1
|
Lurain KA, Ramaswami R, Krug LT, Whitby D, Ziegelbauer JM, Wang HW, Yarchoan R. HIV-associated cancers and lymphoproliferative disorders caused by Kaposi sarcoma herpesvirus and Epstein-Barr virus. Clin Microbiol Rev 2024; 37:e0002223. [PMID: 38899877 PMCID: PMC11391709 DOI: 10.1128/cmr.00022-23] [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] [Indexed: 06/21/2024] Open
Abstract
SUMMARYWithin weeks of the first report of acquired immunodeficiency syndrome (AIDS) in 1981, it was observed that these patients often had Kaposi sarcoma (KS), a hitherto rarely seen skin tumor in the USA. It soon became apparent that AIDS was also associated with an increased incidence of high-grade lymphomas caused by Epstein-Barr virus (EBV). The association of AIDS with KS remained a mystery for more than a decade until Kaposi sarcoma-associated herpesvirus (KSHV) was discovered and found to be the cause of KS. KSHV was subsequently found to cause several other diseases associated with AIDS and human immunodeficiency virus (HIV) infection. People living with HIV/AIDS continue to have an increased incidence of certain cancers, and many of these cancers are caused by EBV and/or KSHV. In this review, we discuss the epidemiology, virology, pathogenesis, clinical manifestations, and treatment of cancers caused by EBV and KSHV in persons living with HIV.
Collapse
Affiliation(s)
- Kathryn A Lurain
- The HIV and AIDS Malignancy Branch, Center for Cancer Research, Bethesda, Maryland, USA
| | - Ramya Ramaswami
- The HIV and AIDS Malignancy Branch, Center for Cancer Research, Bethesda, Maryland, USA
| | - Laurie T Krug
- The HIV and AIDS Malignancy Branch, Center for Cancer Research, Bethesda, Maryland, USA
| | - Denise Whitby
- Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Joseph M Ziegelbauer
- The HIV and AIDS Malignancy Branch, Center for Cancer Research, Bethesda, Maryland, USA
| | - Hao-Wei Wang
- Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland, USA
| | - Robert Yarchoan
- The HIV and AIDS Malignancy Branch, Center for Cancer Research, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Ciani L, Libonati A, Dri M, Pomella S, Campanella V, Barillari G. About a Possible Impact of Endodontic Infections by Fusobacterium nucleatum or Porphyromonas gingivalis on Oral Carcinogenesis: A Literature Overview. Int J Mol Sci 2024; 25:5083. [PMID: 38791123 PMCID: PMC11121237 DOI: 10.3390/ijms25105083] [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: 03/21/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
Periodontitis is linked to the onset and progression of oral squamous cell carcinoma (OSCC), an epidemiologically frequent and clinically aggressive malignancy. In this context, Fusobacterium (F.) nucleatum and Porphyromonas (P.) gingivalis, two bacteria that cause periodontitis, are found in OSCC tissues as well as in oral premalignant lesions, where they exert pro-tumorigenic activities. Since the two bacteria are present also in endodontic diseases, playing a role in their pathogenesis, here we analyze the literature searching for information on the impact that endodontic infection by P. gingivalis or F. nucleatum could have on cellular and molecular events involved in oral carcinogenesis. Results from the reviewed papers indicate that infection by P. gingivalis and/or F. nucleatum triggers the production of inflammatory cytokines and growth factors in dental pulp cells or periodontal cells, affecting the survival, proliferation, invasion, and differentiation of OSCC cells. In addition, the two bacteria and the cytokines they induce halt the differentiation and stimulate the proliferation and invasion of stem cells populating the dental pulp or the periodontium. Although most of the literature confutes the possibility that bacteria-induced endodontic inflammatory diseases could impact on oral carcinogenesis, the papers we have analyzed and discussed herein recommend further investigations on this topic.
Collapse
Affiliation(s)
- Luca Ciani
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (L.C.); (S.P.); (V.C.)
| | - Antonio Libonati
- Department of Surgical Sciences, Catholic University of Our Lady of Good Counsel of Tirane, 1001 Tirana, Albania;
| | - Maria Dri
- Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Silvia Pomella
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (L.C.); (S.P.); (V.C.)
| | - Vincenzo Campanella
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (L.C.); (S.P.); (V.C.)
| | - Giovanni Barillari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (L.C.); (S.P.); (V.C.)
| |
Collapse
|
3
|
Lacunza E, Ahuja A, Coso OA, Abba M, Ramos JC, Cesarman E, Mesri EA, Naipauer J. Unveiling the role of KSHV-infected human mesenchymal stem cells in Kaposi's sarcoma initiation. J Med Virol 2024; 96:e29684. [PMID: 38773828 DOI: 10.1002/jmv.29684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/24/2024]
Abstract
Kaposi's sarcoma (KS) may derive from Kaposi's sarcoma herpesvirus (KSHV)-infected human mesenchymal stem cells (hMSCs) that migrate to sites characterized by inflammation and angiogenesis, promoting the initiation of KS. By analyzing the RNA sequences of KSHV-infected primary hMSCs, we have identified specific cell subpopulations, mechanisms, and conditions involved in the initial stages of KSHV-induced transformation and reprogramming of hMSCs into KS progenitor cells. Under proangiogenic environmental conditions, KSHV can reprogram hMSCs to exhibit gene expression profiles more similar to KS tumors, activating cell cycle progression, cytokine signaling pathways, endothelial differentiation, and upregulating KSHV oncogenes indicating the involvement of KSHV infection in inducing the mesenchymal-to-endothelial (MEndT) transition of hMSCs. This finding underscores the significance of this condition in facilitating KSHV-induced proliferation and reprogramming of hMSCs towards MEndT and closer to KS gene expression profiles, providing further evidence of these cell subpopulations as precursors of KS cells that thrive in a proangiogenic environment.
Collapse
Affiliation(s)
- Ezequiel Lacunza
- Centro de Investigaciones Inmunologicas Basicas y Aplicadas, Facultad de Ciencias Medicas, Universidad Nacional de La Plata, La Plata, Argentina
- University of Miami-Centre for AIDS Research/Sylvester Cancer Comprehensive Center Argentina Consortium for Research and Training in Virally Induced AIDS-Malignancies, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Anuj Ahuja
- Tumor Biology Program, Sylvester Comprehensive Cancer Center and Miami Center for AIDS Research, Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Omar A Coso
- University of Miami-Centre for AIDS Research/Sylvester Cancer Comprehensive Center Argentina Consortium for Research and Training in Virally Induced AIDS-Malignancies, University of Miami Miller School of Medicine, Miami, Florida, USA
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Martin Abba
- Centro de Investigaciones Inmunologicas Basicas y Aplicadas, Facultad de Ciencias Medicas, Universidad Nacional de La Plata, La Plata, Argentina
- University of Miami-Centre for AIDS Research/Sylvester Cancer Comprehensive Center Argentina Consortium for Research and Training in Virally Induced AIDS-Malignancies, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Juan Carlos Ramos
- University of Miami-Centre for AIDS Research/Sylvester Cancer Comprehensive Center Argentina Consortium for Research and Training in Virally Induced AIDS-Malignancies, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
- Center for AIDS Research, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ethel Cesarman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Enrique A Mesri
- University of Miami-Centre for AIDS Research/Sylvester Cancer Comprehensive Center Argentina Consortium for Research and Training in Virally Induced AIDS-Malignancies, University of Miami Miller School of Medicine, Miami, Florida, USA
- Tumor Biology Program, Sylvester Comprehensive Cancer Center and Miami Center for AIDS Research, Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Julian Naipauer
- University of Miami-Centre for AIDS Research/Sylvester Cancer Comprehensive Center Argentina Consortium for Research and Training in Virally Induced AIDS-Malignancies, University of Miami Miller School of Medicine, Miami, Florida, USA
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
4
|
Taheri M, Tehrani HA, Dehghani S, Rajabzadeh A, Alibolandi M, Zamani N, Arefian E, Ramezani M. Signaling crosstalk between mesenchymal stem cells and tumor cells: Implications for tumor suppression or progression. Cytokine Growth Factor Rev 2024; 76:30-47. [PMID: 38341337 DOI: 10.1016/j.cytogfr.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024]
Abstract
Mesenchymal stem cells (MSCs) have been extensively used in various therapeutic applications over the last two decades, particularly in regenerative medicine and cancer treatment. MSCs have the ability to differentiate into mesodermal and non-mesodermal lineages, which makes them a popular choice in tissue engineering and regenerative medicine. Studies have shown that MSCs have inherent tumor-suppressive properties and can affect the behavior of multiple cells contributing to tumor development. Additionally, MSCs possess a tumor tropism property and have a hypoimmune nature. The intrinsic features of MSCs along with their potential to undergo genetic manipulation and be loaded with various anticancer therapeutics have motivated researchers to use them in different cancer therapy approaches without considering their complex dynamic biological aspects. However, despite their desirable features, several reports have shown that MSCs possess tumor-supportive properties. These contradictory results signify the sophisticated nature of MSCs and warn against the potential therapeutic applications of MSCs. Therefore, researchers should meticulously consider the biological properties of MSCs in preclinical and clinical studies to avoid any undesirable outcomes. This manuscript reviews preclinical studies on MSCs and cancer from the last two decades, discusses how MSC properties affect tumor progression and explains the mechanisms behind tumor suppressive and supportive functions. It also highlights critical cellular pathways that could be targeted in future studies to improve the safety and effectiveness of MSC-based therapies for cancer treatment. The insights obtained from this study will pave the way for further clinical research on MSCs and development of more effective cancer treatments.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Rajabzadeh
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nina Zamani
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Lu ZZ, Sun C, Zhang X, Peng Y, Wang Y, Zeng Y, Zhu N, Yuan Y, Zeng MS. Neuropilin 1 is an entry receptor for KSHV infection of mesenchymal stem cell through TGFBR1/2-mediated macropinocytosis. SCIENCE ADVANCES 2023; 9:eadg1778. [PMID: 37224259 DOI: 10.1126/sciadv.adg1778] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/19/2023] [Indexed: 05/26/2023]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) has been implicated in the pathogenesis of Kaposi's sarcoma (KS) and other malignancies. The cellular origin of KS has been suggested to be either mesenchymal stem cells (MSCs) or endothelial cells. However, receptor(s) for KSHV to infect MSCs remains unknown. By combining bioinformatics analysis and shRNA screening, we identify neuropilin 1 (NRP1) as an entry receptor for KSHV infection of MSCs. Functionally, NRP1 knockout and overexpression in MSCs significantly reduce and promote, respectively, KSHV infection. Mechanistically, NRP1 facilitated the binding and internalization of KSHV by interacting with KSHV glycoprotein B (gB), which was blocked by soluble NRP1 protein. Furthermore, NRP1 interacts with TGF-β receptor type 2 (TGFBR2) through their respective cytoplasmic domains and thus activates the TGFBR1/2 complex, which facilitates the macropinocytosis-mediated KSHV internalization via the small GTPases Cdc42 and Rac1. Together, these findings implicate that KSHV has evolved a strategy to invade MSCs by harnessing NRP1 and TGF-beta receptors to stimulate macropinocytosis.
Collapse
Affiliation(s)
- Zheng-Zhou Lu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center and Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cong Sun
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center and Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaolin Zhang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center and Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yingying Peng
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Wang
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Zeng
- Precision clinical laboratory, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong 524037, China
- Key Laboratory of Xinjiang Endemic and Ethnic Disease, School of Medicine, Shihezi University, Shihezi 832000, China
| | - Nannan Zhu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center and Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Yuan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center and Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- Institute for Advanced Medical Research, Shandong University, Jinan, Shandong, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center and Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Damania B, Dittmer DP. Today's Kaposi sarcoma is not the same as it was 40 years ago, or is it? J Med Virol 2023; 95:e28773. [PMID: 37212317 PMCID: PMC10266714 DOI: 10.1002/jmv.28773] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/20/2023] [Accepted: 04/22/2023] [Indexed: 05/23/2023]
Abstract
This review will provide an overview of the notion that Kaposi sarcoma (KS) is a disease that manifests under diverse and divergent circumstances. We begin with a historical introduction of KS and KS-associated herpesvirus (KSHV), highlight the diversity of clinical presentations of KS, summarize what we know about the cell of origin for this tumor, explore KSHV viral load as a potential biomarker for acute KSHV infections and KS-associated complications, and discuss immune modulators that impact KSHV infection, KSHV persistence, and KS disease.
Collapse
Affiliation(s)
- Blossom Damania
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, 450 West Drive CB#7295, Rm 12-048, Chapel Hill, NC 27599
| | - Dirk P. Dittmer
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, 450 West Drive CB#7295, Rm 12-048, Chapel Hill, NC 27599
| |
Collapse
|
7
|
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
|
8
|
Tourlaki A, Nazzaro G, Wei Y, Buffon S, Mattioli MA, Marzano AV, Brambilla L. Clinical, Dermoscopic, Ultrasonographic, and Histopathologic Correlations in Kaposi's Sarcoma Lesions and Their Differential Diagnoses: A Single-Center Prospective Study. J Clin Med 2022; 12:278. [PMID: 36615078 PMCID: PMC9821103 DOI: 10.3390/jcm12010278] [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: 11/16/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
(1) Background: Kaposi's sarcoma (KS) is an angioproliferative neoplasm typically appearing as angiomatous patches, plaques, and/or nodules on the skin. Dermoscopy and ultrasonography have been suggested as an aid in the diagnosis of KS, but there is little evidence in the literature, especially regarding its possible differential diagnoses. Our aim is to describe and compare the clinical, dermoscopic, and ultrasonographic features of KS and KS-like lesions. (2) Methods: we conducted a prospective study on 25 consecutive patients who were first referred to our tertiary care center from January to May 2021 for a possible KS. (3) Results: 41 cutaneous lesions were examined by means of dermoscopy, Doppler ultrasonography, and pathology, 32 of which were KS-related, while the remaining 9 were lesions with clinical resemblance to KS. On dermoscopy, a purplish-red pigmentation, scaly surface, and the collarette sign were the most common features among KS lesions (81.3%, 46.9%, and 28.1%, respectively). On US, all 9 KS plaques and 21 KS nodules presented a hypoechoic image. Dermoscopic and Doppler ultrasonographic findings of KS-like lesions, such as cherry angioma, venous lake, glomus tumor, pyogenic granuloma, and angiosarcoma were also analyzed. (4) Conclusions: dermoscopy and Doppler ultrasonography can be useful to better assess the features of KS lesions and in diagnosing equivocal KS-like lesions.
Collapse
Affiliation(s)
- Athanasia Tourlaki
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Gianluca Nazzaro
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Yiran Wei
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Stefano Buffon
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Maria A. Mattioli
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Angelo V. Marzano
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Lucia Brambilla
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
9
|
Lebeau G, Ah-Pine F, Daniel M, Bedoui Y, Vagner D, Frumence E, Gasque P. Perivascular Mesenchymal Stem/Stromal Cells, an Immune Privileged Niche for Viruses? Int J Mol Sci 2022; 23:ijms23148038. [PMID: 35887383 PMCID: PMC9317325 DOI: 10.3390/ijms23148038] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) play a critical role in response to stress such as infection. They initiate the removal of cell debris, exert major immunoregulatory activities, control pathogens, and lead to a remodeling/scarring phase. Thus, host-derived ‘danger’ factors released from damaged/infected cells (called alarmins, e.g., HMGB1, ATP, DNA) as well as pathogen-associated molecular patterns (LPS, single strand RNA) can activate MSCs located in the parenchyma and around vessels to upregulate the expression of growth factors and chemoattractant molecules that influence immune cell recruitment and stem cell mobilization. MSC, in an ultimate contribution to tissue repair, may also directly trans- or de-differentiate into specific cellular phenotypes such as osteoblasts, chondrocytes, lipofibroblasts, myofibroblasts, Schwann cells, and they may somehow recapitulate their neural crest embryonic origin. Failure to terminate such repair processes induces pathological scarring, termed fibrosis, or vascular calcification. Interestingly, many viruses and particularly those associated to chronic infection and inflammation may hijack and polarize MSC’s immune regulatory activities. Several reports argue that MSC may constitute immune privileged sanctuaries for viruses and contributing to long-lasting effects posing infectious challenges, such as viruses rebounding in immunocompromised patients or following regenerative medicine therapies using MSC. We will herein review the capacity of several viruses not only to infect but also to polarize directly or indirectly the functions of MSC (immunoregulation, differentiation potential, and tissue repair) in clinical settings.
Collapse
Affiliation(s)
- Grégorie Lebeau
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
| | - Franck Ah-Pine
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Service Anatomo-Pathologie, CHU de la Réunion, 97400 Saint-Denis, France
| | - Matthieu Daniel
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
| | - Yosra Bedoui
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
| | - Damien Vagner
- Service de Médecine Interne, CHU de la Réunion, 97400 Saint-Denis, France;
| | - Etienne Frumence
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
| | - Philippe Gasque
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
- Correspondence:
| |
Collapse
|
10
|
Abere B, Zhou H, Shuda M, Stolz DB, Rapchak K, Moore PS, Chang Y. Replication Kinetics for a Reporter Merkel Cell Polyomavirus. Viruses 2022; 14:473. [PMID: 35336880 PMCID: PMC8950423 DOI: 10.3390/v14030473] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/18/2022] [Accepted: 02/20/2022] [Indexed: 01/27/2023] Open
Abstract
Merkel cell polyomavirus (MCV) causes one of the most aggressive human skin cancers, but laboratory studies on MCV replication have proven technically difficult. We report the first recombinase-mediated MCV minicircle (MCVmc) system that generates high levels of circularized virus, allowing facile MCV genetic manipulation and characterization of viral gene expression kinetics during replication. Mutations to Fbw7, Skp2, β-TrCP and hVam6p interaction sites, or to the stem loop sequence for the MCV-encoded miRNA precursor, markedly increase viral replication, whereas point mutation to an origin-binding site eliminates active virus replication. To further increase the utility of this system, an mScarlet fusion protein was inserted into the VP1 c-terminus to generate a non-infectious reporter virus for studies on virus kinetics. When this reporter virus genome is heterologously expressed together with MCV VP1 and VP2, virus-like particles are generated. The reporter virus genome is encapsidated and can be used at lower biosafety levels for one-round infection studies. Our findings reveal that MCV has multiple, self-encoded viral restriction mechanisms to promote viral latency over lytic replication, and these mechanisms are now amenable to examination using a recombinase technology.
Collapse
Affiliation(s)
- Bizunesh Abere
- Cancer Virology Program, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA; (B.A.); (H.Z.); (M.S.); (K.R.)
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Hongzhao Zhou
- Cancer Virology Program, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA; (B.A.); (H.Z.); (M.S.); (K.R.)
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Masahiro Shuda
- Cancer Virology Program, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA; (B.A.); (H.Z.); (M.S.); (K.R.)
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Donna B. Stolz
- Department Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Kyle Rapchak
- Cancer Virology Program, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA; (B.A.); (H.Z.); (M.S.); (K.R.)
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Patrick S. Moore
- Cancer Virology Program, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA; (B.A.); (H.Z.); (M.S.); (K.R.)
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yuan Chang
- Cancer Virology Program, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA; (B.A.); (H.Z.); (M.S.); (K.R.)
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
11
|
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV)-associated primary effusion lymphomas (PEL) are traditionally viewed as homogenous regarding viral transcription and lineage of origin, but so far this contention has not been explored at the single-cell level. Single-cell RNA sequencing of latently infected PEL supports the existence of multiple subpopulations even within a single cell line. At most 1% of the cells showed evidence of near-complete lytic transcription. The majority of cells only expressed the canonical viral latent transcripts: those originating from the latency locus, the viral interferon regulatory factor locus, and the viral lncRNA nut-1/Pan/T1.1; however, a significant fraction of cells showed various degrees of more permissive transcription, and some showed no evidence of KSHV transcripts whatsoever. Levels of viral interleukin-6 (IL-6)/K2 mRNA emerged as the most distinguishing feature to subset KSHV-infected PEL. One newly uncovered phenotype is the existence of BCBL-1 cells that readily adhered to fibronectin and that displayed mesenchymal lineage-like characteristics. IMPORTANCE Latency is the defining characteristic of the Herpesviridae and central to the tumorigenesis phenotype of Kaposi's sarcoma-associated herpesvirus (KSHV). KSHV-driven primary effusion lymphomas (PEL) rapidly develop resistance to therapy, suggesting tumor instability and plasticity. At any given time, a fraction of PEL cells spontaneously reactivate KSHV, suggesting transcriptional heterogeneity even within a clonal cell line under optimal growth conditions. This study employed single-cell mRNA sequencing to explore the within-population variability of KSHV transcription and how it relates to host cell transcription. Individual clonal PEL cells exhibited differing patterns of viral transcription. Most cells showed the canonical pattern of KSHV latency (LANA, vCyc, vFLIP, Kaposin, and vIRFs), but a significant fraction evidenced extended viral gene transcription, including of the viral IL-6 homolog, open reading frame K2. This study suggests new targets of intervention for PEL. It establishes a conceptual framework to design KSHV cure studies analogous to those for HIV.
Collapse
|
12
|
Méndez-Solís O, Bendjennat M, Naipauer J, Theodoridis PR, Ho JJD, Verdun RE, Hare JM, Cesarman E, Lee S, Mesri EA. Kaposi's sarcoma herpesvirus activates the hypoxia response to usurp HIF2α-dependent translation initiation for replication and oncogenesis. Cell Rep 2021; 37:110144. [PMID: 34965440 PMCID: PMC9121799 DOI: 10.1016/j.celrep.2021.110144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/19/2021] [Accepted: 11/29/2021] [Indexed: 12/19/2022] Open
Abstract
Kaposi's sarcoma herpesvirus (KSHV) is an angiogenesis-inducing oncovirus whose ability to usurp the oxygen-sensing machinery is central to its oncogenicity. By upregulating the hypoxia-inducible factors (HIFs), KSHV reprograms infected cells to a hypoxia-like state, triggering angiogenesis. Here we identify a link between KSHV replicative biology and oncogenicity by showing that KSHV's ability to regulate HIF2α levels and localization to the endoplasmic reticulum (ER) in normoxia enables translation of viral lytic mRNAs through the HIF2α-regulated eIF4E2 translation-initiation complex. This mechanism of translation in infected cells is critical for lytic protein synthesis and contributes to KSHV-induced PDGFRA activation and VEGF secretion. Thus, KSHV regulation of the oxygen-sensing machinery allows virally infected cells to initiate translation via the mTOR-dependent eIF4E1 or the HIF2α-dependent, mTOR-independent, eIF4E2. This "translation initiation plasticity" (TRIP) is an oncoviral strategy used to optimize viral protein expression that links molecular strategies of viral replication to angiogenicity and oncogenesis.
Collapse
Affiliation(s)
- Omayra Méndez-Solís
- Tumor Biology Program, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Miami Center for AIDS Research, Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Mourad Bendjennat
- Tumor Biology Program, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Miami Center for AIDS Research, Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Julian Naipauer
- Tumor Biology Program, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Miami Center for AIDS Research, Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Phaedra R Theodoridis
- Tumor Biology Program, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - J J David Ho
- Tumor Biology Program, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ramiro E Verdun
- Cancer Epigenetics Program, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua M Hare
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ethel Cesarman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Stephen Lee
- Tumor Biology Program, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Enrique A Mesri
- Tumor Biology Program, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Miami Center for AIDS Research, Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
13
|
Kaposi's sarcoma herpesvirus is associated with osteosarcoma in Xinjiang populations. Proc Natl Acad Sci U S A 2021; 118:2016653118. [PMID: 33674386 DOI: 10.1073/pnas.2016653118] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Osteosarcoma is the most common malignant tumor of bone predominately affecting adolescents and young adults. Based on animal studies, a viral etiology of osteosarcoma was proposed more than a half-century ago, but no viral association with human osteosarcoma has been found. The Uyghur ethnic population in Xinjiang, China, has an unusually high prevalence of Kaposi's sarcoma-associated herpesvirus (KSHV) infection and elevated incidence of osteosarcoma. In the current study, we explored the possible association of KSHV infection and osteosarcoma occurrence. Our seroepidemiological study revealed that KSHV prevalence was significantly elevated in Uyghur osteosarcoma patients versus the general Uyghur population (OR, 10.23; 95%CI, 4.25, 18.89). The KSHV DNA genome and viral latent nuclear antigen LANA were detected in most osteosarcoma tumor cells. Gene expression profiling analysis showed that KSHV-positive osteosarcoma represents a distinct subtype of osteosarcomas with viral gene-activated signaling pathways important for osteosarcoma development. We conclude that KSHV infection is a risk factor for osteosarcoma, and KSHV is associated with some osteosarcomas, representing a newly identified viral-associated endemic cancer.
Collapse
|
14
|
Gao X, Cao Z. Gingiva-derived Mesenchymal Stem Cells and Their Potential Applications in Oral and Maxillofacial Diseases. Curr Stem Cell Res Ther 2020; 15:43-53. [PMID: 31702517 DOI: 10.2174/1574888x14666191107100311] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/02/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Stem cells are undifferentiated cells with multilineage differentiation potential. They can be collected from bone marrow, fat, amniotic fluid, and teeth. Stem cell-based therapies have been widely used to treat multiple diseases, such as cardiac disease, and hematological disorders. The cells may also be beneficial for controlling the disease course and promoting tissue regeneration in oral and maxillofacial diseases. Oral-derived gingival mesenchymal stem cells are easy to access and the donor sites heal rapidly without a scar. Such characteristics demonstrate the beneficial role of GMSCs in oral and maxillofacial diseases. OBJECTIVE We summarize the features of GMSCs, including their self-renewal, multipotent differentiation, immunomodulation, and anti-inflammation properties. We also discuss their applications in oral and maxillofacial disease treatment and tissue regeneration. CONCLUSION GMSCs are easily harvestable adult stem cells with outstanding proliferation, differentiation, and immunomodulation characteristics. A growing body of evidence indicates that GMSCs have strong potential use in accelerating wound healing and promoting the regeneration of bone defects, periodontium, oral neoplasms, salivary glands, peri-implantitis, and nerves. Moreover, alginate, polylactic acid and polycaprolactone can be used as biodegradable scaffolds for GMSC encapsulation. Various growth factors can be applied to the corresponding scaffolds to obtain the desired GMSC differentiation and phenotypes. Three-dimensional spheroid culture systems could optimize GMSC properties and improve the performance of the cells in tissue engineering. The immunomodulatory property of GMSCs in controlling oral and maxillofacial inflammation needs further research.
Collapse
Affiliation(s)
- Xudong Gao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory for Oral Biomedical Engineering of Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengguo Cao
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Li W, Wang Q, Qi X, Guo Y, Lu H, Chen Y, Lu Z, Yan Q, Zhu X, Jung JU, Tosato G, Gao SJ, Lu C. Viral interleukin-6 encoded by an oncogenic virus promotes angiogenesis and cellular transformation by enhancing STAT3-mediated epigenetic silencing of caveolin 1. Oncogene 2020; 39:4603-4618. [PMID: 32393833 PMCID: PMC7970339 DOI: 10.1038/s41388-020-1317-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 04/20/2020] [Accepted: 04/24/2020] [Indexed: 12/27/2022]
Abstract
Kaposi's sarcoma (KS) caused by oncogenic Kaposi's sarcoma-associated herpesvirus (KSHV) is a highly angiogenic and invasive vascular tumor and the most common AIDS-associated cancer. KSHV-encoded viral interleukin-6 (vIL-6) is implicated in the development of KSHV-induced malignancies; however, the mechanisms underlying vIL-6-induced angiogenesis and tumorigenesis remain undefined. Here, we show that vIL-6 promotes angiogenesis, cell proliferation, and invasion by downregulating caveolin 1 (CAV1) that plays a pivotal and versatile role in multiple cancer-associated processes. Mechanistically, vIL-6 signaling led to the phosphorylation and acetylation of STAT3 that targeted DNA methyltransferase 1 (DNMT1) in a sequential manner. Specifically, the vIL-6-induced phosphorylated form of STAT3 transcriptionally activated DNMT1 expression. Furthermore, vIL-6-induced acetylated form of STAT3 interacted with DNMT1 to form a transcription factor complex that bound to and methylated the CAV1 promoter, leading to CAV1 expression silencing. In fact, downregulation of CAV1 expression resulted in the activation of AKT signaling, promoting cell invasion, and growth transformation induced by KSHV. Finally, genetic deletion of vIL-6 from the KSHV genome abolished KSHV-induced cellular transformation and impaired angiogenesis. Our results reveal that vIL-6 epigenetically silences CAV1 expression to promote angiogenesis and tumorigenesis by regulating the formation of STAT3-DNMT1 complex. These novel findings define a mechanism by which KSHV inhibits the CAV1 pathway and establish the scientific basis for targeting this pathway to treat KSHV-associated cancers.
Collapse
Affiliation(s)
- Wan Li
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, PR China
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Hospital, Nanjing Medical University, Nanjing, 210029, PR China
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, PR China
| | - Qingxia Wang
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, PR China
| | - Xiaoyu Qi
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, PR China
| | - Yuanyuan Guo
- The College of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Hongmei Lu
- Department of Obstetrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210036, PR China
| | - Yuheng Chen
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, PR China
| | - Zhongmou Lu
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, PR China
| | - Qin Yan
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, PR China
| | - Xiaofei Zhu
- Department of Laboratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China.
| | - Jae U Jung
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Giovanna Tosato
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892-1906, USA
| | - Shou-Jiang Gao
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, 15232, USA
| | - Chun Lu
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, PR China.
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Hospital, Nanjing Medical University, Nanjing, 210029, PR China.
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, PR China.
| |
Collapse
|
16
|
Eason AB, Sin SH, Shah M, Yuan H, Phillips DJ, Droste M, Shamshiev A, Dittmer DP. DLX1008 (brolucizumab), a single-chain anti-VEGF-A antibody fragment with low picomolar affinity, leads to tumor involution in an in vivo model of Kaposi Sarcoma. PLoS One 2020; 15:e0233116. [PMID: 32407363 PMCID: PMC7224538 DOI: 10.1371/journal.pone.0233116] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
Kaposi Sarcoma (KS) is among the most angiogenic cancers in humans and an AIDS-defining condition. KS-associated herpesvirus (KSHV) is necessary for KS development, as is vascular endothelial growth factor (VEGF-A). DLX1008 is a novel anti-VEGF-A antibody single-chain variable fragment (scFv) with low picomolar affinity for VEGF-A. In vivo imaging techniques were used to establish the efficacy of DLX1008 and to establish the mechanism of action; this included non-invasive imaging by ultrasound and optical fluorescence, verified by post-mortem histochemistry. The results showed that DLX1008 was efficacious in a KS mouse model. The NSG mouse xenografts suffered massive internal necrosis or involution, consistent with a lack of blood supply. We found that imaging by ultrasound was superior to external caliper measurements in the validation of the angiogenesis inhibitor DLX1008. Further development of DLX1008 against VEGF-dependent sarcomas is warranted.
Collapse
Affiliation(s)
- Anthony B. Eason
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Sang-Hoon Sin
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Mohsin Shah
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Hong Yuan
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | | | | | | | - Dirk P. Dittmer
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
17
|
PDGFRA defines the mesenchymal stem cell Kaposi's sarcoma progenitors by enabling KSHV oncogenesis in an angiogenic environment. PLoS Pathog 2019; 15:e1008221. [PMID: 31881074 PMCID: PMC6980685 DOI: 10.1371/journal.ppat.1008221] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 01/24/2020] [Accepted: 11/15/2019] [Indexed: 11/19/2022] Open
Abstract
Kaposi's sarcoma (KS) is an AIDS-defining cancer caused by the KS-associated herpesvirus (KSHV). Unanswered questions regarding KS are its cellular ontology and the conditions conducive to viral oncogenesis. We identify PDGFRA(+)/SCA-1(+) bone marrow-derived mesenchymal stem cells (Pα(+)S MSCs) as KS spindle-cell progenitors and found that pro-angiogenic environmental conditions typical of KS are critical for KSHV sarcomagenesis. This is because growth in KS-like conditions generates a de-repressed KSHV epigenome allowing oncogenic KSHV gene expression in infected Pα(+)S MSCs. Furthermore, these growth conditions allow KSHV-infected Pα(+)S MSCs to overcome KSHV-driven oncogene-induced senescence and cell cycle arrest via a PDGFRA-signaling mechanism; thus identifying PDGFRA not only as a phenotypic determinant for KS-progenitors but also as a critical enabler for viral oncogenesis.
Collapse
|
18
|
He M, Cheng F, da Silva SR, Tan B, Sorel O, Gruffaz M, Li T, Gao SJ. Molecular Biology of KSHV in Relation to HIV/AIDS-Associated Oncogenesis. Cancer Treat Res 2019; 177:23-62. [PMID: 30523620 DOI: 10.1007/978-3-030-03502-0_2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Discovered in 1994, Kaposi's sarcoma-associated herpesvirus (KSHV) has been associated with four human malignancies including Kaposi's sarcoma, primary effusion lymphoma, a subset of multicentric Castleman's disease, and KSHV inflammatory cytokine syndrome. These malignancies mostly occur in immunocompromised patients including patients with acquired immunodeficiency syndrome and often cause significant mortality because of the lack of effective therapies. Significant progresses have been made to understand the molecular basis of KSHV infection and KSHV-induced oncogenesis in the last two decades. This chapter provides an update on the recent advancements focusing on the molecular events of KSHV primary infection, the mechanisms regulating KSHV life cycle, innate and adaptive immunity, mechanism of KSHV-induced tumorigenesis and inflammation, and metabolic reprogramming in KSHV infection and KSHV-transformed cells.
Collapse
Affiliation(s)
- Meilan He
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Fan Cheng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Suzane Ramos da Silva
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Brandon Tan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Océane Sorel
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Marion Gruffaz
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Tingting Li
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA.
| |
Collapse
|
19
|
Li H, Zhong C, Wang Q, Chen W, Yuan Y. Curcumin is an APE1 redox inhibitor and exhibits an antiviral activity against KSHV replication and pathogenesis. Antiviral Res 2019; 167:98-103. [PMID: 31034848 DOI: 10.1016/j.antiviral.2019.04.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 01/06/2023]
Abstract
Curcumin, a polyphenol, is the main bioactive compound in dietary spice turmeric curcuma longa. It possesses anti-inflammatory, anti-oxidant and anti-neoplastic properties and shows potentials in treating or preventing particular diseases such as oxidative and inflammatory conditions, metabolic syndrome, arthritis, anxiety, hyperlipidemia and cancers. The diverse range and potential health beneficial effects has generated enthusiasm leading to intensive investigation into the phytochemical. However, a concern has been also raised if curcumin has a promiscuous bioassay profile and is a Pan-Assay INterference compound (PAINS). Here we present evidence indicating that curcumin is not a PAINS, but an inhibitor to APE1 redox function that affects many genes and pathways. This discovery explains the wide range of effects of curcumin on diverse human diseases and predicts a potential application in treatment of viral infection and virus-associated cancer. As a proof-of-concept, we demonstrated that curcumin is able to efficiently block Kaposi's sarcoma-associated herpesvirus replication and inhibit the pathogenic processes of angiogenesis and cell invasion.
Collapse
Affiliation(s)
- He Li
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, 19104, USA; Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, China
| | - Canrong Zhong
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qian Wang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Weikang Chen
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yan Yuan
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, 19104, USA; Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
20
|
Yan L, Majerciak V, Zheng ZM, Lan K. Towards Better Understanding of KSHV Life Cycle: from Transcription and Posttranscriptional Regulations to Pathogenesis. Virol Sin 2019; 34:135-161. [PMID: 31025296 PMCID: PMC6513836 DOI: 10.1007/s12250-019-00114-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/14/2019] [Indexed: 02/08/2023] Open
Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus-8 (HHV-8), is etiologically linked to the development of Kaposi’s sarcoma, primary effusion lymphoma, and multicentric Castleman’s disease. These malignancies often occur in immunosuppressed individuals, making KSHV infection-associated diseases an increasing global health concern with persistence of the AIDS epidemic. KSHV exhibits biphasic life cycles between latent and lytic infection and extensive transcriptional and posttranscriptional regulation of gene expression. As a member of the herpesvirus family, KSHV has evolved many strategies to evade the host immune response, which help the virus establish a successful lifelong infection. In this review, we summarize the current research status on the biology of latent and lytic viral infection, the regulation of viral life cycles and the related pathogenesis.
Collapse
Affiliation(s)
- Lijun Yan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Vladimir Majerciak
- National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Zhi-Ming Zheng
- National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA.
| | - Ke Lan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
21
|
Jeon H, Lee J, Lee S, Kang SK, Park SJ, Yoo SM, Lee MS. Extracellular Vesicles From KSHV-Infected Cells Stimulate Antiviral Immune Response Through Mitochondrial DNA. Front Immunol 2019; 10:876. [PMID: 31068945 PMCID: PMC6491682 DOI: 10.3389/fimmu.2019.00876] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/05/2019] [Indexed: 12/24/2022] Open
Abstract
Kaposi's Sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma, which is the most common cancer in acquired immune deficiency syndrome patients. KSHV contains a variety of immunoregulatory proteins. There have been many studies on the modulation of antiviral response by these immunoregulatory proteins of KSHV. However, the antiviral effects of extracellular vesicles (EVs) during de novo KSHV infection have not been investigated to our best knowledge. In this study, we showed that KSHV-infected cells induce interferon-stimulated genes (ISGs) response but not type I interferon in uninfected bystander cells using EVs. mRNA microarray analysis showed that ISGs and IRF-activating genes were prominently activated in EVs from KSHV-infected cells (KSHV EVs)-treated human endothelial cells, which were validated by RT-qPCR and western blot analysis. We also found that this response was not associated with cell death or apoptosis by virus infection. Mechanistically, the cGAS-STING pathway was linked with these KSHV EVs-mediated ISGs expressions, and mitochondrial DNA on the surface of KSHV EVs was one of the causative factors. Besides, KSHV EVs-treated cells showed lower infectivity for KSHV and viral replication activity than mock EVs-treated cells. Our results indicate that EVs from KSHV-infected cells could be an initiating factor for the innate immune response against viral infection, which may be critical to understanding the microenvironment of virus-infected cells.
Collapse
Affiliation(s)
- Hyungtaek Jeon
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Jisu Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Suhyuk Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Su-Kyung Kang
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Sang June Park
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Seung-Min Yoo
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Myung-Shin Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| |
Collapse
|
22
|
Deryabin P, Griukova A, Shatrova A, Petukhov A, Nikolsky N, Borodkina A. Optimization of lentiviral transduction parameters and its application for CRISPR-based secretome modification of human endometrial mesenchymal stem cells. Cell Cycle 2019; 18:742-758. [PMID: 30880567 PMCID: PMC6464586 DOI: 10.1080/15384101.2019.1593650] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/08/2019] [Accepted: 02/19/2019] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) hold a great promise for successful development of regenerative medicine. Among the plenty of uncovered MSCs sources, desquamated endometrium collected from the menstrual blood probably remains the most accessible. Though numerous studies have been published on human endometrium-derived mesenchymal stem cells (hMESCs) properties in the past years, there are only a few data regarding their genetic modulation. Moreover, there is a lack of information about the fate of the transduced hMESCs. The present study aimed to optimize hMESCs transduction parameters and apply Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 technology for genome and secretome modification. The fate of hMESCs transduced either in presence of polybrene (Pb) or protamine sulfate (Ps) was assessed by alterations in CD expression profile, growth rate, cell size, migration capability, osteogenic, adipogenic, and decidual differential potentials. Here, we postulated that the use of Ps for hMESCs genetic manipulations is preferable, as it has no impact on the stem-cell properties, whereas Pb application is undesirable, as it induces cellular senescence. Plasminogen activator inhibitor-1 was selected for further targeted hMESCs genome and secretome modification using CRISPR/Cas9 systems. The obtained data provide optimized transduction scheme for hMESCs and verification of its effectiveness by successful hMESCs genome editing via CRISPR/Cas9 technology.
Collapse
Affiliation(s)
- Pavel Deryabin
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - Anastasiia Griukova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - Alla Shatrova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - Alexey Petukhov
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - Nikolay Nikolsky
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | | |
Collapse
|
23
|
McNamara RP, Chugh PE, Bailey A, Costantini LM, Ma Z, Bigi R, Cheves A, Eason AB, Landis JT, Host KM, Xiong J, Griffith JD, Damania B, Dittmer DP. Extracellular vesicles from Kaposi Sarcoma-associated herpesvirus lymphoma induce long-term endothelial cell reprogramming. PLoS Pathog 2019; 15:e1007536. [PMID: 30716130 PMCID: PMC6361468 DOI: 10.1371/journal.ppat.1007536] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/17/2018] [Indexed: 01/08/2023] Open
Abstract
Extracellular signaling is a mechanism that higher eukaryotes have evolved to facilitate organismal homeostasis. Recent years have seen an emerging interest in the role of secreted microvesicles, termed extracellular vesicles (EV) or exosomes in this signaling network. EV contents can be modified by the cell in response to stimuli, allowing them to relay information to neighboring cells, influencing their physiology. Here we show that the tumor virus Kaposi's Sarcoma-associated herpesvirus (KSHV) hijacks this signaling pathway to induce cell proliferation, migration, and transcriptome reprogramming in cells not infected with the virus. KSHV-EV activates the canonical MEK/ERK pathway, while not alerting innate immune regulators, allowing the virus to exert these changes without cellular pathogen recognition. Collectively, we propose that KSHV establishes a niche favorable for viral spread and cell transformation through cell-derived vesicles, all while avoiding detection.
Collapse
Affiliation(s)
- Ryan P McNamara
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Pauline E Chugh
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Q2 Solutions-EA Genomics, Morrisville, North Carolina
| | - Aubrey Bailey
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Lindsey M Costantini
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, North Carolina
| | - Zhe Ma
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Rachele Bigi
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Avery Cheves
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Anthony B Eason
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Justin T Landis
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kurtis M Host
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jie Xiong
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jack D Griffith
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Blossom Damania
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Dirk P Dittmer
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
24
|
Abstract
RNA modifications have generated much interest in the virology field, as recent works have shown that many viruses harbor these marks and modify cellular marks. The most abundant mRNA modification in eukaryotic cells, N6-methyladenosine (m6A), has been examined extensively at the genome-wide scale in both cellular and viral contexts. This Gem discusses the role of m6A in gene regulation and describes recent advancements in Kaposi's sarcoma-associated herpesvirus (KSHV) and simian virus 40 (SV40) research. We provide insights into future research related to m6A in DNA viruses.
Collapse
Affiliation(s)
- Brandon Tan
- Department of Systems Biology, City of Hope, Monrovia, California, USA
| | - Shou-Jiang Gao
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| |
Collapse
|
25
|
Tan B, Gao SJ. RNA epitranscriptomics: Regulation of infection of RNA and DNA viruses by N 6 -methyladenosine (m 6 A). Rev Med Virol 2018; 28:e1983. [PMID: 29698584 PMCID: PMC6339815 DOI: 10.1002/rmv.1983] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/24/2018] [Accepted: 03/29/2018] [Indexed: 02/05/2023]
Abstract
N6 -methyladenosine (m6 A) was discovered 4 decades ago. However, the functions of m6 A and the cellular machinery that regulates its changes have just been revealed in the last few years. m6 A is an abundant internal mRNA modification on cellular RNA and is implicated in diverse cellular functions. Recent works have demonstrated the presence of m6 A in the genomes of RNA viruses and transcripts of a DNA virus with either a proviral or antiviral role. Here, we first summarize what is known about the m6 A "writers," "erasers," "readers," and "antireaders" as well as the role of m6 A in mRNA metabolism. We then review how the replications of numerous viruses are enhanced and restricted by m6 A with emphasis on the oncogenic DNA virus, Kaposi sarcoma-associated herpesvirus (KSHV), whose m6 A epitranscriptome was recently mapped. In the context of KSHV, m6 A and the reader protein YTHDF2 acts as an antiviral mechanism during viral lytic replication. During viral latency, KSHV alters m6 A on genes that are implicated in cellular transformation and viral latency. Lastly, we discuss future studies that are important to further delineate the functions of m6 A in KSHV latent and lytic replication and KSHV-induced oncogenesis.
Collapse
Affiliation(s)
- Brandon Tan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
- Department of Microbiology, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
26
|
K15 Protein of Kaposi's Sarcoma Herpesviruses Increases Endothelial Cell Proliferation and Migration through Store-Operated Calcium Entry. Viruses 2018; 10:v10060282. [PMID: 29795033 PMCID: PMC6024707 DOI: 10.3390/v10060282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 05/06/2018] [Accepted: 05/09/2018] [Indexed: 01/11/2023] Open
Abstract
Kaposi's sarcoma (KS) is a tumor of the vascular endothelium that is caused by Kaposi's sarcoma-associated herpesvirus (KSHV). K15 of KSHV is a specific gene encoding a transmembrane protein. Two highly different forms of K15, the predominant (K15P) and minor (K15M) have been identified in different KSHV strains. In genomic locations and protein topology, two K15 alleles resemble the latent membrane protein (LMP) 1 and LMP2A of Epstein⁻Barr virus. Both K15 proteins have motifs similar to those found in LMP1 and LMP2A. K15 therefore seems to be a hybrid of a distant evolutionary relative of LMP1 and LMP2A. Ca2+ is a second messenger and participates in numerous activities in cells, like proliferation, migration and metastasis. It has been found previously that LMP1 increased Ca2+ influx through store-operated calcium channels and blockade of LMP1 reduced store-operated Ca2+ entry (SOCE). LMP2A has similar activity. So we sought to determine whether K15 had similar activity. We showed that K15P induced Ca2+ influx and enhanced expression of Orail1, which is a vital protein in SOCE, and overexpression of K15P improved cell motility. Mutant K15P did not show these activities in HEK-293T and EA.hy 926 cells. Our results showed that K15P increased cell proliferation and migration though SOCE and established a novel mechanism for the development of KS and KSHV-associated diseases.
Collapse
|
27
|
Tan B, Liu H, Zhang S, da Silva SR, Zhang L, Meng J, Cui X, Yuan H, Sorel O, Zhang SW, Huang Y, Gao SJ. Viral and cellular N 6-methyladenosine and N 6,2'-O-dimethyladenosine epitranscriptomes in the KSHV life cycle. Nat Microbiol 2018; 3:108-120. [PMID: 29109479 PMCID: PMC6138870 DOI: 10.1038/s41564-017-0056-8] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 10/09/2017] [Indexed: 02/05/2023]
Abstract
N6-methyladenosine (m6A) and N6,2'-O-dimethyladenosine (m6Am) modifications (m6A/m) of messenger RNA mediate diverse cellular functions. Oncogenic Kaposi's sarcoma-associated herpesvirus (KSHV) has latent and lytic replication phases that are essential for the development of KSHV-associated cancers. To date, the role of m6A/m in KSHV replication and tumorigenesis is unclear. Here, we provide mechanistic insights by examining the viral and cellular m6A/m epitranscriptomes during KSHV latent and lytic infection. KSHV transcripts contain abundant m6A/m modifications during latent and lytic replication, and these modifications are highly conserved among different cell types and infection systems. Knockdown of YTHDF2 enhanced lytic replication by impeding KSHV RNA degradation. YTHDF2 binds to viral transcripts and differentially mediates their stability. KSHV latent infection induces 5' untranslated region (UTR) hypomethylation and 3'UTR hypermethylation of the cellular epitranscriptome, regulating oncogenic and epithelial-mesenchymal transition pathways. KSHV lytic replication induces dynamic reprogramming of epitranscriptome, regulating pathways that control lytic replication. These results reveal a critical role of m6A/m modifications in KSHV lifecycle and provide rich resources for future investigations.
Collapse
Affiliation(s)
- Brandon Tan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hui Liu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- School of Information and Electrical Engineering, China University of Mining and Technology, Xuzhou, Jiangsu, China
| | - Songyao Zhang
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, TX, USA
- School of Automation, Northwestern Polytechnic University, Xi'an, Shaanxi, China
| | - Suzane Ramos da Silva
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lin Zhang
- School of Information and Electrical Engineering, China University of Mining and Technology, Xuzhou, Jiangsu, China
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| | - Jia Meng
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, China
| | - Xiaodong Cui
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| | - Hongfeng Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Océane Sorel
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shao-Wu Zhang
- School of Automation, Northwestern Polytechnic University, Xi'an, Shaanxi, China
| | - Yufei Huang
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, TX, USA.
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
28
|
Li Y, Zhong C, Liu D, Yu W, Chen W, Wang Y, Shi S, Yuan Y. Evidence for Kaposi Sarcoma Originating from Mesenchymal Stem Cell through KSHV-induced Mesenchymal-to-Endothelial Transition. Cancer Res 2017; 78:230-245. [PMID: 29066510 DOI: 10.1158/0008-5472.can-17-1961] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/13/2017] [Accepted: 10/18/2017] [Indexed: 12/22/2022]
Abstract
The major transmission route for Kaposi sarcoma-associated herpesvirus (KSHV) infection is the oral cavity through saliva. Kaposi sarcoma (KS) frequently occurs in the oral cavity in HIV-positive individuals and is often the first presenting sign of AIDS. However, the oral target cells for KSHV infection and the cellular origin of Kaposi sarcoma remain unknown. Here we present clinical and experimental evidences that Kaposi sarcoma spindle cells may originate from virally modified oral mesenchymal stem cells (MSC). AIDS-KS spindle cells expressed neuroectodermal stem cell marker (Nestin) and oral MSC marker CD29, suggesting an oral/craniofacial MSC lineage of AIDS-associated Kaposi sarcoma. Furthermore, oral MSCs were highly susceptible to KSHV infection, and infection promoted multilineage differentiation and mesenchymal-to-endothelial transition (MEndT). KSHV infection of oral MSCs resulted in expression of a large number of cytokines, a characteristic of Kaposi sarcoma, and upregulation of Kaposi sarcoma signature and MEndT-associated genes. These results suggest that Kaposi sarcoma may originate from pluripotent MSC and KSHV infection transforms MSC to Kaposi sarcoma-like cells through MEndT.Significance: These findings indicate that Kaposi sarcomas, which arise frequently in AIDS patients, originate from neural crest-derived mesenchymal stem cells, with possible implications for improving the clnical treatment of this malignancy. Cancer Res; 78(1); 230-45. ©2017 AACR.
Collapse
Affiliation(s)
- Yuqing Li
- Institute of Human Virology and Ministry of Education Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Canrong Zhong
- Institute of Human Virology and Ministry of Education Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dawei Liu
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenjing Yu
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
| | - Weikang Chen
- Institute of Human Virology and Ministry of Education Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Wang
- Institute of Human Virology and Ministry of Education Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Songtao Shi
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
| | - Yan Yuan
- Institute of Human Virology and Ministry of Education Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China. .,Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Zhong C, Xu M, Wang Y, Xu J, Yuan Y. An APE1 inhibitor reveals critical roles of the redox function of APE1 in KSHV replication and pathogenic phenotypes. PLoS Pathog 2017; 13:e1006289. [PMID: 28380040 PMCID: PMC5381946 DOI: 10.1371/journal.ppat.1006289] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 03/11/2017] [Indexed: 01/04/2023] Open
Abstract
APE1 is a multifunctional protein with a DNA base excision repair function in its C-terminal domain and a redox activity in its N-terminal domain. The redox function of APE1 converts certain transcription factors from inactive oxidized to active reduced forms. Given that among the APE1-regulated transcription factors many are critical for KSHV replication and pathogenesis, we investigated whether inhibition of APE1 redox function blocks KSHV replication and Kaposi’s sarcoma (KS) phenotypes. With an shRNA-mediated silencing approach and a known APE-1 redox inhibitor, we demonstrated that APE1 redox function is indeed required for KSHV replication as well as KSHV-induced angiogenesis, validating APE1 as a therapeutic target for KSHV-associated diseases. A ligand-based virtual screening yielded a small molecular compound, C10, which is proven to bind to APE1. C10 exhibits low cytotoxicity but efficiently inhibits KSHV lytic replication (EC50 of 0.16 μM and selective index of 165) and KSHV-mediated pathogenic phenotypes including cytokine production, angiogenesis and cell invasion, demonstrating its potential to become an effective drug for treatment of KS. As a major AIDS-associated malignancy, Kaposi’s sarcoma (KS) is caused by Kaposi’s sarcoma-associated herpesvirus (KSHV). Currently there is no definitive cure for KS. In this study, we identified a cellular protein, namely APE1, as an effective therapeutic target for blocking KSHV replication and inhibiting the development of KS phenotypes. We showed that the redox function of APE1 is absolutely required for KSHV replication, virally induced cytokine secretion and angiogenesis. Blockade of APE1 expression or inhibition of APE1 redox activity led to inhibition of KSHV replication and reduction of cytokine release and angiogenesis. Furthermore, we identified a novel small molecular compound, C10, which exhibited specific inhibitory activity on APE1 redox function and was demonstrated to efficiently inhibit KSHV replication and paracrine-mediated KS phenotypes such as angiogenesis and cell invasion. As a potent inhibitor of APE1 redox, C10 not only has value in development of a novel therapeutics for KS, but also may be used in therapies for other human diseases such as leukemia, pancreatic cancer and macular degeneration.
Collapse
Affiliation(s)
- Canrong Zhong
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Mengyang Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yan Wang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jun Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
- * E-mail: (YY); (JX)
| | - Yan Yuan
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail: (YY); (JX)
| |
Collapse
|
30
|
Qin J, Li W, Gao SJ, Lu C. KSHV microRNAs: Tricks of the Devil. Trends Microbiol 2017; 25:648-661. [PMID: 28259385 DOI: 10.1016/j.tim.2017.02.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 01/23/2017] [Accepted: 02/06/2017] [Indexed: 01/02/2023]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma (KS), a vascular tumor frequently found in immunodeficient individuals. KSHV encodes 12 pre-microRNAs (pre-miRNAs), which are processed into 25 mature microRNAs (miRNAs). KSHV miRNAs maintain KSHV latency, enhance angiogenesis and dissemination of the infected cells, and interfere with the host immune system by regulating viral and cellular gene expression, ultimately contributing to KS development. In this review, we briefly introduce the biogenesis of miRNAs and then describe the recent advances in defining the roles and mechanisms of action of KSHV miRNAs in KS development.
Collapse
Affiliation(s)
- Jie Qin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, P.R. China; Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, P.R. China; Department of Microbiology, Nanjing Medical University, Nanjing 211166, P.R. China
| | - Wan Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, P.R. China; Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, P.R. China; Department of Microbiology, Nanjing Medical University, Nanjing 211166, P.R. China
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Chun Lu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, P.R. China.
| |
Collapse
|
31
|
Lee S, Jang J, Jeon H, Lee J, Yoo SM, Park J, Lee MS. Latent Kaposi's sarcoma-associated herpesvirus infection in bladder cancer cells promotes drug resistance by reducing reactive oxygen species. J Microbiol 2016; 54:782-788. [PMID: 27796928 DOI: 10.1007/s12275-016-6388-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 09/19/2016] [Accepted: 09/19/2016] [Indexed: 02/05/2023]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the major etiologic agent of Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. Recent studies have indicated that KSHV can be detected at high frequency in patient-derived bladder cancer tissue and might be associated with the pathogenesis of bladder cancer. Bladder cancer is the second most common cancer of the genitourinary tract, and it has a high rate of recurrence. Because drug resistance is closely related to chemotherapy failure and cancer recurrence, we investigated whether KSHV infection is associated with drug resistance of bladder cancer cells. Some KSHV-infected bladder cancer cell lines showed resistance to an anti-cancer drug, cisplatin, possibly as a result of down-regulation of reactive oxygen species. Additionally, drug resistance acquired from KSHV infection could partly be overcome by HDAC1 inhibitors. Taken together, the data suggest the possible role of KSHV in chemo-resistant bladder cancer, and indicate the therapeutic potential of HDAC1 inhibitors in drug-resistant bladder cancers associated with KSHV infection.
Collapse
Affiliation(s)
- Suhyuk Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, 34824, Republic of Korea
| | - Jaehyuk Jang
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, 34824, Republic of Korea
| | - Hyungtaek Jeon
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, 34824, Republic of Korea
| | - Jisu Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, 34824, Republic of Korea
| | - Seung-Min Yoo
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, 34824, Republic of Korea
| | - Jinsung Park
- Department of Urology, Eulji University Hospital, Eulji University School of Medicine, Daejeon, 34824, Republic of Korea
| | - Myung-Shin Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, 34824, Republic of Korea.
| |
Collapse
|
32
|
Dittmer DP, Damania B. Kaposi sarcoma-associated herpesvirus: immunobiology, oncogenesis, and therapy. J Clin Invest 2016; 126:3165-75. [PMID: 27584730 DOI: 10.1172/jci84418] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Kaposi sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8, is the etiologic agent underlying Kaposi sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. This human gammaherpesvirus was discovered in 1994 by Drs. Yuan Chang and Patrick Moore. Today, there are over five thousand publications on KSHV and its associated malignancies. In this article, we review recent and ongoing developments in the KSHV field, including molecular mechanisms of KSHV pathogenesis, clinical aspects of KSHV-associated diseases, and current treatments for cancers associated with this virus.
Collapse
|
33
|
Song L, Zhou X, Jia HJ, Du M, Zhang JL, Li L. Effect of hGC-MSCs from human gastric cancer tissue on cell proliferation, invasion and epithelial-mesenchymal transition in tumor tissue of gastric cancer tumor-bearing mice. ASIAN PAC J TROP MED 2016; 9:796-800. [PMID: 27569891 DOI: 10.1016/j.apjtm.2016.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 06/16/2016] [Accepted: 06/21/2016] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE To study the effect of hGC-MSCs from human gastric cancer tissue on cell proliferation, invasion and epithelial-mesenchymal transition in tumor tissue of gastric cancer tumor-bearing mice. METHODS BABL/c nude mice were selected as experimental animals and gastric cancer tumor-bearing mice model were established by subcutaneous injection of gastric cancer cells, randomly divided into different intervention groups. hGC-MSCs group were given different amounts of gastric cancer cells for subcutaneous injection, PBS group was given equal volume of PBS for subcutaneous injection. Then tumor tissue volume were determined, tumor-bearing mice were killed and tumor tissues were collected, mRNA expression of proliferation, invasion, EMT-related molecules were determined. RESULTS 4, 8, 12, 16, 20 d after intervention, tumor tissue volume of hGC-MSCs group were significantly higher than those of PBS group and the more the number of hGC-MSCs, the higher the tumor tissue volume; mRNA contents of Ki-67, PCNA, Bcl-2, MMP-2, MMP-7, MMP-9, MMP-14, N-cadherin, vimentin, Snail and Twist in tumor tissue of hGC-MSCs group were higher than those of PBS group, and mRNA contents of Bax, TIMP1, TIMP2 and E-cadherin were lower than those of PBS group. CONCLUSION hGC-MSCs from human gastric cancer tissue can promote the tumor growth in gastric cancer tumor-bearing mice, and the molecular mechanism includes promoting cell proliferation, invasion and epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Lin Song
- Oncology Department No. 2, Linyi People's Hospital of Shandong Province, Linyi City, Shandong Province, 276000, China
| | - Xin Zhou
- Oncology Department No. 2, Linyi People's Hospital of Shandong Province, Linyi City, Shandong Province, 276000, China
| | - Hong-Jun Jia
- Radiotherapy Technology Department, Linyi People's Hospital of Shandong Province, Linyi City, Shandong Province, 276000, China.
| | - Mei Du
- Oncology Department No. 2, Linyi People's Hospital of Shandong Province, Linyi City, Shandong Province, 276000, China
| | - Jin-Ling Zhang
- Oncology Department No. 2, Linyi People's Hospital of Shandong Province, Linyi City, Shandong Province, 276000, China
| | - Liang Li
- Oncology Department No. 2, Linyi People's Hospital of Shandong Province, Linyi City, Shandong Province, 276000, China
| |
Collapse
|
34
|
An Oncogenic Virus Promotes Cell Survival and Cellular Transformation by Suppressing Glycolysis. PLoS Pathog 2016; 12:e1005648. [PMID: 27187079 PMCID: PMC4871371 DOI: 10.1371/journal.ppat.1005648] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/29/2016] [Indexed: 11/19/2022] Open
Abstract
Aerobic glycolysis is essential for supporting the fast growth of a variety of cancers. However, its role in the survival of cancer cells under stress conditions is unclear. We have previously reported an efficient model of gammaherpesvirus Kaposi’s sarcoma-associated herpesvirus (KSHV)-induced cellular transformation of rat primary mesenchymal stem cells. KSHV-transformed cells efficiently induce tumors in nude mice with pathological features reminiscent of Kaposi’s sarcoma tumors. Here, we report that KSHV promotes cell survival and cellular transformation by suppressing aerobic glycolysis and oxidative phosphorylation under nutrient stress. Specifically, KSHV microRNAs and vFLIP suppress glycolysis by activating the NF-κB pathway to downregulate glucose transporters GLUT1 and GLUT3. While overexpression of the transporters rescues the glycolytic activity, it induces apoptosis and reduces colony formation efficiency in softagar under glucose deprivation. Mechanistically, GLUT1 and GLUT3 inhibit constitutive activation of the AKT and NF-κB pro-survival pathways. Strikingly, GLUT1 and GLUT3 are significantly downregulated in KSHV-infected cells in human KS tumors. Furthermore, we have detected reduced levels of aerobic glycolysis in several KSHV-infected primary effusion lymphoma cell lines compared to a Burkitt’s lymphoma cell line BJAB, and KSHV infection of BJAB cells reduced aerobic glycolysis. These results reveal a novel mechanism by which an oncogenic virus regulates a key metabolic pathway to adapt to stress in tumor microenvironment, and illustrate the importance of fine-tuning the metabolic pathways for sustaining the proliferation and survival of cancer cells, particularly under stress conditions. KSHV is causally associated with the development of Kaposi’s sarcoma and primary effusion lymphoma; however, the mechanism underlying KSHV-induced malignant transformation remains unclear. The recent development of an efficient KSHV-induced cellular transformation model of primary rat mesenchymal stem cells should facilitate the delineation of KSHV-induced oncogenesis. In this report, we have used this model to investigate the metabolic pathways mediating the proliferation and survival of KSHV-transformed cells. In contrast to most other cancers that depend on aerobic glycolysis for their fast growth, we demonstrate that KSHV suppresses aerobic glycolysis and oxidative phosphorylation in the transformed cells. Significantly, suppression of aerobic glycolysis enhances the survival of the KSHV-transformed cells under nutrient deprivation. Mechanistically, KSHV-encoded microRNAs and vFLIP suppress aerobic glycolysis by activating the NF-κB pathway to downregulate glucose transporters GLUT1 and GLUT3. We have further shown that GLUT1 and GLUT3 inhibit constitutive activation of the AKT and NF-κB pro-survival pathways. Strikingly, GLUT1 and GLUT3 are significantly downregulated in KSHV-infected cells in human KS tumors. Furthermore, we have detected reduced levels of aerobic glycolysis in several KSHV-infected primary effusion lymphoma cell lines and a KSHV-infected Burkitt’s lymphoma cell line BJAB. Our results reveal a novel mechanism by which an oncogenic virus regulates a key metabolic pathway to adapt to stress in tumor microenvironment, and illustrate the importance of fine-tuning the metabolic pathways for sustaining the proliferation and survival of cancer cells, particularly under nutrient stress microenvironment.
Collapse
|
35
|
A Toolbox for Herpesvirus miRNA Research: Construction of a Complete Set of KSHV miRNA Deletion Mutants. Viruses 2016; 8:v8020054. [PMID: 26907327 PMCID: PMC4776209 DOI: 10.3390/v8020054] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/04/2016] [Accepted: 02/14/2016] [Indexed: 12/11/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) encodes 12 viral microRNAs (miRNAs) that are expressed during latency. Research into KSHV miRNA function has suffered from a lack of genetic systems to study viral miRNA mutations in the context of the viral genome. We used the Escherichia coli Red recombination system together with a new bacmid background, BAC16, to create mutants for all known KSHV miRNAs. The specific miRNA deletions or mutations and the integrity of the bacmids have been strictly quality controlled using PCR, restriction digestion, and sequencing. In addition, stable viral producer cell lines based on iSLK cells have been created for wildtype KSHV, for 12 individual miRNA knock-out mutants (ΔmiR-K12-1 through -12), and for mutants deleted for 10 of 12 (ΔmiR-cluster) or all 12 miRNAs (ΔmiR-all). NGS, in combination with SureSelect technology, was employed to sequence the entire latent genome within all producer cell lines. qPCR assays were used to verify the expression of the remaining viral miRNAs in a subset of mutants. Induction of the lytic cycle leads to efficient production of progeny viruses that have been used to infect endothelial cells. Wt BAC16 and miR mutant iSLK producer cell lines are now available to the research community.
Collapse
|