1
|
Lage SL, Ramaswami R, Rocco JM, Rupert A, Davis DA, Lurain K, Manion M, Whitby D, Yarchoan R, Sereti I. Inflammasome activation in patients with Kaposi sarcoma herpesvirus-associated diseases. Blood 2024; 144:1496-1507. [PMID: 38941593 PMCID: PMC11474434 DOI: 10.1182/blood.2024024144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/02/2024] [Revised: 05/30/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024] Open
Abstract
ABSTRACT Kaposi sarcoma herpesvirus (KSHV)-associated diseases include Kaposi sarcoma (KS), primary effusion lymphoma (PEL), KSHV-associated multicentric Castleman disease (MCD), and KS inflammatory cytokine syndrome (KICS). PEL, MCD, and KICS are associated with elevated circulating inflammatory cytokines. However, activation of the inflammasome, which generates interleukin-1β (IL-1β) and IL-18 via active caspase-1/4/5, has not been evaluated in patients with KSHV-associated diseases (KADs). Herein we report that patients with HIV and ≥1 KAD present with higher plasma levels of IL-18 and increased caspase-1/4/5 activity in circulating monocytes compared with HIV-negative healthy volunteers (HVs) or people with HIV (PWH) without KAD. Within KAD subtypes, KICS and MCD shared enhanced caspase-1/4/5 activity and IL-18 production compared with HVs and PWH, whereas patients with PEL showed remarkably high levels of inflammasome complex formation (known as apoptosis-associated speck-like protein containing a caspase recruitment domain). Moreover, caspase-1/4/5 activity and IL-18 plasma levels correlated with KSHV viral load, indicating KSHV-driven inflammasome activation in KAD. Accordingly, factors released by cells latently infected with KSHV triggered inflammasome activation and cytokine production in bystander monocytes in vitro. Finally, both supervised and unsupervised analyses with inflammasome measurements and other inflammatory biomarkers demonstrate a unique inflammatory profile in patients with PEL, MCD, and KICS as compared with KS. Our data indicate that detrimental inflammation in patients with KAD is at least partially driven by KSHV-induced inflammasome activation in monocytes, thus offering novel approaches to diagnose and treat these complex disorders. These trials were registered at www.ClinicalTrials.gov as #NCT01419561, NCT00092222, NCT00006518, and NCT02147405.
Collapse
Affiliation(s)
- Silvia Lucena Lage
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Ramya Ramaswami
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Joseph M. Rocco
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Adam Rupert
- AIDS Monitoring Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD
| | - David A. Davis
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kathryn Lurain
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Maura Manion
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Denise Whitby
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD
| | - Robert Yarchoan
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Irini Sereti
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
2
|
Cui L, Saeed Y, Li H, Yang J. Regenerative medicine and traumatic brain injury: from stem cell to cell-free therapeutic strategies. Regen Med 2021; 17:37-53. [PMID: 34905963 DOI: 10.2217/rme-2021-0069] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/26/2022] Open
Abstract
Traumatic brain injury (TBI) is a serious health concern, yet there is a lack of standardized treatment to combat its long-lasting effects. The objective of the present study was to provide an overview of the limitation of conventional stem-cell therapy in the treatment of TBI and to discuss the application of novel acellular therapies and their advanced strategies to enhance the efficacy of stem cells derived therapies in the light of published study data. Moreover, we also discussed the factor to optimize the therapeutic efficiency of stem cell-derived acellular therapy by overcoming the challenges for its clinical translation. Hence, we concluded that acellular therapy possesses the potential to bring a breakthrough in the field of regenerative medicine to treat TBI.
Collapse
Affiliation(s)
- Lianxu Cui
- Department of Neurosurgery, The First People's Hospital of Foshan, 81 North Lingnan Road, Foshan, Guangdong, 528300, PR China
| | - Yasmeen Saeed
- Guangdong VitaLife Biotechnology Co., LTD, 61 Xiannan Road, Nanhai District, Foshan, Guangdong, 528200, PR China
| | - Haomin Li
- Department of Neurosurgery, The First People's Hospital of Foshan, 81 North Lingnan Road, Foshan, Guangdong, 528300, PR China
| | - Jingli Yang
- School of medicine, Foshan University, 18 Jiangwan Road, Foshan, Guangdong, 528000, PR China
| |
Collapse
|
3
|
Barrett L, Chen J, Dai L, Plaisance-Bonstaff K, Del Valle L, Qin Z. Role of Interleukin-1 Family Members and Signaling Pathways in KSHV Pathogenesis. Front Cell Infect Microbiol 2020; 10:587929. [PMID: 33194830 PMCID: PMC7662392 DOI: 10.3389/fcimb.2020.587929] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/27/2020] [Accepted: 10/13/2020] [Indexed: 01/22/2023] Open
Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) represents the etiological agent for several human malignancies, including Kaposi’s sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman’s disease (MCD), which are mostly seen in immunocompromised patients. In fact, KSHV has developed many strategies to hijack host immune response, including the regulation of inflammatory cytokine production. Interleukin-1 (IL-1) family represents a major mediator for inflammation and plays an important role in both innate and adaptive immunity. Furthermore, a broadening list of diseases has revealed the pathologic role of IL-1 mediated inflammation. In the current mini-review, we have summarized recent findings about how this oncogenic virus is able to manipulate the activities of IL-1 signaling pathway to facilitate disease progression. We also discuss the therapeutic potential of IL-1 blockade against KSHV-related diseases and several unsolved questions in this interesting field.
Collapse
Affiliation(s)
- Lindsey Barrett
- Department of Pathology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jungang Chen
- Department of Pathology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Lu Dai
- Department of Pathology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Karlie Plaisance-Bonstaff
- Department of Medicine, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA, United States
| | - Luis Del Valle
- Department of Pathology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA, United States
| | - Zhiqiang Qin
- Department of Pathology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
4
|
Alomari N, Totonchy J. Cytokine-Targeted Therapeutics for KSHV-Associated Disease. Viruses 2020; 12:E1097. [PMID: 32998419 PMCID: PMC7600567 DOI: 10.3390/v12101097] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/01/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) also known as human herpesvirus 8 (HHV-8), is linked to several human malignancies including Kaposi sarcoma (KS), primary effusion lymphoma (PEL), multicentric Castleman's disease (MCD) and recently KSHV inflammatory cytokine syndrome (KICS). As with other diseases that have a significant inflammatory component, current therapy for KSHV-associated disease is associated with significant off-target effects. However, recent advances in our understanding of the pathogenesis of KSHV have produced new insight into the use of cytokines as potential therapeutic targets. Better understanding of the role of cytokines during KSHV infection and tumorigenesis may lead to new preventive or therapeutic strategies to limit KSHV spread and improve clinical outcomes. The cytokines that appear to be promising candidates as KSHV antiviral therapies include interleukins 6, 10, and 12 as well as interferons and tumor necrosis factor-family cytokines. This review explores our current understanding of the roles that cytokines play in promoting KSHV infection and tumorigenesis, and summarizes the current use of cytokines as therapeutic targets in KSHV-associated diseases.
Collapse
Affiliation(s)
| | - Jennifer Totonchy
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA;
| |
Collapse
|
5
|
Rivera-Soto R, Damania B. Modulation of Angiogenic Processes by the Human Gammaherpesviruses, Epstein-Barr Virus and Kaposi's Sarcoma-Associated Herpesvirus. Front Microbiol 2019; 10:1544. [PMID: 31354653 PMCID: PMC6640166 DOI: 10.3389/fmicb.2019.01544] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/03/2019] [Accepted: 06/20/2019] [Indexed: 12/25/2022] Open
Abstract
Angiogenesis is the biological process by which new blood vessels are formed from pre-existing vessels. It is considered one of the classic hallmarks of cancer, as pathological angiogenesis provides oxygen and essential nutrients to growing tumors. Two of the seven known human oncoviruses, Epstein–Barr virus (EBV) and Kaposi’s sarcoma-associated herpesvirus (KSHV), belong to the Gammaherpesvirinae subfamily. Both viruses are associated with several malignancies including lymphomas, nasopharyngeal carcinomas, and Kaposi’s sarcoma. The viral genomes code for a plethora of viral factors, including proteins and non-coding RNAs, some of which have been shown to deregulate angiogenic pathways and promote tumor growth. In this review, we discuss the ability of both viruses to modulate the pro-angiogenic process.
Collapse
Affiliation(s)
- Ricardo Rivera-Soto
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Blossom Damania
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
6
|
Pontejo SM, Murphy PM, Pease JE. Chemokine Subversion by Human Herpesviruses. J Innate Immun 2018; 10:465-478. [PMID: 30165356 DOI: 10.1159/000492161] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/17/2018] [Accepted: 07/11/2018] [Indexed: 12/30/2022] Open
Abstract
Viruses use diverse molecular mechanisms to exploit and evade the immune response. Herpesviruses, in particular, encode functional chemokine and chemokine receptor homologs pirated from the host, as well as secreted chemokine-binding proteins with unique structures. Multiple functions have been described for herpesvirus chemokine components, including attraction of target cells, blockade of leukocyte migration, and modulation of gene expression and cell entry by the virus. Here we review current concepts about how human herpesvirus chemokines, chemokine receptors, and chemokine-binding proteins may be used to shape a proviral state in the host.
Collapse
Affiliation(s)
- Sergio M Pontejo
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Philip M Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - James E Pease
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United
| |
Collapse
|
7
|
Pontejo SM, Murphy PM. Chemokines encoded by herpesviruses. J Leukoc Biol 2017; 102:1199-1217. [PMID: 28848041 DOI: 10.1189/jlb.4ru0417-145rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/11/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/15/2022] Open
Abstract
Viruses use diverse strategies to elude the immune system, including copying and repurposing host cytokine and cytokine receptor genes. For herpesviruses, the chemokine system of chemotactic cytokines and receptors is a common source of copied genes. Here, we review the current state of knowledge about herpesvirus-encoded chemokines and discuss their possible roles in viral pathogenesis, as well as their clinical potential as novel anti-inflammatory agents or targets for new antiviral strategies.
Collapse
Affiliation(s)
- Sergio M Pontejo
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Philip M Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
8
|
Li S, Bai L, Dong J, Sun R, Lan K. Kaposi's Sarcoma-Associated Herpesvirus: Epidemiology and Molecular Biology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1018:91-127. [PMID: 29052134 DOI: 10.1007/978-981-10-5765-6_7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV), also known as Human herpesvirus 8 (HHV-8), is a member of the lymphotropic gammaherpesvirus subfamily and a human oncogenic virus. Since its discovery in AIDS-associated KS tissues by Drs. Yuan Chang and Patrick Moore, much progress has been made in the past two decades. There are four types of KS including classic KS, endemic KS, immunosuppressive therapy-related KS, and AIDS-associated KS. In addition to KS, KSHV is also involved in the development of primary effusion lymphoma (PEL) and certain types of multicentric Castleman's disease. KSHV manipulates numerous viral proteins to promote the progression of angiogenesis and tumorigenesis. In this chapter, we review the epidemiology and molecular biology of KSHV and the mechanisms underlying KSHV-induced diseases.
Collapse
Affiliation(s)
- Shasha Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Lei Bai
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Jiazhen Dong
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Rui Sun
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Ke Lan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, People's Republic of China.
| |
Collapse
|
9
|
|
10
|
Szpakowska M, Chevigné A. vCCL2/vMIP-II, the viral master KEYmokine. J Leukoc Biol 2015; 99:893-900. [DOI: 10.1189/jlb.2mr0815-383r] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/27/2015] [Accepted: 11/20/2015] [Indexed: 11/24/2022] Open
|
11
|
Rincon MY, VandenDriessche T, Chuah MK. Gene therapy for cardiovascular disease: advances in vector development, targeting, and delivery for clinical translation. Cardiovasc Res 2015; 108:4-20. [PMID: 26239654 PMCID: PMC4571836 DOI: 10.1093/cvr/cvv205] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/13/2015] [Accepted: 07/22/2015] [Indexed: 01/06/2023] Open
Abstract
Gene therapy is a promising modality for the treatment of inherited and acquired cardiovascular diseases. The identification of the molecular pathways involved in the pathophysiology of heart failure and other associated cardiac diseases led to encouraging preclinical gene therapy studies in small and large animal models. However, the initial clinical results yielded only modest or no improvement in clinical endpoints. The presence of neutralizing antibodies and cellular immune responses directed against the viral vector and/or the gene-modified cells, the insufficient gene expression levels, and the limited gene transduction efficiencies accounted for the overall limited clinical improvements. Nevertheless, further improvements of the gene delivery technology and a better understanding of the underlying biology fostered renewed interest in gene therapy for heart failure. In particular, improved vectors based on emerging cardiotropic serotypes of the adeno-associated viral vector (AAV) are particularly well suited to coax expression of therapeutic genes in the heart. This led to new clinical trials based on the delivery of the sarcoplasmic reticulum Ca2+-ATPase protein (SERCA2a). Though the first clinical results were encouraging, a recent Phase IIb trial did not confirm the beneficial clinical outcomes that were initially reported. New approaches based on S100A1 and adenylate cyclase 6 are also being considered for clinical applications. Emerging paradigms based on the use of miRNA regulation or CRISPR/Cas9-based genome engineering open new therapeutic perspectives for treating cardiovascular diseases by gene therapy. Nevertheless, the continuous improvement of cardiac gene delivery is needed to allow the use of safer and more effective vector doses, ultimately bringing gene therapy for heart failure one step closer to reality.
Collapse
Affiliation(s)
- Melvin Y Rincon
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Building D, room D306, Laarbeeklaan 103, Brussels, Belgium Centro de Investigaciones, Fundacion Cardiovascular de Colombia, Floridablanca, Colombia
| | - Thierry VandenDriessche
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Building D, room D306, Laarbeeklaan 103, Brussels, Belgium Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Marinee K Chuah
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Building D, room D306, Laarbeeklaan 103, Brussels, Belgium Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
12
|
Gramolelli S, Schulz TF. The role of Kaposi sarcoma-associated herpesvirus in the pathogenesis of Kaposi sarcoma. J Pathol 2015; 235:368-80. [PMID: 25212381 DOI: 10.1002/path.4441] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/07/2014] [Revised: 09/05/2014] [Accepted: 09/06/2014] [Indexed: 01/07/2023]
Abstract
Kaposi sarcoma (KS) is an unusual vascular tumour caused by an oncogenic-herpesvirus, Kaposi sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8 (HHV 8). KS lesions are characterized by an abundant inflammatory infiltrate, the presence of KSHV-infected endothelial cells that show signs of aberrant differentiation, as well as faulty angiogenesis/ vascularization. Here we discuss the molecular mechanisms that lead to the development of these histological features of KS, with an emphasis on the viral proteins that are responsible for their development.
Collapse
Affiliation(s)
- Silvia Gramolelli
- Institute of Virology, Hannover Medical School, Carl Neuberg Strasse 1, 30625 Hannover, Germany; German Centre for Infection Research, Hannover-Braunschweig Site, Germany
| | | |
Collapse
|
13
|
Ojala PM, Schulz TF. Manipulation of endothelial cells by KSHV: implications for angiogenesis and aberrant vascular differentiation. Semin Cancer Biol 2014; 26:69-77. [PMID: 24486643 DOI: 10.1016/j.semcancer.2014.01.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/14/2013] [Revised: 01/16/2014] [Accepted: 01/16/2014] [Indexed: 12/21/2022]
Abstract
Kaposi sarcoma (KS), a viral cancer associated to Kaposi sarcoma herpesvirus (KSHV) infection, is currently the most common tumor in men in sub-Saharan Africa. KS is an angiogenic tumor and characterized by the presence of aberrant vascular structures in the lesion. Although our understanding of how KSHV causes the aberrant differentiation of endothelial cells and the typical vascular abnormalities in KS tumors is far from complete, the experimental evidence reviewed here provides a comprehensive description of the role of KSHV in the pathogenesis of this unusual tumor. In contrast to other tumor viruses, whose interference with cellular processes relating to cell cycle, apoptosis and DNA damage may be at the heart of their oncogenic properties, KSHV may cause KS primarily by its ability to engage with the differentiation and function of endothelial cells. Although the intracellular pathways engaged by KSHV in the endothelial cells are being explored as drug targets, a better understanding of the impact of KSHV on endothelial cell differentiation and vasculogenesis is needed before the encouraging findings can form the basis for new targeted therapeutic approaches to KS.
Collapse
Affiliation(s)
- Päivi M Ojala
- Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014 University of Helsinki, Finland; Foundation for the Finnish Cancer Institute, Helsinki, Finland; Section of Virology, Imperial College Faculty of Medicine, Norfolk Place, London W2 1PG, UK.
| | - Thomas F Schulz
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
14
|
Babczyk P, Conzendorf C, Klose J, Schulze M, Harre K, Tobiasch E. Stem Cells on Biomaterials for Synthetic Grafts to Promote Vascular Healing. J Clin Med 2014; 3:39-87. [PMID: 26237251 PMCID: PMC4449663 DOI: 10.3390/jcm3010039] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/06/2013] [Revised: 10/28/2013] [Accepted: 11/16/2013] [Indexed: 12/25/2022] Open
Abstract
This review is divided into two interconnected parts, namely a biological and a chemical one. The focus of the first part is on the biological background for constructing tissue-engineered vascular grafts to promote vascular healing. Various cell types, such as embryonic, mesenchymal and induced pluripotent stem cells, progenitor cells and endothelial- and smooth muscle cells will be discussed with respect to their specific markers. The in vitro and in vivo models and their potential to treat vascular diseases are also introduced. The chemical part focuses on strategies using either artificial or natural polymers for scaffold fabrication, including decellularized cardiovascular tissue. An overview will be given on scaffold fabrication including conventional methods and nanotechnologies. Special attention is given to 3D network formation via different chemical and physical cross-linking methods. In particular, electron beam treatment is introduced as a method to combine 3D network formation and surface modification. The review includes recently published scientific data and patents which have been registered within the last decade.
Collapse
Affiliation(s)
- Patrick Babczyk
- Department of Natural Science, Bonn-Rhein-Sieg University of Applied Science, Von-Liebig-Street 20, Rheinbach 53359, Germany.
| | - Clelia Conzendorf
- Faculty of Mechanical Engineering/Process Engineering, University of Applied Science Dresden, Friedrich-List-Platz 1, Dresden 01069, Germany.
| | - Jens Klose
- Faculty of Mechanical Engineering/Process Engineering, University of Applied Science Dresden, Friedrich-List-Platz 1, Dresden 01069, Germany.
| | - Margit Schulze
- Department of Natural Science, Bonn-Rhein-Sieg University of Applied Science, Von-Liebig-Street 20, Rheinbach 53359, Germany.
| | - Kathrin Harre
- Faculty of Mechanical Engineering/Process Engineering, University of Applied Science Dresden, Friedrich-List-Platz 1, Dresden 01069, Germany.
| | - Edda Tobiasch
- Department of Natural Science, Bonn-Rhein-Sieg University of Applied Science, Von-Liebig-Street 20, Rheinbach 53359, Germany.
| |
Collapse
|
15
|
Vrancken K, Vervaeke P, Balzarini J, Liekens S. Viruses as key regulators of angiogenesis. Rev Med Virol 2011; 21:181-200. [PMID: 21538666 DOI: 10.1002/rmv.688] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022]
Abstract
Angiogenesis is an important physiological process that is controlled by a precise balance of growth and inhibitory factors in healthy tissues. However, environmental and genetic factors may disturb this delicate balance, resulting in the development of angiogenic diseases, tumour growth and metastasis. During the past decades, extensive research has led to the identification and characterization of genes, proteins and signalling pathways that are involved in neovascularization. Moreover, increasing evidence indicates that viruses may also regulate angiogenesis either directly, by (i) producing viral chemokines, growth factors and/or receptors or (ii) activating blood vessels as a consequence of endothelial cell tropism, or indirectly, by (iii) modulating the activity of cellular proteins and/or (iv) inducing a local or systemic inflammatory response, thereby creating an angiogenic microenvironment. As such, viruses may modulate several signal transduction pathways involved in angiogenesis leading to changes in endothelial cell proliferation, migration, adhesion, vascular permeability and/or protease production. Here, we will review different mechanisms that may be applied by viruses to deregulate the angiogenic balance in healthy tissues and/or increase the angiogenic potential of tumours.
Collapse
|
16
|
Zhao B, Liwang PJ. Characterization of the interactions of vMIP-II, and a dimeric variant of vMIP-II, with glycosaminoglycans. Biochemistry 2010; 49:7012-22. [PMID: 20712376 DOI: 10.1021/bi100549y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/28/2022]
Abstract
Chemokines are important immune proteins, carrying out their function by binding to glycosaminoglycans (GAGs) on the endothelial surface and to cell surface chemokine receptors. A unique viral chemokine analogue, viral macrophage inflammatory protein-II (vMIP-II), encoded by human herpesvirus-8, has garnered interest because of its ability to bind to multiple chemokine receptors, including both HIV coreceptors. In addition, vMIP-II binds to cell surface GAGs much more tightly than most human chemokines, which may be the key to its anti-inflammatory function in vivo. The goal of this work was to determine the mechanism of binding of GAG by vMIP-II. The interaction of vMIP-II with a heparin-derived disaccharide was characterized using NMR. Important binding sites were further analyzed by mutagenesis studies, in which corresponding vMIP-II mutants were tested for GAG binding ability using heparin chromatography and NMR. We found that despite having many more basic residues than some chemokines, vMIP-II shares a characteristic binding site similar to that of its human analogues, utilizing basic residues R18, R46, and R48. Interestingly, a particular mutation (Leu13Phe) caused vMIP-II to form a pH-dependent CC chemokine-type dimer as determined by analytical ultracentrifugation and NMR. To the best of our knowledge, this is the first example of engineering a naturally predominantly monomeric chemokine into a dissociable dimer by a single mutation. This dimeric vMIP-II mutant binds to heparin much more tightly than wild-type vMIP-II and provides a new model for studying the relationship between chemokine quaternary structure and various aspects of function. Structural differences between monomeric and dimeric vMIP-II upon GAG binding were characterized by NMR and molecular docking.
Collapse
Affiliation(s)
- Bo Zhao
- University of California, 5200 North Lake Road, Merced, California 95343, USA
| | | |
Collapse
|
17
|
Schussler O, Chachques JC, Mesana TG, Suuronen EJ, Lecarpentier Y, Ruel M. 3-dimensional structures to enhance cell therapy and engineer contractile tissue. Asian Cardiovasc Thorac Ann 2010; 18:188-98. [PMID: 20304859 DOI: 10.1177/0218492310361531] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022]
Abstract
Experimental studies in animals and recent human clinical trials have revealed the current limitations of cellular transplantation, which include poor cell survival, lack of cell engraftment, and poor differentiation. Evidence in animals suggests that use of a 3-dimensional scaffold may enhance cell therapy and engineer myocardial tissue by improving initial cell retention, survival, differentiation, and integration. Several scaffolds of synthetic or natural origin are under development. Until now, contractility has been demonstrated in vitro only in biological scaffolds prepared from decellularized organs or tissue, or in collagenic porous scaffold obtained by crosslinking collagen fibers. While contractility of a cellularized collagen construct is poor, it can be greatly enhanced by tumor basement membrane extract. Recent advances in biochemistry have shown improved cell-matrix interactions by coupling adhesion molecules to achieve an efficient and safe bioartificial myocardium with no tumoral component. Fixation of adhesion molecules may also be a way to enhance cell homing and/or differentiation to increase local angiogenesis. Whatever the clinically successful combination ultimately proves to be, it is likely that cell therapy will require providing a supportive biochemical, physical, and spatial environment that will allow the cells to optimally differentiate and integrate within the target myocardial tissue.
Collapse
Affiliation(s)
- Olivier Schussler
- Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Suite 3403, Ottawa, ON, K1Y 4W7, Canada.
| | | | | | | | | | | |
Collapse
|
18
|
Magkouta S, Stathopoulos GT, Psallidas I, Papapetropoulos A, Kolisis FN, Roussos C, Loutrari H. Protective Effects of Mastic Oil FromPistacia LentiscusVariationChiaAgainst Experimental Growth of Lewis Lung Carcinoma. Nutr Cancer 2009; 61:640-8. [DOI: 10.1080/01635580902825647] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/20/2022]
|
19
|
Miljkovic-Licina M, Hammel P, Garrido-Urbani S, Bradfield PF, Szepetowski P, Imhof BA. Sushi repeat protein X-linked 2, a novel mediator of angiogenesis. FASEB J 2009; 23:4105-16. [PMID: 19667118 DOI: 10.1096/fj.09-135202] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/25/2022]
Abstract
On appropriate stimuli, quiescent endothelial cells start to proliferate and form de novo blood vessels through angiogenesis. To further define molecular mechanisms accompanying the activation of endothelial cells during angiogenesis, we identified genes that were differentially regulated during this process using microarray analyses. In this work, we established a regulatory role for Sushi repeat protein X-linked 2 (Srpx2) in endothelial cell remodeling during angiogenesis. In particular, silencing of Srpx2 using small interfering RNAs (siRNAs) specifically attenuated endothelial cell migration and delayed angiogenic sprout formation. In vivo, Srpx2 expression was detected in de novo formation of blood vessels in angiogenic tissues by in situ mRNA hybridization and immunostaining. Pulldown experiments identified Srpx2 as a ligand for vascular uPAR, a key molecule involved in invasive migration of angiogenic endothelium. Immunostaining revealed coexpression of the Srpx2 and uPAR on vascular endothelium. These findings suggest that Srpx2 regulates endothelial cell migration and tube formation and provides a new target for modulating angiogenesis.
Collapse
Affiliation(s)
- Marijana Miljkovic-Licina
- Department of Pathology and Immunology, Centre Médical Universitaire, Rue Michel-Servet 1, CH-1211 Geneva 4, Switzerland
| | | | | | | | | | | |
Collapse
|
20
|
Nifontova IN, Sats NV, Surin VL, Svinareva DA, Gasparian ME, Drize NJ. Infection of stromal and hemopoietic precursor cells with lentivirus vector in vivo and in vitro. Bull Exp Biol Med 2008; 145:133-6. [PMID: 19024021 DOI: 10.1007/s10517-008-0030-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/22/2023]
Abstract
We developed a method for gene transfer into mesenchymal stromal cells. Lentivirus vector containing green fluorescent protein gene for labeling stromal and hemopoietic precursor cells was obtained using two plasmid sets from different sources. The vector was injected into the femur of mice in vivo and added into culture medium for in vitro infection of the stromal sublayer of long-term bone marrow culture. From 25 to 80% hemopoietic stem cells forming colonies in the spleen were infected with lentivirus vector in vivo and in vitro. Fibroblast colony-forming cells from the femoral bones of mice injected with the lentivirus vector carried no marker gene. The marker gene was detected in differentiated descendants from mesenchymal stem cells (bone cavity cells from the focus of ectopic hemopoiesis formed after implantation of the femoral bone marrow cylinder infected with lentivirus vector under the renal capsule of syngeneic recipient). In in vitro experiments, the marker gene was detected in sublayers of long-term bone marrow cultures infected after preliminary 28-week culturing, when hemopoiesis was completely exhausted. The efficiency of infection of stromal precursor cells depended on the source of lentivirus. The possibility of transfering the target gene into hemopoietic precursor cells in vivo is demonstrated. Stromal precursor cells can incorporate the provirus in vivo and in vitro, but conditions and infection system for effective infection should be thoroughly selected.
Collapse
Affiliation(s)
- I N Nifontova
- Hematology Research Center, Russian Academy of Medical Sciences.
| | | | | | | | | | | |
Collapse
|
21
|
Efklidou S, Bailey R, Field N, Noursadeghi M, Collins MK. vFLIP from KSHV inhibits anoikis of primary endothelial cells. J Cell Sci 2008; 121:450-7. [DOI: 10.1242/jcs.022343] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/20/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV or HHV-8) infection of endothelial cells is an early event in the aetiology of the endothelial cell tumour Kaposi's sarcoma (KS). We have examined the effect of the KSHV latent protein viral FLICE-like inhibitory protein (vFLIP) on dermal microvascular endothelial cell (MVEC) survival as vFLIP is expressed in the KSHV-infected cells within KS lesions. To do this, we have used a lentiviral vector to express vFLIP in MVECs in the absence of other KSHV proteins. vFLIP activates the classical NF-κB pathway in MVECs and causes nuclear translocation of RelA/p65. This NF-κB activation prevents detachment-induced apoptosis (anoikis) of MVECs but does not inhibit apoptosis induced by removal of essential survival factors, including vascular endothelial growth factor (VEGF). vFLIP expression inhibits anoikis in part by inducing the secretion of an additional paracrine survival factor(s). The implications of these results for KS development are discussed.
Collapse
Affiliation(s)
- Sofia Efklidou
- Infection and Immunity, University College London, Windeyer Building, 46 Cleveland Street, London W1T 4JF, UK
| | - Ranbir Bailey
- Infection and Immunity, University College London, Windeyer Building, 46 Cleveland Street, London W1T 4JF, UK
| | - Nigel Field
- Infection and Immunity, University College London, Windeyer Building, 46 Cleveland Street, London W1T 4JF, UK
| | - Mahdad Noursadeghi
- Infection and Immunity, University College London, Windeyer Building, 46 Cleveland Street, London W1T 4JF, UK
| | - Mary K. Collins
- Infection and Immunity, University College London, Windeyer Building, 46 Cleveland Street, London W1T 4JF, UK
| |
Collapse
|