1
|
Chu PY, Hsu CL, Lin YA, Pan YC, Dai YH, Yu YC, Yang JC, Ma WL, Chen YJL, Lee CL, Wu YC. Effects of Citrus depressa Hayata juice on high-fat diet-induced obesity in HBV transgenic mice. Heliyon 2024; 10:e24438. [PMID: 38312542 PMCID: PMC10835261 DOI: 10.1016/j.heliyon.2024.e24438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/20/2023] [Accepted: 01/09/2024] [Indexed: 02/06/2024] Open
Abstract
The present study investigated the potential anti-obesity properties of Citrus depressa Hayata (CDH) juice in HBV transgenic mice, as well as the impact of fermentation on the effectiveness of the juice. The results revealed that fermentation increased the levels of polyphenols and hesperidin in CDH juice. The animal study demonstrated that both juices were effective in mitigating the weight gain induced by a high-fat diet by correcting metabolic parameter imbalances, reducing hepatic lipid accumulation, and reversing hepatic immune suppression. Furthermore, fermented juice exhibited superior efficacy in managing body weight and inhibiting the expansion of white adipose tissue (WAT). Fermented juice significantly enhanced adiponectin production and PPARγ expression in WAT, while also reducing hypertrophy. This study offers valuable insights into the potential role of CDH juices in combating obesity associated with high fat consumption and underscores the promise of CDH juice as a functional beverage.
Collapse
Affiliation(s)
- Pei-Yi Chu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
| | - Chang-Lu Hsu
- Department of Business Administration, National Chiayi University, Chiayi, Taiwan
| | - Yen-An Lin
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Yi-Cheng Pan
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
- .Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan
| | - Yun-Hao Dai
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Ying-Chun Yu
- Department of Medical Research, and Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan
| | - Juan-Cheng Yang
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Wen-Lung Ma
- Department of Medical Research, and Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan
| | | | - Chia-Lin Lee
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
- Department of Cosmeceutics, China Medical University, Taichung 40604, Taiwan
| | - Yang-Chang Wu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
2
|
Protocol for chronic hepatitis B virus infection mouse model development by patient-derived orthotopic xenografts. PLoS One 2022; 17:e0264266. [PMID: 35196351 PMCID: PMC8865695 DOI: 10.1371/journal.pone.0264266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 02/01/2022] [Indexed: 12/03/2022] Open
Abstract
Background According to the World Health Organization, more than 250 million people worldwide are chronically infected with the hepatitis B virus, and almost 800.000 patients die annually of mediated liver disorders. Therefore, adequate biological test systems are needed that could fully simulate the course of chronic hepatitis B virus infection, including in patients with hepatocellular carcinoma. Methods In this study, we will assess the effectiveness of existing protocols for isolation and cultivation of primary cells derived from patients with hepatocellular carcinoma in terms of the yield of viable cells and their ability to replicate the hepatitis B virus using isolation and cultivation methods for adhesive primary cells, flow cytometry and quantitative polymerase chain reaction. Another part of our study will be devoted to evaluating the effectiveness of hepatocellular carcinoma grafting methods to obtain patient-derived heterotopic and orthotopic xenograft mouse avatars using animal X-ray irradiation and surgery procedures and in vivo fluorescent signals visualization and measurements. Our study will be completed by histological methods. Discussion This will be the first extensive comparative study of the main modern methods and protocols for isolation and cultivation primary hepatocellular carcinoma cells and tumor engraftment to the mice. All protocols will be optimized and characterized using the: (1) efficiency of the method for isolation cells from removed hepatocellular carcinoma in terms of their quantity and viability; (2) efficiency of the primary cell cultivation protocol in terms of the rate of monolayer formation and hepatitis B virus replication; (3) efficiency of the grafting method in terms of the growth rate and the possibility of hepatitis B virus persistence and replication in mice. The most effective methods will be recommended for use in translational biomedical research.
Collapse
|
3
|
Kruse RL, Barzi M, Legras X, Pankowicz FP, Furey N, Liao L, Xu J, Bissig-Choisat B, Slagle BL, Bissig KD. A hepatitis B virus transgenic mouse model with a conditional, recombinant, episomal genome. JHEP Rep 2021; 3:100252. [PMID: 33733079 PMCID: PMC7940981 DOI: 10.1016/j.jhepr.2021.100252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 12/27/2020] [Accepted: 01/04/2021] [Indexed: 01/21/2023] Open
Abstract
Background & Aims Development of new and more effective therapies against hepatitis B virus (HBV) is limited by the lack of suitable small animal models. The HBV transgenic mouse model containing an integrated overlength 1.3-mer construct has yielded crucial insights, but this model unfortunately lacks covalently closed circular DNA (cccDNA), the episomal HBV transcriptional template, and cannot be cured given that HBV is integrated in every cell. Methods To solve these 2 problems, we generated a novel transgenic mouse (HBV1.1X), which generates an excisable circular HBV genome using Cre/LoxP technology. This model possesses a HBV1.1-mer cassette knocked into the ROSA26 locus and is designed for stable expression of viral proteins from birth, like the current HBV transgenic mouse model, before genomic excision with the introduction of Cre recombinase. Results We demonstrated induction of recombinant cccDNA (rcccDNA) formation via viral or transgenic Cre expression in HBV1.1X mice, and the ability to regulate HBsAg and HBc expression with Cre in mice. Tamoxifen-inducible Cre could markedly downregulate baseline HBsAg levels from the integrated HBV genome. To demonstrate clearance of HBV from HBV1.1X mice, we administered adenovirus expressing Cre, which permanently and significantly reduced HBsAg and core antigen levels in the murine liver via rcccDNA excision and a subsequent immune response. Conclusions The HBV1.1X model is the first Cre-regulatable HBV transgenic mouse model and should be of value to mimic chronic HBV infection, with neonatal expression and tolerance of HBV antigens, and on-demand modulation of HBV expression. Lay summary Hepatitis B virus (HBV) can only naturally infect humans and chimpanzees. Mouse models have been developed with the HBV genome integrated into mouse chromosomes, but this prevents mice from being cured. We developed a new transgenic mouse model that allows for HBV to be excised from mouse chromosomes to form a recombinant circular DNA molecule resembling the natural circular HBV genome. HBV expression could be reduced in these mice, enabling curative therapies to be tested in this new mouse model.
Collapse
Affiliation(s)
- Robert L Kruse
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA.,Center for Stem Cells and Regenerative Medicine, Baylor College of Medicine, Houston, TX, USA.,Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX, USA.,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Mercedes Barzi
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC, USA.,Y.T. and Alice Chen Pediatric Genetics and Genomics Research Center, Duke University, Durham, NC, USA
| | - Xavier Legras
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC, USA.,Y.T. and Alice Chen Pediatric Genetics and Genomics Research Center, Duke University, Durham, NC, USA
| | - Francis P Pankowicz
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA.,Center for Stem Cells and Regenerative Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Nika Furey
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA.,Center for Stem Cells and Regenerative Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC, USA.,Y.T. and Alice Chen Pediatric Genetics and Genomics Research Center, Duke University, Durham, NC, USA
| | - Lan Liao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Janming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Beatrice Bissig-Choisat
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA.,Center for Stem Cells and Regenerative Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC, USA.,Y.T. and Alice Chen Pediatric Genetics and Genomics Research Center, Duke University, Durham, NC, USA
| | - Betty L Slagle
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Karl-Dimiter Bissig
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA.,Center for Stem Cells and Regenerative Medicine, Baylor College of Medicine, Houston, TX, USA.,Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX, USA.,Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC, USA.,Y.T. and Alice Chen Pediatric Genetics and Genomics Research Center, Duke University, Durham, NC, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Duke Center for Virology, Duke University, Durham, NC, USA.,Duke Cancer Institute, Duke University, Durham, NC, USA
| |
Collapse
|
4
|
Burwitz BJ, Zhou Z, Li W. Animal models for the study of human hepatitis B and D virus infection: New insights and progress. Antiviral Res 2020; 182:104898. [PMID: 32758525 DOI: 10.1016/j.antiviral.2020.104898] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/09/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022]
Abstract
Hepatitis B virus (HBV) is a member of the Hepadnaviridae family and infects hepatocytes, leading to liver pathology in acutely and chronically infected individuals. Co-infection with Hepatitis D virus (HDV), which requires the surface proteins of HBV to replicate, can exacerbate this disease progression. Thus, the >250 million people living with chronic HBV infection, including 13 million co-infected with HDV, would significantly benefit from an effective and affordable curative treatment. Animal models are crucial to the development of innovative disease therapies, a paradigm repeated again and again throughout the fields of immunology, neurology, reproduction, and development. Unfortunately, HBV has a highly-restricted species tropism, infecting limited species including humans, chimpanzees, and treeshrews. The first experimentally controlled studies of HBV infection were following inoculation of human volunteers in 1942, which identified the transmissibility of hepatitis through serum transfer and led to the hypothesis that the etiological agent was viral. Subsequent research in chimpanzees (Desmyter et al., 1971; Lichter, 1969) and later in other species, such as the treeshrews (Walter et al., 1996; Yan et al., 1996), further confirmed the viral origin of hepatitis B. Shortly thereafter, HBV-like viral infections were identified in woodchucks (Summers et al., 1978; Werner et al., 1979) and ducks, and much of our understanding of HBV replication can be attributed to these important models. However, with the exodus of chimpanzees from research and the limited reagents and historical data for treeshrews and other understudied species, there remains an urgent need to identify physiologically relevant models of chronic HBV infection. While large strides have been made in generating such models, particularly over the past two decades, there is still no available model that faithfully recapitulates the immunity and pathogenesis of HBV infection. Here, we discuss recent advancements in the generation of murine and non-human primate (NHP) models of HBV/HDV infection.
Collapse
Affiliation(s)
- Benjamin J Burwitz
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, 97006, USA.
| | - Zhongmin Zhou
- College of Life Sciences, Beijing Normal University, Beijing, 100875, China; National Institute of Biological Sciences, Beijing, 102206, China.
| | - Wenhui Li
- National Institute of Biological Sciences, Beijing, 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China.
| |
Collapse
|
5
|
Nucleic acid vaccines for hepatitis B and C virus. INFECTION GENETICS AND EVOLUTION 2019; 75:103968. [PMID: 31325609 DOI: 10.1016/j.meegid.2019.103968] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 05/25/2019] [Accepted: 07/15/2019] [Indexed: 02/07/2023]
Abstract
Hepatitis B virus (HBV) and Hepatitis C virus (HCV) infections accounts for an important global health problem affecting over 250 million people all around the world. They can cause acute, transient and chronic infections in the human liver. Chronic infection of liver can lead to its failure or cancer. To deal with this problem, alternative approaches or strategies to inhibit these infections have already been started. DNA and mRNA-based vaccination will increase the efficacy and reduce toxicity in patients with Hepatitis B virus (HBV) and Hepatitis C virus (HCV) infections. Gene vaccines represent a promising alternative to conventional vaccine approaches because of their high potency, capacity for rapid development, low-cost manufacture and safe administration. MRNA-based vaccination is a method to elicit potent antigen-specific humoral and cell-mediated immune responses with a superior safety profile compared with DNA vaccines. Exploring the intricacies of these pathways can potentially help the researchers to explore newer vaccines. In this study, DNA and mRNA-based vaccination are introduced as an approach to treat Hepatitis B virus (HBV) and Hepatitis C virus (HCV) infections. DNA and mRNA-based vaccines as one of the most successful therapeutics are introduced and the clinical outcomes of their exploitation are explained.
Collapse
|
6
|
Non-alcoholic hepatic steatosis attenuates hepatitis B virus replication in an HBV-immunocompetent mouse model. Hepatol Int 2018; 12:438-446. [DOI: 10.1007/s12072-018-9877-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/01/2018] [Indexed: 12/23/2022]
|
7
|
Yi X, Yuan Y, Li N, Yi L, Wang C, Qi Y, Gong L, Liu G, Kong X. A mouse model with age-dependent immune response and immune-tolerance for HBV infection. Vaccine 2018; 36:794-801. [PMID: 29306503 DOI: 10.1016/j.vaccine.2017.12.071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 12/10/2017] [Accepted: 12/27/2017] [Indexed: 01/04/2023]
Abstract
BACKGROUND Viral clearance of human HBV infection largely depends on the age of exposure. Thus, a mouse model with age-dependent immune response and immune-tolerance for HBV infection was established. METHODS HBVRag1 mice were generated by crossing Rag1-/- mice with HBV-Tg mice. Following adoptive transfer of splenocytes adult (8-9 weeks old) and young (3 weeks old) HBVRag1 mice were named as HBVRag-ReA and HBVRag-ReY mice respectively. The biochemical parameters that were associated with viral load and immune function, as well as the histological evaluation of the liver tissues between the two mouse models were detected. The immune tolerance of HBVRag-ReY mice that were reconstituted at the early stages of life was evaluated by quantitative hepatitis B core antibody assay, adoptive transfer, and modulation of gut microbiota with the addition of antibiotics. RESULTS HBVRag-ReA mice indicated apparent hepatocytes damage, clearance of HBsAg and production of HBsAb and HBcAb. HBVRag-ReY mice did not develop ALT elevation, and produced HBcAb and HBsAg. A higher number of hepatic CD8+ T and B cells promoted clearance of HBsAg in HBVRag-ReA mice following 30 days of lymphocyte transfer. In contrast to HBVRag-ReA mice, HBVRag-ReY mice exhibited higher levels of Th1/Th2 cytokines. HBVRag-ReY mice exhibited significantly higher (P < .01, approximately 10-fold) serum quantitative anti-HBc levels than HBV-Tg mice, which might be similar to the phase of immune clearance and immune tolerance in human HBV infection. Furthermore, the age-related tolerance in HBVRag-ReY mice that were sensitive to antibiotic treatment was different from that noted in HBV-Tg mice. GS-9620 could inhibit the production of HBsAg, whereas HBV vaccination could induce sustained seroconversion in HBVRag-ReY mice with low levels of HBsAg. CONCLUSIONS The present study described a mouse model with age-dependent immunity and immune-tolerance for HBV infection in vivo, which may mimic chronic HBV infection in humans.
Collapse
Affiliation(s)
- Xuerui Yi
- Liver Diseases Research Center, The 458th Hospital of Guangzhou, Guangzhou 510600, China.
| | - Youcheng Yuan
- Liver Diseases Research Center, The 458th Hospital of Guangzhou, Guangzhou 510600, China
| | - Na Li
- Liver Diseases Research Center, The 458th Hospital of Guangzhou, Guangzhou 510600, China
| | - Lu Yi
- Liver Diseases Research Center, The 458th Hospital of Guangzhou, Guangzhou 510600, China
| | - Cuiling Wang
- Liver Diseases Research Center, The 458th Hospital of Guangzhou, Guangzhou 510600, China
| | - Ying Qi
- Liver Diseases Research Center, The 458th Hospital of Guangzhou, Guangzhou 510600, China
| | - Liang Gong
- Liver Diseases Research Center, The 458th Hospital of Guangzhou, Guangzhou 510600, China
| | - Guangze Liu
- Liver Diseases Research Center, The 458th Hospital of Guangzhou, Guangzhou 510600, China
| | - Xiangping Kong
- Liver Diseases Research Center, The 458th Hospital of Guangzhou, Guangzhou 510600, China
| |
Collapse
|
8
|
Zhao W, Zhou X, Zhao G, Lin Q, Wang X, Yu X, Wang B. Enrichment of Ly6C hi monocytes by multiple GM-CSF injections with HBV vaccine contributes to viral clearance in a HBV mouse model. Hum Vaccin Immunother 2017; 13:2872-2882. [PMID: 28699816 PMCID: PMC5718782 DOI: 10.1080/21645515.2017.1344797] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Adjuvants are considered a necessary component for HBV therapeutic vaccines but few are licensed in clinical practice due to concerns about safety or efficiency. In our recent study, we established that a combination protocol of 3-day pretreatments with GM-CSF before a vaccination (3 × GM-CSF+VACCINE) into the same injection site could break immune tolerance and cause over 90% reduction of HBsAg level in the HBsAg transgenic mouse model. Herein, we further investigated the therapeutic potential of the combination in AAV8–1.3HBV-infected mice. After 4 vaccinations, both serum HBeAg and HBsAg were cleared and there was a 95% reduction of HBV-positive hepatocytes, in addition to the presence of large number of infiltrating CD8+ T cells in the livers. Mechanistically, the HBV-specific T-cell responses were elicited via a 3 × GM-CSF+VACCINE-induced conversion of CCR2-dependent CD11b+ Ly6Chi monocytes into CD11b+CD11c+ DCs. Experimental depletion of Ly6Chi monocytes resulted in a defective HBV-specific immune response thereby abrogating HBV eradication. This vaccination strategy could lead to development of an effective therapeutic protocol against chronic HBV in infected patients.
Collapse
Affiliation(s)
- Weidong Zhao
- a Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences , Fudan University , Shanghai , China
| | - Xian Zhou
- a Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences , Fudan University , Shanghai , China
| | - Gan Zhao
- a Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences , Fudan University , Shanghai , China
| | - Qing Lin
- a Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences , Fudan University , Shanghai , China
| | - Xianzheng Wang
- a Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences , Fudan University , Shanghai , China
| | - Xueping Yu
- b Department of Infectious Diseases, Huashan Hospital , Fudan University , Shanghai , China
| | - Bin Wang
- a Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences , Fudan University , Shanghai , China
| |
Collapse
|
9
|
Dandri M, Petersen J. Animal models of HBV infection. Best Pract Res Clin Gastroenterol 2017; 31:273-279. [PMID: 28774409 DOI: 10.1016/j.bpg.2017.04.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 04/28/2017] [Indexed: 01/31/2023]
Abstract
The mechanisms determining hepatitis B virus (HBV) persistence and pathogenesis are not fully elucidated, but appear to be multi-factorial. Current medication to repress viral replication is available; however, the unique replication strategies employed by HBV enable the virus to persist within the infected hepatocytes. Consequently, cure is rarely achieved. Progresses in HBV research and preclinical testing of antiviral agents have been limited by the narrow species- and tissue-tropism of the virus, the paucity of infection models available and the restrictions imposed by the use of chimpanzees, the only animals fully susceptible to HBV infection. Mice are not HBV permissive but major efforts have focused on the development of mouse models of HBV replication and infection, such as the generation of humanized mice. By presenting the different animal models available, this review will highlight the most important and clinically relevant findings that have been retrieved from the respective systems.
Collapse
Affiliation(s)
- Maura Dandri
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Infection Research (DZIF) Hamburg-Lübeck-Borstel Site, Germany.
| | - Joerg Petersen
- IFI Institute for Interdisciplinary Medicine at Asklepios Clinic St. Georg, Hamburg, Germany
| |
Collapse
|
10
|
Sang X, Wang R, Han Y, Zhang C, Shen H, Yang Z, Xiong Y, Liu H, Liu S, Li R, Yang R, Wang J, Wang X, Bai Z, Xiao X. T cell--associated immunoregulation and antiviral effect of oxymatrine in hydrodynamic injection HBV mouse model. Acta Pharm Sin B 2017; 7:311-318. [PMID: 28540167 PMCID: PMC5430867 DOI: 10.1016/j.apsb.2017.02.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/05/2017] [Accepted: 01/18/2017] [Indexed: 01/05/2023] Open
Abstract
Although oxymatrine (OMT) has been shown to directly inhibit the replication of hepatitis B virus (HBV) in vitro, limited research has been done with this drug in vivo. In the present study, the antiviral effect of OMT was investigated in an immunocompetent mouse model of chronic HBV infection. The infection was achieved by tail vein injection of a large volume of DNA solution. OMT (2.2, 6.7 and 20 mg/kg) was administered by daily intraperitoneal injection for 6 weeks. The efficacy of OMT was evaluated by the levels of HBV DNA, hepatitis B surface antigen (HBsAg), hepatitis B e antigen (HBeAg) and hepatitis B core antigen (HBcAg). The immunoregulatory activity of OMT was evaluated by serum ELISA and flow cytometry. Results shows that OMT at 20 mg/kg inhibited HBV replication, and it was more efficient than entecavir (ETV) in the elimination of serum HBsAg and intrahepatic HBcAg. In addition, OMT accelerated the production of interferon-γ (IFN-γ) in a dose-dependent manner in CD4+ T cells. Our findings demonstrate the beneficial effects of OMT on the enhancement of immunological function and in the control of HBV antigens. The findings suggest this drug to be a good antiviral therapeutic candidate for the treatment of HBV infection.
Collapse
Key Words
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- CD4+ T cell
- CHB, chronic hepatitis B
- ETV, entecavir
- HBV
- HBV, hepatitis B virus
- HBcAg, hepatitis B core antigen
- HBeAg, hepatitis B e antigen
- HBsAg, hepatitis B surface antigen
- HE, hematoxylin and eosin
- IFN-γ
- IFN-γ, interferon-γ
- IL-4, interleukin-4
- Mouse
- NAs, nucleoside and nucleotide analogs
- OMT, oxymatrine
- Oxymatrine
- TCMs, traditional Chinese medicines
- TNF-α, tumor necrosis factor-α
Collapse
|
11
|
Li J, Bao M, Ge J, Ren S, Zhou T, Qi F, Pu X, Dou J. Research progress of therapeutic vaccines for treating chronic hepatitis B. Hum Vaccin Immunother 2017; 13:986-997. [PMID: 28118084 DOI: 10.1080/21645515.2016.1276125] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hepatitis B virus (HBV) is a member of Hepadnavirus family, which leads to chronic infection in around 5% of patients with a high risk of developing liver cirrhosis, liver failure, and hepatocellular carcinoma. 1 Despite the availability of prophylactic vaccines against hepatitis B for over 3 decades, there are still more than 2 billion people have been infected and 240 million of them were chronic. Antiviral therapies currently used in the treatment of CHB (chronic hepatitis B) infection include peg-interferon, standard α-interferon and nucleos/tide analogs (NAs), but none of them can provide sustained control of viral replication. As an alternative strategy, therapeutic vaccines for CHB patients have been widely studied and showed some promising efficacies in dozens of preclinical and clinical trials. In this article, we review current research progress in several types of therapeutic vaccines for CHB treatment, including protein-based vaccines, DNA-based vaccines, live vector-based vaccines, peptide-based vaccines and cell-based therapies. These researches may provide some clues for developing new treatments in CHB infection.
Collapse
Affiliation(s)
- Jianqiang Li
- a Jiangsu Theravac Bio-pharmaceutical Co., Ltd. , Nanjing , China
| | - Mengru Bao
- a Jiangsu Theravac Bio-pharmaceutical Co., Ltd. , Nanjing , China
| | - Jun Ge
- a Jiangsu Theravac Bio-pharmaceutical Co., Ltd. , Nanjing , China
| | - Sulin Ren
- a Jiangsu Theravac Bio-pharmaceutical Co., Ltd. , Nanjing , China
| | - Tong Zhou
- a Jiangsu Theravac Bio-pharmaceutical Co., Ltd. , Nanjing , China
| | - Fengchun Qi
- a Jiangsu Theravac Bio-pharmaceutical Co., Ltd. , Nanjing , China
| | - Xiuying Pu
- b School of Life Science and Engineering, Lanzhou University of Technology , Lanzhou , China
| | - Jia Dou
- c Dalian Institute for Drug Control , Dalian , China
| |
Collapse
|
12
|
Liu Y, Wang B, Wang L, Vikash V, Wang Q, Roggendorf M, Lu M, Yang D, Liu J. Transcriptome Analysis and Comparison of Marmota monax and Marmota himalayana. PLoS One 2016; 11:e0165875. [PMID: 27806133 PMCID: PMC5091844 DOI: 10.1371/journal.pone.0165875] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 10/19/2016] [Indexed: 12/12/2022] Open
Abstract
The Eastern woodchuck (Marmota monax) is a classical animal model for studying hepatitis B virus (HBV) infection and hepatocellular carcinoma (HCC) in humans. Recently, we found that Marmota himalayana, an Asian animal species closely related to Marmota monax, is susceptible to woodchuck hepatitis virus (WHV) infection and can be used as a new mammalian model for HBV infection. However, the lack of genomic sequence information of both Marmota models strongly limited their application breadth and depth. To address this major obstacle of the Marmota models, we utilized Illumina RNA-Seq technology to sequence the cDNA libraries of liver and spleen samples of two Marmota monax and four Marmota himalayana. In total, over 13 billion nucleotide bases were sequenced and approximately 1.5 billion clean reads were obtained. Following assembly, 106,496 consensus sequences of Marmota monax and 78,483 consensus sequences of Marmota himalayana were detected. For functional annotation, in total 73,603 Unigenes of Marmota monax and 78,483 Unigenes of Marmota himalayana were identified using different databases (NR, NT, Swiss-Prot, KEGG, COG, GO). The Unigenes were aligned by blastx to protein databases to decide the coding DNA sequences (CDS) and in total 41,247 CDS of Marmota monax and 34,033 CDS of Marmota himalayana were predicted. The single nucleotide polymorphisms (SNPs) and the simple sequence repeats (SSRs) were also analyzed for all Unigenes obtained. Moreover, a large-scale transcriptome comparison was performed and revealed a high similarity in transcriptome sequences between the two marmota species. Our study provides an extensive amount of novel sequence information for Marmota monax and Marmota himalayana. This information may serve as a valuable genomics resource for further molecular, developmental and comparative evolutionary studies, as well as for the identification and characterization of functional genes that are involved in WHV infection and HCC development in the woodchuck model.
Collapse
Affiliation(s)
- Yanan Liu
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baoju Wang
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Wang
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Vikash Vikash
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Wang
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Michael Roggendorf
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Mengji Lu
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Dongliang Yang
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail: (DY); (JL)
| | - Jia Liu
- Department of Infectious Diseases, Institute of Infection and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail: (DY); (JL)
| |
Collapse
|
13
|
Hirai-Yuki A, Hensley L, McGivern DR, González-López O, Das A, Feng H, Sun L, Wilson JE, Hu F, Feng Z, Lovell W, Misumi I, Ting JPY, Montgomery S, Cullen J, Whitmire JK, Lemon SM. MAVS-dependent host species range and pathogenicity of human hepatitis A virus. Science 2016; 353:1541-1545. [PMID: 27633528 PMCID: PMC5068972 DOI: 10.1126/science.aaf8325] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/02/2016] [Indexed: 12/24/2022]
Abstract
Hepatotropic viruses are important causes of human disease, but the intrahepatic immune response to hepatitis viruses is poorly understood because of a lack of tractable small- animal models. We describe a murine model of hepatitis A virus (HAV) infection that recapitulates critical features of type A hepatitis in humans. We demonstrate that the capacity of HAV to evade MAVS-mediated type I interferon responses defines its host species range. HAV-induced liver injury was associated with interferon-independent intrinsic hepatocellular apoptosis and hepatic inflammation that unexpectedly resulted from MAVS and IRF3/7 signaling. This murine model thus reveals a previously undefined link between innate immune responses to virus infection and acute liver injury, providing a new paradigm for viral pathogenesis in the liver.
Collapse
Affiliation(s)
- Asuka Hirai-Yuki
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Lucinda Hensley
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - David R McGivern
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Medicine, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Olga González-López
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Anshuman Das
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Hui Feng
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Lu Sun
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Justin E Wilson
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Genetics, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Fengyu Hu
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Zongdi Feng
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA
| | - William Lovell
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Ichiro Misumi
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Genetics, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Jenny P-Y Ting
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Genetics, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Stephanie Montgomery
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27517, USA
| | - John Cullen
- Department of Population Health and Pathobiology, North Carolina State University College of Veterinary Medicine, Raleigh, NC 27607, USA
| | - Jason K Whitmire
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Genetics, University of North Carolina, Chapel Hill, NC 27517, USA
| | - Stanley M Lemon
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27517, USA. Department of Medicine, University of North Carolina, Chapel Hill, NC 27517, USA.
| |
Collapse
|
14
|
Experimental in vitro and in vivo models for the study of human hepatitis B virus infection. J Hepatol 2016; 64:S17-S31. [PMID: 27084033 DOI: 10.1016/j.jhep.2016.02.012] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/29/2016] [Accepted: 02/10/2016] [Indexed: 12/13/2022]
Abstract
Chronic infection with the hepatitis B virus (HBV) affects an estimate of 240 million people worldwide despite the availability of a preventive vaccine. Medication to repress viral replication is available but a cure is rarely achieved. The narrow species and tissue tropism of the virus and the lack of reliable in vitro models and laboratory animals susceptible to HBV infection, have limited research progress in the past. As a result, several aspects of the HBV life cycle as well as the network of virus host interactions occurring during the infection are not yet understood. Only recently, the identification of the functional cellular receptor enabling HBV entry has opened new possibilities to establish innovative infection systems. Regarding the in vivo models of HBV infection, the classical reference was the chimpanzee. However, because of the strongly restricted use of great apes for HBV research, major efforts have focused on the development of mouse models of HBV replication and infection such as the generation of humanized mice. This review summarizes the animal and cell culture based models currently available for the study of HBV biology. We will discuss the benefits and caveats of each model and present a selection of the most important findings that have been retrieved from the respective systems.
Collapse
|
15
|
Fibronectin EDA and CpG synergize to enhance antigen-specific Th1 and cytotoxic responses. Vaccine 2016; 34:2453-2459. [PMID: 27016652 PMCID: PMC4859824 DOI: 10.1016/j.vaccine.2016.03.057] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 01/28/2016] [Accepted: 03/17/2016] [Indexed: 12/15/2022]
Abstract
FNIII 11-EDA and CpG synergize in vitro to enhance activation of dendritic cells. Immunization with both adjuvants induces a potent antigen-specific Th1 response in vivo. Co-adjuvanted OVA mediates regression of E.G7-OVA tumors through CTL response. Co-adjuvanted HBsAg induces seroconversion and clearance of circulating virus in HBV-Tg mice.
Subunit vaccines, employing purified protein antigens rather than intact pathogens, require the addition of adjuvants for enhanced immunogenicity with a correct balance between strong activation of the immune system and low toxicity. Here we show that the endogenous (i.e., autologous) non-toxic TLR4 agonist extra domain A type III repeat of fibronectin (FNIII EDA) can synergize with the exogenous (i.e., bacterial), toxic-at-high-dose, TLR9 agonist CpG to induce efficient cellular immune responses while keeping the dose of CpG low. The efficacy of the combined TLR agonists, even at half-doses, led to stronger dendritic cell activation, enhanced cytotoxic T lymphocyte activation as well as stronger humoral response, compared to the individual agonists given at full doses. Immune cells induced after vaccination with the co-adjuvanted formulation could mediate tumor regression in an E.G7-OVA tumor model, and eradicate circulating hepatitis B virus (HBV) in a transgenic HBV model. Together, these results show that endogenous TLR agonists, such as variants of FNIII EDA, can synergize with exogenous TLR ligands, such as CpG, and strongly enhance cellular immune responses, while improving their safety profile.
Collapse
|