1
|
Zeng Q, Sun P, Li W, Tang Y, Hu Y, Zhou J, Zhou Y, Chen L, Yimou W. Protective immunity induced by a novel P1 adhesin C-terminal anchored mRNA vaccine against Mycoplasma pneumoniae infection in BALB/c mice. Microbiol Spectr 2025; 13:e0214024. [PMID: 39831768 DOI: 10.1128/spectrum.02140-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025] Open
Abstract
Mycoplasma pneumoniae (Mp), a unique pathogen devoid of a cell wall, is naturally impervious to penicillin antibiotics. This bacterium is the causative agent of M. pneumoniae pneumonia, an acute pulmonary affliction marked by interstitial lung damage. Non-macrolide medications may have potential adverse effects on the developmental trajectory of children, thereby establishing macrolides as the preferred treatment for M. pneumoniae in pediatric patients. However, the emergence of macrolide-resistant and multidrug-resistant strains of M. pneumoniae presents significant challenges to clinical management and public health. Vaccines, particularly those based on mRNA technology, are regarded as a promising avenue for preventing and controlling M. pneumoniae infections due to their inherent safety, immunogenicity, and adaptability. Our research delves into the P1 adhesin of M. pneumoniae, a protein that binds to host cell receptors with its immunodominant epitopes located at the carboxyl terminus, known to provoke robust immune responses and pulmonary inflammation. We have developed an mRNA vaccine harnessing this dominant antigenic epitope and assessed its protective immunity in BALB/c mice against M. pneumoniae infection. The vaccine elicited potent humoral and cellular immune responses, effectively diminishing inflammation. It notably decreased IL-6 levels in the lungs of infected mice and concurrently elevated IL-4, IL-10, and IFN-γ levels post-immunization. The vaccine also reduced pathological changes in the lungs and the M. pneumoniae DNA copy numbers in the infected animals. Collectively, these findings underscore the mRNA vaccine's remarkable immunogenicity and protective potential against M. pneumoniae infections, offering valuable insights for the development of mRNA vaccines targeting mycoplasma infections.IMPORTANCEM. pneumoniae, a bacteria without a cell wall, is known for causing pneumonia and is resistant to penicillin. The increasing prevalence of macrolide-resistant strains has complicated treatment options, emphasizing the need for new strategies. Our research explores an mRNA vaccine candidate that targets the P1 adhesin of M. pneumoniae, a protein critical for the bacteria's interaction with host cells. In a mouse model, this vaccine has shown potential by inducing immune responses and suggesting a possible reduction in inflammation, as indicated by changes in cytokine levels and lung pathology. While further research is required, the vaccine's preliminary results hint at a potential new direction in managing mycoplasma infections, offering a promising avenue for future therapeutic development. This study contributes to the ongoing search for effective preventive measures against M. pneumoniae.
Collapse
MESH Headings
- Animals
- Mice, Inbred BALB C
- Mycoplasma pneumoniae/immunology
- Mycoplasma pneumoniae/genetics
- Mice
- Pneumonia, Mycoplasma/immunology
- Pneumonia, Mycoplasma/prevention & control
- Pneumonia, Mycoplasma/microbiology
- Adhesins, Bacterial/immunology
- Adhesins, Bacterial/genetics
- Female
- Bacterial Vaccines/immunology
- Bacterial Vaccines/genetics
- Bacterial Vaccines/administration & dosage
- mRNA Vaccines
- Antibodies, Bacterial/immunology
- Antibodies, Bacterial/blood
- Immunity, Cellular
- Cytokines/metabolism
- Immunity, Humoral
- Humans
- Lung/microbiology
- Lung/immunology
- Lung/pathology
Collapse
Affiliation(s)
- Qilin Zeng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| | - Peiyuan Sun
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| | - Weiwei Li
- Department of Clinical Laboratory, The Second People's Hospital of Foshan, Foshan, China
| | - Yuanyuan Tang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| | - Yuxuan Hu
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Jun Zhou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| | - Yanxia Zhou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| | - Liesong Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| | - Wu Yimou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, China
| |
Collapse
|
2
|
Sharma P, Chaudhary NR, Devi S, Negi S, Tandel N, Tyagi RK. Oleuropein mediated autophagy begets antimalarial drug resistance. Front Microbiol 2024; 15:1453998. [PMID: 39228384 PMCID: PMC11369837 DOI: 10.3389/fmicb.2024.1453998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024] Open
Abstract
Drug resistance in Plasmodium falciparum presents a formidable challenge to the humanity. And, unavailability of an effective vaccine worsens the situation further. Autophagy is one of the mechanisms employed by parasite to evade drug pressure to survive. Autophagy induced by the P. falciparum in response to the oleuropein pressure may answer many questions related to the parasite survival as well as evolving drug tolerance. The survival/autophagy axis could be an important avenue to explore in order to address certain questions related to the evolution of drug resistance. In addition, humanized mouse model of P. falciparum infection could serve as an important preclinical tool to investigate the oleuropein-induced autophagy, potentially helping to dissect the mechanisms underlying the development of antimalarial drug resistance.
Collapse
Affiliation(s)
- Prakriti Sharma
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-Immunology Lab, CSIR Institute of Microbial Technology (IMTECH), Chandigarh, India
| | - Neil Roy Chaudhary
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-Immunology Lab, CSIR Institute of Microbial Technology (IMTECH), Chandigarh, India
| | - Sonia Devi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-Immunology Lab, CSIR Institute of Microbial Technology (IMTECH), Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sushmita Negi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-Immunology Lab, CSIR Institute of Microbial Technology (IMTECH), Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Nikunj Tandel
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Rajeev K. Tyagi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-Immunology Lab, CSIR Institute of Microbial Technology (IMTECH), Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
3
|
Li YT, Mei KC, Liam-Or R, Wang JTW, Faruqu FN, Zhu S, Wang YL, Lu Y, Al-Jamal KT. Graphene Oxide Nanosheets Toxicity in Mice Is Dependent on Protein Corona Composition and Host Immunity. ACS NANO 2024; 18:22572-22585. [PMID: 39110092 PMCID: PMC11342366 DOI: 10.1021/acsnano.4c08561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024]
Abstract
Two-dimension graphene oxide (GO) nanosheets with high and low serum protein binding profiles (high/low hard-bound protein corona/HChigh/low) are used in this study as model materials and screening tools to investigate the underlying roles of the protein corona on nanomaterial toxicities in vivo. We proposed that the in vivo biocompatibility/nanotoxicity of GO is protein corona-dependent and host immunity-dependent. The hypothesis was tested by injecting HChigh/low GO nanosheets in immunocompetent ICR/CD1 and immunodeficient NOD-scid II2rγnull mice and performed histopathological and hematological evaluation studies on days 1 and 14 post-injection. HClow GO induced more severe acute lung injury compared to HChigh GO in both immunocompetent and immunodeficient mice, with the effect being particularly pronounced in immunocompetent animals. Additionally, HClow GO caused more significant liver injury in both types of mice, with immunodeficient mice being more susceptible to its hepatotoxic effects. Moreover, administration of HClow GO resulted in increased hematological toxicity and elevated levels of serum pro-inflammatory cytokines in immunocompromised and immunocompetent mice, respectively. Correlation studies were conducted to explore the impact of distinct protein corona compositions on resulting toxicities in both immunocompetent and immunodeficient mice. This facilitated the identification of consistent patterns, aligning with those observed in vitro, thus indicating a robust in vitro-in vivo correlation. This research will advance our comprehension of how hard corona proteins interact with immune cells, leading to toxicity, and will facilitate the development of improved immune-modulating nanomaterials for therapeutic purposes.
Collapse
Affiliation(s)
- Yue-ting Li
- School
of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences
& Medicine, King’s College London, Franklin-Wilkins Building, London SE1 9NH, U.K.
- State
Key Laboratory of Functions and Applications of Medicinal Plants,
Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, No. 9, Beijing Road, Yunyan District, Guiyang 550004, China
| | - Kuo-Ching Mei
- School
of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences
& Medicine, King’s College London, Franklin-Wilkins Building, London SE1 9NH, U.K.
- School
of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, 96 Corliss Avenue, Johnson City, New York 13790, United States
| | - Revadee Liam-Or
- School
of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences
& Medicine, King’s College London, Franklin-Wilkins Building, London SE1 9NH, U.K.
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong SAR, China
| | - Julie Tzu-Wen Wang
- School
of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences
& Medicine, King’s College London, Franklin-Wilkins Building, London SE1 9NH, U.K.
| | - Farid N. Faruqu
- School
of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences
& Medicine, King’s College London, Franklin-Wilkins Building, London SE1 9NH, U.K.
| | - Shengzhang Zhu
- Qiannan
People’s Hospital, No. 9, Wenfeng Road, Duyun 558000, China
| | - Yong-lin Wang
- State
Key Laboratory of Functions and Applications of Medicinal Plants,
Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, No. 9, Beijing Road, Yunyan District, Guiyang 550004, China
| | - Yuan Lu
- School
of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences
& Medicine, King’s College London, Franklin-Wilkins Building, London SE1 9NH, U.K.
- State
Key Laboratory of Functions and Applications of Medicinal Plants,
Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, No. 9, Beijing Road, Yunyan District, Guiyang 550004, China
| | - Khuloud T. Al-Jamal
- School
of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences
& Medicine, King’s College London, Franklin-Wilkins Building, London SE1 9NH, U.K.
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong SAR, China
| |
Collapse
|
4
|
Wadhwa A, Moreno-Villanueva M, Crucian B, Wu H. Synergistic interplay between radiation and microgravity in spaceflight-related immunological health risks. Immun Ageing 2024; 21:50. [PMID: 39033285 PMCID: PMC11264846 DOI: 10.1186/s12979-024-00449-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 06/21/2024] [Indexed: 07/23/2024]
Abstract
Spaceflight poses a myriad of environmental stressors to astronauts´ physiology including microgravity and radiation. The individual impacts of microgravity and radiation on the immune system have been extensively investigated, though a comprehensive review on their combined effects on immune system outcomes is missing. Therefore, this review aims at understanding the synergistic, additive, and antagonistic interactions between microgravity and radiation and their impact on immune function as observed during spaceflight-analog studies such as rodent hindlimb unloading and cell culture rotating wall vessel models. These mimic some, but not all, of the physiological changes observed in astronauts during spaceflight and provide valuable information that should be considered when planning future missions. We provide guidelines for the design of further spaceflight-analog studies, incorporating influential factors such as age and sex for rodent models and standardizing the longitudinal evaluation of specific immunological alterations for both rodent and cellular models of spaceflight exposure.
Collapse
Affiliation(s)
- Anna Wadhwa
- Harvard Medical School, Boston, MA, 02115, USA.
- NASA Johnson Space Center, Houston, TX, 77058, USA.
| | | | | | - Honglu Wu
- NASA Johnson Space Center, Houston, TX, 77058, USA
| |
Collapse
|
5
|
Bhargavi G, Subbian S. The causes and consequences of trained immunity in myeloid cells. Front Immunol 2024; 15:1365127. [PMID: 38665915 PMCID: PMC11043514 DOI: 10.3389/fimmu.2024.1365127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Conventionally, immunity in humans has been classified as innate and adaptive, with the concept that only the latter type has an immunological memory/recall response against specific antigens or pathogens. Recently, a new concept of trained immunity (a.k.a. innate memory response) has emerged. According to this concept, innate immune cells can exhibit enhanced responsiveness to subsequent challenges, after initial stimulation with antigen/pathogen. Thus, trained immunity enables the innate immune cells to respond robustly and non-specifically through exposure or re-exposure to antigens/infections or vaccines, providing enhanced resistance to unrelated pathogens or reduced infection severity. For example, individuals vaccinated with BCG to protect against tuberculosis were also protected from malaria and SARS-CoV-2 infections. Epigenetic modifications such as histone acetylation and metabolic reprogramming (e.g. shift towards glycolysis) and their inter-linked regulations are the key factors underpinning the immune activation of trained cells. The integrated metabolic and epigenetic rewiring generates sufficient metabolic intermediates, which is crucial to meet the energy demand required to produce proinflammatory and antimicrobial responses by the trained cells. These factors also determine the efficacy and durability of trained immunity. Importantly, the signaling pathways and regulatory molecules of trained immunity can be harnessed as potential targets for developing novel intervention strategies, such as better vaccines and immunotherapies against infectious (e.g., sepsis) and non-infectious (e.g., cancer) diseases. However, aberrant inflammation caused by inappropriate onset of trained immunity can lead to severe autoimmune pathological consequences, (e.g., systemic sclerosis and granulomatosis). In this review, we provide an overview of conventional innate and adaptive immunity and summarize various mechanistic factors associated with the onset and regulation of trained immunity, focusing on immunologic, metabolic, and epigenetic changes in myeloid cells. This review underscores the transformative potential of trained immunity in immunology, paving the way for developing novel therapeutic strategies for various infectious and non-infectious diseases that leverage innate immune memory.
Collapse
Affiliation(s)
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| |
Collapse
|
6
|
Zhao G, Bu G, Liu G, Kong X, Sun C, Li Z, Dai D, Sun H, Kang Y, Feng G, Zhong Q, Zeng M. mRNA-based Vaccines Targeting the T-cell Epitope-rich Domain of Epstein Barr Virus Latent Proteins Elicit Robust Anti-Tumor Immunity in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302116. [PMID: 37890462 PMCID: PMC10724410 DOI: 10.1002/advs.202302116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 09/04/2023] [Indexed: 10/29/2023]
Abstract
Epstein-Barr virus (EBV) is associated with various malignancies and infects >90% of the global population. EBV latent proteins are expressed in numerous EBV-associated cancers and contribute to carcinogenesis, making them critical therapeutic targets for these cancers. Thus, this study aims to develop mRNA-based therapeutic vaccines that express the T-cell-epitope-rich domain of truncated latent proteins of EBV, including truncatedlatent membrane protein 2A (Trunc-LMP2A), truncated EBV nuclear antigen 1 (Trunc-EBNA1), and Trunc-EBNA3A. The vaccines effectively activate both cellular and humoral immunity in mice and show promising results in suppressing tumor progression and improving survival time in tumor-bearing mice. Furthermore, it is observed that the truncated forms of the antigens, Trunc-LMP2A, Trunc-EBNA1, and Trunc-EBNA3A, are more effective than full-length antigens in activating antigen-specific immune responses. In summary, the findings demonstrate the effectiveness of mRNA-based therapeutic vaccines targeting the T-cell-epitope-rich domain of EBV latent proteins and providing new treatment options for EBV-associated cancers.
Collapse
Affiliation(s)
- Ge‐Xin Zhao
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer. MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma, Diagnosis, and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Guo‐Long Bu
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer. MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma, Diagnosis, and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Gang‐Feng Liu
- Department of Head and Neck Surgery Section IIThe Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Hospital519 Kunzhou RoadKunming650118China
| | - Xiang‐Wei Kong
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer. MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma, Diagnosis, and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Cong Sun
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer. MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma, Diagnosis, and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Zi‐Qian Li
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer. MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma, Diagnosis, and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Dan‐Ling Dai
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer. MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma, Diagnosis, and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Hai‐Xia Sun
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer. MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma, Diagnosis, and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Yin‐Feng Kang
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer. MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma, Diagnosis, and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Guo‐Kai Feng
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer. MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma, Diagnosis, and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Qian Zhong
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer. MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma, Diagnosis, and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Mu‐Sheng Zeng
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer. MedicineGuangdong Key Laboratory of Nasopharyngeal Carcinoma, Diagnosis, and TherapySun Yat‐sen University Cancer CenterGuangzhou510060China
- Guangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen University Cancer CenterGuangzhou510060China
| |
Collapse
|
7
|
Marchante M, Ramirez-Martin N, Buigues A, Martinez J, Pellicer N, Pellicer A, Herraiz S. Deciphering reproductive aging in women using a NOD/SCID mouse model for distinct physiological ovarian phenotypes. Aging (Albany NY) 2023; 15:10856-10874. [PMID: 37847151 PMCID: PMC10637815 DOI: 10.18632/aging.205086] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/08/2023] [Indexed: 10/18/2023]
Abstract
Female fertility is negatively correlated with age, with noticeable declines in oocyte quantity and quality until menopause. To understand this physiological process and evaluate human approaches for treating age-related infertility, preclinical studies in appropriate animal models are needed. Thus, we aimed to characterize an immunodeficient physiological aging mouse model displaying ovarian characteristics of different stages during women's reproductive life. NOD/SCID mice of different ages (8-, 28-, and 36-40-week-old) were employed to mimic ovarian phenotypes of young, Advanced Maternal Age (AMA), and old women (~18-20-, ~36-38-, and >45-years-old, respectively). Mice were stimulated, mated, and sacrificed to recover oocytes and embryos. Then, ovarian reserve, follicular growth, ovarian stroma, mitochondrial dysfunction, and proteomic profiles were assessed. Age-matched C57BL/6 mice were employed to cross-validate the reproductive outcomes. The quantity and quality of oocytes were decreased in AMA and Old mice. These age-related effects associated spindle and chromosome abnormalities, along with decreased developmental competence to blastocyst stage. Old mice had less follicles, impaired follicle activation and growth, an ovarian stroma inconducive to growth, and increased mitochondrial dysfunctions. Proteomic analysis corroborated these histological findings. Based on that, NOD/SCID mice can be used to model different ovarian aging phenotypes and potentially test human anti-aging treatments.
Collapse
Affiliation(s)
- María Marchante
- IVIRMA Global Research Alliance, IVI Foundation, Valencia 46026, Spain
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia 46010, Spain
| | - Noelia Ramirez-Martin
- IVIRMA Global Research Alliance, IVI Foundation, Valencia 46026, Spain
- Reproductive Medicine Research Group, Instituto Investigación Sanitaria La Fe (IIS La Fe), Valencia 46026, Spain
| | - Anna Buigues
- IVIRMA Global Research Alliance, IVI Foundation, Valencia 46026, Spain
- Reproductive Medicine Research Group, Instituto Investigación Sanitaria La Fe (IIS La Fe), Valencia 46026, Spain
| | - Jessica Martinez
- IVIRMA Global Research Alliance, IVI Foundation, Valencia 46026, Spain
- Reproductive Medicine Research Group, Instituto Investigación Sanitaria La Fe (IIS La Fe), Valencia 46026, Spain
| | - Nuria Pellicer
- IVIRMA Global Research Alliance, IVI Foundation, Valencia 46026, Spain
- IVIRMA Valencia, Valencia 46015, Spain
| | - Antonio Pellicer
- IVIRMA Global Research Alliance, IVI Foundation, Valencia 46026, Spain
- Reproductive Medicine Research Group, Instituto Investigación Sanitaria La Fe (IIS La Fe), Valencia 46026, Spain
- IVIRMA Rome, Rome 00197, Italy
| | - Sonia Herraiz
- IVIRMA Global Research Alliance, IVI Foundation, Valencia 46026, Spain
- Reproductive Medicine Research Group, Instituto Investigación Sanitaria La Fe (IIS La Fe), Valencia 46026, Spain
| |
Collapse
|
8
|
Li K, Wang M, Akoglu M, Pollard AC, Klecker JB, Alfonso P, Corrionero A, Prendiville N, Qu W, Parker MFL, Turkman N, Cohen JA, Tonge PJ. Synthesis and Preclinical Evaluation of a Novel Fluorine-18-Labeled Tracer for Positron Emission Tomography Imaging of Bruton's Tyrosine Kinase. ACS Pharmacol Transl Sci 2023; 6:410-421. [PMID: 36926452 PMCID: PMC10012250 DOI: 10.1021/acsptsci.2c00215] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Indexed: 02/12/2023]
Abstract
Bruton's tyrosine kinase (BTK) is a target for treating B-cell malignancies and autoimmune diseases. To aid in the discovery and development of BTK inhibitors and improve clinical diagnoses, we have developed a positron emission tomography (PET) radiotracer based on a selective BTK inhibitor, remibrutinib. [18F]PTBTK3 is an aromatic, 18F-labeled tracer that was synthesized in 3 steps with a 14.8 ± 2.4% decay-corrected radiochemical yield and ≥99% radiochemical purity. The cellular uptake of [18F]PTBTK3 was blocked up to 97% in JeKo-1 cells using remibrutinib or non-radioactive PTBTK3. [18F]PTBTK3 exhibited renal and hepatobiliary clearance in NOD SCID (non-obese diabetic/severe combined immunodeficiency) mice, and the tumor uptake of [18F]PTBTK3 in BTK-positive JeKo-1 xenografts (1.23 ± 0.30% ID/cc) was significantly greater at 60 min post injection compared to the tumor uptake in BTK-negative U87MG xenografts (0.41 ± 0.11% ID/cc). In the JeKo-1 xenografts, tumor uptake was blocked up to 62% by remibrutinib, indicating the BTK-dependent uptake of [18F]PTBTK3 in tumors.
Collapse
Affiliation(s)
- Kaixuan Li
- Center
for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, John S. Toll Drive, Stony
Brook, New York 11794-3400, United States
| | - Mingqian Wang
- Center
for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, John S. Toll Drive, Stony
Brook, New York 11794-3400, United States
| | - Melike Akoglu
- Center
for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, John S. Toll Drive, Stony
Brook, New York 11794-3400, United States
| | - Alyssa C. Pollard
- Center
for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, John S. Toll Drive, Stony
Brook, New York 11794-3400, United States
| | - John B. Klecker
- Center
for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, John S. Toll Drive, Stony
Brook, New York 11794-3400, United States
| | - Patricia Alfonso
- Enzymlogic
S.L., QUBE Technology
Park, C/Santiago Grisolía, 2, 28760 Madrid, Spain
| | - Ana Corrionero
- Enzymlogic
S.L., QUBE Technology
Park, C/Santiago Grisolía, 2, 28760 Madrid, Spain
| | - Niall Prendiville
- Enzymlogic
S.L., QUBE Technology
Park, C/Santiago Grisolía, 2, 28760 Madrid, Spain
| | - Wenchao Qu
- Center
for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, John S. Toll Drive, Stony
Brook, New York 11794-3400, United States
- Department
of Psychiatry, Department of Radiology, Department of Medicine, Stony Brook Cancer
Center, and Facility of Experimental Radiopharmaceutical Manufacturing (FERM), Stony Brook Renaissance School of Medicine, Stony
Brook University, Stony
Brook, New York 11794, United States
| | - Matthew F. L. Parker
- Center
for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, John S. Toll Drive, Stony
Brook, New York 11794-3400, United States
- Department
of Psychiatry, Department of Radiology, Department of Medicine, Stony Brook Cancer
Center, and Facility of Experimental Radiopharmaceutical Manufacturing (FERM), Stony Brook Renaissance School of Medicine, Stony
Brook University, Stony
Brook, New York 11794, United States
| | - Nashaat Turkman
- Department
of Psychiatry, Department of Radiology, Department of Medicine, Stony Brook Cancer
Center, and Facility of Experimental Radiopharmaceutical Manufacturing (FERM), Stony Brook Renaissance School of Medicine, Stony
Brook University, Stony
Brook, New York 11794, United States
| | - Jules A. Cohen
- Department
of Psychiatry, Department of Radiology, Department of Medicine, Stony Brook Cancer
Center, and Facility of Experimental Radiopharmaceutical Manufacturing (FERM), Stony Brook Renaissance School of Medicine, Stony
Brook University, Stony
Brook, New York 11794, United States
| | - Peter J. Tonge
- Center
for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, John S. Toll Drive, Stony
Brook, New York 11794-3400, United States
- Department
of Psychiatry, Department of Radiology, Department of Medicine, Stony Brook Cancer
Center, and Facility of Experimental Radiopharmaceutical Manufacturing (FERM), Stony Brook Renaissance School of Medicine, Stony
Brook University, Stony
Brook, New York 11794, United States
| |
Collapse
|
9
|
Brunetti JE, Kitsera M, Muñoz-Fontela C, Rodríguez E. Use of Hu-PBL Mice to Study Pathogenesis of Human-Restricted Viruses. Viruses 2023; 15:228. [PMID: 36680271 PMCID: PMC9866769 DOI: 10.3390/v15010228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/07/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Different humanized mouse models have been developed to study human diseases such as autoimmune illnesses, cancer and viral infections. These models are based on the use of immunodeficient mouse strains that are transplanted with human tissues or human immune cells. Among the latter, mice transplanted with hematopoietic stem cells have been widely used to study human infectious diseases. However, mouse models built upon the transplantation of donor-specific mature immune cells are still under development, especially in the field of viral infections. These models can retain the unique immune memory of the donor, making them suitable for the study of correlates of protection upon natural infection or vaccination. Here, we will review some of these models and how they have been applied to virology research. Moreover, the future applications and the potential of these models to design therapies against human viral infections are discussed.
Collapse
Affiliation(s)
| | - Maksym Kitsera
- Bernhard-Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - César Muñoz-Fontela
- Bernhard-Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Borstel-Lübeck, 38124 Braunschweig, Germany
| | - Estefanía Rodríguez
- Bernhard-Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Borstel-Lübeck, 38124 Braunschweig, Germany
| |
Collapse
|
10
|
Chen J, Liao S, Xiao Z, Pan Q, Wang X, Shen K, Wang S, Yang L, Guo F, Liu HF, Pan Q. The development and improvement of immunodeficient mice and humanized immune system mouse models. Front Immunol 2022; 13:1007579. [PMID: 36341323 PMCID: PMC9626807 DOI: 10.3389/fimmu.2022.1007579] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/07/2022] [Indexed: 12/02/2022] Open
Abstract
Animal models play an indispensable role in the study of human diseases. However, animal models of different diseases do not fully mimic the complex internal environment of humans. Immunodeficient mice are deficient in certain genes and do not express these or show reduced expression in some of their cells, facilitating the establishment of humanized mice and simulation of the human environment in vivo. Here, we summarize the developments in immunodeficient mice, from the initial nude mice lacking T lymphocytes to NOD/SCID rgnull mice lacking T, B, and NK cell populations. We describe existing humanized immune system mouse models based on immunodeficient mice in which human cells or tissues have been transplanted to establish a human immune system, including humanized-peripheral blood mononuclear cells (Hu-PBMCs), humanized hematopoietic stem cells (Hu-HSCs), and humanized bone marrow, liver, thymus (Hu-BLT) mouse models. The different methods for their development involve varying levels of complexity and humanization. Humanized mice are widely used in the study of various diseases to provide a transitional stage for clinical research. However, several challenges persist, including improving the efficiency of reconstructing the human B cell immune response, extending lifespan, improving the survival rate of mice to extend the observation period, and improving the development of standardized commercialized models and as well as their use. Overall, there are many opportunities and challenges in the development of humanized immune system mouse models which can provide novel strategies for understanding the mechanisms and treatments of human disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Qingjun Pan
- *Correspondence: Hua-feng Liu, ; Qingjun Pan,
| |
Collapse
|
11
|
Negi S, Saini S, Tandel N, Sahu K, Mishra RP, Tyagi RK. Translating Treg Therapy for Inflammatory Bowel Disease in Humanized Mice. Cells 2021; 10:1847. [PMID: 34440615 PMCID: PMC8393385 DOI: 10.3390/cells10081847] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023] Open
Abstract
Crohn's disease and ulcerative colitis, two major forms of inflammatory bowel disease (IBD) in humans, afflicted in genetically predisposed individuals due to dysregulated immune response directed against constituents of gut flora. The defective immune responses mounted against the regulatory mechanisms amplify and maintain the IBD-induced mucosal inflammation. Therefore, restoring the balance between inflammatory and anti-inflammatory immunepathways in the gut may contribute to halting the IBD-associated tissue-damaging immune response. Phenotypic and functional characterization of various immune-suppressive T cells (regulatory T cells; Tregs) over the last decade has been used to optimize the procedures for in vitro expansion of these cells for developing therapeutic interventional strategies. In this paper, we review the mechanisms of action and functional importance of Tregs during the pathogenesis of IBD and modulating the disease induced inflammation as well as role of mouse models including humanized mice repopulated with the human immune system (HIS) to study the IBD. "Humanized" mouse models provide new tools to analyze human Treg ontogeny, immunobiology, and therapy and the role of Tregs in developing interventional strategies against IBD. Overall, humanized mouse models replicate the human conditions and prove a viable tool to study molecular functions of human Tregs to harness their therapeutic potential.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Colitis, Ulcerative/genetics
- Colitis, Ulcerative/immunology
- Colitis, Ulcerative/metabolism
- Colitis, Ulcerative/therapy
- Crohn Disease/genetics
- Crohn Disease/immunology
- Crohn Disease/metabolism
- Crohn Disease/therapy
- Disease Models, Animal
- Hematopoietic Stem Cell Transplantation
- Humans
- Mice, Transgenic
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Sushmita Negi
- Biomedical Parasitology and Nano-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; (S.N.); (S.S.); (K.S.)
- BERPDC Department, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| | - Sheetal Saini
- Biomedical Parasitology and Nano-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; (S.N.); (S.S.); (K.S.)
| | - Nikunj Tandel
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India;
| | - Kiran Sahu
- Biomedical Parasitology and Nano-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; (S.N.); (S.S.); (K.S.)
| | - Ravi P.N. Mishra
- BERPDC Department, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| | - Rajeev K. Tyagi
- Biomedical Parasitology and Nano-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; (S.N.); (S.S.); (K.S.)
| |
Collapse
|
12
|
Human Somatostatin SST 4 Receptor Transgenic Mice: Construction and Brain Expression Pattern Characterization. Int J Mol Sci 2021; 22:ijms22073758. [PMID: 33916620 PMCID: PMC8038480 DOI: 10.3390/ijms22073758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/13/2022] Open
Abstract
Somatostatin receptor subtype 4 (SST4) has been shown to mediate analgesic, antidepressant and anti-inflammatory functions without endocrine actions; therefore, it is proposed to be a novel target for drug development. To overcome the species differences of SST4 receptor expression and function between humans and mice, we generated an SST4 humanized mouse line to serve as a translational animal model for preclinical research. A transposon vector containing the hSSTR4 and reporter gene construct driven by the hSSTR4 regulatory elements were created. The vector was randomly inserted in Sstr4-deficient mice. hSSTR4 expression was detected by bioluminescent in vivo imaging of the luciferase reporter predominantly in the brain. RT-qPCR confirmed the expression of the human gene in the brain and various peripheral tissues consistent with the in vivo imaging. RNAscope in situ hybridization revealed the presence of hSSTR4 transcripts in glutamatergic excitatory neurons in the CA1 and CA2 regions of the hippocampus; in the GABAergic interneurons in the granular layer of the olfactory bulb and in both types of neurons in the primary somatosensory cortex, piriform cortex, prelimbic cortex and amygdala. This novel SST4 humanized mouse line might enable us to investigate the differences of human and mouse SST4 receptor expression and function and assess the effects of SST4 receptor agonist drug candidates.
Collapse
|
13
|
Kim YY, Kim JS, Che JH, Ku SY, Kang BC, Yun JW. Comparison of Genetically Engineered Immunodeficient Animal Models for Nonclinical Testing of Stem Cell Therapies. Pharmaceutics 2021; 13:130. [PMID: 33498509 PMCID: PMC7909568 DOI: 10.3390/pharmaceutics13020130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 12/23/2022] Open
Abstract
For the recovery or replacement of dysfunctional cells and tissue-the goal of stem cell research-successful engraftment of transplanted cells and tissues are essential events. The event is largely dependent on the immune rejection of the recipient; therefore, the immunogenic evaluation of candidate cells or tissues in immunodeficient animals is important. Understanding the immunodeficient system can provide insights into the generation and use of immunodeficient animal models, presenting a unique system to explore the capabilities of the innate immune system. In this review, we summarize various immunodeficient animal model systems with different target genes as valuable tools for biomedical research. There have been numerous immunodeficient models developed by different gene defects, resulting in many different features in phenotype. More important, mice, rats, and other large animals exhibit very different immunological and physiological features in tissue and organs, including genetic background and a representation of human disease conditions. Therefore, the findings from this review may guide researchers to select the most appropriate immunodeficient strain, target gene, and animal species based on the research type, mutant gene effects, and similarity to human immunological features for stem cell research.
Collapse
Affiliation(s)
- Yoon-Young Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Korea; (Y.-Y.K.); (S.-Y.K.)
| | - Jin-Soo Kim
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Korea;
| | - Jeong-Hwan Che
- Biomedical Center for Animal Resource and Development, Seoul National University College of Medicine, Seoul 03080, Korea;
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Korea; (Y.-Y.K.); (S.-Y.K.)
| | - Byeong-Cheol Kang
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jun-Won Yun
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Korea;
| |
Collapse
|
14
|
Blümich S, Zdimerova H, Münz C, Kipar A, Pellegrini G. Human CD34 + Hematopoietic Stem Cell-Engrafted NSG Mice: Morphological and Immunophenotypic Features. Vet Pathol 2020; 58:161-180. [PMID: 32901581 DOI: 10.1177/0300985820948822] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immunodeficient mice engrafted with human immune cells represent an innovative tool to improve translatability of animal models for the study of human diseases. Immunophenotyping in these mice focuses on engraftment rates and cellular differentiation in blood and secondary lymphoid organs, and is predominantly carried out by FACS (fluorescent activated cell sorting) analysis; information on the morphological aspects of engraftment and the prevalence of histologic lesions is limited. We histologically examined 3- to 6-month-old NSG mice, naïve or engrafted with CD34+ human hemopoietic stem cells (HSC), and employed a quantitative immunohistochemical approach to identify human and murine cell compartments, comparing the results with the FACS data. NSG mice mainly exhibited incidental findings in lungs, kidneys, testes, and adrenal glands. A 6-month-old NSG mouse had a mediastinal lymphoblastic lymphoma. The lymphoid organs of NSG mice lacked typical lymphoid tissue architecture but frequently exhibited small periarteriolar leukocyte clusters in the spleen. Mice engrafted with human HSC frequently showed nephropathy, ovarian atrophy, cataract, and abnormal retinal development, lesions considered secondary to irradiation. In addition, 20% exhibited multisystemic granulomatous inflammatory infiltrates, dominated by human macrophages and T cells, leading to the observed 7% mortality and morbidity. Immunophenotypic data revealed variable repopulation of lymphoid organs with hCD45+ human cells, which did not always parallel the engraftment levels measured via FACS. The study describes the most common pathological features in young NSG mice after human HSC engraftment. As some of these lesions contribute to morbidity, morphological assessment of the engraftment at tissue level might help improve immunophenotypic evaluations of this animal model.
Collapse
Affiliation(s)
- Sandra Blümich
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, 27217University of Zurich, Zurich, Switzerland
| | - Hana Zdimerova
- Viral Immunobiology, Institute of Experimental Immunology, 27217University of Zurich, Zurich, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, 27217University of Zurich, Zurich, Switzerland
| | - Anja Kipar
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, 27217University of Zurich, Zurich, Switzerland
| | - Giovanni Pellegrini
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, 27217University of Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
Lee K, Farrell K, Uh K. Application of genome-editing systems to enhance available pig resources for agriculture and biomedicine. Reprod Fertil Dev 2020; 32:40-49. [PMID: 32188556 DOI: 10.1071/rd19273] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traditionally, genetic engineering in the pig was a challenging task. Genetic engineering of somatic cells followed by somatic cell nuclear transfer (SCNT) could produce genetically engineered (GE) pigs carrying site-specific modifications. However, due to difficulties in engineering the genome of somatic cells and developmental defects associated with SCNT, a limited number of GE pig models were reported. Recent developments in genome-editing tools, such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and the clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated (Cas) 9 system, have markedly changed the effort and time required to produce GE pig models. The frequency of genetic engineering in somatic cells is now practical. In addition, SCNT is no longer essential in producing GE pigs carrying site-specific modifications, because direct injection of genome-editing systems into developing embryos introduces targeted modifications. To date, the CRISPR/Cas9 system is the most convenient, cost-effective, timely and commonly used genome-editing technology. Several applicable biomedical and agricultural pig models have been generated using the CRISPR/Cas9 system. Although the efficiency of genetic engineering has been markedly enhanced with the use of genome-editing systems, improvements are still needed to optimally use the emerging technology. Current and future advances in genome-editing strategies will have a monumental effect on pig models used in agriculture and biomedicine.
Collapse
Affiliation(s)
- Kiho Lee
- Department of Animal and Poultry Sciences, Litton-Reaves Hall, Virginia Tech, Blacksburg, Virgina 24061, USA; and Corresponding author.
| | - Kayla Farrell
- Department of Animal and Poultry Sciences, Litton-Reaves Hall, Virginia Tech, Blacksburg, Virgina 24061, USA
| | - Kyungjun Uh
- Department of Animal and Poultry Sciences, Litton-Reaves Hall, Virginia Tech, Blacksburg, Virgina 24061, USA
| |
Collapse
|
16
|
Lee K, Uh K, Farrell K. Current progress of genome editing in livestock. Theriogenology 2020; 150:229-235. [PMID: 32000993 DOI: 10.1016/j.theriogenology.2020.01.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 01/18/2020] [Indexed: 12/12/2022]
Abstract
Historically, genetic engineering in livestock proved to be challenging. Without stable embryonic stem cell lines to utilize, somatic cell nuclear transfer (SCNT) had to be employed to produce many of the genetically engineered (GE) livestock models. Through the genetic engineering of somatic cells followed by SCNT, GE livestock models could be generated carrying site-specific modifications. Although successful, only a few GE livestock models were generated because of low efficiency and associated birth defects. Recently, there have been major strides in the development of genome editing tools: Zinc-Finger Nucleases (ZFNs), Transcription activator-like effector nucleases (TALENS), and Clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated 9 (Cas9) system. These tools rely on the generation of a double strand DNA break, followed by one of two repair pathways: non-homologous end joining (NHEJ) or homology directed repair (HDR). Compared to the traditional approaches, these tools dramatically reduce time and effort needed to establish a GE animal. Another benefit of utilizing genome editing tools is the application of direct injection into developing embryos to induce targeted mutations, therefore, eliminating side effects associated with SCNT. Emerging technological advancements of genome editing systems have dramatically improved efficiency to generate GE livestock models for both biomedical and agricultural purposes. Although the efficiency of genome editing tools has revolutionized GE livestock production, improvements for safe and consistent application are desired. This review will provide an overview of genome editing techniques, as well as examples of GE livestock models for agricultural and biomedical purposes.
Collapse
Affiliation(s)
- Kiho Lee
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA.
| | - Kyungjun Uh
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Kayla Farrell
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
17
|
Gbyli R, Song Y, Halene S. Humanized mice as preclinical models for myeloid malignancies. Biochem Pharmacol 2020; 174:113794. [PMID: 31926939 DOI: 10.1016/j.bcp.2020.113794] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/07/2020] [Indexed: 02/07/2023]
Abstract
Humanized mice have proven to be invaluable for human hematological translational research since they offer essential tools to dissect disease biology and to bridge the gap between pre-clinical testing of novel therapeutics and their clinical applications. Many efforts have been placed to advance and optimize humanized mice to support the engraftment, differentiation, and maintenance of hematopoietic stem cells (HSCs) and the human hematological system in order to broaden the scope of applications of such models. This review covers the background of humanized mice, how they are used as platforms to model myeloid malignancies, and the various current and potential approaches to further enhance their utilization in biomedical research.
Collapse
Affiliation(s)
- Rana Gbyli
- Section of Hematology, Department of Internal Medicine and Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Yuanbin Song
- Section of Hematology, Department of Internal Medicine and Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine and Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
18
|
Curran M, Mairesse M, Matas-Céspedes A, Bareham B, Pellegrini G, Liaunardy A, Powell E, Sargeant R, Cuomo E, Stebbings R, Betts CJ, Saeb-Parsy K. Recent Advancements and Applications of Human Immune System Mice in Preclinical Immuno-Oncology. Toxicol Pathol 2019; 48:302-316. [PMID: 31847725 DOI: 10.1177/0192623319886304] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significant advances in immunotherapies have resulted in the increasing need of predictive preclinical models to improve immunotherapeutic drug development, treatment combination, and to prevent or minimize toxicity in clinical trials. Immunodeficient mice reconstituted with human immune system (HIS), termed humanized mice or HIS mice, permit detailed analysis of human immune biology, development, and function. Although this model constitutes a great translational model, some aspects need to be improved as the incomplete engraftment of immune cells, graft versus host disease and the lack of human cytokines and growth factors. In this review, we discuss current HIS platforms, their pathology, and recent advances in their development to improve the quality of human immune cell reconstitution. We also highlight new technologies that can be used to better understand these models and how improved characterization is needed for their application in immuno-oncology safety, efficacy, and new modalities therapy development.
Collapse
Affiliation(s)
- Michelle Curran
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.,Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Maelle Mairesse
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Alba Matas-Céspedes
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.,Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Bethany Bareham
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Giovanni Pellegrini
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ardi Liaunardy
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Edward Powell
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Rebecca Sargeant
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Emanuela Cuomo
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Richard Stebbings
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Catherine J Betts
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
19
|
Mesenchymal Stromal Cells Modulate Peripheral Stress-Induced Innate Immune Activation Indirectly Limiting the Emergence of Neuroinflammation-Driven Depressive and Anxiety-like Behaviors. Biol Psychiatry 2019; 86:712-724. [PMID: 31521333 DOI: 10.1016/j.biopsych.2019.07.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 07/05/2019] [Accepted: 07/15/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Hyperactivation of innate immunity has been implicated in the etiology of mood disorders, including major depressive disorder (MDD). Mesenchymal stromal cells (MSCs) have demonstrated potent immunomodulatory capabilities in the context of chronic inflammatory disease and injury but have yet to be evaluated in stress-based preclinical models of MDD. We sought to test the ability of intravenous MSCs to modulate innate immune activation and behavioral patterns associated with repeated social defeat (RSD). METHODS Murine RSD-induced innate immune activation as well as depressive and anxiety-like behaviors were assessed in unstressed, RSD, and RSD + human MSC groups. Biodistribution and fate studies were performed to inform potential mechanisms of action. RESULTS MSCs reduced stress-induced circulating proinflammatory cytokines, monocytes, neuroinflammation, and depressive and anxiety-like behaviors. Biodistribution analyses indicated that infused MSCs distributed within peripheral organs without homing to the brain. Murine neutrophils targeted MSCs in the lungs within hours of administration. MSCs and recipient neutrophils were cleared by recipient macrophages promoting a switch toward a regulatory phenotype and systemic resolution of inflammation. CONCLUSIONS Peripheral delivery of MSCs modulates central nervous system inflammatory processes and aberrant behavioral patterns in a stress-based rodent model of MDD and anxiety. Recent studies suggest that host immune cell-mediated phagocytosis of MSCs in vivo can trigger an immunomodulatory cascade, resulting in resolution of inflammation. Our data suggest that similar mechanisms may protect distal organs, including the brain, from systemic, stress-induced proinflammatory spikes and may uncover unexpected targets in the periphery for novel or adjunct treatment for a subset of patients with MDD.
Collapse
|
20
|
Deng H, Liu H, de Silva T, Xue Y, Mohamud Y, Ng CS, Qu J, Zhang J, Jia WW, Lockwood WW, Luo H. Coxsackievirus Type B3 Is a Potent Oncolytic Virus against KRAS-Mutant Lung Adenocarcinoma. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:266-278. [PMID: 31463367 PMCID: PMC6709373 DOI: 10.1016/j.omto.2019.07.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 07/13/2019] [Indexed: 02/05/2023]
Abstract
KRAS mutant (KRASmut) lung adenocarcinoma is a refractory cancer without available targeted therapy. The current study explored the possibility to develop coxsackievirus type B3 (CVB3) as an oncolytic agent for the treatment of KRASmut lung adenocarcinoma. In cultured cells, we discovered that CVB3 selectively infects and lyses KRASmut lung adenocarcinoma cells (A549, H2030, and H23), while sparing normal lung epithelial cells (primary, BEAS2B, HPL1D, and 1HAEo) and EGFRmut lung adenocarcinoma cells (HCC4006, PC9, H3255, and H1975). Using stable cells expressing a single driver mutation of either KRASG12V or EGFRL858R in normal lung epithelial cells (HPL1D), we further showed that CVB3 specifically kills HPL1D-KRASG12V cells with minimal harm to HPL1D-EGFRL858R and control cells. Mechanistically, we demonstrated that aberrant activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and compromised type I interferon immune response in KRASmut lung adenocarcinoma cells serve as key factors contributing to the sensitivity to CVB3-induced cytotoxicity. Lastly, we conducted in vivo xenograft studies using two immunocompromised mouse models. Our results revealed that intratumoral injection of CVB3 results in a marked tumor regression of KRASmut lung adenocarcinoma in both non-obese diabetic (NOD) severe combined immunodeficiency (SCID) gamma (NSG) and NOD-SCID xenograft models. Together, our findings suggest that CVB3 is an excellent candidate to be further developed as a targeted therapy for KRASmut lung adenocarcinoma.
Collapse
Affiliation(s)
- Haoyu Deng
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Vascular Surgery, RenJi Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huitao Liu
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tanya de Silva
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - YuanChao Xue
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yasir Mohamud
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Chen Seng Ng
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada
| | - Junyan Qu
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Centre for Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Jingchun Zhang
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - William W.G. Jia
- Department of Surgery, Division of Neurosurgery, University of British Columbia, Vancouver, BC, Canada
| | - William W. Lockwood
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
- Corresponding author: William W. Lockwood, Department of Integrative Oncology, British Columbia Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada.
| | - Honglin Luo
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Corresponding author: Honglin Luo, Centre for Heart Lung Innovation, St. Paul’s Hospital, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada.
| |
Collapse
|
21
|
Perrier-Groult E, Pérès E, Pasdeloup M, Gazzolo L, Duc Dodon M, Mallein-Gerin F. Evaluation of the biocompatibility and stability of allogeneic tissue-engineered cartilage in humanized mice. PLoS One 2019; 14:e0217183. [PMID: 31107916 PMCID: PMC6527235 DOI: 10.1371/journal.pone.0217183] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/07/2019] [Indexed: 11/18/2022] Open
Abstract
Articular cartilage (AC) has poor capacities of regeneration and lesions often lead to osteoarthritis. Current AC reconstruction implies autologous chondrocyte implantation which requires tissue sampling and grafting. An alternative approach would be to use scaffolds containing off-the-shelf allogeneic human articular chondrocytes (HACs). To investigate tolerance of allogeneic HACs by the human immune system, we developed a humanized mouse model implanted with allogeneic cartilage constructs generated in vitro. A prerequisite of the study was to identify a scaffold that would not provoke inflammatory reaction in host. Therefore, we first compared the response of hu-mice to two biomaterials used in regenerative medicine, collagen sponge and agarose hydrogel. Four weeks after implantation in hu-mice, acellular collagen sponges, but not acellular agarose hydrogels, showed positive staining for CD3 (T lymphocytes) and CD68 (macrophages), suggesting that collagen scaffold elicits weak inflammatory reaction. These data led us to deepen our evaluation of the biocompatibility of allogeneic tissue-engineered cartilage by using agarose as scaffold. Agarose hydrogels were combined with allogeneic HACs to reconstruct cartilage in vitro. Particular attention was paid to HLA-A2 compatibility between HACs to be grafted and immune human cells of hu-mice: HLA-A2+ or HLA-A2- HACs agarose hydrogels were cultured in the presence of a chondrogenic cocktail and implanted in HLA-A2+ hu-mice. After four weeks implantation and regardless of the HLA-A2 phenotype, chondrocytes were well-differentiated and produced cartilage matrix in agarose. In addition, no sign of T-cell or macrophage infiltration was seen in the cartilaginous constructs and no significant increase in subpopulations of T lymphocytes and monocytes was detected in peripheral blood and spleen. We show for the first time that humanized mouse represents a useful model to investigate human immune responsiveness to tissue-engineered cartilage and our data together indicate that allogeneic cartilage constructs can be suitable for cartilage engineering.
Collapse
Affiliation(s)
- Emeline Perrier-Groult
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS-UMR5305, Lyon, France
- * E-mail:
| | - Eléonore Pérès
- Laboratory of Biology and Modeling of the Cell, Ecole Normale Supérieure (ENS) de Lyon, INSERM U1210, CNRS UMR5239, Lyon, France
| | - Marielle Pasdeloup
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS-UMR5305, Lyon, France
| | - Louis Gazzolo
- Laboratory of Biology and Modeling of the Cell, Ecole Normale Supérieure (ENS) de Lyon, INSERM U1210, CNRS UMR5239, Lyon, France
| | - Madeleine Duc Dodon
- Laboratory of Biology and Modeling of the Cell, Ecole Normale Supérieure (ENS) de Lyon, INSERM U1210, CNRS UMR5239, Lyon, France
| | - Frédéric Mallein-Gerin
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS-UMR5305, Lyon, France
| |
Collapse
|
22
|
Villano JS, Vleck SE, Felt SA, Myers DD, Lester PA. Safety Considerations When Working with Humanized Animals. ILAR J 2018; 59:150-160. [PMID: 30541024 DOI: 10.1093/ilar/ily012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 08/31/2018] [Indexed: 01/05/2023] Open
Abstract
Research using laboratory animals has been revolutionized by the creation of humanized animal models, which are immunodeficient animals engrafted with human cells, tissues, or organs. These animal models provide the research community a unique and promising opportunity to mimic a wide variety of disease conditions in humans, from infectious disease to cancer. A vast majority of these models are humanized mice like those injected with human CD34+ hematopoietic stem cells and patient-derived xenografts. With this technology comes the need for the animal research enterprise to understand the inherent and potential risks, such as exposure to bloodborne pathogens, associated with the model development and research applications. Here, we review existing humanized animal models and provide recommendations for their safe use based on regulatory framework and literature. A risk assessment program-from handling the human material to its administration to animals and animal housing-is a necessary initial step in mitigating risks associated with the use of humanized animals in research. Ultimately, establishing institutional policies and guidelines to ensure personnel safety is a legal and ethical responsibility of the research institution as part of the occupational health and safety program and overall animal care and use program.
Collapse
Affiliation(s)
- Jason S Villano
- Unit for Laboratory Animal Medicine, University of Michigan Medical School in Ann Arbor, Michigan
| | - Susan E Vleck
- Department of Environmental Health and Safety at Stanford University in Stanford, California
| | - Stephen A Felt
- Department of Comparative Medicine, Stanford University, Stanford, California
| | - Daniel D Myers
- Unit for Laboratory Animal Medicine, University of Michigan Medical School in Ann Arbor, Michigan.,Department of Surgery, University of Michigan Medical School, University of Michigan, Ann Arbor, Michigan
| | - Patrick A Lester
- Unit for Laboratory Animal Medicine, University of Michigan Medical School in Ann Arbor, Michigan
| |
Collapse
|
23
|
Tyagi RK, Tandel N, Deshpande R, Engelman RW, Patel SD, Tyagi P. Humanized Mice Are Instrumental to the Study of Plasmodium falciparum Infection. Front Immunol 2018; 9:2550. [PMID: 30631319 PMCID: PMC6315153 DOI: 10.3389/fimmu.2018.02550] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/17/2018] [Indexed: 02/05/2023] Open
Abstract
Research using humanized mice has advanced our knowledge and understanding of human haematopoiesis, non-adaptive and adaptive immunity, autoimmunity, infectious disease, cancer biology, and regenerative medicine. Challenges posed by the human-malaria parasite Plasmodium falciparum include its complex life cycle, the evolution of drug resistance against anti-malarials, poor diagnosis, and a lack of effective vaccines. Advancements in genetically engineered and immunodeficient mouse strains, have allowed for studies of the asexual blood stage, exoerythrocytic stage and the transition from liver-to-blood stage infection, in a single vertebrate host. This review discusses the process of "humanization" of various immunodeficient/transgenic strains and their contribution to translational biomedical research. Our work reviews the strategies employed to overcome the remaining-limitations of the developed human-mouse chimera(s).
Collapse
Affiliation(s)
- Rajeev K. Tyagi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Biomedical parasitology Unit, Institute Pasteur, Paris, France
- Department of Global Health, College of Public Health, University of South Florida, Tampa, FL, United States
| | - Nikunj Tandel
- Institute of Science, Nirma University, Ahmedabad, India
| | | | - Robert W. Engelman
- Department of Pediatrics, Pathology and Cell Biology, University of South Florida, Tampa, FL, United States
| | | | - Priyanka Tyagi
- Department of Basic and Applied Sciences, School of Engineering, GD Goenka University, Gurgaon, India
| |
Collapse
|
24
|
Yang X, Zhou J, He J, Liu J, Wang H, Liu Y, Jiang T, Zhang Q, Fu X, Xu Y. An Immune System-Modified Rat Model for Human Stem Cell Transplantation Research. Stem Cell Reports 2018; 11:514-521. [PMID: 29983387 PMCID: PMC6092637 DOI: 10.1016/j.stemcr.2018.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 02/04/2023] Open
Abstract
Due to its lack of both innate and acquired immune responses to human cells, the NODSCIDIl2rγ−/− (NSG) mouse model has become an important tool for human stem cell research. When compared with the mouse, the rat is physiologically more similar to humans and offers advantages in preclinical efficacy studies on human stem cells, particularly in evaluating neural, hepatic, and cardiac functions. Therefore, we generated a human SIRPα+Prdkc−/−Il2rγ−/− rat model, denoted NSG-like (NSGL) rat, which expresses human SIRPα and is abolished in the development of B, T, and natural killer cells. When compared with Prdkc−/−Il2rγ−/− (SG) rats, NSGL rats allow more efficient engraftment of human cancer cells and human pluripotent stem cells. In addition, only NSGL rats, but not SG rats, can be engrafted with human hematopoietic stem cells to reconstitute the human immune system. Therefore, NSGL rats represent an improved xenotransplantation model for efficacy studies of human stem cells. Generation of human SIRPα+Prkdc−/−Il2rγ−/− NSG-like (NSGL) rat model NSGL rats lack B, T, and NK cells but express human SIRPα NSGL rats can be efficiently engrafted with human stem cells NSGL rats can be reconstituted by human HSCs to generate a human immune system
Collapse
Affiliation(s)
- Xinglong Yang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jianlong Zhou
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jingjin He
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China; The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518033, China
| | - Jingfeng Liu
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hui Wang
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518033, China
| | - Yachen Liu
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Tao Jiang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Qianbing Zhang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xuemei Fu
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518033, China.
| | - Yang Xu
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China; The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518033, China; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
25
|
Xu R, Dagnaes-Hansen F, Wogensen L, Axelsen SM, Seliktar D, Chen M. Fibrogenic and angiogenic commitments of human induced pluripotent stem cells derived mesenchymal stem cells in connective tissue growth factor-delivering scaffold in an immune-deficient mice model. J Biomed Mater Res B Appl Biomater 2017; 106:2266-2274. [DOI: 10.1002/jbm.b.34030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/17/2017] [Accepted: 09/24/2017] [Indexed: 01/03/2023]
Affiliation(s)
- Ruodan Xu
- Department of Engineering; Aarhus University; Aarhus C Denmark
| | | | - Lise Wogensen
- Research Laboratory of Biochemical Pathology; The Institute of Clinical Medicine, Health, Aarhus University; Aarhus C Denmark
| | | | - Dror Seliktar
- The Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology; Haifa Israel
| | - Menglin Chen
- Department of Engineering; Aarhus University; Aarhus C Denmark
| |
Collapse
|
26
|
Humanized mouse model for assessing the human immune response to xenogeneic and allogeneic decellularized biomaterials. Biomaterials 2017; 129:98-110. [PMID: 28334641 DOI: 10.1016/j.biomaterials.2017.03.016] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 03/10/2017] [Indexed: 12/27/2022]
Abstract
Current assessment of biomaterial biocompatibility is typically implemented in wild type rodent models. Unfortunately, different characteristics of the immune systems in rodents versus humans limit the capability of these models to mimic the human immune response to naturally derived biomaterials. Here we investigated the utility of humanized mice as an improved model for testing naturally derived biomaterials. Two injectable hydrogels derived from decellularized porcine or human cadaveric myocardium were compared. Three days and one week after subcutaneous injection, the hydrogels were analyzed for early and mid-phase immune responses, respectively. Immune cells in the humanized mouse model, particularly T-helper cells, responded distinctly between the xenogeneic and allogeneic biomaterials. The allogeneic extracellular matrix derived hydrogels elicited significantly reduced total, human specific, and CD4+ T-helper cell infiltration in humanized mice compared to xenogeneic extracellular matrix hydrogels, which was not recapitulated in wild type mice. T-helper cells, in response to the allogeneic hydrogel material, were also less polarized towards a pro-remodeling Th2 phenotype compared to xenogeneic extracellular matrix hydrogels in humanized mice. In both models, both biomaterials induced the infiltration of macrophages polarized towards a M2 phenotype and T-helper cells polarized towards a Th2 phenotype. In conclusion, these studies showed the importance of testing naturally derived biomaterials in immune competent animals and the potential of utilizing this humanized mouse model for further studying human immune cell responses to biomaterials in an in vivo environment.
Collapse
|
27
|
Gopalakrishnapillai A, Kolb EA, Dhanan P, Bojja AS, Mason RW, Corao D, Barwe SP. Generation of Pediatric Leukemia Xenograft Models in NSG-B2m Mice: Comparison with NOD/SCID Mice. Front Oncol 2016; 6:162. [PMID: 27446808 PMCID: PMC4921874 DOI: 10.3389/fonc.2016.00162] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 06/15/2016] [Indexed: 01/22/2023] Open
Abstract
Generation of orthotopic xenograft mouse models of leukemia is important to understand the mechanisms of leukemogenesis, cancer progression, its cross talk with the bone marrow microenvironment, and for preclinical evaluation of drugs. In these models, following intravenous injection, leukemic cells home to the bone marrow and proliferate there before infiltrating other organs, such as spleen, liver, and the central nervous system. Moreover, such models have been shown to accurately recapitulate the human disease and correlate with patient response to therapy and prognosis. Thus, various immune-deficient mice strains have been used with or without recipient preconditioning to increase engraftment efficiency. Mice homozygous for the severe combined immune deficiency (SCID) mutation and with non-obese diabetic background (NOD/SCID) have been used in the majority of leukemia xenograft studies. Later, NOD/SCID mice deficient for interleukin 2 receptor gamma chain (IL2Rγ) gene called NSG mice became the model of choice for leukemia xenografts. However, engraftment of leukemia cells without irradiation preconditioning still remained a challenge. In this study, we used NSG mice with null alleles for major histocompatibility complex class I beta2-microglobulin (β2m) called NSG-B2m. This is a first report describing the 100% engraftment efficiency of pediatric leukemia cell lines and primary samples in NSG-B2m mice in the absence of host preconditioning by sublethal irradiation. We also show direct comparison of the engraftment efficiency and growth rate of pediatric acute leukemia cells in NSG-B2m and NOD/SCID mice, which showed 80–90% engraftment efficiency. Secondary and tertiary xenografts in NSG-B2m mice generated by injection of cells isolated from the spleens of leukemia-bearing mice also behaved similar to the primary patient sample. We have successfully engrafted 25 acute lymphoblastic leukemia (ALL) and 5 acute myeloid leukemia (AML) patient samples with distinct cytogenetic characteristics in NSG-B2m mice, with the purpose of generating pediatric ALL and AML xenografts for preclinical evaluation of drugs. Thus, our data support the use of NSG-B2m mouse model for leukemia engraftment and in vivo preclinical drug efficacy studies.
Collapse
Affiliation(s)
| | - E Anders Kolb
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children , Wilmington, DE , USA
| | - Priyanka Dhanan
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children , Wilmington, DE , USA
| | - Aruna Sri Bojja
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children , Wilmington, DE , USA
| | - Robert W Mason
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children , Wilmington, DE , USA
| | - Diana Corao
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children , Wilmington, DE , USA
| | - Sonali P Barwe
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children , Wilmington, DE , USA
| |
Collapse
|
28
|
|
29
|
Adams AB, Kitchens WH, Newell KA. Experimental models in discovery and translational studies. Transpl Immunol 2015. [DOI: 10.1002/9781119072997.ch16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
30
|
Sonntag K, Eckert F, Welker C, Müller H, Müller F, Zips D, Sipos B, Klein R, Blank G, Feuchtinger T, Schumm M, Handgretinger R, Schilbach K. Chronic graft-versus-host-disease in CD34(+)-humanized NSG mice is associated with human susceptibility HLA haplotypes for autoimmune disease. J Autoimmun 2015; 62:55-66. [PMID: 26143958 DOI: 10.1016/j.jaut.2015.06.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 05/27/2015] [Accepted: 06/07/2015] [Indexed: 11/26/2022]
Abstract
Chronic graft-versus-host disease (cGVHD) is a significant hurdle to long-term hematopoietic stem-cell transplantation success. Insights into the pathogenesis and mechanistical investigations of novel therapeutic strategies are limited as appropriate animal models are missing. The immunodeficient NSG mouse - when humanized with human bone marrow, fetal liver and thymus (BLT NSG) - is prone for cGVHD, yet mainly affects the skin. In contrast, the NSG mouse humanized exclusively with CD34(+)-selected, CD3(+)-depleted stem cells (CD34(+)NSG) has neither been described for acute nor chronic GVHD so far. This is the first report about the development of systemic autoimmune cGVHD ≥24 weeks post stem cell receipt involving lung, liver, skin, gingiva and intestine in two NSG cohorts humanized with CD34(+) grafts from different donors. Affected mice presented with sclerodermatous skin, fibrotic lung, severe hepatitis, and massive dental malformation/loss. CD4(+)-dominated, TH2-biased, bulky T-cell infiltrates featured highly skewed T cell receptor (TCR) repertoires, clonal expansions, and autoreactive TCRs. In affected tissues profibrotic IL-13 and -4 dominated over TH1 cytokines IFN-γ and TNF-α. Thus, the time point of manifestation and the phenotype match human systemic pleiotropic sclerodermatous GVHD. The CD34(+)NSG-model's intrinsic deficiency of thymus, thymus-derived regulatory T cells (nTreg) and B cells emphasizes the role of the genetic polymorphism and the cytokines in the pathogenesis of cGVHD. Importantly, the only factor discriminating diseased versus non-diseased CD34(+)NSG cohorts were two risk HLA haplotypes that in human mediate susceptibility for autoimmune disease (psoriasis). Thus, the CD34(+)NSG model may serve as a platform for addressing issues related to the pathophysiology and treatment of human autoimmunity and chronic GVHD.
Collapse
Affiliation(s)
- Katja Sonntag
- Department of General Pediatrics, Oncology/Hematology, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany
| | - Franziska Eckert
- Department of General Pediatrics, Oncology/Hematology, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany; Department of Radiation Oncology, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany
| | - Christian Welker
- Department of General Pediatrics, Oncology/Hematology, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany
| | - Hartmut Müller
- Institute of Pathology, Eberhard Karls University Tübingen, Liebermeisterstraße 8, 72076 Tübingen, Germany
| | - Friederike Müller
- Department of General Pediatrics, Oncology/Hematology, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany
| | - Bence Sipos
- Institute of Pathology, Eberhard Karls University Tübingen, Liebermeisterstraße 8, 72076 Tübingen, Germany
| | - Reinhild Klein
- Laboratory for Immunopathology, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Gregor Blank
- Department of General Pediatrics, Oncology/Hematology, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany
| | - Tobias Feuchtinger
- Pediatric Hematology, Oncology and Stem Cell Transplantation Dr. von Hauner'sches Kinderspital, Ludwig-Maximilian-University Munich, Lindwurmstraße 4, 80337 München, Germany
| | - Michael Schumm
- Department of General Pediatrics, Oncology/Hematology, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany
| | - Rupert Handgretinger
- Department of General Pediatrics, Oncology/Hematology, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany
| | - Karin Schilbach
- Department of General Pediatrics, Oncology/Hematology, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany.
| |
Collapse
|
31
|
Fransolet M, Henry L, Labied S, Masereel MC, Blacher S, Noël A, Foidart JM, Nisolle M, Munaut C. Influence of mouse strain on ovarian tissue recovery after engraftment with angiogenic factor. J Ovarian Res 2015; 8:14. [PMID: 25824856 PMCID: PMC4377049 DOI: 10.1186/s13048-015-0142-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 03/10/2015] [Indexed: 11/17/2022] Open
Abstract
Background For women facing gonadotoxic treatment, cryopreservation of ovarian tissue with subsequent retransplantation during remission is a promising technique for fertility preservation. However, follicle loss within grafted ovarian tissue can be caused by ischemia and progressive revascularization. Several xenograft models using different immunodeficient rodent lines are suitable for studying ovarian tissue survival and follicular viability after frozen-thawed ovarian cortex transplantation. SCID mice, which are deficient for functional B and T cells, are the most commonly used mice for ovarian xenograft studies. However, due to incomplete immunosuppression, NOD-SCID mice displaying low NK cell function and an absence of circulating complement might be more appropriate. The present study aims to define the most appropriate immunodeficient mouse strain for ovarian tissue xenotransplantation by comparing ovarian graft recovery in SCID and NOD-SCID mice following engraftment in the presence of isoform 111 of vascular endothelial growth factor. Methods Sheep ovarian cortex fragments were embedded in a collagen matrix, with or without VEGF111, before being stitched onto the ovaries of SCID and NOD-SCID mice. Transplants were recovered after 3 days to study early revascularization or after 3 weeks to evaluate follicle preservation and tissue fibrosis through histological analyses. Results At day 3, vessels were largely reorganized in the ovarian grafts of both mouse strains. After 3 weeks, the cortical tissue was clearly identifiable in SCID mice but not in NOD-SCID mice. Upon VEGF111 treatment, vascularization was significantly improved 3 days after transplantation in SCID mice. This increase in vessel density was correlated with better follicular preservation in SCID mice 3 weeks after transplantation. Fibrosis was not decreased by VEGF treatment in either mouse strain. Conclusions Tissue architecture and follicular morphology were better preserved in ovarian tissues grafted in SCID mice in comparison with NOD-SCID mice. Moreover, tissue revascularization was improved in SCID mice by VEGF111 graft treatment. Thus, we consider SCID mice to be the best murine model for studying ovarian tissue xenografts.
Collapse
|
32
|
Katano I, Ito R, Kamisako T, Eto T, Ogura T, Kawai K, Suemizu H, Takahashi T, Kawakami Y, Ito M. NOD-Rag2null IL-2Rγnull mice: an alternative to NOG mice for generation of humanized mice. Exp Anim 2015; 63:321-30. [PMID: 25077762 PMCID: PMC4206736 DOI: 10.1538/expanim.63.321] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
We have developed NOD-Rag2(null) IL-2Rγ(null) (NR2G) mice similar to NOD-scidIL-2Rγ(null) (NOG) mice that are known as an excellent host to generate humanized mice. To evaluate the usefulness of NR2G mice as a host for humanized mice, the engraftment rates and differentiation of human cells after human hematopoietic stem cell (HSC) transplantation were compared among NR2G, NOG, and NOD-scid mice. For this purpose, the appropriate irradiation doses to expand the niche for human stem cells in the bone marrow were first determined. As a result, 8 and 2.5 Gy in adult, and 4 and 1 Gy in newborn NR2G and NOG mice, respectively, were found to be appropriate. Next, 5 × 10(4) human umbilical cord blood CD34(+) cells were intravenously inoculated into irradiated adult or newborn of the immunodeficient mice. These HSC transplantation experiments demonstrated that both NR2G and NOG mice showed high engraftment rates compared with NOD-scid mice, although NOG mice showed a slightly higher engraftment rate than that for NR2G mice. However, no difference was found in the human cell populations differentiated from HSCs between NR2G and NOG mice. The HSC transplantation experiments to adults and newborns of two immunodeficient mice also revealed that the HSC transplantation into newborn mice resulted in higher engraftment rate than those into adults. These results showed that NR2G mice could be used as an alternative host to NOG mice to generate humanized mice.
Collapse
Affiliation(s)
- Ikumi Katano
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 216-0001, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Rong Z, Wang M, Hu Z, Stradner M, Zhu S, Kong H, Yi H, Goldrath A, Yang YG, Xu Y, Fu X. An effective approach to prevent immune rejection of human ESC-derived allografts. Cell Stem Cell 2014; 14:121-30. [PMID: 24388175 DOI: 10.1016/j.stem.2013.11.014] [Citation(s) in RCA: 182] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/07/2013] [Accepted: 11/15/2013] [Indexed: 01/03/2023]
Abstract
Human embryonic stem cells (hESCs) hold great promise for cell therapy as a source of diverse differentiated cell types. One key bottleneck to realizing such potential is allogenic immune rejection of hESC-derived cells by recipients. Here, we optimized humanized mice (Hu-mice) reconstituted with a functional human immune system that mounts a vigorous rejection of hESCs and their derivatives. We established knockin hESCs that constitutively express CTLA4-Ig and PD-L1 before and after differentiation, denoted CP hESCs. We then demonstrated that allogenic CP hESC-derived teratomas, fibroblasts, and cardiomyocytes are immune protected in Hu-mice, while cells derived from parental hESCs are effectively rejected. Expression of both CTLA4-Ig, which disrupts T cell costimulatory pathways, and PD-L1, which activates T cell inhibitory pathway, is required to confer immune protection, as neither was sufficient on their own. These findings are instrumental for developing a strategy to protect hESC-derived cells from allogenic immune responses without requiring systemic immune suppression.
Collapse
Affiliation(s)
- Zhili Rong
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0322, USA
| | - Meiyan Wang
- Shenzhen Children's Hospital, Shenzhen, Guangdong 518026, China; Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0322, USA
| | - Zheng Hu
- First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Martin Stradner
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0322, USA
| | - Shengyun Zhu
- Shenzhen Children's Hospital, Shenzhen, Guangdong 518026, China; Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0322, USA
| | - Huijuan Kong
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0322, USA
| | - Huanfa Yi
- First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ananda Goldrath
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0322, USA
| | - Yong-Guang Yang
- First Hospital of Jilin University, Changchun, Jilin 130021, China; Columbia Center for Translational Immunology, Columbia University Medical School, New York, NY 10032, USA
| | - Yang Xu
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0322, USA.
| | - Xuemei Fu
- Shenzhen Children's Hospital, Shenzhen, Guangdong 518026, China; Children's Hospital, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
34
|
Tanner A, Taylor SE, Decottignies W, Berges BK. Humanized mice as a model to study human hematopoietic stem cell transplantation. Stem Cells Dev 2013; 23:76-82. [PMID: 23962058 DOI: 10.1089/scd.2013.0265] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hematopoietic stem cell (HSC) transplantation has the potential to treat a variety of human diseases, including genetic deficiencies, immune disorders, and to restore immunity following cancer treatment. However, there are several obstacles that prevent effective HSC transplantation in humans. These include finding a matched donor, having a sufficient number of cells for the transplant, and the potency of the cells in the transplant. Ethical issues prevent effective research in humans that could provide insight into ways to overcome these obstacles. Highly immunodeficient mice can be transplanted with human HSCs and this process is accompanied by HSC homing to the murine bone marrow. This is followed by stem cell expansion, multilineage hematopoiesis, long-term engraftment, and functional human antibody and cellular immune responses. As such, humanized mice serve as a model for human HSC transplantation. A variety of conditions have been analyzed for their impact on HSC transplantation to produce humanized mice, including the type and source of cells used in the transplant, the number of cells transplanted, the expansion of cells with various protocols, and the route of introduction of cells into the mouse. In this review, we summarize what has been learned about HSC transplantation using humanized mice as a recipient model and we comment on how these models may be useful to future preclinical research to determine more effective ways to expand HSCs and to determine their repopulating potential in vivo.
Collapse
Affiliation(s)
- Anne Tanner
- Department of Microbiology and Molecular Biology, Brigham Young University , Provo, Utah
| | | | | | | |
Collapse
|
35
|
Zhang Y, Chen L, Yang J, Fleming JB, Chiao PJ, Logsdon CD, Li M. Study human pancreatic cancer in mice: how close are they? Biochim Biophys Acta Rev Cancer 2012; 1835:110-8. [PMID: 23147198 DOI: 10.1016/j.bbcan.2012.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 10/31/2012] [Accepted: 11/01/2012] [Indexed: 12/15/2022]
Abstract
Pancreatic cancer is the fourth leading cause of cancer deaths and is characterized by dismal prognosis. Xenograft and genetically engineered mouse (GEM) models have recapitulated critical elements of human pancreatic cancer, providing useful tools to probe the underlying cause of cancer etiology. In this review, we provide a brief description of the common genetic lesions that occur during the development of pancreatic cancer. Next, we describe the strengths and weaknesses of these two models and highlight key discoveries each has made. Although the relative merits of GEM and xenograft pancreatic cancer mouse models are subject to debate, both systems have and will continue to yield essential insights in understanding pancreatic cancer etiology. This information is critical for the development of new methods to screen, treat, and prevent pancreatic cancer.
Collapse
Affiliation(s)
- Yuqing Zhang
- Department of Cancer Biology, the University of Texas MD Anderson Cancer Center, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Graft versus host disease in the bone marrow, liver and thymus humanized mouse model. PLoS One 2012; 7:e44664. [PMID: 22957096 PMCID: PMC3434179 DOI: 10.1371/journal.pone.0044664] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 08/06/2012] [Indexed: 02/06/2023] Open
Abstract
Mice bearing a “humanized” immune system are valuable tools to experimentally manipulate human cells in vivo and facilitate disease models not normally possible in laboratory animals. Here we describe a form of GVHD that develops in NOD/SCID mice reconstituted with human fetal bone marrow, liver and thymus (NS BLT mice). The skin, lungs, gastrointestinal tract and parotid glands are affected with progressive inflammation and sclerosis. Although all mice showed involvement of at least one organ site, the incidence of overt clinical disease was approximately 35% by 22 weeks after reconstitution. The use of hosts lacking the IL2 common gamma chain (NOD/SCID/γc−/−) delayed the onset of disease, but ultimately did not affect incidence. Genetic analysis revealed that particular donor HLA class I alleles influenced the risk for the development of GVHD. At a cellular level, GVHD is associated with the infiltration of human CD4+ T cells into the skin and a shift towards Th1 cytokine production. GVHD also induced a mixed M1/M2 polarization phenotype in a dermal murine CD11b+, MHC class II+ macrophage population. The presence of xenogenic GVHD in BLT mice both presents a major obstacle in the use of humanized mice and an opportunity to conduct preclinical studies on GVHD in a humanized model.
Collapse
|
37
|
Jaeger LB, Nath A. Modeling HIV-associated neurocognitive disorders in mice: new approaches in the changing face of HIV neuropathogenesis. Dis Model Mech 2012; 5:313-22. [PMID: 22563057 PMCID: PMC3339825 DOI: 10.1242/dmm.008763] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It is well established that infection with the human immunodeficiency virus (HIV) leads to immune suppression. Less well known is the fact that long-term, progressive HIV disease is associated with the development of cognitive deficits. Since the introduction of combined antiretroviral therapy (cART), the clinical presentation of HIV infection has evolved into a chronic illness with very low levels of viral replication and chronic immune activation, with compliant affected individuals surviving for decades with a high quality of life. Despite these advances, many HIV-infected individuals develop some degree of neurodegeneration and cognitive impairment. The underlying pathophysiological mechanisms are not well understood, and there are no effective treatments. Thus, there is an unmet need for animal models that enable the study of HIV-associated neurocognitive disorders (HAND) and the testing of new therapeutic approaches to combat them. Here, we review the pros and cons of existing mouse models of HIV infection for addressing these aims and propose a detailed strategy for developing a new mouse model of HIV infection.
Collapse
Affiliation(s)
- Laura B Jaeger
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-1296, USA
| | | |
Collapse
|
38
|
Abstract
Humanized mouse models that have received human cells or tissue transplants are extremely useful in basic and applied human disease research. Highly immunodeficient mice, which do not reject xenografts and support cell and tissue differentiation and growth, are indispensable for generating additional appropriate models. Since the early 2000s, a series of immunodeficient mice appropriate for generating humanized mice has been successively developed by introducing the IL-2Rγ(null) gene (e.g., NOD/SCID/γc(null) and Rag2(null)γc(null) mice). These strains show not only a high rate of human cell engraftment, but also generate well-differentiated multilineage human hematopoietic cells after human hematopoietic stem cell (HSC) transplantation. These humanized mice facilitate the analysis of human hematology and immunology in vivo. However, human hematopoietic cells developed from HSCs are not always phenotypically and functionally identical to those in humans. More recently, a new series of immunodeficient mice compensates for these disadvantages. These mice were generated by genetically introducing human cytokine genes into NOD/SCID/γc(null) and Rag2(null)γc(null) mice. In this review, we describe the current knowledge of human hematopoietic cells developed in these mice. Various human disease mouse models using these humanized mice are summarized.
Collapse
|
39
|
Abstract
Pharmaceutical companies must find a better way to increase their output of truly new drugs for the benefit of patients and for their business survival. Here, I highlight a general perspective from within pharmaceutical research as it pertains to research advances in chemistry, biology, pharmacology, pharmacokinetics and toxicology that, if well integrated, stands to put the industry on a productive path. In addition, I provide a complementary perspective on the corporate culture aspect of innovation. I also introduce a new concept, termed 'innovation ASAP' (iASAP; asking powerful questions, seeking the outliers, accepting defeat and populating astutely) and provide support for it using examples of several successful drugs.
Collapse
|
40
|
Mora-Lee S, Sirerol-Piquer MS, Gutiérrez-Pérez M, López T, Casado-Nieto M, Jauquicoam C, Abizanda G, Romaguera-Ros M, Gomez-Pinedo U, Prósper F, García-Verdugo JM. Histological and ultrastructural comparison of cauterization and thrombosis stroke models in immune-deficient mice. J Inflamm (Lond) 2011; 8:28. [PMID: 22008614 PMCID: PMC3221623 DOI: 10.1186/1476-9255-8-28] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 10/18/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Stroke models are essential tools in experimental stroke. Although several models of stroke have been developed in a variety of animals, with the development of transgenic mice there is the need to develop a reliable and reproducible stroke model in mice, which mimics as close as possible human stroke. METHODS BALB/Ca-RAG2-/-γc-/- mice were subjected to cauterization or thrombosis stroke model and sacrificed at different time points (48hr, 1wk, 2wk and 4wk) after stroke. Mice received BrdU to estimate activation of cell proliferation in the SVZ. Brains were processed for immunohistochemical and EM. RESULTS In both stroke models, after inflammation the same glial scar formation process and damage evolution takes place. After stroke, necrotic tissue is progressively removed, and healthy tissue is preserved from injury through the glial scar formation. Cauterization stroke model produced unspecific damage, was less efficient and the infarct was less homogeneous compared to thrombosis infarct. Finally, thrombosis stroke model produces activation of SVZ proliferation. CONCLUSIONS Our results provide an exhaustive analysis of the histopathological changes (inflammation, necrosis, tissue remodeling, scarring...) that occur after stroke in the ischemic boundary zone, which are of key importance for the final stroke outcome. This analysis would allow evaluating how different therapies would affect wound and regeneration. Moreover, this stroke model in RAG 2-/- γC -/- allows cell transplant from different species, even human, to be analyzed.
Collapse
Affiliation(s)
- Silvia Mora-Lee
- Hematology and Cell Therapy Area, Clinica Universidad de Navarra and Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | | | - María Gutiérrez-Pérez
- Hematology and Cell Therapy Area, Clinica Universidad de Navarra and Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Tania López
- Hematology and Cell Therapy Area, Clinica Universidad de Navarra and Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Mayte Casado-Nieto
- Department of Comparative Neurobiology. Cavanilles Institute. CIPF. CIBERNED, Valencia, Spain
| | - Carlos Jauquicoam
- Hematology and Cell Therapy Area, Clinica Universidad de Navarra and Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Gloria Abizanda
- Hematology and Cell Therapy Area, Clinica Universidad de Navarra and Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Miriam Romaguera-Ros
- Department of Comparative Neurobiology. Cavanilles Institute. CIPF. CIBERNED, Valencia, Spain
| | | | - Felipe Prósper
- Hematology and Cell Therapy Area, Clinica Universidad de Navarra and Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | | |
Collapse
|
41
|
Bennani YL. Drug discovery in the next decade: innovation needed ASAP. Drug Discov Today 2011; 16:779-92. [PMID: 21704185 DOI: 10.1016/j.drudis.2011.06.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 05/02/2011] [Accepted: 06/06/2011] [Indexed: 01/18/2023]
Abstract
Pharmaceutical companies must find a better way to increase their output of truly new drugs for the benefit of patients and for their business survival. Here, I highlight a general perspective from within pharmaceutical research as it pertains to research advances in chemistry, biology, pharmacology, pharmacokinetics and toxicology that, if well integrated, stands to put the industry on a productive path. In addition, I provide a complementary perspective on the corporate culture aspect of innovation. I also introduce a new concept, termed 'innovation ASAP' (iASAP; asking powerful questions, seeking the outliers, accepting defeat and populating astutely) and provide support for it using examples of several successful drugs.
Collapse
|
42
|
Nanni P, Nicoletti G, Landuzzi L, Croci S, Murgo A, Palladini A, Antognoli A, Ianzano ML, Stivani V, Grosso V, Maira SM, García-Echeverría C, Scotlandi K, De Giovanni C, Lollini PL. High metastatic efficiency of human sarcoma cells in Rag2/gammac double knockout mice provides a powerful test system for antimetastatic targeted therapy. Eur J Cancer 2009; 46:659-68. [PMID: 20031388 DOI: 10.1016/j.ejca.2009.11.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 11/13/2009] [Accepted: 11/19/2009] [Indexed: 11/20/2022]
Abstract
Immunodeficient animal models are invaluable tools to investigate the metastatic propensity of human tumours. However residual immune responses, in particular natural killer (NK) cells, severely hamper the traffic and growth of human tumour cells. We studied whether a genetically modified mouse host lacking T, B and NK immunity allowed an improved expression of the metastatic phenotype of malignant human tumours. Metastatic spread of a panel of human sarcoma cell lines was studied in double knockout Rag2(-/-);gammac(-/-) mice in comparison with NK-depleted nude mice. Rag2(-/-);gammac(-/-) mice receiving intravenous (i.v.) or subcutaneous (s.c.) human sarcoma cell lines developed extensive multiorgan metastases. Metastatic efficiency in Rag2(-/-);gammac(-/-) was superior than in nude mice in terms of both metastatic sites and metastasis number. Metastatic growth in Rag2(-/-);gammac(-/-) mice was faster than that in nude mice, thus allowing an earlier metastasis evaluation. Most human sarcomas metastasised in the liver of Rag2(-/-);gammac(-/-) mice, a kind of organ preference undetectable in nude mice and specific of sarcomas, as several carcinoma cell lines failed to colonise the liver of Rag2(-/-);gammac(-/-) mice, independently of their metastatic spread to other sites. In vitro analysis of the molecular mechanisms of liver metastasis of sarcomas implicated liver-produced growth and motility factors, in particular the insulin-like growth factor (IGF) axis. NVP-BEZ235, a specific inhibitor of downstream signal transduction targeting PI3K and mTOR, strongly inhibited liver metastasis of human sarcoma cells. In conclusion, the Rag2(-/-);gammac(-/-) mouse model allowed the expression of human metastatic phenotypes inapparent in conventional immunodeficient mice and the preclinical testing of appropriate targeted therapies.
Collapse
Affiliation(s)
- Patrizia Nanni
- Cancer Research Section, Department of Experimental Pathology, Viale Filopanti 22, University of Bologna, 40126 Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Mucosal innate and adaptive immune responses against herpes simplex virus type 2 in a humanized mouse model. J Virol 2009; 83:10664-76. [PMID: 19656896 DOI: 10.1128/jvi.02584-08] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Genital herpes, caused by herpes simplex virus type 2 (HSV-2), is one of the most prevalent sexually transmitted diseases worldwide and a risk factor for acquiring human immunodeficiency virus. Although many vaccine candidates have shown promising results in animal models, they have failed to be effective in human trials. In this study, a humanized mouse strain was evaluated as a potential preclinical model for studying human immune responses to HSV-2 infection and vaccination. Immunodeficient mouse strains were examined for their abilities to develop human innate and adaptive immune cells after transplantation of human umbilical cord stem cells. A RAG2(-/-) gammac(-/-) mouse strain with a BALB/c background was chosen as the most appropriate model and was then examined for its ability to mount innate and adaptive immune responses to intravaginal HSV-2 infection and immunization. After primary infection, human cells in the lymph nodes were able to generate a protective innate immune response and produce gamma interferon (IFN-gamma). After intravaginal immunization and infection, human T cells and NK cells were found in the genital tract and iliac lymph nodes. In addition, human T cells in the spleen, lymph nodes, and vaginal tract were able to respond to stimulation with HSV-2 antigens by replicating and producing IFN-gamma. Human B cells were also able to produce HSV-2-specific immunoglobulin G. These adaptive responses were also shown to be protective and reduce local viral replication in the genital tract. This approach provides a means for studying human immune responses in vivo using a small-animal model and may become an important preclinical tool.
Collapse
|
44
|
Le Dévédec SE, van Roosmalen W, Maria N, Grimbergen M, Pont C, Lalai R, van de Water B. An improved model to study tumor cell autonomous metastasis programs using MTLn3 cells and the Rag2(-/-) gammac (-/-) mouse. Clin Exp Metastasis 2009; 26:673-84. [PMID: 19466569 PMCID: PMC2776159 DOI: 10.1007/s10585-009-9267-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Accepted: 04/24/2009] [Indexed: 01/03/2023]
Abstract
The occurrence of metastases is a critical determinant of the prognosis for breast cancer patients. Effective treatment of breast cancer metastases is hampered by a poor understanding of the mechanisms involved in the formation of these secondary tumor deposits. To study the processes of metastasis, valid in vivo tumor metastasis models are required. Here, we show that increased expression of the EGF receptor in the MTLn3 rat mammary tumor cell-line is essential for efficient lung metastasis formation in the Rag mouse model. EGFR expression resulted in delayed orthotopic tumor growth but at the same time strongly enhanced intravasation and lung metastasis. Previously, we demonstrated the critical role of NK cells in a lung metastasis model using MTLn3 cells in syngenic F344 rats. However, this model is incompatible with human EGFR. Using the highly metastatic EGFR-overexpressing MTLn3 cell-line, we report that only Rag2−/−γc−/− mice, which lack NK cells, allow efficient lung metastasis from primary tumors in the mammary gland. In contrast, in nude and SCID mice, the remaining innate immune cells reduce MTLn3 lung metastasis formation. Furthermore, we confirm this finding with the orthotopic transplantation of the 4T1 mouse mammary tumor cell-line. Thus, we have established an improved in vivo model using a Rag2−/− γc−/− mouse strain together with MTLn3 cells that have increased levels of the EGF receptor, which enables us to study EGFR-dependent tumor cell autonomous mechanisms underlying lung metastasis formation. This improved model can be used for drug target validation and development of new therapeutic strategies against breast cancer metastasis formation.
Collapse
Affiliation(s)
- Sylvia E Le Dévédec
- Division of Toxicology, Leiden Amsterdam Center for Drug Research (LACDR), Leiden University, RA, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|