<i>Dclre1c</i>-Mutation-Induced Immunocompromised Mice Are a Novel Model for Human Xenograft Research

Severe combined immunodeficient (SCID) mice serve as a critical model for human xenotransplantation studies, yet they often suffer from low engraftment rates and susceptibility to graft-versus-host disease (GVHD). Moreover, certain SCID strains demonstrate ‘immune leakage’, underscoring the need for...

Full description

Bibliographic Details
Main Authors: Yixiao Bin, Sanhua Wei, Ruo Chen, Haowei Zhang, Jing Ren, Peijuan Liu, Zhiqian Xin, Tianjiao Zhang, Haijiao Yang, Ke Wang, Zhuan Feng, Xiuxuan Sun, Zhinan Chen, Hai Zhang
Format: Article
Language:English
Published: MDPI AG 2024-02-01
Series:Biomolecules
Subjects:
Online Access:https://www.mdpi.com/2218-273X/14/2/180
_version_ 1797298788207951872
author Yixiao Bin
Sanhua Wei
Ruo Chen
Haowei Zhang
Jing Ren
Peijuan Liu
Zhiqian Xin
Tianjiao Zhang
Haijiao Yang
Ke Wang
Zhuan Feng
Xiuxuan Sun
Zhinan Chen
Hai Zhang
author_facet Yixiao Bin
Sanhua Wei
Ruo Chen
Haowei Zhang
Jing Ren
Peijuan Liu
Zhiqian Xin
Tianjiao Zhang
Haijiao Yang
Ke Wang
Zhuan Feng
Xiuxuan Sun
Zhinan Chen
Hai Zhang
author_sort Yixiao Bin
collection DOAJ
description Severe combined immunodeficient (SCID) mice serve as a critical model for human xenotransplantation studies, yet they often suffer from low engraftment rates and susceptibility to graft-versus-host disease (GVHD). Moreover, certain SCID strains demonstrate ‘immune leakage’, underscoring the need for novel model development. Here, we introduce an SCID mouse model with a targeted disruption of the <i>dclre1c</i> gene, encoding Artemis, which is essential for V(D)J recombination and DNA repair during T cell receptor (TCR) and B cell receptor (BCR) assembly. Artemis deficiency precipitates a profound immunodeficiency syndrome, marked by radiosensitivity and compromised T and B lymphocyte functionality. Utilizing CRISPR/Cas9-mediated gene editing, we generated <i>dclre1c</i>-deficient mice with an NOD genetic background. These mice exhibited a radiosensitive SCID phenotype, with pronounced DNA damage and defective thymic, splenic and lymph node development, culminating in reduced T and B lymphocyte populations. Notably, both cell lines and patient-derived tumor xenografts were successfully engrafted into these mice. Furthermore, the human immune system was effectively rebuilt following peripheral blood mononuclear cells (PBMCs) transplantation. The <i>dclre1c</i>-knockout NOD mice described herein represent a promising addition to the armamentarium of models for xenotransplantation, offering a valuable platform for advancing human immunobiological research.
first_indexed 2024-03-07T22:41:06Z
format Article
id doaj.art-e7faabe2416a4baf92bcef238975f733
institution Directory Open Access Journal
issn 2218-273X
language English
last_indexed 2024-03-07T22:41:06Z
publishDate 2024-02-01
publisher MDPI AG
record_format Article
series Biomolecules
spelling doaj.art-e7faabe2416a4baf92bcef238975f7332024-02-23T15:09:18ZengMDPI AGBiomolecules2218-273X2024-02-0114218010.3390/biom14020180<i>Dclre1c</i>-Mutation-Induced Immunocompromised Mice Are a Novel Model for Human Xenograft ResearchYixiao Bin0Sanhua Wei1Ruo Chen2Haowei Zhang3Jing Ren4Peijuan Liu5Zhiqian Xin6Tianjiao Zhang7Haijiao Yang8Ke Wang9Zhuan Feng10Xiuxuan Sun11Zhinan Chen12Hai Zhang13Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, ChinaDepartment of Obstetrics and Gynecology, Reproductive Medicine Center, Tang Du Hospital, Fourth Military Medical University, Xi’an 710038, ChinaDepartment of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, ChinaDepartment of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi’an 710032, ChinaDepartment of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, ChinaDepartment of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, ChinaDepartment of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, ChinaDepartment of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, ChinaDepartment of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, ChinaDepartment of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, ChinaDepartment of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, ChinaDepartment of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, ChinaDepartment of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, ChinaDepartment of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, ChinaSevere combined immunodeficient (SCID) mice serve as a critical model for human xenotransplantation studies, yet they often suffer from low engraftment rates and susceptibility to graft-versus-host disease (GVHD). Moreover, certain SCID strains demonstrate ‘immune leakage’, underscoring the need for novel model development. Here, we introduce an SCID mouse model with a targeted disruption of the <i>dclre1c</i> gene, encoding Artemis, which is essential for V(D)J recombination and DNA repair during T cell receptor (TCR) and B cell receptor (BCR) assembly. Artemis deficiency precipitates a profound immunodeficiency syndrome, marked by radiosensitivity and compromised T and B lymphocyte functionality. Utilizing CRISPR/Cas9-mediated gene editing, we generated <i>dclre1c</i>-deficient mice with an NOD genetic background. These mice exhibited a radiosensitive SCID phenotype, with pronounced DNA damage and defective thymic, splenic and lymph node development, culminating in reduced T and B lymphocyte populations. Notably, both cell lines and patient-derived tumor xenografts were successfully engrafted into these mice. Furthermore, the human immune system was effectively rebuilt following peripheral blood mononuclear cells (PBMCs) transplantation. The <i>dclre1c</i>-knockout NOD mice described herein represent a promising addition to the armamentarium of models for xenotransplantation, offering a valuable platform for advancing human immunobiological research.https://www.mdpi.com/2218-273X/14/2/180<i>dclre1c</i>CRISPR/Cas9knockouttumor xenograft modelimmune reconstitution
spellingShingle Yixiao Bin
Sanhua Wei
Ruo Chen
Haowei Zhang
Jing Ren
Peijuan Liu
Zhiqian Xin
Tianjiao Zhang
Haijiao Yang
Ke Wang
Zhuan Feng
Xiuxuan Sun
Zhinan Chen
Hai Zhang
<i>Dclre1c</i>-Mutation-Induced Immunocompromised Mice Are a Novel Model for Human Xenograft Research
Biomolecules
<i>dclre1c</i>
CRISPR/Cas9
knockout
tumor xenograft model
immune reconstitution
title <i>Dclre1c</i>-Mutation-Induced Immunocompromised Mice Are a Novel Model for Human Xenograft Research
title_full <i>Dclre1c</i>-Mutation-Induced Immunocompromised Mice Are a Novel Model for Human Xenograft Research
title_fullStr <i>Dclre1c</i>-Mutation-Induced Immunocompromised Mice Are a Novel Model for Human Xenograft Research
title_full_unstemmed <i>Dclre1c</i>-Mutation-Induced Immunocompromised Mice Are a Novel Model for Human Xenograft Research
title_short <i>Dclre1c</i>-Mutation-Induced Immunocompromised Mice Are a Novel Model for Human Xenograft Research
title_sort i dclre1c i mutation induced immunocompromised mice are a novel model for human xenograft research
topic <i>dclre1c</i>
CRISPR/Cas9
knockout
tumor xenograft model
immune reconstitution
url https://www.mdpi.com/2218-273X/14/2/180
work_keys_str_mv AT yixiaobin idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch
AT sanhuawei idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch
AT ruochen idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch
AT haoweizhang idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch
AT jingren idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch
AT peijuanliu idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch
AT zhiqianxin idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch
AT tianjiaozhang idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch
AT haijiaoyang idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch
AT kewang idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch
AT zhuanfeng idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch
AT xiuxuansun idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch
AT zhinanchen idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch
AT haizhang idclre1cimutationinducedimmunocompromisedmiceareanovelmodelforhumanxenograftresearch