1
|
Stabell AR, Lee GE, Jia Y, Wong KN, Wang S, Ling J, Nguyen SD, Sen GL, Nie Q, Atwood SX. Single-cell transcriptomics of human-skin-equivalent organoids. Cell Rep 2023; 42:112511. [PMID: 37195865 PMCID: PMC10348600 DOI: 10.1016/j.celrep.2023.112511] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 03/07/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023] Open
Abstract
Several methods for generating human-skin-equivalent (HSE) organoid cultures are in use to study skin biology; however, few studies thoroughly characterize these systems. To fill this gap, we use single-cell transcriptomics to compare in vitro HSEs, xenograft HSEs, and in vivo epidermis. By combining differential gene expression, pseudotime analyses, and spatial localization, we reconstruct HSE keratinocyte differentiation trajectories that recapitulate known in vivo epidermal differentiation pathways and show that HSEs contain major in vivo cellular states. However, HSEs also develop unique keratinocyte states, an expanded basal stem cell program, and disrupted terminal differentiation. Cell-cell communication modeling shows aberrant epithelial-to-mesenchymal transition (EMT)-associated signaling pathways that alter upon epidermal growth factor (EGF) supplementation. Last, xenograft HSEs at early time points post transplantation significantly rescue many in vitro deficits while undergoing a hypoxic response that drives an alternative differentiation lineage. This study highlights the strengths and limitations of organoid cultures and identifies areas for potential innovation.
Collapse
Affiliation(s)
- Adam R Stabell
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Grace E Lee
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Yunlong Jia
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Kirsten N Wong
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Shuxiong Wang
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
| | - Ji Ling
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sandrine D Nguyen
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - George L Sen
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Qing Nie
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Scott X Atwood
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Dermatology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
2
|
Assessment of Angiogenesis and Cell Survivability of an Inkjet Bioprinted Biological Implant in an Animal Model. MATERIALS 2022; 15:ma15134468. [PMID: 35806588 PMCID: PMC9267737 DOI: 10.3390/ma15134468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/01/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023]
Abstract
The rapidly growing field of tissue engineering hopes to soon address the shortage of transplantable tissues, allowing for precise control and fabrication that could be made for each specific patient. The protocols currently in place to print large-scale tissues have yet to address the main challenge of nutritional deficiencies in the central areas of the engineered tissue, causing necrosis deep within and rendering it ineffective. Bioprinted microvasculature has been proposed to encourage angiogenesis and facilitate the mobility of oxygen and nutrients throughout the engineered tissue. An implant made via an inkjet printing process containing human microvascular endothelial cells was placed in both B17-SCID and NSG-SGM3 animal models to determine the rate of angiogenesis and degree of cell survival. The implantable tissues were made using a combination of alginate and gelatin type B; all implants were printed via previously published procedures using a modified HP inkjet printer. Histopathological results show a dramatic increase in the average microvasculature formation for mice that received the printed constructs within the implant area when compared to the manual and control implants, indicating inkjet bioprinting technology can be effectively used for vascularization of engineered tissues.
Collapse
|
3
|
Tyagi RK. Plasmodium falciparum-infected humanized mice: a viable preclinical tool. Immunotherapy 2021; 13:1345-1353. [PMID: 34424053 DOI: 10.2217/imt-2021-0102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
Extensive research conducted on mouse-human chimeras has advanced our understanding on infectious diseases including the human-malaria parasite, Plasmodium falciparum. In vitro culture of asexual-blood stage infection of P. falciparum does not answer all questions related to parasitology, pharmacology and immunology, and complex life cycle, complicated genome, evolution of drug resistance and poor diagnosis makes it difficult to understand the patho-biology of parasite. Unavailability of effective-vaccine and issues of drug resistance advocates the use of human cell/tissues reconstituted immunodeficient-mice to P. falciparum. A number of immunodeficient-strains (TK/NOG, FRG/NOD, NOD/SCID/IL-2 receptor γ chain null, NOD severe combined immunodeficiency gamma [NSG] mouse and NOD.Rag1-/- IL2Rγ-/- [NRG; DRAG]) are used for humanization purposes. Additionally, human-hematopoietic stem cells (CD34 reconstituted-NSG [human immune system]) mice support the engraftment and repopulation of immune effecters to study systemic inflammatory diseases.
Collapse
Affiliation(s)
- Rajeev K Tyagi
- Division of Cell Biology & Immunology, Biomedical Parasitology & Nano-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh, 160036, India
| |
Collapse
|
4
|
Ajith A, Mulloy LL, Musa MA, Bravo-Egana V, Horuzsko DD, Gani I, Horuzsko A. Humanized Mouse Model as a Novel Approach in the Assessment of Human Allogeneic Responses in Organ Transplantation. Front Immunol 2021; 12:687715. [PMID: 34177940 PMCID: PMC8226140 DOI: 10.3389/fimmu.2021.687715] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/28/2021] [Indexed: 02/02/2023] Open
Abstract
The outcome of organ transplantation is largely dictated by selection of a well-matched donor, which results in less chance of graft rejection. An allogeneic immune response is the main immunological barrier for successful organ transplantation. Donor and recipient human leukocyte antigen (HLA) mismatching diminishes outcomes after solid organ transplantation. The current evaluation of HLA incompatibility does not provide information on the immunogenicity of individual HLA mismatches and impact of non-HLA-related alloantigens, especially in vivo. Here we demonstrate a new method for analysis of alloimmune responsiveness between donor and recipient in vivo by introducing a humanized mouse model. Using molecular, cellular, and genomic analyses, we demonstrated that a recipient's personalized humanized mouse provided the most sensitive assessment of allogeneic responsiveness to potential donors. In our study, HLA typing provided a better recipient-donor match for one donor among two related donors. In contrast, assessment of an allogeneic response by mixed lymphocyte reaction (MLR) was indistinguishable between these donors. We determined that, in the recipient's humanized mouse model, the donor selected by HLA typing induced the strongest allogeneic response with markedly increased allograft rejection markers, including activated cytotoxic Granzyme B-expressing CD8+ T cells. Moreover, the same donor induced stronger upregulation of genes involved in the allograft rejection pathway as determined by transcriptome analysis of isolated human CD45+cells. Thus, the humanized mouse model determined the lowest degree of recipient-donor alloimmune response, allowing for better selection of donor and minimized immunological risk of allograft rejection in organ transplantation. In addition, this approach could be used to evaluate the level of alloresponse in allogeneic cell-based therapies that include cell products derived from pluripotent embryonic stem cells or adult stem cells, both undifferentiated and differentiated, all of which will produce allogeneic immune responses.
Collapse
Affiliation(s)
- Ashwin Ajith
- Georgia Cancer Center, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Laura L. Mulloy
- Nephrology Division, Department of Medicine, Augusta University, Augusta, GA, United States
| | - Md. Abu Musa
- Georgia Cancer Center, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Valia Bravo-Egana
- Histocompatibility and Immunology Laboratory, Department of Surgery, Medical College of Georgia, Augusta University Medical Center, Augusta, GA, United States
| | - Daniel David Horuzsko
- Program of Osteopathic Medicine, Philadelphia College of Osteopathic Medicine South Georgia, Moultrie, GA, United States
| | - Imran Gani
- Nephrology Division, Department of Medicine, Augusta University, Augusta, GA, United States
| | - Anatolij Horuzsko
- Georgia Cancer Center, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
5
|
Lee MW, Miljanic M, Triplett T, Ramirez C, Aung KL, Eckhardt SG, Capasso A. Current methods in translational cancer research. Cancer Metastasis Rev 2021; 40:7-30. [PMID: 32929562 PMCID: PMC7897192 DOI: 10.1007/s10555-020-09931-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/04/2020] [Indexed: 12/22/2022]
Abstract
Recent developments in pre-clinical screening tools, that more reliably predict the clinical effects and adverse events of candidate therapeutic agents, has ushered in a new era of drug development and screening. However, given the rapid pace with which these models have emerged, the individual merits of these translational research tools warrant careful evaluation in order to furnish clinical researchers with appropriate information to conduct pre-clinical screening in an accelerated and rational manner. This review assesses the predictive utility of both well-established and emerging pre-clinical methods in terms of their suitability as a screening platform for treatment response, ability to represent pharmacodynamic and pharmacokinetic drug properties, and lastly debates the translational limitations and benefits of these models. To this end, we will describe the current literature on cell culture, organoids, in vivo mouse models, and in silico computational approaches. Particular focus will be devoted to discussing gaps and unmet needs in the literature as well as current advancements and innovations achieved in the field, such as co-clinical trials and future avenues for refinement.
Collapse
Affiliation(s)
- Michael W Lee
- Department of Medical Education, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Mihailo Miljanic
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Todd Triplett
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Craig Ramirez
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Kyaw L Aung
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - S Gail Eckhardt
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Anna Capasso
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA.
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
6
|
Peelen DM, Hoogduijn MJ, Hesselink DA, Baan CC. Advanced in vitro Research Models to Study the Role of Endothelial Cells in Solid Organ Transplantation. Front Immunol 2021; 12:607953. [PMID: 33664744 PMCID: PMC7921837 DOI: 10.3389/fimmu.2021.607953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/21/2021] [Indexed: 12/26/2022] Open
Abstract
The endothelium plays a key role in acute and chronic rejection of solid organ transplants. During both processes the endothelium is damaged often with major consequences for organ function. Also, endothelial cells (EC) have antigen-presenting properties and can in this manner initiate and enhance alloreactive immune responses. For decades, knowledge about these roles of EC have been obtained by studying both in vitro and in vivo models. These experimental models poorly imitate the immune response in patients and might explain why the discovery and development of agents that control EC responses is hampered. In recent years, various innovative human 3D in vitro models mimicking in vivo organ structure and function have been developed. These models will extend the knowledge about the diverse roles of EC in allograft rejection and will hopefully lead to discoveries of new targets that are involved in the interactions between the donor organ EC and the recipient's immune system. Moreover, these models can be used to gain a better insight in the mode of action of the currently prescribed immunosuppression and will enhance the development of novel therapeutics aiming to reduce allograft rejection and prolong graft survival.
Collapse
Affiliation(s)
- Daphne M Peelen
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Martin J Hoogduijn
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Dennis A Hesselink
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Carla C Baan
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
7
|
Bak I, Kim DJ, Kim HC, Shin HJ, Yu E, Yoo KW, Yu DY. Two base pair deletion in IL2 receptor γ gene in NOD/SCID mice induces a highly severe immunodeficiency. Lab Anim Res 2020; 36:27. [PMID: 32817844 PMCID: PMC7427935 DOI: 10.1186/s42826-020-00048-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/13/2020] [Indexed: 11/17/2022] Open
Abstract
Genome editing has recently emerged as a powerful tool for generating mutant mice. Small deletions of nucleotides in the target genes are frequently found in CRISPR/Cas9 mediated mutant mice. However, there are very few reports analyzing the phenotypes in small deleted mutant mice generated by CRISPR/Cas9. In this study, we generated a mutant by microinjecting sgRNAs targeting the IL2 receptor γ gene and Cas9 protein, into the cytoplasm of IVF-derived NOD.CB17/Prkdcscid/JKrb (NOD/SCID) mice embryos, and further investigated whether a 2 bp deletion of the IL2 receptor γ gene affects severe deficiency of immune cells as seen in NOD/LtSz-scid IL2 receptor γ−/− (NSG) mice. Our results show that the thymus weight of mutant mice is significantly less than that of NOD/SCID mice, whereas the spleen weight was marginally less. T and B cells in the mutant mice were severely deficient, and NK cells were almost absent. In addition, tumor growth was exceedingly increased in the mutant mice transplanted with HepG2, Raji and A549 cells, but not in nude and NOD/SCID mice. These results suggest that the NOD/SCID mice with deletion of 2 bp in the IL2 receptor γ gene shows same phenotype as NSG mice. Taken together, our data indicates that small deletions by genome editing is sufficient to generate null mutant mice.
Collapse
Affiliation(s)
- Inseon Bak
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Korea.,Genome engineering laboratory, GHBIO Inc., C406, 17 Techno4-ro Yuseong-gu, Daejeon, 34013 Korea
| | - Doo-Jin Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Korea
| | - Hyoung-Chin Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju, Chungcheongbukdo 28116 Korea
| | - Hye-Jun Shin
- Genome engineering laboratory, GHBIO Inc., C406, 17 Techno4-ro Yuseong-gu, Daejeon, 34013 Korea
| | - Eunhye Yu
- Genome engineering laboratory, GHBIO Inc., C406, 17 Techno4-ro Yuseong-gu, Daejeon, 34013 Korea
| | - Kyeong-Won Yoo
- Genome engineering laboratory, GHBIO Inc., C406, 17 Techno4-ro Yuseong-gu, Daejeon, 34013 Korea
| | - Dae-Yeul Yu
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 Korea
| |
Collapse
|
8
|
Tyagi RK, Li J, Jacobse J, Snapper SB, Shouval DS, Goettel JA. Humanized mouse models of genetic immune disorders and hematological malignancies. Biochem Pharmacol 2020; 174:113671. [PMID: 31634456 PMCID: PMC7050416 DOI: 10.1016/j.bcp.2019.113671] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023]
Abstract
The immune system is quite remarkable having both the ability to tolerate innocuous and self-antigens while possessing a robust capacity to recognize and eradicate infectious pathogens and foreign entities. The genetics that encode this delicate balancing act include multiple genes and specialized cell types. Over the past several years, whole exome and whole genome sequencing has uncovered the genetics driving many human immune-mediated diseases including monogenic disorders and hematological malignancies. With the advent of genome editing technologies, the ability to correct genetic immune defects in autologous cells holds great promise for a number of conditions. Since assessment of novel therapeutic strategies have been difficult in mice, in recent years, immunodeficient mice capable of engrafting human cells and tissue have been developed and utilized for a variety of research applications. In this review, we discuss immune-humanized mice as a research tool to study human immunobiology and genetic immune disorders in vivo and the promise of future applications.
Collapse
Affiliation(s)
- Rajeev K Tyagi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jing Li
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Justin Jacobse
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Scott B Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| | - Dror S Shouval
- Pediatric Gastroenterology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jeremy A Goettel
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
9
|
Ghandhi SA, Smilenov L, Shuryak I, Pujol-Canadell M, Amundson SA. Discordant gene responses to radiation in humans and mice and the role of hematopoietically humanized mice in the search for radiation biomarkers. Sci Rep 2019; 9:19434. [PMID: 31857640 PMCID: PMC6923394 DOI: 10.1038/s41598-019-55982-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 12/05/2019] [Indexed: 12/12/2022] Open
Abstract
The mouse (Mus musculus) is an extensively used model of human disease and responses to stresses such as ionizing radiation. As part of our work developing gene expression biomarkers of radiation exposure, dose, and injury, we have found many genes are either up-regulated (e.g. CDKN1A, MDM2, BBC3, and CCNG1) or down-regulated (e.g. TCF4 and MYC) in both species after irradiation at ~4 and 8 Gy. However, we have also found genes that are consistently up-regulated in humans and down-regulated in mice (e.g. DDB2, PCNA, GADD45A, SESN1, RRM2B, KCNN4, IFI30, and PTPRO). Here we test a hematopoietically humanized mouse as a potential in vivo model for biodosimetry studies, measuring the response of these 14 genes one day after irradiation at 2 and 4 Gy, and comparing it with that of human blood irradiated ex vivo, and blood from whole body irradiated mice. We found that human blood cells in the hematopoietically humanized mouse in vivo environment recapitulated the gene expression pattern expected from human cells, not the pattern seen from in vivo irradiated normal mice. The results of this study support the use of hematopoietically humanized mice as an in vivo model for screening of radiation response genes relevant to humans.
Collapse
Affiliation(s)
- Shanaz A Ghandhi
- Columbia University Irving Medical Center, 630 W 168th street, VC11-237, New York, NY, 10032, USA.
| | - Lubomir Smilenov
- Columbia University Irving Medical Center, 630 W 168th street, VC11-237, New York, NY, 10032, USA
| | - Igor Shuryak
- Columbia University Irving Medical Center, 630 W 168th street, VC11-237, New York, NY, 10032, USA
| | - Monica Pujol-Canadell
- Columbia University Irving Medical Center, 630 W 168th street, VC11-237, New York, NY, 10032, USA
| | - Sally A Amundson
- Columbia University Irving Medical Center, 630 W 168th street, VC11-237, New York, NY, 10032, USA
| |
Collapse
|
10
|
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
|
11
|
Couto JP, Bentires-Alj M. Mouse Models of Breast Cancer: Deceptions that Reveal the Truth. Breast Cancer 2017. [DOI: 10.1007/978-3-319-48848-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
12
|
Walsh NC, Kenney LL, Jangalwe S, Aryee KE, Greiner DL, Brehm MA, Shultz LD. Humanized Mouse Models of Clinical Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 12:187-215. [PMID: 27959627 DOI: 10.1146/annurev-pathol-052016-100332] [Citation(s) in RCA: 402] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunodeficient mice engrafted with functional human cells and tissues, that is, humanized mice, have become increasingly important as small, preclinical animal models for the study of human diseases. Since the description of immunodeficient mice bearing mutations in the IL2 receptor common gamma chain (IL2rgnull) in the early 2000s, investigators have been able to engraft murine recipients with human hematopoietic stem cells that develop into functional human immune systems. These mice can also be engrafted with human tissues such as islets, liver, skin, and most solid and hematologic cancers. Humanized mice are permitting significant progress in studies of human infectious disease, cancer, regenerative medicine, graft-versus-host disease, allergies, and immunity. Ultimately, use of humanized mice may lead to the implementation of truly personalized medicine in the clinic. This review discusses recent progress in the development and use of humanized mice and highlights their utility for the study of human diseases.
Collapse
Affiliation(s)
- Nicole C Walsh
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Laurie L Kenney
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Sonal Jangalwe
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Ken-Edwin Aryee
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Dale L Greiner
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Michael A Brehm
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | | |
Collapse
|
13
|
Morton JJ, Bird G, Refaeli Y, Jimeno A. Humanized Mouse Xenograft Models: Narrowing the Tumor-Microenvironment Gap. Cancer Res 2016; 76:6153-6158. [PMID: 27587540 DOI: 10.1158/0008-5472.can-16-1260] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/27/2016] [Indexed: 12/11/2022]
Abstract
Cancer research has long been hampered by the limitations of the current model systems. Both cultured cells and mouse xenografts grow in an environment highly dissimilar to that of their originating tumor, frequently resulting in promising treatments that are ultimately clinically ineffective. The development of highly immunodeficient mouse strains into which human immune systems can be engrafted can help bridge this gap. Humanized mice (HM) allow researchers to examine xenograft growth in the context of a human immune system and resultant tumor microenvironment, and recent studies have highlighted the increased similarities in attendant tumor structure, metastasis, and signaling to those features in cancer patients. This setting also facilitates the examination of investigational cancer therapies, including new immunotherapies. This review discusses recent advancements in the generation and application of HM models, their promise in cancer research, and their potential in generating clinically relevant treatments. This review also focuses on current efforts to improve HM models by engineering mouse strains expressing human cytokines or HLA proteins and implanting human bone, liver, and thymus tissue to facilitate immune cell maturation and trafficking. Finally, we discuss how these improvements may help direct future HM model cancer studies. Cancer Res; 76(21); 6153-8. ©2016 AACR.
Collapse
Affiliation(s)
- J Jason Morton
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Gregory Bird
- Department of Dermatology, University of Colorado School of Medicine, Aurora, Colorado
| | - Yosef Refaeli
- Department of Dermatology, University of Colorado School of Medicine, Aurora, Colorado.,Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado School of Medicine, Aurora, Colorado
| | - Antonio Jimeno
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado. .,Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
14
|
Kenney LL, Shultz LD, Greiner DL, Brehm MA. Humanized Mouse Models for Transplant Immunology. Am J Transplant 2016; 16:389-97. [PMID: 26588186 PMCID: PMC5283075 DOI: 10.1111/ajt.13520] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/02/2015] [Accepted: 09/04/2015] [Indexed: 01/25/2023]
Abstract
Our understanding of the molecular pathways that control immune responses, particularly immunomodulatory molecules that control the extent and duration of an immune response, have led to new approaches in the field of transplantation immunology to induce allograft survival. These molecular pathways are being defined precisely in murine models and translated into clinical practice; however, many of the newly available drugs are human-specific reagents. Furthermore, many species-specific differences exist between mouse and human immune systems. Recent advances in the development of humanized mice, namely, immunodeficient mice engrafted with functional human immune systems, have led to the availability of a small animal model for the study of human immune responses. Humanized mice represent an important preclinical model system for evaluation of new drugs and identification of the mechanisms underlying human allograft rejection without putting patients at risk. This review highlights recent advances in the development of humanized mice and their use as preclinical models for the study of human allograft responses.
Collapse
Affiliation(s)
- Laurie L Kenney
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605
| | | | - Dale L Greiner
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605,Corresponding Author: Dale L. Greiner, PhD, University of Massachusetts Medical School, 368 Plantation Street, AS7-2051, Worcester, MA 01605, Office: 508-856-1911, Fax: 508-856-4093,
| | - Michael A. Brehm
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605
| |
Collapse
|
15
|
Waide EH, Dekkers JCM, Ross JW, Rowland RRR, Wyatt CR, Ewen CL, Evans AB, Thekkoot DM, Boddicker NJ, Serão NVL, Ellinwood NM, Tuggle CK. Not All SCID Pigs Are Created Equally: Two Independent Mutations in the Artemis Gene Cause SCID in Pigs. THE JOURNAL OF IMMUNOLOGY 2015; 195:3171-9. [PMID: 26320255 DOI: 10.4049/jimmunol.1501132] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/28/2015] [Indexed: 01/07/2023]
Abstract
Mutations in >30 genes are known to result in impairment of the adaptive immune system, causing a group of disorders collectively known as SCID. SCID disorders are split into groups based on their presence and/or functionality of B, T, and NK cells. Piglets from a line of Yorkshire pigs at Iowa State University were shown to be affected by T(-)B(-)NK(+) SCID, representing, to our knowledge, the first example of naturally occurring SCID in pigs. In this study, we present evidence for two spontaneous mutations as the molecular basis for this SCID phenotype. Flow cytometry analysis of thymocytes showed an increased frequency of immature T cells in SCID pigs. Fibroblasts from these pigs were more sensitive to ionizing radiation than non-SCID piglets, eliminating the RAG1 and RAG2 genes. Genetic and molecular analyses showed that two mutations were present in the Artemis gene, which in the homozygous or compound heterozygous state cause the immunodeficient phenotype. Rescue of SCID fibroblast radiosensitivity by human Artemis protein demonstrated that the identified Artemis mutations are the direct cause of this cellular phenotype. The work presented in the present study reveals two mutations in the Artemis gene that cause T(-)B(-)NK(+) SCID in pigs. The SCID pig can be an important biomedical model, but these mutations would be undesirable in commercial pig populations. The identified mutations and associated genetic tests can be used to address both of these issues.
Collapse
Affiliation(s)
- Emily H Waide
- Department of Animal Sciences, Iowa State University, Ames, IA 50011
| | - Jack C M Dekkers
- Department of Animal Sciences, Iowa State University, Ames, IA 50011
| | - Jason W Ross
- Department of Animal Sciences, Iowa State University, Ames, IA 50011
| | - Raymond R R Rowland
- College of Veterinary Medicine, Kansas State University, Manhattan, KS 66502; and
| | - Carol R Wyatt
- College of Veterinary Medicine, Kansas State University, Manhattan, KS 66502; and
| | - Catherine L Ewen
- College of Veterinary Medicine, Kansas State University, Manhattan, KS 66502; and
| | - Alyssa B Evans
- Department of Animal Sciences, Iowa State University, Ames, IA 50011
| | - Dinesh M Thekkoot
- Department of Animal Sciences, Iowa State University, Ames, IA 50011
| | | | - Nick V L Serão
- Department of Animal Sciences, Iowa State University, Ames, IA 50011
| | | | | |
Collapse
|
16
|
Diverging biological roles among human monocyte subsets in the context of tuberculosis infection. Clin Sci (Lond) 2015; 129:319-30. [PMID: 25858460 DOI: 10.1042/cs20150021] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Circulating monocytes (Mo) play an essential role in the host immune response to chronic infections. We previously demonstrated that CD16(pos) Mo were expanded in TB (tuberculosis) patients, correlated with disease severity and were refractory to dendritic cell differentiation. In the present study, we investigated whether human Mo subsets (CD16(neg) and CD16(pos)) differed in their ability to influence the early inflammatory response against Mycobacterium tuberculosis. We first evaluated the capacity of the Mo subsets to migrate and engage a microbicidal response in vitro. Accordingly, CD16(neg) Mo were more prone to migrate in response to different mycobacteria-derived gradients, were more resistant to M. tuberculosis intracellular growth and produced higher reactive oxygen species than their CD16(pos) counterpart. To assess further the functional dichotomy among the human Mo subsets, we carried out an in vivo analysis by adapting a hybrid mouse model (SCID/Beige, where SCID is severe combined immunodeficient) to transfer each Mo subset, track their migratory fate during M. tuberculosis infection, and determine their impact on the host immune response. In M. tuberculosis-infected mice, the adoptively transferred CD16(neg) Mo displayed a higher lung migration index, induced a stronger pulmonary infiltration of murine leucocytes expressing pro- and anti-inflammatory cytokines, and significantly decreased the bacterial burden, in comparison with CD16(pos) Mo. Collectively, our results indicate that human Mo subsets display divergent biological roles in the context of M. tuberculosis infection, a scenario in which CD16(neg) Mo may contribute to the anti-mycobacterial immune response, whereas CD16(pos) Mo might promote microbial resilience, shedding light on a key aspect of the physiopathology of TB disease.
Collapse
|
17
|
Abstract
During the past decade, the development of humanized mouse models and their general applications in biomedical research greatly accelerated the translation of outcomes obtained from basic research into potential diagnostic and therapeutic strategies in clinic. In this chapter, we firstly present an overview on the history and current progress of diverse humanized mouse models and then focus on those equipped with reconstituted human immune system. The update advancement in the establishment of humanized immune system mice and their applications in the studies of the development of human immune system and the pathogenesis of multiple human immune-related diseases are intensively reviewed here, while the shortcoming and perspective of these potent tools are discussed as well. As a valuable bridge across the gap between bench work and clinical trial, progressive humanized mouse models will undoubtedly continue to play an indispensable role in the wide area of biomedical research.
Collapse
|
18
|
Koboziev I, Jones-Hall Y, Valentine JF, Webb CR, Furr KL, Grisham MB. Use of Humanized Mice to Study the Pathogenesis of Autoimmune and Inflammatory Diseases. Inflamm Bowel Dis 2015; 21:1652-73. [PMID: 26035036 PMCID: PMC4466023 DOI: 10.1097/mib.0000000000000446] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Animal models of disease have been used extensively by the research community for the past several decades to better understand the pathogenesis of different diseases and assess the efficacy and toxicity of different therapeutic agents. Retrospective analyses of numerous preclinical intervention studies using mouse models of acute and chronic inflammatory diseases reveal a generalized failure to translate promising interventions or therapeutics into clinically effective treatments in patients. Although several possible reasons have been suggested to account for this generalized failure to translate therapeutic efficacy from the laboratory bench to the patient's bedside, it is becoming increasingly apparent that the mouse immune system is substantially different from the human. Indeed, it is well known that >80 major differences exist between mouse and human immunology; all of which contribute to significant differences in immune system development, activation, and responses to challenges in innate and adaptive immunity. This inconvenient reality has prompted investigators to attempt to humanize the mouse immune system to address important human-specific questions that are impossible to study in patients. The successful long-term engraftment of human hematolymphoid cells in mice would provide investigators with a relatively inexpensive small animal model to study clinically relevant mechanisms and facilitate the evaluation of human-specific therapies in vivo. The discovery that targeted mutation of the IL-2 receptor common gamma chain in lymphopenic mice allows for the long-term engraftment of functional human immune cells has advanced greatly our ability to humanize the mouse immune system. The objective of this review is to present a brief overview of the recent advances that have been made in the development and use of humanized mice with special emphasis on autoimmune and chronic inflammatory diseases. In addition, we discuss the use of these unique mouse models to define the human-specific immunopathological mechanisms responsible for the induction and perpetuation of chronic gut inflammation.
Collapse
Affiliation(s)
- Iurii Koboziev
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Yava Jones-Hall
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, IN 47907-2027
| | - John F. Valentine
- Department of Internal Medicine, Gastroenterology, Hepatology and Nutrition, University of Utah, Salt Lake City, UT 84132-2410
| | - Cynthia Reinoso Webb
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Kathryn L. Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Matthew B. Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| |
Collapse
|
19
|
Chang DK, Moniz RJ, Xu Z, Sun J, Signoretti S, Zhu Q, Marasco WA. Human anti-CAIX antibodies mediate immune cell inhibition of renal cell carcinoma in vitro and in a humanized mouse model in vivo. Mol Cancer 2015; 14:119. [PMID: 26062742 PMCID: PMC4464115 DOI: 10.1186/s12943-015-0384-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/11/2015] [Indexed: 12/17/2022] Open
Abstract
Background Carbonic anhydrase (CA) IX is a surface-expressed protein that is upregulated by the hypoxia inducible factor (HIF) and represents a prototypic tumor-associated antigen that is overexpressed on renal cell carcinoma (RCC). Therapeutic approaches targeting CAIX have focused on the development of CAIX inhibitors and specific immunotherapies including monoclonal antibodies (mAbs). However, current in vivo mouse models used to characterize the anti-tumor properties of fully human anti-CAIX mAbs have significant limitations since the role of human effector cells in tumor cell killing in vivo is not directly evaluated. Methods The role of human anti-CAIX mAbs on CAIX+ RCC tumor cell killing by immunocytes or complement was tested in vitro by antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) and antibody-dependent cellular phagocytosis (ADCP) as well as on CAIX+ RCC cellular motility, wound healing, migration and proliferation. The in vivo therapeutic activity mediated by anti-CAIX mAbs was determined by using a novel orthotopic RCC xenograft humanized animal model and analyzed by histology and FACS staining. Results Our studies demonstrate the capacity of human anti-CAIX mAbs that inhibit CA enzymatic activity to result in immune-mediated killing of RCC, including nature killer (NK) cell-mediated ADCC, CDC, and macrophage-mediated ADCP. The killing activity correlated positively with the level of CAIX expression on RCC tumor cell lines. In addition, Fc engineering of anti-CAIX mAbs was shown to enhance the ADCC activity against RCC. We also demonstrate that these anti-CAIX mAbs inhibit migration of RCC cells in vitro. Finally, through the implementation of a novel orthotopic RCC model utilizing allogeneic human peripheral blood mononuclear cells in NOD/SCID/IL2Rγ−/− mice, we show that anti-CAIX mAbs are capable of mediating human immune response in vivo including tumor infiltration of NK cells and activation of T cells, resulting in inhibition of CAIX+ tumor growth. Conclusions Our findings demonstrate that these novel human anti-CAIX mAbs have therapeutic potential in the unmet medical need of targeted killing of HIF-driven CAIX+RCC. The orthotopic tumor xenografted humanized mouse provides an improved model to evaluate the in vivo anti-tumor capabilities of fully human mAbs for RCC therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0384-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- De-Kuan Chang
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| | - Raymond J Moniz
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| | - Zhongyao Xu
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA.
| | - Jiusong Sun
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Quan Zhu
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| | - Wayne A Marasco
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Experimental models have contributed enormously to basic immunology. However, the use of reductionist experiments has produced results that are not always successfully translated into the clinic. Recently, incorporation of more realistic clinical parameters in experimental designs has produced new insights relevant to cardiac transplantation. RECENT FINDINGS Experiments in mice have provided crucial insights into the concept that T cell responses to pathogens generate memory cells with cross-reactive specificities for histocompatibility antigens. These memory T cells are resistant to current immunosuppressive strategies. Memory T cells infiltrate grafts within hours after transplantation, and grafts subjected to clinically relevant periods of cold ischemia are more susceptible to injury by this cellular infiltrate. Early immune responses now can be investigated with improved 'humanized' mice. Mice with multiple knock-in genes for human cytokines support development of human monocytes, macrophages and natural killer cells in increased numbers and with better function. SUMMARY Better and more clinically relevant experimental designs are providing animal models tailored to address clinic exigencies.
Collapse
|
21
|
Blank G, Welker C, Haarer J, Sterk M, Nadalin S, Yañez VAC, Joos TO, Menrad A, Snell D, LaCorcia G, Königsrainer A, Handgretinger R, Schilbach K. Selective, efficient modulation of activated CD4+ αβT cells by the novel humanized antibody GZ-αβTCR targeting human αβTCR. Bone Marrow Transplant 2014; 50:390-401. [DOI: 10.1038/bmt.2014.263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 10/02/2014] [Accepted: 10/04/2014] [Indexed: 11/09/2022]
|
22
|
Heterelogous expression of mutated HLA-G decreases immunogenicity of human embryonic stem cells and their epidermal derivatives. Stem Cell Res 2014; 13:342-54. [PMID: 25218797 DOI: 10.1016/j.scr.2014.08.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/29/2014] [Accepted: 08/08/2014] [Indexed: 12/28/2022] Open
Abstract
Human embryonic stem cells (hESCs) are capable of extensive self-renewal and expansion and can differentiate into any somatic tissue, making them useful for regenerative medicine applications. Allogeneic transplantation of hESC-derived tissues from results in immunological rejection absent adjunctive immunosuppression. The goal of our study was to generate a universal pluripotent stem cell source by nucleofecting a mutated human leukocyte antigen G (mHLA-G) gene into hESCs using the PiggyBac transposon. We successfully generated stable mHLA-G(EF1α)-hESC lines using chEF1α promoter system that stably expressed mHLA-G protein during prolonged undifferentiated proliferation andin differentiated embryoid bodies as well as teratomas. Morphology, karyotype, and telomerase activity of mHLA-G expressing hESC were normal. Immunofluorescence staining and flow cytometry analysis revealed persistent expression of pluripotent markers, OCT-3/4 and SSEA-4, in undifferentiated mHLA-G(EF1α)-hESC. Nucleofected hESC formed teratomas and when directed to differentiate into epidermal precursors, expressed high levels of mHLA-G and keratinocyte markers K14 and CD29. Natural killer cell cytotoxicity assays demonstrated a significant decrease in lysis of mHLA-G(EF1a)-hESC targets relative to control cells. Similar results were obtained with mHLA-G(EF1α)-hESC-derived epidermal progenitors (hEEP). One way mixed T lymphocyte reactions unveiled that mHLA-G(EF1a)-hESC and -hEEP restrained the proliferative activity of mixed T lymphocytes. We conclude that heterologous expression of mHLA-G decreases immunogenicity of hESCs and their epidermal differentiated derivatives.
Collapse
|
23
|
Li F, Cowley DO, Banner D, Holle E, Zhang L, Su L. Efficient genetic manipulation of the NOD-Rag1-/-IL2RgammaC-null mouse by combining in vitro fertilization and CRISPR/Cas9 technology. Sci Rep 2014; 4:5290. [PMID: 24936832 PMCID: PMC4894429 DOI: 10.1038/srep05290] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 05/27/2014] [Indexed: 01/20/2023] Open
Abstract
Humanized mouse models have become increasingly important and widely used in modeling human diseases in biomedical research. Immunodeficient mice such as NOD-Rag1-/-IL2RgammaC-null (NRG) or NOD-SCID-IL2RgammaC-null (NSG) mice are critical for efficient engraftment of human cells or tissues. However, their genetic modification remains challenging due to a lack of embryonic stem cells and difficulty in the collection of timed embryos after superovulation. Here, we report the generation of gene knockout NRG mice by combining in vitro fertilization (IVF) and CRISPR/Cas9 technology. Sufficient numbers of fertilized embryos were produced through IVF, and a high rate of Fah gene targeting was achieved with microinjection of Cas9 mRNA, gRNA and single strand oligonucleotide DNA (ssDNA) into the embryos. The technology paves the way to construct NRG or NSG mutant mice to facilitate new humanized mouse models. The technology can also be readily adapted to introduce mutations in other species such as swine and non-human primates.
Collapse
Affiliation(s)
- Feng Li
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dale O. Cowley
- Animal Models Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- TransViragen, Inc., Research Triangle Park, NC 27709, USA
| | - Debra Banner
- TransViragen, Inc., Research Triangle Park, NC 27709, USA
| | - Eric Holle
- Animal Models Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Liguo Zhang
- Key Lab of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Lishan Su
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Key Lab of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Department of translational medicine, Department of surgery, Department of medicine, the first hospital, Jilin University, Changchun 130061, China
| |
Collapse
|
24
|
Hogenes M, Huibers M, Kroone C, de Weger R. Humanized mouse models in transplantation research. Transplant Rev (Orlando) 2014; 28:103-10. [PMID: 24636846 DOI: 10.1016/j.trre.2014.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/09/2014] [Indexed: 11/30/2022]
Abstract
The interest in the use of humanized mouse models for research topics like Graft versus Host Disease (GvHD), allograft studies and other studies to the human immune system is growing. The design of these models is still improving and enables even more complicated studies to these topics. For researchers it can be difficult to choose the best option from the current pool of available models. The decision will depend on which hypothesis needs to be tested, in which field of interest, and therefore 'the best model' will differ from one to another. In this review, we provide a guide to the most common available humanized mouse models, with regards to different mouse strains, transplantation material, transplantation techniques, pre- and post-conditioning and references to advantages and disadvantages. Also, an evaluation of experiences with humanized mouse models in studies on GvHD and allograft rejection is provided.
Collapse
Affiliation(s)
- Marieke Hogenes
- Department of Pathology, University Medical Centre Utrecht, PO box 85500, 3508 GA Utrecht, The Netherlands
| | - Manon Huibers
- Department of Pathology, University Medical Centre Utrecht, PO box 85500, 3508 GA Utrecht, The Netherlands
| | - Chantal Kroone
- Department of Pathology, University Medical Centre Utrecht, PO box 85500, 3508 GA Utrecht, The Netherlands
| | - Roel de Weger
- Department of Pathology, University Medical Centre Utrecht, PO box 85500, 3508 GA Utrecht, The Netherlands.
| |
Collapse
|
25
|
Cai YJ, Huang L, Leung TY, Burd A. A study of the immune properties of human umbilical cord lining epithelial cells. Cytotherapy 2013; 16:631-9. [PMID: 24364910 DOI: 10.1016/j.jcyt.2013.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 10/11/2013] [Accepted: 10/23/2013] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS Our previous study has demonstrated the stem cell-like properties of human umbilical cord lining epithelial cells (CLECs) and their capability for epidermal reconstitution in organotypic skin culture; however, the immunogenicity of these cells has not been clearly defined. We assessed several aspects of the immune properties of CLECs in vitro. METHODS We examined CLECs for their immunoregulatory function in a mixed lymphocyte culture experiment. We characterized the expression patterns of the major histocompatibility complex (MHC), co-stimulatory molecules and the pro-/anti-inflammatory cytokines and growth factors in CLECs by means of reverse transcription-polymerase chain reaction, Western blotting, flow cytometry and FlowCytomix multiple analyte detection assays. RESULTS CLECs were found not to induce but to suppress the proliferation response of the peripheral blood mononuclear cells in a mixed lymphocyte culture assay. They did not express the MHC class II antigen HLA-DR but the non-classic MHC class I antigens HLA-G and HLA-E and lacked the expression of the co-stimulatory molecules CD40, CD80 and CD86. In addition, they produced less interleukin-1β and transforming growth factor-β1 but more interleukin-4 and hepatocyte growth factor than did adult keratinocytes, a pattern in favor of wound healing with less inflammation response. CONCLUSIONS Our data suggest that CLECs have an immunosuppressive function in addition to their low immunogenicity. This could be at least partially explained by their expression of HLA-G and HLA-E molecules associated with immune tolerance and absence of HLA-DR and co-stimulatory molecules. The demonstration that CLECs produce a favorable pattern of cytokines and growth factors for wound healing provides further support for their potential clinical application in allogeneic cell therapy.
Collapse
Affiliation(s)
- Yi-Jun Cai
- Division of Plastic, Reconstructive and Aesthetic Surgery, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, SAR
| | - Lin Huang
- Division of Plastic, Reconstructive and Aesthetic Surgery, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, SAR
| | - Tak-Yeung Leung
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR
| | - Andrew Burd
- Division of Plastic, Reconstructive and Aesthetic Surgery, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, SAR.
| |
Collapse
|