1
|
Jeong PS, Yang HJ, Park YH, Jin YB, Song BS, Hong JJ, Lee SH, Lee JH, Lim KS, Jeong KJ, Kang P, Lee HY, Son HC, Kim HN, Ha SM, Hwang EH, Cha JJ, Jung Y, Choi SA, Lee S, Lee SR, Lee SC, Kang KS, Hur CG, Jung YW, Koo DB, Choo YK, Kim JM, Sim BW, Kim SU. JAK3-deficient mini-pigs exhibit impaired lymphoid organogenesis, intestinal structure, and leukocyte/cytokine production. J Adv Res 2025:S2090-1232(25)00280-2. [PMID: 40280544 DOI: 10.1016/j.jare.2025.04.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 01/09/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION Severe combined immunodeficiency (SCID) mini-pigs are a highly versatile model for human disease research and regenerative medicine. OBJECTIVES This study aims to generate a novel JAK3-deficient mini-pig model with a human-like immune system and to elucidate how JAK3 plays an important role in immune system. METHODS JAK3 and RAG2 knockout (KO) mini-pigs were generated using CRISPR/Cas9 and somatic cell nuclear transfer. These mini-pigs were transferred to a sterilized isolator within a specific pathogen-free facility. Phenotypic characteristics and clinical manifestations were analyzed through histological and hematological analysis of SCID mini-pigs to explore the unique role of JAK3 in immune functions. RESULTS JAK3 KO was characterized by defects in T and NK cells, very low levels of B cells, and a complete absence of thymus and lymph nodes. Notably, JAK3 KO mini-pigs had significantly reduced numbers of monocytes in peripheral blood, macrophages in tissue, and inflammatory cytokines, suggesting that JAK3 KO can induce a broad immunodeficiency that extends to the myeloid system as well as the lymphoid. Moreover, JAK3 KO mini-pigs had intestinal abnormalities similar to those of patients. CONCLUSION These results suggest that JAK3 KO mini-pigs can be used as an effective model for the development of therapies for SCID patients, as well as for regenerative medicine applications such as the development of patient-specific artificial organs.
Collapse
Affiliation(s)
- Pil-Soo Jeong
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Biotechnology, Daegu University, Gyeongsan, Republic of Korea
| | - Hae-Jun Yang
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Biological Science, College of National Sciences, Wonkwang University, Iksan, Republic of Korea
| | - Young-Ho Park
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Yeung Bae Jin
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Bong-Seok Song
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Jung Joo Hong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Seung Hwan Lee
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Jong-Hee Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Kyung Seob Lim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Kang-Jin Jeong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Philyong Kang
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Hwal-Yong Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Hee-Chang Son
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Han-Na Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Seung-Min Ha
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Eun-Ha Hwang
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Jae-Jin Cha
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Yena Jung
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Research Institute, huMetaCELL Inc., Bucheon, Republic of Korea
| | - Seon-A Choi
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Companion Animals, Chungcheong University, Cheongju, Republic of Korea
| | - Sanghoon Lee
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sang-Rae Lee
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Seung-Chan Lee
- Bio Division, APURES Inc., Pyeongtaek, Republic of Korea
| | - Kyung Soo Kang
- Bio Division, APURES Inc., Pyeongtaek, Republic of Korea; Department of Bio Life Sciences, Shingu College, Seongnam, Republic of Korea
| | - Chang-Gi Hur
- Bio Division, APURES Inc., Pyeongtaek, Republic of Korea
| | - Yong Woo Jung
- College of Pharmacy, Korea University, Sejong, Republic of Korea
| | - Deog-Bon Koo
- Department of Biotechnology, Daegu University, Gyeongsan, Republic of Korea
| | - Young-Kug Choo
- Department of Biological Science, College of National Sciences, Wonkwang University, Iksan, Republic of Korea
| | - Jin-Man Kim
- Department of Pathology, Cancer Research Institute and Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Bo-Woong Sim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea.
| | - Sun-Uk Kim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea.
| |
Collapse
|
2
|
Sefik E, Xiao T, Chiorazzi M, Odell I, Zhang F, Agrawal K, Micevic G, Flavell RA. Engineering Mice to Study Human Immunity. Annu Rev Immunol 2025; 43:451-487. [PMID: 40020225 DOI: 10.1146/annurev-immunol-082523-124415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Humanized mice, which carry a human hematopoietic and immune system, have greatly advanced our understanding of human immune responses and immunological diseases. These mice are created via the transplantation of human hematopoietic stem and progenitor cells into immunocompromised murine hosts further engineered to support human hematopoiesis and immune cell growth. This article explores genetic modifications in mice that enhance xeno-tolerance, promote human hematopoiesis and immunity, and enable xenotransplantation of human tissues with resident immune cells. We also discuss genetic editing of the human immune system, provide examples of how humanized mice with humanized organs model diseases for mechanistic studies, and highlight the roles of these models in advancing knowledge of organ biology, immune responses to pathogens, and preclinical drugs tested for cancer treatment. The integration of multi-omics and state-of-the art approaches with humanized mouse models is crucial for bridging existing human data with causality and promises to significantly advance mechanistic studies.
Collapse
Affiliation(s)
- Esen Sefik
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
| | - Tianli Xiao
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut, USA
| | - Michael Chiorazzi
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ian Odell
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Fengrui Zhang
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut, USA
| | - Kriti Agrawal
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Computational Biology and Bioinformatics Program, Yale University, New Haven, Connecticut, USA
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Goran Micevic
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
3
|
Wang C, Zai W, Zhao K, Li Y, Shi B, Wu M, Zhou X, Kozlowski M, Zhang X, Fang Z, Yuan Z. Potential role of liver-resident CD3 + macrophages in HBV clearance in a mouse hepatitis B model. JHEP Rep 2025; 7:101323. [PMID: 40143948 PMCID: PMC11937660 DOI: 10.1016/j.jhepr.2024.101323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 03/28/2025] Open
Abstract
Background & Aims Chronic HBV infection usually causes cirrhosis and hepatocellular carcinoma. Comparative investigations of acute and chronic HBV cases would help determine the immune responses crucial for viral clearance. Methods A fast-cleared HBV mouse model was established in Alb-Cre mice via hydrodynamic injection of HBV plasmid, while persistent HBV model mice were generated via recombinant covalently closed circular DNA-adeno-associated virus 8 infection. The single-cell transcriptomes of CD45+ intrahepatic non-parenchymal cells from these mice were conducted. Multiplexed immunohistochemistry and flow cytometry were used to confirm the findings from single-cell transcriptomes. Transwell, coculture, and adoptive transfer experiments were performed to study the generation and functions of macrophages. Results Twenty-four clusters of immune cells were identified. Myeloid cells, including granulocytes, monocytes, and dendritic cells, are activated early in HBV fast-cleared mice. Significantly, a cluster of CD3+ macrophages was found in the viral clearance phase, which was confirmed in liver tissue from five acute patients with HBV. These cells highly expressed CXCL1, tumor necrosis factor alpha, and HBsAg-specific T cell receptors. The transwell assay revealed that CD3+ macrophages originate from macrophages (n = 6). T cells and anti-HBsAg antibodies are indispensable for their differentiation, which was further confirmed in T- and/or B-cell-deficient mice. Interestingly, these CD3+ macrophages capable of killing peptide-loaded hepatocytes and engulfing IgG-coated beads were persistently detectable in the mouse liver for 10 weeks after HBV clearance. The expression levels of CD5L and Bcl2, two classical antiapoptotic proteins, increased (p <0.001), suggesting that the CD3+ macrophages are long-term resident populations. Finally, adoptive transfer of CD3+ macrophages accelerated HBV clearance in mice (n = 5, p <0.01). Conclusions We identified long-term polyfunctional CD3+ macrophages residing in HBV fast-cleared livers that could help elucidate the immune responses involved in eliminating HBV. Impact and implications The liver is a special organ with unique immune characteristics and tolerance to foodborne antigens. Chronic infections can develop in newborns after exposure to HBV; however, acute infections usually occur in adults, indicating that immune cells in the liver tissue microenvironment can also effectively fight against the virus. Nevertheless, the mechanisms involved in acute HBV infection have rarely been studied. In this study, we identified a macrophage population with both T cell and macrophage characteristics in the livers of acute HBV model mice and revealed that these macrophages play important roles in HBV clearance. Moreover, we confirmed that this population is derived from macrophages in the presence of virus-specific T cells and antibodies. This finding highlights the complexity of antiviral immune responses in liver microenvironments.
Collapse
Affiliation(s)
- Cong Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Wenjing Zai
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kuangjie Zhao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yaming Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bisheng Shi
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Min Wu
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaohui Zhou
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Maya Kozlowski
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaonan Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Zhong Fang
- Liver Cancer Institute of Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Craft K, Amanor A, Barnett I, Donaldson C, Anegon I, Madduri S, Tang Q, Bility MT. Can Humanized Immune System Mouse and Rat Models Accelerate the Development of Cytomegalovirus-Based Vaccines Against Infectious Diseases and Cancers? Int J Mol Sci 2025; 26:3082. [PMID: 40243710 PMCID: PMC11988357 DOI: 10.3390/ijms26073082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/20/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
Over the past three decades, immunodeficient mouse models carrying human immune cells, with or without human lymphoid tissues, termed humanized immune system (HIS) rodent models, have been developed to recapitulate the human immune system and associated immune responses. HIS mouse models have successfully modeled many human-restricted viral infections, including those caused by human cytomegalovirus (HCMV) and human immunodeficiency virus (HIV). HIS mouse models have also been used to model human cancer immunobiology, which exhibits differences from murine cancers in traditional mouse models. Variants of HIS mouse models that carry human liver cells, lung tissue, skin tissue, or human patient-derived tumor xenografts and human hematopoietic stem cells-derived-human immune cells with or without lymphoid tissue xenografts have been developed to probe human immune responses to infections and human tumors. HCMV-based vaccines are human-restricted, which poses limitations for mechanistic and efficacy studies using traditional animal models. The HCMV-based vaccine approach is a promising vaccine strategy as it induces robust effector memory T cell responses that may be critical in preventing and rapidly controlling persistent viral infections and cancers. Here, we review novel HIS mouse models with robust human immune cell development and primary and secondary lymphoid tissues that could address many of the limitations of HIS mice in their use as animal models for HCMV-based vaccine research. We also reviewed novel HIS rat models, which could allow long-term (greater than one year) vaccinology studies and better recapitulate human pathophysiology. Translating laboratory research findings to clinical application is a significant bottleneck in vaccine development; HIS rodents and related variants that more accurately model human immunology and diseases could increase the translatability of research findings.
Collapse
Affiliation(s)
- Kaci Craft
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (K.C.); (A.A.); (I.B.); (C.D.); (Q.T.)
| | - Athina Amanor
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (K.C.); (A.A.); (I.B.); (C.D.); (Q.T.)
| | - Ian Barnett
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (K.C.); (A.A.); (I.B.); (C.D.); (Q.T.)
| | - Clarke Donaldson
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (K.C.); (A.A.); (I.B.); (C.D.); (Q.T.)
| | - Ignacio Anegon
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France;
| | - Srinivas Madduri
- Bioengineering and Neuroregeneration Laboratory, Department of Surgery, University of Geneva, 1211 Geneva, Switzerland;
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (K.C.); (A.A.); (I.B.); (C.D.); (Q.T.)
| | - Moses T. Bility
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (K.C.); (A.A.); (I.B.); (C.D.); (Q.T.)
| |
Collapse
|
5
|
Liu Z, Li G, Li X, Wang Y, Liao L, Yang T, Han C, Huang K, Chen C, Li X, Liu H, Zhang X. CD163 impairs HBV clearance in mice by regulating intrahepatic T cell immune response via an IL-10-dependent mechanism. Antiviral Res 2025; 235:106093. [PMID: 39855274 DOI: 10.1016/j.antiviral.2025.106093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/05/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND & AIMS Chronic hepatitis B (CHB) arises from a persistent hepatitis B virus (HBV) infection, complicating efforts for a functional cure. Kupffer cells (KCs), liver-resident macrophages, are pivotal in mediating immune tolerance to HBV. Although CD163 marks M2-polarized KCs, its precise role in HBV infection remains unclear and warrants further investigation. METHODS CD163 expression in liver tissues of patients with CHB was analyzed using the Gene Expression Omnibus (GEO) database. Cd163 knockout mice were utilized to establish HBV-persistent mouse model, and CD163 deficiency effect on HBV viral markers and T cell immune responses were examined in vivo and in vitro. RESULTS CD163 expression was elevated and correlated with ALT levels in the liver of patients with CHB. In HBV-persistent mouse model, CD163 deficiency facilitated the clearance of HBsAg, HBeAg, HBV DNA, and HBcAg. Additionally, CD163 deficiency promoted the differentiation of naïve T cells into HBV-specific effector T cells. Further, we found that CD163 deficiency reduces KCs-derived IL-10 secretion, and blocking IL-10 further strengthens the enhanced HBV-specific T cell response due to CD163 deficiency. CONCLUSIONS Our findings indicate that CD163 deficiency enhances the HBV-specific T cell response, thereby facilitating HBV clearance through reducing KCs-derived IL-10 secretion. This suggests that CD163 may serve as a potential target for the restoration of exhausted T cell function.
Collapse
MESH Headings
- Animals
- CD163 Antigen
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/immunology
- Antigens, Differentiation, Myelomonocytic/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/metabolism
- Hepatitis B virus/immunology
- Interleukin-10/immunology
- Interleukin-10/metabolism
- Mice
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Mice, Knockout
- Hepatitis B, Chronic/immunology
- Hepatitis B, Chronic/virology
- T-Lymphocytes/immunology
- Humans
- Disease Models, Animal
- Liver/immunology
- Liver/virology
- Kupffer Cells/immunology
- Male
- Female
- Mice, Inbred C57BL
- Hepatitis B Surface Antigens
Collapse
Affiliation(s)
- Ziying Liu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Guiping Li
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Xiaoran Li
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Yiran Wang
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Leyi Liao
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ti Yang
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Chao Han
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Kuiyuan Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Chuyuan Chen
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Xuanyi Li
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Hongyan Liu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Xiaoyong Zhang
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China.
| |
Collapse
|
6
|
Tang X, Zhang W, Zhang Z. Developing T Cell Epitope-Based Vaccines Against Infection: Challenging but Worthwhile. Vaccines (Basel) 2025; 13:135. [PMID: 40006681 PMCID: PMC11861332 DOI: 10.3390/vaccines13020135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
T cell epitope-based vaccines are designed to elicit long-lived pathogen-specific memory T cells that can quickly activate protective effector functions in response to subsequent infections. These vaccines have the potential to provide sustained protection against mutated variants, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which are increasingly capable of evading neutralizing antibodies. Recent advancements in epitope discovery, T cell receptor analysis, and bioinformatics have enabled the precise selection of epitopes and the sophisticated design of epitope-based vaccines. This review outlines the development process for T cell epitope-based vaccines. We summarize the current progress in T cell epitope discovery technologies, highlighting the advantages and disadvantages of each method. We also examine advancements in the design and optimization of epitope-based vaccines, particularly through bioinformatics tools. Additionally, we discuss the challenges of validating the accurate processing and presentation of individual epitopes and establishing suitable rodent models to evaluate vaccine immunogenicity and protective efficacy.
Collapse
Affiliation(s)
- Xian Tang
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen 518112, China; (X.T.); (W.Z.)
| | - Wei Zhang
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen 518112, China; (X.T.); (W.Z.)
| | - Zheng Zhang
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen 518112, China; (X.T.); (W.Z.)
- Guangdong Key Laboratory for Anti-Infection Drug Quality Evaluation, Shenzhen 518112, China
| |
Collapse
|
7
|
Durazzo M, Ferro A, Navarro-Tableros VM, Gaido A, Fornengo P, Altruda F, Romagnoli R, Moestrup SK, Calvo PL, Fagoonee S. Current Treatment Regimens and Promising Molecular Therapies for Chronic Hepatobiliary Diseases. Biomolecules 2025; 15:121. [PMID: 39858515 PMCID: PMC11763965 DOI: 10.3390/biom15010121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/06/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Chronic hepatobiliary damage progressively leads to fibrosis, which may evolve into cirrhosis and/or hepatocellular carcinoma. The fight against the increasing incidence of liver-related morbidity and mortality is challenged by a lack of clinically validated early-stage biomarkers and the limited availability of effective anti-fibrotic therapies. Current research is focused on uncovering the pathogenetic mechanisms that drive liver fibrosis. Drugs targeting molecular pathways involved in chronic hepatobiliary diseases, such as inflammation, hepatic stellate cell activation and proliferation, and extracellular matrix production, are being developed. Etiology-specific treatments, such as those for hepatitis B and C viruses, are already in clinical use, and efforts to develop new, targeted therapies for other chronic hepatobiliary diseases are ongoing. In this review, we highlight the major molecular changes occurring in patients affected by metabolic dysfunction-associated steatotic liver disease, viral hepatitis (Delta virus), and autoimmune chronic liver diseases (autoimmune hepatitis, primary biliary cholangitis, and primary sclerosing cholangitis). Further, we describe how this knowledge is linked to current molecular therapies as well as ongoing preclinical and clinical research on novel targeting strategies, including nucleic acid-, mesenchymal stromal/stem cell-, and extracellular vesicle-based options. Much clinical development is obviously still missing, but the plethora of promising potential treatment strategies in chronic hepatobiliary diseases holds promise for a future reversal of the current increase in morbidity and mortality in this group of patients.
Collapse
Affiliation(s)
- Marilena Durazzo
- Department of Medical Sciences, University of Turin, C.so A.M. Dogliotti 14, 10126 Turin, Italy; (M.D.); (A.F.); (A.G.); (P.F.)
| | - Arianna Ferro
- Department of Medical Sciences, University of Turin, C.so A.M. Dogliotti 14, 10126 Turin, Italy; (M.D.); (A.F.); (A.G.); (P.F.)
| | - Victor Manuel Navarro-Tableros
- 2i3T, Società per la Gestione dell’Incubatore di Imprese e per il Trasferimento Tecnologico, University of Turin, 10126 Turin, Italy;
| | - Andrea Gaido
- Department of Medical Sciences, University of Turin, C.so A.M. Dogliotti 14, 10126 Turin, Italy; (M.D.); (A.F.); (A.G.); (P.F.)
| | - Paolo Fornengo
- Department of Medical Sciences, University of Turin, C.so A.M. Dogliotti 14, 10126 Turin, Italy; (M.D.); (A.F.); (A.G.); (P.F.)
| | - Fiorella Altruda
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre “Guido Tarone”, University of Turin, 10126 Turin, Italy;
| | - Renato Romagnoli
- General Surgery 2U-Liver Transplant Unit, Department of Surgical Sciences, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Turin, Corso Bramante 88-90, 10126 Turin, Italy;
| | - Søren K. Moestrup
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark;
- Department of Clinical Biochemistry, Aarhus University Hospital, 8000 Aarhus, Denmark
| | - Pier Luigi Calvo
- Pediatric Gastroenterology Unit, Regina Margherita Children’s Hospital, Città della Salute e della Scienza, 10126 Turin, Italy;
| | - Sharmila Fagoonee
- Institute for Biostructure and Bioimaging, National Research Council, Molecular Biotechnology Centre “Guido Tarone”, 10126 Turin, Italy
| |
Collapse
|
8
|
de Jong YP. Mice Engrafted with Human Liver Cells. Semin Liver Dis 2024; 44:405-415. [PMID: 39265638 PMCID: PMC11620938 DOI: 10.1055/s-0044-1790601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
Rodents are commonly employed to model human liver conditions, although species differences can restrict their translational relevance. To overcome some of these limitations, researchers have long pursued human hepatocyte transplantation into rodents. More than 20 years ago, the first primary human hepatocyte transplantations into immunodeficient mice with liver injury were able to support hepatitis B and C virus infections, as these viruses cannot replicate in murine hepatocytes. Since then, hepatocyte chimeric mouse models have transitioned into mainstream preclinical research and are now employed in a diverse array of liver conditions beyond viral hepatitis, including malaria, drug metabolism, liver-targeting gene therapy, metabolic dysfunction-associated steatotic liver disease, lipoprotein and bile acid biology, and others. Concurrently, endeavors to cotransplant other cell types and humanize immune and other nonparenchymal compartments have seen growing success. Looking ahead, several challenges remain. These include enhancing immune functionality in mice doubly humanized with hepatocytes and immune systems, efficiently creating mice with genetically altered grafts and reliably humanizing chimeric mice with renewable cell sources such as patient-specific induced pluripotent stem cells. In conclusion, hepatocyte chimeric mice have evolved into vital preclinical models that address many limitations of traditional rodent models. Continued improvements may further expand their applications.
Collapse
Affiliation(s)
- Ype P de Jong
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York
| |
Collapse
|
9
|
Spalding VA, Fellenstein BA, Ahodantin J, Jeyarajan AJ, Wang Y, Khan SK, Xu M, Lin W, Alatrakchi N, Su L, Chung RT, Salloum S. YAP mediates HIV-related liver fibrosis. JHEP Rep 2024; 6:101163. [PMID: 39524207 PMCID: PMC11544392 DOI: 10.1016/j.jhepr.2024.101163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 11/16/2024] Open
Abstract
Background & Aims HIV accelerates liver fibrosis attributable to multiple etiologies, including HCV, HBV, and steatotic liver disease. Evidence also suggests that HIV infection itself is associated with liver fibrogenesis. Recent studies have implicated Yes-associated protein 1 (YAP1) and the upstream lysophosphatidic acid (LPA)/PI3K/AKT pathway as critical regulators of hepatic fibrogenesis, and suggest a connection to HIV-related liver fibrosis. However, the relationship between YAP/PI3K/AKT pathway activation and HIV-related liver fibrosis remains uncertain. Methods qPCR, western blot, immunofluorescence, and ELISA (replicates n ≥3) were performed in an unbiased humanized mouse model (NRG-hu HSC mice, n = 6), the precision cut liver slice ex vivo model, and both traditional in vitro models as well as a 3D spheroid system. Results YAP target gene mRNA and protein levels (ANKRD, CTGF, CYR61) were upregulated across all models exposed to HIV. Humanized mice infected with HIV had significant increases in the percentage of YAP-positive nuclei (2.2-fold) and the percentage area of Sirius Red collagen staining (3.3-fold) compared to control mice. Serum concentrations of LPA were increased 5.8-fold in people living with HIV compared to healthy controls. Modulation of LPAR1, PI3K, and AKT by either inhibitors or small-interfering RNAs abrogated the fibrotic effects of HIV exposure and downregulated YAP target genes within cultured liver cells. Conclusions The LPAR/PI3K/AKT axis is vital for the activation of YAP and hepatic fibrogenesis due to HIV infection. This novel mechanistic insight suggests new pharmacologic targets for treatment of liver fibrosis in people living with HIV. Impact and implications There are currently no FDA-approved treatments for cirrhosis, while liver disease is the second leading cause of mortality among people living with HIV after AIDS. Increased lysophosphatidic acid concentrations and AKT activation after HIV infection found in recent work suggest that the Hippo pathway may be a key regulator of HIV-related fibrogenesis. By linking lysophosphatidic acid signaling, YAP activation, and HIV-related fibrogenesis, this mechanism presents a target for future research into therapeutic interventions for not only HIV but also other liver diseases, e.g. metabolic dysfunction- or alcohol-associated liver disease.
Collapse
Affiliation(s)
- Volney A. Spalding
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Brian A. Fellenstein
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - James Ahodantin
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andre J. Jeyarajan
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Yongtao Wang
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Sanjoy K. Khan
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Min Xu
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Wenyu Lin
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Nadia Alatrakchi
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Lishan Su
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Raymond T. Chung
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Shadi Salloum
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| |
Collapse
|
10
|
Echevarria-Lima J, Moles R. Monocyte and Macrophage Functions in Oncogenic Viral Infections. Viruses 2024; 16:1612. [PMID: 39459945 PMCID: PMC11512331 DOI: 10.3390/v16101612] [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: 09/10/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Monocytes and macrophages are part of innate immunity and constitute the first line of defense against pathogens. Bone marrow-derived monocytes circulate in the bloodstream for one to three days and then typically migrate into tissues, where they differentiate into macrophages. Circulatory monocytes represent 5% of the nucleated cells in normal adult blood. Following differentiation, macrophages are distributed into various tissues and organs to take residence and maintain body homeostasis. Emerging evidence has highlighted the critical role of monocytes/macrophages in oncogenic viral infections, mainly their crucial functions in viral persistence and disease progression. These findings open opportunities to target innate immunity in the context of oncogenic viruses and to explore their potential as immunotherapies.
Collapse
Affiliation(s)
- Juliana Echevarria-Lima
- Laboratório de Imunologia Básica e Aplicada, Department of Immunology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil;
| | - Ramona Moles
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Center for Immunology and Microbial Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
11
|
Marrufo AM, Flores-Mireles AL. Macrophage fate: to kill or not to kill? Infect Immun 2024; 92:e0047623. [PMID: 38829045 PMCID: PMC11385966 DOI: 10.1128/iai.00476-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
Macrophages are dynamic innate immune cells that either reside in tissue, serving as sentinels, or recruited as monocytes from bone marrow into inflamed and infected tissue. In response to cues in the tissue microenvironment (TME), macrophages polarize on a continuum toward M1 or M2 with diverse roles in progression and resolution of disease. M1-like macrophages exhibit proinflammatory functions with antimicrobial and anti-tumorigenic activities, while M2-like macrophages have anti-inflammatory functions that generally resolve inflammatory responses and orchestrate a tissue healing process. Given these opposite phenotypes, proper spatiotemporal coordination of macrophage polarization in response to cues within the TME is critical to effectively resolve infectious disease and regulate wound healing. However, if this spatiotemporal coordination becomes disrupted due to persistent infection or dysregulated coagulation, macrophages' inappropriate response to these cues will result in the development of diseases with clinically unfavorable outcomes. Since plasticity and heterogeneity are hallmarks of macrophages, they are attractive targets for therapies to reprogram toward specific phenotypes that could resolve disease and favor clinical prognosis. In this review, we discuss how basic science studies have elucidated macrophage polarization mechanisms in TMEs during infections and inflammation, particularly coagulation. Therefore, understanding the dynamics of macrophage polarization within TMEs in diseases is important in further development of targeted therapies.
Collapse
Affiliation(s)
- Armando M. Marrufo
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | | |
Collapse
|
12
|
Gangrade A, Zehtabi F, Ohe JY, Kouchehbaghi NH, Voskanian L, Haghniaz R, Shepes M, Rashad A, Ermis M, Khademhosseini A, Barros NRD. Engineered Regenerative and Adhesive Hydrogel for Concurrent Sealing and Healing of Enterocutaneous Fistulas. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39046205 DOI: 10.1021/acsami.4c05154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
In addressing the intricate challenges of enterocutaneous fistula (ECF) treatment, such as internal bleeding, effluent leakage, inflammation, and infection, our research is dedicated to introducing a regenerative adhesive hydrogel that can seal and expedite the healing process. A double syringe setup was utilized, with dopagelatin and platelet-rich plasma (PRP) in one syringe and Laponite and sodium periodate in another. The hydrogel begins to cross-link immediately after passing through a mixing tip and exhibits tissue adhesive properties. Results demonstrated that PRP deposits within the pores of the cross-linked hydrogel and releases sustainably, enhancing its regenerative capabilities. The addition of PRP further improved the mechanical properties and slowed down the degradation of the hydrogel. Furthermore, the hydrogel demonstrated cytocompatibility, hemostatic properties, and time-dependent macrophage M1 to M2 phase transition, suggesting the anti-inflammatory response of the material. In an in vitro bench test simulating high-pressure fistula conditions, the hydrogel effectively occluded pressures up to 300 mmHg. In conclusion, this innovative hydrogel holds promise for ECF treatment and diverse fistula cases, marking a significant advancement in its therapeutic approaches.
Collapse
Affiliation(s)
- Ankit Gangrade
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California 90095, United States
| | - Fatemeh Zehtabi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California 90095, United States
| | - Joo-Young Ohe
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California 90095, United States
- Department of Oral & Maxillofacial Surgery, College of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Negar Hosseinzadeh Kouchehbaghi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California 90095, United States
- Department of Textile Engineering, Amirkabir University of Technology (Tehran Polytechnic), Hafez Avenue, 1591634311 Tehran, Iran
| | - Leon Voskanian
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California 90095, United States
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California 90095, United States
| | - Matan Shepes
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California 90095, United States
| | - Ahmad Rashad
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California 90095, United States
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California 90095, United States
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California 90095, United States
| | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California 90095, United States
- National Laboratory of Bioscience (LNBio), National Center of Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil
| |
Collapse
|
13
|
Sheikhrobat SB, Mahmoudvand S, Kazemipour-Khabbazi S, Ramezannia Z, Baghi HB, Shokri S. Understanding lactate in the development of Hepatitis B virus-related hepatocellular carcinoma. Infect Agent Cancer 2024; 19:31. [PMID: 39010155 PMCID: PMC11247867 DOI: 10.1186/s13027-024-00593-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/26/2024] [Indexed: 07/17/2024] Open
Abstract
Hepatitis B Virus (HBV) is a hepatotropic virus that can establish a persistent and chronic infection in humans. Chronic hepatitis B (CHB) infection is associated with an increased risk of hepatic decompensation, cirrhosis, and hepatocellular carcinoma (HCC). Lactate level, as the end product of glycolysis, plays a substantial role in metabolism beyond energy production. Emerging studies indicate that lactate is linked to patient mortality rates, and HBV increases overall glucose consumption and lactate production in hepatocytes. Excessive lactate plays a role in regulating the tumor microenvironment (TME), immune cell function, autophagy, and epigenetic reprogramming. The purpose of this review is to gather and summarize the existing knowledge of the lactate's functions in the dysregulation of the immune system, which can play a crucial role in the development of HBV-related HCC. Therefore, it is reasonable to hypothesize that lactate with intriguing functions can be considered an immunomodulatory metabolite in immunotherapy.
Collapse
Affiliation(s)
- Sheida Behzadi Sheikhrobat
- Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shahab Mahmoudvand
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Virology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Salva Kazemipour-Khabbazi
- Department of English Language and Persian Literature, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Zahra Ramezannia
- Department of Virology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hossein Bannazadeh Baghi
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Somayeh Shokri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Virology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
14
|
Zhi X, Ren C, Li Q, Xi H, Li D, Chen Q, Lv X, Gao X, Wu X, Wang C, Jiang B, Mao Z, Jiang H, Liu K, Zhao X, Li Y. Therapeutic potential of Angelica sinensis in addressing organ fibrosis: A comprehensive review. Biomed Pharmacother 2024; 173:116429. [PMID: 38490157 DOI: 10.1016/j.biopha.2024.116429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/17/2024] Open
Abstract
Fibrosis-related diseases (FRD) include conditions like myocardial fibrosis, pulmonary fibrosis, hepatic fibrosis, renal fibrosis, and others. The impact of fibrosis can be severe, causing organ dysfunction, reduced functionality, and even organ failure, leading to significant health issues. Currently, there is a lack of effective modern anti-fibrosis drugs in clinical practice. However, Chinese medicine has a certain beneficial effect on the treatment of such diseases. Angelica sinensis, with its considerable medicinal value, has garnered attention for its anti-fibrosis properties in recent investigations. In the past few years, there has been a growing number of experimental inquiries into the impact of angelica polysaccharide (ASP), angelica water extract, angelica injection, and angelica compound preparation on fibrosis-associated ailments, piquing the interest of researchers. This paper aims to consolidate recent advances in the study of Angelica sinensis for the treatment of fibrosis-related disorders, offering insights for prospective investigations. Literature retrieval included core electronic databases, including Baidu Literature, CNKI, Google-Scholar, PubMed, and Web of Science. The applied search utilized specified keywords to extract relevant information on the pharmacological and phytochemical attributes of plants. The investigation revealed that Angelica sinensis has the potential to impede the advancement of fibrotic diseases by modulating inflammation, oxidative stress, immune responses, and metabolism. ASP, Angelica sinensis extract, Angelica sinensis injection, and Angelica sinensis compound preparation were extensively examined and discussed. These constituents demonstrated significant anti-fibrosis activity. In essence, this review seeks to gain a profound understanding of the role of Angelica sinensis in treating fiber-related diseases. Organ fibrosis manifests in nearly all tissues and organs, posing a critical challenge to global public health due to its widespread occurrence, challenging early diagnosis, and unfavorable prognosis. Despite its prevalence, therapeutic options are limited, and their efficacy is constrained. Over the past few years, numerous studies have explored the protective effects of traditional Chinese medicine on organ fibrosis, with Angelica sinensis standing out as a multifunctional natural remedy. This paper provides a review of organ fibrosis pathogenesis and summarizes the recent two decades' progress in treating fibrosis in various organs such as the liver, lung, kidney, and heart. The review highlights the modulation of relevant signaling pathways through multiple targets and channels by the effective components of Angelica sinensis, whether used as a single medicine or in compound prescriptions.
Collapse
Affiliation(s)
- Xiaodong Zhi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Chunzhen Ren
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Qianrong Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Huaqing Xi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Dong Li
- Qingyang Hospital of Traditional Chinese Medicine, Qingyang 745000, China
| | - Qilin Chen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Xinfang Lv
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Xiang Gao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Xue Wu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China; The second hospital of Lanzhou University, Lanzhou 730000, China
| | - Chunling Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Bing Jiang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Zhongnan Mao
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Hugang Jiang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Kai Liu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Xinke Zhao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000, China.
| | - Yingdong Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000, China.
| |
Collapse
|
15
|
Pinto E, Meneghel P, Farinati F, Russo FP, Pelizzaro F, Gambato M. Efficacy of immunotherapy in hepatocellular carcinoma: Does liver disease etiology have a role? Dig Liver Dis 2024; 56:579-588. [PMID: 37758610 DOI: 10.1016/j.dld.2023.08.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/18/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023]
Abstract
The systemic treatment of hepatocellular carcinoma (HCC) is changing rapidly. After a decade of tyrosine kinase inhibitors (TKIs), as the only therapeutic option for the treatment of advanced HCC, in the last few years several phase III trials demonstrated the efficacy of immune checkpoint inhibitors (ICIs). The combination of the anti-PD-L1 atezolizumab and the anti-vascular endothelial growth factor (VEGF) bevacizumab demonstrated the superiority over sorafenib and currently represents the standard of care treatment for advanced HCC. In addition, the combination of durvalumab (an anti-PD-L1) and tremelimumab (an anti-CTLA4) proved to be superior to sorafenib, and in the same trial durvalumab monotherapy showed non-inferiority compared to sorafenib. However, early reports suggest an influence of HCC etiology in modulating the response to these drugs. In particular, a lower effectiveness of ICIs has been suggested in patients with non-viral HCC (in particular non-alcoholic fatty liver disease). Nevertheless, randomized controlled trials available to date have not been stratified for etiology and data suggesting a possible impact of etiology in the outcome of patients managed with ICIs derive from subgroup not pre-specified analyses. In this review, we aim to examine the potential impact of HCC etiology on the response to immunotherapy regimens for HCC.
Collapse
Affiliation(s)
- Elisa Pinto
- Gastroenterology and Multivisceral Transplant Unit, Padua University Hospital, Padua, Italy
| | - Paola Meneghel
- Gastroenterology and Multivisceral Transplant Unit, Padua University Hospital, Padua, Italy
| | - Fabio Farinati
- Gastroenterology and Multivisceral Transplant Unit, Padua University Hospital, Padua, Italy
| | - Francesco Paolo Russo
- Gastroenterology and Multivisceral Transplant Unit, Padua University Hospital, Padua, Italy.
| | - Filippo Pelizzaro
- Gastroenterology and Multivisceral Transplant Unit, Padua University Hospital, Padua, Italy
| | - Martina Gambato
- Gastroenterology and Multivisceral Transplant Unit, Padua University Hospital, Padua, Italy.
| |
Collapse
|
16
|
Sadek A, Abd El Hady AM, Olfat H, Abd El Hady AA, Wafy W, Zaki M, Youness ER. Effect of ozone gas on viral kinetics and liver histopathology in hepatitis C patients. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2024; 21:131-138. [PMID: 38190482 DOI: 10.1515/jcim-2023-0239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/30/2023] [Indexed: 01/10/2024]
Abstract
OBJECTIVES We examine how well ozone/oxygen gas therapy treats chronic hepatitis C patients with varying degrees of liver fibrosis. Also to study the effect of giving multiple anti-oxidants with the ozone/oxygen gas mixture, to see if this addition would have any additive or synergistic effect. METHODS Two hundred and twenty three patients with chronic hepatitis C. Liver biopsies were carried out at after 12 weeks of administering an ozone/oxygen gas mixture. RESULTS The mean stage of fibrosis decreased from 1.98 to 1.41 and the mean grade of inflammation decreased from 10.08 to 7.94, both with a p value less than 0.001. After 12 weeks of treatment, mean PCR values increased. No single significant complication was recorded in a total of >9,000 settings of ozone therapy. CONCLUSIONS Ozone oxygen gas mixture is safe and effective in treatment of hepatic fibrosis due to chronic viral hepatitis C.
Collapse
Affiliation(s)
- Ahmed Sadek
- Departments of Hepatology & Gastroenterology, Theodor Bilharz Research Institute, Giza, Egypt
| | | | - Hammam Olfat
- Departments of Pathology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Afaf A Abd El Hady
- Departments of Clinical Chemistry, Theodor Bilharz Research Institute, Giza, Egypt
| | - Wafaa Wafy
- Departments of Public Health, Theodor Bilharz Research Institute, Giza, Egypt
| | - Moushira Zaki
- Department of Biological Anthropology, Medical Researches and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Eman R Youness
- Department of Medical Biochemistry, Medical Researches and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| |
Collapse
|
17
|
Li Y, Wu C, Lee J, Ning Q, Lim J, Eoh H, Wang S, Hurrell BP, Akbari O, Ou JHJ. Hepatitis B virus e antigen induces atypical metabolism and differentially regulates programmed cell deaths of macrophages. PLoS Pathog 2024; 20:e1012079. [PMID: 38466743 PMCID: PMC10957081 DOI: 10.1371/journal.ppat.1012079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/21/2024] [Accepted: 02/28/2024] [Indexed: 03/13/2024] Open
Abstract
Macrophages can undergo M1-like proinflammatory polarization with low oxidative phosphorylation (OXPHOS) and high glycolytic activities or M2-like anti-inflammatory polarization with the opposite metabolic activities. Here we show that M1-like macrophages induced by hepatitis B virus (HBV) display high OXPHOS and low glycolytic activities. This atypical metabolism induced by HBV attenuates the antiviral response of M1-like macrophages and is mediated by HBV e antigen (HBeAg), which induces death receptor 5 (DR5) via toll-like receptor 4 (TLR4) to induce death-associated protein 3 (DAP3). DAP3 then induces the expression of mitochondrial genes to promote OXPHOS. HBeAg also enhances the expression of glutaminases and increases the level of glutamate, which is converted to α-ketoglutarate, an important metabolic intermediate of the tricarboxylic acid cycle, to promote OXPHOS. The induction of DR5 by HBeAg leads to apoptosis of M1-like and M2-like macrophages, although HBeAg also induces pyroptosis of the former. These findings reveal novel activities of HBeAg, which can reprogram mitochondrial metabolism and trigger different programmed cell death responses of macrophages depending on their phenotypes to promote HBV persistence.
Collapse
Affiliation(s)
- Yumei Li
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Christine Wu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Jiyoung Lee
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Qiqi Ning
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Juhyeon Lim
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Hyungjin Eoh
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Sean Wang
- Michael Amini Transfusion Medicine Center, City of Hope, Duarte, California, United States of America
| | - Benjamin P. Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Jing-hsiung James Ou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
18
|
Rong N, Liu J. Development of animal models for emerging infectious diseases by breaking the barrier of species susceptibility to human pathogens. Emerg Microbes Infect 2023; 12:2178242. [PMID: 36748729 PMCID: PMC9970229 DOI: 10.1080/22221751.2023.2178242] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Outbreaks of emerging infectious diseases pose a serious threat to public health security, human health and economic development. After an outbreak, an animal model for an emerging infectious disease is urgently needed for studying the etiology, host immune mechanisms and pathology of the disease, evaluating the efficiency of vaccines or drugs against infection, and minimizing the time available for animal model development, which is usually hindered by the nonsusceptibility of common laboratory animals to human pathogens. Thus, we summarize the technologies and methods that induce animal susceptibility to human pathogens, which include viral receptor humanization, pathogen-targeted tissue humanization, immunodeficiency induction and screening for naturally susceptible animal species. Furthermore, the advantages and deficiencies of animal models developed using each method were analyzed, and these will guide the selection of susceptible animals and potentially reduce the time needed to develop animal models during epidemics.
Collapse
Affiliation(s)
- Na Rong
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Jiangning Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, People’s Republic of China, Jiangning Liu
| |
Collapse
|
19
|
Zheng Y, Ji S, Li X, Wen L. Qijia rougan formula ameliorates ECM deposition in hepatic fibrosis by regulating the JAK1/STAT6-microRNA-23a feedback loop in macrophage M2 polarization. Biomed Pharmacother 2023; 168:115794. [PMID: 37922651 DOI: 10.1016/j.biopha.2023.115794] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/15/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Hepatic fibrosis is the critical pathological stage in the progression of chronic liver disease to cirrhosis and hepatocellular carcinoma (HCC). However, no approved anti-hepatic fibrosis drugs are available currently. Qijia Rougan Formula (QRF) is a traditional Chinese medicine (TCM) with significant clinical efficacy on hepatic fibrosis. It was derived from Sanjiasan, a famous decoction documented in the Book of Treatise on the Pestilence in the Ming Dynasty of China. However, the underlying regulatory mechanisms remain elusive. This study further confirmed the therapeutic effects of QRF on hepatic fibrosis and dissected its underlying molecular mechanisms from the perspective of macrophage M2 polarization, one of the critical events in hepatic fibrosis. Experimentally, QRF significantly improved extracellular matrix (ECM) deposition and fibrosis in the liver of model rats. QRF diminished the proportion of M2 macrophages, decreased the levels of TGF-β, PDGFB and IL-10, and regulated the expression of p-JAK1, p-STAT6, JAK1 and microRNA-23a both in vitro and in vivo. Collectively, it was confirmed that QRF effectively improves liver function and hepatocyte damage, and reduces ECM deposition. QRF ameliorates hepatic fibrosis by regulating JAK1/STAT6-microRNA-23a negative feedback loop to inhibit macrophage M2 polarization and thus reduce ECM deposition. Our study illustrates the potential of QRF for hepatic fibrosis therapy, suggesting that QRF is a promising anti-hepatic fibrosis drug candidate.
Collapse
Affiliation(s)
- Yanfeng Zheng
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shaoxiu Ji
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xia Li
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Li Wen
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
20
|
Li Y, Ou JHJ. Regulation of Mitochondrial Metabolism by Hepatitis B Virus. Viruses 2023; 15:2359. [PMID: 38140600 PMCID: PMC10747323 DOI: 10.3390/v15122359] [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: 11/09/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Mitochondria play important roles in the synthesis of ATP, the production of reactive oxygen species, and the regulation of innate immune response and apoptosis. Many viruses perturb mitochondrial activities to promote their replication and cause cell damage. Hepatitis B virus (HBV) is a hepatotropic virus that can cause severe liver diseases, including cirrhosis and hepatocellular carcinoma (HCC). This virus can also alter mitochondrial functions and metabolism to promote its replication and persistence. In this report, we summarize recent research progress on the interaction between HBV and mitochondrial metabolism, as well as the effect this interaction has on HBV replication and persistence.
Collapse
Affiliation(s)
| | - Jing-hsiung James Ou
- Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA;
| |
Collapse
|
21
|
Genshaft AS, Subudhi S, Keo A, Sanchez Vasquez JD, Conceição-Neto N, Mahamed D, Boeijen LL, Alatrakchi N, Oetheimer C, Vilme M, Drake R, Fleming I, Tran N, Tzouanas C, Joseph-Chazan J, Arreola Villanueva M, van de Werken HJG, van Oord GW, Groothuismink ZMA, Beudeker BJ, Osmani Z, Nkongolo S, Mehrotra A, Spittaels K, Feld J, Chung RT, de Knegt RJ, Janssen HLA, Aerssens J, Bollekens J, Hacohen N, Lauer GM, Boonstra A, Shalek AK, Gehring AJ. Single-cell RNA sequencing of liver fine-needle aspirates captures immune diversity in the blood and liver in chronic hepatitis B patients. Hepatology 2023; 78:1525-1541. [PMID: 37158243 PMCID: PMC10581444 DOI: 10.1097/hep.0000000000000438] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND AND AIMS HBV infection is restricted to the liver, where it drives exhaustion of virus-specific T and B cells and pathogenesis through dysregulation of intrahepatic immunity. Our understanding of liver-specific events related to viral control and liver damage has relied almost solely on animal models, and we lack useable peripheral biomarkers to quantify intrahepatic immune activation beyond cytokine measurement. Our objective was to overcome the practical obstacles of liver sampling using fine-needle aspiration and develop an optimized workflow to comprehensively compare the blood and liver compartments within patients with chronic hepatitis B using single-cell RNA sequencing. APPROACH AND RESULTS We developed a workflow that enabled multi-site international studies and centralized single-cell RNA sequencing. Blood and liver fine-needle aspirations were collected, and cellular and molecular captures were compared between the Seq-Well S 3 picowell-based and the 10× Chromium reverse-emulsion droplet-based single-cell RNA sequencing technologies. Both technologies captured the cellular diversity of the liver, but Seq-Well S 3 effectively captured neutrophils, which were absent in the 10× dataset. CD8 T cells and neutrophils displayed distinct transcriptional profiles between blood and liver. In addition, liver fine-needle aspirations captured a heterogeneous liver macrophage population. Comparison between untreated patients with chronic hepatitis B and patients treated with nucleoside analogs showed that myeloid cells were highly sensitive to environmental changes while lymphocytes displayed minimal differences. CONCLUSIONS The ability to electively sample and intensively profile the immune landscape of the liver, and generate high-resolution data, will enable multi-site clinical studies to identify biomarkers for intrahepatic immune activity in HBV and beyond.
Collapse
Affiliation(s)
- Alex S. Genshaft
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Massachusetts, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Sonu Subudhi
- Liver Center, Division of Gastroenterology and Liver Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Arlin Keo
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Cancer Computational Biology Center, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Juan Diego Sanchez Vasquez
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Nádia Conceição-Neto
- Infectious Diseases Biomarkers, Janssen Research and Development, Beerse, Belgium
| | - Deeqa Mahamed
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Lauke L. Boeijen
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Nadia Alatrakchi
- Liver Center, Division of Gastroenterology and Liver Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Chris Oetheimer
- Liver Center, Division of Gastroenterology and Liver Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mike Vilme
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Massachusetts, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Riley Drake
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Massachusetts, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Ira Fleming
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Massachusetts, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Nancy Tran
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Massachusetts, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Constantine Tzouanas
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Massachusetts, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Jasmin Joseph-Chazan
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Massachusetts, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Martin Arreola Villanueva
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Massachusetts, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Harmen J. G. van de Werken
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Cancer Computational Biology Center, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
- Department of Immunology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Gertine W. van Oord
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Zwier M. A. Groothuismink
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Boris J. Beudeker
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Zgjim Osmani
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Shirin Nkongolo
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Aman Mehrotra
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kurt Spittaels
- Infectious Diseases Biomarkers, Janssen Research and Development, Beerse, Belgium
| | - Jordan Feld
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Raymond T. Chung
- Liver Center, Division of Gastroenterology and Liver Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Robert J. de Knegt
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Harry L. A. Janssen
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Jeroen Aerssens
- Infectious Diseases Biomarkers, Janssen Research and Development, Beerse, Belgium
| | - Jacques Bollekens
- Infectious Diseases Biomarkers, Janssen Research and Development, Beerse, Belgium
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Georg M. Lauer
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, Massachusetts, USA
| | - Andre Boonstra
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Alex K. Shalek
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Massachusetts, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Adam J. Gehring
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Ozdarendeli A. Crimean-Congo Hemorrhagic Fever Virus: Progress in Vaccine Development. Diagnostics (Basel) 2023; 13:2708. [PMID: 37627967 PMCID: PMC10453274 DOI: 10.3390/diagnostics13162708] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV), a member of the Nairoviridae family and Bunyavirales order, is transmitted to humans via tick bites or contact with the blood of infected animals. It can cause severe symptoms, including hemorrhagic fever, with a mortality rate between 5 to 30%. CCHFV is classified as a high-priority pathogen by the World Health Organization (WHO) due to its high fatality rate and the absence of effective medical countermeasures. CCHFV is endemic in several regions across the world, including Africa, Europe, the Middle East, and Asia, and has the potential for global spread. The emergence of the disease in new areas, as well as the presence of the tick vector in countries without reported cases, emphasizes the need for preventive measures to be taken. In the past, the lack of a suitable animal model susceptible to CCHFV infection has been a major obstacle in the development of vaccines and treatments. However, recent advances in biotechnology and the availability of suitable animal models have significantly expedited the development of vaccines against CCHF. These advancements have not only contributed to an enhanced understanding of the pathogenesis of CCHF but have also facilitated the evaluation of potential vaccine candidates. This review outlines the immune response to CCHFV and animal models utilized for the study of CCHFV and highlights the progress made in CCHFV vaccine studies. Despite remarkable advancements in vaccine development for CCHFV, it remains crucial to prioritize continued research, collaboration, and investment in this field.
Collapse
Affiliation(s)
- Aykut Ozdarendeli
- Department of Microbiology, Faculty of Medicine, Erciyes University, 38039 Kayseri, Türkiye;
- Vaccine Research, Development and Application Centre (ERAGEM), Erciyes University, 38039 Kayseri, Türkiye
| |
Collapse
|
23
|
Strizova Z, Benesova I, Bartolini R, Novysedlak R, Cecrdlova E, Foley L, Striz I. M1/M2 macrophages and their overlaps - myth or reality? Clin Sci (Lond) 2023; 137:1067-1093. [PMID: 37530555 PMCID: PMC10407193 DOI: 10.1042/cs20220531] [Citation(s) in RCA: 164] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 08/03/2023]
Abstract
Macrophages represent heterogeneous cell population with important roles in defence mechanisms and in homoeostasis. Tissue macrophages from diverse anatomical locations adopt distinct activation states. M1 and M2 macrophages are two polarized forms of mononuclear phagocyte in vitro differentiation with distinct phenotypic patterns and functional properties, but in vivo, there is a wide range of different macrophage phenotypes in between depending on the microenvironment and natural signals they receive. In human infections, pathogens use different strategies to combat macrophages and these strategies include shaping the macrophage polarization towards one or another phenotype. Macrophages infiltrating the tumours can affect the patient's prognosis. M2 macrophages have been shown to promote tumour growth, while M1 macrophages provide both tumour-promoting and anti-tumour properties. In autoimmune diseases, both prolonged M1 activation, as well as altered M2 function can contribute to their onset and activity. In human atherosclerotic lesions, macrophages expressing both M1 and M2 profiles have been detected as one of the potential factors affecting occurrence of cardiovascular diseases. In allergic inflammation, T2 cytokines drive macrophage polarization towards M2 profiles, which promote airway inflammation and remodelling. M1 macrophages in transplantations seem to contribute to acute rejection, while M2 macrophages promote the fibrosis of the graft. The view of pro-inflammatory M1 macrophages and M2 macrophages suppressing inflammation seems to be an oversimplification because these cells exploit very high level of plasticity and represent a large scale of different immunophenotypes with overlapping properties. In this respect, it would be more precise to describe macrophages as M1-like and M2-like.
Collapse
Affiliation(s)
- Zuzana Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, 15006, Prague, Czech Republic
| | - Iva Benesova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, 15006, Prague, Czech Republic
| | - Robin Bartolini
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, U.K
| | - Rene Novysedlak
- Third Department of Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, 15006, Prague, Czech Republic
| | - Eva Cecrdlova
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Lily Koumbas Foley
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, U.K
| | - Ilja Striz
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
24
|
Zeng DY, Chen Z, Hong MZ, Jiang LP, Chen XN, Xue HX, Pan JS, Zhu Y. Traditional Chinese medicine invigorating the spleen and kidney promotes HBsAg seroclearance in the mouse model. J Med Virol 2023; 95:e28979. [PMID: 37522253 DOI: 10.1002/jmv.28979] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 08/01/2023]
Abstract
Traditional Chinese medicine (TCM) is often used as an adjuvant or alternative therapy for abnormal liver biochemistry or liver fibrosis associated with chronic hepatitis B (CHB). However, the role of TCM in HBsAg seroclearance remains unclear. We aimed at exploring the role and possible mechanisms of TCM in HBsAg seroclearance. Fifteen widely used TCM granules invigorating the spleen and kidneys were screened. C57BL/6J mice were administered daily with TCM granules by gavage for 1 week. The effect of TCM on the M1 polarization of macrophages was measured using a CD86 assay. According to the principles of formulating prescriptions, three single TCM with the most noticeable effect on M1 polarization, accompanied by two other TCM granules, were used to develop a TCM formula. The hepatitis B virus-expressing mouse model was constructed by hydrodynamic injection of the pAAV/HBV1.2 plasmid. Hepatitis B virus-expressing mice were gavaged daily with phosphate-buffered saline (PBS), TCM formula, or Codonopsis Radix, for 1 week. HBsAg, HBeAg, and hepatitis B virus DNA levels were measured. In addition, gut microbiota was profiled using 16S rDNA sequencing. Several TCM granules showed significant effects on M1 polarization. The TCM formula accelerated HBsAg seroclearance compared with the Codonopsis Radix and PBS groups. Intrahepatic M1 polarization, as indicated by flow cytometry and immunohistochemistry, was induced in the TCM formula and Codonopsis Radix groups. The abundance of Alloprevotella significantly increased in the TCM formula and Codonopsis Radix groups. These results demonstrate that the TCM formula for invigorating the spleen and kidney can accelerate HBsAg seroclearance. This effect can be attributed, at least in part, to M1 polarization of intrahepatic macrophages.
Collapse
Affiliation(s)
- Dan-Yi Zeng
- Department of Hepatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Hepatology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhan Chen
- Department of Hepatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Hepatology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Mei-Zhu Hong
- Department of Traditional Chinese Medicine, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Ling-Ping Jiang
- Department of Hepatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Hepatology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiao-Ning Chen
- Department of Hepatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Hepatology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Han-Xin Xue
- Department of Hepatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Hepatology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Jin-Shui Pan
- Department of Hepatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Hepatology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yueyong Zhu
- Department of Hepatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Hepatology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
25
|
Feng L, Chen X, Huang Y, Zhang X, Zheng S, Xie N. Immunometabolism changes in fibrosis: from mechanisms to therapeutic strategies. Front Pharmacol 2023; 14:1243675. [PMID: 37576819 PMCID: PMC10412938 DOI: 10.3389/fphar.2023.1243675] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
Immune cells are essential for initiating and developing the fibrotic process by releasing cytokines and growth factors that activate fibroblasts and promote extracellular matrix deposition. Immunometabolism describes how metabolic alterations affect the function of immune cells and how inflammation and immune responses regulate systemic metabolism. The disturbed immune cell function and their interactions with other cells in the tissue microenvironment lead to the origin and advancement of fibrosis. Understanding the dysregulated metabolic alterations and interactions between fibroblasts and the immune cells is critical for providing new therapeutic targets for fibrosis. This review provides an overview of recent advances in the pathophysiology of fibrosis from the immunometabolism aspect, highlighting the altered metabolic pathways in critical immune cell populations and the impact of inflammation on fibroblast metabolism during the development of fibrosis. We also discuss how this knowledge could be leveraged to develop novel therapeutic strategies for treating fibrotic diseases.
Collapse
Affiliation(s)
- Lixiang Feng
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xingyu Chen
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yujing Huang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xiaodian Zhang
- Hainan Cancer Clinical Medical Center of the First Affiliated Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province and Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Shaojiang Zheng
- Hainan Cancer Clinical Medical Center of the First Affiliated Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province and Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
- Department of Pathology, Hainan Women and Children Medical Center, Hainan Medical University, Haikou, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
26
|
Yi Q, Yang J, Wu Y, Wang Y, Cao Q, Wen W. Immune microenvironment changes of liver cirrhosis: emerging role of mesenchymal stromal cells. Front Immunol 2023; 14:1204524. [PMID: 37539053 PMCID: PMC10395751 DOI: 10.3389/fimmu.2023.1204524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/21/2023] [Indexed: 08/05/2023] Open
Abstract
Cirrhosis is a progressive and diffuse liver disease characterized by liver tissue fibrosis and impaired liver function. This condition is brought about by several factors, including chronic hepatitis, hepatic steatosis, alcohol abuse, and other immunological injuries. The pathogenesis of liver cirrhosis is a complex process that involves the interaction of various immune cells and cytokines, which work together to create the hepatic homeostasis imbalance in the liver. Some studies have indicated that alterations in the immune microenvironment of liver cirrhosis are closely linked to the development and prognosis of the disease. The noteworthy function of mesenchymal stem cells and their paracrine secretion lies in their ability to promote the production of cytokines, which in turn enhance the self-repairing capabilities of tissues. The objective of this review is to provide a summary of the alterations in liver homeostasis and to discuss intercellular communication within the organ. Recent research on MSCs is yielding a blueprint for cell typing and biomarker immunoregulation. Hopefully, as MSCs researches continue to progress, novel therapeutic approaches will emerge to address cirrhosis.
Collapse
Affiliation(s)
- Qiuyun Yi
- National Center for Liver Cancer, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Jinxian Yang
- National Center for Liver Cancer, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ying Wu
- Department of Breast and Thyroid Surgery, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Ying Wang
- Department of Laboratory Diagnosis, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Qiqi Cao
- National Center for Liver Cancer, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wen Wen
- National Center for Liver Cancer, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Laboratory Diagnosis, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
27
|
Hogan G, Winer BY, Ahodantin J, Sellau J, Huang T, Douam F, Funaki M, Chiriboga L, Su L, Ploss A. Persistent hepatitis B virus and HIV coinfections in dually humanized mice engrafted with human liver and immune system. J Med Virol 2023; 95:e28930. [PMID: 37403703 PMCID: PMC11298785 DOI: 10.1002/jmv.28930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 07/06/2023]
Abstract
Chronic hepatitis B (CHB), caused by hepatitis B virus (HBV), remains a major medical problem. HBV has a high propensity for progressing to chronicity and can result in severe liver disease, including fibrosis, cirrhosis, and hepatocellular carcinoma. CHB patients frequently present with viral coinfection, including human immunodeficiency virus type (HIV) and hepatitis delta virus. About 10% of chronic HIV carriers are also persistently infected with HBV, which can result in more exacerbated liver disease. Mechanistic studies of HBV-induced immune responses and pathogenesis, which could be significantly influenced by HIV infection, have been hampered by the scarcity of immunocompetent animal models. Here, we demonstrate that humanized mice dually engrafted with components of a human immune system and a human liver supported HBV infection, which was partially controlled by human immune cells, as evidenced by lower levels of serum viremia and HBV replication intermediates in the liver. HBV infection resulted in priming and expansion of human HLA-restricted CD8+ T cells, which acquired an activated phenotype. Notably, our dually humanized mice support persistent coinfections with HBV and HIV, which opens opportunities for analyzing immune dysregulation during HBV and HIV coinfection, and preclinical testing of novel immunotherapeutics.
Collapse
Affiliation(s)
- Glenn Hogan
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Benjamin Y Winer
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - James Ahodantin
- Division of Virology, Pathogenesis and Cancer, Institute of Human Virology, Departments of Pharmacology, Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Julie Sellau
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Tiffany Huang
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Florian Douam
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Masaya Funaki
- Division of Virology, Pathogenesis and Cancer, Institute of Human Virology, Departments of Pharmacology, Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Luis Chiriboga
- Department of Pathology, New York University Medical Center, New York, New York, USA
| | - Lishan Su
- Division of Virology, Pathogenesis and Cancer, Institute of Human Virology, Departments of Pharmacology, Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alexander Ploss
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
28
|
Liu Y, Cafiero TR, Park D, Biswas A, Winer BY, Cho CH, Bram Y, Chandar V, Connell AKO, Gertje HP, Crossland N, Schwartz RE, Ploss A. Targeted viral adaptation generates a simian-tropic hepatitis B virus that infects marmoset cells. Nat Commun 2023; 14:3582. [PMID: 37328459 PMCID: PMC10276007 DOI: 10.1038/s41467-023-39148-3] [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: 10/19/2022] [Accepted: 05/26/2023] [Indexed: 06/18/2023] Open
Abstract
Hepatitis B virus (HBV) only infects humans and chimpanzees, posing major challenges for modeling HBV infection and chronic viral hepatitis. The major barrier in establishing HBV infection in non-human primates lies at incompatibilities between HBV and simian orthologues of the HBV receptor, sodium taurocholate co-transporting polypeptide (NTCP). Through mutagenesis analysis and screening among NTCP orthologues from Old World monkeys, New World monkeys and prosimians, we determined key residues responsible for viral binding and internalization, respectively and identified marmosets as a suitable candidate for HBV infection. Primary marmoset hepatocytes and induced pluripotent stem cell-derived hepatocyte-like cells support HBV and more efficient woolly monkey HBV (WMHBV) infection. Adapted chimeric HBV genome harboring residues 1-48 of WMHBV preS1 generated here led to a more efficient infection than wild-type HBV in primary and stem cell derived marmoset hepatocytes. Collectively, our data demonstrate that minimal targeted simianization of HBV can break the species barrier in small NHPs, paving the path for an HBV primate model.
Collapse
Affiliation(s)
- Yongzhen Liu
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Thomas R Cafiero
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Debby Park
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Abhishek Biswas
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Research Computing, Office of Information Technology, Princeton University, Princeton, NJ, 08544, USA
| | - Benjamin Y Winer
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | | | - Yaron Bram
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Vasuretha Chandar
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Aoife K O' Connell
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02118, USA
| | - Hans P Gertje
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02118, USA
| | - Nicholas Crossland
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02118, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Robert E Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
29
|
Gutierrez-Barbosa H, Medina-Moreno S, Perdomo-Celis F, Davis H, Coronel-Ruiz C, Zapata JC, Chua JV. A Comparison of Lymphoid and Myeloid Cells Derived from Human Hematopoietic Stem Cells Xenografted into NOD-Derived Mouse Strains. Microorganisms 2023; 11:1548. [PMID: 37375051 DOI: 10.3390/microorganisms11061548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Humanized mice are an invaluable tool for investigating human diseases such as cancer, infectious diseases, and graft-versus-host disease (GvHD). However, it is crucial to understand the strengths and limitations of humanized mice and select the most appropriate model. In this study, we describe the development of the human lymphoid and myeloid lineages using a flow cytometric analysis in four humanized mouse models derived from NOD mice xenotransplanted with CD34+ fetal cord blood from a single donor. Our results showed that all murine strains sustained human immune cells within a proinflammatory environment induced by GvHD. However, the Hu-SGM3 model consistently generated higher numbers of human T cells, monocytes, dendritic cells, mast cells, and megakaryocytes, and a low number of circulating platelets showing an activated profile when compared with the other murine strains. The hu-NOG-EXL model had a similar cell development profile but a higher number of circulating platelets with an inactivated state, and the hu-NSG and hu-NCG developed low frequencies of immune cells compared with the other models. Interestingly, only the hu-SGM3 and hu-EXL models developed mast cells. In conclusion, our findings highlight the importance of selecting the appropriate humanized mouse model for specific research questions, considering the strengths and limitations of each model and the immune cell populations of interest.
Collapse
Affiliation(s)
| | - Sandra Medina-Moreno
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Federico Perdomo-Celis
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Harry Davis
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Carolina Coronel-Ruiz
- Vice-Chancellor of Research, Virology Group, Universidad El Bosque, Bogotá 110121, Colombia
| | - Juan C Zapata
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joel V Chua
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
30
|
Zhou Z, Li C, Tan Z, Sun G, Peng B, Ren T, He J, Wang Y, Sun Y, Wang F, Li W. A spatiotemporally controlled recombinant cccDNA mouse model for studying HBV and developing drugs against the virus. Antiviral Res 2023:105642. [PMID: 37253400 DOI: 10.1016/j.antiviral.2023.105642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/08/2023] [Accepted: 05/23/2023] [Indexed: 06/01/2023]
Abstract
Covalently closed circular (ccc) DNA is the template for hepatitis B virus (HBV) replication. The lack of small animal models for characterizing chronic HBV infection has hampered research progress in HBV pathogenesis and drug development. Here, we generated a spatiotemporally controlled recombinant cccDNA (rcccDNA) mouse model by combining Cre/loxP-mediated DNA recombination with the liver-specific "Tet-on/Cre" system. The mouse model harbors three transgenes: a single copy of the HBV genome (integrated at the Rosa26 locus, RHBV), H11-albumin-rtTA (spatiotemporal conditional module), and (tetO)7-Cre (tetracycline response element), and is named as RHTC mouse. By supplying the RHTC mice with doxycycline (DOX)-containing drinking water for two days, the animals generate rcccDNA in hepatocytes, and the rcccDNA supports active HBV gene expression and can maintain HBV viremia persistence for over 60 weeks. Persistent HBV gene expression induces intrahepatic inflammation, fibrosis, and dysplastic pathology, which closely mirrors the disease progression in clinical patients. Bepirovirsen, an antisense oligonucleotide (ASO) targeting all HBV RNA species, showed dose-dependent antiviral effects in the RHTC mouse model. The spatiotemporally controlled rcccDNA mouse is convenient and reliable, providing versatile small animal model for studying cccDNA-centric HBV biology as well as evaluating antiviral therapeutics.
Collapse
Affiliation(s)
- Zhongmin Zhou
- College of Life Sciences, Beijing Normal University, Beijing, China; National Institute of Biological Sciences, Beijing, China
| | - Cong Li
- National Institute of Biological Sciences, Beijing, China; Graduate Program in School of Life Sciences, Peking University, Beijing, China
| | - Zexi Tan
- National Institute of Biological Sciences, Beijing, China
| | - Guoliang Sun
- National Institute of Biological Sciences, Beijing, China; Graduate Program in School of Life Sciences, Peking University, Beijing, China
| | - Bo Peng
- National Institute of Biological Sciences, Beijing, China; Graduate Program in School of Life Sciences, Peking University, Beijing, China
| | - Tengfei Ren
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Jiabei He
- National Institute of Biological Sciences, Beijing, China; College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yixue Wang
- College of Life Sciences, Beijing Normal University, Beijing, China; National Institute of Biological Sciences, Beijing, China
| | - Yinyan Sun
- National Institute of Biological Sciences, Beijing, China
| | - Fengchao Wang
- National Institute of Biological Sciences, Beijing, China
| | - Wenhui Li
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| |
Collapse
|
31
|
You H, Wang X, Ma L, Zhang F, Zhang H, Wang Y, Pan X, Zheng K, Kong F, Tang R. Insights into the impact of hepatitis B virus on hepatic stellate cell activation. Cell Commun Signal 2023; 21:70. [PMID: 37041599 PMCID: PMC10088164 DOI: 10.1186/s12964-023-01091-7] [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: 11/14/2022] [Accepted: 02/26/2023] [Indexed: 04/13/2023] Open
Abstract
During chronic hepatitis B virus (HBV) infection, hepatic fibrosis is a serious pathological condition caused by virus-induced liver damage. The activation of hepatic stellate cells (HSCs) is a central event in the occurrence and progression of liver fibrosis. Although accumulating evidence has shown that HBV directly stimulates HSC activation, whether the virus infects and replicates in HSCs remains controversial. Inflammation is one of the obvious characteristics of chronic HBV infection, and it has been demonstrated that persistent inflammation has a predominant role in triggering and maintaining liver fibrosis. In particular, the regulation of HSC activation by HBV-related hepatocytes via various inflammatory modulators, including TGF-β and CTGF, in a paracrine manner has been reported. In addition to these inflammation-related molecules, several inflammatory cells are essential for the progression of HBV-associated liver fibrosis. Monocytes, macrophages, Th17 cells, NK cells, as well as NKT cells, participate in the modulation of HBV-related liver fibrosis by interacting with HSCs. This review summarizes current findings on the effects of HBV and the relevant molecular mechanisms involved in HSC activation. Because HSC activation is essential for liver fibrosis, targeting HSCs is an attractive therapeutic strategy to prevent and reverse hepatic fibrosis induced by HBV infection. Video abstract.
Collapse
Affiliation(s)
- Hongjuan You
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xing Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lihong Ma
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fulong Zhang
- Imaging Department, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Huanyang Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuxin Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiucheng Pan
- Department of Infectious Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fanyun Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
32
|
Uchida T, Teraoka Y, Imamura M, Abe-Chayama H, Makokha GN, Hayes CN, Aikata H, Hamamura S, Ishida Y, Tateno C, Shirouzu T, Kawai S, Tanaka Y, Ohdan H, Okada S, Chayama K. A novel cDNA-uPA/SCID/Rag2 -/- /Jak3 -/- mouse model for hepatitis virus infection and reconstruction of human immune system. J Viral Hepat 2023; 30:262-272. [PMID: 36575861 DOI: 10.1111/jvh.13793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/26/2022] [Accepted: 11/20/2022] [Indexed: 12/29/2022]
Abstract
Although human hepatocyte-transplanted immunodeficient mice support infection with hepatitis viruses, these mice fail to develop viral hepatitis due to the lack of an adaptive immune system. In this study, we generated new immunodeficiency cDNA-urokinase-type plasminogen activator (uPA)/SCID/Rag2-/- /Jak3-/- mice and established a mouse model with both a humanized liver and immune system. Transplantation of human hepatocytes with human leukocyte antigen (HLA)-A24 resulted in establishment of a highly replaced liver in cDNA-uPA/SCID/Rag2-/- /Jak3-/- mice. These mice were successfully infected with hepatitis B virus (HBV) and hepatitis C virus (HCV) for a prolonged period and facilitate analysis of the effect of anti-HCV drugs. Administration of peripheral blood mononuclear cells (PBMCs) obtained from an HLA-A24 donor resulted in establishment of 22.6%-81.3% human CD45-positive mononuclear cell chimerism in liver-infiltrating cells without causing graft-versus-host disease in cDNA-uPA/SCID/Rag2-/- /Jak3-/- mice without human hepatocyte transplantation. When mice were transplanted with human hepatocytes and then administered HLA-A24-positive human PBMCs, an alloimmune response between transplanted human hepatocytes and PBMCs occurred, with production of transplanted hepatocyte-specific anti-HLA antibody. In conclusion, we succeeded in establishing a humanized liver/immune system characterized by an allo-reaction between transplanted human immune cells and human liver using a novel cDNA-uPA/SCID/Rag2-/- /Jak3-/- mouse. This mouse model can be used to generate a chronic hepatitis mouse model with a human immune system with application not only to hepatitis virus virology but also to investigation of the pathology of post-transplantation liver rejection.
Collapse
Affiliation(s)
- Takuro Uchida
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Research Center for Hepatology and Gastroenterology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Yuji Teraoka
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Research Center for Hepatology and Gastroenterology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Michio Imamura
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Research Center for Hepatology and Gastroenterology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Hiromi Abe-Chayama
- Research Center for Hepatology and Gastroenterology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
- Center for Medical Specialist Graduate Education and Research, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Grace Naswa Makokha
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Research Center for Hepatology and Gastroenterology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Clair Nelson Hayes
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Research Center for Hepatology and Gastroenterology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Hiroshi Aikata
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Research Center for Hepatology and Gastroenterology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Satoko Hamamura
- Research Center for Hepatology and Gastroenterology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
- PhoenixBio Co., Ltd., Higashihiroshima, Japan
| | - Yuji Ishida
- Research Center for Hepatology and Gastroenterology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
- PhoenixBio Co., Ltd., Higashihiroshima, Japan
| | - Chise Tateno
- Research Center for Hepatology and Gastroenterology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
- PhoenixBio Co., Ltd., Higashihiroshima, Japan
| | - Takayuki Shirouzu
- Molecular Diagnostics Division, Wakunaga Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Shintaro Kawai
- Molecular Diagnostics Division, Wakunaga Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Yuka Tanaka
- Research Center for Hepatology and Gastroenterology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Hideki Ohdan
- Research Center for Hepatology and Gastroenterology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection and Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Kazuaki Chayama
- Research Center for Hepatology and Gastroenterology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
- Collaborative Research Laboratory of Medical Innovation, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Hiroshima Institute of Life Sciences, Hiroshima, Japan
| |
Collapse
|
33
|
Sun S, Yang H, Xin J, Yao H, Yuan L, Ren K, Jiang J, Shi D, Li J, Zhou Q, An Z, Guo B, Chen J, He L, Liang X, Cheng T, Xia N, Li J. Transcriptomics confirm the establishment of a liver-immune dual-humanized mouse model after transplantation of a single type of human bone marrow mesenchymal stem cell. Liver Int 2023; 43:1345-1356. [PMID: 36810858 DOI: 10.1111/liv.15546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/20/2022] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND AND AIMS Human bone marrow mesenchymal stem cells (hBMSCs) are important for developing a dual-humanized mouse model to clarify disease pathogenesis. We aimed to elucidate the characteristics of hBMSC transdifferentiation into liver and immune cells. METHODS A single type of hBMSCs was transplanted into immunodeficient Fah-/- Rag2-/- IL-2Rγc-/- SCID (FRGS) mice with fulminant hepatic failure (FHF). Liver transcriptional data from the hBMSC-transplanted mice were analysed to identify transdifferentiation with traces of liver and immune chimerism. RESULTS Mice with FHF were rescued by implanted hBMSCs. Human albumin/leukocyte antigen (HLA) and CD45/HLA double-positive hepatocytes and immune cells were observed in the rescued mice during the initial 3 days. The transcriptomics analysis of liver tissues from dual-humanized mice identified two transdifferentiation phases (cellular proliferation at 1-5 days and cellular differentiation/maturation at 5-14 days) and ten cell lineages transdifferentiated from hBMSCs: human hepatocytes, cholangiocytes, stellate cells, myofibroblasts, endothelial cells and immune cells (T/B/NK/NKT/Kupffer cells). Two biological processes, hepatic metabolism and liver regeneration, were characterized in the first phase, and two additional biological processes, immune cell growth and extracellular matrix (ECM) regulation, were observed in the second phase. Immunohistochemistry verified that the ten hBMSC-derived liver and immune cells were present in the livers of dual-humanized mice. CONCLUSIONS A syngeneic liver-immune dual-humanized mouse model was developed by transplanting a single type of hBMSC. Four biological processes linked to the transdifferentiation and biological functions of ten human liver and immune cell lineages were identified, which may help to elucidate the molecular basis of this dual-humanized mouse model for further clarifying disease pathogenesis.
Collapse
Affiliation(s)
- Suwan Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Endocrinology and Metabolic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaojiao Xin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Heng Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lunzhi Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences and School of Public Health, Xiamen University, Xiamen, China
| | - Keke Ren
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongyan Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Li
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhanglu An
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Beibei Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaxian Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lulu He
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xi Liang
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences and School of Public Health, Xiamen University, Xiamen, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences and School of Public Health, Xiamen University, Xiamen, China
| | - Jun Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
34
|
Basak M, Das K, Mahata T, Sengar AS, Verma SK, Biswas S, Bhadra K, Stewart A, Maity B. RGS7-ATF3-Tip60 Complex Promotes Hepatic Steatosis and Fibrosis by Directly Inducing TNFα. Antioxid Redox Signal 2023; 38:137-159. [PMID: 35521658 DOI: 10.1089/ars.2021.0174] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Aims: The pathophysiological mechanism(s) underlying non-alcoholic fatty liver disease (NAFLD) have yet to be fully delineated and only a single drug, peroxisome proliferator-activated receptor (PPAR) α/γ agonist saroglitazar, has been approved. Here, we sought to investigate the role of Regulator of G Protein Signaling 7 (RGS7) in hyperlipidemia-dependent hepatic dysfunction. Results: RGS7 is elevated in the livers of NAFLD patients, particularly those with severe hepatic damage, pronounced insulin resistance, and high inflammation. In the liver, RGS7 forms a unique complex with transcription factor ATF3 and histone acetyltransferase Tip60, which is implicated in NAFLD. The removal of domains is necessary for ATF3/Tip60 binding compromises RGS7-dependent reactive oxygen species generation and cell death. Hepatic RGS7 knockdown (KD) prevented ATF3/Tip60 induction, and it provided protection against fibrotic remodeling and inflammation in high-fat diet-fed mice translating to improvements in liver function. Hyperlipidemia-dependent oxidative stress and metabolic dysfunction were largely reversed in RGS7 KD mice. Interestingly, saroglitazar failed to prevent RGS7/ATF3 upregulation but it did partially restore Tip60 levels. RGS7 drives the release of particularly tumor necrosis factor α (TNFα) from isolated hepatocytes, stellate cells and its depletion reverses steatosis, oxidative stress by direct TNFα exposure. Conversely, RGS7 overexpression in the liver is sufficient to trigger oxidative stress in hepatocytes that can be mitigated via TNFα inhibition. Innovation: We discovered a novel non-canonical function for an R7RGS protein, which usually functions to regulate G protein coupled receptor (GPCR) signaling. This is the first demonstration for a functional role of RGS7 outside the retina and central nervous system. Conclusion: RGS7 represents a potential novel target for the amelioration of NAFLD. Antioxid. Redox Signal. 38, 137-159.
Collapse
Affiliation(s)
| | - Kiran Das
- Centre of Biomedical Research, Lucknow, India
| | | | | | | | - Sayan Biswas
- Department of Forensic Medicine, College of Medicine and Sagore Dutta Hospital, Kolkata, India
| | - Kakali Bhadra
- Department of Zoology, University of Kalyani, Kalyani, India
| | - Adele Stewart
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, Florida, USA
| | | |
Collapse
|
35
|
Tourkochristou E, Assimakopoulos SF, Thomopoulos K, Marangos M, Triantos C. NAFLD and HBV interplay - related mechanisms underlying liver disease progression. Front Immunol 2022; 13:965548. [PMID: 36544761 PMCID: PMC9760931 DOI: 10.3389/fimmu.2022.965548] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 11/15/2022] [Indexed: 12/08/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and Hepatitis B virus infection (HBV) constitute common chronic liver diseases with worldwide distribution. NAFLD burden is expected to grow in the coming decade, especially in western countries, considering the increased incidence of diabetes and obesity. Despite the organized HBV vaccinations and use of anti-viral therapies globally, HBV infection remains endemic and challenging public health issue. As both NAFLD and HBV have been associated with the development of progressive fibrosis, cirrhosis and hepatocellular carcinoma (HCC), the co-occurrence of both diseases has gained great research and clinical interest. The causative relationship between NAFLD and HBV infection has not been elucidated so far. Dysregulated fatty acid metabolism and lipotoxicity in NAFLD disease seems to initiate activation of signaling pathways that enhance pro-inflammatory responses and disrupt hepatocyte cell homeostasis, promoting progression of NAFLD disease to NASH, fibrosis and HCC and can affect HBV replication and immune encountering of HBV virus, which may further have impact on liver disease progression. Chronic HBV infection is suggested to have an influence on metabolic changes, which could lead to NAFLD development and the HBV-induced inflammatory responses and molecular pathways may constitute an aggravating factor in hepatic steatosis development. The observed altered immune homeostasis in both HBV infection and NAFLD could be associated with progression to HCC development. Elucidation of the possible mechanisms beyond HBV chronic infection and NAFLD diseases, which could lead to advanced liver disease or increase the risk for severe complications, in the case of HBV-NAFLD co-existence is of high clinical significance in the context of designing effective therapeutic targets.
Collapse
Affiliation(s)
- Evanthia Tourkochristou
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Stelios F. Assimakopoulos
- Division of Infectious Diseases, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Konstantinos Thomopoulos
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Markos Marangos
- Division of Infectious Diseases, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| |
Collapse
|
36
|
Aghamajidi A, Farhangnia P, Pashangzadeh S, Damavandi AR, Jafari R. Tumor-promoting myeloid cells in the pathogenesis of human oncoviruses: potential targets for immunotherapy. Cancer Cell Int 2022; 22:327. [PMID: 36303138 PMCID: PMC9608890 DOI: 10.1186/s12935-022-02727-3] [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: 07/30/2022] [Accepted: 09/25/2022] [Indexed: 11/10/2022] Open
Abstract
Oncoviruses, known as cancer-causing viruses, are typically involved in cancer progression by inhibiting tumor suppressor pathways and uncontrolled cell division. Myeloid cells are the most frequent populations recruited to the tumor microenvironment (TME) and play a critical role in cancer development and metastasis of malignant tumors. Tumor-infiltrating myeloid cells, including tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), tumor-associated dendritic cells (TADCs), and tumor-associated neutrophils (TANs) exert different states from anti-tumorigenic to pro-tumorigenic phenotypes in TME. Although their role in the anti-tumorigenic state is well introduced, their opposing roles, pro-tumorigenic activities, such as anti-inflammatory cytokine and reactive oxygen species (ROS) production, should not be ignored since they result in inflammation, tumor progression, angiogenesis, and evasion. Since the blockade of these cells had promising results against cancer progression, their inhibition might be helpful in various cancer immunotherapies. This review highlights the promoting role of tumor-associated myeloid cells (TAMCs) in the pathophysiology of human virus tumorigenesis.
Collapse
Affiliation(s)
- Azin Aghamajidi
- grid.411746.10000 0004 4911 7066Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Pooya Farhangnia
- grid.411746.10000 0004 4911 7066Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Salar Pashangzadeh
- grid.411705.60000 0001 0166 0922Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirmasoud Rayati Damavandi
- grid.411705.60000 0001 0166 0922Students’ Scientific Research Center, Exceptional Talents Development Center, Tehran University of Medical Sciences, Tehran, Iran ,grid.411705.60000 0001 0166 0922School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Jafari
- grid.412763.50000 0004 0442 8645Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
37
|
Guo J, Wang S, Gao Q. Can next-generation humanized mice that reconstituted with both functional human immune system and hepatocytes model the progression of viral hepatitis to hepatocarcinogenesis? Front Med (Lausanne) 2022; 9:1002260. [PMID: 36213658 PMCID: PMC9537463 DOI: 10.3389/fmed.2022.1002260] [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: 07/25/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022] Open
Abstract
Hepatitis B virus (HBV) and Hepatitis C virus (HCV) chronic infections cause liver immunopathological diseases such as hepatitis, fibrosis, cirrhosis, and hepatocellular carcinomas, which are difficult to treat and continue to be major health problems globally. Due to the species-specific hepato-tropism of HBV and HCV, conventional rodent models are limited in their utility for studying the infection and associated liver immunopathogenesis. Humanized mice reconstituted with both functional human immune system and hepatocytes (HIS-HuHEP mice) have been extremely instrumental for in vivo studies of HBV or HCV infection and human-specific aspects of the progression of liver immunopathogenesis. However, none of the current HIS-HuHEP mice can model the progression of viral hepatitis to hepatocarcinogenesis which may be a notorious result of HBV or HCV chronic infection in patients, suggesting that they were functionally compromised and that there is still significant space to improve and establish next-generation of HIS-HuHEP mice with more sophisticated functions. In this review, we first summarize the principal requirements to establish HIS-HuHEP mice. We then discuss the respective protocols for current HIS-HuHEP mice and their applications, as well as their advantages and disadvantages. We also raise perspectives for further improving and establishing next-generation HIS-HuHEP mice.
Collapse
Affiliation(s)
- Jinglong Guo
- Department of Cardiovascular Disease, The First Hospital of Jilin University, Changchun, China
| | - Siyue Wang
- Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, United States
| | - Qi Gao
- Department of Cardiovascular Disease, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
38
|
Xiang X, Wu Y, Lv XQ, Xu RQ, Liu Y, Pan SH, He M, Lai GQ. Hepatitis B Virus Infection Promotes M2 Polarization of Macrophages by Upregulating the Expression of B7x In Vivo and In Vitro. Viral Immunol 2022; 35:597-608. [PMID: 36099202 DOI: 10.1089/vim.2022.0029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Several studies have reported that hepatitis B virus (HBV) infection is mediated by macrophages and that the B7x (B7-H4, VTCN-1) protein plays an important role in immune regulation in HBV-associated hepatocellular carcinoma (HBV-HCC). However, the relationship among HBV, macrophages, and B7x has not been studied. In this study, HBV-infected mouse model and coculture of HBV cell lines and macrophages were used to observe the changes in macrophages and the role of B7x after HBV infection. The expression of HBV markers (HBeAg, HBsAg), negative regulator of immunity (B7x), T-helper 17 (Th17)/T-regulatory (Treg)-related cytokines, and macrophage markers, as well as changes in the apoptosis and cell cycle of macrophages were analyzed through reverse transcription quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, western blot, and flow cytometry. The expression of HBsAg, HBeAg, and B7x increased and the levels of macrophage surface marker and Treg cells secrete related cytokines (IL-10 and TGF-β) were altered after HBV infection both in vivo and in vitro. Apoptosis of macrophages increased, and cell cycle arrest occurred in vitro. These effects, except those in the cell cycle, were reversed when B7x was knocked down. Thus, HBV infection can promote the expression of B7x, which in turn regulates the Th17/Treg balance and affects the expression of HBsAg and HBeAg. The mechanism used by B7x likely involves the promotion of macrophage polarization and apoptosis. These results suggest that B7x is a novel target for HBV immunotherapy.
Collapse
Affiliation(s)
- Xia Xiang
- Laboratory Animal Center, Chongqing Medical University, Chongqing, China
| | - Yue Wu
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao-Qin Lv
- Laboratory Animal Center, Chongqing Medical University, Chongqing, China
| | - Ru-Qing Xu
- Laboratory Animal Center, Chongqing Medical University, Chongqing, China
| | - Yang Liu
- Laboratory Animal Center, Chongqing Medical University, Chongqing, China
| | - Suo-Han Pan
- The First Clinical College of Chongqing Medical University, Chongqing, China
| | - Miao He
- Laboratory Animal Center, Chongqing Medical University, Chongqing, China
| | - Guo-Qi Lai
- Laboratory Animal Center, Chongqing Medical University, Chongqing, China
| |
Collapse
|
39
|
Wei L, Cafiero TR, Tseng A, Gertje HP, Berneshawi A, Crossland NA, Ploss A. Conversion of hepatitis B virus relaxed circular to covalently closed circular DNA is supported in murine cells. JHEP Rep 2022; 4:100534. [PMID: 36035363 PMCID: PMC9403495 DOI: 10.1016/j.jhepr.2022.100534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/26/2022] [Accepted: 07/04/2022] [Indexed: 11/05/2022] Open
Abstract
Background & Aims HBV has a narrow host restriction, with humans and chimpanzees representing the only known natural hosts. The molecular correlates of resistance in species that are commonly used in biomedical research, such as mice, are currently incompletely understood. Expression of human NTCP (hNTCP) in mouse hepatocytes enables HBV entry, but subsequently covalently closed circular (cccDNA) does not form in most murine cells. It is unknown if this blockade in cccDNA formation is due to deficiency in repair of relaxed circular DNA (rcDNA) to cccDNA. Methods Here, we deployed both in vivo and in vitro virological and biochemical approaches to investigate if murine cells contain a complete set of repair factors capable of converting HBV rcDNA to cccDNA. Results We demonstrate that HBV cccDNA does form in murine cell culture or in mice when recombinant rcDNA without a protein adduct is directly introduced into cells. We further show that the murine orthologues of core components in DNA lagging strand synthesis, required for the repair of rcDNA to cccDNA in human cells, can support this crucial step in the HBV life cycle. It is worth noting that recombinant HBV rcDNA substrates, either without a protein adduct or containing neutravidin to mimic HBV polymerase, were used in our study; it remains unclear if the HBV polymerase removal processes are the same in mouse and human cells. Conclusions Collectively, our data suggest that the HBV life cycle is blocked post entry and likely before the repair stage in mouse cells, which yields critical insights that will aid in the construction of a mouse model with inbred susceptibility to HBV infection. Lay summary Hepatitis B virus (HBV) is only known to infect humans and chimpanzees in nature. Mouse models are often used in modeling disease pathogenesis and preclinical research to assess the efficacy and safety of interventions before they are then tested in human participants. However, because mice are not susceptible to HBV infection it is difficult to accurately model human infection (and test potential treatments) in mouse models. Herein, we have shown that mice are able to perform a key step in the HBV life cycle, tightening the net around the possible reason why HBV can not efficiently infect and replicate in mice.
Collapse
Key Words
- FEN-1, flap endonuclease 1
- HCC, hepatocellular carcinoma
- HDD, hydrodynamic delivery
- LIG1, DNA ligase 1
- NA-RrcDNA, neutravidin-recombinant relaxed circular DNA
- PCNA, proliferating cell nuclear antigen
- POLδ, DNA polymerase delta
- RFC, replication factor C
- RrcDNA, recombinant relaxed circular DNA
- animal model
- cccDNA, covalently closed circular DNA
- hNTCP, human sodium taurocholate co-transporting polypeptide
- hepatitis B virus
- rcDNA, relaxed circular DNA
- species tropism
- ssDNA, single-stranded DNA
- viral hepatitis
Collapse
Affiliation(s)
- Lei Wei
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Thomas R. Cafiero
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Anna Tseng
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Hans P. Gertje
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Andrew Berneshawi
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Washington Road, Princeton, NJ, 08544, 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, Lewis Thomas Laboratory, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| |
Collapse
|
40
|
Li N, Yu K, Dong M, Wang J, Yang F, Zhu H, Yu J, Yang J, Xie W, Mitra B, Mao R, Wu F, Guo H, Zhang J. Intrahepatic transcriptomics reveals gene signatures in chronic hepatitis B patients responded to interferon therapy. Emerg Microbes Infect 2022; 11:1876-1889. [PMID: 35815389 PMCID: PMC9336496 DOI: 10.1080/22221751.2022.2100831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Chronic hepatitis B virus (HBV) infection remains a substantial public health burden worldwide. Alpha-interferon (IFNα) is one of the two currently approved therapies for chronic hepatitis B (CHB), to explore the mechanisms underlying IFNα treatment response, we investigated baseline and 24-week on-treatment intrahepatic gene expression profiles in 21 CHB patients by mRNA-seq. The data analyses demonstrated that PegIFNα treatment significantly induced antiviral responses. Responders who achieved HBV DNA loss and HBeAg or HBsAg seroconversion displayed higher fold change and larger number of up-regulated interferon-stimulated genes (ISGs). Interestingly, lower expression levels of certain ISGs were observed in responders in their baseline biopsy samples. In HBeAg+ patients, non-responders had relative higher baseline HBeAg levels than responders. More importantly, HBeAg− patients showed higher HBsAg loss rate than HBeAg+ patients. Although a greater fold change of ISGs was observed in HBeAg− patients than HBeAg+ patients, upregulation of ISGs in HBeAg+ responders exceeded HBeAg− responders. Notably, PegIFNα treatment increased monocyte and mast cell infiltration, but decreased CD8 T cell and M1 macrophage infiltration in both responders and non-responders, while B cell infiltration was increased only in responders. Moreover, co-expression analysis identified ribosomal proteins as critical players in antiviral response. The data also indicate that IFNα may influence the production of viral antigens associated with endoplasmic reticulum. Collectively, the intrahepatic transcriptome analyses in this study enriched our understanding of IFN-mediated antiviral effects in CHB patients and provided novel insights into the development of potential strategies to improve IFNα therapy.
Collapse
Affiliation(s)
- Ning Li
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| | - Kangkang Yu
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| | - Minhui Dong
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinyu Wang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| | - Feifei Yang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| | - Haoxiang Zhu
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| | - Jie Yu
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| | - Jingshu Yang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| | - Wentao Xie
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| | - Bidisha Mitra
- Cancer Virology Program, UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, United States
| | - Richeng Mao
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| | - Feizhen Wu
- Key Laboratory of Epigenetics, Institutes of Biomedical Science, Fudan University, China
| | - Haitao Guo
- Cancer Virology Program, UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, United States
| | - Jiming Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Ahodantin J, Nio K, Funaki M, Zhai X, Wilson E, Kottilil S, Cheng L, Li G, Su L. Type I interferons and TGF-β cooperate to induce liver fibrosis during HIV-1 infection under antiretroviral therapy. JCI Insight 2022; 7:e152738. [PMID: 35639478 PMCID: PMC9310524 DOI: 10.1172/jci.insight.152738] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 05/25/2022] [Indexed: 11/17/2022] Open
Abstract
Liver diseases have become a major comorbidity health concern for people living with HIV-1 (PLWH) treated with combination antiretroviral therapy (cART). To investigate if HIV-1 infection and cART interact to lead to liver diseases, humanized mice reconstituted with progenitor cells from human fetal livers were infected with HIV-1 and treated with cART. We report here that chronic HIV-1 infection with cART induced hepatitis and liver fibrosis in humanized mice, associated with accumulation of M2-like macrophages (M2LMs), elevated TGF-β, and IFN signaling in the liver. Interestingly, IFN-I and TGF-β cooperatively activated human hepatic stellate cells (HepSCs) in vitro. Mechanistically, IFN-I enhanced TGF-β-induced SMAD2/3 activation in HepSCs. Finally, blockade of IFN-I signaling reversed HIV/cART-induced liver diseases in humanized mice. Consistent with the findings in humanized mice with HIV-1 and cART, we detected elevated markers of liver injury, M2LMs, and of IFN signaling in blood specimens from PLWH compared with those of healthy individuals. These findings identify the IFN-I/M2LM/HepSC axis in HIV/cART-induced liver diseases and suggest that inhibiting IFN-I signaling or M2LM may provide a novel therapeutic strategy for treating HIV/cART-associated liver diseases in PLWH treated with antiretroviral therapy.
Collapse
Affiliation(s)
- James Ahodantin
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, Departments of Pharmacology and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kouki Nio
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Masaya Funaki
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, Departments of Pharmacology and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Xuguang Zhai
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Eleanor Wilson
- Division of Clinical Care and Research, Institute of Human Virology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Shyamasundaran Kottilil
- Division of Clinical Care and Research, Institute of Human Virology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Liang Cheng
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Guangming Li
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, Departments of Pharmacology and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lishan Su
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, Departments of Pharmacology and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
42
|
Tao L, Li D, Mu S, Tian G, Yan G. LncRNA MAPKAPK5_AS1 facilitates cell proliferation in hepatitis B virus -related hepatocellular carcinoma. J Transl Med 2022; 102:494-504. [PMID: 35264707 DOI: 10.1038/s41374-022-00731-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/14/2021] [Accepted: 12/27/2021] [Indexed: 11/09/2022] Open
Abstract
We explored the biological role of long non-coding RNA (lncRNA) MAPKAPK5_AS1 (MAAS) and the mechanism of its differential expression in hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC). Differentially expressed lncRNAs in HBV-related HCC were determined using bioinformatics analysis. Gain-of-function experiments were conducted to evaluate the effect of MAAS on cell proliferation. A xenograft model was established for in vivo experiments. Dual-luciferase reporter assays, chromatin immunoprecipitation, co-immunoprecipitation, and methylated RNA immunoprecipitation were performed to elucidate the underlying molecular mechanisms. MAAS was upregulated in HBV-related HCC cancerous tissues and its high expression was closely related to the poor survival probability of patients. Functional assays revealed that MAAS overexpression facilitated the proliferation of HBV+HCC cells in vitro and in vivo. Mechanistically, MAAS promoted the MYC proto-oncogene (c-Myc)-induced transcriptional activation of cyclin-dependent kinase 4 (CDK4), CDK6, and S-phase kinase associated protein 2 via stabilizing c-Myc protein, thereby facilitating G1/S transition. The latter contributed to the paradoxical proliferation of HBV+HCC cells. Although MAAS was upregulated in HBV-related HCC cancerous tissues, it was highly expressed in M2 macrophages, a major phenotype of tumor-associated macrophages in HBV-related HCC, instead of in HBV+HCC cells. HBeAg, an HBV-associated antigen, further elevated the MAAS level in M2 macrophages by enhancing the methyltransferase-like 3-mediated N6-methyladenosine modification of MAAS. The increased MAAS in the M2 macrophages was then transferred to HBV+HCC cells through the M2 macrophage-derived exosomes, promoting cell proliferation. Our findings show that HBV+HCC cell-secreted HBeAg upregulates MAAS expression in M2 macrophages by affecting its m6A modification. The upregulated MAAS is then transferred to HBV+HCC cells via exosomes, facilitating the proliferation of HBV+HCC cells by targeting c-Myc.
Collapse
Affiliation(s)
- Lianyuan Tao
- Department of Hepatobiliary Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, 450003, China.,Henan Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Henan Provincial People's Hospital, Zhengzhou, China.,Zhengzhou Key Laboratory of Minimally Invasive Treatment for Liver Cancer, Henan Provincial People's Hospital, Zhengzhou, China
| | - Deyu Li
- Department of Hepatobiliary Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, 450003, China. .,Henan Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Henan Provincial People's Hospital, Zhengzhou, China. .,Zhengzhou Key Laboratory of Minimally Invasive Treatment for Liver Cancer, Henan Provincial People's Hospital, Zhengzhou, China.
| | - Sengmao Mu
- Department of Hepatobiliary Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, 450003, China
| | - Guanjing Tian
- Department of Hepatobiliary Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, 450003, China
| | - Guoyi Yan
- Department of Hepatobiliary Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, 450003, China.,Henan Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Henan Provincial People's Hospital, Zhengzhou, China.,Zhengzhou Key Laboratory of Minimally Invasive Treatment for Liver Cancer, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
43
|
Li J, Yu M, Zong R, Fan C, Ren F, Wu W, Li C. Deacetylation of Notch1 by SIRT1 contributes to HBsAg- and HBeAg-mediated M2 macrophage polarization. Am J Physiol Gastrointest Liver Physiol 2022; 322:G459-G471. [PMID: 35234049 DOI: 10.1152/ajpgi.00338.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatic macrophages are involved in both pathogen clearance and immunopathogenesis. Emerging evidence demonstrates that macrophage polarization plays a critical role in hepatitis B virus (HBV)-induced immune impairment and liver pathology. However, it remains largely unknown as to how HBV infection facilitates M2 macrophage polarization. Here, a mouse HBV infection model was established by hydrodynamic injection with a vector containing 1.3-fold overlength HBV genome via the tail vein. Coculture experiments with HBV-producing HepG2.2.15 cells and macrophages were established in vitro. We found that HBV-inhibited M1 while enhancing M2 markers, which was accompanied by decreased proinflammatory tumor necrosis factor-α (TNF-α) and augmented anti-inflammatory IL-10 expression. Furthermore, both hepatitis B surface antigen (HBsAg) and hepatitis B e antigen (HBeAg) secretion contributed to HBV-triggered macrophage polarization from M1 toward M2 phenotype. Mechanistically, HBsAg and HBeAg could upregulate the sirtuins 1 (SIRT1) deacetylase expression, which in turn promote deacetylation of the Notch1 intracellular domain (NICD), leading to increased Akt phosphorylation and decreased NF-κB nuclear translocation in macrophages. Our findings suggest that NICD deacetylation by SIRT1 contributes to HBsAg- and HBeAg-mediated M2 macrophage polarization, raising the possibility of targeting SIRT1/Notch1 pathway in macrophages to treat HBV immune evasion and chronic HBV infection.NEW & NOTEWORTHY This study identified a previously unrecognized molecular mechanism of HBV-mediated suppression of innate immune responses. We demonstrate that deacetylation of NICD by SIRT1 contributes to HBsAg- and HBeAg-mediated M2 macrophage polarization, which may aid in the development of new macrophage-based immunotherapy for chronic HBV infection and related diseases.
Collapse
Affiliation(s)
- Jiahui Li
- Department of Anatomy and Histology Embryology, Jinzhou Medical University, Jinzhou, People's Republic of China.,Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, People's Republic of China.,Department of Anatomy, Youjiang Medical University for Nationalities, Baise, People's Republic of China
| | - Mengxue Yu
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Ruobin Zong
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Chengpeng Fan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Fu Ren
- Department of Anatomy, Shenyang Medical College, Shenyang, People's Republic of China
| | - Wei Wu
- Institute of Humanities and Social Sciences, Shenyang University, Shenyang, People's Republic of China
| | - Changyong Li
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
44
|
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: 3] [Impact Index Per Article: 1.0] [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
|
45
|
Yu S, Ge H, Li S, Qiu HJ. Modulation of Macrophage Polarization by Viruses: Turning Off/On Host Antiviral Responses. Front Microbiol 2022; 13:839585. [PMID: 35222345 PMCID: PMC8874017 DOI: 10.3389/fmicb.2022.839585] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/12/2022] [Indexed: 11/17/2022] Open
Abstract
Macrophages are professional antigen-presenting cells and serve as the first line of defense against invading pathogens. Macrophages are polarized toward the proinflammatory classical (M1) or anti-inflammatory alternative (M2) phenotype upon viral infections. M1-polarized macrophages exert critical roles in antiviral responses via different mechanisms. Within the long competitive history between viruses and hosts, viruses have evolved various immune evasion strategies, inhibiting macrophage acquisition of an antiviral phenotype, impairing the antiviral responses of activated macrophages, and/or exploiting macrophage phenotypes for efficient replication. This review focuses on the sophisticated regulation of macrophage polarization utilized by viruses and is expected to provide systematic insights into the regulatory mechanisms of macrophage polarization by viruses and further facilitate the design of therapeutic targets for antivirals.
Collapse
|
46
|
Abstract
Hepatitis B virus (HBV) is a hepatotropic virus and an important human pathogen. There are an estimated 296 million people in the world that are chronically infected by this virus, and many of them will develop severe liver diseases including hepatitis, cirrhosis and hepatocellular carcinoma (HCC). HBV is a small DNA virus that replicates via the reverse transcription pathway. In this review, we summarize the molecular pathways that govern the replication of HBV and its interactions with host cells. We also discuss viral and non-viral factors that are associated with HBV-induced carcinogenesis and pathogenesis, as well as the role of host immune responses in HBV persistence and liver pathogenesis.
Collapse
Affiliation(s)
- Yu-Chen Chuang
- Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, CA 90089, USA
| | - Kuen-Nan Tsai
- Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, CA 90089, USA
| | - Jing-Hsiung James Ou
- Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, CA 90089, USA
| |
Collapse
|
47
|
Macrophages activated by hepatitis B virus have distinct metabolic profiles and suppress the virus via IL-1β to downregulate PPARα and FOXO3. Cell Rep 2022; 38:110284. [PMID: 35081341 PMCID: PMC8830375 DOI: 10.1016/j.celrep.2021.110284] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/30/2021] [Accepted: 12/28/2021] [Indexed: 01/15/2023] Open
Abstract
Macrophages display phenotypic plasticity and can be induced by hepatitis B virus (HBV) to undergo either M1-like pro-inflammatory or M2-like anti-inflammatory polarization. Here, we report that M1-like macrophages stimulated by HBV exhibit a strong HBV-suppressive effect, which is diminished in M2-like macrophages. Transcriptomic analysis reveals that HBV induces the expression of interleukin-1β (IL-1β) in M1-like macrophages, which display a high oxidative phosphorylation (OXPHOS) activity distinct from that of conventional M1-like macrophages. Further analysis indicates that OXPHOS attenuates the expression of IL-1β, which suppresses the expression of peroxisome proliferator-activated receptor α (PPARα) and forkhead box O3 (FOXO3) in hepatocytes to suppress HBV gene expression and replication. Moreover, multiple HBV proteins can induce the expression of IL-1β in macrophages. Our results thus indicate that macrophages can respond to HBV by producing IL-1β to suppress HBV replication. However, HBV can also metabolically reprogram macrophages to enhance OXPHOS to minimize this host antiviral response.
Collapse
|
48
|
Luo Y, Lu H, Peng D, Ruan X, Chen YE, Guo Y. Liver-humanized mice: A translational strategy to study metabolic disorders. J Cell Physiol 2022; 237:489-506. [PMID: 34661916 PMCID: PMC9126562 DOI: 10.1002/jcp.30610] [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: 07/14/2021] [Revised: 09/07/2021] [Accepted: 09/11/2021] [Indexed: 01/03/2023]
Abstract
The liver is the metabolic core of the whole body. Tools commonly used to study the human liver metabolism include hepatocyte cell lines, primary human hepatocytes, and pluripotent stem cells-derived hepatocytes in vitro, and liver genetically humanized mouse model in vivo. However, none of these systems can mimic the human liver in physiological and pathological states satisfactorily. Liver-humanized mice, which are established by reconstituting mouse liver with human hepatocytes, have emerged as an attractive animal model to study drug metabolism and evaluate the therapeutic effect in "human liver" in vivo because the humanized livers greatly replicate enzymatic features of human hepatocytes. The application of liver-humanized mice in studying metabolic disorders is relatively less common due to the largely uncertain replication of metabolic profiles compared to humans. Here, we summarize the metabolic characteristics and current application of liver-humanized mouse models in metabolic disorders that have been reported in the literature, trying to evaluate the pros and cons of using liver-humanized mice as novel mouse models to study metabolic disorders.
Collapse
Affiliation(s)
- Yonghong Luo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Haocheng Lu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiangbo Ruan
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins School of Medicine, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA
| | - Y. Eugene Chen
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Center for Advanced Models and Translational Sciences and Therapeutics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yanhong Guo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
49
|
Du Y, Broering R, Li X, Zhang X, Liu J, Yang D, Lu M. In Vivo Mouse Models for Hepatitis B Virus Infection and Their Application. Front Immunol 2021; 12:766534. [PMID: 34777385 PMCID: PMC8586444 DOI: 10.3389/fimmu.2021.766534] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 10/14/2021] [Indexed: 12/19/2022] Open
Abstract
Despite the availability of effective vaccination, hepatitis B virus (HBV) infection continues to be a major challenge worldwide. Research efforts are ongoing to find an effective cure for the estimated 250 million people chronically infected by HBV in recent years. The exceptionally limited host spectrum of HBV has limited the research progress. Thus, different HBV mouse models have been developed and used for studies on infection, immune responses, pathogenesis, and antiviral therapies. However, these mouse models have great limitations as no spread of HBV infection occurs in the mouse liver and no or only very mild hepatitis is present. Thus, the suitability of these mouse models for a given issue and the interpretation of the results need to be critically assessed. This review summarizes the currently available mouse models for HBV research, including hydrodynamic injection, viral vector-mediated transfection, recombinant covalently closed circular DNA (rc-cccDNA), transgenic, and liver humanized mouse models. We systematically discuss the characteristics of each model, with the main focus on hydrodynamic injection mouse model. The usefulness and limitations of each mouse model are discussed based on the published studies. This review summarizes the facts for considerations of the use and suitability of mouse model in future HBV studies.
Collapse
Affiliation(s)
- Yanqin Du
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ruth Broering
- Department of Gastroenterology and Hepatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Xiaoran Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoyong Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jia Liu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongliang Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
50
|
In Vivo Bioluminescence Imaging of HBV Replicating Hepatocytes Allows for the Monitoring of Anti-Viral Immunity. Viruses 2021; 13:v13112273. [PMID: 34835079 PMCID: PMC8619421 DOI: 10.3390/v13112273] [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: 10/01/2021] [Revised: 11/03/2021] [Accepted: 11/11/2021] [Indexed: 11/24/2022] Open
Abstract
Immunity against hepatitis B virus (HBV) infection is complex and not entirely understood so far, including the decisive factors leading to the development of chronic hepatitis B. This lack of a mechanistic understanding of HBV-specific immunity is also caused by a limited number of suitable animal models. Here, we describe the generation of a recombinant adenovirus expressing an HBV 1.3-overlength genome linked to luciferase (Ad-HBV-Luc) allowing for precise analysis of the quantity of infected hepatocytes. This enables sensitive and close-meshed monitoring of HBV-specific CD8 T cells and the onset of anti-viral immunity in mice. A high dose of Ad-HBV-Luc developed into chronic hepatitis B accompanied by dysfunctional CD8 T cells characterized by high expression of PD1 and TOX and low expression of KLRG1 and GzmB. In contrast, a low dose of Ad-HBV-Luc infection resulted in acute hepatitis with CD8 T cell-mediated elimination of HBV-replicating hepatocytes associated with elevated sALT levels and increased numbers of cytotoxic HBV-specific CD8 T cells. Thus, the infectious dose was a critical factor to induce either acute self-limited or chronic HBV infection in mice. Taken together, the new Ad-HBV-Luc vector will allow for highly sensitive and time-resolved analysis of HBV-specific immune responses during acute and chronic infection.
Collapse
|