1
|
Abstract
For decades, scientists have relied on traditional animal models to study viral infection and the immune response. However, these models have limitations, and the search for more accurate and reliable ways to study the human-pathogen interphase has led to the development of humanized mouse systems. These revolutionary models have transformed how we understand viral infection and the human immune system's interactions with viruses to control or exacerbate disease. They are also paving the way for new treatments and therapies. In this article, we explore the history and development of humanized mouse systems and their advantages, limitations, and applications in viral immunology research. We describe the different types of humanized mouse models, including their generation and utility for studying human pathogens, with an emphasis on human-specific viruses. In addition, we discuss areas for further refinement and future applications.
Collapse
Affiliation(s)
- Angela Wahl
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA ; ,
| | - J Victor Garcia
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA ; ,
| |
Collapse
|
2
|
Jia Z, Zhang D, Zhu L, Xue J. Animal models of human herpesvirus infection. Animal Model Exp Med 2025; 8:615-628. [PMID: 39921263 PMCID: PMC12067922 DOI: 10.1002/ame2.12575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/18/2025] [Indexed: 02/10/2025] Open
Abstract
Human herpesvirus, a specific group within the herpesvirus family, is responsible for a variety of human diseases. These viruses can infect humans and other vertebrates, primarily targeting the skin, mucous membranes, and neural tissues, thereby significantly impacting the health of both humans and animals. Animal models are crucial for studying virus pathogenesis, vaccine development, and drug testing. Despite several vaccine candidates being in preclinical and clinical stages, no vaccines are current available to prevent lifelong infections caused by these human herpesviruses, except for varicella-zoster virus (VZV) vaccine. However, the strict host tropism of herpesviruses and other limitations mean that no single animal model can fully replicate all key features of human herpesvirus-associated diseases. This makes it challenging to evaluate vaccines and antivirals against human herpesvirus comprehensively. Herein, we summarize the current animal models used to study the human herpesviruses including α-herpesviruses (herpes simplex virus type 1(HSV-1), HSV-2, VZV), β-herpesviruses (human cytomegalovirus (HCMV), γ-herpesviruses (Epstein-Barr virus (EBV)) and Kaposi's sarcoma herpesvirus (KSHV)). By providing concise information and detailed analysis of the potential, limitations and applications of various models, such as non-human primates, mice, rabbits, guinea pigs, and tree shrews, this summary aims to help researchers efficiently select the most appropriate animal model, offering practical guidance for studying human herpesvirus.
Collapse
Affiliation(s)
- Ziqing Jia
- NHC Key Laboratory of Human Disease Comparative MedicineInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Dong Zhang
- NHC Key Laboratory of Human Disease Comparative MedicineInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lin Zhu
- NHC Key Laboratory of Human Disease Comparative MedicineInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jing Xue
- NHC Key Laboratory of Human Disease Comparative MedicineInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Respiratory Health and MultimorbidityInstitute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Ministry of EducationChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
3
|
Shifflett KW, Dittmer DP. Mouse models of Kaposi sarcoma-associated herpesvirus (KSHV). Virology 2025; 603:110384. [PMID: 39837218 PMCID: PMC11788063 DOI: 10.1016/j.virol.2024.110384] [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: 10/27/2024] [Revised: 12/18/2024] [Accepted: 12/29/2024] [Indexed: 01/23/2025]
Abstract
Infection with Kaposi sarcoma-associated herpesvirus (KSHV) is a prerequisite for the development of several human cancers, including Kaposi sarcoma and primary effusion lymphoma. Efficient long-term infection with KSHV and subsequent virally induced cell transformation is limited to humans, resulting in a lack of small animal models for KSHV-driven malignancies. Various attempts to create a mouse model for KSHV include infection of humanized mice, generating transgenic mice that ectopically express viral proteins, and grafting KSHV-infected tumor, primary, or immortalized cells onto immunodeficient mice. While no single mouse model can recapitulate the full range of KSHV-associated pathologies described in humans, each model adds an essential piece to the complete picture of KSHV infection and oncogenesis.
Collapse
Affiliation(s)
- Kyle W Shifflett
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, USA
| | - Dirk P Dittmer
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, USA.
| |
Collapse
|
4
|
Lee BC. Challenges and innovations in hematopoietic stem cell transplantation: exploring bone marrow niches and new model systems. BMB Rep 2024; 57:352-362. [PMID: 38919014 PMCID: PMC11362137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/27/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) remains an indispensable therapeutic strategy for various hematological diseases. This review discusses the pivotal role of bone marrow (BM) niches in influencing the efficacy of HSCT and evaluates the current animal models, emphasizing their limitations and the need for alternative models. Traditional animal models, mainly murine xenograft, have provided significant insights, but due to species-specific differences, are often constrained from accurately mimicking human physiological responses. These limitations highlight the importance of developing alternative models that can more realistically replicate human hematopoiesis. Emerging models that include BM organoids and BM-on-a-chip microfluidic systems promise enhanced understanding of HSCT dynamics. These models aim to provide more accurate simulations of the human BM microenvironment, potentially leading to improved preclinical assessments and therapeutic outcomes. This review highlights the complexities of the BM niche, discusses the limitations of current models, and suggests directions for future research using advanced model systems. [BMB Reports 2024; 57(8): 352-362].
Collapse
Affiliation(s)
- Byung-Chul Lee
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
- Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| |
Collapse
|
5
|
Muniraju M, Mutsvunguma LZ, Reidel IG, Escalante GM, Cua S, Musonda W, Calero-Landa J, Farelo MA, Rodriguez E, Li Z, Ogembo JG. Kaposi sarcoma-associated herpesvirus complement control protein (KCP) and glycoprotein K8.1 are not required for viral infection in vitro or in vivo. J Virol 2024; 98:e0057624. [PMID: 38767375 PMCID: PMC11237445 DOI: 10.1128/jvi.00576-24] [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: 04/02/2024] [Accepted: 04/21/2024] [Indexed: 05/22/2024] Open
Abstract
Kaposi sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus-8, is the causal agent of Kaposi sarcoma, a cancer that appears as tumors on the skin or mucosal surfaces, as well as primary effusion lymphoma and KSHV-associated multicentric Castleman disease, which are B-cell lymphoproliferative disorders. Effective prophylactic and therapeutic strategies against KSHV infection and its associated diseases are needed. To develop these strategies, it is crucial to identify and target viral glycoproteins involved in KSHV infection of host cells. Multiple KSHV glycoproteins expressed on the viral envelope are thought to play a pivotal role in viral infection, but the infection mechanisms involving these glycoproteins remain largely unknown. We investigated the role of two KSHV envelope glycoproteins, KSHV complement control protein (KCP) and K8.1, in viral infection in various cell types in vitro and in vivo. Using our newly generated anti-KCP antibodies, previously characterized anti-K8.1 antibodies, and recombinant mutant KSHV viruses lacking KCP, K8.1, or both, we demonstrated the presence of KCP and K8.1 on the surface of both virions and KSHV-infected cells. We showed that KSHV lacking KCP and/or K8.1 remained infectious in KSHV-susceptible cell lines, including epithelial, endothelial, and fibroblast, when compared to wild-type recombinant KSHV. We also provide the first evidence that KSHV lacking K8.1 or both KCP and K8.1 can infect human B cells in vivo in a humanized mouse model. Thus, these results suggest that neither KCP nor K8.1 is required for KSHV infection of various host cell types and that these glycoproteins do not determine KSHV cell tropism. IMPORTANCE Kaposi sarcoma-associated herpesvirus (KSHV) is an oncogenic human gamma-herpesvirus associated with the endothelial malignancy Kaposi sarcoma and the lymphoproliferative disorders primary effusion lymphoma and multicentric Castleman disease. Determining how KSHV glycoproteins such as complement control protein (KCP) and K8.1 contribute to the establishment, persistence, and transmission of viral infection will be key for developing effective anti-viral vaccines and therapies to prevent and treat KSHV infection and KSHV-associated diseases. Using newly generated anti-KCP antibodies, previously characterized anti-K8.1 antibodies, and recombinant mutant KSHV viruses lacking KCP and/or K8.1, we show that KCP and K8.1 can be found on the surface of both virions and KSHV-infected cells. Furthermore, we show that KSHV lacking KCP and/or K8.1 remains infectious to diverse cell types susceptible to KSHV in vitro and to human B cells in vivo in a humanized mouse model, thus providing evidence that these viral glycoproteins are not required for KSHV infection.
Collapse
Affiliation(s)
- Murali Muniraju
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Lorraine Z Mutsvunguma
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Ivana G Reidel
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Gabriela M Escalante
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Simeon Cua
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Webster Musonda
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Jonathan Calero-Landa
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
- Irell & Manella Graduate School of Biological Sciences of City of Hope, Duarte, California, USA
| | - Mafalda A Farelo
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Esther Rodriguez
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
- Irell & Manella Graduate School of Biological Sciences of City of Hope, Duarte, California, USA
| | - Zhou Li
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Javier Gordon Ogembo
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, California, USA
| |
Collapse
|
6
|
Caduff N, Rieble L, Böni M, McHugh D, Roshan R, Miley W, Labo N, Barman S, Trivett M, Bosma DMT, Rühl J, Goebels N, Whitby D, Münz C. KSHV infection of B cells primes protective T cell responses in humanized mice. Nat Commun 2024; 15:4841. [PMID: 38844783 PMCID: PMC11156630 DOI: 10.1038/s41467-024-49209-w] [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: 07/03/2023] [Accepted: 05/24/2024] [Indexed: 06/09/2024] Open
Abstract
Kaposi sarcoma associated herpesvirus (KSHV) is associated with around 1% of all human tumors, including the B cell malignancy primary effusion lymphoma (PEL), in which co-infection with the Epstein Barr virus (EBV) can almost always be found in malignant cells. Here, we demonstrate that KSHV/EBV co-infection of mice with reconstituted human immune systems (humanized mice) leads to IgM responses against both latent and lytic KSHV antigens, and expansion of central and effector memory CD4+ and CD8+ T cells. Among these, KSHV/EBV dual-infection allows for the priming of CD8+ T cells that are specific for the lytic KSHV antigen K6 and able to kill KSHV/EBV infected B cells. This suggests that K6 may represent a vaccine antigen for the control of KSHV and its associated pathologies in high seroprevalence regions, such as Sub-Saharan Africa.
Collapse
Affiliation(s)
- Nicole Caduff
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
- Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Lisa Rieble
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Michelle Böni
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Donal McHugh
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
- Pfizer, Medical Department, Schärenmoosstrasse 99, 8052, Zürich, Switzerland
| | - Romin Roshan
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Wendell Miley
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Nazzarena Labo
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Sumanta Barman
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Matthew Trivett
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Douwe M T Bosma
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Julia Rühl
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Norbert Goebels
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Denise Whitby
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
7
|
Wahl A, Yao W, Liao B, Chateau M, Richardson C, Ling L, Franks A, Senthil K, Doyon G, Li F, Frost J, Whitehurst CB, Pagano JS, Fletcher CA, Azcarate-Peril MA, Hudgens MG, Rogala AR, Tucker JD, McGowan I, Sartor RB, Garcia JV. A germ-free humanized mouse model shows the contribution of resident microbiota to human-specific pathogen infection. Nat Biotechnol 2024; 42:905-915. [PMID: 37563299 PMCID: PMC11073568 DOI: 10.1038/s41587-023-01906-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 07/10/2023] [Indexed: 08/12/2023]
Abstract
Germ-free (GF) mice, which are depleted of their resident microbiota, are the gold standard for exploring the role of the microbiome in health and disease; however, they are of limited value in the study of human-specific pathogens because they do not support their replication. Here, we develop GF mice systemically reconstituted with human immune cells and use them to evaluate the role of the resident microbiome in the acquisition, replication and pathogenesis of two human-specific pathogens, Epstein-Barr virus (EBV) and human immunodeficiency virus (HIV). Comparison with conventional (CV) humanized mice showed that resident microbiota enhance the establishment of EBV infection and EBV-induced tumorigenesis and increase mucosal HIV acquisition and replication. HIV RNA levels were higher in plasma and tissues of CV humanized mice compared with GF humanized mice. The frequency of CCR5+ CD4+ T cells throughout the intestine was also higher in CV humanized mice, indicating that resident microbiota govern levels of HIV target cells. Thus, resident microbiota promote the acquisition and pathogenesis of two clinically relevant human-specific pathogens.
Collapse
Affiliation(s)
- Angela Wahl
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Wenbo Yao
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Baolin Liao
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Morgan Chateau
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Cara Richardson
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lijun Ling
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adrienne Franks
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Krithika Senthil
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Genevieve Doyon
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Fengling Li
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Josh Frost
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Comparative Medicine, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christopher B Whitehurst
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY, USA
| | - Joseph S Pagano
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Craig A Fletcher
- Division of Comparative Medicine, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M Andrea Azcarate-Peril
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Microbiome Core, University of North Carolina, Chapel Hill, NC, USA
| | - Michael G Hudgens
- Department of Biostatistics, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Allison R Rogala
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Comparative Medicine, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joseph D Tucker
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Ian McGowan
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, USA
- Orion Biotechnology, Ottawa, Ontario, Canada
| | - R Balfour Sartor
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - J Victor Garcia
- International Center for the Advancement of Translational Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
8
|
Sin SH, Eason AB, Kim Y, Schneider JW, Damania B, Dittmer DP. The complete Kaposi sarcoma-associated herpesvirus genome induces early-onset, metastatic angiosarcoma in transgenic mice. Cell Host Microbe 2024; 32:755-767.e4. [PMID: 38653242 PMCID: PMC11305081 DOI: 10.1016/j.chom.2024.03.012] [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: 06/16/2022] [Revised: 01/16/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
Kaposi sarcoma (KS) is the most common cancer in persons living with HIV. It is caused by KS-associated herpesvirus (KSHV). There exists no animal model for KS. Pronuclear injection of the 170,000-bp viral genome induces early-onset, aggressive angiosarcoma in transgenic mice. The tumors are histopathologically indistinguishable from human KS. As in human KS, all tumor cells express the viral latency-associated nuclear antigen (LANA). The tumors transcribe most viral genes, whereas endothelial cells in other organs only transcribe the viral latent genes. The tumor cells are of endothelial lineage and exhibit the same molecular pattern of pathway activation as KS, namely phosphatidylinositol 3-kinase (PI3K)/Akt/mTOR, interleukin-10 (IL-10), and vascular endothelial growth factor (VEGF). The KSHV-induced tumors are more aggressive than Ha-ras-induced angiosarcomas. Overall survival is increased by prophylactic ganciclovir. Thus, whole-virus KSHV-transgenic mice represent an accurate model for KS and open the door for the genetic dissection of KS pathogenesis and evaluation of therapies, including vaccines.
Collapse
Affiliation(s)
- Sang-Hoon Sin
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anthony B Eason
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yongbaek Kim
- Laboratory of Veterinary Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Johann W Schneider
- National Health Laboratory Service, Division of Anatomical Pathology, Faculty of Medicine and Health Sciences, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
| | - Blossom Damania
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dirk P Dittmer
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
9
|
Stanfield BA, Ruiz E, Chouljenko VN, Kousoulas KG. Guinea pig herpes like virus is a gamma herpesvirus. Virus Genes 2024; 60:148-158. [PMID: 38340271 PMCID: PMC10978641 DOI: 10.1007/s11262-024-02054-x] [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: 09/26/2023] [Accepted: 01/21/2024] [Indexed: 02/12/2024]
Abstract
Guinea Pig Herpes-Like Virus (GPHLV) is a virus isolated from leukemic guinea pigs with herpes virus-like morphology described by Hsiung and Kaplow in 1969. GPHLV transformed embryonic cells from Syrian hamsters or rats, which were tumorigenic in adult animals. Herein, we present the genomic sequence of GPHLV strain LK40 as a reference for future molecular analysis. GPHLV has a broad host tropism and replicates efficiently in Guinea pig, Cat, and Green African Monkey-derived cell lines. GPHLV has a GC content of 35.45%. The genome is predicted to encode at least 75 open-reading frames (ORFs) with 84% (63 ORFs) sharing homology to human Kaposi Sarcoma Associated Herpes Virus (KSHV). Importantly, GPHLV encodes homologues of the KSHV oncogenes, vBCL2 (ORF16), vPK (ORF36), viral cyclin (v-cyclin, ORF72), the latency associated nuclear antigen (LANA, ORF73), and vGPCR (ORF74). GPHLV is a Rhadinovirus of Cavia porcellus, and we propose the formal name of Caviid gamma herpesvirus 1 (CaGHV-1). GPHLV can be a novel small animal model of Rhadinovirus pathogenesis with broad host tropism.
Collapse
Affiliation(s)
- Brent A Stanfield
- Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA.
| | - Emmanuelle Ruiz
- Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Vladimir N Chouljenko
- Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Konstantin G Kousoulas
- Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| |
Collapse
|
10
|
Lohani SC, Ramer-Tait AE, Li Q. High-fat diet feeding exacerbates HIV-1 rectal transmission. mSystems 2024; 9:e0132223. [PMID: 38303112 PMCID: PMC10949459 DOI: 10.1128/msystems.01322-23] [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] [Received: 12/11/2023] [Accepted: 01/07/2024] [Indexed: 02/03/2024] Open
Abstract
High-fat diet (HFD) is well known to impact various aspects of gut health and has been associated with many diseases and inflammation. However, the impact of HFD feeding on HIV-1 rectal transmission has not yet been well addressed. With an increasing threat of HIV-1 infection in men who have sex with men (MSM), where the rectal route is the primary mode of infection, it is imperative to understand the impact of HFD on gut microbiota and inflammation and consequently, its effect on HIV-1 rectal transmission. Here, we utilized our double humanized bone marrow, liver, thymus (dHu-BLT) mouse model to assess the impact of HFD feeding on the host's susceptibility to HIV-1 rectal transmission. We found that feeding an HFD successfully altered the gut microbial composition within 3 weeks in the dHu-BLT mouse model. In addition, levels of inflammatory mediators, specifically IL-12p70, IP-10, ICAM-1, and fecal calprotectin, were significantly higher in HFD-fed mice compared to control mice on a regular chow diet. We also observed that significantly different inflammatory markers (IL-12p70 and ICAM-1) were negatively correlated with the number of observed ASVs, Shannon diversity, and Faith's diversity in the HFD-fed group. Notably, when repeatedly challenged with a low dose of HIV-1 via a rectal route, mice receiving an HFD were significantly more susceptible to HIV-1 rectal infection than control mice. Together, these results underscore the impact of HFD feeding on the gut microbiota and inflammation and suggest the significance of diet-induced gut microbial dysbiosis and inflammation in promoting viral infection.IMPORTANCEHFD induces gut microbial dysbiosis and inflammation and has been associated with many infections and disease progression; however, its impact on HIV-1 rectal transmission is largely unknown. Given the increasing threat of HIV-1 incidence in men who have sex with men (MSM), it has become crucial to comprehend the impact of factors associated with gut health, like HFD consumption, on host susceptibility to HIV-1 rectal transmission. This is particularly important since anal intercourse remains the primary mode of HIV transmission within the MSM group. In this study, utilizing our unique mouse model, featuring both the human immune system and gut microbiota, we showed that HFD feeding led to gut microbial dysbiosis, induced inflammation, and increased HIV-1 rectal transmission. Collectively, our study highlights the significant impact of HFD on gut microbiota and inflammation and suggests an HFD consumption as a potential risk factor for promoting HIV-1 rectal susceptibility.
Collapse
Affiliation(s)
- Saroj Chandra Lohani
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Amanda E. Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Qingsheng Li
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
11
|
White KS, Walker JA, Wang J, Autissier P, Miller AD, Abuelezan NN, Burrack R, Li Q, Kim WK, Williams KC. Simian immunodeficiency virus-infected rhesus macaques with AIDS co-develop cardiovascular pathology and encephalitis. Front Immunol 2023; 14:1240946. [PMID: 37965349 PMCID: PMC10641955 DOI: 10.3389/fimmu.2023.1240946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/03/2023] [Indexed: 11/16/2023] Open
Abstract
Despite effective antiretroviral therapy, HIV co-morbidities remain where central nervous system (CNS) neurocognitive disorders and cardiovascular disease (CVD)-pathology that are linked with myeloid activation are most prevalent. Comorbidities such as neurocogntive dysfunction and cardiovascular disease (CVD) remain prevalent among people living with HIV. We sought to investigate if cardiac pathology (inflammation, fibrosis, cardiomyocyte damage) and CNS pathology (encephalitis) develop together during simian immunodeficiency virus (SIV) infection and if their co-development is linked with monocyte/macrophage activation. We used a cohort of SIV-infected rhesus macaques with rapid AIDS and demonstrated that SIV encephalitis (SIVE) and CVD pathology occur together more frequently than SIVE or CVD pathology alone. Their co-development correlated more strongly with activated myeloid cells, increased numbers of CD14+CD16+ monocytes, plasma CD163 and interleukin-18 (IL-18) than did SIVE or CVD pathology alone, or no pathology. Animals with both SIVE and CVD pathology had greater numbers of cardiac macrophages and increased collagen and monocyte/macrophage accumulation, which were better correlates of CVD-pathology than SIV-RNA. Animals with SIVE alone had higher levels of activated macrophage biomarkers and cardiac macrophage accumulation than SIVnoE animals. These observations were confirmed in HIV infected individuals with HIV encephalitis (HIVE) that had greater numbers of cardiac macrophages and fibrosis than HIV-infected controls without HIVE. These results underscore the notion that CNS and CVD pathologies frequently occur together in HIV and SIV infection, and demonstrate an unmet need for adjunctive therapies targeting macrophages.
Collapse
Affiliation(s)
- Kevin S. White
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Joshua A. Walker
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - John Wang
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Patrick Autissier
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Andrew D. Miller
- Department of Biomedical Sciences, Section of Anatomic Physiology, Cornell University College of Veterinary Medicine, Ithaca, NY, United States
| | - Nadia N. Abuelezan
- Connel School of Nursing, Boston College, Chestnut Hill, MA, United States
| | - Rachel Burrack
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Woong-Ki Kim
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| | | |
Collapse
|
12
|
Julius P, Kang G, Siyumbwa S, Musumali J, Tso FY, Ngalamika O, Kaile T, Maate F, Moonga P, West JT, Angeletti P, Wood C. Co-infection and co-localization of Kaposi sarcoma-associated herpesvirus and Epstein-Barr virus in HIV-associated Kaposi sarcoma: a case report. Front Cell Infect Microbiol 2023; 13:1270935. [PMID: 37928187 PMCID: PMC10623342 DOI: 10.3389/fcimb.2023.1270935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/04/2023] [Indexed: 11/07/2023] Open
Abstract
Kaposi sarcoma (KS), a multifocal vascular neoplasm frequently observed in HIV-positive individuals, primarily affects the skin, mucous membranes, visceral organs, and lymph nodes. KS is associated primarily with Kaposi sarcoma-associated herpesvirus (KSHV) infection. In this case report, we present a rare occurrence of co-infection and co-localization of KSHV and Epstein-Barr virus (EBV) in KS arising from the conjunctiva, which, to our knowledge, has not been reported previously. Immunohistochemistry (IHC), DNA polymerase chain reaction (PCR), and EBV-encoded RNA in situ hybridization (EBER-ISH) were utilized to demonstrate the presence of KSHV and EBV infection in the ocular KS lesion. Nearly all KSHV-positive cells displayed co-infection with EBV. In addition, the KS lesion revealed co-localization of KSHV Latency-Associated Nuclear Antigen (LANA) and EBV Epstein Barr virus Nuclear Antigen-1 (EBNA1) by multi-colored immunofluorescence staining with different anti-EBNA1 antibodies, indicating the possibility of interactions between these two gamma herpesviruses within the same lesion. Additional study is needed to determine whether EBV co-infection in KS is a common or an opportunistic event that might contribute to KS development and progression.
Collapse
Affiliation(s)
- Peter Julius
- Department of Pathology and Microbiology, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Guobin Kang
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center – New Orleans, New Orleans, LA, United States
| | - Stepfanie Siyumbwa
- Department of Pathology and Microbiology, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Jane Musumali
- University Teaching Hospitals, Eye Hospital, Ministry of Health, Lusaka, Zambia
| | - For Yue Tso
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center – New Orleans, New Orleans, LA, United States
| | - Owen Ngalamika
- Department of Pathology and Microbiology, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Trevor Kaile
- Department of Pathology and Microbiology, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Fred Maate
- Department of Pathology and Microbiology, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Phyllis Moonga
- University Teaching Hospitals, Eye Hospital, Ministry of Health, Lusaka, Zambia
| | - John T. West
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center – New Orleans, New Orleans, LA, United States
| | - Peter Angeletti
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Charles Wood
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center – New Orleans, New Orleans, LA, United States
| |
Collapse
|
13
|
Kutle I, Dittrich A, Wirth D. Mouse Models for Human Herpesviruses. Pathogens 2023; 12:953. [PMID: 37513800 PMCID: PMC10384569 DOI: 10.3390/pathogens12070953] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
More than one hundred herpesviruses have been isolated from different species so far, with nine infecting humans. Infections with herpesviruses are characterized by life-long latency and represent a significant challenge for human health. To investigate the consequences of infections and identify novel treatment options, in vivo models are of particular relevance. The mouse has emerged as an economical small animal model to investigate herpesvirus infections. However, except for herpes simplex viruses (HSV-1, HSV-2), human herpesviruses cannot infect mice. Three natural herpesviruses have been identified in mice: mouse-derived cytomegalovirus (MCMV), mouse herpesvirus 68 (MHV-68), and mouse roseolovirus (MRV). These orthologues are broadly used to investigate herpesvirus infections within the natural host. In the last few decades, immunocompromised mouse models have been developed, allowing the functional engraftment of various human cells and tissues. These xenograft mice represent valuable model systems to investigate human-restricted viruses, making them particularly relevant for herpesvirus research. In this review, we describe the various mouse models used to study human herpesviruses, thereby highlighting their potential and limitations. Emphasis is laid on xenograft mouse models, covering the development and refinement of immune-compromised mice and their application in herpesvirus research.
Collapse
Affiliation(s)
- Ivana Kutle
- Research Group Model Systems for Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Anne Dittrich
- Research Group Model Systems for Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- InSCREENeX GmbH, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Dagmar Wirth
- Research Group Model Systems for Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
14
|
Privatt SR, Ngalamika O, Zhang J, Li Q, Wood C, West JT. Upregulation of Cell Surface Glycoproteins in Correlation with KSHV LANA in the Kaposi Sarcoma Tumor Microenvironment. Cancers (Basel) 2023; 15:2171. [PMID: 37046832 PMCID: PMC10093722 DOI: 10.3390/cancers15072171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/29/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
HIV-associated epidemic Kaposi sarcoma (EpKS) remains one of the most prevalent cancers in sub-Saharan Africa despite the widespread uptake of anti-retroviral therapy and HIV-1 suppression. In an effort to define potential therapeutic targets against KS tumors, we analyzed previously published KS bulk tumor transcriptomics to identify cell surface biomarkers. In addition to upregulated gene expression (>6-fold) in the EpKS tumor microenvironment, biomarkers were selected for correlation with KSHV latency-associated nuclear antigen (LANA) expression. The cell surface glycoprotein genes identified were KDR, FLT4, ADAM12, UNC5A, ZP2, and OX40, as well as the endothelial lineage determinants Prox-1 and CD34. Each protein was evaluated for its expression and co-localization with KSHV LANA using multi-color immunofluorescence in KS tissues, KSHV-infected L1T2 cells, uninfected TIVE cells, and murine L1T2 tumor xenografts. Five surface glycoproteins (KDR, FLT4, UNC5A, ADAM12, and CD34) were associated with LANA-positive cells but were also detected in uninfected cells in the KS microenvironment. In vitro L1T2 cultures showed evidence of only FLT4, KDR, and UNC5A, whereas mouse L1T2 xenografts recapitulated human KS cell surface expression profiles, with the exception of CD34 and Prox-1. In KS tumors, most LANA-positive cells co-expressed markers of vascular as well as lymphatic endothelial lineages, suggesting KS-associated dedifferentiation to a more mesenchymal/progenitor phenotype.
Collapse
Affiliation(s)
- Sara R. Privatt
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
- Department of Interdisciplinary Oncology, Louisiana Cancer Research Center, Louisiana State University Health Sciences Center-New Orleans, New Orleans, LA 70112, USA
| | - Owen Ngalamika
- University Teaching Hospital, University of Zambia School of Medicine, Lusaka 10101, Zambia
| | - Jianshui Zhang
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Qinsheng Li
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Charles Wood
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
- Department of Interdisciplinary Oncology, Louisiana Cancer Research Center, Louisiana State University Health Sciences Center-New Orleans, New Orleans, LA 70112, USA
| | - John T. West
- Department of Interdisciplinary Oncology, Louisiana Cancer Research Center, Louisiana State University Health Sciences Center-New Orleans, New Orleans, LA 70112, USA
| |
Collapse
|
15
|
Jondle CN, Sylvester PA, Schmalzriedt DL, Njoya K, Tarakanova VL. The Antagonism between the Murine Gammaherpesvirus Protein Kinase and Global Interferon Regulatory Factor 1 Expression Shapes the Establishment of Chronic Infection. J Virol 2022; 96:e0126022. [PMID: 36169331 PMCID: PMC9599343 DOI: 10.1128/jvi.01260-22] [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: 08/11/2022] [Accepted: 09/09/2022] [Indexed: 11/20/2022] Open
Abstract
Gammaherpesviruses infect most vertebrate species and are associated with B cell lymphomas. Manipulation of B cell differentiation is critical for natural infection and lymphomagenesis driven by gammaherpesviruses. Specifically, human Epstein-Barr virus (EBV) and murine gammaherpesvirus 68 (MHV68) drive differentiation of infected naive B cells into the germinal center to achieve exponential increase in the latent viral reservoir during the establishment of chronic infection. Infected germinal center B cells are also the target of viral lymphomagenesis, as most EBV-positive B cell lymphomas bear the signature of the germinal center response. All gammaherpesviruses encode a protein kinase, which, in the case of Kaposi's sarcoma-associated herpesvirus (KSHV) and MHV68, is sufficient and necessary, respectively, to drive B cell differentiation in vivo. In this study, we used the highly tractable MHV68 model of chronic gammaherpesvirus infection to unveil an antagonistic relationship between MHV68 protein kinase and interferon regulatory factor 1 (IRF-1). IRF-1 deficiency had minimal effect on the attenuated lytic replication of the kinase-null MHV68 in vivo. In contrast, the attenuated latent reservoir of the kinase-null MHV68 was partially to fully rescued in IRF-1-/- mice, along with complete rescue of the MHV68-driven germinal center response. Thus, the novel viral protein kinase-IRF-1 antagonism was largely limited to chronic infection dominated by viral latency and was less relevant for lytic replication during acute infection and in vitro. Given the conserved nature of the viral and host protein, the antagonism between the two, as defined in this study, may regulate gammaherpesvirus infection across species. IMPORTANCE Gammaherpesviruses are prevalent pathogens that manipulate physiological B cell differentiation to establish lifelong infection. This manipulation is also involved in gammaherpesvirus-driven B cell lymphomas, as differentiation of latently infected B cells through the germinal center response targets these for transformation. In this study, we define a novel antagonistic interaction between a conserved gammaherpesvirus protein kinase and a host antiviral and tumor suppressor transcription factor. The virus-host antagonism unveiled in this study was critically important to shape the magnitude of gammaherpesvirus-driven germinal center response. In contrast, the virus-host antagonism was far less relevant for lytic viral replication in vitro and during acute infection in vivo, highlighting the emerging concept that nonoverlapping mechanisms shape the parameters of acute and chronic gammaherpesvirus infection.
Collapse
Affiliation(s)
- C. N. Jondle
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - P. A. Sylvester
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - D. L. Schmalzriedt
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - K. Njoya
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - V. L. Tarakanova
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
16
|
Sylvester PA, Jondle CN, Schmalzriedt DL, Dittel BN, Tarakanova VL. T Cell-Specific STAT1 Expression Promotes Lytic Replication and Supports the Establishment of Gammaherpesvirus Latent Reservoir in Splenic B Cells. mBio 2022; 13:e0210722. [PMID: 35968944 PMCID: PMC9430880 DOI: 10.1128/mbio.02107-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/30/2022] Open
Abstract
Gammaherpesviruses establish lifelong infections in most vertebrate species, including humans and rodents, and are associated with cancers, including B cell lymphomas. While type I and II interferon (IFN) systems of the host are critical for the control of acute and chronic gammaherpesvirus infection, the cell type-specific role(s) of IFN signaling during infection is poorly understood and is often masked by the profoundly altered viral pathogenesis in the hosts with global IFN deficiencies. STAT1 is a critical effector of all classical IFN responses along with its involvement in other cytokine signaling pathways. In this study, we defined the effect of T cell-specific STAT1 deficiency on the viral and host parameters of infection with murine gammaherpesvirus 68 (MHV68). MHV68 is a natural rodent pathogen that, similar to human gammaherpesviruses, manipulates and usurps B cell differentiation to establish a lifelong latent reservoir in B cells. Specifically, germinal center B cells host the majority of latent MHV68 reservoir in the lymphoid organs, particularly at the peak of viral latency. Unexpectedly, T cell-specific STAT1 expression, while limiting the overall expansion of the germinal center B cell population during chronic infection, rendered these B cells more effective at hosting the latent virus reservoir. Further, T cell-specific STAT1 expression in a wild type host limited circulating levels of IFNγ, with corresponding increases in lytic MHV68 replication and viral reactivation. Thus, our study unveils an unexpected proviral role of T cell-specific STAT1 expression during gammaherpesvirus infection of a natural intact host. IMPORTANCE Interferons (IFNs) represent a major antiviral host network vital to the control of multiple infections, including acute and chronic gammaherpesvirus infections. Ubiquitously expressed STAT1 plays a critical effector role in all classical IFN responses. This study utilized a mouse model of T cell-specific STAT1 deficiency to define cell type-intrinsic role of STAT1 during natural gammaherpesvirus infection. Unexpectedly, T cell-specific loss of STAT1 led to better control of acute and persistent gammaherpesvirus replication and decreased establishment of latent viral reservoir in B cells, revealing a surprisingly diverse proviral role of T cell-intrinsic STAT1.
Collapse
Affiliation(s)
- P. A. Sylvester
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - C. N. Jondle
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - D. L. Schmalzriedt
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - B. N. Dittel
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
| | - V. L. Tarakanova
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
17
|
Kenney DJ, O'Connell AK, Turcinovic J, Montanaro P, Hekman RM, Tamura T, Berneshawi AR, Cafiero TR, Al Abdullatif S, Blum B, Goldstein SI, Heller BL, Gertje HP, Bullitt E, Trachtenberg AJ, Chavez E, Nono ET, Morrison C, Tseng AE, Sheikh A, Kurnick S, Grosz K, Bosmann M, Ericsson M, Huber BR, Saeed M, Balazs AB, Francis KP, Klose A, Paragas N, Campbell JD, Connor JH, Emili A, Crossland NA, Ploss A, Douam F. Humanized mice reveal a macrophage-enriched gene signature defining human lung tissue protection during SARS-CoV-2 infection. Cell Rep 2022; 39:110714. [PMID: 35421379 PMCID: PMC8977517 DOI: 10.1016/j.celrep.2022.110714] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/17/2022] [Accepted: 03/29/2022] [Indexed: 01/11/2023] Open
Abstract
The human immunological mechanisms defining the clinical outcome of SARS-CoV-2 infection remain elusive. This knowledge gap is mostly driven by the lack of appropriate experimental platforms recapitulating human immune responses in a controlled human lung environment. Here, we report a mouse model (i.e., HNFL mice) co-engrafted with human fetal lung xenografts (fLX) and a myeloid-enhanced human immune system to identify cellular and molecular correlates of lung protection during SARS-CoV-2 infection. Unlike mice solely engrafted with human fLX, HNFL mice are protected against infection, severe inflammation, and histopathological phenotypes. Lung tissue protection from infection and severe histopathology associates with macrophage infiltration and differentiation and the upregulation of a macrophage-enriched signature composed of 11 specific genes mainly associated with the type I interferon signaling pathway. Our work highlights the HNFL model as a transformative platform to investigate, in controlled experimental settings, human myeloid immune mechanisms governing lung tissue protection during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Devin J Kenney
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Aoife K O'Connell
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Jacquelyn Turcinovic
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA; Bioinformatics Program, Boston University, Boston, MA, USA
| | - Paige Montanaro
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Ryan M Hekman
- Center for Network Systems Biology, Boston University, Boston, MA, USA; Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Tomokazu Tamura
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | | | - Thomas R Cafiero
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Salam Al Abdullatif
- Single Cell RNA Sequencing Core, Boston University, Boston, MA, USA; Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Benjamin Blum
- Center for Network Systems Biology, Boston University, Boston, MA, USA; Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Stanley I Goldstein
- Center for Network Systems Biology, Boston University, Boston, MA, USA; Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Brigitte L Heller
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Hans P Gertje
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Esther Bullitt
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA, USA
| | - Alexander J Trachtenberg
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Elizabeth Chavez
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Evans Tuekam Nono
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Catherine Morrison
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Anna E Tseng
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Amira Sheikh
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Susanna Kurnick
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA; Animal Science Center, Boston University, Boston, MA, USA
| | - Kyle Grosz
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA; Animal Science Center, Boston University, Boston, MA, USA
| | - Markus Bosmann
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz 55131, Germany
| | - Maria Ericsson
- Electron Microscopy Core Facility, Harvard Medical School, Boston, MA, USA
| | - Bertrand R Huber
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Mohsan Saeed
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA; Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | | | | | | | - Neal Paragas
- In Vivo Analytics, Inc., New York, NY, USA; Department of Radiology Imaging Research Lab, University of Washington, Seattle, WA, USA
| | - Joshua D Campbell
- Single Cell RNA Sequencing Core, Boston University, Boston, MA, USA; Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - John H Connor
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Andrew Emili
- Center for Network Systems Biology, Boston University, Boston, MA, USA; Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; Department of Biology, Boston University School of Medicine, Boston, MA, USA
| | - Nicholas A Crossland
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA.
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| | - Florian Douam
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA.
| |
Collapse
|
18
|
Moorad R, Juarez A, Landis JT, Pluta LJ, Perkins M, Cheves A, Dittmer DP. Whole-genome sequencing of Kaposi sarcoma-associated herpesvirus (KSHV/HHV8) reveals evidence for two African lineages. Virology 2022; 568:101-114. [PMID: 35152042 PMCID: PMC8915436 DOI: 10.1016/j.virol.2022.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/07/2022] [Accepted: 01/16/2022] [Indexed: 12/28/2022]
Abstract
Kaposi sarcoma (KS)-associated herpesvirus (KSHV/HHV-8) was first sequenced from the body cavity (BC) lymphoma cell line, BC-1, in 1996. Few other KSHV genomes have been reported. Our knowledge of sequence variation for this virus remains spotty. This study reports additional genomes from historical US patient samples and from African KS biopsies. It describes an assay that spans regions of the virus that cannot be covered by short read sequencing. These include the terminal repeats, the LANA repeats, and the origins of replication. A phylogenetic analysis, based on 107 genomes, identified three distinct clades; one containing isolates from USA/Europe/Japan collected in the 1990s and two of Sub-Saharan Africa isolates collected since 2010. This analysis indicates that the KSHV strains circulating today differ from the isolates collected at the height of the AIDS epidemic. This analysis helps experimental designs and potential vaccine studies.
Collapse
Affiliation(s)
- Razia Moorad
- Lineberger Comprehensive Cancer Center, School of Medicine, Department of Immunology and Microbiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Angelica Juarez
- Lineberger Comprehensive Cancer Center, School of Medicine, Department of Immunology and Microbiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Justin T Landis
- Lineberger Comprehensive Cancer Center, School of Medicine, Department of Immunology and Microbiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Linda J Pluta
- Lineberger Comprehensive Cancer Center, School of Medicine, Department of Immunology and Microbiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Megan Perkins
- Lineberger Comprehensive Cancer Center, School of Medicine, Department of Immunology and Microbiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Avery Cheves
- Lineberger Comprehensive Cancer Center, School of Medicine, Department of Immunology and Microbiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dirk P Dittmer
- Lineberger Comprehensive Cancer Center, School of Medicine, Department of Immunology and Microbiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
19
|
Avdoshina DV, Kondrashova AS, Belikova MG, Bayurova EO. Murine Models of Chronic Viral Infections and Associated Cancers. Mol Biol 2022; 56:649-667. [PMID: 36217336 PMCID: PMC9534466 DOI: 10.1134/s0026893322050028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 11/07/2022]
Abstract
Viruses are now recognized as bona fide etiologic factors of human cancer. Carcinogenic viruses include Epstein– Barr virus (EBV), high-risk human papillomaviruses (HPVs), hepatitis B virus (HBV), hepatitis C virus (HCV), human T-cell leukemia virus type 1 (HTLV-1), human immunodeficiency virus type 1 (HIV-1, indirectly), and several candidate human cancer viruses. It is estimated that 15% of all human tumors worldwide are caused by viruses. Tumor viruses establish long-term persistent infections in humans, and cancer is an accidental side effect of viral replication strategies. Viruses are usually not complete carcinogens, supporting the concept that cancer results from the accumulation of multiple cooperating events, in which human cancer viruses display different, often opposing roles. The laboratory mouse Mus musculus is one of the best in vivo experimental systems for modeling human pathology, including viral infections and cancer. However, mice are unsusceptible to infection with the known carcinogenic viruses. Many murine models were developed to overcome this limitation and to address various aspects of virus-associated carcinogenesis, from tumors resulting from xenografts of human tissues and cells, including cancerous and virus infected, to genetically engineered mice susceptible to viral infections and associated cancer. The review considers the main existing models, analyzes their advantages and drawbacks, describes their applications, outlines the prospects of their further development.
Collapse
Affiliation(s)
- D. V. Avdoshina
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Products, Russian Academy of Sciences (Polio Institute), 108819 Moscow, Russia
| | - A. S. Kondrashova
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Products, Russian Academy of Sciences (Polio Institute), 108819 Moscow, Russia
| | - M. G. Belikova
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Products, Russian Academy of Sciences (Polio Institute), 108819 Moscow, Russia ,Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia ,Peoples’ Friendship University of Russia, 117198 Moscow, Russia
| | - E. O. Bayurova
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Products, Russian Academy of Sciences (Polio Institute), 108819 Moscow, Russia ,Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| |
Collapse
|
20
|
Li D, Baloch Z, Zhao Y, Bai L, Wang X, Wang G, Zhang AM, Lan K, Xia X. Establishment of Tree Shrew Animal Model for Kaposi's Sarcoma-Associated Herpesvirus (HHV-8) Infection. Front Microbiol 2021; 12:710067. [PMID: 34603235 PMCID: PMC8481836 DOI: 10.3389/fmicb.2021.710067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/01/2021] [Indexed: 01/22/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the most common cause of Kaposi's sarcoma (KS) and other malignant growths in humans. However, the lack of a KSHV-infected small animal model has hampered understanding of the mechanisms of KSHV infection, virus replication, pathogenesis, and persistence. This study was designed to explore the susceptibility of tree shrews as a possible KSHV-infected small animal model. A recombinant GFP (latent)/RFP (lytic)-positive rKSHV.219 strain was used to infect primary cells cultured from different tissues of tree shrews as an in vitro model and adult tree shrews as an in vivo model. KSHV latent nuclear antigen (LANA) and DNA were successfully detected in primary cells of tree shrews. Among them, tree shrew kidney epithelial cells (TSKEC) were the most susceptible cells to KSHV infection compared to other cells. KSHV genomic DNA, mRNA, and KSHV-specific proteins were readily detected in the TSKEC cultured up to 32 dpi. Moreover, KSHV DNA and mRNA transcription were also readily detected in the peripheral blood mononuclear cells (PBMCs) and various tissues of tree shrews infected with KSHV. Haematoxylin and eosin (HE) staining showed lymphocyte infiltration, lymphoid tissue focal aggregation, alveolar wall thickening, hepatocyte edema, hepatic necrosis in the spleen, lung, and liver of KSHV-infected animals. Additionally, immune-histochemical (IHC) staining showed that LANA or ORF62-positive cells were present in the spleen, lung, liver, and kidney of KSHV-infected tree shrews. Here, we have successfully established in vitro and in vivo KSHV latent infection in tree shrews. This small animal model is not only useful for studying the pathogenesis of KSHV in vivo but can also be a useful model to study transmission routes of viral infection and a useful platform to characterize the novel therapeutics against KSHV.
Collapse
Affiliation(s)
- Daoqun Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
- Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zulqarnain Baloch
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Yang Zhao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Lei Bai
- State Key Laboratory of Virology, College of Life Sciences, Medical Research Institute, Wuhan University, Wuhan, China
| | - Xing Wang
- State Key Laboratory of Virology, College of Life Sciences, Medical Research Institute, Wuhan University, Wuhan, China
| | - Gang Wang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - A-Mei Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Ke Lan
- State Key Laboratory of Virology, College of Life Sciences, Medical Research Institute, Wuhan University, Wuhan, China
| | - Xueshan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
21
|
Conserved Gammaherpesvirus Protein Kinase Counters the Antiviral Effects of Myeloid Cell-Specific STAT1 Expression To Promote the Establishment of Splenic B Cell Latency. J Virol 2021; 95:e0085921. [PMID: 34132573 DOI: 10.1128/jvi.00859-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gammaherpesviruses establish lifelong infections and are associated with B cell lymphomas. Murine gammaherpesvirus 68 (MHV68) infects epithelial and myeloid cells during acute infection, with subsequent passage of the virus to B cells, where physiological B cell differentiation is usurped to ensure the establishment of a chronic latent reservoir. Interferons (IFNs) represent a major antiviral defense system that engages the transcriptional factor STAT1 to attenuate diverse acute and chronic viral infections, including those of gammaherpesviruses. Correspondingly, global deficiency of type I or type II IFN signaling profoundly increases the pathogenesis of acute and chronic gammaherpesvirus infection, compromises host survival, and impedes mechanistic understanding of cell type-specific role of IFN signaling. Here, we demonstrate that myeloid-specific STAT1 expression attenuates acute and persistent MHV68 replication in the lungs and suppresses viral reactivation from peritoneal cells, without any effect on the establishment of viral latent reservoir in splenic B cells. All gammaherpesviruses encode a conserved protein kinase that antagonizes type I IFN signaling in vitro. Here, we show that myeloid-specific STAT1 deficiency rescues the attenuated splenic latent reservoir of the kinase-null MHV68 mutant. However, despite having gained access to splenic B cells, the protein kinase-null MHV68 mutant fails to drive B cell differentiation. Thus, while myeloid-intrinsic STAT1 expression must be counteracted by the gammaherpesvirus protein kinase to facilitate viral passage to splenic B cells, expression of the viral protein kinase continues to be required to promote optimal B cell differentiation and viral reactivation, highlighting the multifunctional nature of this conserved viral protein during chronic infection. IMPORTANCE IFN signaling is a major antiviral system of the host that suppresses replication of diverse viruses, including acute and chronic gammaherpesvirus infection. STAT1 is a critical member and the primary antiviral effector of IFN signaling pathways. Given the significantly compromised antiviral status of global type I or type II IFN deficiency, unabated gammaherpesvirus replication and pathogenesis hinders understanding of cell type-specific antiviral effects. In this study, a mouse model of myeloid-specific STAT1 deficiency unveiled site-specific antiviral effects of STAT1 in the lungs and peritoneal cavity, but not the spleen, of chronically infected hosts. Interestingly, expression of a conserved gammaherpesvirus protein kinase was required to counteract the antiviral effects of myeloid-specific STAT1 expression to facilitate latent infection of splenic B cells, revealing a cell type-specific virus-host antagonism during the establishment of chronic gammaherpesvirus infection.
Collapse
|
22
|
Low-Density Lipoprotein Receptor Suppresses the Endogenous Cholesterol Synthesis Pathway To Oppose Gammaherpesvirus Replication in Primary Macrophages. J Virol 2021; 95:e0064921. [PMID: 34105999 PMCID: PMC8354329 DOI: 10.1128/jvi.00649-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gammaherpesviruses are ubiquitous pathogens that establish lifelong infections in >95% of adults worldwide and are associated with several cancers. We have shown that endogenous cholesterol synthesis supports gammaherpesvirus replication. However, the role of exogenous cholesterol exchange and signaling during infection remains poorly understood. Extracellular cholesterol is carried in the serum by several lipoproteins, including low-density lipoproteins (LDL). The LDL receptor (LDL-R) mediates the endocytosis of these cholesterol-rich LDL particles into the cell, thereby supplying the cell with cholesterol. We found that LDL-R expression attenuates gammaherpesvirus replication during the early stages of the replication cycle, as evident by increased viral gene expression in LDL-R-/- primary macrophages. This was not observed in primary fibroblasts, indicating that the antiviral effects of LDL-R are cell type specific. Increased viral gene expression in LDL-R-/- primary macrophages was due to increased activity of the endogenous cholesterol synthesis pathway. Intriguingly, despite type I interferon-driven increase in LDL-R mRNA levels in infected macrophages, protein levels of LDL-R continually decreased over the single cycle of viral replication. Thus, our study has uncovered an intriguing tug of war between the LDL-R-driven antiviral effect on cholesterol metabolism and the viral targeting of the LDL-R protein. IMPORTANCE LDL-R is a cell surface receptor that mediates the endocytosis of cholesterol-rich low-density lipoproteins, allowing cells to acquire cholesterol exogenously. Several RNA viruses usurp LDL-R function to facilitate replication; however, the role of LDL-R in DNA virus infection remains unknown. Gammaherpesviruses are double-stranded DNA viruses that are associated with several cancers. Here, we show that LDL-R attenuates gammaherpesvirus replication in primary macrophages by decreasing endogenous cholesterol synthesis activity, a pathway known to support gammaherpesvirus replication. In response, LDL-R protein levels are decreased in infected cells to mitigate the antiviral effects, revealing an intriguing tug of war between the virus and the host.
Collapse
|
23
|
Dash PK, Gorantla S, Poluektova L, Hasan M, Waight E, Zhang C, Markovic M, Edagwa B, Machhi J, Olson KE, Wang X, Mosley RL, Kevadiya B, Gendelman HE. Humanized Mice for Infectious and Neurodegenerative disorders. Retrovirology 2021; 18:13. [PMID: 34090462 PMCID: PMC8179712 DOI: 10.1186/s12977-021-00557-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/22/2021] [Indexed: 12/12/2022] Open
Abstract
Humanized mice model human disease and as such are used commonly for research studies of infectious, degenerative and cancer disorders. Recent models also reflect hematopoiesis, natural immunity, neurobiology, and molecular pathways that influence disease pathobiology. A spectrum of immunodeficient mouse strains permit long-lived human progenitor cell engraftments. The presence of both innate and adaptive immunity enables high levels of human hematolymphoid reconstitution with cell susceptibility to a broad range of microbial infections. These mice also facilitate investigations of human pathobiology, natural disease processes and therapeutic efficacy in a broad spectrum of human disorders. However, a bridge between humans and mice requires a complete understanding of pathogen dose, co-morbidities, disease progression, environment, and genetics which can be mirrored in these mice. These must be considered for understanding of microbial susceptibility, prevention, and disease progression. With known common limitations for access to human tissues, evaluation of metabolic and physiological changes and limitations in large animal numbers, studies in mice prove important in planning human clinical trials. To these ends, this review serves to outline how humanized mice can be used in viral and pharmacologic research emphasizing both current and future studies of viral and neurodegenerative diseases. In all, humanized mouse provides cost-effective, high throughput studies of infection or degeneration in natural pathogen host cells, and the ability to test transmission and eradication of disease.
Collapse
Affiliation(s)
- Prasanta K Dash
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Larisa Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mahmudul Hasan
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Emiko Waight
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chen Zhang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Milica Markovic
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Benson Edagwa
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Xinglong Wang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bhavesh Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
24
|
Lange PT, Damania B. Modeling oncogenic herpesvirus infections in humanized mice. Curr Opin Virol 2020; 44:90-96. [PMID: 32784124 PMCID: PMC7755680 DOI: 10.1016/j.coviro.2020.07.005] [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: 06/15/2020] [Accepted: 07/04/2020] [Indexed: 11/22/2022]
Abstract
The creation of humanized mice generally involves the reconstitution of immunodeficient mice with human immune constituents. Different methodologies have been employed, and significant progress has been made towards the development of robustly humanized mouse models. Some of the techniques used include the injection of mature human immune cells, the injection of human hematopoietic stem cells (HSCs) capable of reconstituting radiation-depleted murine bone marrow, and the implantation of human fetal liver and thymus fragments under the kidney capsule to create a thymic organoid that can support thympoiesis. This review will serve as a brief introduction to the three most commonly utilized humanized mouse models for the study of gammaherpesvirus-driven pathogenesis, and highlight some of the critical discoveries these models have enabled.
Collapse
Affiliation(s)
- Philip T Lange
- Department of Microbiology and Immunology and Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Blossom Damania
- Department of Microbiology and Immunology and Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| |
Collapse
|
25
|
Münz C. Probing Reconstituted Human Immune Systems in Mice With Oncogenic γ-Herpesvirus Infections. Front Immunol 2020; 11:581419. [PMID: 33013936 PMCID: PMC7509489 DOI: 10.3389/fimmu.2020.581419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/17/2020] [Indexed: 12/22/2022] Open
Abstract
Mice with reconstituted human immune systems can mount cell-mediated immune responses against the human tumor viruses Epstein Barr virus (EBV) and Kaposi sarcoma associated herpesvirus (KSHV). Primarily cytotoxic lymphocytes protect the vast majority of persistently infected carriers of these tumor viruses from the respective malignancies for life. Thus, EBV and KSHV infection can teach us how this potent immune control is induced, what phenotype and functions characterize the protective lymphocyte compartments and if similar immune responses could be induced by vaccination. This review will summarize similarities and differences between EBV and KSHV associated pathologies and their immune control in patients and mice with reconstituted human immune systems. Furthermore, it will high-light which aspects of the near perfect immune control can be modeled in the latter preclinical animal models and discuss their relevance for cancer immunology in general.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, Zurich, Switzerland
| |
Collapse
|
26
|
Agarwal Y, Beatty C, Ho S, Thurlow L, Das A, Kelly S, Castronova I, Salunke R, Biradar S, Yeshi T, Richardson A, Bility M. Development of humanized mouse and rat models with full-thickness human skin and autologous immune cells. Sci Rep 2020; 10:14598. [PMID: 32884084 PMCID: PMC7471691 DOI: 10.1038/s41598-020-71548-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 08/13/2020] [Indexed: 12/12/2022] Open
Abstract
The human skin is a significant barrier for protection against pathogen transmission. Rodent models used to investigate human-specific pathogens that target the skin are generated by introducing human skin grafts to immunocompromised rodent strains. Infection-induced immunopathogenesis has been separately studied in humanized rodent models developed with human lymphoid tissue and hematopoietic stem cell transplants. Successful co-engraftment of human skin, autologous lymphoid tissues, and autologous immune cells in a rodent model has not yet been achieved, though it could provide a means of studying the human immune response to infection in the human skin. Here, we introduce the human Skin and Immune System (hSIS)-humanized NOD-scid IL2Rγnull (NSG) mouse and Sprague–Dawley-Rag2tm2hera Il2rγtm1hera (SRG) rat models, co-engrafted with human full-thickness fetal skin, autologous fetal lymphoid tissues, and autologous fetal liver-derived hematopoietic stem cells. hSIS-humanized rodents demonstrate the development of human full-thickness skin, along with autologous lymphoid tissues, and autologous immune cells. These models also support human skin infection following intradermal inoculation with community-associated methicillin-resistant Staphylococcus aureus. The co-engraftment of these human skin and immune system components into a single humanized rodent model could provide a platform for studying human skin infections.
Collapse
Affiliation(s)
- Yash Agarwal
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Cole Beatty
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Sara Ho
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Lance Thurlow
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, USA
| | - Antu Das
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Samantha Kelly
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Isabella Castronova
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Rajeev Salunke
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Shivkumar Biradar
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | | | - Anthony Richardson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, USA
| | - Moses Bility
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
27
|
McHugh D, Caduff N, Murer A, Engelmann C, Deng Y, Zdimerova H, Zens K, Chijioke O, Münz C. Infection and immune control of human oncogenic γ-herpesviruses in humanized mice. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180296. [PMID: 30955487 DOI: 10.1098/rstb.2018.0296] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epstein-Barr virus (EBV) and Kaposi sarcoma-associated herpesvirus (KSHV) comprise the oncogenic human γ-herpesvirus family and are responsible for 2-3% of all tumours in man. With their prominent growth-transforming abilities and high prevalence in the human population, these pathogens have probably shaped the human immune system throughout evolution for near perfect immune control of the respective chronic infections in the vast majority of healthy pathogen carriers. The exclusive tropism of EBV and KSHV for humans has, however, made it difficult in the past to study their infection, tumourigenesis and immune control in vivo. Mice with reconstituted human immune system components (humanized mice) support replication of both viruses with both persisting latent and productive lytic infection. Moreover, B-cell lymphomas can be induced by EBV alone and KSHV co-infection with gene expression hallmarks of human malignancies that are associated with both viruses. Furthermore, cell-mediated immune control by primarily cytotoxic lymphocytes is induced upon infection and can be probed for its functional characteristics as well as putative requirements for its priming. Insights that have been gained from this model and remaining questions will be discussed in this review. This article is part of the theme issue 'Silent cancer agents: multi-disciplinary modelling of human DNA oncoviruses'.
Collapse
Affiliation(s)
- Donal McHugh
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Nicole Caduff
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Anita Murer
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Christine Engelmann
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Yun Deng
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Hana Zdimerova
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Kyra Zens
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Obinna Chijioke
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| |
Collapse
|
28
|
Animal Models for Gammaherpesvirus Infections: Recent Development in the Analysis of Virus-Induced Pathogenesis. Pathogens 2020; 9:pathogens9020116. [PMID: 32059472 PMCID: PMC7167833 DOI: 10.3390/pathogens9020116] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/11/2022] Open
Abstract
Epstein–Barr virus (EBV) is involved in the pathogenesis of various lymphomas and carcinomas, whereas Kaposi’s sarcoma-associated herpesvirus (KSHV) participates in the pathogenesis of endothelial sarcoma and lymphomas. EBV and KSHV are responsible for 120,000 and 44,000 annual new cases of cancer, respectively. Despite this clinical importance, no chemotherapies or vaccines have been developed for virus-specific treatment and prevention of these viruses. Humans are the only natural host for both EBV and KSHV, and only a limited species of laboratory animals are susceptible to their experimental infection; this strict host tropism has hampered the development of their animal models and thereby impeded the study of therapeutic and prophylactic strategies. To overcome this difficulty, three main approaches have been used to develop animal models for human gammaherpesvirus infections. The first is experimental infection of laboratory animals with EBV or KSHV. New-world non-human primates (NHPs) and rabbits have been mainly used in this approach. The second is experimental infection of laboratory animals with their own inherent gammaherpesviruses. NHPs and mice have been mainly used here. The third, a recent trend, employs experimental infection of EBV or KSHV or both to immunodeficient mice reconstituted with human immune system components (humanized mice). This review will discuss how these three approaches have been used to reproduce human clinical conditions associated with gammaherpesviruses and to analyze the mechanisms of their pathogenesis.
Collapse
|
29
|
Johnson KE, Tarakanova VL. Gammaherpesviruses and B Cells: A Relationship That Lasts a Lifetime. Viral Immunol 2020; 33:316-326. [PMID: 31913773 DOI: 10.1089/vim.2019.0126] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gammaherpesviruses are highly prevalent pathogens that establish life-long infection and are associated with diverse malignancies, including lymphoproliferative diseases and B cell lymphomas. Unlike other viruses that either do not infect B cells or infect B cells transiently, gammaherpesviruses manipulate physiological B cell differentiation to establish life-long infection in memory B cells. Disruption of such viral manipulation by genetic or environmental causes is likely to seed viral lymphomagenesis. In this review, we discuss physiological and unique host and viral mechanisms usurped by gammaherpesviruses to fine tune host B cell biology for optimal infection establishment and maintenance.
Collapse
Affiliation(s)
- Kaitlin E Johnson
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vera L Tarakanova
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
30
|
The Role of Dendritic Cells in Immune Control and Vaccination against -Herpesviruses. Viruses 2019; 11:v11121125. [PMID: 31817510 PMCID: PMC6950272 DOI: 10.3390/v11121125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/29/2019] [Accepted: 12/04/2019] [Indexed: 12/21/2022] Open
Abstract
The two human oncogenic -herpesviruses, Epstein Barr virus (EBV) and Kaposi sarcoma-associated herpesvirus (KSHV), are prototypic pathogens that are controlled by T cell responses. Despite their ubiquitous distribution, persistent infections and transforming potential, most carriers' immune systems control them for life. Therefore, they serve as paradigms of how near-perfect cell-mediated immune control can be initiated and maintained for decades. Interestingly, EBV especially quite efficiently avoids dendritic cell (DC) activation, and little evidence exists that these most potent antigen-presenting cells of the human body are involved in the priming of immune control against this tumor virus. However, DCs can be harnessed therapeutically to expand virus-specific T cells for adoptive transfer therapy of patients with virus-associated malignancies and are also currently explored for vaccinations. Unfortunately, despite 55 and 25 years of research on EBV and KSHV, respectively, the priming of their immune control that belongs to the most robust and durable immune responses in humans still remains unclear.
Collapse
|
31
|
Radtke S, Humbert O, Kiem HP. Mouse models in hematopoietic stem cell gene therapy and genome editing. Biochem Pharmacol 2019; 174:113692. [PMID: 31705854 DOI: 10.1016/j.bcp.2019.113692] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/01/2019] [Indexed: 12/26/2022]
Abstract
Gene therapy has become an important treatment option for a variety of hematological diseases. The biggest advances have been made with CAR T cells and many of those studies are now FDA approved as a routine treatment for some hematologic malignancies. Hematopoietic stem cell (HSC) gene therapy is not far behind with treatment approvals granted for beta-hemoglobinopathies and adenosine deaminase severe combined immune deficiency (ADA-SCID), and additional approbations currently being sought. With the current pace of research, the significant investment of biotech companies, and the continuously growing toolbox of viral as well as non-viral gene delivery methods, the development of new ex vivo and in vivo gene therapy approaches is at an all-time high. Research in the field of gene therapy has been ongoing for more than 4 decades with big success stories as well as devastating drawbacks along the way. In particular, the damaging effect of uncontrolled viral vector integration observed in the initial gene therapy applications in the 90s led to a more comprehensive upfront safety assessment of treatment strategies. Since the late 90s, an important read-out to comprehensively assess the quality and safety of cell products has come forward with the mouse xenograft model. Here, we review the use of mouse models across the different stages of basic, pre-clinical and translational research towards the clinical application of HSC-mediated gene therapy and editing approaches.
Collapse
Affiliation(s)
- Stefan Radtke
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Olivier Humbert
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Hans-Peter Kiem
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA; Department of Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
32
|
Alisjahbana A, Mohammad I, Gao Y, Evren E, Ringqvist E, Willinger T. Human macrophages and innate lymphoid cells: Tissue-resident innate immunity in humanized mice. Biochem Pharmacol 2019; 174:113672. [PMID: 31634458 DOI: 10.1016/j.bcp.2019.113672] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/15/2019] [Indexed: 12/17/2022]
Abstract
Macrophages and innate lymphoid cells (ILCs) are tissue-resident cells that play important roles in organ homeostasis and tissue immunity. Their intricate relationship with the organs they reside in allows them to quickly respond to perturbations of organ homeostasis and environmental challenges, such as infection and tissue injury. Macrophages and ILCs have been extensively studied in mice, yet important species-specific differences exist regarding innate immunity between humans and mice. Complementary to ex-vivo studies with human cells, humanized mice (i.e. mice with a human immune system) offer the opportunity to study human macrophages and ILCs in vivo within their surrounding tissue microenvironments. In this review, we will discuss how humanized mice have helped gain new knowledge about the basic biology of these cells, as well as their function in infectious and malignant conditions. Furthermore, we will highlight active areas of investigation related to human macrophages and ILCs, such as their cellular heterogeneity, ontogeny, tissue residency, and plasticity. In the near future, we expect more fundamental discoveries in these areas through the combined use of improved humanized mouse models together with state-of-the-art technologies, such as single-cell RNA-sequencing and CRISPR/Cas9 genome editing.
Collapse
Affiliation(s)
- Arlisa Alisjahbana
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Imran Mohammad
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Yu Gao
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Elza Evren
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Emma Ringqvist
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Tim Willinger
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden.
| |
Collapse
|
33
|
Daharsh L, Zhang J, Ramer-Tait A, Li Q. A Double Humanized BLT-mice Model Featuring a Stable Human-Like Gut Microbiome and Human Immune System. J Vis Exp 2019. [PMID: 31524867 DOI: 10.3791/59773] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Humanized mice (hu-mice) that feature a functional human immune system have fundamentally changed the study of human pathogens and disease. They can be used to model diseases that are otherwise difficult or impossible to study in humans or other animal models. The gut microbiome can have a profound impact on human health and disease. However, the murine gut microbiome is very different than the one found in humans. There is a need for improved pre-clinical hu-mice models that have an engrafted human gut microbiome. Therefore, we created double hu-mice that feature both a human immune system and stable human-like gut microbiome. NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice are one of the best animals for humanization due to their high level of immunodeficiency. However, germ-free NSG mice, and various other important germ-free mice models are not currently commercially available. Further, many research settings do not have access to gnotobiotic facilities, and working under gnotobiotic conditions can often be expensive and time consuming. Importantly, germ-free mice have several immune deficiencies that exist even after the engraftment of microbes. Therefore, we developed a protocol that does not require germ-free animals or gnotobiotic facilities. To generate double hu-mice, NSG mice were treated with radiation prior to surgery to create bone-marrow, liver, thymus-humanized (hu-BLT) mice. The mice were then treated with broad spectrum antibiotics to deplete the pre-existing murine gut microbiome. After antibiotic treatment, the mice were given fecal transplants with healthy human donor samples via oral gavage. Double hu-BLT mice had unique 16S rRNA gene profiles based on the individual human donor sample that was transplanted. Importantly, the transplanted human-like microbiome was stable in the double hu-BLT mice for the duration of the study up to 14.5 weeks post-transplant.
Collapse
Affiliation(s)
- Lance Daharsh
- Nebraska Center for Virology; School of Biological Sciences, University of Nebraska-Lincoln
| | - Jianshui Zhang
- Nebraska Center for Virology; School of Biological Sciences, University of Nebraska-Lincoln
| | - Amanda Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln
| | - Qingsheng Li
- Nebraska Center for Virology; School of Biological Sciences, University of Nebraska-Lincoln;
| |
Collapse
|
34
|
Wahl A, De C, Abad Fernandez M, Lenarcic EM, Xu Y, Cockrell AS, Cleary RA, Johnson CE, Schramm NJ, Rank LM, Newsome IG, Vincent HA, Sanders W, Aguilera-Sandoval CR, Boone A, Hildebrand WH, Dayton PA, Baric RS, Pickles RJ, Braunstein M, Moorman NJ, Goonetilleke N, Victor Garcia J. Precision mouse models with expanded tropism for human pathogens. Nat Biotechnol 2019; 37:1163-1173. [PMID: 31451733 PMCID: PMC6776695 DOI: 10.1038/s41587-019-0225-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 07/12/2019] [Indexed: 12/12/2022]
Abstract
A major limitation of current humanized mouse models is that they primarily enable the analysis of human-specific pathogens that infect hematopoietic cells. However, most human pathogens target other cell types, including epithelial, endothelial and mesenchymal cells. Here, we show that implantation of human lung tissue, which contains up to 40 cell types, including nonhematopoietic cells, into immunodeficient mice (lung-only mice) resulted in the development of a highly vascularized lung implant. We demonstrate that emerging and clinically relevant human pathogens such as Middle East respiratory syndrome coronavirus, Zika virus, respiratory syncytial virus and cytomegalovirus replicate in vivo in these lung implants. When incorporated into bone marrow/liver/thymus humanized mice, lung implants are repopulated with autologous human hematopoietic cells. We show robust antigen-specific humoral and T-cell responses following cytomegalovirus infection that control virus replication. Lung-only mice and bone marrow/liver/thymus-lung humanized mice substantially increase the number of human pathogens that can be studied in vivo, facilitating the in vivo testing of therapeutics. Implantation of lung tissue into humanized mice enables in vivo study of the human immune response to pathogens.
Collapse
Affiliation(s)
- Angela Wahl
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA.
| | - Chandrav De
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA
| | - Maria Abad Fernandez
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Erik M Lenarcic
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Yinyan Xu
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Adam S Cockrell
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Rachel A Cleary
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA
| | - Claire E Johnson
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA
| | - Nathaniel J Schramm
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA
| | - Laura M Rank
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Isabel G Newsome
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC, USA
| | - Heather A Vincent
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Wes Sanders
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Christian R Aguilera-Sandoval
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA.,BD Life Sciences, San Jose, CA, USA
| | - Allison Boone
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
| | - William H Hildebrand
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC, USA
| | - Ralph S Baric
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Raymond J Pickles
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Miriam Braunstein
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Nathaniel J Moorman
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Nilu Goonetilleke
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,UNC HIV Cure Center, University of North Carolina, Chapel Hill, NC, USA
| | - J Victor Garcia
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
35
|
Mulama DH, Mutsvunguma LZ, Totonchy J, Ye P, Foley J, Escalante GM, Rodriguez E, Nabiee R, Muniraju M, Wussow F, Barasa AK, Ogembo JG. A multivalent Kaposi sarcoma-associated herpesvirus-like particle vaccine capable of eliciting high titers of neutralizing antibodies in immunized rabbits. Vaccine 2019; 37:4184-4194. [PMID: 31201053 DOI: 10.1016/j.vaccine.2019.04.071] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/19/2019] [Accepted: 04/24/2019] [Indexed: 12/24/2022]
Abstract
Kaposi sarcoma-associated herpesvirus (KSHV) is an emerging pathogen and the causative agent of multiple cancers in immunocompromised patients. To date, there is no licensed prophylactic KSHV vaccine. In this study, we generated a novel subunit vaccine that incorporates four key KSHV envelope glycoproteins required for viral entry in diverse cell types (gpK8.1, gB, and gH/gL) into a single multivalent KSHV-like particle (KSHV-LP). Purified KSHV-LPs were similar in size, shape, and morphology to KSHV virions. Vaccination of rabbits with adjuvanted KSHV-LPs generated strong glycoprotein-specific antibody responses, and purified immunoglobulins from KSHV-LP-immunized rabbits neutralized KSHV infection in epithelial, endothelial, fibroblast, and B cell lines (60-90% at the highest concentration tested). These findings suggest that KSHV-LPs may be an ideal platform for developing a safe and effective prophylactic KSHV vaccine. We envision performing future studies in animal models that are susceptible to KSHV infection, to determine correlates of immune protection in vivo.
Collapse
Affiliation(s)
- David H Mulama
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States; Biological Sciences Department, Masinde Muliro University of Science and Technology, Kakamega, Kenya
| | - Lorraine Z Mutsvunguma
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | | | - Peng Ye
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Joslyn Foley
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Gabriela M Escalante
- Irell & Manella Graduate School of Biological Sciences of City of Hope, Duarte, CA, United States
| | - Esther Rodriguez
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Ramina Nabiee
- Chapman University, School of Pharmacy, Irvine, CA, United States
| | - Murali Muniraju
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Felix Wussow
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Anne K Barasa
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States; Department of Human Pathology, University of Nairobi, Nairobi, Kenya
| | - Javier Gordon Ogembo
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States.
| |
Collapse
|
36
|
Damania B, Münz C. Immunodeficiencies that predispose to pathologies by human oncogenic γ-herpesviruses. FEMS Microbiol Rev 2019; 43:181-192. [PMID: 30649299 PMCID: PMC6435449 DOI: 10.1093/femsre/fuy044] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/30/2018] [Indexed: 12/13/2022] Open
Abstract
Human γ-herpesviruses include the closely related tumor viruses Epstein Barr virus (EBV) and Kaposi sarcoma-associated herpesvirus (KSHV). EBV is the most growth-transforming pathogen known and is linked to at least seven human malignancies. KSHV is also associated with three human cancers. Most EBV- and KSHV-infected individuals fortunately remain disease-free despite persistent infection and this is likely due to the robustness of the immune control that they mount against these tumor viruses. However, upon immune suppression EBV- and KSHV-associated malignancies emerge at increased frequencies. Moreover, primary immunodeficiencies with individual mutations that predispose to EBV or KSHV disease allow us to gain insights into a catalog of molecules that are required for the immune control of these tumor viruses. Curiously, there is little overlap between the mutation targets that predispose individuals to EBV versus KSHV disease, even so both viruses can infect the same host cell, human B cells. These differences will be discussed in this review. A better understanding of the crucial components in the near-perfect life-long immune control of EBV and KSHV should allow us to target malignancies that are associated with these viruses, but also induce similar immune responses against other tumors.
Collapse
Affiliation(s)
- Blossom Damania
- Lineberger Cancer Research Center and Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| |
Collapse
|
37
|
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: 62] [Impact Index Per Article: 10.3] [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
|
38
|
Rhesus Macaque Rhadinovirus Encodes a Viral Interferon Regulatory Factor To Disrupt Promyelocytic Leukemia Nuclear Bodies and Antagonize Type I Interferon Signaling. J Virol 2019; 93:JVI.02147-18. [PMID: 30626678 DOI: 10.1128/jvi.02147-18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 01/03/2019] [Indexed: 12/17/2022] Open
Abstract
Interferon (IFN) production and the subsequent induction of IFN-stimulated genes (ISGs) are highly effective innate strategies utilized by cells to protect against invading pathogens, including viruses. Critical components involved in this innate process are promyelocytic leukemia nuclear bodies (PML-NBs), which are subnuclear structures required for the development of a robust IFN response. As such, PML-NBs serve as an important hurdle for viruses to overcome to successfully establish an infection. Both Kaposi's sarcoma-associated herpesvirus (KSHV) and the closely related rhesus macaque rhadinovirus (RRV) are unique for encoding viral homologs of IFN regulatory factors (termed vIRFs) that can manipulate the host immune response by multiple mechanisms. All four KSHV vIRFs inhibit the induction of IFN, while vIRF1 and vIRF2 can inhibit ISG induction downstream of the IFN receptor. Less is known about the RRV vIRFs. RRV vIRF R6 can inhibit the induction of IFN by IRF3; however, it is not known whether any RRV vIRFs inhibit ISG induction following IFN receptor signaling. In our present study, we demonstrate that the RRV vIRF R12 aids viral replication in the presence of the type I IFN response. This is achieved in part through the disruption of PML-NBs and the inhibition of robust ISG transcription.IMPORTANCE KSHV and RRV encode a unique set of homologs of cellular IFN regulatory factors, termed vIRFs, which are hypothesized to help these viruses evade the innate immune response and establish infections in their respective hosts. Our work elucidates the role of one RRV vIRF, R12, and demonstrates that RRV can dampen the type I IFN response downstream of IFN signaling, which would be important for establishing a successful infection in vivo.
Collapse
|
39
|
LXR Alpha Restricts Gammaherpesvirus Reactivation from Latently Infected Peritoneal Cells. J Virol 2019; 93:JVI.02071-18. [PMID: 30602604 DOI: 10.1128/jvi.02071-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 12/19/2018] [Indexed: 12/28/2022] Open
Abstract
Gammaherpesviruses are ubiquitous viruses that establish lifelong infections. Importantly, these viruses are associated with numerous cancers and lymphoproliferative diseases. While risk factors for developing gammaherpesvirus-driven cancers are poorly understood, it is clear that elevated viral reactivation from latency often precedes oncogenesis. Here, we demonstrate that the liver X receptor alpha isoform (LXRα) restricts gammaherpesvirus reactivation in an anatomic-site-specific manner. We have previously demonstrated that deficiency of both LXR isoforms (α and β) leads to an increase in fatty acid and cholesterol synthesis in primary macrophage cultures, with a corresponding increase in gammaherpesvirus replication. Interestingly, expression of fatty acid synthesis genes was not derepressed in LXRα-deficient hosts, indicating that the antiviral effects of LXRα are independent of lipogenesis. Additionally, the critical host defenses against gammaherpesvirus reactivation, virus-specific CD8+ T cells and interferon (IFN) signaling, remained intact in the absence of LXRα. Remarkably, using a murine gammaherpesvirus 68 (MHV68) reporter virus, we discovered that LXRα expression dictates the cellular tropism of MHV68 in the peritoneal cavity. Specifically, LXRα-/- mice exhibit reduced latency within the peritoneal B cell compartment and elevated latency within F4/80+ cells. Thus, LXRα restricts gammaherpesvirus reactivation through a novel mechanism that is independent of the known CD8+ T cell-based antiviral responses or changes in lipid synthesis and likely involves changes in the tropism of MHV68 in the peritoneal cavity.IMPORTANCE Liver X receptors (LXRs) are nuclear receptors that mediate cholesterol and fatty acid homeostasis. Importantly, as ligand-activated transcription factors, LXRs represent potential targets for the treatment of hypercholesterolemia and atherosclerosis. Here, we demonstrate that LXRα, one of the two LXR isoforms, restricts reactivation of latent gammaherpesvirus from peritoneal cells. As gammaherpesviruses are ubiquitous oncogenic agents, LXRs may represent a targetable host factor for the treatment of poorly controlled gammaherpesvirus infection and associated lymphomagenesis.
Collapse
|
40
|
Mandal S, Kang G, Prathipati PK, Zhou Y, Fan W, Li Q, Destache CJ. Nanoencapsulation introduces long-acting phenomenon to tenofovir alafenamide and emtricitabine drug combination: A comparative pre-exposure prophylaxis efficacy study against HIV-1 vaginal transmission. J Control Release 2018; 294:216-225. [PMID: 30576746 DOI: 10.1016/j.jconrel.2018.12.027] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/20/2018] [Accepted: 12/15/2018] [Indexed: 02/08/2023]
Abstract
Daily oral antiretroviral (ARV) drugs for pre-exposure prophylaxis (PrEP) has proven efficacy for diverse groups of high-risk individuals. However, daily dosing regimen has augmented non-adherence. These experiments comparatively investigated the long-acting (LA) PrEP potency of subcutaneous (SubQ) administrated tenofovir alafenamide (TAF) and emtricitabine (FTC) loaded nanoparticles (NPs) to solution in humanized (hu) mice. TAF + FTC NPs and TAF + FTC solution (each drug at 200 mg/kg) were administered to hu-CD34-NSG mice (n = 3/time point) for plasma and tissue pharmacokinetic parameter estimation using LC-MS/MS. NP enhanced tissue ARV assimilation compared to plasma. The same dose was administered for PrEP efficacy in HIV-1 challenged hu-BLT mice (n = 5/group). The hu-BLT mice were vaginally challenged with a transmission-founder (T/F) virus at 5 × 105 TCID50 inoculation, on day 4, 7 and 14 post-SubQ treatments (PT) and were compared to infected-untreated-control hu-BLT mice. By 21 days PT, 100% TAF + FTC solution-treated and control-untreated mice were infected. However, TAF + FTC NPs resulted in significant (p = .0002) protection from HIV-1 (day 4: 80%, day 7 and 14: 60%, respectively) compared to control mice. This proof-of-concept study demonstrated detectable TAF/FTC vaginal levels among TAF + FTC NP-treated hu-BLT mice correlating with prolonged PrEP efficacy, thus establishing long-acting TAF + FTC NPs as a potential PrEP modality.
Collapse
Affiliation(s)
- Subhra Mandal
- Creighton University School of Pharmacy & Health Professions, Omaha, NE, United States.
| | - Guobin Kang
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | | | - You Zhou
- Nebraska Center for Virology and Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Wenjin Fan
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | | |
Collapse
|
41
|
Bruce AG, Barcy S, Staheli J, Bielefeldt-Ohmann H, Ikoma M, Howard K, Rose TM. Experimental co-transmission of Simian Immunodeficiency Virus (SIV) and the macaque homologs of the Kaposi Sarcoma-Associated Herpesvirus (KSHV) and Epstein-Barr Virus (EBV). PLoS One 2018; 13:e0205632. [PMID: 30444879 PMCID: PMC6239284 DOI: 10.1371/journal.pone.0205632] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/02/2018] [Indexed: 12/29/2022] Open
Abstract
Macaque RFHV and LCV are close homologs of human KSHV and EBV, respectively. No experimental model of RFHV has been developed due to the lack of a source of culturable infectious virus. Screening of macaques at the Washington National Primate Research Center detected RFHV in saliva of SIV-infected macaques from previous vaccine studies. A pilot experimental infection of two naïve juvenile pig-tailed macaques was initiated by inoculation of saliva from SIV-infected pig-tailed and cynomolgus macaque donors, which contained high levels of DNA (> 10(6) genomes/ml) of the respective species-specific RFHV strain. Both juvenile recipients developed SIV and RFHV infections with RFHV DNA detected transiently in saliva and/or PBMC around week 16 post-infection. One juvenile macaque was infected with the homologous RFHVMn from whole saliva of a pig-tailed donor, which had been inoculated into the cheek pouch. This animal became immunosuppressed, developing simian AIDS and was euthanized 23 weeks after inoculation. The levels of RFHV DNA in saliva and PBMC remained below the level of detection after week 17, showing no reactivation of the RFHVMn infection during the rapid development of AIDS. The other juvenile macaque was infected with the heterologous RFHVMf from i.v. inoculation of purified virions from saliva of a cynomolgus donor. The juvenile recipient remained immunocompetent, developing high levels of persistent anti-RFHV and -SIV antibodies. After the initial presence of RFHVMf DNA in saliva and PBMC decreased to undetectable levels by week 19, all attempts to reactivate the infection through additional inoculations, experimental infection with purified SRV-2 or SIV, or immunosuppressive treatments with cyclosporine or dexamethasone were unsuccessful. An heterologous LCV transmission was also detected in this recipient, characterized by continual high levels of LCVMf DNA from the cynomolgus donor in both saliva (> 10(6) genomes/ml) and PBMC (> 10(4) genomes/million cells), coupled with high levels of anti-LCV antibodies. The macaque was sacrificed 209 weeks after the initial inoculation. Low levels of LCVMf DNA were detected in salivary glands, tonsils and other lymphoid organs, while RFHVMf DNA was below the level of detection. These results show successful co-transmission of RFHV and LCV from saliva and demonstrate differential lytic activation of the different gammaherpesvirus lineages due to presumed differences in biology and tropism and control by the host immune system. Although this initial pilot transmission study utilized only two macaques, it provides the first evidence for experimental transmission of the macaque homolog of KSHV, setting the stage for larger transmission studies to examine the differential activation of rhadinovirus and lymphocryptovirus infections and the pathological effects of immunosuppression.
Collapse
Affiliation(s)
- A. Gregory Bruce
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Department of Pathobiology, University of Washington, Seattle, Washington, United States of America
| | - Serge Barcy
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Jeannette Staheli
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Department of Pathobiology, University of Washington, Seattle, Washington, United States of America
| | - Helle Bielefeldt-Ohmann
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Minako Ikoma
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Kellie Howard
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Department of Pathobiology, University of Washington, Seattle, Washington, United States of America
| | - Timothy M. Rose
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Department of Pathobiology, University of Washington, Seattle, Washington, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
42
|
Samal J, Kelly S, Na-Shatal A, Elhakiem A, Das A, Ding M, Sanyal A, Gupta P, Melody K, Roland B, Ahmed W, Zakir A, Bility M. Human immunodeficiency virus infection induces lymphoid fibrosis in the BM-liver-thymus-spleen humanized mouse model. JCI Insight 2018; 3:120430. [PMID: 30232273 DOI: 10.1172/jci.insight.120430] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 08/07/2018] [Indexed: 12/17/2022] Open
Abstract
A major pathogenic feature associated with HIV infection is lymphoid fibrosis, which persists during antiretroviral therapy (ART). Lymphoid tissues play critical roles in the generation of antigen-specific immune response, and fibrosis disrupts the stromal network of lymphoid tissues, resulting in impaired immune cell trafficking and function, as well as immunodeficiency. Developing an animal model for investigating the impact of HIV infection-induced lymphoid tissue fibrosis on immunodeficiency and immune cell impairment is critical for therapeutics development and clinical translation. Said model will enable in vivo mechanistic studies, thus complementing the well-established surrogate model of SIV infection-induced lymphoid tissue fibrosis in macaques. We developed a potentially novel human immune system-humanized mouse model by coengrafting autologous fetal thymus, spleen, and liver organoids under the kidney capsule, along with i.v. injection of autologous fetal liver-derived hematopoietic stem cells, thus termed the BM-liver-thymus-spleen (BLTS) humanized mouse model. BLTS humanized mouse model supports development of human immune cells and human lymphoid organoids (human thymus and spleen organoids). HIV infection in BLTS humanized mice results in progressive fibrosis in human lymphoid tissues, which was associated with immunodeficiency in the lymphoid tissues, and lymphoid tissue fibrosis persists during ART, thus recapitulating clinical outcomes.
Collapse
|
43
|
Ko A, Kang G, Hattler JB, Galadima HI, Zhang J, Li Q, Kim WK. Macrophages but not Astrocytes Harbor HIV DNA in the Brains of HIV-1-Infected Aviremic Individuals on Suppressive Antiretroviral Therapy. J Neuroimmune Pharmacol 2018; 14:110-119. [PMID: 30194646 PMCID: PMC6391194 DOI: 10.1007/s11481-018-9809-2] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/30/2018] [Indexed: 11/28/2022]
Abstract
The question of whether the human brain is an anatomical site of persistent HIV-1 infection during suppressive antiretroviral therapy (ART) is critical, but remains unanswered. The presence of virus in the brains of HIV patients whose viral load is effectively suppressed would demonstrate not only the potential for CNS to act as an anatomical HIV reservoir, but also the urgent need to understand the factors contributing to persistent HIV behind the blood-brain barrier. Here, we investigated for the first time the presence of cells harboring HIV DNA and RNA in the brains from subjects with undetectable plasma viral load and sustained viral suppression, as identified by the National NeuroAIDS Tissue Consortium. Using new, highly sensitive in situ hybridization techniques, RNAscope and DNAscope, in combination with immunohistochemistry, we were able to detect HIV-1 in the brains of all virally suppressed cases and found that brain macrophages and microglia, but not astrocytes, were the cells harboring HIV DNA in the brain. This study demonstrated that HIV reservoirs persist in brain macrophages/microglia during suppressive ART, which cure/treatment strategies will need to focus on targeting.
Collapse
Affiliation(s)
- Allen Ko
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Guobin Kang
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Julian B Hattler
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Hadiza I Galadima
- Graduate Program in Public Health, Eastern Virginia Medical School, Norfolk, VA, USA.,School of Community and Environmental Health, College of Health Sciences, Old Dominion University, Norfolk, VA, USA
| | - Junfeng Zhang
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA.,Department of Human Anatomy, Xi'an Medical University, Shaanxi, China
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA.
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, USA.
| |
Collapse
|
44
|
Tso FY, Kang G, Kwon EH, Julius P, Li Q, West JT, Wood C. Brain is a potential sanctuary for subtype C HIV-1 irrespective of ART treatment outcome. PLoS One 2018; 13:e0201325. [PMID: 30040863 PMCID: PMC6057662 DOI: 10.1371/journal.pone.0201325] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 07/12/2018] [Indexed: 12/11/2022] Open
Abstract
Subtype C HIV-1 is responsible for the largest proportion of people living with HIV-1 infection. However, there is limited information about the roles of the brain and its cell types as a potential sanctuary for this subtype and how the sanctuary may be affected by the administration of anti-retroviral therapy (ART). To address this issue, we collected postmortem brain tissues from ART treated HIV-1 infected Zambian individuals who experienced complete viral suppression and those who did not. Tissues from various brain compartments were collected from each individual as frozen and formalin-fixed paraffin embedded brain specimens, for detection and quantification of HIV-1 genomes and identification of the infected cell type. Genomic DNA and RNA were extracted from frozen brain tissues. The extracted DNA and RNA were then subjected to droplet digital PCR for HIV-1 quantification. RNA/DNAscope in situ hybridization (ISH) for HIV-1 was performed on formalin-fixed paraffin embedded brain tissues in conjugation with immunohistochemistry to identify the infected cell types. Droplet digital PCR revealed that HIV-1 gag DNA and RNA were detectable in half of the cases studied regardless of ART success or failure. The presence of HIV-1 lacked specific tissue compartmentalization since detection was random among various brain tissues. When combined with immunohistochemistry, RNA/DNAscope ISH demonstrated co-localization of HIV-1 DNA with CD68 expressing cells indicative of microglia or peripheral macrophage. Our study showed that brain is a potential sanctuary for subtype C HIV-1, as HIV-1 can be detected in the brain of infected individuals irrespective of ART treatment outcome and no compartmentalization of HIV-1 to specific brain compartments was evident.
Collapse
Affiliation(s)
- For Yue Tso
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Nebraska, United States of America
| | - Guobin Kang
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Nebraska, United States of America
| | - Eun Hee Kwon
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Nebraska, United States of America
| | - Peter Julius
- Department of Pathology and Microbiology, University Teaching Hospital, Nationalist Road, Lusaka, Zambia
| | - Qingsheng Li
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Nebraska, United States of America
| | - John T. West
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Nebraska, United States of America
| | - Charles Wood
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
45
|
Mandal S, Kang G, Prathipati PK, Fan W, Li Q, Destache CJ. Long-acting parenteral combination antiretroviral loaded nano-drug delivery system to treat chronic HIV-1 infection: A humanized mouse model study. Antiviral Res 2018; 156:85-91. [PMID: 29885378 DOI: 10.1016/j.antiviral.2018.06.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/02/2018] [Accepted: 06/05/2018] [Indexed: 12/14/2022]
Abstract
Human immunodeficiency virus (HIV) patients are often diagnosed in the chronic stage of HIV/AIDS. Combination antiretroviral therapy (cART) has improved quality of life for HIV-infected patients. Present study describes a novel long-acting parenteral formulation of combination antiretroviral (cARV) loaded nano-drugs for treating chronic HIV-1 (cHIV) in a humanized-BLT (hu-BLT) mice model. The cARV (elvitegravir+tenofovir alafenamide+emtricitabine; EVG+TAF+FTC) drugs (mimicking marketed Genvoya® one-pill for HIV-treatment) were encapsulated in poly (lactic-co-glycolic acid) nanoparticles (NPs). To establish cHIV, hu-BLT mice were intravaginally challenged with HIV-1 and maintained for 15 weeks. Plasma viral load (pVL) was monitored by RT-PCR to confirm cHIV. Baseline pVL (week 15) was comparable between treated (n = 10) and control (n = 5) mice groups. Subsequently, treatment hu-BLT mice received 3 subcutaneous doses of cARV NPs (417 mg/kg per dose; n = 10), biweekly, and a fourth/terminal dose a week later. Prior to each treatment and on sacrifice (week 24), pVL was determined. Within three subcutaneous doses of cARV NPs, a non-detectable pVL was established (week 19) and continued until week 22. After the establishment of a non-detectable pVL (week 19-22), 4 treated-mice were sacrificed for tissue drug concentration determination by LC-MS/MS analysis. A considerable amount of cARV was detected at the HIV-infection target and reservoir organs. Subsequently, pVL rebounded comparable to control group by week 24, (7 weeks post-terminal dosage). The present study demonstrated cARV NPs augments sustained ARV efficacy in the cHIV humanized-mouse model. Therefore, cARV NPs could be a novel delivery system to treat cHIV patients, by overcoming drawbacks of conventional cART.
Collapse
Affiliation(s)
- Subhra Mandal
- School of Pharmacy & Health Professions, Creighton University, Omaha, NE 68178, USA.
| | - Guobin Kang
- Center for Virology, University of Nebraska-Lincoln, 4240 Fair St, Lincoln, NE 68583, USA
| | | | - Wenjin Fan
- Center for Virology, University of Nebraska-Lincoln, 4240 Fair St, Lincoln, NE 68583, USA
| | - Qingsheng Li
- Center for Virology, University of Nebraska-Lincoln, 4240 Fair St, Lincoln, NE 68583, USA
| | - Christopher J Destache
- School of Pharmacy & Health Professions, Creighton University, Omaha, NE 68178, USA; School of Medicine, Division of Infectious Diseases, Creighton University, Omaha, NE 68178, USA
| |
Collapse
|
46
|
Barasa AK, Ye P, Phelps M, Arivudainambi GT, Tison T, Ogembo JG. BALB/c mice immunized with a combination of virus-like particles incorporating Kaposi sarcoma-associated herpesvirus (KSHV) envelope glycoproteins gpK8.1, gB, and gH/gL induced comparable serum neutralizing antibody activity to UV-inactivated KSHV. Oncotarget 2018; 8:34481-34497. [PMID: 28404899 PMCID: PMC5470984 DOI: 10.18632/oncotarget.15605] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 01/16/2017] [Indexed: 12/17/2022] Open
Abstract
Infection with Kaposi sarcoma-associated herpesvirus (KSHV) is estimated to account for over 44,000 new cases of Kaposi sarcoma annually, with 84% occurring in Africa, where the virus is endemic. To date, there is no prophylactic vaccine against KSHV. KSHV gpK8.1, gB, and gH/gL glycoproteins, implicated in the virus entry into host cells, are attractive vaccine targets for eliciting potent neutralizing antibodies (nAbs) against virus infection. We incorporated gpK8.1, gB, or gH/gL on the surface of virus-like particles (VLPs) and characterized these VLPs for their composition, size, and functionality. To determine which viral glycoprotein(s) elicit the most effective serum-nAbs, we immunized BALB/c mice with gpK8.1, gB, or gH/gL VLPs individually or in combination. Neutralizing antibody assay revealed that sera from mice immunized with the VLPs inhibited KSHV infection of HEK-293 cells in a dose-dependent manner. As a single immunogen, gpK8.1 VLPs stimulated comparable nAb activity to that of UV-inactivated KSHV (UV-KSHV). In contrast, UV-KSHV stimulated higher titers of nAb compared to gB (p = 0.0316) or gH/gL (p = 0.0486). Mice immunized with the combination of gB and gH/gL VLPs had a better nAb response than those immunized with either gB (p = 0.0268), or gH/gL (p = 0.0397) as single VLP immunogens. Immunization with any VLP combination stimulated comparable nAb activity to UV-KSHV serum. Our data provide the first evidence that KSHV gpK8.1, gB, and gH/gL glycoproteins can be incorporated onto the surface of VLPs and used as prophylactic vaccine candidates, with potential to prevent KSHV infection.
Collapse
Affiliation(s)
- Anne K Barasa
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA.,Department of Human Pathology, University of Nairobi, Nairobi, Kenya
| | - Peng Ye
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Meredith Phelps
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Timelia Tison
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Javier Gordon Ogembo
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA.,Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
47
|
Skelton JK, Ortega-Prieto AM, Dorner M. A Hitchhiker's guide to humanized mice: new pathways to studying viral infections. Immunology 2018; 154:50-61. [PMID: 29446074 PMCID: PMC5904706 DOI: 10.1111/imm.12906] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/09/2018] [Accepted: 02/09/2018] [Indexed: 12/14/2022] Open
Abstract
Humanized mice are increasingly appreciated as an incredibly powerful platform for infectious disease research. The often very narrow species tropism of many viral infections, coupled with the sometimes misleading results from preclinical studies in animal models further emphasize the need for more predictive model systems based on human cells rather than surrogates. Humanized mice represent such a model and have been greatly enhanced with regards to their immune system reconstitution as well as immune functionality in the past years, resulting in their recommendation as a preclinical model by the US Food and Drug Administration. This review aims to give a detailed summary of the generation of human peripheral blood lymphocyte-, CD34+ haematopoietic stem cell- and bone marrow/liver/thymus-reconstituted mice and available improved models (e.g. myeloid- or T-cell-only mice, MISTRG, NSG-SGM3). Additionally, we summarize human-tropic viral infections, for which humanized mice offer a novel approach for the study of disease pathogenesis as well as future perspectives for their use in biomedical, drug and vaccine research.
Collapse
Affiliation(s)
- Jessica Katy Skelton
- Section of Virology, Department of Medicine, Imperial College London, London, UK
| | | | - Marcus Dorner
- Section of Virology, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
48
|
Abstract
KSHV is the etiologic agent of PEL-an aggressive lymphoma. Interestingly, EBV concurrently exists in nearly 70% of PEL cases. In this issue of Cell Host & Microbe, McHugh et al. (2017) develop humanized mouse models for EBV/KSHV co-infection and identify their complementary effect on in vivo tumor formation.
Collapse
|
49
|
McHugh D, Caduff N, Barros MHM, Rämer PC, Raykova A, Murer A, Landtwing V, Quast I, Styles CT, Spohn M, Fowotade A, Delecluse HJ, Papoudou-Bai A, Lee YM, Kim JM, Middeldorp J, Schulz TF, Cesarman E, Zbinden A, Capaul R, White RE, Allday MJ, Niedobitek G, Blackbourn DJ, Grundhoff A, Münz C. Persistent KSHV Infection Increases EBV-Associated Tumor Formation In Vivo via Enhanced EBV Lytic Gene Expression. Cell Host Microbe 2018; 22:61-73.e7. [PMID: 28704654 DOI: 10.1016/j.chom.2017.06.009] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 05/09/2017] [Accepted: 06/20/2017] [Indexed: 11/15/2022]
Abstract
The human tumor viruses Epstein-Barr virus (EBV) and Kaposi sarcoma-associated herpesvirus (KSHV) establish persistent infections in B cells. KSHV is linked to primary effusion lymphoma (PEL), and 90% of PELs also contain EBV. Studies on persistent KSHV infection in vivo and the role of EBV co-infection in PEL development have been hampered by the absence of small animal models. We developed mice reconstituted with human immune system components as a model for KSHV infection and find that EBV/KSHV dual infection enhanced KSHV persistence and tumorigenesis. Dual-infected cells displayed a plasma cell-like gene expression pattern similar to PELs. KSHV persisted in EBV-transformed B cells and was associated with lytic EBV gene expression, resulting in increased tumor formation. Evidence of elevated lytic EBV replication was also found in EBV/KSHV dually infected lymphoproliferative disorders in humans. Our data suggest that KSHV augments EBV-associated tumorigenesis via stimulation of lytic EBV replication.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/virology
- Cell Line, Tumor
- Coinfection
- Cytokines/blood
- DNA, Viral/analysis
- Disease Models, Animal
- Epstein-Barr Virus Infections/blood
- Epstein-Barr Virus Infections/immunology
- Epstein-Barr Virus Infections/virology
- Gene Expression Regulation, Viral
- Genes, Viral/genetics
- Herpesviridae Infections/blood
- Herpesviridae Infections/immunology
- Herpesviridae Infections/virology
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/pathogenicity
- Herpesvirus 8, Human/genetics
- Herpesvirus 8, Human/pathogenicity
- Herpesvirus 8, Human/physiology
- High-Throughput Nucleotide Sequencing
- Humans
- Lymphoma, Primary Effusion/etiology
- Lymphoma, Primary Effusion/virology
- Mice
- Neoplasms/virology
- Spleen/pathology
- Spleen/virology
- Survival Rate
- Virus Replication
Collapse
Affiliation(s)
- Donal McHugh
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Nicole Caduff
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | | | - Patrick C Rämer
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Ana Raykova
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Anita Murer
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Vanessa Landtwing
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Isaak Quast
- Neuroinflammation, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Christine T Styles
- Section of Virology, Faculty of Medicine, Imperial College London, London, UK
| | - Michael Spohn
- Virus Genomics, Heinrich Pette Institute, Hamburg, Germany
| | - Adeola Fowotade
- School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | | | | | - Yong-Moon Lee
- Departments of Pathology and Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jin-Man Kim
- Departments of Pathology and Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jaap Middeldorp
- Department of Pathology, VU University Medical Center and Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Thomas F Schulz
- Institute of Virology, Hannover Medical School, Hannover and German Centre of Infection Research (DZIF), Hannover-Braunschweig Site, Germany
| | - Ethel Cesarman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Andrea Zbinden
- Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Riccarda Capaul
- Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Robert E White
- Section of Virology, Faculty of Medicine, Imperial College London, London, UK
| | - Martin J Allday
- Section of Virology, Faculty of Medicine, Imperial College London, London, UK
| | | | | | - Adam Grundhoff
- Virus Genomics, Heinrich Pette Institute, Hamburg, Germany
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
50
|
Münz C. Human γ-Herpesvirus Infection, Tumorigenesis, and Immune Control in Mice with Reconstituted Human Immune System Components. Front Immunol 2018; 9:238. [PMID: 29483919 PMCID: PMC5816265 DOI: 10.3389/fimmu.2018.00238] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/29/2018] [Indexed: 12/12/2022] Open
Abstract
The human γ-herpesviruses Epstein–Barr virus (EBV or HHV4) and Kaposi sarcoma-associated herpesvirus (KSHV or HHV8) are each associated with around 2% of all tumors in humans worldwide. However, investigations into their infection, oncogenesis, and immune responses that protect from the associated tumors have been hampered by the exclusive tropism of these pathogens for humans. Mice with reconstituted human immune system components (HIS mice) provide the unique opportunity to study persistent infection, virus associated lymphoma formation, and cell-mediated immune control of EBV and KSHV. Moreover, since these pathogens are unique stimuli for cytotoxic human lymphocyte responses, they also allow us to characterize long-lasting cell-mediated immune control and the requirements for its initiation, which would also be desirable to achieve during antitumor vaccination in general. Thus, human γ-herpesvirus infection of HIS mice provides unique insights into the biology of these important human pathogens and human cell-mediated immune responses that are considered to be the main protective entity against tumors.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|