1
|
Denaro N, Indini A, Brambilla L, Marzano AV, Garrone O, Tourlaki A. Management and Future Therapeutic Perspectives of Classic Kaposi's Sarcoma: An Evidence-Based Review. Onco Targets Ther 2024; 17:961-976. [PMID: 39530040 PMCID: PMC11552409 DOI: 10.2147/ott.s468787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/31/2024] [Indexed: 11/16/2024] Open
Abstract
Background Kaposi sarcoma (KS) is a cutaneous neoplasm of endothelial origin. The causative agent is the human herpes virus-8 (HHV-8) which, combined with an immune system impairment, causes cell proliferation. To date, high-quality evidence and treatment recommendations for the management of KS are confined to the acquired immune deficiency syndrome (AIDS)-related KS, while the clinical approach to the treatment of classic KS (CKS) is based on small retrospective case series and the experience of clinicians in selected referral centers. Materials and Methods A search of the English literature was conducted through PubMed/MEDLINE databases for studies regarding CKS diagnosis, staging, and treatment, published between January 1990 and September 2023. Results Overall, 122 out of 565 articles were selected. Based on the results of this literature review, we proposed indications regarding the recommended flow chart for diagnosis, staging, and follow-up of patients with CKS. We assess available evidences regarding topic, locoregional, and systemic treatments of CKS. We also provide a focus on novel treatment strategies and therapeutic approaches currently under evaluation in clinical trials. Conclusion CKS is a rare disease and its management requires a multidisciplinary assessment. Treatment in referral centers and enrolment in clinical trials might impact on outcomes.
Collapse
Affiliation(s)
- Nerina Denaro
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Alice Indini
- Melanoma Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lucia Brambilla
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Angelo Valerio Marzano
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Ornella Garrone
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Athanasia Tourlaki
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
2
|
Krüger-Genge A, Harb K, Braune S, Jung CHG, Westphal S, Bär S, Mauger O, Küpper JH, Jung F. Effects of Arthrospira platensis on Human Umbilical Vein Endothelial Cells. Life (Basel) 2024; 14:1253. [PMID: 39459553 PMCID: PMC11508656 DOI: 10.3390/life14101253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 10/28/2024] Open
Abstract
Atherosclerosis is initiated by injury or damage to the vascular endothelial cell monolayer. Therefore, the early repair of the damaged vascular endothelium by a proliferation of neighbouring endothelial cells is important to prevent atherosclerosis and thrombotic events. Arthrospira platensis (AP) has been used as a dietary supplement, mainly due to its high content of vitamins, minerals, amino acids, and pigments such as chlorophylls, carotenoids, and phycocyanin, ingredients with antioxidant, anti-inflammatory, and anti-thrombotic properties. Therefore, in this prospective, placebo-controlled, data-driven, sample-size-estimated in vitro study, we tested whether an aqueous extract of AP at different concentrations (50, 100, and 200 µg/mL) had an effect on the different cellular parameters of human umbilical vein endothelial cells. Therefore, cell impedance measurement and cell proliferation were measured to investigate the monolayer formation. In addition, cell viability, integrity, and metabolism were analysed to evaluate singular cellular functions, especially the antithrombotic state. Furthermore, cell-cell and cell-substrate interactions were observed. The highest proliferation was achieved after the addition of 100 µg/mL. This was consistently confirmed by two independent optical experiments in cell cultures 48 h and 85 h after seeding and additionally by an indirect test. At this concentration, the activation or dysfunction of HUVECs was completely prevented, as confirmed by prostacyclin and interleukin-6 levels. In conclusion, in this study, AP induced a significant increase in HUVEC proliferation without inducing an inflammatory response but altered the hemostasiological balance in favour of prostacyclin over thromboxane, thereby creating an antithrombotic state. Thus, APE could be applied in the future as an accelerator of endothelial cell proliferation after, e.g., stent placement or atherosclerosis.
Collapse
Affiliation(s)
- Anne Krüger-Genge
- Life Science and Bioprocesses, Fraunhofer Institute for Applied Polymer Research (IAP), 14476 Potsdam, Germany; (K.H.); (S.W.); (S.B.); (O.M.)
| | - Kudor Harb
- Life Science and Bioprocesses, Fraunhofer Institute for Applied Polymer Research (IAP), 14476 Potsdam, Germany; (K.H.); (S.W.); (S.B.); (O.M.)
| | - Steffen Braune
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany (J.-H.K.); (F.J.)
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Conrad H. G. Jung
- Carbon Biotech, Social Enterprise Stiftungs AG, 01968 Senftenberg, Germany
| | - Sophia Westphal
- Life Science and Bioprocesses, Fraunhofer Institute for Applied Polymer Research (IAP), 14476 Potsdam, Germany; (K.H.); (S.W.); (S.B.); (O.M.)
| | - Stefanie Bär
- Life Science and Bioprocesses, Fraunhofer Institute for Applied Polymer Research (IAP), 14476 Potsdam, Germany; (K.H.); (S.W.); (S.B.); (O.M.)
| | - Olivia Mauger
- Life Science and Bioprocesses, Fraunhofer Institute for Applied Polymer Research (IAP), 14476 Potsdam, Germany; (K.H.); (S.W.); (S.B.); (O.M.)
| | - Jan-Heiner Küpper
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany (J.-H.K.); (F.J.)
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
- Carbon Biotech, Social Enterprise Stiftungs AG, 01968 Senftenberg, Germany
| | - Friedrich Jung
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany (J.-H.K.); (F.J.)
| |
Collapse
|
3
|
Li K, Dai M, Sacirovic M, Pagonas N, Ritter O, Kah J, Lauxmann MA, Bramlage P, Bondke Persson A, Buschmann I, Hillmeister P. Association of endothelial function and lower extremity perfusion in peripheral artery disease. VASA 2024; 53:333-340. [PMID: 38979892 DOI: 10.1024/0301-1526/a001131] [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: 07/10/2024]
Abstract
Background: The current study aims to investigate the association between endothelial function and lower extremity perfusion in patients with peripheral artery disease (PAD). Patients and methods: In total 229 patients with PAD (Rutherford stage 0-3) were enrolled in the current study. Endothelial function was assessed by measuring flow-mediated dilation (FMD) and endothelial cell proliferation capacity (ECPC). Lower extremity perfusion was assessed by measuring oscillometry-based ankle brachial index (oABI) and pulse wave index (PWI). In addition, carotid intima-media-thickness (cIMT) was also measured as a surrogate marker for atherosclerosis. Correlations between FMD, ECPC, oABI, PWI, and cIMT were analysed using Pearson correlation coefficient. The relationship between the above variables and the severity of PAD was investigated using ordinal logistic regression analysis. Results: Correlation analysis showed that FMD negatively associated with PWI (r = -0.183, p = 0.005), ECPC positively associated with oABI (r = 0.162, p = 0.014), and oABI negatively associated with PWI (r = -0.264, p < 0.001). Ordinal logistic regression analysis showed that ECPC (β = -0.009, p = 0.048), oABI (β = -5.290, p < 0.001), and age (β = -0.058, p = 0.002) negatively associated with the PAD Rutherford stages. In addition, PWI (β = 0.006, p < 0.001), cIMT (β = 18.363, p = 0.043) positively associated with the PAD Rutherford stages. Conclusions: Endothelial function significantly associates with lower extremity perfusion in patients with PAD, and both are related to the severity of PAD.
Collapse
Affiliation(s)
- Kangbo Li
- Department of Angiology, Center for Internal Medicine I, Deutsches Angiologie-Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mengjun Dai
- Department of Angiology, Center for Internal Medicine I, Deutsches Angiologie-Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mesud Sacirovic
- Department of Angiology, Center for Internal Medicine I, Deutsches Angiologie-Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Nikolaos Pagonas
- Department of Cardiology, Center for Internal Medicine I, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| | - Oliver Ritter
- Department of Cardiology, Center for Internal Medicine I, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| | - Janine Kah
- Department of Gastroenterology, Center for Internal Medicine II, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Martin A Lauxmann
- Institute of Biochemistry, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Peter Bramlage
- Institute for Pharmacology and Preventive Medicine, Cloppenburg, Germany
| | - Anja Bondke Persson
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ivo Buschmann
- Department of Angiology, Center for Internal Medicine I, Deutsches Angiologie-Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| | - Philipp Hillmeister
- Department of Angiology, Center for Internal Medicine I, Deutsches Angiologie-Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| |
Collapse
|
4
|
Hussain MT, Stanfield BA, Bernstein DI. Small Animal Models to Study Herpes Simplex Virus Infections. Viruses 2024; 16:1037. [PMID: 39066200 PMCID: PMC11281376 DOI: 10.3390/v16071037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/14/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
Herpes simplex virus type 1 (HSV-1) and herpes simplex virus type 2 (HSV-2) are two of the most prevalent human viruses worldwide. They are known to cause a variety of diseases including genital herpes, meningitis, encephalitis, cold sores and herpes stromal keratitis. The seropositive rate for HSV-1 is around 90%, whereas for HSV-2 it remains around 20-25% for the general adult population. The infections caused by these viruses remain difficult to study because a large proportion of infected individuals are asymptomatic. Furthermore, given the neurotropic characteristics of the virus, studies aimed at understanding the complex pathogenesis in humans is difficult. As a result, animal models have been developed to understand several characteristics of HSV biology, pathogenesis, disease and host responses to infection. These models are also commonly used as the first evaluation of new drugs and vaccines. There are several well-established animal models to study infection with HSV, including mice, guinea pigs and rabbits. Variables within the animal models depend on the species of animal, route of infection, viral strain, dosage, etc. This review aims at summarizing the most commonly used animal models to study HSV pathogenesis and therapies.
Collapse
Affiliation(s)
- Mohammed Tanveer Hussain
- Division of Biotechnology and Molecular Medicine and Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Brent A. Stanfield
- Division of Biotechnology and Molecular Medicine and Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - David I. Bernstein
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
5
|
Morales AE, Gumenick R, Genovese CM, Jang YY, Ouedraogo A, Ibáñez de Garayo M, Pannellini T, Patel S, Bott ME, Alvarez J, Mun SS, Totonchy J, Gautam A, Delgado de la Mora J, Chang S, Wirth D, Horenstein M, Dao T, Scheinberg DA, Rubinstein PG, Semeere A, Martin J, Godfrey CC, Moser CB, Matining RM, Campbell TB, Borok MZ, Krown SE, Cesarman E. Wilms' tumor 1 (WT1) antigen is overexpressed in Kaposi Sarcoma and is regulated by KSHV vFLIP. PLoS Pathog 2024; 20:e1011881. [PMID: 38190392 PMCID: PMC10898863 DOI: 10.1371/journal.ppat.1011881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 02/27/2024] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
In people living with HIV, Kaposi Sarcoma (KS), a vascular neoplasm caused by KS herpesvirus (KSHV/HHV-8), remains one of the most common malignancies worldwide. Individuals living with HIV, receiving otherwise effective antiretroviral therapy, may present with extensive disease requiring chemotherapy. Hence, new therapeutic approaches are needed. The Wilms' tumor 1 (WT1) protein is overexpressed and associated with poor prognosis in several hematologic and solid malignancies and has shown promise as an immunotherapeutic target. We found that WT1 was overexpressed in >90% of a total 333 KS biopsies, as determined by immunohistochemistry and image analysis. Our largest cohort from ACTG, consisting of 294 cases was further analyzed demonstrating higher WT1 expression was associated with more advanced histopathologic subtypes. There was a positive correlation between the proportion of infected cells within KS tissues, assessed by expression of the KSHV-encoded latency-associated nuclear antigen (LANA), and WT1 positivity. Areas with high WT1 expression showed sparse T-cell infiltrates, consistent with an immune evasive tumor microenvironment. We show that major oncogenic isoforms of WT1 are overexpressed in primary KS tissue and observed WT1 upregulation upon de novo infection of endothelial cells with KSHV. KSHV latent viral FLICE-inhibitory protein (vFLIP) upregulated total and major isoforms of WT1, but upregulation was not seen after expression of mutant vFLIP that is unable to bind IKKƴ and induce NFκB. siRNA targeting of WT1 in latent KSHV infection resulted in decreased total cell number and pAKT, BCL2 and LANA protein expression. Finally, we show that ESK-1, a T cell receptor-like monoclonal antibody that recognizes WT1 peptides presented on MHC HLA-A0201, demonstrates increased binding to endothelial cells after KSHV infection or induction of vFLIP expression. We propose that oncogenic isoforms of WT1 are upregulated by KSHV to promote tumorigenesis and immunotherapy directed against WT1 may be an approach for KS treatment.
Collapse
Affiliation(s)
- Ayana E. Morales
- Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Ruby Gumenick
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Caitlyn M. Genovese
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Yun Yeong Jang
- Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Ariene Ouedraogo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Maite Ibáñez de Garayo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Tania Pannellini
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Sanjay Patel
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Matthew E. Bott
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Julio Alvarez
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Sung Soo Mun
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Jennifer Totonchy
- School of Pharmacy, Chapman University, Irvine, California, United States of America
| | - Archana Gautam
- Department of Allergy and Immunology, Icahn School of Medicine, New York, New York, United States of America
| | - Jesus Delgado de la Mora
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Stephanie Chang
- Cornell University, Ithaca, New York, United States of America
| | - Dagmar Wirth
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research Braunschweig, Germany
| | - Marcelo Horenstein
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Tao Dao
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - David A. Scheinberg
- Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Paul G. Rubinstein
- Section of Hematology/Oncology, John H. Stroger Jr Hospital of Cook County (Cook County Hospital), Ruth M. Rothstein Core Center, University of Illinois, Chicago, Illinois, United States of America
| | - Aggrey Semeere
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Jeffrey Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
| | - Catherine C. Godfrey
- Office of the Global AIDS Coordinator, Department of State, Washington, DC, United States of America
| | - Carlee B. Moser
- Center for Biostatistics in AIDS Research, Harvard T H Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Roy M. Matining
- Center for Biostatistics in AIDS Research, Harvard T H Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Thomas B. Campbell
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Margaret Z. Borok
- Department of Internal Medicine, University of Zimbabwe, Harare, Zimbabwe
| | - Susan E. Krown
- Memorial Sloan Kettering Cancer Center (emerita), New York, New York, United States of America
| | - Ethel Cesarman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| |
Collapse
|
6
|
Kutle I, Dittrich A, Wirth D. Mouse Models for Human Herpesviruses. Pathogens 2023; 12:953. [PMID: 37513800 PMCID: PMC10384569 DOI: 10.3390/pathogens12070953] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
More than one hundred herpesviruses have been isolated from different species so far, with nine infecting humans. Infections with herpesviruses are characterized by life-long latency and represent a significant challenge for human health. To investigate the consequences of infections and identify novel treatment options, in vivo models are of particular relevance. The mouse has emerged as an economical small animal model to investigate herpesvirus infections. However, except for herpes simplex viruses (HSV-1, HSV-2), human herpesviruses cannot infect mice. Three natural herpesviruses have been identified in mice: mouse-derived cytomegalovirus (MCMV), mouse herpesvirus 68 (MHV-68), and mouse roseolovirus (MRV). These orthologues are broadly used to investigate herpesvirus infections within the natural host. In the last few decades, immunocompromised mouse models have been developed, allowing the functional engraftment of various human cells and tissues. These xenograft mice represent valuable model systems to investigate human-restricted viruses, making them particularly relevant for herpesvirus research. In this review, we describe the various mouse models used to study human herpesviruses, thereby highlighting their potential and limitations. Emphasis is laid on xenograft mouse models, covering the development and refinement of immune-compromised mice and their application in herpesvirus research.
Collapse
Affiliation(s)
- Ivana Kutle
- Research Group Model Systems for Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Anne Dittrich
- Research Group Model Systems for Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- InSCREENeX GmbH, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Dagmar Wirth
- Research Group Model Systems for Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
7
|
Lee MJ, Park J, Choi S, Yoo SM, Park C, Kim HS, Lee MS. HMGB1, a potential regulator of tumor microenvironment in KSHV-infected endothelial cells. Front Microbiol 2023; 14:1202993. [PMID: 37520371 PMCID: PMC10374257 DOI: 10.3389/fmicb.2023.1202993] [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: 04/10/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
High-mobility group box 1 (HMGB1) is a protein that binds to DNA and participates in various cellular processes, including DNA repair, transcription, and inflammation. It is also associated with cancer progression and therapeutic resistance. Despite its known role in promoting tumor growth and immune evasion in the tumor microenvironment, the contribution of HMGB1 to the development of Kaposi's sarcoma (KS) is not well understood. We investigated the effect of HMGB1 on KS pathogenesis using immortalized human endothelial cells infected with Kaposi's sarcoma-associated human herpes virus (KSHV). Our results showed that a higher amount of HMGB1 was detected in the supernatant of KSHV-infected cells compared to that of mock-infected cells, indicating that KSHV infection induced the secretion of HMGB1 in human endothelial cells. By generating HMGB1 knockout clones from immortalized human endothelial cells using CRISPR/Cas9, we elucidated the role of HMGB1 in KSHV-infected endothelial cells. Our findings indicate that the absence of HMGB1 did not induce lytic replication in KSHV-infected cells, but the cell viability of KSHV-infected cells was decreased in both 2D and 3D cultures. Through the antibody array for cytokines and growth factors, CXCL5, PDGF-AA, G-CSF, Emmprin, IL-17A, and VEGF were found to be suppressed in HMGB1 KO KSHV-infected cells compared to the KSHV-infected wild-type control. Mechanistically, phosphorylation of p38 would be associated with transcriptional regulation of CXCL5, PDGF-A and VEGF. These observations suggest that HMGB1 may play a critical role in KS pathogenesis by regulating cytokine and growth factor secretion and emphasize its potential as a therapeutic target for KS by modulating the tumor microenvironment.
Collapse
Affiliation(s)
- Myung-Ju Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Joohee Park
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Seokjoo Choi
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Seung-Min Yoo
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, Republic of Korea
- Eulji Biomedical Science Research Institute, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Changhoon Park
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Hong Seok Kim
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Myung-Shin Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, Republic of Korea
- Eulji Biomedical Science Research Institute, Eulji University School of Medicine, Daejeon, Republic of Korea
| |
Collapse
|
8
|
Naipauer J, Mesri EA. The Kaposi's sarcoma progenitor enigma: KSHV-induced MEndT-EndMT axis. Trends Mol Med 2023; 29:188-200. [PMID: 36635149 PMCID: PMC9957928 DOI: 10.1016/j.molmed.2022.12.003] [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: 09/29/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 01/11/2023]
Abstract
Endothelial-to-mesenchymal transition has been described in tumors as a source of mesenchymal stroma, while the reverse process has been proposed in tumor vasculogenesis and angiogenesis. A human oncogenic virus, Kaposi's sarcoma herpes virus (KSHV), can regulate both processes in order to transit through this transition 'boulevard' when infecting KS oncogenic progenitor cells. Endothelial or mesenchymal circulating progenitor cells can serve as KS oncogenic progenitors recruited by inflammatory cytokines because KSHV can reprogram one into the other through endothelial-to-mesenchymal and mesenchymal-to-endothelial transitions. Through these novel insights, the identity of the potential oncogenic progenitor of KS is revealed while gaining knowledge of the biology of the mesenchymal-endothelial differentiation axis and pointing to this axis as a therapeutic target in KS.
Collapse
Affiliation(s)
- Julian Naipauer
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina; Tumor Biology Program, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; University of Miami- Center for AIDS Research (UM-CFAR)/Sylvester Comprehensive Cancer Center (CCC) Argentina Consortium for Research and Training in Virally Induced AIDS-Malignancies, University of Miami Miller School of Medicine, Miami, FL, USA; Miami Center for AIDS Research, Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Enrique A Mesri
- Tumor Biology Program, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; University of Miami- Center for AIDS Research (UM-CFAR)/Sylvester Comprehensive Cancer Center (CCC) Argentina Consortium for Research and Training in Virally Induced AIDS-Malignancies, University of Miami Miller School of Medicine, Miami, FL, USA; Miami Center for AIDS Research, Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
9
|
Lee MJ, Lee J, Kang SK, Wirth D, Yoo SM, Park C, Lee MS. CXCL1 confers a survival advantage in Kaposi's sarcoma-associated herpesvirus-infected human endothelial cells through STAT3 phosphorylation. J Med Virol 2023; 95. [PMID: 35869037 DOI: 10.1002/jmv.28020] [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: 06/03/2022] [Revised: 07/05/2022] [Accepted: 07/20/2022] [Indexed: 01/11/2023]
Abstract
Many cytokines produced by Kaposi's sarcoma-associated herpesvirus (KSHV)-infected cells have been shown to participate in the pathogenesis of KSHV. Determination of the exact role of cytokines in Kaposi's sarcoma (KS) pathogenesis is limited, however, by the difficulty to manipulate the target genes in human endothelial cells. In this study, we sought to elucidate the role of cytokines in KSHV-infected human immortalized endothelial cell line (HuARLT cells) by knockout (KO) of the corresponding target genes using the CRISPR/Cas9 system. The cytokine production profile of KSHV-infected HuARLT cells was analyzed using a protein array, and several cytokines were found to be highly upregulated following KSHV infection. This study focused on CXCL1, which was investigated by knocked out in HuARLT cells. KSHV-infected CXCL1 KO cells underwent increased cell death compared to KSHV-infected wild-type (WT) cells and mock-infected CXCL1 KO cells. Lytic replication was not observed in KSHV-infected WT nor CXCL1 KO cells. Phosphorylation of STAT3 was significantly suppressed in KSHV-infected CXCL1 KO cells. Additionally, inhibitors of STAT3 and CXCL1 induced cell death in KSHV-infected endothelial cells. Our results show that CXCL1 production is required for the survival of KSHV-infected endothelial cells, and the CXCL1 to STAT3 phosphorylation signaling pathway may be a therapeutic target for KS.
Collapse
Affiliation(s)
- Myung-Ju Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Jisu Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Su-Kyung Kang
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Dagmar Wirth
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Seung-Min Yoo
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Changhoon Park
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Myung-Shin Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, Republic of Korea
| |
Collapse
|
10
|
Dubich T, Dittrich A, Bousset K, Geffers R, Büsche G, Köster M, Hauser H, Schulz TF, Wirth D. 3D culture conditions support Kaposi's sarcoma herpesvirus (KSHV) maintenance and viral spread in endothelial cells. J Mol Med (Berl) 2021; 99:425-438. [PMID: 33484281 PMCID: PMC7900040 DOI: 10.1007/s00109-020-02020-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/17/2020] [Accepted: 12/04/2020] [Indexed: 12/27/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is a human tumorigenic virus and the etiological agent of an endothelial tumor (Kaposi's sarcoma) and two B cell proliferative diseases (primary effusion lymphoma and multicentric Castleman's disease). While in patients with late stage of Kaposi's sarcoma the majority of spindle cells are KSHV-infected, viral copies are rapidly lost in vitro, both upon culture of tumor-derived cells or from newly infected endothelial cells. We addressed this discrepancy by investigating a KSHV-infected endothelial cell line in various culture conditions and in tumors of xenografted mice. We show that, in contrast to two-dimensional endothelial cell cultures, KSHV genomes are maintained under 3D cell culture conditions and in vivo. Additionally, an increased rate of newly infected cells was detected in 3D cell culture. Furthermore, we show that the PI3K/Akt/mTOR and ATM/γH2AX pathways are modulated and support an improved KSHV persistence in 3D cell culture. These mechanisms may contribute to the persistence of KSHV in tumor tissue in vivo and provide a novel target for KS specific therapeutic interventions. KEY MESSAGES: In vivo maintenance of episomal KSHV can be mimicked in 3D spheroid cultures 3D maintenance of KSHV is associated with an increased de novo infection frequency PI3K/Akt/mTOR and ATM/ γH2AX pathways contribute to viral maintenance.
Collapse
Affiliation(s)
- Tatyana Dubich
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Anne Dittrich
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Kristine Bousset
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Guntram Büsche
- Hematopathology Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Mario Köster
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Hansjörg Hauser
- Staff Unit Scientific Strategy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas F Schulz
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Hannover, Germany
| | - Dagmar Wirth
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany.
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.
- Cluster of Excellence REBIRTH (EXC 62), Hannover Medical School, Hannover, Germany.
| |
Collapse
|
11
|
Wahlicht T, Vièyres G, Bruns SA, Meumann N, Büning H, Hauser H, Schmitz I, Pietschmann T, Wirth D. Controlled Functional Zonation of Hepatocytes In Vitro by Engineering of Wnt Signaling. ACS Synth Biol 2020; 9:1638-1649. [PMID: 32551516 DOI: 10.1021/acssynbio.9b00435] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Key liver functions, including protein synthesis, carbohydrate metabolism, and detoxification, are performed by specific populations of hepatocytes that are defined by their relative positions within the liver lobules. On a molecular level, the functional heterogeneity with periportal and pericentral phenotypes, so-called metabolic liver zonation, is mainly established by a gradient of canonical Wnt signaling activity. Since the relevant physiological cues are missing in in vitro liver models, they fail to reflect the functional heterogeneity and thus lack many liver functions. We synthetically re-engineered Wnt signaling in murine and human hepatocytes using a doxycycline-dependent cassette for externally controlled digital expression of stabilized β-catenin. Thereby, we achieved adjustable mosaic-like activation of Wnt signaling in in vitro-cultured hepatocytes that was resistant to negative-feedback loops. This allowed the establishment of long-term-stable periportal-like and pericentral-like phenotypes that mimic the heterogeneity observed in vivo. The in vitro-zonated hepatocytes show differential expression of drug-metabolizing enzymes and associated differential toxicity and higher levels of autophagy. Furthermore, recombinant adeno-associated virus and hepatitis C virus preferentially transduce the pericentral-like zonation phenotype, suggesting a bias of these viruses that has been unappreciated to date. These tightly controlled in vivo-like systems will be important for studies evaluating aspects of liver zonation and for the assessment of drug toxicity for mouse and man.
Collapse
Affiliation(s)
- Tom Wahlicht
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Gabrielle Vièyres
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, 30625 Hannover, Germany
| | - Svenja A. Bruns
- Systems-Oriented Immunology and Inflammation Research, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Nadja Meumann
- German Center for Infection Research (DZIF), Hannover−Braunschweig Partner Site, 38124 Braunschweig, Germany
| | - Hildegard Büning
- German Center for Infection Research (DZIF), Hannover−Braunschweig Partner Site, 38124 Braunschweig, Germany
- REBIRTH Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany
| | - Hansjörg Hauser
- Department of Scientific Strategy, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Ingo Schmitz
- Systems-Oriented Immunology and Inflammation Research, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Thomas Pietschmann
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, 30625 Hannover, Germany
| | - Dagmar Wirth
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute of Experimental Hematology, Medical University Hannover, 30625 Hannover, Germany
| |
Collapse
|
12
|
Targeting Kaposi's Sarcoma-Associated Herpesvirus ORF21 Tyrosine Kinase and Viral Lytic Reactivation by Tyrosine Kinase Inhibitors Approved for Clinical Use. J Virol 2020; 94:JVI.01791-19. [PMID: 31826996 DOI: 10.1128/jvi.01791-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/04/2019] [Indexed: 12/20/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the cause of three human malignancies: Kaposi's sarcoma, primary effusion lymphoma, and the plasma cell variant of multicentric Castleman disease. Previous research has shown that several cellular tyrosine kinases play crucial roles during several steps in the virus replication cycle. Two KSHV proteins also have protein kinase function: open reading frame (ORF) 36 encodes a serine-threonine kinase, while ORF21 encodes a thymidine kinase (TK), which has recently been found to be an efficient tyrosine kinase. In this study, we explore the role of the ORF21 tyrosine kinase function in KSHV lytic replication. By generating a recombinant KSHV mutant with an enzymatically inactive ORF21 protein, we show that the tyrosine kinase function of ORF21/TK is not required for the progression of the lytic replication in tissue culture but that it is essential for the phosphorylation and activation to toxic moieties of the antiviral drugs zidovudine and brivudine. In addition, we identify several tyrosine kinase inhibitors, already in clinical use against human malignancies, which potently inhibit not only ORF21 TK kinase function but also viral lytic reactivation and the development of KSHV-infected endothelial tumors in mice. Since they target both cellular tyrosine kinases and a viral kinase, some of these compounds might find a use in the treatment of KSHV-associated malignancies.IMPORTANCE Our findings address the role of KSHV ORF21 as a tyrosine kinase during lytic replication and the activation of prodrugs in KSHV-infected cells. We also show the potential of selected clinically approved tyrosine kinase inhibitors to inhibit KSHV TK, KSHV lytic replication, infectious virion release, and the development of an endothelial tumor. Since they target both cellular tyrosine kinases supporting productive viral replication and a viral kinase, these drugs, which are already approved for clinical use, may be suitable for repurposing for the treatment of KSHV-related tumors in AIDS patients or transplant recipients.
Collapse
|
13
|
Gonzalez-Perez AC, Stempel M, Chan B, Brinkmann MM. One Step Ahead: Herpesviruses Light the Way to Understanding Interferon-Stimulated Genes (ISGs). Front Microbiol 2020; 11:124. [PMID: 32117146 PMCID: PMC7018705 DOI: 10.3389/fmicb.2020.00124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/20/2020] [Indexed: 12/13/2022] Open
Abstract
The host immune system is engaged in a constant battle with microorganisms, with the immediate detection of pathogenic invasion and subsequent signalling acting as crucial deterrents against the establishment of a successful infection. For this purpose, cells are equipped with a variety of sensors called pattern recognition receptors (PRR), which rapidly detect intruders leading to the expression of antiviral type I interferons (IFN). Type I IFN are crucial cytokines which exert their biological effects through the induction of hundreds of IFN-stimulated genes (ISGs). The expression profile of these ISGs varies depending on the virus. For a small subset of ISGs, their anti- or even proviral effects have been revealed, however, the vast majority are uncharacterised. The spotlight is now on herpesviruses, with their large coding capacity and long co-evolution with their hosts, as a key to understanding the impact of ISGs during viral infection. Studies are emerging which have identified multiple herpesviral antagonists specifically targeting ISGs, hinting at the significant role these proteins must play in host defence against viral infection, with the promise of more to come. In this review, we will discuss the current knowledge of the complex interplay between ISGs and human herpesviruses: the antiviral role of selected ISGs during herpesviral infections, how herpesviruses antagonise these ISGs and, in some cases, even exploit them to benefit viral infection.
Collapse
Affiliation(s)
| | - Markus Stempel
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Baca Chan
- Viral Genomics Group, Institute for Respiratory Health, The University of Western Australia, Perth, WA, Australia
| | - Melanie M. Brinkmann
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
14
|
Dubich T, Lieske A, Santag S, Beauclair G, Rückert J, Herrmann J, Gorges J, Büsche G, Kazmaier U, Hauser H, Stadler M, Schulz TF, Wirth D. An endothelial cell line infected by Kaposi's sarcoma-associated herpes virus (KSHV) allows the investigation of Kaposi's sarcoma and the validation of novel viral inhibitors in vitro and in vivo. J Mol Med (Berl) 2019; 97:311-324. [PMID: 30610257 DOI: 10.1007/s00109-018-01733-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 12/18/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi's sarcoma (KS), a tumor of endothelial origin predominantly affecting immunosuppressed individuals. Up to date, vaccines and targeted therapies are not available. Screening and identification of anti-viral compounds are compromised by the lack of scalable cell culture systems reflecting properties of virus-transformed cells in patients. Further, the strict specificity of the virus for humans limits the development of in vivo models. In this study, we exploited a conditionally immortalized human endothelial cell line for establishment of in vitro 2D and 3D KSHV latency models and the generation of KS-like xenograft tumors in mice. Importantly, the invasive properties and tumor formation could be completely reverted by purging KSHV from the cells, confirming that tumor formation is dependent on the continued presence of KSHV, rather than being a consequence of irreversible transformation of the infected cells. Upon testing a library of 260 natural metabolites, we selected the compounds that induced viral loss or reduced the invasiveness of infected cells in 2D and 3D endothelial cell culture systems. The efficacy of selected compounds against KSHV-induced tumor formation was verified in the xenograft model. Together, this study shows that the combined use of anti-viral and anti-tumor assays based on the same cell line is predictive for tumor reduction in vivo and therefore allows faithful selection of novel drug candidates against Kaposi's sarcoma. KEY MESSAGES: Novel 2D, 3D, and xenograft mouse models mimic the consequences of KSHV infection. KSHV-induced tumorigenesis can be reverted upon purging the cells from the virus. A 3D invasiveness assay is predictive for tumor reduction in vivo. Chondramid B, epothilone B, and pretubulysin D diminish KS-like lesions in vivo.
Collapse
Affiliation(s)
- Tatyana Dubich
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Anna Lieske
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Susann Santag
- Institute of Virology, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research, Hannover-Braunschweig, Germany
| | - Guillaume Beauclair
- Institute of Virology, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research, Hannover-Braunschweig, Germany
| | - Jessica Rückert
- Institute of Virology, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research, Hannover-Braunschweig, Germany
| | - Jennifer Herrmann
- German Centre for Infection Research, Hannover-Braunschweig, Germany.,Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research, Saarbrücken, Germany
| | - Jan Gorges
- Institute of Organic Chemistry, Saarland University, Saarbrücken, Germany
| | - Guntram Büsche
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Uli Kazmaier
- Institute of Organic Chemistry, Saarland University, Saarbrücken, Germany
| | - Hansjörg Hauser
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Marc Stadler
- German Centre for Infection Research, Hannover-Braunschweig, Germany.,Microbial Drugs, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas F Schulz
- Institute of Virology, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research, Hannover-Braunschweig, Germany
| | - Dagmar Wirth
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany. .,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
15
|
Ullah S, Seidel K, Türkkan S, Warwas DP, Dubich T, Rohde M, Hauser H, Behrens P, Kirschning A, Köster M, Wirth D. Macrophage entrapped silica coated superparamagnetic iron oxide particles for controlled drug release in a 3D cancer model. J Control Release 2018; 294:327-336. [PMID: 30586597 DOI: 10.1016/j.jconrel.2018.12.040] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/06/2018] [Accepted: 12/21/2018] [Indexed: 12/19/2022]
Abstract
Targeted delivery of drugs is a major challenge in treatment of diverse diseases. Systemically administered drugs demand high doses and are accompanied by poor selectivity and side effects on non-target cells. Here, we introduce a new principle for targeted drug delivery. It is based on macrophages as transporters for nanoparticle-coupled drugs as well as controlled release of drugs by hyperthermia mediated disruption of the cargo cells and simultaneous deliberation of nanoparticle-linked drugs. Hyperthermia is induced by an alternating electromagnetic field (AMF) that induces heat from silica-coated superparamagnetic iron oxide nanoparticles (SPIONs). We show proof-of-principle of controlled release by the simultaneous disruption of the cargo cells and the controlled, AMF induced release of a toxin, which was covalently linked to silica-coated SPIONs via a thermo-sensitive linker. Cells that had not been loaded with SPIONs remain unaffected. Moreover, in a 3D co-culture model we demonstrate specific killing of associated tumour cells when employing a ratio as low as 1:40 (SPION-loaded macrophage: tumour cells). Overall, our results demonstrate that AMF induced drug release from macrophage-entrapped nanoparticles is tightly controlled and may be an attractive novel strategy for targeted drug release.
Collapse
Affiliation(s)
- Sami Ullah
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, Braunschweig 38124, Germany
| | - Katja Seidel
- Institute of Organic Chemistry and Centre of Biomolecular Drug Research (BMWZ), Leibniz Universität Hannover, Hannover, Germany
| | - Sibel Türkkan
- Institute of Organic Chemistry and Centre of Biomolecular Drug Research (BMWZ), Leibniz Universität Hannover, Hannover, Germany
| | - Dawid Peter Warwas
- Institute for Inorganic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Tatyana Dubich
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, Braunschweig 38124, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Hansjörg Hauser
- Scientific Strategy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Peter Behrens
- Institute for Inorganic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Andreas Kirschning
- Institute of Organic Chemistry and Centre of Biomolecular Drug Research (BMWZ), Leibniz Universität Hannover, Hannover, Germany
| | - Mario Köster
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, Braunschweig 38124, Germany
| | - Dagmar Wirth
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Inhoffenstr. 7, Braunschweig 38124, Germany; Institute for Experimental Hematology, Medical University Hannover, Hannover, Germany.
| |
Collapse
|
16
|
Krüger-Genge A, Schulz C, Kratz K, Lendlein A, Jung F. Comparison of two substrate materials used as negative control in endothelialization studies: Glass versus polymeric tissue culture plate. Clin Hemorheol Microcirc 2018; 69:437-445. [PMID: 29843229 DOI: 10.3233/ch-189904] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The endothelialization of synthetic surfaces applied as cardiovascular implant materials is an important issue to ensure the anti-thrombotic quality of a biomaterial. However, the rapid and constant development of a functionally-confluent endothelial cell monolayer is challenging. In order to investigate the compatibility of potential implant materials with endothelial cells several in vitro studies are performed. Here, glass and tissue culture plates (TCP) are often used as reference materials for in vitro pre-testing. However, a direct comparison of both substrates is lacking.Therefore, a comparison of study results is difficult, since results are often related to various reference materials. In this study, the endothelialization of glass and TCP was investigated in terms of adherence, morphology, integrity, viability and function using human umbilical vein endothelial cells (HUVEC).On both substrates an almost functionally confluent HUVEC monolayer was developed after nine days of cell seeding with clearly visible cell rims, decreased stress fiber formation and a pronounced marginal filament band. The viability of HUVEC was comparable for both substrates nine days after cell seeding with only a few dead cells. According to that, the cell membrane integrity as well as the metabolic activity showed no differences between TCP and glass. However, a significant difference was observed for the secretion of IL-6 and IL-8. The concentration of both cytokines, which are associated with migratory activity, was increased in the supernatant of HUVEC seeded on TCP. This result matches well with the slightly increased number of adherent HUVEC on TCP.In conclusion, these findings indicate that both reference materials are almost comparable and can be used equivalently as control materials in in vitro endothelialization studies.
Collapse
Affiliation(s)
- Anne Krüger-Genge
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Christian Schulz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Karl Kratz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Friedrich Jung
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| |
Collapse
|
17
|
Lipps C, Klein F, Wahlicht T, Seiffert V, Butueva M, Zauers J, Truschel T, Luckner M, Köster M, MacLeod R, Pezoldt J, Hühn J, Yuan Q, Müller PP, Kempf H, Zweigerdt R, Dittrich-Breiholz O, Pufe T, Beckmann R, Drescher W, Riancho J, Sañudo C, Korff T, Opalka B, Rebmann V, Göthert JR, Alves PM, Ott M, Schucht R, Hauser H, Wirth D, May T. Expansion of functional personalized cells with specific transgene combinations. Nat Commun 2018. [PMID: 29520052 PMCID: PMC5843645 DOI: 10.1038/s41467-018-03408-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Fundamental research and drug development for personalized medicine necessitates cell cultures from defined genetic backgrounds. However, providing sufficient numbers of authentic cells from individuals poses a challenge. Here, we present a new strategy for rapid cell expansion that overcomes current limitations. Using a small gene library, we expanded primary cells from different tissues, donors, and species. Cell-type-specific regimens that allow the reproducible creation of cell lines were identified. In depth characterization of a series of endothelial and hepatocytic cell lines confirmed phenotypic stability and functionality. Applying this technology enables rapid, efficient, and reliable production of unlimited numbers of personalized cells. As such, these cell systems support mechanistic studies, epidemiological research, and tailored drug development. Personalised medicine requires cell cultures from defined genetic backgrounds, but providing sufficient numbers of cells is a challenge. Here the authors develop gene cocktails to expand primary cells from a variety of different tissues and species, and show that expanded endothelial and hepatic cells retain properties of the differentiated phenotype.
Collapse
Affiliation(s)
- Christoph Lipps
- Model Systems for Infection and Immunity, HZI - Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany.,Experimental Cardiology, Justus-Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany
| | - Franziska Klein
- Department of Gene Regulation and Differentiation, HZI - Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Tom Wahlicht
- Model Systems for Infection and Immunity, HZI - Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Virginia Seiffert
- Department of Gene Regulation and Differentiation, HZI - Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Milada Butueva
- Model Systems for Infection and Immunity, HZI - Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | | | | | - Martin Luckner
- InSCREENeX GmbH, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Mario Köster
- Department of Gene Regulation and Differentiation, HZI - Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Roderick MacLeod
- Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Jörn Pezoldt
- Experimental Immunology, HZI - Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Jochen Hühn
- Experimental Immunology, HZI - Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Qinggong Yuan
- Department of Gastroenterology, Hepatology, Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,Translational Research Group Cell and Gene Therapy, Twincore - Centre for Experimental and Clinical Infection Research GmbH, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Peter Paul Müller
- Department of Gene Regulation and Differentiation, HZI - Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Henning Kempf
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, MHH, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, MHH, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | | | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, 52074, Aachen, Germany
| | - Rainer Beckmann
- Department of Anatomy and Cell Biology, RWTH Aachen University, 52074, Aachen, Germany
| | - Wolf Drescher
- Department of Orthopaedics, Aachen University Hospital, RWTH Aachen University, Aachen, 52074, Germany.,Department of Orthopedic Surgery of the Lower Limb and Arthroplasty, Rummelsberg Hospital, Schwarzenbruck, 90592, Germany
| | - Jose Riancho
- Department of Internal Medicine, Hospital U.M. Valdecilla, University of Cantabria, IDIVAL, 39008, Santander, Spain
| | - Carolina Sañudo
- Department of Internal Medicine, Hospital U.M. Valdecilla, University of Cantabria, IDIVAL, 39008, Santander, Spain
| | - Thomas Korff
- Institute of Physiology and Pathophysiology, RG Blood Vessel Remodeling, University Heidelberg, Im Neuenheimer Feld 326, 69120, Heidelberg, Germany
| | - Bertram Opalka
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Vera Rebmann
- Institute for Transfusion Medicine, University Hospital Essen, Virchowstr. 179, 45147, Essen, Germany
| | - Joachim R Göthert
- Department of Hematology, West German Cancer Center (WTZ), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Paula M Alves
- Instituto de Biologia Experimental e Tecnologica, Universidade Nova de Lisboa, Oeiras, 2781-901, Portugal
| | - Michael Ott
- Department of Gastroenterology, Hepatology, Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,Translational Research Group Cell and Gene Therapy, Twincore - Centre for Experimental and Clinical Infection Research GmbH, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Roland Schucht
- InSCREENeX GmbH, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Hansjörg Hauser
- Department of Gene Regulation and Differentiation, HZI - Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Dagmar Wirth
- Model Systems for Infection and Immunity, HZI - Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany. .,Experimental Hematology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Tobias May
- InSCREENeX GmbH, Inhoffenstr. 7, 38124, Braunschweig, Germany.
| |
Collapse
|
18
|
Bussey KA, Lau U, Schumann S, Gallo A, Osbelt L, Stempel M, Arnold C, Wissing J, Gad HH, Hartmann R, Brune W, Jänsch L, Whitehouse A, Brinkmann MM. The interferon-stimulated gene product oligoadenylate synthetase-like protein enhances replication of Kaposi's sarcoma-associated herpesvirus (KSHV) and interacts with the KSHV ORF20 protein. PLoS Pathog 2018; 14:e1006937. [PMID: 29499066 PMCID: PMC5851652 DOI: 10.1371/journal.ppat.1006937] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 03/14/2018] [Accepted: 02/12/2018] [Indexed: 12/23/2022] Open
Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) is one of the few oncogenic human viruses known to date. Its large genome encodes more than 85 proteins and includes both unique viral proteins as well as proteins conserved amongst herpesviruses. KSHV ORF20 is a member of the herpesviral core UL24 family, but the function of ORF20 and its role in the viral life cycle is not well understood. ORF20 encodes three largely uncharacterized isoforms, which we found were localized predominantly in the nuclei and nucleoli. Quantitative affinity purification coupled to mass spectrometry (q-AP-MS) identified numerous specific interacting partners of ORF20, including ribosomal proteins and the interferon-stimulated gene product (ISG) oligoadenylate synthetase-like protein (OASL). Both endogenous and transiently transfected OASL co-immunoprecipitated with ORF20, and this interaction was conserved among all ORF20 isoforms and multiple ORF20 homologs of the UL24 family in other herpesviruses. Characterization of OASL interacting partners by q-AP-MS identified a very similar interactome to that of ORF20. Both ORF20 and OASL copurified with 40S and 60S ribosomal subunits, and when they were co-expressed, they associated with polysomes. Although ORF20 did not have a global effect on translation, ORF20 enhanced RIG-I induced expression of endogenous OASL in an IRF3-dependent but IFNAR-independent manner. OASL has been characterized as an ISG with antiviral activity against some viruses, but its role for gammaherpesviruses was unknown. We show that OASL and ORF20 mRNA expression were induced early after reactivation of latently infected HuARLT-rKSHV.219 cells. Intriguingly, we found that OASL enhanced infection of KSHV. During infection with a KSHV ORF20stop mutant, however, OASL-dependent enhancement of infectivity was lost. Our data have characterized the interaction of ORF20 with OASL and suggest ORF20 usurps the function of OASL to benefit KSHV infection. The herpesviruses are a family of large double-stranded DNA viruses that cause a variety of illnesses from chicken pox to cancer. Kaposi’s sarcoma-associated herpesvirus (KSHV) is a cancer-causing herpesvirus and can lead to development of Kaposi’s sarcoma, a major form of cancer in HIV-positive patients. As for all herpesviruses, infection with KSHV is lifelong. Exactly how KSHV initiates and maintains its infection is still not well understood, but it must manipulate the host cell to establish favorable conditions. Likewise, the host has developed a complicated system to fight off invaders, which includes the production of interferon-stimulated gene products. We have now found that KSHV exploits one such host cell protein, the oligoadenylate synthetase-like protein (OASL). Rather than OASL acting as an antiviral protein as it does during many other viral infections, KSHV appears to have found a way to utilize OASL for its own benefit. The KSHV protein ORF20 interacts with OASL, they co-localize in nucleoli, and both ORF20 and OASL associate and purify with components of the cellular translational machinery. This may help viral infection by selectively controlling protein production.
Collapse
Affiliation(s)
- Kendra A. Bussey
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Ulrike Lau
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Sophie Schumann
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Antonio Gallo
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Lisa Osbelt
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Markus Stempel
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Christine Arnold
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Josef Wissing
- Cellular Proteomics Research Group, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Hans Henrik Gad
- Center for Structural Biology, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Rune Hartmann
- Center for Structural Biology, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Wolfram Brune
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Lothar Jänsch
- Cellular Proteomics Research Group, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Adrian Whitehouse
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Melanie M. Brinkmann
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|