1
|
Sánchez-Martínez C, Grueso E, Calvo-López T, Martinez-Ortega J, Ruiz A, Almendral JM. VEGF-Virus Interactions: Pathogenic Mechanisms and Therapeutic Applications. Cells 2024; 13:1815. [PMID: 39513922 PMCID: PMC11545703 DOI: 10.3390/cells13211815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Many types of viruses directly or indirectly target the vascular endothelial growth factor (VEGF) system, which is a central regulator of vasculogenesis and angiogenesis in physiological homeostasis, causing diverse pathologies. Other viruses have been developed into effective therapeutic tools for VEGF modulation in conditions such as cancer and eye diseases. Some viruses may alter the levels of VEGF in the pathogenesis of respiratory syndromes, or they may encode VEGF-like factors, promoting vascular disruption and angiogenesis to enable viruses' systemic spread. Oncogenic viruses may express interactive factors that perturb VEGF's functional levels or downstream signaling, which increases the neovascularization and metastasis of tumors. Furthermore, many viruses are being developed as therapeutic vectors for vascular pathologies in clinical trials. Major examples are those viral vectors that inhibit the role of VEGF in the neovascularization required for cancer progression; this is achieved through the induction of immune responses, by exposing specific peptides that block signaling or by expressing anti-VEGF and anti-VEGF receptor-neutralizing antibodies. Other viruses have been engineered into effective pro- or anti-angiogenesis multitarget vectors for neovascular eye diseases, paving the way for therapies with improved safety and minimal side effects. This article critically reviews the large body of literature on these issues, highlighting those contributions that describe the molecular mechanisms, thus expanding our understanding of the VEGF-virus interactions in disease and therapy. This could facilitate the clinical use of therapeutic virus vectors in precision medicine for the VEGF system.
Collapse
Affiliation(s)
- Cristina Sánchez-Martínez
- Biosciences Research Institute, School of Experimental Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain; (C.S.-M.); (E.G.)
| | - Esther Grueso
- Biosciences Research Institute, School of Experimental Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain; (C.S.-M.); (E.G.)
| | - Tania Calvo-López
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Cantoblanco, 28049 Madrid, Spain or (T.C.-L.); (J.M.-O.); (A.R.)
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
- Department of Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Jorge Martinez-Ortega
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Cantoblanco, 28049 Madrid, Spain or (T.C.-L.); (J.M.-O.); (A.R.)
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - Ana Ruiz
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Cantoblanco, 28049 Madrid, Spain or (T.C.-L.); (J.M.-O.); (A.R.)
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - José M. Almendral
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Cantoblanco, 28049 Madrid, Spain or (T.C.-L.); (J.M.-O.); (A.R.)
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
2
|
Pintus D, Cancedda MG, Puggioni G, Scivoli R, Rocchigiani AM, Maestrale C, Coradduzza E, Bechere R, Silva-Flannery L, Bullock HA, Macciocu S, Montesu MA, Marras V, Dore S, Ritter JM, Ligios C. ORF virus causes tumor-promoting inflammation in sheep and goats. Vet Pathol 2024; 61:803-814. [PMID: 38613413 PMCID: PMC11370215 DOI: 10.1177/03009858241241794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2024]
Abstract
ORF virus (ORFV) causes contagious ecthyma ("ORF"), a disease of sheep and goats characterized by lesions ranging from vesicles and pustules to atypical papilloma-like and angiomatous lesions in the skin and mucosae. The authors investigated the molecular factors leading to the ORF-associated atypical tumor-like changes. Fifteen lambs, 15 kids, and an adult ram clinically affected by natural ORFV infection were enrolled in the study and examined by several methods. ORFV was detected by viral culture or real-time polymerase chain reaction (RT-PCR) in the lesioned tissues and in the blood of the clinically affected sheep and goats. Surprisingly, ORFV was also detected in the blood of healthy goats from an affected herd. Microscopically, they found a pseudo-papillomatous proliferation of the epithelium, while the dermis and lamina propria were expanded by a proliferating neovascular component that highly expressed the viral vascular endothelial growth factor (vVEGF) and its host receptor vascular endothelial growth factor receptor 2 (VEGFR2). Immunohistochemistry, immunofluorescence, and in situ hybridization for mRNA showed that epidermal growth factor receptor (EGFR) was expressed in the fibrovascular component, in the infiltrating CD163+ macrophages, and in the basal stratum of the epidermis. Confocal immunofluorescence microscopy demonstrated that CD163+ macrophages were associated with VEGF and VEGFR2. Finally, they found by quantitative RT-PCR the overexpression of the interleukin-6 and VEGFR2 genes in the lesioned tissues. These findings suggest that ORFV activates an inflammatory reaction characterized by CD163+ macrophages expressing EGFR and VEGFR2, which might play an oncogenic role through synergistic action with vVEGF signaling.
Collapse
Affiliation(s)
- Davide Pintus
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Maria G Cancedda
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | | | - Rosario Scivoli
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | | | | | | | - Roberto Bechere
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | | | | | - Simona Macciocu
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | | | | | - Simone Dore
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Jana M Ritter
- Centers for Disease Control and Prevention, Atlanta, GA
| | - Ciriaco Ligios
- Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| |
Collapse
|
3
|
Sabourirad S, Dimitriadis E, Mantamadiotis T. Viruses exploit growth factor mechanisms to achieve augmented pathogenicity and promote tumorigenesis. Arch Microbiol 2024; 206:193. [PMID: 38526562 PMCID: PMC10963461 DOI: 10.1007/s00203-024-03855-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/13/2024] [Accepted: 01/20/2024] [Indexed: 03/26/2024]
Abstract
Cellular homeostasis is regulated by growth factors (GFs) which orchestrate various cellular processes including proliferation, survival, differentiation, motility, inflammation and angiogenesis. Dysregulation of GFs in microbial infections and malignancies have been reported previously. Viral pathogens exemplify the exploitation of host cell GFs and their signalling pathways contributing to viral entry, virulence, and evasion of anti-viral immune responses. Viruses can also perturb cellular metabolism and the cell cycle by manipulation of GF signaling. In some cases, this disturbance may promote oncogenesis. Viral pathogens can encode viral GF homologues and induce the endogenous biosynthesis of GFs and their corresponding receptors or manipulate their activity to infect the host cells. Close investigation of how viral strategies exploit and regulate GFs, a will shed light on how to improve anti-viral therapy and cancer treatment. In this review, we discuss and provide insights on how various viral pathogens exploit different GFs to promote viral survival and oncogenic transformation, and how this knowledge can be leveraged toward the design of more efficient therapeutics or novel drug delivery systems in the treatment of both viral infections and malignancies.
Collapse
Affiliation(s)
- Sarvenaz Sabourirad
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia.
| | - Evdokia Dimitriadis
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women's Hospital, Parkville, VIC, Australia
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Theo Mantamadiotis
- Department of Surgery RMH, The University of Melbourne, Parkville, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
4
|
Pérez-Gutiérrez L, Ferrara N. Biology and therapeutic targeting of vascular endothelial growth factor A. Nat Rev Mol Cell Biol 2023; 24:816-834. [PMID: 37491579 DOI: 10.1038/s41580-023-00631-w] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2023] [Indexed: 07/27/2023]
Abstract
The formation of new blood vessels, called angiogenesis, is an essential pathophysiological process in which several families of regulators have been implicated. Among these, vascular endothelial growth factor A (VEGFA; also known as VEGF) and its two tyrosine kinase receptors, VEGFR1 and VEGFR2, represent a key signalling pathway mediating physiological angiogenesis and are also major therapeutic targets. VEGFA is a member of the gene family that includes VEGFB, VEGFC, VEGFD and placental growth factor (PLGF). Three decades after its initial isolation and cloning, VEGFA is arguably the most extensively investigated signalling system in angiogenesis. Although many mediators of angiogenesis have been identified, including members of the FGF family, angiopoietins, TGFβ and sphingosine 1-phosphate, all current FDA-approved anti-angiogenic drugs target the VEGF pathway. Anti-VEGF agents are widely used in oncology and, in combination with chemotherapy or immunotherapy, are now the standard of care in multiple malignancies. Anti-VEGF drugs have also revolutionized the treatment of neovascular eye disorders such as age-related macular degeneration and ischaemic retinal disorders. In this Review, we emphasize the molecular, structural and cellular basis of VEGFA action as well as recent findings illustrating unexpected interactions with other pathways and provocative reports on the role of VEGFA in regenerative medicine. We also discuss clinical and translational aspects of VEGFA. Given the crucial role that VEGFA plays in regulating angiogenesis in health and disease, this molecule is largely the focus of this Review.
Collapse
Affiliation(s)
- Lorena Pérez-Gutiérrez
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Napoleone Ferrara
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
5
|
Rauniyar K, Bokharaie H, Jeltsch M. Expansion and collapse of VEGF diversity in major clades of the animal kingdom. Angiogenesis 2023; 26:437-461. [PMID: 37017884 PMCID: PMC10328876 DOI: 10.1007/s10456-023-09874-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/17/2023] [Indexed: 04/06/2023]
Abstract
Together with the platelet-derived growth factors (PDGFs), the vascular endothelial growth factors (VEGFs) form the PDGF/VEGF subgroup among cystine knot growth factors. The evolutionary relationships within this subgroup have not been examined thoroughly to date. Here, we comprehensively analyze the PDGF/VEGF growth factors throughout all animal phyla and propose a phylogenetic tree. Vertebrate whole-genome duplications play a role in expanding PDGF/VEGF diversity, but several limited duplications are necessary to account for the temporal pattern of emergence. The phylogenetically oldest PDGF/VEGF-like growth factor likely featured a C-terminus with a BR3P signature, a hallmark of the modern-day lymphangiogenic growth factors VEGF-C and VEGF-D. Some younger VEGF genes, such as VEGFB and PGF, appeared completely absent in important vertebrate clades such as birds and amphibia, respectively. In contrast, individual PDGF/VEGF gene duplications frequently occurred in fish on top of the known fish-specific whole-genome duplications. The lack of precise counterparts for human genes poses limitations but also offers opportunities for research using organisms that diverge considerably from humans. Sources for the graphical abstract: 326 MYA and older [1]; 72-240 MYA [2]; 235-65 MYA [3].
Collapse
Affiliation(s)
- Khushbu Rauniyar
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Biocenter 2, (Viikinkaari 5E), P.O. Box. 56, 00790, Helsinki, Finland
| | - Honey Bokharaie
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Biocenter 2, (Viikinkaari 5E), P.O. Box. 56, 00790, Helsinki, Finland
| | - Michael Jeltsch
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Biocenter 2, (Viikinkaari 5E), P.O. Box. 56, 00790, Helsinki, Finland.
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Wihuri Research Institute, Helsinki, Finland.
- Helsinki One Health, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
6
|
Yu Y, Zhang F, Duan X, Yang C, Cui Y, Yu L. ORFV can carry TRAP gene expression via intracellular CRISPR/Cas9 gene editing technology. J Virol Methods 2023; 312:114652. [PMID: 36493528 DOI: 10.1016/j.jviromet.2022.114652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
Orf is an acute and highly contracted human and animal infection caused by orf virus (ORFV), which mainly affects sheep, goats, and other species. Clinically, opportunistic or conditional pathogens such as Staphylococcus aureus (S. aureus) are often detected in cases of orf, which greatly increases the risk of disease progression and clinical death. It has been reported that TRAP gene products of S. aureus can broadly influence bacterial life and pathogenicity in vivo, and introduction of exogenous TRAP genes may help to inhibit the proliferation of bacteria. In order to achieve the combined control of ORFV and S. aureus, a novel approach to design a S. aureus TRAP gene vaccine using a live attenuated ORFV vector is proposed. In this study, CRISPR/Cas9 gene editing technology was used to disable vascular endothelial growth factor E of ORFV (VEGF-v) and introduced TRAP gene into this position. TRAP gene expression was detected in keratinocytes infected with recombinant virus. The construction and experimental verification of recombinant ORFV (ORFV-v/TRAP) will provide a reference for in-depth studies on the prevention and control of mixed infectious disease.
Collapse
Affiliation(s)
- YongZhong Yu
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China.
| | - Fan Zhang
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Xuyang Duan
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China.
| | - ChaoQun Yang
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - YuDong Cui
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Li Yu
- Division of Livestock Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 427 Maduan Street, Harbin 150001, PR China
| |
Collapse
|
7
|
Deletion of gene OV132 attenuates Orf virus more effectively than gene OV112. Appl Microbiol Biotechnol 2023; 107:835-851. [PMID: 36484827 PMCID: PMC9734686 DOI: 10.1007/s00253-022-12323-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/27/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
Orf virus (ORFV), a Parapoxvirus in Poxviridae, infects sheep and goats resulting in contagious pustular dermatitis. ORFV is regarded as a promising viral vector candidate for vaccine development and oncolytic virotherapy. Owing to their potential clinical application, safety concerns have become increasingly important. Deletion of either the OV132 (encoding vascular endothelial growth factor, VEGF) or OV112 (encoding the chemokine binding protein, CBP) genes reduced ORFV infectivity, which has been independently demonstrated in the NZ2 and NZ7 strains, respectively. This study revealed that the VEGF and CBP gene sequences of the local strain (TW/Hoping) shared a similarity of 47.01% with NZ2 and 90.56% with NZ7. Due to the high sequence divergence of these two immunoregulatory genes among orf viral strains, their contribution to the pathogenicity of Taiwanese ORFV isolates was comparatively characterized. Initially, two ORFV recombinants were generated, in which either the VEGF or CBP gene was deleted and replaced with the reporter gene EGFP. In vitro assays indicated that both the VEGF-deletion mutant ORFV-VEGFΔ-EGFP and the CBP deletion mutant ORFV-CBPΔ-EGFP were attenuated in cells. In particular, ORFV-VEGFΔ-EGFP significantly reduced plaque size and virus yield compared to ORFV-CBPΔ-EGFP and the wild-type control. Similarly, in vivo analysis revealed no virus yield in the goat skin biopsy infected by ORFV-VEGFΔ-EGFP, and significantly reduced the virus yield of ORFV-CBPΔ-EGFP relative to the wild-type control. These results confirmed the loss of virulence of both deletion mutants in the Hoping strain, whereas the VEGF-deletion mutant was more attenuated than the CBP deletion strain in both cell and goat models. KEY POINTS: • VEGF and CBP genes are crucial in ORFV pathogenesis in the TW/Hoping strain • The VEGF-deletion mutant virus was severely attenuated in both cell culture and animal models • Deletion mutant viruses are advantageous vectors for the development of vaccines and therapeutic regimens.
Collapse
|
8
|
Hussain I, Khan MUR, Aslam A, Rabbani M, Masood S, Anjum A. Identification, molecular characterization, and pathological features of orf virus in sheep and goats in Punjab province, Pakistan. Trop Anim Health Prod 2022; 55:24. [PMID: 36562854 DOI: 10.1007/s11250-022-03432-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 11/24/2022] [Indexed: 12/24/2022]
Abstract
Orf virus (ORFV) causes an acute, contagious, skin disease of sheep and goats which is economically important. The objectives of this study were to identify ORFV and to explore its pathological and phylogenetic profiles in 350 goats and 91 sheep of 14 districts of Punjab, Pakistan, from July 2020 to July 2021. Skin scrapings (total no. of samples = 441) of suspected animals were subjected to polymerase chain reactions, phylogenetic analysis, and pathological observations. The partial length of GIF/IL-2 gene (408 bp) was successfully amplified in 58/441 samples. Phylogenetic analysis of GIF/IL2 gene showed that the study isolates belonged to ORFV-cluster I, together with the viruses reported in India and China. Pakistan ORFV isolates were shared 97.6-98.7% nucleotide and 97.6-100% amino acid identities with the reference strain (NC_005336). Moreover, Chinese ORFV-isolates were detected unique multiple amino acid substitutions (F11L, Q21H, D27N, I46V, N49S, N82D, D103N, S129G) with study isolates. Naturally infected animals were anorexic, emaciated, dull, and depressed. The macroscopic lesions included multifocal to coalescing, ulceration followed by proliferative papules, pustules, and crust formation on the epidermis of gums, lips, mouth commissure, muzzles, nose, and udder. Histopathological examination revealed hyperplasia, anastomosing rete ridges formation and degenerative changes, including spongiosis and vacuolation of epidermal cells. Keratinocytes exhibited eosinophilic intracytoplasmic inclusion bodies with pyknotic and karyorrhexis nuclei. This is the first report on molecular characterization of ORFV from Pakistan, with insight into its pathogenesis and comparative analysis of pathological alterations and genetic diversity between ORFV strains reported in different geographical areas.
Collapse
Affiliation(s)
- Irtaza Hussain
- Department of Pathology, Faculty of Veterinary Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
- Department of Pathobiology, Faculty of Veterinary Sciences, Bahauddin Zakariya University Multan, Multan, Pakistan
| | - Muti Ur Rehman Khan
- Department of Pathology, Faculty of Veterinary Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan.
| | - Asim Aslam
- Department of Pathology, Faculty of Veterinary Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Masood Rabbani
- Institute of Microbiology, Faculty of Veterinary Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Saima Masood
- Department of Anatomy and Histology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Ahsan Anjum
- Department of Pathology, Faculty of Veterinary Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| |
Collapse
|
9
|
Saker Z, Rizk M, Bahmad HF, Nabha SM. Targeting Angiogenic Factors for the Treatment of Medulloblastoma. Curr Treat Options Oncol 2022; 23:864-886. [PMID: 35412196 DOI: 10.1007/s11864-022-00981-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2022] [Indexed: 11/24/2022]
Abstract
OPINION STATEMENT Medulloblastoma (MB) is the most frequent pediatric brain tumor. Despite conventional therapy, MB patients have high mortality and morbidity rates mainly due to the incomplete understanding of the molecular and cellular processes involved in development of this cancer. Similar to other solid tumors, MB demonstrated high endothelial cell proliferation and angiogenic activity, wherein new blood vessels arise from the pre-existing vasculature, a process named angiogenesis. MB angiogenesis is considered a hallmark for MB development, progression, and metastasis emphasizing its potential target for antitumor therapy. However, angiogenesis is tightly regulated by a set of angiogenic factors making it a complex process to be targeted. Although agents targeting these factors and their receptors are early in development, the potential for their targeting may translate into improvement in the clinical care for MB patients. In this review, we focus on the most potent angiogenic factors and their corresponding receptors, highlighting their basic properties and expression in MB. We describe their contribution to MB tumorigenesis and angiogenesis and the potential therapeutic targeting of these factors.
Collapse
Affiliation(s)
- Zahraa Saker
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Mahdi Rizk
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hisham F Bahmad
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, 4300 Alton Rd, Miami Beach, FL, 33140, USA.
| | - Sanaa M Nabha
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
10
|
Yu Y, Lian Z, Cui Y. The OH system: A panorama view of the PPV-host interaction. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 98:105220. [PMID: 35066165 DOI: 10.1016/j.meegid.2022.105220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 11/19/2021] [Accepted: 01/17/2022] [Indexed: 06/14/2023]
Abstract
Poxviruses are a family of specialized cytoplasm-parasitic DNA viruses that replicate and assembly in virus factory. In Parapoxvirus (PPV) genus, with the orf virus (ORFV) as a representative species of this genus, their behaviors are significantly different from that of Orthopoxvirus, and the plots of viral practical solutions for evading host immunity are intricate and fascinating, particularly to anti-host and host's antiviral mechanisms. In order to protect the virus factory from immune elimination caused by infection, PPVs attempt to interfere with multiple stress levels of host, mainly by modulating innate immunity response (IIR) and adaptive immunity response (AIR). Given that temporarily constructed by virus infection, ORFV-HOST (OH) system accompanied by viral strategies is carefully managed in the virus factory, thus directing many life-critical events once undergoing the IIR and AIR. Evolutionarily, to reduce the risk of system destruction, ORFV have evolved into a mild-looking mode to avoid overstimulation. Moreover, the current version of development also focus on recognizing and hijacking more than eight antiviral security mechanisms of host cells, such as the 2',5'-oligoadenylate synthetase (OAS)/RNase L and PKR systems, the ubiquitin protease system (UPS), and so on. In summary, this review assessed inescapable pathways as mentioned above, through which viruses compete with their hosts strategically. The OH system provides a panoramic view and a powerful platform for us to study the PPV-Host interaction, as well as the corresponding implications on a great application potential in anti-virus design.
Collapse
Affiliation(s)
- Yongzhong Yu
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China.
| | - Zhengxing Lian
- College of Animal Science and Technology, China Agricultural University, Beijing 100039, PR China
| | - Yudong Cui
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| |
Collapse
|
11
|
Ye X, Gaucher JF, Vidal M, Broussy S. A Structural Overview of Vascular Endothelial Growth Factors Pharmacological Ligands: From Macromolecules to Designed Peptidomimetics. Molecules 2021; 26:6759. [PMID: 34833851 PMCID: PMC8625919 DOI: 10.3390/molecules26226759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 12/27/2022] Open
Abstract
The vascular endothelial growth factor (VEGF) family of cytokines plays a key role in vasculogenesis, angiogenesis, and lymphangiogenesis. VEGF-A is the main member of this family, alongside placental growth factor (PlGF), VEGF-B/C/D in mammals, and VEGF-E/F in other organisms. To study the activities of these growth factors under physiological and pathological conditions, resulting in therapeutic applications in cancer and age-related macular degeneration, blocking ligands have been developed. These have mostly been large biomolecules like antibodies. Ligands with high affinities, at least in the nanomolar range, and accurate structural data from X-ray crystallography and NMR spectroscopy have been described. They constitute the main focus of this overview, which evidences similarities and differences in their binding modes. For VEGF-A ligands, and to a limited extent also for PlGF, a transition is now observed towards developing smaller ligands like nanobodies and peptides. These include unnatural amino acids and chemical modifications for designed and improved properties, such as serum stability and greater affinity. However, this review also highlights the scarcity of such small molecular entities and the striking lack of small organic molecule ligands. It also shows the gap between the rather large array of ligands targeting VEGF-A and the general absence of ligands binding other VEGF members, besides some antibodies. Future developments in these directions are expected in the upcoming years, and the study of these growth factors and their promising therapeutic applications will be welcomed.
Collapse
Affiliation(s)
- Xiaoqing Ye
- Faculté de Pharmacie de Paris, Université de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, 75006 Paris, France; (X.Y.); (M.V.)
| | - Jean-François Gaucher
- Laboratoire de Cristallographie et RMN Biologiques, Faculté de Pharmacie de Paris, Université de Paris, CiTCoM, 8038 CNRS, 75006 Paris, France;
| | - Michel Vidal
- Faculté de Pharmacie de Paris, Université de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, 75006 Paris, France; (X.Y.); (M.V.)
- Service Biologie du Médicament, Toxicologie, AP-HP, Hôpital Cochin, 75014 Paris, France
| | - Sylvain Broussy
- Faculté de Pharmacie de Paris, Université de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, 75006 Paris, France; (X.Y.); (M.V.)
| |
Collapse
|
12
|
Banerjee S, Wei Z, Tian T, Bose D, Shih NNC, Feldman MD, Khoury T, De Michele A, Robertson ES. Prognostic correlations with the microbiome of breast cancer subtypes. Cell Death Dis 2021; 12:831. [PMID: 34482363 PMCID: PMC8418604 DOI: 10.1038/s41419-021-04092-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/18/2022]
Abstract
Alterations to the natural microbiome are linked to different diseases, and the presence or absence of specific microbes is directly related to disease outcomes. We performed a comprehensive analysis with unique cohorts of the four subtypes of breast cancer (BC) characterized by their microbial signatures, using a pan-pathogen microarray strategy. The signature (includes viruses, bacteria, fungi, and parasites) of each tumor subtype was correlated with clinical data to identify microbes with prognostic potential. The subtypes of BC had specific viromes and microbiomes, with ER+ and TN tumors showing the most and least diverse microbiome, respectively. The specific microbial signatures allowed discrimination between different BC subtypes. Furthermore, we demonstrated correlations between the presence and absence of specific microbes in BC subtypes with the clinical outcomes. This study provides a comprehensive map of the oncobiome of BC subtypes, with insights into disease prognosis that can be critical for precision therapeutic intervention strategies.
Collapse
Affiliation(s)
- Sagarika Banerjee
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ, USA
| | - Tian Tian
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ, USA
| | - Dipayan Bose
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Natalie N C Shih
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael D Feldman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thaer Khoury
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Angela De Michele
- Division of Hematology Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erle S Robertson
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Kassa T. A Review on Human Orf: A Neglected Viral Zoonosis. Res Rep Trop Med 2021; 12:153-172. [PMID: 34267574 PMCID: PMC8275206 DOI: 10.2147/rrtm.s306446] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 06/04/2021] [Indexed: 12/30/2022] Open
Abstract
Orf virus (ORFV) is the etiologic agent of Orf or ecthyma contagiosum in humans but primarily affects different domestic and wild animals. The disease mostly affects sheep, goats and other small wild ruminants and spreads to humans through direct contact with infected animals or by way of contaminated fomites worldwide. ORFV is taxonomically classified as a member of the genus Parapoxvirus. It is known to have tolerance to inactivation in a drier environment, and it has been recovered from crusts after several months to years. Among immunocompetent people, the lesions usually resolve by its natural course within a maximum of 8 weeks. In immunosuppressed patients, however, it needs the use of various approaches including antiviral, immune modifier or minor surgical excisions. The virus through its association with divergent host ranges helps to develop a mechanism to evade the immune system. The relative emergence of Orf, diagnosed on clinical ground among human cases, in unusual frequencies in southwest Ethiopia between October 2019 and May 2020, was the driver to write this review. The objective was to increase health care providers' diagnostic curiosity and to bring the attentiveness of public health advisors for prevention, control and the development of schemes for surveillance of Orf zoonosis in a similar setting like Ethiopia.
Collapse
Affiliation(s)
- Tesfaye Kassa
- School of Medical Laboratory Science, Institute of Health, Jimma University, Jimma, Ethiopia
| |
Collapse
|
14
|
Künnapuu J, Bokharaie H, Jeltsch M. Proteolytic Cleavages in the VEGF Family: Generating Diversity among Angiogenic VEGFs, Essential for the Activation of Lymphangiogenic VEGFs. BIOLOGY 2021; 10:167. [PMID: 33672235 PMCID: PMC7926383 DOI: 10.3390/biology10020167] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/24/2022]
Abstract
Specific proteolytic cleavages turn on, modify, or turn off the activity of vascular endothelial growth factors (VEGFs). Proteolysis is most prominent among the lymph-angiogenic VEGF-C and VEGF-D, which are synthesized as precursors that need to undergo enzymatic removal of their C- and N-terminal propeptides before they can activate their receptors. At least five different proteases mediate the activating cleavage of VEGF-C: plasmin, ADAMTS3, prostate-specific antigen, cathepsin D, and thrombin. All of these proteases except for ADAMTS3 can also activate VEGF-D. Processing by different proteases results in distinct forms of the "mature" growth factors, which differ in affinity and receptor activation potential. The "default" VEGF-C-activating enzyme ADAMTS3 does not activate VEGF-D, and therefore, VEGF-C and VEGF-D do function in different contexts. VEGF-C itself is also regulated in different contexts by distinct proteases. During embryonic development, ADAMTS3 activates VEGF-C. The other activating proteases are likely important for non-developmental lymphangiogenesis during, e.g., tissue regeneration, inflammation, immune response, and pathological tumor-associated lymphangiogenesis. The better we understand these events at the molecular level, the greater our chances of developing successful therapies targeting VEGF-C and VEGF-D for diseases involving the lymphatics such as lymphedema or cancer.
Collapse
Affiliation(s)
- Jaana Künnapuu
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; (J.K.); (H.B.)
| | - Honey Bokharaie
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; (J.K.); (H.B.)
| | - Michael Jeltsch
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; (J.K.); (H.B.)
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Wihuri Research Institute, 00290 Helsinki, Finland
| |
Collapse
|
15
|
Koganti R, Suryawanshi R, Shukla D. Heparanase, cell signaling, and viral infections. Cell Mol Life Sci 2020; 77:5059-5077. [PMID: 32462405 PMCID: PMC7252873 DOI: 10.1007/s00018-020-03559-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/17/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022]
Abstract
Heparanase (HPSE) is a multifunctional protein endowed with many non-enzymatic functions and a unique enzymatic activity as an endo-β-D-glucuronidase. The latter allows it to serve as a key modulator of extracellular matrix (ECM) via a well-regulated cleavage of heparan sulfate side chains of proteoglycans at cell surfaces. The cleavage and associated changes at the ECM cause release of multiple signaling molecules with important cellular and pathological functions. New and emerging data suggest that both enzymatic as well as non-enzymatic functions of HPSE are important for health and illnesses including viral infections and virally induced cancers. This review summarizes recent findings on the roles of HPSE in activation, inhibition, or bioavailability of key signaling molecules such as AKT, VEGF, MAPK-ERK, and EGFR, which are known regulators of common viral infections in immune and non-immune cell types. Altogether, our review provides a unique overview of HPSE in cell-survival signaling pathways and how they relate to viral infections.
Collapse
Affiliation(s)
- Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA
| | - Rahul Suryawanshi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA.
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
16
|
Bose D, Banerjee S, Singh RK, Wise LM, Robertson ES. Vascular endothelial growth factor encoded by Parapoxviruses can regulate metabolism and survival of triple negative breast cancer cells. Cell Death Dis 2020; 11:996. [PMID: 33219203 PMCID: PMC7679371 DOI: 10.1038/s41419-020-03203-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022]
Abstract
Dysbiotic microbiomes are linked to many pathological outcomes including different metabolic disorders like diabetes, atherosclerosis and even cancer. Breast cancer is the second leading cause of cancer associated death in women, and triple negative breast cancer (TNBC) is the most aggressive type with major challenges for intervention. Previous reports suggested that Parapoxvirus signatures are one of the predominant dysbiotic viral signatures in TNBC. These viruses encode several genes that are homologs of human genes. In this study, we show that the VEGF homolog encoded by Parapoxviruses, can induce cell proliferation, and alter metabolism of breast cancer and normal breast cells, through alteration of MAPK-ERK and PI3K-AKT signaling. In addition, the activity of the transcription factor FoxO1 was altered by viral-encoded VEGF through activation of the PI3K-AKT pathway, leading to reprogramming of cellular metabolic gene expression. Therefore, this study provides new insights into the function of viral-encoded VEGFs, which promoted the growth of the breast cancer cells and imparted proliferative phenotype with altered metabolism in normal breast cells.
Collapse
Affiliation(s)
- Dipayan Bose
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, Tumor Virology Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sagarika Banerjee
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, Tumor Virology Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rajnish Kumar Singh
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, Tumor Virology Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lyn M Wise
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Erle S Robertson
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, Tumor Virology Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
17
|
Yu Y, Duan X, Liu Y, Ma J, Song B, Lian Z, Cui Y. Laboratory Diagnosis of a NZ7-like Orf Virus Infection and Pathogen Genetic Characterization, Particularly in the VEGF Gene. Front Vet Sci 2020; 7:538. [PMID: 33094105 PMCID: PMC7527421 DOI: 10.3389/fvets.2020.00538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 07/10/2020] [Indexed: 11/13/2022] Open
Abstract
Orf is a widespread contagious epithelial viral disease found particularly in most sheep breeding countries in the world. Recently, an orf virus (ORFV) strain OV-HLJ05 was isolated from an outbreak in northeast China. Three genes of interest including ORFV011 (B2L), ORFV059 (F1L), and ORFV132 (VEGF) of ORFV, were recruited to identify and genetically characterize this newly isolated virus. Amino acid (aa) sequence compared with the ORFV references listed in GenBank, both B2L and F1L of OV-HLJ05 showed less microheterogeneity from their references. In contrast, the VEGF gene was included in the NZ7-VEGF like group as previously considered by Mercer in 2002. Unexpectedly, further multiple VEGF matches were made, using 34 published sequences from China and India, resulting in 27 strains of the NZ7 members. Based on Karki's report in 2020, NZ7-VEGF like viruses are emerging more and more frequently in these two countries, damaging the Asian sheep industry. Obvious heterogeneity with the NZ2, insertion of two oligopeptides TATI(L)QVVVAI(L) and SSSS(S) motif were found in the NZ7-like VEGF protein. These VEGFs are divided mainly into two types and a significant increase in the number of hydrogen bonds within the NZ7-like VEGF dimers was observed. The NZ7-like ORFV apparently favors the goat as a host and an emphasis on this in future epidemiological and pathological studies should be considered, focusing on the NZ7-like virus.
Collapse
Affiliation(s)
- Yongzhong Yu
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
- *Correspondence: Yongzhong Yu
| | - Xuyang Duan
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yuanyuan Liu
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jinzhu Ma
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Baifen Song
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Zhengxing Lian
- College of Animal Science and Technology, China Agricultural University, Beijing, China
- Zhengxing Lian
| | - Yudong Cui
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
- Yudong Cui
| |
Collapse
|
18
|
Hao JH, Kong HJ, Yan MH, Shen CC, Xu GW, Zhang DJ, Zhang KS, Zheng HX, Liu XT. Inhibition of orf virus replication in goat skin fibroblast cells by the HSPA1B protein, as demonstrated by iTRAQ-based quantitative proteome analysis. Arch Virol 2020; 165:2561-2587. [PMID: 32876795 PMCID: PMC7465882 DOI: 10.1007/s00705-020-04789-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 07/26/2020] [Indexed: 02/07/2023]
Abstract
Orf virus (ORFV) infects sheep and goat tissues, resulting in severe proliferative lesions. To analyze cellular protein expression in ORFV-infected goat skin fibroblast (GSF) cells, we used two-dimensional liquid chromatography-tandem mass spectrometry coupled with isobaric tags for relative and absolute quantification (iTRAQ). The proteomics approach was used along with quantitative reverse transcription polymerase chain reaction (RT-qPCR) to detect differentially expressed proteins in ORFV-infected GSF cells and mock-infected GSF cells. A total of 282 differentially expressed proteins were identified. It was found that 222 host proteins were upregulated and 60 were downregulated following viral infection. We confirmed that these proteins were differentially expressed and found that heat shock 70-kDa protein 1B (HSPA1B) was differentially expressed and localized in the cytoplasm. It was also noted that HSPA1B caused inhibition of viral proliferation, in the middle and late stages of viral infection. The differentially expressed proteins were associated with the biological processes of viral binding, cell structure, signal transduction, cell adhesion, and cell proliferation.
Collapse
Affiliation(s)
- Jun-Hong Hao
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Han-Jin Kong
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Ming-Hao Yan
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Chao-Chao Shen
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Guo-Wei Xu
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Da-Jun Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Ke-Shan Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China.
| | - Hai-Xue Zheng
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Xiang-Tao Liu
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| |
Collapse
|
19
|
The Bartonella autotransporter BafA activates the host VEGF pathway to drive angiogenesis. Nat Commun 2020; 11:3571. [PMID: 32678094 PMCID: PMC7366657 DOI: 10.1038/s41467-020-17391-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/26/2020] [Indexed: 12/29/2022] Open
Abstract
Pathogenic bacteria of the genus Bartonella can induce vasoproliferative lesions during infection. The underlying mechanisms are unclear, but involve secretion of an unidentified mitogenic factor. Here, we use functional transposon-mutant screening in Bartonella henselae to identify such factor as a pro-angiogenic autotransporter, called BafA. The passenger domain of BafA induces cell proliferation, tube formation and sprouting of microvessels, and drives angiogenesis in mice. BafA interacts with vascular endothelial growth factor (VEGF) receptor-2 and activates the downstream signaling pathway, suggesting that BafA functions as a VEGF analog. A BafA homolog from a related pathogen, Bartonella quintana, is also functional. Our work unveils the mechanistic basis of vasoproliferative lesions observed in bartonellosis, and we propose BafA as a key pathogenic factor contributing to bacterial spread and host adaptation. Pathogenic bacteria of the genus Bartonella can induce vasoproliferative lesions during infection. Here, Tsukamoto et al. show that this effect is caused by a secreted protein that induces cell proliferation and angiogenesis by acting as an analog of the host’s vascular endothelial growth factor (VEGF).
Collapse
|
20
|
Gallina L, Savini F, Casà G, Bertoletti I, Bianchi A, Gibelli LR, Lelli D, Lavazza A, Scagliarini A. Epitheliotropic Infections in Wildlife Ruminants From the Central Alps and Stelvio National Park. Front Vet Sci 2020; 7:229. [PMID: 32426384 PMCID: PMC7203578 DOI: 10.3389/fvets.2020.00229] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/03/2020] [Indexed: 12/21/2022] Open
Abstract
The mountain chain of the Alps, represents the habitat of alpine fauna where the red deer (Cervus elaphus) population is the outmost numerous, followed by the chamois (Rupicapra rupicapra) and the alpine ibex (Capra ibex) at higher altitudes. Previous reports showed the circulation of epitheliotropic viruses, belonging to the families Papillomaviridae and Poxviridae, causing skin and mucosal lesions in wild ruminants of the Stelvio National Park, situated in the area. To deepen our knowledge on the natural dynamics of the infections, a passive surveillance on all the cases of proliferative skin and mucosal lesions in wild ruminants was performed. Twenty-seven samples (11 chamois, 10 red deer and 6 ibex) collected from 2008 to 2018 were analyzed by negative staining electron microscopy, histology, and PCR followed by genome sequencing and phylogenetic analyses. Results confirmed the spread of Parapoxvirus of Red Deer in New Zealand (PVNZ) in Italy, and its ability to cause severe lesions i.e., erosions and ulcers in the mouth. We showed for the first time a PVNZ/CePV1v (C. elaphus papillomavirus 1 variant) co-infection identified in one red deer. This result supports previous evidence on the ability of papillomavirus and parapoxvirus to mutually infect the same host tissue. Interestingly two ibex and one chamois showing orf virus (OV) skin lesions were shown to be co-infected with bovine papillomavirus type 1 and 2. The presence of bovine papillomavirus, in orf virus induced lesions of chamois and ibex raises the question of its pathogenetic role in these animal species. For the first time, OV/CePV1v co-infection was demonstrated in another chamois. CePV1v is sporadically reported in red deer throughout Europe and is considered species specific, its identification in a chamois suggests its ability of cross-infecting different animal species. Poxviruses and papillomavirus have been simultaneously detected also in the skin lesions of cattle, bird and human suggesting a possible advantageous interaction between these viruses. Taken together, our findings add further information on the epidemiology and pathogenetic role of epitheliotropic viruses in wild ruminants living in the central Alps and in Stelvio National Park.
Collapse
Affiliation(s)
- Laura Gallina
- Dipartimento di Scienze Mediche Veterinarie, Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| | - Federica Savini
- Dipartimento di Scienze Mediche Veterinarie, Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| | - Giovanni Casà
- Dipartimento di Scienze Mediche Veterinarie, Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| | - Irene Bertoletti
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Brescia, Italy
| | - Alessandro Bianchi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Brescia, Italy
| | - Lucia Rita Gibelli
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Brescia, Italy
| | - Davide Lelli
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Brescia, Italy
| | - Antonio Lavazza
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Brescia, Italy
| | - Alessandra Scagliarini
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale, Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| |
Collapse
|
21
|
Shimizu K, Takaiwa A, Takeshima SN, Okada A, Inoshima Y. Genetic Variability of 3'-Proximal Region of Genomes of Orf Viruses Isolated From Sheep and Wild Japanese Serows ( Capricornis crispus) in Japan. Front Vet Sci 2020; 7:188. [PMID: 32391386 PMCID: PMC7193945 DOI: 10.3389/fvets.2020.00188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/23/2020] [Indexed: 12/04/2022] Open
Abstract
Orf virus is a prototype species of the genus Parapoxvirus, subfamily Chordopoxvirinae, family Poxviridae. Japanese orf viruses, infecting sheep and wild Japanese serows (Capricornis crispus), have been considered to be genetically closely related based on the sequence identities of the open reading frames (ORFs) 11, 20, and 132 in their genomes. However, since the genome size of orf viruses is about 140 kbp long, genetic variation among Japanese orf viruses remains unclear. In this study, we analyzed the sequences of ORFs 117, 119, 125, and 127 located in the 3′-proximal region of the viral genome using two strains from sheep and three strains from Japanese serows isolated from 1970 to 2007, and compared them with the corresponding sequences of reference orf viruses from other countries. Sequence analysis revealed that ORFs 125 and 127, which encode the inhibitor of apoptosis and viral interleukin (IL)-10, respectively, were highly conserved among the five Japanese orf viruses. However, high genetic variability with deletions or duplications was observed in ORFs 117 and 119, which encode granulocyte macrophage colony-stimulating factor and IL-2 inhibition factor (GIF), and inducer of cell apoptosis, respectively, in one strain from sheep and two strains from Japanese serows. Our results suggest that genetic variability exists in Japanese orf viruses even in the same host species. This is the first report of genetic variability of orf viruses in Japan.
Collapse
Affiliation(s)
- Kaori Shimizu
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Gifu University, Gifu, Japan
| | - Asari Takaiwa
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Gifu University, Gifu, Japan
| | | | - Ayaka Okada
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Gifu University, Gifu, Japan.,Education and Research Center for Food Animal Health, Gifu University (GeFAH), Gifu, Japan
| | - Yasuo Inoshima
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Gifu University, Gifu, Japan.,Education and Research Center for Food Animal Health, Gifu University (GeFAH), Gifu, Japan.,The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan.,Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| |
Collapse
|
22
|
Yaron JR, Zhang L, Guo Q, Burgin M, Schutz LN, Awo E, Wise L, Krause KL, Ildefonso CJ, Kwiecien JM, Juby M, Rahman MM, Chen H, Moyer RW, Alcami A, McFadden G, Lucas AR. Deriving Immune Modulating Drugs from Viruses-A New Class of Biologics. J Clin Med 2020; 9:E972. [PMID: 32244484 PMCID: PMC7230489 DOI: 10.3390/jcm9040972] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Viruses are widely used as a platform for the production of therapeutics. Vaccines containing live, dead and components of viruses, gene therapy vectors and oncolytic viruses are key examples of clinically-approved therapeutic uses for viruses. Despite this, the use of virus-derived proteins as natural sources for immune modulators remains in the early stages of development. Viruses have evolved complex, highly effective approaches for immune evasion. Originally developed for protection against host immune responses, viral immune-modulating proteins are extraordinarily potent, often functioning at picomolar concentrations. These complex viral intracellular parasites have "performed the R&D", developing highly effective immune evasive strategies over millions of years. These proteins provide a new and natural source for immune-modulating therapeutics, similar in many ways to penicillin being developed from mold or streptokinase from bacteria. Virus-derived serine proteinase inhibitors (serpins), chemokine modulating proteins, complement control, inflammasome inhibition, growth factors (e.g., viral vascular endothelial growth factor) and cytokine mimics (e.g., viral interleukin 10) and/or inhibitors (e.g., tumor necrosis factor) have now been identified that target central immunological response pathways. We review here current development of virus-derived immune-modulating biologics with efficacy demonstrated in pre-clinical or clinical studies, focusing on pox and herpesviruses-derived immune-modulating therapeutics.
Collapse
Affiliation(s)
- Jordan R. Yaron
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Liqiang Zhang
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Qiuyun Guo
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Michelle Burgin
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Lauren N. Schutz
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Enkidia Awo
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Lyn Wise
- University of Otago, Dunedin 9054, New Zealand; (L.W.); (K.L.K.)
| | - Kurt L. Krause
- University of Otago, Dunedin 9054, New Zealand; (L.W.); (K.L.K.)
| | | | - Jacek M. Kwiecien
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S4L8, Canada
| | - Michael Juby
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Masmudur M. Rahman
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Hao Chen
- The Department of Tumor Surgery, Second Hospital of Lanzhou University, Lanzhou 730030, China;
| | - Richard W. Moyer
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA;
| | - Antonio Alcami
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Cantoblanco, 28049 Madrid, Spain;
| | - Grant McFadden
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Alexandra R. Lucas
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
- St Joseph Hospital, Dignity Health, Creighton University, Phoenix, AZ 85013, USA
| |
Collapse
|
23
|
Karki M, Kumar A, Arya S, Venkatesan G. Circulation of orf viruses containing the NZ7-like vascular endothelial growth factor (VEGF-E) gene type in India. Virus Res 2020; 281:197908. [PMID: 32126295 DOI: 10.1016/j.virusres.2020.197908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/25/2020] [Accepted: 02/28/2020] [Indexed: 10/24/2022]
Abstract
Orf, a poxviral skin infection of small ruminants is caused by orf virus (ORFV) of the genus Parapoxvirus of the Poxviridae family. Vascular endothelial growth factor (VEGF) is an important virulence factor that is responsible for proliferative lesions in parapoxviral infections. VEGF gene shows high intra- and inter-species variability. Two variants of VEGF have been described globally in ORFV, viz. NZ2- and NZ7-like. In the present study, ORFV isolates of different geographic regions of India were analysed on the basis of the VEGF gene. Indian ORFV isolates showed 95.7-100 % nucleotide (nt) and 78.4-99.3 % amino acid (aa) identity with each other, except ORFV-Assam/LK/14 and ORFV-Meghalaya/03 which shared 85.1-88.35 % and 79.1-81.8 % identity, at nt and aa levels, respectively with other Indian ORFV isolates. All Indian ORFVs under the study demonstrated 83.5-99.1 % nt and 80.5-97.9 % aa identity with NZ7-like VEGF as compared to 41.2-44.8 % nt and 30.7-38.4 % aa identity with NZ2-like VEGF on comparison with global ORFV strains. Phylogenetic analysis based on the VEGF gene showed two clusters of ORFV in which the Indian ORFVs clustered with NZ7-like VEGF from global ORFV strains, mostly from China. Despite the considerable variation, VEGF protein from Indian ORFV strains showed conserved VEGF homology domain with eight cysteine residues. Homology modeling of Indian ORFV strains predicted the presence of extended Loop 3 similar to NZ7-like VEGF. Therefore, the present study showed the circulation of ORFV strains with comparatively less variable NZ7-like VEGF in India which implicates its importance in the epidemiology of ORFV infections in the country.
Collapse
Affiliation(s)
- Monu Karki
- Division of Virology, ICAR- Indian Veterinary Research Institute, Mukteswar, Uttarakhand, India
| | - Amit Kumar
- Division of Virology, ICAR- Indian Veterinary Research Institute, Mukteswar, Uttarakhand, India
| | - Sargam Arya
- Division of Virology, ICAR- Indian Veterinary Research Institute, Mukteswar, Uttarakhand, India
| | - Gnanavel Venkatesan
- Division of Virology, ICAR- Indian Veterinary Research Institute, Mukteswar, Uttarakhand, India.
| |
Collapse
|
24
|
Pang F, Wang X, Chen Z, Zhang Z, Zhang M, Wang C, Yang X, An Q, Du L, Wang F. Integrated Analysis of Differentially Expressed miRNAs and mRNAs in Goat Skin Fibroblast Cells in Response to Orf Virus Infection Reveals That cfa-let-7a Regulates Thrombospondin 1 Expression. Viruses 2020; 12:v12010118. [PMID: 31963559 PMCID: PMC7019303 DOI: 10.3390/v12010118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 12/30/2022] Open
Abstract
Orf is a zoonotic disease that has caused huge economic losses globally. Systematical analysis of dysregulated cellular micro RNAs (miRNAs) in response to Orf virus (ORFV) infection has not been reported. In the current study, miRNA sequencing and RNA sequencing (RNA-seq) were performed in goat skin fibroblast (GSF) cells at 0, 18, and 30 h post infection (h.p.i). We identified 140 and 221 differentially expressed (DE) miRNAs at 18 and 30 h.p.i, respectively. We also identified 729 and 3961 DE genes (DEGs) at 18 and 30 h.p.i, respectively. GO enrichment analysis indicates enrichment of apoptotic regulation, defense response to virus, immune response, and inflammatory response at both time points. DE miRNAs and DEGs with reverse expression were used to construct miRNA-gene networks. Seven DE miRNAs and seven DEGs related to "negative regulation of viral genome replication" were identified. These were validated by RT-qPCR. Cfa-let-7a, a significantly upregulated miRNA, was found to repress Thrombospondin 1 (THBS1) mRNA and protein expression by directly targeting the THBS1 3' untranslated region. THBS1 has been reported to induce apoptosis; therefore, the cfa-let-7a-THBS1 axis may play an important role in cellular apoptosis during ORFV infection. This study provides new insights into ORFV and host cell interaction mechanisms.
Collapse
Affiliation(s)
- Feng Pang
- College of Animal Science and Technology, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Haikou 570228, China; (F.P.); (Z.C.); (Z.Z.); (M.Z.); (C.W.); (X.Y.); (Q.A.); (L.D.)
| | - Xinying Wang
- Guizhou Institute of Technology, Guiyang, 550003, China;
| | - Zhen Chen
- College of Animal Science and Technology, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Haikou 570228, China; (F.P.); (Z.C.); (Z.Z.); (M.Z.); (C.W.); (X.Y.); (Q.A.); (L.D.)
| | - Zhenxing Zhang
- College of Animal Science and Technology, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Haikou 570228, China; (F.P.); (Z.C.); (Z.Z.); (M.Z.); (C.W.); (X.Y.); (Q.A.); (L.D.)
| | - Mengmeng Zhang
- College of Animal Science and Technology, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Haikou 570228, China; (F.P.); (Z.C.); (Z.Z.); (M.Z.); (C.W.); (X.Y.); (Q.A.); (L.D.)
| | - Chengqiang Wang
- College of Animal Science and Technology, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Haikou 570228, China; (F.P.); (Z.C.); (Z.Z.); (M.Z.); (C.W.); (X.Y.); (Q.A.); (L.D.)
| | - Xiaohong Yang
- College of Animal Science and Technology, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Haikou 570228, China; (F.P.); (Z.C.); (Z.Z.); (M.Z.); (C.W.); (X.Y.); (Q.A.); (L.D.)
| | - Qi An
- College of Animal Science and Technology, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Haikou 570228, China; (F.P.); (Z.C.); (Z.Z.); (M.Z.); (C.W.); (X.Y.); (Q.A.); (L.D.)
| | - Li Du
- College of Animal Science and Technology, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Haikou 570228, China; (F.P.); (Z.C.); (Z.Z.); (M.Z.); (C.W.); (X.Y.); (Q.A.); (L.D.)
| | - Fengyang Wang
- College of Animal Science and Technology, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Haikou 570228, China; (F.P.); (Z.C.); (Z.Z.); (M.Z.); (C.W.); (X.Y.); (Q.A.); (L.D.)
- Correspondence:
| |
Collapse
|
25
|
Sharif S, Ueda N, Nakatani Y, Wise LM, Clifton S, Lateef Z, Mercer AA, Fleming SB. Chemokine-Binding Proteins Encoded by Parapoxvirus of Red Deer of New Zealand Display Evidence of Gene Duplication and Divergence of Ligand Specificity. Front Microbiol 2019; 10:1421. [PMID: 31293551 PMCID: PMC6603201 DOI: 10.3389/fmicb.2019.01421] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/05/2019] [Indexed: 01/20/2023] Open
Abstract
Parapoxvirus of red deer in New Zealand (PVNZ) is a species of the Parapoxvirus genus that causes pustular dermatitis. We identified a cluster of genes in PVNZ that encode three unique chemokine-binding proteins (CBPs) namely ORF112.0, ORF112.3 and ORF112.6. Chemokines are a large family of molecules that direct cell trafficking to sites of inflammation and through lymphatic organs. The PVNZ-CBPs were analyzed by surface plasmon resonance against a broad spectrum of CXC, CC, XC and CX3C chemokines and were found to differ in their specificity and binding affinity. ORF112.0 interacted with chemokines from the CXC, CC and XC classes of chemokines with nM affinities. The ORF112.3 showed a preference for CXC chemokines, while ORF112.6 showed pM affinity binding for CC chemokines. Structural modeling analysis showed alterations in the chemokine binding sites of the CBPs, although the core structure containing two ß-sheets and three α-helices being conserved with the other parapoxvirus CBPs. Chemotaxis assays using neutrophils and monocytes revealed inhibitory impact of the CBPs on cell migration. Our results suggest that the PVNZ-CBPs are likely to have evolved through a process of gene duplication and divergence, and may have a role in suppressing inflammation and the anti-viral immune response.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Stephen B. Fleming
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
26
|
Wang Z, Li S, Yu Y, Yu K, Zhang X, Xiang J, Li F. Identification and characterization of two novel vascular endothelial growth factor genes in Litopenaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2019; 84:259-268. [PMID: 30308291 DOI: 10.1016/j.fsi.2018.10.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/05/2018] [Accepted: 10/07/2018] [Indexed: 06/08/2023]
Abstract
Vascular endothelial growth factor (VEGF) signaling pathway induces endothelial cell proliferation, promotes cell migration, and inhibits apoptosis. Although three VEGF and two VEGF receptor genes have been identified in Litopenaeus vannamei and demonstrated their roles in WSSV infection, another two novel VEGF genes (LvVEGF4, LvVEGF5) were isolated and their involvements in the WSSV infection of shrimp were studied in the present study. The deduced amino acid sequences of both LvVEGF4 and LvVEGF5 contained a signal peptide, a typical PDGF/VEGF domain and a cysteine knot motif (CXCXCX). Tissue distribution analysis showed that LvVEGF4 was predominantly expressed in gill and hemocytes, while LvVEGF5 was mainly detected in hemocytes and intestine. WSSV infection could cause up-regulation of the transcriptional levels of LvVEGF4 and LvVEGF5. Their functions were studied by double-strand RNA interference. The results showed that knock-down of LvVEGF4 and LvVEGF5 led to a decrease of the viral copy number in WSSV infected shrimp. Yeast two-hybrid analysis showed that both LvVEGF4 and LvVEGF5 could interact with LvVEGFR1 rather than LvVEGFR2. In addition, knock-down of LvVEGF4 and LvVEGF5 could reduce the expressional levels of downstream genes FAK and PI3K. The present study provides new clues in demonstrating that the VEGF signaling pathway is involved in the process of WSSV infection in shrimp.
Collapse
Affiliation(s)
- Zhiwei Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Shihao Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, PR China.
| | - Yang Yu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, PR China
| | - Kuijie Yu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, PR China
| | - Xiaojun Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, PR China
| | - Jianhai Xiang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, PR China
| | - Fuhua Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, PR China.
| |
Collapse
|
27
|
Abstract
Receptor tyrosine kinases (RTKs) are essential components of cell communication pathways utilized from the embryonic to adult stages of life. These transmembrane receptors bind polypeptide ligands, such as growth factors, inducing signalling cascades that control cellular processes such as proliferation, survival, differentiation, motility and inflammation. Many viruses have acquired homologs of growth factors encoded by the hosts that they infect. Production of growth factors during infection allows viruses to exploit RTKs for entry and replication in cells, as well as for host and environmental dissemination. This review describes the genetic diversity amongst virus-derived growth factors and the mechanisms by which RTK exploitation enhances virus survival, then highlights how viral ligands can be used to further understanding of RTK signalling and function during embryogenesis, homeostasis and disease scenarios.
Collapse
Affiliation(s)
- Zabeen Lateef
- a Department of Pharmacology and Toxicology, School of Biomedical Sciences , University of Otago , Dunedin , New Zealand
| | - Lyn M Wise
- a Department of Pharmacology and Toxicology, School of Biomedical Sciences , University of Otago , Dunedin , New Zealand
| |
Collapse
|
28
|
VEGF Upregulation in Viral Infections and Its Possible Therapeutic Implications. Int J Mol Sci 2018; 19:ijms19061642. [PMID: 29865171 PMCID: PMC6032371 DOI: 10.3390/ijms19061642] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 12/12/2022] Open
Abstract
Several viruses are recognized as the direct or indirect causative agents of human tumors and other severe human diseases. Vascular endothelial growth factor (VEGF) is identified as a principal proangiogenic factor that enhances the production of new blood vessels from existing vascular network. Therefore, oncogenic viruses such as Kaposi’s sarcoma herpesvirus (KSHV) and Epstein-Barr virus (EBV) and non-oncogenic viruses such as herpes simplex virus (HSV-1) and dengue virus, which lack their own angiogenic factors, rely on the recruitment of cellular genes for angiogenesis in tumor progression or disease pathogenesis. This review summarizes how human viruses exploit the cellular signaling machinery to upregulate the expression of VEGF and benefit from its physiological functions for their own pathogenesis. Understanding the interplay between viruses and VEGF upregulation will pave the way to design targeted and effective therapeutic approaches for viral oncogenesis and severe diseases.
Collapse
|
29
|
Guo S, Martin MG, Tian C, Cui J, Wang L, Wu S, Gu W. Evaluation of Detection Methods and Values of Circulating Vascular Endothelial Growth Factor in Lung Cancer. J Cancer 2018; 9:1287-1300. [PMID: 29675110 PMCID: PMC5907677 DOI: 10.7150/jca.22020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 01/19/2018] [Indexed: 01/08/2023] Open
Abstract
Lung cancer is the deadliest cancer in the world. Angiogenesis plays a crucial role of the incidence, progression, and metastasis in lung cancer. Angiogenesis inhibitors are used to treat non-small cell lung cancer (NSCLC) patients, and the molecular biomarkers are also being assessed to predict treatment response/therapeutic response and patients' prognosis. Vascular endothelial growth factor (VEGF) is a signal protein produced by cells that stimulates angiogenesis. Due to its predictive values of prognosis on NSCLC, a large number of methods have been developed and evaluated to detect VEGF levels in a variety of studies. In this article, we review the detection methods designed to measure the VEGF levels in different body fluids and prognosticate the value of VEGF in treatment, diagnosis and survival in lung cancer.
Collapse
Affiliation(s)
- Sumin Guo
- Department of Oncology, Hebei Chest Hospital, Lung Cancer Control and Prevention Center of Hebei Province, Shijiazhuang, Hebei, 050041, China.,Department of Orthopaedic Surgery- Campbell Clinic and Pathology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Michael G Martin
- West Cancer Center, University of Tennessee Health Science Center, Memphis, Tennessee, 38163, USA
| | - Cheng Tian
- Department of Orthopaedic Surgery- Campbell Clinic and Pathology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Jinglin Cui
- Department of Orthopaedic Surgery- Campbell Clinic and Pathology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Center of Integrative Research, The First Hospital of Qiqihaer City, Qiqihaer, Heilongjiang, 161005, PR China
| | - Lishi Wang
- Department of Basic Medicine (Basic Medical Research), Inner Mongolia Medical University, Inner Mongolia, 010110, PR China
| | - Shucai Wu
- Department of Oncology, Hebei Chest Hospital, Lung Cancer Control and Prevention Center of Hebei Province, Shijiazhuang, Hebei, 050041, China
| | - Weikuan Gu
- Department of Orthopaedic Surgery- Campbell Clinic and Pathology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Research Service, Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis TN 38104, USA
| |
Collapse
|
30
|
Span PN, Grebenchtchikov N, Geurts-Moespot J, Westphal JR, Lucassen AM, Sweep CG. EORTC Receptor and Biomarker Study Group Report: A Sandwich Enzyme-Linked Immunosorbent Assay for Vascular Endothelial Growth Factor in Blood and Tumor Tissue Extracts. Int J Biol Markers 2018; 15:184-91. [PMID: 10883894 DOI: 10.1177/172460080001500210] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A four-antibody sandwich enzyme-linked immunosorbent assay (ELISA) for vascular endothelial growth factor (VEGF) for application in blood (serum and plasma) and tumor tissue extracts was set up within the framework of the EORTC Receptor and Biomarker Study Group (RBSG). Polyclonal antibodies against VEGF165 were raised in chickens and rabbits, and used in a previously described assay format. The assay was validated and characterized for use in serum, plasma and tumor tissue extracts. The resulting VEGF ELISA was found to be specific for VEGF165 and VEGF121, the main isoforms of VEGF. The assay showed good precision and parallelism in serial dilutions of samples. The assay was not susceptible to interference by heterophilic antibodies because avian antibodies (duck anti-chicken and chicken anti-VEGF) were used in the pre-analyte stage and mammalian antibodies (rabbit anti-VEGF and goat anti-rabbit) in the post-analyte stage. In conclusion, a sensitive, robust and specific VEGF ELISA has been developed. Research into the prognostic value of VEGF employing this assay is currently underway.
Collapse
Affiliation(s)
- P N Span
- Department of Chemical Endocrinology, University Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
We recovered the first full-length poxvirus genome, including the terminal hairpin region, directly from complex clinical material using a combination of second generation short read and third generation nanopore sequencing technologies. The complete viral genome sequence was directly recovered from a skin lesion of a grey seal thereby preventing sequence changes due to in vitro passaging of the virus. Subsequent analysis of the proteins encoded by this virus identified genes specific for skin adaptation and pathogenesis of parapoxviruses. These data warrant the classification of seal parapoxvirus, tentatively designated SePPV, as a new species within the genus Parapoxvirus.
Collapse
|
32
|
Bergqvist C, Kurban M, Abbas O. Orf virus infection. Rev Med Virol 2017; 27. [DOI: 10.1002/rmv.1932] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/17/2017] [Accepted: 03/13/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Christina Bergqvist
- Dermatology Department; American University of Beirut Medical Center; Beirut Lebanon
| | - Mazen Kurban
- Dermatology Department; American University of Beirut Medical Center; Beirut Lebanon
| | - Ossama Abbas
- Dermatology Department; American University of Beirut Medical Center; Beirut Lebanon
| |
Collapse
|
33
|
Fleming SB, McCaughan C, Lateef Z, Dunn A, Wise LM, Real NC, Mercer AA. Deletion of the Chemokine Binding Protein Gene from the Parapoxvirus Orf Virus Reduces Virulence and Pathogenesis in Sheep. Front Microbiol 2017; 8:46. [PMID: 28174562 PMCID: PMC5258736 DOI: 10.3389/fmicb.2017.00046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/06/2017] [Indexed: 12/25/2022] Open
Abstract
Orf virus (ORFV) is the type species of the Parapoxvirus genus of the family Poxviridae and infects sheep and goats, often around the mouth, resulting in acute pustular skin lesions. ORFV encodes several secreted immunomodulators including a broad-spectrum chemokine binding protein (CBP). Chemokines are a large family of secreted chemotactic proteins that activate and regulate inflammation induced leukocyte recruitment to sites of infection. In this study we investigated the role of CBP in vivo in the context of ORFV infection of sheep. The CBP gene was deleted from ORFV strain NZ7 and infections of sheep used to investigate the effect of CBP on pathogenesis. Animals were either infected with the wild type (wt) virus, CBP-knockout virus or revertant strains. Sheep were infected by scarification on the wool-less area of the hind legs at various doses of virus. The deletion of the CBP gene severely attenuated the virus, as only few papules formed when animals were infected with the CBP-knock-out virus at the highest dose (107 p.f.u). In contrast, large pustular lesions formed on almost all animals infected with the wt and revertant strains at 107 p.f.u. The lesions for the CBP-knock-out virus resolved approximately 5–6 days p.i, at a dose of 107 pfu whereas in animals infected with the wt and revertants at this dose, lesions began to resolve at approximately 10 days p.i. Few pustules developed at the lowest dose of 103 p.f.u. for all viruses. Immunohistochemistry of biopsy skin-tissue from pustules showed that the CBP-knockout virus replicated in all animals at the highest dose and was localized to the skin epithelium while haematoxylin and eosin staining showed histological features of the CBP-knockout virus typical of the parent virus with acanthosis, elongated rete ridges and orthokeratotic hyperkeratosis. MHC-II immunohistochemistry analysis for monocytes and dendritic cells showed greater staining within the papillary dermis of the CBP-knock-out virus compared with the revertant viruses, however this was not the case with the wt where staining was similar. Our results show that the CBP gene encodes a secreted immunodulator that has a critical role in virulence and pathogenesis.
Collapse
Affiliation(s)
- Stephen B Fleming
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| | - Catherine McCaughan
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| | - Zabeen Lateef
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| | - Amy Dunn
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| | - Lyn M Wise
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| | - Nicola C Real
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| | - Andrew A Mercer
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| |
Collapse
|
34
|
Structural basis of GM-CSF and IL-2 sequestration by the viral decoy receptor GIF. Nat Commun 2016; 7:13228. [PMID: 27819269 PMCID: PMC5103067 DOI: 10.1038/ncomms13228] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 09/14/2016] [Indexed: 12/22/2022] Open
Abstract
Subversion of the host immune system by viruses is often mediated by molecular decoys that sequester host proteins pivotal to mounting effective immune responses. The widespread mammalian pathogen parapox Orf virus deploys GIF, a member of the poxvirus immune evasion superfamily, to antagonize GM-CSF (granulocyte macrophage colony-stimulating factor) and IL-2 (interleukin-2), two pleiotropic cytokines of the mammalian immune system. However, structural and mechanistic insights into the unprecedented functional duality of GIF have remained elusive. Here we reveal that GIF employs a dimeric binding platform that sequesters two copies of its target cytokines with high affinity and slow dissociation kinetics to yield distinct complexes featuring mutually exclusive interaction footprints. We illustrate how GIF serves as a competitive decoy receptor by leveraging binding hotspots underlying the cognate receptor interactions of GM-CSF and IL-2, without sharing any structural similarity with the cytokine receptors. Our findings contribute to the tracing of novel molecular mimicry mechanisms employed by pathogenic viruses. Viruses often subvert the host immune system using molecular decoys to prevent an effective immune response. Here, the authors examine the structural details of the viral decoy receptor GIF and its antagnosim of GM-CSF and IL-2.
Collapse
|
35
|
|
36
|
Fagiani E, Lorentz P, Bill R, Pavotbawan K, Kopfstein L, Christofori G. VEGF receptor-2-specific signaling mediated by VEGF-E induces hemangioma-like lesions in normal and in malignant tissue. Angiogenesis 2016; 19:339-58. [PMID: 27038485 DOI: 10.1007/s10456-016-9508-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/22/2016] [Indexed: 12/24/2022]
Abstract
UNLABELLED Viral VEGF-E (ovVEGF-E), a homolog of VEGF-A, was discovered in the genome of Orf virus. Together with VEGF-A, B, C, D, placental growth factor (PlGF) and snake venom VEGF (svVEGF), ovVEGF-E is a member of the VEGF family of potent angiogenesis factors with a bioactivity similar to VEGF-A it induces proliferation, migration and sprouting of cultured vascular endothelial cells and proliferative lesions in the skin of sheep, goat and man that are characterized by massive capillary proliferation and dilation. These biological functions are mediated exclusively via its interaction with VEGF receptor-2 (VEGFR-2). Here, we have generated transgenic mice specifically expressing ovVEGF-E in β-cells of the endocrine pancreas (Rip1VEGF-E; RVE). RVE mice show an increase in number and size of the islets of Langerhans and a distorted organization of insulin and glucagon-expressing cells. Islet endothelial cells of RVE mice hyper-proliferate and form increased numbers of functional blood vessels. In addition, the formation of disorganized lymphatic vessels and increased immune cell infiltration is observed. Upon crossing RVE single-transgenic mice with Rip1Tag2 (RT2) transgenic mice, a well-studied model of pancreatic β-cell carcinogenesis, double-transgenic mice (RT2;RVE) display hyper-proliferation of endothelial cells resulting in the formation of hemangioma-like lesions. In addition, RT2;RVE mice exhibit activated lymphangiogenesis at the tumor periphery and increased neutrophil and macrophage tumor infiltration and micro-metastasis to lymph nodes and lungs. These phenotypes markedly differ from the phenotypes observed with the transgenic expression of the other VEGF family members in β-cells of normal mice and of RT2 mice.
Collapse
Affiliation(s)
- Ernesta Fagiani
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058, Basel, Switzerland.
| | - Pascal Lorentz
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058, Basel, Switzerland
| | - Ruben Bill
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058, Basel, Switzerland
| | - Kirusigan Pavotbawan
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058, Basel, Switzerland
| | - Lucie Kopfstein
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058, Basel, Switzerland
| | - Gerhard Christofori
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058, Basel, Switzerland
| |
Collapse
|
37
|
Wang Z, Li S, Li F, Yang H, Yang F, Xiang J. Characterization of two types of vascular endothelial growth factor from Litopenaeus vannamei and their involvements during WSSV infection. FISH & SHELLFISH IMMUNOLOGY 2015; 47:824-832. [PMID: 26492995 DOI: 10.1016/j.fsi.2015.10.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/24/2015] [Accepted: 10/17/2015] [Indexed: 06/05/2023]
Abstract
Vascular endothelial growth factors (VEGFs) are important signaling proteins in VEGF signaling pathway which play key roles in inducing endothelial cell proliferation, migration, angiogenesis, vascular permeability, inhibition of apoptosis and virus infection. In the present study, we isolated and characterized two VEGF genes, LvVEGF1 and LvVEGF2 from Litopenaeus vannamei. The deduced amino acid sequences of both LvVEGF1 and LvVEGF2 contained a signal peptide, a typical PDGF/VEGF domain and a cysteine knot motif (CXCXC). Tissue distribution analysis showed that LvVEGF1 was predominantly expressed in lymphoid organ (Oka) while LvVEGF2 was mainly detected in gill and hemocytes. The transcriptional levels of LvVEGF1 in Oka and LvVEGF2 in gill or hemocytes were apparently up-regulated during WSSV infection. Double-stranded RNA interference was used for further functional studies. The data showed that silencing of LvVEGF1 and LvVEGF2 caused a decrease of the copy numbers of the virus in WSSV infected shrimp and a reduction of the cumulative mortality rate of shrimp during WSSV infection. The present study indicated that LvVEGF1 and LvVEGF2 might facilitate WSSV infection, which provided new evidence to understand the function of VEGF signaling pathway during WSSV infection in shrimp.
Collapse
Affiliation(s)
- Zhiwei Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shihao Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; National & Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao 266071, China
| | - Fuhua Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; National & Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao 266071, China.
| | - Hui Yang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fusheng Yang
- Hangzhou Xiaoshan Donghai Aquaculture Company Limited, Hangzhou 311200, China
| | - Jianhai Xiang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| |
Collapse
|
38
|
Friederichs S, Krebs S, Blum H, Lang H, Büttner M. Parapoxvirus (PPV) of red deer reveals subclinical infection and confirms a unique species. J Gen Virol 2015; 96:1446-1462. [DOI: 10.1099/vir.0.000080] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 02/01/2015] [Indexed: 11/18/2022] Open
Affiliation(s)
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Centre, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Centre, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Heike Lang
- Bavarian Health and Food Safety Authority, Oberschleißheim, Germany
| | - Mathias Büttner
- Bavarian Health and Food Safety Authority, Oberschleißheim, Germany
| |
Collapse
|
39
|
An immature B cell population from peripheral blood serves as surrogate marker for monitoring tumor angiogenesis and anti-angiogenic therapy in mouse models. Angiogenesis 2015; 18:327-45. [PMID: 26021306 DOI: 10.1007/s10456-015-9470-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 05/18/2015] [Indexed: 12/18/2022]
Abstract
Tumor growth depends on the formation of new blood vessels (tumor angiogenesis) either from preexisting vessels or by the recruitment of bone marrow-derived cells. Despite encouraging results obtained with preclinical cancer models, the therapeutic targeting of tumor angiogenesis has thus far failed to deliver an enduring clinical response in cancer patients. One major obstacle for improving anti-angiogenic therapy is the lack of validated biomarkers, which allow patient stratification for suitable treatment and a rapid assessment of therapy response. Toward these goals, we have employed several mouse models of tumor angiogenesis to identify cell populations circulating in their blood that correlated with the extent of tumor angiogenesis and therapy response. Flow cytometry analyses of different combinations of cell surface markers that define subsets of bone marrow-derived cells were performed on peripheral blood mononuclear cells from tumor-bearing and healthy mice. We identified one cell population, CD45(dim)VEGFR1(-)CD31(low), that was increased in levels during active tumor angiogenesis in a variety of transgenic and syngeneic transplantation mouse models of cancer. Treatment with various anti-angiogenic drugs did not affect CD45(dim)VEGFR1(-)CD31(low) cells in healthy mice, whereas in tumor-bearing mice, a consistent reduction in their levels was observed. Gene expression profiling of CD45(dim)VEGFR1(-)CD31(low) cells characterized these cells as an immature B cell population. These immature B cells were then directly validated as surrogate marker for tumor angiogenesis and of pharmacologic responses to anti-angiogenic therapies in various mouse models of cancer.
Collapse
|
40
|
Nagarajan G, Swami SK, Dahiya SS, Narnaware SD, Mehta SC, Singh PK, Singh R, Tuteja FC, Patil NV. Characterization of GM-CSF-inhibitory factor and Uracil DNA glycosylase encoding genes from camel pseudocowpoxvirus. Res Vet Sci 2015; 100:291-6. [PMID: 25816930 DOI: 10.1016/j.rvsc.2015.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 02/13/2015] [Accepted: 03/06/2015] [Indexed: 11/29/2022]
Abstract
The present study describes the PCR amplification of GM-CSF-inhibitory factor (GIF) and Uracil DNA glycosylase (UDG) encoding genes of pseudocowpoxvirus (PCPV) from the Indian Dromedaries (Camelus dromedarius) infected with contagious ecthyma using the primers based on the corresponding gene sequences of human PCPV and reindeer PCPV, respectively. The length of GIF gene of PCPV obtained from camel is 795 bp and due to the addition of one cytosine residue at position 374 and one adenine residue at position 516, the open reading frame (ORF) got altered, resulting in the production of truncated polypeptide. The ORF of UDG encoding gene of camel PCPV is 696 bp encoding a polypeptide of 26.0 kDa. Comparison of amino acid sequence homologies of GIF and UDG of camel PCPV revealed that the camel PCPV is closer to ORFV and PCPV (reference stains of both human and reindeer), respectively.
Collapse
Affiliation(s)
- G Nagarajan
- National Research Centre on Camel, Post Bag No.7, Jorbeer, Bikaner, Rajasthan 334 001, India.
| | - Shelesh Kumar Swami
- National Research Centre on Camel, Post Bag No.7, Jorbeer, Bikaner, Rajasthan 334 001, India
| | - Shyam Singh Dahiya
- National Research Centre on Camel, Post Bag No.7, Jorbeer, Bikaner, Rajasthan 334 001, India
| | - S D Narnaware
- National Research Centre on Camel, Post Bag No.7, Jorbeer, Bikaner, Rajasthan 334 001, India
| | - S C Mehta
- National Research Centre on Camel, Post Bag No.7, Jorbeer, Bikaner, Rajasthan 334 001, India
| | - P K Singh
- National Research Centre on Camel, Post Bag No.7, Jorbeer, Bikaner, Rajasthan 334 001, India
| | - Raghvendar Singh
- National Research Centre on Camel, Post Bag No.7, Jorbeer, Bikaner, Rajasthan 334 001, India
| | - F C Tuteja
- National Research Centre on Camel, Post Bag No.7, Jorbeer, Bikaner, Rajasthan 334 001, India
| | - N V Patil
- National Research Centre on Camel, Post Bag No.7, Jorbeer, Bikaner, Rajasthan 334 001, India
| |
Collapse
|
41
|
Fleming SB, Wise LM, Mercer AA. Molecular genetic analysis of orf virus: a poxvirus that has adapted to skin. Viruses 2015; 7:1505-39. [PMID: 25807056 PMCID: PMC4379583 DOI: 10.3390/v7031505] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 03/17/2015] [Accepted: 03/19/2015] [Indexed: 12/17/2022] Open
Abstract
Orf virus is the type species of the Parapoxvirus genus of the family Poxviridae. It induces acute pustular skin lesions in sheep and goats and is transmissible to humans. The genome is G+C rich, 138 kbp and encodes 132 genes. It shares many essential genes with vaccinia virus that are required for survival but encodes a number of unique factors that allow it to replicate in the highly specific immune environment of skin. Phylogenetic analysis suggests that both viral interleukin-10 and vascular endothelial growth factor genes have been "captured" from their host during the evolution of the parapoxviruses. Genes such as a chemokine binding protein and a protein that binds granulocyte-macrophage colony-stimulating factor and interleukin-2 appear to have evolved from a common poxvirus ancestral gene while three parapoxvirus nuclear factor (NF)-κB signalling pathway inhibitors have no homology to other known NF-κB inhibitors. A homologue of an anaphase-promoting complex subunit that is believed to manipulate the cell cycle and enhance viral DNA synthesis appears to be a specific adaptation for viral-replication in keratinocytes. The review focuses on the unique genes of orf virus, discusses their evolutionary origins and their role in allowing viral-replication in the skin epidermis.
Collapse
Affiliation(s)
- Stephen B Fleming
- Department of Microbiology and Immunology, 720 Cumberland St, University of Otago, Dunedin 9016, New Zealand.
| | - Lyn M Wise
- Department of Microbiology and Immunology, 720 Cumberland St, University of Otago, Dunedin 9016, New Zealand.
| | - Andrew A Mercer
- Department of Microbiology and Immunology, 720 Cumberland St, University of Otago, Dunedin 9016, New Zealand.
| |
Collapse
|
42
|
Qian N, Gao L, Dong L, Liu H, Fu J, Meng D, Gao X, Zhang J, Gao Y, Song H. Construction, expression, purification, and characterization of a dual-targeting PD-1/VEGF-A fusion protein (P-V). Protein Expr Purif 2015; 109:1-6. [PMID: 25616235 DOI: 10.1016/j.pep.2015.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/09/2015] [Accepted: 01/12/2015] [Indexed: 12/30/2022]
Abstract
Targeting programmed death-1 (PD-1) is regarded as a novel and promising means for the treatment of many types of solid tumor. In the tumor microenvironment (TME), VEGF expression is dramatically up-regulated, and compounds that neutralize VEGF or block the interaction of VEGF with its receptors exhibit potent antitumor activity, and blocking PD-1 might promote T cell infiltration into TME and significantly enhance local immune activation. Thus, we fused domain II and domain III of kinase-insert domain receptor (KDR), the receptor of VEGF-A, to the Fc side of an anti-PD-1 monoclonal antibody with a (Gly4Ser)3 linker to generate a dual targeting fusion protein. The recombinant plasmid was successfully constructed and the fusion protein was expressed in 293E cells. Protein purification was performed in a single step by using protein A affinity chromatography. The molecular weight of the fusion protein was approximately 220kDa, and the yield was approximately 2.97g/L. Specific binding of recombinant protein to PD-1 and VEGF was detected by enzyme-linked immunosorbent assay (ELISA) analysis. Half maximal effective concentration (EC50) values were 0.561nM for PD-1 and 0.682nM for VEGF-A; accordingly, half maximal inhibitory concentration (IC50) values were 0.914nM and 0.583nM, respectively. Proliferation inhibition assays indicated that the fusion protein could inhibit the growth of human umbilical vein endothelial cells effectively. Taken together, the results indicate that this novel fusion protein can simultaneously target PD-1 and VEGF and may be beneficial for combining anti-angiogenesis with immunotherapeutic approaches for the treatment of patients with cancer.
Collapse
Affiliation(s)
- Niliang Qian
- Anhui Medical University, Hefei 230032, China; Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Liucun Gao
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Lihou Dong
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hongchuan Liu
- Suzhou Junmeng Biosciences Co., Ltd, Suzhou 215200, China
| | - Jie Fu
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Dan Meng
- Suzhou Junmeng Biosciences Co., Ltd, Suzhou 215200, China
| | - Xin Gao
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jing Zhang
- Suzhou Junmeng Biosciences Co., Ltd, Suzhou 215200, China
| | - Yucai Gao
- Suzhou Junmeng Biosciences Co., Ltd, Suzhou 215200, China
| | - Haifeng Song
- Anhui Medical University, Hefei 230032, China; Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
43
|
Martins M, Cargnelutti JF, Weiblen R, Flores EF. Pathogenesis in lambs and sequence analysis of putative virulence genes of Brazilian orf virus isolates. Vet Microbiol 2014; 174:69-77. [PMID: 25293399 DOI: 10.1016/j.vetmic.2014.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 09/09/2014] [Accepted: 09/12/2014] [Indexed: 11/18/2022]
Abstract
The parapoxvirus orf virus (ORFV) is the agent of contagious ecthyma, an ubiquitous mucocutaneous disease of sheep and goats that may present variable clinical presentations. We herein studied the pathogenesis of ORFV infection in lambs and analyzed three putative virulence genes of four Brazilian ORFV isolates. Lambs inoculated in the labial commissures with each ORFV isolate (n=4, viral titer 10(5.6) TCID50/ml) developed classical orf lesions, characterized by a progressive course of erythema/macules, vesicles, pustules and proliferative scabs. Lesions lasted an average of 22.9 days (18-26) and virus shedding was detected for approximately 24.6 days (18-30). Two isolates (SV269/11 and SV820/10) produced more severe, long-lasting lesions resulting in highest clinical scores. Lambs inoculated with isolate SV581/11 developed lesions markedly milder (lower clinical scores [p<0.05]) and more limited than the other groups. Virus shedding by SV581/11 group, however, lasted similarly or even longer than the other groups. Sequence analysis of three virulence genes (VEGF, VIR and IL-10v) revealed amino acid deletions and mutations in VEGF and IL-10v genes of SV581/11 and SV252/11, the isolate(s) producing milder lesions. Additionally, the VEGF gene of isolate SV581/11 presented the lowest amino acid identity with the other isolates and with ORFV standard strain OV-IA82. Thus, these results demonstrate that ORFV isolates may display differential virulence in lambs and these differences might be associated with genetic changes in putative virulence genes.
Collapse
Affiliation(s)
- Mathias Martins
- Setor de Virologia, Department of Preventive Veterinary Medicine, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, RS 97105-900, Brazil
| | - Juliana F Cargnelutti
- Setor de Virologia, Department of Preventive Veterinary Medicine, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, RS 97105-900, Brazil
| | - Rudi Weiblen
- Setor de Virologia, Department of Preventive Veterinary Medicine, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, RS 97105-900, Brazil
| | - Eduardo F Flores
- Setor de Virologia, Department of Preventive Veterinary Medicine, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, RS 97105-900, Brazil.
| |
Collapse
|
44
|
Nakamura H, Murakami T, Imamura T, Toriba M, Chijiwa T, Ohno M, Oda-Ueda N. Discovery of a novel vascular endothelial growth factor (VEGF) with no affinity to heparin in Gloydius tsushimaensis venom. Toxicon 2014; 86:107-15. [DOI: 10.1016/j.toxicon.2014.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 05/08/2014] [Accepted: 05/12/2014] [Indexed: 10/25/2022]
|
45
|
Chen S, Feng J, Ma L, Liu Z, Yuan W. RNA interference technology for anti-VEGF treatment. Expert Opin Drug Deliv 2014; 11:1471-80. [DOI: 10.1517/17425247.2014.926886] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
46
|
Anziliero D, Kreutz L, Flores E, Weiblen R. Effects of inactivated parapoxvirus ovis on cellular and humoral events of the innate immune response in mice. Cell Immunol 2014; 289:36-41. [DOI: 10.1016/j.cellimm.2014.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 03/06/2014] [Accepted: 03/14/2014] [Indexed: 12/17/2022]
|
47
|
Lacek K, Bauer B, Bieńkowska-Szewczyk K, Rziha HJ. Orf virus (ORFV) ANK-1 protein mitochondrial localization is mediated by ankyrin repeat motifs. Virus Genes 2014; 49:68-79. [PMID: 24743940 DOI: 10.1007/s11262-014-1069-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 04/03/2014] [Indexed: 10/25/2022]
Abstract
Orf virus (ORFV) strain D1701-V, a Parapoxvirus belonging to the family Poxviridae, became attractive as a novel virus vector system that we successfully used for the generation of recombinant vaccines. Therefore, the identification of viral genes involved in host tropisms or immune modulation is of great interest, as for instance the ORFV-encoded ankyrin-repeat (AR) containing proteins. The present study shows for the first time that the ANK-1 designated gene product of ORFV126 is targeted to mitochondria of ORFV-infected and in ANK-1 transiently expressing cells. Taking advantage of ANK-1 EGFP fusion proteins and confocal fluorescence microscopy mutational and deletion analyses indicated the importance of AR8 and AR9, which may contain a novel class of mitochondria-targeting sequence (MTS) in the central to C-terminal part of this AR-containing protein. The fluorescent findings were corroborated by cell fractionation and Western blotting experiments. The presented results open the avenue for more detailed investigations on cellular binding partners and the function of ANK-1 in viral replication or virulence.
Collapse
Affiliation(s)
- Krzysztof Lacek
- Laboratory of Virus Molecular Biology, University of Gdańsk, 80-822, Gdańsk, Poland
| | | | | | | |
Collapse
|
48
|
Hosamani M, Scagliarini A, Bhanuprakash V, McInnes CJ, Singh RK. Orf: an update on current research and future perspectives. Expert Rev Anti Infect Ther 2014; 7:879-93. [PMID: 19735227 DOI: 10.1586/eri.09.64] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Madhusudan Hosamani
- Indian Veterinary Research Institute, Mukteswar-263138, Nainital Distt., India and Indian Veterinary Research Institute, Hebbal, Bangalore-24, India.
| | | | | | | | | |
Collapse
|
49
|
Senthilkumar K, Venkatesan J, Manivasagan P, Kim SK. Antiangiogenic effects of marine sponge derived compounds on cancer. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2013; 36:1097-1108. [PMID: 24148290 DOI: 10.1016/j.etap.2013.09.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 09/17/2013] [Accepted: 09/20/2013] [Indexed: 06/02/2023]
Abstract
The term "angiogenic switch" refers to a time-restricted event during tumor progression where the balance between pro- and anti-angiogenic factors, resulting in the transition from dormant avascularized hyperplasia to outgrowing vascularized tumor and eventually to malignant tumor progression. Targeting angiogenesis and its mechanistic pathways are critical target for cancer therapy. Recently, marine derived compounds, plays major role in cancer research. Several sponge derived compounds such as alkaloids, terpenes, macrocylic lactone and polyketide are leading drugs in the treatment of different types of diseases including cancer. Those marine sponge compounds inhibit cancer cell proliferation and tumor angiogenesis. Hence, this review sheds light on angiogenic regulators and marine sponge derived antiangiogenic compounds for cancer.
Collapse
Affiliation(s)
- Kalimuthu Senthilkumar
- Marine Bioprocess Research Center, Department of Chemistry, Pukyong National University, Busan, 608-737, Republic of Korea
| | | | | | | |
Collapse
|
50
|
Nieminen T, Toivanen PI, Rintanen N, Heikura T, Jauhiainen S, Airenne KJ, Alitalo K, Marjomäki V, Ylä-Herttuala S. The impact of the receptor binding profiles of the vascular endothelial growth factors on their angiogenic features. Biochim Biophys Acta Gen Subj 2013; 1840:454-63. [PMID: 24112971 DOI: 10.1016/j.bbagen.2013.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 09/12/2013] [Accepted: 10/01/2013] [Indexed: 01/13/2023]
Abstract
BACKGROUND Vascular endothelial growth factors (VEGFs) are potential therapeutic agents for treatment of ischemic diseases. Their angiogenic effects are mainly mediated through VEGF receptor 2 (VEGFR2). METHODS Receptor binding, signaling, and biological efficacy of several VEGFR2 ligands were compared to determine their characteristics regarding angiogenic activity and vascular permeability. RESULTS Tested VEGFR2 ligands induced receptor tyrosine phosphorylation with different efficacy depending on their binding affinities. However, the tyrosine phosphorylation pattern and the activation of the major downstream signaling pathways were comparable. The maximal angiogenic effect stimulated by different VEGFR2 ligands was dependent on their ability to bind to co-receptor Neuropilin (Nrp), which was shown to form complexes with VEGFR2. The ability of these VEGFR2 ligands to induce vascular permeability was dependent on their concentration and VEGFR2 affinity, but not on Nrp binding. CONCLUSIONS VEGFR2 activation alone is sufficient for inducing endothelial cell proliferation, formation of tube-like structures and vascular permeability. The level of VEGFR2 activation is dependent on the binding properties of the ligand used. However, closely similar activation pattern of the receptor kinase domain is seen with all VEGFR2 ligands. Nrp binding strengthens the angiogenic potency without increasing vascular permeability. GENERAL SIGNIFICANCE This study sheds light on how different structurally closely related VEGFR2 ligands bind to and signal via VEGFR2/Nrp complex to induce angiogenesis and vascular permeability. The knowledge of this study could be used for designing VEGFR2/Nrp ligands with improved therapeutic properties.
Collapse
Affiliation(s)
- Tiina Nieminen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, FI-70211 Kuopio, Finland.
| | | | | | | | | | | | | | | | | |
Collapse
|