, involves endothelial responses to ischemia/hypoxia and oxidative stress. Autophagy is a clearance system enabling bulk degradation of intracellular components and is implicated in cellular adaptation to stressful conditions. Here, we addressed the role of the ATG5 (autophagy-related protein 5) in endothelial cells in the context of pathological ischemia-related neovascularization in the murine model of retinopathy of prematurity.
Approach and Results—
Autophagic vesicles accumulated in neovascular tufts of the retina of retinopathy of prematurity mice. Endothelium-specific
Atg5
deletion reduced pathological neovascularization in the retinopathy of prematurity model. In contrast, no alterations in physiological retina vascularization were observed in endothelial-specific ATG5 deficiency, suggesting a specific role of endothelial ATG5 in pathological hypoxia/reoxygenation–related angiogenesis. Consistently, in an aortic ring angiogenesis assay, endothelial ATG5 deficiency resulted in impaired angiogenesis under hypoxia/reoxygenation conditions. As compared to ATG5-sufficient endothelial cells, ATG5-deficient cells displayed impaired mitochondrial respiratory activity, diminished production of mitochondrial reactive oxygen species and decreased phosphorylation of the VEGFR2 (vascular endothelial growth factor receptor 2). Consistently, ATG5-deficient endothelial cells displayed decreased oxidative inactivation of PTPs (phospho-tyrosine phosphatases), likely due to the reduced reactive oxygen species levels resulting from ATG5 deficiency.
Conclusions—
Our data suggest that endothelial ATG5 supports mitochondrial function and proangiogenic signaling in endothelial cells in the context of pathological hypoxia/reoxygenation–related neovascularization. Endothelial ATG5, therefore, represents a potential target for the treatment of pathological neovascularization-associated diseases, such as retinopathies.
Collapse
Affiliation(s)
- David Sprott
- From the Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty (D.S., D.M.P., I.K., A.Z., J.P., A.C., I.M., T.C., A.K.-v.A.), Technische Universität Dresden, Germany
| | - David M. Poitz
- From the Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty (D.S., D.M.P., I.K., A.Z., J.P., A.C., I.M., T.C., A.K.-v.A.), Technische Universität Dresden, Germany
- Department of Internal Medicine and Cardiology (D.M.P.), Technische Universität Dresden, Germany
| | - Irina Korovina
- From the Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty (D.S., D.M.P., I.K., A.Z., J.P., A.C., I.M., T.C., A.K.-v.A.), Technische Universität Dresden, Germany
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine (I.K.), Technische Universität Dresden, Germany
| | - Athanasios Ziogas
- From the Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty (D.S., D.M.P., I.K., A.Z., J.P., A.C., I.M., T.C., A.K.-v.A.), Technische Universität Dresden, Germany
| | - Julia Phieler
- From the Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty (D.S., D.M.P., I.K., A.Z., J.P., A.C., I.M., T.C., A.K.-v.A.), Technische Universität Dresden, Germany
| | - Antonios Chatzigeorgiou
- From the Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty (D.S., D.M.P., I.K., A.Z., J.P., A.C., I.M., T.C., A.K.-v.A.), Technische Universität Dresden, Germany
| | - Ioannis Mitroulis
- From the Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty (D.S., D.M.P., I.K., A.Z., J.P., A.C., I.M., T.C., A.K.-v.A.), Technische Universität Dresden, Germany
| | - Andreas Deussen
- Institute for Physiology (A.D., A.K.-v.A.), Technische Universität Dresden, Germany
| | - Triantafyllos Chavakis
- From the Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty (D.S., D.M.P., I.K., A.Z., J.P., A.C., I.M., T.C., A.K.-v.A.), Technische Universität Dresden, Germany
| | - Anne Klotzsche-von Ameln
- From the Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty (D.S., D.M.P., I.K., A.Z., J.P., A.C., I.M., T.C., A.K.-v.A.), Technische Universität Dresden, Germany
- Institute for Physiology (A.D., A.K.-v.A.), Technische Universität Dresden, Germany
| |
Collapse
|
13
|
Troullinaki M, Alexaki V, Mitroulis I, Witt A, Klotzsche–von Ameln A, Chung K, Chavakis T, Economopoulou M. Nerve growth factor regulates endothelial cell survival and pathological retinal angiogenesis. J Cell Mol Med 2019; 23:2362-2371. [PMID: 30680928 PMCID: PMC6433692 DOI: 10.1111/jcmm.14002] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 09/28/2018] [Accepted: 10/15/2018] [Indexed: 01/01/2023] Open
Abstract
The mechanism underlying vasoproliferative retinopathies like retinopathy of prematurity (ROP) is hypoxia-triggered neovascularisation. Nerve growth factor (NGF), a neurotrophin supporting survival and differentiation of neuronal cells may also regulate endothelial cell functions. Here we studied the role of NGF in pathological retinal angiogenesis in the course of the ROP mouse model. Topical application of NGF enhanced while intraocular injections of anti-NGF neutralizing antibody reduced pathological retinal vascularization in mice subjected to the ROP model. The pro-angiogenic effect of NGF in the retina was mediated by inhibition of retinal endothelial cell apoptosis. In vitro, NGF decreased the intrinsic (mitochondria-dependent) apoptosis in hypoxia-treated human retinal microvascular endothelial cells and preserved the mitochondrial membrane potential. The anti-apoptotic effect of NGF was associated with increased BCL2 and reduced BAX, as well as with enhanced ERK and AKT phosphorylation, and was abolished by inhibition of the AKT pathway. Our findings reveal an anti-apoptotic role of NGF in the hypoxic retinal endothelium, which is involved in promoting pathological retinal vascularization, thereby pointing to NGF as a potential target for proliferative retinopathies.
Collapse
Affiliation(s)
- Maria Troullinaki
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Clinic Carl Gustav Carus, TU DresdenDresdenGermany
| | - Vasileia‐Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Clinic Carl Gustav Carus, TU DresdenDresdenGermany
| | - Ioannis Mitroulis
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Clinic Carl Gustav Carus, TU DresdenDresdenGermany
| | - Anke Witt
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Clinic Carl Gustav Carus, TU DresdenDresdenGermany
| | - Anne Klotzsche–von Ameln
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Clinic Carl Gustav Carus, TU DresdenDresdenGermany
| | - Kyoung‐Jin Chung
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Clinic Carl Gustav Carus, TU DresdenDresdenGermany
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Clinic Carl Gustav Carus, TU DresdenDresdenGermany
| | - Matina Economopoulou
- Department of OphthalmologyUniversity Clinic Carl Gustav Carus, TU DresdenDresdenGermany
| |
Collapse
|
14
|
Korovina I, Neuwirth A, Sprott D, Weber S, Sardar Pasha SPB, Gercken B, Breier G, El-Armouche A, Deussen A, Karl MO, Wielockx B, Chavakis T, Klotzsche-von Ameln A. Hematopoietic hypoxia-inducible factor 2α deficiency ameliorates pathological retinal neovascularization via modulation of endothelial cell apoptosis. FASEB J 2018; 33:1758-1770. [PMID: 30156910 DOI: 10.1096/fj.201800430r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A hallmark of proliferative retinopathies, such as retinopathy of prematurity (ROP), is a pathological neovascularization orchestrated by hypoxia and the resulting hypoxia-inducible factor (HIF)-dependent response. We studied the role of Hif2α in hematopoietic cells for pathological retina neovascularization in the murine model of ROP, the oxygen-induced retinopathy (OIR) model. Hematopoietic-specific deficiency of Hif2α ameliorated pathological neovascularization in the OIR model, which was accompanied by enhanced endothelial cell apoptosis. That latter finding was associated with up-regulation of the apoptosis-inducer FasL in Hif2α-deficient microglia. Consistently, pharmacological inhibition of the FasL reversed the reduced pathological neovascularization from hematopoietic-specific Hif2α deficiency. Our study found that the hematopoietic cell Hif2α contributes to pathological retina angiogenesis. Our findings not only provide novel insights regarding the complex interplay between immune cells and endothelial cells in hypoxia-driven retina neovascularization but also may have therapeutic implications for proliferative retinopathies.-Korovina, I., Neuwirth, A., Sprott, D., Weber, S., Sardar Pasha, S. P. B., Gercken, B., Breier, G., El-Armouche, A., Deussen, A., Karl, M. O., Wielockx, B., Chavakis, T., Klotzsche-von Ameln, A. Hematopoietic hypoxia-inducible factor 2α deficiency ameliorates pathological retinal neovascularization via modulation of endothelial cell apoptosis.
Collapse
Affiliation(s)
- Irina Korovina
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ales Neuwirth
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - David Sprott
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Silvio Weber
- Institute of Pharmacology and Toxicology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sheik Pran Babu Sardar Pasha
- Deutsche Forschungsgemeinschaft (DFG) Center for Regenerative Therapies, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Bettina Gercken
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Georg Breier
- Medical Biology, Department of Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ali El-Armouche
- Institute of Pharmacology and Toxicology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Andreas Deussen
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Mike O Karl
- Deutsche Forschungsgemeinschaft (DFG) Center for Regenerative Therapies, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany.,German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Deutsche Forschungsgemeinschaft (DFG) Center for Regenerative Therapies, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Anne Klotzsche-von Ameln
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
15
|
Xu D, Liao C, Zhang B, Tolbert WD, He W, Dai Z, Zhang W, Yuan W, Pazgier M, Liu J, Yu J, Sansonetti PJ, Bevins CL, Shao Y, Lu W. Human Enteric α-Defensin 5 Promotes Shigella Infection by Enhancing Bacterial Adhesion and Invasion. Immunity 2018; 48:1233-1244.e6. [PMID: 29858013 PMCID: PMC6051418 DOI: 10.1016/j.immuni.2018.04.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 02/18/2018] [Accepted: 04/13/2018] [Indexed: 01/07/2023]
Abstract
Shigella is a Gram-negative bacterium that causes bacillary dysentery worldwide. It invades the intestinal epithelium to elicit intense inflammation and tissue damage, yet the underlying mechanisms of its host selectivity and low infectious inoculum remain perplexing. Here, we report that Shigella co-opts human α-defensin 5 (HD5), a host defense peptide important for intestinal homeostasis and innate immunity, to enhance its adhesion to and invasion of mucosal tissues. HD5 promoted Shigella infection in vitro in a structure-dependent manner. Shigella, commonly devoid of an effective host-adhesion apparatus, preferentially targeted HD5 to augment its ability to colonize the intestinal epithelium through interactions with multiple bacterial membrane proteins. HD5 exacerbated infectivity and Shigella-induced pathology in a culture of human colorectal tissues and three animal models. Our findings illuminate how Shigella exploits innate immunity by turning HD5 into a virulence factor for infection, unveiling a mechanism of action for this highly proficient human pathogen.
Collapse
Affiliation(s)
- Dan Xu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China,Center for Translational Medicine, Frontier Institute of Science and Technology, Xi’an Jiaotong University,Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Chongbing Liao
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China,Center for Translational Medicine, Frontier Institute of Science and Technology, Xi’an Jiaotong University
| | - Bing Zhang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - W. David Tolbert
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Wangxiao He
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China,Center for Translational Medicine, Frontier Institute of Science and Technology, Xi’an Jiaotong University,Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Zhijun Dai
- The Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine
| | - Wei Zhang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Weirong Yuan
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Marzena Pazgier
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jiankang Liu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Jun Yu
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | | | - Charles L. Bevins
- Department of Microbiology and Immunology, University of California, School of Medicine, Davis, California, USA
| | - Yongping Shao
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China,Center for Translational Medicine, Frontier Institute of Science and Technology, Xi’an Jiaotong University,Correspondence to: (lead contact) or
| | - Wuyuan Lu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China,Center for Translational Medicine, Frontier Institute of Science and Technology, Xi’an Jiaotong University,Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA,Correspondence to: (lead contact) or
| |
Collapse
|
16
|
Konovalova MV, Zubareva AA, Lutsenko GV, Svirshchevskaya EV. Antimicrobial Peptides in Health and Disease (Review). APPL BIOCHEM MICRO+ 2018. [DOI: 10.1134/s0003683818030079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
Howell K, de Leeuw E. Cell adhesion properties of human defensins. Biochem Biophys Res Commun 2018; 502:238-242. [PMID: 29800568 DOI: 10.1016/j.bbrc.2018.05.150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/20/2018] [Indexed: 10/16/2022]
Abstract
Effector peptides of innate immunity play an important role in host defense. They act directly by inactivating microbes but also link innate to adaptive immunity. A variety of innate immune functions has been described for these peptides, including chemoattraction and cytokine release. In this study, we describe the effect on cell morphology and cell adhesion of human defensins. We find that Human Defensin 5, the major product of specialized gut epithelial cells, causes changes in cell morphology. HD-5 induces cell adhesion, binds to fibronectin and facilitates binding of T cells to intestinal epithelial cells. These effects were found also for a second prominent defensing, termed Human Neutrophil peptide-1, but not for other human defensins.
Collapse
Affiliation(s)
- Katie Howell
- Integrated Biotherapeutics, Inc., Rockville, MD 20850, USA
| | - Erik de Leeuw
- Institute of Human Virology of the University of Maryland Baltimore School of Medicine, 725 West Lombard Street, Baltimore, MD 21201, USA.
| |
Collapse
|
18
|
A Critical Analysis of the Available In Vitro and Ex Vivo Methods to Study Retinal Angiogenesis. J Ophthalmol 2017; 2017:3034953. [PMID: 28848677 PMCID: PMC5564124 DOI: 10.1155/2017/3034953] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/20/2017] [Accepted: 06/28/2017] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis is a biological process with a central role in retinal diseases. The choice of the ideal method to study angiogenesis, particularly in the retina, remains a problem. Angiogenesis can be assessed through in vitro and in vivo studies. In spite of inherent limitations, in vitro studies are faster, easier to perform and quantify, and typically less expensive and allow the study of isolated angiogenesis steps. We performed a systematic review of PubMed searching for original articles that applied in vitro or ex vivo angiogenic retinal assays until May 2017, presenting the available assays and discussing their applicability, advantages, and disadvantages. Most of the studies evaluated migration, proliferation, and tube formation of endothelial cells in response to inhibitory or stimulatory compounds. Other aspects of angiogenesis were studied by assessing cell permeability, adhesion, or apoptosis, as well as by implementing organotypic models of the retina. Emphasis is placed on how the methods are applied and how they can contribute to retinal angiogenesis comprehension. We also discuss how to choose the best cell culture to implement these methods. When applied together, in vitro and ex vivo studies constitute a powerful tool to improve retinal angiogenesis knowledge. This review provides support for researchers to better select the most suitable protocols in this field.
Collapse
|
19
|
Klotzsche-von Ameln A, Cremer S, Hoffmann J, Schuster P, Khedr S, Korovina I, Troullinaki M, Neuwirth A, Sprott D, Chatzigeorgiou A, Economopoulou M, Orlandi A, Hain A, Zeiher AM, Deussen A, Hajishengallis G, Dimmeler S, Chavakis T, Chavakis E. Endogenous developmental endothelial locus-1 limits ischaemia-related angiogenesis by blocking inflammation. Thromb Haemost 2017; 117:1150-1163. [PMID: 28447099 DOI: 10.1160/th16-05-0354] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 03/05/2017] [Indexed: 12/23/2022]
Abstract
We have recently identified endothelial cell-secreted developmental endothelial locus-1 (Del-1) as an endogenous inhibitor of β2-integrin-dependent leukocyte infiltration. Del-1 was previously also implicated in angiogenesis. Here, we addressed the role of endogenously produced Del-1 in ischaemia-related angiogenesis. Intriguingly, Del-1-deficient mice displayed increased neovascularisation in two independent ischaemic models (retinopathy of prematurity and hind-limb ischaemia), as compared to Del-1-proficient mice. On the contrary, angiogenic sprouting in vitro or ex vivo (aortic ring assay) and physiological developmental retina angiogenesis were not affected by Del-1 deficiency. Mechanistically, the enhanced ischaemic neovascularisation in Del-1-deficiency was linked to higher infiltration of the ischaemic tissue by CD45+ haematopoietic and immune cells. Moreover, Del-1-deficiency promoted β2-integrin-dependent adhesion of haematopoietic cells to endothelial cells in vitro, and the homing of hematopoietic progenitor cells and of immune cell populations to ischaemic muscles in vivo. Consistently, the increased hind limb ischaemia-related angiogenesis in Del-1 deficiency was completely reversed in mice lacking both Del-1 and the β2-integrin LFA-1. Additionally, enhanced retinopathy-associated neovascularisation in Del-1-deficient mice was reversed by LFA-1 blockade. Our data reveal a hitherto unrecognised function of endogenous Del-1 as a local inhibitor of ischaemia-induced angiogenesis by restraining LFA-1-dependent homing of pro-angiogenic haematopoietic cells to ischaemic tissues. Our findings are relevant for the optimisation of therapeutic approaches in the context of ischaemic diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Emmanouil Chavakis
- Emmanouil Chavakis, MD, Dept. of Internal Medicine III, Goethe University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany, Tel.: +49 69 6301 4131, +49 69 6301 87965, Fax: +49 69 6301 83462, E-mail:
| |
Collapse
|
20
|
Niyonsaba F, Kiatsurayanon C, Chieosilapatham P, Ogawa H. Friends or Foes? Host defense (antimicrobial) peptides and proteins in human skin diseases. Exp Dermatol 2017; 26:989-998. [PMID: 28191680 DOI: 10.1111/exd.13314] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2017] [Indexed: 12/14/2022]
Abstract
Host defense peptides/proteins (HDPs), also known as antimicrobial peptides/proteins (AMPs), are key molecules in the cutaneous innate immune system. AMPs/HDPs historically exhibit broad-spectrum killing activity against bacteria, enveloped viruses, fungi and several parasites. Recently, AMPs/HDPs were shown to have important biological functions, including inducing cell proliferation, migration and differentiation; regulating inflammatory responses; controlling the production of various cytokines/chemokines; promoting wound healing; and improving skin barrier function. Despite the fact that AMPs/HDPs protect our body, several studies have hypothesized that these molecules actively contribute to the pathogenesis of various skin diseases. For example, AMPs/HDPs play crucial roles in the pathological processes of psoriasis, atopic dermatitis, rosacea, acne vulgaris, systemic lupus erythematosus and systemic sclerosis. Thus, AMPs/HDPs may be a double-edged sword, promoting cutaneous immunity while simultaneously initiating the pathogenesis of some skin disorders. This review will describe the most common skin-derived AMPs/HDPs (defensins, cathelicidins, S100 proteins, ribonucleases and dermcidin) and discuss the biology and both the positive and negative aspects of these AMPs/HDPs in skin inflammatory/infectious diseases. Understanding the regulation, functions and mechanisms of AMPs/HDPs may offer new therapeutic opportunities in the treatment of various skin disorders.
Collapse
Affiliation(s)
- François Niyonsaba
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Faculty of International Liberal Arts, Juntendo University, Tokyo, Japan
| | - Chanisa Kiatsurayanon
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Medical Services, Institute of Dermatology, Ministry of Public Health, Bangkok, Thailand
| | - Panjit Chieosilapatham
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hideoki Ogawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
21
|
Shafee TMA, Lay FT, Phan TK, Anderson MA, Hulett MD. Convergent evolution of defensin sequence, structure and function. Cell Mol Life Sci 2017; 74:663-682. [PMID: 27557668 PMCID: PMC11107677 DOI: 10.1007/s00018-016-2344-5] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/27/2016] [Accepted: 08/15/2016] [Indexed: 02/06/2023]
Abstract
Defensins are a well-characterised group of small, disulphide-rich, cationic peptides that are produced by essentially all eukaryotes and are highly diverse in their sequences and structures. Most display broad range antimicrobial activity at low micromolar concentrations, whereas others have other diverse roles, including cell signalling (e.g. immune cell recruitment, self/non-self-recognition), ion channel perturbation, toxic functions, and enzyme inhibition. The defensins consist of two superfamilies, each derived from an independent evolutionary origin, which have subsequently undergone extensive divergent evolution in their sequence, structure and function. Referred to as the cis- and trans-defensin superfamilies, they are classified based on their secondary structure orientation, cysteine motifs and disulphide bond connectivities, tertiary structure similarities and precursor gene sequence. The utility of displaying loops on a stable, compact, disulphide-rich core has been exploited by evolution on multiple occasions. The defensin superfamilies represent a case where the ensuing convergent evolution of sequence, structure and function has been particularly extreme. Here, we discuss the extent, causes and significance of these convergent features, drawing examples from across the eukaryotes.
Collapse
Affiliation(s)
- Thomas M A Shafee
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Fung T Lay
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Thanh Kha Phan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Marilyn A Anderson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
| | - Mark D Hulett
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
22
|
Paulin N, Döring Y, Kooijman S, Blanchet X, Viola JR, de Jong R, Mandl M, Hendrikse J, Schiener M, von Hundelshausen P, Vogt A, Weber C, Bdeir K, Hofmann SM, Rensen PCN, Drechsler M, Soehnlein O. Human Neutrophil Peptide 1 Limits Hypercholesterolemia-induced Atherosclerosis by Increasing Hepatic LDL Clearance. EBioMedicine 2017; 16:204-211. [PMID: 28111237 PMCID: PMC5474437 DOI: 10.1016/j.ebiom.2017.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 01/05/2017] [Accepted: 01/05/2017] [Indexed: 11/20/2022] Open
Abstract
Increases in plasma LDL-cholesterol have unequivocally been established as a causal risk factor for atherosclerosis. Hence, strategies for lowering of LDL-cholesterol may have immediate therapeutic relevance. Here we study the role of human neutrophil peptide 1 (HNP1) in a mouse model of atherosclerosis and identify its potent atheroprotective effect both upon transgenic overexpression and therapeutic delivery. The effect was found to be due to a reduction of plasma LDL-cholesterol. Mechanistically, HNP1 binds to apolipoproteins enriched in LDL. This interaction facilitates clearance of LDL particles in the liver via LDL receptor. Thus, we here identify a non-redundant mechanism by which HNP1 allows for reduction of LDL-cholesterol, a process that may be therapeutically instructed to lower cardiovascular risk. Mice with transgenic expression of human neutrophil peptide 1 (HNP1) exhibit lower plasma VLDL/LDL levels and smaller atherosclerotic lesion sizes. Repetitive HNP1 delivery is atheroprotective by reducing hypercholesterolemia. HNP1 binds to apolipoproteins in LDL and facilitates LDL clearance in the liver involving LDL receptor.
Increased plasma lipid levels (i.e. hypercholesterolemia) are a primary risk factor for atherosclerosis, the pathology underlying myocardial infarction and stroke. Here we show that human neutrophil peptide 1 (HNP1, also known as α-defensin), an antimicrobial protein typically released from activated neutrophils, binds to apolipoproteins within plasma lipoproteins and facilitates the clearance of plasma lipids in the liver. As a consequence, repeated injection of hypercholesterolemic mice with HNP1 reduces atherosclerotic lesion formation. Thus, this study provides an innovative strategy to reduce hypercholesterolemia and hence a way to potentially reduce cardiovascular risk.
Collapse
MESH Headings
- Animals
- Apolipoproteins/blood
- Apolipoproteins/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/prevention & control
- Cholesterol, LDL/blood
- Cholesterol, LDL/metabolism
- Female
- Hep G2 Cells
- Humans
- Hypercholesterolemia/genetics
- Hypercholesterolemia/metabolism
- Hypercholesterolemia/prevention & control
- Immunohistochemistry
- Lipoproteins, LDL/blood
- Lipoproteins, LDL/metabolism
- Lipoproteins, LDL/pharmacokinetics
- Liver/drug effects
- Liver/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Microscopy, Confocal
- Protein Binding
- RNA Interference
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- alpha-Defensins/administration & dosage
- alpha-Defensins/genetics
- alpha-Defensins/metabolism
Collapse
Affiliation(s)
- Nicole Paulin
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich 80336, Germany
| | - Yvonne Döring
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich 80336, Germany
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Einthoven Laboratory for Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Xavier Blanchet
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich 80336, Germany
| | - Joana R Viola
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich 80336, Germany; Department of Pathology, AMC, 1105 AZ Amsterdam, The Netherlands
| | - Renske de Jong
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich 80336, Germany; Department of Pathology, AMC, 1105 AZ Amsterdam, The Netherlands
| | - Manuela Mandl
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich 80336, Germany
| | - Jeffrey Hendrikse
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich 80336, Germany; Department of Pathology, AMC, 1105 AZ Amsterdam, The Netherlands
| | - Maximilian Schiener
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich 80336, Germany
| | | | - Anja Vogt
- Medizinische Klinik und Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich 80336, Germany; DZHK, Partner Site Munich Heart Alliance, Munich 80336, Germany
| | - Khalil Bdeir
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Susanna M Hofmann
- Medizinische Klinik und Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany; Institute for Diabetes and Regeneration, Helmholtz Center Munich, Germany; German Center for Diabetes Research (DZD) München-Neuherberg, Germany
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Einthoven Laboratory for Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Maik Drechsler
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich 80336, Germany; Department of Pathology, AMC, 1105 AZ Amsterdam, The Netherlands; DZHK, Partner Site Munich Heart Alliance, Munich 80336, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich 80336, Germany; Department of Pathology, AMC, 1105 AZ Amsterdam, The Netherlands; DZHK, Partner Site Munich Heart Alliance, Munich 80336, Germany.
| |
Collapse
|
23
|
|
24
|
Immune and regulatory functions of neutrophils in inflammatory bone loss. Semin Immunol 2016; 28:146-58. [PMID: 26936034 DOI: 10.1016/j.smim.2016.02.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 02/06/2016] [Accepted: 02/14/2016] [Indexed: 02/06/2023]
Abstract
Although historically viewed as merely anti-microbial effectors in acute infection or injury, neutrophils are now appreciated to be functionally versatile with critical roles also in chronic inflammation. Periodontitis, a chronic inflammatory disease that destroys the tooth-supporting gums and bone, is particularly affected by alterations in neutrophil numbers or function, as revealed by observations in monogenic disorders and relevant mouse models. Besides being a significant debilitating disease and health burden in its own right, periodontitis is thus an attractive model to dissect uncharted neutrophil-associated (patho)physiological pathways. Here, we summarize recent evidence that neutrophils can contribute to inflammatory bone loss not only through the typical bystander injury dogma but intriguingly also through their absence from the affected tissue, where they normally perform important immunomodulatory functions. Moreover, we discuss recent advances in the interactions of neutrophils with the vascular endothelium and - upon extravasation - with bacteria, and how the dysregulation of these interactions leads to inflammatory tissue damage. Overall, neutrophils have both protective and destructive roles in periodontitis, as they are involved in both the maintenance of periodontal tissue homeostasis and the induction of inflammatory bone loss. This highlights the importance of developing approaches that promote or sustain a fine balance between homeostatic immunity and inflammatory pathology.
Collapse
|
25
|
Economopoulou M, Avramovic N, Klotzsche-von Ameln A, Korovina I, Sprott D, Samus M, Gercken B, Troullinaki M, Grossklaus S, Funk RH, Li X, Imhof BA, Orlova VV, Chavakis T. Endothelial-specific deficiency of Junctional Adhesion Molecule-C promotes vessel normalisation in proliferative retinopathy. Thromb Haemost 2015; 114:1241-9. [PMID: 26311310 DOI: 10.1160/th15-01-0051] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 07/09/2015] [Indexed: 12/29/2022]
Abstract
In proliferative retinopathies, like proliferative diabetic retinopathy and retinopathy of prematurity (ROP), the hypoxia response is sustained by the failure of the retina to revascularise its ischaemic areas. Non-resolving retina ischaemia/hypoxia results in upregulation of pro-angiogenic factors and pathologic neovascularisation with ectopic, fragile neovessels. Promoting revascularisation of the retinal avascular area could interfere with this vicious cycle and lead to vessel normalisation. Here, we examined the function of endothelial junctional adhesion molecule-C (JAM-C) in the context of ROP. Endothelial-specific JAM-C-deficient (EC-JAM-C KO) mice and littermate JAM-C-proficient (EC-JAM-C WT) mice were subjected to the ROP model. An increase in total retinal vascularisation was found at p17 owing to endothelial JAM-C deficiency, which was the result of enhanced revascularisation and vessel normalisation, thereby leading to significantly reduced avascular area in EC-JAM-C KO mice. In contrast, pathologic neovessel formation was not affected by endothelial JAM-C deficiency. Consistent with improved vessel normalisation, tip cell formation at the interface between vascular and avascular area was higher in EC-JAM-C KO mice, as compared to their littermate controls. Consistently, JAM-C inactivation in endothelial cells resulted in increased spreading on fibronectin and enhanced sprouting in vitro in a manner dependent on β1-integrin and on the activation of the small GTPase RAP1. Together, endothelial deletion of JAM-C promoted endothelial cell sprouting, and consequently vessel normalisation and revascularisation of the hypoxic retina without altering pathologic neovascularisation. Thus, targeting endothelial JAM-C may provide a novel therapeutic strategy for promoting revascularisation and vessel normalisation in the treatment of proliferative retinopathies.
Collapse
Affiliation(s)
- Matina Economopoulou
- Matina Economopoulou, Department of Ophthalmology, Univ. Hospital Carl Gustav Carus, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany, Tel.: +49 351 45819291, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Defensins: “Simple” antimicrobial peptides or broad-spectrum molecules? Cytokine Growth Factor Rev 2015; 26:361-70. [DOI: 10.1016/j.cytogfr.2014.12.005] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 12/17/2014] [Indexed: 11/19/2022]
|
27
|
Smith J, Liu F, Beyer B, Morales K, Reilly A, Cole R, Herron BJ. Angiogenesis QTL on Mouse Chromosome 8 Colocalizes with Differential β-Defensin Expression. J Biomol Tech 2015; 26:45-53. [PMID: 25802489 DOI: 10.7171/jbt.15-2602-002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Identification of genetic factors that modify complex traits is often complicated by gene-environment interactions that contribute to the observed phenotype. In model systems, the phenotypic outcomes quantified are typically traits that maximize observed variance, which in turn, should maximize the detection of quantitative trait loci (QTL) in subsequent mapping studies. However, when the observed trait is dependent on multiple interacting factors, it can complicate genetic analysis, reducing the likelihood that the modifying mutation will ultimately be found. Alternatively, by focusing on intermediate phenotypes of a larger condition, we can reduce a model's complexity, which will, in turn, limit the number of QTL that contribute to variance. We used a novel method to follow angiogenesis in mice that reduces environmental variance by measuring endothelial cell growth from culture of isolated skin biopsies that varies depending on the genetic source of the tissue. This method, in combination with a backcross breeding strategy, is intended to reduce genetic complexity and limit the phenotypic effects to fewer modifier loci. We determined that our approach was an efficient means to generate recombinant progeny and used this cohort to map a novel s.c. angiogenesis QTL to proximal mouse chromosome (Chr.) 8 with suggestive QTL on Chr. 2 and 7. Global mRNA expression analysis of samples from parental reference strains revealed β-defensins as potential candidate genes for future study.
Collapse
Affiliation(s)
- Jason Smith
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Fang Liu
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Barbara Beyer
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Krista Morales
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Andrew Reilly
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Richard Cole
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Bruce J Herron
- 1 Wadsworth Center, New York State Department of Health, Albany, New York, USA; and 2 Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| |
Collapse
|
28
|
Abstract
Ocular neovascularization can affect almost all the tissues of the eye: the cornea, the iris, the retina, and the choroid. Pathological neovascularization is the underlying cause of vision loss in common ocular conditions such as diabetic retinopathy, retinopathy of prematurity and age-related macular neovascularization. Glycosylation is the most common covalent posttranslational modification of proteins in mammalian cells. A growing body of evidence demonstrates that glycosylation influences the process of angiogenesis and impacts activation, proliferation, and migration of endothelial cells as well as the interaction of angiogenic endothelial cells with other cell types necessary to form blood vessels. Recent studies have provided evidence that members of the galectin class of β-galactoside-binding proteins modulate angiogenesis by novel carbohydrate-based recognition systems involving interactions between glycans of angiogenic cell surface receptors and galectins. This review discusses the significance of glycosylation and the role of galectins in the pathogenesis of ocular neovascularization.
Collapse
Affiliation(s)
- Anna I Markowska
- Departments of Ophthalmology and Developmental, Molecular & Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA Ymir Genomics LLC, Cambridge, MA 02139, USA
| | - Zhiyi Cao
- Departments of Ophthalmology and Developmental, Molecular & Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA New England Eye Center, Boston, MA 02111, USA
| | - Noorjahan Panjwani
- Departments of Ophthalmology and Developmental, Molecular & Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA New England Eye Center, Boston, MA 02111, USA
| |
Collapse
|
29
|
Basavarajappa HD, Lee B, Fei X, Lim D, Callaghan B, Mund JA, Case J, Rajashekhar G, Seo SY, Corson TW. Synthesis and mechanistic studies of a novel homoisoflavanone inhibitor of endothelial cell growth. PLoS One 2014; 9:e95694. [PMID: 24752613 PMCID: PMC3994091 DOI: 10.1371/journal.pone.0095694] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 03/30/2014] [Indexed: 12/13/2022] Open
Abstract
Preventing pathological ocular angiogenesis is key to treating retinopathy of prematurity, diabetic retinopathy and age-related macular degeneration. At present there is no small molecule drug on the market to target this process and hence there is a pressing need for developing novel small molecules that can replace or complement the present surgical and biologic therapies for these neovascular eye diseases. Previously, an antiangiogenic homoisoflavanone was isolated from the bulb of a medicinal orchid, Cremastra appendiculata. In this study, we present the synthesis of a novel homoisoflavanone isomer of this compound. Our compound, SH-11052, has antiproliferative activity against human umbilical vein endothelial cells, and also against more ocular disease-relevant human retinal microvascular endothelial cells (HRECs). Tube formation and cell cycle progression of HRECs were inhibited by SH-11052, but the compound did not induce apoptosis at effective concentrations. SH-11052 also decreased TNF-α induced p38 MAPK phosphorylation in these cells. Intriguingly, SH-11052 blocked TNF-α induced IκB-α degradation, and therefore decreased NF-κB nuclear translocation. It decreased the expression of NF-κB target genes and the pro-angiogenic or pro-inflammatory markers VCAM-1, CCL2, IL8, and PTGS2. In addition SH-11052 inhibited VEGF induced activation of Akt but not VEGF receptor autophosphorylation. Based on these results we propose that SH-11052 inhibits inflammation induced angiogenesis by blocking both TNF-α and VEGF mediated pathways, two major pathways involved in pathological angiogenesis. Synthesis of this novel homoisoflavanone opens the door to structure-activity relationship studies of this class of compound and further evaluation of its mechanism and potential to complement existing antiangiogenic drugs.
Collapse
Affiliation(s)
- Halesha D. Basavarajappa
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Bit Lee
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Xiang Fei
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Daesung Lim
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Breedge Callaghan
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Julie A. Mund
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States Of America
| | - Jamie Case
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States Of America
| | - Gangaraju Rajashekhar
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Seung-Yong Seo
- College of Pharmacy, Gachon University, Incheon, South Korea
- * E-mail: (S-YS); (TWC)
| | - Timothy W. Corson
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States Of America
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail: (S-YS); (TWC)
| |
Collapse
|
30
|
LI DAN, QIN QING, WANG XIAOYI, SHI HUASHAN, LUO MIN, GUO FUCHUN, WANG WEI, WANG YONGSHENG. Intratumoral expression of mature human neutrophil peptide-1 potentiates the therapeutic effect of doxorubicin in a mouse 4T1 breast cancer model. Oncol Rep 2013; 31:1287-95. [DOI: 10.3892/or.2013.2947] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 10/10/2013] [Indexed: 11/06/2022] Open
|
31
|
Schaal JB, Tran D, Tran P, Ösapay G, Trinh K, Roberts KD, Brasky KM, Tongaonkar P, Ouellette AJ, Selsted ME. Rhesus macaque theta defensins suppress inflammatory cytokines and enhance survival in mouse models of bacteremic sepsis. PLoS One 2012; 7:e51337. [PMID: 23236475 PMCID: PMC3516535 DOI: 10.1371/journal.pone.0051337] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 11/07/2012] [Indexed: 02/08/2023] Open
Abstract
Theta-defensins (θ-defensins) are macrocyclic antimicrobial peptides expressed in leukocytes of Old World monkeys. The peptides are broad spectrum microbicides in vitro and numerous θ-defensin isoforms have been identified in granulocytes of rhesus macaques and Olive baboons. Several mammalian α- and β-defensins, genetically related to θ-defensins, have proinflammatory and immune-activating properties that bridge innate and acquired immunity. In the current study we analyzed the immunoregulatory properties of rhesus θ-defensins 1–5 (RTDs 1–5). RTD-1, the most abundant θ-defensin in macaques, reduced the levels of TNF, IL-1α, IL-1β, IL-6, and IL-8 secreted by blood leukocytes stimulated by several TLR agonists. RTDs 1–5 suppressed levels of soluble TNF released by bacteria- or LPS-stimulated blood leukocytes and THP-1 monocytes. Despite their highly conserved conformation and amino acid sequences, the anti-TNF activities of RTDs 1–5 varied by as much as 10-fold. Systemically administered RTD-1 was non-toxic for BALB/c mice, and escalating intravenous doses were well tolerated and non-immunogenic in adult chimpanzees. The peptide was highly stable in serum and plasma. Single dose administration of RTD-1 at 5 mg/kg significantly improved survival of BALB/c mice with E. coli peritonitis and cecal ligation-and-puncture induced polymicrobial sepsis. Peptide treatment reduced serum levels of several inflammatory cytokines/chemokines in bacteremic animals. Collectively, these results indicate that the anti-inflammatory properties of θ-defensins in vitro and in vivo are mediated by the suppression of numerous proinflammatory cytokines and blockade of TNF release may be a primary effect.
Collapse
Affiliation(s)
- Justin B. Schaal
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Dat Tran
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Patti Tran
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - George Ösapay
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, California, United States of America
| | - Katie Trinh
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Kevin D. Roberts
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Kathleen M. Brasky
- Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Prasad Tongaonkar
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - André J. Ouellette
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Kenneth Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Michael E. Selsted
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Kenneth Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
32
|
Chan JK, Roth J, Oppenheim JJ, Tracey KJ, Vogl T, Feldmann M, Horwood N, Nanchahal J. Alarmins: awaiting a clinical response. J Clin Invest 2012; 122:2711-9. [PMID: 22850880 DOI: 10.1172/jci62423] [Citation(s) in RCA: 355] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Alarmins are endogenous molecules that are constitutively available and released upon tissue damage and activate the immune system. Current evidence indicates that uncontrolled and excessive release of alarmins contributes to the dysregulated processes seen in many inflammatory and autoimmune conditions, as well as tumorigenesis and cancer spread. Conversely, alarmins have also been found to play a major role in the orchestration of tissue homeostasis, including repair and remodeling in the heart, skin, and nervous system. Here, we provide an update and overview on alarmins, highlighting the areas that may benefit from this clinical translation.
Collapse
Affiliation(s)
- James K Chan
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
High fidelity processing and activation of the human α-defensin HNP1 precursor by neutrophil elastase and proteinase 3. PLoS One 2012; 7:e32469. [PMID: 22448222 PMCID: PMC3308943 DOI: 10.1371/journal.pone.0032469] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 01/31/2012] [Indexed: 01/28/2023] Open
Abstract
The azurophilic granules of human neutrophils contain four α-defensins called human neutrophil peptides (HNPs 1–4). HNPs are tridisulfide-linked antimicrobial peptides involved in the intracellular killing of organisms phagocytosed by neutrophils. The peptides are produced as inactive precursors (proHNPs) which are processed to active microbicides by as yet unidentified convertases. ProHNP1 was expressed in E. coli and the affinity-purified propeptide isolated as two species, one containing mature HNP1 sequence with native disulfide linkages (“folded proHNP1”) and the other containing non-native disulfide linked proHNP1 conformers (misfolded proHNP1). Native HNP1, liberated by CNBr treatment of folded proHNP1, was microbicidal against Staphylococcus aureus, but the peptide derived from misfolded proHNP1 was inactive. We hypothesized that neutrophil elastase (NE), proteinase 3 (PR3) or cathepsin G (CG), serine proteases that co-localize with HNPs in azurophil granules, are proHNP1 activating convertases. Folded proHNP1 was converted to mature HNP1 by both NE and PR3, but CG generated an HNP1 variant with an N-terminal dipeptide extension. NE and PR3 cleaved folded proHNP1 to produce a peptide indistinguishable from native HNP1 purified from neutrophils, and the microbicidal activities of in vitro derived and natural HNP1 peptides were equivalent. In contrast, misfolded proHNP1 conformers were degraded extensively under the same conditions. Thus, NE and PR3 possess proHNP1 convertase activity that requires the presence of the native HNP1 disulfide motif for high fidelity activation of the precursor in vitro.
Collapse
|
34
|
Al-Benna S, Shai Y, Jacobsen F, Steinstraesser L. Oncolytic activities of host defense peptides. Int J Mol Sci 2011; 12:8027-51. [PMID: 22174648 PMCID: PMC3233454 DOI: 10.3390/ijms12118027] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 11/07/2011] [Indexed: 12/16/2022] Open
Abstract
Cancer continues to be a leading source of morbidity and mortality worldwide in spite of progress in oncolytic therapies. In addition, the incidence of cancers affecting the breast, kidney, prostate and skin among others continue to rise. Chemotherapeutic drugs are widely used in cancer treatment but have the serious drawback of nonspecific toxicity because these agents target any rapidly dividing cell without discriminating between healthy and malignant cells. In addition, many neoplasms eventually become resistant to conventional chemotherapy due to selection for multidrug-resistant variants. The limitations associated with existing chemotherapeutic drugs have stimulated the search for new oncolytic therapies. Host defense peptides (HDPs) may represent a novel family of oncolytic agents that can avoid the shortcomings of conventional chemotherapy because they exhibit selective cytotoxicity against a broad spectrum of malignant human cells, including multi-drug-resistant neoplastic cells. Oncolytic activity by HDPs is usually via necrosis due to cell membrane lysis, but some HDPs can trigger apoptosis in cancer cells via mitochondrial membrane disruption. In addition, certain HDPs are anti-angiogenic which may inhibit cancer progression. This paper reviews oncolytic HDP studies in order to address the suitability of selected HDPs as oncolytic therapies.
Collapse
Affiliation(s)
- Sammy Al-Benna
- Laboratory for Molecular Oncology and Wound Healing, Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum 44789, Germany; E-Mails: (S.A.-B.); (F.J.)
| | - Yechiel Shai
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot 76100, Israel; E-Mail:
| | - Frank Jacobsen
- Laboratory for Molecular Oncology and Wound Healing, Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum 44789, Germany; E-Mails: (S.A.-B.); (F.J.)
| | - Lars Steinstraesser
- Laboratory for Molecular Oncology and Wound Healing, Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum 44789, Germany; E-Mails: (S.A.-B.); (F.J.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +49-234-302-3442; Fax: +49-234-302-6379
| |
Collapse
|
35
|
Gold nanoparticles inhibit vascular endothelial growth factor-induced angiogenesis and vascular permeability via Src dependent pathway in retinal endothelial cells. Angiogenesis 2011; 14:29-45. [PMID: 21061058 DOI: 10.1007/s10456-010-9193-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Accepted: 10/25/2010] [Indexed: 02/06/2023]
Abstract
The purpose of this study was to investigate the effect of gold nanoparticles on the signaling cascade related to angiogenesis and vascular permeability induced by Vascular Endothelial Growth Factor (VEGF) in Bovine retinal endothelial cells (BRECs). The effect of VEGF and gold nanoparticles on cell viability, migration and tubule formation was assessed. PP2 (Src Tyrosine Kinase inhibitor) was used as the positive control and the inhibitor assay was performed to compare the effect of AuNPs on VEGF induced angiogenesis. The transient transfection assay was performed to study the VEGFR2/Src activity during experimental conditions and was confirmed using western blot analysis. Treatment of BRECs with VEGF significantly increased the cell proliferation, migration and tube formation. Furthermore, gold nanoparticles (500 nM) significantly inhibited the proliferation, migration and tube formation, in the presence of VEGF in BRECs. The gold nanoparticles also inhibited VEGF induced Src phosphorylation through which their mode of action in inhibiting angiogenic pathways is revealed. The fate of the gold nanoparticles within the cells is being analyzed using the TEM images obtained. The potential of AuNPs to inhibit the VEGF165-induced VEGFR-2 phosphorylation is also being confirmed through the receptor assay which elucidates one of the possible mechanism by which AuNPs inhibit VEGF induced angiogenesis. These results indicate that gold nanoparticles can block VEGF activation of important signaling pathways, specifically Src in BRECs and hence modulation of these pathways may contribute to gold nanoparticles ability to block VEGF-induced retinal neovascularization.
Collapse
|
36
|
Motegi SI, Leitner WW, Lu M, Tada Y, Sárdy M, Wu C, Chavakis T, Udey MC. Pericyte-derived MFG-E8 regulates pathologic angiogenesis. Arterioscler Thromb Vasc Biol 2011; 31:2024-34. [PMID: 21737783 DOI: 10.1161/atvbaha.111.232587] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE MFG-E8 (also called lactadherin and SED1) is a secreted glycoprotein that has been previously implicated in enhancement of vascular endothelial growth factor-dependent angiogenesis. Major sources of MFG-E8 in vivo and precise mechanisms of MFG-E8 action remain undetermined. The objective of this study was to identify important sources of MFG-E8 in vivo and further elucidate the role(s) of MFG-E8 in the regulation of angiogenesis. METHODS AND RESULTS We used knockout mice and anti-MFG-E8 antibodies to study MFG-E8 function in vivo. In melanomas and in retinas of mice with oxygen-induced retinopathy, MFG-E8 colocalized with pericytes rather than endothelial cells, and platelet-derived growth factor receptor β+ pericytes/pericyte precursors purified from tumors contained large amounts of MFG-E8 mRNA. Tumor- and retinopathy-associated angiogenesis was diminished in MFG-E8 knockout mice, and pericyte coverage of neovessels was reduced. Inhibition of MFG-E8 production by 10T1/2 cells (surrogate pericyte/pericyte precursors) using small interfering RNAs and short hairpin RNAs, or inhibition of MFG-E8 action with some anti-MFG-E8 antibodies, selectively attenuated migration in vitro. Significantly, the anti-MFG-E8 antibodies that inhibited 10T1/2 cell migration in vitro also inhibited pathological angiogenesis in vivo. CONCLUSIONS These studies strongly implicate MFG-E8 in pericyte/pericyte precursor function and indicate that MFG-E8-directed therapeutics may merit further development.
Collapse
Affiliation(s)
- Sei-ichiro Motegi
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20802-1908, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Kolar SS, McDermott AM. Role of host-defence peptides in eye diseases. Cell Mol Life Sci 2011; 68:2201-13. [PMID: 21584809 PMCID: PMC3637883 DOI: 10.1007/s00018-011-0713-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 04/26/2011] [Accepted: 04/26/2011] [Indexed: 10/18/2022]
Abstract
The eye and its associated tissues including the lacrimal system and lids have evolved several defence mechanisms to prevent microbial invasion. Included among this armory are several host-defence peptides. These multifunctional molecules are being studied not only for their endogenous antimicrobial properties but also for their potential therapeutic effects. Here the current knowledge of host-defence peptide expression in the eye will be summarised. The role of these peptides in eye disease will be discussed with the primary focus being on infectious keratitis, inflammatory conditions including dry eye and wound healing. Finally the potential of using host-defence peptides and their mimetics/derivatives for the treatment and prevention of eye diseases is addressed.
Collapse
Affiliation(s)
- Satya S. Kolar
- College of Optometry, University of Houston, 4901 Calhoun Road, 505 J Davis Armistead Bldg, Houston, TX 77204-2020 USA
| | - Alison M. McDermott
- College of Optometry, University of Houston, 4901 Calhoun Road, 505 J Davis Armistead Bldg, Houston, TX 77204-2020 USA
| |
Collapse
|
38
|
Chabot S, Jabrane-Ferrat N, Bigot K, Tabiasco J, Provost A, Golzio M, Noman MZ, Giustiniani J, Bellard E, Brayer S, Aguerre-Girr M, Meggetto F, Giuriato S, Malecaze F, Galiacy S, Jaïs JP, Chose O, Kadouche J, Chouaib S, Teissié J, Abitbol M, Bensussan A, Le Bouteiller P. A novel antiangiogenic and vascular normalization therapy targeted against human CD160 receptor. ACTA ACUST UNITED AC 2011; 208:973-86. [PMID: 21482699 PMCID: PMC3092350 DOI: 10.1084/jem.20100810] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A monoclonal anti-CD160 antibody inhibits the growth of new vessels in pathological ocular and tumor neoangiogenesis but not in healthy tissues. Angiogenesis plays an essential role in several diseases of the eye and in the growth of solid tumors, but existing antiangiogenic therapies have limited benefits in several cases. We report the antiangiogenic effects of a monoclonal antibody, CL1-R2, in several animal models of neovascularization. CL1-R2 recognizes human CD160, a membrane receptor which is conserved in various mammal species. We show that CD160 is expressed on the endothelial cells of newly formed blood vessels in human colon carcinoma and mouse B16 melanoma but not in vessels of healthy tissues. CL1-R2 reduced fibroblast growth factor 2–induced neovascularization in the rabbit cornea, in a mouse model of oxygen-induced retinopathy, and in a mouse Matrigel plug assay. Treatment of B16 melanoma-bearing mice with CL1-R2 combined with cyclophosphamide chemotherapy caused regression of the tumor vasculature and normalization of the remaining vessels as shown by Doppler ultrasonography, intravital microscopy, and histology. These studies validate CD160 as a potential new target in cases of human pathological ocular and tumor neoangiogenesis that do not respond or become resistant to existing antiangiogenic drugs.
Collapse
Affiliation(s)
- Sophie Chabot
- Institut National de la Santé et de la Recherche Médicale U1043, F-31300 Toulouse, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Amenomori M, Mukae H, Ishimatsu Y, Sakamoto N, Kakugawa T, Hara A, Hara S, Fujita H, Ishimoto H, Hayashi T, Kohno S. Differential effects of human neutrophil peptide-1 on growth factor and interleukin-8 production by human lung fibroblasts and epithelial cells. Exp Lung Res 2010; 36:411-9. [DOI: 10.3109/01902141003714049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Misato Amenomori
- 1Second Department of Internal Medicine, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Hiroshi Mukae
- 2Second Department of Internal Medicine, Nagasaki University School of Medicine, Nagasaki, Japan; and Department of Respiratory Diseases, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Yuji Ishimatsu
- 1Second Department of Internal Medicine, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Noriho Sakamoto
- 1Second Department of Internal Medicine, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Tomoyuki Kakugawa
- 1Second Department of Internal Medicine, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Atsuko Hara
- 1Second Department of Internal Medicine, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Shintaro Hara
- 1Second Department of Internal Medicine, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Hanako Fujita
- 1Second Department of Internal Medicine, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Hiroshi Ishimoto
- 1Second Department of Internal Medicine, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Tomayoshi Hayashi
- 3Department of Pathology, Nagasaki University Hospital, Nagasaki, Japan
| | - Shigeru Kohno
- 1Second Department of Internal Medicine, Nagasaki University School of Medicine, Nagasaki, Japan
| |
Collapse
|
40
|
Complement-mediated inhibition of neovascularization reveals a point of convergence between innate immunity and angiogenesis. Blood 2010; 116:4395-403. [PMID: 20625009 DOI: 10.1182/blood-2010-01-261503] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Beyond its role in immunity, complement mediates a wide range of functions in the context of morphogenetic or tissue remodeling processes. Angiogenesis is crucial during tissue remodeling in multiple pathologies; however, the knowledge about the regulation of neovascularization by the complement components is scarce. Here we studied the involvement of complement in pathological angiogenesis. Strikingly, we found that mice deficient in the central complement component C3 displayed increased neovascularization in the model of retinopathy of prematurity (ROP) and in the in vivo Matrigel plug assay. In addition, antibody-mediated blockade of C5, treatment with C5aR antagonist, or C5aR deficiency in mice resulted in enhanced pathological retina angiogenesis. While complement did not directly affect angiogenesis-related endothelial cell functions, we found that macrophages mediated the antiangiogenic activity of complement. In particular, C5a-stimulated macrophages were polarized toward an angiogenesis-inhibitory phenotype, including the up-regulated secretion of the antiangiogenic soluble vascular endothelial growth factor receptor-1. Consistently, macrophage depletion in vivo reversed the increased neovascularization associated with C3- or C5aR deficiency. Taken together, complement and in particular the C5a-C5aR axes are potent inhibitors of angiogenesis.
Collapse
|
41
|
Abstract
Angiogenesis is the formation of new blood vessels from pre-existing vasculature. Pathologic angiogenesis in the eye can lead to severe visual impairment. In our review, we discuss the roles of both pro-angiogenic and anti-angiogenic molecular players in corneal angiogenesis, proliferative diabetic retinopathy, exudative macular degeneration and retinopathy of prematurity, highlighting novel targets that have emerged over the past decade.
Collapse
Affiliation(s)
- Yureeda Qazi
- Department of Ophthalmology, John Moran Eye Center, University of Utah, Salt Lake City, UT-84132, USA
| | | | | |
Collapse
|
42
|
Szymborska-Kajanek A, Strzelczyk JK, Karasek D, Rawwash HA, Biniszkiewicz T, Cieślar G, Hajdrowska B, Sieroń-Stołtny K, Sieroń A, Wiczkowski A, Grzeszczak W, Strojek K, Wróbel M. Impact of low-frequency pulsed magnetic fields on defensin and CRP concentrations in patients with painful diabetic polyneuropathy and in healthy subjects. Electromagn Biol Med 2010; 29:19-25. [PMID: 20230295 DOI: 10.3109/15368371003635376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIM The aim was to assess whether magnetic field influences defensin and CRP concentrations in patients with diabetic polyneuropathy and in healthy subjects. METHODS 61 diabetic patients were randomly divided into 2 groups: study group-32 patients exposed to low-frequency magnetic field; and control group-29 patients with sham exposure. Additionally, 20 healthy subjects exposed to low-frequency magnetic field. Exposures were performed during 3 weeks, 5 days in a week. Defensin and CRP concentrations were measured at baseline, after 3 weeks and at the end of the study. RESULTS There were no significant changes in defensin concentration in patients with diabetes in both the real and sham exposure group. We observed increased concentration of defensin in healthy subjects in week 5 vs. baseline value (P<0.02). CONCLUSIONS Magnetic field has no impact on defensin concentration in diabetic patients but has positive influence on this parameter in healthy subjects.
Collapse
Affiliation(s)
- Aleksandra Szymborska-Kajanek
- Diabetological Unit of the Department of Internal Medicine, Diabetology and Nephrology, Medical University of Silesia, Zabrze, Poland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kumar A, Hou X, Lee C, Li Y, Maminishkis A, Tang Z, Zhang F, Langer HF, Arjunan P, Dong L, Wu Z, Zhu LY, Wang L, Min W, Colosi P, Chavakis T, Li X. Platelet-derived growth factor-DD targeting arrests pathological angiogenesis by modulating glycogen synthase kinase-3beta phosphorylation. J Biol Chem 2010; 285:15500-15510. [PMID: 20231273 DOI: 10.1074/jbc.m110.113787] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Platelet-derived growth factor-DD (PDGF-DD) is a recently discovered member of the PDGF family. The role of PDGF-DD in pathological angiogenesis and the underlying cellular and molecular mechanisms remain largely unexplored. In this study, using different animal models, we showed that PDGF-DD expression was up-regulated during pathological angiogenesis, and inhibition of PDGF-DD suppressed both choroidal and retinal neovascularization. We also demonstrated a novel mechanism mediating the function of PDGF-DD. PDGF-DD induced glycogen synthase kinase-3beta (GSK3beta) Ser(9) phosphorylation and Tyr(216) dephosphorylation in vitro and in vivo, leading to increased cell survival. Consistently, GSK3beta activity was required for the antiangiogenic effect of PDGF-DD targeting. Moreover, PDGF-DD regulated the expression of GSK3beta and many other genes important for angiogenesis and apoptosis. Thus, we identified PDGF-DD as an important target gene for antiangiogenic therapy due to its pleiotropic effects on vascular and non-vascular cells. PDGF-DD inhibition may offer new therapeutic options to treat neovascular diseases.
Collapse
Affiliation(s)
- Anil Kumar
- NEI, National Institutes of Health, Bethesda, Maryland 20852
| | - Xu Hou
- NEI, National Institutes of Health, Bethesda, Maryland 20852; Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Chunsik Lee
- NEI, National Institutes of Health, Bethesda, Maryland 20852
| | - Yang Li
- NEI, National Institutes of Health, Bethesda, Maryland 20852
| | | | - Zhongshu Tang
- NEI, National Institutes of Health, Bethesda, Maryland 20852
| | - Fan Zhang
- NEI, National Institutes of Health, Bethesda, Maryland 20852
| | - Harald F Langer
- Experimental Immunology Branch, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892; Department of Cardiovascular Medicine, University of Tuebingen, 72076 Tuebingen, Germany
| | | | - Lijin Dong
- NEI, National Institutes of Health, Bethesda, Maryland 20852
| | - Zhijian Wu
- NEI, National Institutes of Health, Bethesda, Maryland 20852
| | - Linda Y Zhu
- NEI, National Institutes of Health, Bethesda, Maryland 20852
| | - Lianchun Wang
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| | - Wang Min
- Department of Pathology, Vascular Biology, and Therapeutics, Yale University, New Haven, Connecticut 06520
| | - Peter Colosi
- NEI, National Institutes of Health, Bethesda, Maryland 20852
| | - Triantafyllos Chavakis
- Experimental Immunology Branch, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Xuri Li
- NEI, National Institutes of Health, Bethesda, Maryland 20852.
| |
Collapse
|
44
|
Bdeir K, Higazi AAR, Kulikovskaya I, Christofidou-Solomidou M, Vinogradov SA, Allen TC, Idell S, Linzmeier R, Ganz T, Cines DB. Neutrophil alpha-defensins cause lung injury by disrupting the capillary-epithelial barrier. Am J Respir Crit Care Med 2010; 181:935-46. [PMID: 20093642 DOI: 10.1164/rccm.200907-1128oc] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
RATIONALE The involvement of neutrophil activation in the sentinel, potentially reversible, events in the pathogenesis of acute lung injury (ALI) is only partially understood. alpha-Defensins are the most abundant proteins secreted by activated human neutrophils, but their contribution to ALI in mouse models is hindered by their absence from murine neutrophils and the inability to study their effects in isolation in other species. OBJECTIVES To study the role of alpha-defensins in the pathogenesis of ALI in a clinically relevant setting using mice transgenic for polymorphonuclear leukocyte expression of alpha-defensins. METHODS Transgenic mice expressing polymorphonuclear leukocyte alpha-defensins were generated. ALI was induced by acid aspiration. Pulmonary vascular permeability was studied in vivo using labeled dextran and fibrin deposition. The role of the low-density lipoprotein-related receptor (LRP) in permeability was examined. MEASUREMENTS AND MAIN RESULTS Acid aspiration induced neutrophil migration and release of alpha-defensins into lung parenchyma and airways. ALI was more severe in alpha-defensin-expressing mice than in wild-type mice, as determined by inspection, influx of neutrophils into the interstitial space and airways, histological evidence of epithelial injury, interstitial edema, extravascular fibrin deposition, impaired oxygenation, and reduced survival. Within 4 hours of insult, alpha-defensin-expressing mice showed greater disruption of capillary-epithelial barrier function and ALI that was attenuated by systemic or intratracheal administration of specific inhibitors of the LRP. CONCLUSIONS alpha-Defensins mediate ALI through LRP-mediated loss of capillary-epithelial barrier function, suggesting a potential new approach to intervention.
Collapse
Affiliation(s)
- Khalil Bdeir
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA l9l04, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Alpha-defensins secreted by dysplastic granulocytes inhibit the differentiation of monocytes in chronic myelomonocytic leukemia. Blood 2009; 115:78-88. [PMID: 19864642 DOI: 10.1182/blood-2009-05-224352] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic disorder that occurs in elderly patients. One of the main diagnostic criteria is the accumulation of heterogeneous monocytes in the peripheral blood. We further explored this cellular heterogeneity and observed that part of the leukemic clone in the peripheral blood was made of immature dysplastic granulocytes with a CD14(-)/CD24(+) phenotype. The proteome profile of these cells is dramatically distinct from that of CD14(+)/CD24(-) monocytes from CMML patients or healthy donors. More specifically, CD14(-)/CD24(+) CMML cells synthesize and secrete large amounts of alpha-defensin 1-3 (HNP1-3). Recombinant HNPs inhibit macrophage colony-stimulating factor (M-CSF)-driven differentiation of human peripheral blood monocytes into macrophages. Using transwell, antibody-mediated depletion, suramin inhibition of purinergic receptors, and competitive experiments with uridine diphosphate (UDP)/uridine triphosphate (UTP), we demonstrate that HNP1-3 secreted by CD14(-)/CD24(+) cells inhibit M-CSF-induced differentiation of CD14(+)/CD24(-) cells at least in part through P2Y6, a receptor involved in macrophage differentiation. Altogether, these observations suggest that a population of immature dysplastic granulocytes contributes to the CMML phenotype through production of alpha-defensins HNP1-3 that suppress the differentiation capabilities of monocytes.
Collapse
|
46
|
Wang YS, Li D, Shi HS, Wen YJ, Yang L, Xu N, Chen XC, Chen X, Chen P, Li J, Deng HX, Wang CT, Xie G, Huang S, Mao YQ, Chen LJ, Zhao X, Wei YQ. Intratumoral expression of mature human neutrophil peptide-1 mediates antitumor immunity in mice. Clin Cancer Res 2009; 15:6901-11. [PMID: 19861439 DOI: 10.1158/1078-0432.ccr-09-0484] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE Human neutrophil peptides (HNP1-3), small molecular antimicrobial peptides, are expressed within tumors and associated with tumor necrosis and inhibition of angiogenesis. Recent investigations have suggested that HNP1-3 are likely to be involved in the host immune responses to tumors. EXPERIMENTAL DESIGN We used recombinant pSec-HNP1, which expresses a secretable form of HNP1, to obtain expression of HNP1 in the tumor milieu in immunocompetent mice to explore the possible roles of HNP1 in tumor immunity. The antitumor effects were investigated in established CT26 colon cancer and 4T1 breast cancer models. RESULTS HNP1-mediated chemotactic and activating effects on immature dendritic cells were detected both in vitro and in vivo. Intratumoral expression of HNP1 resulted in not only significant tumor growth inhibition but also increased CTL infiltration within tumors. Adoptive transfer of splenocytes and a (51)Cr release assay revealed specific cellular immune responses. Furthermore, increased antibodies were also found in sera from pSec-HNP1-treated mice supporting specific humoral immune responses. Increased apoptosis and decreased angiogenesis were also shown in treated tumors. CONCLUSIONS These findings indicate that HNP1 can exert multiple antitumor effects through different mechanisms; more importantly, HNP1 mediates host immune responses to tumors in situ through the recruitment and subsequent activation of immature dendritic cells and thus shows promising potential in cancer therapy.
Collapse
Affiliation(s)
- Yong-Sheng Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Department of Gynecology and Obstetrics, Second West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Seki M, Soussou W, Manabe SI, Lipton SA. Protection of retinal ganglion cells by caspase substrate-binding peptide IQACRG from N-methyl-D-aspartate receptor-mediated excitotoxicity. Invest Ophthalmol Vis Sci 2009; 51:1198-207. [PMID: 19815732 DOI: 10.1167/iovs.09-4102] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
PURPOSE This study investigated whether the enzymatically inactive caspase mimetic IQACRG protects rat retinal ganglion cells (RGCs) from excitotoxic insults. Minimally invasive delivery of the peptide to the retina was explored, and the mechanisms of neuroprotection were elucidated. METHODS IQACRG was linked to penetratin (P-IQACRG) to facilitate cellular uptake. RGC labeling by biotinylated-P-IQACRG delivered via intravitreal or subconjunctival injection was demonstrated by avidin-biotin chemistry. The authors used histologic and electrophysiological measures to evaluate the neuroprotective potential of P-IQACRG against RGC death induced by N-methyl-D-aspartate (NMDA) in vitro and in vivo. In addition, they monitored activity of an enzyme that is downstream of caspase-1, matrix metalloproteinase-9 (MMP-9), and protein levels of the caspase-3/7 substrate, myocyte enhancer factor 2C (MEF2C), to determine the effectiveness of IQACRG in blocking excessive caspase activity. RESULTS IQACRG significantly reduced NMDA-induced RGC death in culture and in vivo. Ex vivo electrophysiological recording of the retina on multielectrode arrays demonstrated functional rescue of RGCs by IQACRG. The authors also found that delivery of IQACRG to the retina inhibited NMDA-triggered MMP-9 activity and prevented cleavage of MEF2C protein that would otherwise have been engendered by caspase activation preceding RGC death. Strikingly, subconjunctival injection of P-IQACRG was very effective in preventing NMDA-induced RGC death in vivo. CONCLUSIONS These data demonstrate that IQACRG protects RGCs from excitotoxicity in vitro and in vivo. The positive results with subconjunctival administration of P-IQACRG suggest that in the future this treatment may be useful clinically in diseases such as glaucoma and retinal ischemia.
Collapse
Affiliation(s)
- Masaaki Seki
- Del E. Webb Center for Neuroscience, Aging, and Stem Cell Research, Burnham Institute for Medical Research, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
48
|
Maier AKB, Kociok N, Zahn G, Vossmeyer D, Stragies R, Muether PS, Joussen AM. Modulation of Hypoxia-Induced Neovascularization by JSM6427, an Integrin α5β 1 Inhibiting Molecule. Curr Eye Res 2009; 32:801-12. [PMID: 17882713 DOI: 10.1080/02713680701553052] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
PURPOSE Integrin alpha5beta1, a fibronectin receptor, is involved in endothelial cell migration and proliferation. Here we investigate the effect of JSM6427, an integrin alpha5beta1 inhibiting molecule, on the development of retinal vascular system using the mouse model of oxygen-induced retinopathy (OIR). METHODS Endothelial cell migration and sprouting was analyzed in vitro using a 2D migration assay and a 3D sprouting/angiogenesis assay in fibrin gel. C57BL/C6 mice were exposed to 75% oxygen from postnatal day 7 (P7) to P12 and returned to room air thereafter. Intravitreal injection of 40 microg JSM6427 was performed in each one eye on P14. On P17, vascular area, avascularized area, and neovascular blood vessel tufts were quantified after perfusion with fluorescein-coupled concanavalin A. The number of retinal neovascular cell nuclei was determined in hematoxylin-stained cross sections of the eyes. Integrin alpha 5 expression was determined by immunohistochemistry. RESULTS In vitro, JSM6427 inhibits the migration of HUVEC and the tube formation induced by both bFGF and VEGF. In vivo, integrin alpha 5 expression was detectable in neovascular retinal blood vessels. Oxygen treatment (positive control) in comparison with no oxygen treatment (negative control) reduced significantly the vascularized area and increased the avascularized area. A single intravitreal injection of 40 microg JSM6427 resulted in a significant reduction of the vascularized area and the number of preretinal nuclei in comparison with the intravitreal injection of the vehicle while the avascularized area increased significantly. CONCLUSIONS These results imply an essential role of integrin alpha5beta1 in the refining of the retinal vasculature in OIR and suggest JSM6427 may have a possible therapeutic function for neovascular disease.
Collapse
Affiliation(s)
- Anna-Karina B Maier
- Department of Vitreoretinal Surgery, Center of Ophthalmology, University of Cologne, Cologne, Germany
| | | | | | | | | | | | | |
Collapse
|
49
|
Histone H2AX is integral to hypoxia-driven neovascularization. Nat Med 2009; 15:553-8. [PMID: 19377486 DOI: 10.1038/nm.1947] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Accepted: 02/27/2009] [Indexed: 12/24/2022]
Abstract
H2A histone family member X (H2AX, encoded by H2AFX) and its C-terminal phosphorylation (gamma-H2AX) participates in the DNA damage response and mediates DNA repair. Hypoxia is a physiological stress that induces a replication-associated DNA damage response. Moreover, hypoxia is the major driving force for neovascularization, as the hypoxia-mediated induction of vascular growth factors triggers endothelial cell proliferation. Here we studied the role of the hypoxia-induced DNA damage response in endothelial cell function and in hypoxia-driven neovascularization in vivo. Hypoxia induced replication-associated generation of gamma-H2AX in endothelial cells in vitro and in mice. Both in cultured cells and in mice, endothelial cell proliferation under hypoxic conditions was reduced by H2AX deficiency. Whereas developmental angiogenesis was not affected in H2afx(-/-) mice, hypoxia-induced neovascularization during pathologic proliferative retinopathy, in response to hind limb ischemia or during tumor angiogenesis was substantially lower in H2afx(-/-) mice. Moreover, endothelial-specific H2afx deletion resulted in reduced hypoxia-driven retina neovascularization and tumor neovascularization. Our findings establish that H2AX, and hence activation of the DNA repair response, is needed for endothelial cells to maintain their proliferation under hypoxic conditions and is crucial for hypoxia-driven neovascularization.
Collapse
|
50
|
Droin N, Hendra JB, Ducoroy P, Solary E. Human defensins as cancer biomarkers and antitumour molecules. J Proteomics 2009; 72:918-27. [PMID: 19186224 DOI: 10.1016/j.jprot.2009.01.002] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Revised: 01/06/2009] [Accepted: 01/07/2009] [Indexed: 12/20/2022]
Abstract
Human defensins, which are small cationic peptides produced by neutrophils and epithelial cells, form two genetically distinct alpha and beta subfamilies. They are involved in innate immunity through killing microbial pathogens or neutralizing bacterial toxins and in adaptive immunity by serving as chemoattractants and activators of immune cells. alpha-defensins are mainly packaged in neutrophil granules (HNP1, HNP2, HNP3) or secreted by intestinal Paneth cells (HD5, HD6), while beta-defensins are expressed in mucosa and epithelial cells. Using surface enhanced laser desorption/ionisation time-of-flight (SELDI-TOF) mass spectrometry (MS), alpha-defensins were found to be expressed in a variety of human tumours, either in tumour cells or at their surface. HNP1-3 peptides are also secreted and their accumulation in biological fluids was proposed as a tumour biomarker. Conversely, beta-defensin-1 (HBD-1) is down-regulated in some tumour types in which it could behave as a tumour suppressor protein. Alpha-defensins promote tumour cell growth or, at higher concentration, provoke cell death. These peptides also inhibit angiogenesis, which, in addition to immunomodulation, indicates a complex role in tumour development. This review summarizes current knowledge of defensins to discuss their role in tumour growth, tumour monitoring and cancer treatment.
Collapse
|